1
|
Paltenghi C, van Leeuwen J. Genetic suppression interactions are highly conserved across genetically diverse yeast isolates. G3 (BETHESDA, MD.) 2025; 15:jkaf047. [PMID: 40037589 PMCID: PMC12060245 DOI: 10.1093/g3journal/jkaf047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/25/2025] [Indexed: 03/06/2025]
Abstract
Genetic suppression occurs when the phenotypic defects caused by a deleterious mutation are rescued by another mutation. Suppression interactions are of particular interest for genetic diseases, as they identify ways to reduce disease severity, thereby potentially highlighting avenues for therapeutic intervention. To what extent suppression interactions are influenced by the genetic background in which they operate remains largely unknown. However, a high degree of suppression conservation would be crucial for developing therapeutic strategies that target suppressors. To gain an understanding of the effect of the genetic context on suppression, we isolated spontaneous suppressor mutations of temperature-sensitive alleles of SEC17, TAO3, and GLN1 in 3 genetically diverse natural isolates of the budding yeast Saccharomyces cerevisiae. After identifying and validating the genomic variants responsible for suppression, we introduced the suppressors in all 3 genetic backgrounds, as well as in a laboratory strain, to assess their specificity. Ten out of 11 tested suppression interactions were conserved in the 4 yeast strains, although the extent to which a suppressor could rescue the temperature-sensitive mutant varied across genetic backgrounds. These results suggest that suppression mechanisms are highly conserved across genetic contexts, a finding that is potentially reassuring for the development of therapeutics that mimic genetic suppressors.
Collapse
Affiliation(s)
- Claire Paltenghi
- Center for Integrative Genomics, University of Lausanne, Génopode Building, 1015 Lausanne, Switzerland
| | - Jolanda van Leeuwen
- Center for Integrative Genomics, University of Lausanne, Génopode Building, 1015 Lausanne, Switzerland
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, United States
| |
Collapse
|
2
|
Zhu H, Zhang J, Rao S, Durbin MD, Li Y, Lang R, Liu J, Xiao B, Shan H, Meng Z, Wang J, Tang X, Shi Z, Cox LL, Zhao S, Ware SM, Tan TY, de Silva M, Gallacher L, Liu T, Mi J, Zeng C, Zheng HF, Zhang Q, Antonarakis SE, Cox TC, Zhang YB. Common cis-regulatory variation modifies the penetrance of pathogenic SHROOM3 variants in craniofacial microsomia. Genome Res 2025; 35:1065-1079. [PMID: 40234029 PMCID: PMC12047249 DOI: 10.1101/gr.280047.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 03/10/2025] [Indexed: 04/17/2025]
Abstract
Pathogenic coding variants have been identified in thousands of genes, yet the mechanisms underlying the incomplete penetrance in individuals carrying these variants are poorly understood. In this study, in a cohort of 2009 craniofacial microsomia (CFM) patients of Chinese ancestry and 2625 Han Chinese controls, we identified multiple predicted pathogenic coding variants in SHROOM3 in both CFM patients and healthy individuals. We found that the penetrance of CFM correlates with specific haplotype combinations containing likely pathogenic-coding SHROOM3 variants and CFM-associated expression quantitative trait loci (eQTLs) of SHROOM3 expression. Further investigations implicate specific eQTL combinations, such as rs1001322 or rs344131, in combination with other significant CFM-associated eQTLs, which we term combined eQTL phenotype modifiers (CePMods). We additionally show that rs344131, located within a regulatory enhancer region of SHROOM3, demonstrates allele-specific effects on enhancer activity and thus impacts expression levels of the associated SHROOM3 allele harboring any rare coding variant. Our findings also suggest that CePMods may serve as pathogenic determinants, even in the absence of rare deleterious coding variants in SHROOM3 This highlights the critical role of allelic expression in determining the penetrance and severity of craniofacial abnormalities, including microtia and facial asymmetry. Additionally, using quantitative phenotyping, we demonstrate that both microtia and facial asymmetry are present in two separate Shroom3 mouse models, the severity of which is dependent on gene dosage. Our study establishes SHROOM3 as a likely pathogenic gene for CFM and demonstrates eQTLs as determinants of modified penetrance in the manifestation of the disease in individuals carrying likely pathogenic rare coding variants.
Collapse
Affiliation(s)
- Hao Zhu
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Jiao Zhang
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Beijing 100144, China
| | - Soumya Rao
- Department of Oral & Craniofacial Sciences, University of Missouri-Kansas City, Kansas City, Missouri 64108, USA
| | - Matthew D Durbin
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | - Ying Li
- Department of Otolaryngology-Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing 100051, China
| | - Ruirui Lang
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Jiqiang Liu
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Baichuan Xiao
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Hailin Shan
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Ziqiu Meng
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Jinmo Wang
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Xiaokai Tang
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Zhenni Shi
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Liza L Cox
- Department of Oral & Craniofacial Sciences, University of Missouri-Kansas City, Kansas City, Missouri 64108, USA
| | - Shouqin Zhao
- Department of Otolaryngology-Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing 100051, China
| | - Stephanie M Ware
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | - Tiong Y Tan
- Victorian Clinical Genetics Service, Royal Children's Hospital and Department of Pediatrics, University of Melbourne, Victoria 3052, Australia
| | - Michelle de Silva
- Victorian Clinical Genetics Service, Royal Children's Hospital and Department of Pediatrics, University of Melbourne, Victoria 3052, Australia
| | - Lyndon Gallacher
- Victorian Clinical Genetics Service, Royal Children's Hospital and Department of Pediatrics, University of Melbourne, Victoria 3052, Australia
| | - Ting Liu
- Department of Ophthalmology, Daping Hospital, Army Medical University, Chongqing 400000, China
| | - Jie Mi
- Center for Non-Communicable Disease Management, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Changqing Zeng
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Hou-Feng Zheng
- Center for Health and Data Science (CHDS), the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
- Diseases & Population (DaP) Geninfo Laboratory, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
| | - Qingguo Zhang
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Beijing 100144, China
| | - Stylianos E Antonarakis
- Department of Genetic Medicine and Development, University of Geneva Medical Faculty, Geneva 1211, Switzerland;
- Medigenome, Swiss Institute of Genomic Medicine, 1207 Geneva, Switzerland
- iGE3 Institute of Genetics and Genomes in Geneva, Geneva 1211, Switzerland
| | - Timothy C Cox
- Department of Oral & Craniofacial Sciences, University of Missouri-Kansas City, Kansas City, Missouri 64108, USA;
- Department of Pediatrics, University of Missouri-Kansas City, Kansas City, Missouri 64108, USA
| | - Yong-Biao Zhang
- School of Engineering Medicine, Beihang University, Beijing 100191, China;
- Key Laboratory of Big Data-Based Precision Medicine and Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing 100191, China
- National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), Beihang University, Beijing 100083, China
| |
Collapse
|
3
|
Simchoni N, Koide S, Likhite M, Kuchitsu Y, Kadirvel S, Law CS, Elicker BM, Kurra S, Wong MMK, Yuan B, Grossi A, Laxer RM, Volpi S, Dissanayake D, Taguchi T, Beck DB, Vogel TP, Shum AK. The common HAQ STING allele prevents clinical penetrance of COPA syndrome. J Exp Med 2025; 222:e20242179. [PMID: 40014299 PMCID: PMC11867111 DOI: 10.1084/jem.20242179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 02/28/2025] Open
Abstract
COPA syndrome, an autosomal-dominant inborn error of immunity, is nonpenetrant in ∼20% of individuals, with no known mediators of protection. Recent studies implicate STING in the pathogenesis of COPA syndrome. We show that the common HAQ STING allele mediates complete clinical protection. We sequenced 35 individuals with COPA mutations, 26 affected patients and 9 unaffected carriers, finding HAQ STING co-segregation with clinical nonpenetrance. Exome sequencing identified only the mutations comprising HAQ STING as variants shared by unaffected carriers and absent in patients. Experimentally, we found that HAQ STING acts dominantly to dampen COPA-dependent STING signaling. Expressing HAQ STING in patient cells rescued the molecular phenotype of COPA syndrome. Our study is the first report of a common and well-tolerated allele mediating complete clinical protection from a severe genetic disorder. Our findings redefine the diagnostic criteria for COPA syndrome, expose functional differences among STING alleles with broad scientific and clinical implications, and reveal a potential universal gene therapy approach for patients.
Collapse
Affiliation(s)
- Noa Simchoni
- Division of Pulmonary, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Shogo Koide
- Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Maryel Likhite
- Center for Human Genetics and Genomics, NYU Grossman School of Medicine, New York, NY, USA
| | - Yoshihiko Kuchitsu
- Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | | | - Christopher S. Law
- Division of Pulmonary, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Brett M. Elicker
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Santosh Kurra
- Division of Pulmonary, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Margaret Mei-Kay Wong
- Center for Human Genetics and Genomics, NYU Grossman School of Medicine, New York, NY, USA
| | - Bo Yuan
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Alice Grossi
- Laboratorio Genetica e Genomica Delle Malattie Rare, Istituto Giannina Gaslini, Genoa, Italy
| | - Ronald M. Laxer
- Division of Rheumatology, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Canada
- Division of Rheumatology, Department of Medicine, St. Michael’s Hospital, Toronto, Canada
| | - Stefano Volpi
- Unità Operativa Complessa Reumatologia e Malattie Autoinfiammatorie, Istituto di Ricovero e Cura a Carattere Scientifico Istituto Giannina Gaslini, Genoa, Italy
- Dipartimento Di Neuroscienze, Riabilitazione, Oftalmologia, Genetica e Scienze Materno-Infantili, Università Degli Studi Di Genova, Genoa, Italy
| | - Dilan Dissanayake
- Division of Rheumatology, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Tomohiko Taguchi
- Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - David B. Beck
- Center for Human Genetics and Genomics, NYU Grossman School of Medicine, New York, NY, USA
- Division of Rheumatology, Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Tiphanie P. Vogel
- Division of Rheumatology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Center for Human Immunobiology, Texas Children’s Hospital, Houston, TX, USA
| | - Anthony K. Shum
- Division of Pulmonary, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
4
|
Bruselles A, Mancini C, Chiriatti L, Carvetta M, Baroni MC, Cappelletti C, Caraffi SG, Celario M, Ciolfi A, Cordeddu V, De Falco A, Ferilli M, Garavelli L, Leoni C, Meossi C, Niceta M, Onesimo R, Peluso F, Politano D, Priolo M, Radio FC, Santorelli F, Signorini S, Sirchia F, Valente EM, Zampino G, Tartaglia M. Expanding the mutational spectrum of ReNU syndrome: insights into 5' Stem-loop variants. Eur J Hum Genet 2025; 33:432-440. [PMID: 40011755 PMCID: PMC11986017 DOI: 10.1038/s41431-025-01820-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/05/2025] [Accepted: 02/13/2025] [Indexed: 02/28/2025] Open
Abstract
A narrow spectrum of heterozygous variants in RNU4-2, encoding the small nuclear RNA (snRNA) U4, underlies ReNU syndrome, a neurodevelopmental disorder (NDD) characterized by moderate to severe developmental delay (DD), intellectual disability (ID), a distinctive facial gestalt, and multisystem involvement. Pathogenic variants have primarily been reported within an 18-nt critical region contributing to stabilizing the U4/U6 snRNA duplex and proper spliceosome assembly. By combining whole genome sequencing reanalysis and targeted direct sequencing in 190 molecularly unexplained NDD cases, we report on five affected individuals carrying pathogenic/putative pathogenic RNU4-2 variants (2.6%). Three individuals harbored the recurrent pathogenic n.64_65insT variant, while two were heterozygous for private/rare maternally inherited variants (n.30 A > T and n.43_44insT) within the 5' Stem-loop region. Deep clinical phenotyping confirmed a homogeneous constellation of features in all individuals, with global DD, ID, brain malformations, and a recognizable facial gestalt representing core findings. Based on structural homology models and available cryo-EM data, n.30 A > T and n.43_44insT were predicted to disrupt key intra- and inter-molecular interactions critical for spliceosome function. Our findings expand the mutational spectrum of ReNU syndrome, and confirm the 5' Stem-loop as a second mutational hotspot in RNU4-2. We propose that a more complex genetics likely underlies the inheritance of a subset of disease-causing RNU4-2 variants from an apparently unaffected parent. We anticipate a relatively high proportion of pathogenic RNU4-2 variants among individuals with unclassified NDD despite extensive genomic testing, and propose a set of facial gestalt core features as a clinical screening tool to prioritize patients for RNU4-2 analysis.
Collapse
Affiliation(s)
- Alessandro Bruselles
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Cecilia Mancini
- Molecular Genetics and Functional Genomics, Bambino Gesù Children's Hospital, IRCCS, 00146, Rome, Italy
| | - Luigi Chiriatti
- Molecular Genetics and Functional Genomics, Bambino Gesù Children's Hospital, IRCCS, 00146, Rome, Italy
| | - Mattia Carvetta
- Molecular Genetics and Functional Genomics, Bambino Gesù Children's Hospital, IRCCS, 00146, Rome, Italy
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University, 00185, Rome, Italy
| | - Maria Chiara Baroni
- Medical Genetics Unit, Azienda USL-IRCCS di Reggio Emilia, 42123, Reggio Emilia, Italy
- Department of Medical and Surgical Science, University of Bologna, 40126, Bologna, Italy
| | - Camilla Cappelletti
- Molecular Genetics and Functional Genomics, Bambino Gesù Children's Hospital, IRCCS, 00146, Rome, Italy
- Department of Biomedicine and Prevention, University "Tor Vergata", 00173, Rome, Italy
| | | | - Massimiliano Celario
- Department of Brain and Behavioural Sciences, University of Pavia, 27100, Pavia, Italy
- Child Neurology and Psychiatry Unit, IRCCS Mondino Foundation, 27100, Pavia, Italy
| | - Andrea Ciolfi
- Molecular Genetics and Functional Genomics, Bambino Gesù Children's Hospital, IRCCS, 00146, Rome, Italy
| | - Viviana Cordeddu
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Alessandro De Falco
- Molecular Medicine for Neurodegenerative and Neuromuscular Diseases Unit, IRCCS Fondazione Stella Maris, 56128, Calambrone, Pisa, Italy
| | - Marco Ferilli
- Molecular Genetics and Functional Genomics, Bambino Gesù Children's Hospital, IRCCS, 00146, Rome, Italy
- Department of Computer, Control and Management Engineering, Sapienza University, 00185, Rome, Italy
| | - Livia Garavelli
- Medical Genetics Unit, Azienda USL-IRCCS di Reggio Emilia, 42123, Reggio Emilia, Italy
| | - Chiara Leoni
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168, Rome, Italy
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Camilla Meossi
- Molecular Medicine for Neurodegenerative and Neuromuscular Diseases Unit, IRCCS Fondazione Stella Maris, 56128, Calambrone, Pisa, Italy
| | - Marcello Niceta
- Molecular Genetics and Functional Genomics, Bambino Gesù Children's Hospital, IRCCS, 00146, Rome, Italy
| | - Roberta Onesimo
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168, Rome, Italy
| | - Francesca Peluso
- Medical Genetics Unit, Azienda USL-IRCCS di Reggio Emilia, 42123, Reggio Emilia, Italy
| | - Davide Politano
- Department of Brain and Behavioural Sciences, University of Pavia, 27100, Pavia, Italy
- Child Neurology and Psychiatry Unit, IRCCS Mondino Foundation, 27100, Pavia, Italy
| | - Manuela Priolo
- Operative Unit of Medical Genetics and Laboratory of Genetics, AORN A Cardarelli, 80131, Naples, Italy
| | | | - Filippo Santorelli
- Molecular Medicine for Neurodegenerative and Neuromuscular Diseases Unit, IRCCS Fondazione Stella Maris, 56128, Calambrone, Pisa, Italy
| | - Sabrina Signorini
- Department of Brain and Behavioural Sciences, University of Pavia, 27100, Pavia, Italy
| | - Fabio Sirchia
- Department of Molecular Medicine, University of Pavia, 27100, Pavia, Italy
- Medical Genetics Unit, IRCCS San Matteo Foundation, Pavia, 27100, Pavia, Italy
| | - Enza Maria Valente
- Department of Molecular Medicine, University of Pavia, 27100, Pavia, Italy
- Neurogenetics Research Center, IRCCS Mondino Foundation, 27100, Pavia, Italy
| | - Giuseppe Zampino
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168, Rome, Italy
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Marco Tartaglia
- Molecular Genetics and Functional Genomics, Bambino Gesù Children's Hospital, IRCCS, 00146, Rome, Italy.
| |
Collapse
|
5
|
Cowan QT, Gu S, Gu W, Ranzau BL, Simonson TS, Komor AC. Development of multiplexed orthogonal base editor (MOBE) systems. Nat Biotechnol 2025; 43:593-607. [PMID: 38773305 PMCID: PMC11579250 DOI: 10.1038/s41587-024-02240-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 04/10/2024] [Indexed: 05/23/2024]
Abstract
Base editors (BEs) enable efficient, programmable installation of point mutations while avoiding the use of double-strand breaks. Simultaneous application of two or more different BEs, such as an adenine BE (which converts A·T base pairs to G·C) and a cytosine BE (which converts C·G base pairs to T·A), is not feasible because guide RNA crosstalk results in non-orthogonal editing, with all BEs modifying all target loci. Here we engineer both adenine BEs and cytosine BEs that can be orthogonally multiplexed by using RNA aptamer-coat protein systems to recruit the DNA-modifying enzymes directly to the guide RNAs. We generate four multiplexed orthogonal BE systems that enable rates of precise co-occurring edits of up to 7.1% in the same DNA strand without enrichment or selection strategies. The addition of a fluorescent enrichment strategy increases co-occurring edit rates up to 24.8% in human cells. These systems are compatible with expanded protospacer adjacent motif and high-fidelity Cas9 variants, function well in multiple cell types, have equivalent or reduced off-target propensities compared with their parental systems and can model disease-relevant point mutation combinations.
Collapse
Affiliation(s)
- Quinn T Cowan
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA
| | - Sifeng Gu
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA
| | - Wanjun Gu
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep Medicine, and Physiology, University of California San Diego, La Jolla, CA, USA
| | - Brodie L Ranzau
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA
| | - Tatum S Simonson
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep Medicine, and Physiology, University of California San Diego, La Jolla, CA, USA
| | - Alexis C Komor
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
6
|
Zaicenoka M, Ramensky VE, Kiseleva AV, Bukaeva AA, Blokhina AV, Ershova AI, Meshkov AN, Drapkina OM. On Penetrance Estimation in Family, Clinical, and Population Cohorts. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2025; 18:e004816. [PMID: 40151935 DOI: 10.1161/circgen.124.004816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
In recent years, there has been a considerable influx of publications assessing the penetrance of pathogenic variants associated with monogenic diseases with dominant inheritance. As large and diverse groups have been sequenced, it has become clear that incomplete penetrance is common to most hereditary diseases, as numerous molecular, genetic, or environmental factors can cause clinical diversity among the carriers of the same variant. In this review, we discuss some of these factors and focus on the existing approaches to estimating penetrance, depending on the data available and their application to different data sets. We also list some currently available large-scale data sets with penetrance estimates.
Collapse
Affiliation(s)
- Marija Zaicenoka
- Genome and Medical Bioinformatics Laboratory, Institute of Personalized Therapy and Prevention (M.Z., V.E.R.), Moscow, Russia
- Moscow Center for Advanced Studies, Moscow, Russia (M.Z.)
| | - Vasily E Ramensky
- Genome and Medical Bioinformatics Laboratory, Institute of Personalized Therapy and Prevention (M.Z., V.E.R.), Moscow, Russia
- Faculty of Bioengineering and Bioinformatics (V.E.R.), Lomonosov Moscow State University, Moscow, Russia
- Institute for Artificial Intelligence (V.E.R.), Lomonosov Moscow State University, Moscow, Russia
| | - Anna V Kiseleva
- Laboratory of Molecular Genetics, Institute of Personalized Therapy and Prevention (A.V.K.), Moscow, Russia
| | - Anna A Bukaeva
- Laboratory of Clinomics (A.A.B., A.V.B., A.I.E.), Moscow, Russia
| | | | | | - Alexey N Meshkov
- Institute of Personalized Therapy and Prevention (A.N.M.), Moscow, Russia
- National Medical Research Center for Cardiology, Moscow, Russia (A.N.M.)
- Research Center for Medical Genetics, Moscow, Russia (A.N.M.)
- Pirogov Russian National Research Medical University, Moscow, Russia, Moscow, Russia (A.N.M.)
| | - Oxana M Drapkina
- Department of Fundamental and Applied Aspects of Obesity (O.M.D.), Moscow, Russia
- National Medical Research Center for Therapy and Preventive Medicine, Moscow, Russia (O.M.D.)
| |
Collapse
|
7
|
Alonso-Castellano P, Tugores A, Mariño Z, Olveira A, Berenguer M, Huarte MP, Fernández-Ramos JR, Lázaro-Ríos M, González-Diéguez ML, Moreno-Planas JM, Hernández-Guerra M, Fernández-Álvarez P, Delgado-Blanco M, Pinazo-Bandera JM, Romero M, Ampuero J, Masnou-Ridaura H, Cachero A, Vargas V, Gómez-Camarero J, Morillas-Ariño MJ, Molina-Pérez E, Miralpeix A, García-Villarreal L. Low penetrance of frequent ATP7B mutations explains the low prevalence of Wilson disease. Lessons from real-life registries. Dig Liver Dis 2025; 57:443-449. [PMID: 39322449 DOI: 10.1016/j.dld.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/03/2024] [Accepted: 09/03/2024] [Indexed: 09/27/2024]
Abstract
BACKGROUND & AIMS Wilson disease (WD) is a copper metabolism disorder caused by mutations in ATP7B gene, with significant clinical variability. Several studies have analyzed the prevalence and penetrance of mutations. We evaluated both characteristics for our more frequent mutations. METHODS Evaluation of 260 patients from the National Registry: clinical, analytical and genetic data. Estimation of homozygotes and total cases according to Hardy-Weinberg equilibrium and comparison with Registry records. RESULTS The estimated number of homozygotes were higher than registered: p.Met645Arg (1949/6), p.His1069Gln (20/8), p.Leu708Pro (63/24) and p.Gly869Arg (147/0). p.Met645Arg homozygotes presented less cirrhosis at diagnosis, extrahepatic disease and Kayser-Fleischer ring (KFR) and more presymptomatic cases and diagnosis after 40 years of age than p.Leu708Pro and p.His1069Gln homozygotes. p.Met645Arg homozygotes presented more late diagnosis than p.Met645Arg compound heterozygotes. Compound heterozygotes carrying p.Met645Arg or p.Gly869Arg showed less cirrhosis at diagnosis, KFR and neurological symptoms and more hepatic and presymptomatic cases, despite clearly low ceruloplasmin levels. The estimated prevalence was 1:3.785, predicting more than 10.500 patients. CONCLUSIONS The widespread mutations p.Met645Arg and p.Gly869Arg show low penetrance. WD might be underdiagnosed in Spain due to less severe phenotype of the most frequent mutations, a crucial fact to avoid misdiagnosis and to offer early therapy.
Collapse
Affiliation(s)
- Pablo Alonso-Castellano
- Universidad Las Palmas Gran Canaria. Servicio Digestivo, Complejo Hospitalario Universitario Insular Materno Infantil (CHUIMI), Las Palmas de Gran Canaria, Spain.
| | - Antonio Tugores
- Unidad Apoyo Investigación, CHUIMI, Las Palmas de Gran Canaria, Spain.
| | - Zoe Mariño
- LiverUnit, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain.
| | - Antonio Olveira
- Servicio Digestivo, Hospital Universitario La Paz, Madrid, Spain.
| | - Marina Berenguer
- Hospital Universitari i Politècnic La Fe, IISLaFe, Universidad de Valencia y Ciberehd, Valencia, Spain.
| | | | | | | | | | - José M Moreno-Planas
- Servicio de Aparato Digestivo, Complejo Hospitalario Universitario de Albacete, Facultad de Medicina Universidad de Castilla La Mancha, Spain.
| | | | | | | | - José M Pinazo-Bandera
- Unidad de Hepatología, Unidad de Gestión Clínica de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga-Plataforma Bionand, Málaga, Spain.
| | - Marta Romero
- Hospital Universitario de Toledo, Toledo, Spain.
| | - Javier Ampuero
- Hospital Universitario Virgen del Rocío, Sevilla, Spain.
| | | | - Alba Cachero
- Hospital Universitario de Bellvitge, Hospitalet de Llobregat, Spain.
| | - Víctor Vargas
- LiverUnit, Hospital Vall d'Hebron, Universitat Autónoma Barcelona, CIBERehd, Barcelona, Spain.
| | | | | | | | - Anna Miralpeix
- LiverUnit, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain.
| | - Luis García-Villarreal
- Grupo de Investigación Patología Médica, Instituto de Investigaciones Biomédicas y Sanitarias. Universidad de Las Palmas de Gran Canaria. Servicio Digestivo, Complejo Hospitalario Universitario Insular Materno Infantil (CHUIMI), Las Palmas de Gran Canaria, Spain.
| |
Collapse
|
8
|
Nienaber-Rousseau C. Understanding and applying gene-environment interactions: a guide for nutrition professionals with an emphasis on integration in African research settings. Nutr Rev 2025; 83:e443-e463. [PMID: 38442341 PMCID: PMC11723160 DOI: 10.1093/nutrit/nuae015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024] Open
Abstract
Noncommunicable diseases (NCDs) are influenced by the interplay between genetics and environmental exposures, particularly diet. However, many healthcare professionals, including nutritionists and dietitians, have limited genetic background and, therefore, they may lack understanding of gene-environment interactions (GxEs) studies. Even researchers deeply involved in nutrition studies, but with a focus elsewhere, can struggle to interpret, evaluate, and conduct GxE studies. There is an urgent need to study African populations that bear a heavy burden of NCDs, demonstrate unique genetic variability, and have cultural practices resulting in distinctive environmental exposures compared with Europeans or Americans, who are studied more. Although diverse and rapidly changing environments, as well as the high genetic variability of Africans and difference in linkage disequilibrium (ie, certain gene variants are inherited together more often than expected by chance), provide unparalleled potential to investigate the omics fields, only a small percentage of studies come from Africa. Furthermore, research evidence lags behind the practices of companies offering genetic testing for personalized medicine and nutrition. We need to generate more evidence on GxEs that also considers continental African populations to be able to prevent unethical practices and enable tailored treatments. This review aims to introduce nutrition professionals to genetics terms and valid methods to investigate GxEs and their challenges, and proposes ways to improve quality and reproducibility. The review also provides insight into the potential contributions of nutrigenetics and nutrigenomics to the healthcare sphere, addresses direct-to-consumer genetic testing, and concludes by offering insights into the field's future, including advanced technologies like artificial intelligence and machine learning.
Collapse
Affiliation(s)
- Cornelie Nienaber-Rousseau
- Centre of Excellence for Nutrition, North-West University, Potchefstroom, South Africa
- SAMRC Extramural Unit for Hypertension and Cardiovascular Disease, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
| |
Collapse
|
9
|
Lee CL, Chuang CK, Chiu HC, Chang YH, Tu YR, Lo YT, Lin HY, Lin SP. Understanding Genetic Screening: Harnessing Health Information to Prevent Disease Risks. Int J Med Sci 2025; 22:903-919. [PMID: 39991772 PMCID: PMC11843151 DOI: 10.7150/ijms.101219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 12/17/2024] [Indexed: 02/25/2025] Open
Abstract
Genetic screening analyzes an individual's genetic information to assess disease risk and provide personalized health recommendations. This article introduces the public to genetic screening, explaining its definition, principles, history, and common types, including prenatal, newborn, adult disease risk, cancer, and pharmacogenetic screening. It elaborates on the benefits of genetic screening, such as early risk detection, personalized prevention, family risk assessment, and reproductive decision-making. The article also notes limitations, including result interpretation uncertainty, psychological and ethical issues, and privacy and discrimination risks. It provides advice on selecting suitable screening, consulting professionals, choosing reliable institutions, and understanding screening purposes and limitations. Finally, it discusses applying screening results through lifestyle adjustments, regular check-ups, and preventive treatments. By comprehensively introducing genetic screening, the article aims to raise public awareness and encourage utilizing this technology to prevent disease and maintain health.
Collapse
Affiliation(s)
- Chung-Lin Lee
- Department of Pediatrics, MacKay Memorial Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
- International Rare Disease Center, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
- Mackay Junior College of Medicine, Nursing and Management, Taipei, Taiwan
| | - Chih-Kuang Chuang
- Division of Genetics and Metabolism, Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
- College of Medicine, Fu-Jen Catholic University, Taipei, Taiwan
| | - Huei-Ching Chiu
- Department of Pediatrics, MacKay Memorial Hospital, Taipei, Taiwan
| | - Ya-Hui Chang
- Department of Pediatrics, MacKay Memorial Hospital, Taipei, Taiwan
- International Rare Disease Center, MacKay Memorial Hospital, Taipei, Taiwan
| | - Yuan-Rong Tu
- Division of Genetics and Metabolism, Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | - Yun-Ting Lo
- International Rare Disease Center, MacKay Memorial Hospital, Taipei, Taiwan
| | - Hsiang-Yu Lin
- Department of Pediatrics, MacKay Memorial Hospital, Taipei, Taiwan
- International Rare Disease Center, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
- Mackay Junior College of Medicine, Nursing and Management, Taipei, Taiwan
- Division of Genetics and Metabolism, Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Shuan-Pei Lin
- Department of Pediatrics, MacKay Memorial Hospital, Taipei, Taiwan
- International Rare Disease Center, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
- Division of Genetics and Metabolism, Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Infant and Child Care, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| |
Collapse
|
10
|
Cox M, Vitello DJ, Chawla A. The Current Role of Circulating Tumor DNA in the Management of Pancreatic Cancer. J Gastrointest Cancer 2025; 56:44. [PMID: 39808248 DOI: 10.1007/s12029-024-01129-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2024] [Indexed: 01/16/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is projected to be the second leading cause of cancer-related death by 2030. Early identification is rare, with a 5-year overall survival (OS) of less than 10%. Advances in the understanding of PDAC tumor biology are needed to improve these outcomes. Circulating tumor DNA (ctDNA) represents a promising novel biomarker in the identification and management of PDAC. Drawn from peripheral blood and analyzed using a variety of techniques, the detection of ctDNA in PDAC has been associated with shorter OS, minimal residual disease presence, and shorter recurrence-free survival. The use of ctDNA has also been examined as an indicator of therapeutic resistance, susceptibility to targeted therapy, and therapeutic response. While promising, ctDNA analysis is limited by its low rates of detection in some settings and lack of predictive ability in others. Many studies examining the utility of ctDNA for the management of PDAC have been relatively small retrospective cohort studies. The current findings will need to be validated by incorporation of ctDNA analysis into cancer registries and larger prospective studies. Given the current, rapid evolution in the field, it is possible that with time, ctDNA will be more routinely incorporated into the clinical management of PDAC.
Collapse
Affiliation(s)
- Madison Cox
- Division of Surgical Oncology, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Northwestern Medicine Cancer Centers, Northwestern Medicine Regional Medical Group, Winfield, IL, USA
| | - Dominic J Vitello
- Division of Surgical Oncology, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Northwestern Quality Improvement, Research and Education in Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Akhil Chawla
- Division of Surgical Oncology, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA.
- Northwestern Medicine Cancer Centers, Northwestern Medicine Regional Medical Group, Winfield, IL, USA.
- Northwestern Quality Improvement, Research and Education in Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
11
|
Goh S, Thiyagarajan L, Dudding-Byth T, Pinese M, Kirk EP. A systematic review and pooled analysis of penetrance estimates of copy-number variants associated with neurodevelopment. Genet Med 2025; 27:101227. [PMID: 39092588 DOI: 10.1016/j.gim.2024.101227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024] Open
Abstract
PURPOSE Many copy-number variants (CNVs) are reported to cause a variety of neurodevelopmental disabilities including intellectual disability, developmental delay, autism, and other phenotypes with incomplete penetrance. Therefore, not all individuals with a pathogenic CNV are affected. Penetrance estimates vary between studies. A systematic review was conducted to clarify CNV penetrance for 83 recurrent CNVs. METHODS A systematic review using PRISMA guidelines (PROSPERO #CRD42021253955) was conducted to identify penetrance estimates for CNVs associated with neurodevelopment. Pooled analysis was performed using forest plots. The Ottawa Risk of Bias Assessment facilitated evaluation. RESULTS Fifteen studies were reviewed in detail with 9 affected cohorts pooled and compared with the gnomAD v4.0 CNV control cohort of 269,885 individuals. Several CNVs previously associated with nonstatistically significant penetrance estimates now exhibit statistically significant differences, contributing to emerging evidence for their pathogenicity (15q24 duplication [A-D breakpoints], 15q24.2q24.5 deletion and duplication [FBXO22], 17q11.2 duplication [NF1], 17q21.31 duplication [KANSL1] and 22q11.2 distal duplication). Additionally, evidence is presented for the benign nature of some CNVs (15q11.2 duplication [NIPA1] and 2q13 proximal duplication [NPHP1]). CONCLUSION This is a large-scale systematic review of CNVs associated with neurodevelopment. A synopsis analyzing penetrance and pathogenicity is provided for each of the 83 recurrent CNVs.
Collapse
Affiliation(s)
- Shuxiang Goh
- School of Clinical Medicine, University of New South Wales, Sydney, NSW, Australia; New South Wales Health, NSW, Australia.
| | - Lavvina Thiyagarajan
- School of Clinical Medicine, University of New South Wales, Sydney, NSW, Australia; New South Wales Health, NSW, Australia; The Children's Hospital at Westmead, Sydney, NSW, Australia
| | | | - Mark Pinese
- School of Clinical Medicine, University of New South Wales, Sydney, NSW, Australia; Children's Cancer Institute, Sydney, NSW, Australia
| | - Edwin P Kirk
- School of Clinical Medicine, University of New South Wales, Sydney, NSW, Australia; New South Wales Health, NSW, Australia; Centre for Clinical Genetics, Sydney Children's Hospital, Sydney, NSW, Australia; NSW Health Pathology Randwick Genomics Laboratory, Sydney, NSW, Australia
| |
Collapse
|
12
|
Schmidt A, Casadei N, Brand F, Demidov G, Vojgani E, Abolhassani A, Aldisi R, Butler-Laporte G, Alawathurage TM, Augustin M, Bals R, Bellinghausen C, Berger MM, Bitzer M, Bode C, Boos J, Brenner T, Cornely OA, Eggermann T, Erber J, Feldt T, Fuchsberger C, Gagneur J, Göpel S, Haack T, Häberle H, Hanses F, Heggemann J, Hehr U, Hellmuth JC, Herr C, Hinney A, Hoffmann P, Illig T, Jensen BEO, Keitel V, Kim-Hellmuth S, Koehler P, Kurth I, Lanz AL, Latz E, Lehmann C, Luedde T, Maj C, Mian M, Miller A, Muenchhoff M, Pink I, Protzer U, Rohn H, Rybniker J, Scaggiante F, Schaffeldt A, Scherer C, Schieck M, Schmidt SV, Schommers P, Spinner CD, Vehreschild MJGT, Velavan TP, Volland S, Wilfling S, Winter C, Richards JB, Heimbach A, Becker K, Ossowski S, Schultze JL, Nürnberg P, Nöthen MM, Motameny S, Nothnagel M, Riess O, Schulte EC, Ludwig KU. Systematic assessment of COVID-19 host genetics using whole genome sequencing data. PLoS Pathog 2024; 20:e1012786. [PMID: 39715278 PMCID: PMC11706450 DOI: 10.1371/journal.ppat.1012786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 01/07/2025] [Accepted: 11/27/2024] [Indexed: 12/25/2024] Open
Abstract
Courses of SARS-CoV-2 infections are highly variable, ranging from asymptomatic to lethal COVID-19. Though research has shown that host genetic factors contribute to this variability, cohort-based joint analyses of variants from the entire allelic spectrum in individuals with confirmed SARS-CoV-2 infections are still lacking. Here, we present the results of whole genome sequencing in 1,220 mainly vaccine-naïve individuals with confirmed SARS-CoV-2 infection, including 827 hospitalized COVID-19 cases. We observed the presence of autosomal-recessive or likely compound heterozygous monogenic disorders in six individuals, all of which were hospitalized and significantly younger than the rest of the cohort. We did not observe any suggestive causal variants in or around the established risk gene TLR7. Burden testing in the largest population subgroup (i.e., Europeans) suggested nominal enrichments of rare variants in coding and non-coding regions of interferon immune response genes in the overall analysis and male subgroup. Case-control analyses of more common variants confirmed associations with previously reported risk loci, with the key locus at 3p21 reaching genome-wide significance. Polygenic scores accurately captured risk in an age-dependent manner. By enabling joint analyses of different types of variation across the entire frequency spectrum, this data will continue to contribute to the elucidation of COVID-19 etiology.
Collapse
Affiliation(s)
- Axel Schmidt
- Institute of Human Genetics, School of Medicine, University Bonn & University Hospital Bonn, Bonn, Germany
- Department of Pediatric Neurology, School of Medicine, University Bonn & University Hospital Bonn, Bonn, Germany
| | - Nicolas Casadei
- DFG NGS Competence Center Tübingen (NCCT), University of Tübingen, Tübingen, Germany
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Fabian Brand
- Institute of Genomic Statistics and Bioinformatics, School of Medicine, University Bonn & University Hospital Bonn, Bonn, Germany
| | - German Demidov
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
- Institute for Bioinformatics and Medical Informatics (IBMI), University of Tübingen, Tübingen, Germany
| | - Elaheh Vojgani
- Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany
| | - Ayda Abolhassani
- Department of Psychiatry and Psychotherapy, School of Medicine, University Bonn & University Hospital Bonn, Bonn, Germany
| | - Rana Aldisi
- Institute of Genomic Statistics and Bioinformatics, School of Medicine, University Bonn & University Hospital Bonn, Bonn, Germany
| | - Guillaume Butler-Laporte
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | | | | | - Max Augustin
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Robert Bals
- Department of Internal Medicine V, Saarland University, Homburg, Germany
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Germany
| | - Carla Bellinghausen
- Department of Internal Medicine, Pneumology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Marc Moritz Berger
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Michael Bitzer
- Center for Personalized Medicine, University Hospital Tübingen, Tübingen, Germany
- Department of Internal Medicine I, University Hospital Tübingen, Tübingen, Germany
| | - Christian Bode
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Jannik Boos
- Institute of Human Genetics, School of Medicine, University Bonn & University Hospital Bonn, Bonn, Germany
| | - Thorsten Brenner
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Oliver A. Cornely
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
- Clinical Trials Center Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute of Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Thomas Eggermann
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Johanna Erber
- Department of Internal Medicine II, University Hospital rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Torsten Feldt
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Duesseldorf, Medical Faculty, Düsseldorf, Germany
| | | | - Julien Gagneur
- Computational Health Center, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
- School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
| | - Siri Göpel
- Department of Internal Medicine I, University Hospital Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
| | - Tobias Haack
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Helene Häberle
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Frank Hanses
- Department for Infection Control and Infectious Diseases, University Hospital Regensburg, Regensburg, Germany
- Emergency Department, University Hospital Regensburg, Regensburg, Germany
| | - Julia Heggemann
- Institute of Human Genetics, School of Medicine, University Bonn & University Hospital Bonn, Bonn, Germany
| | - Ute Hehr
- Center for Human Genetics Regensburg, Regensburg, Germany
| | - Johannes C. Hellmuth
- COVID-19 Registry of the LMU Munich (CORKUM), University Hospital, LMU Munich, Munich, Germany
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Christian Herr
- Department of Internal Medicine V, Saarland University, Homburg, Germany
| | - Anke Hinney
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Per Hoffmann
- Institute of Human Genetics, School of Medicine, University Bonn & University Hospital Bonn, Bonn, Germany
| | - Thomas Illig
- Hannover Unified Biobank, Hannover Medical School, Hannover, Germany
| | - Björn-Erik Ole Jensen
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Duesseldorf, Medical Faculty, Düsseldorf, Germany
| | - Verena Keitel
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Duesseldorf, Medical Faculty, Düsseldorf, Germany
| | - Sarah Kim-Hellmuth
- Department of Pediatrics, Dr. von Hauner Children’s Hospital, University Hospital LMU Munich, Munich, Germany
- Institute of Translational Genomics, Helmholtz Munich, Neuherberg, Germany
| | - Philipp Koehler
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute of Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Ingo Kurth
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Anna-Lisa Lanz
- Department of Pediatrics, Dr. von Hauner Children’s Hospital, University Hospital LMU Munich, Munich, Germany
| | - Eicke Latz
- Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
| | - Clara Lehmann
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Tom Luedde
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Duesseldorf, Medical Faculty, Düsseldorf, Germany
| | - Carlo Maj
- Center for Human Genetics, Philipps University of Marburg, Marburg, Germany
| | - Michael Mian
- Service for Innovation, Research and Teaching, (SABES-ASDAA), Bolzano-Bozen, Italy; Teaching Hospital of Paracelsus Medical University
| | - Abigail Miller
- Institute of Human Genetics, School of Medicine, University Bonn & University Hospital Bonn, Bonn, Germany
| | - Maximilian Muenchhoff
- COVID-19 Registry of the LMU Munich (CORKUM), University Hospital, LMU Munich, Munich, Germany
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, LMU Munich, Munich, Germany
| | - Isabell Pink
- Department of Pneumology, Hannover Medical School, Hannover, Germany
| | - Ulrike Protzer
- German Center for Infection research (DZIF), Partner Site Munich, Munich, Germany
- Institute of Virology, Technical University Munich/Helmholtz Munich, Munich, Germany
| | - Hana Rohn
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Jan Rybniker
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Federica Scaggiante
- Laboratorio di Patologia Clinica di Bressanone, Hospital of Bressanone (SABES-ASDAA), Bressanone-Brixen, Italy; Teaching Hospital of Paracelsus Medical University
| | - Anna Schaffeldt
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Clemens Scherer
- COVID-19 Registry of the LMU Munich (CORKUM), University Hospital, LMU Munich, Munich, Germany
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | | | | | - Philipp Schommers
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Christoph D. Spinner
- Department of Internal Medicine II, University Hospital rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- German Center for Infection research (DZIF), Partner Site Munich, Munich, Germany
| | - Maria J. G. T. Vehreschild
- Department of Internal Medicine, Infectious Diseases, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Thirumalaisamy P. Velavan
- Institute of Tropical Medicine, Universitätsklinikum Tübingen, Tübingen, Germany
- Vietnamese-German Center for Medical Research (VG-CARE), Hanoi, Vietnam
| | - Sonja Volland
- Hannover Unified Biobank, Hannover Medical School, Hannover, Germany
| | - Sibylle Wilfling
- Center for Human Genetics Regensburg, Regensburg, Germany
- Department of Neurology, Bezirksklinikum Regensburg, University of Regensburg, Regensburg, Germany
| | - Christof Winter
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Clinical Chemistry and Pathobiochemistry, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - J. Brent Richards
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- 5 Prime Sciences Inc, Montréal, Québec, Canada
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montréal, Québec, Canada
- Department of Human Genetics, McGill University, Montréal, Québec, Canada
- Department of Twin Research, King’s College London, London, United Kingdom
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | | | - André Heimbach
- Institute of Human Genetics, School of Medicine, University Bonn & University Hospital Bonn, Bonn, Germany
- NGS Core Facility Bonn, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Kerstin Becker
- Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany
- West German Genome Center ‐ Cologne, University of Cologne, Cologne, Germany
| | - Stephan Ossowski
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
- Institute for Bioinformatics and Medical Informatics (IBMI), University of Tübingen, Tübingen, Germany
| | - Joachim L. Schultze
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
- PRECISE Platform for Genomics and Epigenomics, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) e.V. and University of Bonn, Bonn, Germany
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) e.V., Bonn, Germany
| | - Peter Nürnberg
- Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany
| | - Markus M. Nöthen
- Institute of Human Genetics, School of Medicine, University Bonn & University Hospital Bonn, Bonn, Germany
| | - Susanne Motameny
- Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany
- West German Genome Center ‐ Cologne, University of Cologne, Cologne, Germany
| | - Michael Nothnagel
- Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany
| | - Olaf Riess
- DFG NGS Competence Center Tübingen (NCCT), University of Tübingen, Tübingen, Germany
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Eva C. Schulte
- Institute of Human Genetics, School of Medicine, University Bonn & University Hospital Bonn, Bonn, Germany
- Department of Psychiatry and Psychotherapy, School of Medicine, University Bonn & University Hospital Bonn, Bonn, Germany
- Institute of Virology, Technical University Munich/Helmholtz Munich, Munich, Germany
- Department of Psychiatry & Psychotherapy, University of Munich, Munich, Germany
- Institute of Psychiatric Phenomics and Genomics, University of Munich, Munich, Germany
| | - Kerstin U. Ludwig
- Institute of Human Genetics, School of Medicine, University Bonn & University Hospital Bonn, Bonn, Germany
| |
Collapse
|
13
|
Xu J, Zou Z, Liu W, Zhang Q, Shen J, Hao F, Chen G, Yu D, Li Y, Zhang Z, Qin Y, Yang R, Wang Y, Duan L. HAPLN3 p.T34A contributes to incomplete penetrance of moyamoya disease in Chinese carrying RNF213 p.R4810K. Eur J Neurol 2024; 31:e16473. [PMID: 39315749 PMCID: PMC11555006 DOI: 10.1111/ene.16473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/10/2024] [Accepted: 08/26/2024] [Indexed: 09/25/2024]
Abstract
BACKGROUND AND PURPOSE The penetrance of the RNF213 p.R4810K, a founder mutation of moyamoya disease (MMD), is estimated to be only 1/150-1/300. However, the factors affecting its penetrance remain unclear. Therefore, our study aims to identify modifier genes associated with the incomplete penetrance of this founder mutation. METHODS Whole-exome sequencing (WES) was performed on 36 participants, including 22 MMD patients and 14 non-MMD controls with RNF213 p.R4810K mutation. Fisher's exact test was used to assess the presence of genetic variants that differed significantly between MMD patients and non-MMD controls. In order to exclude false-positive results, another 55 carriers were included to perform Fisher's exact test for the selected sites in the WES discovery stage. Subsequently, human brain microvascular endothelial cells were transfected with wild-type and mutant HAPLN3 for tube formation assays and western blotting to explore the impact of candidate genes on angiogenesis. RESULTS Analysis of variants from WES data revealed a total of 12 non-synonymous variants. Through bioinformatics analysis, the focus was on the HAPLN3 p.T34A variant with a significant p value of 0.00731 in Fisher's exact test. Validation through Sanger sequencing confirmed the presence of this variant in the WES data. In vitro experiments revealed that silencing HAPLN3 and transfecting HAPLN3 p.T34A significantly enhanced tube formation and increased the relative protein expression of vascular endothelial growth factor in endothelial cells. CONCLUSIONS These results suggest that HAPLN3 may function as a modifier gene of RNF213 p.R4810K, promoting the development of MMD and contributing to the incomplete penetrance of MMD founder mutations.
Collapse
Affiliation(s)
- Jun Xu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), Department of Nutrition and Food Hygiene, School of Public HealthChina Medical UniversityShenyangChina
| | - Zhengxing Zou
- Department of Neurosurgery, the Fifth Medical CentreChinese PLA General HospitalBeijingChina
| | - Wanyang Liu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), Department of Nutrition and Food Hygiene, School of Public HealthChina Medical UniversityShenyangChina
| | - Qian Zhang
- Department of Neurosurgery, the Fifth Medical CentreChinese PLA General HospitalBeijingChina
| | - Juan Shen
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), Department of Nutrition and Food Hygiene, School of Public HealthChina Medical UniversityShenyangChina
| | - Fangbin Hao
- Department of Neurosurgery, the Fifth Medical CentreChinese PLA General HospitalBeijingChina
| | - Gan Chen
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), Department of Nutrition and Food Hygiene, School of Public HealthChina Medical UniversityShenyangChina
| | - Dan Yu
- Department of Neurosurgery, the Fifth Medical CentreChinese PLA General HospitalBeijingChina
| | - Yunzhu Li
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), Department of Nutrition and Food Hygiene, School of Public HealthChina Medical UniversityShenyangChina
| | - Zhengshan Zhang
- Department of Neurosurgery, the Fifth Medical CentreChinese PLA General HospitalBeijingChina
| | - Yuchen Qin
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), Department of Nutrition and Food Hygiene, School of Public HealthChina Medical UniversityShenyangChina
| | - Rimiao Yang
- Department of Neurosurgery, the Fifth Medical CentreChinese PLA General HospitalBeijingChina
| | - Yue Wang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), Department of Nutrition and Food Hygiene, School of Public HealthChina Medical UniversityShenyangChina
| | - Lian Duan
- Department of Neurosurgery, the Fifth Medical CentreChinese PLA General HospitalBeijingChina
| |
Collapse
|
14
|
de Oliveira TC, Lopes-Cendes I. Population molecular genetics in Brazil: From genomic databases and research to the implementation of precision medicine. J Community Genet 2024:10.1007/s12687-024-00752-5. [PMID: 39557816 DOI: 10.1007/s12687-024-00752-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 11/05/2024] [Indexed: 11/20/2024] Open
Abstract
Precision medicine (PM) stands on the brink of revolutionizing medical practice throughout the world, holding significant potential for enhancing patient outcomes. However, its practical implementation, particularly in resource-limited countries, is not without challenges. The success of PM largely hinges on the availability of extensive datasets, including genetic and genomic information. This paper delves into the PM landscape and the current state of genetic and genomic testing in Brazil. We also shed light on the unique challenges posed by the country's diverse population and discuss ongoing initiatives to tackle these obstacles.
Collapse
Affiliation(s)
- Thais C de Oliveira
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences, University of Campinas - UNICAMP, Tessália Vieira de Camargo, 126. Cidade Universitária "Zeferino Vaz", Campinas, SP, 13083-888, Brazil
- The Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, Brazil
| | - Iscia Lopes-Cendes
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences, University of Campinas - UNICAMP, Tessália Vieira de Camargo, 126. Cidade Universitária "Zeferino Vaz", Campinas, SP, 13083-888, Brazil.
- The Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, Brazil.
| |
Collapse
|
15
|
Gracia B, Montes P, Huang M, Chen J, Karras GI. HSP90 buffers deleterious genetic variations in BRCA1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.15.623783. [PMID: 39605638 PMCID: PMC11601394 DOI: 10.1101/2024.11.15.623783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Protein-folding chaperone HSP90 buffers genetic variation in diverse organisms, but the clinical significance of HSP90 buffering in disease remains unclear. Here, we show that HSP90 buffers mutations in the BRCT domain of BRCA1. HSP90-buffered BRCA1 mutations encode protein variants that retain interactions with partner proteins and rely on HSP90 for protein stability and function in cell survival. Moreover, HSP90-buffered BRCA1 variants confer PARP inhibitor resistance in cancer cell lines. Low-level HSP90 inhibition alleviates this resistance, revealing a cryptic and mutant-specific HSP90-contingent synthetic lethality. Hence, by stabilizing metastable variants across the entirety of the BRCT domain, HSP90 reduces the clinical severity of BRCA1 mutations allowing them to accumulate in populations. We estimate that HSP90 buffers 11% to 28% of known human BRCA1- BRCT missense mutations. Our work extends the clinical significance of HSP90 buffering to a prevalent class of variations in BRCA1 , pioneering its importance in cancer predisposition and therapy resistance.
Collapse
|
16
|
Alotibi RS, Al Eissa MM, Aloraini T, Nasser KK, Al Shammari MJ, Alqahtani AS. Copy Number Variants in 30 Saudi Pediatric Patients with Neurodevelopmental Disorders: From Unknown Significance to Diagnosis. SAUDI JOURNAL OF MEDICINE & MEDICAL SCIENCES 2024; 12:292-298. [PMID: 39539796 PMCID: PMC11556511 DOI: 10.4103/sjmms.sjmms_155_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 05/19/2024] [Accepted: 07/24/2024] [Indexed: 11/16/2024]
Abstract
Background Structural variants (SVs), such as copy number variants (CNVs), insertions, deletions, inversions, and translocations, contribute significantly to genetic diversity and disease etiology. CNVs, which involve the duplication or deletion of DNA segments, are particularly impactful on genes crucial for biological functions and disease processes. Objective To reassess unclassified SVs that may be underlying unresolved neurodevelopmental disorders among Saudi patients. Methodology In this retrospective study conducted at King Saud Medical City, Riyadh, Saudi Arabia, 30 probands with neurodevelopmental disorders and congenital malformations were examined using next-generation sequencing methods-exome sequencing, gene panels, or SNP arrays (the Illumina platform). Reclassification was aided by online tools such as VarSome and ClinVar, with pathogenicity assessments using the ClinGen CNV Pathogenicity Calculator based on American College of Medical Genetics and Genomics criteria for CNV loss and gain, and dosage sensitivity. Results A total of 31 CNVs were analyzed, of which 2 were reclassified: one as benign and the other as pathogenic. The pathogenic CNV, [3p13p12.3 (70411134_75249376) x1], included a deletion of the FOXP1 gene and was associated with an intellectual developmental disorder, language impairment, possible autistic features, psychomotor impairment, developmental regression, and epilepsy. Conclusion This study underscores the importance of continuously documenting and revisiting unclassified CNVs in accessible databases to enhance the diagnosis and understanding of complex genotype-phenotype relationships. Reclassifying these CNVs not only accelerates diagnostic processes but also enriches our insight into their significant roles in health and disease.
Collapse
Affiliation(s)
- Raniah Saleem Alotibi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Mariam M. Al Eissa
- Department of Molecular Genetics, Public Health Authority, Public Health Laboratory, Riyadh, Saudi Arabia
- Department of Medicine, Al Faisal University, Riyadh, Saudi Arabia
| | - Taghrid Aloraini
- Department of Laboratory Medicine, Division of Translational Pathology, King Abdulaziz Medical City, Riyadh, Saudi Arabia
- Department of Genetics, King Abdullah Specialized Children Hospital, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Khalidah Khalid Nasser
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, Centre for Artificial Intelligence in Precision Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Muneera J. Al Shammari
- Department of Pediatrics, King Saud University Medical City, King Saud University, Riyadh, Saudi Arabia
| | - Amerh S. Alqahtani
- Department of Medical Genetics, King Saud Medical City, Riyadh, Saudi Arabia
| |
Collapse
|
17
|
Green DJ, Michaud V, Lasseaux E, Plaisant C, Fitzgerald T, Birney E, Black GC, Arveiler B, Sergouniotis PI. The co-occurrence of genetic variants in the TYR and OCA2 genes confers susceptibility to albinism. Nat Commun 2024; 15:8436. [PMID: 39349469 PMCID: PMC11443028 DOI: 10.1038/s41467-024-52763-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 09/19/2024] [Indexed: 10/02/2024] Open
Abstract
Although rare genetic conditions are mostly caused by DNA sequence alterations that functionally disrupt individual genes, large-scale studies using genome sequencing have started to unmask additional complexity. Understanding how combinations of variants in different genes shape human phenotypes is expected to provide important insights into the clinical and genetic heterogeneity of rare disorders. Here, we use albinism, an archetypal rare condition associated with hypopigmentation, as an exemplar for the study of genetic interactions. We analyse data from the Genomics England 100,000 Genomes Project alongside a cohort of 1120 individuals with albinism, and investigate the effect of dual heterozygosity for the combination of two established albinism-related variants: TYR:c.1205 G > A (p.Arg402Gln) [rs1126809] and OCA2:c.1327 G > A (p.Val443Ile) [rs74653330]. As each of these changes alone is insufficient to cause disease when present in the heterozygous state, we sought evidence of synergistic effects. We show that, when both variants are present, the probability of receiving a diagnosis of albinism is significantly increased (odds ratio 12.8; 95% confidence interval 6.0 - 24.7; p-value 2.1 ×10-8). Further analyses in an independent cohort, the UK Biobank, support this finding and highlight that heterozygosity for the TYR:c.1205 G > A and OCA2:c.1327 G > A variant combination is associated with statistically significant alterations in visual acuity and central retinal thickness (traits that are considered albinism endophenotypes). The approach discussed in this report opens up new avenues for the investigation of oligogenic patterns in apparently Mendelian disorders.
Collapse
Affiliation(s)
- David J Green
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Vincent Michaud
- Department of Medical Genetics, University Hospital of Bordeaux, Bordeaux, France
- INSERM U1211, Rare Diseases, Genetics and Metabolism, University of Bordeaux, Bordeaux, France
| | - Eulalie Lasseaux
- Department of Medical Genetics, University Hospital of Bordeaux, Bordeaux, France
| | - Claudio Plaisant
- Department of Medical Genetics, University Hospital of Bordeaux, Bordeaux, France
| | - Tomas Fitzgerald
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL- EBI), Wellcome Genome Campus, Cambridge, UK
| | - Ewan Birney
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL- EBI), Wellcome Genome Campus, Cambridge, UK
| | - Graeme C Black
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Manchester Centre for Genomic Medicine, Saint Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Benoît Arveiler
- Department of Medical Genetics, University Hospital of Bordeaux, Bordeaux, France
- INSERM U1211, Rare Diseases, Genetics and Metabolism, University of Bordeaux, Bordeaux, France
| | - Panagiotis I Sergouniotis
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL- EBI), Wellcome Genome Campus, Cambridge, UK.
- Manchester Centre for Genomic Medicine, Saint Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, UK.
- Manchester Royal Eye Hospital, Manchester University NHS Foundation Trust, Manchester, UK.
| |
Collapse
|
18
|
Gracheva AS, Kashatnikova DA, Redkin IV, Zakharchenko VE, Kuzovlev AN, Salnikova LE. Genetics and Traumatic Brain Injury: Findings from an Exome-Based Study of a 50-Patient Case Series. Curr Issues Mol Biol 2024; 46:10351-10368. [PMID: 39329968 PMCID: PMC11430351 DOI: 10.3390/cimb46090616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024] Open
Abstract
Traumatic brain injury (TBI) is the leading cause of global mortality and morbidity. Because TBI is accident-related, the role of genetics in predisposing to TBI has been largely unexplored. However, the likelihood of injury may not be entirely random and may be associated with certain physical and mental characteristics. In this study, we analyzed the exomes of 50 patients undergoing rehabilitation after TBI. Patients were divided into three groups according to rehabilitation outcome: improvement, no change, and deterioration/death. We focused on rare, potentially functional missense and high-impact variants in genes intolerant to these variants. The concordant results from the three independent groups of patients allowed for the suggestion of the existence of a genetic predisposition to TBI, associated with rare functional variations in intolerant genes, with a prevalent dominant mode of inheritance and neurological manifestations in the genetic phenotypes according to the OMIM database. Forty-four of the 50 patients had one or more rare, potentially deleterious variants in one or more neurological genes. Comparison of these results with those of a 50-sampled matched non-TBI cohort revealed significant differences: P = 2.6 × 10-3, OR = 4.89 (1.77-13.47). There were no differences in the distribution of the genes of interest between the TBI patient groups. Our exploratory study provides new insights into the impact of genetics on TBI risk and is the first to address potential genetic susceptibility to TBI.
Collapse
Affiliation(s)
- Alesya S Gracheva
- The Department of Population Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia
- The Laboratory of Clinical Pathophysiology of Critical Conditions, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 107031 Moscow, Russia
| | - Darya A Kashatnikova
- The Laboratory of Ecological Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia
- The Laboratory of Molecular Pathophysiology, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia
| | - Ivan V Redkin
- The Laboratory of Organoprotection in Critical Conditions, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 107031 Moscow, Russia
| | - Vladislav E Zakharchenko
- The Department of Clinical Laboratory Diagnostics, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 107031 Moscow, Russia
| | - Artem N Kuzovlev
- The Laboratory of Clinical Pathophysiology of Critical Conditions, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 107031 Moscow, Russia
| | - Lyubov E Salnikova
- The Laboratory of Clinical Pathophysiology of Critical Conditions, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 107031 Moscow, Russia
- The Laboratory of Ecological Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia
- The Laboratory of Molecular Immunology, National Research Center of Pediatric Hematology, Oncology and Immunology, 117997 Moscow, Russia
| |
Collapse
|
19
|
Nam H, Kim D, Jin X, Park S. Metabolic determinants of leukemia onset variability in genetically homogeneous AKR mice. Biochem Biophys Res Commun 2024; 725:150257. [PMID: 38901226 DOI: 10.1016/j.bbrc.2024.150257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 06/10/2024] [Indexed: 06/22/2024]
Abstract
Leukemia is a complex disease shaped by the intricate interplay of genetic and environmental factors. Given our preliminary data showing different leukemia incidence in genetically homogenous AKR mice harboring the spontaneous leukemia-inducing mutation Rmcfs, we sought to unravel the role of metabolites and gut microbiota in the leukemia penetrance. Our metabolomic analysis revealed distinct serum metabolite profiles between mice that developed leukemia and those that did not. We discovered that linoleic acid (LA), an essential ω-6 polyunsaturated fatty acid, was significantly decreased in the leukemia group, with the lower levels observed starting from 25 weeks before the onset. A predictive model based on LA levels demonstrated high accuracy in predicting leukemia development (area under curve 0.82). In vitro experiment confirmed LA's cytotoxic effects against leukemia cells, and in vivo study showed that a diet enriched with LA prolonged survival in AKR mice. Furthermore, gut microbiome analysis identified specific Lachnospiraceae species, that affect host lipid metabolism, are exclusively present in the leukemia group, suggesting their potential influence on LA metabolism and leukemia development. These findings shed light on the complex relationship between metabolites, gut microbiota, and leukemia development, providing valuable insights into the role of non-genetic factors in leukemia penetrance and potential strategies for leukemia prevention.
Collapse
Affiliation(s)
- Hoonsik Nam
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Doyeon Kim
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Xing Jin
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, Republic of Korea.
| | - Sunghyouk Park
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
20
|
Moundir A, Jeddane L, Bousfiha AA. Insights into the genetic theory of infectious diseases. LA TUNISIE MEDICALE 2024; 102:521-528. [PMID: 39287343 PMCID: PMC11459253 DOI: 10.62438/tunismed.v102i9.4872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 06/08/2024] [Indexed: 09/19/2024]
Abstract
Over the past century, classical approaches from microbiology and immunology have produced spectacular results in the control of infectious diseases. However, the recent SARS-COV-2 pandemic has highlighted our continued failure to control some infections. Other microorganisms still pose a threat to humanity such as HIV, Ebola, and influenza viruses. It seems that conventional approaches are not able to solve all the current problems caused by infectious diseases. Human genetics has shown that infections have a strong genetic determinism that can lead to a predisposition or resistance to infections. This explains much of the clinical variability observed in individuals infected with the same pathogen. The identification of the genetic etiology allows a better understanding of the pathogenesis of infectious diseases and, consequently, the consideration of appropriate preventive and therapeutic strategies. This review provides insights into the genetic theory and the concrete evidence to support it. We highlight the role of primary immunodeficiencies in the discovery of Mendelian and monogenic susceptibility to infections, then we show how genetic and phenotypic heterogeneity, redundancy, and resistance to infection manifest in the context of this genetic determinism. To effectively combat the constant threat of microbes, it is essential to integrate human genetics with microbiology to examine the interactions between pathogens and our immune system.
Collapse
Affiliation(s)
- Abderrahmane Moundir
- Laboratory of Clinical Immunology, Inflammation and Allergies LICIA, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Leila Jeddane
- Laboratory of Clinical Immunology, Inflammation and Allergies LICIA, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
- Laboratoire National de Référence, Mohamed VI Health Sciences University, Casablanca, Morocco
| | - Ahmed Aziz Bousfiha
- Laboratory of Clinical Immunology, Inflammation and Allergies LICIA, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
- Ibn-Rochd University Hospital, Casablanca, Morocco
| |
Collapse
|
21
|
Duijvesteijn N, van der Werf JHJ, Kinghorn BP. Segregation GWAS to linearize a non-additive locus with incomplete penetrance: an example of horn status in sheep. Genet Sel Evol 2024; 56:61. [PMID: 39227755 PMCID: PMC11373182 DOI: 10.1186/s12711-024-00928-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 08/23/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND The objective of this study was to introduce a genome-wide association study (GWAS) in conjunction with segregation analysis on monogenic categorical traits. Genotype probabilities calculated from phenotypes, mode of inheritance and pedigree information, are expressed as the expected allele count (EAC) (range 0 to 2), and are inherited additively, by definition, unlike the original phenotypes, which are non-additive and could be of incomplete penetrance. The EAC are regressed on the single nucleotide polymorphism (SNP) genotypes, similar to an additive GWAS. In this study, horn phenotypes in Merino sheep are used to illustrate the advantages of using the segregation GWAS, a trait believed to be monogenic, affected by dominance, sex-dependent expression and likely affected by incomplete penetrance. We also used simulation to investigate whether incomplete penetrance can cause prediction errors in Merino sheep for horn status. RESULTS Estimated penetrance values differed between the sexes, where males showed almost complete penetrance, especially for horned and polled phenotypes, while females had low penetrance values for the horned status. This suggests that females homozygous for the 'horned allele' have a horned phenotype in only 22% of the cases while 78% will be knobbed or have scurs. The GWAS using EAC on 4001 animals and 510,174 SNP genotypes from the Illumina Ovine high-density (600k) chip gave a stronger association compared to using actual phenotypes. The correlation between the EAC and the allele count of the SNP with the highest -log10(p-value) was 0.73 in males and 0.67 in females. Simulations using penetrance values found by the segregation analyses resulted in higher correlations between the EAC and the causative mutation (0.95 for males and 0.89 for females, respectively), suggesting that the most predictive SNP is not in full LD with the causative mutation. CONCLUSIONS Our results show clear differences in penetrance values between males and female Merino sheep for horn status. Segregation analysis for a trait with mutually exclusive phenotypes, non-additive inheritance, and/or incomplete penetrance can lead to considerably more power in a GWAS because the linearized genotype probabilities are additive and can accommodate incomplete penetrance. This method can be extended to any monogenic controlled categorical trait of which the phenotypes are mutually exclusive.
Collapse
Affiliation(s)
- Naomi Duijvesteijn
- Cooperative Research Centre for Sheep Industry Innovation, Armidale, NSW, 2351, Australia
- School of Environmental and Rural Science, University of New England, Armidale, NSW, 2351, Australia
- Hendrix Genetics Research, Technology & Services B.V., P.O. Box 114, 5830 AC, Boxmeer, The Netherlands
| | - Julius H J van der Werf
- Cooperative Research Centre for Sheep Industry Innovation, Armidale, NSW, 2351, Australia.
- School of Environmental and Rural Science, University of New England, Armidale, NSW, 2351, Australia.
| | - Brian P Kinghorn
- School of Environmental and Rural Science, University of New England, Armidale, NSW, 2351, Australia
| |
Collapse
|
22
|
Serpa F, Finn CM, Tahir UA. Navigating the penetrance and phenotypic spectrum of inherited cardiomyopathies. Heart Fail Rev 2024; 29:873-881. [PMID: 38898187 DOI: 10.1007/s10741-024-10405-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/08/2024] [Indexed: 06/21/2024]
Abstract
Inherited cardiomyopathies are genetic diseases that can lead to heart failure and sudden cardiac death. These conditions tend to run in families, following an autosomal dominant pattern where first-degree relatives have a 50% chance of carrying the pathogenic variant. Despite significant advancements and increased accessibility of genetic testing, accurately predicting the phenotypic expression of these conditions remains challenging due to the inherent variability in their clinical manifestations and the incomplete penetrance observed. This poses challenges in providing patient care and effectively communicating the potential risk of future disease to patients and their families. To address these challenges, this review aims to synthesize the available evidence on penetrance, expressivity, and factors influencing disease expression to improve communication and risk assessment for patients with inherited cardiomyopathies and their family members.
Collapse
Affiliation(s)
- Frans Serpa
- Division of Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Caitlin M Finn
- Division of Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Usman A Tahir
- Division of Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
23
|
Zhao C. Genome wide association study gateway-transitioning variants from association to causality in complex diseases. Sleep 2024; 47:zsae116. [PMID: 38752386 DOI: 10.1093/sleep/zsae116] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024] Open
Affiliation(s)
- Chen Zhao
- Institute of Human Genetics, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Neurogenetic Systems Analysis Group, Institute of Neurogenomics, Helmholtz Munich, Germany
| |
Collapse
|
24
|
Cacheiro P, Lawson S, Van den Veyver IB, Marengo G, Zocche D, Murray SA, Duyzend M, Robinson PN, Smedley D. Lethal phenotypes in Mendelian disorders. Genet Med 2024; 26:101141. [PMID: 38629401 PMCID: PMC11232373 DOI: 10.1016/j.gim.2024.101141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/08/2024] [Accepted: 04/09/2024] [Indexed: 04/26/2024] Open
Abstract
PURPOSE Existing resources that characterize the essentiality status of genes are based on either proliferation assessment in human cell lines, viability evaluation in mouse knockouts, or constraint metrics derived from human population sequencing studies. Several repositories document phenotypic annotations for rare disorders; however, there is a lack of comprehensive reporting on lethal phenotypes. METHODS We queried Online Mendelian Inheritance in Man for terms related to lethality and classified all Mendelian genes according to the earliest age of death recorded for the associated disorders, from prenatal death to no reports of premature death. We characterized the genes across these lethality categories, examined the evidence on viability from mouse models and explored how this information could be used for novel gene discovery. RESULTS We developed the Lethal Phenotypes Portal to showcase this curated catalog of human essential genes. Differences in the mode of inheritance, physiological systems affected, and disease class were found for genes in different lethality categories, as well as discrepancies between the lethal phenotypes observed in mouse and human. CONCLUSION We anticipate that this resource will aid clinicians in the diagnosis of early lethal conditions and assist researchers in investigating the properties that make these genes essential for human development.
Collapse
Affiliation(s)
- Pilar Cacheiro
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Samantha Lawson
- ITS Research, Queen Mary University of London, London, United Kingdom
| | - Ignatia B Van den Veyver
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX; Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX
| | - Gabriel Marengo
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - David Zocche
- North West Thames Regional Genetics Service, Northwick Park and St Mark's Hospitals, London, United Kingdom
| | | | - Michael Duyzend
- Massachusetts General Hospital, Boston, MA; Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA; Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, MA
| | - Peter N Robinson
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Damian Smedley
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom.
| |
Collapse
|
25
|
de Masfrand S, Cogné B, Nizon M, Deb W, Goldenberg A, Lecoquierre F, Nicolas G, Bournez M, Vitobello A, Mau-Them FT, le Guyader G, Bilan F, Bauer P, Zweier C, Piard J, Pasquier L, Bézieau S, Gerard B, Faivre L, Saugier-Veber P, Piton A, Isidor B. Penetrance, variable expressivity and monogenic neurodevelopmental disorders. Eur J Med Genet 2024; 69:104932. [PMID: 38453051 DOI: 10.1016/j.ejmg.2024.104932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 03/01/2024] [Accepted: 03/02/2024] [Indexed: 03/09/2024]
Abstract
PURPOSE Incomplete penetrance is observed for most monogenic diseases. However, for neurodevelopmental disorders, the interpretation of single and multi-nucleotide variants (SNV/MNVs) is usually based on the paradigm of complete penetrance. METHOD From 2020 to 2022, we proposed a collaboration study with the French molecular diagnosis for intellectual disability network. The aim was to recruit families for whom the index case, diagnosed with a neurodevelopmental disorder, was carrying a pathogenic or likely pathogenic variant for an OMIM morbid gene and inherited from an asymptomatic parent. Grandparents were analyzed when available for segregation study. RESULTS We identified 12 patients affected by a monogenic neurodevelopmental disorder caused by likely pathogenic or pathogenic variant (SNV/MNV) inherited from an asymptomatic parent. These genes were usually associated with de novo variants. The patients carried different variants (1 splice-site variant, 4 nonsense and 7 frameshift) in 11 genes: CAMTA1, MBD5, KMT2C, KMT2E, ZMIZ1, MN1, NDUFB11, CUL3, MED13, ARID2 and RERE. Grandparents have been tested in 6 families, and each time the variant was confirmed de novo in the healthy carrier parent. CONCLUSION Incomplete penetrance for SNV and MNV in neurodevelopmental disorders might be more frequent than previously thought. This point is crucial to consider for interpretation of variants, family investigation, genetic counseling, and prenatal diagnosis. Molecular mechanisms underlying this incomplete penetrance still need to be identified.
Collapse
Affiliation(s)
- Servane de Masfrand
- Nantes Université, CHU Nantes, Service de Génétique Médicale, 44000, Nantes, France.
| | - Benjamin Cogné
- Nantes Université, CHU Nantes, Service de Génétique Médicale, 44000, Nantes, France; Nantes Université, CHU Nantes, CNRS, INSERM, L'institut Du Thorax, 44000 Nantes, France
| | - Mathilde Nizon
- Nantes Université, CHU Nantes, Service de Génétique Médicale, 44000, Nantes, France; Nantes Université, CHU Nantes, CNRS, INSERM, L'institut Du Thorax, 44000 Nantes, France
| | - Wallid Deb
- Nantes Université, CHU Nantes, Service de Génétique Médicale, 44000, Nantes, France; Nantes Université, CHU Nantes, CNRS, INSERM, L'institut Du Thorax, 44000 Nantes, France
| | - Alice Goldenberg
- CHU Rouen, Service de Génétique et Centre de Référence pour Les Troubles Du Développement, 76183, Rouen, France
| | - François Lecoquierre
- CHU Rouen, Service de Génétique et Centre de Référence pour Les Troubles Du Développement, 76183, Rouen, France
| | - Gaël Nicolas
- CHU Rouen, Service de Génétique et Centre de Référence pour Les Troubles Du Développement, 76183, Rouen, France
| | - Marie Bournez
- Centre de Référence Anomalies Du Développement et Syndromes Malformatifs, FHU TRANSLAD, CHU Dijon, 21000, Dijon, France
| | - Antonio Vitobello
- Unité Fonctionnelle Innovation en Diagnostic Génomique des Maladies Rares, CHU Dijon Bourgogne, Dijon, France; Génétique des Anomalies Du Développement, INSERM 123, Université de Bourgogne, Dijon, France
| | - Frédéric Tran Mau-Them
- Unité Fonctionnelle Innovation en Diagnostic Génomique des Maladies Rares, CHU Dijon Bourgogne, Dijon, France; Génétique des Anomalies Du Développement, INSERM 123, Université de Bourgogne, Dijon, France
| | - Gwenaël le Guyader
- Service de Génétique Clinique, Centre de Compétence Maladies Rares Anomalies Du Développement, CHU de Poitiers, Poitiers, France
| | - Frédéric Bilan
- Service de Génétique Clinique, Centre de Compétence Maladies Rares Anomalies Du Développement, CHU de Poitiers, Poitiers, France
| | | | | | - Juliette Piard
- Centre de Génétique Humaine and Integrative and Cognitive Neuroscience Research Unit EA481, Université de Franche-Comté, Besançon, France
| | | | - Stéphane Bézieau
- Nantes Université, CHU Nantes, Service de Génétique Médicale, 44000, Nantes, France; Nantes Université, CHU Nantes, CNRS, INSERM, L'institut Du Thorax, 44000 Nantes, France
| | - Bénédicte Gerard
- Laboratoire de Diagnostic Génétique, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Laurence Faivre
- Centre de Référence Anomalies Du Développement et Syndromes Malformatifs, FHU TRANSLAD, CHU Dijon, 21000, Dijon, France; Génétique des Anomalies Du Développement, INSERM 123, Université de Bourgogne, Dijon, France
| | - Pascale Saugier-Veber
- CHU Rouen, Service de Génétique et Centre de Référence pour Les Troubles Du Développement, 76183, Rouen, France
| | - Amélie Piton
- Laboratoire de Diagnostic Génétique, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Bertrand Isidor
- Nantes Université, CHU Nantes, Service de Génétique Médicale, 44000, Nantes, France; Nantes Université, CHU Nantes, CNRS, INSERM, L'institut Du Thorax, 44000 Nantes, France.
| |
Collapse
|
26
|
Jiang JH, Tian J, Pan B. Noteworthy phenomena in pediatric inherited cardiomyopathy. World J Pediatr 2024; 20:635-637. [PMID: 38896415 DOI: 10.1007/s12519-024-00825-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Affiliation(s)
- Jin-Hang Jiang
- Department of Pediatric Cardiology, National Clinical Key Cardiovascular Specialty, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, 136 Zhongshan Er Road, Yuzhong Distirct, Chongqing, 400014, China
| | - Jie Tian
- Department of Pediatric Cardiology, National Clinical Key Cardiovascular Specialty, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, 136 Zhongshan Er Road, Yuzhong Distirct, Chongqing, 400014, China
- Key Laboratory of Children's Important Organ Development and Diseases, Chongqing Municipal Health Commission, Chongqing, China
| | - Bo Pan
- Department of Pediatric Cardiology, National Clinical Key Cardiovascular Specialty, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, 136 Zhongshan Er Road, Yuzhong Distirct, Chongqing, 400014, China.
- Key Laboratory of Children's Important Organ Development and Diseases, Chongqing Municipal Health Commission, Chongqing, China.
| |
Collapse
|
27
|
Murray M, Davidson L, Ferenbach AT, Lefeber D, van Aalten DMF. Neuroectoderm phenotypes in a human stem cell model of O-GlcNAc transferase associated with intellectual disability. Mol Genet Metab 2024; 142:108492. [PMID: 38759397 DOI: 10.1016/j.ymgme.2024.108492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/03/2024] [Accepted: 05/07/2024] [Indexed: 05/19/2024]
Abstract
Pathogenic variants in the O-GlcNAc transferase gene (OGT) have been associated with a congenital disorder of glycosylation (OGT-CDG), presenting with intellectual disability which may be of neuroectodermal origin. To test the hypothesis that pathology is linked to defects in differentiation during early embryogenesis, we developed an OGT-CDG induced pluripotent stem cell line together with isogenic control generated by CRISPR/Cas9 gene-editing. Although the OGT-CDG variant leads to a significant decrease in OGT and O-GlcNAcase protein levels, there were no changes in differentiation potential or stemness. However, differentiation into ectoderm resulted in significant differences in O-GlcNAc homeostasis. Further differentiation to neuronal stem cells revealed differences in morphology between patient and control lines, accompanied by disruption of the O-GlcNAc pathway. This suggests a critical role for O-GlcNAcylation in early neuroectoderm architecture, with robust compensatory mechanisms in the earliest stages of stem cell differentiation.
Collapse
Affiliation(s)
- Marta Murray
- Division of Molecular, Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Lindsay Davidson
- Division of Molecular, Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Andrew T Ferenbach
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, DK, Denmark
| | - Dirk Lefeber
- Department of Neurology, Department of Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, NL, the Netherlands
| | - Daan M F van Aalten
- Division of Molecular, Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, UK; Department of Molecular Biology and Genetics, Aarhus University, Aarhus, DK, Denmark.
| |
Collapse
|
28
|
Lu D, Yang J, Shen W, Chen M. Attitudes toward uncertain results from prenatal exome sequencing: a national survey among healthcare professionals working in the prenatal setting. Front Med (Lausanne) 2024; 11:1335649. [PMID: 38813373 PMCID: PMC11133618 DOI: 10.3389/fmed.2024.1335649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 04/30/2024] [Indexed: 05/31/2024] Open
Abstract
Objective The objective of this study was to investigate the attitudes of healthcare professionals (HPs) working in the prenatal setting toward uncertain results (UR) from prenatal exome sequencing (pES) in China. Methods We conducted a national survey among HPs working in the prenatal setting. UR in our study include variants of uncertain significance (VUS), variants with variable penetrance/expressivity (VVPE), and secondary findings unrelated to the indication for testing (SFs). A total of 285 questionnaires that met the inclusion criteria were collected. Data were analyzed using IBM SPSS Statistics 26. Results When performing the pre-test counseling, only 7.4% of HPs mentioned the possibility of VUS, 6.3% discussed the possibility of VVPE, and 7.4% introduced the SFs with parents with the option to not report these variants. In post-test counseling, 73.0-82.8% HPs discussed with the parents but did not make any recommendations for managing the pregnancy after reporting UR (73.0% for VUS, 82.8% for VVPE, 74.7% for SFs, respectively). Conclusion Most parents did not have the option of opting out of reporting UR from pES in pre-test counseling. UR did not influence the pregnancy recommendation made by most HPs. Establishing national guidelines for reporting UR from pES and developing strategies to improve counseling skills may help HPs manage UR.
Collapse
Affiliation(s)
- Dongfang Lu
- Department of Obstetrics, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Jing Yang
- Department of Obstetrics, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Wei Shen
- Department of Obstetrics, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Min Chen
- Department of Obstetrics and Gynecology, Department of Fetal Medicine and Prenatal Diagnosis, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
29
|
Chen C, Liu Y, Yang S, Chen M, Liao J. A bibliometric and visual analysis of research trends and hotspots of familial hypertrophic cardiomyopathy: A review. Medicine (Baltimore) 2024; 103:e37969. [PMID: 38701258 PMCID: PMC11062727 DOI: 10.1097/md.0000000000037969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 03/29/2024] [Indexed: 05/05/2024] Open
Abstract
Familial hypertrophic cardiomyopathy (FHCM) is an inherited cardiac disease caused by mutations of sarcomere proteins and can be the underlining substrate for major cardiovascular events. Early identification and diagnosis of FHCM are essential to reduce sudden cardiac death. So, this paper summarized the current knowledge on FHCM, and displayed the analysis via bibliometrics method. The relevant literature on FHCM were screened searched via the Web of Science Core Collection database from 2012 to 2022. The literatures were was summarized and analyzed via the bibliometrics method analyzed via CiteSpace and VOSviewer according to topic categories, distribution of spatiotemporal omics and authors, as well as references. Since 2012, there are 909 research articles and reviews related to FHCM. The number of publication for the past 10 years have shown that the development of FHCM research has been steady, with the largest amount of literature in 2012. The most published papers were from the United States, followed by the United Kingdom and Italy. The University of London (63 papers) was the institution that published the most research articles, followed by Harvard University (45 papers) and University College London (45 papers). Keywords formed 3 clusters, focused on the pathogenesis of FHCM, the diagnosis of FHCM, FHCM complications, respectively. The bibliometric analysis and visualization techniques employed herein highlight key trends and focal points in the field, predominantly centered around FHCM's pathogenesis, diagnostic approaches, and its complications. These insights are instrumental in steering future research directions in this area.
Collapse
Affiliation(s)
- Cong Chen
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Yang Liu
- College of Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Songwei Yang
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Ming Chen
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Jing Liao
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
30
|
Jenkins D. How do stochastic processes and genetic threshold effects explain incomplete penetrance and inform causal disease mechanisms? Philos Trans R Soc Lond B Biol Sci 2024; 379:20230045. [PMID: 38432317 PMCID: PMC10909503 DOI: 10.1098/rstb.2023.0045] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 01/16/2024] [Indexed: 03/05/2024] Open
Abstract
Incomplete penetrance is the rule rather than the exception in Mendelian disease. In syndromic monogenic disorders, phenotypic variability can be viewed as the combination of incomplete penetrance for each of multiple independent clinical features. Within genetically identical individuals, such as isogenic model organisms, stochastic variation at molecular and cellular levels is the primary cause of incomplete penetrance according to a genetic threshold model. By defining specific probability distributions of causal biological readouts and genetic liability values, stochasticity and incomplete penetrance provide information about threshold values in biological systems. Ascertainment of threshold values has been achieved by simultaneous scoring of relatively simple phenotypes and quantitation of molecular readouts at the level of single cells. However, this is much more challenging for complex morphological phenotypes using experimental and reductionist approaches alone, where cause and effect are separated temporally and across multiple biological modes and scales. Here I consider how causal inference, which integrates observational data with high confidence causal models, might be used to quantify the relative contribution of different sources of stochastic variation to phenotypic diversity. Collectively, these approaches could inform disease mechanisms, improve predictions of clinical outcomes and prioritize gene therapy targets across modes and scales of gene function. This article is part of a discussion meeting issue 'Causes and consequences of stochastic processes in development and disease'.
Collapse
Affiliation(s)
- Dagan Jenkins
- Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| |
Collapse
|
31
|
Gravel B, Renaux A, Papadimitriou S, Smits G, Nowé A, Lenaerts T. Prioritization of oligogenic variant combinations in whole exomes. Bioinformatics 2024; 40:btae184. [PMID: 38603604 PMCID: PMC11037482 DOI: 10.1093/bioinformatics/btae184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 01/29/2024] [Accepted: 04/10/2024] [Indexed: 04/13/2024] Open
Abstract
MOTIVATION Whole exome sequencing (WES) has emerged as a powerful tool for genetic research, enabling the collection of a tremendous amount of data about human genetic variation. However, properly identifying which variants are causative of a genetic disease remains an important challenge, often due to the number of variants that need to be screened. Expanding the screening to combinations of variants in two or more genes, as would be required under the oligogenic inheritance model, simply blows this problem out of proportion. RESULTS We present here the High-throughput oligogenic prioritizer (Hop), a novel prioritization method that uses direct oligogenic information at the variant, gene and gene pair level to detect digenic variant combinations in WES data. This method leverages information from a knowledge graph, together with specialized pathogenicity predictions in order to effectively rank variant combinations based on how likely they are to explain the patient's phenotype. The performance of Hop is evaluated in cross-validation on 36 120 synthetic exomes for training and 14 280 additional synthetic exomes for independent testing. Whereas the known pathogenic variant combinations are found in the top 20 in approximately 60% of the cross-validation exomes, 71% are found in the same ranking range when considering the independent set. These results provide a significant improvement over alternative approaches that depend simply on a monogenic assessment of pathogenicity, including early attempts for digenic ranking using monogenic pathogenicity scores. AVAILABILITY AND IMPLEMENTATION Hop is available at https://github.com/oligogenic/HOP.
Collapse
Affiliation(s)
- Barbara Gravel
- Interuniversity Institute of Bioinformatics in Brussels, Université Libre de Bruxelles-Vrije Universiteit Brussel, 1050 Brussels, Belgium
- Department of Computer Science, Machine Learning Group, Université Libre de Bruxelles, 1050 Brussels, Belgium
- Department of Computer Science, Artificial Intelligence Laboratory, Vrije Universiteit Brussels, 1050 Brussels, Belgium
| | - Alexandre Renaux
- Interuniversity Institute of Bioinformatics in Brussels, Université Libre de Bruxelles-Vrije Universiteit Brussel, 1050 Brussels, Belgium
- Department of Computer Science, Machine Learning Group, Université Libre de Bruxelles, 1050 Brussels, Belgium
- Department of Computer Science, Artificial Intelligence Laboratory, Vrije Universiteit Brussels, 1050 Brussels, Belgium
| | - Sofia Papadimitriou
- Interuniversity Institute of Bioinformatics in Brussels, Université Libre de Bruxelles-Vrije Universiteit Brussel, 1050 Brussels, Belgium
- Department of Computer Science, Machine Learning Group, Université Libre de Bruxelles, 1050 Brussels, Belgium
- Brussels Interuniversity Genomics High Throughput core (BRIGHTcore), UZ Brussel, Vrije Universiteit Brussel (VUB) - Université Libre de Bruxelles (ULB), 1090 Brussels, Belgium
| | - Guillaume Smits
- Interuniversity Institute of Bioinformatics in Brussels, Université Libre de Bruxelles-Vrije Universiteit Brussel, 1050 Brussels, Belgium
- Center of Human Genetics, Hôpital Erasme, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Ann Nowé
- Interuniversity Institute of Bioinformatics in Brussels, Université Libre de Bruxelles-Vrije Universiteit Brussel, 1050 Brussels, Belgium
- Department of Computer Science, Artificial Intelligence Laboratory, Vrije Universiteit Brussels, 1050 Brussels, Belgium
| | - Tom Lenaerts
- Interuniversity Institute of Bioinformatics in Brussels, Université Libre de Bruxelles-Vrije Universiteit Brussel, 1050 Brussels, Belgium
- Department of Computer Science, Machine Learning Group, Université Libre de Bruxelles, 1050 Brussels, Belgium
- Department of Computer Science, Artificial Intelligence Laboratory, Vrije Universiteit Brussels, 1050 Brussels, Belgium
| |
Collapse
|
32
|
Hu X, Wang W, Luo K, Dai J, Zhang Y, Wan Z, He W, Zhang S, Yang L, Tan Q, Li W, Zhang Q, Gong F, Lu G, Tan YQ, Lin G, Du J. Extended application of PGT-M strategies for small pathogenic CNVs. J Assist Reprod Genet 2024; 41:739-750. [PMID: 38263474 PMCID: PMC10957852 DOI: 10.1007/s10815-024-03028-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 01/04/2024] [Indexed: 01/25/2024] Open
Abstract
PURPOSE The preimplantation genetic testing for aneuploidy (PGT-A) platform is not currently available for small copy-number variants (CNVs), especially those < 1 Mb. Through strategies used in PGT for monogenic disease (PGT-M), this study intended to perform PGT for families with small pathogenic CNVs. METHODS Couples who carried small pathogenic CNVs and underwent PGT at the Reproductive and Genetic Hospital of CITIC-Xiangya (Hunan, China) between November 2019 and April 2023 were included in this study. Haplotype analysis was performed through two platforms (targeted sequencing and whole-genome arrays) to identify the unaffected embryos, which were subjected to transplantation. Prenatal diagnosis using amniotic fluid was performed during 18-20 weeks of pregnancy. RESULTS PGT was successfully performed for 20 small CNVs (15 microdeletions and 5 microduplications) in 20 families. These CNVs distributed on chromosomes 1, 2, 6, 7, 13, 15, 16, and X with sizes ranging from 57 to 2120 kb. Three haplotyping-based PGT-M strategies were applied. A total of 89 embryos were identified in 25 PGT cycles for the 20 families. The diagnostic yield was 98.9% (88/89). Nineteen transfers were performed for 17 women, resulting in a 78.9% (15/19) clinical pregnancy rate after each transplantation. Of the nine women who had healthy babies, eight accepted prenatal diagnosis and the results showed no related pathogenic CNVs. CONCLUSION Our results show that the extended haplotyping-based PGT-M strategy application for small pathogenic CNVs compensated for the insufficient resolution of PGT-A. These three PGT-M strategies could be applied to couples with small pathogenic CNVs.
Collapse
Affiliation(s)
- Xiao Hu
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
| | - Weili Wang
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, 410078, China
| | - Keli Luo
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
| | - Jing Dai
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
| | - Yi Zhang
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
| | - Zhenxing Wan
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
| | - Wenbin He
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, 410078, China
- College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Shuoping Zhang
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
| | - Lanlin Yang
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
| | - Qin Tan
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
| | - Wen Li
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
- College of Life Science, Hunan Normal University, Changsha, 410081, China
- Key Laboratory of Stem Cell and Reproduction Engineering, Ministry of Health, Changsha, 410000, China
| | - Qianjun Zhang
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, 410078, China
- College of Life Science, Hunan Normal University, Changsha, 410081, China
- Key Laboratory of Stem Cell and Reproduction Engineering, Ministry of Health, Changsha, 410000, China
| | - Fei Gong
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
- College of Life Science, Hunan Normal University, Changsha, 410081, China
- Key Laboratory of Stem Cell and Reproduction Engineering, Ministry of Health, Changsha, 410000, China
| | - Guangxiu Lu
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
- Key Laboratory of Stem Cell and Reproduction Engineering, Ministry of Health, Changsha, 410000, China
| | - Yue-Qiu Tan
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, 410078, China
- College of Life Science, Hunan Normal University, Changsha, 410081, China
- Key Laboratory of Stem Cell and Reproduction Engineering, Ministry of Health, Changsha, 410000, China
| | - Ge Lin
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China.
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, 410078, China.
- College of Life Science, Hunan Normal University, Changsha, 410081, China.
- Key Laboratory of Stem Cell and Reproduction Engineering, Ministry of Health, Changsha, 410000, China.
| | - Juan Du
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410008, China.
- College of Life Science, Hunan Normal University, Changsha, 410081, China.
- Key Laboratory of Stem Cell and Reproduction Engineering, Ministry of Health, Changsha, 410000, China.
| |
Collapse
|
33
|
Khan S, Drabiak K. Eight Strategies to Engineer Acceptance of Human Germline Modifications. JOURNAL OF BIOETHICAL INQUIRY 2024; 21:81-94. [PMID: 37523056 DOI: 10.1007/s11673-023-10266-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 05/06/2023] [Indexed: 08/01/2023]
Abstract
Until recently, scientific consensus held firm that genetically manipulated embryos created through methods including Mitochondrial Replacement Therapy or human germline genome editing should not be used to initiate a pregnancy. In countries that have relevant laws pertaining to heritable human germline modifications, the vast majority prohibit or restrict this practice. In the last several years, scholars have observed a transformation of scientific and policy restrictions with insistent calls for creating a regulatory pathway. Multiple stakeholders highlight the role of social consensus and public engagement for governance of heritable human germline modifications. However, in the drive to gain public acceptance and lift restrictions, some proponents provide distorted or misleading narratives designed to influence public perception and incrementally shift the consensus. This article describes eight discrete strategies that proponents employ to influence framing, sway public opinion, and revise policymaking of human germline modifications in a manner that undermines honest engagement.
Collapse
Affiliation(s)
- Shoaib Khan
- Morsani College of Medicine, University of South Florida, Tampa, USA
| | | |
Collapse
|
34
|
Vinh DC. From Mendel to mycoses: Immuno-genomic warfare at the human-fungus interface. Immunol Rev 2024; 322:28-52. [PMID: 38069482 DOI: 10.1111/imr.13295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 11/06/2023] [Accepted: 11/14/2023] [Indexed: 03/20/2024]
Abstract
Fungi are opportunists: They particularly require a defect of immunity to cause severe or disseminated disease. While often secondary to an apparent iatrogenic cause, fungal diseases do occur in the absence of one, albeit infrequently. These rare cases may be due to an underlying genetic immunodeficiency that can present variably in age of onset, severity, or other infections, and in the absence of a family history of disease. They may also be due to anti-cytokine autoantibodies. This review provides a background on how human genetics or autoantibodies underlie cases of susceptibility to severe or disseminated fungal disease. Subsequently, the lessons learned from these inborn errors of immunity marked by fungal disease (IEI-FD) provide a framework to begin to mechanistically decipher fungal syndromes, potentially paving the way for precision therapy of the mycoses.
Collapse
Affiliation(s)
- Donald C Vinh
- Infectious Diseases - Hematology/Oncology/Transplant Clinical Program, Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada
- Centre of Excellence for Genetic Research in Infection and Immunity, Research Institute - McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
35
|
Abid R, Nisar H, Chaudhary SU, Hamid M, Sahibzada KI, Firdous S, Mudassar M, Sadaf S. Association of epilepsy and neurological impairments with homozygous recessive missense mutations found in the genes responsible for ganglioside biosynthesis ( ST3GAL5) and calcium voltage-gated channels ( CACNA1H) - insights through molecular dynamic simulations. J Biomol Struct Dyn 2024:1-12. [PMID: 38356142 DOI: 10.1080/07391102.2024.2314751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 01/30/2024] [Indexed: 02/16/2024]
Abstract
With over 2.2 million cases, the incidence rate of epilepsy in Pakistan is far higher than the rest of the world due primarily to the frequent, traditionally imposed cousin marriages. In the present study, comprehensive whole exome sequencing (WES) analyses of a three-generation family with four affected members presenting 'unexplained' childhood absence epilepsy (CAE), seizures and dementia, was performed in a quest to identify heritable, epilepsy-causal gene variants to better aid in carrier screening and genetic counselling. The WES data was generated, analyzed, and validated through Sanger's sequencing, molecular dynamic simulation (MDS) analysis, and molecular mechanics with generalized Born and surface area solvation (MM/GBSA) studies. Two homozygous recessive, missense mutations in ST3GAL5 (c.311A > G, p. His104Arg) and CACNA1H (c.6230G > A, p. Arg2077His) genes, earlier regarded as benign or of uncertain significance, have been identified as a potential etiology. Comparative MDS and free binding energy calculations revealed substantial structural perturbations in mutant forms of ST3GAL5 leading to decreased binding and reduced catalytic activity of the p.His104Arg and two other functional variants (p.Val74Glu and p.Arg288Ter) when compared with wild type. Our findings reinforce that WES analyses may uncover 'hidden', heritable variants and together with MDS and MM/GBSA may provide plausible clues to answer the unexplained causes of epilepsy for an effective management and better patient outcome. Further, revisit of epilepsy-associated mutational landscape in population context is imperative as the variants with 'benign' tags may turn out to be 'non-benign', when exist in combination with other benign.
Collapse
Affiliation(s)
- Rizwan Abid
- School of Biochemistry and Biotechnology, University of the Punjab, Lahore, Pakistan
| | - Haseeb Nisar
- Department of Life-Sciences, University of Management and Technology, Lahore, Pakistan
| | - Safee Ullah Chaudhary
- Biomedical Informatics & Engineering Research Laboratory, Department of Life-Sciences, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Maham Hamid
- Biomedical Informatics & Engineering Research Laboratory, Department of Life-Sciences, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Kashif Iqbal Sahibzada
- Department of Health Professional Technologies, The University of Lahore, Lahore, Pakistan
| | - Safia Firdous
- School of Biochemistry and Biotechnology, University of the Punjab, Lahore, Pakistan
- Faculty of Rehabilitation and Allied Health Sciences, Riphah International University, Lahore, Pakistan
| | | | - Saima Sadaf
- School of Biochemistry and Biotechnology, University of the Punjab, Lahore, Pakistan
| |
Collapse
|
36
|
Tran VK, Cao MH, Nguyen TTH, Le PT, Tran HA, Vu DC, Nguyen HT, Nguyen MTP, Bui TH, Nguyen TB, Ta TV, Tran TH. A novel IGHMBP2 variant and clinical diversity in Vietnamese SMARD1 and CMT2S patients. Front Pediatr 2024; 12:1165492. [PMID: 38415210 PMCID: PMC10896978 DOI: 10.3389/fped.2024.1165492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 01/26/2024] [Indexed: 02/29/2024] Open
Abstract
Background Pathogenic variants in the IGHMBP2 gene are associated with two distinct autosomal recessive neuromuscular disorders: spinal muscular atrophy with respiratory distress type 1 (SMARD1; OMIM #604320) and Charcot-Marie-Tooth type 2S (CMT2S; OMIM #616155). SMARD1 is a severe and fatal condition characterized by infantile-onset respiratory distress, diaphragmatic palsy, and distal muscular weakness, while CMT2S follows a milder clinical course, with slowly progressive distal muscle weakness and sensory loss, without manifestations of respiratory disorder. Methods Whole-exome sequencing of the IGHMBP2 gene was performed for eight Vietnamese patients with IGHMBP2-related neuromuscular disorders including five patients with SMARD1 and the others with CMT2S. Results We identified one novel IGHMBP2 variant c.1574T > C (p.Leu525Pro) in a SMARD1 patient. Besides that, two patients shared the same pathogenic variants (c.1235 + 3A > G/c.1334A > C) but presented completely different clinical courses: one with SMARD1 who deceased at 8 months of age, the other with CMT2S was alive at 3 years old without any respiratory distress. Conclusion This study is the first to report IGHMBP-2-related neuromuscular disorders in Vietnam. A novel IGHMBP2 variant c.1574T > C (p.Leu525Pro) expressing SMARD1 phenotype was detected. The presence of three patients with the same genotype but distinct clinical outcomes suggested the interaction of variants and other factors including relating modified genes in the mechanism of various phenotypes.
Collapse
Affiliation(s)
- Van Khanh Tran
- Center for Gene and Protein Research, Hanoi Medical University, Hanoi, Vietnam
- Department of Molecular Pathology, Faculty of Medical Laboratory Technology, Hanoi Medical University, Hanoi, Vietnam
| | - My Ha Cao
- Center for Gene and Protein Research, Hanoi Medical University, Hanoi, Vietnam
| | | | - Phuong Thi Le
- Center for Gene and Protein Research, Hanoi Medical University, Hanoi, Vietnam
| | - Hai Anh Tran
- Center for Gene and Protein Research, Hanoi Medical University, Hanoi, Vietnam
| | - Dung Chi Vu
- Department of Endocrinology, Metabolism and Genetics, National Hospital of Pediatrics, Hanoi, Vietnam
| | - Ha Thu Nguyen
- Department of Endocrinology, Metabolism and Genetics, National Hospital of Pediatrics, Hanoi, Vietnam
| | | | - The-Hung Bui
- Center for Gene and Protein Research, Hanoi Medical University, Hanoi, Vietnam
- Center for Molecular Medicine, Clinical Genetics Unit, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Thanh Binh Nguyen
- Center for Gene and Protein Research, Hanoi Medical University, Hanoi, Vietnam
| | - Thanh Van Ta
- Department of Biochemistry, Hanoi Medical University, Hanoi, Vietnam
- Clinical Laboratory, Hanoi Medical University Hospital, Hanoi Medical University, Hanoi, Vietnam
| | - Thinh Huy Tran
- Department of Biochemistry, Hanoi Medical University, Hanoi, Vietnam
- Clinical Laboratory, Hanoi Medical University Hospital, Hanoi Medical University, Hanoi, Vietnam
| |
Collapse
|
37
|
Ghaedi H, Davey SK, Feilotter H. Variant Classification Discordance: Contributing Factors and Predictive Models. J Mol Diagn 2024; 26:115-126. [PMID: 38008287 DOI: 10.1016/j.jmoldx.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/04/2023] [Accepted: 11/07/2023] [Indexed: 11/28/2023] Open
Abstract
An ever-growing catalog of human variants is hosted in the ClinVar database. In this database, submissions on a variant are combined into a multisubmitter record; and in the case of discordance in variant classification between submitters, the record is labeled as conflicting. The current study used ClinVar data to identify characteristics that would make variants more likely to be associated with the conflict class of variants. Furthermore, the Extreme Gradient Boosting algorithm was used to train classifier models to provide prediction of classification discordance for single submission variants in ClinVar database. Population allele frequency, the gene harboring the variant, variant type, consequence on protein, variant deleteriousness score, first submitter identity, and submission count were associated with conflict in variant classification. Using such features, the optimized classifier showed accuracy on the test set of 88% with the weighted average of precision, recall, and f1-score of 0.84, 0.88, and 0.85, respectively. There were pronounced associations between variant classification discordance and allele frequency, gene type, and the identity of the first submitter. The study provides the predicted discordance status for single-submitter variants deposited in ClinVar. This approach can be used to assess whether single-submitter variants are likely to be supported, or in conflict with, future entries; this knowledge may help laboratories with clinical variant assessment.
Collapse
Affiliation(s)
- Hamid Ghaedi
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Scott K Davey
- Division of Cancer Biology and Genetics, Department of Pathology and Molecular Medicine, Queen's University Cancer Research Institute, Kingston, Ontario, Canada
| | - Harriet Feilotter
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
38
|
Höglinger GU, Adler CH, Berg D, Klein C, Outeiro TF, Poewe W, Postuma R, Stoessl AJ, Lang AE. A biological classification of Parkinson's disease: the SynNeurGe research diagnostic criteria. Lancet Neurol 2024; 23:191-204. [PMID: 38267191 DOI: 10.1016/s1474-4422(23)00404-0] [Citation(s) in RCA: 202] [Impact Index Per Article: 202.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 09/08/2023] [Accepted: 10/06/2023] [Indexed: 01/26/2024]
Abstract
With the hope that disease-modifying treatments could target the molecular basis of Parkinson's disease, even before the onset of symptoms, we propose a biologically based classification. Our classification acknowledges the complexity and heterogeneity of the disease by use of a three-component system (SynNeurGe): presence or absence of pathological α-synuclein (S) in tissues or CSF; evidence of underlying neurodegeneration (N) defined by neuroimaging procedures; and documentation of pathogenic gene variants (G) that cause or strongly predispose to Parkinson's disease. These three components are linked to a clinical component (C), defined either by a single high-specificity clinical feature or by multiple lower-specificity clinical features. The use of a biological classification will enable advances in both basic and clinical research, and move the field closer to the precision medicine required to develop disease-modifying therapies. We emphasise the initial application of these criteria exclusively for research. We acknowledge its ethical implications, its limitations, and the need for prospective validation in future studies.
Collapse
Affiliation(s)
- Günter U Höglinger
- Department of Neurology, University Hospital, Ludwig-Maximilians-University (LMU) and German Center for Neurodegenerative Diseases, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Charles H Adler
- Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Daniela Berg
- Christian Albrechts University and University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck and University Hospital Schleswig-Holstein, Campus Lübeck, Lüebeck, Germany
| | - Tiago F Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany; Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Werner Poewe
- Medical University Innsbruck, Innsbruck, Austria
| | - Ronald Postuma
- Department of Neurology, McGill University, Montreal Neurological Institute, Montreal, QC, Canada
| | - A Jon Stoessl
- Pacific Parkinson's Research Centre and Parkinson's Foundation Centre of Excellence, University of British Columbia, BC, Canada
| | - Anthony E Lang
- University Health Network's Krembil Brain Institute, Edmond J Safra Program in Parkinson's Disease and the Rossy PSP Centre, Toronto Western Hospital, Toronto, ON, Canada.
| |
Collapse
|
39
|
Cacheiro P, Lawson S, Van den Veyver IB, Marengo G, Zocche D, Murray SA, Duyzend M, Robinson PN, Smedley D. Lethal phenotypes in Mendelian disorders. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.12.24301168. [PMID: 38260283 PMCID: PMC10802756 DOI: 10.1101/2024.01.12.24301168] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Essential genes are those whose function is required for cell proliferation and/or organism survival. A gene's intolerance to loss-of-function can be allocated within a spectrum, as opposed to being considered a binary feature, since this function might be essential at different stages of development, genetic backgrounds or other contexts. Existing resources that collect and characterise the essentiality status of genes are based on either proliferation assessment in human cell lines, embryonic and postnatal viability evaluation in different model organisms, and gene metrics such as intolerance to variation scores derived from human population sequencing studies. There are also several repositories available that document phenotypic annotations for rare disorders in humans such as the Online Mendelian Inheritance in Man (OMIM) and the Human Phenotype Ontology (HPO) knowledgebases. This raises the prospect of being able to use clinical data, including lethality as the most severe phenotypic manifestation, to further our characterisation of gene essentiality. Here we queried OMIM for terms related to lethality and classified all Mendelian genes into categories, according to the earliest age of death recorded for the associated disorders, from prenatal death to no reports of premature death. To showcase this curated catalogue of human essential genes, we developed the Lethal Phenotypes Portal (https://lethalphenotypes.research.its.qmul.ac.uk), where we also explore the relationships between these lethality categories, constraint metrics and viability in cell lines and mouse. Further analysis of the genes in these categories reveals differences in the mode of inheritance of the associated disorders, physiological systems affected and disease class. We highlight how the phenotypic similarity between genes in the same lethality category combined with gene family/group information can be used for novel disease gene discovery. Finally, we explore the overlaps and discrepancies between the lethal phenotypes observed in mouse and human and discuss potential explanations that include differences in transcriptional regulation, functional compensation and molecular disease mechanisms. We anticipate that this resource will aid clinicians in the diagnosis of early lethal conditions and assist researchers in investigating the properties that make these genes essential for human development.
Collapse
Affiliation(s)
- Pilar Cacheiro
- William Harvey Research Institute, Queen Mary University of London, London, UK
| | | | - Ignatia B. Van den Veyver
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
| | - Gabriel Marengo
- William Harvey Research Institute, Queen Mary University of London, London, UK
| | - David Zocche
- North West Thames Regional Genetics Service, Northwick Park & St Mark’s Hospitals, London, UK
| | | | | | - Peter N. Robinson
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Damian Smedley
- William Harvey Research Institute, Queen Mary University of London, London, UK
| |
Collapse
|
40
|
Ciesielski TH, Sirugo G, Iyengar SK, Williams SM. Characterizing the pathogenicity of genetic variants: the consequences of context. NPJ Genom Med 2024; 9:3. [PMID: 38195641 PMCID: PMC10776585 DOI: 10.1038/s41525-023-00386-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 12/15/2023] [Indexed: 01/11/2024] Open
Affiliation(s)
- Timothy H Ciesielski
- The Department of Population and Quantitative Health Sciences at Case Western Reserve University School of Medicine, Cleveland, OH, USA.
- Mary Ann Swetland Center for Environmental Health at Case Western Reserve University School of Medicine, Cleveland, OH, USA.
- Ronin Institute, Montclair, NJ, USA.
| | - Giorgio Sirugo
- The Department of Population and Quantitative Health Sciences at Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Institute of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sudha K Iyengar
- The Department of Population and Quantitative Health Sciences at Case Western Reserve University School of Medicine, Cleveland, OH, USA
- The Department of Genetics and Genome Sciences at Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Cleveland Institute for Computational Biology, Cleveland, OH, USA
| | - Scott M Williams
- The Department of Population and Quantitative Health Sciences at Case Western Reserve University School of Medicine, Cleveland, OH, USA
- The Department of Genetics and Genome Sciences at Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Cleveland Institute for Computational Biology, Cleveland, OH, USA
| |
Collapse
|
41
|
McTague A, Scheffer IE, Kullmann DM, Sisodiya S. Epilepsies. HANDBOOK OF CLINICAL NEUROLOGY 2024; 203:157-184. [PMID: 39174247 DOI: 10.1016/b978-0-323-90820-7.00016-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
Recent advances in genetic diagnosis have revealed the underlying etiology of many epilepsies and have identified pathogenic, causative variants in numerous ion and ligand-gated channel genes. This chapter describes the clinical presentations of epilepsy associated with different channelopathies including classic electroclinical syndromes and emerging gene-specific phenotypes. Also discussed are the archetypal epilepsy channelopathy, SCN1A-Dravet syndrome, considering the expanding phenotype. Clinical presentations where a channelopathy is suspected, such as sleep-related hypermotor epilepsy and epilepsy in association with movement disorders, are reviewed. Channelopathies pose an intriguing problem for the development of gene therapies. Design of targeted therapies requires physiologic insights into the often multifaceted impact of a pathogenic variant, coupled with an understanding of the phenotypic spectrum of a gene. As gene-specific novel therapies come online for the channelopathies, it is essential that clinicians are able to recognize epilepsy phenotypes likely to be due to channelopathy and institute early genetic testing in both children and adults. These findings are likely to have immediate management implications and to inform prognostic and reproductive counseling.
Collapse
Affiliation(s)
- Amy McTague
- Developmental Neurosciences, UCL Great Ormond Street Institute of Child Health, London, United Kingdom; Department of Neurology, Great Ormond Street Hospital for Children, London, United Kingdom.
| | - Ingrid E Scheffer
- Austin Health and Royal Children's Hospital, Florey and Murdoch Children's Research Institutes, University of Melbourne, Melbourne, VIC, Australia
| | - Dimitri M Kullmann
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom; National Hospital for Neurology and Neurosurgery, Queen Square, London, United Kingdom
| | - Sanjay Sisodiya
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom; National Hospital for Neurology and Neurosurgery, Queen Square, London, United Kingdom
| |
Collapse
|
42
|
Van Impe M, Caboor L, Deleeuw V, De Rycke K, Vanhooydonck M, De Backer J, Segers P, Sips P. Application of an automated analysis framework for pulsed-wave Doppler cardiac ultrasound measurements to generate reference data in adult zebrafish. Am J Physiol Regul Integr Comp Physiol 2023; 325:R782-R796. [PMID: 37811715 DOI: 10.1152/ajpregu.00103.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 09/25/2023] [Accepted: 10/04/2023] [Indexed: 10/10/2023]
Abstract
High-frequency cardiac ultrasound is the only well-established method to characterize in vivo cardiovascular function in adult zebrafish noninvasively. Pulsed-wave Doppler imaging allows measurements of blood flow velocities at well-defined anatomical positions, but the measurements and results obtained using this technique need to be analyzed carefully, taking into account the substantial baseline variability within one recording and the possibility for operator bias. To address these issues and to increase throughput by limiting hands-on analysis time, we have developed a fully automated processing pipeline. This framework enables the fast, unbiased analysis of all cardiac cycles in a zebrafish pulsed-wave Doppler recording of both atrioventricular valve flow as well as aortic valve flow without operator-dependent inputs. Applying this automated pipeline to a large number of recordings from wild-type zebrafish shows a strong agreement between the automated results and manual annotations performed by an experienced operator. The reference data obtained from this analysis showed that the early wave peak during ventricular inflow is lower for female compared with male zebrafish. We also found that the peaks of the ventricular inflow and outflow waves as well as the peaks of the regurgitation waves are all correlated positively with body surface area. In general, the presented reference data, as well as the automated Doppler measurement processing tools developed and validated in this study will facilitate future (high-throughput) cardiovascular phenotyping studies in adult zebrafish ultimately leading to a more comprehensive understanding of human (genetic) cardiovascular diseases.
Collapse
Affiliation(s)
- Matthias Van Impe
- IBiTech-BioMMedA, Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
| | - Lisa Caboor
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Violette Deleeuw
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Karo De Rycke
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Michiel Vanhooydonck
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Julie De Backer
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Patrick Segers
- IBiTech-BioMMedA, Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
| | - Patrick Sips
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| |
Collapse
|
43
|
Jakobson CM, Aguilar-Rodríguez J, Jarosz DF. Hsp90 shapes adaptation by controlling the fitness consequences of regulatory variation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.30.564848. [PMID: 37961536 PMCID: PMC10634948 DOI: 10.1101/2023.10.30.564848] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
The essential stress-responsive chaperone Hsp90 impacts development and adaptation from microbes to humans. Yet despite evidence of its role in evolution, pathogenesis, and oncogenic transformation, the molecular mechanisms by which Hsp90 alters the consequences of mutations remain vigorously debated. Here we exploit the power of nucleotide-resolution genetic mapping in Saccharomyces cerevisiae to uncover more than 1,000 natural variant-to-phenotype associations governed by this molecular chaperone. Strikingly, Hsp90 more frequently modified the phenotypic effects of cis-regulatory variation than variants that altered protein sequence. Moreover, these interactions made the largest contribution to Hsp90-dependent heredity. Nearly all interacting variants-both regulatory and protein-coding-fell within clients of Hsp90 or targets of its direct binding partners. Hsp90 activity affected mutations in evolutionarily young genes, segmental deletions, and heterozygotes, highlighting its influence on variation central to evolutionary novelty. Reconciling the diverse epistatic effects of this chaperone, synthetic transcriptional regulation and reconstructions of natural alleles by genome editing revealed a central role for Hsp90 in regulating the fundamental relationship between activity and phenotype. Our findings establish that non-coding variation is a core driver of Hsp90's influence on heredity, offering a mechanistic explanation for the chaperone's strong effects on evolution and development across species.
Collapse
Affiliation(s)
- Christopher M. Jakobson
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
- These authors contributed equally
| | - José Aguilar-Rodríguez
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biology, Stanford University, Stanford, CA, USA
- These authors contributed equally
| | - Daniel F. Jarosz
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
44
|
Meyer BJ, Kunz N, Seki S, Higgins R, Ghosh A, Hupfer R, Baldrich A, Hirsiger JR, Jauch AJ, Burgener AV, Lötscher J, Aschwanden M, Dickenmann M, Stegert M, Berger CT, Daikeler T, Heijnen I, Navarini AA, Rudin C, Yamamoto H, Kemper C, Hess C, Recher M. Immunologic and Genetic Contributors to CD46-Dependent Immune Dysregulation. J Clin Immunol 2023; 43:1840-1856. [PMID: 37477760 PMCID: PMC10661731 DOI: 10.1007/s10875-023-01547-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 06/30/2023] [Indexed: 07/22/2023]
Abstract
Mutations in CD46 predispose to atypical hemolytic uremic syndrome (aHUS) with low penetrance. Factors driving immune-dysregulatory disease in individual mutation carriers have remained ill-understood. In addition to its role as a negative regulator of the complement system, CD46 modifies T cell-intrinsic metabolic adaptation and cytokine production. Comparative immunologic analysis of diseased vs. healthy CD46 mutation carriers has not been performed in detail yet. In this study, we comprehensively analyzed clinical, molecular, immune-phenotypic, cytokine secretion, immune-metabolic, and genetic profiles in healthy vs. diseased individuals carrying a rare, heterozygous CD46 mutation identified within a large single family. Five out of six studied individuals carried a CD46 gene splice-site mutation causing an in-frame deletion of 21 base pairs. One child suffered from aHUS and his paternal uncle manifested with adult-onset systemic lupus erythematosus (SLE). Three mutation carriers had no clinical evidence of CD46-related disease to date. CD4+ T cell-intrinsic CD46 expression was uniformly 50%-reduced but was comparable in diseased vs. healthy mutation carriers. Reconstitution experiments defined the 21-base pair-deleted CD46 variant as intracellularly-but not surface-expressed and haploinsufficient. Both healthy and diseased mutation carriers displayed reduced CD46-dependent T cell mitochondrial adaptation. Diseased mutation carriers had lower peripheral regulatory T cell (Treg) frequencies and carried potentially epistatic, private rare variants in other inborn errors of immunity (IEI)-associated proinflammatory genes, not found in healthy mutation carriers. In conclusion, low Treg and rare non-CD46 immune-gene variants may contribute to clinically manifest CD46 haploinsufficiency-associated immune-dysregulation.
Collapse
Affiliation(s)
- Benedikt J Meyer
- Immunodeficiency Laboratory, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Natalia Kunz
- Immunobiology Laboratory, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
- Complement and Inflammation Research Section, CIRS, DIR, NHLBI, NIH, Bethesda, USA
| | - Sayuri Seki
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | | | - Adhideb Ghosh
- Dermatology, University Hospital Basel, Basel, Switzerland
- Competence Center for Personalized Medicine, University of Zürich/Eidgenössische Technische Hochschule (ETH), Zürich, Switzerland
| | - Robin Hupfer
- Immunodeficiency Laboratory, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Adrian Baldrich
- Immunodeficiency Laboratory, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Julia R Hirsiger
- Translational Immunology, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Annaïse J Jauch
- Immunodeficiency Laboratory, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Anne-Valérie Burgener
- Immunobiology Laboratory, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Jonas Lötscher
- Immunobiology Laboratory, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Markus Aschwanden
- Department of Angiology, University Hospital Basel, Basel, Switzerland
| | - Michael Dickenmann
- Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland
| | - Mihaela Stegert
- Rheumatology Clinic, University Hospital Basel, Basel, Switzerland
| | - Christoph T Berger
- Translational Immunology, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
- University Center for Immunology, University Hospital Basel, Basel, Switzerland
| | - Thomas Daikeler
- Rheumatology Clinic, University Hospital Basel, Basel, Switzerland
- University Center for Immunology, University Hospital Basel, Basel, Switzerland
| | - Ingmar Heijnen
- Division Medical Immunology, Laboratory Medicine, University Hospital Basel, Basel, Switzerland
| | | | - Christoph Rudin
- University Children's Hospital, University of Basel, Basel, Switzerland
| | - Hiroyuki Yamamoto
- Immunodeficiency Laboratory, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Claudia Kemper
- Complement and Inflammation Research Section, CIRS, DIR, NHLBI, NIH, Bethesda, USA
| | - Christoph Hess
- Immunobiology Laboratory, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
- Cambridge Institute of Therapeutic Immunology & Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Mike Recher
- Immunodeficiency Laboratory, Department of Biomedicine, University Hospital Basel, Basel, Switzerland.
- University Center for Immunology, University Hospital Basel, Basel, Switzerland.
| |
Collapse
|
45
|
Maroilley T, Rahit KMTH, Chida AR, Cotra F, Rodrigues Alves Barbosa V, Tarailo-Graovac M. Model Organism Modifier (MOM): a user-friendly Galaxy workflow to detect modifiers from genome sequencing data using Caenorhabditis elegans. G3 (BETHESDA, MD.) 2023; 13:jkad184. [PMID: 37585487 PMCID: PMC10627290 DOI: 10.1093/g3journal/jkad184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 04/21/2023] [Accepted: 08/05/2023] [Indexed: 08/18/2023]
Abstract
Genetic modifiers are variants modulating phenotypic outcomes of a primary detrimental variant. They contribute to rare diseases phenotypic variability, but their identification is challenging. Genetic screening with model organisms is a widely used method for demystifying genetic modifiers. Forward genetics screening followed by whole genome sequencing allows the detection of variants throughout the genome but typically produces thousands of candidate variants making the interpretation and prioritization process very time-consuming and tedious. Despite whole genome sequencing is more time and cost-efficient, usage of computational pipelines specific to modifier identification remains a challenge for biological-experiment-focused laboratories doing research with model organisms. To facilitate a broader implementation of whole genome sequencing in genetic screens, we have developed Model Organism Modifier or MOM, a pipeline as a user-friendly Galaxy workflow. Model Organism Modifier analyses raw short-read whole genome sequencing data and implements tailored filtering to provide a Candidate Variant List short enough to be further manually curated. We provide a detailed tutorial to run the Galaxy workflow Model Organism Modifier and guidelines to manually curate the Candidate Variant Lists. We have tested Model Organism Modifier on published and validated Caenorhabditis elegans modifiers screening datasets. As whole genome sequencing facilitates high-throughput identification of genetic modifiers in model organisms, Model Organism Modifier provides a user-friendly solution to implement the bioinformatics analysis of the short-read datasets in laboratories without expertise or support in Bioinformatics.
Collapse
Affiliation(s)
- Tatiana Maroilley
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Medical Genetics, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - K M Tahsin Hassan Rahit
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Medical Genetics, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Afiya Razia Chida
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Medical Genetics, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Filip Cotra
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Medical Genetics, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Victoria Rodrigues Alves Barbosa
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Medical Genetics, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Maja Tarailo-Graovac
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Medical Genetics, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
46
|
Goodman SJ, Luperchio TR, Ellegood J, Chater-Diehl E, Lerch JP, Bjornsson HT, Weksberg R. Peripheral blood DNA methylation and neuroanatomical responses to HDACi treatment that rescues neurological deficits in a Kabuki syndrome mouse model. Clin Epigenetics 2023; 15:172. [PMID: 37884963 PMCID: PMC10605417 DOI: 10.1186/s13148-023-01582-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 10/08/2023] [Indexed: 10/28/2023] Open
Abstract
BACKGROUND Recent findings from studies of mouse models of Mendelian disorders of epigenetic machinery strongly support the potential for postnatal therapies to improve neurobehavioral and cognitive deficits. As several of these therapies move into human clinical trials, the search for biomarkers of treatment efficacy is a priority. A potential postnatal treatment of Kabuki syndrome type 1 (KS1), caused by pathogenic variants in KMT2D encoding a histone-lysine methyltransferase, has emerged using a mouse model of KS1 (Kmt2d+/βGeo). In this mouse model, hippocampal memory deficits are ameliorated following treatment with the histone deacetylase inhibitor (HDACi), AR-42. Here, we investigate the effect of both Kmt2d+/βGeo genotype and AR-42 treatment on neuroanatomy and on DNA methylation (DNAm) in peripheral blood. While peripheral blood may not be considered a "primary tissue" with respect to understanding the pathophysiology of neurodevelopmental disorders, it has the potential to serve as an accessible biomarker of disease- and treatment-related changes in the brain. METHODS Half of the KS1 and wildtype mice were treated with 14 days of AR-42. Following treatment, fixed brain samples were imaged using MRI to calculate regional volumes. Blood was assayed for genome-wide DNAm at over 285,000 CpG sites using the Illumina Infinium Mouse Methylation array. DNAm patterns and brain volumes were analyzed in the four groups of animals: wildtype untreated, wildtype AR-42 treated, KS1 untreated and KS1 AR-42 treated. RESULTS We defined a DNAm signature in the blood of KS1 mice, that overlapped with the human KS1 DNAm signature. We also found a striking 10% decrease in total brain volume in untreated KS1 mice compared to untreated wildtype, which correlated with DNAm levels in a subset KS1 signature sites, suggesting that disease severity may be reflected in blood DNAm. Treatment with AR-42 ameliorated DNAm aberrations in KS1 mice at a small number of signature sites. CONCLUSIONS As this treatment impacts both neurological deficits and blood DNAm in mice, future KS clinical trials in humans could be used to assess blood DNAm as an early biomarker of therapeutic efficacy.
Collapse
Affiliation(s)
| | - Teresa Romeo Luperchio
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Jacob Ellegood
- Mouse Imaging Centre (MICe), Hospital for Sick Children, Toronto, Canada
| | - Eric Chater-Diehl
- Genetics and Genome Biology, Hospital for Sick Children, Toronto, Canada
| | - Jason P Lerch
- Mouse Imaging Centre (MICe), Hospital for Sick Children, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Wellcome Centre for Integrative Neuroimaging, The University of Oxford, Oxford, UK
- Nuffield Department of Clinical Neuroscience, The University of Oxford, Oxford, UK
| | - Hans Tomas Bjornsson
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, USA
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
- Landspitali University Hospital, Reykjavík, Iceland
| | - Rosanna Weksberg
- Genetics and Genome Biology, Hospital for Sick Children, Toronto, Canada.
- Division of Clinical and Metabolic Genetics, Hospital for Sick Children, Toronto, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, Canada.
- Institute of Medical Science, University of Toronto, Toronto, Canada.
- Department of Paediatrics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
47
|
Keseroglu K, Zinani OQH, Keskin S, Seawall H, Alpay EE, Özbudak EM. Stochastic gene expression and environmental stressors trigger variable somite segmentation phenotypes. Nat Commun 2023; 14:6497. [PMID: 37838784 PMCID: PMC10576776 DOI: 10.1038/s41467-023-42220-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 10/04/2023] [Indexed: 10/16/2023] Open
Abstract
Mutations of several genes cause incomplete penetrance and variable expressivity of phenotypes, which are usually attributed to modifier genes or gene-environment interactions. Here, we show stochastic gene expression underlies the variability of somite segmentation defects in embryos mutant for segmentation clock genes her1 or her7. Phenotypic strength is further augmented by low temperature and hypoxia. By performing live imaging of the segmentation clock reporters, we further show that groups of cells with higher oscillation amplitudes successfully form somites while those with lower amplitudes fail to do so. In unfavorable environments, the number of cycles with high amplitude oscillations and the number of successful segmentations proportionally decrease. These results suggest that individual oscillation cycles stochastically fail to pass a threshold amplitude, resulting in segmentation defects in mutants. Our quantitative methodology is adaptable to investigate variable phenotypes of mutant genes in different tissues.
Collapse
Affiliation(s)
- Kemal Keseroglu
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Oriana Q H Zinani
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Molecular and Developmental Biology Graduate Program, University of Cincinnati, College of Medicine, Cincinnati, OH, 45229, USA
| | - Sevdenur Keskin
- Allergy and Immunology, University of Arkansas for Medical Science and Arkansas Children's Hospital, Little Rock, AR, 72202, USA
| | - Hannah Seawall
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Eslim E Alpay
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Ertuğrul M Özbudak
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA.
| |
Collapse
|
48
|
Jackson L, Weedon MN, Green HD, Mallabar-Rimmer B, Harrison JW, Wood AR, Ruth KS, Tyrrell J, Wright CF. Influence of family history on penetrance of hereditary cancers in a population setting. EClinicalMedicine 2023; 64:102159. [PMID: 37936660 PMCID: PMC10626157 DOI: 10.1016/j.eclinm.2023.102159] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 11/09/2023] Open
Abstract
Background We sought to investigate how penetrance of familial cancer syndromes varies with family history using a population-based cohort. Methods We analysed 454,712 UK Biobank participants with exome sequence and clinical data (data collected between March 2006 and June 2021). We identified participants with a self-reported family history of breast or colorectal cancer and a pathogenic/likely pathogenic variant in the major genes responsible for hereditary breast cancer or Lynch syndrome. We calculated survival to cancer diagnosis (controlled for sex, death, recruitment centre, screening and prophylactic surgery). Findings Women with a pathogenic BRCA1 or BRCA2 variant had an increased risk of breast cancer that was higher in those with a first-degree family history (relative hazard 10.3 and 7.8, respectively) than those without (7.2 and 4.7). Penetrance to age 60 was also higher in those with a family history (44.7%, CI 32.2-59.3 and 24.1%, CI 17.5-32.6) versus those without (22.8%, CI 15.9-32.0 and 17.9%, CI 13.8-23.0). A similar pattern was seen in Lynch syndrome: individuals with a pathogenic MLH1, MSH2 or MSH6 variant had an increased risk of colorectal cancer that was significantly higher in those with a family history (relative hazard 35.6, 48.0 and 9.9) than those without (13.0, 15.4 and 7.2). Penetrance to age 60 was also higher for carriers of a pathogenic MLH1 or MSH2 variant in those with a family history (30.9%, CI 18.1-49.3 and 38.3%, CI 21.5-61.8) versus those without (20.5% CI 9.6-40.5 and 8.3% CI 2.1-30.4), but not for MSH6 (6.5% CI 2.7-15.1 with family history versus 8.3%, CI 5.1-13.2). Relative risk increases were also observed both within and across conditions. Interpretation Individuals with pathogenic cancer syndrome variants may be at a less elevated risk of cancer in the absence of a first-degree family history, so in the context of results return, family history should be considered when counselling patients on the risks and benefits of potential follow-up care. Funding The current work is supported by the MRC (grant no MR/T00200X/1). The MRC had no role in the design and conduct of the study; collection, management, analysis, and interpretation of the data; preparation, review, or approval of the manuscript; and decision to submit the manuscript for publication.
Collapse
Affiliation(s)
- Leigh Jackson
- Institute of Biomedical and Clinical Science, University of Exeter College of Medicine and Health, RILD Building, Royal Devon & Exeter Hospital, Barrack Road, Exeter, EX2 5DW, UK
| | - Michael N. Weedon
- Institute of Biomedical and Clinical Science, University of Exeter College of Medicine and Health, RILD Building, Royal Devon & Exeter Hospital, Barrack Road, Exeter, EX2 5DW, UK
| | - Harry D. Green
- Institute of Biomedical and Clinical Science, University of Exeter College of Medicine and Health, RILD Building, Royal Devon & Exeter Hospital, Barrack Road, Exeter, EX2 5DW, UK
| | - Bethan Mallabar-Rimmer
- Institute of Biomedical and Clinical Science, University of Exeter College of Medicine and Health, RILD Building, Royal Devon & Exeter Hospital, Barrack Road, Exeter, EX2 5DW, UK
| | - Jamie W. Harrison
- Institute of Biomedical and Clinical Science, University of Exeter College of Medicine and Health, RILD Building, Royal Devon & Exeter Hospital, Barrack Road, Exeter, EX2 5DW, UK
| | - Andy R. Wood
- Institute of Biomedical and Clinical Science, University of Exeter College of Medicine and Health, RILD Building, Royal Devon & Exeter Hospital, Barrack Road, Exeter, EX2 5DW, UK
| | - Kate S. Ruth
- Institute of Biomedical and Clinical Science, University of Exeter College of Medicine and Health, RILD Building, Royal Devon & Exeter Hospital, Barrack Road, Exeter, EX2 5DW, UK
| | - Jess Tyrrell
- Institute of Biomedical and Clinical Science, University of Exeter College of Medicine and Health, RILD Building, Royal Devon & Exeter Hospital, Barrack Road, Exeter, EX2 5DW, UK
| | - Caroline F. Wright
- Institute of Biomedical and Clinical Science, University of Exeter College of Medicine and Health, RILD Building, Royal Devon & Exeter Hospital, Barrack Road, Exeter, EX2 5DW, UK
| |
Collapse
|
49
|
Fatemi N, Tu SJ, Chung CC, Moghadam PK, Mojarad EN, Sadeghi A, Totonchi M, Aghdaei HA, Chang JG. Whole exome sequencing identifies MAP3K1, MSH2, and MLH1 as potential cancer-predisposing genes in familial early-onset colorectal cancer. Kaohsiung J Med Sci 2023; 39:896-903. [PMID: 37314251 DOI: 10.1002/kjm2.12715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 05/01/2023] [Accepted: 05/21/2023] [Indexed: 06/15/2023] Open
Abstract
The incidence of early-onset colorectal cancer (CRC), which affects people under 50, is increasing for unknown reasons. Additionally, no underlying genetic cause is found in 20%-30% of patients suspected of having familial CRC syndrome. Whole exome sequencing (WES) has generated evidence for new genes associated with CRC susceptibility, but many patients remain undiagnosed. This study applied WES in five early-onset CRC patients from three unrelated families to identify novel genetic variants that could be linked to rapid disease development. Furthermore, the candidate variants were validated using Sanger sequencing. Two heterozygote variations, c.1077-2A>G and c.199G>A, were found in the MSH2 and the MLH1 genes, respectively. Sanger sequencing analysis confirmed that these (likely) pathogenic mutations segregated in all the affected families' members. In addition, we identified a rare heterozygote variant (c.175C>T) with suspected pathogenic potential in the MAP3K1 gene; formally the variant is of uncertain significance (VUS). Our findings support the hypothesis that CRC onset may be oligogenic and molecularly heterogeneous. Larger and more robust studies are needed to understand the genetic basis of early-onset CRC development, combined with novel functional analyses and omics approaches.
Collapse
Affiliation(s)
- Nayeralsadat Fatemi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Siang-Jyun Tu
- Center for Precision Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Chin-Chun Chung
- Center for Precision Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Pardis Ketabi Moghadam
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ehsan Nazemalhosseini Mojarad
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Sadeghi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Totonchi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jan-Gowth Chang
- Center for Precision Medicine, China Medical University Hospital, Taichung, Taiwan
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| |
Collapse
|
50
|
Li YP, Shen RJ, Cheng YM, Zhao Q, Jin K, Jin ZB, Zhang S. Exome sequencing in retinal dystrophy patients reveals a novel candidate gene ER membrane protein complex subunit 3. Heliyon 2023; 9:e20146. [PMID: 37809982 PMCID: PMC10559921 DOI: 10.1016/j.heliyon.2023.e20146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 09/05/2023] [Accepted: 09/13/2023] [Indexed: 10/10/2023] Open
Abstract
Inherited retinal dystrophies (IRDs) are a heterogeneous group of visual disorders caused by different pathogenic mutations in genes and regulatory sequences. The endoplasmic reticulum (ER) membrane protein complex (EMC) subunit 3 (EMC3) is the core unit of the EMC insertase that integrates the transmembrane peptides into lipid bilayers, and the function of its cytoplasmic carboxyl terminus remains to be elucidated. In this study, an insertional mutation c.768insT in the C-terminal coding region of EMC3 was identified and associated with dominant IRDs in a five-generation family. This mutation caused a frameshift in the coding sequence and a gain of an additional 16 amino acid residues (p.L256F-fs-ext21) to form a helix structure in the C-terminus of the EMC3 protein. The mutation is heterozygous with an incomplete penetrance, and cosegregates in all patients examined. This finding indicates that the C-terminus of EMC3 is essential for EMC functions and that EMC3 may be a novel candidate gene for retinal degenerative diseases.
Collapse
Affiliation(s)
- Yan-Ping Li
- Laboratory for Stem Cell & Retinal Regeneration, The Eye Hospital, Basic Medical College, Wenzhou Medical University, Wenzhou, 325027, China
| | - Ren-Juan Shen
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
| | - You-Min Cheng
- Laboratory for Stem Cell & Retinal Regeneration, The Eye Hospital, Basic Medical College, Wenzhou Medical University, Wenzhou, 325027, China
| | - Qingqing Zhao
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310030, China
| | - Kangxin Jin
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
| | - Zi-Bing Jin
- Laboratory for Stem Cell & Retinal Regeneration, The Eye Hospital, Basic Medical College, Wenzhou Medical University, Wenzhou, 325027, China
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
| | - Shaodan Zhang
- The Eye Hospital of Wenzhou Medical University, National Clinical Research Center for Ocular Diseases, Glaucoma Research Institute of Wenzhou Medical University, Wenzhou, 325027, China
| |
Collapse
|