1
|
Duthoo E, Beyls E, Backers L, Gudjónsson T, Huang P, Jonckheere L, Riemann S, Parton B, Du L, Debacker V, De Bruyne M, Hoste L, Baeyens A, Vral A, Van Braeckel E, Staal J, Mortier G, Kerre T, Pan-Hammarström Q, Sørensen CS, Haerynck F, Claes KB, Tavernier SJ. Replication stress, microcephalic primordial dwarfism, and compromised immunity in ATRIP deficient patients. J Exp Med 2025; 222:e20241432. [PMID: 40029331 PMCID: PMC11874998 DOI: 10.1084/jem.20241432] [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: 08/12/2024] [Revised: 11/13/2024] [Accepted: 12/23/2024] [Indexed: 03/05/2025] Open
Abstract
Ataxia telangiectasia and Rad3-related (ATR) kinase and its interacting protein ATRIP orchestrate the replication stress response. Homozygous splice variants in the ATRIP gene, resulting in ATRIP deficiency, were identified in two patients of independent ancestry with microcephaly, primordial dwarfism, and recurrent infections. The c.829+5G>T patient exhibited lymphopenia, poor vaccine responses, autoimmune features with hemolytic anemia, and neutropenia. Immunophenotyping revealed reduced CD16+/CD56dim NK cells and absent naïve T cells, MAIT cells, and iNKT cells. Lymphocytic defects were characterized by TCR oligoclonality, abnormal class switch recombination, and impaired T cell proliferation. ATRIP deficiency resulted in low-grade ATR activation but impaired CHK1 phosphorylation under genotoxic stress. ATRIP-deficient cells inadequately regulated DNA replication, leading to chromosomal instability, compromised cell cycle control, and impaired cell viability. CRISPR-SelectTIME confirmed reduced cell fitness for both variants. This study establishes ATRIP deficiency as a monogenic cause of microcephalic primordial dwarfism, highlights ATRIP's critical role in protecting immune cells from replication stress, and offers new insights into its canonical functions.
Collapse
Affiliation(s)
- Evi Duthoo
- Primary Immunodeficiency Research Lab (PIRL), Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Center for Primary Immunodeficiency, Jeffrey Modell Diagnosis and Research Center, ERN-RITA Reference Center, Ghent University Hospital, Ghent, Belgium
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Elien Beyls
- Primary Immunodeficiency Research Lab (PIRL), Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Center for Primary Immunodeficiency, Jeffrey Modell Diagnosis and Research Center, ERN-RITA Reference Center, Ghent University Hospital, Ghent, Belgium
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Lynn Backers
- Primary Immunodeficiency Research Lab (PIRL), Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Thorkell Gudjónsson
- Biotech Research and Innovation Centre (BRIC), Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peiquan Huang
- Biotech Research and Innovation Centre (BRIC), Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Leander Jonckheere
- Respiratory Infection and Defense Lab (RIDL), Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Sebastian Riemann
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Bram Parton
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Likun Du
- Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Veronique Debacker
- Primary Immunodeficiency Research Lab (PIRL), Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Center for Primary Immunodeficiency, Jeffrey Modell Diagnosis and Research Center, ERN-RITA Reference Center, Ghent University Hospital, Ghent, Belgium
| | - Marieke De Bruyne
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Levi Hoste
- Primary Immunodeficiency Research Lab (PIRL), Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Department of Pediatrics, Ghent University Hospital, Ghent, Belgium
| | - Ans Baeyens
- Radiobiology Lab, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Anne Vral
- Radiobiology Lab, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Eva Van Braeckel
- Respiratory Infection and Defense Lab (RIDL), Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Jens Staal
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Unit of Molecular Signal Transduction in Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Geert Mortier
- Center for Human Genetics, University Hospitals Leuven, Leuven, Belgium
| | - Tessa Kerre
- Center for Primary Immunodeficiency, Jeffrey Modell Diagnosis and Research Center, ERN-RITA Reference Center, Ghent University Hospital, Ghent, Belgium
- Department of Hematology, Ghent University Hospital, Ghent, Belgium
| | - Qiang Pan-Hammarström
- Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Claus S. Sørensen
- Biotech Research and Innovation Centre (BRIC), Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Filomeen Haerynck
- Primary Immunodeficiency Research Lab (PIRL), Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Center for Primary Immunodeficiency, Jeffrey Modell Diagnosis and Research Center, ERN-RITA Reference Center, Ghent University Hospital, Ghent, Belgium
- Department of Pediatric Respiratory and Infectious Medicine, Ghent University Hospital, Ghent, Belgium
| | - Kathleen B.M. Claes
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Simon J. Tavernier
- Primary Immunodeficiency Research Lab (PIRL), Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Center for Primary Immunodeficiency, Jeffrey Modell Diagnosis and Research Center, ERN-RITA Reference Center, Ghent University Hospital, Ghent, Belgium
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Unit of Molecular Signal Transduction in Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| |
Collapse
|
2
|
Manders TR, Tan CA, Kobayashi Y, Wahl A, Araya C, Colavin A, Facio FM, Metz H, Reuter J, Frésard L, Padigepati SR, Stafford DA, Nussbaum RL, Nykamp K. Harnessing genotype and phenotype data for population-scale variant classification using large language models and bayesian inference. Hum Genet 2025:10.1007/s00439-025-02743-z. [PMID: 40266329 DOI: 10.1007/s00439-025-02743-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 03/31/2025] [Indexed: 04/24/2025]
Abstract
Variants of Uncertain Significance (VUS) in genetic testing for hereditary diseases burden patients and clinicians, yet clinical data that could reduce VUS are underutilized due to a lack of scalable strategies. We assessed whether a machine learning approach using genotype and phenotype data could improve variant classification and reduce VUS. In this cohort study of a multi-step machine learning approach, patient data from test requisition forms were used to distinguish patients with molecular diagnoses from controls ("patient score"). A generative Bayesian model then used patient scores and variant classifications to infer variant pathogenicity ("variant score"). The study included 3.5 million patients referred for clinical genetic testing across various conditions. Primary outcomes were model- and gene-level discrimination, classification performance, probabilistic calibration, and concordance with orthogonal pathogenicity measures. Integration into a semi-quantitative classification framework was based on posterior pathogenicity probabilities matching PPV ≥ 0.99/NPV ≥ 0.95 thresholds, followed by expert review. We generated 1,334 clinical variant models (CVMs); 595 showed high performance in both machine learning steps (AUROCpatient ≥ 0.8 and AUROCvariant ≥ 0.8) on held-out data. High-confidence predictions from these CVMs provided evidence for 5,362 VUS observed in 200,174 patients, representing 23.4% of all VUS observations in these genes. In 17 frequently tested genes, CVMs reclassified over 1,000 unique VUS, reducing VUS report rates by 9-49% per condition. In conclusion, a scalable machine learning approach using underutilized clinical data improved variant classification and reduced VUS.
Collapse
Affiliation(s)
- Toby R Manders
- Labcorp Genetics Inc, 1400 16th Street, San Francisco, CA, 94103, USA.
| | - Christopher A Tan
- Labcorp Genetics Inc, 1400 16th Street, San Francisco, CA, 94103, USA
| | - Yuya Kobayashi
- Labcorp Genetics Inc, 1400 16th Street, San Francisco, CA, 94103, USA
| | - Alexander Wahl
- Labcorp Genetics Inc, 1400 16th Street, San Francisco, CA, 94103, USA
| | - Carlos Araya
- Invitae Corporation, 1400 16th Street, San Francisco, CA, 94103, USA
- Tapanti.org, PO Box #727, 836 Anacapa St, Santa Barbara, CA, 93102, USA
| | - Alexandre Colavin
- Invitae Corporation, 1400 16th Street, San Francisco, CA, 94103, USA
- Present Address: Threshold Health Inc, 1638 Myrtle Ave, San Diego, CA, 92103, USA
| | - Flavia M Facio
- Labcorp Genetics Inc, 1400 16th Street, San Francisco, CA, 94103, USA
- Present Address: GeneDx, 205/207 Perry Parkway, Gaitherburg, MD, 20877, USA
| | - Hillery Metz
- Labcorp Genetics Inc, 1400 16th Street, San Francisco, CA, 94103, USA
| | - Jason Reuter
- Labcorp Genetics Inc, 1400 16th Street, San Francisco, CA, 94103, USA
| | - Laure Frésard
- Labcorp Genetics Inc, 1400 16th Street, San Francisco, CA, 94103, USA
| | | | - David A Stafford
- Labcorp Genetics Inc, 1400 16th Street, San Francisco, CA, 94103, USA
| | - Robert L Nussbaum
- Invitae Corporation, 1400 16th Street, San Francisco, CA, 94103, USA
- Present Address: Division of Medical Genetics, Department of Pediatrics, University of California San Francisco, 1825 4th St, San Francisco, CA, 94158, USA
| | - Keith Nykamp
- Labcorp Genetics Inc, 1400 16th Street, San Francisco, CA, 94103, USA
- Present Address: GeneDx, 205/207 Perry Parkway, Gaitherburg, MD, 20877, USA
| |
Collapse
|
3
|
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
|
4
|
Luppino F, Lenz S, Chow CFW, Toth-Petroczy A. Deep learning tools predict variants in disordered regions with lower sensitivity. BMC Genomics 2025; 26:367. [PMID: 40221640 PMCID: PMC11992697 DOI: 10.1186/s12864-025-11534-9] [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: 01/14/2025] [Accepted: 03/27/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND The recent AI breakthrough of AlphaFold2 has revolutionized 3D protein structural modeling, proving crucial for protein design and variant effects prediction. However, intrinsically disordered regions-known for their lack of well-defined structure and lower sequence conservation-often yield low-confidence models. The latest Variant Effect Predictor (VEP), AlphaMissense, leverages AlphaFold2 models, achieving over 90% sensitivity and specificity in predicting variant effects. However, the effectiveness of tools for variants in disordered regions, which account for 30% of the human proteome, remains unclear. RESULTS In this study, we found that predicting pathogenicity for variants in disordered regions is less accurate than in ordered regions, particularly for mutations at the first N-Methionine site. Investigations into the efficacy of variant effect predictors on intrinsically disordered regions (IDRs) indicated that mutations in IDRs are predicted with lower sensitivity and the gap between sensitivity and specificity is largest in disordered regions, especially for AlphaMissense and VARITY. CONCLUSIONS The prevalence of IDRs within the human proteome, coupled with the increasing repertoire of biological functions they are known to perform, necessitated an investigation into the efficacy of state-of-the-art VEPs on such regions. This analysis revealed their consistently reduced sensitivity and differing prediction performance profile to ordered regions, indicating that new IDR-specific features and paradigms are needed to accurately classify disease mutations within those regions.
Collapse
Affiliation(s)
- Federica Luppino
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307, Dresden, Germany
- Center for Systems Biology Dresden, Pfotenhauerstrasse 108, 01307, Dresden, Germany
| | - Swantje Lenz
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307, Dresden, Germany
- Center for Systems Biology Dresden, Pfotenhauerstrasse 108, 01307, Dresden, Germany
| | - Chi Fung Willis Chow
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307, Dresden, Germany
- Center for Systems Biology Dresden, Pfotenhauerstrasse 108, 01307, Dresden, Germany
- Cluster of Excellence Physics of Life, TU Dresden, 01062, Dresden, Germany
| | - Agnes Toth-Petroczy
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307, Dresden, Germany.
- Center for Systems Biology Dresden, Pfotenhauerstrasse 108, 01307, Dresden, Germany.
- Cluster of Excellence Physics of Life, TU Dresden, 01062, Dresden, Germany.
| |
Collapse
|
5
|
Osler TS, Schoeman M, Pretorius WJS, Mathew CG, Edge J, Urban MF. Application of genetic testing criteria for hereditary breast cancer in South Africa. Breast Cancer Res Treat 2025; 210:477-486. [PMID: 39776011 PMCID: PMC11930871 DOI: 10.1007/s10549-024-07585-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025]
Abstract
PURPOSE Breast cancer (BC) is the commonest cancer in South African women. A proportion are associated with a pathogenic or likely pathogenic (P/LP) variant in a BC susceptibility gene. Clinical guidelines for genetic testing are used to optimise variant detection while containing costs. We assessed the detection rate in women of diverse ancestries who met the South African National Department of Health (NDOH) testing guidelines, and analysed relationships between testing criteria, participant characteristics and presence of a BRCA1/2 P/LP variant. METHODS Records from 376 women with BC who met NDOH criteria and had genetic testing were included. Demographic, clinical and test result data were collated to describe detection rates according to criteria met, and a multivariate analysis conducted to find variables most frequently associated with a P/LP variant. RESULTS P/LP variant prevalence in women meeting NDOH testing criteria was 19.9% (75/376). Women meeting ≥ 2 guideline criteria were over twice as likely to have a P/LP variant (OR 2.27, 95%CI 1.27-4.07, p = 0.006), highlighting the guidelines' capacity to stratify risk. Family history (OR 1.97; 95%CI 1.05-3.70, p = 0.03) and Black African ancestry (OR 2.58; 95%CI 1.28-5.18, p < 0.01) were independently associated with having a BRCA1/2 P/LP variant when controlling for other variables. Notably, although Black African participants were less likely to report a family history, those that did had higher odds of a P/LP variant in BRCA1/2. CONCLUSION These results demonstrate the usefulness of the NDOH guidelines in women of diverse ancestries and provide insight into the factors associated with P/LP variants in understudied African populations.
Collapse
Affiliation(s)
- T S Osler
- Sydney Brenner Institute for Molecular Bioscience and Division of Human Genetics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
- Division of Human Genetics, Faculty of Health Sciences, National Health Laboratory Service and University of the Witwatersrand, Johannesburg, South Africa.
| | - M Schoeman
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, University of Stellenbosch and Tygerberg Hospital, Parow, South Africa
| | - W J S Pretorius
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, University of Stellenbosch and Tygerberg Hospital, Parow, South Africa
| | - C G Mathew
- Sydney Brenner Institute for Molecular Bioscience and Division of Human Genetics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Division of Human Genetics, Faculty of Health Sciences, National Health Laboratory Service and University of the Witwatersrand, Johannesburg, South Africa
| | - J Edge
- Department of Surgery, Charlotte Maxeke Johannesburg Academic Hospital and University of the Witwatersrand, Johannesburg, South Africa
| | - M F Urban
- Division of Human Genetics, Faculty of Health Sciences, National Health Laboratory Service and University of the Witwatersrand, Johannesburg, South Africa
- Department of Obstetrics and Gynaecology, Faculty of Medicine and Health Sciences, University of Stellenbosch and Tygerberg Hospital, Parow, South Africa
| |
Collapse
|
6
|
Lei N, Qiu X, Li W, Liu Y, Hu R, Li C, Lu W. Functional study of three cases with novel TBX19 variants. Endocrine 2025; 88:273-284. [PMID: 39776042 PMCID: PMC11933146 DOI: 10.1007/s12020-024-04153-z] [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: 06/12/2024] [Accepted: 12/25/2024] [Indexed: 01/11/2025]
Abstract
PURPOSE Congenital isolated adrenocorticotropic hormone deficiency (CIAD) is an autosomal recessive disorder. This study identifies novel TBX19 variants for CIAD patients, explores its possible effect mechanism at the structural, functional and protein levels, and guides clinicians better understand the condition. METHODS The clinical characteristics of three CIAD children were summarized. Multiple sequence alignment was performed and five algorithms, PROVEA, PolyPhen2, Mutation Taster, FATHMM, and I Mutant2.0, were used for the pathogenicity prediction. In addition, the three-dimensional protein structure of wild-type TBX19 was generated by Alphafold 3 and its variants were shown using PyMOL. Furthermore, immunoblotting analysis was applied to examine changes in the protein levels and the luciferase reporter assay was performed to further investigate the effects of TBX19 and its variants on pro-opiomelanocortin (POMC) transcriptional activity. RESULTS We describe three Chinese patients with CIAD caused by TBX19 variants. The TBX19 variant, c.856C>T (p.R286*) was classified as pathogenic according to ACMG, whereas the other four variants, c.377C>T (p.P126L), c.602A>T (p.E201V), c.401A>G (p.H134R) and c.299G>A (p.R100H) were predicted to be disease-causing. Variants lead to alter interactions, conformational changes in proteins or truncate protein. TBX19 and PITX1 cooperated, resulting in a strong synergistic activation effect on POMC transcriptional expression. A functional study showed that the variants in our study result in a significant suppression of POMC transcriptional activity compared to wild-type TBX19. CONCLUSIONS Our study identifies five TBX19 loss-of-function variants, two of which are novel and that provides new perspectives into the pathophysiological mechanism and expands the variant spectrum in IAD.
Collapse
Affiliation(s)
- NokI Lei
- Department of Pediatrics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Xiang Qiu
- School of Medicine, Guizhou University, Guiyang, Guizhou, China
| | - Wunying Li
- Department of Pediatrics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Yanlin Liu
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ronggui Hu
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Chuanyin Li
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Wenli Lu
- Department of Pediatrics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
7
|
Khandakar B, Lacy J, Gibson JA. Mismatch Repair Proficient Colorectal Adenocarcinoma in Two Patients With Lynch Syndrome. Clin Genet 2025; 107:469-474. [PMID: 39660603 DOI: 10.1111/cge.14670] [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: 08/16/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/12/2024]
Abstract
Screening for Lynch syndrome (LS) is essential in colorectal carcinoma (CRC) diagnosis. The hallmark of CRC in LS is mismatch repair (MMR) deficiency, a vital biomarkers assessed by microsatellite instability (MSI) analysis and/or immunohistochemistry (IHC) staining of the MMR proteins in the tumor, that also predict response to immune checkpoint inhibitors. We report two LS patients who developed MMR proficient CRCs. Patient A, with a pathogenic MSH6 germline variant, presented with two MMR discordant CRCs: a rectal MMRd/MSI adenocarcinoma, and a sigmoid MMR proficient (MMRp) and microsatellite stable (MSS) adenocarcinoma, leading to metastasis. While the MMRd/MSI carcinoma was recognized early and showed complete pathologic response after pembrolizumab treatment, the MMRp/MSS adenocarcinoma was underrecognized and poorly responsive to treatment. A second patient, with a pathogenic PMS2 variant, also developed a MMRp CRC. These cases highlight the complex biological pathways in CRC development and the impact of molecular classification on treatment.
Collapse
Affiliation(s)
- Binny Khandakar
- Department of Pathology and Laboratory Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New York, New York, USA
| | - Jill Lacy
- Department of Internal Medicine, Medical Oncology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Joanna A Gibson
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
8
|
Vatta M, Regalado E, Parfenov M, Swartzlander D, Nagl A, Mannello M, Lewis R, Clemens D, Garcia J, Ellsworth RE, Morales A, Ting YL, Aradhya S. Analysis of TTN Truncating Variants in >74 000 Cases Reveals New Clinically Relevant Gene Regions. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2025; 18:e004982. [PMID: 39968638 PMCID: PMC11999099 DOI: 10.1161/circgen.124.004982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 12/20/2024] [Indexed: 02/20/2025]
Abstract
BACKGROUND Truncating variants (TTNtvs) in the titin (TTN) gene have been associated with cardiomyopathies or arrhythmias (C/A) and autosomal recessive neuromuscular diseases (NM). However, the clinical significance of TTNtvs across the entire coding sequence of TTN has not been comprehensively assessed. The purpose of this study was to examine the burden of TTNtvs in C/A and NM cases compared with controls in the genome aggregation database. METHODS This was a retrospective study of probands who underwent multigene testing (49 740 C/A panel, 24 514 NM panel) that included TTN from November 2017 to October 2021. Burden testing was performed using controls in the genome aggregation database v3.1.2 database, and the analysis was stratified by exon/band location and exon usage in cardiac or skeletal muscle. Frequency and odds ratio of TTNtv alleles in C/A or NM cases and genome aggregation database controls were measured. RESULTS There were 2446 (4.9%) C/A and 482 (2.0%) NM cases with 2446 and 528 TTNtv alleles, respectively. TTNtvs in all bands were significantly enriched in both C/A and NM cases compared with controls. A significant enrichment of TTNtvs in C/A was observed for exon 358 of the M-band (odds ratio, 2.55 [95% CI, 1.85-3.54]) but not the other M-band exons. CONCLUSIONS In the largest single-site cohort of C/A and NM cases with TTNtvs, an enrichment of TTNtvs across TTN was observed. These findings expand the clinically relevant regions of TTN.
Collapse
Affiliation(s)
- Matteo Vatta
- Labcorp Genetics Inc (formerly Invitae Corporation), San Francisco, CA (M.V., E.R., M.P., D.S., A.N., M.M., R.L., D.C., J.G., R.E.E., A.M., Y.-L.T., S.A.)
| | - Ellen Regalado
- Labcorp Genetics Inc (formerly Invitae Corporation), San Francisco, CA (M.V., E.R., M.P., D.S., A.N., M.M., R.L., D.C., J.G., R.E.E., A.M., Y.-L.T., S.A.)
| | - Michael Parfenov
- Labcorp Genetics Inc (formerly Invitae Corporation), San Francisco, CA (M.V., E.R., M.P., D.S., A.N., M.M., R.L., D.C., J.G., R.E.E., A.M., Y.-L.T., S.A.)
| | - Dan Swartzlander
- Labcorp Genetics Inc (formerly Invitae Corporation), San Francisco, CA (M.V., E.R., M.P., D.S., A.N., M.M., R.L., D.C., J.G., R.E.E., A.M., Y.-L.T., S.A.)
| | - Andrea Nagl
- Labcorp Genetics Inc (formerly Invitae Corporation), San Francisco, CA (M.V., E.R., M.P., D.S., A.N., M.M., R.L., D.C., J.G., R.E.E., A.M., Y.-L.T., S.A.)
| | - Meghan Mannello
- Labcorp Genetics Inc (formerly Invitae Corporation), San Francisco, CA (M.V., E.R., M.P., D.S., A.N., M.M., R.L., D.C., J.G., R.E.E., A.M., Y.-L.T., S.A.)
| | - Rachel Lewis
- Labcorp Genetics Inc (formerly Invitae Corporation), San Francisco, CA (M.V., E.R., M.P., D.S., A.N., M.M., R.L., D.C., J.G., R.E.E., A.M., Y.-L.T., S.A.)
| | - Daniel Clemens
- Labcorp Genetics Inc (formerly Invitae Corporation), San Francisco, CA (M.V., E.R., M.P., D.S., A.N., M.M., R.L., D.C., J.G., R.E.E., A.M., Y.-L.T., S.A.)
| | - John Garcia
- Labcorp Genetics Inc (formerly Invitae Corporation), San Francisco, CA (M.V., E.R., M.P., D.S., A.N., M.M., R.L., D.C., J.G., R.E.E., A.M., Y.-L.T., S.A.)
| | - Rachel E. Ellsworth
- Labcorp Genetics Inc (formerly Invitae Corporation), San Francisco, CA (M.V., E.R., M.P., D.S., A.N., M.M., R.L., D.C., J.G., R.E.E., A.M., Y.-L.T., S.A.)
| | - Ana Morales
- Labcorp Genetics Inc (formerly Invitae Corporation), San Francisco, CA (M.V., E.R., M.P., D.S., A.N., M.M., R.L., D.C., J.G., R.E.E., A.M., Y.-L.T., S.A.)
| | - Yi-Lee Ting
- Labcorp Genetics Inc (formerly Invitae Corporation), San Francisco, CA (M.V., E.R., M.P., D.S., A.N., M.M., R.L., D.C., J.G., R.E.E., A.M., Y.-L.T., S.A.)
| | - Swaroop Aradhya
- Labcorp Genetics Inc (formerly Invitae Corporation), San Francisco, CA (M.V., E.R., M.P., D.S., A.N., M.M., R.L., D.C., J.G., R.E.E., A.M., Y.-L.T., S.A.)
- Department of Pathology, Stanford University School of Medicine, CA (S.A.)
| |
Collapse
|
9
|
Talarico M, de Bellescize J, De Wachter M, Le Guillou X, Le Meur G, Egloff M, Isidor B, Cogné B, Beysen D, Rollier P, Fradin M, Pasquier L, Guella I, Hickey SE, Benke PJ, Shillington A, Kumps C, Vanakker O, Gerkes EH, Lakhani S, Romanova I, Kanivets I, Brugger M, Vill K, Caylor RC, Skinner C, Tinker RJ, Stödberg T, Nümann A, Haack TB, Deininger N, Hengel H, Jury J, Conrad S, Mercier S, Yoon G, Tsuboyama M, Barcia G, Gitiaux C, Rio M, Bevot A, Redon S, Uguen K, Wonneberger A, Schulz A, Timmann D, Karlowicz DH, Chatron N, Carnevale A, Mahida S, Õunap K, Kury S, Cabet S, Lesca G. RORA-neurodevelopmental disorder: A unique triad of developmental disabilities, cerebellar anomalies, and myoclonic seizures. Genet Med 2025; 27:101347. [PMID: 39707840 DOI: 10.1016/j.gim.2024.101347] [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: 06/28/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 12/23/2024] Open
Abstract
PURPOSE RORA encodes the RAR-related orphan receptor-α, playing a pivotal role in cerebellar maturation and function. Here, we report the largest series of individuals with RORA-related-neurodevelopmental disorder. METHODS Forty individuals (30 unrelated; 10 siblings from 4 families) carrying RORA pathogenic/likely pathogenic variants were collected through an international collaboration. RESULTS The 33 variants (29 de novo, 4 inherited, and 1 shared), identified by genome/exome sequencing (n = 21), chromosomal microarray analysis (n = 7), or gene panels (n = 4), included frameshift (n = 18/33), missense (n = 9/33), and stop codon (n = 6/33). Developmental disability (n = 32/37), intellectual disability (n = 22/32), and cerebellar signs (n = 25/34) were the most striking clinical features. Cerebellar symptoms were divided into early-onset, late-onset, and progressive subgroups. Cerebellar hypoplasia, atrophy, or both (n = 16/25) were more frequent in individuals with missense variants in the DNA-binding domain. Epilepsy (n = 18/38), with prominent myoclonic seizure types (n = 11/18), was classified in (1) genetic generalized epilepsy (n = 10/18) with a syndromic diagnosis identifiable for 6: epilepsy with eyelid myoclonia (n = 5/6) and epilepsy with myoclonic absence (n = 1/6); (2) developmental and epileptic encephalopathy (n = 5/18); and (3) unclassified (n = 3/18). A participant with rapid deterioration of visual acuity and cone/rod dystrophy was reported. CONCLUSION Missense variants in DNA-binding domain correlate to a more severe cerebellar phenotype. The RORA-related-neurodevelopmental disorder triad comprises developmental disability, cerebellar features, and a spectrum of myoclonic epilepsy.
Collapse
Affiliation(s)
- Mariagrazia Talarico
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy; Genetics Department, Hospices Civils de Lyon, Member of the ERN EpiCARE, Lyon, France; Neuromyogene Institute, Pathology and Genetics of neuron and muscle, CNRS UMR 5261 INSERM U1315, University of Lyon-Université Claude Bernard Lyon 1, Lyon, France
| | - Julitta de Bellescize
- Department of Clinical Epileptology, Sleep Disorders and Functional Neurology in Children, University Hospital of Lyon (HCL), Member of the ERN EpiCARE, Lyon, France
| | - Matthias De Wachter
- Department of Pediatric Neurology, Antwerp University Hospital, University of Antwerp, - Edegem, Belgium
| | - Xavier Le Guillou
- Poitiers University Hospital, Medical Genetics Department, Poitiers, France; Poitiers University, INSERM U1084, LNEC, Poitiers, France
| | - Guylène Le Meur
- Nantes University, ophthalmology department Nantes University Hospital, Inserm, TARGET, Nantes, France
| | - Matthieu Egloff
- Poitiers University Hospital, Medical Genetics Department, Poitiers, France; Poitiers University, INSERM U1084, LNEC, Poitiers, France
| | - Bertrand Isidor
- Medical Genetics Department, Nantes University, CHU Nantes, Nantes, France; The Thorax Institute, INSERM, CNRS, Nantes University, CHU Nantes, Nantes, France
| | - Benjamin Cogné
- Medical Genetics Department, Nantes University, CHU Nantes, Nantes, France; The Thorax Institute, INSERM, CNRS, Nantes University, CHU Nantes, Nantes, France
| | - Diane Beysen
- Department of Pediatric Neurology, Antwerp University Hospital, University of Antwerp, - Edegem, Belgium
| | - Paul Rollier
- Clinical Genetics Department, "Intellectual Deficiencies of Rare Causes" Reference Center (CRDI), CLAD-Ouest Developmental Abnormalities Reference Center, CHU Rennes, Rennes, France
| | - Melanie Fradin
- Clinical Genetics Department, "Intellectual Deficiencies of Rare Causes" Reference Center (CRDI), CLAD-Ouest Developmental Abnormalities Reference Center, CHU Rennes, Rennes, France
| | - Laurent Pasquier
- Clinical Genetics Department, "Intellectual Deficiencies of Rare Causes" Reference Center (CRDI), CLAD-Ouest Developmental Abnormalities Reference Center, CHU Rennes, Rennes, France
| | - Ilaria Guella
- Centre for Applied Neurogenetics, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Scott E Hickey
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH
| | | | - Amelle Shillington
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Candy Kumps
- Center for Medical Genetics, Ghent University Hospital, Belgium
| | | | - Erica H Gerkes
- Clinical Genetics, University Medical Center Groningen, Groningen, The Netherlands
| | - Shenela Lakhani
- Center for Neurogenetics, Weill Cornell Medicine, Brain and Mind Research Institute, New York, NY
| | - Irina Romanova
- Department of Medical Genetics, Kazan State University, Russia; GENOMED medical-genetic center, Moscow, Russia
| | - Ilya Kanivets
- Department of Neurology, Svt. Luka's Institute of Child Neurology and Epilepsy, Moscow, Russia
| | - Melanie Brugger
- Institute of Human Genetics, School of Medicine and Health, Technical University of Munich, Munich, German; Department of Obstetrics and Gynecology, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Katharina Vill
- Institute of Human Genetics, School of Medicine and Health, Technical University of Munich, Munich, German; Department of Pediatric Neurology and Developmental Medicine and LMU Center for Children with Medical Complexity, Children's Hospital, LMU Hospital, Ludwig-Maximilians-University, Munich, Germany
| | | | | | - Rory J Tinker
- Division of Medical Genetics and Genomic Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Tommy Stödberg
- Pediatric Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Astrid Nümann
- Department of Neurology, Charity University Medicine Berlin, Berlin, Germany
| | - Tobias B Haack
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Natalie Deininger
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Holger Hengel
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany; Center for Neurology and Hertie-Institute for Clinical Brain Research, Tübingen
| | - Jeanne Jury
- Medical Genetics Department, Nantes University, CHU Nantes, Nantes, France; The Thorax Institute, INSERM, CNRS, Nantes University, CHU Nantes, Nantes, France
| | - Solène Conrad
- Medical Genetics Department, Nantes University, CHU Nantes, Nantes, France; The Thorax Institute, INSERM, CNRS, Nantes University, CHU Nantes, Nantes, France
| | - Sandra Mercier
- Medical Genetics Department, Nantes University, CHU Nantes, Nantes, France; The Thorax Institute, INSERM, CNRS, Nantes University, CHU Nantes, Nantes, France
| | - Grace Yoon
- Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Melissa Tsuboyama
- Boston Children's Hospital, department of neurology, Boston Children's Hospital, Boston, MA
| | - Giulia Barcia
- Federation of Genetics, Molecular Genetics Department, Necker-Enfants Malades Hospital, Paris, France
| | - Cyril Gitiaux
- Federation of Genetics, Molecular Genetics Department, Necker-Enfants Malades Hospital, Paris, France
| | - Marlène Rio
- Federation of Genetics, Molecular Genetics Department, Necker-Enfants Malades Hospital, Paris, France
| | - Andrea Bevot
- Neuropediatrics, University Children's Hospital Tübingen, Tübingen, Germany
| | - Sylvia Redon
- Medical Genetics Department, Brest University Hospital, Brest, France; Reference Center for Intellectual Disabilities, Pediatric Department, Brest University Hospital, Brest, France; University of Brest, Inserm, EFS, UMR 1078, GGB, Brest, France
| | - Kevin Uguen
- Medical Genetics Department, Brest University Hospital, Brest, France; Reference Center for Intellectual Disabilities, Pediatric Department, Brest University Hospital, Brest, France; University of Brest, Inserm, EFS, UMR 1078, GGB, Brest, France
| | - Antje Wonneberger
- Department of Neuropediatrics, Jena University Hospital, Jena, Germany
| | - Alexander Schulz
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
| | - Dagmar Timmann
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Essen University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Danielle Hays Karlowicz
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC
| | - Nicolas Chatron
- Genetics Department, Hospices Civils de Lyon, Member of the ERN EpiCARE, Lyon, France; Neuromyogene Institute, Pathology and Genetics of neuron and muscle, CNRS UMR 5261 INSERM U1315, University of Lyon-Université Claude Bernard Lyon 1, Lyon, France
| | - Amanda Carnevale
- Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Sonal Mahida
- Boston Children's Hospital, department of neurology, Boston Children's Hospital, Boston, MA
| | - Katrin Õunap
- Department of Clinical Genetics, Genetics and Personalized Medicine Clinic, Tartu University Hospital, Tartu, Estonia; Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Sébastien Kury
- Medical Genetics Department, Nantes University, CHU Nantes, Nantes, France; The Thorax Institute, INSERM, CNRS, Nantes University, CHU Nantes, Nantes, France
| | - Sara Cabet
- Neuromyogene Institute, Pathology and Genetics of neuron and muscle, CNRS UMR 5261 INSERM U1315, University of Lyon-Université Claude Bernard Lyon 1, Lyon, France; Pediatric imaging department, Femme-Mere-Enfant hospital, Lyon 1 University, Lyon, France
| | - Gaetan Lesca
- Genetics Department, Hospices Civils de Lyon, Member of the ERN EpiCARE, Lyon, France; Neuromyogene Institute, Pathology and Genetics of neuron and muscle, CNRS UMR 5261 INSERM U1315, University of Lyon-Université Claude Bernard Lyon 1, Lyon, France.
| |
Collapse
|
10
|
Pentz R, Hough R, Li C, Tarnopolsky M, Jones K, RamachandranNair R, Whitney R. Biallelic SCN1A variants with divergent epilepsy phenotypes. Seizure 2025; 127:88-93. [PMID: 40120363 DOI: 10.1016/j.seizure.2025.03.009] [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: 11/19/2024] [Revised: 03/09/2025] [Accepted: 03/12/2025] [Indexed: 03/25/2025] Open
Abstract
PURPOSE Pathogenic SCN1A variants most commonly cause autosomal dominant Dravet syndrome and genetic epilepsy with febrile seizures plus (GEFS+). However, rare homozygous SCN1A variants have also been reported. We report two new cases of homozygous SCN1A variants associated with divergent epilepsy phenotypes. METHODS We retrospectively reviewed the charts of two unrelated patients with different homozygous SCN1A variants. We also reviewed all published cases of biallelic SCN1A pathogenic variants, focusing on the epilepsy phenotypes. RESULTS Patient 1 had a homozygous c. 1676T>A, (p. Ile559Asn) variant of uncertain significance, inherited from asymptomatic parents. Patient 1 exhibited early afebrile seizures controlled by first-line anti-seizure medications and no febrile seizures or status epilepticus, as well as profound developmental delay, macrocephaly, and mild dysmorphic features. Patient 2 had a homozygous pathogenic c. 4970G>A, (p. Arg1657His) variant carried by asymptomatic parents. This patient presented with early, recurrent, and prolonged febrile seizures, moderate developmental delay, and motor dysfunction and was diagnosed with Dravet syndrome. We identified 16 further cases from the literature. Including our cases, 9/18 (50 %) were diagnosed with Dravet syndrome and 6/18 (33 %) with GEFS+. The mean age of seizure onset was 7 months (range 3-19 months). Phenotypes ranged from intact neurodevelopment with controlled epilepsy to profound developmental delay and refractory epilepsy. CONCLUSION These cases highlight and expand the phenotypic spectrum associated with biallelic SCN1A variants. While some patients present typically for Dravet/GEFS+, others may present with developmental delay in the absence of febrile seizures or status epilepticus. Further studies are needed to confirm genotype-phenotype relationships.
Collapse
Affiliation(s)
- Rowan Pentz
- The Division of Neurology, Department of Pediatrics, McMaster University, Hamilton, ON, Canada
| | - Rebecca Hough
- The Division of Neurology, Department of Pediatrics, McMaster University, Hamilton, ON, Canada; The Division of Genetics and Metabolics, Department of Pediatrics, McMaster University, Hamilton, Canada
| | - Chumei Li
- The Division of Genetics and Metabolics, Department of Pediatrics, McMaster University, Hamilton, Canada
| | - Mark Tarnopolsky
- The Division of Neuromuscular and Neurometabolic Disease, Department of Pediatrics, McMaster University, Hamilton, Canada
| | - Kevin Jones
- The Division of Neurology, Department of Pediatrics, McMaster University, Hamilton, ON, Canada
| | - Rajesh RamachandranNair
- The Division of Neurology, Department of Pediatrics, McMaster University, Hamilton, ON, Canada
| | - Robyn Whitney
- The Division of Neurology, Department of Pediatrics, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
11
|
Frees M, Carter JN, Wheeler MT, Reuter C. The current landscape of clinical exome and genome reanalysis in the U.S. J Genet Couns 2025; 34:e1968. [PMID: 39285507 DOI: 10.1002/jgc4.1968] [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: 04/08/2024] [Revised: 08/21/2024] [Accepted: 08/26/2024] [Indexed: 03/30/2025]
Abstract
The majority of patients undergoing exome or genome sequencing receive a nondiagnostic result. Periodic reanalysis is known to increase diagnostic yield from exome sequencing, yet laboratory reanalysis practices are obscure. We sought to define the landscape of exome and genome reanalysis across clinical laboratories. Genetic testing registries were queried to identify eligible clinical genetic laboratories offering exome and/or genome sequencing in the United States. A survey administered to lab representatives investigated reanalysis offerings, policies, perceived uptake, bioinformatic steps, and billing options. The analysis consisted of descriptive statistics. Survey data were collected from 30 of 32 eligible laboratories (93%), comprising 28 exome products and 13 genome products. Reanalysis was widely available for both exomes (n = 27/28, 96%) and genomes (n = 12/13, 92%). Most participating laboratories required ordering providers to initiate reanalysis (n = 24/28, 86%). Most respondents estimated providers initiated reanalysis in less than 10% of all exomes (n = 12/22) or genomes (n = 6/9) sequenced. The approach to reanalysis varied greatly by laboratory. Laboratory approaches to exome and genome reanalysis are highly variable and typically require provider initiation. This could contribute to low reanalysis uptake and increased administrative burden on providers. Further work should emphasize development of clinical exome and genome reanalysis standards.
Collapse
Affiliation(s)
- Michelle Frees
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
- Division of Genomic Medicine, UC Davis Medical Center, Sacramento, California, USA
| | - Jennefer N Carter
- Center for Undiagnosed Diseases, Stanford University, Stanford, California, USA
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
- Center for Inherited Cardiovascular Diseases, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Matthew T Wheeler
- Center for Undiagnosed Diseases, Stanford University, Stanford, California, USA
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
- Center for Inherited Cardiovascular Diseases, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Chloe Reuter
- Center for Undiagnosed Diseases, Stanford University, Stanford, California, USA
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
- Center for Inherited Cardiovascular Diseases, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
12
|
Huang S, Chen J, Wang Q, Zhang R, Zhuang J, Huang R, Yu C, Fang M, Zhao H, Lei L. Identification and functional validation of a novel FBN1 variant in a Marfan syndrome family using a zebrafish model. BMC Genomics 2025; 26:288. [PMID: 40128660 PMCID: PMC11931800 DOI: 10.1186/s12864-025-11471-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 03/11/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Marfan syndrome (MFS) is an inherited autosomal dominant disorder that affects connective tissue with an incidence of about 1 in 5,000 to 10,000 people. 90% of MFS is caused by mutations in the fibrillin-1 (FBN1) gene. We recruited a family with MFS phenotype in South China and identified a novel variant. This study investigated whether this genetic variant is pathogenic and the potential pathway related to lipid metabolism in MFS. METHODS A three-generation consanguineous family was recruited for this study. Whole exome sequencing (WES) was utilized on family members. The 3D structure of the protein was predicted using AlphaFold. CRISPR/Cas9 was applied to generate a similar fbn1 nonsense mutation (fbn1+/-) in zebrafish. RNA-seq analysis on zebrafish was performed to identify potential pathways related to MFS pathogenesis. RESULTS Our study identified a novel variant [NM_000138.5; c.7764 C > G: p.(Y2588*)] in FBN1 gene from the family and identified the same site mutation among the proband along with her son and daughter. Structural modeling showed the p.Y2588* mutation resulted from a truncated protein. Compared to wild-type zebrafish, the F2 generation fbn1+/- zebrafish exhibited MFS phenotype. RNA-seq analysis indicated that many genes related to leptin are up-regulating, which could affect bone development and adipose homeostasis. CONCLUSION A novel variant was identified in FBN1 gene. In a zebrafish model, we found functional evidence supporting the pathogenicity of the detected nonsense mutation. Our research proposes a possible mechanism underlying the relationship between lipid metabolism and MFS. These findings can help improve the clinical diagnosis and treatment of MFS.
Collapse
Affiliation(s)
- Shitong Huang
- Department of Cardiac Surgical Intensive Care Unit, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Department of Cardiovascular Surgery, Guangdong Provincial People's Hospital's Nanhai Hospital, The Second People's Hospital of Nanhai District Foshan City, Foshan, Guangdong, 528200, China
| | - Jiansong Chen
- Department of Cardiovascular Surgery, Guangdong Provincial People's Hospital's Nanhai Hospital, The Second People's Hospital of Nanhai District Foshan City, Foshan, Guangdong, 528200, China
| | - Qiuyu Wang
- Department of Cardiac Surgical Intensive Care Unit, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Ruyue Zhang
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Jian Zhuang
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Ruiyuan Huang
- Department of Cardiac Surgical Intensive Care Unit, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Changjiang Yu
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Miaoxian Fang
- Department of Cardiac Surgical Intensive Care Unit, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Haishan Zhao
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| | - Liming Lei
- Department of Cardiac Surgical Intensive Care Unit, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
- Department of Cardiovascular Surgery, Guangdong Provincial People's Hospital's Nanhai Hospital, The Second People's Hospital of Nanhai District Foshan City, Foshan, Guangdong, 528200, China.
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| |
Collapse
|
13
|
Chang Y, Liu L, Cui C, He J, Li C, Jia Y, Zhang R, Wu W, Zhou J, Xiao J, Xu Z, Qin T, Sun Q, Wang H, Xiao Z. Impact of ASXL1 Gene Alterations on Myelodysplastic Syndrome With Isolated 20q Deletion. Cancer Med 2025; 14:e70747. [PMID: 40047093 PMCID: PMC11883421 DOI: 10.1002/cam4.70747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 02/11/2025] [Accepted: 02/25/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Isolated 20q deletion [del(20q)] is a recurrent favorable abnormality in myelodysplastic syndrome (MDS) and may cause deletion of the ASXL1 gene. Meanwhile, ASXL1 mutations are also common in individuals with MDS. This study aimed to describe the biological and clinical implications of ASXL1 mutations and deletion in newly diagnosed MDS patients with isolated del(20q). METHODS Gene mutation and copy number alterations in 178 newly diagnosed MDS patients with isolated del(20q) were analyzed using DNA next generation sequencing. RESULTS Twenty-five (14%) of 178 patients were found to have ASXL1 mutations, which exhibited lower absolute neutrophil counts (ANC) (p = 0.006), a higher percentage of bone marrow blasts (p = 0.001), more mutant genes (p < 0.001), higher IPSS-R (p = 0.038) and IPSS-M (p = 0.001) risk groups. Furthermore, ASXL1 mutations were preferentially associated with mutations in U2AF1, and most ASXL1 mutations (68%) were observed as subclonal lesions. ASXL1 frameshift mutations were associated with a worse prognosis in MDS patients with low blasts (MDS-LB) (p = 0.043), but not in those with increased blasts (MDS-IB). Twenty-two (26.8%) of 82 patients were found to have ASXL1 deletion, which exhibited a lower IPSS-M risk group, lower platelet counts, higher ANC levels, and higher hemoglobin levels compared to ASXL1 patientsonly-mut and ASXL1wt patients. Two (2.4%) of the 82 patients exhibited biallelic ASXL1 inactivation (ASXL1mut&del). CONCLUSIONS ASXL1 mutations are one of the late genetic events in MDS patients with isolated 20q deletion, and different types of ASXL1 gene alterations have distinct clinical and biological characteristics.
Collapse
Affiliation(s)
- Yanan Chang
- Hematologic Pathology CenterInstitute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Linlin Liu
- State Key Laboratory of Experimental HematologyInstitute of Hematology & Blood Disease Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Chenghua Cui
- Hematologic Pathology CenterInstitute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Jiange He
- Hand and Foot MicrosurgeryThe Second Hospital of Tianjin Medical UniversityTianjinChina
| | - Chengwen Li
- Hematologic Pathology CenterInstitute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Yujiao Jia
- Hematologic Pathology CenterInstitute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Ruixue Zhang
- Hematologic Pathology CenterInstitute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Wanyun Wu
- Peking University People's HospitalBeijingChina
| | - Ji Zhou
- Hematologic Pathology CenterInstitute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Jigang Xiao
- Hematologic Pathology CenterInstitute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Zefeng Xu
- MDS and MPN CentreInstitute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Tiejun Qin
- MDS and MPN CentreInstitute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Qi Sun
- Hematologic Pathology CenterInstitute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Huijun Wang
- Hematologic Pathology CenterInstitute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Zhijian Xiao
- Hematologic Pathology CenterInstitute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
- State Key Laboratory of Experimental HematologyInstitute of Hematology & Blood Disease Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
- MDS and MPN CentreInstitute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| |
Collapse
|
14
|
Navabi K, Doraghi K. A 59-Year-Old Man with Fatigue and Abdominal Pain. NEJM EVIDENCE 2025; 4:EVIDmr2400067. [PMID: 39998306 DOI: 10.1056/evidmr2400067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
AbstractMorning Report is a time-honored tradition where physicians-in-training present cases to their colleagues and clinical experts to collaboratively examine an interesting patient presentation. The Morning Report section seeks to carry on this tradition by presenting a patient's chief concern and story, inviting the reader to develop a differential diagnosis and discover the diagnosis alongside the authors of the case. This report examines the story of a 59-year-old man with a history of nephrolithiasis and gastroesophageal reflux who sought evaluation for fatigue and abdominal pain. Using questions, physical examination, and testing, an illness script for the presentation emerges. As the clinical course progresses, the differential is iteratively refined until a final diagnosis is made.
Collapse
|
15
|
Narasimhan M, Cao J, Jones PM. Markedly Elevated Citrulline in a Neonate: Citrin Deficiency due to a Previously Unreported Solute Carrier Family 25 Member 13 Variant. Clin Chim Acta 2025; 568:120109. [PMID: 39800183 DOI: 10.1016/j.cca.2024.120109] [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: 11/18/2024] [Revised: 12/11/2024] [Accepted: 12/20/2024] [Indexed: 01/15/2025]
Abstract
BACKGROUND Citrin deficiency (CD) is an autosomal recessive metabolic disorder affecting the urea cycle and energy production. Diagnosis involves measuring ammonia and amino acid levels (eg: citrulline), with confirmation through solute carrier family 25 member 13 (SLC25A13) gene mutation analysis. Herein, we present a case report of a variant in the SLC25A13 gene that has not been previously reported in the literature. CASE REPORT AND RESULTS The subject was a full-term Hispanic girl infant who was provisionally diagnosed with amino aciduria/urea cycle disorder with citrullinemia of unidentified type based on the second newborn screen performed at the 10th day of life. Sequence analysis and deletion/duplication testing using a panel consisting of 5 genes pertaining to citrullinemia revealed the patient carried a frameshift variant in the SLC25A13 gene (c.429_430del; pArg144fs) consistent with elevated citrulline results. The variant is not found in population databases (gnomAD). While ClinVar has only one entry for this variant (Variation ID: 1076508) and classifies it as pathogenic/likely pathogenic, no case report association exists between this variant and citrullinemia/CD or any SLC25A13-related conditions. CONCLUSION This case study expands the CD variant spectrum and describes a frameshift variant in the SLC25A13 gene in a patient linked to pathology. The finding emphasizes the importance of integrating clinical features with biochemical and genetic analysis to better understand genotype-phenotype correlations in CD and improve management strategies.
Collapse
Affiliation(s)
| | - Jing Cao
- Department of Pathology University of Texas Southwestern Medical Center Dallas USA; Chemistry and Metabolic Disease Laboratory Children's Health Children's Medical Center Dallas USA
| | - Patricia M Jones
- Department of Pathology University of Texas Southwestern Medical Center Dallas USA; Chemistry and Metabolic Disease Laboratory Children's Health Children's Medical Center Dallas USA
| |
Collapse
|
16
|
Ng CA, O’Neill MJ, Padigepati SR, Ting YL, Facio FM, Vatta M, Poll SR, Reuter J, Vandenberg JI, Kroncke BM. Calibrated Functional Data Decreases Clinical Uncertainty for Tier 1 Monogenic Disease: Application to Long QT Syndrome. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.05.25321617. [PMID: 39974038 PMCID: PMC11838680 DOI: 10.1101/2025.02.05.25321617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Rare missense variants are often classified as variants of uncertain significance (VUS) due to insufficient evidence for classification. These ambiguous findings create anxiety and frequently lead to inappropriate workup, colloquially referred to as the 'diagnostic odyssey'. Well-validated high-throughput experimental data have the potential to significantly reduce the number of VUS identified by clinical genetic testing, though the extent of this reduction and the optimal strategies to achieve it remain unclear.1.
Collapse
Affiliation(s)
- Chai-Ann Ng
- Mark Cowley Lidwill Research Program in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- School of Clinical Medicine, UNSW Sydney, NSW, Australia
| | | | | | - Yi-Lee Ting
- Labcorp (formerly Invitae Corporation), San Francisco, CA, USA
| | - Flavia M. Facio
- Labcorp (formerly Invitae Corporation), San Francisco, CA, USA
| | - Matteo Vatta
- Labcorp (formerly Invitae Corporation), San Francisco, CA, USA
| | - Sarah R. Poll
- Labcorp (formerly Invitae Corporation), San Francisco, CA, USA
| | - Jason Reuter
- Labcorp (formerly Invitae Corporation), San Francisco, CA, USA
| | - Jamie I. Vandenberg
- Mark Cowley Lidwill Research Program in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- School of Clinical Medicine, UNSW Sydney, NSW, Australia
| | - Brett M. Kroncke
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
17
|
Ruglioni M, Petrini I, Crucitta S, Sbrana A, Luculli GI, Sadeghi Gol L, Forte C, Chella A, Rolfo C, Danesi R, Del Re M. Clinical characteristics of EGFR-ctDNA shedders in EGFR-mutant NSCLC patients. Transl Oncol 2025; 52:102228. [PMID: 39709717 PMCID: PMC11832947 DOI: 10.1016/j.tranon.2024.102228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/17/2024] [Accepted: 11/28/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Circulating tumor DNA (ctDNA) revolutionized the molecular diagnostics of lung cancer by enabling non-invasive, sensitive identification of actionable mutations. However, ctDNA analysis may be challenging due to tumor shedding variability, leading to false negative results. This study aims to understand the determinants for ctDNA shedding based on clinical characteristics of lung cancer patients, for a better interpretation of false negative results to be considered when ordering ctDNA analysis for clinical practice. METHODS Blood samples were collected from patients with stage IV EGFR-mutated (mEGFR) NSCLC before treatment and monitored until disease progression. EGFR was assessed on tissue by standard procedures, while EGFR status on ctDNA was tested using dPCR at baseline and at the first reassessment. NGS was used to evaluate patients mutational status at the progression of the disease. RESULTS A total of 40 mEGFR tissue samples were collected. Plasma samples were analyzed for mEGFR before starting the first line, 65 % of patients had detectable mEGFR in ctDNA ("shedders"). Higher ECOG PS (p = 0.04), bilateral localization of primary tumor (p = 0.04), and the presence of intrathoracic/extrathoracic disease (p = 0.05), were associated to mEGFR shedding. Shedders had shorter PFS compared to non-shedders (p = 0.03). Patients with detectable mEGFR in ctDNA at the first radiological assessment exhibited worse PFS compared to patients with ctDNA clearance (p = 0.05). CONCLUSION Our preliminary data demonstrate that specific clinical characteristics predict mEGFR shedding in ctDNA of NSCLC, suggesting a potential clinical applicability for understanding potential false negative results and appropriate reporting in clinical practice.
Collapse
Affiliation(s)
- Martina Ruglioni
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Iacopo Petrini
- Unit of Pneumology, Department of Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Stefania Crucitta
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Andrea Sbrana
- Unit of Pneumology, Department of Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Giovanna Irene Luculli
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Leila Sadeghi Gol
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Carola Forte
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Antonio Chella
- Unit of Pneumology, Department of Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Christian Rolfo
- Department of Internal Medicine, Division of Medical Oncology, The Arthur G. James Comprehensive Cancer Center, Columbus, OH, USA
| | - Romano Danesi
- Department of Oncology and Hemato-Oncology, University of Milan, Italy.
| | - Marzia Del Re
- Saint Camillus International University of Medical and Health Sciences, Rome, Italy; Direzione Scientifica Fondazione Policlinico A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
18
|
Ben-Ruby D, Atias-Varon D, Kagan M, Chowers G, Shlomovitz O, Slabodnik-Kaner K, Mano N, Avayou S, Atsmony Y, Levin D, Dotan E, Calderon-Margalit R, Shnaider A, Haviv YS, Birk OS, Hadar N, Anikster Y, Berar Yanay N, Chernin G, Kruzel-Davila E, Beckerman P, Rozen-Zvi B, Doctor GT, Stanescu HC, Shemer R, Pras E, Reznik-Wolf H, Nahum AH, Dominissini D, Skorecki K, Vivante A. Multiethnic prevalence of the APOL1 G1 and G2 variants among the Israeli dialysis population. Clin Kidney J 2025; 18:sfae397. [PMID: 39927257 PMCID: PMC11803305 DOI: 10.1093/ckj/sfae397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Indexed: 02/11/2025] Open
Abstract
Background and hypothesis The two apolipoprotein L1 (APOL1) variants, G1 and G2, are common in populations of sub-Saharan African ancestry. Individuals with two of these alleles (G1 or G2) have an increased risk for a spectrum of non-diabetic chronic kidney diseases. However, these variants are typically not observed outside of populations that self-identify as current continental Africans or having clear recent African ancestry such as, most notably, African Americans, and other large population groups in the Americas and several European countries. We hypothesized that the diverse ethnic groups within the Israeli population may exhibit varying levels of recent African ancestry. Therefore, it is plausible that APOL1 risk alleles might be present even in individuals who do not self-identify as being of sub-Saharan African descent. Methods We non-selectively screened people with kidney failure across Israel for APOL1 risk variants using restriction fragment length polymorphism. Results We recruited 1744 individuals from 38 dialysis units in Israel. We identified eight patients of Moroccan Jewish, Bedouin, or Muslim Arab ancestry, who carry at least one G1 or G2 allele. None of the eight patients carried the protective APOL1 p.N264K variant. Furthermore, despite all Bedouin individuals being G2 heterozygous, the G2 minor allele frequency was significantly enriched in kidney failure cases compared to ethnically matched controls (P = .006). Conclusions These findings show that APOL1 G1 and G2 allelic variants are present in populations previously not appreciated to possess recent sub-Saharan ancestry and suggest that a single G2 risk variant may confer increased risk for chronic kidney disease in certain population contexts.
Collapse
Affiliation(s)
- Dror Ben-Ruby
- Genetic Kidney Disease Research Laboratory, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
- Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Danit Atias-Varon
- Genetic Kidney Disease Research Laboratory, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
- Department of Pediatrics B, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Maayan Kagan
- Genetic Kidney Disease Research Laboratory, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
- Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
- Department of Pediatrics B, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Guy Chowers
- Genetic Kidney Disease Research Laboratory, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
- Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
- Department of Pediatrics B, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Omer Shlomovitz
- Genetic Kidney Disease Research Laboratory, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
- Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
- Department of Pediatrics B, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Keren Slabodnik-Kaner
- Genetic Kidney Disease Research Laboratory, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
- Department of Pediatrics B, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Neta Mano
- Genetic Kidney Disease Research Laboratory, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
- Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
- Arrow Project, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Shany Avayou
- Genetic Kidney Disease Research Laboratory, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Yariv Atsmony
- Genetic Kidney Disease Research Laboratory, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
- Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
- Department of Pediatrics B, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Dana Levin
- Genetic Kidney Disease Research Laboratory, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Edo Dotan
- The George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
- The Taub Faculty of Computer Science, Technion Israel Institute of Technology, Haifa, Israel
| | - Ronit Calderon-Margalit
- Braun School of Public Health, Hadassah Medical Center, Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | - Alla Shnaider
- Department of Nephrology, Soroka University Medical Center, Beer-Sheva, Israel
- Faculty of Health Sciences, Ben Gurion University, Beer-Sheva, Israel
| | - Yosef S Haviv
- Department of Nephrology, Soroka University Medical Center, Beer-Sheva, Israel
- Faculty of Health Sciences, Ben Gurion University, Beer-Sheva, Israel
| | - Ohad S Birk
- Faculty of Health Sciences, Ben Gurion University, Beer-Sheva, Israel
- Genetics Institute at Soroka Medical Center, Beer-Sheva, Israel
| | - Noam Hadar
- Faculty of Health Sciences, Ben Gurion University, Beer-Sheva, Israel
| | - Yair Anikster
- Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
- Metabolic Diseases Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Noa Berar Yanay
- Nephrology Department, Hillel Yaffe Medical Center, Hadera, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Gil Chernin
- Department of Nephrology and Hypertension, Kaplan Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Rehovot, Israel
| | - Etty Kruzel-Davila
- Nephrology Department, Galilee Medical Center, Nahariya, Israel
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Pazit Beckerman
- Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
- Institute of Nephrology and Hypertension, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Benaya Rozen-Zvi
- Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
- Department of Nephrology and Hypertension, Rabin Medical Center, Petah Tikva, Israel
| | - Gabriel T Doctor
- Centre for Genetics and Genomics, Department of Renal Medicine, UCL Division of Medicine, University College London, London, UK
| | - Horia C Stanescu
- Centre for Genetics and Genomics, Department of Renal Medicine, UCL Division of Medicine, University College London, London, UK
| | - Revital Shemer
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Elon Pras
- Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
- The Danek Gertner Institute of Human Genetics, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Haike Reznik-Wolf
- Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
- The Danek Gertner Institute of Human Genetics, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Ayelet Hashahar Nahum
- The Danek Gertner Institute of Human Genetics, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Dan Dominissini
- Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
- Institute of Hematology, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
- Cancer Research Center, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Karl Skorecki
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
- Rambam Health Care Campus, Haifa, Israel
| | - Asaf Vivante
- Genetic Kidney Disease Research Laboratory, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
- Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
- Department of Pediatrics B, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
- Pediatric Nephrology Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| |
Collapse
|
19
|
Zhu L, Xia Y, Ding H, Zhang T, Li J, Li B. Infantile epileptic spasms syndrome: an etiologic study of 361 patients with infantile epileptic spasms syndrome. Front Pediatr 2025; 12:1522079. [PMID: 39850204 PMCID: PMC11754263 DOI: 10.3389/fped.2024.1522079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 12/11/2024] [Indexed: 01/25/2025] Open
Abstract
Introduction Infantile Epileptic Spasms Syndrome (IESS) typically has a profound impact on the neurodevelopment of patients. The study on IESS indicates possible geographical variation in etiology and a lack of data from China. Our study intends to summarize the etiology of IESS and analyze its characteristics. Methods A retrospective analysis was performed to gather clinical data from patients diagnosed with IESS at the Department of Neurology of Qilu Hospital of Shandong University and the Children's Hospital Affiliated to Shandong University between June 2017 to May 2024. Results A total of 361 patients with IESS were included, comprising 115 structural cases (31.9%), 37 genetic cases (10.2%), 32 genetic-structural cases (8.9%), 9 metabolic cases (2.5%), 3 infectious cases (0.8%), and 165 cases with unknown etiology (45.7%). No immunological cause was determined. The primary cause of the condition was linked to hypoxic-ischemic encephalopathy (HIE), with structural brain abnormalities following closely. The predominant pathogenic genes identified were TSC2, NF1, SCN8A, and KCNQ2. Male gender, preterm infants, low birth weight infants, and developmental regression in patients were associated with a higher likelihood of structural etiology. Patients exhibiting developmental regression before the commencement demonstrated inferior outcomes. Patients administered adrenocorticotropic hormone (ACTH) exhibited a higher likelihood of attaining seizure control, and those who responded favorably to the medication saw improved results. Conclusion The predominant etiology of IESS is structural, succeeded by genetic factors, with significant pathogenic genes comprising TSC2, NF1, SCN8A, and KCNQ2. The genetic classifications exhibit geographic variability. Genetic and structural etiologies are frequently linked to an unfavorable prognosis. Genetic testing can help clarify the etiology of IESS when metabolic screening and brain MRI results are negative. The advancement of genetic testing is crucial for future targeted and individualized diagnosis and therapy.
Collapse
Affiliation(s)
- Linghui Zhu
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yuan Xia
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Hao Ding
- Children's Hospital Affiliated to Shandong University, Jinan Children's Hospital, Jinan, Shandong, China
| | - Tong Zhang
- Children's Hospital Affiliated to Shandong University, Jinan Children's Hospital, Jinan, Shandong, China
| | - Jun Li
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Baomin Li
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| |
Collapse
|
20
|
Dias MQ, Gouveia N, Félix R, Estrela-Silva S, Cabral D, Carvalho AL, Murta J, Silva R, Marques JP. Mutational spectrum and deep phenotyping in Pseudoxanthoma Elasticum: Findings from a Portuguese cohort. Eur J Ophthalmol 2025; 35:112-118. [PMID: 38602027 DOI: 10.1177/11206721241247676] [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: 04/12/2024]
Abstract
INTRODUCTION Pseudoxanthoma Elasticum (PXE) is a rare autosomal recessive disorder originated by disease-causing variants in ABCC6 gene. The purpose of this study was to characterize the genetic landscape, phenotypic spectrum and genotype-phenotype correlations in a Portuguese cohort of PXE patients. METHODS Multicentric cross-sectional study conducted in patients with a clinical and genetic diagnosis of PXE. Patients were identified using the IRD-PT registry (www.retina.com.pt). Genotypes were classified into 3 groups: (1) two truncating variants, (2) two non-truncating variants, or (3) mixed variants. Deep phenotyping comprised a comprehensive ophthalmologic and systemic evaluation using the updated Phenodex Score (PS). RESULTS Twenty-seven patients (23 families) were included. Sixteen different ABCC6 variants were identified, 7 of which are novel. The most prevalent variant was the nonsense variant c.3421C > T p.(Arg1141*) with an allele frequency of 18.5%. All patients exhibited ocular manifestations. Cutaneous manifestations were present in most patients (88.9%, n = 24/27). A PS score > E2 was strongly associated with worse visual acuity (B = -29.02; p = 0.001). No association was found between genotypic groups and cutaneous, vascular or cardiac manifestations. CONCLUSIONS This study describes the genetic spectrum of patients with PXE for the first time in a Portuguese cohort. A total of 16 different variants in ABCC6 were found (7 of which are novel), thus highlighting the genotypic heterogeneity associated with this condition and expanding its mutational spectrum. Still, no major genotype-phenotype associations could be established.
Collapse
Affiliation(s)
- Margarida Q Dias
- Ophthalmology Unit, Centro Hospitalar e Universitário de Coimbra, (CHUC), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Nuno Gouveia
- Ophthalmology Unit, Centro Hospitalar e Universitário de Coimbra, (CHUC), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Raquel Félix
- Ophthalmology Unit, Centro Hospitalar e Universitário de Coimbra, (CHUC), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Sérgio Estrela-Silva
- Ophthalmology Unit, Centro Hospitalar e Universitário de São João (CHUSJ), Porto, Portugal
- Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Diogo Cabral
- Ophthalmology Unit, Hospital Garcia de Orta (HGO), Lisbon, Portugal
| | - Ana Luísa Carvalho
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- Medical Genetics Unit, Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal
- University Clinic of Medical Genetics, Faculty of Medicine, University of Coimbra (FMUC), Coimbra, Portugal
| | - Joaquim Murta
- Ophthalmology Unit, Centro Hospitalar e Universitário de Coimbra, (CHUC), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- University Clinic of Ophthalmology, Faculty of Medicine, University of Coimbra (FMUC), Coimbra, Portugal
| | - Rufino Silva
- Ophthalmology Unit, Centro Hospitalar e Universitário de Coimbra, (CHUC), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- University Clinic of Ophthalmology, Faculty of Medicine, University of Coimbra (FMUC), Coimbra, Portugal
| | - João Pedro Marques
- Ophthalmology Unit, Centro Hospitalar e Universitário de Coimbra, (CHUC), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- University Clinic of Ophthalmology, Faculty of Medicine, University of Coimbra (FMUC), Coimbra, Portugal
| |
Collapse
|
21
|
Talarico M, Procopio R, Gagliardi M, Sarubbi MC, Fortunato F, Sammarra I, Lesca G, Malanga D, Annesi G, Gambardella A. Novel KCNQ2 Variants Related to a Variable Phenotypic Spectrum Ranging from Epilepsy with Auditory Features to Severe Developmental and Epileptic Encephalopathies. Int J Mol Sci 2024; 26:295. [PMID: 39796146 PMCID: PMC11719710 DOI: 10.3390/ijms26010295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 12/24/2024] [Accepted: 12/28/2024] [Indexed: 01/13/2025] Open
Abstract
Pathogenic KCNQ2 variants are associated with neonatal epilepsies, ranging from self-limited neonatal epilepsy to KCNQ2-developmental and epileptic encephalopathy (DEE). In this study, next-generation sequencing was performed, applying a panel of 142 epilepsy genes on three unrelated individuals and affected family members, showing a wide variability in the epileptic spectrum. The genetic analysis revealed two likely pathogenic missense variants (c.1378G>A and c.2251T>G) and the already-reported pathogenic splice site (c.1631+1G>A) in KCNQ2 (HGNC:6296). The phenotypes observed in the affected members of family 1, which shared the c.2251T>G variant, were epilepsy with auditory features (EAFs), focal epilepsy, and generalized epilepsy, and none of them suffered from neonatal seizures. The gene panel contained further genes related to EAFs (LGI1, RELN, SCN1A, and DEPDC5), which were tested with negative results. The phenotypes observed in family 2 members, sharing the splice site variant, were neonatal seizures and focal epilepsy in childhood. The last unrelated proband, harboring the de novo missense c.1378G>A, presented a clinical phenotype consistent with DEE. In conclusion, we identified two unreported KCNQ2 variants, and report a proband with EAFs and individuals without typical KCNQ2 neonatal seizures. Our study underscores the extreme variability in the phenotypic spectrum of KCNQ2-related epilepsies and unveils the prospect of its inclusion in screening panels for EAFs.
Collapse
Affiliation(s)
- Mariagrazia Talarico
- Institute of Neurology, Department of Medical and Surgical Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (M.T.); (I.S.); (A.G.)
| | - Radha Procopio
- Neuroscience Research Centre, Medical and Surgical Sciences, 88100 Catanzaro, Italy; (R.P.); (M.G.)
| | - Monica Gagliardi
- Neuroscience Research Centre, Medical and Surgical Sciences, 88100 Catanzaro, Italy; (R.P.); (M.G.)
| | - Maria Chiara Sarubbi
- Laboratory of Molecular Oncology, Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy; (M.C.S.); (D.M.)
| | - Francesco Fortunato
- Institute of Neurology, Department of Medical and Surgical Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (M.T.); (I.S.); (A.G.)
| | - Ilaria Sammarra
- Institute of Neurology, Department of Medical and Surgical Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (M.T.); (I.S.); (A.G.)
| | - Gaetan Lesca
- Genetics Department, Hospices Civils de Lyon, 69000 Lyon, France;
- Neuromyogene Institute, Pathology and Genetics of Neuron and Muscle, CNRS UMR 5261 INSERM U1315, University of Lyon—Université Claude Bernard Lyon 1, 69100 Lyon, France
| | - Donatella Malanga
- Laboratory of Molecular Oncology, Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy; (M.C.S.); (D.M.)
- Interdepartmental Center of Services (CIS), University Magna Graecia, 88100 Catanzaro, Italy
| | - Grazia Annesi
- Biomedical Research and Innovation, National Research Council, 87050 Mangone, Italy;
| | - Antonio Gambardella
- Institute of Neurology, Department of Medical and Surgical Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (M.T.); (I.S.); (A.G.)
| |
Collapse
|
22
|
Gerashchenko G, Gulkovskyi R, Melnichuk N, Hryshchenko N, Marchyshak T, Mankovska O, Bezverkhiy A, Kotuza I, Rosha L, Kotuza A, Tkachuk Z, Kashuba V, Tukalo M. IDENTIFICATION OF CLINICALLY RELEVANT GENE VARIANTS IN COLON ADENOCARCINOMA SAMPLES OF UKRAINIAN PATIENTS USING A COMPREHENSIVE CANCER PANEL: A PILOT STUDY. Exp Oncol 2024; 46:221-227. [PMID: 39704459 DOI: 10.15407/exp-oncology.2024.03.221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Indexed: 12/21/2024]
Abstract
The study aimed to identify the clinically relevant gene variants in colon adenocarcinoma samples of Ukrainian patients using the NGS Comprehensive Cancer Panel (CCP) to implement them conveniently in clinical practice. METHODS We have studied 20 samples of Ukrainian patients with colorectal adenocarcinomas of various differentiation grades. To identify the clinically relevant gene variants, the CCP data were filtered using the Franklin by Genoox database. RESULTS A total of 79 clinically relevant gene variant alterations (SNVs, INDELs) were found in 28 of 409 genes. The largest number of mutations was found in 3 genes, APC, TP53, and KRAS (16, 14, and 8, accordingly). We revealed 4 variants in PTEN and SMAD4, 3 variants in CHEK2, ERBB2, and PIK3CA genes, and 2 variants in AKT1, ATM, DST, IDH1, and TCF12. Mutations for 7 genes, KRAS, TP53, CHEK2, PTEN, AKT1, APC, and SMAD4, were found in more than 1 tumor tissue sample. Tier 1-2 gene variants rate was about 50% of all genetic variants. The therapeutic significance was found in more than 55% of mutations. Additionally, 11 novel genetic mutations in 9 genes have been identified, including G6PD, APC, DST, SINE1, SMAD2, and FLCN. CONCLUSIONS These data suggest a high level of clinical relevance of the NGS CCP approach. Further confirmation on a larger number of samples and using a deeper analysis by other approaches is required.
Collapse
Affiliation(s)
- G Gerashchenko
- Institute of Molecular Biology and Genetics NAS of Ukraine, Kyiv, Ukraine
| | - R Gulkovskyi
- Institute of Molecular Biology and Genetics NAS of Ukraine, Kyiv, Ukraine
| | - N Melnichuk
- Institute of Molecular Biology and Genetics NAS of Ukraine, Kyiv, Ukraine
| | - N Hryshchenko
- Institute of Molecular Biology and Genetics NAS of Ukraine, Kyiv, Ukraine
| | - T Marchyshak
- Institute of Molecular Biology and Genetics NAS of Ukraine, Kyiv, Ukraine
| | - O Mankovska
- Institute of Molecular Biology and Genetics NAS of Ukraine, Kyiv, Ukraine
| | - A Bezverkhiy
- Institute of Molecular Biology and Genetics NAS of Ukraine, Kyiv, Ukraine
| | - I Kotuza
- Feofaniya Clinical Hospital of the State Management of Affairs, Kyiv, Ukraine
| | - L Rosha
- Feofaniya Clinical Hospital of the State Management of Affairs, Kyiv, Ukraine
| | - A Kotuza
- Feofaniya Clinical Hospital of the State Management of Affairs, Kyiv, Ukraine
| | - Z Tkachuk
- Institute of Molecular Biology and Genetics NAS of Ukraine, Kyiv, Ukraine
| | - V Kashuba
- Institute of Molecular Biology and Genetics NAS of Ukraine, Kyiv, Ukraine
| | - M Tukalo
- Institute of Molecular Biology and Genetics NAS of Ukraine, Kyiv, Ukraine
| |
Collapse
|
23
|
Gebbia M, Zimmerman D, Jiang R, Nguyen M, Weile J, Li R, Gavac M, Kishore N, Sun S, Boonen RA, Hamilton R, Dines JN, Wahl A, Reuter J, Johnson B, Fowler DM, Couch FJ, van Attikum H, Roth FP. A missense variant effect map for the human tumor-suppressor protein CHK2. Am J Hum Genet 2024; 111:2675-2692. [PMID: 39642869 PMCID: PMC11639082 DOI: 10.1016/j.ajhg.2024.10.013] [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: 02/29/2024] [Revised: 10/18/2024] [Accepted: 10/22/2024] [Indexed: 12/09/2024] Open
Abstract
The tumor suppressor CHEK2 encodes the serine/threonine protein kinase CHK2 which, upon DNA damage, is important for pausing the cell cycle, initiating DNA repair, and inducing apoptosis. CHK2 phosphorylation of the tumor suppressor BRCA1 is also important for mitotic spindle assembly and chromosomal stability. Consistent with its cell-cycle checkpoint role, both germline and somatic variants in CHEK2 have been linked to breast and other cancers. Over 90% of clinical germline CHEK2 missense variants are classified as variants of uncertain significance, complicating diagnosis of CHK2-dependent cancer. We therefore sought to test the functional impact of all possible missense variants in CHK2. Using a scalable multiplexed assay based on the ability of human CHK2 to complement DNA sensitivity of Saccharomyces cerevisiae cells lacking the CHEK2 ortholog, RAD53, we generated a systematic "missense variant effect map" for CHEK2 missense variation. The map reflects known biochemical features of CHK2 while offering new biological insights. It also provides strong evidence toward pathogenicity for some clinical missense variants and supporting evidence toward benignity for others. Overall, this comprehensive missense variant effect map contributes to understanding of both known and yet-to-be-observed CHK2 variants.
Collapse
Affiliation(s)
- Marinella Gebbia
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Daniel Zimmerman
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Rosanna Jiang
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Maria Nguyen
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Jochen Weile
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Roujia Li
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Michelle Gavac
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Nishka Kishore
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Song Sun
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Rick A Boonen
- Leiden University Medical Center, Leiden, the Netherlands
| | - Rayna Hamilton
- Woods Hole Oceanographic Institution, Woods Hole, MA, USA
| | - Jennifer N Dines
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | | | | | | | - Douglas M Fowler
- Department of Genome Sciences, University of Washington, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA
| | | | | | - Frederick P Roth
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada; Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
24
|
Gutierrez-Rodrigues F, Groarke EM, Thongon N, Rodriguez-Sevilla JJ, Catto LFB, Niewisch MR, Shalhoub R, McReynolds LJ, Clé DV, Patel BA, Ma X, Hironaka D, Donaires FS, Spitofsky N, Santana BA, Lai TP, Alemu L, Kajigaya S, Darden I, Zhou W, Browne PV, Paul S, Lack J, Young DJ, DiNardo CD, Aviv A, Ma F, De Oliveira MM, de Azambuja AP, Dunbar CE, Olszewska M, Olivier E, Papapetrou EP, Giri N, Alter BP, Bonfim C, Wu CO, Garcia-Manero G, Savage SA, Young NS, Colla S, Calado RT. Clonal landscape and clinical outcomes of telomere biology disorders: somatic rescue and cancer mutations. Blood 2024; 144:2402-2416. [PMID: 39316766 PMCID: PMC11862815 DOI: 10.1182/blood.2024025023] [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: 04/17/2024] [Revised: 08/19/2024] [Accepted: 09/05/2024] [Indexed: 09/26/2024] Open
Abstract
ABSTRACT Telomere biology disorders (TBDs), caused by pathogenic germ line variants in telomere-related genes, present with multiorgan disease and a predisposition to cancer. Clonal hematopoiesis (CH) as a marker of cancer development and survival in TBDs is poorly understood. Here, we characterized the clonal landscape of a large cohort of 207 patients with TBD with a broad range of age and phenotype. CH occurred predominantly in symptomatic patients and in signature genes typically associated with cancers: PPM1D, POT1, TERT promoter (TERTp), U2AF1S34, and/or TP53. Chromosome 1q gain (Chr1q+) was the commonest karyotypic abnormality. Clinically, multiorgan involvement and CH in TERTp, TP53, and splicing factor genes were associated with poorer overall survival. Chr1q+ and splicing factor or TP53 mutations significantly increased the risk of hematologic malignancies, regardless of clonal burden. Chr1q+ and U2AF1S34 mutated clones were premalignant events associated with the secondary acquisition of mutations in genes related to hematologic malignancies. Similar to the known effects of Chr1q+ and TP53-CH, functional studies demonstrated that U2AF1S34 mutations primarily compensated for aberrant upregulation of TP53 and interferon pathways in telomere-dysfunctional hematopoietic stem cells, highlighting the TP53 pathway as a canonical route of malignancy in TBD. In contrast, somatic POT1/PPM1D/TERTp mutations had distinct trajectories unrelated to cancer development. With implications beyond TBD, our data show that telomere dysfunction is a strong selective pressure for CH. In TBD, CH is a poor prognostic marker associated with worse overall survival. The identification of key regulatory pathways that drive clonal transformation in TBD allows for the identification of patients at a higher risk of cancer development.
Collapse
Affiliation(s)
| | - Emma M. Groarke
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Natthakan Thongon
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Luiz Fernando B. Catto
- Department of Medical Imaging, Hematology, and Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Marena R. Niewisch
- Division of Cancer Epidemiology and Genetics, Clinical Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Ruba Shalhoub
- Office of Biostatistics Research, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Lisa J. McReynolds
- Division of Cancer Epidemiology and Genetics, Clinical Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Diego V. Clé
- Department of Medical Imaging, Hematology, and Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Bhavisha A. Patel
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Xiaoyang Ma
- Office of Biostatistics Research, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Dalton Hironaka
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Flávia S. Donaires
- Department of Medical Imaging, Hematology, and Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Nina Spitofsky
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Barbara A. Santana
- Department of Medical Imaging, Hematology, and Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Tsung-Po Lai
- Center of Human Development and Aging, Rutgers New Jersey Medical School, The State University of New Jersey, Newark, New Jersey
| | - Lemlem Alemu
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Sachiko Kajigaya
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Ivana Darden
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Weiyin Zhou
- Division of Cancer Epidemiology and Genetics, Clinical Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Paul V. Browne
- Department of Haematology, Trinity College Dublin, Dublin, Ireland
| | - Subrata Paul
- Integrated Data Sciences Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Justin Lack
- Integrated Data Sciences Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - David J. Young
- Translational Stem Cell Biology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Courtney D. DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Abraham Aviv
- Center of Human Development and Aging, Rutgers New Jersey Medical School, The State University of New Jersey, Newark, New Jersey
| | - Feiyang Ma
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | | | | | - Cynthia E. Dunbar
- Translational Stem Cell Biology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Malgorzata Olszewska
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Emmanuel Olivier
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Eirini P. Papapetrou
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Neelam Giri
- Division of Cancer Epidemiology and Genetics, Clinical Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Blanche P. Alter
- Division of Cancer Epidemiology and Genetics, Clinical Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Carmem Bonfim
- Bone Marrow Transplantation Unit, Federal University of Parana, Curitiba, Brazil
- Pediatric Blood and Marrow Transplantation Program, Pequeno Principe Hospital, Curitiba, Brazil
| | - Colin O. Wu
- Office of Biostatistics Research, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | | | - Sharon A. Savage
- Division of Cancer Epidemiology and Genetics, Clinical Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Neal S. Young
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Simona Colla
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Rodrigo T. Calado
- Department of Medical Imaging, Hematology, and Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
25
|
Chung CT, Lee NC, Lin IT, Chen PY, Jao T. The role of genetic testing in adult patients with unexplained epilepsy. Epileptic Disord 2024; 26:814-826. [PMID: 39283677 DOI: 10.1002/epd2.20286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 09/01/2024] [Indexed: 12/18/2024]
Abstract
OBJECTIVE Genetic causes are often overlooked in patients with epilepsy of unknown etiology, particularly in adults. We aimed to evaluate clinical features of genetic epilepsy and the utility of genetic testing. METHODS We retrospectively screened consecutive unrelated adult epilepsy patients at an epilepsy clinic from April 2022 to May 2023. Patients with unknown etiology or special brain lesions were classified as unexplained epilepsy. In them, patients with young-onset seizures or family history of seizures who were recommended for and ultimately underwent genetic testing using either panel next-generation sequencing (NGS) or whole-exome sequencing (WES) were enrolled. A definite or probable genetic diagnosis was established through genotype-phenotype correlation. We compared the demographic characteristics between genetic epilepsy and other etiologies. RESULTS Of the 374 adult epilepsy patients, 258 were classified as unexplained epilepsy, 129 were suspected of having genetic epilepsy due to young-onset seizures or a positive family history, 33 underwent genetic testing; 13 harbored variants classified as pathogenic, and 6 reached a definite genetic diagnosis, resulting in a yield of 18%. Among the 27 patients without a definite genetic diagnosis, 7 had a nongenetic structural etiology. Patients with genetic etiology exhibited greater multisystem involvement particularly multiple structural anomalies and early childhood-onset seizures, but wasn't directly correlated with young-onset seizures or a positive family history. The diagnostic yield was comparable between panel NGS and WES. SIGNIFICANCE In adult patients with unexplained epilepsy, genetic epilepsy is more associated with multisystem involvement and multiple structural anomalies but not family history of seizures or young-onset seizures.
Collapse
Affiliation(s)
- Chi-Ting Chung
- Department of Neurology, En Chu Kong Hospital, New Taipei City, Taiwan
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ni-Chung Lee
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
- Department of Medical Genetics, National Taiwan University, Taipei, Taiwan
| | - I-Ting Lin
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Pin-Yu Chen
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
- Epilepsy Center, National Taiwan University Hospital, Taipei, Taiwan
| | - Tun Jao
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
- Epilepsy Center, National Taiwan University Hospital, Taipei, Taiwan
- Department of Neurology, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
26
|
Hussein Y, Weisblum‐Neuman H, Ben Zeev B, Stern S. Previously defined variants of uncertain significance may play an important role in epilepsy and interactions between certain variants may become pathogenic. Epilepsia Open 2024; 9:2443-2453. [PMID: 39509559 PMCID: PMC11633689 DOI: 10.1002/epi4.13085] [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: 05/26/2024] [Revised: 09/28/2024] [Accepted: 10/09/2024] [Indexed: 11/15/2024] Open
Abstract
OBJECTIVE Epilepsy is a chronic neurological disorder related to various etiologies, and the prevalence of active epilepsy is estimated to be between 4 and 10 per 1000 individuals having a significant role in genetic mutations. Next-Generation Sequencing (NGS) panels are utilized for genetic testing, but a substantial proportion of the results remain uncertain and are not considered directly causative of epilepsy. This study aimed to reevaluate pediatric patients diagnosed with epilepsy who underwent genetic investigation using NGS panels, focusing on inconclusive variant findings or multiple variants of uncertain significance (VUSs). METHODS A subgroup of pediatric patients aged 0-25 years, diagnosed with epilepsy, who underwent genetic investigation with an NGS epilepsy panel at the Child Neurology Unit, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, between 2018 and 2022 through Invitae, was reevaluated. Patients with inconclusive variant findings or multiple VUSs in their test results were included. Genetic data were analyzed to identify potentially pathogenic variants and frequent genetic combinations. RESULTS Two unrelated potentially pathogenic variants were identified in the SCN9A and QARS1 genes. A frequent genetic combination, RANBP2&RYR3, was also observed among other combinations. The RANBP2 gene consistently co-occurred with RYR3 variants in uncertain results, suggesting potential pathogenicity. Analysis of unaffected parents' data revealed certain combinations inherited from different parents, suggesting specific gene combinations as possible risk factors for the disease. SIGNIFICANCE This study highlights the importance of reevaluating genetic data from pediatric epilepsy patients with inconclusive variant findings or multiple VUSs. Identification of potentially pathogenic variants and frequent genetic combinations, such as RANBP2&RYR3, could aid in understanding the genetic basis of epilepsy and identifying potential hotspots. PLAIN LANGUAGE SUMMARY We have performed a retrospective analysis on a subpopulation of pediatric patients diagnosed with epilepsy. We found that specific genetic variants were repeatable, indicating their potential pathogenicity to the disease.
Collapse
Affiliation(s)
- Yara Hussein
- Sagol Department of Neurobiology, Faculty of Natural SciencesUniversity of HaifaHaifaIsrael
| | - Hila Weisblum‐Neuman
- Pediatric Neurology Unit, The Edmond and Lily Safra Children's HospitalSheba Medical CenterRamat GanIsrael
| | - Bruria Ben Zeev
- Pediatric Neurology Unit, The Edmond and Lily Safra Children's HospitalSheba Medical CenterRamat GanIsrael
- Faculty of MedicineTel Aviv UniversityTel AvivIsrael
| | - Shani Stern
- Sagol Department of Neurobiology, Faculty of Natural SciencesUniversity of HaifaHaifaIsrael
| |
Collapse
|
27
|
Yan L, Sun X, Lou B, Zhang Y, Zhuang D, Jia J, Zhang L, He Y, Xu L, Wu S, Zhou Q, Chen C, Yu X, Li H. Carrier frequency and incidence estimation of deficiency of adenosine deaminase 2 in the Chinese population based on massive exome sequencing data. Clin Immunol 2024; 269:110394. [PMID: 39481740 DOI: 10.1016/j.clim.2024.110394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/25/2024] [Accepted: 10/27/2024] [Indexed: 11/02/2024]
Abstract
Deficiency of adenosine deaminase 2 (DADA2) is an autosomal recessive autoinflammatory disease characterised by early onset stroke, recurrent fever, and diverse vascular pathologies, caused by loss-of-function homozygous or compound heterozygous variants of ADA2. This research aimed to determine the carrier frequency and expected incidence of DADA2 in China, using massive exome sequencing (ES) data. A total of 50 likely pathogenic/pathogenic variants (LP/PVs) were identified among 69,413 Chinese individuals, including 20 novel and rare variants (<0.0022 % allele frequency), expanding the known spectrum of PVs in ADA2. The overall carrier frequency in the Chinese population was 1.05 % (732/69,413) and the estimated incidence of DADA2 was approximately one in 92,251 individuals. The present study provides an accurate estimation of the prevalence of DADA2 in China, supporting genetic counseling, early diagnosis treatment, and prognostic evaluation.
Collapse
Affiliation(s)
- Lulu Yan
- The Central Laboratory of Birth Defects Prevention and Control, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, China; Ningbo Key Laboratory for the Prevention and Treatment of Embryogenic Diseases, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, China; Ningbo Key Laboratory of Genomic Medicine and Birth Defects Prevention, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, China
| | - Xiangwei Sun
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
| | - Biying Lou
- Department of Pediatrics, Jinhua Maternal and Child Health Care Hospital, Jinhua, China
| | - Yuxin Zhang
- The Central Laboratory of Birth Defects Prevention and Control, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, China; Ningbo Key Laboratory for the Prevention and Treatment of Embryogenic Diseases, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, China; Ningbo Key Laboratory of Genomic Medicine and Birth Defects Prevention, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, China
| | - Danyan Zhuang
- The Central Laboratory of Birth Defects Prevention and Control, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, China; Ningbo Key Laboratory for the Prevention and Treatment of Embryogenic Diseases, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, China; Ningbo Key Laboratory of Genomic Medicine and Birth Defects Prevention, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, China
| | - Jia Jia
- Fujun Genetics Biotechnology Co., LTD, Shanghai, China
| | - Li Zhang
- Fujun Genetics Biotechnology Co., LTD, Shanghai, China
| | - Yan He
- Department of Pediatrics, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, China
| | - Limin Xu
- Department of Pediatrics, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, China
| | - Shanshan Wu
- Ningbo Key Laboratory of Genomic Medicine and Birth Defects Prevention, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, China; Paediatric Surgery Centre, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, China
| | - Qing Zhou
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
| | - Changshui Chen
- Ningbo Key Laboratory for the Prevention and Treatment of Embryogenic Diseases, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, China
| | - Xiaomin Yu
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China; Department of Rheumatology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Haibo Li
- The Central Laboratory of Birth Defects Prevention and Control, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, China; Ningbo Key Laboratory for the Prevention and Treatment of Embryogenic Diseases, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, China; Ningbo Key Laboratory of Genomic Medicine and Birth Defects Prevention, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, China.
| |
Collapse
|
28
|
Geraghty R, Lovegrove C, Howles S, Sayer JA. Role of Genetic Testing in Kidney Stone Disease: A Narrative Review. Curr Urol Rep 2024; 25:311-323. [PMID: 39096463 PMCID: PMC11374836 DOI: 10.1007/s11934-024-01225-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2024] [Indexed: 08/05/2024]
Abstract
PURPOSE OF REVIEW Kidney stone disease (KSD) is a common and potentially life-threatening condition, and half of patients experience a repeat kidney stone episode within 5-10 years. Despite the ~50% estimate heritability of KSD, international guidelines have not kept up with the pace of discovery of genetic causes of KSD. The European Association of Urology guidelines lists 7 genetic causes of KSD as 'high risk'. RECENT FINDINGS There are currently 46 known monogenic (single gene) causes of kidney stone disease, with evidence of association in a further 23 genes. There is also evidence for polygenic risk of developing KSD. Evidence is lacking for recurrent disease, and only one genome wide association study has investigated this phenomenon, identifying two associated genes (SLC34A1 and TRPV5). However, in the absence of other evidence, patients with genetic predisposition to KSD should be treated as 'high risk'. Further studies are needed to characterize both monogenic and polygenic associations with recurrent disease, to allow for appropriate risk stratification. Durability of test result must be balanced against cost. This would enable retrospective analysis if no genetic cause was found initially. We recommend genetic testing using a gene panel for all children, adults < 25 years, and older patients who have factors associated with high risk disease within the context of a wider metabolic evaluation. Those with a genetic predisposition should be managed via a multi-disciplinary team approach including urologists, radiologists, nephrologists, clinical geneticists and chemical pathologists. This will enable appropriate follow-up, counselling and potentially prophylaxis.
Collapse
Affiliation(s)
- Robert Geraghty
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK.
- Department of Urology, The Newcastle upon Tyne NHS Foundation Trust, Newcastle upon Tyne, UK.
| | - Catherine Lovegrove
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
- Department of Urology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Sarah Howles
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
- Department of Urology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - John A Sayer
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
- Renal Services, The Newcastle upon Tyne NHS Foundation Trust, Newcastle upon Tyne, UK
| |
Collapse
|
29
|
Gavaz M, Aslan ES, Tekeş S. Clinical application of whole-exome sequencing analysis in childhood epilepsy. J Neurogenet 2024; 38:187-194. [PMID: 39654149 DOI: 10.1080/01677063.2024.2434869] [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: 12/26/2023] [Accepted: 11/22/2024] [Indexed: 01/11/2025]
Abstract
The swift updates of public databases and advancements in next-generation sequencing (NGS) technologies have enhanced the genetic identification capacities of epilepsy clinics. This study aimed to evaluate the diagnostic efficacy of NGS in pediatric epilepsy patients as a whole and to present the data obtained in the whole exome sequence analysis. We enrolled 40 children with suspected childhood epilepsy in this study. All patients underwent evaluation by a clinical geneticist or pediatric neurologist and the molecular genetic analysis of those children was performed by whole-exome sequencing (WES). Out of the 40 patients, 12 (30%) received a genetic diagnosis, involving 14 mutations across 13 genes. The cumulative positive diagnostic yield was 30%. Twelve of these patients were identified to have 5 variants previously documented as pathogenic, 9 variants classified as likely pathogenic, and 5 novel variants that have not been reported before. The outcomes indicate that whole-exome sequencing offers great benefits in clinical patient diagnosis, particularly in terms of detecting diagnostic variants. This study underscored the significance of whole exome sequencing (WES) studies, where only a broad gene set is examined in epilepsy patients. This approach has the potential to establish gene-specific phenotypic profiles, particularly by uncovering novel candidate genes in epilepsy patients with well-defined phenotypes. Additionally, conducting validation studies on variants of uncertain clinical significance could enhance the outcome yield.
Collapse
Affiliation(s)
- Meral Gavaz
- Department of Molecular and Medical Genetics, Biruni University, Istanbul, Turkey
| | - Elif S Aslan
- Department of Molecular and Medical Genetics, Biruni University, Istanbul, Turkey
| | - Selahattin Tekeş
- Faculty of Medicine, Department of Medical Genetics, Dicle University, Diyarbakir, Turkey
| |
Collapse
|
30
|
Casula M, Marchetti D, Trevisan L, Pezzoli L, Bellini M, Patrone S, Zingarelli A, Gotta F, Iascone M, Mandich P. Genetics architecture of spontaneous coronary artery dissection in an Italian cohort. Front Cardiovasc Med 2024; 11:1486273. [PMID: 39654947 PMCID: PMC11625805 DOI: 10.3389/fcvm.2024.1486273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 11/01/2024] [Indexed: 12/12/2024] Open
Abstract
Spontaneous coronary artery dissection (SCAD) is a relevant non-atherosclerotic cause of acute coronary syndrome with a complex genetic architecture. Recent discoveries have highlighted the potential role of miRNAs and protein-coding genes involved in the processing of small RNAs in the pathogenesis of SCAD. Furthermore, there may be a connection between SCAD and the increased cardiovascular risk observed in fragile X premutation carriers as well as a correlation with pathogenetic variants in genes encoding for collagen and extracellular matrix, which are related to connective tissue disorders (CTDs). In our cohort of 15 Italian SCAD patients, a total of 37 rare variants were identified in 34 genes using whole exome sequencing (WES) and TRIO-WES analysis when both parents were available. Three likely pathogenic/pathogenetic variants were found in genes previously associated with SCAD and CTDs (COL3A1, COL1A2, and SMAD3) and 26 variants of uncertain significance in genes previously associated with SCAD and CTDs. TRIO-WES analysis revealed 7 de novo variants, 1 of which was found in a potential novel candidate gene (DROSHA). In addition, a premutation allele of 55 ± 2 CGG repeats in the promoter of the FMR1 gene was identified in two related SCAD patients by test for CGG-repeat expansions in the 5'-UTR of the FMR1 gene. Our findings suggest various potential mechanisms such as mRNA toxicity, miRNA regulation, alteration of collagen, and the extracellular matrix architecture, all of which could disrupt vascular homeostasis, and finally, WES and TRIO-WES have proven to be the most powerful approaches for characterizing the genetic background of SCAD.
Collapse
Affiliation(s)
- Marta Casula
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Daniela Marchetti
- Laboratory of Medical Genetics, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Lucia Trevisan
- Medical Genetics Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Laura Pezzoli
- Laboratory of Medical Genetics, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Matteo Bellini
- Laboratory of Medical Genetics, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Serena Patrone
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Antonio Zingarelli
- Cardiological Unit, Ospedale Policlinico IRCSS San Martino, Genoa, Italy
| | - Fabio Gotta
- Medical Genetics Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Maria Iascone
- Laboratory of Medical Genetics, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Paola Mandich
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
- Medical Genetics Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
31
|
Manirakiza AV, Baichoo S, Uwineza A, Dukundane D, Uwinkindi F, Ngendahayo E, Rubagumya F, Muhawenimana E, Nsabimana N, Nzeyimana I, Maniragaba T, Ntirenganya F, Rurangwa E, Mugenzi P, Mutamuliza J, Runanira D, Niyibizi BA, Rugengamanzi E, Besada J, Nielsen SM, Bucknor B, Nussbaum RL, Koeller D, Andrews C, Mutesa L, Fadelu T, Rebbeck TR. Germline sequence variation in cancer genes in Rwandan breast and prostate cancer cases. NPJ Genom Med 2024; 9:61. [PMID: 39582020 PMCID: PMC11586404 DOI: 10.1038/s41525-024-00446-4] [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: 04/09/2024] [Accepted: 11/06/2024] [Indexed: 11/26/2024] Open
Abstract
Cancer genetic data from Sub-Saharan African (SSA) are limited. Patients with female breast (fBC), male breast (mBC), and prostate cancer (PC) in Rwanda underwent germline genetic testing and counseling. Demographic and disease-specific information was collected. A multi-cancer gene panel was used to identify germline Pathogenic Variants (PV) and Variants of Uncertain Significance (VUS). 400 patients (201 with BC and 199 with PC) were consented and recruited to the study. Data was available for 342 patients: 180 with BC (175 women and 5 men) and 162 men with PC. PV were observed in 18.3% fBC, 4.3% PC, and 20% mBC. BRCA2 was the most common PV. Among non-PV carriers, 65% had ≥1 VUS: 31.8% in PC and 33.6% in BC (female and male). Our findings highlight the need for germline genetic testing and counseling in cancer management in SSA.
Collapse
Affiliation(s)
- Achille Vc Manirakiza
- Oncology Unit, Department of Medicine, King Faisal Hospital, Kigali, Rwanda.
- College of Medicine and Health Sciences, University of Rwanda, Kigali, Rwanda.
| | - Shakuntala Baichoo
- Department of Digital Technologies, University of Mauritius, Réduit, Mauritius
| | - Annette Uwineza
- Centre for Human Genetics, College of Medicine and Health Sciences, University of Rwanda, Kigali, Rwanda
- Kigali University Teaching Hospital, Kigali, Rwanda
| | - Damas Dukundane
- Oncology Unit, Department of Medicine, King Faisal Hospital, Kigali, Rwanda
| | | | - Edouard Ngendahayo
- Urology Unit, Department of Surgery, King Faisal Hospital, Kigali, Rwanda
| | - Fidel Rubagumya
- College of Medicine and Health Sciences, University of Rwanda, Kigali, Rwanda
- Rwanda Cancer Center, Rwanda Military Hospital, Kigali, Rwanda
| | | | | | | | | | - Faustin Ntirenganya
- College of Medicine and Health Sciences, University of Rwanda, Kigali, Rwanda
- Kigali University Teaching Hospital, Kigali, Rwanda
| | - Ephrem Rurangwa
- Rwanda Military Referral and Teaching Hospital, Kigali, Rwanda
| | | | | | | | | | | | | | | | | | - Robert L Nussbaum
- Invitae, Inc, San Francisco, CA, USA
- Department of Pediatrics, Division of Medical Genetics, University of California in San Francisco, San Francisco, CA, USA
| | | | | | - Leon Mutesa
- Centre for Human Genetics, College of Medicine and Health Sciences, University of Rwanda, Kigali, Rwanda
| | - Temidayo Fadelu
- Dana Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Timothy R Rebbeck
- Dana Farber Cancer Institute, Boston, MA, USA
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
32
|
Kilic S, Sukruoglu Erdogan O, Tuncer SB, Celik Demirbas B, Yalniz Kayim Z, Yazici H. RNA Splicing Aberrations in Hereditary Cancer: Insights from Turkish Patients. Curr Issues Mol Biol 2024; 46:13252-13266. [PMID: 39590384 PMCID: PMC11593161 DOI: 10.3390/cimb46110790] [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: 10/17/2024] [Revised: 11/14/2024] [Accepted: 11/17/2024] [Indexed: 11/28/2024] Open
Abstract
The process of RNA splicing is fundamental in contributing to proteomic diversity and regulating gene expression. Dysregulation of splicing is associated with various human disorders, including cancer. Through functional studies, this study sought to examine the potential impact of seven variants within six inherited cancer-related genes on RNA splicing patterns in Turkish cancer patients. Upon detecting variants using Next-Generation Sequencing (NGS), we used Reverse Transcriptase Polymerase Chain Reaction (RT-PCR) and Sanger sequencing to elucidate the effects of these variants on splicing. Three of the seven variants demonstrated no discernible effect on RNA, while four exhibited pathogenic characteristics. Specifically, the variants APC c.532-1G>A rs1554072547, BRCA1c.4358-3A>G rs1567779966, BRCA2c.7436-1G>C rs81002830 and MSH3c.1897-1G>A rs1744149615 were identified as pathogenic, while the variants BLMc.4076+4T>G rs183176301, RB1c.2489+2T>C rs1555294636 and RB1c.1050-2A>G rs? were found to be benign from a splicing perspective. These findings highlight the importance of verifying the precise consequences of splice-site variants through experimental analysis, given their potential implications for genetic disorders and cancer predisposition. This research contributes to the understanding of splice-site variants in inherited cancer predisposition, particularly among Turkish cancer patients. It emphasizes the necessity for further exploration into the mechanisms and functional consequences of alternative splicing for potential therapeutic interventions in cancer.
Collapse
Affiliation(s)
- Seda Kilic
- Cancer Genetics Department, Division of Basic Oncology, Institute of Oncology, Istanbul University, 34093 Istanbul, Türkiye; (O.S.E.); (S.B.T.); (B.C.D.); (Z.Y.K.); (H.Y.)
| | - Ozge Sukruoglu Erdogan
- Cancer Genetics Department, Division of Basic Oncology, Institute of Oncology, Istanbul University, 34093 Istanbul, Türkiye; (O.S.E.); (S.B.T.); (B.C.D.); (Z.Y.K.); (H.Y.)
| | - Seref Bugra Tuncer
- Cancer Genetics Department, Division of Basic Oncology, Institute of Oncology, Istanbul University, 34093 Istanbul, Türkiye; (O.S.E.); (S.B.T.); (B.C.D.); (Z.Y.K.); (H.Y.)
| | - Betul Celik Demirbas
- Cancer Genetics Department, Division of Basic Oncology, Institute of Oncology, Istanbul University, 34093 Istanbul, Türkiye; (O.S.E.); (S.B.T.); (B.C.D.); (Z.Y.K.); (H.Y.)
| | - Zubeyde Yalniz Kayim
- Cancer Genetics Department, Division of Basic Oncology, Institute of Oncology, Istanbul University, 34093 Istanbul, Türkiye; (O.S.E.); (S.B.T.); (B.C.D.); (Z.Y.K.); (H.Y.)
| | - Hulya Yazici
- Cancer Genetics Department, Division of Basic Oncology, Institute of Oncology, Istanbul University, 34093 Istanbul, Türkiye; (O.S.E.); (S.B.T.); (B.C.D.); (Z.Y.K.); (H.Y.)
- Medical Biology and Genetics Department, Medical Faculty, Istanbul Health and Technology University, 34275 Istanbul, Türkiye
| |
Collapse
|
33
|
Gohlke J, Lindqvist J, Hourani Z, Rich K, Arnold WD, Heintzman S, Tonino P, Elsheikh B, Morales A, Vatta M, Burghes A, Granzier H, Roggenbuck J. Pathomechanisms of Monoallelic variants in TTN causing skeletal muscle disease. Hum Mol Genet 2024; 33:2003-2023. [PMID: 39277846 PMCID: PMC11578113 DOI: 10.1093/hmg/ddae136] [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: 04/29/2024] [Revised: 07/01/2024] [Accepted: 09/06/2024] [Indexed: 09/17/2024] Open
Abstract
Pathogenic variants in the titin gene (TTN) are known to cause a wide range of cardiac and musculoskeletal disorders, with skeletal myopathy mostly attributed to biallelic variants. We identified monoallelic truncating variants (TTNtv), splice site or internal deletions in TTN in probands with mild, progressive axial and proximal weakness, with dilated cardiomyopathy frequently developing with age. These variants segregated in an autosomal dominant pattern in 7 out of 8 studied families. We investigated the impact of these variants on mRNA, protein levels, and skeletal muscle structure and function. Results reveal that nonsense-mediated decay likely prevents accumulation of harmful truncated protein in skeletal muscle in patients with TTNtvs. Splice variants and an out-of-frame deletion induce aberrant exon skipping, while an in-frame deletion produces shortened titin with intact N- and C-termini, resulting in disrupted sarcomeric structure. All variant types were associated with genome-wide changes in splicing patterns, which represent a hallmark of disease progression. Lastly, RNA-seq studies revealed that GDF11, a member of the TGF-β superfamily, is upregulated in diseased tissue, indicating that it might be a useful therapeutic target in skeletal muscle titinopathies.
Collapse
Affiliation(s)
- Jochen Gohlke
- Department of Cellular and Molecular Medicine, University of Arizona, 1656 E. Mabel St., Tucson, AZ 85724, United States
| | - Johan Lindqvist
- Department of Cellular and Molecular Medicine, University of Arizona, 1656 E. Mabel St., Tucson, AZ 85724, United States
| | - Zaynab Hourani
- Department of Cellular and Molecular Medicine, University of Arizona, 1656 E. Mabel St., Tucson, AZ 85724, United States
| | - Kelly Rich
- Harvard Medical School Department of Genetics - Blavatnik Institute 77 Avenue Louis Pasteur, Boston MA, 02115 USA
| | - W David Arnold
- Department of Physical Medicine and Rehabilitation, University of Missouri, Columbia, MO, USA; NextGen Precision Health, University of Missouri, Columbia, MO, USA; Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA; Department of Neurology, University of Missouri, Columbia, MO, USA
| | - Sarah Heintzman
- Department of Neurology, The Ohio State University Wexner Medical Center, 395 W. 12th Ave, Columbus, OH 43210, United States
| | - Paola Tonino
- Research, Innovation and Impact Core Facilities Department, University of Arizona, 1333 N. Martin Ave, Tucson, AZ 85719, United States
| | - Bakri Elsheikh
- Department of Neurology, The Ohio State University Wexner Medical Center, 395 W. 12th Ave, Columbus, OH 43210, United States
| | - Ana Morales
- Invitae Corporation, 1400 16th St., San Francisco, CA 94103, United States
| | - Matteo Vatta
- Invitae Corporation, 1400 16th St., San Francisco, CA 94103, United States
| | - Arthur Burghes
- Department of Biological Chemistry and Pharmacology, The Ohio State University Wexner Medical Center, 370 W 9th Ave, Columbus, OH 43210, United States
| | - Henk Granzier
- Department of Cellular and Molecular Medicine, University of Arizona, 1656 E. Mabel St., Tucson, AZ 85724, United States
| | - Jennifer Roggenbuck
- Department of Neurology, The Ohio State University Wexner Medical Center, 395 W. 12th Ave, Columbus, OH 43210, United States
| |
Collapse
|
34
|
Kobayashi Y, Chen E, Facio FM, Metz H, Poll SR, Swartzlander D, Johnson B, Aradhya S. Clinical Variant Reclassification in Hereditary Disease Genetic Testing. JAMA Netw Open 2024; 7:e2444526. [PMID: 39504018 PMCID: PMC11541632 DOI: 10.1001/jamanetworkopen.2024.44526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 09/17/2024] [Indexed: 11/08/2024] Open
Abstract
Importance Because accurate and consistent classification of DNA sequence variants is fundamental to germline genetic testing, understanding patterns of initial variant classification (VC) and subsequent reclassification from large-scale, empirical data can help improve VC methods, promote equity among race, ethnicity, and ancestry (REA) groups, and provide insights to inform clinical practice. Objectives To measure the degree to which initial VCs met certainty thresholds set by professional guidelines and quantify the rates of, the factors associated with, and the impact of reclassification among more than 2 million variants. Design, Setting, and Participants This cohort study used clinical multigene panel and exome sequencing data from diagnostic testing for hereditary disorders, carrier screening, or preventive genetic screening from individuals for whom genetic testing was performed between January 1, 2015, and June 30, 2023. Exposure DNA variants were classified into 1 of 5 categories: benign, likely benign, variant of uncertain significance (VUS), likely pathogenic, or pathogenic. Main Outcomes and Measures The main outcomes were accuracy of classifications, rates and directions of reclassifications, evidence contributing to reclassifications, and their impact across different clinical areas and REA groups. One-way analysis of variance followed by post hoc pairwise Tukey honest significant difference tests were used to analyze differences among means, and pairwise Pearson χ2 tests with Bonferroni corrections were used to compare categorical variables among groups. Results The cohort comprised 3 272 035 individuals (median [range] age, 44 [0-89] years; 2 240 506 female [68.47%] and 1 030 729 male [31.50%]; 216 752 Black [6.62%]; 336 414 Hispanic [10.28%]; 1 804 273 White [55.14%]). Among 2 051 736 variants observed over 8 years in this cohort, 94 453 (4.60%) were reclassified. Some variants were reclassified more than once, resulting in 105 172 total reclassification events. The majority (64 752 events [61.65%]) were changes from VUS to either likely benign, benign, likely pathogenic, or pathogenic categories. An additional 37.66% of reclassifications (39 608 events) were gains in classification certainty to terminal categories (ie, likely benign to benign and likely pathogenic to pathogenic). Only a small fraction (663 events [0.63%]) moved toward less certainty, or very rarely (61 events [0.06%]) were classification reversals. When normalized by the number of individuals tested, VUS reclassification rates were higher among specific underrepresented REA populations (Ashkenazi Jewish, Asian, Black, Hispanic, Pacific Islander, and Sephardic Jewish). Approximately one-half of VUS reclassifications (37 074 of 64 840 events [57.18%]) resulted from improved use of data from computational modeling. Conclusions and Relevance In this cohort study of individuals undergoing genetic testing, the empirically estimated accuracy of pathogenic, likely pathogenic, benign, and likely benign classifications exceeded the certainty thresholds set by current VC guidelines, suggesting the need to reevaluate definitions of these classifications. The relative contribution of various strategies to resolve VUS, including emerging machine learning-based computational methods, RNA analysis, and cascade family testing, provides useful insights that can be applied toward further improving VC methods, reducing the rate of VUS, and generating more definitive results for patients.
Collapse
Affiliation(s)
- Yuya Kobayashi
- Labcorp Genetics Inc (formerly Invitae Corporation), San Francisco, California
| | - Elaine Chen
- Invitae Corporation (now part of Labcorp Genetics), San Francisco, California
- Now with Midi Health, Los Altos Hills, California
| | - Flavia M. Facio
- Labcorp Genetics Inc (formerly Invitae Corporation), San Francisco, California
| | - Hillery Metz
- Labcorp Genetics Inc (formerly Invitae Corporation), San Francisco, California
| | - Sarah R. Poll
- Labcorp Genetics Inc (formerly Invitae Corporation), San Francisco, California
| | - Dan Swartzlander
- Labcorp Genetics Inc (formerly Invitae Corporation), San Francisco, California
| | - Britt Johnson
- Invitae Corporation (now part of Labcorp Genetics), San Francisco, California
- Now with GeneDx, Stamford, Connecticut
| | - Swaroop Aradhya
- Invitae Corporation (now part of Labcorp Genetics), San Francisco, California
- Now with Illumina, San Diego, California
- Department of Pathology, Stanford University, Stanford, California
| |
Collapse
|
35
|
Dursun F, Turan İ, Bitkin EÇ, Bayramoğlu E, Çayır A, Erdeve ŞS, Çakır EDP, Çamtosun E, Dilek SO, Kırmızıbekmez H, Eser M, Türkyılmaz A, Karagüzel G. Natural history of ENPP1 deficiency: Nationwide Turkish Cohort Study of autosomal-recessive hypophosphataemic rickets type 2. Clin Endocrinol (Oxf) 2024; 101:475-484. [PMID: 38324408 DOI: 10.1111/cen.15028] [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: 09/12/2023] [Revised: 12/09/2023] [Accepted: 01/24/2024] [Indexed: 02/09/2024]
Abstract
OBJECTIVE Autosomal-recessive hypophosphataemic rickets type 2 (ARHR2) is a rare disease that is reported in survivors of generalized arterial calcification of infancy (GACI). DESIGN, PATIENTS AND MEASUREMENT The objective of this study was to characterize a multicenter paediatric cohort with ARHR2 due to ectonucleotide pyrophosphatase/phosphodiesterase family member 1 (ENPP1) deficiency and with a diagnosis of GACI or GACI-related findings. The clinical, biochemical and genetic characteristics of the patients were retrospectively retrieved. RESULTS We identified 18 patients from 13 families diagnosed with ARHR2. Fifteen of the patients had an ENPP1 variation confirmed with genetic analyses, and three were siblings of one of these patients, who had clinically diagnosed hypophosphataemic rickets (HRs) with the same presentation. From nine centres, 18 patients, of whom 12 (66.7%) were females, were included in the study. The mean age at diagnosis was 4.2 ± 2.2 (1.6-9) years. The most frequently reported clinical findings on admission were limb deformities (66.6%) and short stature (44.4%). At diagnosis, the mean height SD was -2.2 ± 1.3. Five of the patients were diagnosed with GACI in the neonatal period and treated with bisphosphonates. Other patients were initially diagnosed with ARHR2, but after the detection of a biallelic variant in the ENPP1 gene, it was understood that they previously had clinical findings associated with GACI. Three patients had hearing loss, and two had cervical fusion. After the treatment of HRs, one patient developed calcification, and one developed intimal proliferation. CONCLUSION ARHR2 represents one manifestation of ENPP1 deficiency that usually manifests later in life than GACI. The history of calcifications or comorbidities that might be associated with GACI will facilitate the diagnosis in patients with ARHR2, and patients receiving calcitriol and phosphate medication should be carefully monitored for signs of calcification or intimal proliferation.
Collapse
Affiliation(s)
- Fatma Dursun
- Department of Pediatric Endocrinology, SBU Umraniye Training and Research Hospital, University of Health Science, Istanbul, Türkiye
| | - İhsan Turan
- Department of Pediatric Endocrinology, Faculty of Medicine, Cukurova University, Adana, Türkiye
| | - Eda Çelebi Bitkin
- Department of Pediatric Endocrinology, Bakırkoy Dr Sadi Konuk Training and Research Hospital, Istanbul, Türkiye
| | - Elvan Bayramoğlu
- Department of Pediatric Endocrinology, Cerrahpaşa Faculty of Medicine, Istanbul University, İstanbul, Türkiye
| | - Atilla Çayır
- Department of Pediatric Endocrinology and Diabetes, Erzurum Education and Research Hospital, University of Health Science, Erzurum, Türkiye
| | - Şenay Savaş Erdeve
- Department of Pediatric Endocrinology, Ankara Etlik City Hospital, University of Health Science, Ankara, Türkiye
| | - Esra Deniz Papatya Çakır
- Department of Pediatric Endocrinology, Bakırkoy Dr Sadi Konuk Training and Research Hospital, Istanbul, Türkiye
| | - Emine Çamtosun
- Department of Pediatric Endocrinology, Faculty of Medicine, Inönü University, Malatya, Türkiye
| | - Semine Ozdemir Dilek
- Department of Pediatric Endocrinology, Adana City Training and Research Hospital, University of Health Science, Adana, Türkiye
| | - Heves Kırmızıbekmez
- Department of Pediatric Endocrinology, SBU Umraniye Training and Research Hospital, University of Health Science, Istanbul, Türkiye
| | - Metin Eser
- Department of Medical Genetic, Umraniye Training and Research Hospital, Istanbul, Türkiye
| | - Ayberk Türkyılmaz
- Department of Medical Genetic, Faculty of Medicine, Karadeniz Technical University, Trabzon, Türkiye
| | - Gülay Karagüzel
- Department of Pediatric Endocrinology, Faculty of Medicine, Karadeniz Technical University, Trabzon, Türkiye
| |
Collapse
|
36
|
Procopio R, Gagliardi M, Talarico M, Fortunato F, Sammarra I, Procopio AC, Roncada P, Malanga D, Annesi G, Gambardella A. Two Novel Variants in the CHRNA2 and SCN2A Genes in Italian Patients with Febrile Seizures. Genes (Basel) 2024; 15:1407. [PMID: 39596607 PMCID: PMC11593345 DOI: 10.3390/genes15111407] [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: 08/29/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Febrile seizures (FSs) are the most common form of epilepsy in children aged between six months and five years. The exact cause is unknown, but several studies have demonstrated the importance of genetic predisposition, with increasing involvement of receptors and ion channels. The present study aims to identify novel pathogenic variants in Italian patients with FSs. METHODS We performed targeted panel sequencing in a cohort of 21 patients with FSs. In silico analysis was performed to predict the pathogenic role of the resulting variants. RESULTS We found two novel variants segregating in two families with FSs: c.1021C>G (p.Leu341Val) in the CHRNA2 gene and c.140A>G (p.Glu47Gly) in SCN2A. CONCLUSIONS The c.1021C>G (p.Leu341Val) variant leads to a codon change of highly conserved leucine to valine at position 341 and is located in segments M3 of the subunit, which is important for channel gating. The c.140A>G (p.Glu47Gly) variant causes a substitution of glutamic acid with glycine at position 47 of the protein, which is highly conserved across the species. Moreover, it is located in the N-terminal domain, a region commonly affected in ASD, which impacts the inactivation kinetics and voltage dependence of steady-state activation. Further analyses are needed to better explain the role of CHRNA2 and SCN2A in the development of febrile seizures.
Collapse
Affiliation(s)
- Radha Procopio
- Department of Medical and Surgical Sciences, Neuroscience Research Center, Magna Graecia University, 88100 Catanzaro, Italy;
| | - Monica Gagliardi
- Department of Medical and Surgical Sciences, Neuroscience Research Center, Magna Graecia University, 88100 Catanzaro, Italy;
| | - Mariagrazia Talarico
- Institute of Neurology, Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (M.T.); (F.F.); (I.S.); (A.G.)
| | - Francesco Fortunato
- Institute of Neurology, Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (M.T.); (F.F.); (I.S.); (A.G.)
| | - Ilaria Sammarra
- Institute of Neurology, Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (M.T.); (F.F.); (I.S.); (A.G.)
| | - Anna Caterina Procopio
- Department of Health Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (A.C.P.); (P.R.)
| | - Paola Roncada
- Department of Health Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (A.C.P.); (P.R.)
| | - Donatella Malanga
- Laboratory of Molecular Oncology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy;
- Interdepartmental Center of Services (CIS), Magna Graecia University, 88100 Catanzaro, Italy
| | - Grazia Annesi
- Institute for Biomedical Research and Innovation, National Research Council, 87050 Mangone, Italy;
| | - Antonio Gambardella
- Institute of Neurology, Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (M.T.); (F.F.); (I.S.); (A.G.)
| |
Collapse
|
37
|
Abdel-Razeq H, Tamimi F, Sharaf B, Nielsen SM, Heald B, Hatchell KE, Esplin ED, Bani Hani H, Al-Azzam K, Alkyam M, Mustafa R, Al-Atary A. Multi-Gene Panel Testing for Hereditary Cancer Predisposition Among Patients Sixty-Five Years and Above Diagnosed With Breast Cancer. World J Oncol 2024; 15:777-783. [PMID: 39328331 PMCID: PMC11424113 DOI: 10.14740/wjon1919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 07/22/2024] [Indexed: 09/28/2024] Open
Abstract
Background The availability and affordability of germline genetic testing (GGT) has resulted in a broader utilization in daily clinical practice. However, adherence to testing guidelines is low, especially among older patients, where testing is often not offered. Methods In this study, consecutive, newly diagnosed patients with breast cancer (BC) aged ≥ 65 years and eligible for GGT, as per the National Comprehensive Cancer Network (NCCN) guidelines (version 1, 2021), were invited to participate, from March 2021 to December 2022. Patients were offered a restricted (two- or 20-gene panel), or an expanded 84-gene panel. Results During the study period, 204 patients were enrolled. The mean (standard deviation (SD)) age at BC diagnosis was 70.5 (5.13) years, ranging 65 - 81 years. All patients were Arab and the majority were Jordanian. The majority (n = 188, 92.2%) had early-stage (stages I and II) disease. One hundred three (50.5%) patients were tested with a restricted two-gene (n = 13) or 20-gene (n = 90) panel, while the remaining 101 (49.5%) patients had an expanded 84-gene panel. Family history of close blood relative(s) with BC was the most common indication for testing (n = 110, 53.9%). Among the entire study cohort, 22 (10.8%) had pathogenic/likely pathogenic germline variants (PGVs) and another 97 (47.5%) had ≥ 1 variants of uncertain significance (VUS). PGV rates were significantly higher with the expanded panel (14.9%) compared to restricted testing (6.8%) (P = 0.032). Similarly, VUS rates were significantly higher with the expanded panel (64.4%) compared to the restricted panel (31.1%) (P < 0.001). The most prevalent genes with PGVs were BRCA1/2 (31.3% of all PGV-positive patients), CHEK2 (23.1%) and ATM (19.2%). Conclusion GGT should not be overlooked in older BC patients, as this study demonstrates that > 10% of patients have PGVs, largely in potentially actionable genes.
Collapse
Affiliation(s)
- Hikmat Abdel-Razeq
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
- School of Medicine, the University of Jordan, Amman, Jordan
| | - Faris Tamimi
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Baha Sharaf
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | | | | | | | | | - Hira Bani Hani
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Khansa Al-Azzam
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Mais Alkyam
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Rawan Mustafa
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Areej Al-Atary
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| |
Collapse
|
38
|
Barbitoff YA, Khmelkova DN, Pomerantseva EA, Slepchenkov AV, Zubashenko NA, Mironova IV, Kaimonov VS, Polev DE, Tsay VV, Glotov AS, Aseev MV, Shcherbak SG, Glotov OS, Isaev AA, Predeus AV. Expanding the Russian allele frequency reference via cross-laboratory data integration: insights from 7452 exome samples. Natl Sci Rev 2024; 11:nwae326. [PMID: 39498263 PMCID: PMC11533896 DOI: 10.1093/nsr/nwae326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 08/17/2024] [Accepted: 09/12/2024] [Indexed: 11/07/2024] Open
Abstract
Population allele frequency is crucially important for accurate interpretation of known and novel variants in medical genetics. Recently, several large allele frequency databases, such as the Genome Aggregation Database (gnomAD), have been created to serve as a global reference for such studies. However, frequencies of many rare alleles vary dramatically between populations, and population-specific allele frequency is often more informative than the global one. Many countries and regions, including Russia, remain poorly studied from the genetic perspective. Here, we report the first successful attempt to integrate genetic information between major medical genetic laboratories in Russia. We construct RUSeq, an open, large-scale reference set of genetic variants by analyzing 7452 exome samples collected in two major Russian cities-Moscow and St. Petersburg. An ∼10-fold increase in sample size compared to previous studies allowed us to characterize extensive genetic diversity within the admixed Russian population with contributions from several major ancestral groups. We highlight 51 known pathogenic variants that are overrepresented in Russia compared to other European countries. We also identify several dozen high-impact variants that are present in healthy donors despite being annotated as pathogenic in ClinVar and falling within genes associated with autosomal dominant disorders. The constructed database of genetic variant frequencies in Russia has been made available to the medical genetics community through a variant browser available at http://ruseq.ru.
Collapse
Affiliation(s)
- Yury A Barbitoff
- CerbaLab Ltd., St. Petersburg 199106, Russia
- Bioinformatics Institute, St. Petersburg 197342, Russia
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynaecology and Reproductology, St. Petersburg 199034, Russia
| | - Darya N Khmelkova
- Genetics and Reproductive Medicine Center “GENETICO” Ltd., Moscow 121205, Russia
| | | | | | - Nikita A Zubashenko
- Genetics and Reproductive Medicine Center “GENETICO” Ltd., Moscow 121205, Russia
| | - Irina V Mironova
- Genetics and Reproductive Medicine Center “GENETICO” Ltd., Moscow 121205, Russia
| | - Vladimir S Kaimonov
- Genetics and Reproductive Medicine Center “GENETICO” Ltd., Moscow 121205, Russia
| | - Dmitrii E Polev
- CerbaLab Ltd., St. Petersburg 199106, Russia
- Metagenomics Research Group, St. Petersburg Pasteur Institute, St. Petersburg 197101, Russia
| | - Victoria V Tsay
- CerbaLab Ltd., St. Petersburg 199106, Russia
- FGBE “Children's Scientific and Clinical Center for Infectious Diseases of the Federal Medical and Biological Agency”, St. Petersburg 197022, Russia
| | - Andrey S Glotov
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynaecology and Reproductology, St. Petersburg 199034, Russia
| | - Mikhail V Aseev
- CerbaLab Ltd., St. Petersburg 199106, Russia
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynaecology and Reproductology, St. Petersburg 199034, Russia
| | | | - Oleg S Glotov
- CerbaLab Ltd., St. Petersburg 199106, Russia
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynaecology and Reproductology, St. Petersburg 199034, Russia
- FGBE “Children's Scientific and Clinical Center for Infectious Diseases of the Federal Medical and Biological Agency”, St. Petersburg 197022, Russia
- City Hospital No. 40, St. Petersburg 197706, Russia
| | - Arthur A Isaev
- Genetics and Reproductive Medicine Center “GENETICO” Ltd., Moscow 121205, Russia
| | | |
Collapse
|
39
|
Thomas CP. Screening of Living Kidney Donors for Genetic Diseases: CON. KIDNEY360 2024; 5:1413-1415. [PMID: 38748490 PMCID: PMC11556938 DOI: 10.34067/kid.0000000000000353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 01/04/2024] [Indexed: 05/19/2024]
Affiliation(s)
- Christie P Thomas
- Department of Internal Medicine and Iowa Institute of Human Genetics, University of Iowa Carver College of Medicine, Iowa City, Iowa and VA Medical Center, Iowa City, Iowa
| |
Collapse
|
40
|
Abu Hijlih R, Sharaf B, Salah S, Bani Hani H, Nielsen SM, Heald B, Esplin ED, Ghanem R, Alzibdeh A, Al-Batsh T, Al-Masri Y, Abdel-Razeq H. Patterns and Frequency of Pathogenic Germline Variants Among Prostate Cancer Patients Utilizing Multi-Gene Panel Genetic Testing. World J Oncol 2024; 15:801-808. [PMID: 39328335 PMCID: PMC11424115 DOI: 10.14740/wjon1896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 06/21/2024] [Indexed: 09/28/2024] Open
Abstract
Background Germline genetic testing (GGT) has significant implications in the management of patients with prostate cancer (PCa). Herein, we report on patterns and frequency of pathogenic/likely pathogenic germline variants (P/LPGVs) among newly diagnosed Arab patients with PCa. Methods Patients meeting the National Comprehensive Cancer Network (NCCN) eligibility criteria for GGT were offered a 19-gene PCa panel or an expanded 84-gene multi-cancer panel. Results During the study period, 231 patients were enrolled; 107 (46.3%) had metastatic disease at diagnosis. In total, 17 P/LPGVs were detected in 17 patients (7.4%). Among the 113 (48.9%) patients who underwent GGT with the 19-gene panel, eight (7.1%) had P/LPGVs, compared to nine (7.6%) of the 118 (51.1%) who did GGT through the expanded 84-gene panel (P = 0.88). Variant of uncertain significance (VUS) rate was higher (n = 73, 61.9%) among the group who underwent expanded 84-gene panel testing compared to those who underwent the 19-gene PCa panel (n = 35, 30.9%) (P = 0.001). P/LPGVs in DNA damage repair (DDR) genes, most frequently BRCA2, CHEK2 and TP53, were the most common P/LPGVs findings. Conclusion This study is the first to characterize the germline genetic profile of an Arab population with PCa. All detected P/LPGVs were potentially actionable, with most variants able to be detected with a PCa-specific panel.
Collapse
Affiliation(s)
- Ramiz Abu Hijlih
- Department of Radiation Oncology, King Hussein Cancer Center, Amman, Jordan
| | - Baha Sharaf
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Samer Salah
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Hira Bani Hani
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | | | | | | | - Rami Ghanem
- Department of Surgery, King Hussein Cancer Center, Amman, Jordan
| | - Abdulla Alzibdeh
- Department of Radiation Oncology, King Hussein Cancer Center, Amman, Jordan
| | - Tamer Al-Batsh
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Yosra Al-Masri
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Hikmat Abdel-Razeq
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
- School of Medicine, the University of Jordan, Amman, Jordan
| |
Collapse
|
41
|
Li Q, Zheng Y, Guo X, Xue J. Extremely Low Birth Weight Infant (Gestational Age of 29 Weeks) With Kabuki Syndrome Type I: Case Report and Literature Review. Mol Genet Genomic Med 2024; 12:e70025. [PMID: 39400990 PMCID: PMC11476741 DOI: 10.1002/mgg3.70025] [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: 04/27/2023] [Revised: 09/02/2024] [Accepted: 09/25/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND This paper aimed to investigate the clinical phenotype of Kabuki syndrome (KS) in premature infants. METHODS This paper presents a case of an extremely low birth weight infant (gestational age 29 weeks) with KS1 caused by a variant in the KMT2D gene. The clinical, pathological, and differential diagnostic findings were comprehensively analyzed. A thorough literature review was also performed to enhance the understanding of KS, revealing its unique features and prognostic significance. RESULTS The infant was a male with a gestational age of 29 weeks and a birth weight of 850 g. He had intrauterine growth retardation, characterized by cleft palate, sacrococcygeal skin depressions, and recurrent metabolic acidosis. Whole-exome sequencing revealed the c.4267C > T (p.Arg1423Cys) variant in the KMT2D gene, which was absent in his parents. The patient was discharged after 67 days of treatment, and he was followed up to 19 months of corrected gestational age, with growth retardation and expression language delay. Ten previous studies on preterm infants were retrieved, with 10 preterm infants. They all had characteristic facial features, such as long blepharophimosis, sparse and lateral 1/3 eyebrows, and large and prominent/cupped ears. Other manifestations were extrauterine growth delay (7/10), abnormal development of the cardiovascular system (7/10), abnormal development of the nervous system (5/10), and cleft palate (2/10). CONCLUSIONS Kabuki syndrome is a rare hereditary disorder involving multiple organs and systems. Genetic assessment for preterm infants with congenital abnormalities is recommended.
Collapse
Affiliation(s)
- Qi Li
- Neonatal PediatricsShandong University Second HospitalJinanShandongChina
- PediatricThe Fourth Affiliated of Soochow UniversitySuzhouJiangsuChina
| | - Yuzhu Zheng
- Pediatric EndocrinologyShandong University Second HospitalJinanShandongChina
| | - Xinyuan Guo
- Neonatal PediatricsShandong University Second HospitalJinanShandongChina
| | - Jiang Xue
- Neonatal PediatricsShandong University Second HospitalJinanShandongChina
| |
Collapse
|
42
|
Li Q, Troutman C, Peckiconis M, Wurst T, Terry SF. Inorganic pyrophosphate plasma levels in patients with GGCX-associated PXE-like phenotypes. Front Genet 2024; 15:1429320. [PMID: 39399214 PMCID: PMC11466855 DOI: 10.3389/fgene.2024.1429320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 09/16/2024] [Indexed: 10/15/2024] Open
Abstract
Introduction Pseudoxanthoma elasticum (PXE) is an autosomal recessive ectopic calcification disorder clinically affecting the skin, eyes, and vascular system. Most cases of PXE are caused by inactivating pathogenic variants in the ABCC6 gene encoding a hepatic transmembrane efflux transporter, which facilitates the extracellular release of ATP, the precursor of inorganic pyrophosphate (PPi), a potent endogenous inhibitor of calcification. Pathogenic variants in GGCX, encoding γ-glutamyl carboxylase required for activation of vitamin K-dependent coagulation factors as well as matrix Gla protein (MGP) and Gla-rich protein (GRP), two inhibitors of ectopic calcification, have also been reported to cause cutaneous changes like those seen in PXE. While ectopic calcification in ABCC6 deficiency has been associated with reduced plasma levels of PPi due to loss of ABCC6 transport activity in the liver, plasma PPi levels have not been reported in patients with GGCX-associated phenotypes. Methods We analyzed five patients from three unrelated families on their clinical, laboratory, and molecular findings who carry biallelic variants in GGCX and present with phenotypic characteristics associated with PXE. The variants were identified using a next-generation sequencing panel consisting of 29 genes associated with ectopic calcification. Results and conclusion This study demonstrates that in addition to ABCC6, GGCX variants can cause the PXE phenotype, expanding PXE and perhaps other heritable ectopic calcification disorders' clinical and genetic heterogeneity. The results also show that the plasma concentrations of PPi in these patients are not reduced compared to healthy control individuals, suggesting that plasma PPi does not govern ectopic calcification in GGCX deficiency.
Collapse
Affiliation(s)
- Qiaoli Li
- Department of Biochemistry and Molecular Biology, Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Philadelphia, PA, United States
- PXE International Center of Excellence in Research and Clinical Care, Thomas Jefferson University, Philadelphia, PA, United States
| | | | | | - Tamara Wurst
- PXE International, Inc., Damascus, MD, United States
| | | |
Collapse
|
43
|
Osler TS, Brandenburg JT, Schoeman M, Chen WC, Urban MF, Mathew CG. Prevalence and Reclassification of Genetic Variants in South African Populations with Breast Cancer. Genes Chromosomes Cancer 2024; 63:e23275. [PMID: 39324485 DOI: 10.1002/gcc.23275] [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: 08/22/2024] [Accepted: 09/06/2024] [Indexed: 09/27/2024] Open
Abstract
Concurrent testing of numerous genes for hereditary breast cancer (BC) is available but can result in management difficulties. We evaluated use of an expanded BC gene panel in women of diverse South African ancestries and assessed use of African genomic data to reclassify variants of uncertain significance (VUS). A total of 331 women of White, Black African, or Mixed Ancestry with BC had a 9-gene panel test, with an additional 75 genes tested in those without a pathogenic/likely pathogenic (P/LP) variant. The proportion of VUS reclassified using ClinGen gene-specific allele frequency (AF) thresholds or an AF > 0.001 in nonguidelines genes in African genomic data was determined. The 9-gene panel identified 58 P/LP variants, but only two of the P/LP variants detected using the 75-gene panel were in confirmed BC genes, resulting in a total of 60 (18.1%) in all participants. P/LP variant prevalence was similar across ancestry groups, but VUS prevalence was higher in Black African and Mixed Ancestry than in White participants. In total, 611 VUS were detected, representing 324 distinct variants. 10.8% (9/83) of VUS met ClinGen AF thresholds in genomic data while 10.8% (26/240) in nonguideline genes had an AF > 0.001. Overall, 27.0% of VUS occurrences could potentially be reclassified using African genomic data. Thus, expanding the gene panel yielded few clinically actionable variants but many VUS, particularly in participants of Black African and Mixed Ancestry. However, use of African genomic data has the potential to reclassify a significant proportion of VUS.
Collapse
Affiliation(s)
- Tabitha S Osler
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Division of Human Genetics, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Jean-Tristan Brandenburg
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Strengthening Oncology Services Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Mardelle Schoeman
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, University of Stellenbosch and Tygerberg Hospital, Cape Town, Parow, South Africa
| | - Wenlong Carl Chen
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Strengthening Oncology Services Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- National Cancer Registry, National Institute for Communicable Diseases a Division of the National Health Laboratory Service, Johannesburg, South Africa
| | - Michael F Urban
- Division of Human Genetics, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Obstetrics and Gynaecology, Faculty of Medicine and Health Sciences, University of Stellenbosch and Tygerberg Hospital, Cape Town, Parow, South Africa
| | - Christopher G Mathew
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Division of Human Genetics, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King's College London, London, UK
| |
Collapse
|
44
|
MacCarrick G, Aradhya S, Bailey M, Chu D, Hunt A, Izzo E, Krakow D, Mackenzie W, Poll S, Raggio C, Shediac R, White KK, McLaughlin HM, Seratti G. Clinical utility of comprehensive gene panel testing for common and rare causes of skeletal dysplasia and other skeletal disorders: Results from the largest cohort to date. Am J Med Genet A 2024; 194:e63646. [PMID: 38702915 DOI: 10.1002/ajmg.a.63646] [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: 08/16/2023] [Revised: 04/03/2024] [Accepted: 04/18/2024] [Indexed: 05/06/2024]
Abstract
Molecular genetics enables more precise diagnoses of skeletal dysplasia and other skeletal disorders (SDs). We investigated the clinical utility of multigene panel testing for 5011 unrelated individuals with SD in the United States (December 2019-April 2022). Median (range) age was 8 (0-90) years, 70.5% had short stature and/or disproportionate growth, 27.4% had a positive molecular diagnosis (MDx), and 30 individuals received two MDx. Genes most commonly contributing to MDx were FGFR3 (16.9%), ALPL (13.0%), and COL1A1 (10.3%). Most of the 112 genes associated with ≥1 MDx were primarily involved in signal transduction (n = 35), metabolism (n = 23), or extracellular matrix organization (n = 17). There were implications associated with specific care/treatment options for 84.4% (1158/1372) of MDx-positive individuals; >50% were linked to conditions with targeted therapy approved or in clinical development, including osteogenesis imperfecta, achondroplasia, hypophosphatasia, and mucopolysaccharidosis. Forty individuals with initially inconclusive results became MDx-positive following family testing. Follow-up mucopolysaccharidosis enzyme activity testing was positive in 14 individuals (10 of these were not MDx-positive). Our findings showed that inclusion of metabolic genes associated with SD increased the clinical utility of a gene panel and confirmed that integrated use of comprehensive gene panel testing with orthogonal testing reduced the burden of inconclusive results.
Collapse
Affiliation(s)
- Gretchen MacCarrick
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Mitch Bailey
- BioMarin Pharmaceutical Inc, Novato, California, USA
| | - Dorna Chu
- BioMarin Pharmaceutical Inc, Novato, California, USA
| | - Abigail Hunt
- BioMarin Pharmaceutical Inc, Novato, California, USA
| | - Emanuela Izzo
- BioMarin Pharmaceutical Inc, Novato, California, USA
| | - Deborah Krakow
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - William Mackenzie
- Department of Orthopaedic Surgery, Nemours Children's Hospital, Wilmington, Delaware, USA
| | - Sarah Poll
- Invitae Corporation, San Francisco, California, USA
| | - Cathleen Raggio
- Department of Orthopedic Surgery, Hospital for Special Surgery, New York, New York, USA
| | - Renée Shediac
- BioMarin Pharmaceutical Inc, Novato, California, USA
| | - Klane K White
- Department of Pediatric Orthopedic Surgery, Children's Hospital Colorado, Aurora, Colorado, USA
| | | | | |
Collapse
|
45
|
Abramowicz S, Meunier A, Postelmans L, Caspers L, Corazza F, De Bruyne M, Van de Sompele S, De Baere E, Leroy BP, Willermain F, Draganova D. DIAGNOSTIC YIELD OF AN INHERITED RETINAL DISEASE GENE PANEL IN RETINOPATHY OF UNKNOWN ORIGIN. Retina 2024; 44:1597-1607. [PMID: 39167581 DOI: 10.1097/iae.0000000000004155] [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] [Indexed: 08/23/2024]
Abstract
PURPOSE Evaluating the presence of class 3, 4, and 5 genetic variants in inherited retinal disease (IRD) genes in patients with retinopathy of unknown origin (RUO). METHODS Multicentric retrospective study of RUO cases diagnosed between January 2012 and February 2022. General and ophthalmologic history, complete ophthalmologic examination, antiretinal antibodies, and IRD gene panel results were analyzed in every patient. Four RUO categories were defined: nonparaneoplastic autoimmune retinopathy, unilateral pigmentary retinopathy, asymmetrical pigmentary retinopathy, and acute zonal occult outer retinopathy. RESULTS The authors included 12 patients (9 females) across these four RUO categories. Mean age at inclusion was 45.6 years (20-68 years). Seven patients demonstrated class 3 variants in IRD genes. Of these, two also demonstrated class 5 variants in other IRD genes. The remaining five patients had negative panel results. IRD gene panel analysis allowed diagnosis refinement in 1 (8.3%) nonparaneoplastic autoimmune retinopathy patient in the RUO cohort. When considering the nonparaneoplastic autoimmune retinopathy subpopulation only, a higher diagnostic yield of 20% (1/5 patients) was achieved. CONCLUSION Every suspected nonparaneoplastic autoimmune retinopathy patient should benefit from gene panel testing to not overlook undiagnosed IRDs. By contrast, unilateral pigmentary retinopathy, asymmetrical pigmentary retinopathy, and acute zonal occult outer retinopathy subpopulations did not benefit from genetic testing in this study.
Collapse
Affiliation(s)
- Stéphane Abramowicz
- Department of Ophthalmology, Saint-Pierre University Hospital, Université Libre de Bruxelles, Brussels, Belgium
- Department of Ophthalmology, Brugmann University Hospital, Université Libre de Bruxelles, Brussels, Belgium
- Department of Ophthalmology, Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Audrey Meunier
- Department of Ophthalmology, Saint-Pierre University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Laurence Postelmans
- Department of Ophthalmology, Brugmann University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Laure Caspers
- Department of Ophthalmology, Saint-Pierre University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Francis Corazza
- Department of Immunology, LHUB-ULB, Université Libre de Bruxelles, Brussels, Belgium
| | - Marieke De Bruyne
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium ; and
| | - Stijn Van de Sompele
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium ; and
| | - Elfride De Baere
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium ; and
| | - Bart P Leroy
- Department of Ophthalmology, Ghent University Hospital, Ghent University, Ghent, Belgium
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
- Division of Ophthalmology, The Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania
| | - François Willermain
- Department of Ophthalmology, Saint-Pierre University Hospital, Université Libre de Bruxelles, Brussels, Belgium
- Department of Ophthalmology, Brugmann University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Dafina Draganova
- Department of Ophthalmology, Saint-Pierre University Hospital, Université Libre de Bruxelles, Brussels, Belgium
- Department of Ophthalmology, Brugmann University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
46
|
Roggenbuck J, Morales A, Ellis CA, Dratch L, Stetler M, Tan CA, Bucknor B, Hatchell KE, Aradhya S, Esplin ED, Ting YL, Scherer SS. Diagnostic and clinical utility of comprehensive multigene panel testing for patients with neuropathy. J Peripher Nerv Syst 2024; 29:363-367. [PMID: 39140136 DOI: 10.1111/jns.12651] [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] [Accepted: 07/25/2024] [Indexed: 08/15/2024]
Abstract
BACKGROUND AND AIMS Prior to next-generation sequencing (NGS), the evaluation of a patient with neuropathy typically consisted of screening for acquired causes, followed by clinical genetic testing of PMP22, MFN2, GJB1, and MPZ in patients with a positive family history and symptom onset prior to age 50. In this study, we examined the clinical utility of NGS in a large cohort of patients analyzed in a commercial laboratory. METHODS A cohort of 6849 adult patients underwent clinician-ordered peripheral neuropathy multigene panel testing ranging from 66 to 111 genes that included NGS and intragenic deletion/duplication analysis. RESULTS A molecular diagnosis was identified for 8.4% of the cohort (n = 573/6849). Variants in PMP22, MFN2, GJB1, MPZ, and TTR accounted for 73.8% of molecular diagnoses. Results had potential clinical actionability for 398 (69.5%) patients. Our results suggest that 225/573 (39.3%) of molecular diagnoses and 113/398 (28.4%) of clinical interventions would have been missed if the testing approach had been restricted to older guidelines. INTERPRETATION Our results highlight the need for expanded genetic testing guidelines that account for the increased number of genes associated with hereditary neuropathy, address the overlap of acquired and hereditary neuropathy, and provide broader access to genetic diagnosis for patients.
Collapse
Affiliation(s)
| | - Ana Morales
- Invitae Corporation, San Francisco, California, USA
| | - Colin A Ellis
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Laynie Dratch
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | | | | | - Swaroop Aradhya
- Invitae Corporation, San Francisco, California, USA
- Department of Pathology, Stanford University, Stanford, California, USA
| | | | - Yi-Lee Ting
- Invitae Corporation, San Francisco, California, USA
| | - Steven S Scherer
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
47
|
Bauwens M, De Man V, Audo I, Balikova I, Zein WM, Smirnov V, Held S, Vermeer S, Loos E, Jacob J, Casteels I, Désir J, Depasse F, Van de Sompele S, Van Heetvelde M, De Bruyne M, Andrieu C, Condroyer C, Antonio A, Hufnagel R, Carvalho AL, Marques JP, Zeitz C, De Baere E, Damme M. Expanding the genetic landscape of Usher syndrome type IV caused by pathogenic ARSG variants. Clin Genet 2024. [PMID: 39199020 DOI: 10.1111/cge.14614] [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: 05/13/2024] [Revised: 07/26/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024]
Abstract
Usher syndrome (USH) is the most common cause of deafblindness. USH is autosomal recessively inherited and characterized by rod-cone dystrophy or retinitis pigmentosa (RP), often accompanied by sensorineural hearing loss. Variants in >15 genes have been identified as causative for clinically and genetically distinct subtypes. Among the ultra-rare and recently discovered genes is ARSG, coding for the lysosomal sulfatase Arylsulfatase G. This subtype was assigned as "USH IV" with a late onset of RP and usually late-onset progressive SNHL without vestibular involvement. Here, we describe nine new subjects and the clinical description of four cases with the USH IV phenotype bearing seven novel and two known pathogenic variants. Functional experiments indicated the complete loss of sulfatase enzymatic activity upon ectopic expression of mutated ARSG cDNA. Interestingly, we identified a homozygous missense variant, p.(Arg99His), previously described in dogs with neuronal ceroid lipofuscinosis. Our study expands the genetic landscape of ARSG-USH IV and the number of known subjects by more than 30%. These findings highlight that USH IV likely has been underdiagnosed and emphasize the need to test molecularly unresolved subjects with deafblindness syndrome. Finally, testing of ARSG should be considered for the genetic work-up of apparent isolated inherited retinal diseases.
Collapse
Affiliation(s)
- Miriam Bauwens
- Center for Medical Genetics, Ghent University Hospital & Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Vincent De Man
- Department of Ophthalmology, University Hospital Leuven, Leuven, Belgium
| | - Isabelle Audo
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
- Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, Centre de Référence Maladies Rares REFERET and INSERM-DGOS CIC 1423, Paris, France
| | - Irina Balikova
- Department of Ophthalmology, University Hospital Leuven, Leuven, Belgium
| | - Wadih M Zein
- Ophthalmic Genetics and Visual Function Branch, NEI, NIH, Bethesda, Maryland, USA
| | - Vasily Smirnov
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
- Exploration de la Vision et Neuro-Ophtalmologie, CHU de Lille, Lille, France
| | - Sebastian Held
- Christian-Albrechts-University Kiel, Institute of Biochemistry, Kiel, Germany
| | - Sascha Vermeer
- Center for Human Genetics, University Hospitals Leuven, Leuven, Belgium
| | - Elke Loos
- Department of Otorhinolaryngology-Head and Neck Surgery, University Hospitals Leuven, Leuven, Belgium
- Department of Neurosciences, Research Group Experimental Oto-Rhino-Laryngology (ExpORL), KU Leuven, University of Leuven, Leuven, Belgium
| | - Julie Jacob
- Department of Ophthalmology, University Hospital Leuven, Leuven, Belgium
| | - Ingele Casteels
- Department of Ophthalmology, University Hospital Leuven, Leuven, Belgium
- Biomedical Sciences Group, Department of Neurosciences, Research Group Ophthalmology, Leuven, Belgium
| | - Julie Désir
- Center for Medical Genetics, Institut de Pathologie et de Génétique Gosselies, Charleroi, Belgium
| | - Fanny Depasse
- Department of Pediatric Ophthalmology, Queen Fabiola Children's University Hospital (HUDERF), Brussels, Belgium
| | - Stijn Van de Sompele
- Center for Medical Genetics, Ghent University Hospital & Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Mattias Van Heetvelde
- Center for Medical Genetics, Ghent University Hospital & Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Marieke De Bruyne
- Center for Medical Genetics, Ghent University Hospital & Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Camille Andrieu
- Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, Centre de Référence Maladies Rares REFERET and INSERM-DGOS CIC 1423, Paris, France
| | | | - Aline Antonio
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Robert Hufnagel
- Ophthalmic Genetics and Visual Function Branch, NEI, NIH, Bethesda, Maryland, USA
- Pathology Department, Genetics Department, Center for Integrated Healthcare Research, Kaiser Permanente, Honolulu, Hawaii, USA
| | - Ana Luísa Carvalho
- Medical Genetics Unit, Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- University Clinic of Medical Genetics, Faculty of Medicine, University of Coimbra (FMUC), Coimbra, Portugal
| | - João Pedro Marques
- University Clinic of Medical Genetics, Faculty of Medicine, University of Coimbra (FMUC), Coimbra, Portugal
- Ophthalmology Unit, Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal
- University Clinic of Ophthalmology, Faculty of Medicine, University of Coimbra (FMUC), Coimbra, Portugal
- Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra (FMUC), Coimbra, Portugal
| | - Christina Zeitz
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Elfride De Baere
- Center for Medical Genetics, Ghent University Hospital & Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Markus Damme
- Christian-Albrechts-University Kiel, Institute of Biochemistry, Kiel, Germany
| |
Collapse
|
48
|
Ramsey ML, Heald B, Gokun Y, Baker J, Groce JR, Han S, Hart PA, Krishna SG, Lara LF, Lee PJ, Papachristou GI, Pearlman R, Poll S, Roberts ME, Stanich PP. Germline multigene panel testing in acute and chronic pancreatitis. PLoS One 2024; 19:e0307076. [PMID: 39172977 PMCID: PMC11341018 DOI: 10.1371/journal.pone.0307076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 06/29/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND/OBJECTIVES Germline genetic testing is recommended for younger patients with idiopathic pancreatitis but there has been a lack of consensus in recommendations for those over age 35. We aimed to analyze the results of genetic testing among subjects of varying ages. METHODS Individuals who underwent germline multigene testing for pancreatitis susceptibility genes (CASR, CFTR, CPA1, CTRC, PRSS1, SPINK1) through a large commercial laboratory between 2017 and 2022 were included. Test results and information collected from test requisition forms were evaluated. Multivariable logistic regression models were performed to identify factors associated with a positive pancreatitis panel (pathogenic, likely pathogenic, and/or increased risk variants) in pancreatitis-related genes. RESULTS Overall, 2,468 subjects with primary indication of acute pancreatitis (AP) (n = 401), chronic pancreatitis (CP) (n = 631), pancreatic cancer (n = 128), or other indications (n = 1,308) completed germline testing. Among patients with AP or CP, the prevalence of any positive result for those <35 versus ≥35 years of age was 32.1% and 24.5% (p = 0.007), and the prevalence of a clinically meaningful result was 10.8% and 5.4%, respectively (p = 0.001). Positive family history of pancreatitis was associated with increased odds ratio (OR) of 8.59 (95% confidence interval (CI) 2.92-25.25) for a clinically significant panel result while each 5-year increase in age at test completion had lower odds (OR 0.89, 95% CI 0.83-0.95). CONCLUSIONS The highest prevalence of pathogenic variants is seen in younger individuals with a positive family history of pancreatitis. However, clinically meaningful results are identified in older subjects, suggesting that genetic counseling and testing should be considered for all age groups.
Collapse
Affiliation(s)
- Mitchell L. Ramsey
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Brandie Heald
- Medical Affairs, Invitae Corporation, San Francisco, California, United States of America
| | - Yevgeniya Gokun
- Department of Biomedical Informatics, Center for Biostatistics, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Josie Baker
- Division of Human Genetics, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - J. Royce Groce
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Samuel Han
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Phil A. Hart
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Somashekar G. Krishna
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Luis F. Lara
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Peter J. Lee
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Georgios I. Papachristou
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Rachel Pearlman
- Division of Human Genetics, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Sarah Poll
- Medical Affairs, Invitae Corporation, San Francisco, California, United States of America
| | - Maegan E. Roberts
- Division of Human Genetics, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Peter P. Stanich
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| |
Collapse
|
49
|
Balla C, Margutti A, De Carolis B, Canovi L, Di Domenico A, Vivaldi I, Vitali F, De Raffele M, Malagù M, Sassone B, Biffi M, Selvatici R, Ferlini A, Gualandi F, Bertini M. Cardiac conduction disorders in young adults: Clinical characteristics and genetic background of an underestimated population. Heart Rhythm 2024; 21:1363-1369. [PMID: 38467355 DOI: 10.1016/j.hrthm.2024.03.008] [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: 01/09/2024] [Revised: 03/04/2024] [Accepted: 03/05/2024] [Indexed: 03/13/2024]
Abstract
BACKGROUND Cardiac conduction disorder (CCD) in patients <50 years old is a rare and mostly unknown condition. OBJECTIVE We aimed to assess clinical characteristics and genetic background of patients <50 years old with CCD of unknown origin. METHODS We retrospectively reviewed a consecutive series of patients with a diagnosis of CCD before the age of 50 years referred to our center between January 2019 and December 2021. Patients underwent complete clinical examination and genetic evaluation. RESULTS We enrolled 39 patients with a median age of 40 years (28-47 years) at the onset of symptoms. A cardiac implantable electronic device was implanted in 69% of the patients. In 15 of 39 CCD index patients (38%), we found a total of 13 different gene variations (3 pathogenic, 6 likely pathogenic, and 4 variants of uncertain significance), mostly in 3 genes (SCN5A, TRPM4, and LMNA). In our cohort, genetic testing led to the decision to implant an implantable cardioverter-defibrillator in 2 patients for the increased risk of sudden cardiac death. CONCLUSION Patients with the occurrence of CCD before the age of 50 years present with a high rate of pathologic gene variations, mostly in 3 genes (SCN5A, TRPM4, and LMNA). The presence of pathogenic variations may add information about the prognosis and lead to an individualized therapeutic approach.
Collapse
Affiliation(s)
- Cristina Balla
- Cardiology Unit, Azienda Ospedaliero-Universitaria di Ferrara, Cona (Ferrara), Italy.
| | - Alice Margutti
- Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Beatrice De Carolis
- Cardiology Unit, Azienda Ospedaliero-Universitaria di Ferrara, Cona (Ferrara), Italy
| | - Luca Canovi
- Cardiology Unit, Azienda Ospedaliero-Universitaria di Ferrara, Cona (Ferrara), Italy
| | - Assunta Di Domenico
- Cardiology Unit, Azienda Ospedaliero-Universitaria di Ferrara, Cona (Ferrara), Italy
| | - Ilaria Vivaldi
- Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Francesco Vitali
- Cardiology Unit, Azienda Ospedaliero-Universitaria di Ferrara, Cona (Ferrara), Italy
| | - Martina De Raffele
- Cardiology Unit, Azienda Ospedaliero-Universitaria di Ferrara, Cona (Ferrara), Italy
| | - Michele Malagù
- Cardiology Unit, Azienda Ospedaliero-Universitaria di Ferrara, Cona (Ferrara), Italy
| | - Biagio Sassone
- Division of Cardiology, SS.ma Annunziata Hospital, Department of Emergency, AUSL Ferrara, Cento (Ferrara), Italy
| | - Mauro Biffi
- Cardiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Rita Selvatici
- Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Alessandra Ferlini
- Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Francesca Gualandi
- Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Matteo Bertini
- Cardiology Unit, Azienda Ospedaliero-Universitaria di Ferrara, Cona (Ferrara), Italy
| |
Collapse
|
50
|
Goutman SA, Goyal NA, Payne K, Paisán‐Ruiz C, Kupelian V, Kang ML, Mitchell AA, Fecteau TE. ALS Identified: two-year findings from a sponsored ALS genetic testing program. Ann Clin Transl Neurol 2024; 11:2201-2211. [PMID: 39044379 PMCID: PMC11330217 DOI: 10.1002/acn3.52140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 06/21/2024] [Accepted: 06/22/2024] [Indexed: 07/25/2024] Open
Abstract
OBJECTIVE To report initial results from the Amyotrophic Lateral Sclerosis (ALS) Identified genetic testing (GT) program on characteristics of individuals tested and frequency of reported disease-causing variants. METHODS ALS Identified used the Invitae Amyotrophic Lateral Sclerosis panel (Invitae, San Francisco, CA, USA) to assay 22 ALS-associated genes. Sponsored by Biogen (Cambridge, MA, USA), the program was launched in June 2021 and was available at no charge to individuals ≥18 years in the United States and Puerto Rico with an ALS diagnosis or a known family history of ALS. Deidentified data were available to Biogen. RESULTS As of 26 October 2023, 998 healthcare professionals ordered the panel at 681 unique care sites. Of 8054 individuals examined, 7483 (92.9%) were reported to have a clinical diagnosis of ALS, while 571 (7.1%) were asymptomatic relatives. Of the individuals with a clinical ALS diagnosis, 57.7% were male (n = 4319) and 42.3% female (n = 3164). Mean (SD) age at diagnosis is 62 (13) years. Out of the 7483 clinically diagnosed individuals, 1810 (24.2%) showed genetic variations in ALS-associated genes. Among these, 865 individuals (47.8%) carried pathogenic variants, and 44 (2.4%) had likely pathogenic variants, totaling 12.1% of the clinically diagnosed population. INTERPRETATION Since 2021 there has been robust uptake and sustained use of the ALS Identified program, one of the largest samples of people with ALS to date across the United States, demonstrating the interest and need for genetic ALS testing.
Collapse
|