Volume XX | Supplement X
Berlin, Germany
June 1-4, 2024
© European Society of Human Genetics 2024
The ESHG conference delivered the latest findings in the field of human genetics, both basic and applied.
Additional information about the event may be found on the conference website:
Sponsorship: Publication of this supplement was sponsored by the European Society of Human Genetics. All content was reviewed and approved by the ESHG Scientific Programme Committee, which held full responsibility for the abstract selections.
Disclosure Information: In order to help readers, form their own judgments of potential bias in published abstracts, authors are asked to declare any competing financial interests.
Contributions of up to EUR 10 000.- (Ten thousand Euros, or equivalent value in kind) per year per company are considered “Modest”. Contributions above EUR 10 000.- per year are considered “Significant”.
Presenting author names are underlined in the contributor lists.
Plenary Sessions PL1 ELPAG Award Lecture
PL1.1 ESHG Award Lecture
Angus Clarke
As is always true, my career has been shaped by circumstances; in my case, these included the constraints of the 1980s molecular genetics lab. My involvement with genetics research began through exposure to four key conditions: (i) Xp21 (Duchenne and Becker) muscular dystrophy; (ii) X-linked hypohidrotic ectodermal dysplasia (XHED); (iii) Rett syndrome, and (iv) Huntington’s disease (HD), which (along with myotonic dystrophy) was the air we breathed in Cardiff in the age of gene mapping and cloning as it was pursued within the framework of a long-term relationship with each family and with a commitment to their social and medical welfare, enabled by a multidisciplinary genetic counselling team. I must mention Professor Peter Harper, and also Professor John Burn, as key professional influences, who provided great support and guidance.
I had already been involved with Duchenne muscular dystrophy through work in the Cardiff Muscle Clinic, in my earlier role in paediatrics. This led to an interest in newborn screening for Duchenne, a social science evaluation of its impact on families, and an interest in consent. Work with Rett syndrome led to a search for other families with more than one affected person, and that led to a close association with the UK family support group, a focus on the needs of those affected by rare diseases, and how to provide effective support without causing damage from the ‘hype’ of inflated claims for new treatments. My work with HD began more recently, when I took over the Cardiff HD predictive testing clinic from Peter Harper and then took part in projects with the UK HD predictive testing consortium, with its strong interest in genetic counselling.
However, it is perhaps my work with XHED that best illustrates my developing interests in the ethical and social aspects of human genetics. In my project on XHED, from summer 1986 to the end of 1987, I acquired: (i) blood samples from many family members across the UK for a linkage study; (ii) phenotypic information about many patients; (iii) in-depth knowledge of the motorways and housing estates of mainland Britain; and (iv) I heard the stories of many members of XHED families. This contributed to mapping the EDA gene, identifying the spectrum of pathogenic variants, and to my returning some years later with an audio recorder to listen to the stories more systematically. These studies led me to an interest in stigmatisation and how the effects of this on family life are often shaped by the mode of inheritance of the condition.
With others - Heather Skirton, Clara Gaff, Marion McAllister and Nicki Taverner - I have worked to promote a model of genetic counselling practice and to develop the Cardiff University MSc course in Genetic (and Genomic) Counselling.
Clinical genetics trainees today face different circumstances from the 1980s but i would encourage them to follow the new paths of engagement with disease support groups, the genetic counselling process, and the ethical and social issues that emerge in the context of human genetics research and service provision. Be enthusiastic in developing treatments, where feasible, but be scrupulously honest about the limitations of what is achievable. Avoid hype, raising expectations beyond what is realistic, to avoid the destructiveness of dashed hopes.
PL2 Opening Plenary
PL2.1 Advancing biology through single cell phenomics
Bart Deplancke
In my presentation, I will discuss recent advancements from our laboratory in single cell phenomic analysis technologies. These include: 1) IRIS for ‘Interconnecting a Robotic Image of a cell to its ScRNA-seq profile’. This is a novel, so far unpublished technology that pairs the transcriptomic fingerprint of a single cell with its high-resolution image. I will present proof-of-concept examples showhing how IRIS provides an unprecedented opportunity to i) leverage machine learning approaches to uncover the molecular determinants underlying single cell phenotypes and vice versa, contributing to an integrated view of cellular function and structure, and ii) transform clinical practices through paired single cell imaging and transcriptomics; 2) Live-seq: a methodology for extracting single cell transcriptomes without compromising cell viability, allowing temporal analyses of cellular responses and bridging the gap between transcriptomic profiling and functional behavior over time; and 3) (if time permits) scTF-seq: an approach to quantify transcriptomic changes based on transcription factor dose in single cells, providing insights into cell fate determination and the underlying heterogeneity in cellular reprogramming. These breakthroughs significantly enhance our ability to unravel complex biological processes at the single-cell level with precision and temporal resolution. As such, they may have broad implications for gene regulation, cellular reprogramming, and developmental biology, opening new avenues for research and therapeutic strategies.
PL3 What’s new? Highlight session
PL3.1 Zero Childhood Cancer National Precision Medicine Program: Improving outcomes for children with high risk cancer cancer utilising comprehensive, integrated multiomic profiling
Vanessa Tyrrell 1;2, Chelsea Mayoh2;3, Paul Ekert2;3;4, Mark Cowley2;3, David Ziegler2;3;5, Marie Wong2;3, Paulette Barahona2, Mark Pinese2;3, Michelle Haber2;3, Rob Salomon2, Eliza Courtney2;5, Richard Lock2;3, Noemi Fuentes-Bolanos2;3;5, Glenn Marshall2;3;5, Meera Warby5, Toby Trahair2;3;5, Dong Anh Khuong Quang6;7, Claire Wakefield3, Katherine Tucker3;5;8, Kristine Barlow-Stewart2;9, Bhavna Padhye10, Loretta Lau2;3;5
1UNSW Sydney, School of Clinical Medicine, UNSW Medicine and Health, Sydney, Australia; 2Children’s Cancer Institute, Randwick, Australia; 3UNSW Sydney, School of Clinical Medicine, UNSW Medicine & Health, Sydney, Australia; 4Peter MacCallum Cancer Centre, Melbourne, Australia; 5Sydney Children’s Hospital, Randwick, Kids Cancer Centre, Randwick, Australia; 6The Royal Children’s Hospital Melbourne, Children’s Cancer Centre, Parkville, Australia; 7Murdoch Children’s Research Institute, Parkville, Australia; 8Prince Of Wales Hospital, Hereditary Cancer Clinic, Randwick, Australia; 9University of Sydney School of Medicine, Camperdown, Australia; 10The Children’s Hospital at Westmead, Cancer Centre for Children, Westmead, Australia
Consortium: Zero Childhood Cancer National Precision Medicine Program
Background/Objectives: Precision medicine is revolutionising the way cancers are detected, characterised, and treated. The Zero Childhood Cancer program (ZERO) represents one of the world’s largest and most comprehensive high-risk (<30% survival) paediatric cancer cohorts, employing integrated multiomic profiling to identify precision guided treatment (PGT) approaches, to improve outcomes and minimise long-term side effects in survivors.
Methods: ZERO performs integrated somatic whole genome, whole transcriptome and methylation profiling, and germline whole genome analyses, to identify genomic aberrations, and determine potential genomically guided treatment options for each child. These are discussed at a national multidisciplinary tumour board, and a clinical research report is issued to treating clinicians.
Results: Since September 2017, over 1500 children with cancer have been enrolled. In the first 250 patients with high-risk cancer, the molecular basis for the cancer was identified in 94% of patients, with 70% being potentially actionable (Wong et al, Nature Medicine, 2020). Recent analyses of 384 consecutively enrolled high-risk child cancer patients, with >18mths of follow up, demonstrated that those receiving a precision-guided treatment, informed by comprehensive molecular profiling, led to improved two year progression free survival when compared to unguided novel therapies or standard-of-care cytotoxic therapies (27% vs 11%).
Conclusion: These findings demonstrate the power of multiomic profiling to improve outcomes for children with high-risk cancer. ZERO is now open to all Australian children diagnosed with cancer, aiming to demonstrate utility of this multiomic precision medicine model, irrespective of type, stage, or risk.
Grants: Medical Research Future Fund (Australia), Minderoo Foundation
Conflict of Interest: None declared
PL3.2 All by All of Us: common and rare variant association testing in 250,000 whole genomes across diverse ancestry groups
Konrad Karczewski 1;2, Wenhan Lu2, Robert Carroll3, Ying Wang1;2, Riley Grant2, Matthew Solomonson2, Megan He3, Michael Lyons3, Wei Zhou1;2, Anjene Musick4, Daniel King2, Alicia Martin1;2, Dan Roden3, Benjamin Neale1;2
1Massachusetts General Hospital, Boston, United States; 2Broad Institute, Cambridge, United States; 3Vanderbilt University Medical Center, Nashville, United States; 4National Institutes of Health, Bethesda, United States
Background/Objectives: Whole genome sequencing of population biobanks provide opportunities for comprehensive phenome- and genome-wide (“all x all”) association analyses, including rare burden and common variant testing of coding and non-coding variation, across many human phenotypes. Additionally, the increased diversity of the All of Us cohort, in which 80% of the 245,400 participants are underrepresented in genomic research, enhances gene discovery, and multi-phenotype analysis identifies pleiotropy relevant to human disease.
Methods: We built a comprehensive multi-ancestry phenome- and genome-wide analysis framework, using a generalized mixed model framework to perform common variant association tests and rare variant burden tests, followed by meta-analysis.
Results: We applied this framework to 3,348 anthropometric, biomarker, disease, and medication phenotypes derived from electronic health records and surveys across 6 genetic ancestry groups for each quantitative trait and binary trait with over 200 cases. We find thousands of associations for rare variant burden analysis and common variant associations enabled by this diverse cohort. We show consistency with previous efforts such as UK Biobank, and demonstrate how multi-ancestry approaches improve gene discovery.
Conclusion: We present a roadmap for iterative releases of the data up to 1 million individuals that will empower more well-powered association tests for diseases with less than 1% prevalence. We release the full dataset of summary statistics to the public, via bulk data downloads as well as an interactive public-facing browser.
Grants: NIH grants OT2OD035404 and OT2OD002750
Conflict of Interest: Konrad Karczewski Tome, Vor, Nurture Genomics, Wenhan Lu: None declared, Robert Carroll: None declared, Ying Wang: None declared, Riley Grant: None declared, Matthew Solomonson: None declared, Megan He: None declared, Michael Lyons: None declared, Wei Zhou: None declared, Anjene Musick: None declared, Daniel King: None declared, Alicia Martin: None declared, Dan Roden: None declared, Benjamin Neale Deep Genomics, Neumora
PL3.3 A phenome-wide association study of methylated GC-rich repeats identifies a GCC repeat expansion in AFF3 as a significant cause of intellectual disability
Bharati Jadhav1, Paras Garg1, Joke van Vugt2, Kristina Ibañez3, Delia Gagliardi3;4, William Lee1, Mariya Shadrina1, Tom Mokveld5, Egor Dolzhenko5, Alejandro Martin Trujillo1, Scott Gies1, Mafalda Barbosa1, Miten Jain6, Henry Houlden4, Benedict Paten6, Jan Veldink2, Arianna Tucci3, Andrew Sharp 1
1Icahn School of Medicine at Mount Sinai, Department of Genetics and Genomic Sciences and Mindich Child Health and Development Institute, New York, United States; 2Utrecht University, Department of Neurology, UMC Utrecht Brain Center, Utrecht, Netherlands; 3Queen Mary University of London, London, United Kingdom; 4University College London, Department of Neuromuscular Diseases, London, United Kingdom; 5Pacific Biosciences, Menlo Park, United States; 6University of California, Santa Cruz, United States
Consortium: Genomics England Research Consortium, Project MinE ALS Sequencing Consortium
GC-rich tandem repeat expansions (TREs) are often associated with DNA hypermethylation, gene silencing, and folate-sensitive fragile sites and underlie several congenital and late-onset disorders, including fragile X and ALS. Through a combination of DNA methylation profiling and tandem repeat genotyping, we identified 24 methylated TREs. We investigated their effects on human traits using PheWAS in 168,641 individuals from the UK Biobank, identifying 156 significant TRE:trait associations (10% FDR) involving 17 TREs. Of these, a GCC expansion in the promoter of AFF3 was linked with a 2.4-fold reduced probability of completing secondary education, an effect size comparable to several recurrent pathogenic microdeletions. Given its strong negative influence on educational attainment, we hypothesized that this AFF3 expansion might represent the pathogenic mutation in some patients with neurodevelopmental disorders. In support of this, in a cohort of 6,371 probands with neurodevelopmental problems of unknown etiology from the 100,000 Genomes Project, we observed a significant enrichment of AFF3 expansions compared to controls. Finally, we identified SNVs tagging AFF3 expansion; many of these are the same SNVs reported at this locus in prior GWAS for educational attainment. Our observations, therefore, indicate that a subset of GWAS signals in the genome are likely driven by underlying TREs that preferentially occur on specific founder haplotypes. With a population prevalence that is at least 5-fold higher than the TRE causing fragile X syndrome and 3- to 8-fold higher than recurrent microdeletions of 1q21.1, 15q13.3, and 16p11.2, AFF3 expansions therefore represent a significant cause of neurodevelopmental delay.
Conflict of Interest: None declared
PL3.4 In-silico prioritization of open chromatin sequences increases the detection of activating variants
Kilian Salomon 1, Max Schubach1, Mohan Dash1, Chengyu Deng2, Nadav Ahituv2, Jay Shendure3, Martin Kircher1
1Berlin Institute of Health at Charité (BIH), Computational Genome Biology, Berlin, Germany; 2University of California San Francisco, Department of Bioengineering and Therapeutic Sciences, San Francisco, United States; 3University of Washington, Department of Genome Sciences, Seattle, United States
Consortium: NIH Impact of Genomic Variation on Function consortium (IGVF)
Background/Objectives: A substantial reservoir of variants associated with pathological phenotypes resides in non-coding sequences, especially proximal and distal regulatory sequences. As part of the NIH Impact of Genomic Variation on Function (IGVF) consortium, we investigate functional consequences of variation using Massively Parallel Reporter Assays (MPRAs).
Methods: For this purpose, we tested over 28,000 candidate cis-regulatory regions (cCREs) from the proximity (50kb) of 526 genes associated with neurological, cardiac or clinically actionable diseases as well as a random gene set. Within these cCREs, we included >46,000 variants from gnomAD with an allele frequency (AF) of at least 1% and samples of lower frequency variants. Specifically, we ranked sequences based on the Enformer sequence model (Avsec Ž et al. 2021) to select 70% potentially activating, 15% repressing, and 15% random variants.
Results: We measured variant and cCRE expression in neural progenitor cells derived from WTC11. In an initial analysis, we observed 150 significant variant effects, including 81 activating effects. This enrichment of expression-activating variants is only present in the computationally prioritized set, but not in variants above the AF > 1% threshold.
Conclusion: Previous work of saturation mutagenesis MPRAs showed that it is more likely to observe variants with negative effects (Kircher M et al. 2019). Our large set of significant activating variant effects implies that Enformer prioritized these effects successfully. Further investigations are required to test the robustness of in-silico tools to support MPRA designs or whether rare variants more frequently introduce activating binding sites.
Grants: IGVF (1UM1HG011966)
Conflict of Interest: None declared
PL3.5 Understanding the genetic complexity of puberty timing across the allele frequency spectrum.
Katherine Kentistou1
1MRC Epidemiology Unit, University of Cambridge, School of Clinical Medicine, Cambridge, United Kingdom
Consortium: the ReproGen Consortium
Background/Objectives: Pubertal onset is highly polygenic and has been linked to later life disease, including obesity, type 2 diabetes, and hormone-sensitive cancers.
Methods: We performed a GWAS meta-analysis of age at menarche including data from 44 cohorts of European and East-Asian ancestry (n ~ 800,000) and rare exome variant associations (n∼220,000). We developed a GWAS causal-gene identification method, based on omics data and enhancer maps.
Results: Our multi-ancestry GWAS identified 1,080 associations with age at menarche. Women at the top/bottom centiles of polygenic risk exhibited a ∼11/∼14-fold higher risk of delayed/precocious puberty. We identified 665 high-confidence menarche genes supported by multiple functional predictors. These were enriched for genes underlying the embryonic migration of GnRH neurons in RNAseq data. We identified an uncharacterized G-protein coupled receptor, which we demonstrate can amplify canonical MC3R signaling in vitro and genetically. Using data from a longitudinal growth cohort we showed that 45% of the menarche signals exert a primary effect on childhood growth. We identified 6 genes harbouring rare loss-of-function variants that associate with menarche, three of which were novel; KDM4C, PDE10A, and ZNF483. The impact of polygenic risk was abolished by rare variants in ZNF483, which affected the protein’s zinc finger domains and thus its ability to correctly bind to its downstream targets.
Conclusion: These findings extend our understanding of the biological complexity of pubertal timing, highlight body size dependent and independent mechanisms, and demonstrate how common genetic variants can influence the clinical extremes of pubertal development.
Grants: Medical Research Council, MC_UU_12015/2.
Conflict of Interest: None declared
PL3.6 Decoding ALG13-CDG: multi-omics profiling of ALG13-CDG brain organoids reveals distinct glycosylation defects impacting axon growth, neuronal migration, synaptic plasticity, and seizure susceptibility.
Rameen Shah1, Silvia Radenkovic2, Rohit Budhraja3, Graeme Preston1, Akhilesh Pandey3, Steven Sloan4, Eva Morava1, Tamas Kozicz 1
1, Genetics and Genomic Sciences, New York, United States; 2, Clinical Genomics, Rochester, United States; 3, Department of Laboratory Medicine and Pathology, Rochester, United States; 4, Human Genetics, Atlanta, United States
Background/Objectives: Asparagine-linked glycosylation 13 (ALG13) is an X-linked congenital disorder of glycosylation (CDG). Interestingly, unlike most other N-linked CDGs, investigations on ALG13-CDG patients’ blood and fibroblast samples have not revealed defects in glycosylation. ALG13-CDG presents with neurological symptoms such as seizures, intellectual disability, central hypotonia, and developmental delays. These observations have led us to hypothesize that ALG13-CDG might primarily affect the brain.
Methods: To test this hypothesis, we developed a brain model of ALG13-CDG by generating induced pluripotent stem cells from fibroblasts and differentiating them into cortical brain organoids (BOs). At 77-92 days of differentiation, we conducted comprehensive multiomics profiling on these BOs, including single-cell RNA-seq, proteomics, glycoproteomic, and metabolomics analyses.
Results: Our investigation unveiled a significant reduction in the glycosylation of several proteins crucial for axon growth, neuronal migration, and synaptic plasticity. We observed a parallel reduction in glycosylation for proteins implicated in seizures due to lipid accumulation and disturbances in calcium transport. Our proteomics studies revealed alterations in the expression of genes associated with neuronal migration and nucleotide synthesis. By employing single-cell RNA-seq, we elucidated distinct changes in gene expression among GABAergic neurons, glutamatergic neurons, and radial glia, with significant changes in genes important for neuronal migration, axon guidance, calcium ion concentration, and lipid metabolism. Our metabolomics investigations highlighted an elevation in GlcNAc levels and downregulation in metabolites associated with nucleotide synthesis.
Conclusion: These multi-omics studies on ALG13-deficient BOs provide valuable insights into specific brain-related disturbances inherent to ALG13-CDG and offer promising avenues for potential therapeutic interventions.
Conflict of Interest: None declared
Concurrent Sessions C01 Genetics and Multiomics
C01.1 Identification of microbiota components correlated with host lifestyle, molecular, biochemical, immunophenotypic measurements and genotype in a deeply phenotyped Sardinian cohort
Maria Antonietta Diana 1;2, Maristella Pitzalis1, Marco Masala1, Andrea Maschio1, Fabio Busonero1, Monia Lobina1, Marcella Devoto1;3, Myriam Gorospe4, David Schlessinger4, Valeria Orrù1, Edoardo Fiorillo1, Federico Santoni1;5, Francesco Cucca1;2, Mauro Pala1
1C.N.R. - Istituto di Ricerca Genetica e Biomedica, Monserrato, Italy; 2University of Sassari, Scienze Biomediche, Sassari, Italy; 3Sapienza University of Rome, Medicina Traslazione e di Precisione, Roma, Italy; 4National Institute On Aging, Baltimore, United States; 5Lausanne University Hospital, Lausanne, Switzerland
Background/Objectives: Existing literature already explored the association between microbiota and host traits, but suffers from limitations such as small sample sizes, few covariates and reliance on non-whole-genome methods. We aim to overcome these by analyzing metagenomic data in a large cohort. Our goal is to correlate microbiota with diverse host measurements, including genetics, lifestyle, and disease markers.
Methods: We leverage whole-genome shotgun sequencing of fecal microbiota from 2,700 ProgeNIA cohort volunteers where high-resolution genotypes and phenotypes were available from previous projects. Measured phenotypes include blood markers, anthropometric traits, disease status, lifestyle factors (e.g., alcohol consumption, coffee intake, smoking), immunophenotypic data, leukocyte transcriptional profiling, and high-resolution genetic characterization.
Results: We have identified more than 100 taxonomic levels displaying significant correlations with factors such as alcohol consumption, smoking, Proton Pump Inhibitors (PPI), BMI, anti-diabetics intake, cancer, diabetes, glycemia, sex and age. We conducted a Quantitative Trait Loci (QTL) GWAS on 5566 taxa using REGENIE obtaining 330 taxa with at least one significant association with the phenotypic traits. 54 of those associations colocalize with sentinel variants linked to other traits and diseases such as Lymphocyte percentage of white cells, Diastolic blood pressure, Gamma glutamyl transferase levels, Asthma and Lung adenocarcinoma among others. Causality has been inspected with one and two-steps Mendelian randomization methods.
Conclusion: Our study, empowered by a significant sample size and broad phenotyping, enabled the discovery of novel causal interactions between specific taxa and several factors increasing our knowledge about the mutual influence of microbiota on human health and disease.
Conflict of Interest: None declared
C01.2 Multi-trait genetic colocalization of urine protein levels provide insights into renal protein handling and related clinical outcomes.
Oleg Borisov 1, Stefan Haug1, Nora Scherer1, Sara Monteiro-Martins1, Yong Li1, Anna Köttgen1
1Institute of Genetic Epidemiology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
Background/Objectives: Genetic association studies of protein levels provide a valuable link between molecular processes and human phenotypes including diseases. We generated urine proteomics data and integrated it with multiomics datasets and phenome-wide association studies to elucidate genetic mechanisms of protein handling in the kidney and its connection to diseases.
Methods: We performed genetic colocalization analyses (“coloc” package) to assess whether urine protein levels measured in the German Chronic Kidney Disease study (N > 1000) share common genetic architecture with orthogonal omics and clinical datasets. We colocalized urine protein genetic associations with multi-tissue transcriptomics, plasma proteomics, and clinical outcome datasets (UK Biobank and FinnGen).
Results: We performed 3.5 million colocalization tests in more than 100 genetic loci and identified 9,000 pairs of traits with strong colocalization evidence (posterior probability H4 > 80%). The majority of loci (>90%) showed evidence of multi-trait colocalization (up to 1000 colocalizing traits per protein). One example was Prostate Stem Cell Antigen (PSCA) – where cis associations (P < 1e-300) with urine PSCA levels colocalized with PSCA expression in kidney tissues and urogenital clinical manifestations, including bladder cancer and urinary tract infections.
Conclusion: We report colocalization results between urine protein associations and phenome-wide association studies including molecular and clinical traits. The generated resource will facilitate a deeper understanding of genetic factors influencing protein handling by the kidney and will help to reveal circulating disease biomarkers which are also informative when quantified in urine.
Grants: This work was supported by the German Research Foundation (DFG) project ID 431984000 (SFB 1453).
Conflict of Interest: None declared
C01.3 Single-cell eQTL mapping in healthy and IBD-afflicted terminal ileum offers novel insights into disease susceptibility
Tobi Alegbe 1, Monika Krzak1, Mennatallah Ghouraba1, Michelle Strickland1, Marcus Tutert1, Bradley Harris1, Matiss Ozols1, Lucia Ramirez-Navarro1, Jason Skelton1, Jasmin Ostermayer1, May Xueqi Hu1, Noor Wana1, Vivek Iyer1, Cristina Cotobal-Martin1, Miles Parkes2, Gareth-Rhys Jones3, Chris Wallace4, Rebecca McIntyre1, Tim Raine2, Carl Anderson1
1Wellcome Sanger Institute, Hinxton, United Kingdom; 2Addenbrooke’s Hospital, United Kingdom; 3University of Edinburgh, United Kingdom; 4MRC Biostatistics Unit, Cambridge, United Kingdom
Background/Objectives: Inflammatory bowel diseases (IBD) are complex polygenic diseases of the gastrointestinal tract with over 320 genetic susceptibility loci associated. For many loci, the associated variants are non-coding making the causal genes and cell types unclear. Expression quantitative trait loci (eQTL) can help link genes to GWAS variants. However, existing efforts have been hampered by the limited resolution afforded by bulk RNA sequencing.
Methods: We generated single-cell RNA-sequencing data from terminal ileal biopsies of 152 non-IBD and 88 Crohn’s-afflicted individuals. Our dataset consisted of over 1 million single-cell transcriptomes, characterising 49 cell types. We performed pseudobulk sc-eQTL mapping finding 6,853 eGenes (genes with an eQTL, FDR < 0.05), 44% of which were undetectable without cell type annotation.
Results: We reproduced eQTL effects from similar studies (GTEX, CEDAR, and OneK1K) but also 15% of our eGenes were only present in our dataset. We were able to map sc-eQTL interactions with our phenotypes such as inflammation finding 253 eGenes with an interaction effect. Colocalisation with CD, UC and IBD GWAS resulted in 22%, 20% and 17% of GWAS loci, respectively, colocalising (PPH4 > 0.75). Amongst these was a novel colocalisation, WRAP73 (PPH4 = 0.97), which has previously been shown to be differentially methylated in IBD.
Conclusion: We demonstrate the value of mapping eQTLs with tissue-derived single-cell data. As our sample size grows, we will gain more insight into IBD biology bringing us a step closer to novel, genetically motivated therapeutics.
Grants: Wellcome Trust [206194 and 108413/A/15/D], The Crohn’s Colitis Foundation [612986 and 997266] and Open Targets [OTAR2057].
Conflict of Interest: Tobi Alegbe: None declared, Monika Krzak: None declared, Mennatallah Ghouraba: None declared, Michelle Strickland: None declared, Marcus Tutert: None declared, Bradley Harris: None declared, Matiss Ozols: None declared, Lucia Ramirez-Navarro: None declared, Jason Skelton: None declared, Jasmin Ostermayer: None declared, May Xueqi Hu: None declared, Noor Wana: None declared, Vivek Iyer: None declared, Cristina Cotobal-Martin: None declared, Miles Parkes: None declared, Gareth-Rhys Jones: None declared, Chris Wallace Part-time employee for GlaxoSmithKline, Rebecca McIntyre Full-time employee of Relation Therapeutics, Tim Raine Has received research/educational grants and/or speaker/consultation fees from Abbvie, Arena, Aslan, AstraZeneca, Boehringer-Ingelheim, BMS, Celgene, Ferring, Galapagos, Gilead, GSK, Heptares, LabGenius, Janssen, Mylan, MSD, Novartis, Pfizer, Sandoz, Takeda and UCB, Carl Anderson Has received consultancy or speaker fees from Genomics plc, BridgeBio and GSK
C01.4 C-STEM: a fast and powerful method for robust trans-eQTL mapping in multi context studies
Lena Krockenberger 1, Mike Thompson2, Noah Zaitlen3;4;5, Xuanyao Liu6, Brunilda Balliu7;8
1University of California, Los Angeles, Bioinformatics Interdepartmental Graduate Program, Los Angeles, United States; 2Centre for Genomic Regulation (CRG), Systems Biology Department, Barcelona, Spain; 3University of California, Los Angeles, Department of Computer Science, Samueli School of Engineering, Los Angeles, United States; 4University of California, Los Angeles, Department of Neurology, David Geffen School of Medicine, Los Angeles, United States; 5University of California, Los Angeles, Department of Computational Medicine, David Geffen School of Medicine, Los Angeles, United States; 6The University of Chicago, Section of Genetic Medicine, Chicago, United States; 7University of California, Los Angeles, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, Los Angeles, United States; 8University of California, Los Angeles, Department of Biostatistics, Fielding School of Public Health, Los Angeles, United States
Background/Objectives: Most disease-associated genetic variants lie outside exons, fueling extensive research into genetic mechanisms governing transcriptional regulation. While progress has been made in identifying cis effects on gene expression, detecting high-quality distal associations (trans-eQTLs) remains challenging. Existing methods for trans-eQTL mapping overlook the inherent intra-individual correlation of gene expression found in multi context studies with repeated sampling. These oversights can significantly diminish the power to detect trans-eQTLs.
Methods: We introduce C-STEM, a fast and powerful method for multi-context trans-eQTL mapping. C-STEM accounts for intra-individual correlation by decomposing gene expression into shared and context-specific components, builds cross-validated cis-genetic predictors (CVGP) for each component, and constructs a final predictor using both components. C-STEM tests for association between all CVGP-trans gene pairs, reducing the number of tests and enhancing power. We utilize hierarchical testing to control FDR across and within contexts, boosting power when significant CVGP-trans gene pairs exist in multiple contexts.
Results: Simulations demonstrate C-STEM’s increased power in detecting trans-gene regulation compared to other methods. Applied to bulk RNA-seq data from the GTEx consortium (N = 948) and single-cell RNA-Seq data from the CLUES (N = 234) and OneK1K (N = 937) cohorts, C-STEM identifies 89% of trans-eQTLs mapped by existing approaches, while providing a 65% increase in the number of trans-gene regulations identified. Existing approaches overestimate context specificity of trans-eQTL effects; 12% of trans-eQTLs appear unique to a single context using existing methods, compared to only 6% using C-STEM.
Conclusion: C-STEM enables construction of multi-context trans-eQTL maps, enhancing understanding of cross-context gene regulatory networks underlying complex human traits.
Grants: NIH U01HG012079
Conflict of Interest: None declared
C01.5 ProteoVAE: guided disentangled Variational Autoencoder for the multivariate analysis of the blood plasma proteome
Michela Carlotta Massi 1, Nathaniel Robert Nethercott2, Solène Cadiou1, Francesca Ieva1;2, Emanuele Di Angelantonio1;3
1Human Technopole, Italy; 2Politecnico di Milano, Milano, Italy; 3University of Cambridge, United Kingdom
Background/Objectives: Understanding the genetic basis of variation in plasma protein levels, essential for human biology and drug targeting, remains a challenge due to the complexity of the plasma proteome and limitations in existing pQTL studies, which fail to capture shared regulatory patterns and face a high multiplicity burden. This study introduces a novel Deep Learning-based method to perform multivariate association tests, i.e. latentQTL analyses, to explore the genetic determinants of protein level variability through interdependent protein sets.
Methods: We present proteoVAE, a disentangled Variational AutoEncoder (VAE) that maps protein level profiles into lower-dimensional spaces, where each dimension represents an independent latent factor driving variation in the plasma proteome, devoid of technical confounders. We also propose a novel method to associate protein sets to each dimension and study factors’ functional meaning; Finally, we exploit these latent factors to perform Genome-Wide Association tests, uncovering latentQTLs.
Results: Applied to 54,219 UKBiobank participants’ plasma proteomic profiles from The Pharma Proteomics Project, proteoVAE efficiently mapped 2,923 protein levels into low-dimensional latent spaces (from 10 to 128 dimensions). We associate protein-sets to each latent factor, revealing significant enrichment in intra-group protein correlation. Notably, each protein-set represented meaningful and disentangled biological pathways. latentQTL results replicated known pQTL signals, while uncovering new associations with the identified pathways.
Conclusion: proteoVAE offers an innovative, interpretable framework for linking genetic factors to functionally coherent protein groups, simplifying the genetic architecture of the plasma proteome, reducing the burden of multiple testing and identifying new therapeutic targets and drug repurposing opportunities.
Conflict of Interest: None declared
C01.6 Genetic prediction of UK Biobank proteomics data for the OmicsPred portal
Douglas Loesch 1, Yu Xu2, Henry Taylor2;3, Samuel Lambert2, Scott C. Ritchie2, Xilin Jiang2, Carles Foguet Coll2, Benjamin B. Sun4, Heiko Runz4, Christopher D. Whelan5, Slavé Petrovski1, Abhishek Nag1, Dirk Paul1, Michael Inouye2
1AstraZeneca, Centre for Genomics Research, Cambridge, United Kingdom; 2University of Cambridge, British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, Cambridge, United Kingdom; 3National Human Genome Research Institute (NHGRI), National Center for Precision Health Research, Bethesda, United States; 4Biogen, Cambridge, United States; 5Janssen Pharmaceuticals, Boston, United States
Background/Objectives: Multi-omic analyses are a powerful tool for uncovering the molecular underpinnings of disease. Genetic prediction of omics data can extend omics information into biobank-scale studies where financial restraints have typically imposed significant hurdles.
Methods: We have expanded the OmicsPred portal, a repository of omics genetic scores, to include proteomics data from the UK Biobank (N = 54,219), dramatically increasing both the sample size and the number of Olink-characterized proteins over currently available scores. We trained genetic scores for proteins using Bayesian ridge regression and imputed genotype data on a training set of 34,557 participants, followed by testing the scores on the remaining participants. We then performed phenome-wide association studies (PheWAS) with the genetic scores using Cox regression and the time to diagnosis.
Results: Out of the 2,612 models we trained, 1,712 had an R2 of at least 0.01 in the withheld set and 166 had an R2 of at least 0.3. To evaluate score portability, we stratified the withheld set by genetically predicted ancestry and compared the variance explained (R2) in the European ancestry subset with four other ancestry groups using the slope of the regression line (0.88, 087, 0.71, and 0.44 for Admixed American, South Asian, East Asian, and African, respectively).
Conclusion: To demonstrate the utility of our scores, we performed PheWAS in the UK Biobank and All of Us, encompassing over 900,000 participants from diverse backgrounds. Other potential applications include causal inference and pathway-aware genetic risk prediction. Genetic scores will be made available on the OmicsPred portal (https://www.omicspred.org/).
Grants:
Conflict of Interest: Douglas Loesch Employee of AstraZeneca, Yu Xu: None declared, Henry Taylor: None declared, Samuel Lambert: None declared, Scott C. Ritchie: None declared, Xilin Jiang: None declared, Carles Foguet Coll: None declared, Benjamin B. Sun May own stock or interests in Biogen., Employee of Biogen., Heiko Runz Holds stock in Biogen, Employee of Biogen, Christopher D. Whelan May hold stock in Janssen Pharmaceuticals or Johnson and Johnson, Employee of Janssen Pharmaceuticals, a Johnson and Johnson company, Slavé Petrovski May own stocks or interests in AstraZeneca, Employee of AstraZeneca, Abhishek Nag May own AstraZeneca stocks or interests., Employee of AstraZeneca, Dirk Paul May own stock or interests in AstraZeneca, Employee of AstraZeneca, Michael Inouye Research collaborations with Nightingale Health and Pfizer., Grants include British Heart Foundation (RG/18/13/33946), NIHR Cambridge Biomedical Research Centre (BRC-1215-20014; NIHR203312), Cambridge BHF Centre of Research Excellence (RE/18/1/34212), BHF Chair Award (CH/12/2/29428) and the Health Data Research UK (Molecules to Health Records programme), Trustee of the Public Heath Genomics Foundation, member of the Scientific Advisory Board of Open Targets
C02 Reproductive genetics
C02.1 Mapping the prevalence and origin of (mosaic) chromosomal abnormalities in human blastocysts: findings and potential value for PGT
Machteld Baetens 1, Lisa De Witte2, Kelly Tilleman3, Frauke Vanden Meerschaut3, Sandra Janssens1, Ariane Van Tongerloo1, Virginie Szymczak1, Dominic Stoop3, Annelies Dheedene1, Sofie Symoens1, Björn Menten4
1Ghent University Hospital, Center for Medical Genetics, Ghent; 2Ghent University, Center for Medical Genetics, Ghent, Belgium; 3Ghent University Hospital, Department for Reproductive Medicine, Ghent; 4Ghent University Hospital - Ghent University, Center for Medical Genetics, Ghent
Background/Objectives: Diagnosing (mosaic) chromosomal aneuploidy in blastocysts relies mostly on copy number analysis of single trophectoderm (TE) biopsies, with no cell-division information. Whereas meiotic errors generally affect all embryonic cells, mitotic errors lead to embryo mosaicism, linked with healthy live birth, albeit with lower implantation and higher miscarriage rates than euploid embryos. Combining copy number and cell-division origin analysis, mitotic errors could more reliably be identified, resulting in more transferable embryos.
Methods: TE biopsies of 1,479 embryos were processed by GENType (De Witte et al., 2022) for copy number analysis and comprehensive single nucleotide polymorphism (SNP) analysis.
Results: 68% of the biopsies were euploid, others presented with whole-chromosome aneuploidy (22%), segmental aneuploidy (7%) or both (2%). The overall incidence of whole-chromosome mosaicism was 8%. Origin analysis revealed that uniform aneuploidies predominantly occurred from meiotic errors (97%) in the oocyte, with a higher prevalence in acrocentric and smaller chromosomes (P < 0.0001) and advancing female age (P < 0.0001). Conversely, mosaic aneuploidy originated from mitotic errors (92%), on both parental alleles (P > 0.5), distributed across all chromosomes (P > 0.5), with no relation to maternal age (P > 0.5).
These findings demonstrate high concordance (96%) between cell-division origin and copy number. Interestingly, 8% of aneuploidies with an intermediate copy number resulted from a meiotic error, while 3% with a uniform aneuploidy were of mitotic origin.
Conclusion: Our study provides more insight into the underlying mechanisms of (mosaic) chromosomal abnormalities at the blastocyst stage and demonstrates that integrating cell-division origin information improves diagnosing true mosaicism.
Grants: Research Foundation Flanders (FWO; 1S74619N)
Conflict of Interest: None declared
C02.2 Clinical utility of preimplantation genetic testing for aneuploidy origin (PGT-AO) to enhance embryo selection for monogenic disorders: a retrospective pan-country integrative genomic and clinical study
Rick Essers 1;2, Sajjad Biglari1;2, Anouk Janssen1;2, Servi Stevens1;2, Jeroen Meekels1, Bart de Koning1, Jos C.M.F. Dreessen1, Merryn Macville1;2, Sonja de Munnik1, Malou Heijligers1, Ron van Golde1;2;3, Marianne L van Buul-van Zwet4, Math H. E. Pieters4, Koen van Zomeren5, Jannie van Echten-Arends5, Alwin Derijck6;7, Sebastiaan Mastenbroek6;7, Arthur van den Wijngaard1;2, Edith Coonen1;2;3, Aimee D.C. Paulussen1;2, Masoud Zamani Esteki1;2
1Maastricht University Medical Center MUMC + , Department of Clinical Genetics, Maastricht, Netherlands; 2GROW Research Institute for Oncology and Reproduction, Department of Genetics and Cell Biology, Maastricht, Netherlands; 3Maastricht University Medical Center MUMC + , Department of Obstetrics and Gynecology, Maastricht, Netherlands; 4University Medical Center (UMC) Utrecht, IVF Center, Utrecht, Netherlands; 5University Medical Center Groningen, Section of Reproductive Medicine, Department of Obstetrics and Gynecology, Groningen, Netherlands; 6Amsterdam UMC location University of Amsterdam, Center for Reproductive Medicine, Amsterdam, Netherlands; 7Amsterdam Reproduction and Development Research Institute, Amsterdam, Netherlands
Background/Objectives: Randomized controlled trials revealed that PGT-A does not increase live birth rates. PGT-A overlooks the mosaic nature and segregational origin of aneuploidies, causing potentially viable mosaic embryos to not be transferred. Our novel PGT- A approach, termed PGT-AO, provides a nuanced understanding of early embryonic development in clinical practice and potentially leads to viable mosaic embryo transfer.
Methods: This retrospective study includes 816 embryos from 323 couples counseled by clinical geneticists at University Medical Centers in Maastricht, Amsterdam, Utrecht, and Groningen. We examined 284 embryos with genome haplarithmisis-based PGT-AO to assess if (mosaic) aneuploidies and segregational origin would have altered transfer decisions and subsequent clinical outcomes.
Results: Of transferred embryos, 26.7% (48/180) resulted in live-births. PGT-AO shows that 14.6% (7/48) carried abnormalities, including embryos with mosaic mitotic trisomies (n = 2), mosaic monosomies (n = 2), and mosaic duplications (n = 2) and a deletion (n = 1). Of the transferred embryos, 73.3% (132/180) did not result in live-births, and 36.4% (48/132) contained an aberration. A total of 35 trisomies were found in 41.7% (20/48), including meiotic (n = 9), mitotic (n = 5), meiotic and mitotic origins (n = 1), and 5 were indeterminable.
Conclusion: Our preliminary results show PGT-AO can avoid transferring embryos with aneuploidies of meiotic origin and non-mosaic status, while potentially considering embryos with aneuploidies of mosaic and mitotic origin for transfer. This increases pregnancy rate per embryo transfer and minimizes transfers with poor outcomes, potentially avoiding termination of aneuploid pregnancies.
Grants:
-
EVA (KP111513), MUMC+
-
Horizon Europe (NESTOR, 101120075), European Commission
-
Horizon 2020 Innovation (ERIN, EU952516)
Conflict of Interest: None declared
C02.3 The potential of long-read whole genome sequencing based preimplantation genetic testing
Yan Zhao 1, Greet Peeters1, Olga Tšuiko1, Eftychia Dimitriadou1, Erika Souche1, Tatjana Jatsenko1, Joris Robert Vermeesch1
1KU Leuven, Department of Human Genetics, Leuven, Belgium
Background/Objectives: Haplotype linkage analysis is commonly employed to enable preimplantation genetic testing for monogenic disorders (PGT-M). To haplotype, DNA from close relatives need to be accessible. However, close relatives are not always available, and this requirement can delay and increase costs for PGT-M. To overcome the need for relatives, we hypothesized that long-read whole genome sequencing (lrWGS) of parental samples and trophectoderm/single blastomere biopsies could enable genome-wide haplotype-base PGT-M analysis.
Methods: We used cell lines and available sequencing data from the Genome in a Bottle (GIAB) Ashkenazi trio. For the parents, publicly available lrWGS data were downloaded. For the son, multi-cell (10 cells) and single-cell samples were collected and sequenced to simulate trophectoderm and single blastomere biopsies, respectively. Variant calling and phasing was performed for all individuals, with a pedigree-based analysis strategy employed specifically for the son to improve accuracy. PGT-M was simulated by comparing haplotypes of the son with parental haplotypes.
Results: Leveraging phased haplotypes of the trio, parental haplotypes inherited by the son were inferred across ~ 99% of protein-coding gene regions for both multi-cell and single-cell samples. Furthermore, haplotype based PGT-M exhibited a diagnosis accuracy of ~99% for SNV loci within protein-coding gene regions for both multi-cell and single-cell samples.
Conclusion: Our study demonstrates that lrWGS-based PGT provides an alternative to current methods, eliminating the reliance on family members for phasing.
Grants: Marie Skłodowska-Curie grant agreement no. 813707 (MATER).
Conflict of Interest: None declared
C02.4 Pre-implantation genetic testing by polygenic risk scores selects different embryos across score construction methods with randomness
Shinichi Namba 1;2;3, Masato Akiyama4, Haruka Hamanoue5, Kazuto Kato2, Minae Kawashima6, Itaru Kushima7, Koichi Matsuda1, Masahiro Nakatochi7, Soichi Ogishima8, Kyuto Sonehara1;2;3, Ken Suzuki2, Atsushi Takata3, Gen Tamiya3;8, Chizu Tanikawa1, Ken Yamamoto2, Natsuko Yamamoto2, Norio Ozaki7, Yukinori Okada1;2;3
1The University of Tokyo; 2Osaka University; 3RIKEN Center; 4Kyushu University; 5Yokohama City University Hospital; 6ROID-DS; 7Nagoya University; 8Tohoku University
Consortium: The BioBank Japan Project
Background/Objectives: Private enterprises offer pre-implantation genetic testing by polygenic risk scores (PRS) to select embryos with ‘desirable’ potential, prior to clinical implementation of PRS for disease prevention. Here, we question the testing’s validity; selected embryos can differ across PRS methods and replicates of the same method.
Methods: Using six popular methods, we constructed PRS for adult height, a well-known target of mating preferences. Using biobank resources, we virtually simulated mating process and generated ten embryo genotypes per mate-pair to evaluate the concordance of selected embryos across methods. We repeated the simulation for type 2 diabetes and saturated summary statistics that we simulated with a sample size larger than any studies to date (N = 1×107).
Results: In any combination of PRS methods, the same embryo was selected as the top-ranked one with only a 30.0% chance for adult height (median; range, 20.4–41.6%). The top-ranked embryo by a particular method was lowest-ranked by at least one other method with a median 5.9% chance (3.4–8.0%), and vice versa (median 5.2% [4.0–7.8%]). This inconsistency was not affected by assortative mating. We observed consistent results for type 2 diabetes and the simulated phenotype, with the latter indicating that the inconsistency cannot be solved in the current or near-future sample sizes. Furthermore, simply repeating the same method produced a different embryo ranking.
Conclusion: The selected embryo substantially relied on the choice of PRS method and the random fluctuation in PRS construction, raising serious ethical concerns about selecting embryos and consequently discarding other embryos in an unreliable way.
Grants: None.
Conflict of Interest: None declared
C02.5 Spatiotemporal transcriptomic atlas of mouse placentation
Yanting Wu1;2;3;4, Kaizhen Su 5, Ying Zhang6;7, Langchao Liang6;8;9, Fei Wang7, Ling Gao2, Qiutong Zheng2, Cheng Li2, siyue chen2, Yunfei Su2, Yiting Mao2, Simeng Zhu5, Chaochao Chai6;8;9, Qing Lan7, Man Zhai7, Xin Jin7, Jinglan Zhang2;3;4, Xun Xu7;10, Yu Zhang2, Ya Gao7, Hefeng Huang1;2;3;4;5;11
1Zhejiang University School of Medicine, Key Laboratory of Reproductive Genetics (Ministry of Education), Hangzhou, China; 2Fudan University, Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Shanghai, China; 3Chinese Academy of Medical Sciences, Research Units of Embryo Original Diseases, Shanghai, China; 4Shanghai Key Laboratory of Reproduction and Development, Shanghai, China; 5Shanghai Jiao Tong University, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai, China; 6BGI Research, Wuhan, China; 7BGI Research, Shenzhen, China; 8BGI Research, Qingdao, China; 9University of Chinese Academy of Sciences, College of Life Sciences, Beijing, China; 10Guangdong Provincial Key Laboratory of Genome Read and Write, Shenzhen, China; 11Zhejiang University School of Medicine, Department of Reproductive Endocrinology, Women’s Hospital, Hangzhou, China
Background/Objectives: The placenta, a transient yet vital organ for gestational support, undergoes intricate morphological and functional transformations throughout gestation. Disruptions in placentation are always associated with adverse perinatal outcomes and birth defects. Establishing a spatiotemporal transcriptome atlas of mouse placentation across different stages is crucial for providing valuable research resources and enhancing our understanding of placentation.
Methods: We collected 13 mouse uterine segments, each containing an embryo and its placenta, at 6 different time points spanning from E7.5 to E14.5. We then performed Stereo-seq and utilized the spatial transcriptome data to generate an atlas for placentation.
Results: Stereo-seq was used to construct a Mouse Placentation Spatiotemporal Transcriptomic Atlas (MPSTA). Focusing on the labyrinth region, spatiotemporal ligand-receptor dynamics unveiled pivotal modulators of trophoblast development and placental angiogenesis. At the invasion front, we identified interface-specific transcription factor regulons involved in gestational maintenance. Our analysis also indicated that a maternal high-fat diet preferentially impacts this interface, exacerbating inflammatory responses and perturbing angiogenic homeostasis. Finally, we discovered a compartmentalized expression pattern of certain imprinted genes, with paternal alleles predominantly expressed in the labyrinth and maternal genes in the interface and decidua. We also observed some of the heart defects-associated genes exhibited higher expression in the placenta than in the foetal heart, especially in syncytiotrophoblasts and glycogen trophoblasts across E10.5-E14.5, providing research clues for the mechanisms of placenta-heart axis.
Conclusion: Our findings furnish a comprehensive, spatially resolved atlas, offering invaluable insights and benchmarks for future explorations into placental development.
Grants: NSFC (82088102); National Key Research and Development Program of China (2021YFC2700701).
Conflict of Interest: Yanting Wu National Key Research and Development Program of China (No. 2021YFC2700701), Full time, Kaizhen Su: None declared, Ying Zhang Full time, Langchao Liang: None declared, Fei Wang: None declared, Ling Gao Full time, Qiutong Zheng: None declared, Cheng Li Full time, siyue chen: None declared, Yunfei Su: None declared, Yiting Mao: None declared, Simeng Zhu: None declared, Chaochao Chai: None declared, Qing Lan Full time, Man Zhai Full time, Xin Jin Full time, Jinglan Zhang Full time, Xun Xu Full time, Yu Zhang Full time, Ya Gao Full time, Hefeng Huang National Natural
Science Foundation of China (No. 82088102), Full time
C02.6 Comparison of seven carrier screening panels: results from a large multicenter study with more than 24000 patients (virtual)
Mónica Fabbro 1, Filippo Zambelli2, Denny Sakkas3, Irene Miguel Escalada4, Juanjo Guillen Quilez5, Sebastián Menazzi1, Micaela Galain1, Eloisa Caviglia1, Yannina Diaz Cano1, Sergio Papier1, Karinna Lattes6, Aline Lorenzon7, Jose Buratini8, Frederikke Lindenberg9, Mina Popovic2, Cecilia Fernandez1
1Cegyr - Eugin Group, Novagen Genetics Laboratory, Buenos Aires, Argentina; 2Eugin Group, Research and Development, Barcelona, Spain; 3Boston IVF Fertility Clinic, IVF Laboratory, Boston, United States; 4Clínica Eugin, Research and Development, Barcelona, Spain; 5Clínica Eugin, Medical, Barcelona, Spain; 6Center for Infertility and Human Reproduction CIRH - Eugin Group, Research and Development, Barcelona, Spain; 7Huntington Medicina Reprodutiva - Eugin Group, Research and Development, São Paulo, Brazil; 8Biogenesi-Istituti Clinici Zucchi - Eugin Group, Research and Development, Monza, Italy; 9Copenhaguen Fertility Center - Eugin Group, Research and Development, Copenhaguen, Denmark
Background/Objectives: Carrier screening (CS) tests have evolved with diverse methodological approaches and variability in gene inclusion. The lack of consensus on the most suitable genetic panel for reproductive medicine presents a challenge in clinical applications. This study aimed to conduct a comprehensive review and comparison of various CS panels utilized by different providers.
Methods: This study included 8 IVF centers from 6 countries. Results from 24038 individuals that were tested using one of 7 different CS panels (designated A to G) were included. Panels were evaluated based on included genes, methodology, carrier rate (CR), and most frequently detected diseases (MFDD). Manual curation of reported MFDD variants was conducted, and panels were compared to the 113 genes recommended by the ACMG.
Results: Panels included a different number of genes: 283(A), 299(B), 302(C,D), 310(E), 484(F), and 546(G). Providers used different methodologies: genotyping by Array (C) or NGS (B,G), full-exon NGS (D,F) or a combination (A,E). Significant (p < 0.05) CR variations were observed: 58.8%(A), 69.4%(B), 44.9%(C), 79.4%(D), 81.7%(E), 79.2%(F), and 88%(G). Observed MFDD and carrier frequencies for common diseases like Cystic fibrosis (CFTR), GJB2 nonsyndromic hearing loss, Congenital adrenal hyperplasia (CYP21A2), Familial mediterranean fever (MEFV), and Spinal muscular atrophy (SMN1) displayed notable variations. Importantly, curation of variants led to significant changes in disease CRs. Panels only included 65 to 79 ACMG genes.
Conclusion: Panel variability affects carriers and disorders detected, impacting the number of at-risk couples to be identified. Curation of reported variants should be considered. Healthcare professionals should take this into consideration when implementing CS.
Grants:
Conflict of Interest: Mónica Fabbro Full-time employee at Novagen, Filippo Zambelli Full-time employee at Eugin Clinic, Denny Sakkas Full-time employee at Boston IVF, Irene Miguel Escalada Full-time employee at Eugin Clinic, Juanjo Guillen Quilez Full-time employee at Eugin Clinic, Sebastián Menazzi Full-time employee at Novagen, Micaela Galain Full-time employee at Novagen, Eloisa Caviglia Full-time employee at Novagen, Yannina Diaz Cano Full-time employee at Novagen, Sergio Papier Shareholder of CEGYR, Karinna Lattes Full-time employee at CIRH, Aline Lorenzon Full-time employee at Huntungton, Jose Buratini Full-time employee at Biogenesi-Istituti Clinici Zucchi - Eugin Group, Frederikke Lindenberg Full-time employee at Copenhaguen Fertility Center - Eugin Group, Mina Popovic Full-time employee at Clínica Eugin, Cecilia Fernandez Full-time employee at Novagen
C03 Structural Variants
C03.1 Copy number analysis from genome sequencing data of 11,754 rare disease parent-child trios: a model for identifying autosomal recessive human gene knockouts including a novel gene for autosomal recessive retinopathy
Eric Olinger 1;2, Ian Wilson3, Sarah Orr2, Miguel Barroso-Gil2, Ruxandra Neatu2, Denize Atan4;5, John Sayer2;6
1Cliniques universitaires Saint-Luc (UCLouvain), Center for Human Genetics, Bruxelles, Belgium; 2Newcastle University, Translational and Clinical Research Institute, Newcastle, United Kingdom; 3Newcastle University, Biosciences Institute, Newcastle, United Kingdom; 4Bristol Eye Hospital, Bristol, United Kingdom; 5Bristol Medical School, Translational Health Sciences, Bristol, United Kingdom; 6Newcastle Upon Tyne Hospitals NHS Foundation Trust, Renal Services, Newcastle
Consortium: Genomics England Research Consortium
Background: In parent-child trios with genome sequencing data, we investigated inherited biallelic deletions to identify known and novel genetic disorders.
Methods: We developed a CNV analysis pipeline based on autosomal genome sequencing read depth of Genomics England 100,000 Genomes Project data from 11,754 parent-child trios and additional 18,875 non-trios. A control cohort of 15,440 cancer patients provided independent deletion frequencies.
Results: Autosomal recessive (AR) modelling detected 34 distinct rare deletions that were homozygous in the proband and heterozygous in both parents. These inherited biallelic deletions were only detected in 52 trios. These “knockout” regions included 37 genes among them 8 with an OMIM AR annotation. Deletions of NPHP1, followed by OTOA, both within segmental duplications, were the only recurrent findings explaining phenotypes in a total of 10 and 3 patients, respectively. Recurrent heterozygous NPHP1 deletions were detected in 0.3%-0.5% of controls. We reviewed “knockout” patients for the remaining 29 genes without disease associations and identified SLC66A1 as a likely novel cause for AR rod-cone dystrophy in 4 families.
Conclusion: A tailored CNV analysis of genome sequencing trio data shows that biallelic inherited gene deletions are rare, with NPHP1 biallelic deletions causing nephronophthisis the leading finding. We propose SLC66A1 as a novel cause for AR retinopathy.
Grants: Kidney Research UK Grants Paed_RP_001_20180925, ST_001_20171120; LifeArc, MRC (MR/Y007808/1); Northern Counties Kidney Research Fund (20/01); Swiss National Science Foundation Grants P2ZHP3_195181 and P500PB_206851
Conflict of Interest: None declared
C03.2 Mechanisms of misexpression-associated structural variants in whole blood
Thomas Vanderstichele 1, Katie L Burnham1, Niek de Klein1, Manuel Tardaguila2, Brittany Howell1, Klaudia Walter1, Kousik Kundu1, Wanseon Lee1, Jonas Koeppel1, Alex Tokolyi1, Elodie Persyn3, Artika Nath4, Jonathan Marten3, Slavé Petrovski5, David Roberts6, Emanuele Di Angelantonio3, John Danesh3, Alix Berton7, Adam Platt8, Adam Butterworth3, Nicole Soranzo2, Leopold Parts1, Michael Inouye3, Dirk Paul5, Emma Davenport1
1Wellcome Sanger Institute, Hinxton, United Kingdom; 2Human Technopole, Milan, Italy; 3University of Cambridge, British Heart Foundation Cardiovascular Epidemiology Unit, Cambridge, United Kingdom; 4Baker Heart and Diabetes Institute, Cambridge Baker Systems Genomics Initiative, Melbourne, Australia; 5AstraZeneca, Centre for Genomics Research, Cambridge, United Kingdom; 6University of Oxford, Radcliffe Department of Medicine, Oxford, United Kingdom; 7AstraZeneca, Translational Science and Experimental Medicine, Gothenburg, Sweden; 8AstraZeneca, Translational Science and Experimental Medicine, Cambridge, United Kingdom
Consortium: INTERVAL consortium
Background: Gene misexpression is the unexpected transcription of a gene in a context where it is usually inactive. In this study, we aimed to establish the genetic mechanisms by which gene misexpression occurs in healthy individuals.
Methods: Using 2,804 paired RNA and whole-genome sequencing samples (INTERVAL cohort), we identified 105 rare structural variants (SVs) associated with consistent gene misexpression. To understand the underlying mechanisms we combined STAR-fusion, Salmon, and regulatory annotations, and visualised their effects using Sashimi and coverage plots.
Results: Overall, we identified mechanisms for 42% of misexpression events with a nearby associated SV. First, we identified 12 deletions and 5 tandem duplications associated with highly aberrant read counts and coverage across the intergenic regions upstream of the misexpressed gene. We concluded that these variants resulted in transcriptional readthrough leading to downstream gene misexpression. Interestingly, 2 deletion carriers also showed evidence of intergenic splicing involving the misexpressed genes RTP1 and OTP. Next, we identified 7 deletions and 3 tandem duplications that created fusion transcripts, including a duplication associated with misexpression of myosin heavy chain 1, a gene normally expressed exclusively in skeletal muscle. Finally, we identified a partial gene inversion leading to transcript-specific misexpression of ROPN1B. Surprisingly, we did not observe misexpression occurring via 3D chromatin architecture rearrangements as reported in certain rare diseases.
Conclusion: We have identified putative mechanisms for a subset of misexpression-associated SVs, including transcriptional readthrough, transcript fusions and gene inversion, thus increasing understanding of the impact of rare genetic variants on gene expression.
Grants: BB/V509425/1
Conflict of Interest: Thomas Vanderstichele Has received PhD studentship funding from AstraZeneca, Katie L Burnham: None declared, Niek de Klein: None declared, Manuel Tardaguila: None declared, Brittany Howell: None declared, Klaudia Walter: None declared, Kousik Kundu Stockholder of AstraZeneca, Current employee at AstraZeneca, Wanseon Lee: None declared, Jonas Koeppel: None declared, Alex Tokolyi: None declared, Elodie Persyn: None declared, Artika Nath: None declared, Jonathan Marten Completed this work while employed by the University of Cambridge, but is now an employee of Genomics plc, Slavé Petrovski Stockholder of AstraZeneca, Current employee at AstraZeneca, David Roberts Employee of NHS Blood and Transplant, Emanuele Di Angelantonio: None declared, John Danesh Received multiple grants from academic, charitable and industry sources outside of the submitted work, Scientific advisory boards for AstraZeneca, Novartis, and UK Biobank, Alix Berton Currently an employee of Bayer AG, Research and Early Development Precision Medicine, Research & Development, Pharmaceutical Division, Wuppertal, DE, Adam Platt Stockholder of AstraZeneca, Current employee at AstraZeneca, Adam Butterworth: None declared, Nicole Soranzo: None declared, Leopold Parts: None declared, Michael Inouye Research collaboration with AstraZeneca which is unrelated to this study, Trustee of the Public Health Genomics (PHG) Foundation and a member of the Scientific Advisory Board of Open Targets, Dirk Paul Stockholder of AstraZeneca, Current employee at AstraZeneca, Emma Davenport: None declared
C03.3 Optical genome mapping identifies hidden structural variants in previously undiagnosed rare disease cases in Solve-RD
Kornelia Neveling1, Ronald van Beek1, Eveline Kamping1, Marloes Steehouwer1, Bart van der Sanden 1, Lydia Sagath1, Laura Batlle-Masó2, Roger Colobran2, Lisenka Vissers1, Alexander Hoischen1;3
1Radboud university medical center, Department of Human Genetics, Research Institute for Medical Innovation, Nijmegen, Netherlands; 2Vall d’Hebron University Hospital (HUVH), Autonomous University of Barcelona (UAB), Vall Hebron Research Institute (VHIR), Barcelona, Spain; 3Radboud university medical center, Department of Internal Medicine; Radboud Expertise Center for Immunodeficiency and Autoinflammation and Radboud Center for Infectious Disease (RCI), Nijmegen, Netherlands
Consortium: Solve-RD DITF-ITHACA, Solve-RD DITF-Euro-NMD, Solve-RD DITF-RND, Solve-RD ERN-RITA, Solve-RD DITF-EpiCARE, SOLVE-RD DATF on behalf of the Solve RD OGM working group and the Solve-RD consortium
Background/Objectives: Optical genome mapping (OGM) allows genome-wide detection of structural variants (SV) as small as 500bp. Here, we systematically applied OGM to identify disease-causing SVs in rare disease (RD) patients that remained undiagnosed after standard-of-care analysis in the respective RD-expert centers.
Methods: 159 individuals from 83 families, with 98 affected individuals have been successfully analyzed using OGM. These included 40 singletons, 11 duos, 26 parent-child trios with expected de novo origin of disease, and 6 other family constellations. The largest groups of samples were selected by clinical experts of ERN-ITHACA (52 families, of which 25 trios), followed by 13 NMD-, 8 RND-, 6 RITA-, and 4 EpiCare- families.
Results: Ongoing data analysis revealed 6 de novo SVs in 26 trios (SV mutation rate: ~1/4 genomes), confirming an expected low SV mutation rate, but also implying every de novo SV as a potential disease candidate. Additionally, we focused on rare (segregating) SVs in singleton cases (30-50/genome). In total, we identified (candidate) disease-causing SVs in >10% of index cases. This includes SVs in well-established disease genes (e.g. SERPING1 in two hereditary angioedema families), as well as rare inversions (n = 1), complex chromosomal aberrations (n = 1) or overlapping non-coding SVs (n = 5) each suggesting novel disease mechanisms. These SVs were usually missed by the initial test modalities or were identified in parallel by Solve-RDs short- and long-read genome sequencing efforts.
Conclusion: This large-scale pan-European study successfully demonstrates the identification of so far hidden SVs in RD by OGM.
Grants: Solve-RD received EU Horizon 2020 funding (779257).
Conflict of Interest: Kornelia Neveling The Solve-RD project has received funding from the European Union’s Horizon 2020 research and innovation program under grant agreement No. 779257, Ronald van Beek The Solve-RD project has received funding from the European Union’s Horizon 2020 research and innovation program under grant agreement No. 779257, Eveline Kamping The Solve-RD project has received funding from the European Union’s Horizon 2020 research and innovation program under grant agreement No. 779257, Marloes Steehouwer The Solve-RD project has received funding from the European Union’s Horizon 2020 research and innovation program under grant agreement No. 779257, Bart van der Sanden The Solve-RD project has received funding from the European Union’s Horizon 2020 research and innovation program under grant agreement No. 779257, Lydia Sagath The Solve-RD project has received funding from the European Union’s Horizon 2020 research and innovation program under grant agreement No. 779257, Laura Batlle-Masó The Solve-RD project has received funding from the European Union’s Horizon 2020 research and innovation program under grant agreement No. 779257, Roger Colobran The Solve-RD project has received funding from the European Union’s Horizon 2020 research and innovation program under grant agreement No. 779257, Lisenka Vissers The Solve-RD project has received funding from the European Union’s Horizon 2020 research and innovation program under grant agreement No. 779257, Alexander Hoischen The Solve-RD project has received funding from the European Union’s Horizon 2020 research and innovation program under grant agreement No. 779257
C03.4 Unraveling undiagnosed rare disease families by HiFi long-read genome sequencing
Lydia Sagath 1, Wouter Steyaert1, german demidov2, Nick Zomer1, Marloes Steehouwer1, Ronny Derks1, Marjan Weiss1, Amber den Ouden1, Simone Van Den Heuvel1, Hilde Swinkels1, Kornelia Neveling1, Stephan Ossowski2, Lisenka Vissers1, Christian Gilissen1, Alexander Hoischen1;3
1Radboud University Medical Center, Department of Human Genetics, Research Institute for Medical Innovation, Nijmegen, Netherlands; 2Universitätsklinikum Tübingen – Institut für Medizinische Genetik und angewandte Genomik, Tübingen, Germany; 3Radboud University Medical Center, Department of Internal Medicine; Radboud Expertise Center for Immunodeficiency and Autoinflammation and Radboud Center for Infectious Disease (RCI), Nijmegen, Netherlands
Consortium: Solve-RD long-read sequencing working group, Solve-RD DITF-ITHACA, Solve-RD DITF-Euro-NMD, Solve-RD DITF-RND, Solve-RD DITF-EpiCARE, Solve-RD DATF, and the Solve-RD consortium
Background/Objectives: Solve-RD aims to identify the disease-causing genetic variant in previously undiagnosed rare disease (RD) families; we present one of the largest long-read sequencing (LRS) studies to date. We evaluated the effectiveness of LRS in resolving genetic causes in RD families lacking a clear molecular diagnosis despite previous testing.
Methods: We studied 293 individuals from 114 genetically undiagnosed RD families selected by European Rare Disease Networks experts. The 93 families in the ‘unsolved’ cohort were affected by rare neurological, neuromuscular, or epilepsy disorders, and 21 families by clinically recognizable ‘unsolvable syndromes’ for which genetic causes remain unknown. Using 10x coverage HiFi-LRS we detected small variants, SNVs, structural variants (SVs), and short tandem repeats (STRs), with de novo analysis in trios.
Results: Variant analysis in known disease genes resulted in novel genetic diagnoses in ten families with neurological or neuromuscular disorders, involving de novo and inherited SVs, SNVs, and STRs in DMD, NOP56, DAB1, RFC1, SPAST, and TTN. Additionally, we found potential disease-causing SVs in three families (affecting MCF2/FGF13, REEP1, and PSMA3). In an ‘unsolvable syndromes’ patient, we identified a known disease-causing SNV in TUBA1A, re-classifying the patient’s diagnosis.
Conclusion: While we did not identify a common genetic cause in any of the ‘unsolvable syndromes’, we identified causal genetic variants in 10% of families from the ‘unsolved’ cohort, and candidate variants in an additional 5%. Our study shows the potential and effectiveness of even modest-coverage LRS in solving RDs.
Grants: European Union’s Horizon 2020 (779257) and Sigrid Jusélius Stiftelse (220540).
Conflict of Interest: None declared
C03.5 Dosage imbalance of the chromatin remodeler CHD1L contributes to the 1q21.1-CNV associated mirrored neurodevelopmental phenotypes
Marianne Lemée 1, Maria Nicla Loviglio1, Tao Ye1, Céline Keime1, Peggy Tilly1, Pernelle Klein1, Chantal Weber1, Olivia Wendling1, Hugues Jacobs1, Delphine Duteil1, Juliette D Godin1, Christophe Romier1, Christelle Golzio1
1I.G.B.M.C. - Institut de génétique et de biologie moléculaire et cellulaire, Illkirch-Graffenstaden, France
Background/objectives: Deletions and duplications of the distal 1q21.1 region are associated with syndromic forms of autism (MIM612474, 612475). Variable phenotypes have been reported, including congenital heart defects, autism, schizophrenia, head circumference and height defects.
Methods: To elucidate which gene(s) are responsible for the 1q21.1 duplication/deletion-associated phenotypes, we performed gene manipulation in zebrafish and mice. We deciphered the function of candidate driver(s) by multi-omics approaches on human iPSC-derived neural progenitor cells (NPC) and cerebral organoids (CO).
Results: We modeled duplication of the 1q21.1 region by overexpressing the 8 genes in the region in zebrafish. We found that only overexpression of CHD1L led to macrocephaly and increased larval body length. Conversely, deletion of the CHD1L zebrafish ortholog resulted in microcephaly and decreased larval body length. These mirror phenotypes are also observed when Chd1l expression is modulated in a mammalian model, notably with a variation in the number of mature Tbr1-positive neurons in the mouse embryo. NPCs derived from control and CHD1L Knock-out isogenic hiPSCs were subjected to transcriptomic, cistromic and chromatin accessibility analyses. These approaches revealed that CHD1L regulates the expression levels and accessibility of genes involved in neuronal differentiation and synaptogenesis, including autism susceptibility genes such as UNC5D and DPP6. Strikingly, absence of CHD1L shifts the cellular identity from forebrain to retinal fate in CO. Finally, pathogenic missense and truncating variants of CHD1L were found in individuals with autism and/or height defects.
Conclusion: CHD1L is a major contributor of the 1q21.1 duplication/deletion-associated microcephaly and growth defects. CHD1L dosage is required during human brain regionalization.
Grants: ANR, FNSNF.
Conflict of Interest: None declared
C03.6 Multiomics and deep phenotyping in MECP2 Duplication Syndrome: Insight into disease severity, expression variability, and nucleic acid therapeutics
Davut Pehlivan 1;2;3, Jesse Bentsson4, Sameer Bajikar2;3, Christopher M. Grochowski2;5, Ming Yin Lun4, Mira Gandhi2, Angad Jolly2, Holly Harris6, Bernhard Suter1, Sukru Aras7, Melissa Ramocki8, Haowei Du2, Ellen Wilkey4, Cemal Karakas9, Jesper Eisfeldt10, Maria Pettersson10, Shalini Jhangiani5, Lynn Liu11, Marwan Shinawi12, Virginia Kimonis13, Wojciech Wiszniewski14, Kyle Mckenzie15, Timo Roser16, Angela Morgante17, Alberto Cornier18, Ahmed Abdelmoity19, Donna Muzny5, Tadahiro Mitani20, Kazuhiro Muramatsu20, Shin Nabatame21, Daniel Glaze1, Jawid Fatih2, Richard Gibbs2;5, Zhandong Liu1;3, Anna Lindstrand10, Fritz Sedlazeck5, James Lupski2;5, Huda Yahya Zoghbi1;2;3, Claudia Carvalho4
1Baylor College of Medicine, Department of Pediatrics Section of Neurology and Developmental Neuroscience, Houston, United States; 2Baylor College of Medicine, Molecular and Human Genetics, Houston, United States; 3Texas Children’s Hospital, Jan and Dan Duncan Neurological Research Institute, Houston, United States; 4Pacific Northwest Research Institute, Seattle, United States; 5Baylor College of Medicine, Human Genome Sequencing Center, Houston, United States; 6Baylor College of Medicine, Developmental and Behavioral Pediatrics, Houston; 71250 Moursund St, Houston, United States; 8University Otolaryngology, Greenwich, United States; 9University of Louisville, Department of Pediatrics Division of Neurology, Louisville, United States; 10Karolinska Institute, Department of Molecular Medicine and Surgery, Sweden; 11University of North Carolina, Department of Pediatrics - Neurology, Chapel Hill, United States; 12Washington University, Department of Genetics and Genomic Medicine, Town and Country, United States; 13University of California Irvine, Department of Pediatrics, Division of Genetics and Genomic Medicine, Orange, United States; 14Oregon Health Science University, Department of Molecular and Medical Genetics, Portland; 15University of Alberta, Department of Pediatrics, Alberta, Canada; 16Ludwig Maximilian University of Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Munich, Germany; 17Institute of Biosciences, Department of Genetics and Evolutionary Biology, Sao Paolo, Brazil; 18San Jorge Children’s Hospital, Department of Genetics, San Juan, Puerto Rico; 19Children’s Mercy Kansas City, Division of Neurology, Department of Pediatrics, Kansas City, United States; 20Jichi Medical University, Department of Pediatrics, Tochigi, Japan; 21Osaka University Graduate School of Medicine, Department of Pediatrics, Osaka, Japan
Background/Objectives: Copy number gains spanning MECP2 cause X-linked intellectual developmental disorder Lubs-type (MRXSL). It is a severe neurodevelopmental disorder with diverse clinical manifestations. Each individual has a unique genomic size of duplication and varying genomic rearrangement structures. Our hypothesis posits that these genomic rearrangement structures contribute to interindividual clinical variability and disease severity.
Methods: We conducted extensive genomic studies including custom-designed aCGH, optical genome mapping and short-read/long-read genome sequencing on 137 MRXSL individuals. We correlated genomic structures with transcriptomic and deep phenotyping analyses including Human Phenotype Ontology (HPO) semantic similarity scores.
Results: CNV sizes ranged from 64 kb to 16 Mb with the following structural distribution: 48% tandem duplications, 22% terminal, 20% Duplication-Triplication-Duplication (DUP-TRP/INV-DUP) structure and 10% other complex genomic structures. De novo events overwhelmingly occurred in the terminal duplication group (65%) compared to the tandem duplication (17%) and CGR groups. RNAseq data from lymphoblastoid cell lines revealed that the MECP2 transcript in MECP2 triplications is statistically different from all duplications, but not between other genomic structures. Genotype-phenotype analyses indicated a gradual worsening of phenotypic features, including overall survival, developmental levels, microcephaly, epilepsy, and genitourinary/eye abnormalities in the following order: tandem duplications, other CGRs, terminal duplications/translocations, and triplications encompassing MECP2. HPO analysis revealed specific patterns between different structures.
Conclusion: MECP2 copy number alterations alone do not solely influence the disease phenotype; the genomic rearrangement structure also plays crucial role in clinical manifestations and severity. Employing comprehensive analytical approach enhances our understanding of genomic disorder impact, guiding more effective therapeutic design.
Conflict of Interest: None declared
C04 Novel genes in neurogenetic disorders
C04.1 The spliceosomal factor CRNKL1 is a novel disease gene required for brain development
Sankalita Ray1, Rosie Sullivan1, Mischa Ruegg1, Gregory Gimenez2, Julia Horsfield2, Gemma Poke3, Sarah Duerinckx4, Muzhirah Haniffa5, Wei-Teik Keng5, Gaik-Siew Ch’ng6, David Parry7, Masamune Sakamoto8, Naomichi MATSUMOTO8, Emma Wakeling9, Ghaydda Mirzaa10;11, Kimberly A. Aldinger12;13, William Dobyns14, Marc Abramowicz15, Louise Bicknell 1
1University of Otago, Department of Biochemistry, Dunedin, New Zealand; 2University of Otago, Department of Pathoogy, Dunedin, New Zealand; 3Genetic Health Service New Zealand, Central Hub, Wellington, New Zealand; 4Université Libre de Bruxelles (ULB), Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Brussels, Belgium; 5Hospital Kuala Lumpur, Department of Genetics, Kuala Lumpur, Malaysia; 6Penang Hospital, Department of Genetics, Penang, Malaysia; 7Institute of Genetics and Cancer, MRC Human Genetics Unit, Edinburgh, United Kingdom; 8Yokohama City University Graduate School of Medicine, Department of Human Genetics, Yokohama, Japan; 9Great Ormond Street Hospital for Children NHS Foundation Trust, North East Thames Regional Genetics Service, London, United Kingdom; 10Brotman Baty Institute for Precision Medicine, Seattle, United States; 11University of Washington, Department of Pediatrics, Seattle, United States; 12Seattle Children’s Research Institute, Center for Integrative Brain Research, Seattle, United States; 13University of Washington, Division of Genetic Medicine, Department of Pediatrics, Seattle, United States; 14University of Minnesota, Department of Pediatrics, Division of Genetics and Metabolism, Minneapolis, United States; 15Université de Geneve, Department of Genetic Medicine and Development, Faculty of Medicine, Geneva, Switzerland
Background/Objectives: Splicing is a complex process, requiring for cell functioning and to create the transcriptomic diversity required for specialised functions in higher eukaryotes. Despite the ubiquitous nature, pathogenic variants in components associated with genetic disease often cause a tissue-specific phenotype, hinting at further complexities which are not yet fully understood.
Methods: Exome sequencing and clinical collaborations or Genematcher partners identified a patient cohort. In vitro and zebrafish experiments were performed to explore the pathogenicity of identified variants, including RNA-seq analysis for the effects on splicing.
Results: We have identified eight families with de novo missense variants in a spliceosomal component, CRNKL1. Significantly, seven individuals have one of two variants that all affect the same amino acid, while the eighth individual has a similar substitution affecting an amino acid in the same position in a neighbouring a-helix. All patients share a specific phenotype: profound pre- and post-natal microcephaly, with pontocerebellar hypoplasia, seizures and severe intellectual disability. Patient-derived fibroblasts show no obvious effects, with minimal splicing alterations and healthy growth. A zebrafish model demonstrated an obvious lack of brain development with a significant reduction in proliferating cells. RNA-seq analysis indicated widespread splicing changes, with altered expression of neuronal and cell cycle genes.
Conclusion: We have identified CRNKL1 as a novel disease gene and demonstrate the requirement for this protein in brain development and functioning. Our findings contribute to a growing disease cluster within the spliceosome which causes a severe brain-specific phenotype.
Grants: Neurological Foundation of New Zealand, Cure Kids New Zealand
Conflict of Interest: None declared
C04.2 A genetic link between neurodevelopmental disorders and activation of transposable elements: TRIM28 as a novel disease causal gene
Laura Castilla-Vallmanya 1, Ninoslav Pandiloski1, Carrie Davis-Hansson1, Fereshteh Dorazehi1, Christopher Douse1, Susanna Balcells2, Raquel Rabionet2, Johan Jakobsson1
1Lund University, Stem Cell Center, Lund, Sweden; 2University of Barcelona, Genetics, Microbiology and Statistics, Barcelona, Spain
Background/Objectives: TRIM28 is an epigenetic co-repressor protein that acts as a master regulator of transposable elements (TEs). These mobile genetic sequences are normally epigenetically silenced to protect genome integrity. TRIM28 silences specific TEs during brain development in neural progenitors. Perturbations of this pathway in mice result in aberrant neurodevelopment and an inflammatory response in the adult brain. Here, we aim to provide functional proof of the pathogenicity of TRIM28 mutations in humans.
Methods: We identified heterozygous missense variants in TRIM28 in four individuals with moderate intellectual disability through whole exome sequencing. We generated CRISPR-edited induced pluripotent stem cells (iPSCs) lines carrying two of these mutations, which affect the PHD-Bromodomain, and a TRIM28 knock-down iPSC model using CRISPRi. We differentiated the TRIM28-mutant iPSCs into forebrain neural progenitors and unguided neural organoids. The transcriptional and epigenetic profile of our models was analyzed by RNA-seq and CUT&RUN histone mark profiling.
Results: We found that TRIM28-mutant iPSCs show a loss of the histone mark H3K9me3 coupled with overexpression of genes and TEs, predominantly endogenous retroviruses (ERVs), an abundant type of TE. These transcriptional changes highly overlap with those observed in the TRIM28-CRISPRi cells, suggesting a loss-of-function effect of the mutations. TRIM28-mutant differentiated neural progenitor cells also showed epigenetic and transcriptional changes related to abnormal neurodevelopment.
Conclusion: Our results show that TRIM28 mutations lead to epigenetic dysregulation causing aberrant TE and gene expression and could explain the neurodevelopmental phenotype of the patients.
Grants: Lindhés Advokatbyrå, Nilsson-Ehle, Vetenskapsrådet, Cancerfonden, Barncancerfonden, Hjärnfonden, Ministerio Ciencia, Innovación y Universidades.
Conflict of Interest: None declared
C04.3 Somatic Short-Tandem Repeats (STR) amplification in neurodevelopmental disorders with regression: the role of FAN1 and DNA repair pathways deficiency
Maria Cerminara 1;2, Giovanni Spirito3;4, Silvia Boeri5, Luca Pandolfini4, Giulia Rosti2;6, Margherita Mancardi5, Livia Pisciotta7;8, Marco Fontana1;2, Alessandra Bianchi1;2, Loretta Ferrera1, Francesco Caroli1, Marco Di Duca1, Maria Teresa Divizia6, Elisa De Grandis2;5, Sara Trova3, Diego Vozzi4, Remo Sanges4;9, Lino Nobili2;5, Federico Zara1;2, Stefano Gustincich4;9, Aldamaria Puliti1;2
1IRCCS Istituto Giannina Gaslini, Medical Genetics Unit, Genoa, Italy; 2University of Genoa, DiNOGMI Dipartimento di Neuroscienze, Riabilitazione, Oftalmologia, Genetica e Scienze Materno-Infantili, Genoa, Italy; 3CMP3vda, Aosta, Italy; 4Istituto Italiano di Tecnologia – IIT, Central RNA Laboratory, Genoa, Italy; 5IRCCS Istituto Giannina Gaslini, Child Neuropsychiatry Unit, ERN EpiCARE Centre, Genoa, Italy; 6IRCCS Istituto Giannina Gaslini, Clinical Genetics and Genomics Unit, Genoa, Italy; 7ASST Fatebenefratelli Sacco, Child Neuropsychiatry Unit, Milan, Italy; 8University of Milan, Milan, Italy; 9Scuola Internazionale Superiore di Studi Avanzati (SISSA), Area of Neuroscience, Trieste, Italy
Background/Objectives: Some patients with neurodevelopmental disorders (NDDs) exhibit regression, losing acquired developmental milestones. DNA repair genes, including FAN1, have been proposed as age-modifier of neurological/psychiatric diseases acting through STRs instability. In NDDs, this mechanism remains unexplored. We aim to advance our understanding on FAN1 and STRs amplification in regressive NDDs employing advanced integrated approaches.
Methods: We used multiple genomic technologies, including exome sequencing, short-read (SR-GS) and long-read genome sequencing (LR-GS) (Oxford Nanopore Technologies), to detect SNVs/indels and STR expantions from blood DNA, Mitomycin C (MMC)-induced cytotoxicity to evaluate DNA damage in lymphoblastoid cells, and capillary electrophoresis-based PCR assay to verify STRs amplification in possible target genes.
Results: Among 7 sporadic cases with heterozygous FAN1 loss-of-function variants, we selected three patients who showed regression. All three patients showed increased sensitivity to MMC-mediated cytotoxicity. SR-GS data indicated additional deleterious variants in other DNA-repair genes, and an increased burden of somatic SNV/indels compared to their parents. In one patient, SR-GS and LR-GS data showed increased amount of short-reads mapping inside repeats, and long-reads, supporting potentially deleterious STR expansions especially in brain-related Gene-Ontologies and NDD genes (e.g. SHANK2). This patient had a maternal FAN1 nonsense variant, and paternally inherited ERCC2 missense and SFR1 nonsense variants. These genes interact in DNA repair pathways via RAD51, central for homologous recombination.
Conclusion: Our study suggests FAN1 deficiency and simultaneous alterations in other DNA repair genes, can lead to somatic STRs expansion, and accumulation in NDD genes, potentially contributing to regressive phenotypes.
Grants: PRIN-20203P8C3X_001
Conflict of Interest: None declared
C04.4 Molecular and functional characterization of a novel tubulinopathy caused by recessive variants in the Tubulin Tyrosine Ligase gene
Valentina Serpieri 1, Alessia Orsi1, Satish Bodakuntla2;3, Veronique Henriot2;3, Roberta De Mori4, Monia Ginevrino5, Patrizia Accorsi6, Lorenzo Pinelli7, Abdulaziz Alsemari8, Mohanned Alsuhaibani9, Stefan T Arold10, Caroline Bénech11, Tommaso Biagini12, Renato Borgatti13;14, Lucio Giordano6, Joseph Gleeson15;16, Maria Magdalena Magiera2;3, Tommaso Mazza12, Sylvia Quemener11;17;18, Rocio Rius19;20, Romina Romaniello14, Silvia Tardivo4, Kevin Uguen11;17;18, Sue White19;20, Maha Zaki21, Carsten Janke2;3, Enza Maria Valente22
1University of Pavia, Department of Molecular Medicine, Pavia, Italy; 2Institut Curie, Université PSL, CNRS UMR3348, Orsay, France; 3Université Paris-Saclay, CNRS UMR3348, Orsay, France; 4IRCCS Santa Lucia Foundation, Induced Pluripotent Stem Cells Unit, Rome, Italy; 5IRCCS Bambino Gesù Children’s Hospital, Laboratory of Medical Genetics, Translational Cytogenomics Reseach Unit, Rome, France; 6ASST-Spedali Civili of Brescia, Unit of Child Neurology and Psychiatry, Brescia, Italy; 7ASST-Spedali Civili of Brescia, Neuroradiology Unit, Pediatric Neuroradiology Section, Brescia, France; 8King Faisal Specialist Hospital and Research Centre, Department of Neurosciences, Riyadh, Saudi Arabia; 9King Faisal Specialist Hospital and Research Centre, Department of Radiology, Riyadh, Saudi Arabia; 10King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Centre (CBRC), Riyadh, Saudi Arabia; 11Univ Brest, Inserm, EFS, UMR 1078, GGB, Brest, France; 12Fondazione IRCCS Casa Sollievo della Sofferenza, Bioinformatics Unit, San Giovanni Rotondo, Foggia, Italy; 13University of Pavia, Department of Brain and Behavioral Sciences, Pavia, Italy; 14IRCCS Mondino Foundation, Unit of Child Neurology and Psychiatry, Pavia, Italy; 15University of California, Department of Neurosciences, San diego, La Jolla, United States; 16Rady Children’s Institute for Genomic Medicine, Rady Children’s Hospital, San Diego, La Jolla, United States; 17CHU de Brest, Service de Génétique Médicale et Biologie de la Reproduction, Brest, France; 18Centre de Référence Déficiences Intellectuelles de causes rares, Brest, France; 19Murdoch Children’s Research Institute, Victorian Clinical Genetics Service, Melbourne, Australia; 20University of Melbourne, Department of Paediatrics, Melbourne, Australia; 21Human Genetics and Genome Research Institute, National Research Centre, Clinical Genetics Department, Cairo, Egypt; 22IRCCS Mondino Foundation, Neurogenetics Research Center, Pavia, Italy
Background/Objectives: Tubulinopathies are complex brain malformation syndromes caused by pathogenic heterozygous variants in genes encoding tubulin subunits. So far, genes implicated in post-translational modifications of tubulins have not been shown to cause tubulinopathy-related phenotypes. By exome sequencing and GeneMatcher search, we identified 8 patients from 5 unrelated families carrying biallelic pathogenic variants in TTL, a gene encoding tubulin tyrosine ligase. This is a ubiquitous enzyme that catalyzes the conversion of detyrosinated to tyrosinated α-tubulin. The phenotype is characterized by a complex neurological presentation (global developmental delay, intellectual disability, ataxia, spasticity, microcephaly) and a peculiar brain malformative pattern of the cerebral cortex, basal ganglia, commissures and posterior fossa, resembling a tubulinopathy.
Methods: Functional studies focused so far on TTL p.Cys338Tyr, detected in homozygosity in two sisters.
Results: Molecular dynamics simulation showed that mutant TTL protein is abnormally flexible, potentially leading to impaired interaction with α-tubulin. An in vitro enzymatic assay demonstrated almost abolished enzymatic activity. Moreover, TTL Cys338Tyr overexpression in mouse hippocampal neurons resulted in abnormally increased axonal length compared to controls. Preliminary studies in fibroblasts from the two sisters disclosed markedly reduced levels of the mutant protein, a significant reduction of primary cilia length and proportion of ciliated cells, an increased percentage of aberrant mitotic spindles, and abnormal cell shape indicating reduced polarization of lamellipodium extension.
Conclusion: Our preliminary work indicates that TTL loss of function causes a novel recessive tubulinopathy, suggesting that mutations in genes involved in tubulin post-translational modifications may account for a proportion of undiagnosed patients with tubulinopathies.
Grants:
Conflict of Interest: None declared
C04.5 Hypomorphic endocytic component REPS2 interferes with neuronal differentiation and causes X-linked cerebral palsy and cognitive impairment
Hao Hu1
1Guangzhou Women and Children’s Medical Center, Laboratory of Medical Systems Biology, Guangzhou, China
Background/Objectives: Clathrin-mediated endocytosis (CME) plays an important role in neuronal maturation and synaptic integration. Here, we identified hypomorphic variants of REPS2 on chromosome X, encoding a regulatory component of CME, from four unrelated male patients with defects in motor and cognition.
Methods: bioinformatics and omics technology; cell, zebrafish and mice models
Results: REPS2 was found to be ubiquitously expressed in multiple brain regions with enrichment in cortical neurons at different developmental stages. In zebrafish models, reps2-knockdown led to brain abnormalities and behavioral alteration, which could be effectively rescued by the human wild-type REPS2 transcript but not by transcripts with the hypomorphic variants. The mouse knockin (KI) models with a hypomorphic Reps2 variant showed impaired performances in motor, cognition, and social behaviors. The defective Reps2 hindered migration of cortical neuronal cells in embryos and interfered with cortical neurite development during adulthood. The defective Reps2 also impeded the formation of clathrin-coated vesicles and thus the progression of cortical neuronal endocytosis.Via single-cell transcriptomics and proteomics analysis, we identified a batch of critical genes affected by the Reps2 defect and confirmed their mutual interactions at cellular level. Furthermore, we found the deficit of dopamine in specific brain regions of the mice KI models, and ameliorated the behavioral abnormalities by supplementing L-dopa accordingly.
Conclusion: In this work, we established for the first time the phenotype–genotype correlation for Reps2 in brain development, and provided novel mechanistic and therapeutic insights into CME-dependent neuronal differentiation.
Grants: This work was supported by the National Natural Science Foundation of China (81974163, 82001121).
Conflict of Interest: None declared
C04.6 Association of rare functionally deficient SMPD1 mutations with Parkinson’s disease
Elaine Chew 1, Zhehao Liu2;3, Zheng Li3, Sun Ju Chung4, Michelle Lian1, Moses Tandiono1, Yue Jing Heng1, Ebonne Ng5, Louis Tan6, Wee Ling Chng2, Tiak Ju Tan3, Esther Peh7, Ying Swan Ho7, Xiao Yin Chen3, Erin Lim3, Chu Hua Chang1, Jonavan Leong1, Ting Xuan Peh3, Ling-Ling Chan8, Yinxia Chao5, Wing-Lok Au6, Kumar Prakash5, Jia Lun Lim9, Yi Wen Tay9, Vincent Mok10, Anne Chan10, Juei-Jueng Lin11, Beomseok Jeon12, Kyuyong Song13, Clement Tham14, Chi Pui Pang14, Jeeyun Ahn15, Kyu Hyung Park16, Janey L. Wiggs17, Tin Aung2;18, Ai Huey Tan19, Azlina Ahmad Annuar9, Mary Makarious20;21, Cornelis Blauwendraat20, Mike Nalls20;22, Laurie Robak23;24, roy alcalay25, ziv gan-or26;27;28, Shen-Yang Lim19, chiea chuen khor2;3, Eng-King Tan2;6, Zhenxun Wang2;3, Jia Nee Foo1;3
1Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore; 2Duke-National University of Singapore Medical School, Singapore, Singapore; 3Genome Institute of Singapore, Singapore, Singapore; 4Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea, Rep. of South; 5Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Singapore, Singapore; 6Department of Neurology, National Neuroscience Institute, Singapore, Singapore; 7Bioprocessing Technology Institute, Singapore, Singapore; 8Department of Diagnostic Radiology, Singapore General Hospital,, Singapore, Singapore; 9Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; 10The Chinese University of Hong Kong, Hong Kong, Hong Kong; 11Department of Neurology, Chushang Show-Chwan Hospital, Nantou, Taiwan; 12Department of Neurology, Seoul National University Hospital, Seoul, Korea, Rep. of South; 13Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, Korea, Rep. of South; 14Department of Ophthalmology and Visual Sciences, Chinese University of Hong Kong, Hong Kong, Hong Kong; 15Department of Ophthalmology, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, Korea, Rep. of South; 16Department of Ophthalmology, Seoul National University Hospital,, Seoul, Korea, Rep. of South; 17Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, United States; 18Singapore Eye Research Institute, Singapore, Singapore; 19Department of Medicine and the Mah Pooi Soo and Tan Chin Nam Centre for Parkinson’s and Related Disorders, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; 20Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, United States; 21Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom; 22Data Tecnica International LLC, Glen Echo, United States; 23Department of Molecular and Human Genetics, Baylor College of Medicine, Texas, United States; 24Jan and Dan Duncan Neurologic Research Institute, Texas Children’s Hospital, Houston, United States; 25Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; 26The Neuro (Montréal Neurological Institute-Hospital), McGill University, Montréal, Canada; 27Department of Human Genetics, McGill University, Montréal, Canada; 28Department of Neurology and Neurosurgery, McGill University, Montréal, Canada
Background/Objectives: Parkinson’s disease is a progressive neurodegenerative disease that adversely affects balance, muscle control and movement. We seek to improve understanding of Parkinson’s disease pathogenesis mechanisms by gleaning mechanistic insights from rare protein-altering genetic variants associated with the disease.
Methods: Whole-exome sequencing was performed on 9,810 individuals (Discovery cohort: 4,298 patients and 5,512 controls) of East Asian ancestry. We tested gene-based association of rare protein-altering variants with Parkinson’s disease status and tested findings in another 11,227 individuals (Replication cohort: 5,585 patients and 5,642 controls) of Asian and European ancestry. Functional assays were conducted to delineate pathogenicity of observed rare protein-altering SMPD1 variants.
Results: Exome-wide significant association (P < 2.5×10-6) of both GBA1 and SMPD1 were observed in the Discovery cohort and subsequently replicated. We established that rare protein-altering SMPD1 variants with less than 44% of normal acid sphingomyelinase activity were associated with Parkinson’s disease risk in both Discovery (odds ratio (OR) = 2.37, 95% CI = 1.68 - 3.35, P = 4.35 × 10-7) and Replication cohorts (OR = 2.18, 95% CI = 1.69 - 2.81, P = 4.80 × 10-10), thus resulting in highly significant association observed when all cohorts were meta-analyzed (OR = 2.24, 95% CI = 1.83 - 2.76, P = 1.25 × 10-15).
Conclusion: Our findings show that rare functionally deficient SMPD1 variants are associated with increased Parkinson’s disease risk and emphasize the significance of sphingomyelin metabolism in Parkinson’s disease pathobiology. Our approach also underscored the utility of functional assays to delineate pathogenic variants in exome sequencing investigations.
Grant: NRF-NRFF2016-03, MOH-000559, MOE-MOET32020-0004
Conflict of Interest: None declared
C05 Genetic contributions to cancer risk and clinical management
C05.1 Characterising the contribution of rare protein-coding variants to prostate cancer risk and severity in 36,866 cases
Jonathan Mitchell 1, Niedzica Camacho1, Patrick R. Shea2, Konrad Stopsack3, Vijay Joseph4, Oliver Burren1, Kathryn Penney3, Jacob Berchuck5, Quanli Wang1, David Goldstein2, Athena Matakidou6, Carolina Haefliger1, Ruth March7, Brian Dougherty8, Keren Carss1, Slavé Petrovski1, Vaidehi Jobanputra2, Philip Kantoff4, Kenneth Offit4, Lorelei Mucci3, Mark Pomerantz5, Margarete Fabre1
1AstraZeneca, Centre for Genomics Research, Cambridge, United Kingdom; 2Columbia University, New York, United States; 3Harvard University, Cambridge, United States; 4Memorial Sloan Kettering Cancer Center, New York, United States; 5Dana-Farber Cancer Institute, Boston, United States; 6GSK, Stevenage, United Kingdom; 7AstaZeneca, Cambridge, United Kingdom; 8AstraZeneca, Watham, United States
Background/Objectives: The pathogenesis of prostate cancer has a strong heritable component. While rare coding germline variants in several genes have been identified as risk factors from candidate gene and linkage studies, the full spectrum of causal rare variants exome-wide remains largely unexplored. To address this, we meta-analysed five exome/genome sequenced cohorts in combination with imputed array data from a population with enriched low-frequency deleterious variants.
Methods: Firstly, we performed a gene-level collapsing analysis, utilising global biobanks, curated disease cohorts and AstraZeneca clinical trial participants with germline whole exome (WES) or whole genome sequencing (WGS) data (total 19,644 cases; 154,025 controls). Secondly, we performed an ExWAS, additionally incorporating imputed array data from the FinnGen cohort (total 33,326 cases; 273,642 controls).
Results: In the gene-level collapsing analysis we identified rare damaging variants as associated with the development of prostate cancer in the DNA damage response pathway genes BRCA2, ATM and CHEK2, and additionally SAMHD1. In the association analysis with prostate cancer severity, we discovered that rare damaging variants in AOX1 increased severity, as do rare damaging variants in BRCA2. At the single-variant level, we found rare non-synonymous variants significantly associated with increased risk of prostate cancer in three genes (HOXB13, CHEK2, BIK), and with decreased risk in four genes (ANO7, SPDL1, AR, TERT).
Conclusion: Our study clarifies the role of known and identifies novel genes associated with prostate cancer risk and severity, providing deeper insight into disease pathogenesis, with the potential for improved risk stratification and novel therapeutic approaches.
Grants: NA
Conflict of Interest: Jonathan Mitchell AstaZeneca, Niedzica Camacho AstraZeneca, Patrick R. Shea: None declared, Konrad Stopsack: None declared, Vijay Joseph: None declared, Oliver Burren AstraZeneca, Kathryn Penney: None declared, Jacob Berchuck: None declared, Quanli Wang AstraZeneca, David Goldstein Actiobio, Athena Matakidou GSK, Carolina Haefliger Debiopharm, Ruth March AstraZeneca, Brian Dougherty AstraZeneca, Keren Carss AstraZeneca, Slavé Petrovski AstraZeneca, Vaidehi Jobanputra: None declared, Philip Kantoff Convergent Therapeutics, Kenneth Offit: None declared, Lorelei Mucci: None declared, Mark Pomerantz: None declared, Margarete Fabre AstraZeneca
C05.2 Deciphering dual clinical entities associated with TP53 pathogenic variants: insights from 53,085 HBOC panels analysis in French laboratories
Edwige Kasper 1, Flavie boulouard2;3, BASSET NOEMIE4, Golmard Lisa5, Hela Sassi6, Emilie Bouvignies7, Maud Brauchaud7, Charbonnier Camille8, Nathalie Parodi7, marion rolain7, Juliette Albuisson9, Ayman AL SAATI10, Marie Bidart11;12, celine bonnet13;14, Ahmed Bouras15, Nadia Boutry-Kryza16, Virginie Bubien17, Adrien Buisson18, Laurent Castera2;3, Chrystelle Colas5, Florence Coulet4, Capucine DELNATTE19, Valentin Derangère20, Celine Garrec21, Marion Gauthier-Villars5, Mathilde Gay-Bellile22, Vincent Goussot23, JESSICA LE GALL5, Mathis Lepage22, Anna Lokchine2, Etienne ROULEAU6, Nicolas Sevenet17;24, Dominique Stoppa-Lyonnet5;25;26, Pierre VANDE PERRE27, Dominique Vaur2;3, Paul Vilquin28, Gaelle Bougeard29, Stéphanie Baert-Desurmont8, Théry Jean-Christophe30, Claude Houdayer1
1Univ Rouen Normandie, Normandie Univ, Inserm U1245 and CHU Rouen, Department of Genetics, F-76000 Rouen, France; 2Laboratory of cancer biology and genetics, Centre François Baclesse, Caen, France; 3Inserm U1245, Cancer and Brain Genomics, Normandie Univ, UNICAEN, FHU G4 génomique, Rouen, France; 4Département de Génétique médicale APHP Sorbonne Université U.F. d’Onco-angiogénétique et Génomique des tumeurs solides Hôpitaux Universitaires Pitié Salpêtrière - Charles Foix, Paris; 5Department of Genetics, Institut Curie, Paris, France PSL University, Paris, France; 6Service de Génétique des Tumeurs, Institut Gustave Roussy, Villejuif; 7Univ Rouen Normandie, Normandie Univ, CHU Rouen, Department of Genetics, F-76000 Rouen, France; 8Univ Rouen Normandie, Normandie Univ, Inserm U1245 and CHU Rouen, Department of Biostatistics and CNRMAJ, F-76000 Rouen, France.; 9Plateforme de Transfert en Biologie Cancérologique, Centre Georges Francois Leclerc, 1 rue Pr Marion, 21000 DIJON; 10Oncogenetics Laboratory, Oncopole Claudius Regaud, IUCT-Oncopole, Toulouse, France et Université de Toulouse, Université Toulouse III-Paul Sabatier, Toulouse, France; 111-CHU Grenoble Alpes, Institut de Biologie et Pathologie, Laboratoire de Génétique Moléculaire : Maladies Héréditaires et Oncologie; 122-Université Grenoble Alpes, Institut Albert Bonniot, INSERM 1209, CNRS UMR 5309, Equipe “Génétique, Epigénétique et Thérapies de l’Infertilité”; 13Genetics Laboratory, University Hospital of Nancy, Nancy, France;; 14INSERM UMR_S 1256, Nutrition, Genetics, and Environmental Risk Exposure (NGERE), Faculty of Medicine of Nancy, University of Lorraine, Nancy, France; 15Centre Léon Bérard, Laboratory of Constitutional Genetics for Frequent Cancer HCL-CLB, Lyon, France; 16Genetic Department, Hospices civils de Lyon, Bron, France; 17Unité d’Oncogénétique, Institut Bergonié, 229 cours de l’Argonne, 33076 Bordeaux cedex; 18Bio-pathology department, Centre Léon Bérard, Lyon, France; 19Institut de Cancérologie de l’Ouest, Unité d’Oncogénétique, Bd Professeur Jacques Monod, 44800 Saint Herblain, France (les 2 pour CD et juste le premier pour CG); 20Plateforme de Transfert en Biologie Cancérologique (CGFL), Centre Georges Francois Leclerc, 1 rue Pr Marion, 21000 DIJON, INSERM UMR1231, Université de Bourgogne; 21Laboratoire de Génétique Moléculaire, Service de Génétique Médicale, Centre Hospitalier Universitaire (CHU) de Nantes, 9 quai Moncousu Nantes 44093, France); 22Department of Oncogenetics, Centre Jean Perrin, INSERM U1240, Université Clermont Auvergne, Clermont-Ferrand 63000, France.; 23Département de Biologie et Pathologie des Tumeurs, Centre Georges Francois Leclerc, 1 rue Pr Marion, 21000 DIJON; 24INSERM U1312, BRIC, Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux cedex; 25INSERM U830, Institut Curie, Paris, France; 26Université Paris Cité, Paris, France; 27Oncogenetics Laboratory, Oncopole Claudius Regaud, IUCT-Oncopole, Toulouse, France; 28Service de Génomique des Tumeurs et Pharmacologie, AP-HP. Paris Cité Université, Hôpital Saint-Louis, Paris, France; 29Univ Rouen Normandie, Inserm U1245, F-76000 Rouen, France; 30Univ Rouen Normandie, Normandie Univ, Inserm U1245, Centre Henri Becquerel, F-76000 Rouen, France
Background/Objectives: Germline TP53 variants predispose to Li-Fraumeni syndrome (LFS), characterized by a wide tumor spectrum at an early age. Excess risk of early onset breast cancer (<31y) has motivated its inclusion in Hereditary Breast and Ovarian Cancer (HBOC) panels. Aim of this work is to provide new insights about TP53 in HBOC patients.
Methods: We collected personal and familial cancer history of 398 patients harboring a TP53 variant identified by 53,085 HBOC panels sequencing in 15 French laboratories.
Results: Heterozygous TP53 variant was identified in 0.44% of HBOC panels, evenly distributed between pathogenic variants (PV) and VUS. Breast cancers associated with TP53 were predominantly triple positive particularly Her2+ breast cancer (p < 0.0001), in situ cancer (p < 0.0001) or phyllode tumors (p < 0.0001). Interestingly, TP53 PV were identified across all ages in breast cancer patients with a median age of onset at 37y [22;82], without enrichment before 31y. We demonstrated that LOF variants were linked with HBOC phenotype, and missense variants, especially with dominant negative effect, with LFS phenotype (p = 0.0096). Patients with breast cancer harboring LOF variants displayed an earlier age of onset compared to missense (p = 0.0030). Surprisingly, we identified, in late onset cancer patients,TP53 hotspot PV usually identified in classic LFS which underlines variable penetrance.
Conclusion: This study suggests the existence of two phenotypic entities associated with TP53 PV: clinical LFS and TP53-related breast cancer. The type of TP53 variant and modifying factors, reflected in familial history, influence these phenotypes and both should be considered to define clinical follow-up of patients and relatives.
Conflict of Interest: None declared
C05.3 Estimating the frequency of Birt-Hogg-Dubé syndrome from genomics and population cohorts
Bryndis Yngvadottir 1;2, Lucy Richman3, Avgi Andreou2, Jessica Woodley4;5, Anita Luharia4;5, Derek Lim6, Eamonn Maher2;7, Stefan Marciniak1;3;8;9
1University of Cambridge, Department of Respiratory Medicine, Cambridge, United Kingdom; 2University of Cambridge, Department of Medical Genetics, Cambridge, United Kingdom; 3Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, United Kingdom; 4Central and South Genomic Laboratory Hub, Birmingham Women’s and Children’s NHS Foundation Trust, Birmingham, United Kingdom; 5West Midlands Regional Genomics Laboratory, Birmingham, United Kingdom; 6Birmingham Women’s and Children’s NHS Foundation Trust, Department of Clinical Genetics, Birmingham, United Kingdom; 7Aston University, Aston Medical School, Birmingham; 8University of Cambridge, Cambridge Institute for Medical Research (CIMR), Cambridge, United Kingdom; 9Royal Papworth Hospital, Cambridge, United Kingdom
Consortium: Genomics England Research Consortium
Genes & Health Research Team
Background/Objectives: Birt-Hogg-Dubé syndrome (BHDS) is an autosomal dominant disorder characterised by benign skin lesions, lung cysts, pneumothorax and an increased lifetime risk for kidney cancer. The syndrome is likely clinically underdiagnosed and its prevalence in the general population underestimated. Most affected families have pathogenic variants in the FLCN gene.
Methods: We extracted pathogenic loss-of-function FLCN variants from >550,000 exomes/genomes of participants in the 100,000 Genomes Project (100kGP), the UK Biobank (UKB) and East London Genes & Health (ELGH). We then calculated age-related risks of pneumothorax and kidney cancer in UKB and compared this to a clinical series of BHDS patients.
Results: The mutation frequency of loss-of-function FLCN variants was 1 in 2,714 in the 100kGP, 1 in 4,189 in UKB and 1 in 1,487 in ELGH. The lifetime risk of pneumothorax in FLCN mutation carriers in the population was substantial with no significant difference between the BHDS patient cohort and UKB participants (p = 0.2154) while the lifetime risk of kidney cancer was significantly lower in UKB compared to BHDS patients (p = 0.0005).
Conclusion: Our frequency estimates for pathogenic FLCN variants are at least ~48 times higher than previous clinical estimates of BHDS frequency (1 in 200,000). These findings highlight the importance of clinical context in managing individuals with FLCN mutations.
Grants: Myrovlytis Trust, Cambridge BRC
This research was made possible through access to data and findings in the National Genomic Research Library via the Genomics England Research Environment.
This research has been conducted using the UK Biobank Resource under application number 95547.
Conflict of Interest: None declared
C05.4 Effect of a Polygenic Risk Score for colorectal cancer incidence in patients with early-onset, familial, or hereditary colorectal cancer
Isabel Spier1;2;3, Hannah Klinkhammer 4;5, Claudia Perne1, Per Hoffmann1, Reinhard Büttner6, Friederike David1, Evelin Schröck3;7;8;9, Silke Redler10, Marcus Franke3;7;8, Gabriela Möslein11, Matthias Kloor12;13, Börge Schmidt14, Annette Erle1;2;3, Deepak Vangala15;16, Huu Phuc Nguyen16, Verena Steinke-Lange3;17;18, Elke Holinski-Feder3;17;18, Andreas Forstner1;19, Robert Hüneburg2;3;20, Carlo Maj21, Christoph Engel22, Andreas Mayr4, Stefan Aretz1;2;3
1Institute of Human Genetics, Medical Faculty, University of Bonn, Bonn, Germany; 2National Center for Hereditary Tumour Syndromes, University Hospital Bonn, Bonn, Germany; 3European Reference Network on Genetic Tumour Risk Syndromes (ERN GENTURIS); 4Institute for Medical Biometry, Informatics and Epidemiology, Medical Faculty, University of Bonn, Bonn, Germany; 5Institute for Genomic Statistics and Bioinformatics, Medical Faculty, University of Bonn, Bonn, Germany; 6Institute of Pathology, University Hospital Cologne, Cologne, Germany; 7Institute for Clinical Genetics, Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany; 8National Center for Tumor Diseases Dresden (NCT/UCC), Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; 9German Cancer Consortium (DKTK), Dresden, Germany; 10Institute of Human Genetics, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany; 11Center for Hereditary Tumors, Bethesda Hospital, Duisburg, Germany; 12Department of Applied Tumor Biology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany; 13Clinical Cooperation Unit Applied Tumor Biology, German Cancer Research Center Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany; 14Institute for Medical Informatics, Biometry and Epidemiology (IMIBE), University Hospital Essen, Essen, Germany; 15Gemeinschaftspraxis für Hämatologie und Onkologie, Ruhr-University Comprehensive Cancer Center, Dortmund, Germany; 16Department of Human Genetics, Ruhr-University Bochum, Bochum, Germany; 17Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany; 18MGZ - Medical Genetics Center, Munich, Germany; 19Institute of Neuroscience and Medicine (INM-1), Research Center Jülich, Jülich, Germany; 20Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany; 21Center for Human Genetics, University of Marburg, Marburg, Germany; 22Institute for Medical Informatics, Statistics and Epidemiology (IMISE), University of Leipzig, Leipzig, Germany
Consortium: German Consortium for Familial Intestinal Cancer
Background/Objectives: The study investigates how a polygenic risk score (PRS) affects colorectal cancer (CRC) risk in individuals of different, clinically or molecularly defined risk groups, using a large cohort from the German Consortium for Familial Intestinal Cancer.
Methods: A total of 1,839 European-descendant individuals were stratified according to low, intermediate, or high PRS for the following risk groups: (i) Lynch syndrome [LS; n = 679 with CRC and n = 422 CRC-free carriers]; (ii) sporadic early-onset CRC [SEO-CRC; n = 518], (iii) CRC patients with positive family history [F-CRC; n = 220], and (iv) MSI/dMMR CRC [n = 144] vs. pMMR CRC [n = 485]. Logistic regression and Cox proportional hazards models were applied to estimate odds ratios and to compute lifetime incidences in comparison to 3,119 population based controls and sporadic late-onset CRC patients (≥60 years) [SLO-CRC; n = 97]).
Results: In all risk groups, CRC individuals showed significantly increased PRS compared to population controls (p < 0.02). The SEO-CRC and F-CRC risk groups both showed a two times increased CRC risk in the high PRS category (p < 0.001) corresponding to lifetime incidences of 23% and 13%, respectively. PRS did not differ significantly between SEO-CRC and SLO-CRC cases or between LS CRC patients and CRC-free LS carriers. Finally, non-LS individuals with MSI/dMMR tumors had a significantly lower PRS than individuals with pMMR tumors (p < 0.001), but no difference compared to LS CRC individuals.
Conclusion: Preliminary findings suggest PRS effectively stratifies CRC risk in specific groups, but larger studies are needed, particularly for SEO and SLO CRC cohorts, to further explore a PRS-based CRC risk stratification.
Grants:
Conflict of Interest: None declared
C05.5 Personalized peptide vaccination among 173 patients with glioblastoma
Pauline Latzer 1, Henning Zelba1, Florian Battke2, Annekathrin Reinhardt1, Borong Shao1, Saskia Biskup1
1Zentrum für Humangenetik Tübingen; 2CeGaT GmbH
Background/Objectives: Current treatment outcome of patients with glioblastoma (GBM) remains poor. Following standard therapy, recurrence is universal with limited survival.
Methods: GBM tumors from 173 patients were analysed for somatic mutations to generate a personalized peptide vaccine targeting tumor-specific neoepitopes. In agreement with their treating physician, patients added a personalized peptide vaccine to their treatment as an individual healing attempt. Patients were monitored from October 2015 until August 2023. We retrospectively evaluated their clinical courses and the results of their immune monitoring data.
Results: Among all patients, including 70 treated prior to progression (primary) and 103 treated after progression (recurrent), the median overall survival from first diagnosis was 31.9 months (95% CI: 25.0-36.5). Adverse events were infrequent and were predominantly grade 1 or 2. An immune response to at least one of the vaccinated peptides was detected in blood samples of 87 of 99 (88%) monitored patients. T-cell responses to vaccinated neoepitope peptides were durable in most patients. Significantly prolonged survival was observed for patients with multiple vaccine-induced immune responses (53 months) compared to those with no/low induced responses (27 months; P = 0.03).
Conclusion: Altogether, our results highlight that the application of personalized neoantigen-targeting peptide vaccine is feasible and represents a promising potential treatment option for GBM patients. This is the largest real-world observation involving GBM patients safely treated with a personalized peptide vaccine to date. This real-world observation will be translated into a clinical trial entitled NeoGBM to evaluate the specific contribution of the neoantigen vaccine.
Grants: none.
Conflict of Interest: None declared
C05.6 Comprehensive guidelines for the diagnosis, counselling, surveillance and clinical management of people with constitutional mismatch repair deficiency: a join effort from ERN GENTURIS and the European consortium Care for CMMRD
Chrystelle Colas1, Léa Guerrini-Rousseau2, Manon Suerink3, Richard Gallon4, Christian Kratz5, Eloise Ayuso6, Laurence Brugières2, Katharina Wimmer 7
1Institut Curie, France; 2Gustave Roussy Cancer Center, Villejuif, France; 3Leiden University Medical Centre, Leiden, Netherlands; 4Newcastle University, Translational and Clinical Research Institute, Newcastle, United Kingdom; 5Hannover Medical School, Pediatric Hematology and Oncology, Hannover, Germany; 6Patient representative, Paris, France; 7Medical University Innsbruck, Institute of Human Genetics, Innsbruck, Austria
Consortium: ERN Genturis CMMRD Guideline Group
Background/Objectives: Constitutional mismatch repair deficiency (CMMRD) firstly described 25 years ago confers an extraordinarily high and lifelong cancer risk, including hematologic, brain, and gastrointestinal tract malignancies, and is associated with several non-neoplastic features. Our understanding of this condition has improved and novel assays to assist CMMRD diagnosis have been developed. Surveillance protocols need adjustment taking into account recent observational prospective studies assessing their effectiveness. Response to immune checkpoint inhibitors and the effectiveness and toxicity of other treatments have been described. An update and collation of the different guidelines on diagnosis and clinical management of CMMRD into one comprehensive guideline was needed.
Methods: Seventy-two expert members of the European Reference Network GENTURIS and/or the European care for CMMRD consortium and one patient representative developed recommendations for CMMRD diagnosis, genetic counselling, surveillance, quality of life, and clinical management based on a thorough literature review and a modified Delphi process.
Results: Recommendations for the diagnosis of CMMRD provide indication criteria and strategies for testing, and define criteria for diagnosis. Recommendations for surveillance cover each CMMRD-associated tumour type and contain information on starting age, frequency, and surveillance modality. Recommendations for clinical management cover cancer treatment, management of benign tumours or non-neoplastic features, and chemoprevention. Recommendations also address genetic counselling and quality of life.
Conclusion: Based on existing guidelines and all currently available data, we present 82 recommendations to improve and standardise the care of CMMRD patients in Europe. These recommendations are not meant to be prescriptive and may be adjusted based on individual decisions.
Conflict of Interest: Chrystelle Colas honoraria or consultation fees from AstraZeneca, Léa Guerrini-Rousseau: None declared, Manon Suerink: None declared, Richard Gallon grants/research support from Cancer Research UK Catalyst and UK National Health Service, Christian Kratz: None declared, Eloise Ayuso: None declared, Laurence Brugières honoraria or consultation fees from ESAI and TAKEDA, Katharina Wimmer: None declared
C06 Skin and Bones
C06.1 Liquid Biospy identifies causes of cutaneous mosaicism and hemihypertrophy
Maximilian Witzel 1, Thomas Keßler1, Ariane Hallermayr1, Angela Abicht1, Teresa Neuhann1
1MGZ München, München, Germany
Background/Objectives: Mosaicism of skin with or without overgrowth remains a challenge in the pediatric and genetic practice. Sampling errors of blood DNA, skin biopsy, cultured fibroblast can hamper a correct diagnosis. Liquid biopsy offers a noninvasive tool, sampling whole body cfDNA.
Methods: Duplex Sequencing technology for detection of low frequency variants in plasma was applied in children with mosaic conditions. The multi-gene panel included AKT3, BRAF, CCND2, EPHB4, FGFR1, GNA11, GNAQ, GNAS, HRAS, KRAS, MAP2K1, MTOR, NRAS, PIK3CA, PIK3R2, RASA1, SMO and others (>20 genes).
Results: We identified relevant pathogenic variants with suballelic VAF in 6 patients: patient (P1) with the clinical diagnosis Klippel-Trenaunay syndrome (AKT1 gene, NM_001382430.1:c.49G>A, p.(Glu17Lys), VAF 0.6%; class 5), patient (P2) with cavernoma of basal ganglia and prominent vessels of the eye sclera (PIK3CA gene, NM_006218.4:c.277C>T, p.(Arg93Trp), VAF 0.14%, class 4), patient (P3) with hemihypertrophy, Naevus flammaeus and syndaktyly (PIK3CA gene, NM_006218.4:c.316G>T, p.(Gly106Cys), VAF 2%, class 4), patient (P4) with hemihypertrophy, hyperpigmentation (KRAS gene, NM_004985.5, c.35G>A p.(Gly12Asp), VAF 1%, class 5), patient (P5) with diffuse capillary malformation with overgrowth (PIK3CA gene, NM_006218.4:c.353G>A p.(Gly118Asp) class 4/5, VAF 5.1%) and patient (P6) with premature puberty, asymmetrical breast development, subcutaneous lipoma, diffuse hyperpigmentation and café-au-lait spots (PIK3CA gene, NM_006218.4:c.1093G>A p.(Glu365Lys), VAF 0.12%, class 4/5). Additional NGS sequencing (panel, exom or trio exome) of blood and skin fibroblast had remained unconclusive in all patients.
Conclusion: Liquid biopsy is noninvasive, precise and significantly increases diagnostic yield in patients with mosaic diseases with profound therapeutical implications, e.g. targeted therapy, cancer surveillance, counseling.
Conflict of Interest: Maximilian Witzel full time employed by MGZ München, Thomas Keßler full time employed by MGZ München, Ariane Hallermayr full time employed by MGZ München, Angela Abicht full time employed by MGZ München, Teresa Neuhann full time employed by MGZ München
C06.2 A novel form of Ehlers-Danlos syndrome with prominent vascular features in humans and mice caused by a heterozygous THBS2 mutation
Noam Hadar 1;2, Omri Porgador1;2, Idan Cohen2, Hilla Levi2, Vadim Dolgin1;2, Yuval Yogev1;2, Sufa Sued-Hendrickson1;2, Ilan Shelef3, Elena Didkovsky4, Ohad Shmuel Birk1;2;5, Marina Eskin-Shwartz1;5;6
1The Morris Kahn Laboratory of Human Genetics, Faculty of Health Sciences, Ben-Gurion University, Beer Sheva, Israel; 2The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University, Beer Sheva, Israel; 3Department of Radiology, Soroka Medical Center, and Faculty of Health Sciences, Ben-Gurion University, Beer Sheva, Israel; 4Department of Pathology, Rabin Medical Center, Petah-Tikva, and Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; 5Genetics Institute, Soroka Medical Center, Beer Sheva, Israel; 6Faculty of Health Sciences, Ben-Gurion University, Beer Sheva, Israel
Background/Objectives: Ehlers-Danlos syndromes (EDS) are a group of connective tissue disorders caused by collagen and collagen-interacting gene mutations. We studied a novel distinct type of dominantly inherited EDS with vascular features.
Methods: Clinical and histopathological phenotyping and genetic human studies followed by generation and analyses of CRISPR/Cas9 knock-in (KI) mice.
Results: We studied a three-generation pedigree displaying an apparently autosomal dominant phenotype of frequent joint dislocations, joint hypermobility, atrophic scarring, prolonged bleeding time, and age-related aortic dilatation and rupture. Light microscopy revealed highly disorganized collagen fibers in the reticular dermis and transmission electron microscopy (TEM) displayed irregularly shaped fibroblasts and endothelial cells, with abundant amorphous extracellular matrix (ECM) substance, especially near blood vessels. Genetic analysis demonstrated a disease-causing heterozygous variant in THBS2 (NM_003247.5:c.2686T>C, p.Cys896Arg). We generated knock-in (KI) mice harboring this mutation in the mouse ortholog. Heterozygous THBS2Cys896Arg KI mice exhibited clinical features similar to the human disease, as evidenced by comparable morphologic, histologic, and TEM findings, and prolonged bleeding times.
Conclusion: We delineate a new form of EDS with vascular features caused by a dominant THBS2 missense mutation. The phenotype observed in heterozygous THBS2Cys896Arg KI mice mirrors that in the human disease and that of Thbs2 homozygous null-mutant mice. THBS2 encodes Thrombospondin-2, a secreted homotrimeric matricellular protein that directly interacts with Matrix Metalloproteinase 2 (MMP2), an ECM-shaping protein, and mediates its clearance. THBS2 loss-of-function hinders MMP2 clearance, causing an increase in MMP2-mediated proteoglycan cleavage, which leads to ECM abnormalities as seen in the human and mouse disease.
Conflict of Interest: None declared
C06.3 Generation of alpl zebrafish models for detailed pathogenic analysis of the rare disease hypophosphatasia.
Regina Hark 1, Lukas Hekel1, Angela Borst1, Barbara Ohlebusch1;2, Franz Jakob3, Eva Klopocki1, Stephanie Graser3, Daniel Liedtke1
1Institute of Human Genetics, Biocenter, University of Würzburg, Würzburg, Germany; 2Zoological Institute, University of Cologne, Cologne, Germany; 3Bernhard-Heine-Centrum for Locomotion Research, Department of Musculoskeletal Tissue Regeneration, Orthopaedic Hospital König-Ludwig-Haus, University of Würzburg, Würzburg, Germany
Background/Objectives: Hypophosphatasia (HPP), caused by mutations in the gene ALPL, is a rare metabolic disorder resulting in osteogenic and neuronal symptoms. Established human in vitro and mouse models entail experimental limitations, which could be resolved by using zebrafish (Danio rerio) as a supplemental HPP model for in vivo analyses. Our work aims to establish and investigate transgenic zebrafish alpl knockout lines and thereby, provide insights into developmental deficits that have been poorly elucidated in patients so far.
Methods: We generated different alpl knockout zebrafish models using the CRISPR/Cas9 system and characterized these using macroscopic, microscopic, and molecular approaches (Sanger sequencing, in-situ hybridization, immunostaining, bone/cartilage staining, in-vivo observations).
Results: Successful alpl knockout was confirmed in these lines. Homozygous zebrafish develop a lethal phenotype at larval stage. Early developmental abnormalities were observed, including progressive body axis curving, lack of swimming orientation, jaw deformations and decreased mineralization of bone and teeth. Furthermore, immunostaining revealed altered axonal growth within the telencephalon, the dorsal root ganglia and in sensory neurons.
Conclusion: We successfully generated novel alpl knockout zebrafish lines, which display altered craniofacial and neuronal development, resembling severe HPP forms. The detailed characterization of these in vivo models will help to gain a deeper understanding of the associated neurological phenotype, which is currently neglected in HPP patients.
Grants: The project is funded by DFG grant nr. 397519724.
Conflict of Interest: Regina Hark: None declared, Lukas Hekel: None declared, Angela Borst: None declared, Barbara Ohlebusch: None declared, Franz Jakob: None declared, Eva Klopocki: None declared, Stephanie Graser: None declared, Daniel Liedtke DFG research grant
C06.4 Human embryonic single-cell RNA sequencing data identifies potential involvement of epithelial cells in non-syndromic cleft lip with/without cleft palate
Anna Siewert 1, Elisabeth Mangold1, Kerstin Ludwig1
1Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn
Background/Objectives: Non-syndromic cleft lip with/without cleft palate (nsCL/P) is one of the most common birth defects. Although over 45 risk loci have been identified, the function of the risk alleles at these loci and affected cell types are mostly unknown. Here, we identified nsCL/P-related cell types based on candidate genes from GWAS loci and generated co-expression networks to suggest novel candidate genes based on joint expression patterns.
Methods: We re-analyzed published single-cell RNA sequencing (scRNA-seq) data from unaffected human embryos (4-6 weeks) using Seurat. We investigated the association of cell types with the expression of nsCL/P GWAS candidate genes using single-cell disease relevance scores (scDRS) and generated co-expression networks from these cell types using hdWGCNA.
Results: We found that nsCL/P candidate genes are significantly associated with the epithelium (p < 0.0001), which furthermore showed significant within-cell type scDRS heterogeneity (p = 0.007). The marker genes of the associated epithelial cells comprised known nsCL/P candidate genes (e.g. IRF6, TFAP2A, ESRP1). Within the epithelium, we identified 18 co-expression modules, three of which we prioritized based on their nsCL/P candidate genes (e.g. IRF6, TFAP2A, ARHGAP29, etc. in one module) and which include interesting new candidate genes (e.g. CALD1, PDGFC).
Conclusion: Our results suggest the existence of a specific subset of epithelial cells involved in nsCL/P development. For some nsCL/P candidate genes, genetic interaction has already been shown (e.g. IRF6 and TFAP2A). Our co-expression networks, however, indicate further genetic associations and interactions between known and novel candidate genes that should be investigated.
Grants: DFG: LU-1944/3-1
Conflict of Interest: None declared
C06.5 Understanding the molecular bases of a new Chondrodysplasia with Multiple Dislocations case.
Miriam Villegas Villarroel 1, Céline Huber1, Genevieve Baujat2, Adeline Bonnard3, Corinne Collet1;2, Valérie Cormier-Daire1;2
1Université de Paris Cité, INSERM UMR1163, Imagine Institut, Molecular and physiopathological bases of osteochondrodysplasia, Paris, France; 2AP-HP, Necker Hospital, Service de médecine génomique des maladies rares, Centre de référence pour les maladies osseuses constitutionnelles, Paris, France; 3Robert-Debré Hospital, Genetics, Paris, France
Background/Objectives: Chondrodysplasia with multiple dislocations (CLM) form a group of rare disorders mainly characterized by joint laxity and large joint dislocations. Most of these disorders have been associated with pathogenic variants in genes encoding proteins involved in the proteoglycan biosynthesis. Our aim was to confirm the pathogenicity of variants identified in a novel gene in a patient with CLM.
Methods: Whole Exome Sequencing as well as protein expression and GAG production through DMMB assay in patient fibroblasts were performed. Using a specific DDR1 inhibitor (DDR1-IN-1), we further studied the consequences in human chondrocytes and osteoblast differentiation and mineralization through ALP and alizarin red staining.
Results: We identified a homozygous nonsense mutation (exon 13, c.1825C>T) in the DDR1 gene (Discoidin Domain Receptor Family, Member 1; MIM #600408) in one patient with hyperlaxity, multiple dislocations, and cerebellar hypoplasia.
We demonstrated in patient fibroblasts an important decrease in DDR1 mRNA, absence of the protein and a GAG production defect. Using DDR1-IN-1, we found an impairment of the osteoblast differentiation and mineralization, and a defect in WNT signaling pathway. We also identified an alteration in the IHH signaling pathway in chondrocytes.
Conclusion: Our results suggest that DDR1 is a new gene involved in the CLM group, through a proteoglycan biosynthesis defect.
Grants: FRM and ANR
Conflict of Interest: None declared
C06.6 RHOA-related mosaic ectodermal dysplasia: a large international cohort study and natural history data
Elodie Javey 1;2, Bénédicte DEMEER3, Evgenia Sklirou4, David Geneviève5, Laurence Perrin6, Didier Bessis7, Odile Boute8, jean-luc alessandri9, Marta Spodenkiewicz10, JACQUEMONT Marie-Line9, Julie Servel11, Tiffany Busa12, Bertille Bonniaud13, LYSE RUAUD14, BOURRAT Emmanuelle14, Juliette Dupont15, Erica Gerkes16, Roelineke J Lunsing17, Gareth Baynam18, Seema Lalani19, Ian Glass20, Ashley Lahr4, Aimee Allworth20, Haley Streff19, Irene Valenzuela Palafoll21, Marta Codina21, Philippe Kerschen22, sophie scheidecker1, Elise Schaefer23, Audrey Schalk1, Nicolas Chassaing24, Alice Goldenberg25, Daphné Lehalle26, Diana Rodriguez27, jehanne martel28, Pierre Vabres28;29, Paul Kuentz30, Juliette Piard30, Arthur Sorlin31
1CHU de Strasbourg - Hôpital Civil, Laboratoire de diagnostic génétique, Strasbourg, France; 2CHU de Besançon, Centre de Génétique Humaine, Besançon, France; 3CHU Amiens-Picardie, Centre d’activité de Génétique clinique et Oncogénétique, Amiens, France; 4UPMC Children’s Hospital of Pittsburgh, Pittsburgh, United States; 5Hospital Center University De Montpellier, Département Génétique Médicale, Maladies rares et médecine personnalisée, Montpellier, France; 6Hospital Robert Debré Ap-Hp, Département de génétique, Paris, France; 7Hospital Center University De Montpellier, Service Dermatologie, Montpellier, France; 8Hospital Center University De Lille, Service de génétique clinique, Lille, France; 9Hospital Center Félix Guyon, Saint-Denis, Reunion; 10C.h.u. De Saint-Pierre, Service de Génétique, Saint-Pierre, Reunion; 11C.h.u. De Saint-Pierre, Plateforme de coordination des Maladies Rares Réunion-Mayotte, Saint-Pierre, Reunion; 12Marseille University Hospital Timone, Département de génétique médicale, Marseille, France; 13Chu Dijon, Dijon, France; 14Hospital Robert Debré Ap-Hp, Service de dermatologie, Paris, France; 15Hospital de Santa Maria - Urgências Centrais, Serviço de Genética, Lisboa, Portugal; 16University Medical Center Groningen, Department of clinical genetics, Groningen, Netherlands; 17University Medical Center Groningen, Department of Child Neurology, Groningen, Netherlands; 18King Edward Memorial Hospital, Subiaco, Australia; 19Baylor College of Medicine, Houston, United States; 20Seattle Children’s Hospital, Seattle, United States; 21Vall d’Hebron University Hospital, Medical genetics department, Barcelona, Spain; 22Centre Hospitalier de Luxembourg, Service de Neurologie, Luxembourg, Luxembourg; 23Igma Institute Genetic Medical D’alsace, Service de génétique médicale, Strasbourg, France; 24Hospital Paule De Viguier, Service de Génétique Médicale, Toulouse, France; 25Hospital Center University De Rouen, Service de génétique, Rouen, France; 26Hospital Armand Trousseau Ap-Hp, Service de Génétique, Paris, France; 27Hospital Armand Trousseau Ap-Hp, Service de Neuropédiatrie, Paris, France; 28Chu Dijon, Centre de référence Maladies Rares Génétiques à Expression Cutanée (MAGEC), Dijon, France; 29Chu Dijon, Service de dermatologie, Dijon, France; 30CHU de Besançon, Génétique biologique constitutionnelle et cytogénétique, Besançon, France; 31Laboratoire national de sante, National Center of Genetics, Dudelange, Luxembourg
Background/Objectives: RHOA-related mosaic ectodermal dysplasia (MIM:618727) was first described in 2019 in 7 patients exhibiting syndromic pigmentary mosaicism, resulting from a postzygotic variation in the RHOA gene. Three subsequent articles described the phenotype of six other patients.
Clinical features included linear hypopigmentation, hemihypotrophy with facial asymmetry, scarring alopecia, acral, ocular and teeth anomalies. Hearing impairment was occasionally associated. Brain imaging revealed consistent morphological abnormalities, described as non progressive comprising leukoencephalopathy, ventricular dilation and dilation of Virchow-Robin spaces, without cognitive impairment.
Identification of a RHOA mosaic pathogenic variation is based on high-depth sequencing using DNA extracted from a biopsy of affected skin.
The aim of our work is to define and extend the clinical phenotype associated with pathogenic RHOA mosaic variants and to elucidate a possible progressive brain deterioration.
Methods: We established an international collaboration to collect detailed clinical and molecular data, brain images and photographs from patients with RHOA-related mosaic ectodermal dysplasia.
Results: We report a clinical and molecular study of 18 new and 13 previously published patients, including the first fetal case. Our results show new frequent clinical signs, digestive anomalies (67% of the patients) and neurodevelopmental disorders (40%). Other newly described clinical manifestations are focal epilepsy, nystagmus and corpus callosum abnormalities. Brain MRI shows progressive worsening in 70% of cases.
Conclusion: RHOA-related mosaic ectodermal dysplasia is a clinically recognizable entity, with a possible neurodevelopmental, cerebral and digestive involvement. Diagnosing and monitoring older patients could help better anticipate the progression of cerebral involvement.
Grants: none
Conflict of Interest: None declared
C07 Cardiovascular genetics
C07.1 Deciphering 5’UTR variants in ENG revealed for the first time a uCUG-creating variant in patients with Hereditary Hemorrhagic Telangiectasia and associated with Endoglin deficiency.
Omar Soukarieh 1;2, Clemence deiber2, Caroline Meguerditchian2, Carole Proust2, Maud Tusseau3, Alexandre Guilhem3, Shirine Mohamed4, Béatrice Jaspard-Vinassa1, Aurélie Goyenvalle5, Sophie Dupuis-Girod3, David-Alexandre Tregouët2
1Inserm U1034, Pessac, France; 2Bordeaux Population Health, Bordeaux, France; 3Hospices Civils De Lyon, Lyon, France; 4Chru Brabois Umj, Vandœuvre-lès-Nancy, France; 5Paris-Saclay University, Orsay, France
Background/Objectives: Upstream Open Reading Frames (upORFs) located in the 5′UTR of coding genes are key regulators of translation. Their alteration by non-coding 5’UTR variants can be pathogenic. Recently, we characterized 5 single nucleotide variants (SNVs) creating AUGs (uAUG-SNVs) in the 5′UTR of ENG causing Hereditary Hemorrhagic Telangiectasia (HHT). The created uAUGs are all in frame with the same stop codon located in the coding sequence (ustop-c.125). We here extend our analysis to all SNVs altering upORFs in the 5’UTR of ENG.
Methods: We in silico mutated all positions of the 5’UTR of ENG (n = 909 SNVs) and annotated those creating canonical and non-canonical upstream translation initiation sites (uTIS), and/or creating/deleting stop codons, by using an updated version of the MORFEE tool. We also performed a functional assay based on the expression of constructs containing the wild-type and mutant ENG transcript in human cells on a selection of SNVs to assess their effect on ENG protein.
Results: MORFEE annotated 328 variants creating or modifying upORFs, among which 8 additional variants create uAUGs and 102 create non-canonical uTIS in frame with the ustop-c.125. We demonstrated that 6/8 of uAUG-SNVs decrease ENG levels. Moreover, one of these 6 variants and one uCUG-SNV (c.-76C > T) have been identified in 2 unrelated French HHT patients. This latter, reported in ClinVar as variant of unkown significance, moderately decreases ENG levels in our functional assay.
Conclusion: Our results contributed to the molecular diagnosis of HHT. Our strategy could be applied in any other gene implicated in human diseases.
Grants: Lefoulon Delalande, ANR.
Conflict of Interest: None declared
C07.2 Somatic loss-of-function PIK3R1 and activating non-hotspot PIK3CA mutations associated with Capillary Malformation with Dilated Veins (CMDV)
Martina De Bortoli1, Angela Queisser1, Van Cuong Pham2, Anne Dompmartin-Blanchere3, Raphaël Helaers1, Simon Boutry1;4, Cathy Claus5, An-Katrien De Roo5;6;7, Frank Hammer8, Pascal Brouillard1, Salim Abdelilah-Seyfried2, Laurence Boon1;5, Miikka Vikkula 1;9;10
1De Duve Institute UCLouvain, Human Molecular Genetics, Woluwe-Saint-Lambert, Belgium; 2Institute of Biochemistry and Biology, Postdam, Germany; 3Caen-Normandy University, Department of Dermatology, Caen, France; 4Interuniversity Institute Of Bioinformatics, Brussels, Belgium; 5Center for Vascular Anomalies Saint-Luc UCLouvain, Division of Pathology, Brussels, Belgium; 6University Clinics Saint-Luc, Service d’anatomopathologie, Brussels, Belgium; 7Institute of Experimental and Clinical Research UCLouvain, Brussels, Belgium; 8University Clinics Saint-Luc, Department of Medical Imaging, Brussels, Belgium; 9Cliniques universitaires Saint-Luc (UCLouvain), Center for Vascular Anomalies, Bruxelles, Belgium; 10WEL Research Institute, WELBIO department, Wavre, Belgium
Consortium: Revamp - Leducq network
V.A. cure ITN network
Background/Objectives: Common capillary malformations (CMs) are red vascular skin lesions, most commonly associated with somatic activating GNAQ or GNA11 mutations. We focused on CMs lacking such a mutation to identify novel genetic causes.
Methods: We used targeted next-generation-sequencing on 82 lesions. Bioinformatic analysis was performed with Highlander to pinpoint likely pathogenic variants. Primary cells were isolated from two lesions and PI3K-AKT-mTOR and RAS-RAF-MAPK signaling were assessed by western-blot. Zfish was used to model lack of PIK3R1 and effects of identified likely pathogenic variants.
Results: We identified 9 somatic pathogenic variants in PIK3R1 and PIK3CA, encoding for the regulatory and catalytic subunits of the PI3K kinase. Re-characterization of the lesions unraveled a common phenotype: a pale Capillary Malformation associated with visible Dilated Veins (CMDV). Primary-endothelial cells from PIK3R1-mutated lesions unveiled an abnormal increase in AKT phosphorylation, effectively reduced by mTOR, AKT and PIK3CA-inhibitors. Endothelium-specific expression of PIK3R1 mutants in zebrafish embryos resulted in abnormal development of the posterior capillary-venous plexus.
Conclusion: CMDV emerges as a clinical entity associated with somatic pathogenic variants in PIK3R1 or PIK3CA (non-hotspot). The activated AKT signaling can be effectively reversed by PI3K-pathway inhibitors. The zebrafish model holds promise as a valuable tool for future drug screens aimed at developing patient-tailored treatments.
Grants: Fonds de la Recherche Scientifique - FNRS Grants T.0240.23, P.C005.22, T.0146.16 and P.C013.20; Fund Generet managed by the King Baudouin Foundation (Grant 2018-J1810250-211305); Walloon Region through the FRFS-WELBIO strategic research programme (WELBIO-CR-2019C-06); European Union’s Horizon 2020 research and innovation programme under grant agreement No 874708 (Theralymph).
Conflict of Interest: None declared
C07.3 SHOX2-dependent arrhythmias: Mechanistic insights from human iPSC-derived cardiomyocytes
Sandra Hoffmann 1, Kristin Rädecke1, Annette Löwen1, Simon A. Sumer1, Micha Böckers1, Fangfang Zhang2, Birgit Campbell2, Amelie Paasche3, Christian Goetz3, Sarah Huecker4, Adrian Chang5, Volker Eckstein6, Tatjana Dorn2, Christoph Dieterich5, Stefan Kirsch4, Christian Schaaf1, Constanze Schmidt3, Alessandra Moretti2, Gudrun Rappold1
1Institute of Human Genetics, Heidelberg, Germany; 2Klinikum rechts der Isar der Technischen Universität München, First Department of Medicine, Cardiology, München, Germany; 3University Hospital Heidelberg, Heidelberg Centre for Heart Rhythm Disorders, Heidelberg, Germany; 4Fraunhofer-Institute for Toxicology and Experimental Medicine, Division of Personalized Tumor Therapy, Regensburg, Germany; 5Klaus Tschira Institute for Integrative Computational Cardiology, Section of Bioinformatics and Systems Cardiology, Heidelberg, Germany; 6University Hospital Heidelberg, FACS Core Facility, Department of Medicine V, Heidelberg, Germany
Background/Objectives: The orchestrated contraction of cardiac muscle relies on regulatory networks, where transcription factors guide downstream effectors for proper cardiac conduction. The transcription factor SHOX2 plays a pivotal role in the development and function of the sinoatrial node, emerging as a key pacemaker gene linked to conduction-related diseases. With the identification of SHOX2 variants in patients with early-onset atrial fibrillation and sinus node dysfunction, these variants have undergone functional characterization in various animal model systems.
Methods: To simulate SHOX2-related conduction dysfunction in a human system, iPSCs were generated from two different atrial fibrillation SHOX2-deficient patients, along with isogenic control lines. Differentiation of these cells into nodal and atrial cardiomyocytes enabled a comprehensive analysis using gene expression profiling, scRNA sequencing, and electrical phenotyping to uncover variant-specific changes contributing to the disease phenotype.
Results: Our data revealed deregulated gene expression patterns and impaired differentiation capability in patient cells. Furthermore, action potential characteristics in the patient lines showed changes attributed to differential ion channel expression, particularly deregulation of Na+ and K+ channels, unveiling novel disease mechanisms and therapeutic targets.
Conclusion: The SHOX2 human iPSC model mirrors the phenotypes observed in patients with atrial fibrillation and Shox2 animal models. This allowed us to gain valuable insights into the underlying molecular mechanisms that are affected by SHOX2 variant carriers and influence the occurrence and progression of conduction disorders.
Grants: Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) – 493726818 (to S.H.) and the Ministerium für Ernährung, ländlicher Raum und Verbraucherschutz, Baden Württemberg – 16(34)-0802/0415E (to S.H.).
Conflict of Interest: None declared
C07.4 Utility of genome sequencing and group-enrichment to support splice variant interpretation in Marfan syndrome
Alistair Pagnamenta 1, Susan Walker2, Dave Bunyan3, N Simon Thomas3, John Holloway4, Htoo Wai4, Dimitris Vavoulis1, Chris Kershaw5, Siddharth Banka5, Ed Blair6, Dahlia HopMeier7, Stephanie L. Curtis8, Catherine Fielden9, Julie Evans9, Rebecca Whittington9, Sarah Smithson10, Helen Cox11, Paul Clift12, Edward Sanders13, Meriel McEntagart14, Matina Prapa14, Suzanne Alsters14, Deborah Morris-Rosendahl14, John Dean15, Vivienne McConnell16, Abhijit Dixit17, Claire Turner18, Sian Ellard18, Emma Baple18, Diana Baralle4, ian berry9, Jenny Taylor1
1University of Oxford, Centre for Human Genetics, Oxford, United Kingdom; 2Genomics England, London, United Kingdom; 3Salisbury NHS Foundation Trust, Wessex Regional Genetics Laboratory, Salisbury, United Kingdom; 4University of Southampton, Human Development and Health, Faculty of Medicine, Southampton, United Kingdom; 5Manchester University Hospitals NHS Foundation Trust, Manchester Centre for Genomic Medicine, Manchester, United Kingdom; 6Oxford University Hospitals NHS Foundation Trust, Oxford Centre for Genomic Medicine, Oxford, United Kingdom; 7Chapel Allerton Hospital, Leeds, United Kingdom; 8University Hospitals Bristol and Weston NHS Foundation Trust, Bristol Heart Institute, Bristol, United Kingdom; 9North Bristol NHS Trust, Bristol Genetics Laboratory, Bristol, United Kingdom; 10University Hospitals Bristol NHS Foundation Trust, Department of Clinical Genetics, Bristol, United Kingdom; 11Birmingham Women’s and Children’s Hospital, West Midlands Clinical Genetics Service, Birmingham, United Kingdom; 12Queen Elizabeth Hospital, Grown Up Congenital Heart Disease Unit, Birmingham, United Kingdom; 13University of Oxford, MRC Weatherall Institute of Molecular Medicine, Oxford, United Kingdom; 14St George’s University Hospitals NHS Foundation Trust, SW Thames Centre for Genomic Medicine, London, United Kingdom; 15NHS Grampian, Department of Medical Genetics, Aberdeen, United Kingdom; 16Belfast Health and Social Care Trust, Regional Genetics Service, Belfast, United Kingdom; 17Nottingham University Hospitals NHS Trust, Department of Clinical Genetics, Nottingham, United Kingdom; 18Royal Devon University Healthcare NHS Trust, Department of Clinical Genetics, Exeter, United Kingdom
Background/Objectives: Marfan syndrome (MFS) is a connective tissue disorder associated with skeletal abnormalities, lens dislocation and Hereditary Thoracic Aortopathy (HTA). Pathogenic/likely pathogenic FBN1 variants are found in >90% of affected individuals and as the gene comprises 66 exons, splicing variants represent a significant fraction of these.
Methods: To better quantify the impact of aberrant FBN1 splicing in MFS we performed a systematic analysis of ultra-rare variants using short-read data from the 100k genomes project.
Results: Utilising aggregate data for 78,195 individuals, 13,864 singleton SNVs in FBN1 were identified of which 21 were predicted to impact splicing (SpliceAI>0.5). Incidence in individuals recruited under HTA (9/703) was significantly elevated compared to that seen in non-HTA participants (12/77,492; OR = 84, p = 9.7x10-14), a finding which prompted several variants to be re-assessed. Although RT-PCRs for an individual harbouring c.1961-3T > G previously showed no splicing effect, repeating this with intronic primers detected a 133bp exon extension, (p.Asp654AlafsTer17). Another variant (c.-182 + 1G > A) predicted exon1 extension and creates an uAUG. Examples of SNVs that simultaneously alter transcription/translation are rare. Additional analyses uncovered a further 14 families harbouring 11 different FBN1 splice variants. 14/20 variants lay beyond the +/-8 splice regions and 4 families harboured the previously reported recurrent c.7820-2034A > G.
Conclusion: Together with reports of cryptic SVs disrupting FBN1, our findings highlight the importance of incorporating the analysis of FBN1 introns into first-line MFS testing. The enrichment seen is an exemplar that we hope will stimulate further discussions about how statistical support for specific variant classes should be considered as part of interpretation frameworks.
Grants: MRC (MR/W01761X/1)
Conflict of Interest: Alistair Pagnamenta: None declared, Susan Walker Employed by Genomics England, Dave Bunyan: None declared, N Simon Thomas: None declared, John Holloway: None declared, Htoo Wai: None declared, Dimitris Vavoulis: None declared, Chris Kershaw: None declared, Siddharth Banka: None declared, Ed Blair: None declared, Dahlia HopMeier: None declared, Stephanie L. Curtis: None declared, Catherine Fielden: None declared, Julie Evans: None declared, Rebecca Whittington: None declared, Sarah Smithson: None declared, Helen Cox: None declared, Paul Clift: None declared, Edward Sanders: None declared, Meriel McEntagart: None declared, Matina Prapa: None declared, Suzanne Alsters: None declared, Deborah Morris-Rosendahl: None declared, John Dean: None declared, Vivienne McConnell: None declared, Abhijit Dixit: None declared, Claire Turner: None declared, Sian Ellard: None declared, Emma Baple: None declared, Diana Baralle: None declared, ian berry: None declared, Jenny Taylor: None declared
C07.5 A proteome-wide Mendelian randomization study identifies potentially causal associations with ischemic stroke
Lazaros Belbasis 1, Jemma Hopewell1, Cornelia M. van Dujin1
1University of Oxford, Nuffield Department of Population Health (NDPH), Oxford, United Kingdom
Consortium: N/A
Background/Objectives: Identification of novel drug targets for ischemic stroke (IS) is important to tackle its increasing global burden. Proteins participate in multiple biological processes and serve as drug targets. We aim to identify proteins associated with IS using a Mendelian randomisation approach.
Methods: We identified the lead cis protein quantitative trait locus (pQTL) for each plasma protein in the two largest proteo-genomic studies in populations of European ancestry (Olink platform in the UK Biobank and SomaScan platform in the deCODE Health Study). We retrieved the genetic effect of the selected pQTLs on IS from the latest GWAS from GIGASTROKE consortium. We used the Wald ratio to estimate the causal effects of proteins on IS and assessed the confounding by linkage disequilibrium through co-localisation.
Results: We tested 2,671 proteins for an association with IS, and 18 were statistically significant at a 5% false discovery rate. Sixteen proteins showed evidence of co-localisation with IS. ABO, F11, FURIN, KNG1, LRP4, FGF5, TMPRSS5, and WARS were associated with a higher IS risk. GRK5, MMP12, MMUT, CEP85, KIAA0319, PROCR, SH3BGRL3 and TMEM106B were associated with lower IS risk.
Conclusion: We provided a comprehensive analysis of the effect of plasma proteome on IS, with two of the identified proteins (F11 and PROCR) being tested in clinical trials. Our additional work considering IS subtypes and vascular risk factors further elucidates the potentially causal role of proteins in stroke aetiology and its mediating mechanisms.
Grants: The study is supported by an Oxford BHF–CRE Intermediate Transition Research Fellowship.
Conflict of Interest: Lazaros Belbasis Lazaros Belbasis is supported by an NDPH Early Career Research Fellowship (until July 2024) and a BHF-CRE Intermediate Transition Research Fellowship after August 2024., Jemma Hopewell: None declared, Cornelia M. van Dujin: None declared
C07.6 Enhancing 10-year cardiovascular disease prediction using polygenic risk scores and metabolomic biomarker scores
Scott C. Ritchie 1, Xilin Jiang1, Lisa Pennells1, Yu Xu1, Claire Coffey1, Yang Liu1, Praveen Surendran1, Savita Karthikeyan1, Samuel Lambert1, John Danesh1, Adam Butterworth1, Angela Wood1, Stephen Kaptoge1, Emanuele Di Angelantonio1, Michael Inouye1
1University of Cambridge, Public Health and Primary Care, Cambridge, United Kingdom
Background/Objectives: Polygenic risk scores (PRS) and metabolomic biomarker scores have shown promise for improving cardiovascular disease (CVD) prediction, but have not yet been evaluated in the context of European-recommended clinical risk factor prediction models (SCORE2) and guidelines for statin initiation for 10-year CVD prevention.
Methods: PRS were compared and combined with metabolomics scores in 168,517 UK Biobank participants (5,096 non-fatal and fatal CVD over 10-years of follow-up) 40–69 years of age without previous CVD, diabetes, or statin treatment. Improvement over SCORE2 in risk discrimination was assessed using Harrel’s C-index and stratification at clinically relevant risk thresholds using categorical net reclassification. Population modelling was subsequently applied to estimate the impact on 10-year CVD prevention if applied at scale.
Results: Risk discrimination of SCORE2 (C-index: 0.718) was similarly improved by both PRSs (ΔC-index 0.009 [0.007–0.012]) and metabolomics biomarker scores (ΔC-index: 0.011 [0.009–0.014]); capturing orthogonal information that combined yielded ΔC-index 0.019 (0.016–0.022) for 10-year CVD. Concomitant improvements in risk stratification following guidelines for statin initiation were observed, with PRSs and NMR scores yielding a combined improvement in net case reclassification of 13.04% (11.67–14.41%). Applied in population modelling, the number of CVD events prevented per 100,000 screened increased from 201 to 370 (ΔCVDprevented: 170 [158–182]) while essentially maintaining the number of statins prescribed per CVD event prevented.
Conclusion: Combining PRSs and metabolic biomarker scores with SCORE2 moderately enhances prediction of first-onset CVD, and could have substantial population health benefit.
Grants: British Heart Foundation (RG/18/13/33946)
Conflict of Interest: Scott C. Ritchie: None declared, Xilin Jiang: None declared, Lisa Pennells: None declared, Yu Xu: None declared, Claire Coffey: None declared, Yang Liu: None declared, Praveen Surendran During the course of this project Dr. Praveen Surendran became a full-time employee of GSK Plc. All significant contributions to this study were made prior to this role and GSK Plc had no input to the study., Savita Karthikeyan: None declared, Samuel Lambert: None declared, John Danesh Professor John Danesh has received multiple grants from academic, charitable and industry sources outside of the submitted work., Professor John Danesh serves on scientific advisory boards for AstraZeneca, Novartis, and UK Biobank., Adam Butterworth Professor Adam S. Butterworth reports institutional grants from AstraZeneca, Bayer, Biogen, BioMarin, Bioverativ, Novartis, Regeneron and Sanofi., Angela Wood: None declared, Stephen Kaptoge: None declared, Emanuele Di Angelantonio: None declared, Michael Inouye: None declared
C08 Implementing genomic screening
C08.1 Perspectives of preimplantation genetic testing users in Belgium on the ethics of PGT for polygenic conditions
Maria Siermann 1;2, Joris Robert Vermeesch3, Taneli Raivio2, Arne Vanhie4;5, Karen Peeraer6, Olga Tsuiko3;7, Pascal Borry1
1KU Leuven, Department of Public Health and Primary Care, Centre for Biomedical Ethics and Law, Leuven, Belgium; 2University of Helsinki, Faculty of Medicine, Department of Physiology, Helsinki, Finland; 3KU Leuven, Department of Human Genetics, Leuven, Belgium; 4University Hospital Leuven, Department of Gynaecology and Obstetrics, Leuven University Fertility Centre, Leuven, Belgium; 5KU Leuven, Group Biomedical Sciences, Department of Development and Regeneration, Endometriosis and Reproductive Medicine (LEERM), Laboratory of Endometrium, Leuven, Belgium; 6KU Leuven, Department of Development and Regeneration, Leuven, Belgium; 7University Hospital Leuven, Center for Human Genetics, Reproductive Genetics Unit, Leuven, Belgium
Background/Objectives: Preimplantation genetic testing using polygenic risk scores (PGT-P) is an ethically controversial technology that aims to screen embryos for risk of developing polygenic conditions. It is crucial to investigate ethical considerations and the perspectives of PGT users regarding ethics of embryo selection procedures.
Methods: We performed a qualitative interview study with PGT-M/SR users in Belgium, consisting of 18 interviews (10 couples, 8 women, n = 28). The aim was to get insights into the viewpoints of PGT-M/SR users on the ethics of PGT-P.
Results: Participants emphasized the physical, psychological and practical difficulties of their PGT-M/SR trajectories. Because of these burdens and the probabilistic nature of PGT-P, most participants did not find PGT-P an attractive option. They were largely negative about PGT-P without indication and about knowing risk information. PGT-P was seen as introducing unnecessary worries, complex choices and additional responsibilities. Many participants felt that PGT-P was going too far and that not everything should be controlled. If PGT-P would be implemented, participants generally wanted it to be limited to serious familial genetic conditions and decision-making to be left to healthcare professionals.
Conclusion: Our findings demonstrate that, from the perspective of those with experience with PGT-M/SR, PGT-P is seen as distinctly different from PGT-M/SR and as ethically contentious. It is important that these viewpoints are considered with regards to potential PGT-P implementation and guidelines.
Grants: This project has received funding from the European Union’s Horizon 2020 research and innovation programme under the Marie Skłodowska-Curie grant agreement No 813707.
Conflict of Interest: None declared
C08.2 “This is going to change everything”: A mixed-methods study examining outcomes of reproductive genetic carrier screening within the first 12-months post-result
Erin Tutty 1;2, Alison Archibald1;2;3, Nigel Laing4;5;6, Edwin Kirk7;8;9, Belinda McClaren1;2, Sharon Lewis2;10, Martin Delatycki2;3;11
1Murdoch Children’s Research Institute, Genomics in Society, Parkville, Australia; 2The University of Melbourne, Department of Paediatrics, Parkville, Australia; 3Victorian Clinical Genetics Services, Parkville, Australia; 4Harry Perkins Institute of Medical Research, Nedlands, Australia; 5University of Western Australia, School of Biomedical Sciences, Crawley, Australia; 6PathWest Laboratory Medicine, Department of Diagnostic Genomics, Nedlands, Australia; 7Sydney Children’s Hospital, Centre for Clinical Genetics, Randwick, Australia; 8NSW Health Pathology Randwick, Genomics Laboratory, Randwick, Australia; 9University of New South Wales, School of Clinical Medicine, Sydney, Australia; 10Murdoch Children’s Research Institute, Reproductive Epidemiology, Parkville, Australia; 11Murdoch Children’s Research Institute, Bruce Lefroy Centre, Parkville, Australia
Background/Objectives: Responsibly implementing reproductive genetic carrier screening (RGCS) involves promoting reproductive autonomy and psychological adaption to results. To inform implementation, this study explored psychosocial and reproductive outcomes from an Australian project (‘Mackenzie’s Mission’) at 12-months post-result.
Methods: Mackenzie’s Mission offered RGCS to >10,000 reproductive couples, 9107 had RGCS. A survey captured psychosocial (reproductive confidence, anxiety, and regret) and reproductive outcomes. Data were analysed descriptively. Participants with an increased chance result were interviewed. Data were analysed using reflexive thematic analysis.
Results: 4984 individuals completed the survey, including 116 with an increased chance (representing 96 couples, response = 55.0%). Most had high reproductive confidence and minimal regret. Anxiety was elevated for increased chance participants (median = 38.3, scores 40-80 = clinically meaningful). 53.1% of increased chance couples felt results impacted the timing/method of conception. 30/96 were pregnant at result disclosure; 16 had prenatal diagnosis. 30 conceived post-result; 17 used pre-implantation genetic testing. Interviewees (N = 19) felt their result “change[d] everything” about their reproductive plans. Reproductive decision-making was an evolving “journey”. Making a decision increased reproductive confidence and aided psychological adaptation. Emotions came in “stages” based on participants’ reproductive pathway. For example, those using pre-implantation genetic testing expressed anxious anticipation to conceive. Most felt “grateful” for the result.
Conclusion: By 12-months post-result, people have found value in their result given the reproductive confidence and autonomy it provides. Raised anxiety while people with an increased chance make and enact reproductive decisions suggests the need for longitudinal care to promote long-term well-being.
Grants: Medical Research Future Fund (GHFM73390), Research Training Program scholarship.
Conflict of Interest: Erin Tutty: None declared, Alison Archibald Employed by VCGS, a not-for-profit provider of genetic testing., Nigel Laing Employed by PathWest, a genetic testing provider, Edwin Kirk Employed by NSW Health Pathology, a service that provides reproductive genetic carrier screening on a fee-for-service basis, and employed part time by Genea, a fertility company that refers patients for reproductive genetic carrier screening., Belinda McClaren: None declared, Sharon Lewis: None declared, Martin Delatycki Employed by VCGS, a not-for-profit provider of genetic testing.
C08.3 Defining next steps in the implementation of polygenic scores in Australia’s cancer genetics clinics: Professionals’ views on determinants, strategies and needs
Rebecca Purvis 1;2, Mary-Anne Young3;4, Natalie Taylor5, Paul James1;2, Laura Forrest1;2
1Peter MacCallum Cancer Centre and the Royal Melbourne Hospital, Parkville Familial Cancer Centre, Melbourne, Australia; 2The University of Melbourne, Sir Peter MacCallum Department of Oncology, Melbourne, Australia; 3Garvan Institute of Medical Research, Clinical Translation and Engagement Platform, Sydney, Australia; 4UNSW Medicine and Health, St Vincent Clinical Campus, School of Clinical Medicine, Sydney, Australia; 5University of New South Wales, School of Population Health, Sydney, Australia
Background/Objectives: Polygenic scores (PGS) capture a proportion of the genomic liability for cancer. However, there are significant evidence gaps regarding clinical implementation, with scant evidence regarding determinants and strategies. The study objective was to explore genetic healthcare professionals’ views on the clinical implementation of PGS in cancer genetics clinics, including expected barriers, enablers, and priorities.
Methods: We conducted multi-modal, semi-structured interviews with genetic healthcare professionals purposively sampled using professional networks across Australia. A process map was used as a co-design tool to encourage engagement. Deductive content analyses were conducted, informed by the Consolidated Framework for Implementation Research 2022 and the Theoretical Domains Framework.
Results: 27 professionals participated across sector roles. Participants were majority female (85.2%) with 14.6 years of experience on average (range 0.5-35 years). Participants raised concerns regarding compatibility with current care pathways, the relative advantage, cost-effectiveness and complexity of PGS, capability of deliverers and equity of access. Having a defined model of care supported by national guidelines, standardised reporting, stakeholder education and resources were top implementation priorities. Whilst professionals highlighted key outcomes, such as quick turnaround times, as indicators of implementation success, few discussed defined implementation strategies.
Conclusion: Overall, professionals were optimistic about the clinical implementation of PGS. There was complementation between professionals‘ implicit understanding of determinants and implementation frameworks, however, the steps towards clinical implementation in the sector were poorly defined. Leaders can utilitse prioritised determinants and outcome indicators to direct research resourcing and better define future implementation outcomes.
Grants: Peter Mac Foundation Lester Peters Award and an Australian Government RTP Scholarship.
Conflict of Interest: None declared
C08.4 Feedback from two major national studies involving over 2500 individuals on the issue of secondary findings from exome or genome sequencing
Eléonore Viora-Dupont 1;2, Françoise Robert3, Benedicte Gerard4, Aline Chassagne5;6, Aurore pelissier6;7, Marion Bouctot8, Marie-Laure Humbert8, Dominique Salvi7, Elodie Gauthier2;6, Stephanie Staraci9;10, Nicolas Meunier-Beillard6;8, Amandine Cadénes3, Myrtille Spetchian11, Baurand Amandine2, Caroline Sawka2, Geoffrey Bertolone2, Delphine Heron11, Boris Keren11, Damien Sanlaville3, Gaetan Lesca3;12, Christel Thauvin-Robinet1;2, Christophe Philippe1, Sylvie Odent13;14, Dominique Bonneau15, Didier LACOMBE16, Roseline Caumes17, Patrick Edery3;18, Sabine Sigaudy19, David Geneviève20, Bertrand Isidor21;22, Gael Nicolas23;24, Julien Thevenon25, Catherine Lejeune6;8, Christine Peyron6;7, Hélène Dollfus4, Marcela Gargiulo26;27, Christine Binquet6;8, Laurence Faivre1;2
1Hospital Center University Dijon Bourgogne, FHU TRANSLAD et équipe GAD INSERM UMR 1231, Dijon, France; 2Hospital Center University Dijon Bourgogne, Centre de Référence Anomalies du Développement et Syndromes Malformatifs Est, Dijon, France; 3Hospices civils de Lyon, Service de génétique clinique, Centre de Référence Anomalies du Développement et Syndromes Malformatifs Sud Est, Bron, France; 4CHU Strasbourg, Laboratoire de Biologie Moléculaire, Strasbourg, France; 5University of Burgundy Franche-Comté, Laboratory of Sociology and Anthropology (LaSA, EA3189), Besançon, France; 6Hospital Center University Dijon Bourgogne, FHU TRANSLAD, Dijon, France; 7University of Burgundy Franche-Comté, Laboratory of economy (LEDI), EA7467, Dijon, France; 8Hospital Center University Dijon Bourgogne, INSERM, CIC1432, module épidémiologie Clinique, Dijon, France; 9University of Paris, Sorbonne Paris City, Clinical Psychology Laboratory, Psychopathology, Psychoanalysis (EA4056, ED 261), Paris, France; 10GH APHP, Genetics Department, Reference Center for Hereditary Cardiac Disorders, Paris, France; 11APHP, Centre de Référence Anomalies du Développement et Syndromes Malformatifs, Paris, France; 12Univ Lyon, Univ Lyon 1, CNRS, INSERM, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène, Lyon, France; 13Université de Rennes, ERN Ithaca, Institut de Génétique et Développement de Rennes, CNRS, UMR 6290, Rennes, France; 14CHU de Rennes, Service de Génétique Clinique, Centre labélisé pour les Anomalies du Développement ouest, Rennes, France; 15University Hospital of Angers, Biochemistry and Genetics Department, Angers, France; 16CHU de Bordeaux, Reference Center AD SOOR, AnDDI-RARE, INSERM U 1211, Medical Genetics Department, Bordeaux, France; 17CHU de Lille, Clinique de Génétique, Lille, France; 18University of Claude Bernard Lyon 1, INSERM U1028, CNRS UMR5292, CRNL, GENDEV Team, Bron, France; 19Hôpital Timone Enfant, Département de Génétique Médicale, Marseille, France; 20CHU Montpellier, Reference Center for Rare Disease, Medical Genetic Department for Rare Disease and Personalize Medicine, Montpellier, France; 21CHU Nantes, Service de Génétique Médicale, Nantes, France; 22Université de Nantes, CNRS, INSERM, L’Institut du Thorax, Nantes, France; 23Université de Rouen Normandie, Inserm U1245, Rouen, France; 24CHU Rouen, Department of Genetics, Rouen, France; 25CHU de Grenoble, Service de génétique médicale, Grenoble, France; 26Université Paris Cité, Laboratoire de Psychologie Clinique, Psychopathologie, Psychanalyse, Boulogne-Billancourt, France; 27Pitié-Salpêtrière University Hospital, Institute of Myologie, Paris, France
Consortium: FIND Consortium and DEFIDIAG-DS Consortium
Background/objectives: In the context of global debates on reporting additional findings (AF) such as incidental or secondary findings, particularly in Europe and France, our team has conducted extensive studies. The FIND study, involving 340 patients across three centers and using exome sequencing, examined patient preferences and the medical, organizational, and psychological challenges associated with reporting three categories of SF: “actionable” diseases, genetic counseling, and pharmacogenetics. After this first study, the DEFIDIAG-DS study, involving 2150 participants across 13 centers and using genome sequencing, delved into the impact of medical discourse and the decision-making process of parents concerning their health in the context of one category of “actionable” SF.
Methods: Mixed methodology combining quantitative and qualitative approaches over a longitudinal period of up to 1 to 3 years post-results disclosure, including interviews and specific questionnaires.
Result: Both studies revealed high patient satisfaction and a lack of regret, despite potential psychological impacts, especially in situations of vulnerability. Patients emphasized the importance of separate information and reporting sessions for primary diagnosis (PD) and AF within the same day. Coordination by geneticists in their care pathway with specialists was also underscored.
Conclusion: the results of these studies provide valuable insights into the perception and impact of AF: importance of information, reporting organization and care pathways coordination. Emphasizing the importance of multidisciplinary support for “opportunistic presymptomatic diagnosis,” these data underline the challenging nature of anticipating effects on patients, given that their primary motivation remains the search for an etiological diagnosis for PD and potential actionability.
Grant: FIND: PREPS (DGOS); DEFIDIAG-DS: DEFIDIAG (PFMG)
Conflict of Interest: None declared
C08.5 Opportunistic genomic screening in acute care settings: pilot and process evaluation of a two-step model
Melissa Martyn 1;2;3, Ling Lee1;2, Alli Jan2;4, Chole Mighton5;6, Sophie Bouffler7, Marc Clausen5;6, Yvonne Bombard5;6, Clara Gaff1;2;3, Zornitza Stark3;7;8
1Melbourne Genomics Health Alliance, Melbourne, Australia; 2Murdoch Children’s Research Institute, Parkville, Australia; 3The University of Melbourne, Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, Melbourne, Australia; 4NSW Health, Central Coast Local Health District, Gosford, Australia; 5St. Michael’s Hospital, Unity Health Toronto, Genomics Health Services Research Program, Toronto, Canada; 6University of Toronto, Institute of Health Policy, Management and Evaluation, Toronto, Canada; 7Australian Genomics, Parkville, Australia; 8Victorian Clinical Genetics Services, Parkville, Australia
Background/Objectives: Opportunistic genomic screening (OGS) is the analysis of clinical genomic data for additional health information. OGS has primarily been offered concurrent with diagnostic testing. An alternative approach is needed in acute care, where parents of children undergoing ultra-rapid sequencing report ‘feeling overwhelmed’. The Acute Care Genomics study piloted and evaluated an alternative approach.
Methods: Families (n = 232) were offered OGS after return of ultra-rapid diagnostic sequencing results: adult-onset for parents; childhood onset for (living) children; couple carrier screening. Parents had access to an online patient platform ‘Genetics Adviser’, modified for this study, followed by genetic counselling. Surveys were administered at 3 timepoints. Genetic counselling sessions were recorded and transcribed. Descriptive statistic and qualitative content analyses of Genetics Adviser utilisation, survey and transcript data focused on uptake and decision-making, understanding and service delivery preferences.
Results: Uptake of OGS was 40-42% across categories. Couple screening was an easier decision than paediatric or adult OGS (p = 0.007). Genetics Adviser users had higher knowledge scores than non-users, before and after counselling (p < 0.02). Differences in counselling sessions were noted, with less information seeking/giving and value clarification for Genetics Adviser users. After results return, 54% supported delaying OGS; 18% preferred a concurrent offer.
Conclusion: Two-step OGS in acute care is acceptable to parents. Couple screening is an easier decision than paediatric or adult OGS; concurrent couple screening merits further investigation. Genetics Advisor improves knowledge and impacts counselling; further research into the potential role of online patient platforms in OGS is warranted.
Grants: Medical Research Future Fund: GHFM76747. Royal Children’s Hospital Foundation: 2020-1259
Conflict of Interest: Melissa Martyn: None declared, Ling Lee: None declared, Alli Jan: None declared, Chole Mighton: None declared, Sophie Bouffler: None declared, Marc Clausen Genetics Adviser, Inc (part owner), Genetics Adviser Creative Director (part-time), Yvonne Bombard Genetics Adviser, Inc, Canadian Institutes of Health Research, Canadian cancer society, Ontario institute of Cancer research, Terry fox research institute, McLaughlin Centre of university of Toronto, Canada Research Chairs Program, Takeda, ASCO, ACMG, Clara Gaff: None declared, Zornitza Stark: None declared
C08.6 Challenges and legislation influencing genetic counselling practice in EU Member States
J. Matt McCrary 1, Denis Horgan2, Els Van Valckenborgh3, Marc Van Den Bulcke3, Anke Katharina Bergmann1
1Hannover Medical School, Department of Human Genetics, Hannover, Germany; 2European Alliance of Personalized Medicine, Brussels, Belgium; 3Sciensano, Cancer Center, Department of Epidemiology and Health, Bussels, Belgium
Consortium: CAN.HEAL Consortium
Background/Objectives: Integration of genetic testing into cancer care requires expert genetic counselling, a human interface point between patients and complex genetic technologies. Genetic counselling facilitates informed decision-making by patients and their families. However, its delivery is complicated by growing healthcare workforce shortages coinciding with an increasing inclusion of genetic testing in cancer prevention, diagnosis, and care. This project aims to provide an overview of genetic counselling practice, challenges, and legislation across EU Member States and a foundation for consensus European recommendations for delivering sustainable and equitable genetic counselling.
Methods: National legislative databases were searched in all 27 Member States. Interviews with relevant experts (medical geneticists, genetic counsellors, oncologists) and an online survey of cancer patient organizations from each EU Member State provide detailed insights into genetic counselling practice in each country.
Results: Legislation mandating genetic counselling exists in 22 of 27 Member States, albeit with significant variation in language addressing the ‘who, what, when, and how’ of counselling. Practice is similarly varied. Key barriers to counselling delivery noted by health professionals were remarkably consistent: genetic literacy of patients and non-geneticist health professionals (all 27 Member States); workforce capacity (25 of 27 Member States). Collection of data regarding key barriers from the patient perspective is ongoing, and will be presented at the conference.
Conclusion: This project highlights substantial variability in genetic counselling legislation and practice across the EU, but common challenges. Recommendations will focus on opportunities for collective European action to address these common challenges.
Grants: European Union Grant: 101080009
Conflict of Interest: None declared
C09 Omics and Pharmacogenetics
C09.1 Clinical pharmacokinetic study of novel CYP2C19 and CYP2D6 alleles identified by genome and exome sequencing in the Estonian Biobank
Jana Lass1;2, Laura Birgit Luitva1, Anette Caroline Kõre3, Raul Kokassaar4, Kristi Krebs1, Elisabet Størset5, Kadri Maal1, Magnus Ingelman-Sundberg6, Espen Molden5, Alar Irs7, Kersti Oselin4, Lili Milani 1;8
1Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia; 2Department of Pharmacy, Tartu University Hospital, Tartu, Estonia; 3Institute of Clinical Medicine, University of Tartu, Tartu, Estonia; 4Clinic of Oncology and Haematology, North Estonia Medical Center, Tallinn, Estonia; 5Center for Psychopharmacology, Diakonhjemmet Hospital, Oslo, Norway; 6Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institutet, Stockholm, Sweden; 7Heart Clinic, Tartu University Hospital, Tartu, Estonia; 8Genetics and Personalized Medicine Clinic, Tartu University Hospital, Tartu, Estonia
Background/Objectives: CYP2C19 and CYP2D6 metabolize over 30% of drugs. While the impact of common genetic variants on drug metabolism is well-established, understanding the effects of rare genetic variants remains challenging due to their low prevalence and absence of intermediate phenotypes for robust testing. In this context, we initiated a clinical study within the Estonian Biobank (EstBB) to investigate the effect of novel CYP2C19 and CYP2D6 alleles identified by genome and exome sequencing (GS and ES).
Methods: Novel rare genetic variants were identified in GS (n = 3,000) and ES (n = 2,500) data from EstBB participants. Putative loss-of-function and missense variants were annotated using LOFTEE and ANNOVAR. We used phased genotype data to call star alleles and invited individuals with different combinations of star alleles and novel variants to participate in the clinical study. The functional effect of the alleles was assessed by intake of omeprazole and metoprolol as probe drugs for CYP2C19 and CYP2D6 activity, respectively, among 114 eligible study participants.
Results: Probe drug and metabolite concentrations from ten time points per participant were quantified by mass-spectrometry. Based on differences in drug/metabolite area-under-the-curve ratios we confirmed the predicted effect of novel missense variants in CYP2C19 and CYP2D6, shifting normal metabolizers into intermediate metabolizers. All deletions in CYP2C19 had clear loss-of-function effects.
Conclusion: This study emphasizes the importance of rare genetic variants in explaining the ‘missing heritability’ in drug response, and how biobanks with the possibility to recall participants can contribute to discoveries in pharmacogenomics.
Grants: Estonian Research Council (PRG184), Swedish Research Council (2021-02732).
Conflict of Interest: None declared
C09.2 A method to estimate the effect of medications using genome-wide data
Ricky Lali 1;2, Wei Deng3;4, Shihong Mao2, Matteo Di Scipio2;5, Guillame Paré1;2;6
1McMaster University, Department of Health Research Methods, Evidence, and Impact, Hamilton, Canada; 2Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, Canada; 3McMaster University, Psychiatry and Behavioural Neurosciences, Hamilton, Canada; 4Peter Boris Centre for Addictions Research, Hamilton, Canada; 5McMaster University, Medicine, Hamilton, Canada; 6McMaster University, Pathology and Molecular Medicine, Hamilton, Canada
Background/Objectives: Understanding the effect of medications on clinically relevant biomarkers is crucial to assess their safety and efficacy. However, the only reliable way to estimate these effects is through comparison of pre and post dose levels, ideally in randomized populations. Though large-scale biobanks house rich data on baseline medication use, effect estimation is subject to indication bias.
Methods: We propose GENEPIE (GENotype-based Effect Prediction for Interventions and Exposures), an unbiased method to estimate medication effects using ultra-fast genome-wide outcome heritability optimization using a restricted maximum likelihood model. We tested the effect of 100 medications on lipids and waist-to-hip ratio in 408,069 British UK Biobank participants at baseline.
Results: Consistent with clinical trial data, GENEPIE determined that rosuvastatin conferred the strongest low-density lipoprotein cholesterol (LDL-C) reduction (-46.5%; 95% CI, -47.0 to -45.9%), atorvastatin the second strongest reduction (-45.4%; 95% CI, -45.6 to -45.1%) and that ezetimibe demonstrated the weakest (-25.1%; 95% CI, -25.9 to -24.3%). Standard epidemiological approaches resulted in indication bias, with marked attenuation of expected effects on LDL-C (e.g., -5.20%; 95% CI, -6.06 to -4.34% for ezetimibe). GENEPIE also revealed that metformin and orlistat decrease waist-to-hip ratio, as expected, and also showed a potential role of fibrates in weight loss.
Conclusion: GENEPIE leverages cross-sectional biobank-scale data to produce drug effect estimates robust to indication bias. Our results recapitulate known medication effects and show a potential effect of fibrates on weight, an observation that currently has basis only in model systems.
Grants: Not applicable
Conflict of Interest: Ricky Lali: None declared, Wei Deng: None declared, Shihong Mao: None declared, Matteo Di Scipio: None declared, Guillame Paré Received consulting fees from Bayer, Sanofi, Amgen, Novartis, and Illumina.
C09.3 Gene-environment interplay of socioeconomic indices and complex diseases
Fiona Hagenbeek 1, Max Tamlander1, Zhiyu Yang1, Tuomo Hartonen1, Kira Detrois1, Pekka Martikainen2;3, Nina Mars1;4, Andrea Ganna1;5, Samuli Ripatti1;5;6
1Institute for Molecular Medicine Finland, Helsinki, Finland; 2Helsinki Institute for Demography and Population Health, Faculty of Social Sciences, University of Helsinki, Helsinki, Finland; 3Max Planck – University of Helsinki Center for Social Inequalities in Population Health, Helsinki, Finland; 4Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, United States; 5Massachusetts General Hospital and Broad Institute of MIT and Harvard, Cambridge, United States; 6University of Helsinki, Department of Public Health, Helsinki, Finland
Consortium: FinnGen
Background/Objectives: Low socioeconomic status (SES) is a risk factor for several diseases. Here, we aim to explore whether differences in SES (educational attainment (EA) and occupation) modify polygenic risk for 19 complex diseases.
Methods: We used genomics, SES, and health data from approximately 280,000 Finnish individuals in the FinnGen study aged 35-80 at cohort entry. We considered 19 common diseases for which we calculated polygenic scores (PGS). EA was classified according to the 1997 international standard classification of education (ISCED) and categorized into low (ISCED ≤ 4), or high (ISCED ≥ 5) EA. Occupational information was categorized as either manual labor, self-employment, lower-level or high-level administrative occupations.
Results: Using Cox proportional hazard models, we observed higher PGS hazard ratios (per SD change in PGS) for the low EA group in 5 diseases (type 2 diabetes (T2D), rheumatoid arthritis, lung cancer, depression, and alcohol use disorder) and for the high EA group in breast, prostate, and all cancers. For example, in T2D the PGS hazard ratios per SD were 1.71 [95% CI 1.70,1.73] in low EA and 1.65 [1.63,1.67] in high EA groups and in breast cancer 1.53 [1.50,1.56] and 1.71 [1.67,1.74], respectively. Occupation results mostly mirrored the EA results. We are currently performing replication across European biobanks from the INTERVENE consortium.
Conclusion: We observed heterogeneity in PGS effects across SES groups which may improve equitable use of genetic information in healthcare.
Grants: This project is funded by the European Union’s Horizon 2020 research and innovation programme under grant agreement No 101016775.
Conflict of Interest: None declared
C09.4 From ATP7B variant spectrum to underdiagnosed cases: Estonian biobank supports preventing the preventable
Miriam Nurm 1, Anu Reigo1, Tarmo Annilo1, Toomas Toomsoo2;3, Margit Nõukas1, Tiit Nikopensius1, Vasili Pankratov1, Tuuli Reisberg4, Georgi Hudjašov1, Toomas Haller1, Neeme Tonisson1;5
1Institute of Genomics, University of Tartu, Tartu, Estonia; 2School of Natural Sciences and Health, Tallinn University, Tallinn, Estonia; 3Confido Medical Center, Tallinn, Estonia; 4Core Facility of Genomics, University of Tartu, Tartu, Estonia; 5Tartu University Hospital, Genetics and Personalized Medicine clinic, Tartu, Estonia
Background/Objectives: Wilson’s disease (WD) is a rare copper metabolism disorder with highly variable phenotypic presentation, caused by deleterious variants in the ATP7B gene. WD often remains underdiagnosed and undertreated, but could lead to debilitating health outcomes without treatment. The Estonian Biobank (EstBB) holds genotype and phenotype data of over 213,000 adult participants, thereby providing an excellent platform for studying the ATP7B variation spectrum in the Estonian population.
Methods: We identified and classified according to ACMG/AMP guidelines 49 pathogenic or likely pathogenic ATP7B variants in the cohort. We performed a comprehensive analysis of all available electronic health records (lab measurement results, ICD-10 codes, drug prescription data, epicrises) for EstBB participants diagnosed with copper metabolism disorders (ICD-10 code E83.0) (n = 59) or at high risk for developing WD (n = 17). Our study culminated in a recall project for biobank participants at high risk of WD for precise phenotyping (including family history, clinical biochemistry, neurological examination, etc.).
Results: The predominant pathogenic ATP7B variant in Europe, p.His1069Gln, has significantly higher prevalence in the Estonian population (AF 0.006) compared to other non-isolated populations (gnomAD global AF 0.001). Based on electronic health records, thirteen EstBB participants were ascertained as probably undiagnosed WD cases. Regardless of age, genotype, and prior WD diagnosis, all recall participants showed serum copper and ceruloplasmin values below reference, and 86.7% had signs of early to late neurodegeneration.
Conclusion: Population-based biobanks are excellent resources for combining continuous multilevel data and providing personalized feedback on disease risks, enabling better disease diagnostics in the future.
Grants: Estonian Research Council (#PRG555)
Conflict of Interest: None declared
C09.5 The multi-omics landscape of systemic inflammation in people living with HIV
Nienke van Unen 1;2, Javier Botey Bataller1;2;3, Jessica Dos Santos3, Maartje Cleophas3, Marc Blaauw3, Willem Vos3, Louise van Eekeren3, Albert Groenendijk3;4, Xun Jiang1;2, manoj Kumar Gupta1;2, Nhan Nguyen1;2, Cheng-Jian Xu1;2, Leo A.B. Joosten3, Mihai Gheorghe Netea3, Andre van der Ven3, Yang Li1;2;3
1Centre for Individualised Infection Medicine (CiiM), Hannover, Germany; 2Twincore, Zentrum für Experimentelle und Klinische Infektionsforschung GmbH, Hannover, Germany; 3Radboud University Medical Center, Nijmegen, Netherlands; 4Erasmus MC, Rotterdam, Netherlands
Consortium: 2000HIV
Background: People living with HIV (PLHIV) with viral suppression because of HIV-treatment have a higher susceptibility to non-AIDS co-morbidities, such as cardiovascular diseases (CVD), due to systemic inflammation. Systemic inflammation is potentially caused by immune activation by viral glycoproteins, but its underlying mechanisms are largely unknown.
Methods: We analyzed 1342 virally suppressed PLHIV participating in the 2000HIV project. By integrating five different omics layers together with immune function profiling, we deciphered mechanisms underlying persistent inflammation.
Results: Integrating epigenomics, transcriptomics, proteomics, metabolomics, and cytokine responses resulted in a set of co-morbidity related factors capturing inter-individual variation across layers. Specifically, we identified latent factors associated with CVD and accelerated aging. Furthermore, we used genomics to understand the sources of inter-individual variation in the different omics layers and identified 4,766 expression-QTLs, 1,155 protein-QTLs, 22 metabolite-QTLs and 2 cytokine-QTLs at study-wide significance level. The identified QTLs were largely concordant with existing QTL studies performed in healthy cohorts. Mendelian randomization analysis revealed baseline determinants of immune responses, such as gene TNFRSF8 and protein IL17D as regulators of immune responses to HIV. Lastly, by linking genetic variation to the latent factors we identified, we discovered that the NLRP12 locus regulates the inflammasome complex at different layers. This suggests a causal factor in determining systemic inflammation in PLHIV.
Conclusion: All in all, we discovered intricate molecular mechanisms underlying inter-individual variation in systemic inflammation in 1342 virally suppressed PLHIV, identified molecular QTLs in PLHIV, and pinpointed baseline regulators orchestrating the immune response to HIV stimulation.
Conflict of Interest: None declared
C09.6 Discovery of core genes for type 1 diabetes via genome-wide aggregation of trans-effects using UK Biobank proteomics study.
Xuan Zhou1, Andrii Iakovliev 2, Stuart McGurnaghan2, Athina Spiliopoulou1;2, Helen M Colhoun2, Paul McKeigue1;2
1Usher institute, Collage of Medicine and Veterinary Medicine, Edinburgh, United Kingdom; 2The Institute of Genetics and Cancer, College of Medicine and Veterinary Medicine, Edinburgh, United Kingdom
Background/Objectives: Type 1 diabetes (T1D) is a complex autoimmune disease involving destruction of insulin-producing beta cells. Our objective is to identify putative “core” effector genes for T1D using genome-wide aggregated trans-effects (GATE) analysis to inform drug discovery.
Methods: We used pQTL summary statistics from UKB-PPP and deCODE to compute GATE scores in a target dataset (4964/7497 cases/controls) from the SDRNT1BIO and the Generation Scotland respectively. We tested for association between T1D and GATE scores, one protein at a time, and identified core genes based on the strength of the association, the number of independent trans-loci contributing to each score and retained core genes with uncorrelated GATE scores.
Results: We identified 16 putative core genes at P values < 10-6, 13 were novel compared with our previous analysis. The strongest finding is PDCD1 (LogOR = 0.19, P = 10-24) followed by CXCL10, LGALS9, PRSS27, FASLG, IDO1, SDK2, AGR2, FGF19, SELL, TCN1, and KLRB1. Of these, 7 are in immune pathways, 6 are expressed on immune cells, 2 are expressed on NK-cells. Expression of PD-1, encoded by PDCD1, measured among 253 T1D cases in UKB-PPP is strongly associated with T1D (LogOR = 0.47, P = 2x10-15).
Conclusion: We highlight the role of adaptive (PDCD1, LGALS9, IDO1) as well as innate (FASLG, KLRB1) immune pathways in T1D, and suggest new targets for drug development including PDCD1 -- immune checkpoint in a clinical trial for rheumatoid arthritis. Our investigation substantiates the “omnigenic” model and sheds light on the pathogenesis of T1D.
Grants: Diabetes UK Project Grant [15/0005301]; Cross Disciplinary Fellowship (XDF) Programme [MC_FE_00035]
Conflict of Interest: None declared
C10 Genomes for the undiagnosed
C10.1 The Australian national genomic autopsy study: a summary of results, outcomes and instructive families from 406 trios/quads.
Thuong Ha1;2, Alicia Byrne1;3;4, Peer Arts1;5, Karin Kassahn5;6, Lynn Pais4;7, Anne O’Donnell-Luria4;7, Milena Babic8, Mahalia Frank1, Jinghua Feng3;9, Paul Wang2, David Lawrence2;6, Leila Eshraghi1, Luis Arriola1;2, John Toubia2, Hung Nguyen6, Alison Gardiner1;5, Jarrad Dearman10, Hannah Kovilpillai10, Genomic Autopsy Study Research Network11, George McGillivray12, Jason Pinner13, Fiona McKenzie14;15, Rebecca Morrow1;16, Jill Lipsett16, Nick Manton16, Yee Khong5;16, Theresa Power16, Lynette Moore5;16, Jan Liebelt5;10, Suzanne Sallevelt10, Lesley McGregor10, Shannon LeBlanc10, Arjan Bouman10, Andreas Schreiber2;3;17, Sarah King-Smith1;11, Tristan Hardy5;6, Matilda Jackson1;11, Hamish S. Scott1;2;3;5;11, Christopher P. Barnett 5;10;11
1Centre for Cancer Biology, Genetics and Molecular Pathology, Adelaide, Australia; 2Centre for Cancer Biology, ACRF Genomics Facility, Adelaide, Australia; 3University of South Australia, UniSA Clinical and Health Sciences, Adelaide, Australia; 4Broad Institute of MIT and Harvard, Program in Medical and Population Genetics, Cambridge, United States; 5University of Adelaide, Adelaide Medical School, Adelaide, Australia; 6SA Pathology, Genetics and Molecular Pathology, Adelaide, Australia; 7Boston Children’s Hospital, Division of Genetics, Boston, United States; 8Centre for Cancer biology, Genetics and molecular pathology, Adelaide, Australia; 9Centre for Cancer biology, ACRF Genomics Facility, Adelaide, Australia; 10Women’s and Children’s Hospital, Paediatric and Reproductive Genetics Unit, North Adelaide, Australia; 11Australian Genomics, Melbourne, Australia; 12Murdoch Children’s Research Institute, Victorian Clinical Genetics Services and Royal Women’s Hospital, Melbourne, Australia; 13Sydney Childre’s Hospital Network, Centre for Clinical Genetics, Randwick Sydney, Australia; 14Genetics Services of Western Australia, Perth, Australia; 15University of Western Australia, School of Paediatrics and Child Health, Perth, Australia; 16SA Pathology at Women’s and Children’s Hospital, Anatomical Pathology, North Adelaide, Australia; 17University of Adelaide, School of Biological Sciences, Adelaide, Australia
Consortium: Genomic Autopsy Study Research Network
Background/Objectives: The cause of pregnancy loss and perinatal death remains unexplained in ~ 25% of cases, despite a high perinatal autopsy rate in Australia. The genomic autopsy study is a national collaborative study using WES and WGS to identify genetic causes of fetal/newborn abnormalities that result in termination of pregnancy, death in utero or in the newborn period, in view to providing families with answers regarding cause and likelihood of recurrence.
Methods: WES and/or WGS was performed on prospective families referred to the Genetics unit (parent-fetus trios/quads). Priority cases were consanguineous families, fetuses with multiple malformations, and unexplained fetal/newborn death. Statistical, bioinformatic and experimental laboratory techniques were used to confirm variant causality.
Results: 406 prospective trios or quads have been recruited and sequenced from around Australia. 170/406 (42%) are either solved or have a strong candidate gene identified. Of the 170 solved families, 87 (51.2%) have been clearly solved by identification of a pathogenic mutation in a known gene. An additional 33/170 (19.4%) have a novel VUS in a known OMIM disease gene. Numerous instructive cases involving numerous genes (e.g., KIF14, SIK3, LAMC3, ARSL) will be presented.
Conclusion: Extensive genomic investigation of pregnancy loss and perinatal death should be offered as standard care, particularly when congenital abnormalities are present. Many families have benefitted from a clear diagnosis which could not be made on clinical grounds. Fetal genetic causes of late stillbirth are rare.
Grants: MRFF-GHFM Project Grant GHFM76777; NHMRC Project Grant ID APP1123341 CI Barnett and Scott are co-senior authors
Conflict of Interest: None declared
C10.2 Scalable automated reanalysis in rare disease: achieving high performance while limiting curation burden in diverse clinical and research cohorts
Matthew Welland1;2, Kaileigh Ahlquist3;4, Paul de Fazio5, Christina Austin-Tse3;6, Lynn Pais4, Laura Wedd1;2, Samantha Bryen1;2, Rocio Rius1;2, Michael Franklin2;7, Giles Hall6, Laura Gauthier3, Andrew Mallett8, Amali Mallawaarachchi7, Paul Lockhart9, Richard Leventer9;10, Ingrid Scheffer9;10, Katherine Howell9;10, Karin Kassahn11, Hamish S. Scott11, Christopher Semsarian12, Julie McGaughran13, Bryony Thompson14, Chirag Patel13, Greg Smith15, Anne O’Donnell-Luria3;4, Simon Sadedin5, Heidi Rehm3;4, Sebastian Lunke5, Jeremiah Wander15, Kaitlin Samocha3;4, Cas Simons1;2, Daniel MacArthur1;2, Zornitza Stark 5
1Garvan Institute of Medical Research, Centre for Population Genomics, Sydney, Australia; 2Murdoch Children’s Research Institute, Center for Population Genomics, Melbourne, Australia; 3Massachusetts General Hospital, Center for Genomic Medicine, Boston, United States; 4Broad Institute of MIT and Harvard, Program in Medical and Population Genetics, Cambridge, United States; 5Murdoch Children’s Research Institute, Victorian Clinical Genetics Services, Melbourne, Australia; 6Broad Institute of MIT and Harvard, Data Sciences Platform, Cambridge, United States; 7Garvan Institute of Medical Research, Sydney, Australia; 8James Cook University, College of Medicine and Dentistry, Townsville, Australia; 9Murdoch Children’s Research Institute, Melbourne, Australia; 10Royal Children’s Hospital, Melbourne, Australia; 11SA Pathology, Department of Genetics and Molecular Pathology, Adelaide, Australia; 12Royal Prince Alfred Hospital, Department of Cardiology, Sydney, Australia; 13Royal Brisbane and Women’s Hospital, Genetic Health Queensland, Brisbane, Australia; 14Royal Melbourne Hospital, Melbourne, Australia; 15Microsoft Research, Redmond, United States
Background: Reanalysis of existing genomic data in rare disease is highly effective in increasing diagnostic yield but remains limited by current manual approaches.
Methods: We developed an automated reanalysis framework, leveraging new, publicly available data on gene-disease relationships (PanelApp Australia) and variant pathogenicity (ClinVar). The framework was validated in a phenotypically diverse cohort of 400 rare disease patients from the USA and Australia. It was then applied prospectively in 2,002 unselected patients, who remained undiagnosed following clinical testing between 2018-2020 (1761 singletons, 241 trios; 573 genomes, 1429 exomes).
Results: In the validation cohort, our framework identified 159/196 (81%) of known diagnoses with data analysed as trios, and 145/196 (74%) if analysed as singletons. On average, it returned one candidate variant per trio, and two variants per singleton. Iterative reanalysis of unsolved patients (N = 204) identified five additional diagnoses in 12 months (2.5%). In the prospective cohort, the framework has identified 80 new diagnoses to date (4.0% yield): 42% due to new gene-disease relationships, 13% due to additional variant-level evidence, 40% due to process issues in the original analysis, and 5% due to bioinformatic improvements. All but one of the variants returned to diagnostic laboratories for evaluation have been classified as pathogenic or likely pathogenic.
Conclusion: We demonstrate the feasibility of an automated reanalysis model, optimised for high specificity and scalable to thousands of unsolved rare disease patients across multiple clinical and research environments. The model operates iteratively, raising the near-term prospect of delivering continuous and systematic automated reanalysis at scale.
Grants: GHFM APP2008820
Conflict of Interest: Matthew Welland: None declared, Kaileigh Ahlquist: None declared, Paul de Fazio: None declared, Christina Austin-Tse: None declared, Lynn Pais: None declared, Laura Wedd: None declared, Samantha Bryen: None declared, Rocio Rius: None declared, Michael Franklin: None declared, Giles Hall: None declared, Laura Gauthier: None declared, Andrew Mallett: None declared, Amali Mallawaarachchi: None declared, Paul Lockhart: None declared, Richard Leventer: None declared, Ingrid Scheffer: None declared, Katherine Howell: None declared, Karin Kassahn: None declared, Hamish S. Scott: None declared, Christopher Semsarian: None declared, Julie McGaughran: None declared, Bryony Thompson: None declared, Chirag Patel: None declared, Greg Smith Microsoft, Anne O’Donnell-Luria: None declared, Simon Sadedin: None declared, Heidi Rehm: None declared, Sebastian Lunke: None declared, Jeremiah Wander Microsoft, Kaitlin Samocha: None declared, Cas Simons: None declared, Daniel MacArthur GlaxoSmithKline, Insitro, Foresite Labs, Zornitza Stark: None declared
C10.3 Genome sequencing of a Pan-European cohort of 883 rare disease cases leads to new diagnoses in the Solve-RD project
Steven Laurie 1;2, Leslie Matalonga1;2, Ida Paramonov1;2, german demidov3, Galuh Astuti4, Wouter Steyaert4, Kornelia Ellwanger3, Stephan Ossowski3, Christian Gilissen4, Katja Lohmann5, Holm Graessner3, Richarda de Voer4, Ana Töpf6, Lisenka Vissers4, Alexander Hoischen4, Sergi Beltran1;7
1Centro Nacional de Análisis Genómico - CNAG, Barcelona, Spain; 2Universitat de Barcelona, Barcelona, Spain; 3Universitätsklinikum Tübingen – Institut für Medizinische Genetik und angewandte Genomik, IMGAG, Tübingen, Germany; 4Radboud University Medical Center, Nijmegen, Netherlands; 5University of Lübeck, Institute of Neurogenetics, Lübeck, Germany; 6John Walton Muscular Dystrophy Research Centre (JWMDRC), Translational and Clinical Research Institute, Newcastle, United Kingdom; 7Universitat de Barcelona, Departament de Genètica, Microbiologia I Estadística, Facultat, de Biologia,, Barcelona, Spain
Consortium: Solve-RD consortium
Background/Objectives: A key objective of the H2020 Solve-RD project is to provide a diagnosis for thousands of families affected by rare diseases (RD) across Europe, the majority of which have previously undergone exhaustive analysis of exome sequencing (ES) data, without resolution. To this end, 883 families underwent short-read genome sequencing (GS), with the goal that the broader and more even coverage provided by GS will allow the identification of previously missed causative variants.
Methods: DNA from affected probands (n = 883), and relatives (970) was sequenced to generate 30X coverage on a BGI-DNBseq platform. In parallel, deep phenotypic descriptions of probands were collated using Human Phenotype Ontology terms. Alignment and short variant calling were performed using BWA-MEM and GATK, and a suite of tools was applied to identify rare de novo, mtDNA, copy-number, structural, and short tandem repeat variants. Variants were prioritised for clinical interpretation by Solve-RD partnering European Reference Network (ERN) experts if they impacted upon candidate genes in curated lists provided by the submitting ERNs
Results: While clinical interpretation of identified variants remains ongoing, to date >10% of families for which prioritised variants have been interpreted have had their diagnostic odysseys brought to a conclusion as a result of this pan-European effort. We report here on some interesting cases highlighting why they were not resolved through analyses of the original genetic data.
Conclusion: Systematic analysis of short-read GS data, accompanied by detailed patient descriptions, can elevate the diagnostic rate in RD cases markedly, even where prior comprehensive analysis of genetic data proved inconclusive.
Grants: EU H2020-779257
Conflict of Interest: None declared
C10.4 Rare disease gene association discovery from burden analysis of the 100,000 Genomes Project data
Valentina Cipriani 1;2;3, Letizia Vestito1, Damian Smedley1
1William Harvey Research Institute, Queen Mary University of London; 2UCL Institute of Ophthalmology, University College London; 3UCL Genetics Institute (UGI), University College London
Consortium: the Genomics England Clinical Interpretation Partnership (GeCIP) leads and collaborators
Background/Objectives: High-throughput sequencing has accelerated gene discovery and molecular diagnosis in rare diseases. However, the genetic cause for 50% of Mendelian diseases remains unknown. We set out to address the “missing heritability” in rare diseases.
Methods: We have developed an Exomiser-based gene burden analytical framework to identify enrichments of rare, protein-coding, predicted pathogenic and segregating variants in probands of families with a specific phenotype relative to control families and applied it to 35,008 rare disease families recruited to the 100,000 Genomes Project (100KGP).
Results: Following in silico and clinical expert triaging, we identified 88 novel associations (38 with existing experimental evidence; 5 interim published confirmations via collaboration/independent study). Here, we describe 7 compelling associations: hypertrophic cardiomyopathy with DYSF and SLC4A3 (high/specific heart expression; existing associations to skeletal dystrophies/short QT syndrome); monogenic diabetes with UNC13A (known role in regulation of β cells; mouse model with impaired glucose tolerance); epilepsy with KCNQ1 (mouse model shows seizures; existing long QT syndrome association may be linked); early onset Parkinson’s disease with RYR1 (existing links to tremor pathophysiology; mouse model with neurological phenotypes); anterior segment ocular abnormalities associated with POMK (expression in corneal cells; zebrafish model with developmental ocular abnormalities); and cystic kidney disease with COL4A3 (high renal expression; prior evidence for a digenic/modifying role in renal disease).
Conclusion: Confirmation of our findings would lead to potential diagnoses in 456 molecularly undiagnosed cases within the 100KGP and additional patients worldwide, highlighting the clinical impact of a large-scale statistical approach to rare disease gene discovery.
Grants: 1R01HD103805-01;GACR 24-10324S;ASAP-000478;ASAP-000509.
Conflict of Interest: Valentina Cipriani: None declared, Letizia Vestito: None declared, Damian Smedley 1R01HD103805-01, D.S. was seconded to and received salary from Genomics England, a wholly owned Department of Health and Social Care company, from 2016 to 2018.
C10.5 Structural and non-coding variants increase the diagnostic yield of clinical whole genome sequencing for rare diseases
Jenny Taylor 1, Alistair Pagnamenta1, Carme Camps1, Edoardo Giacopuzzi2, John Taylor3, Eduardo Calpena4, Usha Kini3, Andrew Wilkie4, Niko Popitsch5
1University of Oxford, Centre for Human Genetics, Oxford, United Kingdom; 2Human Technopole, Milan, Italy; 3Oxford Universities Hospital Trust, Oxford, United Kingdom; 4University of Oxford, Weatherall Institute of Molecular Medicine, Oxford, United Kingdom; 5University of Vienna, Department of Biochemistry and Cell Biology, Vienna, Austria
Consortium: OxClinWGS Consortium
Background/Objectives: Whole genome sequencing (WGS) has revolutionised the diagnosis of patients with rare diseases. However, the diagnostic yields of large-scale clinical programmes are still modest at ~25-30%, in part because the full genome is not interrogated.
Methods: We undertook WGS on a cohort of 122 unrelated rare disease patients and their relatives (300 genomes) who were referred with a broad spectrum of rare diseases and had been pre-screened by arrays, panel and/or exome sequencing. We incorporated algorithms SVRare, ALTSPLICE and GREEN-DB in the bioinformatics pipeline to enhance the annotation of structural, splicing and regulatory variants, respectively.
Results: We identified pathogenic or likely pathogenic variants in 43/122 cases (35%). Seven structural variants were identified, three of which were in genes novel for disease or phenotype, whilst the remaining four had not been detected by prior clinical testing. Two were complex rearrangements.
Of sixteen splice site variants identified, seven were in non-canonical sites of known genes and two, were in genes, MCM10 and DOCK7, novel at the time of discovery. Our WGS results expanded the clinical spectrum for RMND1, changed the clinical diagnoses for six patients and informed treatment in a further eight patients, which for five patients was considered life-saving. Incidental findings were discovered in FBN1 and KCNQ1 genes and led to diagnoses of Marfan and long QT syndromes, respectively.
Conclusion: Our results emphasize the importance of analyzing the full genome sequence, including introns, to maximize clinical diagnoses for rare disease patients.
Grants: Health Innovation Challenge Fund [R6-388 / WT 100127]. Oxford NIHR Biomedical Research Centre
Conflict of Interest: None declared
C10.6 Genome sequencing: efficiently feeding the health care system.
Pierre Blanc 1, Boris Keren1;2, Benjamin Cogne1;3, Thomas Smol1;4, Jean-Madeleine de Sainte Agathe1;2, Jonathan Levy1;5, Audrey Briand-Suleau1, Lamisse Mansour-Hendili1, Paul Gueguen1;6, Tania Attie-Bitach1;7, Sophie Rondeau1;7, Giulia Barcia1;7, marion lesieur-sebellin1;7, Sophie Valleix1;7, François Lecoquierre1;8, Christele Dubourg1;9, Cyril Burin Des Roziers1;7, VALÉRIE MALAN1;7, Corinne Collet1;7, Olivier Grunewald1;4, Jérémy Bertrand1, Jérôme Champ1, Sylvain Dugat10, Amélie Souchet1, Jérôme Chansard1, Floriane Carallis1, Steve Ho Tam Chay1, Béréthé Diarra1, Ines Detrait1, Audrey Adechokan1, Marielle Letchimy1, Leslie Delachaux1, Camille Barette1, Sami Mahamoudi1, Nicolas Derive1, Thomas Rambaud1, Zinara Lidamahasolo1, Pierre Marijon1, Virginie Saillour1, Anaïs Lharidon1, Aurélien De Reynies1, Antonio RAUSELL1, Sacha Schutz1;11, Laurent Frobert1, Mouhamadou Niang1, Kevin Jousselin1, Marion Ducrotverdun1, Lara Vinauger1, Alban Lermine1, Michel Vidaud1
1Laboratoire SeqOIA; 2APHP.SU; 3CHU de Nantes; 4CHRU de Lille; 5APHP.Nord; 6CHU de Tours; 7APHP.Centre; 8CHU de Rouen; 9CHU de Rennes; 10Integragen; 11CHU de Brest
Background/Objectives: Genomic medicine is a cutting edge and rapidly expanding domain in the health care landscape. However, if genome sequencing data production is no longer a challenge, the clinical pathway back to the patient still represents a burden. In 2018, the French government launched a genomic medicine plan (PFMG 2025) and set up two national laboratories, accepting the idea that they could adopt distinct tools and strategies. We present the core output and choices of SeqOIA, the lab that operates in the northern half of France.
Methods: Between 2020 and 2023, short-read genome sequencing was performed on ~31,000 rare disease participants including ~12,000 affected individuals in 60 medical conditions.
Results: The overall diagnostic rate is 31%. Intellectual disability and congenital abnormalities are the most represented conditions, respectively with 3,142 and 2,674 prescriptions and a raw diagnostic yield of 35% and 31%. Over the first 3 years, the total pending results fraction is 10% and by the end of 2023, 82% of the prescriptions of the first semester of the year led to a report with a median turnaround time of 3.4 months. Interestingly, among our strategies some may be considered as orthogonal to worldwide current trends. Using these, the mean time from genome variant analysis to report delivery felt down to 50 minutes.
Conclusion: We propose an alternative operational lab framework that enables genome sequencing results to efficiently return to the clinicians.
Grants: The SeqOIA Laboratory is funded by the French Ministry for Solidarity and Health.
Conflict of Interest: None declared
C11 Novel genes in Intellectual disability
C11.1 KICS2 dysfunction impairs KICSTOR complex-mediated mTORC1 activation causing intellectual disability and epilepsy
Rebecca Buchert 1, Martin Burkhalter2, Linda Sofan1, Timo Roser3, Kirsten Cremer4, Javeria Raza Alvi5, Stephanie Efthymiou6, Tawfiq Froukh7, Sughra Guliyeva8, Ulviyya Guliyeva8, Moath Hamdallah9, Muriel Holder10, Rauan Kaiyrzhanov11, Doreen Klingler1, Mahmoud Koko12, Reza Maroofian13, Lars Matthies4, Joohyun Park1, Ana Velic14, Stephanie Spranger15, Tipu Sultan16, Hartmut Engels4, Holger Lerche12, Henry Houlden13, Alistair Pagnamenta17, Ingo Borggräfe18, Yvonne Weber12, Penelope Bonnen19, Olaf Riess1, Melanie Philipp2, Tobias Haack1
1University of Tübingen, Institute of Medical Genetics and Applied Genomics, Tübingen, Germany; 2University of Tübingen, Department of Experimental and Clinical Pharmacology and Pharmacogenomics, Division of Pharmacogenomics, Tübingen, Germany; 3Ludwig-Maximilians-University, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Munich, Germany; 4University of Bonn, Institute of Human Genetics, School of Medicine & University Hospital Bonn, Bonn, Germany; 5Children’s Hospital Lahore, Department of Pediatric Neurology, Institute of Child Health, Lahore, Pakistan; 6UCL Queen Square Institute of Neurology, Department of Neuromuscular Disorders, London, United Kingdom; 7Philadelphia University, Department of Biotechnology and Genetic Engineering, Amman, Jordan; 8MediClub Hospital, Baku, Azerbaijan; 9Farah Hospital, Amman, Jordan; 10Guy’s & St Thomas’ NHS Foundation Trust, Clinical Genetics Department, Guy’s Hospital, London, United Kingdom; 11UCL Institute of Neurology, Department of Neuromuscular diseases, London, United Kingdom; 12University of Tübingen, Department of Neurology and Epileptology, Hertie-Institute for Clinical Brain Research, Tübingen, Germany; 13University College London, Department of Neuromuscular Diseases, London, United Kingdom; 14University of Tübingen, Proteome Center Tübingen, Tübingen, Germany; 15Practice of Human Genetics, Bremen, Germany; 16Children’s Hospital & the Institute of Child Health, Lahore, Pakistan; 17University of Oxford, NIHR Oxford Biomedical Research Centre, Centre for Human Genetics, Oxford, United Kingdom; 18Ludwig-Maximilians-University, Dr. von Hauner Children̛s Hospital, Department of Pediatric Neurology and Developmental Medicine, Munich, Germany; 19Baylor College of Medicine, Molecular and Human Genetics, Houston, United States
Background/Objectives: Nutrient-dependent mTORC1 regulation upon amino acid deprivation is mediated by the KICSTOR complex, comprising SZT2, KPTN, ITFG2, and KICS2, which recruits GATOR1 to lysosomes. Previously, pathogenic SZT2 and KPTN variants have been associated with autosomal recessive intellectual disability (ID) and epileptic encephalopathy. Here we report on eight families with ID and biallelic loss of function variants in KICS2 (previously C12orf66).
Methods: Exome or genome sequencing identified five different variants as well as a 1.1 Mb deletion involving KICS2. We generated HEK293T cells carrying the respective variants using CRISPR/Cas9 gene-editing and performed mTORC1 activity assays and dimethyl-labeled phosphoproteomics. Furthermore, we depleted Kics2 in zebrafish using morpholinos and attempted rescue using either wildtype or mutant RNA.
Results: We identified biallelic KICS2 variants in eight index cases presenting with ID and epilepsy. In HEK293T cells, the identified variants demonstrated a deleterious impact on nutrient-dependent mTORC1 regulation, as confirmed by phosphoproteome analysis. Depletion of Kics2 in zebrafish resulted in ciliary dysfunction consistent with a role of mTORC1 in cilia biology which could not be rescued with mutant Kics2. Cilia analysis in zebrafish and human fibroblasts confirmed the pathogenicity of the identified variants.
Conclusion: Our genetic and experimental data provide evidence that KICS2 is a novel factor involved in ID presumably due to its dysfunction impacting mTORC1 regulation and cilia biology.
Grants: German Academic Exchange Service (DAAD, Project-ID 57166498), Fundació La Marató de TV3 (grant number 202019-32) and the Deutsche Forschungsgemeinschaft (German Research Foundation, grant numbers PH144/4-3, 6-1, 418081722, 433158657 and INST 37/1049-1).
Conflict of Interest: None declared
C11.2 De novo variants in the SF3B1 gene are responsible of a new spliceosomopathy with neurodevelopmental disorders.
Kevin Uguen1;2, Tiffany Bergot1, Marie Pier Scott-Boyer3, Solene Chapalain1, Severine Commet1, Claudia Gonzaga-Jauregui4, Susan Hiatt5, Devon Haynes6, Michael Kruer7;8, Patrick Sulem9, Cynthia Curry10, Trine Prescott11, Kohji Kato12, Merry Ferre13, Changlian Zhu14, Theresa Brunet15, Martine Doco-Fenzy16, Thomas Courtin17, Poirsier Céline18, Trine Bjørg Hammer19, Tony Roscioli20, Melissa MacPherson21, Jacqueline Leonard22, Dong Li22, Ian Glass23, Scott Ward24, Philippe M. Campeau25, Eric Lippert1, Laurent Corcos1, Claude Férec1, Arnaud Droit3, Sebastien Küry16, Delphine Bernard 1
1Université de Bretagne Occidentale, Inserm U1078, Brest, France; 2CHRU de Brest, Service de Génétique Médicale, Brest, France; 3Université de Laval, Research Center of Quebec CHU, Quebec, Canada; 4Universidad Nacional Autónoma de México, Laboratorio Internacional de Investigación sobre el Genoma Humano, Juriquilla, Mexico; 5HudsonAlpha Institute for Biotechnology, Huntsville, United States; 6Arnold Palmer Hospital for Children-Orlando Health, Division of Genetics, Orlando, United States; 7Phoenix Children’s Hospital, Phoenix, Pediatric Movement Disorders Program, Division of Pediatric Neurology, Barrow Neurological Institute, Phoenix, United States; 8University of Arizona College of Medicine, Departments of Child Health, Neurology, Cellular & Molecular Medicine and Program in Genetics, Phoenix, United States; 9deCODE Genetics/Amgen, Inc., Reykjavik, Iceland; 10University of California, San Francisco, Genetic Medicine, Fresno, United States; 11Telemark Hospital Trust, Department of Medical Genetics, Skien, Norway; 12Nagoya University, Department of Genetics, Research Institute of Environmental Medicine (RIeM), Nagoya; 13Laboratory Corporation of America Holdings (LabCorp), Durham, United States; 14University of Gothenburg, enter for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Gothenburg, Sweden; 15Technical University of Munich, Institute of Human Genetics, Munich, Germany; 16Nantes Université, CHU de Nantes, Service de Génétique médicale, Nantes, France; 17Institut du Cerveau - Paris Brain Institute, Paris, France; 18CHU Reims, Département de génétique médicale, Reims, France; 19Filadelfia Danish Epilepsy Centre, Department of Epilepsy Genetics, Dianalund, Denmark; 20Prince of Wales Hospital, New South Wales Health Pathology Randwick Genomics, Syndey, Australia; 21Faculty of Medicine and Dentistry, University of Alberta, Department of Medical Genetics, Edmonton, Canada; 22Children’s Hospital of Philadelphia, Divisions of Human Genetics, Philadelphia, United States; 23University of Washington, Department of Pediatrics, Seattle, United States; 24Vanderbilt University Medical Center, Department of Pediatrics, Division of Medical Genetics and Genomic Medicine, Nashville, United States; 25CHU Sainte-Justine, Centre Hospitalier Universitaire Saint-Justine Research Center, Montreal, Canada
Background/Objectives: SF3B1 (splicing factor 3b subunit 1) is an essential splicing factor that plays a role in recognizing the branch point within the snRNP U2 complex. Recurrent missense mutations in the SF3B1 gene are frequently found in cancers. To date, no constitutional mutations have been described. However, mutations in other splicing machinery genes, such as SF3B2, SF3B4, or PUF60, have been associated with various developmental syndromes.
Methods: We describe an international cohort of 23 patients with a syndromic neurodevelopmental disorder and in whom a de novo missense (n = 15) or loss-of-function variation (n = 8) in the SF3B1 gene was identified. We assessed the effect of missense variants on SF3B1 function by targeted splicing analyses using K562 and HEK293 cells. We also performed genome-wide analyses (RNA-Seq) on K562 cells with inducible expression of three different variants.
Results: Targeted analyses show that each missense variant rescues the splicing defect induced by SF3B1 silencing, suggesting that none can inactivate SF3B1 function. Only one variant has an effect on splicing similar to cancer-associated missense variations. Genome-wide analyses suggest that all three variants studied lead to splicing remodeling, primarily involving exon skipping. These data, combined with available information on the effects of SF3B1 loss of function, support the involvement of these variants in a developmental phenotype. We highlight a genotype-phenotype correlation, with a more severe phenotype in patients with missense variants.
Conclusion: In conclusion, this work describes a new syndrome within the spectrum of spliceosomopathies. Further transcriptomic analyses are needed to elucidate the mechanisms associated with these SF3B1 variants.
Conflict of Interest: None declared
C11.3 ANO4 is a novel gene causing developmental and epileptic encephalopathy or GEFS+ and temporal lobe epilepsy
Fang Yang1, Anais Begemann 2, Nadine Reichhart1, Akvile Haeckel3, Katharina Steindl2, Eyk Schellenberger3, Ronja Fini Sturm1, Magalie Barth4, Sissy Bassani2, Paranchai Boonsawat2, Thomas Courtin5;6, Bruno Delobel7, Boudewijn Gunning8, Katia Hardies9, Jennesson Mélanie10, Tarja Linnankivi11, Clément Prouteau4, Marta Spodenkiewicz12, Sandra Toelle13, Koen L. I. van Gassen14, Wim Van Paesschen15, Nienke Verbeek14, Alban Ziegler4, Markus Zweier2, Anselm Horn2;16, Heinrich Sticht16, Holger Lerche17, Sarah Weckhuysen9;18, Olaf Strauß1, Anita Rauch2
1Charité – Universitätsmedizin Berlin, Department of Ophthalmology, Berlin, Germany; 2University of Zurich, Institute of Medical Genetics, Schlieren-Zurich, Switzerland; 3Charité –Universitätsmedizin Berlin, Institute for Radiology and Children’s Radiology, Berlin, Germany; 4University Hospital of Angers, Department of Genetics, Angers, France; 5Sorbonne Université, INSERM, CNRS, Institut du Cerveau, Paris, France; 6Hôpital Pitié-Salpêtrière, DMU BioGe’M, AP-HP, Paris, France; 7Hopital Saint Vincent de Paul, GH de l’Institut Catholique de Lille, Lille, France; 8Stichting Epilepsie Instellingen Nederland, Zwolle, Netherlands; 9University of Antwerp, Applied & Translational Neurogenomics Group, Antwerp, Belgium; 10CHU Reims, Department of Pediatrics, Reims, France; 11University of Helsinki and Helsinki University Hospital, Epilepsia Helsinki, Helsinki, Finland; 12La Réunion University Hospital, Department of Genetics, Saint-Pierre, France; 13Children’s University Hospital Zurich, Department of Pediatric Neurology, Zurich, Switzerland; 14University Medical Center Utrecht, Department of Genetics, Utrecht, Netherlands; 15KU Leuven, Laboratory for Epilepsy Research, Leuven, Belgium; 16Friedrich-Alexander-Universität Erlangen-Nürnberg, Division of Bioinformatics, Institute of Biochemistry, Erlangen, Germany; 17University of Tübingen, Department of Neurology and Epileptology, Tübingen, Germany; 18Antwerp University Hospital, Department of Neurology, Antwerp, Belgium
Background/Objectives: Anoctamins are a family of Ca2+-activated proteins that assemble as homodimers and may act as ion channels and/or phospholipid scramblases with limited understanding of function and disease association. To date, no human disease has been associated with variants in ANO4 (alias TMEM16D).
Methods: We assembled a cohort of 7 patients with ANO4 variants and assessed them by variant mapping and structural modeling. We evaluated functional effects of all variants in a heterologous expression system by patch-clamp recordings, immunocytochemistry, and surface expression of annexin A5 as a measure of phosphatidylserine scramblase activity.
Results: ANO4 missense variants occurred de novo in 5 patients with fever sensitive developmental and epileptic encephalopathy and were inherited with reduced penetrance in 2 families with generalized epilepsy with febrile seizures plus or temporal lobe epilepsy. Variant mapping and structural analysis suggested a preliminary genotype-phenotype correlation. Experimentally, all ANO4 variants showed severe loss of ion channel function and mild loss of surface expression due to impaired plasma membrane trafficking. Increased levels of Ca2+-independent annexin A5 at the cell surface suggested an increased apoptosis rate in most mutant ANO4 expressing cells, but no changes in Ca2+-dependent scramblase activity were observed. Co-transfection with ANO4 wildtype suggested a dominant negative effect.
Conclusion: In summary, we present a novel genetic base for both, encephalopathic sporadic and inherited fever sensitive epilepsies, and a first hereditary disease caused by variants in ANO4.
Grants: SNSF 320020_179547 and University of Zurich ITINERARE to A.R., DFG STR480/14-1 to O.S., and FWO 1861424N and G056122N to SW.
Conflict of Interest: None declared
C11.4 Heterozygous variants in BICRA and BICRAL, sub-units of ncBAF complex, cause neurodevelopmental disorders in human
Yuwei Shi 1, Ruizhi Deng1, Sourav Ghosh2, Anita Nikoncuk1, Scott Barish3, Daryl Scott3, Hugo J Bellen3, Axel Schmidt4, Sarah Dyack5;6, Andrea Rideout5;6, Isabelle Maystadt7, Colombine Meunier7, YLINE CAPRI8, Viviana Lupo8, Jonathan Levy9, Alain Verloes8, Nathalie Couque9, Andrea Holubová10, Miroslava Hančárová10, Didier LACOMBE11;12, Vincent MIchaud11;12, Miquel Raspall13, Clément Prouteau14, Vincent Milon14, Katharina Steindl15, Francesco Brancati16, Antonio Novelli17, Dario Cocciadiferro17, Maria Cristina Digilio18, Kai Muru19;20, Hilde Van Esch21, Anita Gorrie22, Jessica Planner22, Hülya Kayserili23, Gael Nicolas24, Anne-Marie Guerrot24, Mona Grimmel25, Tobias Haack25, Haley McConkey2, Jessica Rzasa2, Jennifer Kerkhof2, Bekim Sadikovic2;26, Kristina Lanko1, Stefan Barakat1
1Erasmus University Medical Center, Department of Clinical Genetics, Rotterdam, Netherlands; 2London Health Sciences Centre, Verspeeten Clinical Genome Centre, London, Canada; 3Baylor College of Medicine, Department of Molecular and Human Genetics, Houston, United States; 4University Hospital Bonn, Institute of Human Genetics, Bonn, Germany; 5Dalhousie University, Department of Pediatrics, Halifax, Canada; 6IWK Health Centre, Maritime Medical Genetics Service, Halifax, Canada; 7Institute of Pathology and Genetics, Gosselies, Center for Human Genetics, Gosselies, Belgium; 8Hospital Robert Debré Ap-Hp, Clinical genetics unit, Genetic Department, Paris, France; 9Hospital Robert Debré Ap-Hp, Cytogenetics unit, Genetic Department, Paris, France; 10Charles University Second Faculty of Medicine and University Hospital Motol, Prague, Czech Republic; 11Hospital Center University De Bordeaux, Service de Génétique Médicale, Centre de Référence Anomalies du Développement et Syndrome Malformatifs, Bordeaux, France; 12Université de Bordeaux, Laboratoire Maladies Rares: Génétique et Métabolisme, INSERM U1211,, Bordeaux, France; 13Vall d’Hebron University Hospital, Barcelona, Spain; 14Angers University Hospital Center, Department of Medical Genetics and Mitovasc INSERM 1083, CNRS 6015, Angers, France; 15University of Zurich, Instiute of Medical Genetics, Zürich, Switzerland; 16University of L’Aquila, Department of Life, Health and Environmental Sciences, L’Aquila, Italy; 17Bambino Gesù Children’s Hospital, Translational Cytogenomics Research Unit, Roma, Italy; 18Bambino Gesù Children’s Hospital, Medical Genetics Unit, Medical Genetics Laboratory, Pediatric Cardiology Department, Scientific Rectorate, Roma, Italy; 19Tartu University Hospital, Genetics and Personalized Medicine Clinic, Tartu, Estonia; 20Tartu University, Institute of Clinical Medicine, Tartu, Estonia; 21UZ Leuven, Center for Human Genetics, Leuven, Belgium; 22Monash Medical Centre, Melbourne, Australia; 23Koç University Hospital, Istanbul, Türkyie; 24Normandie Université, UNIROUEN, Inserm U1245, CHU Rouen, Department of Genetics and reference center for developmental disorders, FHU G4 Génomique, Rouen, France; 25University of Tuebingen, Institute of Medical Genetics and Applied Genomics, Tübingen, Germany; 26Western University, Department of Pathology and Laboratory Medicine, London, Canada
Background/Objectives: SWI/SNF-related intellectual disability disorders (SSRIDDs) are rare neurodevelopmental disorders caused by variants in several members of SWI/SNF chromatin-remodeling complexes. A new ncBAF complex with unique core subunits BICRA (a.k.a. GLTSCR1) and its homolog BICRAL (a.k.a. GLTSCR1L) were recently discovered. Interestingly we previously showed that variants in BICRA cause a unique SSRIDD in human.
Methods: Through international collaborations, we collected clinical data of 33 additional individuals carrying variants in BICRA and 5 individuals with variants in BICRAL. Blood-derived DNA was used to generate an episignature. Human embryonic stem cells based disease models were generated and differentiated towards neural stem cells and excitatory neurons, followed by RNA-sequencing and morphology analysis.
Results: Individuals carrying rare variants in BICRA present with neurodevelopmental phenotypes, including intellectual disability, developmental delay, autism spectrum disorders and behavioral abnormalities as well as dysmorphic features.
Similarly, individuals with variants BICRAL, showed overlapping clinical phenotypes.
A predominantly hypomethylated DNA methylation profile was identified in patients with pathogenic BICRA variants. The RNA-sequencing analysis revealed that either reduced BICRAL or complete absence of BICRA leads to transcriptome change in neural stem cells, altering pathways related to axon guidance. Both BICRA and BICRAL haploinsufficiency result in morphologically increased branching and decreased neurites’ length in excitatory neurons.
Conclusion: Our findings showed that haploinsufficiency of both BICRA and BICRAL are causes of NDDs, that are related to a specific episignature. Furthermore, our functional studies provide insights in the pathomechanisms of these novel NDDs.
Grants: China Scholarship Council (CSC), ZonMw Vidi 09150172110002
Conflict of Interest: None declared
C11.5 A recurrent homozygous single amino acid deletion (p.Glu133del) in MED22 leads to a progressive neurodevelopmental disorder with microcephaly, dystonia, seizures and neurodegeneration
Elisa Cali 1, Lea Lescouzeres2, Mikhail Savitskiy3, MariaSavina Severino4, Sara Fortuna5, Miguel Soler6, Vladimir Katanaev3, Dylan Taatjes7, Henry Houlden1, Shunmoogum A. Patten2, Reza Maroofian1
1University College London; 2Université de Montréal; 3University of Geneva, Geneva, Switzerland; 4Giannina Gaslini Institute, Genova, Italy; 5IIT - Center for Robotics and Intelligent Systems, Genova, Italy; 6Università degli Studi di Udine, Udine, Italy; 7University of Colorado Boulder, Boulder, United States
Background/Objectives: Proper neurodevelopment relies on gene expression programs influenced by signalling molecules, transcription factors (TFs), and epigenetic processes. In this study, we investigate an ultra-rare homozygous single amino acid deletion (p.Glu133del) in MED22, a component of Mediator, a key regulator of transcription, gene expression and posttranslational epigenetic modifications. Disruption of Mediator has been implicated in various neurodevelopmental and neurodegenerative disorders.
Methods: We analysed the variant’s impact into the Mediator structure and function through cellular studies (Western blotting and IP-MS) and computational simulations. We investigated MED22 protein function generating knock-out zebrafish model and knock-in Drosophila model. We performed scRNA-seq on zebrafish mutants.
Results: We identified eight affected individuals from six unrelated families with a severe progressive neurodevelopmental disorder, characterized by global developmental delay, microcephaly, dystonia, seizures, and microcephaly. In vivo models of Drosophila melanogaster and zebrafish demonstrate significant developmental disruption, mirroring major aspects of the human disorder and displaying Parkinson-like features. scRNA-sequencing data obtained from zebrafish mutants implicate MED22 in dopaminergic network dysfunction.
Conclusion: We establish the first association of MED22 with a human disorder, through integration of clinical, genetic and neuroradiological data. We provide insights in the molecular dynamics of Mediator dysfunction and in the neuronal pathways that might be affected by MED22 dysfunction.
Grants: Wellcome Trust (WT093205MA and WT104033AIA).
Conflict of Interest: None declared
C11.6 Elucidating the clinical and genetic spectrum of inositol polyphosphate phosphatase INPP4A-related neurodevelopmental disorder
Lettie Rawlins 1;2, Reza Maroofian3, Stuart Cannon1, Muhannad Daana4, Mina Zamani5;6, Shamsul Ghani1;7, Joseph Leslie1, Nishanka Ubeyratna1, Nasar Khan8, Hamid Khan7, Annarita Scardamaglia3, Robin Cloarec9, Shujaat Ali Khan7, Muhammad Umair10;11, Saeid Sadeghian12, Hamid Galehdari5, Almundher Al-Maawali13;14, Adila Al-Kindi13;14, Reza Azizimalamiri12, Gholamreza Shariati6;15, Faraz Ahmed16, Amna Mohammed Al-Futaisi16, Alireza Sedaghat6;17, Mohammad Hamid18, Maha Zaki19, Abdullah Alhashem20, Makaram Obeid21, Amjad Khan22;23, Ahmad Beydoun21, Marwan Najjar21, Homa Tajsharghi24, Giovanni Zifarelli25, Peter Bauer25, Rose-Mary Boustany21, lydie BURGLEN26;27, Adam Gunning1, Ehsan Ghayoor Karimiani28, Joseph Gleeson29;30, Mathieu MILH9;31, Somaya Salah32, Jahangir Khan7, Volker Haucke33;34, Caroline Wright1, Lucy McGavin35;36, Orly Elpeleg32, Muhammad Shabbir7, Henry Houlden3, Michael Ebner33, Emma Baple1;2, Andrew Crosby1
1University of Exeter, Clinical and Biomedical Sciences, Exeter, United Kingdom; 2Royal Devon and Exeter Hospital, Peninsula Clinical Genetics Service, Exeter, United Kingdom; 3University College London, Department of Neuromuscular Diseases, London, United Kingdom; 4Clalit Health Care Services, Child Development Center, Jerusalem, Israel; 5Shahid Chamran University of Ahvaz, Department of Biology, Ahvaz, Iran; 6Narges Medical Genetic & PND Laboratory, Ahvaz, Iran; 7International Islamic University Islamabad (IIUI), Department of Biological Sciences, Islamabad, Pakistan; 8Aarhus University, Interdisciplinary Nanoscience Center, Aarhus, Denmark; 9Timone Infant Hospital, APHM, Marseille, France; 10University of Management and Technology, Department of Life Sciences, Lahore, Pakistan; 11King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia; 12Ahvaz Jondishapur University of Medical Sciences, Department of Pediatric Neurology, Ahvaz, Iran; 13Sultan Qaboos University, Department of Genetics, Muscat, Oman; 14Sultan Qaboos University Hospital, Genetic and Developmental Medicine Clinic, Muscat, Oman; 15Ahvaz Jondishapur University of Medical Sciences, Department of Medical Genetics, Ahvaz, Iran; 16Sultan Qaboos University, Deaprtment of Child Health, Muscat, Oman; 17Ahvaz Jondishapur University of Medical Sciences, Diabetes Research Center, Ahvaz, Iran; 18Pasteur Institute of Iran, Department of Molecular Medicine, Tehran, Iran; 19National Research Centre (NRC), Department of Clinical Genetics, Cairo, Egypt; 20Prince Sultan Military Medical City, Department of Radiology, Riyadh, Saudi Arabia; 21American University of Beirut, Beirut, Lebanon; 22University of Lakki Marwat, Department of Zoology, Lakki Marwat, Pakistan; 23University of Tübingen, Institute for Medical Genetics and Applied Genomics, Tübingen, Germany; 24University of Skövde, School of Health and Education, Skovde, Sweden; 25CENTOGENE GmbH, Rostock, Germany; 26Inserm, Developmental Brain Disorders Laboratory, Paris, France; 27Sorbonne University, Department of Genetics, Paris, France; 28St George’s University of London, Molecular and Clinical Sciences Institute, London, United Kingdom; 29University of California San Diego, Department of Neurosciences, La Jolla, United States; 30Rady Children’s Institute for Genomic Medicine, San Diego, United States; 31Aix-Marseille University, APHM, Marseille, France; 32Hebrew University Medical Center, Department of Genetics, Jerusalem, Israel; 33Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany; 34Free University Berlin, Department of Biology, Berlin, Germany; 35University Hospitals Plymouth NHS Trust, Plymouth, United Kingdom; 36University of Plymouth, Plymouth, United Kingdom
Background/Objectives: Biallelic INPP4A variants have been associated with severe neurodevelopmental disease in single case reports. Our studies expand and elucidate the clinical-genetic spectrum and provide a pathomechanistic explanation for genotype-phenotype correlations.
Methods: Clinical and genomic investigations of 26 individuals with INPP4A-related neurodevelopmental disorder were undertaken alongside molecular and in silico modelling and translation reinitiation studies.
Results: We characterize a clinically variable disorder with cardinal features including global developmental delay, severe-profound intellectual disability, microcephaly, limb weakness, cerebellar signs and short stature. A more severe presentation associated with biallelic INPP4A variants downstream of exon 4 has additional features of (ponto)cerebellar hypoplasia, reduced cerebral volume, peripheral spasticity, contractures, intractable seizures and cortical visual impairment. Our studies identify the likely pathomechanism of this genotype-phenotype correlation entailing translational reinitiation in exon 4 resulting in an N-terminal truncated INPP4A protein retaining partial functionality, associated with less severe disease. We also identified identical reinitiation site conservation in Inpp4a-/- mouse models displaying similar genotype-phenotype correlation. Additionally, we show fibroblasts from affected individuals exhibit disrupted endocytic trafficking pathways with largely unaffected Akt and mTORC1 signalling pathways, defining the likely biological basis of disease. Lastly, our work defined a new MANE Plus Clinical transcript (NM_001351425.2) as the most relevant to this neurodevelopmental disorder.
Conclusion: Our studies comprehensively characterise INPP4A-related neurodevelopmental disorder and suggest genotype-specific clinical assessment guidelines. We propose the potential mechanistic basis of observed genotype-phenotype correlations entails exon 4 translation reinitiation.
Grants: Wellcome Trust: 209083/Z/17/Z, 221951/Z/20/Z, 226083/Z/22/Z. Medical Research Council: G1002279, MR/S01165X/1, MR/S005021/1. Academy of Medical Sciences: SGL029\1079.
Conflict of Interest: None declared
C12 Germline and somatic mechanisms in cancer
C12.1 Comprehensive molecular analysis in advanced thymic epithelial tumors uncovers germline alterations in one fifth of patients impacting therapy recommendations
Lino Möhrmann1;2, Lysann Rostock1, Maximilian Werner1;2, Małgorzata Oleś3, Jonas Arnold 4;5;6, Nagarajan Paramasivam3, Korinna Jöhrens7, Luise Rupp8, Marc Schmitz8, Daniela Richter1;2;9, Sebastian Uhrig3, Martina Fröhlich3, Barbara Hutter3, Jennifer Hüllein3, Arne Jahn4;5;6, Marie Arlt4;5;6, Elena Möhrmann1;2, Dorothea Hanf1;2, Laura Gieldon10, Simon Kreutzfeldt11;12, Christoph Heilig11;12, Maria-Veronica Teleanu11;12, Daniel Lipka13, Katja Beck11;12;14, Annika Baude-Müller11;12;14, Andreas Mock15, Ivan Jelas16, Damian Rietke16, Marcel Wiesweg17, Christian Brandts18;19;20, Melanie Börries9;21, Anna Lena Illert22, Alexander Desuki23;24;25, Thomas Kindler23;24;25, Angela Krackhardt26, Christoph Benedikt Westphalen27;28, Petros Christopoulos29;30, Leonidas Apostolidis31, Albrecht Stenzinger32, Michael Allgäuer32, Olaf Neumann32, Irina Kerle1;2;33, Peter Horak11;12, Christoph Heining1;2, Heidrun Grosch30;34, Evelin Schröck4;5;6, Daniel Hübschmann3, Stefan Fröhling11;35;36, Hanno Glimm1;2;37
1TMO, NCT/UCC, a partnership between DKFZ, Faculty of Medicine and University Hospital Dresden and HZDR; 2TMO, Faculty of Medicine and University Hospital Dresden, Germany; DKTK, Dresden, Germany; 3Computational Oncology Group, Molecular Precision Oncology Program, NCT Heidelberg and DKFZ, Heidelberg, Germany; 4Institute for Clinical Genetics, University Hospital Dresden, Dresden, Germany; TMO, (NCT/UCC), a partnership between DKFZ, Faculty of Medicine and University Hospital Dresden and HZDR; 5ERN GENTURIS, Hereditary Cancer Syndrome Center Dresden, Germany; TMO, Faculty of Medicine and University Hospital Dresden, Germany; 6DKTK, Dresden, Germany; Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany; 7Department of Pathology, University Hospital Carl Gustav Carus, Dresden, Germany; 8Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany; 9German Cancer Research Center (DKFZ) Heidelberg, Germany; 10Institute of Medical Genetics, Carl von Ossietzky University, Oldenburg, Germany; 11Division of Translational Medical Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany; 12National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany; 13Section of Translational Cancer Epigenomics, Division of Translational Medical Oncology, German Cancer Research Center (DKFZ) and NCT Heidelberg, Heidelberg, Germany; 14Molecular Precision Oncology Program, NCT Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany; 15Institute of Pathology, Ludwig-Maximilians-Universität München (LMU), Munich, Germany; 16Charité Comprehensive Cancer Center, Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; 17Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Germany; 18Department of Medicine, Hematology/Oncology, University Hospital, Goethe University, Frankfurt, Germany; 19University Cancer Center Frankfurt (UCT), University Hospital, Goethe University, Frankfurt, Germany; 20German Cancer Consortium (DKTK), Heidelberg, Germany; 21Institute of Medical Bioinformatics and Systems Medicine, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg and DKTK, partner site Freiburg, a partnership between DKFZ and Medical Center - University of Freiburg; 22Department of Internal Medicine I, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany; 23University Cancer Center and Department of Internal Medicine III, University Medical Center Mainz, Germany; 24TRON-Translational Oncology, University Medical Center, Johannes Gutenberg University Mainz, Germany; 25German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, a partnership between DKFZ and University Medical Center Mainz, Germany; 26Klinik und Poliklinik für Innere Medizin III, Klinikum rechts der Isar, School of Medicine, Technical University of Munich (TUM), Munich, Germany; 27Department of Internal Medicine III, University Hospital, LMU Munich and Comprehensive Cancer Center, Munich, Germany; 28German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and University of Munich, Germany; 29Translational Lung Research Center, member of the German Center for Lung Research (DZL), Heidelberg, Germany; 30Department of Thoracic Oncology, Thoraxklinik, Heidelberg University Hospital and National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Germany; 31Department of Medical Oncology, NCT Heidelberg and Heidelberg University Hospital, Heidelberg, Germany; 32Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany; 33DKTK, Dresden, Germany; 34Translational Lung Research Center, member of the German Center for Lung Research (DZL), Heidelberg, Germany; 35National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany; DKTK, Heidelberg, Germany; 36Institute of Human Genetics, Heidelberg University, Heidelberg, Germany; 37Translational Functional Cancer Genomics, German Cancer Research Center (DKFZ) Heidelberg, Germany
Background/Objectives: The molecular landscape and genetic risk factors of malignant thymic epithelial tumors (TETs), a clinically challenging entity, are largely unknown. We characterized a cohort of patients with advanced TETs clinically and molecularly using a broad multiomics approach.
Methods: 46 thymic carcinomas (TC), 8 thymic neuroendocrine neoplasms (TNEN) and 27 thymomas from the prospective observational German multicenter precision oncology study DKFZ/NCT/DKTK MASTER were included. Parallel tumor and control genome or exome sequencing, RNA analysis and DNA methylation profiling were performed. A molecular tumor board assessed clinical actionability of somatic and germline alterations.
Results: We observed mutational heterogeneity between entities with targetable alterations being more frequent in TC. Assessment of germline variants within 142 cancer predisposition genes (CPGs) among 76 patients with available germline data revealed 14 pathogenic germline variants (PGVs) in 14 patients (18%) with WHO type B2 thymoma or higher. PGVs in eight autosomal dominant CPGs occurred in nine patients. PGVs in BRCA1 and MUTYH occurred more than once and there was one biallelic pathogenic MUTYH variant. Loss of heterozygosity was observed in five tumors with PGVs in autosomal dominant CPGs. PGVs impacted therapy recommendations in seven patients, all of them targeting impaired DNA damage response. We detected BRCAness-signature SBS3 in 24/67 (36%) samples. Therapy recommendations were applied in 28/68 (41%) cases.
Conclusion: Comprehensive molecular analysis provides clinically relevant information. Based on the higher PGV yield and potential clinical impact on patients and their relatives, we propose germline testing parallel to molecular tumor characterization for all patients with TC, TNEN and B2/B3 thymoma.
Grants: DKFZ-HIPO-H021
Conflict of Interest: None declared
C12.2 Looking beyond coding mutations: the role of regulatory noncoding sequences in hereditary diffuse gastric cancer
Celina São José 1, Marta Ferreira1;2, Lilian Cordova3, Ana Maria Pedro1, Raquel Santos1, Ricardo Amorim1, ana andré1, J. Garcia-Pelaez1, janine senz3, Pardeep Kaurah3, Fiona Puntieri4, Juliane Glaser4, david huntsman3;5, Stefan Mundlos4;6;7, Kasmintan Schrader3, Carla Oliveira1;8
1Instituto de Investigação e Inovação em Saúde, Porto, Portugal; 2Faculty of Science of the University of Porto, Computer Science, Porto, Portugal; 3University of British Columbia, Pathology and Laboratory Medicine, Vancouver, British Columbia, Canada; 4Max Planck Institute for Molecular Genetics, Berlin, Germany; 5Genetic Pathology Evaluation Centre, University of British Columbia and Vancouver General, Vancouver, British Columbia, Canada; 6Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany; 7Institute of Medical and Human Genetics, Charité Universitätsmedizin Berlin, Berlin, Germany; 8Faculty of Medicine of the University of Porto, Porto, Portugal
Background/Objectives: CDH1 or CTNNA1 germline inactivation explains 10-40% of families with hereditary diffuse gastric cancer (HDGC). Missing heritability prevents proper management and disease prevention in HDGC-suspected families. We hypothesized that defects in CDH1-regulatory elements contribute to HDGC missing heritability.
Methods: We called single-nucleotide/copy-number variants (SNV/CNV) from 19 HDGC probands whole-genome sequencing data, performed gene-ontology analysis, 4C-seq and ATAC-seq in normal stomach epithelia, CRISPR-Cas9, RT-PCR and flow cytometry in cell lines, immunohistochemistry and Microsatellite instability (MSI) analysis in tumours and enhancer assays in mouse embryos.
Results: We found a MLH1 2.7Kb germline deletion overlapping a stomach-specific regulatory element in 1/19 HDGC-suspected families. Gastric tumour displayed MLH1 protein loss and MSI. We crossed ATAC-seq (46.249 peaks) and 4C-seq (370 CDH1 interactor-regions) data from normal stomach with WGS data (1.882 rare CNVs) from 19 HDGC-suspected families. A CDH3 20kb-deletion and a 39bp-intergenic deletion downstream of CDH1 were mimicked with CRISPR-Cas9 (homozygous) triggering 100% or 50% CDH1 mRNA downregulation, respectively, similarly to a CDH1 coding deletion. Two sequences within the CDH3 CNV displayed enhancer activity in mouse embryos. Combined analysis of deleted accessible chromatin regions revealed a pattern of impaired immune-related pathways in 9 families.
Conclusion: We identified regulatory regions within a MLH1 stomach-specific accessible sequence, and within the CDH1 TAD that harbour CNVs potentially explaining the missing heritability in three HDGC-suspected families. Germline CNVs in stomach-specific regulatory regions favouring an immune-deficient phenotype may predispose 9 additional families to HDGC.
Grants: FCT: PTDC/BTM-TEC/30164/2017, PTDC/BTM-TEC/6706/2020, SFRH/BD/140796/2018; EU: SOLVE-RD_Ref 22184, HORIZON_HORIZON-HLTH-2022-CARE-08.
Conflict of Interest: None declared
C12.3 Identification and functional characterization of MSH2 variants with partial effects at RNA splicing and/or protein level: implications for Lynch syndrome diagnosis
Manon Quilan 1, Nawel Malouche2, Abhijit Rath3, Marine Cauchois-Lemière1, Laetitia Meulemans1, Aurelie Drouet1, Julie Leclerc4, Marie-Pierre Buisine4, Florence Coulet2, Stéphanie Baert-Desurmont5, Pascaline Gaildrat1, Christopher D. Heinen3, Martine Muleris2, Alexandra Martins1
1Inserm U1245, Inserm, Univ Rouen Normandie, Rouen, France; 2Département de Génétique Médicale et Inserm UMR-S 958, Hôpital Pitié-Salpêtrière, AP-HP, Sorbonne Université, Paris, France; 3Center for Molecular Oncology, UConn Health, Farmington, CT, United States; 4UMR9020-U1277 CANTHER, Univ. Lille, CNRS, Inserm, CHU Lille, and Molecular Oncogenetics Unit, Department of Biochemistry and Molecular Biology, Lille University Hospital, Lille, France; 5Inserm U1245 et Département de Génétique Moléculaire, CHU Rouen, Univ Rouen Normandie, Rouen, France
Background/Objectives: MSH2 is a major gene involved in Lynch syndrome (LS), an inherited predisposition to cancer, especially to colorectal/endometrial tumours. Identification of a causal variant is essential for LS diagnosis. However, ≥30% of the variants detected in MSH2 remain classified as of uncertain significance (VUS). It is therefore critical to develop strategies to improve their interpretation, particularly of those with partial/dual effects at RNA and/or protein level.
Methods: We identified and characterized such VUS using three complementary approaches: (i) cell-based minigene splicing assays (ii) protein functional tests based on cDNA complementation of MSH2-deficient cells and methylation tolerance assessment, and (iii) integrated RNA/protein analyses using CRISPR/Cas9-modified human embryonic stem cells (hESCs-tests). We chose MSH2 exon 5 (MSH2ex5) as a model-system because this exon is essential for MSH2 function.
Results: Our minigene assays uncovered multiple non-coding (intronic/synonymous) and missense MSH2ex5 variants causing RNA splicing defects of different severities (weak to total exon skipping). Selected missense variants were subsequently analysed for their impact on protein function, which revealed a large spectrum of outcomes (none, intermediate, and drastic defects). Preliminary results from the hESCs-tests on the non-coding spliceogenic variants indicate that hESCs can tolerate a substantial decrease in full-length MSH2 transcript produced by the mutant MSH2 alleles without significantly impacting DNA damage response nor DNA mismatch repair functions. Three missense variants with partial impacts on splicing, combined or not with intermediate protein effects, were also selected for hESCs-tests.
Conclusion: This work contributes to the interpretation of MSH2 VUS, notably those with partial/dual effects at RNA/protein levels.
Grants: RIN2021-100, INCA_16094.
Conflict of Interest: None declared
C12.4 BPTF Mutation Unravels Viral Oncogenicity in Heritable Kaposi Sarcoma
Yuval Yogev 1;2, Moshe Schaffer3;4, Mark Shlapobersky4;5, Matan M. Jean1, Ohad Wormser1, Max Drabkin1, Daniel Halperin1, Riad Kassem6;7, Alexandra A. Tsitrina8, Noam Asna3;4, Alejandro Livoff4;5, Ohad Shmuel Birk1;9
1Ben-Gurion University of the Negev Faculty of Health Sciences, The Morris Kahn Laboratory of Human Genetics at the National Institute of BiotechnologyThe Morris Kahn Laboratory of Human Genetics at the National Institute of Biotechnology, Beer-Sheva, Israel; 2The Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Ramat Gan, Israel; 3Barzilai University Medical Center, Department of Oncology, Ashkelon, Israel; 4Ben-Gurion University of the Negev, Faculty of Health Sciences, Beer-Sheva, Israel; 5Barzilai University Medical Center, Department of Pathology, Ashkelon, Israel; 6Sheba Medical Center, Department of Dermatology, Ramat Gan, Israel; 7Tel Aviv University, Sackler School of Medicine, Tel Aviv, Israel; 8Ben-Gurion University of the Negev, Ilse Katz Institute for Nanoscale Science and Technology, Beer-Sheva, Israel; 9Soroka University Medical Center, Genetics Institute, Beer-Sheva, Israel
Background/Objectives: Kaposi sarcoma (KS), a rare neoplasm caused by Kaposi’s sarcoma-associated herpesvirus (KSHV), exhibits distinct sub-forms with varied clinical presentations. While classical KS (cKS) is predominantly sporadic, rare familial cases suggest genetic predisposition.
Methods: We utilize WES for variant characterization, and functional assays using CRISPR-KI alongside with reporter viruses, confocal0microscopy, flow-cytometry and transcriptomics.
Results: We report two Moroccan Jewish families with seven individuals across two generations affected by cKS, segregating with a heterozygous missense mutation (p.I2012T) in Bromodomain and PHD finger-containing transcription factor (BPTF), known to interact with the KSHV protein LANA-1. Functional studies using a rKSHV.219-infected CRISPR-KI model reveal that the BPTFI2012T variant promotes a higher latent-to-lytic infection ratio, with increased LANA expression and decreased virion production. Transcriptomic analysis highlights dysregulated pathways related to tumorigenesis, immune response, blood vessel development pathways and viral genome replication, consistent with cKS. Notably, BPTFI2012T enriched pathways of viral genome regulation and replication, immune response, and chemotaxis, including downregulation of IFI16, SHFL HLAs, TGFB1 and HSPA5, all previously associated with KSHV infection and tumorigenesis, and influences NF-κB pathway, suggesting its role in KSHV oncogenesis.
Conclusion: Our findings suggest that BPTF plays a crucial role in KSHV-induced tumorigenesis by modulating viral life cycle and immune response. Understanding the genetic basis of heritable KS sheds light on the molecular mechanisms underlying viral oncogenicity and may lead to targeted therapies for this rare malignancy
Grants: Israel Science Foundation (grant no. 2034/18) the Foulkes foundation, ISCORT grant, the National Knowledge Center for Rare/Orphan Diseases of the Israel Ministry of Science, Technology and Space.
Conflict of Interest: None declared
C12.5 A FGFR1 susceptibility allele modulates the oncogenic potential of driver mutations in low-grade brain tumors
Jacopo Boni 1, Míriam Fernández-González1, Hyerim Han1, Cassandra Wong2, Jonathan Boulais3, Islam Elkholi3, Carla Roca1, Cristina Rioja1, Clara Nogué1, Cristina Jou4, Anne-Claude Gingras2, Jean-François Côté3, Barbara Rivera1;5;6
1IDIBELL Institut d’Investigació Biomèdica de Bellvitge, L’Hospitalet de Llobregat, Spain; 2Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Canada; 3Institut de recherches cliniques de Montréal (IRCM), Montréal, Canada; 4Sant Joan de Déu Barcelona Hospital, Pathology department, Esplugues de Llobregat, Spain; 5Lady Davis Institute at the Jewish General Hospital, Montréal, Canada; 6McGill University, Gerald Bronfman Department of Oncology, Montréal, Canada
Background/Objectives: Alterations in the FGFR1 receptor are associated with a wide range of human diseases, including different types of brain tumors. We identified a germline FGFR1 variant (p.R661P) in a family with DNETs (low-grade glioneuronal tumors). Secondary oncogenic somatic hits (p.N546K or p.K656E) in-cis with the p.R661P were identified in the tumors of these individuals. This pattern of multiple FGFR1 mutants was validated in therapy-naïve sporadic cases, suggesting intrinsic mechanisms of selective pressure that promote FGFR1 mutational events in the context of a quiet genome.
Methods: To gain insights into the molecular mechanisms triggered by different FGFR1 mutations, we mapped the proximal interactome of FGFR1 mutants through a BioID-MS approach. We generated FGFR1-mutant human oligodendroglioma cell lines by CRISPR-Cas9 editing to assess cell phenotypic changes.
Results: The study revealed similar profiles for the germline p.R661P and the WT condition, whereas all oncogenic mutants show pronounced differences in their proximal interactome. We identified a modulating effect played by the R661P variant, hampering the oncogenic potential of N546K and K656E mutants by reducing affinity for downstream effector PLC-y and enhancing receptor degradation, respectively. The p.R661P mutation alone abolished the self-renewing capacity of these cells, while in double mutants (N546K-R661P, K656E-R661P) weaking the proliferative potential of the oncogenic N546K and K656E clones.
Conclusion: These results reveal a severe impairment phenotype associated to the susceptibility variant, likely compensated in patients by the presence of the wt copy, and, suggest oncogenic modulation potential exerted by multiple FGFR1 alterations during low-grade brain tumorigenesis.
Grants: ALSF YIA award & la Caixa (LCF/BQ/PI19/11690009) award.
Conflict of Interest: None declared
C12.6 Genomic mechanisms of aggressive and metastatic phenotypes in non-melanoma skin cancers
Andrey Yurchenko 1, Andrea Forschner2, Fatemeh Rajabi1, Lukas Flatz2, Sergey Nikolaev1
1Gustave Roussy Cancer Campus, Université Paris Saclay, France, INSERM U981, Villejuif, France; 2University Hospital Tübingen, University of Tübingen, Department of Dermatology, Tübingen, Germany
Background/Objectives: Basal Cell Carcinoma (BCC) and cutaneous Squamous Cell Carcinoma (cSCC) are the most common human malignancies of keratinocyte origin with increasing incidence. Usually, these cancers can be effectively treated, but rarely, they can be aggressive or metastatic with very poor prognosis and still unknown genomic mechanisms.
Methods: We have assembled the largest to date cohorts of BCC (n = 389) and cSCC (n = 223) representing major histological subtypes, lymph node metastasis and distant metastasis. The samples were profiled with WES/WGS/RNA-seq and clinically annotated.
Results: The mutational profiles of BCC and cSCC were determined by UV-light exposure with high TMB (80 and 100 mt/mb respectively) and a median of six cancer driver alterations per sample. We found that loss of TP53 is typically the first driver event in keratinocyte carcinomas and is associated with a dramatic increase in successive driver mutation acquisition. Aggressive morpheaphorm BCC samples did not demonstrate specific genetic alterations but had fibrotic TME and increased activity of HIPPO-YAP pathway according to RNA-seq analysis and immunohistochemistry. Metastatic BCC were characterised by high genomic instability and aneuploidy not typical for this type of cancer. Metastatic cSCC tumours had an increased frequency of MAPK pathway alterations and high levels of aneuploidy and whole genome duplication events. These findings were confirmed by an independent cohort of the GENIE project. Lymph node metastasis of cSCC samples demonstrated lymphocyte-enriched TME favourable for immunotherapeutic treatment.
Conclusion: Genomic instability and transcriptional activation of HIPPO-YAP and MAPK pathways define the high-risk and metastatic potential of keratinocyte carcinomas.
Grants:
Conflict of Interest: None declared
C13 Exploring cellular signatures
C13.1 Uncovering cell-type specific gene-regulation using single-cell multiomics in the sc-eQTLGen consortium
Maryna Korshevniuk 1, Dan Kaptijn1, Roy Oelen1, Martijn van der Werff1, Jelmer Niewold1, Harm-Jan Westra1, Lude Franke1, Monique van der Wijst1, Marc Jan Bonder1
1University Medical Center Groningen, Genetics Department, Groningen, Netherlands
Consortium: sc-eQTLGen
Background/Objectives: One approach to interpreting disease-related genetic variation is through the identification of expression quantitative trait loci (eQTLs). Linking genetic variation to single-cell expression data (scRNA-seq) enables the identification of cell type-specific eQTLs (ct-eQTLs), thus providing insight into the relevant context in which these variants act. Overlaying ct-eQTLs with cell type-specific DNA accessibility data (scATACseq) allows us to gain insight into the mechanisms underlying ct-eQTLs and suggest potential disease mechanisms.
Methods: Within the sc-eQTLGen consortium we conducted the largest ct-eQTL mapping to date in PBMC (n-donors 2,000). Furthermore, we generated a large single-cell multiomics (scMO) dataset on PBMCs derived from 271 healthy individuals to gain mechanistic insights into the ct-eQTL SNPs.
Results: Here we present the first results of the sc-eQTLGen consortium where we identified in total 14,749 eQTLs (FDR 10%) for the six major PBMC cell types corresponding to 7,310 eGenes and 2,810 unique ct-eQTLs. In the preliminary analysis of the scMO data we identified several overlapping ct-specific ATAC peaks and ct-eQTLs. Among them is a monocytes-specific eQTL on the TREM1 which overlaps a KLF family motive differentially accessible in monocytes. Leveraging our complete sc-MO dataset, we aim to identify ct-specific regulators of gene expression, thus providing deeper insights into the regulatory mechanisms of the ct-eQTLs.
Conclusion: Integrating scMO data enhances our understanding of cell-type-specific gene regulation mediated by eQTLs. This study underscores the importance of such integrative approaches in elucidating the genetic mechanisms underlying complex traits and diseases, with potential implications for precision medicine and disease research.
Conflict of Interest: None declared
C13.2 Spatial dissection of colorectal cancer initiation using whole transcriptome imaging
helena crowell1, hu zedong2, Michael Rhodes3, Joseph Beechem3, sabine tejpar2, holger heyn1, Anna Pascual Reguant 4
1Centre Nacional d’Anàlisi Genòmica, Single Cell Genomics, Barcelona, Spain; 2KU Leuven, Laboratory of Molecular Digestive Oncology, Leuven, Belgium; 3NanoString® Technologies, Seattle, United States; 4Centre Nacional d’Anàlisi Genòmica, Spatial Genomics, Barcelona, Spain
Background/Objectives: Revolutionizing clinical pathology requires new technologies that excel currently applied approaches that multiplex selected biomarkers to guide clinical decisions. While higher-plex imaging techniques recently became available, none is scalable to appreciate the full spectrum of molecular biomarkers holistically. We argue that biomarker discovery requires substantially more features to cope with the complexity of diseases. Here, thousands of genes or proteins, imaged at single-cell resolution, will be required to comprehensively identify diagnostic and therapy-prognostic biomarkers.
Methods: Spatial Molecular Imaging (SMI) allows for transcriptome-wide analyses at molecule resolution, enabling spatial single-cell profiling at a definition that was only achieved through scRNA-seq until now. Through digitalization of full transcriptomes in tissue sections and improving the current state-of-the-art methods in pathology by orders of magnitude, next-generation pathology tools are in sight.
Results: We applied SMI’s whole transcriptomics to spatially resolve tumor evolution within colon samples, progressing from premalignant lesions (tubullovillous adenoma) to colorectal cancer (CRC). We generated spatially resolved single-cell landscapes of these tissues and detected vast spatial heterogeneity that matched H&E pathologist annotations, with intertwined cell state niches resembling stem cells or differentiated colonocytes, phenotypes previously linked to the progression into different CRC subtypes. We associated gene programs of stemness, fetal development and immunogenicity, the latter spatially correlated with immune-cell infiltration patterns.
Conclusion: Importantly, the enrichment of clinically relevant pre-malignant cell states at discrete niches could only be resolved using whole transcriptome spatial data and not through scRNA-seq, providing evidence of the method’s superiority in enabling next-generation pathology strategies.
Grants: RYC2022-035848-I
Conflict of Interest: helena crowell: None declared, hu zedong: None declared, Michael Rhodes NanoString Technologies, NanoString Technologies, Joseph Beechem NanoString Technologies, NanoString Technologies, sabine tejpar: None declared, holger heyn co-founder of Omniscope, SAB member of Nanostring and MiRXES, and consultant to Moderna and Singularity, Anna Pascual Reguant: None declared
C13.3 Convergence of neurodevelopmental signatures in Prader-Willi syndrome from an allelic series of CRISPR-engineered neuronal models
Celine De Esch 1, Derek Tai1, Xander Nuttle1, Rachita Yadav1, Yating Liu1, Jaewon Shin1, Nicholas Burt1, Serkan Erdin1, John Lemanski1, Bimal Jana1, Benjamin Currall1, Yulia Mostovoy1, Dadi Gao1, Lisa Burnett1, James Gusella1, Michael Talkowski1
1Massachusetts General Hospital/Harvard Medical School, Center for Genomic Medicine, Boston, United States
Background/Objectives: Prader-Willi Syndrome (PWS) is a neurodevelopmental disorder caused by a loss of expression of paternally inherited genes from the imprinted chromosome 15q11-q13 region. Recurrent deletions spanning 5-6 Mb (Type I/II deletions) are the most common cause of PWS and the affected genomic region includes five paternally expressed protein-coding genes, six small nucleolar RNAs (SNORDs), and IPW, a long non-coding RNA. To investigate the potential drivers of neurodevelopmental changes associated with PWS, we created an allelic series of human induced pluripotent stem cell (hiPSC) models harboring CRISPR-engineered paternal deletions across the PWS locus and characterized derivative neuronal models.
Methods and Results: We differentiated 221 isogenic hiPSC models with PWS-related mutations and matched controls for each edit, including Type I/II deletions, the critical region deletion (CRD, including SNORD116, IPW, and SNORD109A), and individual gene deletions in male and female iPSC backgrounds into Ngn2-induced neurons (iNs) followed by systematic transcriptomic analyses (small RNA, mRNA), along with functional microelectrode array (MEA). Meta-analyses of differentially expressed and co-expressed genes across Type I/II edits were strongly enriched for nervous system and synaptic functions (false discovery rate < 0.1), including retrograde endocannabinoid signaling, GABAergic synapse, serotonergic synapse, and taste transduction pathways. The Type I/II edits displayed significant (p < 0.05) reductions in synaptic activity, synchrony (strength of synaptic connections), and oscillation (network activity).
Conclusion: Collectively, our findings demonstrate the functional impact of PWS-associated gene deletions in human-derived neural models and highlight alterations to neuronal and endocrine pathways associated with PWS pathogenesis.
Conflict of Interest: Celine De Esch PWS grant, MGH, Derek Tai: None declared, Xander Nuttle: None declared, Rachita Yadav: None declared, Yating Liu: None declared, Jaewon Shin: None declared, Nicholas Burt: None declared, Serkan Erdin: None declared, John Lemanski: None declared, Bimal Jana: None declared, Benjamin Currall: None declared, Yulia Mostovoy: None declared, Dadi Gao: None declared, Lisa Burnett: None declared, James Gusella: None declared, Michael Talkowski: None declared
C13.4 Colocalization between human aortic smooth muscle cells molecular QTLs and vascular disease GWAS identified loci with pleiotropic effect
Ilakya Selvarajan1, Nadine Odenthal 2;3;4, Luis Eichelmann2;3;4, Tobias Reinberger2;3;4, Zouhair Aherrahrou2;3;4, Minna Kaikkonen-Määttä1, Redouane Aherrahrou1;2;3;4;5
1A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland; 2Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany; 3DZHK (German Centre for Cardiovascular Research); 4University Heart Centre Lübeck, Lübeck, Germany; 5Center for Public Health Genomics, University of Virginia, Charlottesville, United States
Background/Objectives: Smooth muscle cells (SMCs) play a crucial role in cardiovascular diseases (CVD), undergoing phenotypic changes under pathological conditions, contributing to vascular remodeling and CVD pathogenesis. This study aims to identify genetic variants regulating SMC gene expression and phenotypes associated with vascular disease-related loci from genome-wide association studies (GWAS).
Methods: SMCs from 151 heart transplant donors of various genetic ancestries were cultured in proliferative and quiescent conditions and the expression and splicing quantitative trait loci (eQTL and sQTL) were conducted. Colocalization analysis was performed between these SMCs molecular QTL and GWAS loci associated with vascular disorders to identify effector transcripts for CVD pathogenesis. Potential risk genes were prioritized and validated for their effects on the vascular phenotype using in vivo and in vitro experiments
Results: We found 336 eQTLs and 580 sQTLs co-localizing with CVD loci, showing genetic regulation of mRNA expression and splicing in CVD genetic risk. Filtering these loci for accessibility to chromatin regions and high expression in SMCs revealed key genes, such as MAP3K7CL, which colocalizes with CAD, heart attack, and blood pressure GWAS loci. MAP3K7CL modulates SMC migration, proliferation, and calcification and its knockdown in zebrafish altered blood flow, heart rhythm patterns, and angiogenesis, highlighting its role in CVD pathogenesis.
Conclusion: Our study reveals that SMCs influence vascular disorders, and their genetic regulation is linked to CVD pathogenesis. These findings may guide targeted therapeutic interventions for vascular diseases.
Grants: This work was supported by Finnish Foundation for Cardiovascular Research, DZHK and the CORONA Foundation.
Conflict of Interest: None declared
C13.5 Cell type signatures in cell free DNA fragmentation profiles reveal disease biology
Kate Stanley 1, Joris Vermeesch2, dhanya sudhakaran3, Stefania Tuveri3, Tatjana Jatsenko3, Bernard Thienpont3, Kristel Van Calsteren3, Lore Lannoo3, Kris Van Den Bogaert3, Kevin Punie3, Marie de Borre3, sabine tejpar3, Liesbeth Lenaerts3, Peter Vandenberghe3, Rodrigo Almeida3, Ilse Parijs3, Leen Vancoillie3, Laura Rengifo3
1KU Leuven, Centre for Human Genetics, Leuven; 2KU Leuven, Centre for Human Genetics, Leu, Belgium; 3KU Leuven, Centre for Human Genetics, L, Belgium
Background/Objectives: Circulating cell free DNA (cfDNA) fragments have characteristics that are specific to the cell types that release them. Current methods for cfDNA deconvolution typically use disease tailored marker selection in a limited number of bulk tissues or cell lines. We utilize single cell transcriptome data as a comprehensive cellular reference set for disease-agnostic cfDNA cell-of-origin analysis.
Methods: We performed whole-genome paired-end sequencing of plasma cfDNA from healthy individuals and patients with cancer and pregnancy complications. Sequencing data was aligned to the human reference genome and the position of fragments was used to infer nucleosome spacing. We then correlated cfDNA-inferred nucleosome spacing with gene expression data from the Tabula Sapiens database to rank the relative contribution of over 450 cell types to plasma cfDNA.
Results: In 744 healthy individuals and patients, we uncover cell type signatures in support of emerging disease paradigms in oncology and prenatal care. We trained predictive models that differentiated patients with colorectal cancer (84.7%), early-stage breast cancer (90.1%), multiple myeloma (AUC 95.0%), and preeclampsia (88.3%) from matched controls using cell type ranks. Importantly, our approach performed well in ultra-low coverage cfDNA datasets.
Conclusion: CfDNA fragmentation patterns can be used to estimate the relative abundance of fragments from different cell types. These relative abdundances are altered under certain conditions and can be used to predict disease in diverse clinical settings for the potential expansion of liquid biopsy.
Grants: EU Horizon 2020 (824110, 813707), Kom Op Tegen Kanker (KOTK/2018/11468), FWO-SBO (S003422N), and Agentschap Innoveren en Ondernemen (HBC.2018.2108).
Conflict of Interest: None declared
C13.6 Interpreting GWAS Signals and the Genetic Architecture of Ventricular Hypertrophy in African Americans trough the Context of Cellular Expression Networks in Human iPSC-Cardiomyocytes.
Jakob Blamer1, Andrea Matter1, Elizabeth Virlee1, Amy Turner1, Ulrich Broeckel 1
1Medical College of Wisconsin, Pediatrics, Milwaukee, United States
Background: GWAS studies provide many susceptibility loci for common diseases, yet the underlying genetic architecture is not well understood. Evidence indicates that GWAS-identified genes interact systematically through networks. We explore this concept using results from a GWAS study of Left Ventricular Hypertrophy (LVH) in an African American cohort combined with relevant expression data.
Methods: We generated hiPSC lines from 110 participants in the NHLBI HyperGEN GWAS study and differentiated those into cardiomyocytes (hiPSC-CMs) for expression analysis. We identified a set association signals in the full HyperGEN cohort and mapped variants to respective candidate genes. Subsequently, we paired GWAS results with hiPSC-CM cardiac phenotypes and expression data to prioritize genes using network-based analyses. We identified expression modules and functionally relate those to LVH which are enriched for GWAS genes. Finally, we performed CRISPR gene knockout studies of candidate genes to explore changes in expression and network properties.
Results: We identified two distinct expression modules to be functionally related to LVH and enriched in candidate GWAS genes. Additionally, GWAS genes were local hubs in one of these two modules. We identified two novel (ETV6, KLF2) and one known (BNC2) transcription factor GWAS genes differentially co-expressed between hiPSC-CMs from normal and affected donors. Knock-down of these genes through CRISPR/Cas9 in hiPSC-CM resulted in significant hypertrophy marker expression changes and strong enrichment of cardiac pathways.
Conclusion: Network-based analyses using disease relevant cells allow for the functional annotation of GWAS genes. Human iPSCs combined with CRISPR editing provide a platform for the functional annotation of GWAS results.
Grants: NHLBI R56HL167896
Conflict of Interest: Jakob Blamer: None declared, Andrea Matter: None declared, Elizabeth Virlee: None declared, Amy Turner: None declared, Ulrich Broeckel NHLBI
C14 Sensory disorders
C14.1 A multi-omics approach increases the diagnostic yield ‘beyond the exome’ in unsolved patients with inherited eye diseases
Miriam Bauwens 1;2, Marta Del Pozo-Valero1;2, Marieke De Bruyne1;2, Filip Van den Broeck3;4, Quinten Mahieu1;2, Mattias Van Heetvelde1;2, Stijn Van de Sompele1;2, Alfredo Dueñas Rey1;2, Audrey Meunier5, Thomy de Ravel6, Joke Ruys3, Irina Balikova7, Sascha Vermeer8, Julie De Zaeytijd3;4, Bart Leroy3;4, Elfride De Baere1;2
1Center for Medical Genetics, Ghent University Hospital, Gent, Belgium; 2Department of Biomolecular Medicine, Ghent University, Universiteit Gent, Gent, Belgium; 3Department of Ophthalmology, Ghent University Hospital, Gent, Belgium; 4Department of Head & Skin, Universiteit Gent, Gent, Belgium; 5Department of Ophthalmology, University Hospital Saint-Pierre, Université Libre de Bruxelles (ULB), Brussels, Belgium; 6Center for Medical Genetics, University Hospital Brussels, Brussels, Belgium; 7Department of Ophthalmology, UZ Leuven, Leuven, Vlaams-Brabant, Belgium; 8Center for Human Genetics, UZ Leuven, Leuven, Vlaams-Brabant, Belgium
Background/Objectives: Exome sequencing (ES)-based testing in patients with inherited eye diseases (IED) has an overall diagnostic yield of ~60%. To overcome the missing heritability in molecularly undiagnosed ES-tested IED patients, we adopted a multi-omics approach, combining genomics, transcriptomics and functional assays.
Methods: Short-read genome sequencing (GS) of 111 cases (103 probands) was analyzed using Franklin (Geenox) and Seqplorer (in-house). Coding single nucleotide variants (SNVs) in known IED genes were prioritized, followed by non-coding variants assessed for an effect on splicing or cis-regulation, and by structural variants (SVs). Candidate genes were selected based on single-cell expression profiles and function. GS-data directed whole-blood RNA-seq and/or functional assays.
Results: A likely genetic diagnosis was obtained for 44.5% of probands. Our multi-omics approach pinpointed likely pathogenic non-coding variants in 18.5%, predominantly affecting splicing (16.5%). These include OPA1 variant c.1608+622A > G, leading to pseudo-exon inclusion (PEI) and a novel OPA1 variant, leading to out-of-frame PEI. A first deep-intronic ALMS1 variant and a novel deep-intronic RPGRIP1 variant were also shown to induce aberrant splicing. Moreover, we demonstrated a reduced (40%) regulatory activity of a novel RPGRIP1 promotor variant. Variants in novel candidate genes, including SAMD7 and GPATCH11, were identified in 14.5%, while coding SNVs (8.5%) and SVs (3%) impacting known IED genes were found in 11.5%.
Conclusion: Our multi-omics approach solved missing heritability in 44.5% of molecularly undiagnosed IED cases, mainly attributed to non-coding deep-intronic splicing variants and variants in novel genes. Overall, its added diagnostic value urges its implementation in standard-of-care genetic testing of IED.
Grants: EJPRD19-234, BeSolveRD, BOF20/GOA/023
Conflict of Interest: None declared
C14.2 Mutations in COQ8B are a novel cause of recessive non-syndromic retinitis pigmentosa.
Karolina Kaminska 1;2, Ana Belén Iglesias-Romero1;2, Cristina Santos3;4, Siying Lin5;6, Mathieu Quinodoz1;2, Marc Folcher1;2, Joaquim Calado7, Gavin Arno5;6, Andrew R Webster5;6, Ana Berta Sousa8;9, Luisa Coutinho Santos3, Carlo Rivolta1;2;10
1Institute of Molecular and Clinical Ophthalmology Basel (IOB), Basel, Switzerland; 2University of Basel, Department of Ophthalmology, Basel, Switzerland; 3Instituto de Oftalmologia Dr. Gama Pinto, Lisboa, Portugal; 4NOVA Medical School, iNOVA4Health, Lisboa, Portugal; 5Moorfields Eye Hospital NHS Foundation Trust, National Institute of Health Research Biomedical Research Centre, United Kingdom; 6University College London, UCL Institute of Ophthalmology, United Kingdom; 7NOVA Medical School, ToxOmics, Lisboa, Portugal; 8Hospital de Santa Maria, Department of Medical Genetics, Lisboa, Portugal; 9University of Lisbon, Laboratory of Basic Immunology, Faculty of Medicine, Lisboa, Portugal; 10University of Leicester, Department of Genetics and Genome Biology, United Kingdom
Background/Objectives: Retinitis pigmentosa (RP) is a highly heterogeneous genetic disease characterized by progressive retinal blindness. Mutations in more than 90 genes, with very diverse functions, have been so far identified to be associated with this condition.
The objective of this study was to determine the molecular cause of RP in 5 patients from 4 unrelated families, who were all negative for mutations in previously known disease genes.
Methods: All patients underwent a detailed ophthalmic examination. Genotyping was performed by Next Generation or Sanger sequencing.
Mutant or wild-type versions of COQ8B were obtained by transfecting HEK293T cells with synthetic expression plasmids. Then, an assay using Bioluminescence Resonance Energy Transfer Technology (NanoBRET) was used to assess the functional consequences of the variants on COQ8B activity. Western blots were performed on extracts from human retinas.
Results: Analysis of sequencing data identified biallelic assortments of 5 different variants in COQ8B (3 missense and 2 nonsense), a gene involved in the biosynthesis of coenzyme Q, in all patients. Segregation analysis within families confirmed the recessive nature of the disease.
NanoBRET assays showed that COQ8B bearing the combination of variants detected in patients had significantly reduced ligand binding affinity, compared to the wild-type protein. Western blots showed the presence of COQ8B in the human retina.
Conclusion: We identified a new disease gene associated with recessive and non-syndromic RP, COQ8B. Functional studies indicate that the variants detected in patients likely impair COQ8B protein function, leading to defects in coenzyme Q biosynthesis and, ultimately, to retinal disease.
Grants:
Conflict of Interest: None declared
C14.3 High throughput screening: identification of candidate molecules for the treatment of SLC7A8- dependent age-related hearing loss (ARHL)
Paola Tesolin 1, Paola Bartoccioni2;3;4, Giulia Pianigiani1, Maria Caballero2, Anna Morgan1, Cristina Bon1, Gema Rojas Granado2, Israel Ramos2, Patrick Aloy2, Paolo Gasparini1;5, Virginia Nunes6;7, Giorgia Girotto1;5, Manuel Palacin2;3;4
1Institute for Maternal and Child Health – IRCCS, Burlo Garofolo; 2Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology; 3Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III; 4University of Barcelona, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology; 5University of Trieste, Department of Medicine, Surgery and Health Sciences; 6Genes, Disease and Therapy Program, Molecular Genetics Laboratory - IDIBELL; 7University of Barcelona, Genetics Section, Physiological Sciences Department, Health Sciences and Medicine Faculty
Background/Objectives: ARHL is a common multifactorial sensory impairment, but few causative genes have been identified, hampering the development of targeted therapies. Our previous study (PMID:29355479) proposed SLC7A8 as a novel candidate by showing that knock-out (KO) mice develop ARHL and by identifying damaging variants in humans displaying ARHL. These findings suggest that SLC7A8 is an interesting therapeutic target to investigate.
Methods: We performed a high throughput screening (HTS) to detect SLC7A8 transcription regulators. A reporter line was generated by inserting the luciferase gene in SLC7A8 genomic locus. Subsequently, it was incubated for 48 hours with a library of 2240 approved drugs, 960 drug-like compounds and 44 molecules selected as regulators of SLC7A8 transcription according to literature data.
Results: The HTS led to the identification of 56 hit compounds. The most promising ones were re-screened to define the dose-response curve in luciferase and cell viability assays. Furthermore, three molecules, which are already approved drugs, were finally selected. The Compound-1 mechanism of action is linked with increased SLC7A8 expression. Compound-2 was chosen to represent a drug category over-present among the hits. Finally, Compound-3 is already used to treat other forms of hearing loss. An RT-qPCR confirmed that the treatment significantly increased SLC7A8 expression in the analysed cell lines.
Conclusion: These results highlight the possible use of the selected molecules in treating SLC7A8-dependent ARHL. Subsequently, the hits will be tested in HEK293T carrying the variants identified in humans, in inner ear-related cell lines, and in Slc7a8 heterozygous KO mice.
Conflict of Interest: None declared
C14.4 Whole genome sequencing and functional analysis in families with inherited retinal dystrophies (IRDs)
Valerie Wenzel 1, Tanja Plieger1, Anna-Zoe Häusler1, Christina Kiel1, Heidi Stöhr1, Georg Spital2, Ines Schönbuchner3, Herbert Jägle4, Ulrich Kellner5, Bernhard Weber1;6
1University Regensburg, Institute for Human Genetics, Regensburg, Germany; 2St. Franziskus-Hospital Münster, Eye Center, Münster, Germany; 3Human Genetics Ostbayern, Germany; 4University Hospital Regensburg, Clinic and Polyclinic for Ophthalmology, Regensburg, Germany; 5Eye Center Siegburg, MVZ Ophthalmologic Diagnostic and Therapy Center Siegburg GmbH, Center for Rare Retinal Diseases, Münster, Germany; 6University Hospital Regensburg, Institute of Clinical Human Genetics, Regensburg, Germany
Consortium: Bavarian Genomes Network for Rare Disorders, Munich, Deutschland
Background: Inherited retinal dystrophies (IRDs) are a group of rare diseases characterized by phenotypic and genetic heterogeneity. Despite major advances in molecular genetic diagnosis, approximately 30-40% of IRD cases remain unsolved. Whole Genome Sequencing (WGS) represents a promising approach in routine diagnostics by capturing genetic variants throughout the human genome and addressing structural variation as disease causation. The study aimed to evaluate the efficacy of WGS in routine diagnostics.
Methods: As part of the multicenter study “Bavarian Genomes Network for Rare Disorders”, WGS was performed in 65 IRD families comprising 143 individuals with no clear result after exhaustive analysis with traditional DNA diagnostics tools. WGS data evaluation focused on variant type, allele frequency, computational predictions, and homozygosity analysis. Furthermore, a luciferase reporter assay was established to study the effects of non-coding variants on promoter activity.
Results: WGS analysis determined disease causation in 13 of 65 (20%) families. Moreover, WGS analysis identified at least one disease-causing gene candidate in 10 families (15%). The latter findings include a promising homozygous non-coding variant located in the Rhodopsin (RHO) promoter, found in a patient of consanguineous parents. A luciferase assay studying the functional consequences of the identified variant showed strongly reduced RHO promoter activity.
Conclusion: This study underscores the advantages of WGS for the detection of genetic variants in DNA diagnostics. It should be noted, however, that elaborate functional analyses are generally indispensable to determine pathogenicity of candidate variants.
Grants: The “1,000 clinical genomes” project was funded by “Masterplan Digital II” of the Bavarian State Ministry of Science and Arts.
Conflict of Interest: None declared
C14.5 Biallelic loss-of-function variants in NDUFA7 cause autosomal recessive Leber’s hereditary optic neuropathy by impairing complex I repair
Christiane Michaela Neuhofer 1;2;3;4, Theresa Brunet1;4;5, Robert Kopajtich1;2, Riccardo Berutti1;2;4;6, Thomas Meitinger1;4, Hans A. Mayr3;7, Ilka Wittig8, Thomas Klopstock9, Holger Prokisch1;2;4
1Institute of Human Genetics, Technical University of Munich, Munich, Germany; 2Institute of Neurogenomics, Helmholtz Zentrum München, Neuherberg, Germany; 3Institute of Human Genetics, Paracelsus Medical University, University Hospital Salzburg, Salzburg, Austria; 4Bavarian Genomes Network for Rare Disorders; 5Dr. v. Hauner Children’s Hospital, Department of Pediatric Neurology and Developmental Medicine, LMU- University of Munich, Munich, Germany; 6NEXT - NGS core facility, CRS4 - Center for Advanced Studies, Research and Development in Sardinia, Pula, Italy; 7University Children’s Hospital, Paracelsus Medical University, University Hospital Salzburg, Salzburg, Austria; 8Institute for Cardiovascular Physiology, Goethe-University Frankfurt am Main, Frankfurt am Main, Germany; 9Friedrich-Baur-Institute, Department of Neurology, LMU- University of Munich, Munich, Germany
Background/Objectives: Leber’s hereditary optic neuropathy (LHON) is the most common mitochondrial disorder. It is caused in >90% of cases by one of three mitochondrial point mutations in mitochondrial complex I (CI) subunits. Recently DNAJC30 has been identified as the first nuclear gene to cause LHON and implicated CI repair as underlying pathomechanism. We present genetic and functional findings in a patient clinically diagnosed with LHON to introduce NDUFA7 as a novel nuclear gene causing LHON. The patient showed a classic LHON phenotype with subacute, painless central vision loss first of the right eye, within one week bilaterally, at age 17.
Methods: We performed WGS after targeted LHON diagnostics was unable to reveal the patient`s disease cause. Functional work-up included transcriptome, proteome and complexome turnover analysis.
Results: WGS revealed a homozygous 1bp duplication at a splice donor site of NDUFA7, which encodes a CI N-module subunit that directly interacts with DNAJC30 to enable rapid N-module turnover. All four unaffected siblings of the patient were not homozygous for this NDUFA7 variant. Transcriptome analysis confirmed a 1bp frameshift at the transcript level and proteomics showed a significant reduction of NDUFA7 protein levels in fibroblasts. Dynamic complexome analysis of CI showed a reduction of three N-module subunits and DNAJC30 interactors, NDUFA7, NDUFS6 and NDUFA12, which physiologically show a high turnover rate.
Conclusion: We report NDUFA7 as a novel disease gene, extend the genetic landscape of complex I defects leading to LHON and emphasize the importance of the N-module repair for LHON disease.
Grants: GENOMIT, Bavarian Genomes
Conflict of Interest: Christiane Michaela Neuhofer: None declared, Theresa Brunet: None declared, Robert Kopajtich: None declared, Riccardo Berutti: None declared, Thomas Meitinger PI of Bavarian Genomes that in part funded this project, Hans A. Mayr: None declared, Ilka Wittig: None declared, Thomas Klopstock PI in GENOMIT which partly funded this project, Holger Prokisch PI and coordinator of GENOMIT, which partly funded this project
C14.6 Biallelic variants in GPN2, an essential RNA polymerase assembly factor, result in pleiotropic Perrault syndrome phenotypes
Thomas Smith 1;2, Leigh Demain1;2, Rabia Faridi3, Huw Thomas1;2, Alessandro Rea1;2, Arshia Maqbool4, Isabelle Schrauwen5, Khurram Liaqat5;6, Zubair Ahmed7;8, Sondhya Ghedia9, Andrew Green10;11, Ruth Sheffer12, Hagar Mor-Shaked12, Mathilda Wilding9, Robin Hay9, Sayaka Inagaki3, Saima Riazuddin7;8, Langping He13;14, Glenda Beaman1;2, Wasim Ahmad15, Suzanne Leal5;16, Sheikh Riazuddin4, Robert Taylor14, Thomas Friedman3, Raymond O’Keefe1;2, William Newman1;2
1University of Manchester, School of Biological Sciences, Manchester, United Kingdom; 2University of Manchester, Manchester Centre for Genomic Medicine; 3National Institutes of Health, Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders; 4University of Health Sciences, Jinnah Burn and Reconstructive Surgery Centre, Allama Iqbal Medical Research, Lahore, Pakistan; 5Columbia University Medical Center, Center for Statistical Genetics, Gertrude H. Sergievsky Center, and the Department of Neurology; 6Quaid-i-Azam University, Department of Biotechnology, Faculty of Biological Sciences; 7University of Maryland School of Medicine, Department of Otorhinolaryngology Head and Neck Surgery; 8University of Maryland, Department of Molecular Biology and Biochemistry, School of Medicine; 9Royal North Shore Hospital, Department of Clinical Genetics, St Leonards, Australia; 10Children’s Health Ireland at Crumlin, Department of Clinical Genetics; 11University College Dublin, Ireland School of Medicine and Medical Science; 12Hadassah-Hebrew University Medical Center, Department of Genetics; 13Newcastle upon Tyne Hospitals NHS Foundation Trust, NHS Highly Specialised Rare Mitochondrial Disorders Service; 14Newcastle University, Wellcome Centre for Mitochondrial Research; 15Quaid-i-Azam University, Department of Biochemistry; 16Columbia University Medical Center, Taub Institute for Alzheimer’s Disease and the Aging Brain, New York, United States
Background/Objectives: Perrault syndrome is a rare, autosomal recessive disorder characterised by sensorineural hearing loss and primary ovarian insufficiency. To date, biallelic variants in nine genes affecting mitochondrial and peroxisomal function have been associated with this condition, with presentations encompassing leukodystrophy, intellectual disability and developmental delay. However, in many individuals with Perrault syndrome the genetic cause remains unknown.
Methods: Exome sequencing was undertaken on affected individuals with Perrault syndrome alongside unaffected family members to identify disease-associated variants. Biallelic variants in GPN2, a GTPase involved in RNA polymerase biogenesis and assembly, were identified in four unrelated families. Individuals from two families presented with Perrault syndrome and cerebellar ataxia with homozygosity for p.Asn222Asp (RefSeq 54707). A 662kb common haplotype at the GPN2 locus was defined. A further family of ten affected individuals with Perrault syndrome homozygous for p.His121Gln was identified. Through reverse phenotyping, a child homozygous for p.Ser276Cys, presented with progressive ataxia, intellectual disability and seizures, but no hearing loss.
Results: Previously known to localize to the nucleus, we demonstrate that GPN2 also localizes to the mitochondria using immunofluorescence in patient fibroblasts. Respiratory chain complex activities were not altered in p.Asn222Asp fibroblasts, and GPN2 mRNA and protein levels were unchanged via qPCR and Western blotting, suggesting a subtle molecular phenotype. However, disease-associated variants may lead to altered GTPase activity and protein stability.
Conclusion: We present compelling genetic evidence that biallelic variants in GPN2 result in a multisystem phenotype, with preliminary data suggesting that GPN2 also has a previously unrecognized role in mitochondrial function.
Grants: MRC(MR/W019027/1);RNID&MCF(S60_Newman);AMR(GN2494); NIHR_MBRC(NIHR203308);WCMR_RWT(203105/Z/16/Z);NIDCD_NIH_TBF(DC000039).
Conflict of Interest: None declared
C15 Population and Evolutionary Genetics
C15.1 Insights from 1.2M structural variants across global populations in the Genome Aggregation Database (gnomAD)
Xuefang Zhao 1;2;3, Ryan Collins4, Mark Walker2;3, Alex Yenkin1, Nehir Kurtas1;2;3, Arthur Lee1;2;3, Lily Wang1, Daniel Ben-Isvy1, Jack Fu1;2;3, Cal Liao3;5, Philip Darnowsky3, Katherine Chao3, Stacey Gabriel3;5, Eric Banks3, Anne O’Donnell-Luria2;3, Daniel MacArthur3, Heidi Rehm2;3, Benjamin Neale3;5, Mark Daly3;5, Kaitlin Samocha2;3, Konrad Karczewski3;5, Dadi Gao1;6, Harrison Brand1;2;3, Michael Talkowski1;2;3
1Harvard Medical School, Neurology, Boston, United States; 2Massachusetts General Hospital, Center for Genomic Medicine, Boston, United States; 3Broad Institute, Medical and Population Genetics, Boston, United States; 4Dana-Farber Cancer Institute, Division of Medical Oncolog, Boston, United States; 5Massachusetts General Hospital, The Analytic and Translational Genetics Unit, Boston, United States; 6Massachusetts General Hospital, Neurology, Boston, United States
Consortium: gnomAD consortium
Background/Objectives: Structural variantion (SVs) contribute to the diversity of human genomes and myriad human trait associations.
Methods: As part of the Genome Aggregation Database (gnomAD), we present the completion of an SV atlas in gnomADv4 that encompasses 1,199,117 unique SVs genotyped across 63,046 unrelated individuals. Importantly, over half of these genomes were inferred non-European, enabling unique insights into genetic diversity across global ancestries.
Results: The average genome harbored 11,844 SVs discovered by short-read sequencing, with long-read sequences from 15 matched genomes supporting ~97% of SVs discovered outside of highly repetitive genomic regions. Beyond canonical SVs, we resolved ~45 complex SVs in each genome (most involving inversions) and found a highly complex yet polymorphic variant involving 49 breakpoints across 7 chromosomes in 3 individuals. From these data we estimate that SVs accessible to short-read sequencing cause, on average, loss-of-function (LoF) of 188 genes and copy gain (CG) of 34 genes per genome. The per-gene frequency of LoF and CG in SVs are significantly correlated with metrics of constraint against LoF for short variants, including the surprising finding of selection against SVs that only duplicate transcription start sites or UTRs despite the absence of direct disruption to the genic open reading frames. Further, we find that metrics measuring an excess of singleton SVs over neutral selection show positive correlation with expression levels (r = 0.73) of genes they interrupt.
Conclusion: These preliminary findings underscore the substantial impact of SVs on genome structure and function and the value of publicly accessible reference resources such as gnomAD (https://gnomad.broadinstitute.org/)
Grants: R01MH115957
Conflict of Interest: None declared
C15.2 Whole genome sequencing and analysis of 5750 Indians: Phase-1 insights from the Genome India project. (virtual)
Analabha Basu 1, Bratati Kahali2, Mohd Faruq3, Divya Tej Sowpati4, Kumarasamy Thangaraj4, Yadati Narahari5
1iBRIC-National Institute of BioMedical Genomics, Statistical and Computational Genomics, Kalyani, India; 2Centre for Brain Research - Indian Institute of Science, Bangalore, India; 3CSIR-Institute of Genomics and Integrative Biology, New Delhi, India; 4CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India; 5Centre for Brain Research - Indian Institute of Science Campus, Bengaluru, India
Consortium: GenomeIndia Project
Background/Objectives: India, the most populous country in the world, (1.43billion) is grossly under-represented in global genomic databases and biobanks. India harbours genetic diversity next only to Africa with more than 5000 prominent ethnolinguistic groups. The GenomeIndia Project is initiated by the Department of Biotechnology, Government of India, to catalogue the genomic diversity of India and build a roadmap for genomic-medicine, pharmacogenetics and public health benefits for the subcontinent and the world at large.
Methods: WGS of 10,000 individuals from 99 populations spanning geography, ethno-linguistic identity and social sub-structuring. In phase-1, a preliminary analysis of 5750 individuals from 65 populations has been completed.
Results:
- 1)
Identification of 135.48million SNVs (117.8million biallelic SNVs, 10.6million biallelic INDELs, 6.97million multiallelic).
- 2)
More than 4.7million common variant candidates for association study design.
- 3)
27 million variants, which are overall rare (<1%overall) but common in atleast one population(>5%). About 7million of 27million are novel. This potentially can reclassify the definition of 213 InterVar variants which were identified by ACMG as deleterious.
- 4)
Identified 118 Level1A and 1B variants, classified by PharmGKB as adversely impacting drug response, of which 38 are in high frequency in different populations.
- 5)
More than two-third of the populations analyzed in phase-1, have homozygosity and shared identical-by-descent segments significantly more numerous and extensive compared to the Finnish.
Conclusion:
- 1)
An Unprecedented diversity, unraveling new understanding of population history
- 2)
Possible reclassification of hundreds of variants identified as deleterious.
- 3)
Enormous improvement in imputing power of genotypes using the GenomeIndia Data compared to what was previously available.
Grants: BT/GenomeIndia/2018
Conflict of Interest: None declared
C15.3 A PCA and F-statistics based framework to infer human population history
Divyaratan Popli 1, Benjamin Peter2
1Max Planck Institute for Evolutionary Anthropology, Evolutionary Genetics, Leipzig, Germany; 2Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
Background/Objectives: F-statistics and Principal Component Analysis (PCA) are among the most widely used methods to study human population history. PCA is a dimensionality reduction method to visualize the genetic distance of individuals, and is generally not used for statistical testing. In contrast, F-statistics measure the genetic drift between two, three, and four populations, and can be used to test hypotheses about admixture and structure. Unlike PCA, F-statistics require assignment of individuals to discrete populations.
Methods: In this study we provide a statistical framework to estimate F-statistics from PCA. Our approach is based on geometrical interpretation of F-statistics (Oteo-García 2021 and Peter, 2022). The accuracy of estimated F-statistic depends on the PCA method used. Therefore, using simulations as well as Neandertal genomes, we evaluate estimation of F-statistics from different PCA methods, and compare them to a popular software ADMIXTOOLS 2.
Results: We find that PCA methods like probabilistic PCA (PPCA) and Latent Subspace Estimation (LSE), which model sampling bias, are more accurate compared to classical PCA. An advantage of our framework is that we can reliably estimate F-statistics with low-coverage genomes with missing genotypes, which are common in ancient DNA studies. Additionally, calculating genetic distances on PCA allows us to use the entire dataset without the need to group individuals.
Conclusion: We provide a statistical framework to combine PCA and F-statistics, and show that this framework addresses limitations of both PCA and F-statistics, when applied independently.
Grants: European Research Council (Grant No.: 694707) to Svante Pääbo, European Research Council (Grant No.: 101042421) to Benjamin M. Peter
Conflict of Interest: None declared
C15.4 Recombination enables higher numbers of recessive genes, contributing to the emergence of sexual mating in complex organisms
Luis Aniello La Rocca 1, Peter Krawitz2, Anton Bovier1, Konrad Gerischer2, Hannah Klinkhammer2, Leonie Fohler2
1Institute for Applied Mathematics, University of Bonn, Bonn, Germany; 2Institute for Genomic Statistics and Bioinformatics, University of Bonn, Bonn, Germany
Background/Objectives: The drift-barrier hypothesis states that random genetic drift constraints the refinement of a phenotype under natural selection. The influence of effective population size and the genome-wide deleterious mutation rate were studied theoretically and an inverse relationship between mutation rate and genome size has been observed for many species. However, the effect of the recessive gene count, an important feature of the genomic architecture, is unknown.
Methods: In a Wright-Fisher model we studied the spread of N completely recessive, lethal diseases. Diploid individuals are represented with a 2xN matrix denoting wild type (0) and mutated alleles (1). Analytic results for specific cases were complemented by simulations across a broad parameter regime for number of genes, mutation- and recombination rates.
Results: Simulations revealed a generational transition to higher mutation burden and prevalence, linked to the extinction of least-loaded haplotype classes. Metastability persists over 100,000 generations. Recombination proves pivotal, preventing population collapse and maintaining mutation burden below 10. The drift-barrier-hypothesis is confirmed by a high mutation burden resulting population collapse.
Conclusion: This study advances our understanding of gene pool stability, and particularly the role of number of recessive disorders. Simulations showed the emergence of mutually exclusive haplotypes with lethal equivalents above 0.02 per generation. Recombination’s essential role in curbing mutation burden and stabilizing the gene pool is demonstrated. Insights into Muller’s ratchet dynamics are provided, stressing the significance of recessive gene count in genomic architecture for population stability.
Grants: DFG - German Excellence Strategy GZ 2047/1, Project-ID 390685813; GZ 2151, Project-ID 390873048
Conflict of Interest: None declared
C15.5 Estimating ancestry profiles by nearest neighbor regression in principal component space
Juha Riikonen 1, Sini Kerminen1, Aki Havulinna1;2, Matti Pirinen1;3;4
1Institute for Molecular Medicine Finland, Helsinki, Finland; 2Finnish Institute for Health and Welfare, Helsinki, Finland; 3University of Helsinki, Department of Public Health, Helsinki, Finland; 4University of Helsinki, Department of Mathematics and Statistics, Helsinki, Finland
Background: Haplotype-based tools are ideal for ancestry inference but computationally impractical for biobank-scale analyses. We describe an efficient alternative using principal components (PCs) for individual-level ancestry estimation. Contrary to existing tools like Rye1 that use reference samples with discrete population assignment, our approach enables using reference samples with ancestry proportions to multiple populations. We had data on 18,125 samples from the biobank of the Finnish Institute for Health and Welfare (THL) with PC-coordinates and ancestry profiles estimated using the haplotype-based tool SOURCEFIND2, based on 1,124 reference samples allocated to 10 Finnish source populations2.
Methods: For a test set of 1,000 non-reference samples we computed their pairwise eigenvalue-weighted Euclidean distances to the reference samples in the 10-dimensional PC space. Using k-nearest neighbor (kNN) regression over the reference sample profiles, we estimated the test samples’ ancestry proportions to each source population.
Results: Estimating over k = 25 nearest reference samples, the mean total variation distance (TVD) between the test samples’ SOURCEFIND and kNN-profiles was 0.173. With the same reference samples, Rye showed a slightly larger mean TVD of 0.224. Using kNN-regression on all samples instead of the reference samples alone, reduced the TVD to 0.113.
Conclusion: As measured by TVD, our kNN-approach outperformed Rye. The method relies only on PCs and enables inference based on reference samples with continuous ancestry profiles, making it a promising tool for ancestry estimation in large-scale genomic studies.
Grants: Juselius foundation
1. Kerminen, PLoS Genetics 2021
2. Chacón-Duque, Nature Communications 2018
3. Conley, Nucleic Acids Research 2023
Conflict of Interest: Juha Riikonen: None declared, Sini Kerminen Nightingale Health Plc, stock options, Nightingale Health Plc, Full-time, Aki Havulinna: None declared, Matti Pirinen: None declared
C15.6 Heterozygote selection reduces recessive intellectual disability
Hila Fridman 1;2;3, Gelana Khazeeva1, Ephrat Levy-Lahad2;3, Christian Gilissen1, Han Brunner4;5
1Radboud University Medical Center, Department of Human Genetics, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands; 2Shaare Zedek Medical Center, Medical Genetics Institute, Jerusalem, Israel; 3The Hebrew University of Jerusalem, Faculty of Medicine, Jerusalem, Israel; 4Radboud University Medical Centre, 4Department of Human Genetics and Donders Center for Neuroscience, Nijmegen, Netherlands; 5Maastricht University Medical Center, 5Department of Clinical Genetics, GROW-School for Oncology and Developmental Biology and MHENS School for Mental Health and Neuroscience, Maastricht, Netherlands
Background/Objectives: For recessive conditions, there is strong selection against biallelic pathogenic variants (PVs) resulting in severe disease. Here we investigated whether heterozygosity for PVs in recessive genes has phenotypic consequences that may also be subject to selection.
Methods: We used exome data from 406,194 unrelated European individuals of the UK Biobank, and identified 55,746 different PVs in a curated set of 1929 recessive disease genes. We used three different constraint metrics to perform association analysis of the impact of heterozygosity for PVs in these genes on reproductive success and cognition.
Results: We find that heterozygous carriers in recessive constrained genes are more likely to be childless (OR:1.89, p:1.8E-3 Bonferroni corrected), especially heterozygotes for PVs in genes underlying intellectual disability (ID; OR:2.18, p:4.4E-2). Heterozygous variants in recessive ID genes also affect cognition (OR:0.29, p:1.0E-4). This effect is not seen for genes associated with other disorders. This negative selection is reflected in a threefold reduction in the frequency of heterozygosity for recessive ID in the population relative to other recessive disorders.
Conclusion: We find evidence for fitness effects in heterozygous carriers for recessive genes, especially for variants in constrained genes that underlie intellectual disability (ID), and show that such variants affect cognition. This negative selection is reflected in a drastically altered genetic landscape.
Grants: VIDI grant from the Dutch Research Council (917-17-353 to C.G.) and an AI for Health PhD grant from Radboudumc.
Conflict of Interest: None declared
C16 Neuromuscular omics
C16.1 Investigating germline instability in triplet repeat disorders in 11,415 family trios
Anupriya Dalmia 1;2, Valentina Galassi Deforie3, Kristina Ibañez2, Chris Clarkson2, Davina Hensman Moss4, Peter Holmans5, Arianna Tucci2
1UCL, UK DRI, London, United Kingdom; 2QMUL, Clinical Pharmacology, London; 3UCL, Neuromuscular Diseases; 4St George’s University of London; 5Cardiff University, Division of Psychological Medicine and Clinical Neurosciences
Consortium: 100,000 Genomes Project
Background/Objectives: Polyglutamine diseases stem from abnormal expansions of repetitive CAG trinucleotide repeats, notably in Huntington disease patients where one HTT gene copy exceeds 36 repeats. Intermediate-sized repeats (27-35) occur in ~6% of the population, often remaining asymptomatic but prone to expansion. We investigate intergenerational instability in normal and intermediate allele carriers across various polyglutamine disease genes.
Methods: In this study, we employed Expansion Hunter v3 to genotype the CAG tract in ATN1, ATXN1, ATXN2, ATXN3, ATXN7, CACNA1A, and HTT, utilizing whole-genome sequencing data from 11,415 family trios in the 100,000 Genomes Project. We, then, ascertain parental origin through haplotyping. We used regression-based analyses to identify key factors influencing germline instability.
Results: Consistent with existing literature, 5.9% of our parent population carries an intermediate allele for HTT. The instability percentage ranges from 0.2% to 2.5% across the seven analyzed genes. Although the prevalence of intermediate allele carriers and the clinically defined intermediate range vary among the genes, our analysis indicates that allele size is the primary predictor of instability. We observed a consistent pattern of increasing germline instability across six different genes (ATN1, ATXN1, ATXN2, ATXN3, ATXN7, and HTT), with instability becoming exponential at a common cut-off point.
Conclusion: Our research breaks new ground by exploring germline repeat instability in a largely neurologically healthy population. The consistent rise in instability at a shared threshold across multiple genes carries profound implications in the genetic counselling setting.
Grants: Barts Charity
Conflict of Interest: None declared
C16.2 Biallelic variants in the ribosomal protein S6 kinase C1 (RPS6KC1) cause a complex neurodevelopmental disorder
Laura Planas-Serra 1;2, Mar Rodríguez1;2, Agustí Rodriguez-Palmero1;2;3, Valentina Vélez-Santamaria2;4;5, Agatha Schlüter1;2, Edgard Verdura1;2, Gorka Gereñu6;7;8;9, Andrés Jiménez-Zúñiga6, Adolfo López de Munaín6;8;10, Alejandro Iñañez11;12, Josefina Casas11;12, Joan Josep Bech13, Carolina De La Torre13, Maria Iascone14, Romano Tenconi15, Reza Maroofian16, Ehsan Ghayoor Karimiani17, Suzanne Leal18;19, Isabelle Schrauwen18, Susanna Öhman20, Irma Järvelä21, Johanna Granvik22, Karit Reinson23;24, Elvira Kurvinen23, Katrin Ounap23;24, Tuğba Kalaycı25, Shahrashoub Sharifi25, Carlos Casasnovas2;4;26, Eric Anderson27, Udai Pandey27, Aurora Pujol2;4;28
1Bellvitge Biomedical Research Institute (IDIBELL), Neurometabolic Diseases Laboratory, L’Hospitalet de Llobregat, Spain; 2Instituto de Salud Carlos III, Centre for Biomedical Research on Rare Diseases (CIBERER), Madrid, Spain; 3Hospital Universitari Germans Trias i Pujol, Pediatric Neurology Unit, Barcelona, Spain; 4Bellvitge Biomedical Research Institute (IDIBELL), Neurometabolic Diseases Laboratory, L’Hospitalet de Llobregat, Spain; 5Hospital Universitari de Bellvitge, Neuromuscular Unit, L’Hospitalet de Llobregat, Spain; 6Biodonostia Health Research Institute, Neuromuscular Diseases Group, Donostia, Spain; 7Instituto de Salud Carlos III, Center on Biomedical Research Networking Neurodegenerative Diseases (CIBERNED), Madrid, Spain; 8Basque Foundation for Science, Ikerbasque, Bilbao, Spain; 9University of the Basque Country UPV-EHU, Department of Physiology, Leioa, Spain; 10University of the Basque Country UPV-EHU, Department of Neurosciences, Leioa, Spain; 11Institute of Advanced Chemical of Catalonia of CSIC (IQAC-CSIC), RUBAM, Department of Biological Chemistry, Madrid, Spain; 12Instituto de Salud Carlos III, Liver and Digestive Diseases Networking Biomedical Research Centre (CIBEREHD), Madrid, Spain; 13Josep Carreras Leukaemia Research Institute Barcelona, Proteomics Unit, Barcelona, Spain; 14ASST Papa Giovanni XXIII, Laboratory of Genetic Medicine, Bergamo, Italy; 15Università di Padova, Department of Pediatrics, Genetica Clinica, Padova, Italy; 16University College London, Department of Neuromuscular Diseases UCL Queen Square Institute of Neurology, London, United Kingdom; 17University of London, Molecular and Clinical Sciences Institute, St. George’s, London, United Kingdom; 18Columbia University Medical Center, Center for Statistical Genetics, Sergievsky Center, Department of Neurology, New York, United States; 19Columbia University Medical Center, Taub Institute for Alzheimer’s Disease and the Aging Brain, New York, United States; 20Kårkulla Joint Authority, Kirjala, Finland; 21University of Helsinki, Department of Medical Genetics, Helsinki, Finland; 22Wellbeing services county of Central Ostrobothnia, Kokkola, Finland; 23Tartu University Hospital, Department of Clinical Genetics, Genetic and Personalized Medicine Clinic, Tartu, Estonia; 24University of Tartu, Institute of Clinical Medicine, Tartu, Estonia; 25Istanbul University, Department of Medical Genetics, Istanbul Medical Faculty, Istanbul, Türkyie; 26Hospital Universitari de Bellvitge, Neuromuscular Unit, L’H; 27University of Pittsburgh Medical Center, Department of Pediatrics, Children’s Hospital of Pittsburgh, Pittsburgh, United States; 28Catalan Institution of Research and Advanced Studies (ICREA), Barcelona, Spain
Background/Objectives: The ribosomal protein S6 kinase family members play essential biological functions in disease, from cancer to intellectual disability. Little is known about RPS6KC1, aside from its lack of phosphorylation capacity and its roles in sphingosine-1-phosphate signaling and peroxiredoxin-3 transport to mitochondria.
Methods: We used whole-exome and -genome sequencing of undiagnosed patients with spastic paraplegia in a Spanish cohort and additional patients through GeneMatcher. Functional studies included proteomics, phosphoproteomics, and lipidomics in patients’ PBMCs and plasma, along with disease modeling in HAP1 cells and Drosophila melanogaster.
Results: We identified biallelic RPS6KC1 loss-of-function variants in 9 individuals from 6 independent families. The broad spectrum of phenotypic manifestations included neurodevelopmental delay, hypotonia, spastic paraplegia, and dysmorphic features, overlapping with those of Coffin-Lowry syndrome caused by RPS6KA3 loss.
Functional studies on patients’ samples indicated decreased expression and phosphorylation of RPS6, with subsequent impact on ribosomal protein synthesis, and a decrease in known interactors PRDX3 and SPHK1, along with a marked repression of mTOR and PI3K pathways. We detected an overall dysregulation of the lipidome, particularly phosphoinositides, and the enzymes controlling lipid synthesis. Functional studies in HAP1 RPS6KC1 knockdown cells suggested that RPS6KC1 may regulate PRDX3 and SPHK1 activities by facilitating their anchor at endosomes. In Drosophila melanogaster, RPS6KC1 knockdown resulted in motor dysfunction, defects at neuromuscular junctions, reduced lifespan, and decreased mTOR activity. Overexpression of mTOR improved motor function and lifespan in this model.
Conclusion: RPS6KC1 exerts crucial roles in neurodevelopment by controlling ribosomal protein synthesis, lipid signaling, and the mTOR pathway.
Conflict of Interest: None declared
C16.3 Deciphering the Genetic Landscape of Unsolved Congenital Myopathies: A Multinational Research Approach
Mridul Johari 1, Chiara Folland1, Marion ONNEE2;3, Machteld Oud4, Ana Töpf5, Yoshihiko Saito6, Anita Cairns7, Francesca Clementina Radio8, Adele D Amico9, Thatjana Gardeitchik10, Isabella Herman11, Sergei A. Kurbatov12, Pilar Marti13, Alayne Meyer14, Stefan Nicolau15, Kiran Polavarapu16, Thomas Robertson17, Juan Vilchez13, Gordana Kovacevic>18, Alan Beggs19, Daniel Calame11, Michael C. Fahey20, Nigel Laing1, Hanns Lochmuller16, James Lupski11, Edoardo Malfatti3, Ichizo Nishino6, Atchayaram Nalini21, Vedrana Milic18, Marco Tartaglia22, Volker Straub5, Bjarne Udd23, Nicol Voermans24, Gina Ravenscroft1
1Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands, WA, Australia, Perth, Australia; 2Institut Mondor de Recherche Biomédicale, Université Paris Est Créteil, INSERM U955, Créteil, France; 3Institut Mondor de Recherche Biomédicale, Université Paris Est Créteil, INSERM U955, Créteil, France, Paris, France; 4Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, Netherlands; 5John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom; 6Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan; 7Neurosciences Department, Queensland Children’s Hospital, Brisbane (Meanjin) Queensland, Brisbane, Australia; 8Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù IRCCS, Rome, Italy; 9Unit of Muscular and Neurodegenerative Disorders, Genetics and Rare Diseases Research Division, Department of Neurosciences, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy; 10Department of Human Genetics, Radboud university medical center, Nijmegen, Netherlands; 11Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States; 12Research Institute of Experimental Biology and Medicine, Voronezh N. N. Burdenko State Medical University, Voronezh, Russian Federation; 13Neuromuscular Unit, Department of Neurology, Hospital Universitari i Politècnic La Fe, Valencia, Spain; 14Division of Genetic and Genomic Medicine, Nationwide Children’s Hospital, Columbus, United States; 15Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, United States; 16Children’s Hospital of Eastern Ontario Research Institute; Division of Neurology, Department of Medicine, The Ottawa Hospital; and Brain and Mind Research Institute, University of Ottawa, Ottawa, Canada; 17Anatomical Pathology, Queensland Pathology, Brisbane, Queensland, Australia, Brisbane, Australia; 18Faculty of Medicine, University of Belgrade, Belgrade, Serbia; 19The Manton Center for Orphan Disease Research, Division of Genetics and Genomics, Boston Children’s Hospital, Harvard Medical School, Boston, United States; 20Department of Paediatrics Monash Children’s Hospital, Melbourne, Australia; 21Department of Neurology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India; 22Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy; 23Folkhälsan Research Center, Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland; 24Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, Netherlands
Consortium: Congenital myopathy consortium
Background/Objectives: Congenital myopathies (CM) are a diverse group of inherited neuromuscular disorders most frequently characterized by muscle weakness and structural abnormalities on muscle biopsy. A significant proportion of CM cases remain genetically unsolved at a single-centre level, underscoring the necessity for comprehensive research approaches that leverage international collaborations.
Methods: We analysed exome sequencing and short-read genome sequencing data from unsolved CM cases across multiple centres in Europe, the USA, Asia, and Australia. This analysis was complemented by dedicated expert curation of clinical and histopathological data, along with curation of de novo variants and Variants of Uncertain Significance (VUS). We further explored genetic pathomechanisms through in vitro studies, and bulk and single nucleus RNA sequencing (snRNAseq).
Results: We identified three novel forms of CM. (1) Loss-of-function variants in JPH1 were identified in three unrelated patients with striking bulbar and ocular involvement. (2) Biallelic variants in ASCC3 were identified in eight patients manifesting with a range of overlapping features including early-onset core myopathy, intellectual disability, and respiratory involvement. snRNAseq analysis highlighted the role of ASC-1 complex in translational homeostasis. (3) Our highly collaborative research revealed 12 unique missense variants in TUBA4A in 23 individuals, indicating a novel class of myopathy with de novo, autosomal dominant, and autosomal recessive inheritance. Previously, TUBA4A variants had been associated with FTD, ALS and ataxia. Thus, our study extends the phenotypic landscape of TUBA4A-related disease.
Conclusion: These findings emphasise the critical role of international collaborations in enhancing genetic interpretation and identifying novel disease genes and genotype-phenotype relationships in neuromuscular disease research.
Grants: AFM-Téléthon(24438)_Australian-NHMRC-Ideas-Grant-(APP2002640)
Conflict of Interest: None declared
C16.4 Allele-specific epigenome therapy in COL6-RD patient-derived primary fibroblasts
Franziska Haarich 1;2;3, Malte Spielmann2;4;5, Jeanette Erdmann1;2;3
1Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany; 2DZHK (German Research Centre for Cardiovascular Reasearch), partner site Hamburg/Lübeck/Kiel, Lübeck, Germany; 3University Heart Center Lübeck, Lübeck, Germany; 4Institute of Human Genetics, Universitätsklinikum Schleswig-Holstein, University of Lübeck and University of Kiel, Lübeck and Kiel, Germany; 5Human Molecular Genomics Group, Max Planck Institute for Molecular Genetics, Berlin, Germany
Background/Objectives: Collagen VI-related dystrophies (COL6-RD) are rare disorders caused by mutations in COL6A1, COL6A2 and COL6A3. Common symptoms include muscle weakness, contractures, hyperlaxity, skin abnormalities and respiratory insufficiency. There is no treatment available. Since many disease-causing variants are almost private, a mutation-independent therapy approach would be beneficial. In this project we use allele-specific epigenome editing as a potential therapy to knockdown dominant-negative mutation-carrying alleles to enable the healthy allele to function properly without targeting the mutation itself.
Methods: Variants in the promotor region of COL6A2 were identified and phased by long read sequencing with the disease-causing mutation for two unrelated COL6-RD patients harboring different dominant-negative Glycine substitutions. Allele-specific guideRNAs were designed targeting shared common variants. Cultured primary fibroblasts of these individuals were transfected with synthetic sgRNAs and in vitro transcribed mRNA coding for the epigenome editor CRISPRoff. Transfected cells were analyzed on gene expression and DNA methylation. With immunofluorescence staining the collagen VI secretion and network formation was examined.
Results: The expression of the mutated allele was significantly reduced and the DNA was allele-specifically methylated in the targeted region after the treatment with CRISPRoff and designed gRNAs. Successfully treated cells of both cell lines were enabled to secrete collagen VI into the extracellular matrix leading to a network formation.
Conclusion: Allele-specific and mutation-independent epigenome editing leads to a phenotypic improvement in cultured COL6-RD patient-derived primary fibroblasts. Targeting common variants instead of the mutation enables the treatment of very rare mutations.
Grants: The project is funded by CureCMD, MD Canada and DGM.
Conflict of Interest: None declared
C16.5 Following the clues in the transcripts: differential exon usage provides clinical and prognostic insights in the whole titinopathies spectrum
Maria Francesca Di Feo 1;2;3, Ali Oghabian4, Ella Nippala4, Francesca Forzano5;6, David Gómez-Andrés7, Angela F Brady8, Maria Iascone9, Anna Cereda10, Lidia Pezzani10, Daniel Natera-de Benito11, Andrés Nascimento11, Berta Estevez-Arias11, Sergei A. Kurbatov12, Tania Attie-Bitach13, Sheela Nampoothiri14, Erin Ryan15, Michelle Morrow15, Svetlana Gorokhova16;17, Brigitte Chabrol18, Johanna Nokelainen19, Lauri Snellman20, Francina Munell21, Peter Hackman4, Bjarne Udd4;22, Marco Savarese4;23
1Folkhälsan Research Centre, Biomedicum, Helsinki, Finland; 2Giannina Gaslini Institute, Clinical Genetics and Genomics Unit, Genova, Italy; 3University of Genoa, Department of Neurology, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Genova, Italy; 4Folkhälsan Research Centre, Helsinki, Finland; 5Guy’s and St Thomas’ NHS Foundation Trust, Dep. of Clinical Genetics, London, United Kingdom; 6King’s College London, London, United Kingdom; 7Vall d’Hebron University Hospital, Paediatric Neurology, Barcelona, Spain; 8North West Thames Regional Genetics Service, Northwick Park Hospital, Middlesex, United Kingdom; 9ASST Papa Giovanni XXIII Hospital, Medical Genetics Lab, Bergamo, Italy; 10ASST Papa Giovanni XXIII Hospital, Clinical Genetics Unit, Bergamo, Italy; 11Sant Joan de Déu Barcelona Hospital, Neuromuscular Disorders Unit, Pediatric Neurology Department, Esplugues de Llobregat, Spain; 12Voronezh N.N.Burdenko State Medical University, Voronez, Russian Federation; 13Necker Hospital, Service de Médecine Génomique des Maladies Rares, Paris, France; 14Amrita Institute of Medical Sciences & Research Centre, Dep. of Pediatric Genetics, Kerala, India; 15GeneDx, Gaithersburg, United States; 16Aix-Marseille University, Medical Genetics, Marseille, France; 17Assistance Publique Hôpitaux de Marseille, Dép. de Génétique Médicale, Marseille, France; 18Assistance Publique Hôpitaux de Marseille, Centre de Référence des Maladies Neuromusculaires de l’enfant PACARARE, Marseille, France; 19Helsinki University Hospital, Transplantation Laboratory, Helsinki, Finland; 20University of Helsinki, Coronary Disease Research Group, Helsinki, Finland; 21Hospital Materno-Infantil Vall d’Hebron, Unitat de Recerca Biomèdica, Barcelona, Spain; 22Tampere University Hospital, Dep. of Musculoskeletal Diseases, Tampere, Finland; 23University of Helsinki, Department of Medical Genetics, Helsinki, Finland
Background/Objectives: Titin (TTN), one of the largest genes with its 364 exons, generates numerous isoforms through potentially millions of splice pathways. Despite being well known as the molecular spring of the sarcomere, our understanding of titin’s differential expression has barely scratched the surface. Truncating variants in TTN (TTNtv) cause a wide range of neuromuscular disorders, posing challenges in determining clinical management and prognosis.
Methods: Short and long-read RNA sequencing was performed on a sample cohort including fetal and adult skeletal and heart muscles (over 60 samples) and utilizing ribosomal RNA-depletion for optimal TTN RNA-sequencing. An exon usage database (to be published for free use) was generated by a newly designed software (IntEREst). Then, molecular and clinical data of 12 patients carrying biallelic TTNtv were accrued from multiple hospitals.
Results: By looking at variants’ exon usage in our database, we were able to subdivide the patients in three categories that effectively matched their clinical trajectories: perinatal death, improvement with age, and aggravation with age. In some prenatal cases, exon usage allowed us to exclude titinopathy as a differential diagnosis. Interestingly, some novel fetal isoforms were discovered.
Conclusion: Interpreting TTN variants is crucial in the genomic era, as 1 in 100 individuals carries a TTNtv. Our study provides data for clinical and prognostic application and serves as a model for genetic disease personalized medicine. Almost 70% of human genes have at least 15 transcript isoforms, highlighting the need for a deeper consideration of this finely tuned evolutionary mechanism.
Grants: Academy of Finland, Samfundet Folkhälsan, Sydantutkimussaatio, Magnus Ehrnrooth Foundation
Conflict of Interest: None declared
C16.6 SENEGENE: insights into neuromuscular and neurogenetic diseases in Senegal, West Africa
Pedro M Rodriguez Cruz 1;2;3, Rokhaya Diagne3;4, Ainara Salazar Villacorta5, ALASSANE MAMADOU DIOP6, Alois Benoît Baba Diouf7;8, Marie Emilie Yandé Ndong8, Henriette Senghor4, Seynabou Dieng8, Christelle Rose Oko4, MARIETOU TRAORE9, Maimouna Santos10, Khalifa Ababacar Mbaye11, Lala Bouna Seck4, Mamadou Sy4, Maouly FALL6, Adjaratou Sow4;12, Ngor Side DIAGNE4, Loreto Martorell Sampol13, Daniel Natera-de Benito14, Andrés Nascimento14, Raul Tonda1;2, Steven Laurie1;2, Leslie Matalonga1, Henry Houlden5, Rokhaya Ndiaye15, Moustapha Ndiaye3;4, Sergi Beltran1;16, Amadou Gallo Diop4;15
1Centro Nacional de Análisis Genómico - CNAG, Barcelona, Spain; 2Universitat de Barcelona, Barcelona, Spain; 3Universite Cheikh Anta Diop (UCAD), Dakar, Senegal; 4Centre Hospitalier National Universitaire de Fann, Neurology, Dakar, Senegal; 5UCL Queen Square Institute of Neurology, London, United Kingdom; 6Centre Hospitalier National de Pikine, Neurology, Pikine, Senegal; 7Centre Talibou Dabo, Dakar, Senegal; 8Centre National Hospitalier d’Enfants Albert Royer, Paediatric Neurology, Dakar, Senegal; 9Centre Hospitalier Roi Baudouin de Guediawaye, Neurology, Senegal; 10Centre Hospitalier Régional de Saint-Louis, Neurology, Saint Louis, Senegal; 11Centre Hospitalier Régional de Ziguinchor, Neurology, Ziguinchor, Senegal; 12Centre National Hospitalier des Enfants de Diamniadio, Neurology, Diamniadio, Senegal; 13Sant Joan de Déu Barcelona Hospital, Unidad de Genética Molecular, Esplugues de Llobregat, Spain; 14Sant Joan de Déu Barcelona Hospital, Paediatric Neurology, Esplugues de Llobregat, Spain; 15Universite Cheikh Anta Diop (UCAD), Human genetics, Dakar, Senegal; 16Universitat de Barcelona, Genètica, Microbiologia i Estadística,, Barcelona, Spain
Background/objective: Rare genetic diseases remain largely understudied in sub-Saharan Africa. Here, we develop a global partnership to jointly advance the field of neuromuscular and neurogenetic diseases in Senegal, West Africa.
Methods: Adults and children suspected of neuromuscular or neurogenetic diseases, and relatives, are recruited from the Neurology Department of CHNU de Fann (Dakar) and systematically phenotyped using HPO terms. Sequencing (Barcelona & London) follows a progressive approach with MLPA, WES & WGS. The bioinformatic analysis uses state-of-the-art alignment, variant calling and annotation for SNV & InDels, CNVs, STRs, SVs and dedicated mtDNA analysis. Annotated variants are evaluated based on phenotype relevance, segregation, frequency and functional consequences using the RD-Connect Genome-Phenome Analysis Platform (GPAP).
Results: 841 individuals (401F:440M) have been recruited to date from over 350 families: 55% neuromuscular disorders, 19% neurometabolic diseases, 14% complex motor disorders and ataxia, 4% epilepsy and 8% miscellaneous. A definitive genetic diagnosis has been established in ∼65% of the cases (inheritance: 60% recessive, 10% dominant, 30% X-linked) allowing improved medical care and genetic counselling. Unreported variants of known disease genes were found in 29 cases and 3 novel disease candidate genes have been identified.
Conclusion: Our study unveils the first description of neuromuscular and neurogenetic diseases in Senegal,increasing diversity and novel pathogenic variant knowledge in human genetics. Collaborative efforts like SENEGENE contribute to advance rare diseases in underrepresented populations, improving clinical management, and facilitating connexions of local rare disease communities to available resources and existing networks for the benefit of patients and families.
Grants: AFM-Telethon (25078), La Caixa Foundation (LCF/BQ/PI21/11830012).
Conflict of Interest: Pedro M Rodriguez Cruz La Caixa Foundation Junior Leader Fellowship (LCF/BQ/PI21/11830012). AFM Telethon Trampoline grant (25078), Rokhaya Diagne: None declared, Ainara Salazar Villacorta: None declared, ALASSANE MAMADOU DIOP: None declared, Alois Benoît Baba Diouf: None declared, Marie Emilie Yandé Ndong: None declared, Henriette Senghor: None declared, Seynabou Dieng: None declared, Christelle Rose Oko: None declared, MARIETOU TRAORE: None declared, Maimouna Santos: None declared, Khalifa Ababacar Mbaye: None declared, Lala Bouna Seck: None declared, Mamadou Sy: None declared, Maouly FALL: None declared, Adjaratou Sow: None declared, Ngor Side DIAGNE: None declared, Loreto Martorell Sampol: None declared, Daniel Natera-de Benito: None declared, Andrés Nascimento: None declared, Raul Tonda: None declared, Steven Laurie: None declared, Leslie Matalonga: None declared, Henry Houlden: None declared, Rokhaya Ndiaye: None declared, Moustapha Ndiaye: None declared, Sergi Beltran: None declared, Amadou Gallo Diop: None declared
C18 New technologies to identify causal variants
C18.1 Metabolomic profiling of the entire UK Biobank enables interpretation of autosomal dominant disease mutations
Luke Jostins-Dean 1, Kirsten Schut1, Sini Kerminen1, Peter Wurtz1, Jeffrey Barrett1
1Nightingale Health Plc., Helsinki, Finland
Consortium: Nightingale Health Biobank Collaborative Group
Background/Objectives: We have generated metabolomic profiles of the entire UK Biobank. We sought to find new potential manifestations of rare diseases in human metabolism, and to investigate whether metabolic profiles can aid interpretation of candidate pathogenic variants.
Methods: We quantified 249 metabolite parameters from 490,830 UK Biobank participants and combined this with exome sequences for 200,643 participants. Based on high-confidence pathogenic mutations in ClinVar, we identified 44 autosomal dominant diseases with ≥5 carriers. We tested for association between each metabolic parameter and mutation status. For familial hypercholesterolemia, we also fitted a logistic predictive model to identify pathogenic mutations, using LDL levels, statin use and 17 principal components of the metabolic parameters.
Results: We identified ≥1 metabolite association at FDR < 0.05 for 8 diseases. These included expected associations, including dyslipidemia in familial hypercholesterolemia (FH) and hyperglycemia in monogenic diabetes, and previously unreported associations, including elevated serum acetone levels in neurofibromatosis and lowered levels of glycoprotein acetylation in Noonan syndrome. A model including all metabolites had a higher accuracy at identifying known pathogenic FH mutations (36/56 at FDR < 0.05), compared to LDL and statin use alone (30/56). Our model also identified 8 FH variants marked as uncertain significance in ClinVar with significant (FDR < 0.05) evidence of predicted pathogenicity.
Conclusion: Combined exome sequencing and large-scale metabolomic profiling in population biobanks can identify new mechanisms of metabolic dysregulation in genetic disease and improve our models for interpreting pathogenic mutations.
Grants: Nightingale Health.
Conflict of Interest: Luke Jostins-Dean Stock options in Nightingale Health Plc, Employee of Nightingale Health, Kirsten Schut Stock and stock options at Nightingale Health, Employee of Nightingale Health, Sini Kerminen Stock options for Nightingale Health, Employee at Nightingale Health, Peter Wurtz Stock and stock options at Nightingale Health, Employee of Nightingale Health, Jeffrey Barrett Stock and stock options at Nightingale Health, Employee of Nightingale Health
C18.2 Analysis of RNAseq from over 5000 individuals in the 100,000 Genomes Project identifies new potential diagnoses for patients with rare disease.
Diana Baralle 1, Jenny Lord2, Carolina Jaramillo Oquendo2, Mark Ross3;4, Terena James4, Lily Hoa5, Greg Elgar5
1University of Southampton; 2University of Sheffield; 3; 4Illumina; 5Genomics England
Consortium: N McGiness, A Ho, C Odhams.
Genomics England
Diagnosis of rare disorders has been revolutionised by whole exome and genome sequencing (WES/WGS), but even with WGS, around half of patients’ disorders remain unsolved. Interpretation of non-coding variation, which can affect splicing and gene regulation, has lagged behind coding variation, and improvement in this area will improve diagnostic yields. Here we present analyses of whole blood-based transcriptome sequencing data from over 5,000 probands with rare disorders that underwent WGS in the 100,000 Genomes Project, but did not receive a molecular diagnosis.
We used multiple tools to identify candidate expression (OUTRIDER via DROP) and splicing (LeafCutterMD, FRASER2 via DROP) outliers. PanelApp gene panels relevant to each proband’s phenotypes were applied and cross-referencing with WGS data was utilised to flag candidate diagnostic events.
We will present the 100,000 Genomes Project RNAseq cohort, including information on disease classes and demographics. OUTRIDER identified on average 3.1 expression outliers per proband genome wide, with 8% of the cohort possessing an outlier event in a disease gene in a relevant gene panel. LeafCutterMD with stringent filtering reduced this to 4.0, consistent with FRASER2’s number of identified candidates. Analysis and review of candidates is ongoing, but we estimate around 21% of the cohort has at least one significant outlier in a disease relevant gene. Interesting examples of diagnostic discoveries will be identified and highlighted.
Although work is ongoing, we estimate at least 20% of the sequenced cohort will have a diagnostic candidate, which is likely to bring a significant uplift to diagnostic rates.
Conflict of Interest: None declared
C18.3 Full-length transcript atlas of the developing human cortex uncovers novel candidate diagnoses in developmental disorders
Rosemary Bamford1, SziKay Leung1, Kartik Chundru 1;1, Caroline Wright1, Jonathan Mill1
1University of Exeter, Department of Clinical and Biomedical Sciences, Exeter, United Kingdom
Background/Objectives: Alternative splicing enables multiple RNA isoforms to be produced from a single mRNA precursor, resulting in high levels transcriptomic and proteomic diversity. Long-read sequencing approaches can be used to generate full-length transcript sequences and fully characterise isoform diversity.
Methods: We used Oxford Nanopore Technologies (ONT) transcriptome sequencing to profile transcript diversity in the human cortex across development (n = 26 prenatal; n = 21 postnatal).
Results: We identify widespread transcript diversity in the developing cortex and observe novel transcript isoforms, with 55% of transcripts in protein-coding genes not previously characterised. Of the most abundant transcript within protein coding genes, 19% were novel isoforms.
We identify developmental changes in alternative splicing, with widespread differential transcript expression between human foetal and adult cortex. This has important implications for variant interpretation, as variant consequences may differ between isoforms depending on the open reading frame. We find regions of transcripts predicted to be protein-coding that were previously thought to be non-coding, including novel exons. These novel coding regions are highly conserved with a mean PhyloP conservation of 0.74, significantly higher than GENCODE UTR and intronic regions (0.32 and 0.12 respectively). Within the novel coding regions of genes associated with dominant developmental disorders, we find 15 de novo variants from undiagnosed neurodevelopmental disorder trios in the Genomics England 100,000 genomes project. Of these, 9/15 are predicted to be non-synonymous in the novel transcripts, including two stop-gained variants in patients with phenotypes consistent with the corresponding single gene disorder.
Conclusion: This highlights the potential clinical utility of long-read transcriptomics.
Grants: Simons Foundation [120682]. Wellcome [226083/Z/22/Z]
Conflict of Interest: None declared
C18.4 CRISPR activation: a new tool for RNA diagnostics
Thorkild Terkelsen 1;2, Nanna S Mikkelsen2, Ebbe N Bak3, Johan Vad-Nielsen2, Jenny Blechingberg1, Simone Weiss3, Simon O Drue3, Henning Andersen4, Brage S Andresen5, Rasmus O Bak2, Uffe B Jensen1;2
1Aarhus University Hospital, Department of Clinical Genetics, Aarhus, Denmark; 2Aarhus University, Department of Biomedicine, Aarhus, Denmark; 3Aarhus University Hospital, Department of Molecular Medicine, Aarhus, Denmark; 4Aarhus University Hospital, Department of Neurology, Aarhus, Denmark; 5University of Southern Denmark, Department of Biochemistry and Molecular Biology, Odense, Denmark
Background/Objectives: Inadequate expression of tissue-specific gene transcripts in easily accessible cells from clinical samples is a major challenge for RNA diagnostics. We have recently evaluated the application of CRISPR activation (CRISPRa) to address this issue [1].
Methods: CRISPRa hijacks the capability of an inactive dCas9 protein to lead transcriptional activators to the gene of interest. The applied CRISPRa method uses an in vitro-transcribed mRNA, which encodes the activator complex, combined with guide RNAs that target the gene promotor. Pulsed delivery of this RNA mix to skin fibroblasts by simple electroporation induces gene upregulation within 24 hours. For RNA diagnostics, we have tested CRISPRa with reverse-transcription-PCR, short-read and long-read RNA sequencing platforms.
Results: We have applied CRISPRa in fibroblasts from individuals to revise the classification of suspected splice-altering variants in the myelin protein zero (MPZ) and spastin (SPAST) genes. With short-read RNA sequencing at routine diagnostic sequencing depths, CRISPRa resulted in excellent coverage of otherwise undetectable splice junctions in MPZ. Additionally, CRISPRa facilitated phasing of distant variants in SPAST by long-read RNA sequencing via increased abundance of the gene transcripts. In this way, CRISPRa supported the quantitative measurement of splicing impact.
Conclusion: CRISPRa is a promising new diagnostic tool to rapidly investigate the impact of splice-altering variants using easily accessible cells from individuals with rare diseases.
References: 1. Terkelsen T, Mikkelsen NS, Bak EN, Vad-Nielsen J, Blechingberg J, Weiss S, et al. CRISPR activation to characterize splice-altering variants in easily accessible cells. Am J Hum Genet. 2024;111(2):309-22.
Grants: DFF 9039-00337B
Conflict of Interest: Thorkild Terkelsen: None declared, Nanna S Mikkelsen: None declared, Ebbe N Bak: None declared, Johan Vad-Nielsen: None declared, Jenny Blechingberg: None declared, Simone Weiss: None declared, Simon O Drue: None declared, Henning Andersen: None declared, Brage S Andresen: None declared, Rasmus O Bak Holds equity in Graphite Bio and UNIKUM Tx., Paid consultant of UNIKUM Therapeutics, Uffe B Jensen: None declared
C18.5 Methylation quantitative trait loci mapping in more than 2,500 placentas provides insight into human complex disease.
Ariadna Cilleros-Portet 1, Sergi Marí1, Johanna Lepeule2, Johanna Tuhkanen3, Marta Cosín-Tomàs4, Marika Groleau5, Kelly Bakulski6, charles breeze7, Todd Everson8, Mariana F. Fernandez9;10;11;12, John Dou6, Loreto Santa-Marina9;13;14, barbara heude15, jia chen16, jari lahti3, Marie-France Hivert17;18;19, Mariona Bustamante4, Carmen Marsit8, Jose Ramon Bilbao1, Corina Lesseur16, Nora Fernandez-Jimenez1
1University of the Basque Country (UPV/EHU) and Biobizkaia Health Research Institute, Department of Genetics, Physical Anthropology and Animal Physiology, Leioa, Spain; 2University Grenoble Alpes, Inserm, CNRS, Team of Environmental Epidemiology Applied to Reproduction and Respiratory Health, IAB, Grenoble, France; 3University of Helsinki, Department of Psychology and Logopedics, Helsinki, Finland; 4Institute for Global Health (ISGlobal), Barcelona, Spain; 5Université de Sherbrooke, Département de Biologie, Sherbrooke, Canada; 6University of Michigan, School of Public Health, Department of Epidemiology, Ann Arbor, United States; 7National Cancer Institute, Division of Cancer Epidemiology and Genetics, Rockville, United States; 8Rollins School of Public Health at Emory University, Gangarosa Department of Environmental Health, Atlanta, United States; 9Instituto de Salud Carlos III, Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Madrid, Spain; 10University of Granada, Biomedical Research Center (CIBM), Granada, Spain; 11Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain; 12University of Granada, Department of Radiology and Physical Medicine, School of Medicine, Granada, Spain; 13Biogipuzkoa Health Research Institute, San Sebastian, Spain; 14Subdirectorate of Public Health of Gipuzkoa, Department of Health of the Basque Government, San Sebastian, Spain; 15Université de Paris, Centre for Research in Epidemiology and Statistics (CRESS), INSERM, INRAE, Paris, France; 16Icahn School of Medicine at Mount Sinai, Department of Environmental Medicine and Public Health, New York, United States; 17Harvard Medical School and Harvard Pilgrim Health Care Institute, Department of Population Medicine, Boston, United States; 18Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Sherbrooke, Canada; 19Massachusetts General Hospital, Diabetes Unit, Boston, Spain
Consortium: Pregnancy And Childhood Epigenetics (PACE) Consortium
Background/Objectives: The Developmental Origins of Health and Disease hypothesis (DOHaD) posits that the early-life environment can impact later-life health. The placenta plays a key role in DOHaD as it regulates the intrauterine environment. Placental DNA methylation (DNAm) has been suggested as a mechanistic link between intrauterine exposures, genetics, and adverse later-life health outcomes. Herein, we characterized genetic factors contributing to disease susceptibility through placental DNAm.
Methods: We meta-analysed placental methylation Quantitative Trait Loci (mQTLs) in 2,583 samples from nine cohorts of the international Pregnancy And Childhood Epigenetics (PACE) consortium. Each cohort mapped cis-mQTLs (0.5 Mb window) with TensorQTL. The meta-analysis was performed with the inverse variance weighted method of the software GWAMA. Afterwards, Stratified-LD Score Regression (S-LDSC) and Summary-based Mendelian Randomization (SMR) were performed to estimate the contribution of placental DNAm to 36 complex traits and disorders, by using the summary statistics of the largest GWAS available.
Results: We obtained more than 25 million placental cis-mQTLs, with 210,476 and 3,531,259 unique CpGs and SNPs, respectively. Placental mQTLs were enriched in genomic features with intermediate methylation values, and placenta specific accessible chromatin regions. S-LDSC and SMR analyses showed that SNPs linked to immune and anthropometric traits, and specific diseases such as schizophrenia, myocardial infraction, and Hashimoto’s thyroiditis are enriched for placental mQTLs.
Conclusion: The results obtained highlight the impact of genetics through placental DNAm in a range of different complex traits and disorders, reinforcing the role of the placenta in the DOHaD hypothesis.
Grants: GVSAN2018111086, PI18/01142, GVSAN2019111085, PI20/01116, PI21/01491, and PRB2-ISCIII.
Conflict of Interest: None declared
C18.6 Genetic regulators of whole blood gene expression and splicing and their impact on molecular traits and complex diseases
Elodie Persyn 1;2;3, Alex Tokolyi4, Artika Nath3;5, Katie L Burnham4, Jonathan Marten1;2, Thomas Vanderstichele4, Manuel Tardaguila4;6, David Stacey1;7;8, Ben Farr4, Vivek Iyer4, Xilin Jiang1;2;9, Samuel A. Lambert1;2;3, Guillaume Noell4, Michael Quail4, Diana Rajan4, Scott C. Ritchie1;2;3, Benjamin B. Sun10, Scott A.J. Thurston4, Yu Xu1;2;3, Christopher D. Whelan11, Heiko Runz10, Slavé Petrovski12;13, Daniel J. Gaffney4;14, David Roberts15, Emanuele Di Angelantonio1;2, James Peters16, Nicole Soranzo4;6, John Danesh1;2;4, Adam Butterworth1;2, Michael Inouye1;2;3, Emma Davenport4, Dirk Paul1;2;12
1British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom; 2Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, United Kingdom; 3Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom; 4Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom; 5Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Australia; 6Human Technopole, Fondazione Human Technopole, Milan, Italy; 7Australian Centre for Precision Health, University of South Australia, Adelaide, Australia; 8South Australian Health and Medical Research Institute, Adelaide, Australia; 9Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, United States; 10Translational Sciences, Research & Development, Biogen, Cambridge, United States; 11Neuroscience Data Science, Janssen Research & Development, Cambridge, United States; 12Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom; 13Department of Medicine, University of Melbourne, Austin Health, Melbourne, Australia; 14Genomics, BioMarin Pharmaceutical Inc., Novato, United States; 15Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom; 16Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
Background/Objectives: As many genetic risk factors are localised in non-coding regions of the genome, the integration of multi-omics datasets is often used to better understand downstream molecular mechanisms. In this study, we analysed RNA-sequencing data from the INTERVAL cohort to identify genetic regulators of gene expression and splicing events in whole blood and assess the impact of these genetic loci on other molecular phenotypes (proteins and metabolites) and complex diseases.
Methods: We tested the association between genetic variants and the expression of 19,173 genes and 111,937 splicing events, from the RNA-sequencing data of 4,732 INTERVAL participants. Shared genetic factors with other molecular traits and complex diseases were identified by conducting colocalization analyses. As subsets of INTERVAL participants had multiple -omics data available, we further tested the mediating effect of gene expression and splicing on other molecular traits.
Results: We found cis-quantitative trait loci (cis-QTL) for the expression of 17,233 genes and 29,514 splicing events. Genetic colocalization analyses identified 3,430 proteomic and metabolomic traits and 24 selected diseases with a shared genetic factor with either gene expression or splicing. Focusing on those colocalized signals, we found significant mediated effect on 222 molecular phenotypes.
Conclusion: By assessing the shared genetic aetiology with other molecular traits, our study provides new insights on molecular mechanisms. These results can be integrated in genetic studies to better understand complex diseases pathophysiology and identify new drug targets. The full description of our study is available on medRxiv with available data on https://IntervalRNA.org.uk.
Grants: BHF Programme Grant (RG/18/13/33946)
Conflict of Interest: Elodie Persyn E.P. was funded by the EU/EFPIA Innovative Medicines Initiative Joint Undertaking BigData@Heart grant 116074 and is funded by the NIHR BTRU in Donor Health and Behaviour (NIHR203337), Alex Tokolyi A.T. is supported by the Wellcome Trust (PhD studentship 222548/Z/21/Z), Artika Nath: None declared, Katie L Burnham: None declared, Jonathan Marten: None declared, Thomas Vanderstichele: None declared, Manuel Tardaguila: None declared, David Stacey: None declared, Ben Farr: None declared, Vivek Iyer: None declared, Xilin Jiang: None declared, Samuel A. Lambert: None declared, Guillaume Noell: None declared, Michael Quail M.A.Q. is on the KOL panel for New England Biolabs., Diana Rajan: None declared, Scott C. Ritchie S.C.R was funded by a BHF Programme Grant (RG/18/13/33946) and the NIHR Cambridge BRC (BRC-1215-20014; NIHR203312)., Benjamin B. Sun B.B.S. is a stockholder of Biogen, B.B.S. is an employee of Biogen, Scott A.J. Thurston: None declared, Yu Xu Y.X. is supported by the UK Economic and Social Research Council (ES/T013192/1)., Christopher D. Whelan C.D.W. is a stockholder of Johnson & Johnson, C.D.W. is an employee of Johnson & Johnson, Heiko Runz H.R. is a stockholder of Biogen, H.R. is an employee of Biogen, Slavé Petrovski S.P. is a stockholder of AstraZeneca, S.P. is an employee of AstraZeneca, Daniel J. Gaffney D.J.G. is a stockholder of BioMarin Pharmaceutical, D.J.G is an employee of BioMarin Pharmaceutical, David Roberts D.J.R. is an employee of NHS Blood and Transplant., Emanuele Di Angelantonio: None declared, James Peters J.E.P. has received hospitality and travel expenses to speak at Olink-sponsored academic meetings (none within the past 5 years), J.E.P. is supported by a Medical Research Foundation Fellowship (MRF-057-0003-RG-PETE-C0799), Nicole Soranzo: None declared, John Danesh J.D. holds a British Heart Foundation Professorship and a NIHR Senior Investigator Award., Adam Butterworth A.S.B. has received grants outside of this work from AstraZeneca, Bayer, Biogen, BioMarin and Sanofi, Michael Inouye M.I. is a trustee of the Public Health Genomics (PHG) Foundation. M.I has a research collaboration with AstraZeneca that is unrelated to this study, M.I. is supported by the Munz Chair of Cardiovascular Prediction and Prevention and the NIHR Cambridge Biomedical Research Centre (BRC-1215-20014; NIHR203312). M.I. is also supported by the UK Economic and Social Research Council (ES/T013192/1)., M.I is a member of the Scientific Advisory Board of Open Targets, Emma Davenport: None declared, Dirk Paul D.S.P is a stockholder of AstraZeneca, D.S.P is an employee of AtraZeneca
C19 Treatments for Genetic Disorders
C19.1 Inhibition of phosphodiesterase 10A by a specific antagonist rescues behavioral deficits and normalizes microglial morphology and synaptic pruning in a mouse model of FOXP1 syndrome
Henning Fröhlich 1, Jing Wang1, Ferdinand Althammer1, Tim Schubert1, Nina Kluck1, Valery Grinevich2, Stefanie Schmitteckert1, Christian Schaaf1, Gudrun Rappold1
1Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany; 2Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Department of Neuropeptide Research in Psychiatry, Mannheim, Germany
Background/Objectives: FOXP1 syndrome caused by FOXP1 haploinsufficiency is characterized by intellectual disability, speech and language impairment, autistic traits and neuropsychiatric abnormalities such as anxiety and hyperactivity. Despite research into its pathogenesis, effective treatment options are still lacking. Behavioral changes in patients are mirrored in Foxp1+/- mice. Phosphodiesterases (Pde) have been hypothesized to play a role in the pathophysiology of neurodevelopmental disorders. We investigated in Foxp1+/- animals whether Pde10a inhibition starting immediately after birth has a positive effect on the striatum and alleviates their behavioral deficits.
Methods: Pde10a mRNA and protein levels were determined by quantitative real-time PCR and western blot at P1, P12 and adulthood. Bulk expression of striatal tissue was analyzed with a NanoString nCounter Neuroinflammation Panel. One of the most advanced PDE10A antagonists was administered intraperitoneally and we studied its effect on behavior focusing on ultrasonic vocalization, anxiety and hyperactivity. We assessed astrocyte and microglia morphology on brain sections using 3D reconstruction by IMARIS.
Results: Foxp1+/- striata show significantly decreased Pde10a mRNA and protein levels from birth to adulthood. Moreover, they exhibit an upregulation of the neuroinflammation markers Cd74, Ifi30, Fcgr2b and Rhoa as well as increased microglial cell volume, decreased filament length and branching and decreased synaptic pruning. Pde10a inhibition in Foxp1+/- mice not only corrected the observed behavioral deficits, but also normalized microglial morphology and synaptic pruning without detectable side effects.
Conclusion: Reduced PDE10A expression most likely also contributes to the cognitive and behavioral deficits in FOXP1 syndrome, suggesting that PDE10A antagonists also alleviate neurological dysfunction in these individuals.
Grants: FR 3990/3-1
Conflict of Interest: None declared
C19.2 Nanoparticles-mediated siRNA delivery as a noninvasive personalized therapeutic strategy to hamper premature suture ossification in Crouzon syndrome
FEDERICA TIBERIO 1;2, Martina Salvati2, Elisabetta Falvo3, Luca Polito2, Noah Giacon4, Lorena Di Pietro1;2, Gianpiero Tamburrini5, Giada Tisci6, Pierpaolo Ceci3, Luca Massimi5, Ornella Parolini1;2, Alessandro Arcovito4, Wanda Lattanzi1;2
1Fondazione Policlinico Universatario Agostino Gemelli IRCSS, Rome, Italy; 2Università Cattolica del Sacro Cuore, Scienze della Vita e Sanità Pubblica, Roma; 3Consiglio Nazionale delle Ricerche, Istituto di Biologia Molecolare e Patologia, Rome, Italy; 4Università Cattolica del Sacro Cuore, Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche, e Perioperatorie, Roma, Italy; 5Fondazione Policlinico Universatario Agostino Gemelli IRCSS, Dipartimento di Neuroscienze, Rome, Italy; 6Sapienza Università di Roma, Dipartimento di Scienze Biochimiche, Rome, Italy
Background/Objectives: Crouzon syndrome (CS) is a rare genetic condition caused by heterozygous gain-of-function mutations in the Fibroblast growth factor receptor 2 (FGFR2) gene, which lead to the premature ossification of skull sutures. CS treatment is based on craniectomy, burdened by complications. This study aims at developing a noninvasive personalized FGFR2 knockdown strategy using allele-specific siRNAs, delivered by highly biocompatible polymeric nanoparticles (NPs).
Methods: Calvarial-derived mesenchymal stromal cells (CMSC) were isolated from surgical waste tissues of FGFR2 mutation-positive CS patients. siRNAs targeting mutant FGFR2 alleles have been designed and tested in CMSC. Their silencing and allele-discrimination efficiency were analyzed through gene (qPCR) and protein expression analyses. The intracellular delivery and trafficking of selected siRNAs vehicled through nanoparticles based on human recombinant ferritin and of Poly(d,l-lactide-co-glycolide) (PLGA) were evaluated by Incucyte Live-cells analysis system and confocal microscopy. siRNA-ferritin NPs and siRNA-PLGA complexes were produced and tested in CMSC by qPCR and Western Blot.
Results: For each enrolled patient, we developed specific siRNAs that reduced the expression of FGFR2 mutant allele by at least 50% without affecting the wild-type. The intracellular trafficking analysis showed an efficient internalization of both NPs, widely distributing within the CMSC cytoplasm. Our data demonstrated an efficient NP-mediated delivery and release of siRNA in cells, reducing FGFR2 expression levels by 40%.
Conclusion: Polymeric NPs-mediated siRNAs‘ allele-specific FGFR2 knockdown represents a suitable strategy to dampen the constitutive expression of FGFR2 in CS patients’ cells.
Grants: Funded by AFM-Telethon (Research Grant #23607) and by POR FESR Lazio 2014-2020 (Protocol: A0375-2020- 36576)
Conflict of Interest: FEDERICA TIBERIO Post-doc researcher full time, Martina Salvati PhD student full time, Elisabetta Falvo patent N. WO2016051340A1 entitled “A fusion protein, a nanoparticle composed by a plurality of monomers of said fusion protein and uses thereof”. The patent was sold to the company Thena Biotech S.r.l. in 2017. A Research Activity Agreement between the Institution of team#2 (C.N.R.) and Thena Biotech S.r.l. is currently operative for the projects involving the human ferritins as delivery systems., Researcher full time, Luca Polito Post-doc researcher full time, Noah Giacon PhD student full time, Lorena Di Pietro Researcher full time, Gianpiero Tamburrini MD full time and Full Professor, Giada Tisci PhD student full time, Pierpaolo Ceci patent N. WO2016051340A1 entitled “A fusion protein, a nanoparticle composed by a plurality of monomers of said fusion protein and uses thereof”. The patent was sold to the company Thena Biotech S.r.l. in 2017. A Research Activity Agreement between the Institution of team#2 (C.N.R.) and Thena Biotech S.r.l. is currently operative for the projects involving the human ferritins as delivery systems., Researcher full time, Luca Massimi MD full time, Ornella Parolini Full Professor, Alessandro Arcovito Associate Professor, Wanda Lattanzi Principal Investigator AFM-Telethon (Research Grant #23607) and POR FESR Lazio 2014-2020 (Protocol: A0375-2020- 36576), MD and associate professor full time
C19.3 Modulating splicing in 5’ untranslated regions to treat rare haploinsufficient disorders
Eloise Wells 1;2, Ruebena Dawes2, Stephan Sanders1;3, Nicola Whiffin2
1Institute of Developmental & Regenerative Medicine (IDRM) - University of Oxford, United Kingdom; 2Nuffield Department of Medicine (NDM), University of Oxford, United Kingdom; 3UCSF Joan and Sanford I. Weill Neurosciences Building, San Francisco, United States
Background/Objectives: Rare disorders collectively affect over 300 million people worldwide, but over 95% have no effective treatments. Recent advances in oligonucleotide and CRISPR editing technologies are driving therapeutic development. It is relatively straightforward to down-regulate genes therapeutically, for example with gapmer antisense oligonucleotides (ASOs). However, while haploinsufficiency is a major cause of rare disorders, upregulating protein levels is considerably more difficult.
Methods: We explore a novel approach to upregulation: modulating splicing of 5’ untranslated regions (5’UTRs) to increase protein translation. Specifically, we aim to selectively exclude 5’UTR exons that contain upstream open reading frames (uORFs), which are negative regulators of translation.
Results: We find 2,206 ‘skippable’ 5’UTR exons (i.e. flanked by other 5’UTR encoding exons) in 1,573 MANE transcripts. Using ribosome profiling data from human tissues including brain, heart, and skeletal muscle, we identify start sites of translated uORFs in 650 of these skippable exons (29%). Sixty-three of these uORF-containing exons are in the 5’UTRs of 48 genes with a dominant loss-of-function disease mechanism (from G2P, ClinVar, GenCC, and/or PanelApp). For each candidate gene, we assessed alternative 5’UTR splicing in long-read RNA-sequencing data and predicted changes in secondary structure and uORF translation in the presence and absence of the identified skippable exon(s). Subsequently, we prioritised nine target exons with a CRISPR-editable splice-acceptor site or that could be targeted by a splice-switching ASO for on-going functional validation.
Conclusion: Up-regulation of translation through modifying 5’UTR splicing is a promising therapeutic approach for a subset of haploinsufficient disorders.
Grants: Wellcome/Royal Society (220134/Z/20/Z)
Conflict of Interest: Eloise Wells: None declared, Ruebena Dawes: None declared, Stephan Sanders S.J.S receives research funding from BioMarin Pharmaceuticals, Nicola Whiffin Partner is an exmployee and owns stock options in Vertex Pharmaceuticals., Research funding from Novo Nordisk., Has consulted for ArgoBio.
C19.4 Microparticle sustained delivery of the calcineurin inhibitor FK506 to curtail autophagy and restore vision in an ADOA mouse model
Ana Paula Miranda Mendonca 1, ricardo rampado2, paolo caliceti3, stefano salmaso3, Luca Scorano1
1University of Padova, Department of Biology, Padua, Italy; 2Tel Aviv University, Tel Aviv-Yafo, Israel; 3University of Padova, Department of Pharmaceutical and Pharmacological Sciences, Padua, Italy
Background/Objectives: Autosomal dominant optic atrophy (ADOA) is the most common hereditary neuropathy that affects 1:35.000 people worldwide, and treatment is not available. Mutations in OPA1 in ADOA patients promote low levels of OPA1 protein, causing Retinal Ganglion Cells (RGCs) degeneration leading to blindness. Our group showed fragmented mitochondria, excluded from the axon but combined with autophagosomes in the axonal hillock of the ADOA mouse model. Moreover, genetic autophagy inhibition restores mitochondrial distribution in the RGC axon and curtails visual loss in the ADOA animal. Given that, this project aims to develop a pharmacological therapy through vitreal injection to curtail ADOA mouse visual loss.
Methods: We developed PLGA microparticles loaded with FK506 (PLGA-FK506), an inhibitor of autophagy in RGCs. The ADOA mouse model (Opa1fl/fl::Grik4-Cre mice) was subjected to an PLGA-FK506 intravitreal treatment over 3 months. The visual acuity of mice was measured by optokinetic tests.
Results: The ADOA mouse model loses visual acuity after 3 months. However, PLGA-FK506 treatment prevented loss of visual acuity in ADOA animals, and it was maintained over 3 months.
Conclusion: The treatment with PLGA-FK506 prevented ADOA’s vision loss for 3 months.
Grants: Marie Sklodowska-Curie grant n° 101030965 and Telethon Foundation project n° GGP19089.
Conflict of Interest: None declared
C19.5 Enhanced gene editing in patient-derived hiPSCs as a therapeutic strategy for inherited retinal dystrophies
Laura Siles 1, Sheila Ruiz Nogales1, Pilar Mendez1, Paula Gaudó1, Arnau Navinés-Ferrer1, Esther Pomares1
1IMO Grupo Miranza, Genetics, Barcelona
Background/Objectives: Inherited retinal dystrophies (IRD) are a group of rare disorders triggering blindness and other ocular affectations that have no treatment. They are caused by mutations in near 300 genes resulting in retinal cells degeneration. By using gene editing we aim to precisely correct IRD-related pathogenic variants (PV) as a therapeutic approximation for IRD.
Methods: We used CRISPR/Cas9 and TALEN technologies for the correction of PV in iPSCs derived from patients affected by retinitis pigmentosa, Stargardt disease, Best disease or achromatopsia. Edited cells were differentiated into in vitro retina cell models to analyze the reversion of disease-associated phenotypes.
Results: We have corrected seven hiPS cell lines without detected off-target defects. In most assays, at least 50% of the clones exhibit PV editing without undesired genomic abnormalitites. The highest gene editing results were obtained in homozygous mutations and small insertions/deletions, achieving a maximum efficiency of the 80%. Moreover, sgRNA specificity towards mutant allele decisively influences gene editing outcomes. Surprisingly, some hiPS clones edited the PV through the use of the wild-type allele as repair template in case of heterozygous carriers. Finally, corrected clones exhibit improved retinal cell differentiation and function coinciding with reversion of the diseased condition.
Conclusion: Gene editing represents a promising approach for IRD treatment, with CRISPR/Cas9 postulating as a more robust technology, but TALENs being appropriate in cases where CRISPR not. These results strengthen the study of gene editing as a therapeutic approximation for IRD, and suggest an unexpected potential application for PV repair by using patient’s own wild-type allele.
Grants: Fi-201401
Conflict of Interest: None declared
C19.6 Preventing Cancer in Lynch Syndrome: Vaccination with Mutation-Derived Neoantigen-Loaded Dendritic Cells Eliminates Precancerous Cells
Nicoline Hoogerbrugge 1, asima abidi2, harm westdorp3, mark gorris4, steve boudewijns4, Roland Kuiper5, reno bladergroen5, Tanya Bisseling6, anna de Goede7, Michelle van Rossum8, Alexandra Croockewit9, Marjolijn Ligtenberg1, Iris Nagtegaal10, Carl Figdor11, winald Gerritsen12, Gerty Schreibelt4, Jolanda de Vries4
1Radboudumc, Human Genetics, Nijmegen; 2Radboud; 3Radboudumc, Mecical Oncology, Nijmegen; 4Radboudumc, Medical BioSciences, Nijmegen; 5Princess Máxima Center for Pediatric Oncology, Human Genetics, Utrecht; 6Radboudumc, Gastroenterology, Nijmegen; 7Radboudumc, Pharmacy, Nijmegen; 8Radboudumc, Dermatology, Nijmegen; 9Radboudumc, Hematology, Nijmegen; 10Radboudumc, Pathology, Nijmegen; 11Radboudumc, Medical, Nijmegen; 12Radboudumc, Medical Oncology, Nijmegen
Background/Objectives: Individuals with Lynch syndrome (LS) have a hereditary risk of colorectal cancer and other cancers due to pathogenic variants in DNA mismatch repair genes. Mismatch repair deficiency results in the formation of neoantigens in the tumours. Dendritic cells (DC) loaded with these neoantigens can stimulate immune responses against tumour cells expressing these neoantigens, ultimately leading to tumour eradication. Our aim was to evaluate the potential of using DC-based vaccination in LS.
Methods: From 2012 to 2016, 23 individuals with LS from a phase I/II clinical trial received DC loaded with predicted neoantigen peptides from TGF-βRII, caspase-5, and carcinoembryonic antigen (CEA). Patients received up to 3 vaccination cycles, each consisting of 3 weekly injections.
Results: Neoantigen- and CEA-specific T-cells were detected in post-treatment skin biopsies in 87% of the patients. In-vitro expanded antigen specific T-cells were capable of specifically lysing tumour cells presenting 1) exogenously loaded peptides of all three (neo)antigens loaded on the DC’s and 2) endogenously processed TGF-βRII and CEA.
Higher probability of disease-free 10-year survival post vaccination was found to correlate with the presence of TGF-βRII specific T-cells.
Conclusion: LS patients who mounted a T-cell response against the neoantigen TGF-βRII did not develop cancer in almost 10 years. Within these 10 years, premalignant lesions did not exhibit the mutated form of TGF-βRII. This confirms that T-cells prevent the outgrowth of (pre)cancerous cells expressing the mutation-induced neopeptide, hence prevent the progression from adenoma to carcinoma. Prophylactic DC vaccination offers a new preventive treatment modality for patients with LS.
Conflict of Interest: None declared
C20 Large Scale Genetic Association Studies
C20.1 Exome-wide evidence of compound heterozygous effects across common phenotypes in the UK Biobank
Frederik Heymann Lassen 1, Samvida Venkatesh1, Nikolas Baya1, Wei Zhou2, Alex Bloemendal2, Benjamin Neale2, Benedikt Kessler3, Nicola Whiffin1, Cecilia Lindgren1, Duncan Palmer1
1University of Oxford, Oxford, United Kingdom; 2Broad Institute of MIT and Harvard, Cambridge, United States; 3University of Oxford, Target Discovery Institute, Oxford, United Kingdom
Background/objectives: Exome sequencing has identified rare protein-coding variants underlying many diseases, typically studying heterozygous and homozygous variation. However, the phenotypic impact of deleterious compound heterozygous (CH) variants on trait variation at scale remains largely unexplored.
Methods: We statistically phased rare variants (minor allele frequency (MAF) ~ 0.001%) in the UK Biobank exome sequencing data and analysed recessive effects in 175,587 individuals across 311 common diseases. Using a logistic mixed model framework, we investigated CH effects while accounting for relatedness, polygenic background, nearby common variants, and rare variant burden. We incorporated electronic health record age-at-diagnosis data to explore the longitudinal effects of bi-allelic variation. We replicate our associations in the 233,837 remaining UK Biobank participants. Finally, we developed an additive haplotype-based collapsing model, modelling the number of disrupted gene copies, comparing it with SAIGE-GENE+ burden testing.
Results: We found that 6.5% of individuals carry CH variants, and 90% these occur with a frequency below 0.34%. We find 19 significant (FDR < 0.1) recessive bi-allelic gene-trait associations, of which only six (31.1%) would have been discovered considering homozygosity alone. We find known bi-allelic gene-trait associations such as MUTYH and colorectal cancer (FDR < 0.1) and identify novel relationships such as ATP2C2-COPD (FDR < 0.1) driven by CH variation. Lastly, among our significant additive haplotype-based gene associations, 20/21 were more strongly associated than in SAIGE-GENE+ burden testing.
Conclusion: We demonstrate the utility of phasing large-scale cohorts for identification of the phenome-wide consequences of compound heterozygosity.
Grants: Wellcome Trust award 224894/Z/21/Z
Conflict of Interest: Frederik Heymann Lassen: None declared, Samvida Venkatesh: None declared, Nikolas Baya: None declared, Wei Zhou: None declared, Alex Bloemendal: None declared, Benjamin Neale B.M.N. is a member of the scientific advisory board at Deep Genomics and Neumora., Benedikt Kessler: None declared, Nicola Whiffin: None declared, Cecilia Lindgren: None declared, Duncan Palmer: None declared
C20.2 Unsupervised phenotyping and genetic discovery in 62,495 cardiac MRIs
Sara Ometto 1, Soumick Chatterjee1, Andrea Mario Vergani1, Alessia Visconti1, Emanuele Bianchi1, Edoardo Giacopuzzi1, Arianna Landini1, Sodbo Sharapov1, Francesco Cisternino1, Emanuel Soda1, Francesca Ieva1, Emanuele Di Angelantonio1, Nicola Pirastu1, Craig Anthony Glastonbury1
1Human Technopole, Milan, Italy
Background/Objectives: Temporal cardiac MRI imaging is an information rich modality but manually deriving multiple phenotypes of organ function via annotation is a rate limiting step. Autoencoders can learn high level (latent) semantic concepts from images in an unsupervised manner. These latent representations can capture complex phenotypes that can then be used to drive novel genetic discovery.
Methods: We developed a 3D diffusion autoencoder (3D-diffAE) to extract robust latent phenotypes from four chamber view long-axis cardiac MRIs across 62,495 subjects in UK Biobank (UKB). Latent phenotype-clinical associations were assessed using linear regression. Testing of common SNVs associated with latent phenotypes was performed using REGENIE. Conditional analysis was conducted with COJO and colocalisation performed using multi-trait coloc. Replication was performed in a held-out subset of UKB.
Results: Using 3D-DiffAE, we inferred latent phenotypes that are reproducible, heritable (4%-18%), and significantly associated with atrial fibrillation (P-value = 1.9e-88), myocardial infarction (P-value = 4.4e-71), and other outcomes in concurrently measured EHR data. GWAS (n = 47,827) identified 76 genome-wide significant associations in 182 endophenotypes mapping to 40 independent loci, of which 6 have not been previously described. By integrating GWAS summary statistics from 56 cardiometabolic traits, we find that 10 loci colocalise (PP4 > 0.9) with at least one derived cardiac phenotype and 6 with cardiac diseases.
Conclusion: We performed the largest systematic characterisation of cardiac endophenotypes to date, demonstrating that unsupervised phenotyping of cardiac MRIs using diffusion models provides a tractable means for novel genetic discovery and interpretation.
Grants: N.A
Conflict of Interest: None declared
C20.3 Decomposing sex-different phenotypic correlations in the UK Biobank into genetic and environmental components
Amelie Fritz 1;2;3, Liza Darrous1, Anders G. Pedersen2, Klaus Bønnelykke3, Zoltan Kutalik1
1UNIL - Université de Lausanne, Lausanne, Switzerland; 2Technical University of Denmark, Department of Health Tech, Kgs. Lyngby, Denmark; 3COPSAC, Gentofte, Denmark
Background/Objectives: Sexual dimorphism influences disease patterns, stature, and mental health. The role of genetic versus environmental factors in these sex-specific differences, particularly in traits linked to risk factors like smoking and lung function, are unclear but crucial for understanding disease etiology.
Methods: We developed a method to calculate the environmental correlation (rE) of trait pairs using phenotypic and genetic correlation (rG) and heritabilities. Taking advantage of big sample sizes and multiple phenotypes per individual, we analyzed 299 trait pairs from the UK Biobank exhibiting significantly different phenotypic correlations across sexes and investigated the extent to which this is influenced by environmental and genetic factors.
Results: We observed differing rG and rE across sexes for blood biomarkers e.g. c-reactive-protein~smoking with a greater contribution of genetic components in males (rGm = -0.38;95% CI = [-0.47, -0.29]/rEm = -0.07;95%CI = [-0.073, -0.069]) than females (rGf = -0.15;95%CI = [-0.23,-0.08]/rEf = -0.042;95%CI = [-0.044, -0.04]), whereas for hdl-cholesterol~education females show greater genetic contribution (rGf = 0.38;95%CI = [0.379, 0.46]/rEf = 0.05; 95%CI = [0.048,0.056] - rGm = 0.11;95%CI = [0.02,0.20]/rEm = 0.03;95%CI = [0.027,0.031]). These findings are supported by intra-trait rG between males and females being significantly different from 1, indicating differing genetic mechanisms across sexes.
Sex-specific significant differences were also observed in pairs involving testosterone with sex hormone-binding globulin, urate, waist-hip-ratio, and triglycerides.
Conclusion: We demonstrate that phenotypic correlations between trait pairs are differentially influenced by genetic and environmental effects across sexes, therefore different mechanisms may be responsible for the outcome of certain risk factors on traits or diseases. Future research will focus on clustering traits into groups representing different health aspects and assessing via Mendelian Randomization whether sex-different causal effects may give rise to the observed sex-dependent rG.
Grants: NovoNordiskFoundation
Conflict of Interest: None declared
C20.4 Large-scale exome sequencing identifies an enriched missense variant in HNF4A protective from type 2 diabetes (T2D) in South Asians
Moeen Riaz 1, Joshua Backman1, Bin Ye1, Manav Kapoor1, Jonas Bovijn1, Sameer Malhotra1, SR Chaudhury2, Md Alfazal Khan3, Md Khalequzzaman4, Adam Butterworth5, Luca Lotta1, Jonathan Marchini1, Aris Baras1, Goncalo Abecasis1, Alan R Shuldiner1
1Regeneron Genetics Center, New York, United States; 2National Heart Foundation Hospital & Research Institute, Dhaka, Bangladesh; 3icddrb, Dhaka, Bangladesh; 4Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh; 5University of Cambridge, Hinxton, United Kingdom
Consortium: Regeneron Genetics Center, the BELIEVE study group
Background: South Asians (SA) have a fourfold higher risk of T2D compared to Europeans. Protein-coding genetic variants that affect disease risk or protection can provide clues to disease pathogenesis or therapeutics targets.
Methods: We performed genome and exome-wide association analysis in two South Asian cohorts (BangladEsh Longitudinal Investigation of Emerging Vascular Events study [BELIEVE] and UK Biobank SA) using an additive model for single variants and SKAT/SKATO for gene-based test, adjusted for age, sex and 20 genetic PCs in REGENIE. Inverse-variance weighted meta-analysis was conducted, and GWAS summary statistics from the Genes and Health Study were added, making a total effective sample size of 11,205 T2D cases and 52,135 controls for exome analysis and 21,467 T2D cases and 86,061 controls for imputed GWAS analysis.
Results: 37 genome-wide significant (P < 5e-08) index variants were associated with T2D. These include 4 novel and 33 previously known common variants (MAF > 0.01). Gene burden masks showed no genome-wide significant associations with T2D. A novel protective association of a missense variant rs150776703 (p.Pro437Ser) in HNF4A (P = 1.738e-13, effect = 0.51) was identified. Rs150776703 is enriched in Bangladeshi (MAF 0.01) compared to gnomAD Non-Finnish Europeans (MAF 1.47E-05). We also observed nominal protective association of this variant with self-reported chronic kidney disease (P = 9.5e-4, OR 0.48). HNF4A encodes a transcription factor, and loss-of-function mutations cause MODY1. p.Pro437Ser occurs in the repression domain and is putatively a gain-of-function mutation.
Conclusion: Our findings provide first human genetic evidence for activation of HNF4A as a potential therapeutic approach for T2D.
Conflict of Interest: Moeen Riaz Regeneron Genetics Center or Regeneron Pharmaceuticals., Regeneron Genetics Center and Regeneron Pharmaceuticals., Joshua Backman Regeneron Genetics Center or Regeneron Pharmaceuticals., Regeneron Genetics Center and Regeneron Pharmaceuticals., Bin Ye Regeneron Genetics Center or Regeneron Pharmaceuticals., Regeneron Genetics Center and Regeneron Pharmaceuticals., Manav Kapoor Regeneron Genetics Center or Regeneron Pharmaceuticals., Regeneron Genetics Center and Regeneron Pharmaceuticals., Jonas Bovijn Regeneron Genetics Center or Regeneron Pharmaceuticals., Regeneron Genetics Center and Regeneron Pharmaceuticals., Sameer Malhotra Regeneron Genetics Center or Regeneron Pharmaceuticals., Regeneron Genetics Center and Regeneron Pharmaceuticals., SR Chaudhury: None declared, Md Alfazal Khan: None declared, Md Khalequzzaman: None declared, Adam Butterworth: None declared, Luca Lotta Regeneron Genetics Center or Regeneron Pharmaceuticals., Regeneron Genetics Center and Regeneron Pharmaceuticals., Jonathan Marchini Regeneron Genetics Center or Regeneron Pharmaceuticals., Regeneron Genetics Center and Regeneron Pharmaceuticals., Aris Baras Regeneron Genetics Center or Regeneron Pharmaceuticals., Regeneron Genetics Center and Regeneron Pharmaceuticals., Goncalo Abecasis Regeneron Genetics Center or Regeneron Pharmaceuticals., Regeneron Genetics Center and Regeneron Pharmaceuticals., Alan R Shuldiner Regeneron Genetics Center or Regeneron Pharmaceuticals., Regeneron Genetics Center and Regeneron Pharmaceuticals.
C20.5 An atlas of quantitative trait loci for a hundred gestational phenotypes
Siyang Liu 1, Hao Zheng1;2, yuqin Gu1, Zijing Yang1;3, Yanhong Liu1, Yuandan Wei1;2, Xinxin Guo1, Jianxin Zhen2, Fengxiang Wei3, Shujia Huang4, Xiu Qiu4
1Sun Yat-sen University, School of Public Health (Shenzhen), Shenzhen, China; 2Shenzhen Baoan Women’s and Children’s Hospital, Central Laboratory, Shenzhen, China; 3Longgang District Maternity & Child Healthcare Hospital of Shenzhen City, The Genetics Laboratory, Shenzhen, China; 4Guangzhou Women and Children’s Medical Center, Division of Birth Cohort Study, Guangzhou, China
Background/Objectives: The gestational period, spanning approximately 40 weeks from fertilization to birth, is pivotal in human reproduction. Monitoring the health of pregnant women and newborns during this period involves systematic prenatal and postpartum examinations, guided by indicators established by the national medical insurance system, collectively termed gestational phenotypes. However, our understanding of the genetic basis of these phenotypes and their intricate relationship with short- and long-term health outcomes remains markedly limited.
Methods: We conducted genome-wide association studies on over a hundred gestational phenotypes in 120,671 Chinese pregnancies, covering anthropometric metrics, non-invasive prenatal testing, and a series of biomarker measurements. Mendelian randomization was employed to explore potential causal relationships between the gestational phenotypes and birth outcomes, as well as long-term health outcomes.
Results: We identified 3,828 genetic loci, including 1,368 novel ones. We observed gestation-specific genetic effects, with a predominant enrichment in hematological pathways. Longitudinal trajectory analyses of repeated measurements of 24 phenotypes revealed significant fluctuations in genetic influences throughout gestational and postpartum periods. Intergenerational and two-sample Mendelian Randomization (MR) analyses combined with East Asian Biobanks suggested extensive causal associations between gestational phenotypes, birth outcomes, and the risk of future chronic diseases.
Conclusion: Our findings provide initial insights into the genetic foundations of human gestational phenotypes and their potential causal associations with short- and long-term health outcomes, laying the foundation for advanced population health during gestation.
Grants: This study received support from the Shenzhen Basic Research Foundation and the National Natural Science Foundation of China. The funders had no role in the study design.
Conflict of Interest: None declared
C20.6 Genome-wide association study using health registers in harmonized Nordic cohorts reveals different genetic architecture between early- and late-onset depression
John Shorter 1, Joëlle Pasman2, Andreas Jangmo3, Siim Kurvits4, joonas naamanka5, Espen Hagen6, Arvid Harder2, Jacob Bergstedt2, Patrick Sullivan2, Iiris Hovatta5, Thomas Werge7, Martin Tesli3, Kelli Lehto4, Yi Lu2, Alfonso Buil7
1Roskilde University, Roskilde, Denmark; 2Karolinska Institute, Sweden; 3Folkehelseinstituttet, Norway; 4University of Tartu, Tartu, Estonia; 5University of Helsinki, Helsinki, Finland; 6University of Oslo, Norway; 7Research Institute of Biological Psychiatry, Roskilde, Denmark
Consortium: TRYGGVE, iPSYCH, MoBa, FinnGen, Estonian Biobank
Background/Objectives: Major depressive disorder (MDD) is a common and heterogeneous mental disorder. Heterogeneity in symptoms, severity, and response to treatment hampers our ability to provide a biological characterization of MDD. Studying specific subtypes of more homogeneous patient groups may increase our chances of identifying the underlying genetic causes of the disorder and facilitate our efforts of designing targeted treatment strategies.
Methods: We investigated differences in the genetic architecture of early onset (eoMDD) and late onset (loMDD) depression; two MDD subtypes with potential etiological differences. We performed a meta-analysis using nine cohorts from five countries with detailed electronic healthcare registries and accumulated around 46K early onset (eoMDD, ≤25 years old at diagnosis) and 37K late onset (loMDD, ≥50 years at diagnosis) cases to compare the genetic architecture between these two age-related subtypes of MDD.
Results: We found that eoMDD and loMDD shared no genome-wide significant loci (P < 5x10-8), that genes associated with eoMDD have a role in neurodevelopment and suicide, that the SNP-heritability for eoMDD was nearly twice as high as loMDD, the two subtypes only had a modest genetic correlation, and the polygenic score for eoMDD is a significant predictor for suicide.
Conclusion: This study is the first to identify genetic loci differentially associated with early versus late-onset MDD. eoMDD is associated with a more severe clinical profile than loMDD, suggesting prioritization of treating and preventing severe outcomes in individuals with an early onset.
Grants: US NIMH MH123724
Conflict of Interest: None declared
C21 Late Breaking Abstracts
C21.1 De novo variants in the non-coding spliceosomal snRNA gene RNU4-2 are a frequent cause of syndromic neurodevelopmental disorders
Yuyang Chen 1;2, Ruebena Dawes1;2, Hyung Chul Kim1;2, Sarah Stenton3;4, Susan Walker5, Alicia Ljungdahl6;7, Jenny Lord8, Vijay S. Ganesh3;4;9, Jialan Ma3, Alexandra Martin-Geary1;2, Gabrielle Lemire3;4, Elston N. D’Souza1;2, Shan Dong6;7, Jamie Ellingford5;10;11, John Rubenstein7, Eirene Markenscoff-Papadimitriou7, Sebastian Fica12, Diana Baralle13;14, Christel Depienne15, Daniel MacArthur16;17, Joanna Howson18, Stephan Sanders6;7, Anne O’Donnell-Luria3;4;19, Nicola Whiffin1;2;3
1University of Oxford, Big Data Institute, Oxford, United Kingdom; 2University of Oxford, Centre for Human Genetics, Oxford, United Kingdom; 3Broad Institute of MIT and Harvard, Broad Center for Mendelian Genomics, Cambridge, United States; 4Boston Children’s Hospital, Harvard Medical School, Division of Genetics and Genomics, Boston, United States; 5Genomics England, London, United Kingdom; 6University of Oxford, Department of Paediatrics, Oxford, United Kingdom; 7University of California, UCSF Weill Institute for Neurosciences, San Francisco, United States; 8University of Sheffield, Sheffield Institute for Translational Neuroscience (SITraN), Sheffield, United Kingdom; 9Brigham and Women’s Hospital, Harvard Medical School, Department of Neurology, Boston, United States; 10Manchester University NHS Foundation Trust, Manchester Centre for Genomic Medicine, Manchester, United Kingdom; 11University of Manchester, Faculty of Biology, Medicines and Health, Manchester, United Kingdom; 12University of Oxford, Department of Biochemistry, Oxford, United Kingdom; 13University of Southampton, Faculty of Medicine, Southampton, United Kingdom; 14University Hospital Southampton National Health Service (NHS) Foundation Trust, National Institute for Health Research (NIHR) Southampton Biomedical Research Centre, Southampton, United Kingdom; 15University Duisburg-Essen, Institute of Human Genetics, University Hospital Essen, Essen, Germany; 16Garvan Institute of Medical Research and UNSW Sydney, Centre for Population Genomics, Sydney, Australia; 17Murdoch Children’s Research Institute, Centre for Population Genomics, Melbourne, Australia; 18Novo Nordisk Research Centre, Human Genetics Centre of Excellence, Oxford, United Kingdom; 19Massachusetts General Hospital, Harvard Medical School, Center for Genomic Medicine, Boston, United States
Consortium: RNU4-2 consortium
Background/Objectives: Around 60% of individuals with neurodevelopmental disorders (NDD) remain undiagnosed after comprehensive genetic testing, primarily of protein-coding genes. Increasingly, large genome-sequenced cohorts are improving our ability to discover new diagnoses in the non-coding genome.
Methods: Using a cohort of 8,841 probands with genetically undiagnosed NDD in Genomics England (GEL), we identify the non-coding RNA RNU4-2 as a novel syndromic NDD gene. RNU4-2 encodes the U4 small nuclear RNA (snRNA), which is a critical component of the major spliceosome.
Results: We identify an 18 bp region of RNU4-2 mapping to two structural elements in the U4/U6 snRNA duplex that is severely depleted of variation in the general population, but in which we identify heterozygous variants in 119 individuals with NDD across cohorts. This region is significantly enriched for variants in GEL NDD probands compared to individuals in the UK Biobank (OR = 85.8; 95%CI:56.4-131.6; Fisher’s P = 1.84x10-78). The majority of individuals with NDD (77.3%) have the same highly recurrent single base-pair insertion (n.64_65insT). We estimate that variants in this region explain 0.41% of individuals with NDD. We demonstrate that RNU4-2 is highly expressed in the developing human brain, in contrast to other U4 homologs, supporting RNU4-2’s role as the primary U4 transcript in the brain.
Conclusion: This work underscores the importance of non-coding genes in rare disorders. It will provide a diagnosis to thousands of individuals with NDD worldwide and catalyse development of effective treatments for these individuals.
Grants: due to word count limits, please follow this link (https://docs.google.com/document/d/1aJEbZZKtirtHutC5odTK9Wq40SKkoOSorWJBAYh_7zc/edit?usp=sharing) for grant information.
Conflict of Interest: Yuyang Chen: None declared, Ruebena Dawes: None declared, Hyung Chul Kim: None declared, Sarah Stenton: None declared, Susan Walker: None declared, Alicia Ljungdahl: None declared, Jenny Lord: None declared, Vijay S. Ganesh: None declared, Jialan Ma: None declared, Alexandra Martin-Geary: None declared, Gabrielle Lemire: None declared, Elston N. D’Souza: None declared, Shan Dong: None declared, Jamie Ellingford: None declared, John Rubenstein: None declared, Eirene Markenscoff-Papadimitriou: None declared, Sebastian Fica: None declared, Diana Baralle: None declared, Christel Depienne: None declared, Daniel MacArthur Microsoft, GlaxoSmithKline, Insitro, Overtone Theraputics, Joanna Howson Novo Nordisk, Novo Nordisk, Stephan Sanders BioMarin Pharmaceutical, Anne O’Donnell-Luria PacBio, Congenica, Tome Biosciences, Ono Pharma USA Inc., Nicola Whiffin Novo Nordisk, ArgoBio studio
C21.2 Mutations in the U4 snRNA gene RNU4-2 cause one of the most prevalent monogenic neurodevelopmental disorders
Daniel Greene 1;2, Chantal Thys3, Els Ortibus4;5, Andrew Mumford6, Kathleen Freson3, Ernest Turro1;2;7
1University of Cambridge, Department of Medicine, Cambridge, United Kingdom; 2Icahn School of Medicine at Mount Sinai, Department of Genetics and Genomic Sciences, New York, United States; 3KU Leuven, Department of Cardiovascular Sciences, Leuven, Belgium; 4KU Leuven, Department of Development and Regeneration, Leuven, Belgium; 5KU Leuven, Paediatric Neurology Department, Leuven, Belgium; 6University of Bristol, School of Cellular and Molecular Medicine, Bristol, United Kingdom; 7Icahn School of Medicine at Mount Sinai, Mindich Child Health and Development Institute, New York, United States
Background/Objectives: Although 1,427 genes have been identified confidently as aetiological for intellectual disability (ID), most cases remain unexplained. All but nine of the 1,427 known genes are protein-coding.
Methods: To discover non-coding aetiologies of ID, we performed a genetic association analysis comparing rare variants in 41,132 non-coding genes between 4,619 unrelated ID cases and 48,039 unrelated controls in the 100,000 Genomes Project.
Results: The most strongly associated gene was RNU4-2 (log Bayes factor = 64), one of two genes encoding the major spliceosome component U4 snRNA. We implicated variants in 47 cases in two short regions mapping to U4-U6 interactions in the aetiology of a syndrome characterised by ID, microcephaly, short stature, hypotonia, seizures and motor delay. We replicated our finding in three collections, bringing the number of cases to 72. Trio sequencing of 56 cases showed 55 alleles were de novo (one parental mosaic) and one was inherited. RNU4-2 variants explained more cases of neurodevelopmental abnormalities sequenced through the UK’s diagnostic service than any other gene except MECP2. RNA sequencing of patient fibroblasts (but not of blood) revealed abnormal splicing patterns similar to those in a recently discovered spliceosomopathy caused by loss-of-function of the U4 binding partner WBP4.
Conclusion: RNU4-2 mutations cause the most common neurodevelopmental disorder after Rett syndrome. This has remained concealed because the variants are usually de novo and the gene is soft-masked and omitted by exome sequencing. The observed lineage-specific splicing defects underscore the role of spliceosome dysfunction in the aetiologies of neurodevelopmental disorders.
Grants: R01HL161365.
Conflict of Interest: None declared
C21.3 Diagnostic Utility of Genome-wide DNA Methylation Analysis in Genetically Unsolved Developmental and Epileptic Encephalopathies and Refinement of a CHD2 Episignature
Christy LaFlamme 1, Cassandra Rastin2, Soham Sengupta1, Helen Pennington1, Sophie Russ-Hall3, Amy Schneider3, Emily Bonkowski1, Edith Almanza Fuerte1, Talia Allan3, Methylation Collaborative4, University of Washington Center for Rare Disease Research5, Sam Berkovic3, Lynette Sadleir6, Danny Miller7, Ingrid Scheffer3, Bekim Sadikovic2, Heather Mefford1
1St. Jude Children’s Research Hospital, Center for Pediatric Neurological Disease Research, Memphis, United States; 2Western University, Department of Pathology & Laboratory Medicine, London, Canada; 3The University of Melbourne, Department of Medicine, Parkville, Australia; 4Multiple Centers; 5University of Washington, Seattle, United States; 6University of Otago, Wellington, Department of Paediatrics and Child Health, Wellington, New Zealand; 7University of Washington Medical Center, Department of Laboratory Medicine and Pathology, Seattle, United States
Background/Objectives: Sequence-based genetic testing currently identifies causative genetic variants in ~50% of individuals with developmental and epileptic encephalopathies (DEEs). Aberrant changes in DNA methylation are implicated in various neurodevelopmental disorders but remain unstudied in DEEs. Rare epigenetic variations (“epivariants”) can drive disease by modulating gene expression at single loci. DNA methylation changes at many loci across the genome can result in distinct “episignature” biomarkers for monogenic disorders in a growing number of rare diseases.
Methods: Here, we interrogate the diagnostic utility of genome-wide DNA methylation array analysis on peripheral blood samples from 582 individuals with genetically unsolved DEEs who had previously undergone extensive genetic testing.
Results: We identified rare differentially methylated regions (DMRs) and explanatory episignatures to discover causative and candidate genetic etiologies in 12 individuals. We then used long-read sequencing to identify DNA variants underlying rare DMRs, including one balanced translocation, three CG-rich repeat expansions, and four copy number variants. We also identify pathogenic sequence variants associated with episignatures; some had been missed by previous exome sequencing. Although most DEE genes lack known episignatures, the increase in diagnostic yield for DNA methylation analysis in DEEs is comparable to the added yield of genome sequencing. Finally, we refine an episignature for CHD2 using 850K metharray and at higher CpG resolution using bisulfite sequencing to investigate potential insights into CHD2 pathophysiology.
Conclusion: Our study demonstrates the diagnostic yield of genome-wide DNA methylation analysis to identify causal and candidate genetic causes as 2% (12/582) for unsolved DEE cases.
Grants: CURE Epilepsy, AES
Conflict of Interest: Christy LaFlamme: None declared, Cassandra Rastin: None declared, Soham Sengupta: None declared, Helen Pennington: None declared, Sophie Russ-Hall: None declared, Amy Schneider: None declared, Emily Bonkowski: None declared, Edith Almanza Fuerte: None declared, Talia Allan: None declared, Methylation Collaborative: None declared, University of Washington Center for Rare Disease Research: None declared, Sam Berkovic: None declared, Lynette Sadleir L.G.S. receives funding from the Health Research Council of New Zealand and Cure Kids New Zealand. L.G.S. has received research grants and consultancy fees from Zynerba Pharmaceuticals., L.G.S. has received travel grants from Seqirus and Nutricia., L.G.S. is a consultant for the Epilepsy Consortium. L.G.S. has served on Takeda and Eisai Pharmaceuticals scientific advisory panels., Danny Miller D.E.M holds stock options in MyOme., D.E.M is engaged in a research agreement with ONT., D.E.M is on a scientific advisory board at ONT and has received travel support from ONT to speak on their behalf., Ingrid Scheffer I.E.S. may accrue future revenue on pending patent WO61/010176 (filed: 2008): Therapeutic Compound; has a patent for SCN1A testing held by Bionomics Inc and licensed to various diagnostic companies; has a patent molecular diagnostic/theragnostic target for benign familial infantile epilepsy (BFIE) [PRRT2] 2011904493 & 2012900190 and PCT/AU2012/001321 (TECH ID:2012-009)., I.E.S has served as an investigator for Anavex Life Sciences, Cerevel Therapeutics, Eisai, Encoded Therapeutics, EpiMinder Inc, Epygenyx, ES-Therapeutics, GW Pharma, Marinus, Neurocrine BioSciences, Ovid Therapeutics, Takeda Pharmaceuticals, UCB, Ultragenyx, Xenon Pharmaceuticals, Zogenix and Zynerba., I.E.S. has received speaker honoraria from GlaxoSmithKline, UCB, BioMarin, Biocodex, Chiesi, Liva Nova, Nutricia, Zuellig Pharma, Stoke Therapeutics and Eisai; has received funding for travel from UCB, Biocodex, GlaxoSmithKline, Biomarin, Encoded Therapeutics Stoke Therapeutics and Eisai., I.E.S. has served on scientific advisory boards for BioMarin, Chiesi, Eisai, Encoded Therapeutics, GlaxoSmithKline, Knopp Biosciences, Nutricia, Rogcon, Takeda Pharmaceuticals, UCB, Xenon Pharmaceuticals, Cerecin. I.E.S. has consulted for Care Beyond Diagnosis, Epilepsy Consortium, Atheneum Partners, Ovid Therapeutics, UCB, Zynerba Pharmaceuticals, BioMarin, Encoded Therapeutics and Biohaven Pharmaceuticals; and is a Non-Executive Director of Bellberry Ltd and a Director of the Australian Academy of Health and Medical Sciences and the Australian Council of Learned Academies Limited., Bekim Sadikovic B.S. is a shareholder in EpiSign Inc, a company involved in commercialization of EpiSignTM software., Heather Mefford: None declared
C21.4 Pathogenicity mechanism and gene therapy of hereditary spastic paraplegia caused by recurrent UBAP1 variant
Nicolas James Ho1, Lipeng Ding1, Min Chen1, Yu Shan Fung1, Dexuan Cui1, Linyan Meng2, Shen Gu 1
1The Chinese University of Hong Kong, School of Biomedical Sciences, Faculty of Medicine, Hong Kong, Hong Kong; 2Baylor College of Medicine, Department of Molecular and Human Genetics, Houston, United States
Background: Hereditary spastic paraplegia (HSP) is a group of neurological disorders characterized by upper motor neuron (UMN) degeneration, leading to spasticity and weakness in the lower extremities. We and others identified an autosomal dominant HSP subtype associated with the UBAP1 (ubiquitin-associated protein 1) gene.
Methods: Using CRISPR/Cas9, we established human embryonic stem cells (hESCs) and mouse models harboring the recurrent UBAP1 delGA variant found in 80% of our patients. Neural progenitor cells (NPCs) were further differentiated from hESCs. The three mouse models included a precise delGA mutant (Ubap1delGA/+), a knockout (Ubap1+/-), and a humanized knock-in expressing delGA UBAP1 (Rosa26∆UBAP1/+).
Results: Mutant UBAP1 mRNA escaped nonsense-mediated decay, resulting in stable expression of a truncated protein alongside wildtype (WT) UBAP1. Mutant NPCs exhibited accumulation of ubiquitinated proteins and abnormal endosomal dynamics. Ubap1delGA/+ and Rosa26∆UBAP1/+ mice demonstrated motor deficits. Single nucleus RNA sequencing analysis identified dysregulated gene expression predominantly in UMNs. In contrast, Ubap1+/- mice were phenotypically normal, suggesting loss-of-function is unlikely to cause HSP. These models recapitulated the HSP phenotype and demonstrated a dominant negative disease mechanism.
We developed therapeutics to suppress the mutant protein while preserving WT UBAP1. siRNAs reduced mutant protein levels by 75% while maintaining WT UBAP1 expression. Antisense oligonucleotides (ASOs) targeting mutant UBAP1 mRNA showed similar results. We are evaluating these approaches to rescue the HSP phenotype in our preclinical models.
Conclusion: Our study elucidated a dominant negative mechanism underlying UBAP1-HSP and underscored the importance of allele-specific gene-silencing therapy in treating the disease.
GRF/ECS #24101921, NSFC #82202045
Conflict of Interest: None declared
C21.5 Whole genome association testing and prediction in large-scale biobank data
Al Depope1, Jakub Bajzik1, Marco Mondelli1, Matthew Robinson 1
1Institute of Science and Technology Austria, Klosterneuburg, Austria
Background/Objectives: The recent release of whole-genome sequence (WGS) data for all UK Biobank participants facilitates investigating the impact of rare variants on complex traits. The common statistical approach of single-marker, or single-gene burden score regression, gives marginal associations that do not account for linkage disequilibrium, and in large sample size high density WGS data, even weak associations that are physically distant from causal variants will be discovered as significant. This limits our understanding of the genetic basis of human traits
Methods: We present a new algorithmic paradigm, gVAMP, that (i) directly fine-maps WGS variants and gene burden scores, conditional on all other measured DNA variations genome-wide, and (ii) provides optimal polygenic risk score prediction. This is done in one-shot, in lightning speed, allowing the analysis of datasets considered impossible before.
Results: On DNA Nexus, it takes just over one day to fine-map many thousands of human height-associated WGS variants and create a polygenic risk score with 48% prediction accuracy. We find 60 genes where rare coding mutations significantly influence phenotype, 76 X-chromosome associations, and thousands of autosomal associations localised to the single-locus level for five additional traits. Additionally, gVAMP outperforms summary statistic polygenic score methods, and outperforms REGENIE for standard association testing in a fraction of the compute time across 13 traits in imputed sequence data.
Conclusion: We showcase how to fine-map variants genome-wide in large sequencing datasets and provide open-source software.
Grants: Lopez-Loreta Prize and SNSF Eccellenza Grant PCEGP3-181181
Conflict of Interest: None declared
C21.6 Single-cell transcriptomics of post-implantation embryos: unveiling aneuploidy effects and lineage dynamics
Sheila Kwok 1;2, Lilach Marom Haham2, Stewart Russell2, Hanna Balakier2, Siwei Chen2, Manuel Viotti3, Svetlana Madjunkova2;4, Clifford Librach1;2;5;6;7
1University of Toronto, Department of Physiology, Toronto, Canada; 2CReATe Fertility Centre, Toronto, Canada; 3Zouves Fertility Center, Foster City, United States; 4University of Toronto, Department of Laboratory Medicine and Pathobiology, Toronto, Canada; 5University of Toronto, Department of Obstetrics and Gynaecology, Toronto, Canada; 6University of Toronto, Institute of Medical Science, Toronto, Canada; 7Sunnybrook Health Sciences Centre, Toronto, Canada
Background/Objectives: Embryo aneuploidy is clinically associated with pregnancy abnormalities. However, healthy live births resulting from diploid-aneuploid mosaic embryos indicate the early presence of aneuploid cells might not necessarily compromise embryo developmental competence. Here, we aim to investigate the global ploidy-specific transcriptomic signatures in post-implantation human embryos, revealing the molecular mechanisms determining aneuploid cell fates and survival.
Methods: Donated preimplantation euploid, aneuploid and mosaic embryos were cultured from day 5-6. On day 9-11, embryos were dissociated into single cells and pooled for scRNA-seq (10X Chromium). Sequencing results were analyzed using Freemuxlet to identify embryo-of-origin, Seurat for clustering and InferCNV to determine cell ploidy. Differential expression analyses were performed to identify enriched genetic markers and pathways.
Results: scRNA-seq data from 3159 cells identified five primary lineages, including epiblast, hypoblast, cytotrophoblast, syncytiotrophoblast and extravillous trophoblast. Differential expression analyses showed different intra-embryo environments facilitate distinct gene expression patterns related to energy expenditure, proteostasis, and cellular repair processes in the aneuploid cells. In homogeneous aneuploid embryos, temporary adaptive stress responses were observed, indicated by upregulated unfolded protein response (e.g. ATF4). In mosaic embryos, key machinery contributing to cell fitness is disrupted in aneuploid cells, including glycolytic process (e.g. HIF1A) and DNA damage repair (e.g. DDIT4).
Conclusion: Ploidy-specific transcriptomic analyses pointed to a diminished survival advantage in the aneuploid cells of post-implantation mosaic embryos. Differential fitness levels in the environment potentially induced self-correction by aneuploid cell depletion. These results provide the foundation for interrogating developmental competence of mosaic embryos, potentially expanding the range of viable embryos suitable for IVF treatments.
Grants:
Conflict of Interest: None declared
C22 What is in your brain?
C22.1 Epigenome and transcriptome changes in Kabuki syndrome 1 iPSCs and neuronal progenitors
Sara Cuvertino 1, Evgenii Martirosian1, Peiwen Cheng2, Ian Donaldson3, Terence Garner3, Adam Stevens4, Andrew Sharrocks5, Susan Kimber6, Siddharth Banka6
1University of Manchester, Division of Evolution and Genomic Sciences, Manchester, United Kingdom; 2University of Manchester, Bioinformatics and Genomic Technologies Core Facilities, Manchester, United Kingdom; 3University of Manchester, Division of Developmental Biology & Medicine, Manchester, United Kingdom; 4University of Manchester, Division of Molecular and Cellular function, Manchester, United Kingdom; 5University of Manchester, Division of Cell Matrix Biology and Regenerative Medicine, Manchester, United Kingdom; 6Manchester University Foundation NHS Trust, Manchester Centre for Genomic Medicine, Manchester, United Kingdom
Background/Objectives: Kabuki syndrome type-1 (KS1) is a neurodevelopmental disorder caused by loss-of-function variants in KMT2D which encodes a H3K4 methyltransferase. The mechanisms underlying neurodevelopmental problems in KS1 are unknown.
Methods: Three control and three patient-derived induced pluripotent stem cell (iPSC) lines were differentiated into neuronal progenitors (NPs) and cortical neurons (CNs). RT-PCR, western blots, RNASeq and H3K4me1 ChIPSeq were performed at different stages of differentiation.
Results: KS1 iPSCs showed significantly lower expression of functional KMT2D transcript and protein. We also found significantly lower H3K4me1 and H3K4me2 levels but not significant changes for H3K4me3 levels.
ChIPseq in iPSCs, NPs and CNs showed that the H3K4me1 loss in KS1 is region specific and primarily affects annotated enhancer regions. RNASeq in iPSCs revealed differentially expressed genes (DEGs) related to several KS1 phenotypes such as extracellular structure organization, heart morphogenesis, ear and axon development. In contrast, RNASeq at NP and CNs showed DEGs related to axonal development and ion transport.
Comparison of ChIPSeq and RNASeq data revealed a statistically significant enrichment of differentially downregulated genes in regions corresponding to enhancer regions with H3K4me1 loss across the three differentiation stages.
Motif analysis showed that the binding sites for SUZ12, a subunit of Polycomb Repressive complex 2, were most enriched in DEGs at all three stages of differentiation.
Conclusion: KS1 is characterised by epigenomic and transcriptomic dysregulation at all stages of neuronal differentiation. We present a disease relevant human cellular model that can be used for high throughput drug screening for KS1.
Grants: Great Ormond Street Hospital (GOSH) charity, NewLife
Conflict of Interest: None declared
C22.2 Single-cell genomics and regulatory networks for 388 human brains
Matthew Jensen 1;2, Declan Clarke1;2, Cagatay Dursun1;2, Prashant Emani1;2, Chirag Gupta3;4, Cheyu Lee5, Jason Liu1;2, Shaoke Lou1;2, Ran Meng1;2, Jonathan Warrell1;2, Siwei Xu5, Yuhang Chen1;2, Zhiyuan Chu1;2, Timur Galeev1;2, Michael Gancz1;2, Ahyeon Hwang5, Pengyu Ni1;2, Yan Xia1;2, Xiao Zhou1;2, Trygve Bakken6, Tanima Chatterjee1;2, Sophie Gaynor7;8, Ella Henry1;2, Ao Huang1;2, Yunzhe Jiang1;2, Ting Jin3;4, Yunyang Li1;2, Shuang Liu4, Shaojie Ma9, Eric Nguyen1;2, Ananya Rajagopalan1;2, Tiernon Riesenmy1;2, Brie Wamsley10, Gaoyuan Wang1;2, Andy Yang1;2, Suchen Zheng1;2, Michael Gandal11;12, Gabriel Hoffman13;14, Donghoon Lee13;14, Ed Lein6, Panos Roussos13;14, Nenad Šestan9, Zhiping Weng15, Kevin White16, Hyejung Won17, Matthew Girgenti18, Jing Zhang5, Daifeng Wang3;4, Daniel Geschwind10, Mark Gerstein1;2
1Yale University, Program in Computational Biology and Bioinformatics, New Haven, United States; 2Yale University, Department of Molecular Biophysics and Biochemistry, New Haven, United States; 3University of Wisconsin-Madison, Department of Biostatistics and Medical Informatics, Madison, United States; 4University of Wisconsin-Madison, Waisman Center, Madison, United States; 5University of California, Irvine, Department of Computer Science, Irvine, United States; 6Allen Institute of Brain Science, Seattle, United States; 7Tempus Labs, Inc., Chicago, United States; 8Cornell College, Department of Biology, Mount Vernon, United States; 9Yale University, Department of Neuroscience, New Haven, United States; 10University of California, Los Angeles, Department of Neurology, David Geffen School of Medicine, Los Angeles, United States; 11University of Pennsylvania, Department of Psychiatry, Perelman School of Medicine, Philadelphia, United States; 12The Children’s Hospital of Philadelphia, Lifespan Brain Institute, Philadelphia, United States; 13Icahn School of Medicine at Mount Sinai, Department of Psychiatry, New York, United States; 14Icahn School of Medicine at Mount Sinai, Department of Genetics and Genomic Science, New York, United States; 15UMass Chan Medical School, Worcester, United States; 16National University of Singapore, Yong Loo Lin School of Medicine, Singapore, Singapore; 17University of North Carolina at Chapel Hill, Chapel Hill, United States; 18Yale University, Department of Psychiatry, New Haven, United States
Consortium: PsychENCODE Consortium
Background/Objectives: Single-cell genomics offers a powerful means to understand the role of variants in gene expression and regulation, especially for the diverse cell types in the brain. Using these approaches, we can refine our understanding of how variants and gene regulation affect brain phenotypes, including neuropsychiatric disorders. However, population-scale cohorts with a wide range of brain disorders and traits are needed to infer significant associations between variants, regulatory elements, and expression, and to develop comprehensive models of gene regulation at the single-cell level.
Methods: Using a harmonized cell-typing scheme, we uniformly processed genotype, expression, and chromatin accessibility sequencing data in >2.8M single nuclei from the pre-frontal cortex of 388 adults with brain-related disorders (e.g., autism spectrum disorder, Alzheimer’s disease) and controls.
Results: We identified population and cell-type variation in expression and chromatin across 28 cell types for multiple gene families, including neurotransmitters and neuro-related drug targets. We found >550K cell-type-specific regulatory elements and >1.4M single-cell eQTLs, from which we built cell-type regulatory and cell-to-cell communication networks. These networks exhibit extensive cell-type-specific regulatory changes across traits, including enrichment of TF motifs during aging, and disorders, such as altered WNT and FGF signaling in schizophrenia and bipolar disorder. Using these networks, we constructed an integrative model to accurately impute single-cell expression, quantify disorder risk conditional on genotypes, and simulate perturbations, which prioritized ~250 disease-risk genes and drug targets with their associated cell types.
Conclusion: Our population-scale single-cell resource for the human brain can facilitate discovery of precision medicine approaches for neuropsychiatric disorders.
Grants: Funding provided by NIMH (U01MH116492).
Conflict of Interest: Matthew Jensen: None declared, Declan Clarke: None declared, Cagatay Dursun: None declared, Prashant Emani: None declared, Chirag Gupta: None declared, Cheyu Lee: None declared, Jason Liu: None declared, Shaoke Lou: None declared, Ran Meng: None declared, Jonathan Warrell: None declared, Siwei Xu: None declared, Yuhang Chen: None declared, Zhiyuan Chu: None declared, Timur Galeev: None declared, Michael Gancz: None declared, Ahyeon Hwang: None declared, Pengyu Ni: None declared, Yan Xia: None declared, Xiao Zhou: None declared, Trygve Bakken: None declared, Tanima Chatterjee: None declared, Sophie Gaynor Employee of Tempus Labs, Ella Henry: None declared, Ao Huang: None declared, Yunzhe Jiang: None declared, Ting Jin: None declared, Yunyang Li: None declared, Shuang Liu: None declared, Shaojie Ma: None declared, Eric Nguyen: None declared, Ananya Rajagopalan: None declared, Tiernon Riesenmy: None declared, Brie Wamsley: None declared, Gaoyuan Wang: None declared, Andy Yang: None declared, Suchen Zheng: None declared, Michael Gandal PI for PsychENCODE grants (NIMH), Gabriel Hoffman PI for PsychENCODE grants (NIMH), Donghoon Lee PI for PsychENCODE grants (NIMH), Ed Lein PI for related BICCN grants (NIMH), Panos Roussos PI for PsychENCODE grants (NIMH), Nenad Šestan PI for PsychENCODE grants (NIMH), Zhiping Weng PI for PsychENCODE grants (NIMH), Kevin White PI for PsychENCODE grants (NIMH), Hyejung Won PI for PsychENCODE grants (NIMH), Matthew Girgenti PI on related grants (NIMH), Jing Zhang PI for PsychENCODE grants (NIMH), Daifeng Wang PI for PsychENCODE grants (NIMH), Daniel Geschwind PI for PsychENCODE grants (NIMH), Mark Gerstein PI for PsychENCODE grants (NIMH)
C22.3 Local heterochromatin mitigates the impact of a transposable element insertion causing a mendelian neurodegenerative disorder
Vivien Horvath 1, Raquel Garza1, Marie Jonsson1, Pia Johansson1, Anita Adami1, Georgia Christoforidou1, Ofelia Karlsson1, Laura Castilla-Vallmanya1, Patricia Gerdes1, Ninoslav Pandiloski1, Christopher Douse1, Johan Jakobsson1
1Lund University/Lund Stem Cell Center, Experimental Medical Science, Lund, Sweden
Background/Objectives: X-Linked Dystonia-Parkinsonism (XDP) is a mendelian neurodegenerative disorder. Recently, a polymorphic transposable element (TE) insertion in the 32nd intron of the TAF1 gene has been identified as the genetic factor responsible for this disease. The XDP-TE is associated with TAF1 mis-regulation, but the mechanisms behind this phenomenon remain elusive. We hypothesize that repressive epigenetic marks on the XDP-TE are key players in this process. Thus, here we aim to dissect the molecular intricacies that keep the XDP-TE at bay and identify how it triggers aberrant TAF1 expression, ultimately leading to XDP.
Methods: Leveraging advanced sequencing techniques and XDP patient-derived iPSCs and neural progenitor cells, we employed CUT&RUN and Oxford Nanopore Sequencing to identify epigenetic marks on the XDP-TE. To illuminate what factors establish these marks, and their effect on gene expression, we did CRISPR inhibition of various candidate genes coupled with RNA sequencing.
Results: We demonstrate that ZNF91 - a TE-binding KRAB-Zinc Finger Protein - establishes H3K9me3 and DNA methylation over the XDP-TE in a cell type specific manner in patient derived cells. Moreover, removal of DNA methylation, or both H3K9me3 and DNA methylation, severely aggravates the XDP molecular phenotype, causing a reduced TAF1 expression and increased intron retention.
Conclusion: Our study unveils how a polymorphic TE results in XDP and highlights DNA methylation as potential therapeutic target. Moreover, this work underscores the significance of studying polymorphic TEs as disease triggers, with implications for various disorders.
Grants: Collaborative Center for XDP, Torsten och Elsa Segerfalk Foundation
Conflict of Interest: None declared
C22.4 Dissecting the pleiotropic effects of MORC2 mutations through multi-omics analysis
Fatemeh Peymani 1;2, Tomohiro Ebihara2;3, Dmitrii Smirnov1, Felix Distelmaier4, Daniele Ghezzi5, Robert Kopajtich1;2, Thomas Klopstock6, Costanza Lamperti5, Kei Murayama7;8, Fang Fang9, Agnès Rötig10, Christian Schlein11, Polina Tsygankova12, Aysylu Murtazina12, Thomas Meitinger1, Masaru Shimura2;8, Holger Prokisch1;2
1Technical University of Munich, School of Medicine, Institute of Human Genetics, Munich, Germany; 2Helmholtz Munich, Computational Health Center, Institute of Neurogenomics, Oberschleißheim, Germany; 3Chiba Children’s Hospita, Department of Neonatology,, Funabashi, Japan; 4Heinrich Heine University Düsseldorf, Düsseldorf, Germany; 5Istituto Neurologico “Carlo Besta” | Fondazione IRCCS, Milano, Italy; 6Friedrich-Baur-Institut am LMU Klinikum, Department of Neurology, München, Germany; 7Juntendo University, Diagnostics and Therapeutics of Intractable Diseases, Intractable Disease Research Center, Graduate School of Medicine, Bunkyo City, Japan; 8Chiba Children’s Hospital, Department of Metabolism, Chiba, Japan; 9Beijing Children’s Hospital, Department of Neurology, China; 10Université Paris Descartes, Institut IMAGINE, Paris, France; 11University Medical Center Hamburg-Eppendorf, Institute of Human Genetics, Hamburg, Germany; 12Research Centre for Medical Genetics, Moscow, Russian Federation
Consortium: GENOMIT consortium
Background/Objectives: Variants in the epigenetic silencing factor MORC2 are linked to a spectrum of neurological disorders, encompassing Charcot-Marie-Tooth (CMT), Leigh-spectrum syndrome (LSS), Cockayne-like syndrome (CLS) and DIGFAN. The molecular mechanism underlying diverse clinical manifestations of MORC2-disease remains largely unexplored. We leveraged multi-omics to dissect the phenotypic spectrum.
Methods: We diagnosed 30 patients with de novo mutations in MORC2 and performed a multi-omics comprising RNA-seq, proteomics, and DNA methylation from blood (n = 13) and fibroblasts (n = 13). Epigenome-wide, transcriptome-wide, and proteome-wide association studies were undertaken using >400 diseased and >20 healthy controls for each omics layer. Outliers were called using OUTRIDER. SVM classifiers were trained to establish diagnostic signatures.
Results: Amongst 100 disorders, we identified MORC2-specific signatures in DNA methylation and, for the first time, in transcriptome. MORC2-specific episignature were independently discovered by outliers and EWAS, while RNA-signature were only detected by outliers. Both signatures were driven by the same genes; hypermethylation caused lower expression. Episignature was conserved across tissues. Proteomics confirmed functional consequences of dysregulated genes. Interestingly, among the MORC2-specific outliers, we identified genes associated with LSS, CMT, CSL, and DIGFAN. The level of epigenetic silencing and gene expression of those genes strongly correlates with phenotypic manifestation. This is exemplified by hypermethylation of respiratory chain subunits, reduced expression, lower levels of protein complexes, and diminished enzymatic activity in LSS patients.
Conclusion: We demonstrated the power of multi-omics analysis in deciphering the hitherto unexplained phenotypic spectrum of MORC2-associated disorders. We provide the first example in which molecular signatures are not only diagnostic biomarkers but explain underlying pathomechanisms.
Grants: DMB-1805-0002
Conflict of Interest: None declared
C22.5 Investigating the role of SIRT6 in neurodevelopmental delay using Drosophila Melanogaster as an animal model
Clarissa Rocca 1, Gabriel Aughey1, Reza Maroofian1, Henry Houlden1, James Jepson1
1UCL Queen Square Institute of Neurology, London, United Kingdom
Background/Objectives: NAD-dependent histone deacetylase sirtuin-6 is an enzyme encoded by SIRT6 which is localised in the nucleus and plays a crucial role in chromatin homeostasis regulation, DNA signalling and repair, and is involved in glucose and lipid metabolism. Although the role of SIRT6 in human development remains unclear, studies suggest an association with neurodevelopmental processes. We identified SIRT6 as a novel human disease gene and investigated the effect of protein knock-down in Drosophila Melanogaster.
Methods: Exome analysis was performed on ~35,000 cases. We used the GAL4-UAS system to express the interfering shRNA (RNAi). First, we altered protein expression with the global driver actinGAL4. With the same approach, we then looked at neural stem cells (NSC) and post-mitotic effects. To investigate cell growth and division in specific clones, we employed the FLP/FRT system.
Results: We identified six different biallelic missense mutations in 20 patients from 10 unrelated families. The cases described show severe neurodevelopmental delay, including intellectual disability (n = 8) and focal and generalised seizures (n = 6). Knocking down SIRT6 in adult neurons led to decreased movement in experimental flies compared to controls. In contrast, protein knockdown in neuroblasts had a lethal effect on experimental flies.
Conclusion: In this study, targeted knockdown of the protein in adult neurons resulted in a notable reduction in movement without significant impact on overall viability. Interestingly, lethality occurred consequently to protein knockdown in NSC, a crucial component in the early and critical stages of neurodevelopment, therefore implying an essential role of SIRT6 protein in the neuronal development of Drosophila melanogaster.
Grants:
Conflict of Interest: None declared
C22.6 Long-read genome sequencing improves diagnostic sensitivity and aids gene identification
Karla Figueroa1, Elena Buena-Atienza2, Joohyun Park2, Sharan Paul1, Mandi Gandelman1, Neseebullah Kakar3, Marc Sturm2, Gunnar Schmidt4, Nicolas Casadei2, Yorck Hellenbroich3, Jelena Pozojevic3, Kristian Händler3, Simone Zittel-Dirks5, Dagmar Timmann-Braun6, Friedrich Erdlenbruch6, Claudia Dufke2, Lara Stühn2, Arnulf Koeppen7, Thomas Klopstock8, Florian Kraft9, Martin Zenker10, Nadja Ehmke11, Bernd Auber4, Ingo Kurth9, Malte Spielmann3, Stephan Ossowski2, Stefan M. Pulst1, Olaf Riess2;12, Tobias Haack 2;12
1University of Utah, Department of Neurology,, Salt Lake City, United States; 2University of Tübingen, Institute of Medical Genetics and Applied Genomics, Tübingen, Germany; 3Institute of Human Genetics, Lübeck, Germany; 4University of Hannover, Institute of Human Genetics, Hannover, Germany; 5University Medical Center Hamburg-Eppendorf, Department of Neurology, Hamburg, Germany; 6University of Duisburg-Essen, Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Essen; 7Veterans Affairs Medical Center, Albany, United States; 8University Hospital of Ludwig- Maximilians-Universität München, Department of Neurology with Friedrich-Baur-Institute, Munich, Germany; 9RWTH Aachen University Hospital, Institute for Human Genetics and Genomic Medicine, Aachen, Germany; 10Otto-von-Guericke University, Institute of Human Genetics, Magdeburg, Germany; 11Charité Berlin, Institute of Human Genetics, Berlin; 12University Tübingen, Center for rare diseases, Tübingen
Consortium: lonGER
Background: Clinical long-read genome sequencing (LR-GS) is expected to narrow the diagnostic gap and to identify novel disease genes and mechanisms. We aimed to determine the diagnostic accuracy of LR-GS and to discover the molecular correlate of SCA4, which has escaped identification for more than 25 years.
Methods: LR-GS was benchmarked using reference samples and various disease entities prone to genomic variation that are difficult to detect, including alterations in low complexity regions, pseudogenes, structural variation, repeat expansions, and imprinting disorders. Combined linkage and LR-GS studies were performed on a large Utah SCA4 pedigree, followed by matchmaking and cellular studies.
Results: LR-GS showed advantages in the diagnostic sensitivity across all types of genomic variation, apart from small insertions and deletions, with methylation calling as an additional readout. We identified a heterozygous GGC-repeat expansion (RE) in ZFHX3 as the likely molecular cause of SCA4. A query of 6,495 genomes and exchange with collaborators identified the RE in a further 7 pedigrees. Ultra-rare DNA variants near the RE indicated a common distant founder event in Sweden. The SCA4 phenotype extends from cerebellar and sensory ataxia, to autonomic dysfunction and chronic cough. Abnormal autophagy was observed in fibroblast and iPS cells, which normalized upon siRNA-mediated ZFHX3 knockdown.
Conclusion: LR-GS has the potential to serve as an all-in-one genetic test covering all types of (epi)genetic variation. The coding GGC-RE in an extremely GC-rich region was undetectable by short-read GS and demonstrates the power of LR-GS for variant detection. SCA4 adds to neuronal intranuclear inclusion disease and FXTAS as a polyglycinopathy.
Conflict of Interest: Karla Figueroa: None declared, Elena Buena-Atienza: None declared, Joohyun Park: None declared, Sharan Paul: None declared, Mandi Gandelman: None declared, Neseebullah Kakar: None declared, Marc Sturm: None declared, Gunnar Schmidt: None declared, Nicolas Casadei: None declared, Yorck Hellenbroich: None declared, Jelena Pozojevic: None declared, Kristian Händler: None declared, Simone Zittel-Dirks: None declared, Dagmar Timmann-Braun: None declared, Friedrich Erdlenbruch: None declared, Claudia Dufke: None declared, Lara Stühn: None declared, Arnulf Koeppen: None declared, Thomas Klopstock: None declared, Florian Kraft: None declared, Martin Zenker: None declared, Nadja Ehmke: None declared, Bernd Auber: None declared, Ingo Kurth: None declared, Malte Spielmann: None declared, Stephan Ossowski Received reimbursement for travel expenses and payment for conference presentations from Illumina Inc. and Oxford Nanopore Technologies, Stefan M. Pulst: None declared, Olaf Riess: None declared, Tobias Haack: None declared
C23 Expanding the boundaries of genetic counselling
C23.1 What does a consent conversation for whole genome sequencing look like? An observational study of consent appointments in the NHS Genomic Medicine Service in England
Holly Ellard 1;2, Celine Lewis1;2, Huda Alfardus1;2
1Cardiff University, Centre for Medical Education, Cardiff, United Kingdom; 2UCL Great Ormond Street Institute of Child Health, Population, Policy and Practice Department, London, United Kingdom
Background/Objectives: Consent to whole genome sequencing (WGS) through the NHS Genomic Medicine Service follows a new ‘patient choice’ model, covering clinical testing and an invitation to the National Genomic Research Library (NGRL). Little is known about what the consent conversation looks like in practice. This study analysed the structure and content of WGS consent appointments.
Methods: Participants were healthcare professionals (HCPs) (n = 20) and parents (n = 42) of paediatric patients being offered WGS during 26 consent appointments across seven NHS Trusts. Consent appointments were audio-recorded and analysed using deductive content analysis.
Results: Consent conversations took an average of 13 minutes 59 seconds, with administrative tasks taking an average additional 5 minutes 18 seconds (total average 19 minutes 17 seconds). An average of 29% of the conversation was dedicated to the various result types from WGS, 29% to the NGRL, 17% to incidental findings, and 15% to biological concepts. HCPs frequently discussed the possibility of receiving an uncertain result (25/26) or no diagnosis (24/26), but less commonly discussed the risk of uncovering non-paternity (12/26) or the potential for reanalysis (12/26). Parents asked questions around the logistics of sampling (8/26), testing process (6/26), and data security (3/26). Health-related incidental findings were occasionally the source of questions (2/26) and fears (1/26). Whilst parents expressed few concerns, HCPs did not always ask questions to elicit their thoughts, feelings, and values (12/26).
Conclusion: Parents and patients may benefit from further opportunities to explore their views and concerns during the consent conversation.
Grants: NIHR Advanced Fellowship Grant (NIHR300099)
Conflict of Interest: None declared
C23.2 Are we doing a good job of supporting parents who are making decisions about whole genome sequencing? A cross-sectional survey study conducted in the NHS Genomic Medicine Service
Celine Lewis1, Ria Patel 2, Bettina Friedrich1
1UCL GOS Institute of Child Health, Population, Policy and Practice, London; 2University College London, Medical School, London, United Kingdom
Background/Objectives: Whole genome sequencing (WGS) for paediatric rare disease diagnosis is now available as a first-line test for certain clinical indications in the Genomic Medicine Service (GMS). We conducted a quantitative survey comprising validated measures and bespoke questions to understand decision-making processes. Surveys were sent to parents of children offered WGS within 4 weeks of their appointment.
Methods: 1366 surveys were sent out across 7 NHS Trusts in England; 393 completed surveys were returned (29% response rate). Descriptive and inferential statistics were conducted.
Results: Parents were satisfied with their WGS appointment (mean =24.47; range 0-28). Parents had a positive attitude towards WGS (mean = 18.17; range 0-20), although those who had discussed WGS with a genetic counsellor/genomic associate had significantly more positive attitudes than those seen by genetic consultants (p < 0.001). Decisional conflict was low (mean = 20; range 0-64). Regarding knowledge of WGS, the higher parents scored, the less conflicted they felt about their decision (p = 0.003). There was a significant correlation between parents who had spent longer looking for a diagnosis (p = 0.015), had previously undergone genetic testing (p = 0.004), were White (p = 0.003), and had higher formal educational qualifications (p = 0.005) with higher WGS knowledge scores. Type of health professional discussing WGS was not associated with improved knowledge.
Conclusion: Low decisional conflict, good knowledge, and positive attitudes (coupled with test uptake) are indicators of informed decision-making. Our results provide nuance around those parents who might need additional support. Further work to understand how to improve knowledge of WGS is important.
Grants: Celine Lewis: NIHR Advanced Fellowship Grant (NIHR300099)
Conflict of Interest: None declared
C23.3 Ethical implementation of automated genomic reanalysis: Patient, professional, and public perspectives
Danya Vears 1;2;3;4, Fiona Lynch1;3, Jack Harrison2, Emily King1, Savio Nona1, Zornitza Stark2;5
1Murdoch Children’s Research Institute, Biomedical Ethics Research Group, Parkville, Australia; 2University of Melbourne, Department of Paediatrics, Parkville, Australia; 3University of Melbourne, Melbourne Law School, Carlton, Australia; 4KU Leuven, Centre for Biomedical Ethics and Law, Leuven, Belgium; 5Victorian Clinical Genetics Service, Parkville, Australia
Background/Objectives: Reanalysis of undiagnosed patients‘ genomic data two years after the initial test leads to a 10-15% increase in diagnostic yield. Yet reanalysis is far from routine and many patients miss out due to lack of workforce capacity and haphazard practices required to trigger this process. Automation of reanalysis using artificial intelligence (AI) is the solution proposed to provide routine reanalysis to all undiagnosed patients. However, little is known about how different stakeholders perceive this proposal.
Methods: To address this, we conducted online focus groups with members of the Australian public, patients/their carers, and genetics professionals, exploring their perspectives on the practical and ethical challenges, as well as potential solutions, for implementing automated reanalysis into clinical care.
Results: Seventy-eight stakeholders (23 patients/carers, 34 members of the public, 21 genetics professionals) participated across 20 focus groups. Overall, participants from all stakeholder groups were positive about using automated systems for reanalysis. They highlighted benefits relating to equity of access to the possibility of receiving a diagnosis and reducing the burden placed on patients to be responsible for triggering reanalysis. Genetics professionals raised concerns about how to obtain reconsent and the moral duty to return results to patients lost to follow up. In contrast, patients had mixed opinions regarding the need for reconsent and the public worried about who was accountable if results were inaccurate.
Conclusion: Our findings will be used to guide implementation of automated reanalysis, and also develop policies and educate the public about use of AI in genomics more broadly.
Grants: Australian Government MRFF GHFM APP2008820.
Conflict of Interest: Danya Vears Grant declared, Fiona Lynch: None declared, Jack Harrison: None declared, Emily King Grant declared, Savio Nona Grant declared, Zornitza Stark Grant declared
C23.4 The development and use of polygenic scores for social traits: Biobank participants’ perspectives
Margaret Waltz 1, Kristine Kuczynski1, Shawneequa Callier2, R. Jean Cadigan1
1University of North Carolina at Chapel Hill, Chapel Hill, United States; 2George Washington University, Washington, United States
Background/Objectives: Using large datasets from biobanks, polygenic scores (PGS) are being created for a variety of social traits. These scores allow researchers to study gene-environment interactions, but worries abound about their uses in commercial and social settings. Scant research examines the perspectives of biobank participants, whose data could be used to develop PGS for social traits.
Methods: We conducted a survey with over 400 U.S. based biobank participants and five follow-up focus groups exploring views on various PGS for social traits and their potential uses.
Results: We will focus on respondents’ views of PGS for three traits, educational attainment, aggression, and obesity, as well as their possible uses outside of research. Survey respondents revealed mixed feelings about PGS for these traits, particularly if used to help determine which embryos to use in in vitro fertilization. In focus groups, some could understand the desire to select for traits in the otherwise uncontrollable realm of parenting. But others noted fears of scores being used to stigmatize, discriminate, or eliminate certain traits from the population, especially since scores indicate likelihood, not guarantees, and may be based on problematic measures, like body mass index. As a result, respondents questioned whether they would want their data to be used to develop PGS for social traits.
Conclusion: We will discuss the ethical implications of these findings for biobanks when handling potential researcher requests for specimens as well as the merits of engaging with or potentially reconsenting biobank participants.
Grants: U.S. National Human Genome Research Institute (#R01HG012402)
Conflict of Interest: None declared
C23.5 Emerging issues that call for more inclusive strategies in engaging underrepresented groups regarding human germline gene editing
Diewertje Houtman 1, Wendy Geuverink2;3, Joosje Kist1, Isabel Retel Helmrich1, Martina Cornel2;4, Michelle Kasprzak5, Lidewij Henneman2;3, Sam Riedijk1
1Erasmus Medical Center, Department of Clinical Genetics, Rotterdam, Netherlands; 2Amsterdam UMC, location Vrije Universiteit Amsterdam, Department of Human Genetics, Amsterdam, Netherlands; 3Amsterdam Reproduction and Development Research Institute, Amsterdam, Netherlands; 4Amsterdam Public Health Research Institute, Amsterdam, Netherlands; 5Rotterdam University of Applied Sciences, Willem de Kooning Academy, Rotterdam, Netherlands
Consortium: De DNA dialogen Consortium
Background/Objectives: The methods, conditions and values for public engagement regarding human germline gene editing (HGGE) are increasingly discussed. The WHO framework for governance report on human genome editing (2021) states that ‘specific strategies are needed to engage traditionally underrepresented groups, such as indigenous peoples, minority ethnic groups or faiths, or specific patient groups’. In the Dutch DNA dialogues project (2022-2026), we have started to operationalize, identify and engage underrepresented groups in dialogues about HGGE, uncovering issues that call for more inclusive strategies.
Methods: We work together with societal partners to operationalize and identify underrepresented groups and to co-create tailored dialogue strategies that employ theatre and the qualitative research method Photovoice. We conducted 14 dialogues, including individuals with genetic conditions, physical disabilities, and those identified as being difficult to reach for scientists, and analysed these using thematic analysis.
Results: In preparing and analysing the dialogues, several issues emerged: (1) Our current informed consent procedures were unfit for underrepresented groups. (2) Islamic participants were hesitant to engage in a dialogue when they were unsure about the views of their religious leaders on this new technology. (3) Experiences during the COVID-19 pandemic severely and negatively impacted participants trust in science and governance of new technologies.
Conclusion: To engage underrepresented groups in dialogue and decision-making about new disruptive technologies, researchers and engagement practitioners need more inclusive informed consent procedures, a religious knowledge base in addition to a scientific knowledge base, and to make distrust in science and governance part of the conversation.
Grants: [NWO/NWA.1389.20.075]
Conflict of Interest: Diewertje Houtman As of the 1st September 2022, the authors are involved in a Netherlands Consortium “Public Realm Entrance of Human Germline Gene Editing” funded by The Netherlands Organization for Scientific Research (NWO NWA.1389.20.075)., Wendy Geuverink As of the 1st September 2022, the authors are involved in a Netherlands Consortium “Public Realm Entrance of Human Germline Gene Editing” funded by The Netherlands Organization for Scientific Research (NWO NWA.1389.20.075)., Joosje Kist As of the 1st September 2022, the authors are involved in a Netherlands Consortium “Public Realm Entrance of Human Germline Gene Editing” funded by The Netherlands Organization for Scientific Research (NWO NWA.1389.20.075)., Isabel Retel Helmrich As of the 1st September 2022, the authors are involved in a Netherlands Consortium “Public Realm Entrance of Human Germline Gene Editing” funded by The Netherlands Organization for Scientific Research (NWO NWA.1389.20.075)., Martina Cornel As of the 1st September 2022, the authors are involved in a Netherlands Consortium “Public Realm Entrance of Human Germline Gene Editing” funded by The Netherlands Organization for Scientific Research (NWO NWA.1389.20.075)., Michelle Kasprzak As of the 1st September 2022, the authors are involved in a Netherlands Consortium “Public Realm Entrance of Human Germline Gene Editing” funded by The Netherlands Organization for Scientific Research (NWO NWA.1389.20.075)., Lidewij Henneman As of the 1st September 2022, the authors are involved in a Netherlands Consortium “Public Realm Entrance of Human Germline Gene Editing” funded by The Netherlands Organization for Scientific Research (NWO NWA.1389.20.075)., Sam Riedijk As of the 1st September 2022, the authors are involved in a Netherlands Consortium “Public Realm Entrance of Human Germline Gene Editing” funded by The Netherlands Organization for Scientific Research (NWO NWA.1389.20.075).
C23.6 Genetic counselling for caregivers of children and adolescents diagnosed with psychiatric disorders - a pilot study
Madalina Radu 1, Andra Ciuca1, Sebastian Pintea1, Ramona Moldovan1;2;3
1Babeș-Bolyai University, Department of Psychology, Cluj-Napoca, Romania; 2University of Manchester, Division of Evolution and Genomic Sciences, Manchester, United Kingdom; 3Manchester Academic Health Science Centre, Manchester Centre for Genomic Medicine, St Mary’s Hospital, Manchester, United Kingdom
Consortium: -
Background/Objectives: The efficacy of psychiatric genetic counselling has been well documented for patients with psychiatric disorders. Family members of adult patients also appear to benefit. To date, there is no data available for parents of children and adolescents diagnosed with psychiatric disorders. This study aimed to investigate the impact of psychiatric genetic counselling on parent’s knowledge, empowerment, well-being and stigma.
Methods: Parents of young people diagnosed with psychiatric disorders in an outpatient clinic were invited to take part. Participants were assessed pre and post intervention using standardised questionnaires. A registered genetic counseller offered the parents one genetic counselling session.
Results: Of the 43 paticipants enrolled (i.e. 41 mothers and 2 fathers, mean age 40.2 ± 5.9 years old), 32 completed the post test measures. Yound people’s (mean age 9 ± 4 years old) diagnosis included ASD, ADHD, affective disorders and anxiety disorders. Statistical analyses shows genetic counselling has a significant impact on parents‘ knowledge [t(31) = -7.132, p = .000] and empowerment [t(31) = -2.723, p = .011]. Parents of children with anxiety disorders significantly improved their knowledge (r = .400, p < .05) and parents of children with ASD significantly decreased stigma (r = .379, p < .05). The sooner after the moment of diagnosis parents attended the session, the greater the impact on their knowledge (r = .388, p < .05) and well-being (r = .381, p < 0.05).
Conclusion: Psychiatric genetic counselling improves parents’ knowledge, empowerment, well-being and stigma. This can help families by providing them with the support they need to better understand, adapt, and manage the condition in their family.
Grants: -
Conflict of Interest: None declared
C24 Immunology and Hematopoietic System
C24.1 Diagnostic yield of genome-wide genetic testing and demographic distribution in 741 index patients with inborn errors of immunity
Darja Gauck 1, Marc Sturm1, Ines Brecht2, Ute Grasshoff1, Stefanie Beck-Wödl1, Tatjana Welzel3, Ursula Holzer2, Martin Kehrer1, Olaf Riess1;4, Julia Skokowa5, Jasmin B. Kümmerle-Deschner6, Tobias Haack1
1University of Tübingen, Institute of Medical Genetics and Applied Genomics, Tübingen, Germany; 2University Children’s Hospital, University of Tübingen, Department of Pediatric Hematology and Oncology, Tübingen, Germany; 3University Children’s Hospital Basel, University of Basel, Pediatric-Rheumatology, Basel, Switzerland; 4University of Tübingen, Center for Rare diseases, Tübingen, Germany; 5University Hospital Tübingen, Department of Oncology, Hematology, Clinical Immunology, and Rheumatology, Tübingen, Germany; 6University Hospital Tübingen, Department of Pediatrics, Division of Pediatric Rheumatology and autoinflammation reference center Tübingen (arcT), Tübingen, Germany
Background/Objectives: Human Inborn Errors of Immunity (IEI) encompass a heterogeneous group of genetic disorders associated with versatile clinical features. In this report we provide an overview of the diagnostic yield and demographics in a prospective multi-center cohort of 741 index patients with a suspected clinical diagnosis of IEI.
Methods: Sequencing libraries were generated from genomic DNA for genome (TruSeq DNA PCR-free kit) or exome (TWIST Custom Exome IMGAG V2/SureSelectXT Human All Exon kits V6/V7) sequencing and sequenced on a NovaSeq6000 System (Illumina). Data analysis was conducted using an in-house bioinformatics pipeline (megSAP).
Results: A firm genetic diagnosis was established in 10.8% (n = 68/627) of pediatric cases, with a highest rate of solved cases (31%, n = 9/29) in a group of patients with syndromic features and/or severe congenital neutropenia <1 year of age. A definite molecular diagnosis was made in 11.7% (n = 17/145) of adult patients, mostly in the category of autoinflammatory disorders (n = 5). For 15.5% (n = 117) of the entire cohort with variants in known IEI genes and for 1.8% (n = 14) with suspected candidate genes, genetic outcome was uncertain. In 71.6% (n = 556), the underlying genetic cause of IEI remained unclear.
Conclusion: Our observations illustrate the broad genetic and clinical heterogeneity of IEI and provides insights into the dispersion of solved cases among various age groups and disease categories. The increased usage of genome sequencing has the potential to further narrow the diagnostic gap by expanding the analysis to structural variations and non-coding regions.
Conflict of Interest: None declared
C24.2 p53 Unchained – Constitutional Upregulation of p53 Caused by Germline Variants in MDM4 Leads to Bone Marrow Failure and Insufficient Telomere Maintenance
Robert Meyer 1;2, Senthil Velan Bhoopalan3, Richa Sharma3, Lei Han3, Matthias Begemann1;2, Marcus Jakob4, Daniela Dey1;2, Tarek Hanafee-Alali4, Lara Heller5, Heinz Gabriel5, Kim Kricheldorf2;6, Margherita Vieri2;6, Susanne Isfort2;6;7, Martin Kirschner2;6, Jens Panse2;6, Udo Kontny8, Selim Corbacioglu4, Shondra M. Pruett-Miller9, Claudia Khurana10, Miriam Erlacher11;12, Lise Larcher13;14;15;16, Jean Soulier13;14;15;16, Miriam Elbracht1;2, Ingo Kurth1;2, Tim H. Brümmendorf2;6, Fabian Beier2;6, Marcin Wlodarski3
1Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH Aachen University, Aachen, Germany; 2Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Germany; 3Department of Hematology, St. Jude Children´s Research Hospital, Memphis, United States; 4Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital of Regensburg, Regensburg, Germany; 5Zentrum für Humangenetik Tübingen, Tübingen, Germany; 6Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical Faculty, RWTH Aachen University, Aachen, Germany; 7Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Medical School Hannover, Hannover, Germany; 8Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Medical Faculty, RWTH Aachen University, Aachen, Germany; 9Department of Cell and Molecular Biology, St. Jude Children´s Research Hospital, Memphis, United States; 10Klinik für Kinder- und Jugendmedizin, Evangelisches Klinikum Bethel, Bielefeld, Germany; 11Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center, University of Freiburg, Freiburg, Germany; 12German Cancer Consortium (DKTK), Freiburg, Germany; 13Institut de Recherche Saint-Louis (IRSL), Université Paris Cité, Paris, France; 14INSERM U944/CNRS UMR7212, Paris, France; 15Saint-Louis Hospital, Hematology Laboratory, APHP, Paris, France; 16Centre de Référence Maladies Rares “Aplasie Médullaire”, Saint-Louis and Robert Debré Hospitals, Paris, France
Background/Objectives: Loss of function of p53 – often referred to as the guardian of the genome – has been studied extensively, both in germline and cancer cells. Data on the effects of an overactive p53, however, are rare. Based on clinical and functional data, we describe the effects of constitutional dysregulation of p53 caused by germline variants in MDM4, an important regulator of p53. Until now, a single family carrying a pathogenic missense variant in MDM4 had been described.
Methods: Genome/Exome/RNA sequencing, Flow-FISH telomere length measurement, western blot, CRISPR/Cas9-guided knock-in of MDM4 variants in iPSC lines, deletion of MDM4 in CD34+ cells and mouse xenotransplantation studies.
Results: Cas9-mediated mutation of MDM4 in iPSC and CD34+ cells resulted in increased p53 activity and fewer erythroid and myeloid colonies, which was rescued by TP53 knock-out. Xenotransplant studies showed a p53-dependent bone marrow repopulation defect in MDM4-disrupted CD34+ cells. Six individual patients carrying germline variants in MDM4 presented with a varying degree of bone marrow failure (BMF), a tendency to shortened telomeres and further single features of telomere biology disorders (TBD).
Conclusion: Here, we provide strong evidence from functional and clinical data of six unrelated patients that germline variants in MDM4 cause a disorder of p53 dysregulation resulting in a variable phenotype including BMF and features of TBD. While clinical trials for MDM4 inhibitors to reactivate p53 in tumor cells are ongoing, our work allows an outlook on potential side effects of these drugs and sheds light on an unfamiliar dark side of p53.
Grants:
Conflict of Interest: None declared
C24.3 Combined long- and short-read RNA sequencing of pathogen stimulated primary immune cells reveals the expression of uncharacterized genes and transcripts
Emil Vorsteveld 1, Renee Salz2, Caspar van der Made3, Simone Kersten1, Charlotte Kaffa2, Merel Stemerdink1, Tabea Riepe2, Tsung-han Hsieh4, Musa Mhlanga4, Mihai Gheorghe Netea3, Pieter-Jan Volders5, Peter-Bram t Hoen2, Alexander Hoischen1
1Radboud University Medical Center, Department of Human Genetics, Nijmegen, Netherlands; 2Radboud University Medical Center, Department of Medical BioSciences, Nijmegen, Netherlands; 3Radboud University Medical Center, Department of Internal Medicine, Nijmegen, Netherlands; 4Radboud University Nijmegen, Department of Cell Biology, Nijmegen, Netherlands; 5University of Ghent, Department of Biomolecular Medicine, Gent, Belgium
Background: Immune responses are shaped by the nature of infections and by inter-individual variability, contributing to differential susceptibility to infections and to various diseases with an inflammatory component. Dynamic transcript and protein expression in a range of cells responsible for the innate immune response is important to shape the first line of defense against a wide variety of pathogens.
Methods: Here we perturbed immune-cells with in vitro pathogen exposure. We stimulated PBMCs from 5 healthy donors for 4h or 24h with LPS, S. aureus, Poly(I:C) or C. albicans with RPMI medium as control, resulting in a total of 52 studied samples. We performed short read sequencing using Lexogen QuantSeq in all samples, as well long read sequencing using PacBio IsoSeq in a subset of samples.
Results: Short-read sequencing reveals common and distinct genes and pathways expressed during immune responses to different pathogens. Beside well-established genes, we highlight uncharacterized genes KIAA0040 and FAM49A, which show up to 2.3-fold increased expression after pathogen exposure and are co-expressed with modules of established immune genes. Long-read sequencing revealed 47.7% novelty in the transcriptomes. We found widespread isoform switching induced upon pathogen stimulation. We highlight novel transcripts of NFKB1 and CASP1 that may indicate novel immunological mechanisms.
Conclusions: Transcriptome profiling of pathogen-stimulated immune cells using paired short- and long-read approaches highlights candidate immune genes and identifies novel transcripts, revealing a more complex transcriptome landscape following pathogen exposure than previously appreciated.
Grants: MGN: ERC Advanced (#833247), Spinoza Grant (NWO). AH: Solve-RD (EU H2020 #779257).
Conflict of Interest: None declared
C24.4 A collective influence of genetic background on influenza vaccinees responses
Nhan Nguyen 1;2, Saumya Dileep Kumar1;2, Martijn Zoodsma1;2, Janyn Heisig3, Stephanie Trittel3, Peggy Riese3, Cheng-Jian Xu1;2;4, Frank Pessler1;5, Luis Graca6, Carlos Alberto Guzman3, Yang Li1;2;4;7
1Helmholtz Centre for Infection Research (HZI), Centre for Individualised Infection Medicine (CiiM), Hannover, Germany; 2TWINCORE, Center for Experimental and Clinical Infection Research, Hannover, Germany; 3Helmholtz Centre for Infection Research (HZI), Department Vaccinology and Applied Microbiology, Braunschweig, Germany; 4Radboud University Medical Center, Department of Internal Medicine and Radboud Center for Infectious Diseases, Nijmegen, Netherlands; 5TWINCORE, Center for Experimental and Clinical Infection Research, Research Group Biomarkers for Infectious Diseases, Hannover, Germany; 6Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal; 7Hannover Medical School, Cluster of Excellence Resolving Infection Susceptibility (RESIST; EXC 2155), Hannover, Germany
Background/Objectives: While influenza infection is a significant public health threat leading to about 290,000-650,000 deaths annually worldwide, the effectiveness of influenza vaccines varies between 40 to 80% among individuals. In this study, we developed a reclassification concept to differentiate protected and unprotected vaccinees and identified a plausible genetic determination for unprotected vaccinees.
Methods: From blood samples of 286 healthy donors spanning 4 influenza seasons (45% female, age range: 18-81), we generated a multimodal dataset including genotype, whole blood transcriptome, plasma proteome, metabolome, and serological parameters. Based on the serological correlate for protection, we used pre-vaccination antibody titer and antibody titer foldchange between pre- and post-vaccination to establish a new approach to classify protected and unprotected vaccinees. Thereafter, we analyzed the differences in omics profiles between protected and non-protected vaccinees.
Results: We identified proteins and metabolites that could serve as predictive markers to identify unprotected vaccinees before their vaccine administration. In QTL analysis with post-vaccination antibody titers, we observed 3 loci (p > 10e-7) with nearby genes such as TCERG1L, SHCBP1, ORC6, GTP2, DNAJA2, ITFG1 that are implicated in multiple immune responses. Furthermore, we predicted multi-omics signatures at gene expression, protein, and metabolite levels directly from genotypes using OmicsPred models. Then, we identified omics signatures, such as PTX3 and CD5, which were genetically determined for non-protected vaccinees.
Conclusion: This study demonstrates a new approach to classify protected and unprotected individuals after influenza vaccination, potential biomarkers for predicting unprotected vaccinees, and a collective influence of genetic background on non-protected vaccinees.
Grants: ERC Starting Grant (948207)
Conflict of Interest: None declared
C24.5 A comprehensive genetic map of cytokine responses in Lyme borreliosis
Javier Botey Bataller 1;2;3, Hedwig D. Vrijmoeth1;4, Jeanine Ursinus4;5, Bart Jan Kullberg1, Cees C. van den Wijngaard4, Hadewych ter Hofstede1, Ahmed Alaswad2;3, manoj Kumar Gupta2;3;6, Lennart Rösner6, Jochen Huehn6;7, Thomas Werfel6, Thomas F. Schulz6, Cheng-Jian Xu1;2;3, Mihai Gheorghe Netea1;8, Joppe W.R. Hovius5, Leo A.B. Joosten1;9, Yang Li1;2;3;6
1Radboud University Medical Center, Nijmegen, Netherlands; 2Centre for Individualised Infection Medicine (CiiM), Hannover, Germany; 3Twincore, Zentrum für Experimentelle und Klinische Infektionsforschung GmbH, Hannover, Germany; 4National Institute for Public Health and Environment, Bilthoven, Netherlands; 5Amsterdam UMC, locatie AMC, Amsterdam, Netherlands; 6Hannover Medical School, Hannover, Germany; 7Helmholtz Centre for Infection Research, Braunschweig, Germany; 8LIMES-Institut der Universität Bonn, Bonn, Germany; 9Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
Background: The incidence of Lyme borreliosis is rapidly increasing, leading to more patients experiencing persistent symptoms after antibiotic treatment. The innate immune system and related cytokines play a crucial role in the host response to infection and may be involved in the development of persistent symptoms.
Methods: Previously, we identified genetic loci that contribute to inter-individual variation in cytokine responses (cQTLs) and affect the susceptibility to infection. Here, we used a functional genomics approach to characterize the cytokine profile of 1,060 physician-confirmed Lyme borreliosis patients. We measured cytokine production upon ex vivo pathogen stimulation in samples taken before the start of antibiotic treatment and six weeks thereafter, resulting in a total of 108 cytokine-stimulation conditions.
Results: We confirmed the inhibiting effect of IL-10 on antibody production against B. burgdorferi s.l. and observed enhanced IL-1Ra responses in patients with disseminated disease. Genome-wide mapping the cytokine production allowed us to identify 34 cQTLs, 31 of which were not found in previous studies. We pinpointed the causal variant at the TLR1-6-10 locus and validated the specific regulation of this locus on IL-1Ra responses to Pam3Cys compared to B. burgdorferi s.l. at the transcriptome level using an independent cohort. Our findings show that cQTLs contribute to the susceptibility to Lyme borreliosis, and are relevant to other immune-mediated diseases such as allergy, IBD and multiple sclerosis.
Conclusions: Collectively, our results improve our understanding of the role of cytokine responses in Lyme borreliosis and provide a genetic map of general immune function as an expanded resource, made available at https://lab-li.ciim-hannover.de/apps/lyme_cqtl/.
Conflict of Interest: None declared
C24.6 Effect of parental autoimmune diseases on type 1 diabetes in offspring can be partially explained by HLA and non-HLA polymorphisms: a nationwide registry and biobank study in 7.2M Finns
Feiyi Wang 1, Aoxing Liu1;2;3;4, FinnGen Project1, Tiinamaija Tuomi1;5;6;7, Andrea Ganna1;2;3;4
1Institute for Molecular Medicine Finland, Helsinki, Finland; 2Broad Institute of Harvard and MIT, Program in Medical and Population Genetics, Cambridge, United States; 3Broad Institute of Harvard and MIT, Stanley Center for Psychiatric Research, Cambridge, United States; 4Massachusetts General Hospital, Analytic and Translational Genetics Unit, Boston, United States; 5HUS, Abdominal Center, Endocrinology, Helsinki, Finland; 6Folkhalsan Research Center, Helsinki, Finland; 7Lund University Diabetes Center, Malmo, Sweden
Consortium: FinnGen
Type 1 diabetes (T1D) and some autoimmune diseases (ADs) are observed to co-occur in families, yet it remains largely unknown whether offspring with ADs-affected parents would have elevated risks of T1D and to what extent such intergenerational transmission is attributed to shared genetic factors, especially when partitioning into HLA and non-HLA variations. Leveraging nationwide registers of 7.2M Finns - the population with the highest T1D prevalence globally - we defined 50 parental AIDs in 58,284 family trios and observed 10 parental ADs exerting an increased risk of T1D in offspring. The identified epidemiological associations were further evidenced by comprehensive genetic analyses performed in 470K genotyped Finns of the FinnGen study (N of trios = 10,787). Collectively, our findings suggested that intergenerational transmission of ADs to offspring T1D can be partially explained by HLA and non-HLA polymorphisms, but in a disease-dependent manner. For example, coeliac disease and psoriasis are mainly impacted by HLA while autoimmune hypothyroidism and rheumatoid arthritis also had non-HLA contributors in addition to HLA. We proposed a novel parental polygenic score, integrating variations of both HLA and non-HLA, as a useful predictor to evaluate the risk of developing T1D in offspring and observed parental ADs could impact offspring T1D incidence even among those whose parents were T1D-unaffected. While presenting the power of combining high-dimensional nationwide registers with rich genetic information in studying disease etiology, the analytical framework developed here can also be used to reveal detailed mechanical insights into the transmission of any diseases or traits across generations.
Conflict of Interest: Feiyi Wang: None declared, Aoxing Liu: None declared, FinnGen Project: None declared, Tiinamaija Tuomi: None declared, Andrea Ganna Founder of Real World Genetics
C25 Liver, Kidney and Gonads
C25.1 Redefining the relevance of genetic variants in kidney stone disease through combined genetic and biochemical analyses has prognostic value
Johannes Münch1, Joana Figueiro da Silva2, Elena Cabello2, Michael Papik2, Ivan Ivanovski2, Beatrice Oneda2, Daniel Fuster3, Harald Seeger4, Thomas Ernandez5, Florian Buchkremer6, Grégoire Wuerzner7, Nasser Dhayat8, Alexander Ritter9, Catherine Stoermann5, Stephan Segerer6, Beat Roth7, Anita Rauch2, Manuel Ferraro10, Olivier Bonny11, Carsten Wagner1, Ruxandra Bachmann-Gagescu 2
1University of Zurich, Physiology, ZURICH, Switzerland; 2University of Zurich, Medical Genetics, Schlieren / Zurich, Switzerland; 3University Hospital Bern; 4Kantonsspital Baden; 5University hospital Geneva; 6Kantonsspital Aarau; 7University Hospital Lausanne; 8B. Braun Medical Care AG, Hochfelden; 9Kantonsspital St Gallen; 10Fondazione Policlinico Universitario A. Gemelli IRCCS; 11Hôpital fribourgeois, Departement de médecine
Kidney stone disease (KSD) affects 10% of the population and is predominantly caused by complex interplay of lifestyle and genetic factors. Rare genetic variants can also cause monogenic KSD, which accounts for up to 30% in pediatric cohorts, while its prevalence in unselected adult cohorts is less clear. Challenges with variant classification and variable inheritance modes, with both mono- and bi-allelic variants in the same genes suggested to cause monogenic KSD, complicate the interpretation of their clinical relevance. To clarify this, we studied 736 kidney stone formers (KSFs) and 201 controls from the Swiss kidney stone cohort, a multicenter longitudinal observational study, with WES and detailed biochemical profiling. This combined approach and comparison with controls confirmed the role of several recessive and dominant genes involved mostly in phosphate or cystine handling in monogenic KSD. Heterozygous SLC7A9 variants increased the risk for KSD but required additional factors for disease occurrence. Notably, a common variant in SLC7A9, previously classified as LP/P, has no biochemical consequence and is not enriched in KSFs. Similarly, we show that heterozygous SLC34A1 and SLC9A3R1 LP/P variants do not perturb tubular phosphate reabsorption and are unlikely to cause monogenic KSD. Considering all factors, we find a genetic cause for KSD in 7.2% of KSFs. Importantly, we observed a steeper decline in kidney function in KSFs harboring LP/P variants compared to those without. Taken together, combined genetic and biochemical analyses clarify the clinical relevance of variants in KSD genes and have prognostic value. Funding: NCCR Kidney.CH
Conflict of Interest: None declared
C25.2 INSL3 variants and Insl3 deficiency in human and murine CAKUT
Anne Christians1, Esra Kesdiren1, Helge Martens 1, Lina Werfel1;2, Ulrike Beyer1, Robert Geffers3, Martina Muehlenhoff4, Jan Hinrich Bräsen5, Ibrahim M. Adham6, Anna-Carina Weiss4, Dieter Haffner2, Ruthild G. Weber1
1Hannover Medical School, Department of Human Genetics, Hannover, Germany; 2Hannover Medical School, Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover, Germany; 3Helmholtz Centre for Infection Research, Genome Analytics Research Group, Braunschweig, Germany; 4Hannover Medical School, Institute of Clinical Biochemistry, Hannover, Germany; 5Hannover Medical School, Nephropathology, Department of Pathology, Hannover, Germany; 6University Medical Centre, Institute of Human Genetics, Göttingen, Germany
Background/Objectives: Variants in the insulin-like 3 (INSL3) gene encoding a ligand of relaxin family peptide receptor 2 (RXFP2) and Insl3 deficiency cause cryptorchidism. Here, a cryptorchidism-associated INSL3 missense variant co-segregated with the CAKUT phenotype in females of an index family, instigating the investigation of the role of INSL3 in human and murine CAKUT.
Methods: Whole-exome or targeted INSL3 sequencing was done in 312 CAKUT families. Variant carriers were subjected to reverse phenotyping. INSL3 expression was determined in human fetal and adult tissues and the developing murine kidney by qRT-PCR. A tubulo-morphogenesis assay was done on CRISPR/Cas9-derived Insl3-deficient mIMCD3 cells. Insl3 knock-out mice were characterized morphologically and histologically with respect to urogenital anomalies.
Results: Rare INSL3 variants predicted to be deleterious were identified in 6 of 312 (1.9%) CAKUT families, significantly more frequently than in controls (0.8%, p = 0.04). The four different INSL3 variants detected were heterozygous, and almost exclusively maternally inherited. Cryptorchidism was significantly more frequent in male CAKUT patients with versus without INSL3 variants (67% vs. 9%, p = 0.02). INSL3 was expressed in the fetal and adult human kidney. Insl3 and Rxfp2 transcripts were present in the developing murine kidney. In Insl3+/- and Insl3-/- mIMCD3 cells, fewer tubular structures were observed indicating impaired tubulogenesis processes. The CAKUT spectrum in Insl3+/- and Insl3-/- mice included kidney hypo(dys)plasia, fusion, segmental nephron losses, hydronephrosis, duplex ureter, a narrowed ureteric lumen.
Conclusion: Heterozygous INSL3 variants associate with human CAKUT, and heterozygous and homozygous Insl3 knock-out can cause CAKUT in mice.
Grants: Deutsche Forschungsgemeinschaft (KO5614/2-1, MA9606/1-1), Else Kröner-Fresenius-Stiftung (2018_Kolleg.12, TITUS-Clinician Scientist Program)
Conflict of Interest: None declared
C25.3 Targeted RNA sequencing in monogenic renal diseases
Clément SAUVESTRE 1, Marc Bras2, Zaina Ait Arkoub1, Vincent Moriniere1, Christelle Arrondel3, Nicolas Cagnard2, Patrick Nischke2, Manon Mautret Godefroy1, Laurence Heidet3;4, Corinne Antignac3, Guillaume Dorval1;3
1APHP, Université Paris-Cité, Service de Médecine Génomique des Maladies Rares, Paris, France; 2Institut Imagine, Université de Paris, Plateforme Bio-informatique, Inserm UMR 1163, Paris, France; 3Institut Imagine, Université Paris Cité, Laboratoire des Maladies Rénales Héréditaires, Inserm UMR 1163, Paris, France; 4Hôpital Necker-Enfants Malades, Assistance publique, Hôpitaux de Paris (AP-HP), Service de néphrologie pédiatrique Centre de Référence MARHEA, Paris, France
Background/Objectives: RNA sequencing (RNAseq) is a crucial tool for functional assessment of certain variants affecting splicing in the post-genomic era. However, it can be challenging to use when studying genes expressed in specific, inaccessible tissues, such as kidney tissue. Additionally, transcript analysis requires significant depth of coverage, which can be difficult to achieve.
Methods: This study applies Targeted RNAseq technology to capture a panel of 228 genes involved in monogenic kidney diseases in various tissues (kidney, blood, fibroblasts, and urine) from patients with a previously identified intronic variant affecting splicing. The expression profiles of the captured genes in each tissue were compared, and the impact of each identified variant on RNA splicing was interpreted.
Results: Several samples were studied for each tissue. On average, 11,176 reads were aligned with the exon-exon junctions identified, providing sufficient reading depth to interpret the splicing events (approximately 354 reads per junction). The analysis of different samples in clusters demonstrated the homogeneity and repeatability of analysis in tissues. For instance, the analysis of RNA from the complete urine of three patients allowed for the examination of splicing of 222 out of 237 captured genes in all cases. Specific studies of the transcripts of interest were conducted and confirming all the variants identified.
Conclusion: This work demonstrates that studying RNA from various tissues, enables functional interpretation of variants affecting splicing. In the field of nephrogenesis, RNA from urine can be used to interpret events in almost all of the genes studied.
Grants: No funding
Conflict of Interest: None declared
C25.4 Metabolome and exome (WES) data integration to stratify Non-Alcoholic Fatty Liver disease (NAFLD) patients
Rebecca Filomena 1, Carla Debernardi1, Cecilia Di Primio1, ANGELO SAVOCA1, Gian Paolo Caviglia2, Chiara Catalano1, clara viberti1, Elisabetta Casalone1, Chiara Rosso2, Angelo Armandi2, Alessandra Allione1, Marcello Manfredi3, Miriam Rosselli1, elton jalis herman1, Alessia Russo1, Elisabetta Bugianesi2, Giuseppe Matullo1;4
1University of Turin, Unit of Genomic Variability and Complex diseases, Department of Medical Sciences, Torino, Italy; 2University of Turin, Division of Gastroenterology and Hepatology, Department of Medical Sciences, Torino, Italy; 3University of Piemonte Orientale, Department of Translational Medicine, Novara, Italy; 4AOU Città della Salute e della Scienza, Medical Genetics Unit, Torino, Italy
Background/Objectives: Non-alcoholic fatty liver disease (NAFLD) is defined as the accumulation of excessive fat in the liver in the absence of excessive alcohol consumption or other liver diseases. It is the most common cause of liver disease worldwide and includes a wide range of liver damage such as simple steatosis (non-alcoholic fatty liver; NAFL) and non-alcoholic steatohepatitis (NASH). The aim of this study is to find a potential correlation between the genetic and metabolic component of the disease than can better differentiate the phenotypes based on the severity.
Methods: We performed untargeted metabolomics analysis and Whole Exome Sequencing (WES) on 154 patients with biopsy-proven NAFLD status, enrolled at the Liver Unit of the Department of Medical Sciences, University of Turin.
Results: From the untargeted metabolomics analysis, 8 metabolites out of 380 resulted to be significantly differentially expressed (p-value < 0.05, 0.769<Fold Change<1.3). Pathway analysis showed that all of them are involved in the Aminoacyl-tRNA biosynthesis (FDR = 6.82×10−5).
As a preliminary analysis, aimed to identify metabolite-QTL, we selected the exome variants in the 44 genes related to this pathway. We observed 230 non-synonymous variants, with an average of 14 variants per sample in the investigated genes. Moreover, a higher metabolite expression correlated with an increased average number of variants.
Conclusion: The integration of metabolomics and WES could help to identify genes and metabolites involved in NAFLD progression, improve patients’ stratification and potentially develop new therapies.
Grants: Ministero dell’Istruzione, dell’Università e della Ricerca (MIUR) Project “Dipartimenti di Eccellenza 2018–2022. Project n° D15D18000410001”
Conflict of Interest: None declared
C25.5 The molecular study of 407 Chinese children with 46,XY Disorders of Sex Development (DSD) by Next-generation sequencing (virtual)
Xiaojun Tang 1, Xiaoping Lan1, Xiaozhen Song1, wuhen xu1, Yongchen Yang1, Wenhao Weng1, Shengnan Wu1
1Shanghai Children’s Hospital Luding Rd Yuanqu-Blood Bank, Shanghai Children’s Hospital, Shanghai, China
Objectives 46,XY Disorders of Sex Development (DSD) is the most common and complicated subtype, accounting for approximately 50% of all DSD cases. Molecular diagnoses provide guidance for suitable treatments, prognosis evaluation, and genetic counseling. This study aims to evaluate the diagnostic yield and clinical value efficacy of next-generation sequencing (NGS) in children with 46,XY DSD.
Methods Single/trio whole exome sequencing or targeted panel sequencing were performed on 407 Chinese children with 46,XY DSD. Single-nucleotide variants (SNVs), insertions/deletions and copy number variants (CNVs) were concurrently analyzed and interpreted according to American College of Medical Genetics guideline. The variants detected by NGS were confirmed by Sanger sequencing, QPCR or MLPA accordingly. The clinical presentation and family history were collected.
Results The overall diagnostic yield of this cohort stood at 39.56%, comprising 155 with SNVs, 5 with CNVs and one with both. SNVs were most frequently found in SRD5A2, AR, NR5A1, FGFR1, CYP17A1, HSD17B3 and ANOS1. Notably, 56 variants have not been previously reported, expanding the mutation spectra of DSD related genes. It’s crucial to highlight that intragenic exons deletion/duplication were identified in 6 patients. Interestingly, 35 variants were categorized as variants of uncertain significance due to lack of evidence currently, for which further functional experiments and additional patients are needed to clarify the pathogenicity of these variants.
Conclusion The early diagnosis can provide an accurate molecular etiology, which enables to detect potential phenotypes, guide appropriate therapies and clinical follow-up, which benefiting the clinical management of patients.
Grants Shanghai Children’s Hospital Funding (2021YGZQ07)
Conflict of Interest: None declared
C25.6 Added value of clinical exomes in the management of male infertility - study on ESTonian ANDrology (ESTAND) cohort.
Kristiina Lillepea1, Anna-Grete Juchnewitsch1, Laura Kasak1, Anu Valkna1, Avirup Dutta1, Kristjan Pomm2, Erik Tamp3, Stanislav Tjagur2, Mailis Sütt2, Olev Poolamets2, Margus Punab2, Maris Laan 1
1University of Tartu, Institute of Biomedicine and Translational Medicine, Tartu, Estonia; 2Tartu University Hospital, Andrology Clinic, Tartu, Estonia; 3East-Tallinn Central Hospital, Center of Pathology, Tallinn, Estonia
Background/Objectives: Infertility affects ~10% men and a large fraction of idiopathic cases are thought to have an unknown genetic cause. Although hundreds of genes have been reported as plausible candidates linked to spermatogenic failure (SPGF), current management of infertile men does not include exome sequencing (ES). The main objective was to analyze a large nationwide cohort of SPGF patients for likely pathogenic and pathogenic variants (LP/P) in infertility candidate genes to evaluate the diagnostic yield, genotype-phenotype links and contribution of ES to clinical management, counselling of patients for reproductive and overall health.
Methods: ES was performed to 521 SPGF patients (total sperm counts 0 - 10 million per ejaculate) recruited to ESTAND cohort and the analyzed gene panel comprised of >600 infertility linked loci. Variant pathogenicity was evaluated using the ACMG guidelines and variants were confirmed by Sanger sequencing. We also piloted providing feedback to patients about the finding(s) and their broader impact.
Results: More than 10% of patients received molecular diagnosis linked to diverse spectrum of molecular etiologies, including isolated and syndromic forms of SPGF. The findings were distributed to all inheritance modes and some patients carried >1 LP/P variants. Notably, over 20% of identified variants represented recurrent findings in male infertility either reported in other cohorts and/or carried by two ESTAND patients. Link to SPGF was validated for several recently proposed candidate genes (e.g. ASZ1, RBM5, GLUD2).
Conclusion: Introducing ES to andrology practice will support improved diagnostics and multidisciplinary management of SPGF patients.
Grants: PRG1021 (Estonian Research Council)
Conflict of Interest: None declared
C27 Prenatal diagnosis
C27.1 Enhancing prenatal prognostic accuracy and understanding intrafamilial variability in osteogenesis imperfecta: insights from a retrospective study of 269 cases.
Cristina Peduto 1, sophie monnot1;2, Caroline Michot1;2, Elise Schaefer3, Melanie FRADIN4, Carine Abel5, Genevieve Baujat1;6, Valérie Cormier-Daire1;2
1Centre de Référence Maladies Osseuses Constitutionnelles, Fédération de médecine génomique des maladies rares, APHP, Hôpital Necker-Enfants Malades,, Paris, France; 2Institut Imagine, Université Paris Cité, INSERM UMR1163, Paris, France; 3Service de Génétique Médicale et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, Hôpitaux Universitaires de Strasbourg, Institut de Génétique Médicale d’Alsace, Strasbourg, France; 4Service de Génétique Médicale, CHU de Rennes, Rennes, France; 5Service de Génétique et Centre de Diagnostic Anténatal, CHU de Lyon HCL-GH Nord-Hôpital de La Croix Rousse, Lyon, France; 6Institut IMAGINE and Hôpital Universitaire Necker-Enfants Malades, Département de Génétique, Paris, France
Background/Objectives: Osteogenesis Imperfecta (OI) is due mainly by mutations in COL1A1 and COL1A2 genes. OI exhibits diverse phenotypes, from severe prenatal forms to mild postnatal ones, posing challenges in prognosis, particularly when diagnosed during pregnancy.
Methods: This retrospective study was based on a cohort of 269 cases from the French Constitutional Bone Diseases Reference Centre OI database (January2017-March2023), with the aim of improve prenatal prognostic accuracy by the correlation of prenatal isolated femoral bowing with disease progression, likewise intrafamilial variability.
Results: Out of 119 individuals (COL1A1 = 55; COL1A2 = 64) with prenatal OI diagnosis only, 25 (COL1A1 = 7;COL1A2 = 15) has isolated femoral bowing. COL1A1 patients (1 missense,5 slicing,1 frameshift) mostly display moderate severity (6/7,85%), however none showed favorable progression. COL1A2 patients (7 missense,2 splicing,2 indels) exhibit a range of phenotypes from mild to severe. Notably, two COL1A2 gene variants (c.2026-2A > G;p.? and c.1109G>C;p.Gly370Ala) showed regressive outcomes, highlighting outcome variability with improved femur bowing and fracture numbers. Otherwise, to assess intrafamilial variability 9 patients inherited the condition from asymptomatic parents among 107 cases: 4 COL1A1 (2 missense,1 frameshift,1 splicing) and 5 COL1A2 (4 missense,1 splicing). We roll out a mosaicism by testing on two separate blood samples, grandparent segregation and analysis on buccal cells and urine. We found 5 parents with de novo variants (COL1A1 = 3, COL1A2 = 2), 3 inherited from a grand-parent (COL1A2 = 3) and 1 mosaic.
Conclusion: These findings suggest that modifier genes may influence OI expression, enhancing our understanding of OI phenotype variability and providing clearer guidance for family counseling concerning prognosis.
Grants: No
Conflict of Interest: None declared
C27.2 First elements of comparison between invasive and non-invasive exomes strategies in prenatal diagnosis when ultrasound reveals developmental anomalies: towards implementation in routine strategy?
Frédéric Tran Mau-Them 1;2, Jean Muller3;4;5, Julian Delanne6, Virginie Haushalter3;4, Anne-Sophie Denommé-Pichon1;2, Audrey Schalk3;4, Aurore Garde6, sophie scheidecker3;4, Hana Safraou1;2, Cecile Lang3;4, Caroline Racine6, Ange-Line Bruel1;2, Antonio Vitobello1;2, Sophie Nambot6, Nicolas Bourgon7, Valentin Bourgeois1;2, Victor Couturier1;2, Charlotte Poe1;2, Martin Chevarin1;2, Thierry Rousseau7, Paul Sagot7, Emmanuel Simon7, Sophie Naudion8, Estelle Colin9, Agnes Guichet10, Yosra Halleb11, Alice Goldenberg12, Anne-Marie Guerrot12, Clara Houdayer12, Juliette Coursimault12, Audrey Putoux13, Mathilde Nizon14;15, Solène Conrad14;15, Benjamin Durand16, elise brischoux-boucher17, Laetitia Lambert18, Marie-Laure Humbert19, Melodie Soto2, Thi-Thu Creusvaux2, Céline Bernard2, Christine Binquet19, Yannis Duffourd1;2, Benedicte GERARD3;4, Christophe Philippe1;2, Nadège Calmels3;4, Laurence Faivre2;6, Caroline Schluth-Bolard3;4, Christel Thauvin-Robinet1;2;6
1CHU Dijon, Unité Fonctionnelle Innovation en Diagnostic Génomique des maladies rares, Dijon, France; 2INSERM, UMR1231 GAD, Dijon, France; 3Laboratoire de Génétique Médicale, INSERM, UMR_S 1112, CRBS, Université de Strasbourg, Strasbourg, France; 4Nouvel Hôpital Civil, Laboratoires de Diagnostic Génétique, Strasbourg, France; 5Hôpitaux Universitaires de Strasbourg, Unité Fonctionnelle de Bioinformatique Médicale appliquée au diagnostic (UF7363),, Strasbourg, France; 6CHU Dijon, Centre de Référence Maladies Rares “Anomalies du développement et syndromes malformatifs”, Dijon, France; 7CHU Dijon, Service de Gynécologie Obstétrique, Médecine Fœtale et Stérilité Conjugale, Dijon, France; 8CHU Bordeaux, Service de Génétique Médicale, Bordeaux, France; 9Université Angers, Biochemistry and Genetics Department, Angers; 10CHU Angers, Service de génétique, Plateau de Biologie Hospitaliere, Angers, France; 11CHU Angers, Centre Robert Debré, Angers, France; 12CHU Rouen, Service de génétique - Unité de génétique clinique, Rouen, France; 13GH Est-Hôpital Femme Mère Enfant, Service de génétique, Lyon, France; 14CHU Nantes, Service de Génétique Médicale, Nantes, France; 15INSERM, Institut du thorax, Nantes, France; 16Hopital Universitaire de Strasbourg, Institute Genetic Medical D’alsace Igma, Strasbourg, France; 17Hôpital Saint Jacques, Centre de génétique humaine, Pole de biologie et anatomie pathologie,, Besançon, France; 18CHU de Nancy, Hôpitaux de Brabois, Service de génétique clinique et médicine infantile, hôpital d’enfants, Vandoeuvre les Nancy, France; 19UFR des Sciences de Santé, Université de Bourgogne-Franche-Comté,, Centre d'Investigation Clinique CIC-EC Inserm CIC1432, Dijon, France
Background/Objectives: Prenatal invasive exome sequencing (pES) has become a first-line diagnostic tool to refine fetal prognosis and guide couples in pregnancy management when ultrasound abnormalities are detected. Available non-invasive prenatal testing (NIPT) of circulating-free fetal DNA is currently limited to specific genetic indications. Leveraging our experience with pES, we compared NIPT-ES and pES approaches when ultrasound abnormalities are detected.
Methods: We performed trio pES (70X) and NIPT-ES (300X) in 70 pregnancies with ultrasound abnormalities (10-32 weeks of gestation). Only class 5, 4, and 3+ variants in pES, associated with the ultrasound phenotype, were reported during pregnancy. The results from both blindly interpreted strategies were compared and disclosed after the pregnancy outcome.
Results: Preliminary results showed that pES identified the causal diagnosis in 15/43 (35%), whereas NIPT identified it in 9/43 (21%) pregnancies respectively. pES and NIPT-ES were concordant in 36/43 (84%). Compared with pES, NIPT had a false-positive (FP) and false-negative (FN) rates of 3.5% (1/28) and 40% (6/15), respectively. FP and FN results were due to technical limitations of NIPT (two CNV detection) or the complexity of the prenatal situation (incomplete clinical information).
Conclusion: Preliminary results demonstrate the feasibility of NIPT-ES. However, the 3.5% FP rate suggests that its first-line use may lead to few unnecessary invasive procedures and the 40% FN rate suggests that diagnosis may be missed in some cases. All the 70 inclusions will be presented, but further evaluations of NIPT-ES are essential given the FP and FN rates before considering NIPT-ES as a routine strategy and its positioning in pregnancy management.
Conflict of Interest: None declared
C27.3 Comprehensive noninvasive fetal screening by deep trio-exome sequencing
Ieva Miceikaite 1;2, Qin Hao2, Charlotte Brasch-Andersen2, Christina Fagerberg2, Pernille Torring2, Britta Schlott Kristiansen3, Lilian Ousager1, Lene Sperling3, Mette Holm Ibsen4, Katrin Löser5, Martin Larsen1;2
1University of Southern Denmark, Clinical Research, Odense, Denmark; 2Odense University Hospital, Clinical Genetics, Odense, Denmark; 3Odense University Hospital, Gynecology and Obstetrics, Odense, Denmark; 4University Hospital of Southwestern Jutland, Esbjerg, Denmark; 5Hospital of Southern Jutland, Aabenraa, Denmark
Background/Objectives: Fetal genetic diagnosis is vital for prenatal care. While invasive prenatal trio-exome sequencing is effective, it’s limited to fetuses with structural anomalies, leaving many monogenic disorders undiagnosed. Noninvasive prenatal screening using maternal plasma cell-free DNA (cfDNA) revolutionized screening but mainly targets chromosomal disorders due to its low resolution. desNIPT, a novel noninvasive screening method using deep trio-exome sequencing, aims to address this gap.
Methods: In our study involving 36 pregnancies, deep trio-exome sequencing was performed on maternal plasma cell-free DNA and parental blood samples. The approach leverages ultra-deep, error-corrected, trio-exome sequencing for accurate detection of all fetal de novo variants, including single-nucleotide variants, indels, copy-number variants, and chromosomal aneuploidies. Results were compared with invasive prenatal analysis outcome.
Results: desNIPT showed 100% concordance with invasive methods in detecting pathogenic fetal de novo variants, including single-nucleotide variants, deletions, trisomies, and an unbalanced translocation. Mean sensitivity for detecting fetal sequence variants across protein-coding region was 95.12%. The method also enables additional carrier screening in both parents.
Conclusion: desNIPT offers a comprehensive, noninvasive approach for prenatal screening of a wide range of genetic disorders. Integrating this approach into routine prenatal care could improve early detection rates, reduce invasive procedures, and facilitate timely interventions, with a low false-positive rate and quick turnaround time. These findings support further exploration of deep-exome sequencing of cell-free DNA for screening fetuses without structural anomalies.
Grants: The Region of Southern Denmark (18/17848 and 20/14085), the Aase and Ejnar Danielsens Foundation (19-10-0259), the A.P. Møller Foundation for the Advancement of Medical Science (19-L-0281).
Conflict of Interest: Ieva Miceikaite Nonacus Ltd. is sponsoring my travel to ESHG, no sponsorship or involvement was provided for the study itself., Qin Hao: None declared, Charlotte Brasch-Andersen: None declared, Christina Fagerberg: None declared, Pernille Torring: None declared, Britta Schlott Kristiansen: None declared, Lilian Ousager: None declared, Lene Sperling: None declared, Mette Holm Ibsen: None declared, Katrin Löser: None declared, Martin Larsen: None declared
C27.4 Novel Utility of Non-Invasive Fetal Exome Sequencing
Christopher Whelan 1;2;3, Harrison Brand1;2;3;4;5, Michael Duyzend1;2;6, John Lemanski2, Jessica Giordano7, Monica Salani2, Stephanie Hao2;5, Elise Valkanas1;2, Caroline Cusick3, Diane Lucente2, Krithika Nathamuni1;2, Casie Genetti6, Lori Dobson8, Courtney Studwell8, Kathleen Gianforcaro8, Louise Wilkins-Haug9, Stephanie Guseh9, benjamin Curral2, Ronald Loosen7, Kathryn Gray10, Wendy Chung6, Ronald Wapner7, Michael Talkowski1;2;3;4
1Broad Institute of MIT and Harvard, Program in Medical and Population Genetics, Cambridge, United States; 2Massachusetts General Hospital, Center For Genomic Medicine, Boston, MA, United States; 3Broad Institute of MIT and Harvard, Stanley Center for Psychiatric Research, Cambridge, MA, United States; 4Massachusetts General Hospital and Harvard Medical School, Department of Neurology, Boston, MA, United States; 5Massachusetts General Hospital, Pediatric Surgical Research Laboratory, Boston, MA, United States; 6Boston Children’s Hospital and Harvard Medical School, Department of Pediatrics, Boston, MA, United States; 7Columbia University Medical Center, Department of Obstetrics and Gynecology, New York, NY, United States; 8Brigham and Women’s Hospital, Center for Fetal Medicine and Reproductive Genetics, Boston, MA, United States; 9Brigham and Women’s Hospital and Harvard Medical School, Division of Maternal-Fetal Medicine, Boston, MA, United States; 10University of Washington, Department of Obstetrics & Gynecology, Seattle, WA, United States
Background/Objectives: Analysis of cell-free DNA (cfDNA) has revolutionized prenatal diagnostics, providing a noninvasive screen for aneuploidies. We devised a higher-resolution approach with the ability to capture all coding fetal genetic variation: noninvasive fetal exome sequencing (NIFS). We applied NIFS to 51 pregnancies (gestational ages 9 – 40 weeks) and developed novel methods for detecting single nucleotide variants (SNVs), indels, and copy number variants (CNVs). We are currently performing a validation study that includes over 100 participants with matched NIFS and whole genome sequencing (WGS).
Methods: NIFS exome libraries were prepared from cfDNA extracted from maternal plasma and sequenced to 200x mean coverage on ~22,000 genes. We detected SNVs and indels using machine learning-based variant filtration and a Bayesian mixture model that disentangles maternal and fetal variants. We identified CNVs via analysis of read depth.
Results: In 11 samples with matched invasive sequencing, NIFS detected de novo and paternal variants with high sensitivity (median 96.3%) irrespective of fetal fraction. Across all variants, NIFS demonstrated high median sensitivity for fetal SNVs (93.0%) and indels (77.1%). All 5 clinically confirmed pathogenic variants were detected. NIFS genotyped mothers with high average accuracy (97.6% SNVs; 86.4% indels) and 57% had a reportable carrier variant. Our validation study permits further extensive benchmarking.
Conclusion: NIFS extends prenatal cfDNA screening to SNVs, indels, and CNVs, and potentially offers a rapid reflex test in the presence of ultrasound abnormalities. NIFS demonstrates that fetal exomes are accessible from maternal blood samples collected for prenatal screening.
Grants: NICHD (HD081256, HD105266, HD112084); NIMH (MH115957).
Conflict of Interest: None declared
C27.5 Fetal genome sequencing: what do we miss by prenatal exome sequencing ?
Agnese Feresin 1, Boris Keren2;3, Delphine Heron4, Benjamin Cogne5;6, Sarah Grotto7;8, Christele Dubourg6;9, Anne Dieux10, Sandra Whalen11, Chloe Quelin12, Jonathan Levy6;13, Daphné Lehalle8, Paul Gueguen6;14, Mathilde Nizon15, melanie rama6;16, LYSE RUAUD13, Sophie Rondeau6;17, Anne Brehin18, Corinne Collet6;17, Leila Ghesh15;19, Laurence Heidet6;20, YLINE CAPRI13, lydie BURGLEN6;21, Laurence Perrin13, severine drunat6;13, Catherine Delorme22, Fabienne Escande23;24, Claire Beneteau15, Lucie Boutaud6;25, Florence Petit26, Pascale Saugier-Veber6;27, Audrey Briand-Suleau6;28, Stéphanie Arpin29;30, Andrée Delahaye6;31, Manon Godin32, Marie Faoucher6;9, Anne-Marie Guerrot18, Nathalie Couque6;33, Alice Goldenberg18, Yoann Vial6;13, Alinoe Lavillaureix34, Adeline Bonnard13, Agnes Guichet6;35, Alban Lermine6, Pierre Blanc6;36, Thomas Smol6;37, Odile Boute38, Tania Attie-Bitach6;39
1Service de Médecine Génomique des Maladies Rares, UF MP5, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France; 2Centre de Génétique Moléculaire et Chromosomique, Hôpital de la Pitié-Salpêtrière, UF de génomique du développement, Assistance Publique-Hôpitaux de Paris, Sorbonne Université, Paris, France; 3Laboratoire de Biologie Médicale multisites SeqOIA – PFMG2025, Paris, France; 4Centre de Génétique Moléculaire et Chromosomique, Hôpital de la Pitié-Salpêtrière, UF de génomique du développement, Assistance Publique-Hôpitaux de Paris, Paris, France; 5Service de Génétique Médicale, Centre Hospitalier Universitaire de Nantes, Nantes, France; 6Laboratoire de Biologie Médicale multisites SeqOIA – PFMG2025, Paris, France; 7Maternité Port-Royal, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris, France; 8UF de génétique clinique, Centre de référence anomalies du développement et syndromes malformatifs, Hôpital Trousseau, Assistance Publique-Hôpitaux de Paris, Paris, France; 9Service de Génétique Moléculaire et Génomique, Centre Hospitalier Universitaire de Rennes, Rennes, France; 10Service de Génétique Clinique, Centre Hospitalier Régional Universitaire de Lille, Lille, France; 11UF de génomique du développement, Centre de Génétique Moléculaire et Chromosomique, Hôpital de la Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, France; 12Service de Génétique Clinique, CLAD Ouest, Centre Hospitalier Universitaire de Rennes, Rennes, France; 13Département de Génétique, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France; 14Laboratoire de Génétique Moléculaire · Service de génétique · CHRU de Tours - Hôpital Bretonneau, Tours, France; 15Service de Génétique Médicale, Centre Hospitalier Universitaire de Nantes, Nantes, France; 16Institut de Génétique Médicale, Centre Hospitalier Régional Universitaire de Lille, Université de Lille, Lille, France; 17Service de Médecine Génomique des Maladies Rares - GHU Necker- Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France; 18Service de Génétique-Unité de Génétique Clinique, Centre Hospitalier Universitaire de Rouen, Rouen, France; 19UF de Foetopathologie et Génétique, Centre Hospitalier Universitaire de Nantes, Nantes, France; 20Service de Néphrologie Pédiatrique, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Centre de Référence MARHEA, Paris, France; 21Centre de Référence des Malformations et Maladies Congénitales du Cervelet, Département de Génétique et Embryologie Médicale, Assistance Publique-Hôpitaux de Paris, Hôpital Trousseau, Paris, France; 22Pôle De Biologie Pathologie Génétique, Hôpital Jeanne de Flandre, Centre Hospitalier Régional Universitaire De Lille, Lille, France; 23Laboratoire de Biochimie et Biologie Moléculaire, Centre Hospitalier Régional Universitaire de Lille, Lille, France; 24Laboratoire de Biologie Médicale multisites SeqOIA – PFMG2025; 25Service de Médecine Génomique des Maladies Rares, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France; 26Service De Génétique Clinique, Centre Hospitalier Régional Universitaire De Lille, Lille, France; 27Département de génétique et Centre de référence anomalies du développement, CHU de Rouen, Rouen, France; 28Service de Médecine Génomique des Maladies de Système et d’Organe, Hôpital Cochin, AP-HP Centre-Université de Paris Cité, Hôpital Cochin, Assistance Publique-Hôpitaux De Paris, Paris, France; 29Service De Génétique, Centre Hospitalier Universitaire De Tours, Tours, France; 30Université De Tours, INSERM, Tours, France; 31Unité Fonctionnelle De Médecine Génomique Et Génétique Clinique, Hôpital Jean Verdier, Assistance Publique Des Hôpitaux De Paris, Université Sorbonne Paris Nord, Paris, France; 32Départemet de génétique, Centre de reference anomalies du développement, Centre Hospitalier Universitaire De Caen, Caen, France; 33Service de Génétique, Hôpital Robert-Debré, Assistance Publique-Hôpitaux de Paris, Paris, France; 34Service de Génétique Clinique, Centre Hospitalier Universitaire de Rennes, Rennes, France; 35Génétique, Centre Hospitalier Universitaire D’Angers, Université d’Angers, Angers, France; 36Département de Génétique, Hôpital Armand Trousseau, Assistance Publique-Hôpitaux de Paris City: Paris, Université Sorbonne Université, Paris, France; 37Centre Hospitalier Régional Universitaire de Lille, Institut de Génétique Médicale, Université, Lille, France; 38Service de Génétique Clinique, Hôpital Jeanne de Flandre, CLAD Nord, Lille, France; 39Service de Médecine Génomique des Maladies Rares, UF MP5, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Paris, France
Background/Objectives: Prenatal cytogenetics and chromosomal microarray (CMA) solve 40-45% of congenital anomalies and exome sequencing (ES) improves diagnostic yield in 26-36% of cases. The study aims to describe the input of genome sequencing (GS) in a cohort of postmortem fetuses/newborns with structural anomalies and to compare the efficiency of GS versus prenatal CMA + ES.
Methods: We collected information of 482 index cases sequenced at the French Laboratory SeqOIA (2020-April 2023) including terminations of pregnancy (418), in-utero deaths (39), fetal demises (4), and deceased newborns (21). Prior to GS: all had CMA, pathological examination; some cases had ES (39) or targeted analysis (59).
Results: GS solved 162 cases (34%). Chromosomal abnormalities (5) included mosaic imbalances in fetal tissue (3) - absent in amniocytes (2) or villi (1) – and balanced inversion (1). Monogenic disorders (157) involved 128 disease genes due to structural variants (15), variants in coding (194), and in non-coding regions (2). Eleven solved cases (7%) had previous ES. Eleven variants initially classified as of uncertain significance (VUS) were reclassified, mainly following RNA analysis (7). GS was non conclusive in 298 cases (62%), and with a VUS in 22 cases (4%).
Conclusion: GS is effective for etiology determination of fetal structural anomalies. Overpowering CMA + ES by identifying non coding variants, balanced structural variants, CNVs below the resolution of CMA, and overcoming limitations of ES detection, GS should be rapidly proposed in prenatal diagnosis. Fetal pathology is crucial for deep phenotyping, genotype/phenotype correlations, and tissue sampling/analysis. New strategies are needed for non conclusive cases.
Grants:
Conflict of Interest: None declared
C27.6 Non-invasive cell-free DNA methylation profiling of pregnant women for the diagnosis and early risk determination of preeclampsia
Machteld Baetens1, Bram Van Gaever 2, Stephanie Deblaere3, Andries De Koker4, Nico Callewaert4, Kristien Roelens3, Ellen Roets3, Isabelle Dehaene3, Björn Menten5
1Ghent University Hospital, Center for Medical Genetics, Ghent, Belgium; 2Ghent University, Medical Genetics, Ghent, Belgium; 3Ghent University Hospital, Women’s Clinic, Ghent, Belgium; 4Flanders Institute for Biotechnology, Ghent, Belgium; 5Ghent University Hospital - Ghent University, Medical Genetics, Ghent, Belgium
Background/Objectives: Aberrant embryo implantation and suboptimal placentation can lead to (severe) pregnancy complications, like preeclampsia. Current identification of high-risk pregnancies relies on combining risk factors, biomarkers, and ultrasound examinations, a relatively inaccurate approach. An improved first trimester screening could enhance understanding placental development and pathology, to provide better prenatal care. Given the identification of disease-associated methylation changes in cfDNA, this study explores this potential of prenatal, non-invasive methylation screening.
Methods: We performed genome-wide cfDNA methylation profiling to identify significantly differentially methylated regions (DMRs) between uneventful pregnancies and those complicated with preeclampsia, with two objectives: diagnose preeclampsia at time of maternal symptoms and predict preeclampsia during the pre-symptomatic first trimester. Furthermore, we performed a longitudinal cfDNA methylation profiling of women with a complicated pregnancy and with a normal-term birth to investigate methylation evolution and to identify differences associated with placental dysfunction.
Results: We detected DMRs in pregnancies complicated with preeclampsia as early as 12 weeks of gestation. DMRs could be categorized into two groups, with an observed difference in genomic methylation patterns between early and late pregnancy. We were able to build two separate classification models which performed well in predicting or diagnosing preeclampsia on a small set of validation samples. Investigation of methylation patterns throughout the pregnancy yielded several methylation trends that showed to be significantly different between unaffected and affected pregnancies.
Conclusion: This study demonstrated the efficacy of non-invasive cfDNA methylation profiling in understanding pathology of preeclampsia. Our results offer promising potential for integrating this approach into routine prenatal care.
Grants: Vlaams Industrieel Onderzoeksfonds (F2020/IOF_STarTT035)
Conflict of Interest: None declared
C28 Machine Learning and Statistics
C28.1 Integration of protein language models, regulatory CNNs and other nucleotide-level scores improves genome-wide variant prediction of CADD
Max Schubach 1, Thorben Maass2, Lusiné Nazaretyan1, Sebastian Röner1, Martin Kircher1;2
1Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Exploratory Diagnostic Sciences, Berlin, Germany; 2University Hospital Schleswig-Holstein, University of Lübeck, Institute of Human Genetics, Lübeck, Germany
Background/Objectives: Machine Learning-based scoring and classification of genetic variants aids the assessment of clinical findings and is employed to prioritize variants in diverse genetic studies and analyses. Combined Annotation-Dependent Depletion (CADD) is one of the first methods for the genome-wide prioritization of variants across different molecular functions and has been continuously developed and improved since its original publication.
Methods: Here, we present our most recent release, CADD v1.7. We explored and integrated new annotation features, among them protein language model scores (Meta ESM), regulatory variant effect predictions (from sequence-based convolutional neural networks) and sequence conservation scores (Zoonomia). We evaluated the new version on data sets derived from ClinVar and gnomAD. For coding effects, we tested CADD on 31 Deep Mutational Scanning (DMS) datasets, and, for regulatory effect prediction, we used saturation mutagenesis reporter assay data of promoter and enhancer sequences.
Results: The individual inclusion of new features in CADD improved performance on variant prioritization, respectively. Further, the integration of all features improved the overall performance, and we released the improved model as CADD v1.7. As with previous releases, all data sets, genome-wide CADD v1.7 scores, scripts for on-site scoring and an easy-to-use webserver are provided via https://cadd.bihealth.org/.
Conclusion: With v1.7 we moved CADD further into the direction of prioritizing regulatory variants effects and deep learning models for protein and regulatory grammar. We conclude that CADD highly benefits from these sequence models, keeping it up-to-date with the latest research.
Grants: Deutsche Forschungsgemeinschaft (DFG) [464313370]; Impact of Genomic Variation on Function/NHGRI [1UM1HG011966-01];
Conflict of Interest: None declared
C28.2 NERINE: Network-based association test for rare variants in complex traits and diseases
Sumaiya Nazeen 1;2, Xinyuan Wang2, Autumn Morrow1;2, Vikram Khurana2, Shamil Sunyaev1;3
1Harvard Medical School, Biomedical Informatics, Boston, United States; 2Brigham and Women’s Hospital, Neurology, Boston, United States; 3Brigham and Women’s Hospital, Medicine/Division of Genetics, Boston, United States
Background: Rare protein-coding variants provide a direct path from genetics to biology but are difficult to detect. Traditional single-variant and gene-level tests have limited power. A promising strategy to increase power is to combine variants from multiple genes in a network. However, gene networks can have noisy definitions, with many genes having no effect or varying effects in different directions, posing additional challenges.
Methods: We present NERINE, a new statistical framework that infers the effect of rare variants aggregated across a gene network on a trait using a maximum-likelihood framework. We encode gene-gene relationships in a symmetric positive-definite matrix that can accommodate data from different screens. Genes can have either zero effect or varying degrees of trait-increasing and trait-decreasing effects. We test for network significance by nested hypothesis testing, providing an asymptotic p-value.
Results: NERINE outperforms existing methods in simulations, especially on noisy networks and imbalanced case-control datasets. It also helps us select the most relevant functional screen for a trait. When applied to LDL, HDL, and early MI phenotypes in the UK Biobank, NERINE recovers networks of lipid genes with expected directions of effects. Additionally, it identifies a novel gene module involved in Parkinson’s disease in UKBB and AMP-PD datasets, which we validate in CRISPRi experiments in a synucleinopathy iPSC model.
Conclusion: Overall, NERINE can identify novel disease-associated gene networks with rare variant burden, provide evidence of a screen’s relevance to a given disease, and facilitate a mechanistic understanding of the disease process.
Grants: This project is supported by grants NIH R35-GM127131 and ASAP 124423.
Conflict of Interest: Sumaiya Nazeen Full-time postdoctoral fellow at Harvard Medical School, Consultant for Dacapo Brainscience, Inc., Xinyuan Wang Full-time postdoctoral fellow at Brigham and Women’s Hospital, Consultant at Dacapo Brainscience, Inc., Autumn Morrow Full-time research assistant at Brigham and Women’s Hospital, Vikram Khurana: None declared, Shamil Sunyaev: None declared
C28.3 LDAK-PBAT: A Pathway-Based Analysis Tool for Decoding the Genetics of Complex Diseases
Takiy Berrandou1, Doug Speed1
1Aarhus University, Center for Quantitative Genetics And Genomics (QGG), Aarhus, Denmark
Background/Objectives: The quest for personalized medicine necessitates sophisticated analysis tools capable of deciphering complex disease genetics. We introduce LDAK-PBAT, a novel tool that offers a holistic systems biology approach by simultaneously estimating variant effects.
Methods: Employing a heritability-based framework, LDAK-PBAT collectively analyzes multiple genes within pathways, refined through simulations and validated against datasets like the UK Biobank. Its performance was benchmarked against established tools, including MAGMA.
Results: LDAK-PBAT demonstrates exceptional accuracy in pathway heritability estimation, achieving RMSE values as low as 0.001084 and correlation coefficients up to 0.98. Comparative performance analysis reveals its enhanced sensitivity and precision, outperforming MAGMA and Hypergeometric testing with an F1 score of 0.734 versus 0.636 and 0.570, respectively. At the stringent threshold of 2.78e-06, it maintains an F1 score of 0.518, significantly higher than MAGMA’s 0.206. In analyzing 6000 pathways across 10 UK Biobank traits at the Bonferroni threshold, LDAK-PBAT identifies 344 significant pathways, markedly surpassing MAGMA’s 6, and demonstrating its superior pathway detection capabilities. Validation across increasing sample sizes further confirms LDAK-PBAT’s efficacy, with a significant increase in pathways retaining significance, affirming its reliability in genetic pathway analysis. Moreover, its robustness against variations in SNP lists and reference panels underscores the tool’s consistent performance.
Conclusion: LDAK-PBAT emerges as a potent tool for genetic research, offering advanced insights into complex diseases. Its precise estimation of pathway enrichment and heritability, along with superior performance in detecting significant pathways, underscores its potential to revolutionize genetic analysis and further the advancement of personalized medicine.
Conflict of Interest: None declared
C28.4 Adjusting for environmental factors in multi-ancestry meta-regression improves power to detect genetic associations
Siru Wang 1, Oyesola Ojewunmi2, Tinashe Chikowore3;4;5, Abram Kamiza6;7;8, Michele Ramsay6, Andrew Morris9, Segun Fatumo2;7, Jennifer Asimit1
1University of Cambridge, MRC Biostatistics Unit, Cambridge, United Kingdom; 2London School of Hygiene and Tropical Medicine, Department of Non-Communicable Disease Epidemiology, London, United Kingdom; 3University of the Witwatersrand, MRC/Wits Developmental Pathways for Health Research Unit, Department of Paediatrics, Faculty of Health Sciences, Johannesburg, South Africa; 4Brigham and Women’s Hospital, Channing Division of Network Medicine, Boston, United States; 5Harvard Medical School, Boston, United States; 6University of the Witwatersrand, Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, Johannesburg, South Africa; 7MRC/UVRI and LSHTM, The African Computational Genomic (TACG) Research Group, Entebbe, Uganda; 8Malawi Epidemiology and Intervention Research Unit, Lilongwe, Malawi; 9The University of Manchester, Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Manchester, United Kingdom
Background/Objectives: Meta-analysis of genome-wide association studies (GWAS) across diverse populations offers gains in power to identify associations with small effect sizes through sample size increases. Common models for meta-analysis are grouped into fixed-effects (FE), random-effects (RE), and multi-ancestry meta-regression (MR-MEGA). However, with the growing number of genome-wide association (GWAS) data collected from highly diverse populations, it is likely for heterogeneous effect sizes across the populations to be correlated with not only ancestry but also environmental exposures, such as dietary and lifestyle factors. The current models cannot easily accommodate these factors in a meta-analysis framework that uses GWAS summary statistics.
Methods: To address this challenge, we present an environment-adjusted meta-regression model (env-MR-MEGA) to detect genetic variant associations by adjusting for differing environmental exposures between populations. Additionally, env-MR-MEGA quantifies the extent of heterogeneity due to ancestral and environmental effects.
Results: Through a series of simulations, we demonstrate that env-MR-MEGA increases power over MR-MEGA as high as 16% to detect genetic associations. In genetic association studies with LDL cholesterol in 20,000 individuals across twelve sex-stratified GWAS from Africa, adjusting for sex and overweight status within env-MR-MEGA leads to the detection of three novel loci, of which one has evidence of heterogeneity due to sex and overweight status, and another for ancestral heterogeneity.
Conclusion: The proposed meta-analysis method provides a way to adjust and test for environmental effects using summary-level data, making it a useful tool for meta-analyses without the need to share individual-level data.
Grants: Funded by an MRC-NIHR Better Methods, Better Research grant (MR/W02098X/1).
Conflict of Interest: None declared
C28.5 Understanding nurture via untransmitted genetic alleles
Robin J. Hofmeister 1;2;3, Liza Darrous1;2;3;4, Zoltan Kutalik1;2;3
1Unisanté I Policlinic Medicine Générale Du Bugnon, Lausanne, Switzerland; 2UNIL - Université de Lausanne, Lausanne, Switzerland; 3SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland; 4University of Geneva, Geneva, Switzerland
Background/Objectives: Studies distinguishing transmitted and untransmitted genetic variants revealed that the genetic basis of educational attainment (EA) is substantially driven by the effect of parental alleles acting through parental rearing. While most studies focussed on accounting for these indirect genetic effects (IGEs), our aim is to accurately estimate them, in order to establish parental nurturing effects via Mendelian Randomisation (MR).
Methods: To boost sample size, we introduce a novel approach for estimating IGEs without the need for parental information. For each target individual, we identify close relatives and pinpoint Identity-By-Descent sharing among them to infer untransmitted parental alleles. These alleles are then employed to estimate associations with various offspring phenotypes. Finally, we employ MR to understand how parental characteristics can influence offspring traits.
Results: Using UK Biobank data, we inferred untransmitted alleles for ~130’000 individuals (vs ~5’000 parent-offspring pairs). The inference accuracy depends on relatedness: median R-squared of 0.81, 0.33, and 0.25 for inference from sibling, second-degree and third-degree relatives, respectively. We tested associations between a range of traits and untransmitted parental alleles and successfully replicated these associations using 178’000 sibling pairs from 19 cohorts. Plugging these estimates into MR, we identified a widespread impact of parental characteristics on offspring traits, notably highlighting the influence of parental obesogenic environment and socio-economic status on offspring EA and body mass index.
Conclusion: Overcoming the traditional requirement for parental information by leveraging inter-individual relatedness in biobanks allows us to evaluate nurturing effects at an unprecedented scale.
Grants: Swiss National Science Foundation 310030-189147
Conflict of Interest: None declared
C28.6 Multi-trait reverse regression for gene burden and rare variant association tests.
Andrey Ziyatdinov 1, Joseph Herman1, Benjamin Geraghty1, Karl Landheer1, Joelle Mbatchou1, Jonathan Marchini1
1Regeneron Genetics Center, Tarrytown, NY, United States
Background/Objectives: Understanding the genetic basis of complex traits often requires analyzing multiple interrelated phenotypes. Most genetic association studies are performed for a single quantitative or binary trait at a time. However, joint analysis of multiple traits can boost power and aid interpretation. Existing multi-trait approaches have focused on analysis of common variants, with gene burden and rare variant multi-trait tests having received less attention. The advent of large scale exome sequencing studies of deeply phenotyped cohorts motivates the need for new approaches.
Methods: We have developed a reverse regression multi-trait test that works on rare variants, gene burden and common variants, and on any combination of quantitative and binary traits. The test is implemented within the REGENIE framework so utilizes a whole-genome regression model that accounts for population structure, relatedness and polygenicity. To control Type I error at tests of rare variants and binary traits with imbalanced case control ratios, we use Firth logistic or proportional odds models fitted with a robust pseudo-response step-halving algorithm.
Results: We use UK Biobank imputed genotype and exome sequencing data in up to 414,000 individuals together with simulated phenotypes to demonstrate the Type I error control and power of this approach. We further illustrate the approach using multiple phenotypes derived using MRI and DXA images from the UK Biobank imaging study.
Conflict of Interest: Andrey Ziyatdinov Regeneron Genetics Center or Regeneron Pharmaceuticals, Regeneron Genetics Center or Regeneron Pharmaceuticals, Joseph Herman Regeneron Genetics Center or Regeneron Pharmaceuticals, Regeneron Genetics Center or Regeneron Pharmaceuticals, Benjamin Geraghty Regeneron Genetics Center or Regeneron Pharmaceuticals, Regeneron Genetics Center or Regeneron Pharmaceuticals, Karl Landheer Regeneron Genetics Center or Regeneron Pharmaceuticals, Regeneron Genetics Center or Regeneron Pharmaceuticals, Joelle Mbatchou Regeneron Genetics Center or Regeneron Pharmaceuticals, Regeneron Genetics Center or Regeneron Pharmaceuticals, Jonathan Marchini Regeneron Genetics Center or Regeneron Pharmaceuticals, Regeneron Genetics Center or Regeneron Pharmaceuticals
C29 Fantastic OMICS - Improved methods for diagnostics
C29.1 GestaltMatcher Database - A global reference for the phenotypic variability of rare diseases in humans
Hellen Lesmann 1, Gholson Lyon2, Pilar Caro3, Ibrahim Abdelrazek4, Alexander Hustinx1, Hannah Klinkhammer1, Shahida Moosa5, Jean Tori Pantel6, Annabelle Arlt1, Elaine Marchi2, Hannah Weiland1, Merle ten Hagen1, Sophia Kaptain1, Alexej Knaus1, Behnam Javanmardi1, Rami Abou Jamra7, Rebekah Waikel8, Sebastien Küry9, Frédéric Ebstein9, Sylvia Safwat4, Franziska Schnabel7, Artem Borovikov10, Claudio Graziano11, Amira Nabil4, Vincent Strehlow7, Clara Velmans12, Miriam Elbracht13, Cordula Knopp13, Annette Uwineza14, Luisa Averdunk15, Tinatin Tkemaladze16, Himanshu Goel17, Stefan Barakat18, Nergis Güzel13, Julia Suh13, Theresa Brunet19, Sabine Rudnik20, Christian Schaaf3, Benjamin D. Solomon8, Christoffer Nellaker21, Alain Verloes22, Koen Devriendt23, Martin Mücke6, Aleksandra Jezela24, Karen W. Gripp25, Christian Netzer12, Ebtessam Abdallah4, Markus M. Nöthen26, Peter Krawitz1, Tzung-Chien Hsieh1
1Institute for Genomic Statistics and Bioinformatics, University Hospital Bonn, Bonn, Germany; 2Department of Human Genetics, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, United States; 3Institute of Human Genetics, Heidelberg University, Heidelberg, Germany; 4Department of Human Genetics, Medical Research Institute, Alexandria University, Alexandria, Egypt; 5Division of Molecular Biology and Human Genetics, Stellenbosch University and Medical Genetics, Tygerberg Hospital, Stellenbosch, South Africa; 6Institute for Digitalization and General Medicine, University Hospital RWTH Aachen, Aachen, Germany; 7Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany; 8Medical Genomics Unit, Medical Genetics Branch, National Human Genome Research Institute, Bethesda, United States; 9Service de Génétique Médicale, Nantes Université, CHU Nantes, Nantes, France; 10Research Centre for Medical Genetics (RCMG), Moscow, Russian Federation; 11Medical Genetics Unit, Ausl Romagna, Cesena, Italy; 12Institute of Human Genetics, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; 13Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH Aachen University, Aachen, Germany; 14Department of Clinical Biology, University of Rwanda, Kigali, Rwanda; 15Clinic for General Paediatrics, Neonatology and Paediatric Cardiology, University Hospital Düsseldorf, Düsseldorf, Germany; 16Department of Molecular and Medical Genetics, Tbilisi State Medical University, T’bilisi, Georgia; 17Hunter Genetics, Waratah Campus Newcastle, Newcastle, Australia; 18Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, Netherlands; 19Institute of Human Genetics, Technische Universität München, München, Germany; 20Institute Of Human Genetics, Medical University Innsbruck, Innsbruck, Austria; 21Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, Nuffield Department of Women’s & Reproductive Health, University of Oxford, Oxford, United Kingdom; 22Department of Clinical Genetics, Robert-Debré Hospital, Paris, France; 23Center for Human Genetics, Uni Leuven, Leuven, Belgium; 24Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, Warsaw, Poland; 25Division of Medical Genetics, A.I. du Pont Hospital for Children/Nemours, USA, Wilmington, United States; 26Institute of Human Genetics, University Hospital Bonn, Bonn, Germany
Background: Characteristic facial dysmorphism is more difficult for both humans and AI to recognize when there are no cases of the same ethnicity to learn from. Although portraits in the literature are predominantly of Europeans, most children with rare disorders are now born in Asia and Africa, indicating a need for action.
Methods: We designed GestaltMatcher Database (GMDB) - a platform compliant with the FAIR Data principles. Each case in GMDB can get a Digital Object Identifier (DOI), becoming a citable micro-attribution. We curated data of individuals with dysmorphic features from 2,224 publications to initialize GMDB. Then, we encouraged clinician scientists and patient support groups from everywhere to contribute cases with molecular diagnoses of diverse ethnic backgrounds. Finally, we analyzed the impact of the novel data on the performance of GestaltMatcher AI.
Results: We have compiled a collection of 10,189 frontal images from 7,695 patients representing 683 disorders. These data contain 1,409 novel medical images that registered users can query. Our global collaborators increased the data of underrepresented ethnicities to 20% from Asia and 5% from Africa. Our analysis with next-generation phenotyping (NGP) indicate that higher ethnic diversity enhances the overall performance of GestaltMatcher AI. For all ethnic groups, incorporating non-European cases into the training set increased the top-1-accuracy by 32% and top-5-accuracy by 13%.
Conclusion: GMDB is a resource for medical image data of individuals with rare disorders from diverse ethnic backgrounds that can contribute to the training of dysmorphologists and NGP technology worldwide.
Grants: Eva Luise und Horst Köhler Stiftung. Arbeitsgemeinschaft für Gendiagnostik
Conflict of Interest: None declared
C29.2 The true recurrence risk: parental mosaicism in a large family cohort with de novo variants
Sonja Neuser 1, Natalie Ahmad1, John Wiedenhöft1, Denny Popp1, Johannes Lemke1, Knut Krohn2, Ulrike Klotz3, Toralf Kirsten3, Maike Karnstedt1, Julia Hentschel1, Stephan Drukewitz1, Rami Abou Jamra1
1Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany; 2Core Unit DNA Technologies, Medical Faculty, University Leipzig, Leipzig, Germany; 3Medical Data Science, University of Leipzig Medical Center, Leipzig, Germany
Background/Objectives: Parents of children with genetic disorders due to de novo variants are usually counselled on a recurrence probability estimate of 1-3% for further affected siblings, while the actual probability varies between 0 and 50%. This discrepancy is well-known, but barely investigated.
Methods: We prospectively enrolled 138 families, where a pathogenic putative de novo variant had previously been identified in an affected child. Covering two germ layers, we collected blood (n = 276), buccal (n = 228) and nail samples (n = 229) of both parents and a semen sample of the father (n = 92). We performed deep sequencing achieving >1000x coverage after unique molecular identifiers (UMI) collapsing for all variants across all tissues (n ≈ 130,000 measured values). Sophisticated Bayesian analyses regarding low mosaicism, correlations of allele fractions in different tissues, and probability models are ongoing. Using Nanopore long read sequencing, variants were phased to identify the parental allele of origin.
Results: We identified clear (>1%) mosaicism for six variants in either the mother (n = 1) or the father (n = 5). In two cases, only the semen sample was affected. The alternative allele fraction varied from 1.3% to 23%. Phasing revealed one quarter of variants are located on the maternal allele and, three quarters on the paternal allele.
Conclusion: Our observation suggests that parental mosaicism could be less common than assumed. For families with a child carrying a de novo variant, testing the semen - and not the blood of the father - is an indispensable analysis to give a reliable answer.
Grants: DFG AB393/11-1 and NE2706/2-1
Conflict of Interest: Sonja Neuser DFG NE2706/2-1, Natalie Ahmad: None declared, John Wiedenhöft: None declared, Denny Popp: None declared, Johannes Lemke PTC Therapeutics, GRIN Therapeutics, UCB Pharma, Angelini Pharma, Knut Krohn: None declared, Ulrike Klotz: None declared, Toralf Kirsten: None declared, Maike Karnstedt: None declared, Julia Hentschel: None declared, Stephan Drukewitz: None declared, Rami Abou Jamra DFG AB393/11-1
C29.3 Towards population-scale transcriptomics — RNA-seq using decades-old blood spot samples
Victor Yakimov 1, Andres Ingason2, Christine Søholm1, Marie Bækvad-Hansen1, Preben Bo Mortensen2, merete nordentoft2, Ole Mors2, Anders Børglum2, David Michael Hougaard1, Bjarke Feenstra3, Thomas Werge2, Alfonso Buil2, Jonas Bybjerg-Grauholm1
1Statens Serum Institut, Department for Congenital Disorders, København, Denmark; 2The Lundbeck Foundation Initiative for Integrative Psychiatric Research; 3Statens Serum Institut, Department of Epidemiology Research, Copenhagen, Denmark
Background/Objectives: Biobanks contain millions of dried blood spot (DBS) samples obtained for routine neonatal screening. These samples create an opportunity to study biological signals preceding clinical presentation of disease. To date, no large-scale RNA-seq study in DBS samples has been published. Here, we show that such studies are feasible in samples stored at -20ºC for up to four decades.
Methods: We examine the quality of RNA-seq data obtained from DBS by inspecting transcriptomic profiles as a function of time they have spent in storage, and replicate differential expression analysis on sex, as well as expression-quantitative trait loci (eQTL) analysis performed by the Genotype-Tissue Expression (GTEx) consortium.
Results: We find no evidence that fewer genes can be identified in our samples compared to RNA-sequencing done in fresh whole blood samples. We also show that robust biological signals can be detected by closely replicating two high-quality studies performed by GTEx. While we find evidence of transcript degradation associated with a sample’s age, we find that it does not impact our ability to detect meaningful biological signals, and it can be controlled for by using standard statistical approaches.
Conclusion: Our results open the door to a new wave of experiments that can measure transcriptomic profiles in the blood of newborns and integrate that information with genetic data and life-time medical history of thousands of individuals.
Grants: This study was supported by The Lundbeck Foundation. This research has been conducted using the Danish National Biobank resource, supported by the Novo Nordisk Foundation.
Conflict of Interest: None declared
C29.4 Harnessing RNA-Sequencing in rare disease diagnostics: Insights from a multi-regional pilot project.
Carolina Jaramillo Oquendo 1, Matthew Lyon2, Dave Bunyan2, Jacob Abbott2, Jessica Woodley3, Samantha Court3, Jennie Bell3, Andrew Douglas4, Sarah Willis5, N Simon Thomas2, Anthony Williams5, Sarah Ennis1, Diana Baralle1
1University of Southampton, Southampton, United Kingdom; 2Wessex Genomics Laboratory Service, Salisbury, United Kingdom; 3West Midlands Regional Genetics Laboratory, Birmingham, United Kingdom; 4Oxford Centre for Genomic Medicine, Oxford, United Kingdom; 5Central & South Genomic Medicine Service Alliance, United Kingdom
Background/Objectives: Whole exome and genome sequencing (WES/WGS) have revolutionized the genetic testing of rare disease, however, even with WGS, nearly half of patients remain without a conclusive diagnosis. In 2022, the Central and South Genomic Medicine Service Alliance (GMSA) initiated a project with regional collaborations aiming to evidence RNA-based testing as a technique that increases diagnostic yield and develop proposals for how RNA-based testing could be incorporated into NHS England’s National Genomics Test Directory.
Methods: Clinical leads identified three patient cohorts: patients with splicing variants of uncertain significance (VUS) [n = 76], patients with no genetic diagnosis following DNA testing [n = 41] and patients with a recessive disorder and only one pathogenic variant identified [n = 3]. RNA was extracted from blood collected in PAXgene tubes and sent for sequencing in batches of 16. FASTQ files were aligned to the reference genome using STAR. Aligned files were visually inspected using the Integrative Genomics Viewer to detect splicing abnormalities.
Results: To date 39 VUS cases have been analysed, resolving 28 with RNA-Sequencing alone and 10 using RT-PCR. RNA-based testing aided in the re-classification of 68% of VUSs. For 33% of VUSs, RNA-sequencing identified complex splicing abnormalities missed by RT-PCR. In at least one case results enabled preimplantation genetic diagnosis. We will compare RNA-seq and RT-PCR, noting pros and cons, and share insights on infrastructure and data governance challenges.
Conclusion: We implemented a high-throughput RNA-sequencing workflow within the Central & South GMSA, with forthcoming steps aimed at collating results across regions to inform clinical integration.
Grants: NHSE, NIHR
Conflict of Interest: None declared
C29.5 Using Multiplexed Functional Data to Reduce Variant Classification Disparities in Populations Underrepresented in Genomic Medicine (virtual)
Moez Dawood 1;2;3, Shawn Fayer4;5, Mason Post5, Divya Kalra1, Karynne Patterson4, Eric Venner1;2, Lara Muffley4;5, Douglas Fowler4;5, Alan Rubin6;7, Jennifer Posey2, Sharon E Plon1;2, James Lupski1;2, Richard Gibbs1;2, Lea Starita4;5, Carla Daniela Robles Espinoza8;9, Willow Coyote-Maestas10;11, Irene Gallego Romero12;13;14
1Baylor College of Medicine, Human Genome Sequencing Center, Houston, United States; 2Baylor College of Medicine, Molecular and Human Genetics, Houston, United States; 3Baylor College of Medicine, Medical Scientist Training Program, Houston, United States; 4University of Washington, Genome Sciences, Seattle, United States; 5University of Washington, Brotman Baty Institute, Seattle, United States; 6The Walter and Eliza Hall Institute of Medical Research, Bioinformatics Division, Parkville, Australia; 7University of Melbourne, Medical Biology, Melbourne, Australia; 8Wellcome Sanger Institute, CASM, Hinxton, United Kingdom; 9Universidad Nacional Autónoma de México, Laboratorio Internacional de Investigación sobre el Genoma Humano, Ciudad de México, Mexico; 10University of California San Francisco, Bioengineering and Therapeutic Sciences, San Francisco, United States; 11University of California San Francisco, Quantitative Biosciences Institute, San Francisco, United States; 12University of Tartu, Center for Genomics, Evolution and Medicine, Tartu, Estonia; 13St. Vincent’s Institute of Medical Research, Human Genomics and Evolution, Fitzroy, Australia; 14The University of Melbourne, School of BioSciences and Melbourne Integrative Genomics, Parkville, Australia
Background/Objectives: The saturation-nature of Multiplexed Assays of Variant Effects (MAVEs) may offer a systematic experimental approach to address variant classification disparities between different populations, especially for variants of uncertain significance (VUS).
Methods: We analyzed the distribution of clinical significance classifications in genome sequencing data from 245,394 individuals in All of Us v7 and replicated our findings in two independent population-sale databases of the Genome Aggregation Database (gnomAD), 123,709 exomes of gnomAD v2.1.1 and 51,535 genomes of gnomAD v3.1.2 (non v2). Then, we incorporated clinically calibrated MAVE data into the ClinicalGenome’s Variant Curation Expert Panel rules and automated VUS reclassification.
Results: Using two orthogonal statistical approaches, we show across all 3 databases a higher prevalence (p ≤ 5.95e-06) of VUS in individuals of non-European genetic ancestry across all medical specialties assessed. Further, in the non-European genetic ancestry group, higher rates of Benign or Likely Benign and variants with no clinical designation (p ≤ 2.5e-05) were found across many medical specialties, whereas Pathogenic or Likely Pathogenic assignments were increased in individuals of European genetic ancestry (p ≤ 2.5e-05).
Using publicly-accessible MAVE data, we then reclassified 80.3% of all VUS in BRCA1, TP53, and PTEN across the three databases and reclassified non-European VUS at a significantly higher proportion in comparison to European VUS (p ≤ 8.7e-06) effectively compensating for the original VUS disparity.
Conclusion: These findings demonstrate the potential of MAVEs to reduce disparities in variant classification for underrepresented populations to make genomic medicine more informative and inclusive for all.
Grants: NHGRI-U01HG011758-01, NHGRI-UM1HG011969-03S, CPRIT-RP210027
Conflict of Interest: None declared
C29.6 A benchmarking database of SNPs, indels and SVs created using a four-generation, 28-member pedigree sequenced with multiple technologies
Zev Kronenberg 1, Cillian Nolan1, David Porubsky2, Katy Munson2, William Rowell1, Michelle Noyes2, Brent Pedersen3, Sean Mcgee2, Josh Smith2, Cairbre Fanslow1, primo baybayan1, Nidhi Koundinya2, William Harvey2, Kendra Hoekzema2, Jordan Knuth2, Gage Garcia2, Tom Mokveld1, Egor Dolzhenko1, Scott Watkins3, Deborah Neklason3, Aaron Quinlan3, Lynn Jorde3, Evan Eichler2, Michael Eberle1
1PacBio, Menlo Park, United States; 2University of Washington, Genome Sciences, Seattle, United States; 3The University of Utah, Salt Lake City, United States
Background/Objectives: Accurate long-read sequencing makes it possible to survey the full spectrum of genetic variation. Developing and validating new variant calling methods requires a comprehensive benchmarking dataset but current benchmark datasets have been developed primarily from short-read technologies and cannot adequately quantify the performance of long-read methods.
Methods: We have created a more comprehensive variant truthset by utilizing the power of genetic inheritance within a four-generation family (CEPH 1463) sequenced with multiple technologies (PacBio, ONT, Illumina) from blood-derived DNA. Large kinship pedigrees allow us to unambiguously determine the genetic inheritance of the parental haplotypes. Filtering genotypes with the genetic inheritance provides a means to adjudicate variant accuracy for all the sequencing technologies in our study. Combining these ‘truth’ callsets from three distinct technologies creates a comprehensive truthset that is not affected by the limitations of any individual datatype.
Results: Our approach yielded a truthset containing over six million SNPs and indels, an 18% increase over the NA12878 Genome in a Bottle dataset. Benchmarking revealed HiFi-based variant calls exhibit the highest recall for SNVs and INDELs (0.985/0.952), compared to ONT (0.969/0.733) and Illumina (0.935/0.914). Expanding beyond small variants, we identified 327,226 non-reference tandem repeats and >29,000 structural variants that were inherited. Importantly, we increased the rate of de novo SNVs, STRs and SVs by more than 25% in previously inaccessible regions of the genome.
Conclusion: Combining technologies and the inheritance from a 28-member pedigree we built a much more comprehensive benchmarking dataset that will be a valuable resource for tool developers and geneticists alike.
Grants:
Conflict of Interest: Zev Kronenberg PacBio, and Phase Genomics, PacBio, Cillian Nolan PacBio, PacBio, David Porubsky: None declared, Katy Munson: None declared, William Rowell PacBio, PacBio, Michelle Noyes: None declared, Brent Pedersen: None declared, Sean Mcgee: None declared, Josh Smith: None declared, Cairbre Fanslow PacBio, PacBio, primo baybayan PacBio, PacBio, Nidhi Koundinya: None declared, William Harvey: None declared, Kendra Hoekzema: None declared, Jordan Knuth: None declared, Gage Garcia: None declared, Tom Mokveld PacBio, PacBio, Egor Dolzhenko PacBio, PacBio, Scott Watkins: None declared, Deborah Neklason: None declared, Aaron Quinlan: None declared, Lynn Jorde: None declared, Evan Eichler SAB: VariantBio, Michael Eberle PacBio, PacBio
C30 Complexity of brain phenotypes
C30.1 A unified genome constraint, pathogenicity, and pLoF model identifies new genes associated with epilepsy, autism, and schizophrenia
Oscar Aguilar 1, Manuel Rivas2
1Stanford University, Management Science & Engineering, Stanford, United States; 2Stanford University, Biomedical Data Science, Stanford, United States
Background/Objectives: Highly heterogeneous disorders like epilepsy, autism, and schizophrenia are thought to have strong genetic components. However, identifying these risk factors using whole-exome sequencing studies requires very large sample sizes and good signal-to-noise ratio in order to assess the association between rare variants in any given gene and disease.
Methods: We present a novel approach for predicting constraint in the human genome – sections of the genome where any mutation can cause a severe disorder. Through application of a Hidden Markov Model (HMM) to the Regeneron Genetics Center Million Exome dataset and the AllofUs whole genome sequencing data, we predict the probability of observing no variants across the population for each position in the genome. Next, we aggregate the constraint predictions by gene and assess its association to epilepsy, schizophrenia, and autism, and compare against published results. Finally, we extend our analysis model to incorporate pathogenicity predictions from AlphaMissense (AM) and pLoFs.
Results: We identified a set of (p < 1x10-4) genes with stronger signals than previously published studies including KDM5B, KCNQ2, CACNA1A, CACNA1B, RYR2, and ATP2B2. Our models allow us to evaluate the contribution of constraint, protein structure based pathogenicity prediction from AM, and pLoFs jointly.
Conclusion: We showed that relatively simple sequence-dependent constraint prediction models can complement structure-based missense variant pathogenicity predictions and pLoFs for population cohort studies which require additional statistical power in the identification of gene-based signals for neurogenetic and psychiatric disorders. Finally, we explore the application of transformers to incorporate other features for constraint prediction.
Conflict of Interest: Oscar Aguilar Some of the computing for this project was performed on the Sherlock cluster., Graduate research assistant for Stanford University., Manuel Rivas Co Founder of Broadwing Bio, Some of the computing for this project was performed on the Sherlock cluster., M.A.R. is in part supported by National Human Genome Research Institute (NHGRI) under award R01HG010140, and by the National Institutes of Mental Health (NIMH) under award R01MH124244 both of the National Institutes of Health (NIH), Stanford University, Consultant for Curie.Bio and Mubadala Ventures
C30.2 Unlocking the genetic code of cerebral palsy: exploring clinical and genetic insights in modern era
Tracy Lau 1, charles steward2, Jozef Gecz3, david rowitch4, Jana Vandrovcova1, Reza Maroofian1, Henry Houlden1
1UCL Queen Square Institute of Neurology, Department of Neuromuscular Diseases, London, United Kingdom; 2Genomics England, London, United Kingdom; 3Adelaide Medical School, The University of Adelaide, Adelaide, Australia; 4University of Cambridge, Department of Paediatrics, Cambridge, United Kingdom
Consortium: Genomics England Research Consortium
Queen Square Genomics
Background/Objectives: Cerebral palsy (CP), characterised by non-progressive movement impairments and co-morbidities, was traditionally linked to perinatal environmental risk factors. Recent evidence suggests a genetic component, prompting an investigation into the clinical and genetic aspect of individuals with CP.
Methods: Screened the 100,000 Genomes Project data (100KGP, n = 90,178) and the UCL Queen Square Genomics (QSG, n = ~43,000) database to identify individuals with confirmed or suspected CP. Clinical and genetic data were collected and analysed. Unsolved cases with available exome/genome sequencing data underwent analysis using bioinformatic filtering pipelines.
Results: A total of 2121 individuals (100KGP-CP, n = 1464; QSG-CP, n = 657) were identified, with ~43.9% displaying at least one recorded motor subtype phenotype (spastic/spastic-related: ~21.3%; dyskinetic/dyskinetic-related: ~15.8%; ataxic/ataxic-related: ~6.9%). Neurodevelopmental features like intellectual disability (~41.3%), developmental delay (~39.9%) and seizures/epilepsy (~36.7%) were commonly observed. The diagnostic yield was ~35.9% with around 400 unique genes involved. Most reported in the 100KGP cohort were genes associated with autosomal dominant disorders (CTNNB1, SCN1A, KCNQ2, COL4A1, GNB1 and GRIN1), whereas in the QSG cohort, it was genes associated with autosomal recessive/X-linked disorders (ALDH18A1, VLDLR, SLC16A2, WDR62, EPRS, PIGT and AMPD2). Reanalysis of unsolved cases uncovered both known and potential novel genes implicated in CP.
Conclusion: This study highlights the diverse clinical and genetic aspects of CP, revealing its overlap with rare neurodevelopmental and movement disorders. In addition, it underlines the importance of reanalysing unsolved cases, given the ongoing gene discovery and updated clinical description in the context of CP.
Grant References: Research Foundation, Cerebral Palsy Alliance PhD Research Grant.
Conflict of Interest: None declared
C30.3 The contribution of common and rare exonic variation to cognitive performance across development
Daniel Malawsky 1, Mahmoud Koko1, Wei Huang1, Matthew Hurles1, Hilary Martin1
1Wellcome Sanger Institute, Hinxton, United Kingdom
Background/Objectives: Twin and SNP-based studies suggest the heritability of cognitive performance increases across childhood. However, the contribution of rare variants to cognitive performance has been relatively understudied. Here, we investigated the contribution of common and rare exonic variants to cognitive performance in >6,000 unrelated individuals from the Avon Longitudinal Study of Parents and Children.
Methods: We calculated polygenic indices (PGIs) and deleterious rare variant burden (DRVB), a measure of exome-wide burden of damaging variants that incorporates genic constraint, using SNP array and WES data. Mixed-effects linear modeling was used to assess associations with cognitive performance measured at ages 4, 8, and 16.
Results: DRVB was negatively associated with cognitive performance across all ages (p < 10-8). We found the variance explained by DRVB decreased with age including after controlling for parental DRVB (p < 10-5), suggesting that this was due to changes in direct genetic effects. Using gene expression data, we calculated DRVB stratified by genes predominantly expressed in prenatal (prenatal-DRVB) versus postnatal brain (postnatal-DRVB), or not brain expressed (nonCNS-DRVB). In a conditional analysis, we found both prenatal- and postnatal-DRVB showed associations with cognitive performance across ages (p < 10-4) while nonCNS-DRVB had no significant effects. Fetal-DRVB recapitulated the decreasing effect with age (p < .001), whereas adult-DRVB showed no attenuation. In contrast, we found that educational attainment and cognitive performance PGIs were associated with cognitive performance across development and explained increasing variance with age (p < 10-10), which remained significant after controlling for parental PGIs.
Conclusion: These results reveal a dynamic genetic architecture of cognitive performance across childhood.
Grants: Wellcome Trust grant 220540/Z/20/A
Conflict of Interest: None declared
C30.4 Genomic landscape of Rett syndrome spectrum disorders: genome-wide testing and a multi-omics integrative approach for improved diagnostics
Clara Xiol 1;2, Guerau Fernandez3;4;5, Roger Prats2;6, Núria Brandi2;7, María Heredia2;6, Gonzalo Villanueva-Martin2;6, Silvia Vidal2;6, Dmitrii Smirnov8;9, Robert Kopajtich8;9, Paola Pacheco1;2, Carlota Ros1;2, Marta Tejedor1;2, David De las Heras1;2, Delia Yubero1;2, Laura Martí1;4, Antonio Federico Martínez-Monseny10, Didac Casas-Alba10, mar o’callaghan11, Àngels García-Cazorla5;11;12, Mercedes Pineda Marfa13, Holger Prokisch8;9, Alfonso Oyarzábal6;12, Judith Armstrong1;2;5
1Hospital Sant Joan de Déu, Genomics-NGS, Genetic and Molecular Medicine Department, Barcelona, Spain; 2Institut de Recerca Sant Joan de Déu, Genomics for the Diagnosis of Rare Diseases Lab, Barcelona, Spain; 3Hospital Sant Joan de Déu, Bioinformatics, Genetic and Molecular Medicine Department, Barcelona, Spain; 4Institut de Recerca Sant Joan de Déu, Neurogenetics and Molecular Medicine, Barcelona, Spain; 5Instituto de Salud Carlos III (ICSIII), CIBER-ER (Biomedical Network Research Center for Rare Diseases), Madrid, Spain; 6Fundació de Recerca Sant Joan de Déu, Barcelona, Spain; 7Universitat de Barcelona, Barcelona, Spain; 8Technical University of Munich, Institute of Human Genetics, Munich, Germany; 9Helmholtz Zentrum München, Institute of Neurogenomics, Munich, Germany; 10Hospital Sant Joan de Déu, Clinical Genetics, Genetic and Molecular Medicine Department and Pediatric Institute of Rare Diseases, Barcelona, Spain; 11Hospital Sant Joan de Déu, Neurology Department, Barcelona, Spain; 12Institut de Recerca Sant Joan de Déu, Synaptic Metabolism Lab, Barcelona, Spain; 13Institut de Recerca Sant Joan de Déu, Barcelona, Spain
Background/Objectives: Rett syndrome (RTT) spectrum disorders are a cluster of neurodevelopmental disorders characterised by overlapping phenotypes and diverse genetic aetiologies. We have analysed the outcomes of various NGS genomic tests and implemented a multi-omics approach, integrating transcriptomics and proteomics, to maximise diagnostic yield.
Methods: We recruited 81 patients meeting clinical criteria for RTT with no pathogenic variants in MECP2, CDKL5 and FOXG1. They underwent sequential analysis using clinical exome sequencing (CES), whole exome sequencing (WES), and whole genome sequencing (WGS) coupled with gene expression analysis via RNAseq and proteomics in primary fibroblast cell cultures.
Results: 39 patients (48%) presented pathogenic or likely pathogenic variants in genes associated with RTT-spectrum disorders, while 17 (20%) had candidate variants of unknown significance. Molecular diagnoses were achieved in 24% through CES, 20% through WES, and an additional 4% through the implementation of WGS coupled with multi-omics. Variants were reported in 44 different genes, with only SYNGAP1, TCF4, SHANK3 and DNMT3A harbouring disease-causing variants in more than one patient. Improved coverage in WGS facilitated the detection of previously missed variants, while transcriptomics aided in the biological interpretation and reclassification of variants identified via WGS.
Conclusion: RTT-spectrum disorders, with overlapping phenotypes and heterogeneous genetic aetiologies, require comprehensive genetic testing for efficient genetic diagnosis. An integrated multi-omics approach enhances diagnostic yield, streamlining the process of variant prioritisation and interpretation. Deep phenotyping by expert clinicians in genetic syndromes could facilitate genotype-phenotype correlations.
Grants: FPU18/02152, PI20/0289, PI/23/00108
Conflict of Interest: None declared
C30.5 Rare variant analysis in Epileptic Encephalopathies highlights role for 3′UTR variation (virtual)
Ifeolutembi Fashina 1;2;3, Genomics England Research Consortium4, Simon J. Furney5, Gianpiero Cavalleri1;6
1Royal College of Surgeons in Ireland, School of Pharmacy and Biomolecular Sciences, Ireland; 2University of Galway, Centre for Research Training in Genomic Data Science, Galway, Ireland; 3Royal College of Surgeons in Ireland, Futureneuro, Ireland; 4Genomics England, United Kingdom; 5Royal College of Surgeons in Ireland, Genomic Oncology Research Group, Department of Physiology & Medical Physics, Ireland; 6Royal College of Surgeons in Ireland, FutureNeuro, Ireland
Background/Objectives: The epileptic encephalopathies (EEs) are rare devastating forms of early-onset epilepsy with a largely monogenic cause. Other early-onset epilepsies are often co-morbid with neurodevelopmental disorders (NDDs). Most of the known diagnostic variants are de novo, within coding regions of the genome, but majority of cases return negative diagnosis. New evidence shows that ultra-rare coding variants are enriched in EE cases. However, the role of URVs within non-coding elements of the genome is understudied in the epilepsies. microRNAs are known to be dysregulated in epilepsy. Using whole genome sequencing (WGS) data, we tested the hypothesis that rare microRNA-associated variation contributes to genetic risk for the EEs and epilepsies with NDD features
Methods: We assessed rare variant enrichment in 3,040 individuals (1,807 cases and 1,233 controls) through the 100,000 Genomes Project. We ran gene-set burden and collapsing analysis on ultra-rare variants (URVs) across microRNA-encoding genes and 3 functional domains within 540 epilepsy-associated genes, including conserved 3’UTRs, predicted microRNA-binding sites within 3’UTRs, and the coding sequence (CDS).
Results: We found significant enrichment of URVs within conserved 3’UTRs in people affected by epilepsy with NDD features compared to controls. We also observed this enrichment in the smaller cohort with EE. In addition, we replicated findings on the enrichment of damaging URVs within the CDS.
Conclusion: Using WGS data, we have shown for the first time, that there is an enrichment of rare variants outside protein-coding regions in EE and epilepsy with NDD. The 3’UTR URVs of interest potentially influence polyadenylation and microRNA-binding and are being characterized.
Grants: 18/CRT/214.
Conflict of Interest: None declared
C30.6 Shared genetic links between sleep, neurodevelopmental, and neuropsychiatric conditions: a genome-wide and pathway-based polygenic score analysis
Laura Fahey 1, Cathy Wyse1, Lorna Lopez1
1Maynooth University, Department of Biology, Maynooth, County Kildare, Ireland
Background/Objectives: It has been postulated that circadian dysfunction may contribute to the sleep problems prevalent in neurodevelopmental and neuropsychiatric conditions. Genetic correlations between numerous neurodevelopmental/neuropsychiatric and sleep phenotypes have been observed. We hypothesize that this overlapping genetic variation is enriched in certain biological pathways.
Methods: We used polygenic score analysis to confirm previously reported genome-wide correlations and identify enriched pathways. We created polygenic scores using summary statistics from GWAS of ADHD, autism, bipolar disorder (BP), schizophrenia. We tested the performance of these polygenic scores in predicting chronotype and insomnia status of UK Biobank participants. For the pathway-based polygenic scores, we restricted genetic variation to SNPs that mapped to genes within 451 biological pathways.
Results: Genome-wide polygenic scores for autism, BP and schizophrenia were associated with an evening chronotype, while polygenic scores for ADHD, autism, BP, schizophrenia were associated with insomnia status. Pathway-based polygenic score analysis identified the NRF2 KEAP1 and mRNA splicing minor pathways as being enriched for genetic variation overlapping between chronotype and BP.
Conclusion: These results demonstrate that the overlapping genetic variation between chronotype and BP is enriched in genes involved in the NRF2-KEAP1 and mRNA splicing minor pathways. Previous studies have linked the NRF pathway to the pathology of BP and schizophrenia. Additionally, NRF2 and splicing components have been reported to be rhythmically regulated by circadian clock genes. Our results suggest that these pathways could be involved in mediating disrupted circadian rhythms in BP.
Grants: European Research Council under the European Union’s Horizon 2020 research and innovation programme (grant agreement No 950010).
Conflict of Interest: None declared
C31 Somatic drivers in cancer
C31.1 Structural and functional insights from single cell transcriptional profiles of pituitary tumors
Maxime Brunner 1, Federico Santoni1, Mahmoud Messerer2, Roy Daniel2, Jenny Meylan-Merlini1, Maude Muriset3, Ekkehard Hewer3
1Lausanne University Hospital, EDM, Lausanne, Switzerland; 2Lausanne University Hospital, Neurosurgery, Lausanne, Switzerland; 3Lausanne University Hospital, IUP, Lausanne, Switzerland
Background/Objectives: Pituitary Neuroendocrine Tumors (PitNETs) are frequent in the population (15%) and are almost exclusively benign. Their pathoetiology is still unknown as well as the PITNET microenvironment (PTME).
Methods: We performed WGS and single cell transcriptome analysis on 12 independent tumors for a total of ~70’000 single cells: 4 bi-hormonal (GH-PRL) tumor, 3 corticotropic macro-adenoma, 4 non-secreting adenomas and one prolactinoma.
Results: Characterization of all tumors showed heterogeneous cell populations. We discover that GATA2, a transcription factor for early gonadotrophs differentiation is exclusively expressed in non-secreting adenoma. GATA2 inhibits GNRH receptor, leading to a decrease in LHB and FSHB production possibly explaining the non-secreting phenotype. Furthermore, GATA2 expression profile in single-nuclei RNAseq data from healthy pituitary glands shows restricted transcription in prepubescent individuals suggesting a come-back to a pre-differentiated state for gonadotroph cells in non-secreting adenomas. Next, integration of our tumors with scRNAseq data of adult healthy pituitary gland highlighted the presence of two subtypes of S100B positive/negative stem-like cells which could indicate future relapse. DGEA identified different typical tumor-upregulated pathways such as increased oxidative phosphorylation, Myc targets, E2F targets and DNA repair. To understand PTME, we investigate the exclusive tumoral expression of surface protein and we found promising candidates expressed in tumor endothelial cells.
Conclusion: Taken together, our results give a new perspective on the comprehension of the structural composition and the dynamic progression of pituitary tumors and of adenomas in general and identify new potential gene targets for immunotherapy.
Grants:
Conflict of Interest: None declared
C31.2 Constitutional disease-causing variants of the PTEN-gene are common in patients with papillary tumours of the pineal region, group B
Steffen Hirsch 1;2;3, Ramin Rahmanzade4;5, Kerstin Grund1, Kathrin Schramm3;6, Florian Selt2;3;7, Jonas Ecker2;3;7, Barbara Jones3;6;7, Dominik Sturm2;3;7, Christian Sutter1, Katrin Hinderhofer1, Nicola Dikow1, Maja Hempel1, David Jones3;6, Till Milde2;3;7, Kristian Pajtler2;3;7, Stefan Pfister2;3;7, Felix Sahm4;5, Christian Schaaf1
1Heidelberg University Hospital, Institute of Human Genetics, Heidelberg, Germany; 2German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Clinical Cooperation Unit Pediatric Oncology, Heidelberg, Germany; 3Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany; 4Heidelberg University Hospital, Department of Neuropathology, Institute of Pathology, Heidelberg, Germany; 5German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Clinical Cooperation Unit Neuropathology, Heidelberg, Germany; 6German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Pediatric Glioma Research Group, Heidelberg, Germany; 7Heidelberg University Hospital, Department of Pediatric Oncology, Hematology, Immunology & Pulmonology, Heidelberg, Germany
Background/Objectives: Papillary tumours of the pineal region, group B (PTPR(B)) are rare neuroepithelial CNS malignancies. Somatic deletions of the PTEN gene on chromosome 10 have previously been described in PTPR(B), but PTPR(B) have not been associated with PTEN hamartoma tumour syndrome. Here, we investigate the prevalence of constitutional PTEN alterations in patients with PTPR(B).
Methods: We searched for individuals with PTPR(B) in four cancer patient registry cohorts: MNP2.0 (n = 1,034), MNP Int-R (n = 806), PTT (n = 582), INFORM (n = 2572). All patients in these cohorts have been investigated by a multi-omics approach with tumour DNA-methylation (EPIC-Array) and paired tumour/normal sequencing using a multi-gene panel (MNP2.0, MNP Int-R, PTT) or whole exome/ low coverage whole genome (INFORM). Tumour copy number profiles were generated from raw DNA-methylation data. Germline sequencing data were analysed using a virtual panel of known cancer predisposition genes.
Results: We identified 10 patients with PTPR(B) (age 0-31years). All tumours showed heterozygous deletion of chromosome 10. This was the only copy number alteration in four tumours, while six tumours had additional CNV alterations. Germline sequencing revealed a constitutional heterozygous pathogenic or likely pathogenic (P/LP) PTEN variant in four cases. Germline variants were limited to the cases with isolated deletion of chromosome 10 in the tumour sample.
Conclusion: Constitutional PTEN P/LP variants are common among patients with PTPR(B) and germline testing should be considered in all patients with PTPR(B). Isolated deletion of chromosome 10 seems to be indicative of a relevant PTEN constitutional variant although patient numbers are too small to draw definitive conclusions.
Grants:
Conflict of Interest: None declared
C31.3 The spectrum of SMARCB1-deficient tumors extends to T-cell lymphomas enriched in children and young adults
Anja Fischer 1;2, Natalia Moreno2;3, Marta Interlandi2;3;4, Thomas K. Albert2;3, Jana Mormann3, Selina Glaser1, Archana Verma3, Paurnima Patil1, Sebastian T. Balbach4, Rabea Wagener1, Susanne Bens1, Sonja Dahlum1, Carolin Göbel5;6, Monika Graf3, Eva Kremer3, Flavia W. de Faria3, Viktoria Melcher3, Gioia Di Stefano7, Raffaella Santi7, Alexander Chan8, Ahmet Dogan8, Jonathan Bush9, Martin Hasselblatt10, Sylvia Cheng11, Signe Spetalen12, Alexander Fosså13, Wolfgang Hartmann14, Heidi Herbrüggen3, Stella Robert15, Martin Dugas4;16, Carolin Walter4, Clara Inserte4, Sarah Sandmann4, Julian Varghese4, Claudia Rössig3, Ulrich Schüller5;6;17, Alexandar Tzankov18, Martin Bjerregård Pedersen19, Francesco Annibale d’Amore19;20, Karin Mellgren21, Udo Kontny22, Venkatesh Kancherla23, Luiz Veloza23, Edoardo Missiaglia23, Virginie Fataccioli24;25, Philippe Gaulard24;25, Birgit Burkhardt3, Wolfram Klapper26, Laurence de Leval23, Reiner Siebert1;27, Kornelius Kerl3;27
1Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany; 2These authors contributed equally; 3Department of Pediatric Hematology and Oncology, University Children’s Hospital Münster, Münster, Germany; 4Institute of Medical Informatics, University of Münster, Münster, Germany; 5Department of Pediatric Hematology and Oncology, University Medical Center Hamburg, Hamburg, Germany; 6Research Institute Children’s Cancer Center, Hamburg, Germany; 7Pathological Anatomy Section, Careggi University Hospital, Florence, Italy; 8Department of Pathology, Hematopathology Service, Memorial Sloan Kettering Cancer Center, New York, United States; 9Division of Anatomical Pathology, British Columbia Children’s Hospital and Women’s Hospital and Health Center, Vancouver, Canada; 10Institute of Neuropathology, University Hospital Münster, Münster, Germany; 11Division of Pediatric Hematology/Oncology/BMT, Department of Pediatrics, British Columbia Children’s Hospital, Vancouver, Canada; 12Department of Pathology, Oslo University Hospital, Oslo, Norway; 13Department of Oncology, Oslo University Hospital-Norwegian Radium Hospital, Oslo, Norway; 14Division of Translational Pathology, Gerhard-Domagk-Institut für Pathologie, Universitätsklinikum Münster, Münster, Germany; 15Department of Medicine A, Hematology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany; 16Institute of Medical Informatics, Heidelberg University Hospital, Heidelberg, Germany; 17Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; 18Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland; 19Department of Hematology, Aarhus University Hospital, Aarhus, Denmark; 20Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; 21Department of Pediatric Oncology and Hematology, Sahlgrenska University Hospital, The Queen Silvia Children’s Hospital, Gothenburg, Sweden; 22Section of Pediatric Hematology, Oncology, and Stem Cell Transplantation, Department of Pediatric and Adolescent Medicine, RWTH Aachen University Hospital, Aachen, Germany; 23Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland; 24INSERM U955, Université Paris-Est, Créteil, France; 25Département de Pathologie, Hôpitaux Universitaires Henri Mondor, Créteil, France; 26Department of Pathology, Haematopathology Section and Lymph Node Registry, University of Kiel/University Hospital Schleswig-Holstein, Kiel, Germany; 27These authors jointly supervised this work
Background/Objectives: SMARCB1 is a bona fide tumor suppressor gene associated with the development of tumors classically including malignant rhabdoid tumors and schwannomas, but has also been shown to be involved in lymphocyte development. Peripheral T-cell lymphoma, not otherwise specified (PTCL-NOS) is a heterogeneous group of malignancies with poor outcome. So far, the frequency of SMARCB1 alterations in T-cell lymphomas remains unclear.
Methods: SMARCB1 (INI1) gene and protein expression was assessed in 281 patients with diverse mature T-cell lymphomas by array-based expression profiling and immunohistochemistry. Additionally, the DNA methylome and single-cell transcriptome were analyzed in SMARCB1-negative PTCL-NOS patient samples and corresponding murine T-cell lymphomas using the Infinium BeadChips and 10X Genomics, respectively.
Results: SMARCB1 expression was heterogeneous between and within different human mature T-cell lymphoma entities. Loss of SMARCB1 protein expression in PTCL-NOS patients correlated with young age of the patients. Molecular characterization and DNA methylation analysis revealed that loss of SMARCB1 expression in human PTCL largely occurs via somatic mutation and/or epigenetic silencing. In contrast to malignant rhabdoid tumors, germline SMARCB1 mutations have not yet been observed. scRNA-seq analyses in human and murine SMARCB1-deficient cases revealed an immunosuppressive and pro-inflammatory tumor microenvironment. Treatment of tumor-bearing mice with a pan-HDAC inhibitor promoted replenishment of lymphoid compartments.
Conclusion: Our findings suggest that SMARCB1 mutations are present in a substantial proportion of PTCL-NOS and are especially encountered in children and young adults. The role of germline SMARCB1 variants in T-cell lymphomas needs to be further investigated.
Grants: Deutsche Krebshilfe, TRANSCAN.2-ERA-NET(BMBF)
Conflict of Interest: None declared
C31.4 Somatic EPAS1 Gain-of-Function Pathogenic Variants Identified in Head and Neck Paragangliomas of Pacak-Zhuang Syndrome Patients
Yasemin Cole 1, Jared Rosenblum1, Danielle Dang1, Pashayar Lookian1, Hussam Alkaissi2, Anthony Cappadona1, Abhishek Jha3, Danielle Donahue4, Jeeva Munasinghe4, Herui Wang1, Russell Knutsen5, Alberto Pappo6, Ronald Lechan7, Beth Kozel5, James Smirniotopoulos8;9, Jeffrey Kim10;11, Alexander Vortmeyer12, Markku Miettinen13, John Heiss14, Zhengping Zhuang1, Karel Pacak2
1National Cancer Institute, Neuro-Oncology Branch, Bethesda, United States; 2National Institute of Diabetes and Digestive and Kidney Disease, Bethesda, United States; 3National Institute of Child Health and Human Development, Section on Medical Neuroendocrinology, Bethesda, United States; 4National Institute of Neurological Disorders and Stroke, Mouse Imaging Facility, Bethesday, United States; 5National Heart Lung and Blood Institute, Laboratory of Vascular and Matrix Genetics, Bethesda, United States; 6St Jude Children’s Research Hospital, Division of Solid Tumor,, Memphis, United States; 7Tufts Medical Center, Division of Endocrinology, Diabetes & Metabolism, Boston, United States; 8George Washington University, Radiology, Washington D.C.; 9MedPix, National Library of Medicine, Bethesda, United States; 10Georgetown University School of Medicine, Department of Otolaryngology, Washington D.C., United States; 11National Institute on Deafness and Other Communication Disorders, Office of Clinical Director, Bethesda, United States; 12Indiana University School of Medicine, Clinical Pathology & Laboratory Medicine, Indianapolis, United States; 13National Institutes of Health, General Surgical Pathology Section, Bethesda, United States; 14National Institute of Neurological Disorders and Stroke, Surgical Neurology Branch, Bethesda, United States
Background/Objectives: Head and neck paragangliomas (HNPGLs) are typically slow-growing, hormonally inactive tumours primarily associated with succinate dehydrogenase (SDHx) germline pathogenic variants. We recently discovered a syndrome characterized by paraganglioma/phaeochromocytoma, somatostatinoma, and polycythaemia caused by somatic gain-of-function pathogenic variants in hypoxia-inducible factor-2α (HIF2A, encoded by EPAS1). Recent studies of variants in prolyl-hydroxylase domain-containing 2 (PHD2, encoded by EGLN1) have demonstrated that indirect gain-of-function of hypoxia signalling, mediated by HIF2A, is sufficient to cause hyperplasia of the carotid body. Thus, we hypothesized that EPAS1 gain-of-function pathogenic variants are sufficient for HNPGL formation.
Methods: We assessed 9 patients with EPAS1 gain-of-function variants for HNGPL using magnetic resonance imaging (MRI), positron emission tomography, and computed tomography (CT) and measured carotid body dimensions. We evaluated a corresponding mouse model using high-resolution ex vivo imaging and histopathology.
Results: Three patients had imaging findings consistent with HNPGLs, one of which was histologically confirmed. Three individuals had carotid body enlargement (Z-score >2.0) compared to literature reference values; Three others had carotid artery malformations. Assessment of the mouse model revealed head and neck lesions in adult and post-natal day 8 (P8) mice. We found carotid body tumours in adult mice (9/10) and cranio-caval vein masses in adult (6/8) and P8 mice (4/5). Tyrosine hydroxylase, synaptophysin and chromogranin A positivity of these resected lesions confirmed PGL.
Conclusion: These findings suggest that in addition to germline SDHx predisposition syndromes, somatic variants in EPAS1 are also sufficient to cause HNPGLs. Patients with HNPGLs without SDHx pathogenic variants should be screened for EPAS1 variants.
Grants: None
Conflict of Interest: None declared
C31.5 Identification of large-scale and focal copy number alterations to improve classification and risk stratification in childhood B-cell Acute Lymphoblastic Leukemia
Anastasija Jakjimovska1, Unai Illarregi1, Ander Diaz-Navarro2, Jorge Herrero1, ivan martinez de estibariz1, Idoia Martin-Guerrero1, Nerea Bilbao Aldaiturriaga3, daniel sinnett4;5, Angela Gutierrez-Camino 3
1University of the Basque Country, Department of Genetics, Physical Anthropology & Animal Physiology, Leioa, Spain; 2Ontario Institute for Cancer Research, Adaptative Oncology, Toronto, Canada; 3BioBizkaia Health Research Institute, Pediatric Oncology Group,, Barakaldo, Spain; 4CHU Sainte-Justine, Division of Hematology-Oncology, Montréal, Canada; 5UdeM, Department of Pediatrics, Faculty of Medicine, Montréal, Canada
Background/Objectives: Childhood B-cell acute lymphoblastic leukemia (B-ALL) is characterized by recurrent, prognostic genetic alterations. Recent reports already demonstrated the clinical utility of transcriptome sequencing (RNA-seq) as a diagnostic tool in B-ALL, since it allows the simultaneous identification of rearrangements, mutations and expression profiles. Copy number alterations (CNA) can be highly informative to guide disease classification and risk stratification in ALL, however, they are rarely addressed in bulk RNA-seq. Therefore, we aimed to explore the potential of large-scale and focal CNAs identification to improve B-ALL diagnosis.
Methods: Large-scale CNAs were inferred from RNA-seq datasets of 136 patients using RNAseqCNV and Superfreq algorithms. Focal CNAs of IKZF1, PAX5 and ERG were identified with Toblerone. Resulting CNAs were compared to clinical karyotypes, FISH and Whole Exome Sequencing (WES) data.
Results: Large-scale CNAs identified from RNA-seq resulted in a concordance of 88,9% with clinical data, and 98.2% with WES. Importantly, two patients firstly assigned to the hyperdiploid subtype were reclassified as hypodiploid ALL. Additionally, twelve patients presented a loss of 9p and two patients a loss of 7p, involving PAX5 and IKZF1 locus, respectively. Focal deletions in IKZF1, PAX5 and ERG were identified in 16/20 (80%), 10/12 (83.3%) and 2/3 (66,7%) patients, respectively. The combination of tools helped identify three patients with IKZF1plus profile, a new very-poor prognostic group in B-ALL that requires treatment intensification.
Conclusion: Adding CNA information to bioinformatic pipelines improves classification and risk stratification in B-ALL.
Grants: Health Department of the Basque Government (2022111010), Fundación Vasca de Innovación e Investigación Sanitarias (BIO20/CI/016/BIOEF)
Conflict of Interest: None declared
C31.6 Characterization of the mutational landscape of colorectal tumors from individuals with adenomatous- or serrated polyposis
Anna Sommer 1, Iris te Paske2, Erik Jansen2, Axel Gschwind3, german demidov3, Oliver Hommerding4, Dimo Dietrich5, Wouter Steyaert2, Burcu Yaldiz2, Sophia Peters1, Marloes Steehouwer2, Glen Kristiansen4, Isabel Spier1;6;7, Iris Nagtegaal8, Hendrik Bläker9, Alexander Hoischen2;10, Christian Gilissen2, Rachel van der Post8, Laura Valle7;11;12, Elke Holinski-Feder7;13;14, Verena Steinke-Lange7;13;14, Nicoline Hoogerbrugge2;7, Stephan Ossowski3, Marjolijn Ligtenberg2;7;8, Stefan Aretz1;6;7, Richarda de Voer2;7
1Institute of Human Genetics, Medical Faculty, University of Bonn, Bonn, Germany; 2Department of Human Genetics, Radboud Research Institute for Medical Innovation, Radboud university medical center, Nijmegen, Netherlands; 3Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany; 4Institute of Pathology, Medical Faculty, University of Bonn, Bonn, Germany; 5Department of Otolaryngology, Head and Neck Surgery, Medical Faculty, University of Bonn, Bonn, Germany; 6Center for Hereditary Tumour Syndromes, University Hospital Bonn, Bonn, Germany; 7European Reference Network on Genetic Tumour Risk Syndromes (ERN GENTURIS) – Project ID No 739547; 8Department of Pathology, Radboud Research Institute for Medical Innovation, Radboud university medical center, Nijmegen, Netherlands; 9Institute of Pathology, University of Leipzig, Leipzig, Germany; 10Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud Research Institute for Medical Innovation, Radboud university medical center, Nijmegen, Netherlands; 11Hereditary Cancer Program, Catalan Institute of Oncology, Oncobell Program, IDIBELL, Hospitalet de Llobregat, Barcelona, Spain; 12Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; 13MGZ - Medizinisch Genetisches Zentrum, Munich, Germany; 14Medizinische Klinik und Poliklinik IV – Campus Innenstadt, Klinikum der Universität München, Munich, Germany
Background/Objectives: Germline pathogenic variants in known genes are identified in about 50% of adenomatous polyposis (AP) and a minority of serrated polyposis (SP) cases. Within the EU project Solve-RD (No.779257) the ERN GENTURIS performed a molecular characterization of colorectal tumors from unsolved AP/SP individuals to extend biological insights and identify potential clues for underlying germline causes.
Methods: Whole-exome sequencing was performed on DNA from 215 formalin-fixed paraffin-embedded gastrointestinal tumors (115 adenomas; 81 serrated polyps; 19 carcinomas) from 124 AP/SP patients. Somatic variants were called and annotated using an in-house pipeline. Tumor mutational burden (TMB) and mutational signatures were analyzed and compared to control datasets. Driver gene analysis was based on the COSMIC Cancer-Gene-Census list (n = 736). SP tumors were analyzed for CpG-island-methylator-phenotype (CIMP).
Results: Overall, 99% of tumors had low TMB (<10 mut/Mb) and were microsatellite-stable. APC/CTNNB1 and BRAF mutations were almost exclusively present in AP (86%) and SP (81%) tumors, respectively. APC mosaicism was found in 30% (11/37) of AP patients (≥2 sequenced tumors). Further recurrent (likely) pathogenic somatic mutations were identified in AKAP9, AMER1, BRCA2, KMT2C, LRP1B, and THRAP3. In 30% of SP tumors CIMP-high was detected. APC/CTNNB1-mutated tumors presented with a low contribution of the clock-like signature SBS1 and BRAF-mutated tumors presented with a high contribution of the normal colon tissue-associated signature SBS89.
Conclusion: Distinct (potential) driver genes characterize tumors in AP/SP individuals. Signature analysis suggests that BRAF-mutated tumors are clonal outgrowths of normal colon tissue. Overall, molecular tumor profiling provides further insights into mechanisms of polyp tumorigenesis.
Conflict of Interest: None declared
C32 New syndromes in 2024
C32.1 Biallelic MED16 variants cause a novel MEDopathy with intellectual disability, motor delay and craniofacial, cardiac and limb malformations
Charlotte Guillouet 1, Valeria Agostini1, Genevieve Baujat2, Tommaso Pippucci3, Marion Lesieur-Sebellin1, Mathieu Georget1, Ulrich Schatz4, Christine Fauth4, Raymond Louie5, Curtis Rogers5, Jessica M Davis5, Vassiliki Konstantopoulou6, Hans A. Mayr7, Arjan Bouman8, Kristen Park9, Grace E VanNoy10, Eleina England10, Kathleen Brown11, Margarita Saenz11, Rauan Kaiyrzhanov12, Sughra Guliyeva13, Lala Hasanova14, Deborah Shears15, Ishita Bhatnagar15, Karen Stals16, Oliver Klaas17, Judit Horvath17, Dane Witmer18, Gretchen MacCarrick18, Katarina Vizar Cisarova19, Jean-Marc Good19, Svetlana Gorokhova20, Tiffany Busa20, Odile Boute21, Thomas Smol21, Ange-Line Bruel22, Olivier Patat23, Julia R Broadbent24, Tiong Yang Tan25, Natalie Tan26, Claudio Graziano27, Jeanne Amiel1, Chris Gordon1
1Laboratoire d’Embryologie et Génétique des Malformations, INSERM UMR 1163, Institut Imagine, Université Paris Cité, Paris, France; 2Service de Génétique Médicale, Hôpital Necker-Enfants Malades, AP-HP, Paris, France; 3IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; 4Institute of Human Genetics, Medical University Innsbruck, Innsbruck, Austria; 5Greenwood Genetic Center, Greenwood, United States; 6Department of Pediatrics and Adolescent Medicine, Austrian Newborn Screening, Medical University of Vienna, Vienna, Austria; 7University Children’s Hospital, Salzburger Landeskliniken (SALK), Paracelsus Medical University,, Salzburg, Austria; 8Department of Clinical Genetics, Erasmus University Medical Center,, Rotterdam, Netherlands; 9University of Colorado School of Medicine, Departments of Pediatrics and Neurology, Aurora, United States; 10Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, United States; 11University of Colorado Anschutz Medical Campus, Aurora, United States; 12Department of Neuromuscular diseases, UCL Institute of Neurology,Queen Square, London, United Kingdom; 13MediClub Hospital, Baku, Azerbaijan; 14Caspian International Hospital, Baku, Azerbaijan; 15Oxford Centre for Genomic Medicine, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom; 16Exeter Genomics Laboratory, Royal Devon University Healthcare NHS Foundation Trust, Exeter, United Kingdom; 17Institute for Human Genetics, University Hospital Muenster, Muenster, Germany; 18McKusick-Nathans Department of Genetic Medicine, School of Medicine, Johns Hopkins University, Batimore, United States; 19Division of Genetic Medicine, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland; 20Department of Medical Genetics, Timone Children’s Hospital, AP-HM, Marseille, France; 21Univ. Lille, CHU Lille, ULR 7364 - RADEME - Maladies RAres du DÉveloppement embryonnaire et du Métabolisme, Lille, France; 22Functional Unit of Innovative Diagnosis for Rare Diseases, Dijon Bourgogne University Hospital, Dijon, France; 23Department of Medical Genetics, CHU Toulouse Purpan, Toulouse, France; 24Rare Disease Discovery Group, Murdoch Children’s Research Institute, Melbourne, Australia; 25Department of Paediatrics, University of Melbourne, Melbourne, Australia; 26Victorian Clinical Genetics Services, Melbourne, Australia; 27U.O. Genetica Medica, AUSL della Romagna, Cesena, Italy
Background/Objectives: MED16 is a subunit of the evolutionary-conserved Mediator complex, which regulates protein-coding gene transcription by linking enhancers with RNA Polymerase II. Pathogenic variants in several Mediator subunits are responsible for MEDopathies, with variable clinical presentations and modes of inheritance. MED16 has not previously been implicated in human pathology.
Methods: Trio WES or WGS was performed on patients with undiagnosed multiple congenital anomalies and intellectual disability, with cohort creation facilitated by Genematcher. The effect of missense variants on subcellular localization was tested by transient transfection of MED16-encoding plasmids followed by immunofluorescence. Three-dimensional modelling of variants was performed in Chimera. Two mutant med16 zebrafish strains were generated by CRISPR-Cas9.
Results: Biallelic MED16 variants were identified in 22 individuals from 16 families. Intellectual disability and motor delay were present in all patients, and were associated in variable combinations with craniofacial malformations, limb anomalies and cardiac defects (predominantly tetralogy of Fallot). Growth defects, deafness, visual impairment and MRI abnormalities were also reported.
Six predicted protein-truncating and 17 missense or in-frame duplication variants were identified, with low reported frequencies in gnomAD and high scores for predicted pathogenicity (CADD, AlphaMissense). In silico modelling suggested an impact of missense variants on functional domains of the protein. Immunofluorescence assays show that 15/17 mutant proteins were delocalized to the cytoplasm. med16-/- zebrafish had reduced body length and increased mortality compared with wild-type and heterozygous fish.
Conclusion: We describe a novel autosomal recessive MEDopathy, emphasizing the importance of Mediator in neurodevelopment and suggesting tissue-specific roles for specific subunits.
Conflict of Interest: None declared
C32.2 FKBP4 is a critical developmental gene and its loss causes a recessive syndrome of global developmental delay with sexual differentiation disorder
Rebecca Yarwood 1;2, Annie Godwin3, Maha Zaki4, John McDermott1;5, Najmeh Ahangari6, Adila Al-Kindi7;8, Kristin Baranano9, Kate Chandler1;5, Siobhan Crilly10;11, Sara Cuvertino1;12, Antonios Frantzeskos1;10, Joseph Gleeson13;14, Jan Idkowiak15;16, Rebecca Igbokwe17, Ehsan Ghayoor Karimiani6;18, Inas Mazen4, Ken McElreavey19, Weiyi Mu20, Maryam Najafi21;22, Esma Secen21, Rolph Pfundt22, Hannah Robinson23, Mars Skae24;25, Vijay Tiwari26, Rolf Backofen27, Mehran Beiraghi Toosi28;29, Paria Najarzadeh Torbati30, Miriam Schmidts21;22;31, Martin Lowe2, Reza Maroofian32, Matthew Guille3, Paul R. Kasher10;11, Siddharth Banka1;5
1Division of Evolution, Infection & Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, UK; 2Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, UK; 3European Xenopus Resource Centre, School of Biological Sciences, University of Portsmouth, UK; 4Clinical Genetics and Endocrinology, Department of Clinical Genetics, National Research Center, Egypt; 5Manchester Centre for Genomic Medicine, St Mary’s Hospital, Manchester University NHS Foundation Trust, Health Innovation Manchester, UK; 6Innovative Medical Research Centre, Mashhad Branch, Islamic Azad University, Iran; 7Department of Genetics, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman; 8Genetic and Developmental Medicine Clinic, Sultan Qaboos University Hospital, Muscat, Oman; 9Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, USA; 10Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, UK; 11Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, UK; 12Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine, and Health, The University of Manchester, UK.; 13Department of Neurosciences, University of California, San Diego, USA; 14Rady Children’s Hospital, San Diego, USA; 15Department of Endocrinology and Diabetes, Birmingham Children’s Hospital, Birmingham Women’s and Children’s NHS Foundation Trust, UK; 16Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, UK; 17West Midlands Clinical Genetics, Birmingham Women’s Hospital, UK; 18Molecular and Clinical Sciences Institute, St. George’s, University of London, UK; 19Institut Pasteur, Université Paris Cité, CNRS UMR3738, Human Developmental Genetic Unit, France; 20Department of Genetic Medicine, John Hopkins School of Medicine, Baltimore, USA; 21Center for Pediatrics and Adolescent Medicine, University Hospital Freiburg, Freiburg University Faculty of Medicine, Germany; 22Department of Human Genetics, Radboud University Nijmegen Medical Centre, The Netherlands.; 23Exeter Genomics Laboratory, Royal Devon and Exeter Hospital, Royal Devon University Healthcare NHS Foundation Trust, UK; 24Department of Paediatric Endocrinology, Royal Manchester Children’s Hospital, UK; 25School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, UK; 26School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, UK; 27Bioinformatics Group, Department of Computer Science, University of Freiburg, Germany; 28Department of Pediatrics, School of Medicine, Mashhad University of Medical Sciences, Iran; 29Neuroscience Research Center, School of Medicine, Mashhad University of Medical Sciences, Iran; 30Department of Medical Genetics, Next Generation Genetic Polyclinic, Mashhad, Iran; 31CIBSS-Centre for Integrative Biological Signalling Studies, University of Freiburg, Germany; 32Department of Neuromuscular Disorders, Queen Square Institute of Neurology, University College London, UK
Background/Objectives: FKBP4 encodes an immunophilin required for nuclear translocation of steroid receptors and androgen receptor-mediated signalling. However, its role in development is poorly understood.
Methods: Genomic, clinical, and patient fibroblast studies were performed, scRNA-seq and RNA-Seq data from wild-type mouse and various iPSC models were analysed and a Xenopus tropicalis disease-model was generated.
Results: We identified homozygous FKBP4 truncating variants in 12 individuals with severe-profound developmental delay, craniofacial dysmorphism and variable microcephaly. External genitalia of all 46,XY individuals were ambiguous. Some affected individuals developed aortic dilatation. Patient fibroblasts showed absence of FKBP4 mRNA and protein.
Developing XY murine gonad scRNASeq showed Fkbp4 expression to be highest in the gonadal progenitors. Mouse neocortical cell scRNA-seq from E14.5 and P0 stages showed Fkbp4 expression to be highest in the neural progenitors. Human GW8-26 embryonic prefrontal cortex scRNA-seq showed in vivo FKBP4 expression to be highest in the GW12-13 ventricular zone radial glia cells. RNASeq performed at different stages of differentiation from iPSCs to cortical neurons showed that in vitro FKBP4 expression is highest at the stem cell stage.
Xenopus fkbp4 crispants showed significantly lower survival over the first 28 days of development, and reduced head size and movement recapitulating aspects of the human disorder. RNASeq of the Xenopus crispants is ongoing.
Conclusion: We have identified a clinically recognizable novel recessive FKBP4-related syndrome of global developmental delay and sexual differentiation disorder. This indicates that FKBP4 has critical roles in the early stages of development of the brain and external male genitalia.
Conflict of Interest: None declared
C32.3 Identification and functional analysis of three novel responsible genes for 3C/Ritscher-Schinzel syndrome
Kohji Kato 1;2, Aljazi Al-Maraghi3, Waleed Aamer3, Ajaz Bhat3, Hester Kroes4, Mohamad Abdelhamid5, Maha Zaki6, Kirsty McMillan1, Kate Heesom7, Philip Lewis7, Yuka Murofushi2, Yosuke Nishio2, Ammira Akil3, Kazi Mehrin8, Hisashi Oishi8, Khalid Fakhro3, Shinji Saitoh2, Peter Cullen1
1University of Bristol, School of Biochemistry, United Kingdom; 2Nagoya City University Graduate School of Medical Sciences, Department of Pediatrics and Neonatology, Japan; 3Sidra Medicine, Department of Human Genetics, Qatar; 4UMC Utrecht, Department of Genetics, Netherlands; 5Human Genetics and Genome Research Institute, National Research Centre, Medical Molecular Genetics Department, Egypt; 6Human Genetics and Genome Research Institute, National Research Centre, Clinical Genetics Department, Egypt; 7University of Bristol, Proteomics facility, School of Biochemistry,, United Kingdom; 8Nagoya City University Graduate School of Medical Sciences, Department of Comparative and Experimental Medicine, Japan
Background/Objectives: 3C/Ritscher-Schinzel syndrome (3C/RSS) is a rare congenital malformation syndrome characterized by craniofacial-cerebellar-cardiac defects. Four responsible genes, WASHC5, CCDC22, VPS35L, and DPYSL5 are listed in OMIM database. Except for DPYSL5, all gene are involved in the SNX17/Commander/WASH-related endosomal recycling network. However, the molecular mechanism and disease entity of 3C/RSS is not yet well understood.
Methods: We utilized GeneMatcher for patient recruitment with 3C/RSS-related clinical phenotypes. Cellular analysis was conducted to confirm pathogenicity of identified mutations and the molecular cause of 3C/RSS.
Results: Biallelic variants in the Commander subunits COMMD4, COMMD9, and CCDC93 were identified in patients with overlapping 3C/RSS phenotypes. All variants were confirmed as loss of function. Cellular models deficient in COMMD4, COMMD9, or CCDC93 mirrored the impaired SNX17-mediated cargo recycling observed in WASHC5-, CCDC22-, and VPS35L-deficient cells. Additionally, our literature review identified patients harboring VPS26C and WASHC4 mutations, additional subunits of SNX17/Commander/WASH pathway, with similar clinical phenotype of 3C/RSS. We confirmed that VPS26C-KO and WASHC4-KO also affected SNX17-mediated cargo recycling. Finally, DPYSL5-KO did not impact on this cargo recycling system and a review of clinical phenotypes suggested patients with DPYSL5 variants display different clinical features such as agenesis of corpus callosum.
Conclusion: COMMD4, COMMD9, and CCDC93 are identified as novel responsible genes for 3C/RSS. Furthermore, cellular and clinical analysis suggests mutations in VPS26C and WASHC4 also cause 3C/RSS. This suggests a unified pathogenic mechanism for 3C/RSS, likely rooted in disruptions of the SNX17/Commander/WASH endosomal recycling pathway. This research advances our understanding of 3C/RSS and opens avenues for targeted therapeutic strategies.
Grants:104568/Z/14/Z(WellcomeTrust)_MR/L007363/1(MRC)_RSRP/R1/211004(RoyalSocietyNoreenMurrayResearch).
Conflict of Interest: None declared
C32.4 De novo missense variants in EIF3I cause a neurodevelopmental disorder with dysmorphism, short stature, and midline brain defects.
Marcello Scala 1;2, Amelia Royce3;4, Jo Roobol3, Dong Li5, Jennifer Johnston6, Solveig Heide7, Boris Keren7, Ingrid MBH van de Laar8, Coranne Aarts-Tesselaar9, Louise Wilson10, Adeline Vanderver11, Kayla Muirhead11, Cas Simons11, Ryan Taft12, Kristin G. Monaghan13, Bobbi McGivern13, Julie Cohen14;15, Daniel Wegner16, Kathleen Sisco16, Jorge Luis Granadillo De Luque16, Federico Zara1;2, Valeria Capra17, Yaser Hashem18, Mark Smales3;4
1Università di Genova - Dipartimento di Neuroscienze, Riabilitazione, Oftalmologia, Genetica e Scienze Materno-Infantili, Genova, Italy; 2Giannina Gaslini Institute, Unit of Medical Genetics, Genova, Italy; 3University of Kent, School of Biosciences, Division of Natural Sciences, United Kingdom; 4University College Dublin, National Institute for Bioprocessing Research and Training, Ireland; 5Children’s Hospital of Philadelphia, Philadelphia, United States; 6National Institutes of Health, Center for Precision Health Research, Bethesda, United States; 7University Hospitals Pitié Salpêtrière - Charles Foix, Department of Genetics, Paris, France; 8Erasmus University Medical Center, Department of Clinical Genetics, Rotterdam, Netherlands; 9Amphia Hospital, Breda, Netherlands; 10Great Ormond Street Hospital for Children, United Kingdom; 11The Children’s Hospital Of Philadelphia, Division of Neurology, Philadelphia, United States; 12Illumina, San Diego, United States; 13GeneDx, Gaithersburg, United States; 14Kennedy Krieger Institute, Department of Neurology and Developmental Medicine, Baltimore, United States; 15The Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, United States; 16Washington University School of Medicine, Department of Pediatrics, St. Louis, United States; 17Giannina Gaslini Institute, Genomics and Clinical Genetics Unit, Genova, Italy; 18European Institute of Chemistry and Biology, Pessac, France
Background/Objectives: mRNA translation is a tightly regulated step in gene expression beginning with ribosome assembly, mRNAs recruitment, and subsequent scanning until the start-codon is identified. All stages of eukaryotic canonical translation initiation are tightly coordinated by eukaryotic initiation factors (eIFs). The largest and most complex eIF, eIF3, consists of 13 subunits (a-m) in humans. The i subunit (eIF3i), encoded by the EIF3I gene, is one of the most conserved and participates to the formation of initiation complexes and translation of specific mRNAs. Its overexpression causes an oncogenic phenotype with increased protein synthesis rates.
Methods: We performed exome sequencing to investigate the genetic etiology of overlapping neurodevelopmental phenotypes and brain abnormalities in a cohort of eight unrelated patients. We established a 3D model of EIF3I mutants to predict the consequences on protein structure and stable HEK-293 cell mutants to explore the biological properties of the variants. Co-immunoprecipitation assays were performed to investigate protein-protein interaction perturbances.
Results: Affected individuals showed neurodevelopmental impairment, behavioral abnormalities, short stature, eye and skeletal defects, and recognizable facial dysmorphism. Brain MRI revealed midline defects involving white matter and pituitary gland. Exome sequencing revealed 8 de novo EIF3I missense variants, all rare and affecting conserved residues within WD40 repeats. These variants disrupted protein folding in silico. EIF3I mutants displayed overgrowth phenotypes with decreased protein synthesis rate. Co-immunoprecipitation assays showed that EIF3I variants impair EIF3I interaction with its binding partners.
Conclusion: Our findings support de novo EIF3I variants as the cause of a novel recognizable neurodevelopmental condition.
Grants: none.
Conflict of Interest: Marcello Scala: None declared, Amelia Royce: None declared, Jo Roobol: None declared, Dong Li: None declared, Jennifer Johnston: None declared, Solveig Heide: None declared, Boris Keren: None declared, Ingrid MBH van de Laar: None declared, Coranne Aarts-Tesselaar: None declared, Louise Wilson: None declared, Adeline Vanderver: None declared, Kayla Muirhead: None declared, Cas Simons: None declared, Ryan Taft: None declared, Kristin G. Monaghan KGM and BM are employees of GeneDx, LLC, Bobbi McGivern KGM and BM are employees of GeneDx, LLC, Julie Cohen: None declared, Daniel Wegner: None declared, Kathleen Sisco: None declared, Jorge Luis Granadillo De Luque: None declared, Federico Zara: None declared, Valeria Capra: None declared, Yaser Hashem: None declared, Mark Smales: None declared
C32.5 Elucidating the clinical spectrum and expanding genotype-phenotype associations in PI4KA-related disorder
Claire Salter 1;2, Yiying Cai3;4;5, Bernice Lo6;7, Guy Helman8;9, Richard Caswell10, Francesco Saettini11, Fabiola Guerra12;13;14, Mario Mauri13, Henry Taylor15, Amber McCartney3;4;5, Joseph Leslie2, Andrea Accogli16, Federico Zara16, Monica Traverso16, James Fasham2;17, Joshua Lees18, Matteo Ferla19, Barry Chioza2, Olivia Wenger20, Ethan Scott20, Harold Cross21, Joanna Crawford9, Ilka Warshawsky22, Matthew Keisling22, Dimitris Agamanolis22, Catherine Ward-Melver22, Helen Cox23, Mamoun Elawad24, Tamas Marton25, Matthew Wakeling2, Dirk Holzinger26, Stephan Tippelt26, Deyana Valcheva27, Christin Deal28, Sara Van Meerbeke28, Catherine Walsh Vockley29, Manish Butte30, Utkucan Acar30, Marjo van der Knaap31;32, Georg-Christoph Korenke33, Urania Kotzaeridou34, Tamas Balla35, Cas Simons8;9, Holm Uhlig36;37;38, Pietro De Camilli3;5;39, Nicole Wolf31;32, Andrew Crosby2, Emma Baple2;17
1Wessex Clinical Genetics Service, Princess Anne Hospital, Southampton, United Kingdom; 2University of Exeter Medical School, RILD Wellcome Wolfson Centre, Exeter, United Kingdom; 3Yale University School of Medicine, Neuroscience and cell biology, New Haven, United States; 4Yale University School of Medicine, Program in Cellular Neuroscience Neurodegeneration and Repair, New Haven, United States; 5Yale University School of Medicine, Howard Hughes Medical Institute, New Haven, United States; 6Sidra Medicine, Research Branch, Doha, Qatar; 7Hamad Bin Khalifa University, College of Health and Life Sciences, Doha, Qatar; 8Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia; 9Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia; 10Royal Devon University Healthcare NHS Foundation Trust, Genomics Laboratory, Exeter, United Kingdom; 11Centro Tettamanti, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy; 12Fondazione IRCCS San Gerardo dei Tintori, Centro Tettamanti, Monza, Italy; 13Università degli Studi Milano-Bicocca, Dipartimento di Medicina e Chirurgia, Monza, Italy; 14University of Surrey, Molecular Systems Biology, Faculty of Health and Medical Sciences, Guildford, United Kingdom; 15Imperial College London, Department of Surgery and Cancer, London, United Kingdom; 16IRCCS Istituto Giannina Gaslini, Genova, Italy; 17Peninsula Clinical Genetics Service, Royal Devon and Exeter Hospital, Exeter, United Kingdom; 18Yale University School of Medicine, Department of Cell Biology, New Haven, United States; 19University of Oxford, Wellcome Centre Human Genetics, Oxford, United Kingdom; 20New Leaf Center, Mt. Eaton, United States; 21University of Arizona College of Medicine, Department of Ophthalmology,, Tucson, United States; 22Akron Children’s Hospital, Akron, United Kingdom; 23West Midlands Clinical Genetics Service, Birmingham Women’s and Children’s Hospital, Birmingham, United Kingdom; 24Sidra Medicine, Department of Gastroenterology, Doha, Qatar; 25Birmingham Women’s and Children’s Hospital, West Midlands Perinatal Pathology, Birmingham, United Kingdom; 26University of Duisburg-Essen, Department of Pediatric Haematology-Oncology, Essen, Germany; 27Sana Kliniken Duisburg, Department of Pediatrics, Germany; 28UPMC Children’s Hospital of Pittsburgh, Division of Pediatric Allergy and Immunology, Pittsburgh, United States; 29UPMC Children’s Hospital of Pittsburgh, Division of Genetic and Genomic Medicine, Pittsburgh, United States; 30UCLA, Department of Paediatrics, Los Angeles, United States; 31VU University Amsterdam and Amsterdam Neuroscience, Amsterdam Leukodystrophy Center, Department of Child Neurology, Amsterdam, Netherlands; 32Centre for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Department of Functional Genomics, Amsterdam, Netherlands; 33University Children’s Hospital, Department of Neuropediatrics, Oldenburg, Germany; 34Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Department of Child Neurology and Metabolic Medicine, Heidelberg, Germany; 35Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Section on Molecular Signal Transduction, Bethesda, United States; 36NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Translational Gastroenterology Unit, Oxford, United Kingdom; 37Oxford NIHR Biomedical Research Centre, Oxford, United Kingdom; 38University of Oxford, Department of Paediatrics, Oxford, United Kingdom; 39Yale University School of Medicine, Kavli Institute for Neuroscience, New Haven, United States
Background/Objectives: We recently described biallelic PI4KA (phosphatidylinositol 4-kinase alpha) variants as a cause of a severe syndromic neurodevelopmental disorder. PI4KA encodes PI4KIIIa (phosphatidylinositol 4-kinase III alpha), an essential lipid kinase that functions within the PI4KIIIa-TTC7-HYCC-EFR3 heterotetrameric complex.
Here, we expand our patient group to 46 individuals from 30 unrelated families and reveal a highly variable phenotypic spectrum encompassing gastrointestinal, immunological and neurological presentations.
Methods: Detailed clinical evaluations were undertaken alongside exome/genome sequencing, in-silico modelling, enzymatic and protein expression studies. New affected individuals were identified through GeneMatcher, international collaboration, peer-reviewed literature and the PI4KA Community (https://pi4ka-community.org).
Results: We have identified 45 rare, predicted deleterious PI4KA variants, associated with a phenotype that displays striking overlap with TTC7A-related gastrointestinal defects and immunodeficiency and/or HYCC1-related hypomyelinating leukodystrophy (PI4KIIIa complex proteins).
We define the potentially modifiable immunological features as a predominate B-cell deficiency, and show that the variability in clinical outcomes relates to the impact of each variant on PI4KIIIa complex function, either by directly impacting catalytic activities or altering complex stability in a tissue dependent manner.
Conclusion: Our data defines PI4KA genotype-phenotype correlations and consolidates biallelic variants as a cause of a variable multisystem disorder, most commonly comprising hypomyelinating leukodystrophy with/without inflammatory bowel disease, multiple intestinal atresia and immunodeficiency.
By developing clinical guidelines, a comprehensive patient registry and the PI4KA Community website, our work provides an important resource for improving knowledge of the natural history and pathomolecular basis of this rare disorder, and for supporting newly diagnosed families.
Grants: Wellcome Trust (216279/Z/19/Z)
Conflict of Interest: None declared
C32.6 Dysfunction of the centrosomal protein, CEP76, is associated with syndromic ciliopathies
Kamal Khan1, Katherine Bishara1, Aysegul Ozanturk2, Stephan Frangakis2, Farid Ullah1, Thomas Arbogast1, Mohammed Ayman Al Khateeb3, Léna Damaj4, Beatrice Bocquet5, Isabelle Meunier5, Tobias Bartolomaeus6, LEILA QEBIBO7, Małgorzata Rydzanicz8, Rami Abou Jamra6, Francis Ramond9;10, lydie BURGLEN7;11, Erica Davis 1;12
1Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, United States; 2Center for Human Disease Modeling, Duke University Medical Center, Durham, NC, United States; 3Women’s Wellness Research Center and Neonatal Intensive Care Unit, Hamad Medical Corporation; and Weill Cornell Medicine, Doha, Qatar; 4Department of Pediatrics, Competence Center of Inherited Metabolic Disorders, Rennes Hospital, Rennes, France; 5Referent National Centre for Sensory Genetic Diseases, University Hospital and Vision Team Inserm U1298 Institute for Neurosciences of Montpellier (INM), Univ. Montpellier, INSERM, Montpellier, France; 6Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany; 7Centre de Référence Malformations et Maladies Congénitales du Cervelet, Département de génétique, AP-HP. Sorbonne Université, Hôpital Trousseau, Paris, France; 8Department of Medical Genetics, Medical University of Warsaw, Warsaw, Poland; 9Department of Genetics, Centre Hospitalier Universitaire de Saint-Étienne, Saint-Étienne, France; 10AURAGEN Laboratory, Plan France Medecine Genomique 2025, Lyon, France; 11Developmental Brain Disorders Laboratory, Imagine Institute, INSERM UMR 1163, Paris, France; 12Department of Pediatrics and Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
Background/Objectives: We report CEP76, encoding centrosomal protein 76, as a ciliopathy-associated locus. CEP76 has been shown previously to associate with CP110, a CDK2 substrate, and CEP97, a calcium-binding protein, which localize to the basal body and daughter centriole of primary cilia.
Methods: We used multidisciplinary tools including clinical assessment, exome/genome sequencing, in vivo and in vitro modeling, and mass spectrometry.
Results: Data sharing platforms connected multiple independent groups who performed proband-parent trio sequencing on unrelated pedigrees with suspected genetic disorders. Six unrelated affected children with partially overlapping phenotypes spanning the clinical entities of retinitis pigmentosa, intellectual disability, Bardet-Biedl syndrome and Joubert syndrome harbor rare biallelic variants in CEP76 that segregate with disease and are predicted to impact protein function. Importantly, zebrafish mutants with a homozygous intragenic cep76 deletion display photoreceptor deficits as evidenced by significantly altered response to light-dark stimuli and histological phenotypes including rhodopsin mislocalization and significantly reduced ciliary length compared to controls. Furthermore, RNA silencing of endogenous CEP76 and/or chemical perturbation induced centriolar proliferation phenotypes in U2OS cell lines. These abnormalities were rescued by introduction of wild type CEP76, however, introduction of missense patient variants did not rescue the aberrant centriolar phenotypes. We confirmed known interactors CP110 and CEP97, identified other interacting ciliopathy proteins such as ALMS1, and nominated candidate proteins, such as LUZP1 in the CEP76 centrosomal protein network.
Conclusion: Genetic and functional data provide evidence to support association of CEP76 in a clinically heterogeneous human disorder hallmarked by retinal and neurological features.
Grants: R01DK072301, R01HD042601
Conflict of Interest: None declared
C33 Metabolic and Mitochondrial
C33.1 Biallelic loss-of-function variants in SGMS1 cause a novel metabolic disorder
Johannes Kopp 1;2;3, Hristiana Lyubenova1;2, Timo Sachsenheimer4, Oliver Küchler1;5, Leonard Koch1, Viktor Glaser6;7, Yaolin Pu6;7, Dimitrios Laurin Wagner6;7;8;9, Lena-Luise Becker10;11;12, Jana Marie Schwarz10, Stefan Mundlos1;2, Denise Horn1, Hossein Najmabadi13, Kimia Kahrizi13, Björn Fischer-Zirnsak1;2, Britta Brügger4, Felix Boschann1;14;15
1Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Medical Genetics and Human Genetics, Berlin, Germany; 2Max Planck Institute for Molecular Genetics, FG Development and Disease, Berlin, Germany; 3Institute of Chemistry and Biochemistry, Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Berlin, Germany; 4Heidelberg University Biochemistry Center (BZH), Heidelberg, Germany; 5Exploratory Diagnostic Sciences, Berliner Institut für Gesundheitsforschung, Berlin, Germany; 6.Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany; 7BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany; 8Institute of Transfusion Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Campus Charité Mitte, Charitéplatz 1, 10117, Berlin, Germany; 9Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany.; 10Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Pediatric Neurology, Campus Virchow Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany; 11Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Chronically Sick Children, Campus Virchow Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany; 12Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Cell biology and Neurobiology, Campus Virchow Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany; 13Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran; 14Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, Charitéplatz 1, 10117 Berlin, Germany; 15Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
Background/Objectives: Sphingolipids are critical for the composition of cell membranes thereby facilitating vesicle trafficking and cell signaling. Several lipid storage disorders have been linked to genes involved in sphingolipid metabolism. SGMS1 encodes the sphingomyelinsynthase 1 (SMS1), which catalyses the reaction from ceramide to sphingomyelin.
Methods: Trio exome sequencing (Trio-ES) was performed in the affected child and her unaffected parents. Lipidomic profiles were analysed in patient-derived plasma, cerebrospinal fluid (CSF) and fibroblasts. Further studies in patient-derived dermal fibroblasts included immunoblotting, immunofluorescence and transcriptome sequencing.
Results: The affected girl presented with global developmental delay, sensorineural hearing loss and cerebral dysmyelination. Trio-ES identified compound heterozygous missense variants in SGMS1 (NM_147156.4): c.677G>A, p.R226Q; c.850G>A, p.G284S. Lipidomics analysis in dermal fibroblast, plasma and CSF showed reduced sphingomyelin and increased ceramide levels, thereby confirming SMS1 enzyme deficiency. Abnormal morphology of the Golgi apparatus was observed by immunofluorescence staining in fibroblasts and RNA-seq revealed misexpression of genes involved in lipid metabolism, mitochondrial and autophagic pathways. Additionally, two siblings with moderate intellectual disability, motor delay and coarse facial features, homozygous for the p.R226Q variant, were identified via a GeneMatcher query.
Conclusion: Here, we present three individuals from two unrelated families with biallelic SGMS1 variants. As our data confirm enzyme deficiency as the underlying mechanism, we propose SGMS1 as a candidate gene for a novel sphingolipid-related metabolic disorder.
Conflict of Interest: None declared
C33.2 Antisense oligonucleotides for mitochondrial disorders
Catalina Vasilescu 1;2, Ekaterina Smirnova1;2, Robert Kopajtich1;2, Adela Della Marina3, Peter Freisinger4, Daniele Ghezzi5;6, Costanza Lamperti5, Hans A. Mayr7, Robert Taylor8;9, Arnaud Vanlander10;11, Dmitrii Smirnov1;2, Holger Prokisch1;2
1Technische Universität München, Institute of Human Genetics, School of Medicine, Munich, Germany; 2Helmholtz Zentrum München, Institute of Neurogenomics, Computational Health Center, Munich, Germany; 3University Duisburg-Essen, Department of Pediatric Neurology, Centre for Neuromuscular Disorders, Centre for Translational Neuro- and Behavioral Sciences, Essen, Germany; 4Klinikum am Steinenberg, Children’s Hospital Reutlingen, Reutlingen, Germany; 5Fondazione IRCCS Istituto Neurologico Carlo Besta, Medical Genetics and Neurogenetics Unit, Milan, Italy; 6University of Milan, Department of Pathophysiology and Transplantation (DEPT), Milan, Italy; 7Salzburger Landeskliniken (SALK) and Paracelsus Medical University (PMU), University Children’s Hospital, Salzburg, Austria; 8Newcastle University, Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle Upon Tyne, United Kingdom; 9Newcastle upon Tyne Hospitals NHS Foundation Trust, NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle Upon Tyne, United Kingdom; 10Ghent University, Mitochondrial Investigations Laboratory, Ghent, Belgium; 11Ghent University Hospital, Department of Internal Medicine and Paediatrics, Division of Paediatric Neurology and Metabolism, Ghent, Belgium
Consortium: PerMiM
Background/Objectives: Mitochondrial disorders are clinically heterogeneous genetic disorders characterized by mitochondrial dysfunction. Despite >400 causative genes uncovered, ~50% of patients remain without a genetic diagnosis after whole exome sequencing. To decipher the missing heritability, we routinely integrate exomes with RNAseq and proteomics from patient fibroblasts. Our RNAseq data revealed pathogenic variants inducing aberrant splicing, which hold the potential to be modulated with splice-switching antisense oligonucleotides (SSOs). Here, we design and test SSOs to correct aberrant splicing caused by intronic variants.
Methods: We selected 7 intronic variants causing aberrant splicing in NDUFAF5 (n = 2), NAXE, MRPS25, DGUOK, LIG3, and TIMMDC1. We designed allele-specific SSOs to block the pathogenic variant site sterically. We treated patient fibroblasts with SSOs and extracted total RNA to evaluate the efficacy of the splice correction by qPCR. RNAseq and proteomics are ongoing to depict the extent of functional recovery and potential side effects.
Results: Out of 7 targeted pathogenic variants, we obtained correct transcript rescue for 4. The rescue efficacy varied, ranging between 10-50% of a control normal transcript. Integrating qPCR data with RNAseq and proteomics may indicate the percentage of the transcript recovery needed to correct the proteome profile and identify candidate SSOs for personalized therapies.
Conclusion: Using a rational design of SSOs based on RNAseq, we can correct splicing defects of nuclear-encoded genes causing mitochondrial disorders. RNAseq and proteomics are valuable tools to evaluate the efficacy and safety of SSOs, facilitating personalized therapies in mitochondrial disorders with aberrant splicing.
Grants: Personalized Mitochondrial Medicine (PerMiM)
Conflict of Interest: None declared
C33.3 Unravelling Mitochondrial Diseases Metabolism: Discovery and Validation of Biomarkers in the Largest Patient Cohort Study to Date
Dmitrii Smirnov 1;2, Costanza Lamperti3, Sarah Stenton1;4, Silvia Marchet3, Daniele Ghezzi3, Rafael Artuch5, Marcello Bellusci6, Carole Linster7, Emma L. Schymanski7, Gabi Kastenmüller8, Boriana Buechner9, Ron Wevers10, Michael Witting11;12, Mohit Jain13, Thomas Klopstock9, Holger Prokisch1;2
1Klinikum rechts der Isar der Technischen Universität München, School of Medicine, Institute of Human Genetics, Munich, Germany; 2Helmholtz Munich, Institute of Neurogenomics, Neuherberg, Germany; 3Istituto Neurologico “Carlo Besta” | Fondazione IRCCS, Unit of Medical Genetics and Neurogenetics, Milano, Italy; 4Boston Children’s Hospital, Division of Genetics and Genomics, Boston, MA, United States; 5Institut de Recerca Sant Joan de Déu (IRSJD), Clinical Biochemistry Department, Barcelona, Spain; 6University Hospital October 12, Reference Center for Hereditary Metabolic Disorders, Madrid, Spain; 7University of Luxembourg, Luxembourg Centre for Systems Biomedicine, Belvaux, Luxembourg; 8Helmholtz Munich, Institute of Computational Biology, Neuherberg, Germany; 9University Hospital of the Ludwig-Maximilians-University, Department of Neurology Friedrich-Baur-Institute, München, Germany; 10Radboud University Medical Center, Department Laboratory Medicine, Nijmegen, Netherlands; 11Helmholtz Munich, Metabolomics and Proteomics Core, Neuherberg, Germany; 12TUM School of Life Sciences, Technical University of Munich, Chair of Analytical Food Chemistry, Freising, Germany; 13University of California San Diego, La Jolla, United States
Consortium: mitoNET, GENOMIT
Background/Objectives: Mitochondrial diseases (MDs) encompass a diverse group of genetically and phenotypically heterogeneous disorders that pose diagnostic challenges. Current biomarkers for MDs have limited diagnostic sensitivity/specificity and correlation with severity/progression. We conducted a comprehensive study using the largest untargeted lipidomics dataset for MDs, comprising >1,500 patients.
Methods: Within the GENOMIT consortium, we collected plasma samples, clinical measurements, phenotypic and genetic data from multiple patient visits. Through LC-MS lipidomics, we quantified >20,000 features in 3,211 plasma samples. Biomarker discovery was performed with multivariate regression and validated in independent cohorts and using different analytical platforms. mQTL analysis was performed on 1,087 genotyped samples.
Results: We discovered a class of fatty acids (p-value < 2e-36) significantly increased in MDs. Findings were validated in two independent cohorts, muscle biopsies, fibroblast cell lines, and a mouse model. The biomarker exhibited comprehensive performance for MD diagnosis achieving an area under the curve (AUC) of 0.88, outperforming FGF-21 (AUC = 0.78) and GDF-15 (AUC = 0.72). It effectively distinguished MDs from non-mitochondrial phenocopies and did not show elevated levels in asymptomatic mutation carriers. The biomarker demonstrated even greater discriminatory power for specific MD syndromes, including MELAS (AUC = 0.94), MERRF (AUC = 0.92), and CPEO/CPEOplus (AUC = 0.91) and significant correlation with MD severity. Biomarker GWAS prioritised several loci, including LIPT2, a mitochondrial protein involved in fatty acid metabolism.
Conclusion: The international collaboration allowed, to our knowledge, the largest biomarker investigation performed on MDs to date. The new biomarker exhibits the highest sensitivity and specificity characteristics reported so far for MDs.
Grants: GENOMIT
Conflict of Interest: None declared
C33.4 Biallelic loss-of-function variants in MRPL42 cause a lethal mitochondrial disease
Felix Boschann1;2;3, Susanne Römer4, Johannes Kopp1;5;6, Oliver Küchler1;7, Dominik Seelow1;7, Stefan Mundlos1;5, Sebastian Brachs8;9, Denise Horn1, Markus Schülke-Gerstenfeld10;11;12, Björn Fischer-Zirnsak 1;5
1Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Medical Genetics and Human Genetics, Berlin, Germany.; 2Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, Charitéplatz 1, 10117 Berlin, Germany.; 3Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.; 4Charité - Universitätsmedizin Berlin, Department of Neonatology, Berlin, Germany.; 5Max Planck Institute for Molecular Genetics, FG Development and Disease, Berlin, Germany.; 6Institute of Chemistry and Biochemistry, Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Berlin, Germany.; 7Exploratory Diagnostic Sciences, Berliner Institut für Gesundheitsforschung, Berlin, Germany.; 8Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Endocrinology and Metabolism, 10117 Berlin, Germany.; 9German Centre for Cardiovascular Research, partner site Berlin, Berlin, Germany.; 10Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Department of Pediatric Neurology, Berlin, Germany.; 11Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health (BIH), Center for Chronically Sick Children, Berlin, Germany.; 12Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), NeuroCure Clinical Research Center, Berlin, Germany
Background: Mitochondrial ribosomes consist of a large and a small subunit and are essential for the translation of mitochondrial-encoded proteins. Genetic defects affecting mitochondrial ribosome components cause multisystem diseases, often associated with early lethality.
Methods: Trio genome sequencing (Trio-GS) was performed on DNA samples of the affected boy and his healthy parents. This analysis was complemented by PolyA-enriched RNA-seq and qRT-PCR using patient-derived fibroblasts. These cells were further characterized by immunoblotting, immunofluorescence and OXPHOS activity assays.
Results: The affected boy presented postnatal with a severe multisystem disorder consisting of respiratory insufficiency, muscular hypotonia and hyperlactatemia. In addition, he showed an auditory conduction defect, epileptic seizures, a hypertrophic right ventricle and died nine weeks after birth. Trio-GS identified the intronic homozygous variant (NM_014050.4):c.219+6T > A in MRPL42, which encodes a component of the large mitochondrial ribosome subunit. RNA-seq revealed aberrant splicing leading to skipping of exon 4, resulting in a frameshift and generation of a premature termination codon: p.(Asn46Leufs*18). Immunoblotting showed the absence of MRPL42 and a swollen mitochondrial network was found by immunofluorescence. Investigation of OXPHOS complex activities identified a reduction of complex I and IV. Furthermore, we detected an overall decreased activity of the respiratory chain in his fibroblasts.
Conclusion: Here, we described a biallelic MRPL42 loss-of-function variant as the underlying cause for a lethal form of mitochondrial disease. Since our data show a strong impact on the OXPHOS system, we propose MRPL42 as a candidate gene for a novel form of mitochondrial ribosome-related disorders.
Conflict of Interest: None declared
C33.5 An Inborn Metabolic Disease as the cause of early infant death: a retrospective study in 1569 deceased children
B. Sikkema-Raddatz 1, Gea Kiewiet1, Willemijn J van Rijt2;3, Klaas Bijsterveld4, Pim de Blaauw4, Fjodor van der Sluijs4, Ronald G H J Maatman4, Francjan van Spronsen2, Peter Schielen5, Terry Derks2, Rebecca Heiner4
1University Medical Center Groningen, Department of Genetics, Groningen; 2University Medical Center Groningen, Beatrix Childrens’ Hospital; 3; 4University Medical Center Groningen, Department of Laboratory Medicine, Groningen; 5International Society for Neonatal Screening, Stichtse Vecht
Background/Objectives: Over 43 Inborn Metabolic Diseases (IMDs) are associated with sudden infant death (SID), causing 0.9 - 6.0% of the SID cases. Postmortem, they might remain unrecognized because of non-specific findings. Only some of these IMDs are included in the Dutch Newborn Screening (NBS). Many can be identified by biochemical analysis in dried blood spots (DBS). The aim of this study was to investigate IMD prevalence in a large cohort of Dutch children who died before 5 years-of-age.
Methods: DBS, obtained for NBS between 2013 and 2017, of children deceased within the first 60 months of life were collected (n = 1569). DBS acylcarnitines and amino acids were measured with tandem mass spectrometry. Outlier detection with non-parametric methods on log-transformed data and principal component analyses was performed. Whole exome sequencing (WES) was performed on all identified outliers. A panel of 967 IMD-related genes was analysed. One homozygous or two heterozygous (likely) pathogenic variants in a gene were defined as disease causing and possible cause of death.
Results: Biochemical outliers were identified in 303 (19%) samples. WES failed in 7 of these samples due to insufficient DNA or DNA quality. In 14 cases (0.9% of the total cohort) a causal IMD was detected in 13 different genes.
Conclusion: The estimated prevalence of IMD in these 1569 children who died before the age of 5 years in the Netherlands was at least 0.9%. Even without any further clinical information the combination of a positive biochemical and genetic result allowed identification of IMD as causal for early death.
Conflict of Interest: None declared
C33.6 Diagnosed cases in Solve-RD through reanalysis of mitochondrial DNA in exome and genome sequencing data
Ida Paramonov 1, Thiloka Ratnaike2;3, Leslie Matalonga1, Raul Tonda1, Steven Laurie1, Rita Horvath4, Sergi Beltran1;5
1Centro Nacional de Análisis Genómico (CNAG), Barcelona, Spain; 2University of Cambridge, Department of Paediatrics, United Kingdom; 3Colchester Hospital, East Suffolk and North Essex NHS Foundation Trust, Department of Paediatrics, United Kingdom; 4University of Cambridge, Department of Clinical Neurosciences, School of Clinical Medicine, United Kingdom; 5Universitat de Barcelona (UB), Departament de Genètica, Microbiologia I Estadística, Facultat de Biologia, Spain
Consortium: Solve-RD consortium
Background/Objectives: A main objective of the Solve-RD project is to diagnose patients with undiagnosed rare diseases through reanalysis of exomes and genomes submitted by partnering European Reference Networks (ERNs). Mitochondrial diseases are rare, heterogeneous, and challenging to diagnose. We developed and applied an integrated genotype-phenotype approach for streamlining the diagnosis of mitochondrial disorders, using MToolBox, MITOMAP and a reference database of mitochondrial DNA (mtDNA) disease patients (MitoPhen).
Methods: MToolBox was used to analyse mtDNA in 11,516 datasets from 11,250 undiagnosed individuals with Human Phenotype Ontology (HPO) descriptions submitted by 4 ERNs (ERN-NMD, ERN-RND, ERN-ITHACA and ERN-EpiCARE). 10,157 datasets in which at least 50% of the mtDNA had 5X coverage were considered for further analysis. MtDNA variants were prioritised based on MITOMAP database information, heteroplasmy level, and population frequencies. HPO-based phenotype similarity scoring between probands in MitoPhen and affected participants was computed with R.
Results: 184 rare mtDNA variants were prioritised in 180 undiagnosed individuals. The phenotypic similarity score with previously reported probands in MitoPhen was high or medium in 151 of these individuals, supporting a possible mtDNA disorder. Clinical evaluation resulted in 21 newly diagnosed patients and 8 individuals with candidate mtDNA variants still under evaluation, all with variable blood heteroplasmy fractions. Additionally, 45 individuals had reportable findings (e.g. homoplasmic variants with variable penetrance).
Conclusion: Our analysis demonstrates the importance of reviewing mtDNA variants identifiable from next-generation sequencing data. A tailored phenotype analysis approach specific to mtDNA diseases aids variant prioritisation.
Grants: EU H2020-779257
Conflict of Interest: None declared
Concurrent SymposiaS01 Paleogenomics as a tool to elucidate the genetic basis of modern diseases
S01.1 A Historical Perspective on Disease Risk
Svante Pääbo
A Historical Perspective on Disease Risk
Most medical conditions exhibit some degree of heritability. However, only a subset of these hereditary conditions are Mendelian, i.e., influenced by variants at a single locus. Genetic risk is more accurately understood as the combined probability of carrying risk-increasing or risk-decreasing variants at several or many loci. The likelihood of inheriting such variants is approximated by their frequencies in the population. With advances in our ability to retrieve ancient DNA, we are increasingly able to estimate the frequencies of genetic variants over time and space and determine their origins. Some medically relevant variants emerged among Neandertals. I will discuss how Neandertal-derived medically relevant variants have been introduced into the modern human gene pool and how ancient DNA can be used to trace their frequency over time among modern humans.
Conflict of Interest: None declared
S01.2 Human diseases in archaeological bone
Kirsten Bos
The past decade has demonstrated an impressive contribution from molecular methods for understanding the distribution and evolutionary history of a growing number of historical infectious diseases. This talk will explore the application of next generation sequencing to the study of past disease, and will describe current state of the art techniques for detection of pathogen DNA in archaeological tissues. Drawing upon Mycobacterium tuberculosis as a model organism, I will offer perspectives on molecular preservation, taphonomic influence, as well as the challenges and benefits that come with genome-level ancient pathogen reconstruction and analysis.
Conflict of Interest: None declared
S01.3 Ancient pathogens and modern diseases
Verena Schuenemann
The zoonotic potential of infectious diseases has largely been overlooked, despite its implications. Only recently, with the COVID-19 pandemic, this concept has received unparalleled attention in the wider public. Furthermore, many other diseases, some of which have afflicted humans since millennia, are re-emerging. Both situations require a more thorough approach for the characterization, prediction and eradication of zoonotic pathogens. Our One Health approach focusses, among other aspects, on the evolution and persistence of pathogens, together with their animal hosts. Besides its potential, this concept has largely been ignored in most studies of diseases in the past.
Here we will explore the first insights on medieval and early modern rodent hosts and their connections to plague and leprosy. In particular, we will look into two archaeological sites at Winchester, a medieval English city, well-known for its leprosarium and its connections to fur trade, from which we recovered four medieval Mycobacterium leprae genomes, including one from a red squirrel. The combination of historic sources with archaeological and genetic evidence enabled us to reconstruct details on the transmission of leprosy between humans and squirrels in medieval times including a possible transmission event.
Together, our research represents the first One Health approach on leprosy and plague in the past, which is centered around medieval animal host strains, and highlights the feasibility of such approaches to understand diseases’ zoonotic past and current potential.
Conflict of Interest: None declared
S02 The place of non-directiveness in genetic counselling revisited
S02.1 Nondirectiveness: A global 2020s perspective on a local 1970s decision
Jon Weil
The profession of genetic counseling began in the early 1970s with the Masters Degree Program in the School of Social Work at Sarah Lawrence College, New York. Its adoption of nondirectiveness as a guiding principle involved many factors including: scientific – at that time counseling primarily involved providing occurrence/recurrence risks to support reproductive decisions; societal – patients were primarily white, upper-middle class; institutional – Carl Roger’s theory of nondirective counseling was the basis for a highly regarded interviewing course in the new program; historical – to distance genetic counseling from eugenics and Nazi genocide.
Nondirectiveness has received many critiques. Some involve inherent problems and contradictions, e.g., narrow definitions are too prescriptive and broad definitions can be characterized without use of the term. Others involve changes in the scientific, societal, institutional and historical factors of its adoption, e.g., the expansion of genetic counseling into many areas of healthcare and the increasing diversity of individuals and communities served.
Genetic counseling is now international. It requires counseling techniques appropriate to the globally diverse individuals, communities and cultures that are and will be served. Their scientific, societal, institutional and historical circumstances are critically relevant, reducing nondirectiveness to a historically important but residual feature of genetic counseling.
Conflict of Interest: None declared
S02.2 The ethics of non-directiveness in genetic counselling
Christoph Rehmann-Sutter
Session: The place of non-directiveness in genetic counselling revisited
Presentation Title: The ethical basis of non-directiveness in genetic counselling
Speaker: Christoph Rehmann-Sutter
Abstract
The ethical basis of non-directiveness in counselling (genetic or psychosocial) is the respect for the client’s own vulnerability, competence and responsibility for their own life. “Autonomy” can be understood either negatively as independence or positively as agency. The latter implies responsible decision-making widely considered.
Negative autonomy however implies a narrow idea of the role of the genetic counsellor. The counsellor should provide relevant objective information about the implications of a test, its results and of genetic information in the context of the client’s situation, while the values that guide their decision should be the client’s only. Advice-giving in this framing of the counsellor’s role appears as difficult. Non-directiveness would then be defined by two rules: (i) providing complete unbiased information; (ii) refraining from giving practical advice.
I will argue that this suggests a form of genetic counselling and reproductive autonomy, which is not well prepared to the complexities of genetic counselling the the genomic era (for instance regarding polygenic screening) and is also actually not really worth having. A more spacious, positive and essentially relational understanding of autonomy allows the provision of respectful advice, if the client asks for it. This however does not push non-directiveness over board. We need to reinterpret non-directiveness in the sense of what Carl Rogers (who has introduced the idea of non-directive counselling in the 1940s) has called “client-centeredness”. Support of the clients’ decisions and responsibilty is the basic idea, the ethical core of genetic counselling. This suggests a form of conversation in the counselling situation that is focused on the client’s development of insight and agency. I hope that this wider conception of the ethics of genetic counselling essentially as an ethics of care, based on experiences of counselling practitioners as care providers, can accomodate some of the main concerns that have been raised in regard to dilemmas in the genomic area.
Conflict of Interest: None declared
S02.3 Non-directive communication and meeting patient needs in genetic counselling
Megan Scott
The non-directive approach in genetic counselling (GC) emphasises creating a supportive and collaborative environment to ensure patient autonomy, informed decision-making, and respect for individual values and preferences. Although a cornerstone of GC teaching and theory, the practical implementation of non-directiveness may be more complex. Advances in genetic testing and clinical management, as well as expectations from patients and referring healthcare providers, may create challenges in maintaining neutrality. Differing risk perceptions as well as contextual and cultural influences create further complexity during GC interactions. This paper provides debate on how genetic counsellors balance ethical and professional boundaries while simultaneously providing patient-centred care and meeting individual patient needs. Whilst much has been discussed on this topic from Western perspectives, less is known about how non-directiveness impacts GC communication in other global settings. The paper provides empirical evidence from video-recorded South African prenatal GC interactions showing how context may influence communication processes. Genetic counsellors may grapple with the dilemma of honouring non-directive principles, especially when patients request advice. Self-awareness of personal risk perceptions and how that may impact subtle communication cues, and the need to ensure patient autonomy is further highlighted. The ongoing debate on non-directiveness, decision-making, and patient care in GC is important for reflection and development within the profession. Further insights into this complex and demanding process may assist in providing enhanced professional support and training for genetic counsellors, including those practising in diverse settings. It may also ensure contextually tailored approaches for patients facing sensitive and difficult decisions during GC interactions.
Conflict of Interest: None declared
S03 The urinary bladder: from discovery to treatment
S03.1 The genomic architecture of disorders of the lower urinary tract
Melanie Chan
Conflict of Interest: None declared
S03.3 Gene therapy to treat urofacial syndrome
Neil Roberts
Rare early onset lower urinary tract disorders include defects of functional maturation of the bladder. Current treatments do not target the primary pathobiology of these diseases. Some have a monogenic basis, such as urofacial, or Ochoa, syndrome (UFS). Here, the bladder does not empty fully because of incomplete relaxation of its outflow tract, and subsequent urosepsis can cause kidney failure. UFS is associated with biallelic variants of HPSE2, encoding heparanase-2. This protein is detected in pelvic ganglia, autonomic relay stations that innervate the bladder and control voiding. Bladder outflow tracts of Hpse2 mutant mice display impaired neurogenic relaxation. We hypothesized that HPSE2 gene transfer soon after birth would ameliorate this defect and explored an adeno-associated viral (AAV) vector-based approach. AAV9/HPSE2, carrying human HPSE2 driven by CAG, was administered intravenously into neonatal mice. In the third postnatal week, transgene transduction and expression were sought, and ex vivo myography was undertaken to measure bladder function. In mice administered AAV9/HPSE2, the viral genome was detected in pelvic ganglia. Human HPSE2 was expressed and heparanase-2 became detectable in pelvic ganglia of treated mutant mice. On autopsy, wild-type mice had empty bladders whereas bladders were uniformly distended in mutant mice, a defect ameliorated by AAV9/HPSE2 treatment. Therapeutically, AAV9/HPSE2 significantly ameliorated impaired neurogenic relaxation of Hpse2 mutant bladder outflow tracts. Impaired neurogenic contractility of mutant detrusor smooth muscle was also significantly improved. These results constitute first steps towards curing UFS, a clinically devastating genetic disease featuring a bladder autonomic neuropathy.
Conflict of Interest: None declared
S04 Understanding the non coding genome
S04.1 Decoding the Code of the Non-coding Genome
Axel Visel
Understanding the function of enhancers within the context of genome regulation and in vivo biology continues to be a grand challenge. Currently, our limited ability to predict the consequences of sequence changes at the level of an individual enhancer up to larger enhancer architectures is a particular hurdle for the interpretation of whole-genome sequencing data from patients with a wide spectrum of conditions. We use sequence-based molecular approaches including chromatin mapping, synthesis-enabled modification of enhancer sequences, large-scale transgenic mouse studies (http://enhancer.lbl.gov), and CRISPR genome editing in mice to study the in vivo function of enhancers in developmental and disease-related processes. These methods provide insight not only into the in vivo function of enhancers, but also the impact of sequence and structural disruptions on activity and regulatory network architectures. I will illustrate this with data from ongoing studies of non-coding functions from the nucleotide to the megabase scale. Examples will include systematic tiling mutagenesis studies of individual enhancers to correlate their inner sequence architecture with in vivo activity patterns, as well as deletions of megabase scale gene deserts to explore their in vivo functions.
Conflict of Interest: None declared
S05 GWAS of disease progression: are we there yet?
S05.2 Longitudinal GWAS of early growth phenotypes
Nicole Warrington
Genetic effects on changes in human traits over time are understudied and may have important pathophysiological impact. We propose a framework that enables data quality control, implements mixed models to evaluate trajectories of change in traits, and estimates phenotypes to identify age-varying genetic effects in genome-wide association studies (GWASs). Using childhood body mass index (BMI) as an example, we included 71,336 participants from six cohorts and estimated the slope and area under the BMI curve within four time periods (infancy, early childhood, late childhood and adolescence) for each participant, in addition to the age and BMI at the adiposity peak and the adiposity rebound. GWAS on each of the estimated phenotypes in a subset of 25,640 individuals identified 28 genome-wide significant variants at 13 loci across the 12 estimated phenotypes, one of which was novel (in DAOA) and had not been previously associated with childhood or adult BMI. Genetic studies of changes in human traits over time could uncover novel biological mechanisms influencing quantitative traits.
Conflict of Interest: None declared
S05.3 CNS resilience in progression of Multiple Sclerosis
Stephen Sawcer
Multiple sclerosis (MS) is an autoimmune disease of the central nervous system (CNS) and a common cause of chronic neurological disability in young adults. Clinically the disease is characterized by episodes of temporary neurological dysfunction (“relapses”) which are super imposed on an inexorable accumulation of irreversible neurological disability (“progression”). Little is known about what determines the highly variable rate at which progression develops. In a Genome-wide Association Screen of 12,584 people with MS considering 7.8 million variants we confirmed that progression is highly heritable and identified genome-wide significant association with the common variant rs10191329 on chromosome 2. Carriage of the risk allele at rs10191329 (A) advances the time taken to need a walking aid by an average of 3.7 years. The association with clinical outcome was replicated in an independent cohort of 9,805 people with MS, and the variant was also shown to be associated with worse pathology in postmortem brains. In contrast we found no evidence that previously reported MS susceptibility variants, including HLA-DRB1*15:01, have an influence on the outcome of the disease. Linkage disequilibrium score regression (LDSC) in 205 tissue and cell type specific gene expression data sets showed that heritability is significantly enriched only in CNS tissues. Mendelian randomization demonstrated a protective effect of years in education and a risk from smoking, but no effect from vitamin D. Collectively these data suggest while the risk of developing MS is mediated by genetic effects in the immune system the outcome of the disease likely reflects the resilience of the nervous system.
Conflict of Interest: None declared
S06 Genetic Infertility
S06.1 The genetics of sperm defects
Joris Veltman
Severe forms of male infertility are mostly genetics in origin. Well-known genetic causes include Klinefelter syndrome and AZF microdeletions on the Y chromosome, but together these only explain a minority of all genetic forms of male infertility. Unfortunately, the genomics revolution has largely bypassed this field and diagnostic guidelines for male infertility have not been updated to include next generation sequencing approaches. As a consequence, most patients are not receiving information about the underlying cause of their infertility, and genetics information is rarely used to inform couples about the predicted success of assisted reproductive approaches or about preventable co-morbidities.
I believe that a major bottleneck in the advancement of this field has to do with the large genetic heterogeneity and the need to establish very large genomic datasets in order to identify recurrently mutated genes and reliably associate genes/genomic regions to specific types of reproductive disorders. For this reason, we have established the International Male Infertility Genomics Consortium (IMIGC) with a few colleagues in 2017, aiming to advance gene discoveries by sharing data on candidate genes and identifying recurrently mutated genes in cohorts from collaborators, but also aimed at bringing together clinical and biological expertise critical to the interpretation of genomic variation and the advancement of diagnostics.
In this presentation I will discuss the genetics of male infertility and highlight some recent advances in the field, strongly supported by the collaborative work of the IMIGC. I will highlight recent progress in our own research group, focused on studying the role of de novo as well as inherited mutations (DNMs) and structural variations in severe forms of male infertility.
Key references:
- 1.
Houston et al. A systematic review of the validated monogenic causes of human male infertility: 2020 update and a discussion of emerging gene-disease relationships. Human Reproduction Update 28: 15-29 (2021).
- 2.
Xavier et al. Disease gene discovery in male infertility: Past, Present and Future. Human Genetics (2021).
- 3.
Oud et al. A de novo paradigm for male infertility. Nature Communications 13: 154 (2022).
- 4.
Kimmins et al. Frequency, morbidity and equity - the case for increased research on male fertility. Nature Reviews Urology 21: 102-124 (2024).
- 5.
Wyrwoll et al. Improved phenotypic classification of male infertility to promote discovery of genetic causes. Nature Reviews Urology 21: 91-101 (2024).
Conflict of Interest: None declared
S06.2 Male infertility: When to test what?
Frank Tüttelmann
Male factors play a role in half of all infertile couples. This can often be overcome by medically assisted reproduction (MAR). To date, in the majority of infertile men, the underlying genetic, molecular, cellular, and/or organ defect(s) still remain poorly defined or unknown altogether. Clinical diagnosis of men in infertile couples is mostly based on semen analysis, resulting in descriptive diagnoses such as ‘asthenozoospermia’ (impaired sperm motility) or ‘azoospermia’ (no sperm in the ejaculate). These may partially explain why the couple’s inability to conceive, but the molecular mechanisms remain unclear, precluding evidence-based and individualised treatment decisions.
In recent years, progress has been made in the genetics of male infertility, as the number of genes in which variants are causal for various male infertility phenotypes continuous to increase. These include hormonal disorders such as hypogonadotrophic hypogonadism, impaired sperm motility and/or morphology, and importantly quantitatively reduced or absent sperm production. Thus, the number of male infertility cases that can be ‘solved’ by genetic analyses, in particular exome sequencing, is steadily increasing. The main aim of this talk is to review and dissect the highly heterogeneous phenotypes of male infertility and to provide an overview of the genes/pathways involved. Special attention will indeed be given to “when to test what”, how the genetic/molecular diagnosis can inform treatment decisions, and impact on the general health of the man and his offspring.
Conflict of Interest: None declared
S06.3 Premature Ovarian Insufficiency
Elena Tucker
Genomics for Premature Ovarian Insufficiency – Progress, Promise and Pitfalls
Elena J Tucker
Reproductive Development, Murdoch Children’s Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia
Although genomic testing for many genetic disorders has become part of routine clinical care, this has not occurred for the management of female infertility, including premature ovarian insufficiency (POI). POI is a leading form of female infertility affecting up to 4% of women under the age of 40 and characterized by amenorrhea and elevated gonadotropins.
Genetic diagnosis for POI is challenging because it is highly heterogeneous with over 100 causative genes affecting a wide variety of processes, such as meiosis, DNA damage repair, hormone signalling, folliculogenesis, immune function, mitochondrial function and more. Variants in these known genes only account for a minority of cases with the genetic basis of POI far from fully elucidated.
We have studied a diverse cohort of over 150 girls/women with POI using whole exome sequencing, identifying cause in >20%. We have validated causation using various approaches such as proteomic/transcriptomic analysis of patient cells, modelling in Drosophila or zebrafish and in-vitro functional assays. Our approach has led to multiple novel POI gene discoveries, such as TP63, TFAM, MRPL50, HROB, REC8, GGPS1 and more.
Importantly, we have shown that genomic sequencing can alter and improve patient management and outcomes. For example, we identified causative variants in NBN, EIF2B2 and LARS2 in three different patients presenting with apparently “isolated” POI. These genes are usually associated with syndromic POI in the context of cancer predisposition, neurodegeneration, and hearing loss respectively. In each case genomic sequencing identified syndromic POI before its full clinical manifestation. This enabled early intervention for associated co-morbidities, with the potential to improve patient outcomes.
Although genomic testing for infertility conditions such as POI has clinical utility, the fact that many causative genes have pleiotropic roles means that genetic diagnoses can have broad and unanticipated implications for patient health. Genomic counselling plays a critical role in the implementation of genomic testing for infertility to optimize health outcomes.
Conflict of Interest: None declared
S07 Editing in cardiovascular disease
S07.3 Base editing in dilated cardiomyopathy
Lars Steinmetz
Therapies for human disease, including cardiomyopathies, are rapidly transitioning from one-size-fits-all approaches to personalized and preventive treatments. To catalyze this transition, my lab develops and applies novel technologies for measuring and modifying biological systems across scales. With long-read sequencing, we mapped full-transcriptome alterations in dilated cardiomyopathy (DCM) caused by variants in the cardiac alternative splicing regulator, RBM20. We developed two therapeutic strategies to restore deficient alternative splicing in RBM20 DCM. First, we applied precision genome editing in the heart for correcting pathogenic variants in vivo. Our gene correction approach demonstrated high editing efficacy and tissue specificity, with no detectable systemic off-target effects. To establish a second therapeutic strategy, we dissected the molecular mechanism of pathogenic RBM20 via genome-wide CRISPR screening and high-throughput image-enabled cell sorting. This yielded an orthologous avenue targeted at restoring defective nuclear import of mutant RBM20. Furthermore, we utilize saturation gene editing to catalog disease-causing RBM20 variants into classes based on their pathogenic mechanisms. Together these mechanistic and technological insights help build precision and wellness-focused medicines for DCM.
Conflict of Interest: None declared
S08 Precision Oncology
S08.1 MSK-IMPACT: A Targeted Test for Mutations in Both Rare and Common Cancers
Zsofia K Stadler
Germline genetic risk assessment is becoming an integral part of oncological care. Memorial Sloan Kettering-Integration Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT) is a hybridization capture-based next-generation sequencing tumor-normal clinical assay for cancer patients that has allowed identification of not only somatic variants but also identification of germline cancer susceptibility. With over 40,000 cancer patients having undergone germline genetic risk assessment, both common and rare genetic variants have been identified with important implications for oncological treatment, cancer surveillance, and identification of at-risk family members. Findings from patients with solid tumors as well as those with early-onset cancer will be reviewed with insights for real-world application.
Conflict of Interest: None declared
S09 Assessing variants at scale
S09.2 The impact of substitutions, insertions, deletions and truncations on amyloid beta fibril nucleation
Benedetta Bolognesi
Amyloid fibrils form and precipitate in more than 50 incurable human diseases, including Alzheimer’s (AD) and Parkinson’s disease. Only a small number of human proteins and protein variants are known to form amyloids, limiting our ability to understand, predict and engineer amyloid aggregation from sequence. The initial events in amyloid formation are particularly challenging to study by classic biophysical methods, due to the transient and high-energy nature of transition states, but they are also the most critical steps to understand, as these events control the rate of the aggregation reaction and can be targeted to prevent or slow down amyloid formation for therapeutic purposes.
We have developed a multiplexed assay of variant effects (MAVE) approach that is able to quantify the rate of aggregation of thousands of protein sequences in parallel. We have shown that this MAVE accurately classifies insertions, deletions, and missense variants in Amyloid Beta (Aß), the protein which is mutated in familial forms of AD and which aggregates in all forms of AD. In this talk I will show how, by employing the same approach on different amyloids, we find that mutational effects in one single amyloid are not enough to predict mutational impact in another amyloid forming sequence. This provides a rationale for generating MAVEs for all human amyloids, an approach which will greatly impact clinical variant classification.
Conflict of Interest: None declared
S09.3 Prospectively generating functional data for every coding single nucleotide variant in SLC2A1 could transform our ability to make diagnoses of GLUT1-deficiency syndrome
Elizabeth Radford
Genetic sequencing is a powerful diagnostic tool in rare disease. However, correctly interpreting candidate genetic variants remains challenging. Where there is conflicting or insufficient evidence of a variant’s effect, it is termed a variant of uncertain significance (VUS). VUS prevent access to targeted treatments, prenatal diagnosis, inclusion in clinical trials, and can cause emotional turmoil for families. The number of VUS is rising rapidly, over 70% of missense variants in ClinVar are VUS. VUS are becoming a major impediment to optimal care. Uncertain or incorrect assessment of variant function can also reduce the power of clinical trials.
Early, fast, accurate diagnosis is particularly important where there is a targeted treatment which alters a child’s outcomes. Evidence is accumulating across diverse conditions that such treatments are often most efficacious if provided early in the disease course. As we start to consider expanded screening for such conditions, there is an imperative to improve our ability to interpret the associated genetic variants.
One such condition is GLUT1-deficiency syndrome, caused by loss of function genetic variants in the gene SLC2A1 which encodes the CNS glucose transporter. We employ saturation genome editing to experimentally determine the functional consequence of every possible coding single nucleotide variant (SNV), codon deletion and observed frameshift variants in SLC2A1 - generating a comprehensive, prospectively generated ‘variant effect map’.
To date, we have functionally characterised over 4500 SLC2A1 variants. 99% of synonymous variants appear functionally normal in our assay, while 99% of nonsense variants appear to be non-functional, demonstrating that the assay correctly classifies variant function. Using clinically ascertained pathogenic variants as true positives, and variants observed in population databases and clinically ascertained as benign as true negatives, we estimate 90-95% sensitivity and 97% specificity of these data for GLUT1 deficiency syndrome. We would like to explore whether our quantitative measurement of variant effect in vitro correlates with patients’ clinical phenotypes.
For greatest clinical impact, this approach will need to be scaled across many neurodevelopmental disorders, as these conditions are individually rare. Such direct functional characterisation of genetic variation at scale has the potential to transform clinical diagnosis. I will present our deep mutational scanning approach, progress on SLC2A1, and our broader strategy to address VUS.
Conflict of Interest: None declared
S12 Things that RNAs can do that we did not know about
S12.1 Circular RNAs drive oncogenic chromosomal translocations within the MLL recombinome in leukemia
Simon Conn
The first step of oncogenesis is the acquisition of a repertoire of genetic mutations to initiate and sustain the malignancy. An important example of this initiation phase in acute leukemias is the formation of a potent oncogene by chromosomal translocations between the mixed lineage leukemia (MLL) gene and one of 100 translocation partners, known as the MLL recombinome. Here, we show that circular RNAs (circRNAs)—a family of covalently closed, alternatively spliced RNA molecules—are enriched within the MLL recombinome and can bind DNA, forming circRNA:DNA hybrids (circR loops) at their cognate loci. These circR loops promote transcriptional pausing, proteasome inhibition, chromatin re-organization, and DNA breakage. Importantly, overexpressing circRNAs in mouse leukemia xenograft models results in co-localization of genomic loci, de novo generation of clinically relevant chromosomal translocations mimicking the MLL recombinome, and hastening of disease onset. Our findings provide fundamental insight into the acquisition of chromosomal translocations by endogenous RNA carcinogens in leukemia, via a mechanism we term Endogenous RNA-directed DNA damage (ER3D).
Conflict of Interest: None declared
S12.2 Engineered tRNAs suppress nonsense mutations in cells and in vivo
Zoya Ignatova
Unlocking the unreachable: harnessing tRNA therapeutics for rare genetic conditions
Zoya Ignatova
University of Hamburg. Germany
11% of all genetic mutations diseases are linked to non-sense mutations that convert a sense codon decoded by tRNA into a premature termination or stop codon (PTC), resulting in an abrupt termination of translation. To correct PTCs, we developed a novel strategy which is based on repurposing sense-codon decoding tRNAs to readthrough nonsense mutation-associated premature termination and restore protein synthesis and function. I will discuss our recent advances in the development of tRNA therapeutics with high activity and safety in cell and animal models and discuss different formulation approaches for single or chronic treatment modalities.
Conflict of Interest: None declared
S13 Integrative genomics: opportunities for disease prevention
S13.1 Strategies for risk-based screening and preventive actions using genetic profiles
Samuli Ripatti
Analyses of retrospective large-scale biobank studies have shown that polygenic risk scores stratify individuals efficiently in most common diseases with heritable risk. However, there are still limited number of showing the benefits of utilizing polygenic risk information in prospective settings.
In this talk I will discuss how in some diseases the genetic risks are context dependent and discuss potential uses of polygenic risk scores in picking up the right medicine and optimizing risk-based screening programs. I will also present some early results from prospective follow-up studies utilizing polygenic risk scores to stratify individuals based on their polygenic risks for cardiovascular diseases and breast cancer.
Conflict of Interest: None declared
S13.2 When the exposome meets genomics: tackling the complexity of preventive actions
Roel Vermeulen
An organism’s traits are the product of both genetic and environmental factors. Thanks to the Human Genome Project and ensuing advancements, we have now a thorough understanding of the genetic factors affecting health. However, the environmental aspects are less clear, primarily due to challenges in fully quantifying the environment. This gap significantly limits preventive healthcare, especially considering the critical role of environmental factors in many prevalent chronic diseases. To address this, the European Human Exposome Network was launched in 2019 to advance the measurement of the exposome—defined as the total environmental influences and biological responses over a person’s lifetime. Presently, the integration of exposome data with health records is enhancing the evaluation of both genetic and environmental data. This integration supports innovative research into gene-environment interactions and the development of exposome risk scores. This presentation will discuss recent advancements in exposomic methods and illustrate how these can be used to support primary and secondary health prevention strategies.
Conflict of Interest: None declared
S14 Long-read sequencing in the clinic
S14.3 Assessing long-read sequencing for rare disease diagnostic research
Lisenka Vissers
Long-read sequencing (LRS) technologies, such as PacBio HiFi genome sequencing on Revio, have the potential to revolutionize human genetics as long reads finally allow detection of variation in regions of the genome that have remained inaccessible by short read genome sequencing. This technology could therefore offer a comprehensive first-tier test for germline clinical genetics and rare disease research.
In this presentation I will highlight various studies to assess the clinical utility of long-read sequencing for rare disease diagnosis. Examples will include the use of a series of 100 mutation-positive controls in difficult to assess regions of the genome to determine whether LRS can replace current diagnostic assays, but also cohort studies using individuals whose genetic diagnosis has remained elusive after exhausting routine diagnostic testing, to determine the additional diagnostic yield of LRS in rare disease research.
Conflict of Interest: None declared
S15 Preimplantation genetic testing for polygenic conditions
S15.1 Polygenic embryo screening: epidemiological considerations and stakeholder perspectives
Shai Carmi
The genetic composition of embryos generated by in-vitro fertilization (IVF) can be examined prior to transfer using preimplantation genetic testing (PGT). Recent advances in PGT have made genome-wide genotyping of IVF embryos feasible and affordable, raising the question whether embryos could (or should) be screened for risk of polygenic diseases such as breast cancer, hypertension, diabetes, or schizophrenia using recently developed polygenic risk scores. In this talk, I will review research on the predicted clinical utility of polygenic embryo screening, as well as cover recent studies on the perspectives of clinicians, IVF patients, and the public.
Specifically, after introducing the technology, I will cover a number of modeling approaches predicting that polygenic embryo screening could, under a theoretical best-case scenario, lead to substantial relative risk reductions. I will also review several practical factors that are expected to limit the realized risk reductions, particularly not having enough embryos and a decay of polygenic scores accuracy over time. I will also briefly cover possible personal and social harms that may result from screening. Finally, I will cover recent research suggesting that while clinicians see little utility in polygenic embryo screening and are concerned about harms to patients and society, IVF patients and the American public are interested in the technology.
Conflict of Interest: None declared
S16 Sex and the Single Cell
S16.2 Cis-regulatory control of mouse sex determination
Nitzan Gonen
Cis-Regulatory Control of Mouse Sex Determination
Meshi Ridnik1&, Elisheva Abberbock1&, Veronica Alipov1, Shelly Ziv Lherman1, Shoham Kaufman1, Maor Lubman1, Francis Poulat2, Nitzan Gonen 1*
& These authors contributed equally to this work
1. The Mina and Everard Goodman Faculty of Life Sciences and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan, 5290002, Israel. 2. Group “Development and Pathology of the Gonad”. Department of Genetics and Development, Institute of Human Genetics, CNRS-University of Montpellier UMR9002, Montpellier, France
Male development in mammals depends on the activity of the two SOX gene: Sry and Sox9, in the embryonic testis. As deletion of Enhancer 13 (Enh13) of the Sox9 gene results in XY male-to-female sex reversal, we explored the critical elements necessary for its function and hence, for testis and male development. We demonstrate that while ablation of individual transcription factor binding site (TFBS) in Enh13 leads to normal testicular development, combined ablation of just two SRY/SOX binding motifs can alone fully abolish Enh13 activity leading to XY male-to-female sex reversal. This suggest that male development fully relies on very few nucleotides of non-coding DNA. Interestingly, we show that depending on the nature of these TFBS mutations, dramatically different phenotypic outcomes can occur, providing a molecular explanation for the distinct clinical outcomes observed in patients harboring different variants in the same enhancer.
Conflict of Interest: None declared
S17 Next generation functional genomics: variant effects at the single cell level
S17.3 Characterizing cis-regulatory elements using single-cell epigenomics
Sebastian Preissl
Transcriptional regulation plays a fundamental role in development and disease. Cis-regulatory elements (CRE) in the genome such as enhancers and promoters control cell-type-specific gene expression patterns. CRE and their activation status are characterized by distinct combinations of epigenetic marks. A large fraction of genetic variants associated with common traits and diseases overlap with CREs in the human genome. However, we still lack a comprehensive knowledge of cell-type-specific CRE dynamics and its role in disease pathogenesis. Thus, we have optimized single-cell chromatin accessibility and multiomics workflows to generate CREs atlases for a wide variety of tissues including deep profiling of the brain and heart. These comprehensive maps revealed regulatory programs and key transcription factors in hundreds of cell types and enabled functional validation of noncoding variants associated with complex human traits and diseases. Next, we have applied single-cell multiomics workflows to unravel the gene regulatory changes in beta cell subtypes in the human pancreas underlying type 2 diabetes. Currently, we are optimizing and applying joint profiling of histone modifications and transcriptomes from the same nucleus to gain insight into the interplay of different epigenetic layers in heart cells. I will discuss our most recent progress and findings.
Conflict of Interest: None declared
S18 Advanced statistical methods in human genetics
S18.1 Inference of ancestral recombination graphs enables genealogical analysis of complex traits
Pier Francesco Palamara
Conflict of Interest: None declared
S18.2 GWAS in isolated populations
Ida Moltke
For a long time GWAS have been performed mainly in Europeans. In this talk, I will present results from several studies where we have instead focused on a non-European population, namely the historically small and isolated Greenlandic population. I will show how this different focus has led to new insights into the genetic architecture of complex metabolic traits, like type 2 diabetes. Importantly, in contrast to GWAS in Europeans, we have identified several Inuit-specific genetic variants that have a population level impact, because they are both common and have large effects. For example, we have identified a type 2 diabetes variant with a population frequency of 0.17 and an odds-ratio of 10.3, which explains more than 10% of all diabetes cases in Greenland. Next, Iwill show some of the consequences of these new insights for diagnosis, treatment, and prediction of disease in Greenland. Finally, I will briefly discuss the implications for equity in health care.
Conflict of Interest: None declared
S18.3 Network expansion of genetic associations defines a pleiotropy map of human cell biology
Pedro Beltrao
Interacting proteins tend to have similar functions and when genetically perturbed will often influence the same organismal traits. For this reason, protein-protein interaction networks can be used to expand the list of candidate trait-associated genes from genome-wide association studies (GWAS) or family studies. In a published study we have performed network-based expansion of GWAS trait-associated genes for over 1000 human traits showing that this recovers known disease genes or drug targets. The similarity of network expansion scores was shown able to identify groups of related traits that are likely to share similar underlying genetic and biological process. Based on this analysis we can then defined pleiotropic gene modules linked to multiple traits which were enriched in genes related to protein ubiquitination and RNA processing. This analysis has now been extended to a joint analysis of GWAS, rare disorders and mouse gene-deletion phenotypes. We find that network-based propagation can be used to identify related traits across these different approaches defining a global map of human traits. For rare disorders, and using ciliopathies as an example, we can show trait distances defined by network propagation can split traits by organismal level phenotype and that related traits other than ciliopathies can be used to prioritize novel candidate disease-relevant genes.
Conflict of Interest: None declared
S19 Genomics in Africa
S19.2 Novel discoveries from genome-wide association studies in African populations
Ananyo Choudhury
The distinctive diversity and architecture of African genomes make genome-wide association studies (GWASs) in these populations ideal for discovering novel genetic associations and narrowing down on causal variants. We will begin the presentation by reviewing the current understanding of the origin of this diversity and how it could impact the discovery of signals in GWAS. The presentation will then introduce AWI-Gen, a study nested under the Human Heredity and Health in Africa (H3Africa) consortium, that includes about 12,000 participants from six study sites in Ghana, Burkina Faso, Kenya, and South Africa. The unique features of this cohort such as its geographic spread across East, West, and South Africa, population cross-sectional nature, focus on cardiometabolic disease/traits, multi-omics approach, and longitudinal data will be highlighted. Findings from several GWASs conducted on this cohort will be synthesized to illustrate the promises and importance of large-scale genomic studies in African populations. Moreover, based on the experience gathered from conducting these studies some of the critical considerations and methodological challenges encountered while implementing GWASs in African populations will be discussed.
Conflict of Interest: None declared
S20 Who owns your genome?
S20.1 Genome Defence: Inside the Epic Legal Battle to Determine Who Owns Your DNA
Jorge Contreras
In 2013, the US Supreme Court held that naturally occurring genetic sequences may not be patented, instantly invalidating hundreds, if not thousands, of existing patents and opening the market to genetic screens for cancer and other hereditary diseases. The case, Association for Molecular Pathology v. Myriad Genetics, was remarkable in many ways, not least because it was prosecuted on behalf of twenty plaintiffs -- researchers, professional associations, medical practitioners and individual patients -- by the American Civil Liberties Union and the Public Patent Foundation as a case centered on individual civil rights rather than a technical interpretation of the U.S. Patent Act. The case also sheds light on the poorly understood role of the executive branch and its many offices and agencies in formulating U.S. policy on science-based issues. In The Genome Defense: Inside the Epic Legal Battle to Determine Who Owns Your DNA (Hachette/Algonquin, 2021), Professor Jorge Contreras brings this important and unique case to life. Through nearly 100 interviews with attorneys, advocates, judges, patients and government officials, Contreras peels back the layers of this remarkable episode in American legal history and explains not only what happened, but why and how, and what its implications are for the future of medical science.
Conflict of Interest: None declared
S20.2 Ethical and social concerns related to commercialisation in genomics
Klaus Hoeyer
The European health data space: populations, populism, or public legitimacy?
With its vote on April 24, 2024, the European Parliament took a definitive step towards the realization of a European Health Data Space: a novel legal framework for genetic and other types of health data in the European Union. The framework establishes rights for citizens, facilitate access for secondary use, and designates standards for electronic health information. I will briefly introduce the framework and how it will intervene in debates about rights to control genetic data. The new cross-border health data infrastructure depends on public support for it to deliver the benefits envisioned by policymakers. It is therefore worth considering what we know about public attitudes towards data sharing and perceptions of legitimate rights and duties. By engaging these insights, we see the need for researchers in getting involved as the implementation of the European Health Data Space is translated into practice.
Conflict of Interest: None declared
S20.3 Data Ownership in Genomic Research Consortia (virtual)
Dianne Nicol
TITLE
Data Ownership in Genomic Research Consortia
Presented by Dianne Nicol and Jane Nielsen
ABSTRACT
Discourse around ownership of genomic sequence data has proliferated over recent years. There are likely to be few people who don’t feel a degree of connectedness to their genomic data. The inclusion of individuals’ genomic data in genomic datasets is critical to genomic research, and these datasets are most effective if shared widely. Genomic research consortia are an integral part of the genomic data sharing ecosystem, critical in facilitating data sharing among research groups. This presentation will consider the property status of genomic data at various stages of the research life cycle, and the potential ‘ownership’ claims to be made by various actors in data sharing networks. It does so by describing the legal position in Australia, the US and the UK, and presenting the findings of a study that examined policy documents and guidelines produced by international research consortia. This analysis has enabled us to assess whether consideration of property interests is at the forefront of data sharing efforts, where such property interests might reside and whether more needs to be done to reflect community values.
Conflict of Interest: None declared
S21 Inherited Metabolic Diseases
S21.2 Inherited disorders of glycosylation and deglycosylation
Eva Morava
Congenital disorders of glycosylation (CDG) are a group of more than 190 inborn errors of metabolism affecting N-linked and O-linked glycosylation, multiple pathways of protein glycosylation and lipid glycosylation. CDG patients present with a wide range of symptoms including the central nervous system and almost all organs of the body. Treatment however is only available for very few CDG subtypes. In the past decade the focus of CDG research changed and was redirected towards finding new therapies. These include drug repurposing, small molecules and dietary interventions. Specific nutritional treatment options for certain CDG types include oral supplementation of monosaccharide sugars, manganese, uridine, or pyridoxine. Additional management includes specific diets (i.e., complex carbohydrate or ketogenic diet), iron supplementation, and acute management including fresh frozen plasma supplements or coagulation factor treatment and albumin infusions. We will review the diagnostic flow chart, acute management, novel treatment options, clinical trials and dietary management in CDG with a focus on the most common subgroup: N-linked glycosylation defects.
Conflict of Interest: None declared
S22 Dosage sensitivity of human genetic disorders – clinical implications and therapeutic avenues
S22.2 Modulating gene regulatory elements to treat genetic diseases
Nadav Ahituv
Numerous genetic diseases are caused due to mutations that cause changes in gene dosage. For example, it is estimated that over 660 genes lead to human disease due to haploinsufficiency, having only one functional gene copy. Modulating gene regulatory elements to change the expression levels of the disease-associated gene/s, termed cis-regulation therapy (CRT), could provide a therapeutic option for these diseases. By targeting regulatory elements via CRISPR activation (CRISPRa), we show that this approach could be used to rescue a variety of haploinsufficient disorders, including obesity and autism spectrum disorders (ASD), neurodevelopmental delay and epilepsy. In addition, utilizing CRISPRa to engineer adipocytes and adipose organoids to outcompete tumors for nutrients, we show that gene regulatory modulation can also be used as a novel cancer therapy, termed Adipose Manipulation Transplantation (AMT).
Conflict of Interest: None declared
S23 Predisposition, Molecular signatures and Treatment in Cancer
S23.1 Comprehensive cancer predisposition testing in rare cancers
Arne Jahn
Typically, genetic testing for cancer predisposition is initiated based on clinical parameters (e.g. age of onset, family history for cancers), which lack sensitivity. In precision oncology programs large multi-omics datasets are generated to primarily evaluate targetable molecular alterations, but parallel tumor and control sequencing and data interpretation following international guidelines allow evaluation of cancer predisposition independent of these parameters. The German multi-center and multi-disciplinary precision oncology program MASTER (Molecularly Aided Stratification for Tumor Eradication Research, ClinicalTrial.gov NCT05852522) performs broad multi-omics molecular profiling (DNA, RNA, methylation, proteome) for patients with rare and early onset advanced cancers. Here we share our insights into cancer predisposition, therapy recommendations and somatic alterations (PMID: 35988656) from this cohort. We also discuss considerations on how to use this complex data and harmonize similar workflows for research and improved patient care.
Conflict of Interest: None declared
S23.2 Targetable pathway identification from mutation signatures
Andrew Jackson
With the aspiration of personalised medicine, cancer genome sequencing has been pursued at an industrial scale in the last decade. Alongside the identification of many cancer driver genes, mutational signatures have also been discovered. Such signatures reflect underlying disordered cellular processes, and can point to drug vulnerabilities for particular tumours. However the mechanistic basis for many signatures remains to be elucidated. Here I will describe our work investigating one such signature, linking it to the action of Topoisomerase I and to transcription associated mutagenesis.
Conflict of Interest: None declared
S23.3 Uncovering vulnerabilities in SWI/SNF-deficient SCCOHT
Sidong Huang
Small cell carcinoma of the ovary, hypercalcemic type (SCCOHT), is a rare and aggressive cancer that mainly affects young women. Conventional chemotherapeutics are rarely effective for treating SCCOHT, and most patients succumb to the disease within 2 years of diagnosis. SCCOHT is driven by biallelic deleterious mutations in SMARCA4, a key SWI/SNF chromatin remodeling gene, leading to loss of SMARCA4 (BRG1) protein expression. In contrast to other ovarian cancer subtypes, SCCOHT has a remarkably simple genome that harbors few mutations or chromosomal alterations. While SMARCA4 loss is not directly targetable, the monogenic nature of SCCOHT presents an ideal opportunity to uncover druggable targets that are synthetic lethal with SMARCA4 loss through functional screening. Using this systematic approach, we have uncovered selective vulnerabilities of SMARCA4 deficiency in SCCOHT that can exploited therapeutically, such as using inhibitors targeting CDK4/6, mitochondria respiration, and glutamine metabolism, as well as alanine dietary supplementation. Our work has uncovered potential treatment strategies for SCCOHT patients.
Conflict of Interest: None declared
S24 From single cell maps to function: disentangling the heterogeneity of complex diseases
S24.2 Creating a multi-omics roadmap to link disease variants to actionable biology to inform drug therapies
Gosia Trynka
GWAS variants are the genetic foundation for disease-causing biological processes and a significant avenue for discovering new drug targets. Given that the majority of GWAS variants are non-coding, their role is likely in regulating gene expression, which can be cell type-specific. Hence, the initial and crucial step is identifying the disease-relevant cell type.
CD4 + T cells play a critical role in regulating the immune system. Dysregulation of CD4 + T cell function leads to severe immune conditions. GWAS variants associated with immune diseases strongly implicate the role of T-cell activation in disease biology. Focusing on T-cell function, we are applying a toolkit of genomics approaches coupled with immune phenotyping at scale to link disease genes to dysregulated cell functions. We used single-cell eQTL mapping to define gene expression regulation during the time course of CD4 + T cell activation. We identified disease variants that colocalised with genes dynamically regulated during T-cell activation, i.e. genes whose eQTL effects manifested at specific activation time points. This approach identified known and suggested novel drug targets. Importantly, assaying gene expression at multiple activation time points enabled us to identify 60% more of colocalising genes. This highlights that many effects go unmapped when studying eQTL in steady cell states.
Finally, in addition to the low resolution in the cellular contexts, most approaches to interpreting GWAS variants are limited to transcriptional effects. We developed a high-content imaging assay that captures hundreds of functional and morphological features. Using a combination of transcriptomics and high-content imaging, we are building a multi-modal profiling of T-cell biology at scale to accelerate the translation of immune disease variants to function.
Conflict of Interest: None declared
S24.3 Deciphering Obesity-Linked Brain Circuits with Genetic and Single-Cell Strategies
Tune Pers
Conflict of Interest: This work is partly funded by Novo Nordisk A/S.
Talk title:
Deciphering Obesity-Linked Brain Circuits with Genetic and Single-Cell Strategies
Talk abstract:
Body weight regulation is a polygenic trait driven by currently incompletely understood cellular processes. Work by, among others, the GIANT consortium has previously shown that common genetic risk variants associated with body mass index (BMI), a commonly used proxy for obesity, most likely exert their effect through neuronal cell populations. However, despite the wealth of common genetic variants associated with BMI, the molecular processes mediating the effect of these variants remain mostly unresolved. In my presentation, I will present previous and current efforts leveraging mouse in vivo pharmacology, single-nucleus RNA sequencing and integration with BMI genome-wide association study (GWAS) data to decipher obesity-linked brain circuits. Our analyses identify hindbrain cell populations that respond to semaglutide and cagrilintide treatment, long-acting glucagon-like peptide-1 and amylin receptor analogs with proven effects on weight loss. We find that cagrilintide and semaglutide are likely to exert their action through both joint and complementary molecular pathways.
Conflict of Interest: None declared
S25 Biogeographical definitions of population groups
S25.1 Roots versus legs: How migration shaped human diversity (virtual)
Guido Barbujani
Guido Barbujani
Department of Life Sciences and Biotechnology, University of Ferrara, Italy
g.barbujani@unife.it
The DNA in our cells is a message from our ancestors and, along with fossil and archaeological evidence, has a lot to tell us about our past. Humans have the lowest genomic diversity in primates, suggesting our population sizes were minimal until recently. Homo sapiens populations expanded from Africa between 120,000 to 60,000 years (estimates differ), and have been on the move ever since. We shall explore the genomic consequences of some migration processes inferred from the analysis of both modern and ancient DNA, showing how these processes not only shaped human diversity, but also affected the landscape, plant and animal diversity, our social structure and our culture.
Conflict of Interest: None declared
S25.3 Appearance, ancestry, ‘race’?: Translations and the trouble with racial categories in forensic DNA phenotyping
Roos Hopman
In this talk I discuss the societal implications of (forensic) genetic investigatory technologies that estimate elements of physical appearance from DNA traces. Whereas these technologies, also known under the header forensic DNA phenotyping (FDP), aim at the identification of individual suspects, they can only work through the clustering of genetic and phenotypic data into groups and categories. In the talk I zoom in on the categories that are used in the development and application of FDP technologies, and show that these are not stable but mobile entities that change shape as they move from laboratories to police stations into investigations and society more broadly. Offering examples from forensic investigations and the use of FDP in the Netherlands, I show the risk of racialization adhering to these translations and open up discussion on why and how the categories we use in research matter.
Conflict of Interest: None declared
S26 Complete genomes
S26.1 Structural variation diversity and T2T human genomes
Evan Eichler
Structural variation diversity and T2T human genomes
Evan E. Eichler
Department of Genome Sciences and Howard Hughes Medical Institute, University of Washington, Seattle, WA
The complete resolution of genetic variation is critical to understanding the mutational processes underlying both disease and evolution. I will present our most recent work sequencing diverse human genomes telomere-to-telomere (T2T) and non-human apes using both ultra-long and high-fidelity long-read sequencing technologies. The work has led to the discovery of more complex forms of structural variation and their association with disease and disease susceptibility. We show that >30% of large inversion polymorphisms occur recurrently and these occur preferentially on sex chromosomes and at sites associated with recurrent genomic disorders linked to developmental delay and autism. Long-read sequence assembly of breakpoint regions associated with these genomic disorders (e.g. chromosome 22q11) reveals complex patterns of segmental duplications, both protecting and predisposing different human haplotypes to rearrangement. We also show that complete sequencing of human centromeres is leading to potential new insights into the molecular basis of nondisjunction. T2T assembly of human genomes will be important to understanding differential genetic risks in the human population.
Conflict of Interest: None declared
S26.2 Pangenome based analysis of structural variation
Tobias Marschall
Breakthroughs in long-read sequencing technology and assembly methodology enable the routine de novo assembly of human genomes to near completion. Such assemblies open a door to exploring structural variation in previously inaccessible regions of the genome, but the analysis of complex polymorphic loci comes with considerable computational challenges. The Human Pangenome Reference Consortium (HPRC) has released a draft pangenome based on the assemblies of 47 human genomes, providing a framework for comparative genome analysis using pangenome graphs. In my presentation, I focus on two questions:
First, we will ask how a pangenomic resource like this can be used in order to better analyse structural variants in samples from large cohorts. We present Locityper, a tool capable of genotyping such challenging genes using short and long-read whole genome sequencing. For each target, Locityper recruits and aligns reads to locus haplotypes and finds the likeliest haplotype pair by optimizing read alignment, insert size and read depth profiles. Locityper accurately genotypes up to 194 of 256 challenging medically relevant loci (95% haplotypes at QV33), an 8.8-fold gain compared to 22 genes achieved with standard variant calling pipelines. Furthermore, Locityper provides access to hyperpolymorphic HLA genes and other gene families, including KIR, MUC and FCGR.
Second, we will look at limitations of current pangenomes and how to address them, both in terms of completeness of individual assemblies and in terms of representation of variants at lower allele frequencies. To complement the HPRC resource, we have, in a separate effort, sequenced 1019 samples from the 1000 Genomes Cohort using nanopore sequencing. Introducing a new pangenome graph augmentation strategy, we have integrated structural variant alleles detected by various callers across this cohort with the variation already encoded within the HPRC pangenome. This combined data sets provides a basis to study the spectrum of structural variation and their underlying mechanisms in a much more comprehensive manner.
Conflict of Interest: None declared
S27 Psychiatric Genomics
S27.1 Genomics of ADHD
Anders Børglum
ADHD is a childhood neurodevelopmental disorder affecting around 5% of children and 2.5% of adults. It is characterized by extensive hyperactive, impulsive and/or inattentive behaviors that impair daily functioning, often accompanied by adverse health and socioeconomic outcomes. ADHD has a major genetic component, with an estimated heritability of 0.7-0.8, rooted in both rare and common risk variants.
I will present the results of our recent common variant (GWAS) and rare variant (RVAS) studies of ADHD, based on collaborations with the Psychiatric Genomics Consortium, deCODE Genetics, and the iPSYCH-Broad Consortium.
The recent GWAS of ADHD, comprising 38,691 individuals with ADHD and 186,843 controls, identified 27 genome-wide significant loci and estimated that around 7,300 common variants influence ADHD (explaining 90% of the SNP heritability). Common variant risk was enriched across brain regions, and in excitatory and inhibitory neurons. Additionally, we demonstrated that common variant ADHD risk was associated with decreased complex cognition such as verbal reasoning and a range of executive functions in the general population.
In the largest-to-date RVAS of ADHD, including exomes from 8,895 individuals with ADHD and 53,780 controls, we identified the first exome-wide significant genes with increased burden of rare deleterious variants in ADHD. By investigation of protein-protein interactions of the identified risk genes and integration with functional genomic data, we highlight potential biological mechanisms involved in ADHD.
Finally, I will discus results on additivity of rare and common variant ADHD risk, dissection of the genetic architecture across comorbid conditions, and risk prediction of important outcomes.
Conflict of Interest: None declared
S27.3 The genetics of depression: utilising the deep phenotyping of the Australian Genetics of Depression Study
Brittany Mitchell
Almost two decades ago the World Health Organization (WHO) predicted that by 2030, depression would be the single largest cause of disease burden in the world. As we emerge from a period that has included fires, floods, COVID-19 and the resulting economic consequences, evidence indicates that we may meet this unwelcome outcome even sooner. Although there have been significant advances in treatments for depression, increased treatment uptake has not coincided with a reduction in depression-related disability or prevalence. Instead, in the last two decades, depression prevalence has continued to climb. The major challenge inherent in treating depression is that it is not a single disease with a defined underlying cause. Rather, it is a heterogeneous condition that emerges from a complex interplay of biological, psychological, and social processes.
Genome-wide association studies of hundreds of thousands of people with depression have enabled major breakthroughs in our understanding of the genetic aetiology of depression. However, until recently, available research cohorts have lacked sufficient depth of phenotyping to examine the genetic architecture of the clinical heterogeneity observed within MDD. With this in mind, the Australian Genetics of Depression Study (AGDS) was established in 2015 and has recruited 22,424 participants through two enrolment avenues: a targeted assisted mail out campaign (14%) and an open media campaign (86%). Individuals who had been diagnosed with depression were invited to enrol and complete an online instrument assessing their history of depression and other psychiatric disorders. Supplementary online questionnaires achieved in depth phenotyping for a range of psychiatric and behavioural traits.
Here we show results of various studies that have leveraged the depth of phenotyping in the AGDS to explore questions surrounding the genetic contribution to overall depression risk as well as depression heterogeneity, including age of onset, subtypes and sex differences, treatment response, and the impact of phenotyping in genetic studies of depression. In many ways, the AGDS is a proof-of-principle study, demonstrating the utility of online recruitment in psychiatric genetic studies, resulting in one of the largest and deeply phenotyped studies investigating the genetic of depression. Going forward, it is imperative for genetic studies of neuropsychological traits to aim to achieve as deep a phenotyping as possible while still
Conflict of Interest: None declared
S28 Proteomics in human genetics and precision medicine
S28.1 Proteomics results in over 50,000 UK biobank participants
Heiko Runz
Background/Objectives: Evidence across a broad range of diseases supports that drugs against targets with human genetic evidence have a higher probability of regulatory approval. There is therefore an interest across industry and academia to identify and validate genes and gene products that, when manipulated by drugs, will yield safe, efficacious, and transformative therapies for diseases of large unmet medical need.
Methods: To provide the source data for drug target prioritization from human genetics we and others have led and contributed to substantial data generation efforts. These efforts link genetics with detailed health phenotypes and biomarkers in large numbers of individuals. Most recently we determined the levels of thousands of proteins in blood from participants of UK Biobank and FinnGen.
Results: We will discuss how we are utilizing these and other large-scale population data to generate biological insights through causally linking genetic variation to the risk for CNS-related conditions; identifying predictive disease biomarkers through studying the human plasma proteome; and validating hypotheses for therapeutic interventions through customized recall studies and experimental follow-up.
Conclusion: A particular focus will be given to conditions where the joint analysis of rare and common variation, biomarkers and disease endpoints is increasingly blurring the classic distinction between rare Mendelian and common, complex disorders, and how such insights may benefit drug discovery.
Conflict of Interest: None declared
S28.2 Plasma Proteome Variation and its Genetic Determinants in Children and Adolescents
Lili Niu
Background: The dynamics of the plasma proteome during pediatric development are crucial for understanding both normal growth, including pubertal changes, and developmental disorders. Advances in mass spectrometry (MS)-based proteomics now enable extensive population studies, greatly improving our capacity to identify biomarkers and map protein quantitative trait loci (pQTLs) across various body fluids and tissues.
Objective: This presentation will highlight critical findings from the HOLBAEK study, which examines the plasma proteome dynamics and their genetic underpinnings in pediatric populations. Our goal was to delineate protein signatures that are associated with age, sex, BMI standard deviation score (BMI-SDS), obesity, and single nucleotide polymorphisms (SNPs).
Methods: Using the Orbitrap Astral Mass Spectrometer, we analyzed neat plasma proteomes from over 3,000 children and adolescents aged 5-20 years, as well as over 500 adults aged 18-82 years. This high-throughput approach facilitated the quantification of over 1,200 proteins per sample, making it the most comprehensive single-shot MS-based neat plasma proteomics dataset to date.
Results: Our study identified age-dependent protein alterations associated with the insulin growth factor signaling pathway, skeletal development, and angiogenesis, all essential for pediatric growth. We identified distinct trajectories of protein abundance that vary by sex following puberty and novel protein associations with childhood obesity. Integrating genomic and proteomic data, we identified pQTLs for over 400 proteins, supported by robust peptide-level evidence and confirmed in both a pediatric replication set and an independent adult cohort. Additionally, integrating cis-pQTLs with publicly available GWAS summary statistics revealed that dozens of proteins colocalize with cardiometabolic traits and diseases, suggesting their potential causal roles in disease development.
Conclusion: The HOLBAEK study offers novel insights into the plasma proteome variation of children and adolescents. Our findings highlight the relative contributions of genetic and other factors in shaping the proteomic landscape during pediatric growth and provide pivotal insights into the pathophysiology of obesity in young populations. The pQTLs identified in this study including the novel ones expands the existing list of genetic instruments for biomarker identification and drug validation.
Keywords: Pediatric Obesity, Plasma Proteomics, Genetic Determinants, Mass Spectrometry, Developmental Disorders
Conflict of Interest: None declared
S29 AI genomics: uses, ethics and regulatory issues
S29.3 Artificial intelligence in national genomics research: Legal, ethical, and policy landscape
James Hazel
Artificial intelligence (AI), and its range of emerging subfields, holds great potential for advancing genomics and human health research, but relies on access to large-scale, diverse datasets. National genomics and precision medicine initiatives are a source of such data, encompassing genetic information and a widening array of health, lifestyle, and other sensitive data. This presentation will explore how national genomics initiatives worldwide are incorporating AI technologies, including associated legal and ethical issues, and present preliminary findings from a global survey into current and planned AI applications and associated data governance policies.
Conflict of Interest: None declared
S30 Repeat expansion diseases: progress and puzzles
S30.1 Repeats expansion in ataxias: SCA27b - an illustrative, frequent and likely treatable example
Matthis Synofzik
Repeats expansion in ataxias: SCA27b - an illustrative, frequent and likely treatable example
Matthis Synofzik, Hertie-Institute for Clinical Brain Research, Tübingen
Compared to most other neurological and non-neurological diseases, an exceptionally large number of repeat expansion diseases cause ataxia. They present the most frequent molecular disease cause in the by far largest share of genetic ataxia patients. Over the past few years, the implementation of advanced bioinformatics tools and long-read sequencing has allowed the identification of an even larger number of novel ataxia repeat expansion disorders, such as the recently identified spinocerebellar ataxia 27B (SCA27B), caused by deep-intronic GAA repeat expansion in the fibroblast growth factor 14 (FGF14) gene.
Although only identified in late 2023, SCA27B has already been acknowledged as one of most common - if not THE most common - genetic ataxia, accounting for a substantial number (9-61%) of previously undiagnosed cases from different cohorts, and 20% of autosomal-dominant ataxias, as shown by our findings. Surprisingly, it even genetically explains 48% of patients with seemingly “idiopathic” downbeat nystagmus syndrome; and 12% of seemingly “idiopathic” sporadic late-onset ataxias, as demonstrated by our findings from the large SPORTAX cohort- a stratum of ataxia patients seemingly most de-riched for any genetic cause whatsoever. First natural history studies from our group and others have already outlined the progression and core phenotype of this novel disease, demonstrating that it consists of a late-onset slowly progressive pan-cerebellar syndrome that is frequently associated with cerebellar oculomotor signs, such as downbeat nystagmus, and episodic worsening. Yet – unlike in many other repeat expansion ataxias- the number of GAA repeats correlates only very weakly, if at all, with age of onset. This indicates that it is of primary research priority in SCA27B to identify other genetic and non-genetic factors contributing to age of onset, disease penetrance, and disease progression.
Finally, we will hre present an already substantial body of evidence from our group that this frequent repeat expansion disease is even readily treatable with an already marketed drug: in several case series, including exact digital-motor assessments, and now even first larger cohort analyses, SCA27B patients show substantial symptomatic benefits from 4-aminopyridine – with benefits in gait, stance, and speech.
Conflict of Interest: None declared
S30.3 ATTTT/ATTTC repeat expansions in Familial Adult myoclonic epilepsy
Mark Corbett
Background / Objectives: Familial Adult Myoclonic Epilepsy (FAME) is characterised by cortical tremor, myoclonus and myoclonic and / or generalised tonic-clonic seizures with onset in the 2nd to 3rd decade. FAME is caused by non-coding, intronic expansions of a reference TTTTA DNA motif adjacent to an inserted and repeated TTTCA motif in one of seven different genes with unrelated molecular functions. Guidelines for clinical molecular tests for FAME are currently not available, and the pathogenic limits on repeat length or the effects of different motifs within the repeat are not known.
Methods: Multiple FAME loci were screened using long-range PCR and Oxford nanopore DNA sequencing. Lengths of FAME TTTTA, TTTCA and novel repeat motifs directly from sequence data then correlated these to phenotype.
Results: The average length of the entire repeat increased in successive generations in multiple FAME2 families and correlated with younger age of onset of myoclonus (r = -0.329, p = 0.05). There were no significant differences in the magnitude of changes in repeat lengths between maternal or paternal inheritance. Changes in numbers of both TTTTA and TTTCA repeats over successive passages of patient-derived lymphoblastoid and primary skin fibroblast cell lines were dynamic; potentially modelling somatic instability. High levels of motif and repeat length variation at FAME loci were observed in population controls from the gnomAD database and individuals we sequenced.
Conclusion: Our results collectively suggest that TTTTA and TTTCA repeat expansions are dynamic during both meiosis and mitosis, with longer overall length correlating with earlier disease onset.
Conflict of Interest: None declared
Educational SessionsE01 Telomeres in cancer
E01.1 Telomere transcription, TERRA and telomere maintenance (virtual)
Claus M. Azzalin
Telomere elongation enables cell population immortality by preventing cellular senescence triggered by critically short telomeres. Human somatic cells, lacking sustained mechanisms of de novo telomere synthesis, have limited proliferation potential and cease dividing after a finite number of population doublings. Accumulation of senescent cells with critically short or damaged telomeres has been observed in both proliferative and non-proliferative tissues of aging primates, including humans, suggesting that telomere shortening contributes to organismal aging. Conversely, active telomere lengthening mechanisms are a hallmark of cancer cells. About 85% of human cancers maintain telomere length homeostasis through reactivation of the specialized reverse transcriptase telomerase. The remaining 15% utilize the ‘Alternative Lengthening of Telomeres’ (ALT) mechanism, which relies on homology directed repair of shortened telomeres.
Despite their heterochromatic nature, telomeres are transcribed into long noncoding RNA species known as TERRA (TElomeric Repeat-containing RNA). TERRA is transcribed from multiple chromosome ends by RNA polymerase II and a fraction of it remains tightly associated with telomeric loci throughout the cell cycle. Our laboratory has demonstrated that TERRA and telomere transcription play crucial roles in telomeric functions, including telomere elongation in both telomerase-positive and ALT cells. I will provide an overview of data supporting this hypothesis and present unpublished findings suggesting that TERRA reorganizes the composition of the telomeric nucleoprotein, likely to support the repair and elongation of damaged DNA.
Conflict of Interest: None declared
E02 Germline mosaicism: contribution to disease, diagnostics options and counselling
E02.1 Germline mosaicism and transmission risk of ‘apparently’ de novo mutations
Anne Goriely
In this educational session, we will address the general topic of Germline mosaicism. We will review its biological underpinning, describe our current understanding of its contribution to disease and the associated risk of recurrence of conditions caused by de novo mutations (DNMs), the diagnostics options and how to improve reproductive counselling for families who have had a child with a DNM.
Although germline mosaicism is a well-known phenomenon and has long been recognised as a source of DNMs, its exact contribution to spontaneous disease and the associated recurrence risk in future pregnancies remain poorly defined. As a result, couples who have had a child with a pathogenic DNM are typically given a generic recurrence risk of 1-2%. Importantly, this figure represents a population average that is nearly always incorrect for any specific couple. In most cases, the risk is negligible, whereas in a minority (those in which one of the parents is a germline mosaic) the risk might reach 50%.
To discuss this topic, we will draw on the experience gained from the UK-based PREGCARE (PREcision Genetic Counselling And REproduction) study which has developed a systematic approach to providing personalized recurrence risk stratification for 60 families who had a child with a serious developmental disorder caused by an apparent DNM and were seeking individualized reproductive counselling about recurrence risk prior to a future pregnancy (Bernkopf et al 2023. https://doi.org/10.1038/s41467-023-36606-w).
Providing evidence-based estimation of recurrence risk will allow couples to make informed choices about the different diagnostic options available to them and offers the prospect of driving a major transformation in the practice of genetic counselling.
Conflict of Interest: None declared
E02.2 Clonal Mosaicism in Sperm
Martin Breuss
Genomic mosaicism—where some but not all cells within a tissue carry a unique mutation—is typically considered a somatic phenomenon with no impact on the collective human genome. However, if such mosaic mutations occur within the germ cell lineage they can transmit to the next generation and present as a ‘germline mutation’ in offspring; in rare cases, this can have a disastrous impact on a child and cause congenital disorders, such as intellectual disability, epilepsy, or congenital heart disease. Thus, as a contributor to human health, clinical interest in such mutations and their representation in parental germ cells has consistently grown over the last decade.
A subset of clonal mosaic mutations in sperm are detectable through modern sequencing technologies and represent a quantifiable transmission risk to the next generation. Their patterns—including their difference to or absence from somatic tissues—can be understood through the lens of germ cell development. As mutations naturally accumulate throughout this process, every male carries up to dozens of detectable mosaic mutations in their sperm, some of which may convey disease risk; these can be accurately assessed from ejaculates but not from other biological samples like, for instance, blood.
Based on these considerations, direct sperm mosaicism analysis holds promise for clinical applications: 1) in recurrence risk assessment in families with a history of a pathogenic de novo mutation in a prior child. Rather than a flat 1-2% risk derived from population studies, this allows for a personalized stratification based on mosaicism analysis. This information often represents the final puzzle piece in these families’ diagnostic odyssey and can inform future family planning. 2) For mosaicism analysis across the genome for any male prior to conception to determine their transmission risk for potentially pathogenic variants. Here, deep sequencing and specialized analytical methods can quantify all detectable mosaic variants within an individual’s sperm. While technological advances have put this application within reach, ethical and practical hurdles around variant interpretation must be considered carefully when moving such a framework forward.
Together, this educational session will convey basic concepts of clonal mosaicism in sperm, describe its current and potential clinical utility, and discuss the remaining barriers and challenges to its implementation.
Conflict of Interest: None declared
E03 AI meets genomics
E03.1 What is Human-Centered Artificial Intelligence? Exploring its Potential in Facial Genetics
Elisabeth André
Human-centered Artificial Intelligence posits that Artificial Intelligence (AI) and Machine Learning (ML) algorithms should be developed with an understanding that AI systems and humans complement each other, emphasizing that AI operates within environment that involve human interaction. My talk explores the application of human-centered AI in the area of facial phenotyping, where deep neural networks have shown remarkable progress in recognizing facial genetic syndromes, which are often challenging for even experienced clinicians to diagnose. In clinical genetics, where high-stakes decisions are common, clinicians must critically assess the reliability of machine-generated diagnoses. Studies suggest that less experienced clinicians are particularly vulnerable to automation bias, leading them to overtrust AI diagnoses. It is therefore crucial for AI systems to provide transparent explanations of their decision-making processes. This presentation will explore explanation techniques such as saliency maps and counterfactual explanations, which help professionals calibrate their trust in AI. Additionally, I will present findings from eye-tracking studies that reveal differing attention patterns between human experts and deep learning models. These insights are instrumental in refining AI systems to better align with human diagnostic strategies and, conversely, in enhancing human decision-making by revealing the AI’s attention patterns. The overarching goal of this talk is to illustrate how a collaborative relationship between humans and AI can enhance medical diagnostics through a human-centered approach.
Conflict of Interest: None declared
E03.2 PrimateAI-3D: how to make sense of missense variants
Tobias Hamp
Rare genetic variants can have large effects on disease risk, but their use has been limited owing to the difficulty of distinguishing pathogenic from benign variants and estimating the magnitude of their effects. We obtained whole-genome sequencing data from 233 primate species and cataloged 4.3 million common primate missense variants. Then we trained PrimateAI-3D, a semi-supervised 3D-convolutional neural network operating on voxelized protein structures, to separate common primate variants from matched control variants in 3D space. PrimateAI-3D showed the strongest correlation among existing variant interpretation algorithms for predicting the effects of rare human variants on clinical phenotypes and revealed 73% more significant gene-phenotype associations (FDR < 0.05) compared with not using the method. A new rare-variant polygenic risk score (PRS) model combined PrimateAI-3D scores from all genes associated with a phenotype. It had more power at the end of the distribution than common variants PRS models, thus better identified individuals at the greatest risk for disease and may be more relevant for population genetic screening and risk management. A new model that unified our rare- and common-variant PRS models further improved genetic risk prediction for common diseases. We observed that nearly every person carried at least one rare penetrant variant for the phenotypes we examined, demonstrating the utility of personal genome sequencing for otherwise healthy individuals in the general population.
Conflict of Interest: None declared
E04 Genetic Skeletal Disorders (GSD): from diagnosis to treatment
E04.1 Radiographic clues for the diagnosis of Genetic Skeletal Disorders
Gen Nishimura
This presentation aims to review crucial radiological findings for diagnosing prevalent skeletal dysplasias and their differential diagnosis. Over 700 disorders categorized into 41 groups are featured in the current Nosology and Classification of Genetic Skeletal Disorders. The genetic and phenotypic diversity in the large number of disorders might seem overwhelming. In fact, however, we encounter only a limited number of skeletal dysplasias in daily clinical practice. Commonly seen conditions include FGFR3-related disorders (achondroplasia, thanatophoric dysplasia, and hypochondroplasia), type II collagenopathies (spondyloepiphyseal dysplasia congenita, Stickler dysplasia, and Kniest dysplasia), as well as osteogenesis imperfecta. Additionally, TRPV4-related disorders (metatropic dysplasia and spondylometaphyseal dysplasia Kozlowski type), a group of metaphyseal dysplasias, a group of multiple epiphyseal dysplasia, campomelic dysplasia, and hypophosphatasia are occasionally encountered. Once we are familiar with the overall pattern and core skeletal changes of these disorders and their differential diagnosis, we will be able to make a diagnosis of over 90% in the vast array of skeletal dysplasias. Since most skeletal dysplasias occur as a result of defective endochondral ossification, morphological changes ensue at the chondroosseous junction; i.e., meta-epiphyses of tubular bones and metaphyseal equivalents of the spine and flat bones. The radiological consequences of tubular bones include metaphyseal cupping, metaphyseal broadening or dumbbell deformity, metaphyseal dysplasia (metaphyseal irregularity and wide growth plates), and epiphyseal dysplasia (epiphyseal irregularity and delayed ossification). The spine and flat bones imitate the meta-epiphyseal changes of tubular bones.
Conflict of Interest: None declared
E04.2 Tools of the clinical geneticist for the diagnosis of Genetic Skeletal Disorders
Geert Mortier
Genetic skeletal disorders, also known as skeletal dysplasias, are an extremely diverse and complex group of rare genetic diseases that affect the formation, growth and metabolism of hyaline cartilage and bone. Up to date, more than 700 different disorders have been delineated that range in severity from relatively mild to severe or even lethal phenotypes. Common clinical features include severe short stature (“dwarfism”), early-onset degenerative joint disease, pathological fractures, sensory deficits (hearing and/or visual loss) and chronic pain, often starting in childhood. The rare occurrence of these diseases makes an early and correct diagnosis often challenging. Although radiographic evaluation is very crucial in the diagnostic process, a careful clinical examination still remains very important when evaluating an individual suspected of having a skeletal disorder. In my presentation I will discuss which clinical signs are important and useful indicators of a skeletal disorder and explain how a clinician should approach the differential diagnosis of this complex and heterogenous group of disorders. These aspects will be illustrated in an interactive way by presenting some “classical” cases. Nowadays, the clinical geneticist can also rely on the availability of NGS-based technologies (WES, gene panels) for making a timely and correct diagnosis. However, without clinical data, interpretation of VUS may become very difficult, if not impossible. In addition, the clinical geneticist should also be aware that normal results obtained from these analyses, do not always exclude the possibility of a genetic skeletal disorder.
Conflict of Interest: None declared
E04.3 Is there a treatment after diagnosis?
Carlos Ferreira
Previous lectures in this session addressed various modalities for the diagnosis of genetic skeletal disorders, ranging from imaging to molecular studies. Once a diagnosis is established, the option of therapy remains possible for a number of these conditions. In this lecture, we will explore treatment approaches for eight genetic skeletal disorders, including hypophosphatasia, X-linked hypophosphatemia, tumor-induced osteomalacia, osteopetrosis, achondroplasia, fibrodysplasia ossificans progressiva, cartilage-hair hypoplasia and Morquio syndrome. The therapeutic armamentarium includes small molecules, monoclonal antibodies, enzyme replacement therapy, surgical tumoral resection, and hematopoietic stem cell transplantation. Most targeted therapies for genetic skeletal disorders were approved within the last few years, portending a bright future for the expansion of therapies targeting the skeleton in upcoming years.
Conflict of Interest: None declared
E05 Next generation cytogenetics
E05.1 Next generation cytogenetics by optical genome mapping
Rashmi Kanagal-Shamanna
Optical Genome Mapping (OGM) has emerged as a powerful tool for high-resolution genome-wide analysis of structural chromosomal variations at the gene/exon level, so-called next-generation cytogenetics. This educational session provides an overview of the basic principles of the technology, and workflow including analytical methodologies and interpretation. The talk also discusses the clinical and research applications of OGM, both potential and those already in place, based on evidence published in the literature.
Conflict of Interest: None declared
E06 Genetics of Speech Disorders
E06.1 Speech and language phenotyping for gene discovery
Angela Morgan
Speech and language phenotyping and associated genetic landscape in neurodevelopmental syndromes
Angela T. Morgan1,2, Tjitske Kleefstra3,4
1Murdoch Children’s Research Institute, Melbourne, Australia. 2Speech Pathology, School of Health Sciences, University of Melbourne, Australia. 3Department of Clinical Genetics, Erasmus MC, Rotterdam, Netherlands. 4Center for neuropsychiatry, Vincent van Gogh Institute for Psychiatry, Venray, The Netherlands
Speech and language skills are critical for leading a successful and fulfilled life. Communication disorders are common in neurodevelopmental disorders caused by genetic variants, but remain poorly characterised due to a lack of specific speech and language phenotyping. Speech and language difficulties frequently co-occur in children, but these are actually distinct and dissociable skills. In simple terms, speech can be defined as the motor production of a message; and language the ability to understand and produce a message using vocabulary and grammar, in spoken or written form. In particular for genetic neurodevelopmental disorders (rare syndromes), speech and language characterisation may be extremely challenging given the complex somatic, cognitive and behavioural phenotypes that often co-occur.
The literature on genetic neurodevelopmental disorders is currently restricted by conflation of the terms speech and language. This distinction is important because a striking speech presentation of childhood apraxia of speech has recently been shown to have high genetic diagnostic yield (30-40%). Most of the genes associated with relatively isolated speech disorder have already been described in individuals with more significant clinical phenotypes, expanding the genotype-phenotype spectrum for these rare conditions, such as SETD1A and DDX3X-related conditions, and even Kleefstra Syndrome (EHMT1). We will discuss current findings in the literature and provide an engaging and interactive workshop on speech and language in children with monogenic neurodevelopmental disorders.
Attendees will be able to:
-
Describe and dissociate specific speech and language phenotypes including childhood apraxia of speech and dysarthria
-
Implement deep speech phenotyping using the Human Phenotyping Ontology
-
Identify and describe different speech phenotypes in clinical examples of rare genetic conditions
-
Describe the current genetic landscape of known genes associated with severe speech disorder
-
Identify core speech features signalling a red flag for monogenic conditions
Conflict of Interest: None declared
E07 Mental Health in Academia and Healthcare
E07.1 Raising awareness for the Mental Health of early career researchers
Sandra Naumann
Although many academics love their research and experience fulfillment from various tasks of their profession, mounting evidence suggests that working in academia might contribute to mental health problems. Feelings of being an impostor and other unhelpful core beliefs are no rarity within this work environment. Within this talk, the work of the initiative Scholar Minds, which is a Berlin-based initiative to promote mental health awareness and resources for early career researchers (ECRs) will be displayed. Firstly, data collected by the initiative on the status quo of ECRs’ mental health is presented. Secondly, unhelpful thoughts and habits which ultimately impact researchers’ mental health will be discussed. Lastly, first ideas as well as exercises to challenge and overcome these cognitions will be shared.
Conflict of Interest: None declared
E07.2 The Care Under Pressure experience
Daniele Carrieri
The growing incidence of poor mental health and wellbeing in doctors has been widely recognised across healthcare settings in the world, and has been defined as a global crisis. Doctors’ wellbeing is now seen as a key aspect contributing to the achievement of the other three aims of healthcare: improving patient experience and population health, and reducing costs. Therefore, understanding how to support doctors’ wellbeing at work would benefit doctors, patients and the wider healthcare system. However, despite a growing focus on workforce wellbeing, the problem continues, suggesting that current support initiatives are not working optimally to resolve the problem.
In this presentation I will provide some background on this problem; I will draw on Care Under Pressure - a multidisciplinary research programme in the UK (funded by the NIHR) that aims to understand the causes of mental ill-health and well-being in doctors and health professionals and make informed recommendations - and discuss the findings of current research in this area, using also interactive tools such as cartoons and animations.
For more information about Care Under pressure, please visit: https://sites.exeter.ac.uk/careunderpressure/; https://twitter.com/care_under
Conflict of Interest: None declared
E08 FADS - from phenotype to genotype
E08.1Phenotypic spectrum of Fetal akinesia deformation sequence
Johanna I. P. de Vries
FADS is a rare disorder with prevalence of 1:13,000 pregnancies. Within the spectrum of arthrogryposis multiplex congenita (AMC) FADS is one of the severest forms. Hall et al 2019 divided the various forms of AMC into three groups, I. myogenic (amyoplasia), II. distal athrogryposis, III. other,s a.o. FADS. Prenatal detection of AMC and FADS is about 50% when structural anomalies are examined. Attempts to classify the found structural anomalies within the three AMC groups support counselling. Structural anomalies, however, vary by cause, onset and expression and thus are often not visible during the first ultrasound examination. Serial ultrasound examinations after findings associated with FADS can overcome this problem and should be advised. Human phenotypic ontology also emphasizes the importance of precise phenotypic descriptions over time to enhance insight in the underlying disorder and finding publications of genetic anomalies.
Despite the various etiology from genetic, environmental to unknown, there probably is one common feature and that is reduced motility. There is a plea to include systematic motor assessment to distinguish between isolated contractures, AMC and FADS to increase prenatal detection (Tjon et al 2019)
During this educational session various themes will be addressed:
-
How can the clinical geneticist attribute to the prenatal detection of FADS within the multidisciplinary team of obstetricians, and other specialists (neonatologist, paediatric-- neurologist, -physiatrist, -orthopaedic surgeon, pathologist anatomist, social workers).
-
Knowledge of the wide spectrum of FADS changing over time and request of serial ultrasound with targeted ultrasound examination a.o. anatomical structures influenced by reduced motility, motility, brain and heart
-
Uniformity within the hospital in genetic testing, counselling, follow-up examinations, post-mortem examinations with pathologist anatomist and register families with FADS and AMC.
-
Care pathway specified with possibilities within own hospital concerning FADS and AMC.
-
Inform parents about the national arthrogryposis patient support group for information amd possibility of peer contact with parents.
Ultrasound examples of the wide spectrum of abnormal structural and motor findings will be presented.
The set-up of the presentation will enable interaction per theme.
Conflict of Interest: None declared
E08.2 Genomics of arthrogryposis multiplex congenita
Judith Melki
Genomics of Arthrogyposis Multiplex Congenita
Judith Melki, Inserm, Université Paris Saclay, UMR-1195, Le Kremlin-Bicetre, France
Judith.melki@inserm.fr
Arthrogryposis multiplex congenita (AMC) is a developmental condition characterized by joint contractures in two or more body areas resulting from hypo or akinesia. AMC has an overall incidence of 1 in 3000 to 5000. AMC is the direct consequence of reduced fetal movements which may lead, in addition to AMC, to pterygia, pulmonary hypoplasia, diaphragmatic defect or cleft palate. There are multiple causes of AMC including i) genetic defects, ii) congenital infection, iii) extrinsic causes leading to limitations of fetal movement and iv) maternal immune diseases.
The genetic causes of AMC identified to date includes a large spectrum of diseases which arise as a result of pathogenic variants in genes encoding components required for the formation or the function of neuromuscular junctions, skeletal muscle, motor neurons, myelin of peripheral nerve, connective tissue of tendons and joints, or central nervous system including brain with or without spinal cord anomalies.
Our goals were to improve the diagnostic rates of AMC, to evaluate the added value of whole exome sequencing (WES) compared to targeted exome and to identify new genes in 315 unrelated undiagnosed families. Using WES, we achieved a disease gene identification in 61% of AMC cases including 9 new disease genes (CNTNAP1, GLDN, ADCY6, ADGRG6, LGI4, UNC50, LMOD3, MAGEL2 and SCN1A). New genes recently identified in AMC represent 21% of causing genes in our cohort. The most frequent cause of AMC was a primary involvement of skeletal muscle (40%) followed by brain involvement (23%).
In 47.3% of AMC cases, a genetic cause was not established. Whole genome sequencing has shown its superior diagnostic and analytical sensitivity to whole exome sequencing. This strategy should be taken into consideration to evaluate its added value in the AMC cases undiagnosed using WES.
There was a statistically significant difference in gene identification in familial (74%) when compared to sporadic cases (45%), and in sporadic cases, in consanguineous (67%) compared to non-consanguineous cases (40%). The most frequent mode of inheritance of AMC is autosomal recessive (66%). Interestingly, in sporadic non-consanguineous cases, de novo variants were observed in 50% of cases, a proportion similar to that reported in probands with European ancestry from the Deciphering Developmental Disorders Study (DDD, 49.9%). Therefore, even the phenotype of DDD and AMC patients is quite different, a similar and high proportion of de novo variants was observed indicating that this genetic mechanism plays a prominent part in developmental diseases.
The benefits of an accurate genetic diagnosis include a better understanding of prognosis, more tailored management of AMC and possibly other organ involvement and improved surveillance. A precise genetic diagnosis enables the provision of accurate genetic advice to individuals and their families and may provide them with increased reproductive choice, for example, by enabling pre-implantation diagnosis, non-invasive prenatal testing or prenatal diagnosis.
Conflict of Interest: None declared
E09 Understanding variant penetrance in the era of large-scale population-based sequencing studies
E09.2 Phenotypic effects of genetic variants associated with autism
Thomas Bourgeron
The genetic contribution to autism is high (>80% of heritability), but its architecture involves a complex combination of rare and common variants. Autism shares genetic variations with other conditions such as attention deficit hyperactivity disorders (ADHD), intellectual disability, and epilepsy, but little is known about the factors that contribute to the diversity of the clinical trajectories. Remarkably, if rare variants with strong effects are most often associated with intellectual disability, common variants taken altogether are in contrast positively associated with intelligence. In this presentation, I will introduce recent results that shed new light on the inheritance of autism and on some of the underlying mechanisms. For example, I will illustrate how genes associated with autism shape brain connectivity by regulating gene expression and synaptic function. Finally, I will present how we are currently studying Resilience to understand why some carriers of genetic variants seem to be protected from adverse symptoms while others have more difficulties to thrive in the society.
Conflict of Interest: None declared