1
|
Spineli-Silva S, de Leeuw N, Pontes LB, Leijsten N, Ruiterkamp-Versteeg M, Prota JRM, Marques-de-Faria AP, Vieira TP. A nonsense variant in the C-terminal transactivation domain of the EBF3 gene in an individual with intellectual disability and behavioural disorder: case report and literature review. Psychiatr Genet 2025; 35:75-78. [PMID: 40073166 DOI: 10.1097/ypg.0000000000000386] [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: 03/14/2025]
Abstract
Heterozygous variants in the Early B cell factor 3 ( EBF3 ) have been reported in individuals presenting with hypotonia, ataxia and delayed development syndrome (HADDS) (MIM#617330). However, individuals with pathogenic variants in EBF3 show phenotypic heterogeneity and very few variants in the C-terminal domain have been described. We report on a heterozygous de-novo variant in the EBF3 gene in an individual with neurodevelopmental delay and behavioural problems. The proband presented with speech delay, learning disability and behavioural problems that suggest an oppositional defiant disorder. He also has hyperactivity, irritability, hetero-aggressiveness, visual hallucinations, insomnia and decreased pain sensitivity. Whole exome sequencing revealed a de-novo heterozygous nonsense variant - c.1408C>T (p.Arg470*) - in the EBF3 gene, classified as pathogenic. The patient herein described, with a truncating variant in the C-terminal domain of EBF3 , supports the clinical variability of this condition and contributes to genotype-phenotype correlation of this rare disorder.
Collapse
Affiliation(s)
- Samira Spineli-Silva
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences, Universidade Estadual de Campinas, São Paulo, Brazil
| | - Nicole de Leeuw
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Larissa B Pontes
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences, Universidade Estadual de Campinas, São Paulo, Brazil
| | - Nico Leijsten
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Joana R M Prota
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences, Universidade Estadual de Campinas, São Paulo, Brazil
| | - Antonia P Marques-de-Faria
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences, Universidade Estadual de Campinas, São Paulo, Brazil
| | - Társis P Vieira
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences, Universidade Estadual de Campinas, São Paulo, Brazil
| |
Collapse
|
2
|
Sklenar M, Borecka S, Varga L, Bernardinelli E, Stanik J, Skopkova M, Sabo M, Ugorova D, Dossena S, Gasperikova D. Genetic heterogeneity in patients with enlarged vestibular aqueduct and Pendred syndrome. Mol Med 2025; 31:208. [PMID: 40426046 PMCID: PMC12107780 DOI: 10.1186/s10020-025-01262-x] [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] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Pathogenic variants in the SLC26A4 gene, encoding for Cl-/HCO3- and I- anion transporter pendrin, are associated with non-syndromic hearing loss with enlarged vestibular aqueduct (NSEVA) and Pendred syndrome (PDS). In the Caucasian population, up to 75% of patients fail to identify a genetic cause through biallelic mutations in the SLC26A4 gene. The CEVA haplotype could therefore play an important role in the diagnostics of NSEVA. The aim of the study was to determine the genetic etiology of hearing loss with EVA or with fully developed PDS in 37 probands and the functional characterization of novel variants identified in the SLC26A4 gene. METHODS To determine the genetic etiology, Sanger sequencing, WES and KASP genotyping assay were used. Functional characterization of SLC26A4 variants c.140G>A (p.R47Q), c.415G>A (p.G139R), c.441G>A (p.M147I), c.481T>A (p.F161I), c.1589A>C (p.Y530S) and c.2260del (p.D754Ifs*5) involved determination of iodide influx, total and plasma membrane pendrin expression level and subcellular localization of pendrin by confocal imaging. The nanopore sequencing of nasopharyngeal swab samples was performed to confirm the pathogenic effect of potential splice site variant c.415G>A. RESULTS Biallelic variants in the SLC26A4 gene (M2 genotype) were identified in ten probands and a complete CEVA haplotype was confirmed in three probands harbouring SLC26A4 monoallelic variants (M1 genotype). Fifteen variants in the SLC26A4 gene were identified in total, three of which are novel. The functional characterization of the novel variants and variants which were not yet functionally characterized confirmed the pathogenic potential of five out of six tested variants (p.G139R, p.M147I, p.Y530S, p.D754Ifs*5, and p.F161I). Analysis of nasopharyngeal swab samples confirmed exon 4 skipping due to novel variant SLC26A4:c.415G>A. Probands with biallelic SLC26A4 variants had significantly larger thyroid volume per m2 of body surface area than subjects with monoallelic SLC26A4 variants and the CEVA haplotype. CONCLUSIONS The genetic aetiology was determined in 13 out of 37 probands (35%), seven manifested with PDS and six with NSEVA. The present study highlights the importance of functional testing to confirm the pathogenicity of SLC26A4 variants and the phenotype-genotype correlation in SLC26A4-related disorders.
Collapse
Affiliation(s)
- Marek Sklenar
- Diabgene Laboratory, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava, 845 05, Slovakia
| | - Silvia Borecka
- Diabgene Laboratory, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava, 845 05, Slovakia
| | - Lukas Varga
- Diabgene Laboratory, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava, 845 05, Slovakia
- Department of Otorhinolaryngology - Head and Neck Surgery, Faculty of Medicine and University Hospital Bratislava, Comenius University, Bratislava, Slovakia
| | - Emanuele Bernardinelli
- Institute of Pharmacology and Toxicology, Paracelsus Medical University, Salzburg, 5020, Austria
| | - Juraj Stanik
- Diabgene Laboratory, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava, 845 05, Slovakia
- Department of Paediatrics, Faculty of Medicine, National Institute of Children's Diseases, Bratislava, Slovakia
| | - Martina Skopkova
- Diabgene Laboratory, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava, 845 05, Slovakia
| | - Miroslav Sabo
- Diabgene Laboratory, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava, 845 05, Slovakia
| | - Diana Ugorova
- Department of Otorhinolaryngology - Head and Neck Surgery, Faculty of Medicine and University Hospital Bratislava, Comenius University, Bratislava, Slovakia
| | - Silvia Dossena
- Institute of Pharmacology and Toxicology, Paracelsus Medical University, Salzburg, 5020, Austria
- Research and Innovation Center Regenerative Medicine & Novel Therapies, Paracelsus Medical University, Salzburg, 5020, Austria
| | - Daniela Gasperikova
- Diabgene Laboratory, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava, 845 05, Slovakia.
| |
Collapse
|
3
|
Luo X, Xiao B, Liang L, Zhang K, Xu T, Liu H, Liu Y, Yu Y, Fan Y. Blood RNA-seq in rare disease diagnostics: a comparative study of cases with and without candidate variants. J Transl Med 2025; 23:586. [PMID: 40420094 PMCID: PMC12105386 DOI: 10.1186/s12967-025-06609-w] [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] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Accepted: 05/13/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND Approximately 60% of rare disease cases remain unsolved after exome and genome sequencing (ES/GS). Blood RNA sequencing (RNA-seq) complements DNA-level diagnosis by revealing the functional impact of variants on gene expression and splicing, but to what extent RNA-driven approaches offer diagnostic benefits across different scenarios-with and without pre-existing candidate variants-remains uncertain. METHODS 128 unrelated probands with suspected Mendelian disorders who had previously undergone ES/GS were recruited. A validation cohort (n = 7, with variants expected to alter RNA) and a test cohort (n = 121, including 10 with variants of uncertain significance (VUS) and 111 with no previously identified candidate variants) were analyzed. Blood RNA-seq was performed, and aberrant splicing (AS) and aberrant expression (AE) were detected using the DROP pipeline. SpliceAI predictions were compared with RNA-seq results for splicing-related VUS variants, and pathogenicity was re-evaluated. AS/AE outliers were evaluated for diagnostic potential in cases without candidate variants. The feasibility of an RNA-driven approach was assessed by ranking causal variant-associated aberrant events. RESULTS The pipeline correctly identified all expected AS/AE events in the validation cohort. In the test cohort with candidate VUS, RNA-seq provided a 60% (6/10) diagnostic uplift. Notably, SpliceAI predictions matched RNA-seq observations perfectly only in 40% of these VUS. A 2.7% (3/111) diagnostic uplift was achieved in the test cohort with no prior candidates. Overall, target AS and AE events ranked among the top eight in 14 of the 16 diagnosed cases using a purely RNA-driven approach; however, two cases would have been missed without prior candidate identification from DNA sequencing. CONCLUSION Blood RNA-seq is highly effective in refining the interpretation of splicing VUS, frequently leading to reclassification and diagnosis. Meanwhile, RNA-driven identification of causal variants shows a more modest yield in cases without prior candidates. This study supports an RNA-complementary approach as the preferred strategy for clinical utility.
Collapse
Affiliation(s)
- Xiaomei Luo
- Clinical Genetics Center, Shanghai Institute for Pediatrics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Room 801, Science and Education Building, No.1665, Kong Jiang Road, Shanghai, 200092, China
| | - Bing Xiao
- Clinical Genetics Center, Shanghai Institute for Pediatrics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Room 801, Science and Education Building, No.1665, Kong Jiang Road, Shanghai, 200092, China
| | - Lili Liang
- Department of Pediatric Endocrinology and Genetic Metabolism, Shanghai Institute for Pediatrics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 20092, China
| | - Kaichuang Zhang
- Department of Pediatric Endocrinology and Genetic Metabolism, Shanghai Institute for Pediatrics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 20092, China
| | - Ting Xu
- Clinical Genetics Center, Shanghai Institute for Pediatrics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Room 801, Science and Education Building, No.1665, Kong Jiang Road, Shanghai, 200092, China
| | - Huili Liu
- Clinical Genetics Center, Shanghai Institute for Pediatrics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Room 801, Science and Education Building, No.1665, Kong Jiang Road, Shanghai, 200092, China
| | - Yi Liu
- Clinical Genetics Center, Shanghai Institute for Pediatrics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Room 801, Science and Education Building, No.1665, Kong Jiang Road, Shanghai, 200092, China
| | - Yongguo Yu
- Clinical Genetics Center, Shanghai Institute for Pediatrics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Room 801, Science and Education Building, No.1665, Kong Jiang Road, Shanghai, 200092, China.
- Department of Pediatric Endocrinology and Genetic Metabolism, Shanghai Institute for Pediatrics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 20092, China.
| | - Yanjie Fan
- Clinical Genetics Center, Shanghai Institute for Pediatrics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Room 801, Science and Education Building, No.1665, Kong Jiang Road, Shanghai, 200092, China.
| |
Collapse
|
4
|
Yahia S, Ghozzy D, Wahba Y, Abdelmoneim Z, Yahia S. Neonatal Onset Seizures and Hypotonia Due to D-Bifunctional Protein Deficiency. Indian J Pediatr 2025:10.1007/s12098-025-05582-y. [PMID: 40402439 DOI: 10.1007/s12098-025-05582-y] [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: 12/20/2024] [Accepted: 05/07/2025] [Indexed: 05/23/2025]
Abstract
Peroxisomal disorders (PDs) are a diverse group of inherited conditions arising from impaired function of a specific peroxisomal enzyme, metabolite transporter, or defect in the peroxisome biogenesis system. Peroxisomal D-bifunctional protein (DBP) deficiency is generally classified as a Zellweger-like syndrome. This disorder is caused by mutations in the HSD17B4 gene, and only a limited number of confirmed cases have been reported to date.The authors report case of a 6-mo-old female infant presenting with neonatal-onset intractable seizures, characteristic facial features, hypotonia, and progressive hepatomegaly. An acylcarnitine profile revealed elevated very long-chain fatty acids, prompting the initiation of a medium-chain triglyceride (MCT) formula. Remarkably, this treatment led to seizure control, improved muscle tone, and a reduction in liver size. Whole exome sequencing identified a homozygous missense mutation in the HSD17B4 gene (c.1444A>T).This case suggests that MCT-containing formulas may offer therapeutic potential in the treatment of D-bifunctional protein deficiency.
Collapse
Affiliation(s)
- Sohier Yahia
- Department of Pediatrics, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Dina Ghozzy
- Department of Pediatrics, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Yahya Wahba
- Department of Pediatrics, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Zahraa Abdelmoneim
- Department of Pediatrics, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | - Sohier Yahia
- Department of Pediatrics, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
5
|
Wijnant KN, Nadif Kasri N, Vissers LELM. Systematic analysis of genetic and phenotypic characteristics reveals antisense oligonucleotide therapy potential for one-third of neurodevelopmental disorders. Genome Med 2025; 17:59. [PMID: 40400017 PMCID: PMC12096787 DOI: 10.1186/s13073-025-01477-x] [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] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 04/22/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND Neurodevelopmental disorders (NDDs) are a challenging group of disorders to treat, but promising therapeutic interventions in the form of antisense oligonucleotides (AONs) have emerged in recent years. However, the applicability of AON therapy for NDDs varies based on genetic and phenotypic traits. In this study we systematically evaluated key characteristics for AON therapy suitability in NDDs, to estimate overall therapy potential and identify, both well- and less-studied, targetable NDDs. METHODS An NDD dataset was created and evaluated to identify potentially targetable NDDs for seven AON strategies. This involved examining the presence of a combination of critical factors including disease-gene properties, such as regulatory elements, effects of pathogenic variants, and disease-associated phenotypic features. RESULTS Through the systematic evaluation of the presence of targetable characteristic for each NDD and AON strategy, we identified 711 NDDs (38% of the total) with characteristics favorable for at least one AON strategy and predicted that 18% of affected individuals could benefit from AON therapy. CONCLUSIONS The results from our analysis demonstrate that there might be a more extensive potential for the use of AON therapy in NDDs than was anticipated thus far, underscoring AON therapy as a promising treatment option for NDDs while simultaneously contributing to informed therapy selection.
Collapse
Affiliation(s)
- Kim N Wijnant
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
- Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nael Nadif Kasri
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lisenka E L M Vissers
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands.
- Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
6
|
Vooren EV, den Broeck FV, Mahieu Q, Geens E, Heetvelde MV, De Bruyne M, de Sompele SV, Uppal S, Poliakov E, Dhaenens CM, Gregory-Evans CY, Hoefsloot L, Gonzalez AI, Kohl S, Zuleger T, Demaret T, Dominant RPE65-p.(E519K) Consortium, Tuupanen S, Ruys J, Os LV, Platteau E, Jacob J, Vermeer S, Postelmans L, Dahan K, Maystadt I, Rasquin F, Thiadens AA, Stephenson KA, Sheri N, Smirnov V, MacDonald IM, Gregory-Evans K, Redmond TM, De Zaeytijd J, Leroy BP, Bauwens M, De Baere E. RPE65 variant p.(E519K) causes a novel dominant adult-onset maculopathy in 83 affected individuals. RESEARCH SQUARE 2025:rs.3.rs-5849564. [PMID: 40386434 PMCID: PMC12083654 DOI: 10.21203/rs.3.rs-5849564/v2] [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] [Indexed: 05/25/2025]
Abstract
Recessive RPE65-related retinopathy is an inherited retinal disease (IRD) that is a well-established target for gene therapy. Dominant RPE65-related retinopathy, however, due to Irish founder variant p.(D477G), is extremely rare. Here, we report the discovery, replication and characterization of a novel dominant retinopathy caused by RPE65 variant p.(E519K), identified in 83 individuals of European ancestry across IRD registries (Belgian discovery cohort, n=2,873; replication cohort, n=18,796). Long-read sequencing-based haplotyping revealed a shared region of 464 kb, supporting a founder effect. Genotype-phenotype data support dominant inheritance and phenotypic variability respectively, characterized by late-onset macular dystrophy with two main subtypes, a pathognomonic mottled subtype and a pattern dystrophy subtype. Functional studies showed that the p.(E519K) variant affects RPE65 enzymatic activity, correlating with lower protein expression. Protein modelling and cellular thermal shift assays further supported a destabilizing effect on protein structure. Overall, our work provides strong genetic, clinical, molecular and functional evidence for a novel dominant RPE65 retinopathy in multiple families in Europe and North America due to a Belgian founder variant. This discovery reduces the diagnostic gap in dominant IRD, particularly in individuals of European ancestry. Finally, it lays the foundation for developing therapeutic strategies targeting dominant RPE65 retinopathy.
Collapse
Affiliation(s)
- Eline Van Vooren
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Center for Medical Genetics, Ghent University Hospital, Ghent, 9000, Belgium
| | - Filip Van den Broeck
- Department of Head and Skin, Ghent University, Ghent, 9000, Belgium
- Department of Ophthalmology, Ghent University Hospital, Ghent, 9000, Belgium
| | - Quinten Mahieu
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Center for Medical Genetics, Ghent University Hospital, Ghent, 9000, Belgium
| | - Eline Geens
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Center for Medical Genetics, Ghent University Hospital, Ghent, 9000, Belgium
| | - Mattias Van Heetvelde
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Center for Medical Genetics, Ghent University Hospital, Ghent, 9000, Belgium
| | - Marieke De Bruyne
- Center for Medical Genetics, Ghent University Hospital, Ghent, 9000, Belgium
| | | | - Sheetal Uppal
- Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, NIH, Bethesda, 20892, United States
| | - Eugenia Poliakov
- Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, NIH, Bethesda, 20892, United States
| | - Claire-Marie Dhaenens
- Univ. Lille, Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience & Cognition, F-59000 Lille, 59000, France
| | - Cheryl Y. Gregory-Evans
- University of British Columbia, Department of Ophthalmology & Visual Sciences, Vancouver, BC, V6T 1Z4, Canada
| | - Lies Hoefsloot
- Department of Clinical Genetics, Erasmus Medical Centre, Rotterdam, 3015, The Netherlands
| | | | - Susanne Kohl
- Institute for Ophthalmic Research, Centre for Ophthalmology, University Hospital Tübingen, 72074, Germany
| | - Theresia Zuleger
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, 72074, Germany
| | - Tanguy Demaret
- Centre de Génétique Humaine, Institut de Pathologie et de Génétique, Gosselies, 6041, Belgium
| | | | | | - Joke Ruys
- Department of Ophthalmology, Ghent University Hospital, Ghent, 9000, Belgium
- Department of Ophthalmology, Vitaz, Sint-Niklaas, 9100, Belgium
| | - Luc Van Os
- Department of Ophthalmology, Antwerp University Hospital, Antwerp, 2650, Belgium
| | - Elise Platteau
- Department Ophthalmology, Maria Middelares Hospital, Ghent, 9000, Belgium
| | - Julie Jacob
- Department of Ophthalmology, UZ Leuven, Leuven, 3000, Belgium
| | - Sascha Vermeer
- Center for Human Genetics, UZ Leuven, Leuven, 3000, Belgium
| | | | - Karin Dahan
- Centre de Génétique Humaine, Institut de Pathologie et de Génétique, Gosselies, 6041, Belgium
| | - Isabelle Maystadt
- Centre de Génétique Humaine, Institut de Pathologie et de Génétique, Gosselies, 6041, Belgium
| | - Florence Rasquin
- Department of ophthalmology, Erasme Hospital, Université Libre de Bruxelles, Brussels, 1070, Belgium
| | | | - Kirk A.J. Stephenson
- University of British Columbia, Department of Ophthalmology & Visual Sciences, Vancouver, BC, V6T 1Z4, Canada
| | - Narin Sheri
- Department of Ophthalmology and Visual Sciences, University of Alberta, Edmonton, T6G 2R3, Canada
| | - Vasily Smirnov
- Univ. Lille, Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience & Cognition, F-59000 Lille, 59000, France
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, 75012, France
| | - Ian M. MacDonald
- Department of Ophthalmology and Visual Sciences, University of Alberta, Edmonton, T6G 2R3, Canada
| | - Kevin Gregory-Evans
- University of British Columbia, Department of Ophthalmology & Visual Sciences, Vancouver, BC, V6T 1Z4, Canada
| | - T. Michael Redmond
- Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, NIH, Bethesda, 20892, United States
| | - Julie De Zaeytijd
- Department of Ophthalmology, Ghent University Hospital, Ghent, 9000, Belgium
| | - Bart P. Leroy
- Center for Medical Genetics, Ghent University Hospital, Ghent, 9000, Belgium
- Department of Head and Skin, Ghent University, Ghent, 9000, Belgium
- Department of Ophthalmology, Ghent University Hospital, Ghent, 9000, Belgium
- Division of Ophthalmology and Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, United States
| | - Miriam Bauwens
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Center for Medical Genetics, Ghent University Hospital, Ghent, 9000, Belgium
| | - Elfride De Baere
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Center for Medical Genetics, Ghent University Hospital, Ghent, 9000, Belgium
| |
Collapse
|
7
|
Ying D, Kwok JSL, Chu ATW, Ma W, Tam HYF, Or D, Hue SPY, Li Q, Leung CKS, Chung BHY. Accelerating genetic diagnostics in retinitis pigmentosa: implementation of a semi-automated bespoke cohort analysis workflow for Hong Kong Genome Project. Hum Genet 2025; 144:515-528. [PMID: 40163143 PMCID: PMC12033112 DOI: 10.1007/s00439-025-02737-x] [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: 09/19/2024] [Accepted: 03/13/2025] [Indexed: 04/02/2025]
Abstract
The study aims to enhance the efficiency of the genetic variant curation process at the Hong Kong Genome Institute by developing a Semi-Automated Bespoke Cohort Analysis Workflow (S-BCAW) for patients with, or suspected to have, retinitis pigmentosa (RP) in the Hong Kong Genome Project (HKGP), leveraging advances in next-generation sequencing (NGS). A comparative analysis involving 79 RP patients was conducted using both the conventional manual workflow and the novel S-BCAW, which integrates initial filtering and variant classification based on ACMG guidelines, followed by detailed manual review. The diagnostic yields from both methods were identical, but the bespoke workflow reduced analysis time by approximately 60% (1.5 h/sample). This efficiency increase resulted from automated application of ACMG rules and systematic aggregation of supportive data, including disease-specific information. The study reports 25 positive cases with a diagnostic yield of 32%, including three novel variants. The S-BCAW significantly improves efficiency, helping to end the diagnostic odyssey for patients in the HKGP. This approach facilitates rapid assessment of variant pathogenicity, enhancing the feasibility and timeliness of NGS technology for clinical applications, especially in urgent scenarios.
Collapse
Affiliation(s)
- Dingge Ying
- Hong Kong Genome Institute, Hong Kong, China
| | | | | | - Wei Ma
- Hong Kong Genome Institute, Hong Kong, China
| | | | - Dicky Or
- Hong Kong Genome Institute, Hong Kong, China
| | | | - Qing Li
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Christopher Kai Shun Leung
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
- Department of Ophthalmology, Queen Mary Hospital, Pok Fu Lam, Hong Kong, China.
- Hong Kong Eye Hospital, Kowloon City, Hong Kong, China.
- Grantham Hospital, Hong Kong, China.
| | - Brian Hon Yin Chung
- Hong Kong Genome Institute, Hong Kong, China.
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
8
|
Bertoli-Avella AM, Radefeldt M, Al-Ali R, Pardo LM, Lemke S, Leubauer A, Polla DL, Hörnicke R, Almeida LS, Kandaswamy KK, Beetz C, Pinto Basto J, Bauer P. Beyond genomics: using RNA-seq from dried blood spots to unlock the clinical relevance of splicing variation in a diagnostic setting. Eur J Hum Genet 2025; 33:614-623. [PMID: 39870877 PMCID: PMC12048715 DOI: 10.1038/s41431-025-01792-2] [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: 06/26/2024] [Revised: 12/20/2024] [Accepted: 01/15/2025] [Indexed: 01/29/2025] Open
Abstract
We aimed to assess the impact of splicing variants reported in our laboratory to gain insight into their clinical relevance. A total of 108 consecutive individuals, for whom 113 splicing variants had been reported, were selected for RNA-sequencing (RNA-seq), considering the gene expression in blood. A protocol was developed to perform RNA extraction and sequencing using the same sample (dried blood spots, DBS) provided for the DNA analysis, including library preparation and bioinformatic pipeline analysis. Splicing in genes of interest was inspected using IGV, with at least three unaffected individuals as controls. From the 113 variants, we confirmed an abnormal splicing in 64 variants (57%). In 15 variants (13%), we did not observe a splicing alteration. In the remaining 34 variants, no decision could be made on the splicing effect due to insufficient sample quality (21%) or a low number of reads (9%). The most common event leading to aberrant splicing was exon skipping, identified in 31 variants (48%). Other events included cryptic donor/acceptor site usage (n = 25; 39%), intron retention (n = 4; 6%), and other complex events (n = 4; 6%). In three patients, pathologically reduced enzymatic activity (measured using the same DBS) served as additional confirmation of the abnormal splicing caused by variants in HEXA, GAA, and GLA. We implemented an RNA-seq pipeline using the same sample provided for genomic testing. This multiomic approach, as implemented in our routine diagnostic processes, clarifies the clinical relevance of most of the analyzed variants and delivers more comprehensive genetic testing.
Collapse
|
9
|
Yang S, Li Z, Ren X, Yue J. A Compound Heterozygous Pathogenic Variant in ZP2 Gene Causes Female Infertility. Reprod Sci 2025; 32:1557-1565. [PMID: 39443359 DOI: 10.1007/s43032-024-01729-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: 11/20/2023] [Accepted: 10/10/2024] [Indexed: 10/25/2024]
Abstract
The oocyte maturation defect 6 is an autosomal recessive hereditary disease caused by a homozygous variant in ZP2 gene. It is characterized by female primary infertility due to an abnormally thin zona pellucida (ZP) and defective sperm binding. Here we identified a compound heterozygous variant (c.1924C > T and c.1695-2A > G) in ZP2 gene in a Chinese Han family. Quantitative real-time PCR showed that the variant c.1924C > T significantly decreased the expression of truncated ZP2 message RNA by the nonsense-mediated decay pathway. Minigene assays showed the c.1695-2A > G variant led to an extra-61-nt preservation of intron 15 at the junction between exons 15 and 16 during transcription. Both variants (c.1924C > T and c.1695-2A > G) resulted in truncated ZP2 proteins (p.R642X and p.C566Hfs*2) that lost the transmembrane domain, which prevented the secretion of the mutant ZP2 proteins and produced a structurally abnormal ZP, thus resulting in female infertility. This study further elucidated the pathogenic mechanism of these two variants and provided new support for the genetic diagnosis of female infertility.
Collapse
Affiliation(s)
- Shulin Yang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Zongzhe Li
- Division of Cardiology, Departments of Internal Medicine and Genetic Diagnosis Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinling Ren
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| | - Jing Yue
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
10
|
Rawnsley K, Weisschuh N, Kohl S, Reuter P. Comprehensive functional splicing analysis of non-canonical CNGB3 variants using in vitro minigene splice assays. J Pathol 2025. [PMID: 40304364 DOI: 10.1002/path.6431] [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: 10/29/2024] [Revised: 02/10/2025] [Accepted: 03/26/2025] [Indexed: 05/02/2025]
Abstract
Variants in the CNGB3 gene, encoding the B3-subunit of the cone photoreceptor cyclic nucleotide gated channel, are a major cause of autosomal recessive achromatopsia, a rare inherited retinal disease. The mutation spectrum of achromatopsia-associated CNGB3 variants comprises all types of mutations, including those that are straightforward to evaluate in molecular genetic diagnostics, such as frame-shifting, nonsense, and canonical splice site variants. Additionally, variants have been identified within splice regions outside the conserved ±1,2 splice site dinucleotides, making their potential impact on disease association challenging to interpret. This poses a major hurdle for clinical interpretation of causality between the patient's genotype and the proposed clinical diagnosis, but also for the inclusion of such patients into clinical trials for gene augmentation therapy, for which only patients with confirmed (likely) pathogenic CNGB3 variants are eligible. We here performed comprehensive genetic functional analysis of 21 candidate spliceogenic CNGB3 variants-15 reported and 6 novel variants-by means of in vitro minigene splice assays and cDNA analysis, and characterization of spliceogenic events by subcloning, Sanger-sequencing, and capillary fragment analysis. For 16 variants, an impact on splicing was confirmed, supporting the reclassification of 86% of variants of uncertain significance as likely pathogenic or pathogenic according to the ACMG/AMP guidelines. This reclassification enables the confirmation of patients' genotypes, both retrospectively and prospectively. © 2025 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Katharina Rawnsley
- Institute for Ophthalmic Research, Centre for Ophthalmology, University Hospital Tübingen, Tübingen, Germany
| | - Nicole Weisschuh
- Institute for Ophthalmic Research, Centre for Ophthalmology, University Hospital Tübingen, Tübingen, Germany
| | - Susanne Kohl
- Institute for Ophthalmic Research, Centre for Ophthalmology, University Hospital Tübingen, Tübingen, Germany
| | - Peggy Reuter
- Institute for Ophthalmic Research, Centre for Ophthalmology, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
11
|
Bu X, Yu X, Zeng L, Zeng G, Tan R, Peng C, Zhou S, Linpeng S, Liu J. A retrospective single center analysis of fetuses with region of homozygosity detected by single nucleotide polymorphism array. Sci Rep 2025; 15:13623. [PMID: 40253570 PMCID: PMC12009308 DOI: 10.1038/s41598-025-98497-9] [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: 08/11/2024] [Accepted: 04/11/2025] [Indexed: 04/21/2025] Open
Abstract
We assessed the incidence and clinical significance of the fetal region of homozygosity (ROH) detected using single nucleotide polymorphism (SNP) array by analyzing clinical information and pregnancy outcomes. We collected data on 6176 mid- and late pregnancies. All fetuses were subjected to SNP array analysis. Fetuses with ROH were analyzed by karyotyping, parental SNP array verification, whole-exome sequencing, and/or placental studies. Eighty-seven ROHs met our reporting thresholds. Thirty-four fetuses were detected from noninvasive prenatal testing-positive results, with the most common detection rate (2.03%). Twenty-four cases were diagnosed using ultrasound abnormalities; fetal growth restriction was the indication with the highest diagnostic rate. Fifteen cases of uniparental disomy in mid- and late pregnancy were identified (0.24%). Nine cases were of ROH accompanied by aneuploidy or pathogenic/likely pathogenic copy number variants with an adverse pregnancy outcome rate of 88.9%. Of the remaining 78 cases, 14 carriers had adverse outcomes (including two cases of imprinting syndrome), 63 had normal development after birth, and one was lost to follow-up. ROH is relatively common in mid- and late-term pregnancies; its incidence is higher than that reported previously. SNP array is effective in assessing ROH and should be combined with multiple techniques to evaluate ROH's clinical relevance.
Collapse
Affiliation(s)
- Xiufen Bu
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, China
| | - Xiuyun Yu
- Department of Obstetrics, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, China
| | - Li Zeng
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, China
| | - Guo Zeng
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, China
| | - Rong Tan
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, China
| | - Can Peng
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, China
| | - Shihao Zhou
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, China
| | - Siyuan Linpeng
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, China.
| | - Jing Liu
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, China.
| |
Collapse
|
12
|
Luo Y, Guo N, Wang Y, Li J. Novel homozygous frameshift mutation of ITGB3 in the Glanzmann thrombasthenia patient with abnormal bone metabolism and congenital bone defects. Orphanet J Rare Dis 2025; 20:189. [PMID: 40251671 PMCID: PMC12007121 DOI: 10.1186/s13023-025-03700-9] [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: 10/12/2024] [Accepted: 03/24/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Glanzmann thrombasthenia (GT) is a rare inherited bleeding disorder caused by dysfunction of the integrin αIIbβ3 in platelets. The subunit β3, encoded by ITGB3 also plays a significant role in bone metabolism. Whether GT patients with β3 deficiency also suffer from bone pathology remains unclear. METHOD The 21-year-old female patient presenting with bleeding diathesis and multiple congenital bone defects in her right hand, and her seven family members were included in the study. Whole exome sequencing as well as Sanger sequencing were conducted to identify GT-associated mutations within the family. The platelet function of the family was detected by the platelet aggregation test and thromboelastography (TEG). The expression levels of CD41 (αIIb) and CD61 (β3) on the platelet surface and total in platelet were detected by flow cytometry and Western blot. Bioinformatics analysis was used to evaluate the pathogenicity of mutation sites and their effects on protein structure and function. X-ray imaging, bone densitometry and bone metabolism index were performed to evaluate bone development and metabolism. RESULT A novel homozygous frameshift mutation c.2143_2158delinsCT (p.Lys715Leufs*36) of ITGB3 was found in the proband. Platelet aggregation by ADP, collagen, epinephrine, and arachidonic acid was absent, TEG showed hypocoagulability and decreased platelet function, and the expression levels of αIIb and β3 on the platelet surface and total in platelet were significantly reduced (< 5%) in the proband. The parents, second elder sister and grandmother of proband were heterozygous carriers without bleeding symptoms and had normal platelet aggregation function and αIIb/β3 protein expression. Structural modeling strongly suggested that the mutation creates a truncation in cytoplasmic domains of β3, resulting in the mutant β3/αIIbβ3 inactivated and low expression. The proband was born with partial absence of phalanges in digits 2-4 and the deformity of fingers 1 and 5 in her right hand, bone densitometry indicated significant osteopenia and increased risk of fracture in her right radius, and no other gene mutations related to bone pathology were identified. CONCLUSION A novel mutation of ITGB3 which results in GT was identified. This is the third reported case of GT combined with bone defect. Our work expands ITGB3 mutation spectrum and provide further insights into the potential association between GT and bone development and metabolism.
Collapse
Affiliation(s)
- Yujiao Luo
- Department of Hematology, Section of Hemostasis and Thrombosis, Institute of Molecular Hematology, The Second XiangYa Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, China
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Nina Guo
- Department of Hematology and Oncology, The Third Hospital of Changsha, Changsha, 410011, Hunan, China
| | - Yewei Wang
- Department of Hematology, Section of Hemostasis and Thrombosis, Institute of Molecular Hematology, The Second XiangYa Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Ji Li
- Department of Hematology, Section of Hemostasis and Thrombosis, Institute of Molecular Hematology, The Second XiangYa Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, China.
| |
Collapse
|
13
|
García-Aznar JM, Besada-Cerecedo ML, Castro-Alonso C, Sierra Carpio M, Blasco M, Quiroga B, Červienka M, Mouzo R, Torra R, Ortiz A, de Sequera P. Novel Truncating Variants in PODXL Represent a New Entity to Be Explored Among Podocytopathies. Genes (Basel) 2025; 16:464. [PMID: 40282423 PMCID: PMC12026838 DOI: 10.3390/genes16040464] [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: 03/11/2025] [Revised: 04/12/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES Podocalyxin is a sialoprotein mainly expressed in the kidney cortex and lung tissue, which has been described as a component of the podocyte glycocalyx. This protein promotes the reorganization of the podocyte cytoskeleton, as well as the morphogenesis and differentiation of nascent podocytes, actively participating in glomerular filtration. Previous research has suggested that PODXL haploinsufficiency leads to podocytopathy with development of focal segmental glomerulosclerosis, a disorder that has been demonstrated in Podxl-deficient animal models and proposed as a primary cause in human families affected by this condition. However, only a few families have been reported, which limits the understanding about the spectrum of phenotype and prognosis of the disease. METHODS We performed high-throughput sequencing in a cohort of young adults with CKD, describing the clinical scenario of those who harbored truncating variants in the PODXL gene and testing the families for detected variants. RESULTS The PODXL gene exhibited a slight deviation in loss intolerance probability and moderate deviation in the observed/expected ratio of variation, which is typically observed in dominant genes with age-dependent incomplete penetrance or variable expression. We reported four novel truncating variants in the PODXL gene, along with a collection of previously published monoallelic truncating variants. CONCLUSIONS These findings further support evidence about genetic defects in the PODXL gene associated with a new molecular entity of podocytopathy with adult onset. Additionally, the nucleotide sequence of PODXL contains particularities that require careful analysis to interpret the effect of the variants detected in this gene.
Collapse
Affiliation(s)
| | | | - Cristina Castro-Alonso
- Department of Nephrology, Doctor Peset University Hospital, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO), 46020 Valencia, Spain
| | | | - Miquel Blasco
- Nephrology and Kidney Transplant Department, National Reference Center for Complex Glomerular Diseases (CSUR), Hospital Clínic, Barcelona University, 08036 Barcelona, Spain
- Fundació de Recerca Clínic Barcelona-Institut d’Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), 08036 Barcelona, Spain
| | - Borja Quiroga
- Nephrology Department, Hospital Universitario de la Princesa, 28006 Madrid, Spain
| | - Michal Červienka
- Nephrology Department, Hospital El Bierzo, 24404 Ponferrada, Spain
| | - Ricardo Mouzo
- Nephrology Department, Hospital El Bierzo, 24404 Ponferrada, Spain
| | - Roser Torra
- Nephrology Department, Fundació Puigvert, 08025 Barcelona, Spain
| | - Alberto Ortiz
- Nephrology and Hypertension Department, IIS-Fundación Jiménez Díaz UAM, 28040 Madrid, Spain
- Medicine Department, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain
| | - Patricia de Sequera
- Nephrology Department, Hospital Universitario Infanta Leonor, 28031 Madrid, Spain
- Medicine Department, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| |
Collapse
|
14
|
Veitia RA, Cowles JD, Caburet S. Reclassifying NOBOX variants in primary ovarian insufficiency cases with a corrected gene model and a novel quantitative framework. Hum Reprod 2025:deaf058. [PMID: 40246288 DOI: 10.1093/humrep/deaf058] [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: 06/06/2024] [Revised: 02/19/2025] [Indexed: 04/19/2025] Open
Abstract
STUDY QUESTION How updated expression and genomic data combined with a disease/disorder-specific classification system can be used to correct a gene model for a better evaluation of the pathogenicity of variants found in patients? SUMMARY ANSWER By combining available genomic and transcriptomic data from several species and a quantitative classification framework with primary ovarian insufficiency (POI)-adjusted parameters, we correct the human NOBOX (newborn ovary homeobox) gene model and provide a reclassification of variants previously reported in POI cases. WHAT IS KNOWN ALREADY The NOBOX gene, encoding a gonad-specific transcription factor with a crucial role in early folliculogenesis and considered a major gene involved in POI, is currently described as being expressed as four transcripts, the longest one considered canonical. All the variants identified in POI cases have been evaluated according to this canonical transcript, and the various functional tests have been performed using the corresponding predicted protein. STUDY DESIGN, SIZE, DURATION We refined and corrected the NOBOX gene model using available genomic and RNAseq data in human and 16 other mammalian species. Expression data were selected for tissue specificity, strand specificity, and coverage. The analysis of RNAseq data from different ovarian fetal stages allows for a time-course description of NOBOX isoforms. Literature was scanned to retrieve NOBOX variants reported in POI cases, and NOBOX variants present in ClinVar and GnomAD 4 databases were also retrieved. PARTICIPANTS/MATERIALS, SETTING, METHODS Strand-specific RNAseq data from human fetal ovaries and human adult testes were analysed to infer the correct human NOBOX gene isoforms. The conservation of the gene structure was verified by combining the aligned genomic sequences from 17 mammalian species covering a wide phylogenetic range and the relevant RNAseq data. As changing a gene model implies a reclassification of variants, we set up a quantitative framework with updated variant frequencies from GnomAD4 and POI-adjusted parameters following the American College of Medical Genetics and Genomics/Association for Molecular Pathology (ACMG/AMP) guidelines. Using this framework, we reclassified 44 NOBOX variants reported in POI patients and families, 117 NOBOX variants reported in ClinVar, and 2613 NOBOX variants present in GnomAD4. MAIN RESULTS AND THE ROLE OF CHANCE The corrected NOBOX gene model proposes the invalidation of two transcripts, including the canonical one. The two correct isoforms were present in fetal ovarian samples, and only one was detected in adult testes. Only 14 variants remained as possibly causative for POI. Furthermore, this re-evaluation strongly suggests that NOBOX biallelic variants are the most likely cause of POI. LARGE SCALE DATA Large tables are provided as supplementary data sets on the Zenodo repository. LIMITATIONS, REASONS FOR CAUTION The proposed gene model is robust but relies on available transcriptomic data covering a range of time points and tissues. Our scoring system was manually adjusted and other laboratories can implement it with different parameters. WIDER IMPLICATIONS OF THE FINDINGS For the NOBOX variants that cannot be considered pathogenic or causative anymore, the genome/exome sequencing data of the corresponding patients should be reanalysed. Furthermore, the functional studies performed using the obsolete coding sequence should be reconsidered. The corrected gene model should be taken into account when evaluating novel NOBOX variants identified in POI patients. Our results highlight the importance of the careful assessment of the most updated expression data for validating a gene model, enabling a correct evaluation of the pathogenicity of variants found in patients. The proposed quantitative framework developed here can be used for the classification of variants in other genes underlying POI. Furthermore, the global approach based on quantitatively adjusting the ACMG/AMP guidelines could be extended to other inherited pathologies. STUDY FUNDING/COMPETING INTEREST(S) This project was not funded. All the authors have no conflict of interest to disclose.
Collapse
Affiliation(s)
- Reiner A Veitia
- Department of Life Sciences, Université Paris Cité, CNRS, Institut Jacques Monod, CNRS UMR7592, Paris, France
- Department of Life Sciences, Université Paris Saclay, Gif-sur-Yvette, France
- Institut de Biologie François Jacob, CEA, Fontenay aux Roses, France
| | - Jamie D Cowles
- Department of Life Sciences, Université Paris Cité, CNRS, Institut Jacques Monod, CNRS UMR7592, Paris, France
| | - Sandrine Caburet
- Department of Life Sciences, Université Paris Cité, CNRS, Institut Jacques Monod, CNRS UMR7592, Paris, France
| |
Collapse
|
15
|
Min J, Luo Y, Fu Q, Sun X, Mi L, Shen Y, Wang H. Clinical spectrum, genetics and management insights of PAX2-related disorder in nine children. Eur J Med Res 2025; 30:276. [PMID: 40229647 PMCID: PMC11998127 DOI: 10.1186/s40001-025-02522-6] [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: 02/18/2025] [Accepted: 03/27/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Renal coloboma syndrome (RCS) is a rare autosomal dominant genetic disorder associated with the paired box gene 2 (PAX2). Over the past three decades, PAX2 variants have manifested with vast phenotype and genotype heterogeneity, with limited experience gained regarding its clinical progression and management strategies. METHODS We included nine Chinese children diagnosed with PAX2-related disorder at Beijing Children's Hospital. The medical records were retrospectively reviewed, and the demographic data, clinical manifestations, genetic testing results, imaging examination findings, laboratory tests, and renal biopsy results were recorded and analyzed. RESULTS Eight distinct PAX2 pathogenic variants, including four novel variants, were identified in nine children. All patients exhibited developmental abnormalities of the urinary system, while two presented with ocular abnormalities with retinal myelinated nerve fibers identified as a novel manifestation of PAX2-related disorder. Cranial imaging examinations were performed on three patients, revealing vascular anatomical abnormalities. Five patients received empirical treatment with angiotensin-converting enzyme inhibitors (ACEIs), resulting in varied therapeutic outcomes. CONCLUSION PAX2 variants exhibit significant clinical diversity, and our findings further expand the genotype-phenotype spectrum of PAX2-related disorder. The variable efficacy of ACEIs highlights the importance of personalized evaluation and treatment strategies and extended follow-up periods in the future.
Collapse
Affiliation(s)
- Jie Min
- Department of Nephrology, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, China
- Department of Nephrology, Key Laboratory of Basic and Clinical Pediatric Nephrology, Baoding Hospital of Beijing Children's Hospital, Regional Center for Children's Health, Capital Medical University, Baoding, 071000, China
| | - Yulin Luo
- Department of Nephrology, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, China
- Department of Nephrology, Key Laboratory of Basic and Clinical Pediatric Nephrology, Baoding Hospital of Beijing Children's Hospital, Regional Center for Children's Health, Capital Medical University, Baoding, 071000, China
| | - Qian Fu
- Department of Nephrology, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, China
- Department of Nephrology, Key Laboratory of Basic and Clinical Pediatric Nephrology, Baoding Hospital of Beijing Children's Hospital, Regional Center for Children's Health, Capital Medical University, Baoding, 071000, China
| | - Xiaona Sun
- Department of Ophthalmology, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
| | - Lan Mi
- Department of Nephrology, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, China
- Department of Nephrology, Key Laboratory of Basic and Clinical Pediatric Nephrology, Baoding Hospital of Beijing Children's Hospital, Regional Center for Children's Health, Capital Medical University, Baoding, 071000, China
| | - Yutian Shen
- Department of Nephrology, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, China
| | - Hui Wang
- Department of Nephrology, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, China.
- Department of Nephrology, Key Laboratory of Basic and Clinical Pediatric Nephrology, Baoding Hospital of Beijing Children's Hospital, Regional Center for Children's Health, Capital Medical University, Baoding, 071000, China.
| |
Collapse
|
16
|
Sullivan PJ, Quinn JMW, Ajuyah P, Pinese M, Davis RL, Cowley MJ. Data-driven insights to inform splice-altering variant assessment. Am J Hum Genet 2025; 112:764-778. [PMID: 40056912 DOI: 10.1016/j.ajhg.2025.02.012] [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: 03/11/2024] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 04/06/2025] Open
Abstract
Disease-causing genetic variants often disrupt mRNA splicing, an intricate process that is incompletely understood. Thus, accurate inference of which genetic variants will affect splicing and what their functional consequences will be is challenging, particularly for variants outside of the essential splice sites. Here, we describe a set of data-driven heuristics that inform the interpretation of human splice-altering variants (SAVs) based on the analysis of annotated exons, experimentally validated SAVs, and the currently understood principles of splicing biology. We defined requisite splicing criteria by examining around 202,000 canonical protein-coding exons and 19,000 experimentally validated splicing branchpoints. This analysis defined the sequence, spacing, and motif strength required for splicing, with 95.9% of the exons examined meeting these criteria. By considering over 12,000 experimentally validated variants from the SpliceVarDB, we defined a set of heuristics that inform the evaluation of putative SAVs. To ensure the applicability of each heuristic, only those supported by at least 10 experimentally validated variants were considered. This allowed us to establish a measure of spliceogenicity: the proportion of variants at a location (or motif site) that affected splicing in a given context. This study makes considerable advances toward bridging the gap between computational predictions and the biological process of splicing, offering an evidence-based approach to identifying SAVs and evaluating their impact. Our splicing heuristics enhance the current framework for genetic variant evaluation with a robust, detailed, and comprehensible analysis by adding valuable context over traditional binary prediction tools.
Collapse
Affiliation(s)
- Patricia J Sullivan
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia; School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Sydney, NSW, Australia; University of New South Wales Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia
| | - Julian M W Quinn
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Pamela Ajuyah
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Mark Pinese
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia; School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Sydney, NSW, Australia
| | - Ryan L Davis
- Neurogenetics Research Group, Kolling Institute, University of Sydney and Northern Sydney Local Health District, St. Leonards, NSW, Australia; School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Mark J Cowley
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia; School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Sydney, NSW, Australia.
| |
Collapse
|
17
|
Romo-Aguas JC, de Guimarães TAC, Kalitzeos A, Aychoua N, Tsika C, Robson AG, Fujinami-Yokokawa Y, Fujinami K, Mahroo OA, Webster AR, Michaelides M. Detailed Clinical, Ophthalmic, and Genetic Characterization of MYO7A-Associated Usher Syndrome. Invest Ophthalmol Vis Sci 2025; 66:60. [PMID: 40257781 PMCID: PMC12020961 DOI: 10.1167/iovs.66.4.60] [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: 11/23/2024] [Accepted: 03/22/2025] [Indexed: 04/22/2025] Open
Abstract
Purpose To analyze the clinical spectrum and natural history of MYO7A-associated Usher syndrome type I (USH1). Methods Patients with molecularly confirmed MYO7A-associated USH1 in a single tertiary referral center. Data was extracted from physical and electronic case notes, including imaging and electrophysiology. Genetic results were reviewed, and the detected variants were assessed. Main outcome measures were clinical findings, qualitative and quantitative analysis of retinal imaging, and electrophysiology. Results Eighty patients were identified and evaluated longitudinally. The mean age (±SD) of onset of symptoms was 12.0 ± 5.8 years of age, and a mean follow-up time of 16.2 years. BCVA was 0.4 ± 0.5 LogMAR at baseline, and 0.7 ± 0.8 LogMAR at the last visit for both eyes. The change in BCVA over time was 0.02 LogMAR per year. A hyperautofluorescent (hyperAF) ring was present in 51% of the patients. The mean ellipsoid zone width (EZW) at baseline was 2568.2 ± 1528.9 µm OD and 2527.9 ± 1609.3 µm OS, which decreased to 2012.3 ± 1705.1 µm OD and 1806.3 ± 1647.1 µm OS at last visit. Electrophysiology revealed rod and cone dysfunction with relative or complete sparing of macular function. There were statistically significant changes in BCVA, EZW, and hyperAF ring between baseline and follow-up. Genetic analysis identified 83 variants in MYO7A, including 18 novel variants. Conclusions Longitudinal analysis shows that the majority of patients retain central visual function and structure until the fifth decade of life, which informs advice on prognosis and the window for therapeutic intervention.
Collapse
Affiliation(s)
- Juan C. Romo-Aguas
- Moorfields Eye Hospital, London, United Kingdom
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Thales A. C. de Guimarães
- Moorfields Eye Hospital, London, United Kingdom
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Angelos Kalitzeos
- Moorfields Eye Hospital, London, United Kingdom
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Nancy Aychoua
- Moorfields Eye Hospital, London, United Kingdom
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Chrysanthi Tsika
- Moorfields Eye Hospital, London, United Kingdom
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Anthony G. Robson
- Moorfields Eye Hospital, London, United Kingdom
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Yu Fujinami-Yokokawa
- Moorfields Eye Hospital, London, United Kingdom
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
- Laboratory of Visual Physiology, Division of Vision Research, National Institute of Sensory Organs, NHO Tokyo Medical Center, Meguro-ku, Tokyo, Japan
| | - Kaoru Fujinami
- Moorfields Eye Hospital, London, United Kingdom
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
- Laboratory of Visual Physiology, Division of Vision Research, National Institute of Sensory Organs, NHO Tokyo Medical Center, Meguro-ku, Tokyo, Japan
| | - Omar A. Mahroo
- Moorfields Eye Hospital, London, United Kingdom
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Andrew R. Webster
- Moorfields Eye Hospital, London, United Kingdom
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Michel Michaelides
- Moorfields Eye Hospital, London, United Kingdom
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| |
Collapse
|
18
|
Uner OE, Elsharawi R, Reynolds M, Bacci GM, Bargiacchi S, Birch DG, Chen FK, Jain N, Heath Jeffery RC, Lamey TM, Mustafi D, da Palma MM, Sallum JMF, Torres Soto M, Jones K, Yang P, Pennesi ME, Everett LA. Phosphoribosyl pyrophosphate synthetase 1 ( PRPS1) associated retinal degeneration: an international study. Ophthalmic Genet 2025; 46:133-143. [PMID: 39763288 PMCID: PMC12064003 DOI: 10.1080/13816810.2024.2444619] [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: 07/03/2024] [Revised: 11/06/2024] [Accepted: 12/14/2024] [Indexed: 04/17/2025]
Abstract
INTRODUCTION Phosphoribosyl pyrophosphate synthetase 1 (PRPS1) is an X-linked gene critical for nucleotide metabolism. Pathogenic PRPS1 variants cause three overlapping phenotypes: Arts syndrome (severe neurological disease), Charcot-Marie-Tooth type 5 [CMTX5] (peripheral neuropathy), and non-syndromic sensorineural hearing loss (SNHL). Each may be associated with retinal dystrophy. Multicenter phenotypic studies are limited. METHODS A multicenter retrospective clinical case series of 15 patients from 12 pedigrees with PRPS1-associated retinal degeneration is presented. RESULTS Of 15 patients, 11 (73.3%) were female. Mean age of ocular disease onset was 8.5 years (range, 0.5-35 years). Many were diagnosed with Leber congenital amaurosis prior to genetic testing (n = 5). Five patients had clinical diagnoses of CMTX5 and Arts syndrome, two had isolated ocular disease, and one was asymptomatic. Mean initial VA (LogMAR) was 0.74, 0.74, 0.83, and 0.85 for isolated ocular disease, CMTX5, Arts, and SNHL, respectively. Ten patients were hyperopic and eight had asymmetric VA. Macular atrophy (n = 13), optic atrophy (n = 13), bone spicules (n = 10), and parafoveal outer retinal atrophy (n = 12) were common findings. Electroretinogram showed delayed and attenuated photopic and scotopic responses (n = 10). Median follow-up of 2.9 years (range, 1.5-11.6 years) in six patients showed retinal disease progression in two patients. DISCUSSION PRPS1-associated retinal degeneration predominantly manifests as a bilateral asymmetric cone and rod dystrophy, commonly associated with hyperopia and optic atrophy.
Collapse
Affiliation(s)
- Ogul E Uner
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Radwa Elsharawi
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Margaret Reynolds
- Department of Ophthalmology, St. Louis Children's Hospital, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Giacomo M Bacci
- Pediatric Ophthalmology Unit, Meyer Children's Hospital IRCCS, Firenze, Toscana, Italy
| | - Sara Bargiacchi
- Medical Genetics Unit, Meyer Children's Hospital IRCCS, Firenze, Toscana, Italy
| | - David G Birch
- Ophthalmology, Retina Foundation of the Southwest, Dallas, Texas, USA
| | - Fred K Chen
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Australia
- Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Nieraj Jain
- Department of Ophthalmology, Emory School of Medicine, Atlanta, Georgia, USA
| | - Rachael C Heath Jeffery
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Australia
- Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Tina M Lamey
- Australian Inherited Retinal Diseases Registry, Medical Technology and Physics Department, Sir Charles Gairdner Hospital, Nedlands, Australia
| | - Debarshi Mustafi
- Department of Ophthalmology, Seattle Children's Hospital, University of Washington School of Medicine, Seattle, Washington, USA
| | | | | | - Mariam Torres Soto
- Department of Ophthalmology, Emory School of Medicine, Atlanta, Georgia, USA
| | - Kaylie Jones
- Ophthalmology, Retina Foundation of the Southwest, Dallas, Texas, USA
| | - Paul Yang
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Mark E Pennesi
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, USA
- Ophthalmology, Retina Foundation of the Southwest, Dallas, Texas, USA
| | - Lesley A Everett
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
19
|
Cruz-Criollo L, Dávila-Salazar W, Sarapura-Castro E, Rivera-Valdivia A, Bazalar-Montoya J, Bluske K, Taylor J, Thorpe E, Kesari A, Taft RJ, Cornejo-Olivas M. Delayed diagnosis of ataxia with oculomotor apraxia type 2 in a Peruvian patient, a case report. Clin Neurol Neurosurg 2025; 251:108823. [PMID: 40068357 DOI: 10.1016/j.clineuro.2025.108823] [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: 01/17/2025] [Revised: 03/01/2025] [Accepted: 03/01/2025] [Indexed: 03/30/2025]
Abstract
INTRODUCTION Ataxia with oculomotor apraxia type 2 (AOA2) is a rare autosomal recessive cerebellar ataxia characterized by progressive cerebellar ataxia, sensorimotor peripheral neuropathy, and occasional oculomotor apraxia. CASE REPORT A 50-year-old male with a history of orthopedic shoe use since childhood presented with slowly progressive ataxia and neuropathy. Laboratory tests showed elevated serum alpha-fetoprotein levels and increased total cholesterol. Clinical whole genome sequencing identified a c.4853C > G (p.Ser1618Ter) homozygous pathogenic variant in SETX. CONCLUSION The case highlights the challenges identifying rare disorders like AOA2 due to limited access to genetic testing and socioeconomic and healthcare barriers.
Collapse
Affiliation(s)
- Leonardo Cruz-Criollo
- Neurogenetics Research Center, Instituto Nacional de Ciencias Neurológicas, Lima, Peru; Department of Neurology, University of Iowa Healthcare, Iowa City, IA, United States
| | | | - Elison Sarapura-Castro
- Neurogenetics Research Center, Instituto Nacional de Ciencias Neurológicas, Lima, Peru; Neurogenetics Working Group, Universidad Cientifica del Sur, Lima, Peru
| | - Andrea Rivera-Valdivia
- Neurogenetics Research Center, Instituto Nacional de Ciencias Neurológicas, Lima, Peru; Neurogenetics Working Group, Universidad Cientifica del Sur, Lima, Peru
| | - Jeny Bazalar-Montoya
- Neurogenetics Research Center, Instituto Nacional de Ciencias Neurológicas, Lima, Peru; Facultad de Medicina Humana, Universidad de Piura, Lima, Peru
| | | | | | - Erin Thorpe
- Illumina, Inc., San Diego, CA, United States; Genetic Alliance, Damascus, MD, United States
| | | | - Ryan J Taft
- Illumina, Inc., San Diego, CA, United States; Genetic Alliance, Damascus, MD, United States
| | - Mario Cornejo-Olivas
- Neurogenetics Research Center, Instituto Nacional de Ciencias Neurológicas, Lima, Peru; Neurogenetics Working Group, Universidad Cientifica del Sur, Lima, Peru.
| |
Collapse
|
20
|
Andrade Azevedo de Vasconcelos A, Kyun Oh J, Guan B, Torres VLL, Lynch Gaete MI, Lima de Carvalho JR. A novel frameshift variant in the GJA1 gene is associated with recessive oculodentodigital dysplasia. Ophthalmic Genet 2025; 46:202-206. [PMID: 39833124 DOI: 10.1080/13816810.2024.2447499] [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: 02/16/2024] [Revised: 09/13/2024] [Accepted: 12/20/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND Oculodentodigital dysplasia (ODDD) is a rare syndrome that causes a constellation of facial, ophthalmic, dental, and limb abnormalities. Variants in the gap junction alpha-1 (GJA1) gene have been described in patients with ODDD. Hereby we present the ocular manifestations in a patient with recessive ODDD due to a novel homozygous frameshift variant in GJA1. MATERIAL AND METHODS Detailed ophthalmic manifestation and clinical features of disease were documented through external color photography and ultrasound biomicroscopy (UBM). Genetic testing was performed through a congenital heart disease panel. RESULTS A six-year-old girl was referred for ophthalmic evaluation in the setting of numerous syndromic features compatible with ODDD. Clinical features included nasal thinning, alar hypoplasia, hypotrichosis, microdontia and enamel hypoplasia. Ocular manifestations included microcornea, microphthalmia, posterior synechiae, cataract, and persistent hyperplastic primary vitreous. Genetic testing revealed a novel homozygous variant in the GJA1 gene, c.565del p.(Arg189Glufs *35). This variant disrupts the fourth helical transmembrane domain of the protein as well as its C-terminal cytoplasmic tail. CONCLUSION Here we describe the clinical and ocular manifestations of a Brazilian patient with ODDD, report a novel frameshift homozygous variant in GJA1, and contribute to the ongoing expansion of scientific knowledge regarding ODDD.
Collapse
Affiliation(s)
| | - Jin Kyun Oh
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, New York, USA
| | - Bin Guan
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Virginia Laura Lucas Torres
- Department of Ophthalmology, Hospital das Clínicas - Empresa Brasileira de Serviços Hospitalares, Federal University of Pernambuco, Recife, Brazil
| | - Maria Isabel Lynch Gaete
- Department of Ophthalmology, Hospital das Clínicas - Empresa Brasileira de Serviços Hospitalares, Federal University of Pernambuco, Recife, Brazil
| | - Jose Ronaldo Lima de Carvalho
- Department of Ophthalmology, Hospital das Clínicas - Empresa Brasileira de Serviços Hospitalares, Federal University of Pernambuco, Recife, Brazil
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
21
|
Vanoye CG, Desai RR, John JD, Hoffman SC, Fink N, Zhang Y, Venkatesh OG, Roe J, Adusumilli S, Jairam NP, Sanders CR, Gordon AS, George AL. Functional profiling of KCNE1 variants informs population carrier frequency of Jervell and Lange-Nielsen syndrome type 2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.28.646046. [PMID: 40236191 PMCID: PMC11996308 DOI: 10.1101/2025.03.28.646046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Congenital long-QT syndrome (LQTS) is most often associated with pathogenic variants in KCNQ1 encoding the pore-forming voltage-gated potassium channel subunit of the slow delayed rectifier current ( I Ks ). Generation of I Ks requires assembly of KCNQ1 with an auxiliary subunit encoded by KCNE1 , which is also associated with LQTS but causality of autosomal dominant disease is disputed. By contrast, KCNE1 is an accepted cause of recessive type 2 Jervell and Lange-Nielson syndrome (JLN2). The functional consequences of most KCNE1 variants have not been determined and the population prevalence of JLN2 is unknown. Methods : We determined the functional properties of 95 KCNE1 variants co-expressed with KCNQ1 in heterologous cells using high-throughput voltage-clamp recording. Experiments were conducted with each KCNE1 variant expressed in the homozygous state and then a subset was studied in the heterozygous state. The carrier frequency of JLN2 was estimated by considering the population prevalence of dysfunctional variants. Results : There is substantial overlap between disease-associated and population KCNE1 variants. When examined in the homozygous state, 68 KCNE1 variants exhibited significant differences in at least one functional property compared to WT KCNE1, whereas 27 variants did not significantly affect function. Most dysfunctional variants exhibited loss-of-function properties. We observed no evidence of dominant-negative effects. Most variants were scored as variants of uncertain significance (VUS) and inclusion of functional data resulted in revised classifications for only 14 variants. The population carrier frequency of JLN2 was calculated as 1 in 1034. Peak current density and activation voltage-dependence but no other biophysical properties were correlated with findings from a mutational scan of KCNE1. Conclusions : Among 95 disease-associated or population KCNE1 variants, many exhibit abnormal functional properties but there was no evidence of dominant-negative behaviors. Using functional data, we inferred a population carrier frequency for recessive JLN2. This work helps clarify the pathogenicity of KCNE1 variants.
Collapse
|
22
|
Bengl D, Koparir A, Prastyo WE, Remmele C, Dittrich M, Flandin S, Shehata-Dieler W, Grimm C, Haaf T, Hofrichter MAH. Whole-genome sequencing, as a powerful diagnostic tool in hearing loss, reveals novel variants in PTPRQ missed by whole-exome sequencing. BMC Med Genomics 2025; 18:59. [PMID: 40165225 PMCID: PMC11956499 DOI: 10.1186/s12920-025-02122-7] [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] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 03/10/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND/OBJECTIVES Hearing loss (HL) is one of the most common congenital disorders, affecting 1-2 in 1,000 newborns. Modern genetic diagnostics using large gene panels and/or whole exome analysis (WES) can identify disease-causing mutations in 25-50 % of patients, with higher solve rates in individuals with earlier onset. RESULTS Here, we used whole-genome sequencing (WGS) to reanalyze 14 index patients/families who remained without genetic diagnosis by WES. We were able to identify the genetic cause of HL in 6 families ( ∼ 43 %). Two families were diagnosed with DFNB84A caused by compound heterozygous recessive mutations in PTPRQ. Three of the four underlying variants, including a structural variant, a deep intronic variant, and a splice variant, escaped detection by WES. Minigene assays confirmed the pathogenicity of the intronic and the splice variants. In addition, we used protein 3D structure prediction and rigid ligand docking to study the pathogenicity of variants that escape nonsense-mediated decay. CONCLUSION In our study, we present four novel variants in PTPRQ, three of which were detected only by WGS. To our knowledge, we report here the first pathogenic deep intronic PTPRQ variant causing HL. Our results suggest that the mutational spectrum of PTPRQ is not well covered by standard WES and that PTPRQ-associated hearing loss may be more frequent than previously thought. WGS provides an additional layer of information in the diagnostics of HL.
Collapse
Affiliation(s)
- Daniel Bengl
- Institute of Human Genetics, Julius Maximilians University, Am Hubland, Würzburg, 97074, Bavaria, Germany
| | - Asuman Koparir
- Institute of Human Genetics, Julius Maximilians University, Am Hubland, Würzburg, 97074, Bavaria, Germany.
| | - Wahyu Eka Prastyo
- Institute of Human Genetics, Julius Maximilians University, Am Hubland, Würzburg, 97074, Bavaria, Germany
| | - Christian Remmele
- Institute of Human Genetics, Julius Maximilians University, Am Hubland, Würzburg, 97074, Bavaria, Germany
- Center for Rare Diseases, University Clinics, Josef-Schneider-Straße 2, Würzburg, 97080, Bavaria, Germany
- Bavarian Genomes Network for Rare Diseases, Technical University of Munich, Trogerstraße 32, Munich, 81675, Bavaria, Germany
| | - Marcus Dittrich
- Institute of Human Genetics, Julius Maximilians University, Am Hubland, Würzburg, 97074, Bavaria, Germany
- Department of Bioinformatics, Julius Maximilians University, Am Hubland, Würzburg, 97074, Bavaria, Germany
| | - Sophie Flandin
- Department of Otorhinolaryngology, Comprehensive Hearing Center, Würzburg University Hospital, Josef-Schneider-Straße 11, Würzburg, 97080, Bavaria, Germany
| | - Waafa Shehata-Dieler
- Department of Otorhinolaryngology, Comprehensive Hearing Center, Würzburg University Hospital, Josef-Schneider-Straße 11, Würzburg, 97080, Bavaria, Germany
| | - Clemens Grimm
- Chair of Biochemistry, Theodor-Boveri-Institute at the Biocentre University of Würzburg, Am Hubland, Würzburg, 97074, Bavaria, Germany
| | - Thomas Haaf
- Institute of Human Genetics, Julius Maximilians University, Am Hubland, Würzburg, 97074, Bavaria, Germany
| | - Michaela A H Hofrichter
- Institute of Human Genetics, Julius Maximilians University, Am Hubland, Würzburg, 97074, Bavaria, Germany
| |
Collapse
|
23
|
Chen J, Huang J, Xu H. Association Analysis of ENPP1 Tissue Expression, Polymorphism, and Growth Traits in Xiangsu Pigs. Genes (Basel) 2025; 16:395. [PMID: 40282354 PMCID: PMC12027002 DOI: 10.3390/genes16040395] [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: 03/05/2025] [Revised: 03/25/2025] [Accepted: 03/28/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Pigs are vital agricultural animals, with growth traits serving as key indicators of their quality. Methods: In this study, we examined the mRNA expression of ENPP1 as a candidate gene in heart, liver, spleen, lungs, and kidneys at 3 days and 6 months of age by real-time polymerase chain reaction method and single-nucleotide polymorphism (SNP) loci in 165 Xiangsu pigs by Sanger sequencing. Results: The expression of ENPP1 in different tissues of Xiangsu pigs at different stages was significantly different, and it had high conservation in different species.. Sequence alignment with reference data identified five SNP sites: g.64275T→C and g.64429G→A in intron 19, g.64850T→C and g.64911G→A in intron 20, and g.64527T→C in exon 20. Association analysis revealed that g.64275T→C, g.64429G→A, and g.64527T→C significantly influence the growth performance of Xiangsu pigs (p < 0.05). Conclusions: These findings suggest that ENPP1 polymorphisms are closely associated with growth traits in Xiangsu pigs and may provide valuable insights for molecular breeding of this breed.
Collapse
Affiliation(s)
- Jiaqi Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China; (J.C.); (J.H.)
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Jiajin Huang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China; (J.C.); (J.H.)
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Houqiang Xu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China; (J.C.); (J.H.)
- College of Animal Science, Guizhou University, Guiyang 550025, China
| |
Collapse
|
24
|
Greco A, Martínez-Barrios E, Cruzalegui J, Cesar S, Chipa F, Díez-Escuté N, Cerralbo P, Zschaeck I, Loredo P, Sarquella-Brugada G, Campuzano O. Brugada Syndrome and GPD1L: Definite Genotype-Phenotype Association? CARDIOGENETICS 2025; 15:9. [DOI: 10.3390/cardiogenetics15010009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025] Open
Abstract
The GPD1L gene encodes a small cytoplasmic protein that is involved in the regulation of sodium currents. Alterations in this gene have been associated with Brugada syndrome. This rare arrhythmogenic syndrome is characterized by a typical electrocardiographic pattern, incomplete penetrance, variable expressivity, and risk of sudden cardiac death. To date, few families with a clinical diagnosis of Brugada syndrome caused by a rare alteration in the GPD1L gene have been reported worldwide. The increase in data focused on genetic variants allows us to improve the interpretation of their role in Brugada syndrome. In our study, we have compiled the GPD1L variants reported so far in patients with a definitive clinical diagnosis or suspected Brugada syndrome. We performed an exhaustive update and interpretation of each variant following the guidelines of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Our results showed that none of the variants described to date can be classified as truly harmful in Brugada syndrome. Despite this fact, more clinical and genetic data are needed to definitively rule out the GPD1L gene as a cause of Brugada syndrome. In summary, to date, there is insufficient evidence to conclude a definitive association between GPD1L and Brugada syndrome.
Collapse
Affiliation(s)
- Andrea Greco
- Arrhythmias, Inherited Cardiac Diseases and Sudden Death Unit, Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
- Arrítmies Pediàtriques, Cardiologia Genètica i Mort Sobtada, Malalties Cardiovasculars en el Desenvolupament, Institut de Recerca Sant Joan de Déu (IRSJD), 08950 Esplugues de Llobregat, Spain
| | - Estefanía Martínez-Barrios
- Arrhythmias, Inherited Cardiac Diseases and Sudden Death Unit, Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
- Arrítmies Pediàtriques, Cardiologia Genètica i Mort Sobtada, Malalties Cardiovasculars en el Desenvolupament, Institut de Recerca Sant Joan de Déu (IRSJD), 08950 Esplugues de Llobregat, Spain
- Medical Science Department, School of Medicine, Universitat de Girona, 17003 Girona, Spain
| | - José Cruzalegui
- Arrhythmias, Inherited Cardiac Diseases and Sudden Death Unit, Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
- Arrítmies Pediàtriques, Cardiologia Genètica i Mort Sobtada, Malalties Cardiovasculars en el Desenvolupament, Institut de Recerca Sant Joan de Déu (IRSJD), 08950 Esplugues de Llobregat, Spain
| | - Sergi Cesar
- Arrhythmias, Inherited Cardiac Diseases and Sudden Death Unit, Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
- Arrítmies Pediàtriques, Cardiologia Genètica i Mort Sobtada, Malalties Cardiovasculars en el Desenvolupament, Institut de Recerca Sant Joan de Déu (IRSJD), 08950 Esplugues de Llobregat, Spain
| | - Fredy Chipa
- Arrhythmias, Inherited Cardiac Diseases and Sudden Death Unit, Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
- Arrítmies Pediàtriques, Cardiologia Genètica i Mort Sobtada, Malalties Cardiovasculars en el Desenvolupament, Institut de Recerca Sant Joan de Déu (IRSJD), 08950 Esplugues de Llobregat, Spain
| | - Nuria Díez-Escuté
- Arrhythmias, Inherited Cardiac Diseases and Sudden Death Unit, Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
- Arrítmies Pediàtriques, Cardiologia Genètica i Mort Sobtada, Malalties Cardiovasculars en el Desenvolupament, Institut de Recerca Sant Joan de Déu (IRSJD), 08950 Esplugues de Llobregat, Spain
| | - Patricia Cerralbo
- Arrhythmias, Inherited Cardiac Diseases and Sudden Death Unit, Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
- Arrítmies Pediàtriques, Cardiologia Genètica i Mort Sobtada, Malalties Cardiovasculars en el Desenvolupament, Institut de Recerca Sant Joan de Déu (IRSJD), 08950 Esplugues de Llobregat, Spain
| | - Irene Zschaeck
- Arrhythmias, Inherited Cardiac Diseases and Sudden Death Unit, Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
- Arrítmies Pediàtriques, Cardiologia Genètica i Mort Sobtada, Malalties Cardiovasculars en el Desenvolupament, Institut de Recerca Sant Joan de Déu (IRSJD), 08950 Esplugues de Llobregat, Spain
| | - Paula Loredo
- Arrhythmias, Inherited Cardiac Diseases and Sudden Death Unit, Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
- Arrítmies Pediàtriques, Cardiologia Genètica i Mort Sobtada, Malalties Cardiovasculars en el Desenvolupament, Institut de Recerca Sant Joan de Déu (IRSJD), 08950 Esplugues de Llobregat, Spain
- Universidade Luterana do Brasil (ULBRA), Medical Department, Clínica Médica, Canoas CEP 92425-900, RS, Brazil
| | - Georgia Sarquella-Brugada
- Arrhythmias, Inherited Cardiac Diseases and Sudden Death Unit, Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
- Arrítmies Pediàtriques, Cardiologia Genètica i Mort Sobtada, Malalties Cardiovasculars en el Desenvolupament, Institut de Recerca Sant Joan de Déu (IRSJD), 08950 Esplugues de Llobregat, Spain
- Medical Science Department, School of Medicine, Universitat de Girona, 17003 Girona, Spain
- Pediatrics Department, School of Medicine, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Oscar Campuzano
- Medical Science Department, School of Medicine, Universitat de Girona, 17003 Girona, Spain
- Centro Investigación Biomédica en Red-Cardiovascular (CIBERCV), 28029 Madrid, Spain
- Institut d’Investigació Biomèdiques de Girona (IDIBGI-CERCA), 17190 Salt, Spain
| |
Collapse
|
25
|
Laich Y, Georgiou M, Fujinami K, Daich Varela M, Fujinami-Yokokawa Y, Hashem SA, de Guimaraes TAC, Mahroo OA, Webster AR, Michaelides M. Best Vitelliform Macular Dystrophy Natural History Study Report 2: Fundus Autofluorescence and OCT. Ophthalmol Retina 2025:S2468-6530(25)00103-4. [PMID: 40086732 DOI: 10.1016/j.oret.2025.03.004] [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: 01/04/2025] [Revised: 03/01/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
PURPOSE To analyze the retinal imaging findings and natural history of Best vitelliform macular dystrophy (BVMD). DESIGN Single-center retrospective, consecutive, observational study. PARTICIPANTS Patients with a clinical diagnosis of BVMD, from pedigrees with a likely disease-causing monoallelic variant in BEST1. METHODS Data were extracted from electronic and physical case notes. Retinal imaging with OCT and fundus autofluorescence (FAF) was analyzed cross-sectionally and longitudinally. MAIN OUTCOME MEASURES Qualitative and quantitative OCT and FAF analysis. RESULTS Two hundred twenty-two patients (127 men) from 141 families were included. Mean central retinal thickness on OCT at baseline was 337.2 μm for the right eye and 341.1 μm for the left eye, with a mean annual thickness loss of 5.7 and 5.2 μm, respectively. The presence of the OCT features: previtelliform lesion, solid vitelliform lesion, vitelliform lesion with subretinal fluid, and focal choroidal excavation were associated with a better mean visual acuity (VA), whereas the presence of intraretinal fluid and atrophy/fibrosis were correlated with a worse mean VA. Fundus autofluorescence showed an area of hyperautofluorescence at the posterior pole in 138 eyes (34.7%), a circumscribed area of hyperautofluorescence superior or superotemporal of the optic nerve head in 53 eyes (13.3%), fibrotic changes in 48 eyes (12.1%), and atrophy in 41 eyes (10.3%). CONCLUSIONS Best vitelliform macular dystrophy shows a wide spectrum of phenotypes on OCT and FAF imaging. The slow and variable disease course may pose a challenge in identification of early end points for therapeutic trials aimed at altering kinetics of degeneration. FINANCIAL DISCLOSURE(S) Proprietary or commercial disclosures may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Yannik Laich
- Moorfields Eye Hospital, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom; Eye Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Michalis Georgiou
- Moorfields Eye Hospital, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom; Jones Eye Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Kaoru Fujinami
- Moorfields Eye Hospital, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom; Laboratory of Visual Physiology, Division of Vision Research, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | - Malena Daich Varela
- Moorfields Eye Hospital, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Yu Fujinami-Yokokawa
- UCL Institute of Ophthalmology, University College London, London, United Kingdom; Laboratory of Visual Physiology, Division of Vision Research, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Tokyo, Japan; Department of Health Policy and Management, Keio University School of Medicine, Tokyo, Japan
| | - Shaima Awadh Hashem
- Moorfields Eye Hospital, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Thales A C de Guimaraes
- Moorfields Eye Hospital, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Omar A Mahroo
- Moorfields Eye Hospital, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Andrew R Webster
- Moorfields Eye Hospital, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Michel Michaelides
- Moorfields Eye Hospital, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom.
| |
Collapse
|
26
|
Lokchine A, Bergougnoux A, Servant N, Akloul L, Launay E, Mary L, Cluzeau L, Philippe M, Domin-Bernhard M, Duros S, Odent S, Tucker E, Paris F, Belaud-Rotureau MA, Jaillard S. Identification and Characterization of Novel FSHR Copy Number Variations Causing Premature Ovarian Insufficiency. Am J Med Genet A 2025; 197:e63924. [PMID: 39497500 DOI: 10.1002/ajmg.a.63924] [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: 07/12/2024] [Revised: 10/07/2024] [Accepted: 10/20/2024] [Indexed: 02/13/2025]
Abstract
Follicle stimulating hormone (FSH) is a key pituitary gonadotropic hormone implicated in human fertility and is crucial for folliculogenesis and recruitment of new antral follicles. Variations in its receptor, FSHR, can lead to diverse reproductive phenotypes including ovarian hyperstimulation syndrome (OHSS) and premature ovarian insufficiency (POI). This study reports a novel case of FSHR-related ovarian insufficiency in a patient with primary amenorrhea, subnormal AMH levels, and delayed puberty. Genetic exploration revealed two compound heterozygous intragenic deletions of FSHR. Specifically, the patient inherited a maternally derived deletion spanning exons 5-10 and a paternally derived deletion involving exons 3-6. Through chromosomal microarray analysis (CMA), exome sequencing, long-range PCR, and Sanger sequencing, we characterized the breakpoints and confirmed the compound heterozygous deletions. The findings reveal a complete loss of function of both FSHR alleles, contributing to the patient's POI phenotype. This case emphasizes the complexity of genotype-phenotype correlations in FSHR-related disorders and the role of CNVs in POI phenotypes. Although these events are rare, our results advocate for the inclusion of CNV detection in the diagnostic workup of POI to ensure accurate diagnosis and better patient management.
Collapse
Affiliation(s)
- Anna Lokchine
- Univ Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail)-UMR_S1085, Rennes, France
- CHU Rennes, Service de Cytogénétique et Biologie Cellulaire, Rennes, France
| | - Anne Bergougnoux
- Service de Génétique Moléculaire et de Cytogénomique, CHU de Montpellier, Montpellier, France
- PhyMedExp, Université de Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
| | - Nadège Servant
- Service de Génétique Moléculaire et de Cytogénomique, CHU de Montpellier, Montpellier, France
| | - Linda Akloul
- CHU Rennes, Service de Génétique Clinique, CLAD Ouest, Rennes, France
| | - Erika Launay
- CHU Rennes, Service de Cytogénétique et Biologie Cellulaire, Rennes, France
| | - Laura Mary
- CHU Rennes, Service de Cytogénétique et Biologie Cellulaire, Rennes, France
| | - Laurence Cluzeau
- CHU Rennes, Service de Cytogénétique et Biologie Cellulaire, Rennes, France
| | - Mathieu Philippe
- CHU Rennes, Service de Cytogénétique et Biologie Cellulaire, Rennes, France
| | | | - Solène Duros
- CHU Rennes, Département de Gynécologie Obstétrique et Reproduction Humaine, Rennes, France
| | - Sylvie Odent
- CHU Rennes, Service de Génétique Clinique, CLAD Ouest, Rennes, France
- Institut de Génétique et Développement de Rennes, IGDR UMR 6290 CNRS, University of Rennes, Rennes, France
| | - Elena Tucker
- Reproductive Development, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Françoise Paris
- Service de Génétique Moléculaire et de Cytogénomique, CHU de Montpellier, Montpellier, France
- Unité d'Endocrinologie et Gynécologie Pédiatriques, Service de Pédiatrie I, Hôpital A. de Villeneuve, Montpellier, France
- Développement Embryonnaire, Fertilité et Environnement, Université de Montpellier, INSERM, Montpellier, France
| | - Marc-Antoine Belaud-Rotureau
- Univ Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail)-UMR_S1085, Rennes, France
- CHU Rennes, Service de Cytogénétique et Biologie Cellulaire, Rennes, France
| | - Sylvie Jaillard
- Univ Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail)-UMR_S1085, Rennes, France
- CHU Rennes, Service de Cytogénétique et Biologie Cellulaire, Rennes, France
| |
Collapse
|
27
|
Leon-Quintero FZ, Bowling KM, Dickson A, Corliss MM, Schroeder MC, Neidich JA, Heusel JW, Krysiak K, Polonis K, Parikh BA, Cao Y. Modified Rules for Classification of Variants Associated With Disorders of Somatic Mosaicism. Clin Genet 2025; 107:261-270. [PMID: 39434542 DOI: 10.1111/cge.14636] [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: 08/04/2024] [Revised: 10/01/2024] [Accepted: 10/08/2024] [Indexed: 10/23/2024]
Abstract
Disorders of somatic mosaicism (DoSMs) are rare genetic disorders arising from postzygotic alteration leading to segmental/nonsegmental disease. Current professional guidelines for standardized variant interpretation focus on germline and cancer variants, making them suboptimal for DoSM variant interpretation. The Brain Malformations Variant Curation Expert Panel (BMVCEP) modified existing guidelines to account for brain-specific disorders of somatic mosaicism, but applicability to other DoSM presentations is limited. At Washington University in St. Louis School of Medicine, we have adopted the BMVCEP interpretation framework but suggested alterations that make it more suitable for generalized DoSM variant classification. These modifications include (1) expanding applicability beyond genes associated with brain malformations, (2) introduction of a criterion to interpret truncating variants at the C-terminus of gain of function genes, (3) establishment of a variant allele fraction (VAF) cutoff for applying de novo criteria, and (4) demonstration that in silico prediction tools are relevant to interpretation of gain of function missense variants. Furthermore, modifications to BMVCEP guidelines reduce the number of variants classified as uncertain. The variant classification considerations that we propose have the potential to improve the accuracy of somatic variant classification, better inform clinical care, and may benefit clinical laboratories also conducting DoSM testing.
Collapse
Affiliation(s)
- Fernando Zazueta Leon-Quintero
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
- Pathology Department, Hospital San Javier, Guadalajara, Mexico
| | - Kevin M Bowling
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Alexa Dickson
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Meagan M Corliss
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Molly C Schroeder
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Julie A Neidich
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Jonathan W Heusel
- Department of Pathology, Renaissance School of Medicine, State University of New York at Stony Brook, Stony Brook, New York, USA
| | - Kilannin Krysiak
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Katarzyna Polonis
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Bijal A Parikh
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Yang Cao
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
28
|
Ćomić J, Tilch E, Riedhammer KM, Brugger M, Brunet T, Eyring K, Vill K, Redler S, Tasic V, Schmiedeke E, Schäfer FM, Abazi-Emini N, Jenetzky E, Schwarzer N, Widenmann A, Lacher M, Zech M, Grasshoff-Derr S, Geßner M, Kabs C, Seitz B, Heydweiller AC, Muensterer O, Lange-Sperandio B, Rolle U, Schumacher J, Braunisch MC, Berutti R, Reutter H, Hoefele J. Trio Exome Sequencing in VACTERL Association. Kidney Int Rep 2025; 10:877-891. [PMID: 40225364 PMCID: PMC11993224 DOI: 10.1016/j.ekir.2024.12.006] [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: 06/12/2024] [Revised: 11/12/2024] [Accepted: 12/03/2024] [Indexed: 02/10/2025] Open
Abstract
INTRODUCTION Currently, there is only limited data on monogenic causes of vertebral defects, anorectal malformations, cardiac defects, esophageal atresia or tracheoesophageal fistula, renal malformations, and limb defects (VACTERL) association. The aim of this study was to extend the spectrum of disease-causing variants in known genes, to determine the diagnostic yield of monogenic causes, and to identify candidate genes and rare variants by applying comprehensive genetic testing or rare variant burden. METHODS The total cohort comprised 101 affected individuals and their parents. Trio exome sequencing was only performed in 96 individuals and their parents because of DNA quality reasons and case-control gene and pathway burden tests were calculated and evaluated by quantile-quantile plots, principal component analysis plots and family-based association test (FBAT). RESULTS In 5 of 96 individuals, disease-causing variants in known genes or loci were identified to be associated with the following 4 disorders: Kabuki syndrome, Sotos syndrome, MELAS syndrome, and deletion syndrome encompassing TWIST1. In 91 individuals, no disease-causing variants were found. FBAT showed 14 significant variants, 2 significant genes (LOC645752 and ZNF417), and 8 significant pathways. CONCLUSION This study shows that most individuals with VACTERL association do not have known discrete genetic syndromes, implying that pathomechanisms or variants not identifiable by exome sequencing may exist requiring further investigation.
Collapse
Affiliation(s)
- Jasmina Ćomić
- Institute of Human Genetics, Klinikum rechts der Isar, Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
- Department of Nephrology, Klinikum rechts der Isar, Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
| | - Erik Tilch
- Institute of Human Genetics, Klinikum rechts der Isar, Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
| | - Korbinian M. Riedhammer
- Institute of Human Genetics, Klinikum rechts der Isar, Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
- Department of Nephrology, Klinikum rechts der Isar, Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
- Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Melanie Brugger
- Institute of Human Genetics, Klinikum rechts der Isar, Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
| | - Theresa Brunet
- Institute of Human Genetics, Klinikum rechts der Isar, Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
| | - Katharina Eyring
- Institute of Human Genetics, Klinikum rechts der Isar, Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
| | - Katharina Vill
- Institute of Human Genetics, Klinikum rechts der Isar, Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
- Department of Pediatric Neurology and Developmental Medicine and LMU Center for Children with Medical Complexity, Dr. von Hauner Children's Hospital, Ludwig-Maximilians University, Munich, Germany
| | - Silke Redler
- Institute of Human Genetics, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine University, Düsseldorf, Germany
| | - Velibor Tasic
- University Children’s Hospital, Medical Faculty of Skopje, North Macedonia
| | - Eberhard Schmiedeke
- Clinic for Paediatric Surgery and Paediatric Urology, Klinikum Bremen-Mitte, Bremen, Germany
| | - Frank-Mattias Schäfer
- Department of Pediatric Surgery and Urology, Cnopf'sche Kinderklinik, Nürnberg, Germany
| | - Nora Abazi-Emini
- University Children’s Hospital, Medical Faculty of Skopje, North Macedonia
| | - Ekkehart Jenetzky
- Institute of Integrative Medicine, Witten/Herdecke University, Herdecke, Germany
- Department of Pediatric and Adolescent Psychiatry and Psychotherapy, University Medical Centre, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Nicole Schwarzer
- SoMA, The German Patient Support Organization for Anorectal Malformations and Hirschsprung Disease, Munich, Germany
| | - Anke Widenmann
- Patient Organisation for Esophageal Diseases KEKS e.V., Stuttgart, Germany
| | - Martin Lacher
- Department of Pediatric Surgery, University of Leipzig, Leipzig, Germany
| | - Michael Zech
- Institute of Human Genetics, Klinikum rechts der Isar, Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
- Institute of Neurogenomics, Helmholtz Munich, Neuherberg, Germany
- Institute of Advanced Study, Technical University of Munich, Garching, Germany
| | - Sabine Grasshoff-Derr
- Pediatric Surgery Unit, Buergerhospital and Clementine Kinderhospital, Frankfurt, Germany
| | - Michaela Geßner
- KfH-Board of Trustees for Dialysis and Kidney Transplantation (KfH-Kuratorium für Dialyse und Nierentransplantatione.V.), Munich, Germany
| | - Carmen Kabs
- Department of Paediatrics Surgery, Muenchen KlinikgGmbH, Munich Clinic Schwabing, Munich, Germany
| | - Barbara Seitz
- KfH-Board of Trustees for Dialysis and Kidney Transplantation (KfH-Kuratorium für Dialyse und Nierentransplantatione.V.), Munich, Germany
| | - Andreas C. Heydweiller
- Department of General, Visceral, Vascular and Thoracic Surgery, Unit of Pediatric Surgery, University Hospital Bonn, Bonn, Germany
| | - Oliver Muensterer
- Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, Ludwig-Maximilians University, Munich, Germany
| | - Bärbel Lange-Sperandio
- Division of Pediatric Nephrology, Department of Pediatrics, Dr. v. Hauner Children's Hospital, Ludwig-Maximilians University, Munich, Germany
| | - Udo Rolle
- Department of Pediatric Surgery and Pediatric Urology, University Hospital Frankfurt/M., Frankfurt am Main, Germany
| | - Johannes Schumacher
- Institute of Human Genetics, University Hospital of Marburg, Marburg, Germany
| | - Matthias C. Braunisch
- Institute of Human Genetics, Klinikum rechts der Isar, Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
- Department of Nephrology, Klinikum rechts der Isar, Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
| | - Riccardo Berutti
- Institute of Human Genetics, Klinikum rechts der Isar, Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
| | - Heiko Reutter
- Department of Neonatology and Pediatric Intensive Care, University Erlangen-Nürnberg, Erlangen, Germany
| | - Julia Hoefele
- Institute of Human Genetics, Klinikum rechts der Isar, Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
- Institute of Human Genetics, University Hospital, Ludwig-Maximilians University, Munich, Germany
| |
Collapse
|
29
|
Hashem SA, Georgiou M, Wright G, Fujinami-Yokokawa Y, Laich Y, Daich Varela M, de Guimaraes TAC, Mahroo OA, Webster AR, Fujinami K, Michaelides M. PDE6A-Associated Retinitis Pigmentosa, Clinical Characteristics, Genetics, and Natural History. Ophthalmol Retina 2025; 9:278-287. [PMID: 39218074 DOI: 10.1016/j.oret.2024.08.018] [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: 05/05/2024] [Revised: 08/09/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE To analyze the genetics, clinical characteristics, and natural history of PDE6A-associated retinitis pigmentosa. DESIGN Retrospective, longitudinal, observational cohort study. PARTICIPANTS Patients with molecularly confirmed PDE6A-associated retinal dystrophy in a single tertiary referral center. METHODS Review of medical records and retinal imaging, including fundus autofluorescence (FAF) imaging and spectral-domain OCT. Genetic results were reviewed, and the detected variants were assessed. MAIN OUTCOME MEASURES Molecular genetic testing, clinical findings including best-corrected visual acuity (BCVA), qualitative and quantitative retinal imaging analysis. RESULTS Sixteen patients (32 eyes) were identified and evaluated longitudinally. Genetic analysis identified 14 variants in the PDE6A gene, including 8 novel variants. The mean age (± standard deviation, range) was 34.8 years (± 17.4, 12-76) at baseline, with a mean follow-up time of 4.8 years. Best-corrected visual acuity was 0.45 ± 0.45 logarithm of the minimum angle of resolution (logMAR) (range 0.0-1.6) at baseline and 0.65 ± 0.7 logMAR (range 0.0-2.3) at the last visit. Best-corrected visual acuity was similar among eyes in 88% of patients. A hyperautofluorescent ring was observed on FAF in 50% and 43.8% of the eyes at baseline and follow-up visit, respectively, with a mean area of 9.7 ± 4.5 mm2 at baseline and mean of 8.6 ± 4.8 mm2 at the follow-up visit. Mean horizontal ellipsoid zone width (EZW) at baseline was 1765 ± 1093 μm, which decreased to 1580 ± 1077 μm at follow-up. Eighteen eyes exhibited cystoid macular edema at baseline (56%), and 17 eyes (53%) at follow-up. There were statistically significant changes during the follow-up period in terms of BCVA, hyperautofluorescent ring area, and the EZW. CONCLUSIONS This study highlights the natural history of PDE6A-retinopathy. Most patients in this cohort had mild BCVA loss, and slowly progressive disease, based on FAF and OCT measurements. FINANCIAL DISCLOSURE(S) Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Shaima Awadh Hashem
- Moorfields Eye Hospital, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Michalis Georgiou
- Moorfields Eye Hospital, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom; Jones Eye Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Genevieve Wright
- Moorfields Eye Hospital, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Yu Fujinami-Yokokawa
- UCL Institute of Ophthalmology, University College London, London, United Kingdom; Laboratory of Visual Physiology, Division of Vision Research, National Institute of Sensory Organs, NHO Tokyo Medical Center, Tokyo, Japan; Department of Health Policy and Management, Keio University School of Medicine, Tokyo, Japan
| | - Yannik Laich
- Moorfields Eye Hospital, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom; Eye Center, Faculty of Medicine, University Freiburg, Freiburg im Breisgau, Germany
| | - Malena Daich Varela
- Moorfields Eye Hospital, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Thales A C de Guimaraes
- Moorfields Eye Hospital, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Omar A Mahroo
- Moorfields Eye Hospital, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom; Section of Ophthalmology, King's College London, London, United Kingdom; Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom; Eye Department, St Thomas' Hospital, London, United Kingdom; Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Andrew R Webster
- Moorfields Eye Hospital, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Kaoru Fujinami
- Moorfields Eye Hospital, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom; Laboratory of Visual Physiology, Division of Vision Research, National Institute of Sensory Organs, NHO Tokyo Medical Center, Tokyo, Japan
| | - Michel Michaelides
- Moorfields Eye Hospital, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom.
| |
Collapse
|
30
|
Chandler N, Holder‐Espinasse M, Mone F. The Challenges of Performing Exome Sequencing in Structurally Normal Fetuses. Prenat Diagn 2025; 45:294-298. [PMID: 39394633 PMCID: PMC11893515 DOI: 10.1002/pd.6687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 09/23/2024] [Accepted: 09/30/2024] [Indexed: 10/13/2024]
Affiliation(s)
- Natalie Chandler
- North Thames Genomic Laboratory HubGreat Ormond Street Hospital for Children NHS Foundation TrustLondonUK
| | | | - Fionnuala Mone
- Centre for Public HealthQueen's University BelfastBelfastUK
| |
Collapse
|
31
|
Yang J, Liu C, Geng Q, Chen L, Zhang L, Wu W. Two GNPTAB Variations Caused Mucolipidosis II Alpha/Beta in a Chinese Family. Fetal Pediatr Pathol 2025; 44:157-165. [PMID: 39957256 DOI: 10.1080/15513815.2025.2466057] [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: 07/09/2024] [Revised: 01/26/2025] [Accepted: 02/05/2025] [Indexed: 02/18/2025]
Abstract
Introduction: Mucolipidosis II alpha/beta (ML II) is an autosomal recessive disorder with craniofacial dysmorphism and bone deformities. The variants in GNPTAB are associated with ML II. Materials and Methods: A female pediatric patient presented with bone deformities, mental and motor developmental abnormalities and craniofacial dysmorphism. We performed clinical whole-exome sequencing (WES) and verified the variants via qPCR, gap-PCR and Sanger sequencing. Results: Clinical WES identified a point variant c.1090C > T (p.R364*) and a copy number variation (CNV) in GNPTAB. Compared with normal control, GNPTAB expression was reduced in blood of the proband. Using Gap-PCR and Sanger sequencing, we identified the break point of CNV (NC_000012.11:g.102136912_102142973del), and successfully performed prenatal diagnosis for the proband's mother. Conclusion: To our knowledge, this is the first report of this novel CNV associated with ML II. Our findings expand the genotypes related to ML II and contribute to the gene diagnosis of ML II.
Collapse
Affiliation(s)
- Jingxin Yang
- Medical Genetic Center, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong Province, China
| | - Chao Liu
- Medical Genetic Center, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong Province, China
| | - Qian Geng
- Medical Genetic Center, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong Province, China
| | - Liyuan Chen
- Medical Genetic Center, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong Province, China
| | - Lei Zhang
- Medical Genetic Center, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong Province, China
| | - Weiqing Wu
- Medical Genetic Center, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong Province, China
| |
Collapse
|
32
|
Benn P, Wang Y, Gray J, Dugan EK, Hajjar M, Prigmore B, Souter V, Wolf B. Evaluating reproductive carrier screening using biotinidase deficiency as a model: Variants identified, variant rates, and management. Genet Med 2025; 27:101345. [PMID: 39688110 DOI: 10.1016/j.gim.2024.101345] [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: 07/24/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024] Open
Abstract
PURPOSE To review biotinidase gene (BTD) variants identified in a large, diverse, reproductive carrier screening (RCS) cohort and outline management of heterozygotes with pathogenic or likely pathogenic (P/LP) variants. METHODS This retrospective observational study included samples tested from January 2020 to September 2022 in a 274-gene panel. The study involved females aged 18 to 55 years. Screening was performed using next-generation sequencing covering exons and 10 base-pair flanking introns. The heterozygote frequency was calculated for P/LP variants for the entire population and individual racial/ethnic groups. RESULTS Of the 91,637 women tested, 5625 (6.1%) had a P/LP variant in BTD. NM_000060.4:c.1330G>C p.(Asp444His) (referred to as D444H or D424H) alone, or in combination with another variant, accounted for 5193 (92.3%) of the positive tests. P/LP heterozygote rates differed between racial and ethnic groups. We ascertained 7 novel P/LP variants not previously recorded in databases. CONCLUSION The BTD P/LP variants identified through RCS were substantially compatible with those found through positive newborn screening. Therefore, RCS provides a potential for earlier diagnosis. We observed significant differences in P/LP heterozygote rates for biotinidase deficiency among different racial and ethnic groups. Most reported variants can be interpreted without requiring determination of serum biotinidase activity.
Collapse
Affiliation(s)
- Peter Benn
- University of Connecticut Health Center, Farmington, CT.
| | | | | | | | | | | | | | - Barry Wolf
- Division of Genetics, Birth Defects and Metabolism, Department of Pediatrics, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL; Emeritus, Departments of Medical Genetics and Pediatrics, Henry Ford Hospital, Detroit, MI
| |
Collapse
|
33
|
Vodnjov N, Cerar A, Maver A, Peterlin B, Writzl K. TTN:c.12478del in proximal I-band of titin represents a common molecular cause of dilated cardiomyopathy in Slovenian patients. Orphanet J Rare Dis 2025; 20:92. [PMID: 40022161 PMCID: PMC11871621 DOI: 10.1186/s13023-025-03613-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 02/16/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Titin truncating variants (TTNtv-s) are the most common genetic cause of dilated cardiomyopathy (DCM). Only rare TTNtv-s in the constitutively expressed exons of the A-band of the protein titin are associated with DCM according to the guidelines, however, studies in large cohorts of patients with DCM suggest that the region where TTNtv-s are associated with DCM is wider, extending at least into the I-band. The aim of this study was to describe the molecular pathology of TTNtv-s in Slovenian patients with cardiomyopathy and to clinically characterise the most recurrent TTNtv. RESULTS We collected all TTNtv-s identified in patients with cardiomyopathy using next-generation sequencing genetic testing between 2010 and July 2024, resulting in 42 unique variants identified in 54 patients. The TTN:c.12478del variant, affecting not the A-band but the proximal I-band, specifically the cardiac-specific N2Bus region, was found to be the most recurrent variant, present in seven (11.6%) probands with DCM. Genetic characterisation revealed a probable founder origin of the variant. Clinical characterisation of these probands revealed a phenotype consistent with DCM and severely reduced left ventricular ejection fraction in all probands. Three (43%) of the probands had atrial fibrillation and/or non-sustained ventricular tachycardia. Based on literature reports and evidence supporting the pathogenicity of the TTN:c.12478del variant affecting the proximal I-band, we classified all rare TTNtv-s in constitutively expressed exons of the I-band as (likely) pathogenic. Therefore, 33 (78.6%) TTNtv-s were classified as (likely) pathogenic (13 in the I-band, affecting 19 probands and 20 in the A-band affecting 25 probands), meaning that TTNtv-s were identified in 44 genotype-positive Slovenian probands with DCM, explaining 73.3% of the molecular pathology of DCM. CONCLUSION We report an almost threefold higher diagnostic yield of TTNtv-s in probands with DCM compared to previously reported findings in cohorts of patients with DCM from other populations. We also highlight the need for screening for rare TTNtv-s in the constitutively expressed exons of the I-band and for TTN:c.12478del in patients with DCM in this geographical region.
Collapse
Affiliation(s)
- Nina Vodnjov
- Clinical Institute of Genomic Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Andraž Cerar
- Department of Cardiology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Aleš Maver
- Clinical Institute of Genomic Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Borut Peterlin
- Clinical Institute of Genomic Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Karin Writzl
- Clinical Institute of Genomic Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia.
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.
- European Reference Network for Rare, Low Prevalence, Or Complex Diseases of the Heart (ERN GUARD-Heart), Amsterdam, The Netherlands.
| |
Collapse
|
34
|
Mercè F, Asla Q, Illana FJ, Victòria F, Javier HL, Marta S, Iglesias C, Webb SM, Aulinas A. A novel likely pathogenic germline variant in CDKN1B in a patient with MEN4 and medullary thyroid cancer. Fam Cancer 2025; 24:24. [PMID: 40009226 DOI: 10.1007/s10689-025-00449-9] [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: 08/28/2024] [Accepted: 02/15/2025] [Indexed: 02/27/2025]
Abstract
Multiple endocrine neoplasia type 4 (MEN4) is caused by a germline CDKN1B deleterious variant. CDKN1B encodes p27Kip1, a cyclin-dependent kinase inhibitor that acts as tumor-suppressor. Clinical presentation of MEN4 is similar to multiple endocrine neoplasia type 1 (MEN1) but the diagnosis of MEN4 can only be established once a germline CDKN1B pathogenic variant has been confirmed. We describe a unique case presenting with two -rare endocrine conditions. A 59-year-old female patient was diagnosed with medullary thyroid cancer (MTC) without evidence of a germline pathogenic variant in the RET proto-oncogene. Five years later, she developed Cushing's disease. A heterozygous germline variant was identified in the CDKN1B gene, specifically c.536del (p.Prol179GlnfsTer46), corresponding to a single-nucleotide deletion at position 536. This variant induces a frameshift, leading to an alternative stop codon. Immunostaining of the pituitary and thyroid tumors revealed a weak nuclear expression of p27/Kip1 without significant differences of expression between tumor and non-tumoral tissues. The NGS panel (Oncomine Comprehensive Assay v3) performed in both MTC and pituitary tissues identified the germline CDKN1B variant, as well as a pathogenic missense somatic variant c.182 A > G, p.(Gln61Arg) in HRAS in the MTC, without any RET somatic pathogenic variant. Evaluation of loss of heterozygosity (LOH) in both MTC and pituitary tissues showed compatibility with copy-neutral LOH, although further evidence is required for definitive confirmation. In conclusion, we report a clinical case of MTC coexisting with MEN4 due to a novel CDKN1B germline heterozygote frameshift variant.
Collapse
Affiliation(s)
- Fernández Mercè
- Department of Endocrinology and Nutrition, Institut de Recerca Sant Pau (IR SANTPAU), Endo-ERN. Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.
- Department of Endocrinology and Nutrition, Centre d'Atenció Integral Dos de Maig, Consorci Sanitari Integral, Barcelona, Spain.
| | - Queralt Asla
- Department of Endocrinology and Nutrition, Institut de Recerca Sant Pau (IR SANTPAU), Endo-ERN. Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Faculty of Medicine, University of Vic-Central University of Catalonia, Vic, Spain
| | - Francisco J Illana
- Department of Biochemistry, Hospital de la Santa Creu i Sant Pau, IIB Sant Pau, Barcelona, Spain
| | - Fusté Victòria
- Department of Pathological Anatomy, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Hernández-Losa Javier
- Department of Pathological Anatomy, Vall d'Hebrón Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Sesé Marta
- Department of Pathological Anatomy, Vall d'Hebrón Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Carmela Iglesias
- Department of Pathological Anatomy, Vall d'Hebrón Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Susan M Webb
- Department of Endocrinology and Nutrition, Institut de Recerca Sant Pau (IR SANTPAU), Endo-ERN. Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Anna Aulinas
- Department of Endocrinology and Nutrition, Institut de Recerca Sant Pau (IR SANTPAU), Endo-ERN. Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Faculty of Medicine, University of Vic-Central University of Catalonia, Vic, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
35
|
Sun HS, Huang T, Liu ZX, Xu YT, Wang YQ, Wang MC, Zhang SR, Xu JL, Zhu KY, Huang WK, Huang XF, Li J. Identification of mutations associated with congenital cataracts in nineteen Chinese families. BMC Ophthalmol 2025; 25:94. [PMID: 39994538 PMCID: PMC11853334 DOI: 10.1186/s12886-025-03920-4] [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] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 02/13/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Congenital cataracts (CC) are one of the leading causes of impaired vision or blindness in children, with approximately 8.3-25% being inherited. The aim of this study is to investigate the mutation spectrum and frequency of 9 cataract-associated genes in 19 Chinese families with congenital cataracts. PURPOSE To identify the gene variants associated with congenital cataracts. METHODS This study included a total of 58 patients from 19 pedigrees with congenital cataracts. All probands were initially screened by whole-exome sequencing(WES), and then validated by co-segregation analysis using Sanger sequencing. RESULTS Likely pathogenic variants were detected in 8 families, with a positivity rate of 42.1%. Variants in various genes were identified, including GJA3, CRYGD, CRYBA4, BFSP2, IARS2, CRYAA, CRYBA1, ARL2 and CRYBB3. Importantly, this study identified compound heterozygous variants of IARS2 in one family. CONCLUSIONS Our research findings have revealed multiple gene variants associated with cataracts, providing clinical guidance for improved molecular diagnosis of congenital cataracts in the era of precision medicine.
Collapse
Affiliation(s)
- Hai-Sen Sun
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Teng Huang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Zhe-Xuan Liu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yi-Tong Xu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Ya-Qi Wang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | | | - Shen-Rong Zhang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jia-Lin Xu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Kai-Yi Zhu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Wen-Kai Huang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xiu-Feng Huang
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Jin Li
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China.
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
36
|
Huang T, Liu YN, Ding DT, Wang Q, Xie QL, Miao XC, Qin C, Huang XF, Li J. Identification of a novel single nucleotide deletion in the NHS causing Nance-Horan syndrome. BMC Ophthalmol 2025; 25:92. [PMID: 39994540 PMCID: PMC11854407 DOI: 10.1186/s12886-025-03933-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 02/18/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Nance-Horan syndrome (NHS) is a rare X-linked dominant disorder caused by pathogenic variants in the NHS gene on chromosome Xp22.2-Xp22.13. Clinical manifestations consist of congenital cataracts, along with dysmorphic facial features and dental anomalies and, in certain instances, intellectual disability. This study aimed to identify the genetic cause responsible for NHS in a Chinese family with four individuals primarily presenting with congenital cataracts. METHODS Genomic DNA was collected from six family members, including four affected individuals (three females and one male) from a two-generation family. The family history and clinical data were documented. Whole-exome sequencing was performed on the proband, and candidate pathogenic variants were filtered through a series of screening steps and validated by Sanger sequencing. Co-segregation analysis was conducted to confirm the pathogenicity of the identified variant. RESULTS Genetic analysis revealed a novel frameshift pathogenic variant in NHS gene (c.1735delA: p.R579Gfs*91) present in all four affected members. All affected members exhibited congenital cataracts, congenital ptosis, strabismus, high myopia as well as dental and facial anomalies, and more severe characteristic features observed in the male patient. These clinical manifestations were consistent with the phenotype of NHS. CONCLUSION This study identified a novel NHS pathogenic variant in a Chinese family, expanding the mutational spectrum of NHS. Contrary to previous reports of female carriers exhibiting mild symptoms, we demonstrated severe ocular phenotypes in three affected females. These findings will assist in providing genetic counseling for NHS patients.
Collapse
Affiliation(s)
- Teng Huang
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Ya-Nan Liu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Dan-Tong Ding
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Qiao Wang
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Qiu-Ling Xie
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xue-Chuan Miao
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Chuan Qin
- Institute of PSI Genomics Co., Ltd, Wenzhou, China
| | - Xiu-Feng Huang
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, Wenzhou, 325027, China.
| | - Jin Li
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China.
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China.
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
37
|
Jacovas VC, Zelnick M, McNulty S, Ross JE, Khurana N, Pan X, Nieto A, Martin S, McLean B, Elnagheeb MA, Cowan MJ, Puck JM, Hershfield M, Verbsky J, Walter J, Allenspach E, Chan AY, van Oers NSC, Ghosh R, Piazza M, Yuan B, Notarangelo LD, Johnson BA, Chinn IK. The ClinGen Severe Combined Immunodeficiency Disease Variant Curation Expert Panel: Specifications for classification of variants in ADA , DCLRE1C , IL2RG , IL7R , JAK3 , RAG1 , and RAG2. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.11.25322033. [PMID: 39990552 PMCID: PMC11844601 DOI: 10.1101/2025.02.11.25322033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Purpose This collaborative study, led by the Clinical Genome Resource Severe Combined Immunodeficiency Disease Variant Curation Expert Panel (ClinGen SCID-VCEP), implemented and adapted the American College of Medical Genetics and Genomics/Association for Molecular Pathology (ACMG/AMP) guidelines for interpreting germline variants in genes with established relationships to SCID. The effort focused on the 7 most common SCID-related genes identified by SCID newborn screening in North America: ADA , DCLRE1C , IL2RG , IL7R , JAK3 , RAG1 , and RAG2 . Methods The SCID-VCEP conducted a rigorous review of variants that involved database analyses, literature review, and expert feedback to derive gene-specific modifications to the ACMG/AMP guidelines. These specifications were validated using a pilot set of 90 variants. Results: Of these 90 variants, 25 were classified as pathogenic, 21 as likely pathogenic, 14 as variants of uncertain significance (VUS), 18 as likely benign, and 12 as benign. Seventeen variants with conflicting classifications in ClinVar were successfully resolved. The criteria included modifications to 20 of the 28 original ACMG/AMP criteria specific to SCID-related genes. Conclusion The SCID-specific variant curation guidelines developed by the SCID-VCEP will enhance the precision of SCID genetic diagnosis and provide a robust framework for interpreting variants in SCID-related genes, contributing to appropriate treatment of SCID.
Collapse
|
38
|
Han SY, Cho SH, Jung SH, Kang M, Suh MW, Park MK, Lee JH, Lee SY. Genotypes and clinical phenotypes of pediatric patients with NOG variants: Middle ear surgical outcomes from a Tertiary Center in South Korea. Int J Pediatr Otorhinolaryngol 2025; 189:112230. [PMID: 39837070 DOI: 10.1016/j.ijporl.2025.112230] [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: 10/31/2024] [Revised: 12/31/2024] [Accepted: 01/08/2025] [Indexed: 01/23/2025]
Abstract
OBJECTIVE Although NOG variants are linked to congenital stapes fixation and conductive hearing loss (CHL), little is known about middle ear surgery outcomes and the characteristics of accompanying inner ear anomalies. We explored auditory phenotypes in patients with NOG variants, with a focus on the outcomes of middle ear surgery. METHODS This study included 11 patients from five unrelated Korean families harboring NOG variants. Genomic investigations were conducted using whole-exome sequencing and whole-genome sequencing. The clinical phenotypes, including pre- and postoperative audiological profiles, radiological abnormalities, and other comorbidities, were analyzed. RESULTS The average age at genetic testing was 8.2 years (range, 0-13 years). Two previously reported NOG variants (c.509C > T:p.Pro170Leu and c.252dup:p.Glu85ArgfsTer97) and three novel NOG variants, including the c.187G > T: p.Glu63Ter and two cryptic large deletion within the 17q22.2 region, were identified. All patients exhibited non-progressive CHL. Inner ear anomalies were documented in two patients, with variations such as cochlea and vestibular dysplasia. In this study, seven ears of four patients underwent stapedotomy, resulting in a significantly reduced air-bone gap of 10.18 ± 1.48 dB (P = 0.016), with sustained improvement. Conversely, patients carrying p.Pro170Leu variant, which is associated with poor outcomes for middle ear surgery, were excluded from surgical consideration. CONCLUSION We expanded the spectrum of genotypes and auditory phenotypes associated with NOG variants. Surgical intervention for CHL underlying NOG variants elicits favorable outcomes. However, clinicians should consider the potential for poor prognosis in certain NOG variants. Collectively, identifying NOG variants could guide the treatment strategies to improve CHL.
Collapse
Affiliation(s)
- Sang-Yoon Han
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Sung Ho Cho
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sung Ho Jung
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Myeongsin Kang
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Myung-Whan Suh
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea; Sensory Organ Research Institute, Seoul National University, Medical Research Center, Seoul, Republic of Korea
| | - Moo Kyun Park
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea; Sensory Organ Research Institute, Seoul National University, Medical Research Center, Seoul, Republic of Korea
| | - Jun Ho Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea; Sensory Organ Research Institute, Seoul National University, Medical Research Center, Seoul, Republic of Korea
| | - Sang-Yeon Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea; Sensory Organ Research Institute, Seoul National University, Medical Research Center, Seoul, Republic of Korea.
| |
Collapse
|
39
|
Beaman MM, Yin W, Smith AJ, Sears PR, Leigh MW, Ferkol TW, Kearney B, Olivier KN, Kimple AJ, Clarke S, Huggins E, Nading E, Jung SH, Iyengar AK, Zou X, Dang H, Barrera A, Majoros WH, Rehder CW, Reddy TE, Ostrowski LE, Allen AS, Knowles MR, Zariwala MA, Crawford GE. Promoter Deletion Leading to Allele Specific Expression in a Genetically Unsolved Case of Primary Ciliary Dyskinesia. Am J Med Genet A 2025; 197:e63880. [PMID: 39364610 PMCID: PMC11698635 DOI: 10.1002/ajmg.a.63880] [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: 04/15/2024] [Revised: 08/16/2024] [Accepted: 09/02/2024] [Indexed: 10/05/2024]
Abstract
Variation in the non-coding genome represents an understudied mechanism of disease and it remains challenging to predict if single nucleotide variants, small insertions and deletions, or structural variants in non-coding genomic regions will be detrimental. Our approach using complementary RNA-seq and targeted long-read DNA sequencing can prioritize identification of non-coding variants that lead to disease via alteration of gene splicing or expression. We have identified a patient with primary ciliary dyskinesia with a pathogenic coding variant on one allele of the SPAG1 gene, while the second allele appears normal by whole exome sequencing despite an autosomal recessive inheritance pattern. RNA sequencing revealed reduced SPAG1 transcript levels and exclusive allele specific expression of the known pathogenic allele, suggesting the presence of a non-coding variant on the second allele that impacts transcription. Targeted long-read DNA sequencing identified a heterozygous 3 kilobase deletion of the 5' untranslated region of SPAG1, overlapping the promoter and first non-coding exon. This non-coding deletion was missed by whole exome sequencing and gene-specific deletion/duplication analysis, highlighting the importance of investigating the non-coding genome in patients with "missing" disease-causing variation. This paradigm demonstrates the utility of both RNA and long-read DNA sequencing in identifying pathogenic non-coding variants in patients with unexplained genetic disease.
Collapse
Affiliation(s)
- M. Makenzie Beaman
- Department of Pediatrics, Division of Medical Genetics, Duke University, Durham, NC 27710 USA
- Medical Scientist Training Program, Duke University, Durham, NC 27710 USA
- University Program in Genetics & Genomics, Duke University, Durham, NC 27710 USA
| | - Weining Yin
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Amanda J. Smith
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Patrick R. Sears
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Margaret W. Leigh
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Thomas W. Ferkol
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Brendan Kearney
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC 27710 USA
- Center for Statistical Genetics and Genomics, Duke University, Durham, NC 27710 USA
| | - Kenneth N. Olivier
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Adam J. Kimple
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Department of Otolaryngology/Head & Neck Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Shannon Clarke
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC 27710 USA
- Center for Statistical Genetics and Genomics, Duke University, Durham, NC 27710 USA
| | - Erin Huggins
- Department of Pediatrics, Division of Medical Genetics, Duke University, Durham, NC 27710 USA
| | - Erica Nading
- Department of Pediatrics, Division of Medical Genetics, Duke University, Durham, NC 27710 USA
| | - Seung-Hye Jung
- Department of Pediatrics, Division of Medical Genetics, Duke University, Durham, NC 27710 USA
| | - Apoorva K. Iyengar
- University Program in Genetics & Genomics, Duke University, Durham, NC 27710 USA
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC 27710 USA
- Center for Statistical Genetics and Genomics, Duke University, Durham, NC 27710 USA
| | - Xue Zou
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC 27710 USA
- Center for Statistical Genetics and Genomics, Duke University, Durham, NC 27710 USA
- Program in Computational Biology & Bioinformatics, Duke University, Durham, NC 27710 USA
| | - Hong Dang
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Alejandro Barrera
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC 27710 USA
- Center for Statistical Genetics and Genomics, Duke University, Durham, NC 27710 USA
| | - William H. Majoros
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC 27710 USA
- Center for Statistical Genetics and Genomics, Duke University, Durham, NC 27710 USA
| | | | - Timothy E. Reddy
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC 27710 USA
- Center for Statistical Genetics and Genomics, Duke University, Durham, NC 27710 USA
| | - Lawrence E. Ostrowski
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Andrew S. Allen
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC 27710 USA
- Center for Statistical Genetics and Genomics, Duke University, Durham, NC 27710 USA
| | - Michael R. Knowles
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Maimoona A. Zariwala
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- These authors contributed equally
| | - Gregory E. Crawford
- Department of Pediatrics, Division of Medical Genetics, Duke University, Durham, NC 27710 USA
- These authors contributed equally
| |
Collapse
|
40
|
Deepthi B, Krishnasamy S, Krishnamurthy S, Khandelwal P, Sinha A, Hari P, Jaikumar R, Agrawal P, Saha A, Deepthi RV, Agarwal I, Sinha R, Venkatachari M, Shah MA, Bhatt GC, Krishnan B, Vasudevan A, Bagga A, Krishnamurthy S. Clinical characteristics and genetic profile of children with WDR72-associated distal renal tubular acidosis: a nationwide experience. Pediatr Nephrol 2025; 40:407-416. [PMID: 39150521 DOI: 10.1007/s00467-024-06478-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 07/22/2024] [Accepted: 07/22/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND Limited data, primarily from small case series, exist regarding the clinical profile, genetic variants, and outcomes of WDR72-associated distal renal tubular acidosis (WDR72-dRTA). METHODS Our study enrolled children diagnosed with WDR72-dRTA below 18 years of age from 9 Indian centers and analyzed their clinical characteristics, genetic profiles, and outcomes. Potential genotype-phenotype correlations were explored. RESULTS We report 22 patients (59% female) with WDR72-dRTA who were diagnosed at a median age of 5.3 (3, 8) years with polyuria (n = 17; 77.3%), poor growth (16; 72.7%), and rickets (9; 40.9%). Amelogenesis imperfecta was present in 21 (95.5%) cases. At presentation, all patients had normal anion gap metabolic acidosis; hypokalemia and nephrocalcinosis were seen in 17 (77.3%) patients each. Seven (31.8%) patients had concomitant proximal tubular dysfunction. Genetic analysis identified biallelic nonsense variants in 18 (81.8%) patients, including novel variants in 6 cases. A previously reported variant, c.88C > T, and a novel variant, c.655C > T, were the most frequent variants, accounting for 10 (45.5%) cases. Over a median follow-up of 1.3 (1, 8) years, the height velocity improved by 0.74 (0.2, 1.2) standard deviation scores, while 3 children (13.6%) progressed to chronic kidney disease (CKD) stage 2, with eGFR ranging from 67 to 76 mL/min/1.73 m2, respectively, after 11.3-16 years of follow-up. No specific genotype-phenotype correlation could be established. CONCLUSIONS WDR72-dRTA should be considered in children with typical features of amelogenesis imperfecta and dRTA. Biallelic nonsense variants are common in Asians. While most patients respond well to treatment with improved growth and preserved eGFR, on long-term follow-up, a decline in eGFR may occur.
Collapse
Affiliation(s)
- Bobbity Deepthi
- Pediatric Nephrology Services, Department of Pediatrics, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, India
| | - Sudarsan Krishnasamy
- Pediatric Nephrology Services, Department of Pediatrics, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, India
| | | | - Priyanka Khandelwal
- Division of Pediatric Nephrology, Department of Pediatrics, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Aditi Sinha
- Division of Pediatric Nephrology, Department of Pediatrics, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Pankaj Hari
- Division of Pediatric Nephrology, Department of Pediatrics, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Rohitha Jaikumar
- Division of Pediatric Nephrology, Department of Pediatrics, Lady Hardinge Medical College, New Delhi, India
| | - Prajal Agrawal
- Division of Pediatric Nephrology, Department of Pediatrics, Lady Hardinge Medical College, New Delhi, India
| | - Abhijeet Saha
- Division of Pediatric Nephrology, Department of Pediatrics, Lady Hardinge Medical College, New Delhi, India
| | - R V Deepthi
- Division of Pediatric Nephrology, Department of Pediatrics, Christian Medical College, Vellore, India
| | - Indira Agarwal
- Division of Pediatric Nephrology, Department of Pediatrics, Christian Medical College, Vellore, India
| | - Rajiv Sinha
- Division of Pediatric Nephrology, Institute of Child Health, Kolkata, India
| | - Mahesh Venkatachari
- Department of Pediatrics, All India Institute of Medical Sciences, Mangalagiri, India
| | - Mehul A Shah
- Little Star Children's Hospital, Hyderabad, India
| | - Girish Chandra Bhatt
- Division of Pediatric Nephrology, Department of Pediatrics, All India Institute of Medical Sciences, Bhopal, India
| | - Balasubramanian Krishnan
- Department of Dentistry, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, India
| | - Anil Vasudevan
- Division of Molecular Medicine, St. John's Research Institute, Bangalore, India
- Department of Pediatric Nephrology, St. John's Medical College Hospital, Bangalore, India
| | - Arvind Bagga
- Division of Pediatric Nephrology, Department of Pediatrics, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Sriram Krishnamurthy
- Pediatric Nephrology Services, Department of Pediatrics, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, India.
| |
Collapse
|
41
|
Wu W, Zhang Y, Xu J, Jiang H, Chen X. A novel RP1 truncating mutation that causes autosomal dominant retinitis pigmentosa (ADRP). ADVANCES IN OPHTHALMOLOGY PRACTICE AND RESEARCH 2025; 5:41-48. [PMID: 39911684 PMCID: PMC11795563 DOI: 10.1016/j.aopr.2024.08.005] [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: 05/28/2024] [Revised: 08/10/2024] [Accepted: 08/28/2024] [Indexed: 02/07/2025]
Abstract
Background Retinitis pigmentosa (RP) is a genetically and clinically heterogeneous group of hereditary degenerative disorders affecting approximately one in every 4000 people worldwide. Abnormalities in the retina's photoreceptors can cause night blindness or even complete vision loss. Retinitis Pigmentosa 1 (RP1), also known as the oxygen-regulated protein-1, is a microtubule-associated protein (MAP) that organizes the outer segment of the photoreceptor. Besides, mutations in the RP1 gene are associated with dominant or recessive form of RP. This study aims to identify the potential pathogenic genes in Chinese RP patients and to elucidate the association relationship between the mutant gene and the phenotypes. Methods Multiple ophthalmic examinations, whole-exome sequencing, sanger sequencing, and in silico analysis were performed to evaluate the clinical features and pathogenic genes in a five-generation Chinese family diagnosed with RP. Results Our findings revealed a novel truncating mutation c.2015_2018del p. (Lys672Argfs∗9) in RP1 that may result in the translation of a protein with deleterious effects on photoreceptors. Therefore, resulting in autosomal dominant retinitis pigmentosa (ADRP). Conclusions This study broaden the range of genetic mutations associated with RP1 in ADRP and make a valuable contribution to the ongoing endeavors aimed at characterizing the molecular aspects of Chinese ADRP. Future studies would pay more attention in determining the characterization of the mutantations in RP1 gene and the relationship between genotype and phenotype in RP patients.
Collapse
Affiliation(s)
- Wei Wu
- Eye Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Lab of Ophthalmology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Zhang
- Eye Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Lab of Ophthalmology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Jingjie Xu
- Eye Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Lab of Ophthalmology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Hua Jiang
- Department of Otolaryngology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiangjun Chen
- Eye Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Lab of Ophthalmology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
42
|
Stark Z, Byrne AB, Sampson MG, Lennon R, Mallett AJ. A guide to gene-disease relationships in nephrology. Nat Rev Nephrol 2025; 21:115-126. [PMID: 39443743 DOI: 10.1038/s41581-024-00900-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2024] [Indexed: 10/25/2024]
Abstract
The use of next-generation sequencing technologies such as exome and genome sequencing in research and clinical care has transformed our understanding of the molecular architecture of genetic kidney diseases. Although the capability to identify and rigorously assess genetic variants and their relationship to disease has advanced considerably in the past decade, the curation of clinically relevant relationships between genes and specific phenotypes has received less attention, despite it underpinning accurate interpretation of genomic tests. Here, we discuss the need to accurately define gene-disease relationships in nephrology and provide a framework for appraising genetic and experimental evidence critically. We describe existing international programmes that provide expert curation of gene-disease relationships and discuss sources of discrepancy as well as efforts at harmonization. Further, we highlight the need for alignment of disease and phenotype terminology to ensure robust and reproducible curation of knowledge. These collective efforts to support evidence-based translation of genomic sequencing into practice across clinical, diagnostic and research settings are crucial for delivering the promise of precision medicine in nephrology, providing more patients with timely diagnoses, accurate prognostic information and access to targeted treatments.
Collapse
Affiliation(s)
- Zornitza Stark
- ClinGen, Boston, MA, USA.
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.
- Australian Genomics, Melbourne, Victoria, Australia.
| | - Alicia B Byrne
- ClinGen, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Matthew G Sampson
- ClinGen, Boston, MA, USA
- Division of Nephrology, Boston Children's Hospital, Boston, MA, USA
- Department of Paediatrics, Harvard Medical School, Boston, MA, USA
| | - Rachel Lennon
- ClinGen, Boston, MA, USA
- Wellcome Centre for Cell-Matrix Research, The University of Manchester, Manchester, UK
- Department of Paediatric Nephrology, Royal Manchester Children's Hospital, Manchester, UK
| | - Andrew J Mallett
- ClinGen, Boston, MA, USA.
- Townsville Hospital and Health Service, Townsville, Queensland, Australia.
- College of Medicine and Dentistry, James Cook University, Townsville, Queensland, Australia.
- Institute for Molecular Bioscience and Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
43
|
Rius A, Aguirre N, Erra L, Brunello FG, Biagioli G, Zaiat J, Marti MA. Study of the impact of ClinGen Revisions on ACMG/AMP variant semi-automatic classification for Rare Diseases diagnosis. Clin Chim Acta 2025; 566:120065. [PMID: 39615735 DOI: 10.1016/j.cca.2024.120065] [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: 10/17/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 12/11/2024]
Abstract
With the rapid development of massive sequencing technologies, the analysis of genetic variants for clinical diagnosis has exponentially escalated, particularly in the context of Rare Diseases (RDs). Diagnosing them involves identifying the genetic variants responsible for the underlying pathology development. In 2015, the American College of Medical Genetics (ACMG) established a set of recommendations to assess the evidence associated with each variant, aiming to achieve a standardized five tier classification. Over the past 5 years, ClinGen, the NIH-funded Clinical Genome Resource, has reviewed these criteria in order to make variant classification a more reproducible and rigorous process. This paper examines the impact of ClinGen-Rev modifications on variant classification, comparing them with the ACMG-2015 original recommendations. After analyzing sets of genetic variants, extracted from VCFs samples, using both criteria, we observed a change in 8.0 % of the clinical verdicts for these variants. ClinGen-Rev modifications correctly categorized 89.2 % of the curated variants, representing a significant improvement compared to the 65.6 % achieved by ACMG-2015. We also analyzed the modifications impact in a real like clinical setting, showing a significant overall reduction of VUS variants and thus potential reduction in analysis time. Finally, we discuss the underlying reasons for the most relevant changes in terms of specific labels and present their implications on the prioritization and selection process of variants, identifying some recommendations of key significant importance.
Collapse
Affiliation(s)
- Ana Rius
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (FCEyN-UBA) e Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN) CONICET, Pabellón 2 de Ciudad Universitaria, Ciudad de Buenos Aires C1428EHA, Argentina.
| | - Nicolas Aguirre
- Bitgenia, Análisis de Datos Genómicos, Camino Parque Centenario N° 2565 - La Plata, Alicia Moreau de Justo N° 1750 3° H - CABA, Buenos Aires, Argentina
| | - Lorenzo Erra
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (FCEyN-UBA) e Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN) CONICET, Pabellón 2 de Ciudad Universitaria, Ciudad de Buenos Aires C1428EHA, Argentina
| | - Franco Gino Brunello
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (FCEyN-UBA) e Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN) CONICET, Pabellón 2 de Ciudad Universitaria, Ciudad de Buenos Aires C1428EHA, Argentina
| | - German Biagioli
- Bitgenia, Análisis de Datos Genómicos, Camino Parque Centenario N° 2565 - La Plata, Alicia Moreau de Justo N° 1750 3° H - CABA, Buenos Aires, Argentina
| | - Jonathan Zaiat
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (FCEyN-UBA) e Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN) CONICET, Pabellón 2 de Ciudad Universitaria, Ciudad de Buenos Aires C1428EHA, Argentina
| | - Marcelo A Marti
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (FCEyN-UBA) e Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN) CONICET, Pabellón 2 de Ciudad Universitaria, Ciudad de Buenos Aires C1428EHA, Argentina
| |
Collapse
|
44
|
Ding Y, Zhang Q, Gao S, Li J, Chang G, Wang Y, Wang L, Li X, Chen Y, Yao RE, Yu T, Li N, Lou D, Wang X. Focusing on Rare Variants Related to Maturity-Onset Diabetes of the Young in Children. Pediatr Diabetes 2025; 2025:8155443. [PMID: 40303944 PMCID: PMC12017003 DOI: 10.1155/pedi/8155443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 01/08/2025] [Indexed: 05/02/2025] Open
Abstract
Background: In this study, we analysed the clinical and genetic characteristics and follow-up data of patients with maturity-onset diabetes of the young (MODY). Methods: From January 2015 to December 2022, patients with persistent hyperglycaemia suspected of having monogenic diabetes or diabetes syndrome were recruited, and next-generation sequencing (NGS) was performed at the Shanghai Children's Medical Center. Patients' clinical and laboratory findings were recorded preceding follow-ups. Candidate variants were verified using Sanger sequencing. Variant pathogenicity was evaluated according to the American College of Medical Genetics and Genomics (ACMG) guidelines. Results: Genetic testing was performed in 175 children. MODY-related pathogenic or likely pathogenic gene variants were identified in 30 patients from different families. Of these, 11 were diagnosed with GCK-MODY (36.7%), six with INS-MODY (20%), five with HNF1A-MODY (16.7%), five with ABCC8-MODY (16.7%), two with HNF1B-MODY (6.7%) and one with HNF4A-MODY (3.3%). There was one shift variant and seven splice-site variants, and the rest were missense variants. We discovered six novel variants. Of the 30 patients, 63.3% had a family history of diabetes, 13.3% had diabetic ketoacidosis (DKA), and 16.7% had positive diabetes-associated autoantibodies. The diabetes phenotype of patients with the INS variant was similar to that of patients with type 1 diabetes. All patients, including those having positive autoantibodies, required long-term insulin therapy during follow-ups. Four patients with the ABCC8 variant were unable to switch to oral sulfonylurea therapy and continued insulin therapy. Conclusion: Genetic testing is helpful for the precise diagnosis and treatment of patients with MODY, including those with DKA history and positive diabetes autoantibody. GCK-MODY is the most common type of MODY, and patients with INS variant account for a relatively large proportion of MODY cases in our cohort.
Collapse
Affiliation(s)
- Yu Ding
- Department of Endocrinology and Metabolism, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | - Qianwen Zhang
- Department of Endocrinology and Metabolism, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | - Shiyang Gao
- Department of Endocrinology and Metabolism, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | - Juan Li
- Department of Endocrinology and Metabolism, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | - Guoying Chang
- Department of Endocrinology and Metabolism, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | - Yirou Wang
- Department of Endocrinology and Metabolism, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | - Libo Wang
- Department of Endocrinology and Metabolism, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | - Xin Li
- Department of Endocrinology and Metabolism, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | - Yao Chen
- Department of Endocrinology and Metabolism, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | - Ru-en Yao
- Department of Medical Genetics and Molecular Diagnostic Laboratory, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | - Tingting Yu
- Department of Medical Genetics and Molecular Diagnostic Laboratory, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | - Niu Li
- Department of Reproductive Genetics, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Dan Lou
- Department of Pediatrics, The First Affiliated Hospital of Henan University of Science and Technology, Henan University of Science and Technology, Luoyang 471000, China
| | - Xiumin Wang
- Department of Endocrinology and Metabolism, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| |
Collapse
|
45
|
Ariza MJ, Coca-Prieto I, Rioja J, Muñiz-Grijalvo O, Zambón-Rados D, Blanco-Echevarría A, Arrobas-Velilla T, Delgado-Lista J, León-Jiménez D, Casañas-Martínez M, Álvarez-Sala LA, Gutiérrez-Carrasquilla L, Sánchez-Gil J, Domènech M, González-Jiménez A, Benítez-Toledo MJ, Espíldora-Hernández J, Ortega-Martínez de Victoria E, Sánchez-Chaparro MÁ, Valdivielso P. Pathogenicity assessment of genetic variants identified in patients with severe hypertriglyceridemia: Novel cases of familial chylomicronemia syndrome from the Dyslipidemia Registry of the Spanish Atherosclerosis Society. Genet Med 2025; 27:101365. [PMID: 39873189 DOI: 10.1016/j.gim.2025.101365] [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: 03/21/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 01/30/2025] Open
Abstract
PURPOSE Genetic testing is required to confirm a diagnosis of familial chylomicronemia syndrome (FCS). We assessed the pathogenicity of variants identified in the FCS canonical genes to diagnose FCS cases. METHODS 245 patients with severe hypertriglyceridemia underwent next-generation sequencing. Preliminary variant pathogenicity criteria and classification, based on the American College of Medical Genetics and Genomics guidelines, were obtained online and verified. Phenotype evaluation was based on lipoprotein lipase activity deficiency, a clinical score, and/or type I hyperlipoproteinemia determined in 25 patients. RESULTS Twenty-four biallelic variants were analyzed. Evidence-based criteria allowed the reclassification of 8 likely pathogenic (LP) variants in the LPL, APOA5, and LMF1 genes into pathogenic (P) and the change of 2 variants of uncertain significance (VUS) to LP. Conversely, 2 variations in LMF1 remained as VUS. Additionally, 1 variant in LPL and 2 in GPIHBP1 were likely benign. Twenty FCS cases had biallelic P/LP variants and 1 patient, with an FCS phenotype, harbored biallelic VUS. FCS was excluded from 4 patients with pathogenic/likely benign combinations. CONCLUSION The analysis of the clinical and biochemical features of patients with variants in the FCS canonical genes allowed a confident variant classification that helped in the diagnosis of novel FCS cases.
Collapse
Affiliation(s)
- María José Ariza
- Lipids and Atherosclerosis Laboratory, Department of Medicine and Dermatology, Centro de Investigaciones Médico Sanitarias (CIMES), Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA -Plataforma Bionand), University of Málaga, Málaga, Spain
| | - Inmaculada Coca-Prieto
- Lipid Unit, Internal Medicine Service, University Hospital Virgen de la Victoria, Málaga, Spain.
| | - José Rioja
- Lipids and Atherosclerosis Laboratory, Department of Medicine and Dermatology, Centro de Investigaciones Médico Sanitarias (CIMES), Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA -Plataforma Bionand), University of Málaga, Málaga, Spain
| | - Ovidio Muñiz-Grijalvo
- Lipid Unit, Internal Medicine Service, University Hospital Virgen del Rocío, Seville, Spain
| | | | - Agustín Blanco-Echevarría
- Internal Medicine Department, University Hospital October 12, i+12 Institute, Madrid, Spain; Department of Medicine, School of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Teresa Arrobas-Velilla
- Nutrition and cardiovascular risk laboratory, Clinical Biochemistry Unit, University Hospital Virgen Macarena, Sevilla, Spain
| | - Javier Delgado-Lista
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, University Hospital Reina Sofía, Department of Medical and Surgical Sciences, Universidad de Córdoba, IMIBIC, Córdoba, Spain; CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - David León-Jiménez
- Vascular Risk Unit, Internal Medicine department, Clinical Unit for Comprehensive Medical Care (UCAMI), University Hospital Virgen del Rocío, University of Seville, Seville, Spain; Clinical Epidemiology and Vascular Unit, Instituto de Biomedicina de Sevilla (IBIS), Seville, Spain
| | - Marta Casañas-Martínez
- Lipids and Vascular Risk Unit, Internal Medicine Service, Hospital San Pedro, Logroño, Spain
| | - Luis Antonio Álvarez-Sala
- Lipids and Cardiovascular Risk Unit, Internal Medicine Service, Hospital General Universitario Gregorio Marañón, Instituto de investigaciones Sanitarias Gregorio Marañón (IiSGM), Madrid, Spain; Department of Medicine, School of Medicine, Complutense University of Madrid, Madrid, Spain
| | | | - Justo Sánchez-Gil
- Lipid Unit, Internal Medicine Service, Hospital de Antequera, Málaga, Spain
| | - Mónica Domènech
- Occupational health area, Lipids and Vascular Risk Unit, Hospital Clínic Barcelona, Spain
| | - Andrés González-Jiménez
- Bioinformatics platform, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain
| | | | | | - Emilio Ortega-Martínez de Victoria
- CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; Lipid and Vascular risk Unit, Endocrinology and Nutrition department, IDIBAPS-FCRB, Hospital Clínic Barcelona, Spain
| | - Miguel Ángel Sánchez-Chaparro
- Lipids and Atherosclerosis Laboratory, Department of Medicine and Dermatology, Centro de Investigaciones Médico Sanitarias (CIMES), Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA -Plataforma Bionand), University of Málaga, Málaga, Spain; Lipid Unit, Internal Medicine Service, University Hospital Virgen de la Victoria, Málaga, Spain
| | - Pedro Valdivielso
- Lipids and Atherosclerosis Laboratory, Department of Medicine and Dermatology, Centro de Investigaciones Médico Sanitarias (CIMES), Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA -Plataforma Bionand), University of Málaga, Málaga, Spain; Lipid Unit, Internal Medicine Service, University Hospital Virgen de la Victoria, Málaga, Spain
| |
Collapse
|
46
|
Pan J, Teng H, Liu F, Chen S, Liu Y, Teng Y, Liang D, Li Z, Wu L. Oligogenic effect is associated with the clinical heterogeneity of autosomal dominant deafness-15. Sci Rep 2025; 15:1981. [PMID: 39809934 PMCID: PMC11733205 DOI: 10.1038/s41598-025-85881-8] [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] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 01/07/2025] [Indexed: 01/30/2025] Open
Abstract
Autosomal dominant deafness-15 which is caused by mutation in the POU4F3 gene, has been reported with a wide degree of clinical heterogeneity, even between intrafamilial members. However, the reason is still elusive. In this study, A four-generation Chinese family with 11 patients manifesting late-onset progressive non-syndromic hearing loss was recruited. The phenotype of hearing loss in this family showed a large variability in terms of onset age and progression speed. A novel mutation (c.706 C > T, p.L236F) was identified by the whole exome sequencing, and its pathogenicity was confirmed by altering the subcellular localization of POU4F3. In addition, we found that two individuals with earlier age of onset and more rapid progression of hearing loss carry additional pathogenic variants in other deafness genes (III-7, STRC:c.4057 C > T; IV-1, GJB2:c.109G > A; CDC14A:c.935G > A). By using the real time quantitative PCR, western blot, luciferase assays and electrophoretic mobility-shift assay, POU4F3 was proved to directly regulate the expression of STRC, GJB2 and CDC14A respectively. ChIP-seq further revealed that POU4F3 can also bind to a series of deafness genes. In summary we expanded the mutation spectrum of POU4F3 by identifying a novel mutation and its pathogenicity. Meanwhile, three genes STRC, GJB2 and CDC14A were validated as POU4F3 new targets, implicating that the variants in the three genes may play a role of genetic modifier to generate a synergistic and enhancement effect on the progression of DFNA15.
Collapse
Affiliation(s)
- Jianyan Pan
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, 410000, Hunan, China
- Department of Birth Health and Genetics, The Reproductive Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530000, Guangxi, China
| | - Hua Teng
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, 410000, Hunan, China
| | - Fang Liu
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, 410000, Hunan, China
| | - Siyi Chen
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, 410000, Hunan, China
| | - Yaning Liu
- Laboratory of Molecular Genetics, Hunan Jiahui Genetics Hospital, Changsha, 410000, Hunan, China
| | - Yanling Teng
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, 410000, Hunan, China
| | - Desheng Liang
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, 410000, Hunan, China
- Laboratory of Molecular Genetics, Hunan Jiahui Genetics Hospital, Changsha, 410000, Hunan, China
| | - Zhuo Li
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, 410000, Hunan, China.
| | - Lingqian Wu
- Center for Medical Genetics, Hunan Key Laboratory of Medical Genetics, MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, 410000, Hunan, China.
- Laboratory of Molecular Genetics, Hunan Jiahui Genetics Hospital, Changsha, 410000, Hunan, China.
- Bright Prosperity Institute, Room 1006-2, 10th Floor, Building 1, No. 180 Kecheng Street, Hangzhou, 310000, China.
| |
Collapse
|
47
|
Zhou JY, Wang CY, Li J, Chen GL, Tang XW, Li FT, Jiang F. First Reported Case of Hemoglobin H Disease Caused by the Rare α-Globin Gene Mutation ( HBA2 c.244delT) in a Chinese Family. Hemoglobin 2025; 49:69-71. [PMID: 39748151 DOI: 10.1080/03630269.2024.2444360] [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: 06/09/2023] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 01/04/2025]
Abstract
Microcytosis of red cells and mild anemia are common in thalassemia carriers but those phenotypes are not specific. It is really a challenge for clinical interpretation of those variants. Co-segregation with disease in affected family members or specific phenotypes such as the abnormal Hb H are very helpful to assess the pathogenicity of rare variants. HBA2 c.244delT was only reported in a 19-year-old woman with mild microcytosis and hypochromia. There was no other information about this variant reported before. We first described the case of this variant compounded with SEA deletion who presented with moderate anemia. Co-segregation analysis was confirmed by Sanger sequencing. Our study gave evidence for predicting the pathogenicity of this rare missense variant.
Collapse
Affiliation(s)
- Jian-Ying Zhou
- Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center affiliated with Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Chen-Yu Wang
- Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center affiliated with Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Jian Li
- Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center affiliated with Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Gui-Lan Chen
- Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center affiliated with Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Xue-Wei Tang
- Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center affiliated with Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Fa-Tao Li
- Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center affiliated with Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Fan Jiang
- Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center affiliated with Guangzhou Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
48
|
Patsalis C, Kyriakou S, Georgiadou M, Ioannou L, Constantinou L, Soteriou V, Jossif A, Evangelidou P, Sismani C, Kypri E, Ioannides M, Koumbaris G. Investigating TNNC1 gene inheritance and clinical outcomes through a comprehensive familial study. Am J Med Genet A 2025; 197:e63838. [PMID: 39248034 DOI: 10.1002/ajmg.a.63838] [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: 02/01/2024] [Revised: 07/16/2024] [Accepted: 07/27/2024] [Indexed: 09/10/2024]
Abstract
Hypertrophic cardiomyopathy (HCM) and restrictive cardiomyopathy (RCM) have significant phenotypic overlap and a similar genetic background, both caused mainly by variants in sarcomeric genes. HCM is the most common cardiomyopathy, while RCM is a rare and often underdiagnosed heart condition, with a poor prognosis. This study focuses on a large family with four infants diagnosed with fatal RCM associated with biventricular hypertrophy. Affected infants were found to be homozygous for NM_003280.3(TNNC1):c.23C>T(p.Ala8Val) variant. Interestingly, this variant resulted in a low penetrance and mild form of hypertrophic cardiomyopathy (HCM) in relatives carrying a single copy of the variant. Overall, this study underscores the complex nature of genetic inheritance in cardiomyopathies and the wide range of clinical presentations they can exhibit. This emphasizes the vital role of genetic testing in providing essential insights crucial for diagnosis, prognosis, early intervention, and the development of potential treatment strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Antonis Jossif
- Paedi Center for Specialized Pediatrics, Nicosia, Cyprus
| | - Paola Evangelidou
- Department of Cytogenetics and Genomics, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Carolina Sismani
- Department of Cytogenetics and Genomics, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | | | | | | |
Collapse
|
49
|
Freire MV, Thissen R, Martin M, Fasquelle C, Helou L, Durkin K, Artesi M, Lumaka A, Leroi N, Segers K, Deberg M, Gatot JS, Habran L, Palmeira L, Josse C, Bours V. Genetic evaluation of patients with multiple primary cancers. Oncol Lett 2025; 29:4. [PMID: 39492936 PMCID: PMC11526284 DOI: 10.3892/ol.2024.14750] [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: 04/10/2024] [Accepted: 08/22/2024] [Indexed: 11/05/2024] Open
Abstract
Regarding inherited cancer predisposition, single gene carriers of pathogenic variants (PVs) have been extensively reported on in the literature, whereas the oligogenic coinheritance of heterozygous PVs in cancer-related genes is a poorly studied event. Currently, due to the increased number of cancer survivors, the probability of patients presenting with multiple primary cancers (MPCs) is higher. The present study included patients with MPCs aged ≤45 years without known PVs in common cancer predisposition genes. This study used whole exome sequencing (WES) of germline and tumoral DNA, chromosomal microarray analysis (CMA) of germline DNA (patients 1-7, 9 and 10), and a karyotype test of patient 8 to detect variants associated with the disease. The 10 patients included in the study presented a mean of 3 cancers per patient. CMA showed two microduplications and one microdeletion, while WES of the germline DNA identified 1-3 single nucleotide variants of potential interest to the disease in each patient and two additional copy number variants. Most of the identified variants were classified as variants of uncertain significance. The mapping of the germline variants into their pathways showed a possible additive effect of these as the cause of the cancer. A total of 12 somatic samples from 5 patients were available for sequencing. All of the germline variants were also present in the somatic samples, while no second hits were identified in the same genes. The sequencing of patients with early cancers, family history and multiple tumors is already a standard of care. However, growing evidence has suggested that the assessment of patients should not stop at the identification of one PV in a cancer predisposition gene.
Collapse
Affiliation(s)
- Maria Valeria Freire
- Department of Human Genetics, GIGA Research Center-University of Liège and CHU Liège, 4000 Liège, Belgium
| | - Romain Thissen
- Department of Human Genetics, GIGA Research Center-University of Liège and CHU Liège, 4000 Liège, Belgium
| | - Marie Martin
- Department of Human Genetics, CHU Liège, 4000 Liège, Belgium
| | | | - Laura Helou
- Department of Human Genetics, GIGA Research Center-University of Liège and CHU Liège, 4000 Liège, Belgium
| | - Keith Durkin
- Department of Human Genetics, GIGA Research Center-University of Liège and CHU Liège, 4000 Liège, Belgium
- Department of Human Genetics, CHU Liège, 4000 Liège, Belgium
| | - Maria Artesi
- Department of Human Genetics, GIGA Research Center-University of Liège and CHU Liège, 4000 Liège, Belgium
- Department of Human Genetics, CHU Liège, 4000 Liège, Belgium
| | - Aimé Lumaka
- Department of Human Genetics, GIGA Research Center-University of Liège and CHU Liège, 4000 Liège, Belgium
- Department of Human Genetics, CHU Liège, 4000 Liège, Belgium
| | - Natacha Leroi
- Department of Human Genetics, CHU Liège, 4000 Liège, Belgium
| | - Karin Segers
- Department of Human Genetics, CHU Liège, 4000 Liège, Belgium
| | - Michelle Deberg
- Department of Human Genetics, CHU Liège, 4000 Liège, Belgium
| | | | - Lionel Habran
- Department of Pathology, CHU Liège, 4000 Liège, Belgium
| | - Leonor Palmeira
- Department of Human Genetics, CHU Liège, 4000 Liège, Belgium
| | - Claire Josse
- Department of Medical Oncology, GIGA Research Center-University of Liège and CHU Liège, 4000 Liège, Belgium
| | - Vincent Bours
- Department of Human Genetics, GIGA Research Center-University of Liège and CHU Liège, 4000 Liège, Belgium
- Department of Human Genetics, CHU Liège, 4000 Liège, Belgium
| |
Collapse
|
50
|
Drackley A, Somerville C, Arnaud P, Baudhuin LM, Hanna N, Kluge ML, Kotzer K, Boileau C, Bronicki L, Callewaert B, Cecchi A, Dietz H, Guo D, Harris S, Jarinova O, Lindsay M, Little L, Loeys B, MacCarrick G, Meester J, Milewicz D, Morisaki T, Morisaki H, Murdock D, Renard M, Richer J, Robert L, Ouzounian M, Van Laer L, De Backer J, Muiño-Mosquera L. Interpretation and classification of FBN1 variants associated with Marfan syndrome: consensus recommendations from the Clinical Genome Resource's FBN1 variant curation expert panel. Genome Med 2024; 16:154. [PMID: 39741318 DOI: 10.1186/s13073-024-01423-3] [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: 01/26/2024] [Accepted: 12/06/2024] [Indexed: 01/02/2025] Open
Abstract
BACKGROUND In 2015, the American College of Medical Genetics and Genomics (ACMG) and the Association for Molecular Pathology (AMP) developed standardized variant curation guidelines for Mendelian disorders. Although these guidelines have been widely adopted, they are not gene- or disease-specific. To mitigate classification discrepancies, the Clinical Genome Resource FBN1 variant curation expert panel (VCEP) was established in 2018 to develop adaptations to the ACMG/AMP criteria for FBN1 in association with Marfan syndrome. METHODS The specific recommendations were developed through literature review, surveys, online expert panel discussions, and pilot testing of a set of 60 different variants. Consensus among experts was considered reached if at least 75% of the members agreed with a given rule specification. The final set of rules received approval from the ClinGen Sequence Variant Interpretation Working Group. RESULTS The developed specifications introduce modifications to 14 of the 28 ACMG/AMP evidence criteria and deem 6 criteria non-applicable. Some of these specifications include refining the minor allele frequency thresholds, creating a FBN1-specific flowchart for PVS1, defining functional domains of the protein, developing a point-based system of counting probands and instances of de novo occurrences, recommending a points-based method of accounting for family segregation data, and clarifying the applicable functional assays that should be considered. To date, this VCEP has curated 120 variants which have been deposited to ClinVar with the 3-star review status. CONCLUSIONS Establishing specific adaptations for FBN1 has provided a framework to foster greater classification concordance among clinical laboratories, ultimately improving clinical care for patients with Marfan syndrome.
Collapse
Affiliation(s)
- A Drackley
- Department of Pathology & Laboratory Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - C Somerville
- Genetics Diagnostic Laboratory, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - P Arnaud
- Genetics Department, Hôpital Bichat, Université Paris Cité, Paris, France
- European Reference Network for Rare Multisystemic Vascular Disease (VASCERN), HTAD and MSA Rare Disease, Working Group, Paris, France
| | - L M Baudhuin
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - N Hanna
- Genetics Department, Hôpital Bichat, Université Paris Cité, Paris, France
- European Reference Network for Rare Multisystemic Vascular Disease (VASCERN), HTAD and MSA Rare Disease, Working Group, Paris, France
| | - M L Kluge
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - K Kotzer
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - C Boileau
- Genetics Department, Hôpital Bichat, Université Paris Cité, Paris, France
| | - L Bronicki
- Genetics Diagnostic Laboratory, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
- Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa, ON, Canada
| | - B Callewaert
- Centre for Medical Genetics, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - A Cecchi
- Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - H Dietz
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - D Guo
- Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - S Harris
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - O Jarinova
- Genetics Diagnostic Laboratory, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
- Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa, ON, Canada
| | - M Lindsay
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - L Little
- Genetics Diagnostic Laboratory, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - B Loeys
- European Reference Network for Rare Multisystemic Vascular Disease (VASCERN), HTAD and MSA Rare Disease, Working Group, Paris, France
- Centre of Medical Genetics, Antwerp University Hospital and University of Antwerp, Antwerp, Belgium
| | - G MacCarrick
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - J Meester
- European Reference Network for Rare Multisystemic Vascular Disease (VASCERN), HTAD and MSA Rare Disease, Working Group, Paris, France
- Centre of Medical Genetics, Antwerp University Hospital and University of Antwerp, Antwerp, Belgium
| | - D Milewicz
- Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - T Morisaki
- IMSUT Hospital, The Institute of Medical Science, The University of Tokyo, Minato-Ku, Tokyo, Japan
| | - H Morisaki
- IMSUT Hospital, The Institute of Medical Science, The University of Tokyo, Minato-Ku, Tokyo, Japan
- Department of Medical Genetics, Sakakibara Heart Institute, Fuchu, Tokyo, Japan
| | - D Murdock
- Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - M Renard
- Centre for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - J Richer
- Department of Medical Genetics, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - L Robert
- Department of Cardiology, Guy's and St Thomas' Foundation Trust, London, UK
| | - M Ouzounian
- Division of Cardiac Surgery, University of Toronto, Toronto, ON, Canada
| | - L Van Laer
- European Reference Network for Rare Multisystemic Vascular Disease (VASCERN), HTAD and MSA Rare Disease, Working Group, Paris, France
- Centre of Medical Genetics, Antwerp University Hospital and University of Antwerp, Antwerp, Belgium
| | - J De Backer
- European Reference Network for Rare Multisystemic Vascular Disease (VASCERN), HTAD and MSA Rare Disease, Working Group, Paris, France
- Centre for Medical Genetics, Ghent University Hospital, Ghent, Belgium
- Department of Cardiology, Ghent University Hospital, Ghent, Belgium
- Department of Internal Medicine and Paediatrics, Ghent University, Ghent, Belgium
| | - L Muiño-Mosquera
- European Reference Network for Rare Multisystemic Vascular Disease (VASCERN), HTAD and MSA Rare Disease, Working Group, Paris, France.
- Centre for Medical Genetics, Ghent University Hospital, Ghent, Belgium.
- Department of Internal Medicine and Paediatrics, Ghent University, Ghent, Belgium.
- Division of Paediatric Cardiology, Department of Paediatrics, Ghent University Hospital, Ghent, Belgium.
| |
Collapse
|