1
|
Tsuchida N, Uchiyama Y, Hamanaka K, Okamoto N, Fujimoto A, Enoki H, Koshimizu E, Fujita A, Misawa K, Miyatake S, Mizuguchi T, Matsumoto N. Mosaic deletions detected by genome sequencing in two families. J Hum Genet 2025; 70:307-312. [PMID: 40199966 DOI: 10.1038/s10038-025-01336-y] [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: 02/20/2025] [Revised: 03/18/2025] [Accepted: 03/27/2025] [Indexed: 04/10/2025]
Abstract
Trio-based genome sequencing (GS) is useful for genetic analysis of cases in which exome sequencing failed to resolve the disease-causing variants. In this paper, we report two unrelated families with pathogenic deletions (one outside exome-covering genomic regions and the other involving a single exon) successfully identified by GS. Notably, mosaic deletions were found in both families, which were carefully evaluated in detail by analyzing GS data using Integrative Genomics Viewer, breakpoint PCR, quantitative PCR, and digital PCR. This study emphasizes the benefit of trio-based GS, enabling straightforward interpretation, further aided by other confirmatory experimental methods.
Collapse
Affiliation(s)
- Naomi Tsuchida
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Department of Rare Disease Genomics, Yokohama City University Hospital, Yokohama, Japan
| | - Yuri Uchiyama
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Department of Rare Disease Genomics, Yokohama City University Hospital, Yokohama, Japan
| | - Kohei Hamanaka
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Nobuhiko Okamoto
- Department of Medical Genetics, Osaka Women's and Children's Hospital, Osaka, Japan
| | - Ayataka Fujimoto
- Center of Epilepsy, and Neurological Disorder, Seirei Hamamatsu General Hospital, Hamamatsu, Japan
| | - Hideo Enoki
- Center of Epilepsy, and Neurological Disorder, Seirei Hamamatsu General Hospital, Hamamatsu, Japan
| | - Eriko Koshimizu
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Atsushi Fujita
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kazuharu Misawa
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Satoko Miyatake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Department of Clinical Genetics, Yokohama City University Hospital, Yokohama, Japan
| | - Takeshi Mizuguchi
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
- Department of Rare Disease Genomics, Yokohama City University Hospital, Yokohama, Japan.
- Department of Clinical Genetics, Yokohama City University Hospital, Yokohama, Japan.
| |
Collapse
|
2
|
Ghorbani M, Moosa S, Siddig Z, Farhad R, Naeem H, Harvey WT, Mastrorosa FK, Munson KM, Mohamad Razali R, Aliyev E, Diboun I, Abouelhassan R, Tauro M, Hassan S, Mathew R, Al Hashmi M, Mathew LS, Wang K, Salhab AR, Vempalli FR, El Khouly A, Alazwani I, Tomei S, Fakhro KA, Satti A, Benini R, Rhie A, Eichler EE, Mokrab Y. Near-complete Middle Eastern genomes refine autozygosity and enhance disease-causing and population-specific variant discovery. Nat Genet 2025; 57:1119-1131. [PMID: 40325133 DOI: 10.1038/s41588-025-02173-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 03/18/2025] [Indexed: 05/07/2025]
Abstract
Advances in long-read sequencing have enabled routine complete assembly of human genomes, but much remains to be done to represent broader populations and show impact on disease-gene discovery. Here, we report highly accurate, near-complete and phased genomes from six Middle Eastern (ME) family trios (n = 18) with neurodevelopmental conditions, representing ancestries from Sudan, Jordan, Syria, Qatar and Afghanistan. These genomes revealed 42.2 Mb of new sequence (13.8% impacting known genes), 75 new HLA/KIR alleles and strong signals of inbreeding, with ROH covering up to one-third of chromosomes 6 and 12 in one individual. Using assembly-based variant calling, we identified 23 de novo and recessive variants as strong candidates for causing previously unresolved symptoms in the probands. The ME genomes revealed unique variation relative to existing references, showing enhanced mappability and variant calling. These results underscore the value of de novo assembly for disease variant discovery and the need for sampled ME-specific references to better characterize population-relevant variation.
Collapse
Affiliation(s)
| | | | | | | | | | - William T Harvey
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | | | - Katherine M Munson
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Rozaimi Mohamad Razali
- Department of Biomedical Science, College of Health Sciences, Qatar University, Doha, Qatar
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Khalid A Fakhro
- Sidra Medicine, Doha, Qatar
- Department of Genetic Medicine, Weill Cornell Medicine, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | | | - Ruba Benini
- Sidra Medicine, Doha, Qatar
- Department of Genetic Medicine, Weill Cornell Medicine, Doha, Qatar
| | - Arang Rhie
- National Human Genome Research Institute, Bethesda, MD, USA
| | - Evan E Eichler
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | - Younes Mokrab
- Sidra Medicine, Doha, Qatar.
- Department of Biomedical Science, College of Health Sciences, Qatar University, Doha, Qatar.
- Department of Genetic Medicine, Weill Cornell Medicine, Doha, Qatar.
| |
Collapse
|
3
|
Ishorst N, Hölzel S, Greve C, Yilmaz Ö, Lindenberg T, Lambertz J, Drichel D, Zametica B, Mingardo E, Kalanithy JC, Channab K, Kibris D, Henne S, Degenhardt F, Siewert A, Dixon M, Kruse T, Ongkosuwito E, Girisha KM, Pande S, Nowak S, Hagelueken G, Geyer M, Carels C, van Rooij IALM, Ludwig KU, Odermatt B, Mangold E. Role of ZFHX4 in orofacial clefting based on human genetic data and zebrafish models. Eur J Hum Genet 2025; 33:595-606. [PMID: 39702590 PMCID: PMC7617551 DOI: 10.1038/s41431-024-01775-9] [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: 03/11/2024] [Revised: 12/03/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024] Open
Abstract
Orofacial clefting (OFC) is a frequent congenital anomaly and can occur either in the context of underlying syndromes or in isolation (nonsyndromic). The two common OFC phenotypes are cleft lip with/without cleft palate (CL/P) and cleft palate only (CPO). In this study, we searched for penetrant CL/P genes, by evaluating de novo copy number variants (CNV) from an exome sequencing dataset of 50 nonsyndromic patient-parent trios. We detected a heterozygous 86 kb de novo deletion affecting exons 4-11 of ZFHX4, a gene previously associated with OFC. Genetic and phenotypic data from our in-house and the AGORA cohort (710 and 229 individuals with nonsyndromic CL/P) together with literature and database reviews demonstrate that ZFHX4 variants can lead to both nonsyndromic and syndromic forms not only of CL/P but also CPO. Expression analysis in published single-cell RNA-sequencing data (mouse embryo, zebrafish larva) at relevant time-points support an important role of Zfhx4/zfhx4 in craniofacial development. To characterize the role of zfhx4 in zebrafish craniofacial development, we knocked out/down the zebrafish orthologue. Cartilage staining of the zfhx4 CRISPR F0 knockout and morpholino knockdown at 4 days post-fertilization showed an underdeveloped and abnormally shaped ethmoid plate and cartilaginous jaw (resembling micrognathia). While there is evidence for the dominant inheritance of ZFHX4 variants in OFC, we here present a patient with a possible recessive inheritance. In conclusion, ZFHX4 has a highly heterogeneous phenotypic spectrum and variable mode of inheritance. Our data highlight that ZFHX4 should be considered in genetic testing in patients with nonsyndromic clefting.
Collapse
Affiliation(s)
- Nina Ishorst
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany.
- Institute of Anatomy, Division of Neuroanatomy, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany.
| | - Selina Hölzel
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
- Institute of Anatomy and Cell Biology, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Carola Greve
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
- LOEWE Centre for Translational Biodiversity Genomics, Frankfurt am Main, Germany
| | - Öznur Yilmaz
- Institute of Anatomy, Division of Neuroanatomy, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Tobias Lindenberg
- Institute of Anatomy, Division of Neuroanatomy, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Jessica Lambertz
- Institute of Anatomy, Division of Neuroanatomy, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Dmitriy Drichel
- Cologne Center for Genomics, University of Cologne, Cologne, Germany
| | - Berina Zametica
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Enrico Mingardo
- Institute of Anatomy and Cell Biology, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
- Faculty of Life Science, Nutritional Biochemistry, University of Bayreuth, Bayreuth, Germany
| | - Jeshurun C Kalanithy
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
- Institute of Anatomy, Division of Neuroanatomy, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Khadija Channab
- Institute of Anatomy and Cell Biology, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Duygu Kibris
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
- Institute of Anatomy, Division of Neuroanatomy, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Sabrina Henne
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Franziska Degenhardt
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Duisburg, Germany
| | - Anna Siewert
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Michael Dixon
- Faculty of Biology, Medicine & Health, University of Manchester, Manchester, M13 9PL, UK
| | - Teresa Kruse
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Orthodontics, Cologne, Germany
| | - Edwin Ongkosuwito
- Department of Dentistry, Section of Orthodontics and Craniofacial Biology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Katta M Girisha
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Shruti Pande
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Stefanie Nowak
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | | | - Matthias Geyer
- Institute of Structural Biology, University of Bonn, Bonn, Germany
| | - Carine Carels
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Iris A L M van Rooij
- IQ Health Science Department, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kerstin U Ludwig
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Benjamin Odermatt
- Institute of Anatomy, Division of Neuroanatomy, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany.
- Institute of Anatomy and Cell Biology, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany.
| | - Elisabeth Mangold
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany.
| |
Collapse
|
4
|
Chen L, Li S, Xie F, Hu X, Lv W. Cerebral small vessel disease associated with COL4A1 and COL4A2 duplication: clinical and MRI features resembling CADASIL. Neurol Sci 2025:10.1007/s10072-025-08175-x. [PMID: 40261521 DOI: 10.1007/s10072-025-08175-x] [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: 12/02/2024] [Accepted: 04/06/2025] [Indexed: 04/24/2025]
Abstract
BACKGROUND Large duplications or triplications involving the 13q33-34 chromosomal region, which encompass the COL4A1 and COL4A2 genes, have been reported in association with cerebral small vessel disease (CSVD) in a few patients. Herein, we report an additional case of CSVD linked to a duplication of COL4A1 and COL4A2 and provide a detailed summary of the associated clinical and MRI findings. METHODS A patient with CSVD underwent detailed clinical and neuroimaging evaluations. Targeted next-generation sequencing (NGS) and copy number variation sequencing (CNV-seq) based on whole genome sequencing were used to identify the genetic basis of the disease. RESULTS The patient experienced his first ischemic stroke at age 51. Cranial MRI revealed extensive acute and chronic lacunar infarcts and white matter hyperintensities across both cerebral hemispheres, with involvement of the anterior temporal lobe and the external capsule. Bilateral thalamic microbleeds were also noted. The clinical features and MRI findings are similar to those observed in cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL). Targeted NGS and CNV-seq analysis identified a duplication in the region of chromosome 13, which included the COL4A1 and COL4A2 genes. CONCLUSIONS This case provides further evidence supporting the association of CNVs in COL4A1 and COL4A2 with CSVD. When hereditary CSVD is suspected and no micro-mutations in CSVD-associated genes are identified, CNV analysis of the 13q region should be considered.
Collapse
Affiliation(s)
- Lili Chen
- Department of Neurology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
| | - Shuang Li
- Department of Neurology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
| | - Fei Xie
- Department of Neurology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
| | - Xingyue Hu
- Department of Neurology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China.
| | - Wen Lv
- Department of Neurology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China.
| |
Collapse
|
5
|
Rodilla C, Núñez-Moreno G, Benitez Y, Rodríguez de Alba M, Blanco-Kelly F, López-Alcojor A, Fernández-Caballero L, Perea-Romero I, Del Pozo-Valero M, García-García G, Balanzá M, Villaverde C, Zurita O, Jubin C, Fund C, Delepine M, Leduc A, Deleuze JF, Millán JM, Minguez P, Corton M, Ayuso C. Long-Read Whole-Genome Sequencing as a Tool for Variant Detection in Inherited Retinal Dystrophies. Int J Mol Sci 2025; 26:3825. [PMID: 40332496 PMCID: PMC12027592 DOI: 10.3390/ijms26083825] [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: 02/12/2025] [Revised: 04/07/2025] [Accepted: 04/15/2025] [Indexed: 05/08/2025] Open
Abstract
Advances in whole-genome sequencing (WGS) have significantly enhanced our ability to detect genomic variants underlying inherited diseases. In this study, we performed long-read WGS on 24 patients with inherited retinal dystrophies (IRDs) to validate the utility of nanopore sequencing in detecting genomic variations. We confirmed the presence of all previously detected variants and demonstrated that this approach allows for the precise refinement of structural variants (SVs). Furthermore, we could perform genotype phasing by sequencing only the probands, confirming that the variants were inherited in trans. Moreover, nanopore sequencing enables the detection of complex variants, such as transposon insertions and structural rearrangements. This comprehensive assessment illustrates the power of long-read sequencing in capturing diverse forms of genomic variation and in improving diagnostic accuracy in IRDs.
Collapse
Affiliation(s)
- Cristina Rodilla
- Department of Genetics & Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; (C.R.); (G.N.-M.); (Y.B.); (M.R.d.A.); (F.B.-K.); (A.L.-A.); (L.F.-C.); (I.P.-R.); (M.D.P.-V.); (C.V.); (O.Z.); (P.M.)
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain; (G.G.-G.); (M.B.); (J.M.M.)
| | - Gonzalo Núñez-Moreno
- Department of Genetics & Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; (C.R.); (G.N.-M.); (Y.B.); (M.R.d.A.); (F.B.-K.); (A.L.-A.); (L.F.-C.); (I.P.-R.); (M.D.P.-V.); (C.V.); (O.Z.); (P.M.)
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain; (G.G.-G.); (M.B.); (J.M.M.)
- Bioinformatics Unit, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain
| | - Yolanda Benitez
- Department of Genetics & Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; (C.R.); (G.N.-M.); (Y.B.); (M.R.d.A.); (F.B.-K.); (A.L.-A.); (L.F.-C.); (I.P.-R.); (M.D.P.-V.); (C.V.); (O.Z.); (P.M.)
- Bioinformatics Unit, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain
| | - Marta Rodríguez de Alba
- Department of Genetics & Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; (C.R.); (G.N.-M.); (Y.B.); (M.R.d.A.); (F.B.-K.); (A.L.-A.); (L.F.-C.); (I.P.-R.); (M.D.P.-V.); (C.V.); (O.Z.); (P.M.)
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain; (G.G.-G.); (M.B.); (J.M.M.)
| | - Fiona Blanco-Kelly
- Department of Genetics & Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; (C.R.); (G.N.-M.); (Y.B.); (M.R.d.A.); (F.B.-K.); (A.L.-A.); (L.F.-C.); (I.P.-R.); (M.D.P.-V.); (C.V.); (O.Z.); (P.M.)
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain; (G.G.-G.); (M.B.); (J.M.M.)
| | - Aroa López-Alcojor
- Department of Genetics & Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; (C.R.); (G.N.-M.); (Y.B.); (M.R.d.A.); (F.B.-K.); (A.L.-A.); (L.F.-C.); (I.P.-R.); (M.D.P.-V.); (C.V.); (O.Z.); (P.M.)
| | - Lidia Fernández-Caballero
- Department of Genetics & Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; (C.R.); (G.N.-M.); (Y.B.); (M.R.d.A.); (F.B.-K.); (A.L.-A.); (L.F.-C.); (I.P.-R.); (M.D.P.-V.); (C.V.); (O.Z.); (P.M.)
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain; (G.G.-G.); (M.B.); (J.M.M.)
| | - Irene Perea-Romero
- Department of Genetics & Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; (C.R.); (G.N.-M.); (Y.B.); (M.R.d.A.); (F.B.-K.); (A.L.-A.); (L.F.-C.); (I.P.-R.); (M.D.P.-V.); (C.V.); (O.Z.); (P.M.)
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain; (G.G.-G.); (M.B.); (J.M.M.)
| | - Marta Del Pozo-Valero
- Department of Genetics & Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; (C.R.); (G.N.-M.); (Y.B.); (M.R.d.A.); (F.B.-K.); (A.L.-A.); (L.F.-C.); (I.P.-R.); (M.D.P.-V.); (C.V.); (O.Z.); (P.M.)
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain; (G.G.-G.); (M.B.); (J.M.M.)
| | - Gema García-García
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain; (G.G.-G.); (M.B.); (J.M.M.)
- Molecular, Cellular and Genomics Biomedicine, Health Research Institute La Fe, 46026 Valencia, Spain
| | - Mar Balanzá
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain; (G.G.-G.); (M.B.); (J.M.M.)
- Molecular, Cellular and Genomics Biomedicine, Health Research Institute La Fe, 46026 Valencia, Spain
| | - Cristina Villaverde
- Department of Genetics & Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; (C.R.); (G.N.-M.); (Y.B.); (M.R.d.A.); (F.B.-K.); (A.L.-A.); (L.F.-C.); (I.P.-R.); (M.D.P.-V.); (C.V.); (O.Z.); (P.M.)
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain; (G.G.-G.); (M.B.); (J.M.M.)
| | - Olga Zurita
- Department of Genetics & Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; (C.R.); (G.N.-M.); (Y.B.); (M.R.d.A.); (F.B.-K.); (A.L.-A.); (L.F.-C.); (I.P.-R.); (M.D.P.-V.); (C.V.); (O.Z.); (P.M.)
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain; (G.G.-G.); (M.B.); (J.M.M.)
| | - Claire Jubin
- Centre National de Recherche en Génomique Humaine, Université Paris-Saclay, 91057 Evry, France; (C.J.); (C.F.); (M.D.); (A.L.); (J.-F.D.)
| | - Cedric Fund
- Centre National de Recherche en Génomique Humaine, Université Paris-Saclay, 91057 Evry, France; (C.J.); (C.F.); (M.D.); (A.L.); (J.-F.D.)
| | - Marc Delepine
- Centre National de Recherche en Génomique Humaine, Université Paris-Saclay, 91057 Evry, France; (C.J.); (C.F.); (M.D.); (A.L.); (J.-F.D.)
| | - Aurelie Leduc
- Centre National de Recherche en Génomique Humaine, Université Paris-Saclay, 91057 Evry, France; (C.J.); (C.F.); (M.D.); (A.L.); (J.-F.D.)
| | - Jean-François Deleuze
- Centre National de Recherche en Génomique Humaine, Université Paris-Saclay, 91057 Evry, France; (C.J.); (C.F.); (M.D.); (A.L.); (J.-F.D.)
| | - José M. Millán
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain; (G.G.-G.); (M.B.); (J.M.M.)
- Molecular, Cellular and Genomics Biomedicine, Health Research Institute La Fe, 46026 Valencia, Spain
| | - Pablo Minguez
- Department of Genetics & Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; (C.R.); (G.N.-M.); (Y.B.); (M.R.d.A.); (F.B.-K.); (A.L.-A.); (L.F.-C.); (I.P.-R.); (M.D.P.-V.); (C.V.); (O.Z.); (P.M.)
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain; (G.G.-G.); (M.B.); (J.M.M.)
- Bioinformatics Unit, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain
| | - Marta Corton
- Department of Genetics & Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; (C.R.); (G.N.-M.); (Y.B.); (M.R.d.A.); (F.B.-K.); (A.L.-A.); (L.F.-C.); (I.P.-R.); (M.D.P.-V.); (C.V.); (O.Z.); (P.M.)
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain; (G.G.-G.); (M.B.); (J.M.M.)
| | - Carmen Ayuso
- Department of Genetics & Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; (C.R.); (G.N.-M.); (Y.B.); (M.R.d.A.); (F.B.-K.); (A.L.-A.); (L.F.-C.); (I.P.-R.); (M.D.P.-V.); (C.V.); (O.Z.); (P.M.)
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain; (G.G.-G.); (M.B.); (J.M.M.)
| |
Collapse
|
6
|
Li Q, Keskus AG, Wagner J, Izydorczyk MB, Timp W, Sedlazeck FJ, Klein AP, Zook JM, Kolmogorov M, Schatz MC. Unraveling the hidden complexity of cancer through long-read sequencing. Genome Res 2025; 35:599-620. [PMID: 40113261 PMCID: PMC12047254 DOI: 10.1101/gr.280041.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Cancer is fundamentally a disease of the genome, characterized by extensive genomic, transcriptomic, and epigenomic alterations. Most current studies predominantly use short-read sequencing, gene panels, or microarrays to explore these alterations; however, these technologies can systematically miss or misrepresent certain types of alterations, especially structural variants, complex rearrangements, and alterations within repetitive regions. Long-read sequencing is rapidly emerging as a transformative technology for cancer research by providing a comprehensive view across the genome, transcriptome, and epigenome, including the ability to detect alterations that previous technologies have overlooked. In this Perspective, we explore the current applications of long-read sequencing for both germline and somatic cancer analysis. We provide an overview of the computational methodologies tailored to long-read data and highlight key discoveries and resources within cancer genomics that were previously inaccessible with prior technologies. We also address future opportunities and persistent challenges, including the experimental and computational requirements needed to scale to larger sample sizes, the hurdles in sequencing and analyzing complex cancer genomes, and opportunities for leveraging machine learning and artificial intelligence technologies for cancer informatics. We further discuss how the telomere-to-telomere genome and the emerging human pangenome could enhance the resolution of cancer genome analysis, potentially revolutionizing early detection and disease monitoring in patients. Finally, we outline strategies for transitioning long-read sequencing from research applications to routine clinical practice.
Collapse
Affiliation(s)
- Qiuhui Li
- Department of Computer Science, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Ayse G Keskus
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Justin Wagner
- Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, USA
| | - Michal B Izydorczyk
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Winston Timp
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Fritz J Sedlazeck
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Texas 77030, USA
- Department of Computer Science, Rice University, Houston, Texas 77251, USA
| | - Alison P Klein
- Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins Medicine, Baltimore, Maryland 21031, USA
| | - Justin M Zook
- Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, USA
| | - Mikhail Kolmogorov
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA;
| | - Michael C Schatz
- Department of Computer Science, Johns Hopkins University, Baltimore, Maryland 21218, USA;
- Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins Medicine, Baltimore, Maryland 21031, USA
| |
Collapse
|
7
|
Mahmoud M, Agustinho DP, Sedlazeck FJ. A Hitchhiker's Guide to long-read genomic analysis. Genome Res 2025; 35:545-558. [PMID: 40228901 PMCID: PMC12047252 DOI: 10.1101/gr.279975.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
Over the past decade, long-read sequencing has evolved into a pivotal technology for uncovering the hidden and complex regions of the genome. Significant cost efficiency, scalability, and accuracy advancements have driven this evolution. Concurrently, novel analytical methods have emerged to harness the full potential of long reads. These advancements have enabled milestones such as the first fully completed human genome, enhanced identification and understanding of complex genomic variants, and deeper insights into the interplay between epigenetics and genomic variation. This mini-review provides a comprehensive overview of the latest developments in long-read DNA sequencing analysis, encompassing reference-based and de novo assembly approaches. We explore the entire workflow, from initial data processing to variant calling and annotation, focusing on how these methods improve our ability to interpret a wide array of genomic variants. Additionally, we discuss the current challenges, limitations, and future directions in the field, offering a detailed examination of the state-of-the-art bioinformatics methods for long-read sequencing.
Collapse
Affiliation(s)
- Medhat Mahmoud
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Daniel P Agustinho
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Fritz J Sedlazeck
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA;
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Computer Science, Rice University, Houston, Texas 77005, USA
| |
Collapse
|
8
|
Steyaert W, Sagath L, Demidov G, Yépez VA, Esteve-Codina A, Gagneur J, Ellwanger K, Derks R, Weiss M, den Ouden A, van den Heuvel S, Swinkels H, Zomer N, Steehouwer M, O'Gorman L, Astuti G, Neveling K, Schüle R, Xu J, Synofzik M, Beijer D, Hengel H, Schöls L, Claeys KG, Baets J, Van de Vondel L, Ferlini A, Selvatici R, Morsy H, Saeed Abd Elmaksoud M, Straub V, Müller J, Pini V, Perry L, Sarkozy A, Zaharieva I, Muntoni F, Bugiardini E, Polavarapu K, Horvath R, Reid E, Lochmüller H, Spinazzi M, Savarese M, Matalonga L, Laurie S, Brunner HG, Graessner H, Beltran S, Ossowski S, Vissers LELM, Gilissen C, Hoischen A. Unraveling undiagnosed rare disease cases by HiFi long-read genome sequencing. Genome Res 2025; 35:755-768. [PMID: 40138663 PMCID: PMC12047270 DOI: 10.1101/gr.279414.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 02/21/2025] [Indexed: 03/29/2025]
Abstract
Solve-RD is a pan-European rare disease (RD) research program that aims to identify disease-causing genetic variants in previously undiagnosed RD families. We utilized 10-fold coverage HiFi long-read sequencing (LRS) for detecting causative structural variants (SVs), single-nucleotide variants (SNVs), insertion-deletions (indels), and short tandem repeat (STR) expansions in previously studied RD families without a clear molecular diagnosis. Our cohort includes 293 individuals from 114 genetically undiagnosed RD families selected by European Reference Network (ERN) experts. Of these, 21 families were affected by so-called "unsolvable" syndromes for which genetic causes remain unknown and for which prior testing was not a prerequisite. The remaining 93 families had at least one individual affected by a rare neurological, neuromuscular, or epilepsy disorder without a genetic diagnosis despite extensive prior testing. Clinical interpretation and orthogonal validation of variants in known disease genes yielded 12 novel genetic diagnoses due to de novo and rare inherited SNVs, indels, SVs, and STR expansions. In an additional five families, we identified a candidate disease-causing variant, including an MCF2/FGF13 fusion and a PSMA3 deletion. However, no common genetic cause was identified in any of the "unsolvable" syndromes. Taken together, we found (likely) disease-causing genetic variants in 11.8% of previously unsolved families and additional candidate disease-causing SVs in another 5.4% of these families. In conclusion, our results demonstrate the potential added value of HiFi long-read genome sequencing in undiagnosed rare diseases.
Collapse
Affiliation(s)
- Wouter Steyaert
- Radboud University Medical Center, Department of Human Genetics, Research Institute for Medical Innovation, 6500 HB Nijmegen, Netherlands
| | - Lydia Sagath
- Radboud University Medical Center, Department of Human Genetics, Research Institute for Medical Innovation, 6500 HB Nijmegen, Netherlands
| | - German Demidov
- Universitätsklinikum Tübingen - Institut für Medizinische Genetik und angewandte Genomik, 72076 Tübingen, Germany
| | - Vicente A Yépez
- TUM School of Computation, Information and Technology, Technical University of Munich, 85748 Garching, Germany
| | - Anna Esteve-Codina
- Centro Nacional de Análisis Genómico (CNAG), 08028 Barcelona, Spain
- Universitat de Barcelona (UB), 08007 Barcelona, Spain
| | - Julien Gagneur
- TUM School of Computation, Information and Technology, Technical University of Munich, 85748 Garching, Germany
- Institute of Human Genetics, School of Medicine, Technical University of Munich, 81675 Munich, Germany
- Computational Health Center, Helmholtz Center Munich, 85764 Neuherberg, Germany
| | - Kornelia Ellwanger
- Universitätsklinikum Tübingen - Institut für Medizinische Genetik und angewandte Genomik, 72076 Tübingen, Germany
- Center for Rare Diseases, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Ronny Derks
- Radboud University Medical Center, Department of Human Genetics, Research Institute for Medical Innovation, 6500 HB Nijmegen, Netherlands
| | - Marjan Weiss
- Radboud University Medical Center, Department of Human Genetics, Research Institute for Medical Innovation, 6500 HB Nijmegen, Netherlands
| | - Amber den Ouden
- Radboud University Medical Center, Department of Human Genetics, Research Institute for Medical Innovation, 6500 HB Nijmegen, Netherlands
| | - Simone van den Heuvel
- Radboud University Medical Center, Department of Human Genetics, Research Institute for Medical Innovation, 6500 HB Nijmegen, Netherlands
| | - Hilde Swinkels
- Radboud University Medical Center, Department of Human Genetics, Research Institute for Medical Innovation, 6500 HB Nijmegen, Netherlands
| | - Nick Zomer
- Radboud University Medical Center, Department of Human Genetics, Research Institute for Medical Innovation, 6500 HB Nijmegen, Netherlands
| | - Marloes Steehouwer
- Radboud University Medical Center, Department of Human Genetics, Research Institute for Medical Innovation, 6500 HB Nijmegen, Netherlands
| | - Luke O'Gorman
- Radboud University Medical Center, Department of Human Genetics, Research Institute for Medical Innovation, 6500 HB Nijmegen, Netherlands
| | - Galuh Astuti
- Radboud University Medical Center, Department of Human Genetics, Research Institute for Medical Innovation, 6500 HB Nijmegen, Netherlands
| | - Kornelia Neveling
- Radboud University Medical Center, Department of Human Genetics, Research Institute for Medical Innovation, 6500 HB Nijmegen, Netherlands
| | - Rebecca Schüle
- Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, 72076 Tübingen, Germany
- Division of Neurodegenerative Diseases, Department of Neurology, Heidelberg University Hospital and Faculty of Medicine, 69120 Heidelberg, Germany
| | - Jishu Xu
- Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, 72076 Tübingen, Germany
| | - Matthis Synofzik
- Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, 72076 Tübingen, Germany
- German Center of Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
| | - Danique Beijer
- Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, 72076 Tübingen, Germany
| | - Holger Hengel
- Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, 72076 Tübingen, Germany
| | - Ludger Schöls
- Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, 72076 Tübingen, Germany
- German Center of Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
| | - Kristl G Claeys
- Department of Neurology, University Hospitals Leuven, 3000 Leuven, Belgium
- Department of Neurosciences, Laboratory for Muscle Diseases and Neuropathies, KU Leuven, and Leuven Brain Institute (LBI), 3000 Leuven, Belgium
| | - Jonathan Baets
- Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
- Laboratory of Neuromuscular Pathology, Institute Born-Bunge, University of Antwerp, 2610 Antwerp, Belgium
- Neuromuscular Reference Center, Department of Neurology, Antwerp University Hospital, 2650 Antwerp, Belgium
| | - Liedewei Van de Vondel
- Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
- Laboratory of Neuromuscular Pathology, Institute Born-Bunge, University of Antwerp, 2610 Antwerp, Belgium
| | - Alessandra Ferlini
- Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Rita Selvatici
- Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Heba Morsy
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London WC1N 3BG, United Kingdom
| | - Marwa Saeed Abd Elmaksoud
- Neurology Unit, Department of Pediatrics, Faculty of Medicine, Alexandria University, Alexandria 5372066, Egypt
| | - Volker Straub
- John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 3BZ, United Kingdom
| | - Juliane Müller
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Hospital, London WC1N 3JH, United Kingdom
| | - Veronica Pini
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Hospital, London WC1N 3JH, United Kingdom
| | - Luke Perry
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Hospital, London WC1N 3JH, United Kingdom
- NIHR Great Ormond Street Hospital Biomedical Research Centre, London WC1N 1EH, United Kingdom
| | - Anna Sarkozy
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Hospital, London WC1N 3JH, United Kingdom
| | - Irina Zaharieva
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Hospital, London WC1N 3JH, United Kingdom
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Hospital, London WC1N 3JH, United Kingdom
- NIHR Great Ormond Street Hospital Biomedical Research Centre, London WC1N 1EH, United Kingdom
| | - Enrico Bugiardini
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London WC1N 3BG, United Kingdom
| | - Kiran Polavarapu
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa and Division of Neurology, Department of Medicine, The Ottawa Hospital, Ottawa, ON K1H 8L1, Canada
| | - Rita Horvath
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0PY, United Kingdom
| | - Evan Reid
- Cambridge Institute for Medical Research and Department of Medical Genetics, University of Cambridge, Cambridge CB2 0XY, United Kingdom
| | - Hanns Lochmüller
- Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, Ontario, ON K1H 8M8, Canada
- Brain and Mind Research Institute, University of Ottawa, Ottawa, Ontario, ON K1H 8M5, Canada
- The Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, ON K1Y 4E9, Canada
| | - Marco Spinazzi
- Department of Neurology, Centre Hospitalier Universitaire d'Angers, 49933 Angers, France
| | - Marco Savarese
- Folkhälsan Research Center, 00250 Helsinki, Uusimaa, Finland
- Faculty of Medicine, University of Helsinki, 00014 University of Helsinki, Uusimaa, Finland
| | - Leslie Matalonga
- Centro Nacional de Análisis Genómico (CNAG), 08028 Barcelona, Spain
- Universitat de Barcelona (UB), 08007 Barcelona, Spain
| | - Steven Laurie
- Centro Nacional de Análisis Genómico (CNAG), 08028 Barcelona, Spain
- Universitat de Barcelona (UB), 08007 Barcelona, Spain
| | - Han G Brunner
- Radboud University Medical Center, Department of Human Genetics, Research Institute for Medical Innovation, 6500 HB Nijmegen, Netherlands
- Department of Clinical Genetics, Maastricht University Medical Center, 6229 HX Maastricht, Netherlands
| | - Holm Graessner
- Universitätsklinikum Tübingen - Institut für Medizinische Genetik und angewandte Genomik, 72076 Tübingen, Germany
- Center for Rare Diseases, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Sergi Beltran
- Centro Nacional de Análisis Genómico (CNAG), 08028 Barcelona, Spain
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona (UB), 08028 Barcelona, Spain
| | - Stephan Ossowski
- Universitätsklinikum Tübingen - Institut für Medizinische Genetik und angewandte Genomik, 72076 Tübingen, Germany
| | - Lisenka E L M Vissers
- Radboud University Medical Center, Department of Human Genetics, Research Institute for Medical Innovation, 6500 HB Nijmegen, Netherlands
| | - Christian Gilissen
- Radboud University Medical Center, Department of Human Genetics, Research Institute for Medical Innovation, 6500 HB Nijmegen, Netherlands
| | - Alexander Hoischen
- Radboud University Medical Center, Department of Human Genetics, Research Institute for Medical Innovation, 6500 HB Nijmegen, Netherlands;
- Radboud University Medical Center, Department of Internal Medicine; Radboud Expertise Center for Immunodeficiency and Autoinflammation and Radboud Center for Infectious Disease (RCI), 6500 HB Nijmegen, Netherlands
| |
Collapse
|
9
|
Buianova AA, Lashkova YS, Kulichenko TV, Kuznetsov IS, Ivanov AA, Parshina OP, Suchalko ON, Vakhlyarskaya SS, Korostin DO. Heterozygous deletion of 10q24.31-q24.33- a new syndrome associated with multiple congenital anomalies: case report and literature review. Neurol Res Pract 2025; 7:22. [PMID: 40189573 PMCID: PMC11974182 DOI: 10.1186/s42466-025-00378-z] [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: 12/09/2024] [Accepted: 02/19/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND Congenital anomalies and neurodevelopmental disorders are complex conditions often requiring comprehensive diagnostic approaches. Next-generation sequencing (NGS), particularly whole-exome sequencing (WES), has greatly improved the detection of pathogenic variants, including copy number variations (CNVs), which account for up to 35% of genetic causes in neurological patients. Combining CNV and single nucleotide variant (SNV) analysis through WES enhances diagnostic accuracy, especially in cases with unclassified congenital anomalies. CASE PRESENTATION AND LITERATURE REVIEW This study reports a 14-year-old male patient with multiple congenital anomalies, including hypospadias, complete cleft palate, and recurrent pneumonia. His clinical presentation includes significant physical and intellectual developmental delays, autism-like symptoms, and spastic diplegia. Whole-exome sequencing (WES) was performed due to these complex symptoms, revealing a novel heterozygous deletion on chromosome 10q24.31-q24.33. Laboratory findings indicated agammaglobulinemia, leading to prophylactic antibiotic therapy and immunoglobulin replacement. Additional imaging studies showed cystic malformation of the middle lobe of the right lung, sliding hiatal hernia with prolapse of the gastric mucosa, and brain anomalies consistent with Joubert syndrome. CONCLUSIONS This case underscores the importance of genetic analysis in understanding the etiology of congenital anomalies and neurodevelopmental disorders, providing critical insights into the molecular mechanisms driving complex phenotypes. The identified chromosomal deletion contributes to the existing literature on genomic imbalances associated with similar phenotypes.
Collapse
Affiliation(s)
- Anastasiia A Buianova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 1, bld. 1, Ostrovityanova St., Moscow, 117513, Russia.
| | - Yulia S Lashkova
- Russian Children's Clinical Hospital, 117 Leninsky Prospect, Moscow, 119571, Russia.
| | - Tatiana V Kulichenko
- Russian Children's Clinical Hospital, 117 Leninsky Prospect, Moscow, 119571, Russia
| | - Ivan S Kuznetsov
- Russian Children's Clinical Hospital, 117 Leninsky Prospect, Moscow, 119571, Russia
| | - Artem A Ivanov
- Russian Children's Clinical Hospital, 117 Leninsky Prospect, Moscow, 119571, Russia
| | - Olga P Parshina
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 1, bld. 1, Ostrovityanova St., Moscow, 117513, Russia
| | - Oleg N Suchalko
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 1, bld. 1, Ostrovityanova St., Moscow, 117513, Russia
| | | | - Dmitriy O Korostin
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 1, bld. 1, Ostrovityanova St., Moscow, 117513, Russia
| |
Collapse
|
10
|
Twumasi Aboagye E, Adadey SM, Alves de Souza Rios L, Esoh KK, Wonkam-Tingang E, Xhakaza L, De Kock C, Schrauwen I, Amenga-Etego L, Lang D, Awandare GA, Leal SM, Mowla S, Wonkam A. Bi-Allelic MARVELD2 Variant Identified with Exome Sequencing in a Consanguineous Multiplex Ghanaian Family Segregating Non-Syndromic Hearing Loss. Int J Mol Sci 2025; 26:3337. [PMID: 40244166 PMCID: PMC11989440 DOI: 10.3390/ijms26073337] [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: 02/28/2025] [Revised: 03/20/2025] [Accepted: 03/28/2025] [Indexed: 04/18/2025] Open
Abstract
Genetic studies and phenotypic expansion of hearing loss (HL) for people living in Africa are greatly needed. We evaluated the clinical phenotypes of three affected siblings presenting non-syndromic (NS) HL and five unaffected members of a consanguineous Ghanaian family. Analysis of exome sequence data was performed for all affected and one unaffected family members. In-depth genetic and cellular characterization studies were performed to investigate biological significance of the implicated variant using bioinformatic tools and cell-based experimentation. Audiological examinations showed severe-to-profound, bilateral, symmetrical, and post-lingual onset. The whole-exome sequencing (WES) identified a homozygous frameshift variant: MARVEL domain containing 2 (MARVELD2):c.1058dup;p.(Val354Serfs*5) in all affected siblings. This frameshift variant leads to an early stop codon insertion and predicted to be targeted by nonsense medicated decay (mutant protein predicted to lack conserved C-terminal domain if translated). Cell immunofluorescence and immunocytochemistry studies exposed the functional impact of the mutant protein's expression, stability, localization, protein-protein binding, barrier function, and actin cytoskeleton architecture. The identified variant segregates with NSHL in the index Ghanaian family. The data support this nonsense variant as pathogenic, likely to impact the homeostasis of ions, solutes, and other molecules, compromising membrane barrier and signaling in the inner ear spaces.
Collapse
Affiliation(s)
- Elvis Twumasi Aboagye
- Department of Pathology, Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa or (E.T.A.); (S.M.A.); (K.K.E.); (E.W.-T.); (L.X.); (C.D.K.)
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Legon, Accra LG 54, Ghana; (L.A.-E.); (G.A.A.)
| | - Samuel Mawuli Adadey
- Department of Pathology, Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa or (E.T.A.); (S.M.A.); (K.K.E.); (E.W.-T.); (L.X.); (C.D.K.)
| | - Leonardo Alves de Souza Rios
- Department of Pathology, Division of Haematology, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa; (L.A.d.S.R.); (S.M.)
| | - Kevin K. Esoh
- Department of Pathology, Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa or (E.T.A.); (S.M.A.); (K.K.E.); (E.W.-T.); (L.X.); (C.D.K.)
- McKusick-Nathans Institute & Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Edmond Wonkam-Tingang
- Department of Pathology, Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa or (E.T.A.); (S.M.A.); (K.K.E.); (E.W.-T.); (L.X.); (C.D.K.)
| | - Lettilia Xhakaza
- Department of Pathology, Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa or (E.T.A.); (S.M.A.); (K.K.E.); (E.W.-T.); (L.X.); (C.D.K.)
| | - Carmen De Kock
- Department of Pathology, Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa or (E.T.A.); (S.M.A.); (K.K.E.); (E.W.-T.); (L.X.); (C.D.K.)
| | - Isabelle Schrauwen
- Department of Translational Neurosciences, University of Arizona College of Medicine Phoenix, Phoenix, AZ 85004, USA;
| | - Lucas Amenga-Etego
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Legon, Accra LG 54, Ghana; (L.A.-E.); (G.A.A.)
| | - Dirk Lang
- Department of Human Biology, Division of Cell Biology, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa;
| | - Gordon A. Awandare
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Legon, Accra LG 54, Ghana; (L.A.-E.); (G.A.A.)
| | - Suzanne M. Leal
- Center for Statistical Genetics, Gertrude H. Sergievsky Center, Department of Neurology, Columbia University Medical Centre, New York, NY 10032, USA;
- Taub Institute, Columbia University Medical Center, New York, NY 10032, USA
| | - Shaheen Mowla
- Department of Pathology, Division of Haematology, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa; (L.A.d.S.R.); (S.M.)
| | - Ambroise Wonkam
- Department of Pathology, Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa or (E.T.A.); (S.M.A.); (K.K.E.); (E.W.-T.); (L.X.); (C.D.K.)
- McKusick-Nathans Institute & Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
11
|
Gjoni K, Zhang S, Yan RE, Zhang B, Miller D, Resnick A, Dahmane N, Pollard KS. Machine learning-predicted chromatin organization landscape across pediatric tumors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.28.645984. [PMID: 40236019 PMCID: PMC11996386 DOI: 10.1101/2025.03.28.645984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Structural variants (SVs) are increasingly recognized as important contributors to oncogenesis through their effects on 3D genome folding. Recent advances in whole-genome sequencing have enabled large-scale profiling of SVs across diverse tumors, yet experimental characterization of their individual impact on genome folding remains infeasible. Here, we leveraged a convolutional neural network, Akita, to predict disruptions in genome folding caused by somatic SVs identified in 61 tumor types from the Children's Brain Tumor Network dataset. Our analysis reveals significant variability in SV-induced disruptions across tumor types, with the most disruptive SVs coming from lymphomas and sarcomas, metastatic tumors, and germline cell tumors. Dimensionality reduction of disruption scores identified five recurrently disrupted regions enriched for high-impact SVs across multiple tumors. Some of these regions are highly disrupted despite not being highly mutated, and harbor tumor-associated genes and transcriptional regulators. To further interpret the functional relevance of high-scoring SVs, we integrated epigenetic data and developed a modified Activity-by-Contact scoring approach to prioritize SVs with disrupted genome contacts at active enhancers. This method highlighted highly disruptive SVs near key oncogenes, as well as novel candidate loci potentially implicated in tumorigenesis. These findings highlight the utility of machine learning for identifying novel SVs, loci, and genetic mechanisms contributing to pediatric cancers. This framework provides a foundation for future studies linking SV-driven regulatory changes to cancer pathogenesis.
Collapse
|
12
|
Daida K, Yoshino H, Malik L, Baker B, Ishiguro M, Genner R, Paquette K, Li Y, Nishioka K, Masuzugawa S, Hirano M, Takahashi K, Kolmogorov M, Billingsley KJ, Funayama M, Blauwendraat C, Hattori N. The Utility of Long-Read Sequencing in Diagnosing Early Onset Parkinson's Disease. Ann Neurol 2025; 97:753-765. [PMID: 39699073 PMCID: PMC11889530 DOI: 10.1002/ana.27155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/27/2024] [Accepted: 11/27/2024] [Indexed: 12/20/2024]
Abstract
OBJECTIVE Variants in PRKN and PINK1 are the leading cause of early-onset autosomal recessive Parkinson's disease, yet many cases remain genetically unresolved. We previously identified a 7 megabases complex structural variant in a pair of monozygotic twins using Oxford Nanopore Technologies (ONT) long-read sequencing. This study aims to determine if ONT long-read sequencing can detect a second variant in other unresolved early-onset Parkinson's disease (EOPD) cases with 1 heterozygous PRKN or PINK1 variant. METHODS ONT long-read sequencing was performed on EOPD patients with 1 reported PRKN/PINK1 pathogenic variant, with onset age under 50. Positive controls included EOPD patients with 2 known PRKN pathogenic variants. Initial testing involved short-read targeted panel sequencing for single nucleotide variants and multiplex ligation-dependent probe amplification for copy number variants. RESULTS A total of 47 patients were studied (PRKN "one-variant," n = 23; PINK1 "one-variant," n = 12; PRKN "two-variants," n = 12). ONT long-read sequencing identified a second pathogenic variant in 26% of PRKN "one-variant" patients (6/23), but none in PINK1 "one-variant" patients (0/12). Detected variants included 1 complex inversion, 2 structural variant overlaps, and 3 duplications. In the PRKN "two-variants" group, both variants were identified in all patients (100%, 12/12). INTERPRETATION ONT long-read sequencing effectively identifies pathogenic structural variants in the PRKN locus missed by conventional methods. It should be considered for unresolved EOPD cases when a second variant is not detected through conventional approaches. ANN NEUROL 2025;97:753-765.
Collapse
Affiliation(s)
- Kensuke Daida
- Integrative Neurogenomics Unit, Laboratory of NeurogeneticsNational Institute on Aging, National Institutes of HealthBethesdaMDUSA
- Center for Alzheimer's and Related Dementias (CARD)National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMDUSA
- Department of Neurology, Faculty of MedicineJuntendo UniversityTokyoJapan
| | - Hiroyo Yoshino
- Research Institute for Diseases of Old Age, Graduate School of MedicineJuntendo UniversityTokyoJapan
| | - Laksh Malik
- Center for Alzheimer's and Related Dementias (CARD)National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMDUSA
| | - Breeana Baker
- Center for Alzheimer's and Related Dementias (CARD)National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMDUSA
| | - Mayu Ishiguro
- Department of Neurology, Faculty of MedicineJuntendo UniversityTokyoJapan
| | - Rylee Genner
- Center for Alzheimer's and Related Dementias (CARD)National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMDUSA
| | - Kimberly Paquette
- Center for Alzheimer's and Related Dementias (CARD)National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMDUSA
| | - Yuanzhe Li
- Department of Neurology, Faculty of MedicineJuntendo UniversityTokyoJapan
- Department of Diagnosis, Prevention and Treatment of Dementia, Graduate School of MedicineJuntendo UniversityTokyoJapan
| | - Kenya Nishioka
- Department of NeurologyJuntendo Tokyo Koto Geriatric Medical CenterTokyoJapan
| | | | - Makito Hirano
- Department of NeurologyKindai University Faculty of MedicineOsakaJapan
| | - Kenta Takahashi
- Division of Neurology and Gerontology, Department of Internal Medicine, School of MedicineIwate Medical UniversityMoriokaJapan
| | - Mikhail Kolmogorov
- Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMDUSA
| | - Kimberley J. Billingsley
- Molecular Genetics Section, Laboratory of NeurogeneticsNational Institute on Aging, National Institutes of HealthBethesdaMDUSA
| | - Manabu Funayama
- Department of Neurology, Faculty of MedicineJuntendo UniversityTokyoJapan
- Research Institute for Diseases of Old Age, Graduate School of MedicineJuntendo UniversityTokyoJapan
| | - Cornelis Blauwendraat
- Integrative Neurogenomics Unit, Laboratory of NeurogeneticsNational Institute on Aging, National Institutes of HealthBethesdaMDUSA
- Center for Alzheimer's and Related Dementias (CARD)National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMDUSA
| | - Nobutaka Hattori
- Department of Neurology, Faculty of MedicineJuntendo UniversityTokyoJapan
- Research Institute for Diseases of Old Age, Graduate School of MedicineJuntendo UniversityTokyoJapan
- Neurodegenerative Disorders Collaborative Laboratory, RIKEN Center for Brain ScienceWakoJapan
| |
Collapse
|
13
|
Rudaks LI, Stevanovski I, Yeow D, Reis ALM, Chintalaphani SR, Cheong PL, Gamaarachchi H, Worgan L, Ahmad K, Hayes M, Hannaford A, Kim S, Fung VSC, Halmagyi GM, Martin A, Manser D, Tchan M, Ng K, Kennerson ML, Deveson IW, Kumar KR. Targeted Long-Read Sequencing as a Single Assay Improves the Diagnosis of Spastic-Ataxia Disorders. Ann Clin Transl Neurol 2025; 12:832-841. [PMID: 40007153 PMCID: PMC12040508 DOI: 10.1002/acn3.70008] [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: 12/26/2024] [Accepted: 01/19/2025] [Indexed: 02/27/2025] Open
Abstract
OBJECTIVE The hereditary spastic-ataxia spectrum disorders are a group of disabling neurological diseases. The traditional genetic testing pathway is complex, multistep and leaves many cases unsolved. We aim to streamline and improve this process using long-read sequencing. METHODS We developed a targeted long-read sequencing strategy with the capacity to characterise the genetic variation of all types and sizes within 469 disease-associated genes, in a single assay. We applied this to a cohort of 34 individuals with unsolved spastic-ataxia. An additional five individuals with a known genetic diagnosis were included as positive controls. RESULTS We identified causative pathogenic variants that would be sufficient for genetic diagnosis in 14/34 (41%) unsolved participants. The success rate was 5/11 (45%) in those who were naïve to genetic testing and 9/23 (39%) in those who were undiagnosed after prior genetic testing, completed on a clinical basis. Short tandem repeat expansions in FGF14 were the most common (7/34, 21%). Two individuals (2/34, 6%) had biallelic pathogenic expansions in RFC1 and one individual had a monoallelic pathogenic expansion in ATXN8OS/ATXN8. Causative pathogenic sequence variants other than short tandem repeat expansions were found in four individuals, including in VCP, STUB1, ANO10 and SPG7. Furthermore, all five positive controls were identified. INTERPRETATION Our results demonstrate the utility of targeted long-read sequencing in the genetic evaluation of patients with spastic-ataxia spectrum disorders, highlighting both the capacity to increase overall diagnostic yield and to streamline the testing pathway by capturing all known genetic causes in a single assay.
Collapse
|
14
|
Qian G, Yang N, Deng F, Zhang M, Pan X, Tan B, Liu L, Zhang X, Yao H, Dong X. SNV/Indel and CNV Analysis in Trio-WES for Intellectual and Developmental Disabilities: Diagnostic Yield & Cost-Effectiveness. Clin Genet 2025; 107:402-412. [PMID: 39829082 DOI: 10.1111/cge.14677] [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: 08/26/2024] [Revised: 11/29/2024] [Accepted: 12/07/2024] [Indexed: 01/22/2025]
Abstract
Intellectual and developmental disabilities (IDD) are clinically and genetically heterogeneous disorders of global concern. While whole exome sequencing (WES) is used to identify single nucleotide variants (SNVs) and small insertions/deletions (Indels) in IDD patients, its detection rate is limited. This study evaluated the value of integrating copy number variation (CNV) analysis into traditional SNV/Indel analysis based on trio-WES. One hundred eighty seven patients with IDD in 140 families from southwest China were incorporated into the study cohort. The overall diagnostic rate was 40.11% (75/187), with 33.16% (62/187) from SNV/Indel analysis and 6.95% (13/187) from CNV analysis. SNV/Indel analysis identified 52 variants in 42 genes, including 30 novel and 22 reported variants; CNV analysis identified 11 CNVs, comprising 1 repeat and 10 deletions, with sizes ranging from 1313 to 55 184 kb. 39.29% (55/140) families benefited from this study for their clinical diagnosis, treatment, and reproduction. Furthermore, our strategy, with an incremental cost-effectiveness ratio (ICER) of $2546.22/diagnosis, had demonstrated significant advantages in terms of cost-effectiveness and detection speed compared to previous methods. In general, by incorporating SNV/Indel and CNV analysis based on trio-WES, a robust, cost-effective, and time-saving approach for diagnosing IDD has been developed.
Collapse
Affiliation(s)
- Guanhua Qian
- Obstetrics and Gynecology Department, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Nanyan Yang
- Obstetrics and Gynecology Department, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fang Deng
- Obstetrics and Gynecology Department, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mingze Zhang
- Obstetrics and Gynecology Department, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xin Pan
- Obstetrics and Gynecology Department, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bo Tan
- Obstetrics and Gynecology Department, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Liu
- Obstetrics and Gynecology Department, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xu Zhang
- Obstetrics and Gynecology Department, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hong Yao
- Obstetrics and Gynecology Department, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaojing Dong
- Obstetrics and Gynecology Department, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
15
|
Costa CIS, Madanelo L, Wang JYT, da Silva Campos G, De Sanctis Girardi AC, Scliar M, Monfardini F, de Cássia Mingroni Pavanello R, Cória VR, Vibranovski MD, Krepischi AC, Lourenço NCV, Zatz M, Yamamoto GL, Zachi EC, Passos-Bueno MR. Understanding rare variant contributions to autism: lessons from dystrophin-deficient model. NPJ Genom Med 2025; 10:18. [PMID: 40050609 PMCID: PMC11885547 DOI: 10.1038/s41525-025-00469-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 01/28/2025] [Indexed: 03/09/2025] Open
Abstract
Duchenne and Becker Muscular Dystrophy are dystrophinopathies with a prevalence of 1:5000-6000 males, caused by pathogenic variants in DMD. These conditions are often accompanied by neurodevelopmental disorders (NDDs) like autism (ASD; ~20%) and intellectual disability (ID; ~30%). However, their low penetrance in dystrophinopathies suggests additional contributing factors. In our study, 83 individuals with dystrophinopathies were clinically evaluated and categorized based on ASD (36 individuals), ID risk (12 individuals), or controls (35 individuals). Exome sequencing analysis revealed an enrichment of risk de novo variants (DNVs) in ASD-DMD individuals (adjusted p value = 0.0356), with the number of DNVs correlating with paternal age (p value = 0.0133). Additionally, DMD-ASD individuals showed a higher average of rare risk variants (RRVs) compared to DMD-Controls (adjusted p value = 0.0285). Gene ontology analysis revealed an enrichment of extracellular matrix-related genes, especially collagens, and Ehlers-Danlos syndrome genes in ASD-DMD and DMD-ID groups. These findings support an oligogenic model for ASD in dystrophinopathies, highlighting the importance of investigating homogenized samples to elucidate ASD's genetic architecture.
Collapse
Affiliation(s)
- Claudia Ismania Samogy Costa
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Luciana Madanelo
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Jaqueline Yu Ting Wang
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Gabriele da Silva Campos
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Ana Cristina De Sanctis Girardi
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Marília Scliar
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Frederico Monfardini
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Rita de Cássia Mingroni Pavanello
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Vivian Romanholi Cória
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Maria Dulcetti Vibranovski
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Ana Cristina Krepischi
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Naila Cristina Vilaça Lourenço
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Mayana Zatz
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Guilherme Lopes Yamamoto
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Elaine Cristina Zachi
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Maria Rita Passos-Bueno
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil.
| |
Collapse
|
16
|
Beyer T, Diwan GD, Leonhard T, Dahlke K, Klose F, Stehle IF, Seda M, Bolz S, Woerz F, Russell RB, Jenkins D, Ueffing M, Boldt K. Ciliopathy-Associated Missense Mutations in IFT140 are Tolerated by the Inherent Resilience of the IFT Machinery. Mol Cell Proteomics 2025; 24:100916. [PMID: 39880085 PMCID: PMC11907452 DOI: 10.1016/j.mcpro.2025.100916] [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: 04/25/2024] [Revised: 11/26/2024] [Accepted: 01/23/2025] [Indexed: 01/31/2025] Open
Abstract
Genotype-phenotype correlations of rare diseases are complicated by low patient number, high phenotype variability, and compound heterozygosity. Mutations may cause instability of single proteins, and affect protein complex formation or overall robustness of a specific process in a given cell. Ciliopathies offer an interesting case for studying genotype-phenotype correlations as they have a spectrum of severity and include diverse phenotypes depending on different mutations in the same protein. For instance, mutations in the intraflagellar transport protein IFT140 cause a vast spectrum of ciliopathies ranging from isolated retinal dystrophy to severe skeletal abnormalities and multi-organ diseases such as Mainzer-Saldino and Jeune syndrome. Here, the quantitative effects of 23 missense mutations in IFT140, which forms part of the crucial IFT-A complex of the ciliary machinery, were analyzed using affinity purification coupled with mass spectrometry (AP-MS). A subset of 10 mutations led to a significant and domain-specific reduction in IFT140-IFT-A complex interaction indicating complex formation issues and potentially hampering its molecular function. Knockout of IFT140 led to loss of cilia, as shown before. However, phenotypically only mild effects concerning cilia assembly were observed for two out of four tested IFT140 missense mutations. Therefore, our results demonstrate the utility of AP-MS in discerning pathogenic MMs from polymorphisms, and we postulate that reduced function is tolerated by the evolutionarily highly conserved IFT-A system.
Collapse
Affiliation(s)
- Tina Beyer
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany.
| | - Gaurav D Diwan
- BioQuant, University of Heidelberg, Heidelberg, Germany; Biochemistry Center (BZH), Heidelberg University, Heidelberg, Germany
| | - Tobias Leonhard
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Katrin Dahlke
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Franziska Klose
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Isabel F Stehle
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Marian Seda
- UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Sylvia Bolz
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Franziska Woerz
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Robert B Russell
- BioQuant, University of Heidelberg, Heidelberg, Germany; Biochemistry Center (BZH), Heidelberg University, Heidelberg, Germany
| | - Dagan Jenkins
- UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom.
| | - Marius Ueffing
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Karsten Boldt
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
17
|
Liu X, Gu L, Hao C, Xu W, Leng F, Zhang P, Li W. Systematic assessment of structural variant annotation tools for genomic interpretation. Life Sci Alliance 2025; 8:e202402949. [PMID: 39658089 PMCID: PMC11632063 DOI: 10.26508/lsa.202402949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 11/30/2024] [Accepted: 12/02/2024] [Indexed: 12/12/2024] Open
Abstract
Structural variants (SVs) over 50 base pairs play a significant role in phenotypic diversity and are associated with various diseases, but their analysis is complex and resource-intensive. Numerous computational tools have been developed for SV prioritization, yet their effectiveness in biomedicine remains unclear. Here we benchmarked eight widely used SV prioritization tools, categorized into knowledge-driven (AnnotSV, ClassifyCNV) and data-driven (CADD-SV, dbCNV, StrVCTVRE, SVScore, TADA, XCNV) groups in accordance with the ACMG guidelines. We assessed their accuracy, robustness, and usability across diverse genomic contexts, biological mechanisms and computational efficiency using seven carefully curated independent datasets. Our results revealed that both groups of methods exhibit comparable effectiveness in predicting SV pathogenicity, although performance varies among tools, emphasizing the importance of selecting the appropriate tool based on specific research purposes. Furthermore, we pinpointed the potential improvement of expanding these tools for future applications. Our benchmarking framework provides a crucial evaluation method for SV analysis tools, offering practical guidance for biomedical research and facilitating the advancement of better genomic research tools.
Collapse
Affiliation(s)
- Xuanshi Liu
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute; MOE Key Laboratory of Major Diseases in Children; Genetics and Birth Defects Control Center, National Center for Children's Health; Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Lei Gu
- Epigenetics Laboratory, Max-Planck Institute for Heart and Lung Research, Cardiopulmonary Institute, Bad Nauheim, Germany
| | - Chanjuan Hao
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute; MOE Key Laboratory of Major Diseases in Children; Genetics and Birth Defects Control Center, National Center for Children's Health; Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Wenjian Xu
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute; MOE Key Laboratory of Major Diseases in Children; Genetics and Birth Defects Control Center, National Center for Children's Health; Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Fei Leng
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute; MOE Key Laboratory of Major Diseases in Children; Genetics and Birth Defects Control Center, National Center for Children's Health; Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Peng Zhang
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute; MOE Key Laboratory of Major Diseases in Children; Genetics and Birth Defects Control Center, National Center for Children's Health; Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Wei Li
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute; MOE Key Laboratory of Major Diseases in Children; Genetics and Birth Defects Control Center, National Center for Children's Health; Beijing Children's Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
18
|
Atallah I, Cisarova K, Guenot C, Dubruc E, Superti-Furga A, Campos-Xavier B, Unger S. Recurrent Increased Nuchal Translucency Led to the Identification of Novel NUP107 Variants. Am J Med Genet A 2025; 197:e63920. [PMID: 39473271 DOI: 10.1002/ajmg.a.63920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 10/12/2024] [Indexed: 02/13/2025]
Abstract
Five percent of fetuses presents increased fetal nuchal translucency. It is a well-known marker for aneuploidy (T21, Turner syndrome) and a variety of monogenic syndromes such as Noonan syndrome and certain skeletal dysplasias, as well as associated with structural malformations such as congenital heart disease. Current diagnostic algorithms for increased nuchal translucency include a rapid test for aneuploidy (fluorescence in situ hybridization, FISH, or quantitative PCR), a cytogenetic analysis (karyotype or chromosomal microarray, CMA) followed by or concurrent with targeted gene panel analysis for RASopathies/Noonan syndrome. Some centers now propose whole exome sequencing as an adjunct, but its usefulness in isolated increased nuchal translucency remains debated. We describe the recurrence of apparently isolated increased nuchal translucency in 2 euploid fetuses. Whole genome sequencing identified two compound heterozygous variants in the NUP107 gene in both fetuses. Biallelic variants in NUP107 are responsible for severe steroid-resistant nephrotic syndrome, either isolated or syndromic (Galloway-Mowat syndrome); in addition to the renal phenotype, the latter also includes intellectual deficiency and dysmorphic features. Pregnancy termination made it impossible to assess whether the NUP107 variants found would have resulted in isolated or syndromic steroid-resistant nephrotic syndrome. However, identifying the responsible gene improved the accuracy of the genetic counseling. This family is an example of the added benefit of introducing WES/WGS in standardized protocols for prenatal diagnosis of euploid fetuses in "isolated" increased nuchal translucency.
Collapse
Affiliation(s)
- Isis Atallah
- Division of Genetic Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Katarina Cisarova
- Division of Genetic Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Cécile Guenot
- Materno-Fetal and Obstetrics Research Unit, Department of Obstetrics and Gynecology, University Hospital, Lausanne, Switzerland
| | - Estelle Dubruc
- Service of Clinical Pathology, Institute of Pathology, Lausanne University Hospital, Lausanne, Switzerland
| | - Andrea Superti-Furga
- Division of Genetic Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Belinda Campos-Xavier
- Division of Genetic Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Sheila Unger
- Division of Genetic Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
19
|
Furukawa S, Kushima I, Kato H, Kimura H, Nawa Y, Aleksic B, Banno M, Yamamoto M, Uematsu M, Nagasaki Y, Ogi T, Ozaki N, Ikeda M. Whole-genome sequencing analysis of Japanese autism spectrum disorder trios. Psychiatry Clin Neurosci 2025; 79:87-97. [PMID: 39610113 PMCID: PMC11874045 DOI: 10.1111/pcn.13767] [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: 09/05/2024] [Revised: 10/23/2024] [Accepted: 11/04/2024] [Indexed: 11/30/2024]
Abstract
AIM Autism spectrum disorder (ASD) is a genetically and phenotypically heterogeneous neurodevelopmental disorder with a strong genetic basis. Conducting the first comprehensive whole-genome sequencing (WGS) analysis of Japanese ASD trios, this study aimed to elucidate the clinical significance of pathogenic variants and enhance the understanding of ASD pathogenesis. METHODS WGS was performed on 57 Japanese patients with ASD and their parents, investigating variants ranging from single-nucleotide variants to structural variants (SVs), short tandem repeats (STRs), mitochondrial variants, and polygenic risk score (PRS). RESULTS Potentially pathogenic variants that could explain observed phenotypes were identified in 18 patients (31.6%) overall and in 10 of 23 patients (43.5%) with comorbid intellectual developmental disorder (IDD). De novo variants in PTEN, CHD7, and HNRNPH2 were identified in patients referred for genetic counseling who exhibited previously reported phenotypes, including one patient with ASD who had profound IDD and macrocephaly with PTEN L320S. Analysis of the AlphaFold3 protein structure indicated potential inhibition of intramolecular interactions within PTEN. SV analysis identified deletions in ARHGAP11B and TMLHE. A pathogenic de novo mitochondrial variant was identified in a patient with ASD who had a history of encephalitis and cognitive decline. GO enrichment analysis of genes with nonsense variants and missense variants (Missense badness, PolyPhen-2, and Constraint >1) showed associations with regulation of growth and ATP-dependent chromatin remodeler activity. No reportable results were obtained in the analysis of STR and PRS. CONCLUSION Characterizing the comprehensive genetic architecture and phenotypes of ASD is a fundamental step towards unraveling its complex biology.
Collapse
Affiliation(s)
- Sawako Furukawa
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
| | - Itaru Kushima
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
- Medical Genomics CenterNagoya University HospitalNagoyaJapan
| | - Hidekazu Kato
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
- Department of Psychiatry for Parents and ChildrenNagoya University HospitalNagoyaJapan
| | - Hiroki Kimura
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
| | - Yoshihiro Nawa
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
- Department of Psychiatry for Parents and ChildrenNagoya University HospitalNagoyaJapan
| | - Branko Aleksic
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
| | | | - Maeri Yamamoto
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
| | - Mariko Uematsu
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
| | - Yukako Nagasaki
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
| | - Tomoo Ogi
- Department of Genetics, Research Institute of Environmental Medicine (RIeM)Nagoya UniversityNagoyaJapan
| | - Norio Ozaki
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
- Pathophysiology of Mental DisordersNagoya University Graduate School of Medicine
| | - Masashi Ikeda
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaJapan
| |
Collapse
|
20
|
Margalit S, Tulpová Z, Detinis Zur T, Michaeli Y, Deek J, Nifker G, Haldar R, Gnatek Y, Omer D, Dekel B, Baris Feldman H, Grunwald A, Ebenstein Y. Long-read structural and epigenetic profiling of a kidney tumor-matched sample with nanopore sequencing and optical genome mapping. NAR Genom Bioinform 2025; 7:lqae190. [PMID: 39781516 PMCID: PMC11704781 DOI: 10.1093/nargab/lqae190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 12/12/2024] [Accepted: 12/31/2024] [Indexed: 01/12/2025] Open
Abstract
Carcinogenesis often involves significant alterations in the cancer genome, marked by large structural variants (SVs) and copy number variations (CNVs) that are difficult to capture with short-read sequencing. Traditionally, cytogenetic techniques are applied to detect such aberrations, but they are limited in resolution and do not cover features smaller than several hundred kilobases. Optical genome mapping (OGM) and nanopore sequencing [Oxford Nanopore Technologies (ONT)] bridge this resolution gap and offer enhanced performance for cytogenetic applications. Additionally, both methods can capture epigenetic information as they profile native, individual DNA molecules. We compared the effectiveness of the two methods in characterizing the structural, copy number and epigenetic landscape of a clear cell renal cell carcinoma tumor. Both methods provided comparable results for basic karyotyping and CNVs, but differed in their ability to detect SVs of different sizes and types. ONT outperformed OGM in detecting small SVs, while OGM excelled in detecting larger SVs, including translocations. Differences were also observed among various ONT SV callers. Additionally, both methods provided insights into the tumor's methylome and hydroxymethylome. While ONT was superior in methylation calling, hydroxymethylation reports can be further optimized. Our findings underscore the importance of carefully selecting the most appropriate platform based on specific research questions.
Collapse
Affiliation(s)
- Sapir Margalit
- School of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, 6997801 Tel Aviv, Israel
- Department of Biomedical Engineering, Tel Aviv University, 6997801 Tel Aviv, Israel
| | - Zuzana Tulpová
- School of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, 6997801 Tel Aviv, Israel
- Department of Biomedical Engineering, Tel Aviv University, 6997801 Tel Aviv, Israel
- Institute of Experimental Botany of the Czech Academy of Sciences, Olomouc, Czech Republic
| | - Tahir Detinis Zur
- School of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, 6997801 Tel Aviv, Israel
- Department of Biomedical Engineering, Tel Aviv University, 6997801 Tel Aviv, Israel
| | - Yael Michaeli
- School of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, 6997801 Tel Aviv, Israel
- Department of Biomedical Engineering, Tel Aviv University, 6997801 Tel Aviv, Israel
| | - Jasline Deek
- School of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, 6997801 Tel Aviv, Israel
- Department of Biomedical Engineering, Tel Aviv University, 6997801 Tel Aviv, Israel
| | - Gil Nifker
- School of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, 6997801 Tel Aviv, Israel
- Department of Biomedical Engineering, Tel Aviv University, 6997801 Tel Aviv, Israel
| | - Rita Haldar
- School of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, 6997801 Tel Aviv, Israel
- Department of Biomedical Engineering, Tel Aviv University, 6997801 Tel Aviv, Israel
| | - Yehudit Gnatek
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, 52621 Ramat Gan, Israel
| | - Dorit Omer
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, 52621 Ramat Gan, Israel
| | - Benjamin Dekel
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, 52621 Ramat Gan, Israel
- Pediatric Nephrology Unit, The Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, 52621 Ramat Gan, Israel
- School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, 6997801 Tel Aviv, Israel
| | - Hagit Baris Feldman
- School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, 6997801 Tel Aviv, Israel
- The Genetics Institute and Genomics Center, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Assaf Grunwald
- School of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, 6997801 Tel Aviv, Israel
- Department of Biomedical Engineering, Tel Aviv University, 6997801 Tel Aviv, Israel
| | - Yuval Ebenstein
- School of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, 6997801 Tel Aviv, Israel
- Department of Biomedical Engineering, Tel Aviv University, 6997801 Tel Aviv, Israel
| |
Collapse
|
21
|
Choi SH, Jurgens SJ, Xiao L, Hill MC, Haggerty CM, Sveinbjörnsson G, Morrill VN, Marston NA, Weng LC, Pirruccello JP, Arnar DO, Gudbjartsson DF, Mantineo H, von Falkenhausen AS, Natale A, Tveit A, Geelhoed B, Roselli C, Van Wagoner DR, Darbar D, Haase D, Soliman EZ, Davogustto GE, Jun G, Calkins H, Anderson JL, Brody JA, Halford JL, Barnard J, Hokanson JE, Smith JD, Bis JC, Young K, Johnson LSB, Risch L, Gula LJ, Kwee LC, Chaffin MD, Kühne M, Preuss M, Gupta N, Nafissi NA, Smith NL, Nilsson PM, van der Harst P, Wells QS, Judy RL, Schnabel RB, Johnson R, Smit RAJ, Gabriel S, Knight S, Furukawa T, Blackwell TW, Nauffal V, Wang X, Min YI, Yoneda ZT, Laksman ZWM, Bezzina CR, Alonso A, Psaty BM, Albert CM, Arking DE, Roden DM, Chasman DI, Rader DJ, Conen D, McManus DD, Fatkin D, Benjamin EJ, Boerwinkle E, Marcus GM, Christophersen IE, Smith JG, Roberts JD, Raffield LM, Shoemaker MB, Cho MH, Cutler MJ, Rienstra M, Chung MK, S Olesen M, Sinner MF, Sotoodehnia N, Kirchhof P, Loos RJF, Nazarian S, Mohanty S, Damrauer SM, Kaab S, Heckbert SR, Redline S, Shah SH, Tanaka T, Ebana Y, Holm H, Stefansson K, Ruff CT, Sabatine MS, et alChoi SH, Jurgens SJ, Xiao L, Hill MC, Haggerty CM, Sveinbjörnsson G, Morrill VN, Marston NA, Weng LC, Pirruccello JP, Arnar DO, Gudbjartsson DF, Mantineo H, von Falkenhausen AS, Natale A, Tveit A, Geelhoed B, Roselli C, Van Wagoner DR, Darbar D, Haase D, Soliman EZ, Davogustto GE, Jun G, Calkins H, Anderson JL, Brody JA, Halford JL, Barnard J, Hokanson JE, Smith JD, Bis JC, Young K, Johnson LSB, Risch L, Gula LJ, Kwee LC, Chaffin MD, Kühne M, Preuss M, Gupta N, Nafissi NA, Smith NL, Nilsson PM, van der Harst P, Wells QS, Judy RL, Schnabel RB, Johnson R, Smit RAJ, Gabriel S, Knight S, Furukawa T, Blackwell TW, Nauffal V, Wang X, Min YI, Yoneda ZT, Laksman ZWM, Bezzina CR, Alonso A, Psaty BM, Albert CM, Arking DE, Roden DM, Chasman DI, Rader DJ, Conen D, McManus DD, Fatkin D, Benjamin EJ, Boerwinkle E, Marcus GM, Christophersen IE, Smith JG, Roberts JD, Raffield LM, Shoemaker MB, Cho MH, Cutler MJ, Rienstra M, Chung MK, S Olesen M, Sinner MF, Sotoodehnia N, Kirchhof P, Loos RJF, Nazarian S, Mohanty S, Damrauer SM, Kaab S, Heckbert SR, Redline S, Shah SH, Tanaka T, Ebana Y, Holm H, Stefansson K, Ruff CT, Sabatine MS, Lunetta KL, Lubitz SA, Ellinor PT. Sequencing in over 50,000 cases identifies coding and structural variation underlying atrial fibrillation risk. Nat Genet 2025; 57:548-562. [PMID: 40050430 DOI: 10.1038/s41588-025-02074-9] [Show More Authors] [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: 03/03/2022] [Accepted: 01/02/2025] [Indexed: 03/15/2025]
Abstract
Atrial fibrillation (AF) is a prevalent and morbid abnormality of the heart rhythm with a strong genetic component. Here, we meta-analyzed genome and exome sequencing data from 36 studies that included 52,416 AF cases and 277,762 controls. In burden tests of rare coding variation, we identified novel associations between AF and the genes MYBPC3, LMNA, PKP2, FAM189A2 and KDM5B. We further identified associations between AF and rare structural variants owing to deletions in CTNNA3 and duplications of GATA4. We broadly replicated our findings in independent samples from MyCode, deCODE and UK Biobank. Finally, we found that CRISPR knockout of KDM5B in stem-cell-derived atrial cardiomyocytes led to a shortening of the action potential duration and widespread transcriptomic dysregulation of genes relevant to atrial homeostasis and conduction. Our results highlight the contribution of rare coding and structural variants to AF, including genetic links between AF and cardiomyopathies, and expand our understanding of the rare variant architecture for this common arrhythmia.
Collapse
Grants
- K24HL105780 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- U01 AG068221 NIA NIH HHS
- K08HL153950 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- 75N92019D00031 NHLBI NIH HHS
- 18SFRN34110082 American Heart Association (American Heart Association, Inc.)
- R01HL141989 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- 648131 EC | EU Framework Programme for Research and Innovation H2020 | H2020 Priority Excellent Science | H2020 European Research Council (H2020 Excellent Science - European Research Council)
- 847770 EC | EU Framework Programme for Research and Innovation H2020 | H2020 Priority Excellent Science | H2020 European Research Council (H2020 Excellent Science - European Research Council)
- 18SFRN34230127 American Heart Association (American Heart Association, Inc.)
- R01HL157635 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- 1U01AG068221-01A1 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R01HL147148 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R01HL111314 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R01HL155197 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- 9SFRN34830063 American Heart Association (American Heart Association, Inc.)
- 1U01AG058589-01A1 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- 2019-0526 Hjärt-Lungfonden (Swedish Heart-Lung Foundation)
- R01HL092577 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R35HL135818 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- 18SFRN34250007 American Heart Association (American Heart Association, Inc.)
- IRC15-0067 Stiftelsen för Strategisk Forskning (Swedish Foundation for Strategic Research)
- R01HL137927 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- 32473B_176178 Schweizerischer Nationalfonds zur Förderung der Wissenschaftlichen Forschung (Swiss National Science Foundation)
- AA/18/2/34218 British Heart Foundation (BHF)
- 1R01HL164824-01 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- HL113338 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R01HL111024 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R01HL141901 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- PG/20/22/35093 British Heart Foundation (BHF)
- HL116690 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- 961045 American Heart Association (American Heart Association, Inc.)
- 18SFRN34110067 American Heart Association (American Heart Association, Inc.)
- P01HL158505 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R01HL089856 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- IK2-CX001780 U.S. Department of Veterans Affairs (Department of Veterans Affairs)
- 349-2006-237 Vetenskapsrådet (Swedish Research Council)
- K08HL159346 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- 17K07251 MEXT | Japan Society for the Promotion of Science (JSPS)
- 2009-1039 Vetenskapsrådet (Swedish Research Council)
- 32003B_197524 Schweizerischer Nationalfonds zur Förderung der Wissenschaftlichen Forschung (Swiss National Science Foundation)
- 03-007-2022-0035 Hartstichting (Dutch Heart Foundation)
- 33CS30_177520 Schweizerischer Nationalfonds zur Förderung der Wissenschaftlichen Forschung (Swiss National Science Foundation)
- JP18H02804 MEXT | Japan Society for the Promotion of Science (JSPS)
- 2019-0354 Svenska Sällskapet för Medicinsk Forskning (Swedish Society for Medical Research)
- 19SFRN34830063 American Heart Association (American Heart Association, Inc.)
- 2021-02273 Vetenskapsrådet (Swedish Research Council)
- 18SFRN34110067. American Heart Association (American Heart Association, Inc.)
- PG/17/30/32961 British Heart Foundation (BHF)
- 33CS30_148474 Schweizerischer Nationalfonds zur Förderung der Wissenschaftlichen Forschung (Swiss National Science Foundation)
- R01HL149352 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- 1R01HL139731 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- 2R01HL127564-05A1 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- 20CDA35260081 American Heart Association (American Heart Association, Inc.)
- HL-093613 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- 1R01HL128914 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- HL43680 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- Ki 731/4-1 Deutsche Forschungsgemeinschaft (German Research Foundation)
Collapse
Affiliation(s)
- Seung Hoan Choi
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Sean J Jurgens
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Experimental Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences (Heart Failure & Arrhythmias), Amsterdam UMC, Amsterdam, The Netherlands
| | - Ling Xiao
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachsetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Matthew C Hill
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachsetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | - Valerie N Morrill
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Nicholas A Marston
- TIMI Study Group, Boston, MA, USA
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lu-Chen Weng
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachsetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - James P Pirruccello
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachsetts General Hospital, Harvard Medical School, Boston, MA, USA
- Division of Cardiology, University of California, San Francisco, San Francisco, CA, USA
| | - David O Arnar
- deCODE genetics/Amgen, Reykjavik, Iceland
- Cardiovascular Center, Landspitali - The National University Hospital of Iceland, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Daniel Fannar Gudbjartsson
- deCODE genetics/Amgen, Reykjavik, Iceland
- Faculty of Electical and Computer Engineering and School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | - Helene Mantineo
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Aenne S von Falkenhausen
- Department of Medicine I, University Hospital Munich, Ludwig Maximilian University, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Andrea Natale
- Texas Cardiac Arrhythmia Institute, St David's Medical Center, Austin, TX, USA
| | - Arnljot Tveit
- Department of Medical Research, Bærum Hospital, Vestre Viken Hospital Trust, Gjettum, Norway
| | - Bastiaan Geelhoed
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Carolina Roselli
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - David R Van Wagoner
- Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Dawood Darbar
- Division of Cardiology, University of Illinois at Chicago, Chicago, IL, USA
| | - Doreen Haase
- Atrial Fibrillation NETwork (AFNET), Münster, Germany
| | - Elsayed Z Soliman
- Epidemiological Cardiology Research Center (EPICARE), Department of Internal Medicine, Cardiology Section, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Giovanni E Davogustto
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Goo Jun
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Hugh Calkins
- Cardiac Arrhythmia Service, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jeffrey L Anderson
- Intermountain Heart Institute, Intermountain Medical Center, Murray, UT, USA
- Division of Cardiology, University of Utah, Salt Lake City, UT, USA
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Jennifer L Halford
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachsetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - John Barnard
- Departments of Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - John E Hokanson
- Department of Epidemiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Jonathan D Smith
- Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Kendra Young
- Department of Epidemiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Linda S B Johnson
- Department of Clinical Physiology, Department of Clinical Sciences, Skåne University Hospital and Lund University, Lund, Sweden
| | - Lorenz Risch
- Institute of Laboratory Medicine, Faculty of Medical Sciences, Private University of the Principality of Liechtenstein, Triesen, Liechtenstein
- Center of Laboratory Medicine, University Institute of Clinical Chemistry, University of Bern, Inselspital, Bern, Switzerland
| | - Lorne J Gula
- Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine, Western University, London, Ontario, Canada
| | - Lydia Coulter Kwee
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Mark D Chaffin
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michael Kühne
- Cardiology/Electrophysiology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Michael Preuss
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Namrata Gupta
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Navid A Nafissi
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Nicholas L Smith
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Peter M Nilsson
- Department of Clinical Sciences, Clinical Research Center, Lund University, Malmö, Sweden
- Department of Internal Medicine, Skåne University Hospital, Malmö, Sweden
| | - Pim van der Harst
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Quinn S Wells
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Departments of Medicine, Pharmacology, and Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Renae L Judy
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Surgery, Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Renate B Schnabel
- Atrial Fibrillation NETwork (AFNET), Münster, Germany
- Department of Cardiology, University Heart and Vascular Center Hamburg, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg/Kiel/Lübeck, Germany
| | - Renee Johnson
- Molecular Cardiology Division, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Roelof A J Smit
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stacey Gabriel
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Stacey Knight
- Intermountain Heart Institute, Intermountain Medical Center, Murray, UT, USA
- Department of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Tetsushi Furukawa
- Department of Bio-Informational Pharmacology, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Thomas W Blackwell
- Center for Statistical Genetics, Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Victor Nauffal
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachsetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Xin Wang
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachsetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yuan-I Min
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Zachary T Yoneda
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Zachary W M Laksman
- Department of Medicine and the School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Connie R Bezzina
- Department of Experimental Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences (Heart Failure & Arrhythmias), Amsterdam UMC, Amsterdam, The Netherlands
| | - Alvaro Alonso
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Christine M Albert
- Department of Cardiology, Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Dan E Arking
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dan M Roden
- Departments of Medicine, Pharmacology, and Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Daniel I Chasman
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Divisions of Preventive Medicine and Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniel J Rader
- Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - David Conen
- Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - David D McManus
- University of Massachusetts Chan Medical School Worcester, Worcester, MA, USA
| | - Diane Fatkin
- Molecular Cardiology Division, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
- Cardiology Department, St. Vincent's Hospital, Sydney, New South Wales, Australia
| | - Emelia J Benjamin
- NHLBI and Boston University's Framingham Heart Study, Framingham, MA, USA
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA, USA
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Gregory M Marcus
- Division of Cardiology, University of California, San Francisco, San Francisco, CA, USA
| | - Ingrid E Christophersen
- Department of Medical Research, Bærum Hospital, Vestre Viken Hospital Trust, Gjettum, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - J Gustav Smith
- Department of Cardiology, Lund University Diabetes Center and Wallenberg Center for Molecular Medicine, Clinical Sciences, Lund University and Skåne University Hospital, Lund, Sweden
- The Wallenberg Laboratory/Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University and the Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jason D Roberts
- Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine, Western University, London, Ontario, Canada
- Population Health Research Institute, McMaster University, and Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Laura M Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - M Benjamin Shoemaker
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael H Cho
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Michael J Cutler
- Intermountain Heart Institute, Intermountain Medical Center, Murray, UT, USA
| | - Michiel Rienstra
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Mina K Chung
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Morten S Olesen
- Laboratory for Molecular Cardiology, Department of Cardiology, Centre for Cardiac, Vascular, Pulmonary and Infectious Diseases, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Moritz F Sinner
- Department of Medicine I, University Hospital Munich, Ludwig Maximilian University, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Paulus Kirchhof
- Atrial Fibrillation NETwork (AFNET), Münster, Germany
- Department of Cardiology, University Heart and Vascular Center Hamburg, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg/Kiel/Lübeck, Germany
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Saman Nazarian
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Sanghamitra Mohanty
- Texas Cardiac Arrhythmia Institute, St David's Medical Center, Austin, TX, USA
- Dell Medical School, Austin, TX, USA
| | - Scott M Damrauer
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Surgery, Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Stefan Kaab
- Department of Medicine I, University Hospital Munich, Ludwig Maximilian University, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Susan R Heckbert
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
- Cardiovascular Health Research Unit, Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Svati H Shah
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Toshihiro Tanaka
- Department of Human Genetics and Disease Diversity, Tokyo Medical and Dental University (TMDU) Graduate School of Medical and Dental Sciences, Tokyo, Japan
- BioResource Research Center, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yusuke Ebana
- Life Science and Bioethics Research Center, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hilma Holm
- deCODE genetics/Amgen, Reykjavik, Iceland
| | - Kari Stefansson
- deCODE genetics/Amgen, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Christian T Ruff
- TIMI Study Group, Boston, MA, USA
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Marc S Sabatine
- TIMI Study Group, Boston, MA, USA
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kathryn L Lunetta
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- NHLBI and Boston University's Framingham Heart Study, Framingham, MA, USA
| | - Steven A Lubitz
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachsetts General Hospital, Harvard Medical School, Boston, MA, USA
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA
| | - Patrick T Ellinor
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Cardiovascular Research Center, Massachsetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
22
|
Motelow JE, Malakar A, Murthy SBK, Verbitsky M, Kahn A, Estrella E, Kunkel L, Wiesenhahn M, Becket J, Harris N, Lee R, Adam R, Kiryluk K, Gharavi AG, Brownstein CA. Interstitial Cystitis: a phenotype and rare variant exome sequencing study: Interstitial Cystitis: a phenotype and exome sequencing study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.16.25322147. [PMID: 40034785 PMCID: PMC11875234 DOI: 10.1101/2025.02.16.25322147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Interstitial cystitis/bladder pain syndrome (IC/BPS) is a poorly understood and underdiagnosed syndrome of chronic bladder/pelvic pain with urinary frequency and urgency. Though IC/BPS can be hereditary, little is known of its genetic etiology. Using the eMERGE data, we confirmed known phenotypic associations such as gastroesophageal reflux disease and irritable bowel syndrome and detected new associations, including osteoarthrosis/osteoarthritis and Barrett's esophagus. An exome wide ultra-rare variants analysis in 348 IC/BPS and 11,981 controls extended the previously reported association with ATP2C1 and ATP2A2, implicated in Mendelian desquamating skin disorders, but did not provide evidence for other previously proposed pathogenic pathways such as bladder development, nociception or inflammation. Pathway analysis detected new associations with "anaphase-promoting complex-dependent catabolic process", the "regulation of MAPK cascade" and "integrin binding". These findings suggest perturbations in biological networks for epithelial integrity and cell cycle progression in IC/BPS pathogenesis, and provide a roadmap for its future investigation.
Collapse
Affiliation(s)
- Joshua E Motelow
- Division of Critical Care and Hospital Medicine, Department of Pediatrics, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY
| | - Ayan Malakar
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY
- Center for Precision Medicine and Genomics, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Sarath Babu Krishna Murthy
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY
- Center for Precision Medicine and Genomics, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Miguel Verbitsky
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY
- Center for Precision Medicine and Genomics, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Atlas Kahn
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY
| | - Elicia Estrella
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA
| | - Louis Kunkel
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA
- The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston MA
| | - Madelyn Wiesenhahn
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA
- The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston MA
| | - Jaimee Becket
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY
- Center for Precision Medicine and Genomics, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Natasha Harris
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY
- Center for Precision Medicine and Genomics, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Richard Lee
- Department of Urology, Boston Children's Hospital, Harvard Medical School, Boston MA
| | - Rosalyn Adam
- Department of Urology, Boston Children's Hospital, Harvard Medical School, Boston MA
- Department of Surgery, Harvard Medical School, Boston, MA
| | - Krzysztof Kiryluk
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY
| | - Ali G Gharavi
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY
- Center for Precision Medicine and Genomics, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Catherine A Brownstein
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA
- The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston MA
| |
Collapse
|
23
|
Iyer KR, Clarke SL, Guarischi‐Sousa R, Gjoni K, Heath AS, Young EP, Stitziel NO, Laurie C, Broome JG, Khan AT, Lewis JP, Xu H, Montasser ME, Ashley KE, Hasbani NR, Boerwinkle E, Morrison AC, Chami N, Do R, Rocheleau G, Lloyd‐Jones DM, Lemaitre RN, Bis JC, Floyd JS, Kinney GL, Bowden DW, Palmer ND, Benjamin EJ, Nayor M, Yanek LR, Kral BG, Becker LC, Kardia SLR, Smith JA, Bielak LF, Norwood AF, Min Y, Carson AP, Post WS, Rich SS, Herrington D, Guo X, Taylor KD, Manson JE, Franceschini N, Pollard KS, Mitchell BD, Loos RJF, Fornage M, Hou L, Psaty BM, Young KA, Regan EA, Freedman BI, Vasan RS, Levy D, Mathias RA, Peyser PA, Raffield LM, Kooperberg C, Reiner AP, Rotter JI, Jun G, de Vries PS, Assimes TL. Unveiling the Genetic Landscape of Coronary Artery Disease Through Common and Rare Structural Variants. J Am Heart Assoc 2025; 14:e036499. [PMID: 39950338 PMCID: PMC12074758 DOI: 10.1161/jaha.124.036499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 10/21/2024] [Indexed: 02/17/2025]
Abstract
BACKGROUND Genome-wide association studies have identified several hundred susceptibility single nucleotide variants for coronary artery disease (CAD). Despite single nucleotide variant-based genome-wide association studies improving our understanding of the genetics of CAD, the contribution of structural variants (SVs) to the risk of CAD remains largely unclear. METHOD AND RESULTS We leveraged SVs detected from high-coverage whole genome sequencing data in a diverse group of participants from the National Heart Lung and Blood Institute's Trans-Omics for Precision Medicine program. Single variant tests were performed on 58 706 SVs in a study sample of 11 556 CAD cases and 42 907 controls. Additionally, aggregate tests using sliding windows were performed to examine rare SVs. One genome-wide significant association was identified for a common biallelic intergenic duplication on chromosome 6q21 (P=1.54E-09, odds ratio=1.34). The sliding window-based aggregate tests found 1 region on chromosome 17q25.3, overlapping USP36, to be significantly associated with coronary artery disease (P=1.03E-10). USP36 is highly expressed in arterial and adipose tissues while broadly affecting several cardiometabolic traits. CONCLUSIONS Our results suggest that SVs, both common and rare, may influence the risk of coronary artery disease.
Collapse
Affiliation(s)
- Kruthika R. Iyer
- Data Science and Biotechnology, Gladstone InstitutesSan FranciscoCAUSA
- Department of Medicine, Division of Cardiovascular MedicineStanford University School of MedicineStanfordCAUSA
| | - Shoa L. Clarke
- Department of Medicine, Division of Cardiovascular MedicineStanford University School of MedicineStanfordCAUSA
- Department of Medicine, Stanford Prevention Research CenterStanford University School of MedicineStanfordCAUSA
| | - Rodrigo Guarischi‐Sousa
- Department of Medicine, Division of Cardiovascular MedicineStanford University School of MedicineStanfordCAUSA
| | - Ketrin Gjoni
- Data Science and Biotechnology, Gladstone InstitutesSan FranciscoCAUSA
- Department of Epidemiology and BiostatisticsUniversity of CaliforniaSan FranciscoCAUSA
| | - Adam S. Heath
- Department of Epidemiology, Human Genetics Center, School of Public HealthThe University of Texas Health Science Center at HoustonHoustonTXUSA
| | - Erica P. Young
- Department of Medicine, Division of CardiologyWashington University School of MedicineSaint LouisMOUSA
- McDonnell Genome Institute, Washington University School of MedicineSaint LouisMOUSA
| | - Nathan O. Stitziel
- Department of Medicine, Division of CardiologyWashington University School of MedicineSaint LouisMOUSA
- McDonnell Genome Institute, Washington University School of MedicineSaint LouisMOUSA
- Department of GeneticsWashington University School of MedicineSaint LouisMOUSA
| | - Cecelia Laurie
- Department of BiostatisticsUniversity of WashingtonSeattleWAUSA
| | - Jai G. Broome
- Department of BiostatisticsUniversity of WashingtonSeattleWAUSA
- Department of Medicine, Division of Internal MedicineUniversity of WashingtonSeattleWAUSA
| | - Alyna T. Khan
- Department of BiostatisticsUniversity of WashingtonSeattleWAUSA
| | - Joshua P. Lewis
- Department of MedicineUniversity of Maryland School of MedicineBaltimoreMDUSA
| | - Huichun Xu
- Department of MedicineUniversity of Maryland School of MedicineBaltimoreMDUSA
| | - May E. Montasser
- Department of MedicineUniversity of Maryland School of MedicineBaltimoreMDUSA
| | - Kellan E. Ashley
- Department of MedicineUniversity of Mississippi Medical CenterJacksonMSUSA
| | - Natalie R. Hasbani
- Department of Epidemiology, Human Genetics Center, School of Public HealthThe University of Texas Health Science Center at HoustonHoustonTXUSA
| | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics Center, School of Public HealthThe University of Texas Health Science Center at HoustonHoustonTXUSA
- Human Genome Sequencing CenterBaylor College of MedicineHoustonTXUSA
| | - Alanna C. Morrison
- Department of Epidemiology, Human Genetics Center, School of Public HealthThe University of Texas Health Science Center at HoustonHoustonTXUSA
| | - Nathalie Chami
- The Charles Bronfman Institute for Personalized MedicineIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Ron Do
- The Charles Bronfman Institute for Personalized MedicineIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Ghislain Rocheleau
- The Charles Bronfman Institute for Personalized MedicineIcahn School of Medicine at Mount SinaiNew YorkNYUSA
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | | | - Rozenn N. Lemaitre
- Department of Medicine, Cardiovascular Health Research UnitUniversity of WashingtonSeattleWAUSA
| | - Joshua C. Bis
- Department of Medicine, Cardiovascular Health Research UnitUniversity of WashingtonSeattleWAUSA
| | - James S. Floyd
- Department of Medicine, Cardiovascular Health Research UnitUniversity of WashingtonSeattleWAUSA
- Department of EpidemiologyUniversity of WashingtonSeattleWAUSA
| | - Gregory L. Kinney
- Department of EpidemiologyColorado School of Public HealthAuroraCOUSA
| | - Donald W. Bowden
- Department of BiochemistryWake Forest University School of MedicineWinston‐SalemNCUSA
| | - Nicholette D. Palmer
- Department of BiochemistryWake Forest University School of MedicineWinston‐SalemNCUSA
| | - Emelia J. Benjamin
- Department of Medicine, Cardiovascular Medicine, Boston Medical CenterBoston University Chobanian & Avedisian School of MedicineBostonMAUSA
- Department of EpidemiologyBoston University School of Public HealthBostonMAUSA
| | - Matthew Nayor
- Department of Medicine, Cardiovascular MedicineBoston University Chobanian & Avedisian School of MedicineBostonMAUSA
- Department of Medicine, Preventive Medicine & EpidemiologyBoston University Chobanian & Avedisian School of MedicineBostonMAUSA
| | - Lisa R. Yanek
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Brian G. Kral
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Lewis C. Becker
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Sharon L. R. Kardia
- Department of EpidemiologyUniversity of Michigan School of Public HealthAnn ArborMIUSA
| | - Jennifer A. Smith
- Department of EpidemiologyUniversity of Michigan School of Public HealthAnn ArborMIUSA
- Institute for Social ResearchSurvey Research Center, University of MichiganAnn ArborMIUSA
| | - Lawrence F. Bielak
- Department of EpidemiologyUniversity of Michigan School of Public HealthAnn ArborMIUSA
| | - Arnita F. Norwood
- Department of MedicineUniversity of Mississippi Medical CenterJacksonMSUSA
| | - Yuan‐I Min
- Department of MedicineUniversity of Mississippi Medical CenterJacksonMSUSA
| | - April P. Carson
- Department of MedicineUniversity of Mississippi Medical CenterJacksonMSUSA
| | - Wendy S. Post
- Department of Medicine, Division of CardiologyJohns Hopkins UniversityBaltimoreMDUSA
| | - Stephen S. Rich
- Department of Genome SciencesUniversity of Virginia School of MedicineCharlottesvilleVAUSA
| | - David Herrington
- Department of MedicineWake Forest University School of MedicineWinston‐SalemNCUSA
| | - Xiuqing Guo
- Department of Pediatrics, The Institute for Translational Genomics and Population SciencesThe Lundquist Institute for Biomedical Innovation at Harbor‐UCLA Medical CenterTorranceCAUSA
| | - Kent D. Taylor
- Department of Pediatrics, The Institute for Translational Genomics and Population SciencesThe Lundquist Institute for Biomedical Innovation at Harbor‐UCLA Medical CenterTorranceCAUSA
| | - JoAnn E. Manson
- Department of MedicineBrigham and Women’s Hospital, Harvard Medical SchoolBostonMAUSA
| | - Nora Franceschini
- Department of EpidemiologyUniversity of North Carolina at Chapel HillChapel HillNCUSA
| | - Katherine S. Pollard
- Data Science and Biotechnology, Gladstone InstitutesSan FranciscoCAUSA
- Department of Epidemiology and BiostatisticsUniversity of CaliforniaSan FranciscoCAUSA
- Chan Zuckerberg BiohubSan FranciscoCAUSA
| | - Braxton D. Mitchell
- Department of MedicineUniversity of Maryland School of MedicineBaltimoreMDUSA
- Geriatric Research and Education Clinical CenterBaltimore Veterans Administration Medical CenterBaltimoreMDUSA
| | - Ruth J. F. Loos
- The Charles Bronfman Institute for Personalized MedicineIcahn School of Medicine at Mount SinaiNew YorkNYUSA
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic ResearchUniversity of CopenhagenCopenhagenDenmark
| | - Myriam Fornage
- Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science Center at HoustonHoustonTXUSA
| | - Lifang Hou
- Department of Preventive MedicineNorthwestern UniversityChicagoILUSA
| | - Bruce M. Psaty
- Department of Medicine, Cardiovascular Health Research UnitUniversity of WashingtonSeattleWAUSA
- Department of EpidemiologyUniversity of WashingtonSeattleWAUSA
- Department of Health Systems and Population HealthUniversity of WashingtonSeattleWAUSA
| | - Kendra A. Young
- Department of EpidemiologyColorado School of Public HealthAuroraCOUSA
| | | | - Barry I. Freedman
- Department of Internal Medicine, Section on NephrologyWake Forest University School of MedicineWinston‐SalemNCUSA
| | | | - Daniel Levy
- Division of Intramural Research, Population Sciences BranchNational Heart, Lung, and Blood Institute, National Institutes of HealthBethesdaMDUSA
| | - Rasika A. Mathias
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Patricia A. Peyser
- Department of EpidemiologyUniversity of Michigan School of Public HealthAnn ArborMIUSA
| | - Laura M. Raffield
- Department of GeneticsUniversity of North Carolina at Chapel HillChapel HillNCUSA
| | | | - Alex P. Reiner
- Division of Public HealthFred Hutchinson Cancer CenterSeattleWAUSA
| | - Jerome I. Rotter
- Department of Pediatrics, The Institute for Translational Genomics and Population SciencesThe Lundquist Institute for Biomedical Innovation at Harbor‐UCLA Medical CenterTorranceCAUSA
| | - Goo Jun
- Department of Epidemiology, Human Genetics Center, School of Public HealthThe University of Texas Health Science Center at HoustonHoustonTXUSA
| | - Paul S. de Vries
- Department of Epidemiology, Human Genetics Center, School of Public HealthThe University of Texas Health Science Center at HoustonHoustonTXUSA
| | - Themistocles L. Assimes
- Department of Medicine, Division of Cardiovascular MedicineStanford University School of MedicineStanfordCAUSA
- VA Palo Alto Healthcare SystemPalo AltoCAUSA
| |
Collapse
|
24
|
Wang C, Li J, Chen J, Wang Z, Zhu G, Song L, Wu J, Li C, Qiu R, Chen X, Zhang L, Li W. Multi-omics analyses reveal biological and clinical insights in recurrent stage I non-small cell lung cancer. Nat Commun 2025; 16:1477. [PMID: 39929832 PMCID: PMC11811181 DOI: 10.1038/s41467-024-55068-2] [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: 06/27/2022] [Accepted: 11/26/2024] [Indexed: 02/13/2025] Open
Abstract
Post-operative recurrence rates of stage I non-small cell lung cancer (NSCLC) range from 20% to 40%. Nonetheless, the molecular mechanisms underlying recurrence hitherto remain largely elusive. Here, we generate genomic, epigenomic and transcriptomic profiles of paired tumors and adjacent tissues from 122 stage I NSCLC patients, among which 57 patients develop recurrence after surgery during follow-up. Integrated analyses illustrate that the presence of predominantly solid or micropapillary histological subtypes, increased genomic instability, and APOBEC-related signature are associated with recurrence. Furthermore, TP53 missense mutation in DNA-binding domain could contribute to shorter time to recurrence. DNA hypomethylation is pronounced in recurrent NSCLC, and PRAME is the significantly hypomethylated and overexpressed gene in recurrent lung adenocarcinoma (LUAD). Mechanistically, hypomethylation at TEAD1 binding site facilitates the transcriptional activation of PRAME. Inhibition of PRAME restrains the tumor metastasis via downregulation of epithelial-mesenchymal transition-related genes. We also identify that enrichment of AT2 cells with higher copy number variation burden, exhausted CD8 + T cells and Macro_SPP1, along with the reduced interaction between AT2 and immune cells, is essential for the formation of ecosystem in recurrent LUAD. Finally, multi-omics clustering could stratify the NSCLC patients into 4 subclusters with varying recurrence risk and subcluster-specific therapeutic vulnerabilities. Collectively, this study constitutes a promising resource enabling insights into the biological mechanisms and clinical management for post-operative recurrence of stage I NSCLC.
Collapse
Affiliation(s)
- Chengdi Wang
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Jingwei Li
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jingyao Chen
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhoufeng Wang
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Guonian Zhu
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lujia Song
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiayang Wu
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Changshu Li
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Rong Qiu
- Department of Respiratory and Critical Care Medicine, Suining Central Hospital, Suining, China
| | - Xuelan Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Chengdu, Sichuan, China
| | - Li Zhang
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Weimin Li
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
25
|
Gong J, Sun H, Wang K, Zhao Y, Huang Y, Chen Q, Qiao H, Gao Y, Zhao J, Ling Y, Cao R, Tan J, Wang Q, Ma Y, Li J, Luo J, Wang S, Wang J, Zhang G, Xu S, Qian F, Zhou F, Tang H, Li D, Sedlazeck FJ, Jin L, Guan Y, Fan S. Long-read sequencing of 945 Han individuals identifies structural variants associated with phenotypic diversity and disease susceptibility. Nat Commun 2025; 16:1494. [PMID: 39929826 PMCID: PMC11811171 DOI: 10.1038/s41467-025-56661-9] [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: 03/05/2024] [Accepted: 01/22/2025] [Indexed: 02/13/2025] Open
Abstract
Genomic structural variants (SVs) are a major source of genetic diversity in humans. Here, through long-read sequencing of 945 Han Chinese genomes, we identify 111,288 SVs, including 24.56% unreported variants, many with predicted functional importance. By integrating human population-level phenotypic and multi-omics data as well as two humanized mouse models, we demonstrate the causal roles of two SVs: one SV that emerges at the common ancestor of modern humans, Neanderthals, and Denisovans in GSDMD for bone mineral density and one modern-human-specific SV in WWP2 impacting height, weight, fat, craniofacial phenotypes and immunity. Our results suggest that the GSDMD SV could serve as a rapid and cost-effective biomarker for assessing the risk of cisplatin-induced acute kidney injury. The functional conservation from human to mouse and widespread signals of positive natural selection suggest that both SVs likely influence local adaptation, phenotypic diversity, and disease susceptibility across diverse human populations.
Collapse
Affiliation(s)
- Jiao Gong
- State Key Laboratory of Genetic Engineering, Lab for Evolutionary Synthesis, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Huiru Sun
- State Key Laboratory of Genetic Engineering, Lab for Evolutionary Synthesis, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Kaiyuan Wang
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Yanhui Zhao
- State Key Laboratory of Genetic Engineering, Lab for Evolutionary Synthesis, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Yechao Huang
- State Key Laboratory of Genetic Engineering, Lab for Evolutionary Synthesis, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Qinsheng Chen
- State Key Laboratory of Genetic Engineering, Lab for Evolutionary Synthesis, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Hui Qiao
- State Key Laboratory of Genetic Engineering, Lab for Evolutionary Synthesis, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Yang Gao
- State Key Laboratory of Genetic Engineering, Lab for Evolutionary Synthesis, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Jialin Zhao
- State Key Laboratory of Genetic Engineering, Lab for Evolutionary Synthesis, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Yunchao Ling
- Bio-Med Big Data Center, Chinese Academy of Sciences Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ruifang Cao
- Bio-Med Big Data Center, Chinese Academy of Sciences Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jingze Tan
- State Key Laboratory of Genetic Engineering, Lab for Evolutionary Synthesis, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Qi Wang
- State Key Laboratory of Genetic Engineering, Lab for Evolutionary Synthesis, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Yanyun Ma
- Department of Anthropology and Human Genetics, Institute for Six-sector Economy, and MOE Key Laboratory of Contemporary Anthropology, Fudan University, Shanghai, China
| | - Jing Li
- State Key Laboratory of Genetic Engineering, Lab for Evolutionary Synthesis, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Jingchun Luo
- State Key Laboratory of Genetic Engineering, Lab for Evolutionary Synthesis, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Sijia Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering, Lab for Evolutionary Synthesis, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
- Research Unit of dissecting the population genetics and developing new technologies for treatment and prevention of skin phenotypes and dermatological diseases (2019RU058), Chinese Academy of Medical Sciences, Shanghai, China
| | - Guoqing Zhang
- Bio-Med Big Data Center, Chinese Academy of Sciences Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Shuhua Xu
- State Key Laboratory of Genetic Engineering, Lab for Evolutionary Synthesis, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Feng Qian
- State Key Laboratory of Genetic Engineering, Lab for Evolutionary Synthesis, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Fang Zhou
- School of Data Science and Engineering, East China Normal University, Shanghai, China
| | - Huiru Tang
- State Key Laboratory of Genetic Engineering, Lab for Evolutionary Synthesis, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Dali Li
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Fritz J Sedlazeck
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA.
| | - Li Jin
- State Key Laboratory of Genetic Engineering, Lab for Evolutionary Synthesis, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China.
- Research Unit of dissecting the population genetics and developing new technologies for treatment and prevention of skin phenotypes and dermatological diseases (2019RU058), Chinese Academy of Medical Sciences, Shanghai, China.
| | - Yuting Guan
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China.
| | - Shaohua Fan
- State Key Laboratory of Genetic Engineering, Lab for Evolutionary Synthesis, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China.
| |
Collapse
|
26
|
Corbett RJ, Kaufman RS, McQuaid SW, Vaksman Z, Phul S, Brown MA, Mason JL, Waszak SM, Zhang B, Zhong C, Desai H, Hausler R, Naqvi AS, Chroni A, Geng Z, Gonzalez EM, Zhu Y, Heath AP, Li M, Storm PB, Resnick AC, Maxwell KN, Cole KA, Waanders AJ, Bornhorst M, MacFarland SP, Rokita JL, Diskin SJ. Germline pathogenic variation impacts somatic alterations and patient outcomes in pediatric CNS tumors. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.04.25321499. [PMID: 39974082 PMCID: PMC11838646 DOI: 10.1101/2025.02.04.25321499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The contribution of rare pathogenic/likely pathogenic (P/LP) germline variants to pediatric central nervous system (CNS) tumor development remains understudied. Here, we characterized the prevalence and clinical significance of germline P/LP variants in cancer predisposition genes across 830 CNS tumor patients from the Pediatric Brain Tumor Atlas (PBTA). We identified germline P/LP variants in 24.2% (201/830) of patients and the majority (154/201) lacked clinical reporting of genetic tumor syndromes. Among P/LP carriers, 30.7% had putative somatic second hits or loss of function tumor alterations. Finally, we linked pathogenic germline variation with novel somatic events and patient survival to highlight the impact of germline variation on tumorigenesis and patient outcomes.
Collapse
Affiliation(s)
- Ryan J Corbett
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Rebecca S Kaufman
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Shelly W McQuaid
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Division of Hematology, Oncology, Neuro-Oncology, and Stem Cell Transplantation, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Zalman Vaksman
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Saksham Phul
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Miguel A Brown
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jennifer L Mason
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sebastian M Waszak
- Laboratory of Computational Neuro-Oncology, Swiss Institute for Experimental Cancer Research, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Bo Zhang
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Chuwei Zhong
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Heena Desai
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ryan Hausler
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ammar S Naqvi
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Antonia Chroni
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Zhuangzhuang Geng
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Elizabeth M Gonzalez
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Yuankun Zhu
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Allison P Heath
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Marilyn Li
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Phillip B Storm
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Adam C Resnick
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kara N Maxwell
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kristina A Cole
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Oncology, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Angela J Waanders
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Division of Hematology, Oncology, Neuro-Oncology, and Stem Cell Transplantation, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Miriam Bornhorst
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Division of Hematology, Oncology, Neuro-Oncology, and Stem Cell Transplantation, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Suzanne P MacFarland
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Oncology, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jo Lynne Rokita
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA
- Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Sharon J Diskin
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Oncology, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
27
|
Negi S, Stenton SL, Berger SI, Canigiula P, McNulty B, Violich I, Gardner J, Hillaker T, O'Rourke SM, O'Leary MC, Carbonell E, Austin-Tse C, Lemire G, Serrano J, Mangilog B, VanNoy G, Kolmogorov M, Vilain E, O'Donnell-Luria A, Délot E, Miga KH, Monlong J, Paten B. Advancing long-read nanopore genome assembly and accurate variant calling for rare disease detection. Am J Hum Genet 2025; 112:428-449. [PMID: 39862869 PMCID: PMC11866955 DOI: 10.1016/j.ajhg.2025.01.002] [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: 08/21/2024] [Revised: 12/22/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
More than 50% of families with suspected rare monogenic diseases remain unsolved after whole-genome analysis by short-read sequencing (SRS). Long-read sequencing (LRS) could help bridge this diagnostic gap by capturing variants inaccessible to SRS, facilitating long-range mapping and phasing and providing haplotype-resolved methylation profiling. To evaluate LRS's additional diagnostic yield, we sequenced a rare-disease cohort of 98 samples from 41 families, using nanopore sequencing, achieving per sample ∼36× average coverage and 32-kb read N50 from a single flow cell. Our Napu pipeline generated assemblies, phased variants, and methylation calls. LRS covered, on average, coding exons in ∼280 genes and ∼5 known Mendelian disease-associated genes that were not covered by SRS. In comparison to SRS, LRS detected additional rare, functionally annotated variants, including structural variants (SVs) and tandem repeats, and completely phased 87% of protein-coding genes. LRS detected additional de novo variants and could be used to distinguish postzygotic mosaic variants from prezygotic de novos. Diagnostic variants were established by LRS in 11 probands, with diverse underlying genetic causes including de novo and compound heterozygous variants, large-scale SVs, and epigenetic modifications. Our study demonstrates LRS's potential to enhance diagnostic yield for rare monogenic diseases, implying utility in future clinical genomics workflows.
Collapse
Affiliation(s)
- Shloka Negi
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Sarah L Stenton
- Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Seth I Berger
- Children's National Research Institute, Washington, DC, USA
| | | | - Brandy McNulty
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Ivo Violich
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Joshua Gardner
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Todd Hillaker
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Sara M O'Rourke
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Melanie C O'Leary
- Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Elizabeth Carbonell
- Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Christina Austin-Tse
- Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Gabrielle Lemire
- Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jillian Serrano
- Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Brian Mangilog
- Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Grace VanNoy
- Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Mikhail Kolmogorov
- Cancer Data Science Laboratory, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Eric Vilain
- Institute for Clinical and Translational Science, University of California, Irvine, Irvine, CA, USA
| | - Anne O'Donnell-Luria
- Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Emmanuèle Délot
- Institute for Clinical and Translational Science, University of California, Irvine, Irvine, CA, USA
| | - Karen H Miga
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Jean Monlong
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France.
| | - Benedict Paten
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA.
| |
Collapse
|
28
|
Laurie S, Steyaert W, de Boer E, Polavarapu K, Schuermans N, Sommer AK, Demidov G, Ellwanger K, Paramonov I, Thomas C, Aretz S, Baets J, Benetti E, Bullich G, Chinnery PF, Clayton-Smith J, Cohen E, Danis D, de Sainte Agathe JM, Denommé-Pichon AS, Diaz-Manera J, Efthymiou S, Faivre L, Fernandez-Callejo M, Freeberg M, Garcia-Pelaez J, Guillot-Noel L, Haack TB, Hanna M, Hengel H, Horvath R, Houlden H, Jackson A, Johansson L, Johari M, Kamsteeg EJ, Kellner M, Kleefstra T, Lacombe D, Lochmüller H, López-Martín E, Macaya A, Marcé-Grau A, Maver A, Morsy H, Muntoni F, Musacchia F, Nelson I, Nigro V, Olimpio C, Oliveira C, Paulasová Schwabová J, Pauly MG, Peterlin B, Peters S, Pfundt R, Piluso G, Piscia D, Posada M, Reich S, Renieri A, Ryba L, Šablauskas K, Savarese M, Schöls L, Schütz L, Steinke-Lange V, Stevanin G, Straub V, Sturm M, Swertz MA, Tartaglia M, Te Paske IBAW, Thompson R, Torella A, Trainor C, Udd B, Van de Vondel L, van de Warrenburg B, van Reeuwijk J, Vandrovcova J, Vitobello A, Vos J, Vyhnálková E, Wijngaard R, Wilke C, William D, Xu J, Yaldiz B, Zalatnai L, Zurek B, Brookes AJ, Evangelista T, Gilissen C, Graessner H, Hoogerbrugge N, Ossowski S, Riess O, Schüle R, Synofzik M, et alLaurie S, Steyaert W, de Boer E, Polavarapu K, Schuermans N, Sommer AK, Demidov G, Ellwanger K, Paramonov I, Thomas C, Aretz S, Baets J, Benetti E, Bullich G, Chinnery PF, Clayton-Smith J, Cohen E, Danis D, de Sainte Agathe JM, Denommé-Pichon AS, Diaz-Manera J, Efthymiou S, Faivre L, Fernandez-Callejo M, Freeberg M, Garcia-Pelaez J, Guillot-Noel L, Haack TB, Hanna M, Hengel H, Horvath R, Houlden H, Jackson A, Johansson L, Johari M, Kamsteeg EJ, Kellner M, Kleefstra T, Lacombe D, Lochmüller H, López-Martín E, Macaya A, Marcé-Grau A, Maver A, Morsy H, Muntoni F, Musacchia F, Nelson I, Nigro V, Olimpio C, Oliveira C, Paulasová Schwabová J, Pauly MG, Peterlin B, Peters S, Pfundt R, Piluso G, Piscia D, Posada M, Reich S, Renieri A, Ryba L, Šablauskas K, Savarese M, Schöls L, Schütz L, Steinke-Lange V, Stevanin G, Straub V, Sturm M, Swertz MA, Tartaglia M, Te Paske IBAW, Thompson R, Torella A, Trainor C, Udd B, Van de Vondel L, van de Warrenburg B, van Reeuwijk J, Vandrovcova J, Vitobello A, Vos J, Vyhnálková E, Wijngaard R, Wilke C, William D, Xu J, Yaldiz B, Zalatnai L, Zurek B, Brookes AJ, Evangelista T, Gilissen C, Graessner H, Hoogerbrugge N, Ossowski S, Riess O, Schüle R, Synofzik M, Verloes A, Matalonga L, Brunner HG, Lohmann K, de Voer RM, Töpf A, Vissers LELM, Beltran S, Hoischen A. Genomic reanalysis of a pan-European rare-disease resource yields new diagnoses. Nat Med 2025; 31:478-489. [PMID: 39825153 PMCID: PMC11835725 DOI: 10.1038/s41591-024-03420-w] [Show More Authors] [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: 05/01/2023] [Accepted: 11/14/2024] [Indexed: 01/20/2025]
Abstract
Genetic diagnosis of rare diseases requires accurate identification and interpretation of genomic variants. Clinical and molecular scientists from 37 expert centers across Europe created the Solve-Rare Diseases Consortium (Solve-RD) resource, encompassing clinical, pedigree and genomic rare-disease data (94.5% exomes, 5.5% genomes), and performed systematic reanalysis for 6,447 individuals (3,592 male, 2,855 female) with previously undiagnosed rare diseases from 6,004 families. We established a collaborative, two-level expert review infrastructure that allowed a genetic diagnosis in 506 (8.4%) families. Of 552 disease-causing variants identified, 464 (84.1%) were single-nucleotide variants or short insertions/deletions. These variants were either located in recently published novel disease genes (n = 67), recently reclassified in ClinVar (n = 187) or reclassified by consensus expert decision within Solve-RD (n = 210). Bespoke bioinformatics analyses identified the remaining 15.9% of causative variants (n = 88). Ad hoc expert review, parallel to the systematic reanalysis, diagnosed 249 (4.1%) additional families for an overall diagnostic yield of 12.6%. The infrastructure and collaborative networks set up by Solve-RD can serve as a blueprint for future further scalable international efforts. The resource is open to the global rare-disease community, allowing phenotype, variant and gene queries, as well as genome-wide discoveries.
Collapse
Grants
- The Solve-RD consortium is grateful to all involved rare disease patients and their families as well as other contributors to Solve-RD. The Solve-RD project has received funding from the European Union’s Horizon 2020 research and innovation programme under grant agreement No 779257 (to all authors). This research is supported (not financially) by four ERNs: (1) The ERN for Intellectual Disability, Telehealth and Congenital Anomalies (ERN ITHACA)—Project ID No 101085231; (2) The ERN on Rare Neurological Diseases (ERN RND)—Project ID No 101155994; (3) The ERN for Neuromuscular Diseases (ERN Euro-NMD)—Project ID No 101156434; (4) The ERN on Genetic Tumour Risk Syndromes (ERN GENTURIS)—Project ID No 101155809. The ERNs are co-funded by the European Union within the framework of the Third Health Programme. The RD-Connect Genome-Phenome Analysis platform developed under FP7/2007–2013 funded project (grant agreement n° 305444) and ongoing funding from EJP-RD (grant numbers H2020 779257, H2020 825575), Instituto de Salud Carlos III (Grant numbers PT13/0001/0044, PT17/0009/0019; Instituto Nacional de Bioinformática, INB), ELIXIR-EXCELERATE (Grant number EU H2020 #676559) and ELIXIR Implementation Studies (Remote real-time visualisation of human rare disease genomics data (RD-Connect) stored at the EGA ELIXIR. 2017-2018; ELIXIR IT-2017-INTEGRATION, Rare Disease Infrastructure ELIXIR, 2019-2020 and the Beacon ELIXIR, 2019-2021). The RD-Connect GPAP has leveraged developments funded through project VEIS (001-P-001647 co-financed by the European Regional Development Fund of the European Union in the framework of the Operational Program FEDER of Catalonia 2014-2020 with the support of the Secretaria d'Universitats i Recerca del Departament d'Empresa i Coneixement de la Generalitat de Catalunya) and URD-Cat (PERIS SLT002/16/00174, Departament de Salut, Generalitat de Catalunya). The Spanish academic and research network RedIris (https://www.rediris.es/) provided the Aspera service used for uploading raw data for processing to the RD-Connect GPAP, and for transferring data between centres. Netherlands Science Organisations (NWO VIDI 917.164.55 to C.G.). Ministero della Salute (Genoma mEdiciNa pERsonalizzatA, T3-AN-04, to V.N., A.R., and M.T.). The “Network for Italian Genomes - NIG”, “Cell lines and DNA bank of Rett Syndrome, X-linked mental retardation and other genetic diseases”, member of the Telethon Network of Genetic Biobanks (project no. GTB12001), and EuroBioBank network. H.L. receives support from the Canadian Institutes of Health Research (CIHR) for Foundation Grant FDN-167281 (Precision Health for Neuromuscular Diseases), Transnational Team Grant ERT-174211 (ProDGNE) and Network Grant OR2-189333 (NMD4C), from the Canada Foundation for Innovation (CFI-JELF 38412), the Canada Research Chairs program (Canada Research Chair in Neuromuscular Genomics and Health, 950-232279), the European Commission (grant number 101080249) and the Canada Research Coordinating Committee New Frontiers in Research Fund (NFRFG-2022-00033) for SIMPATHIC, and from the Government of Canada Canada First Research Excellence Fund (CFREF) for the Brain-Heart Interconnectome (CFREF-2022-00007). K.P. is a recipient of a Canadian Institutes of Health Research (CIHR) postdoctoral fellowship award under award no: MFE-491707 This work was furthermore supported by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) No 441409627, as part of the PROSPAX consortium under the frame of EJP RD, the European Joint Programme on Rare Diseases, under the EJP RD COFUND-EJP N° 825575 (to M.Sy., R.S., and R.H.,) and the Clinician Scientist programme "PRECISE.net" funded by the Else Kröner-Fresenius-Stiftung (to C.W., M.K., R.S. and M.Sy.). J.P.S. was financed by Programme EXCELES, (ID Project No. LX22NPO5107) - Funded by the European Union – Next Generation EU. B.v.d.W. is supported by ZonMW, the Gossweiler Foundation, and the ‘Hersenstichting’. The work of F.Mun. was also supported by Muscular Dystrophy UK, and Muscular Dystrophy USA. H.G. and T.B.H. are supported by the European Union's Horizon 2020 research and innovation program project Recon4IMD (grant number 101080997). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.
Collapse
Affiliation(s)
- Steven Laurie
- Centro Nacional de Análisis Genómico (CNAG), Barcelona, Spain
- Universitat de Barcelona (UB), Barcelona, Spain
| | - Wouter Steyaert
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Radboud Institute for Medical Innovation, Nijmegen, the Netherlands
| | - Elke de Boer
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Kiran Polavarapu
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Nika Schuermans
- Program for Undiagnosed Rare Diseases (UD-PrOZA), Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Anna K Sommer
- Institute of Human Genetics, Medical Faculty, University of Bonn, Bonn, Germany
| | - German Demidov
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Kornelia Ellwanger
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Ida Paramonov
- Centro Nacional de Análisis Genómico (CNAG), Barcelona, Spain
- Universitat de Barcelona (UB), Barcelona, Spain
| | - Coline Thomas
- European Bioinformatics Institute, European Molecular Biology Laboratory, Cambridge, UK
| | - Stefan Aretz
- Institute of Human Genetics, Medical Faculty, University of Bonn, Bonn, Germany
- Center for Hereditary Tumor Syndromes, University Hospital Bonn, Bonn, Germany
| | - Jonathan Baets
- Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- Laboratory of Neuromuscular Pathology, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
- Neuromuscular Reference Centre, Department of Neurology, Antwerp University Hospital, Antwerp, Belgium
| | - Elisa Benetti
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy
- Medical Genetics, University of Siena, Siena, Italy
| | - Gemma Bullich
- Centro Nacional de Análisis Genómico (CNAG), Barcelona, Spain
- Universitat de Barcelona (UB), Barcelona, Spain
| | - Patrick F Chinnery
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Jill Clayton-Smith
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University Hospitals NHS Foundation Trust, Health Innovation Manchester, Manchester, UK
| | - Enzo Cohen
- Centre de Recherche en Myologie, Sorbonne Université, Inserm, Institut de Myologie, Paris, France
| | - Daniel Danis
- Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Jean-Madeleine de Sainte Agathe
- Department of Genetics, Assistance Publique-Hôpitaux de Paris, Sorbonne Université, Pitié-Salpêtrière University Hospital, Paris, France
| | - Anne-Sophie Denommé-Pichon
- University of Burgundy, Dijon, France
- Functional Unit for Diagnostic Innovation in Rare Diseases, Dijon Bourgogne University Hospital, Dijon, France
| | - Jordi Diaz-Manera
- John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Stephanie Efthymiou
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Laurence Faivre
- University of Burgundy, Dijon, France
- Genetics Department, Dijon University Hospital, Dijon, France
- Centre of Reference for Rare Diseases: Development Disorders and Malformation Syndromes, Dijon University Hospital, Dijon, France
- University of Burgundy-Franche Comté, Dijon, France
- GIMI institute, Dijon University Hospital, Dijon, France
| | - Marcos Fernandez-Callejo
- Centro Nacional de Análisis Genómico (CNAG), Barcelona, Spain
- Universitat de Barcelona (UB), Barcelona, Spain
| | - Mallory Freeberg
- European Bioinformatics Institute, European Molecular Biology Laboratory, Cambridge, UK
| | - José Garcia-Pelaez
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
| | | | - Tobias B Haack
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Mike Hanna
- MRC Centre for Neuromuscular Diseases and National Hospital for Neurology and Neurosurgery, UCL Queen Square Institute of Neurology, London, UK
| | - Holger Hengel
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research (HIH), University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Rita Horvath
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Henry Houlden
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Adam Jackson
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University Hospitals NHS Foundation Trust, Health Innovation Manchester, Manchester, UK
| | - Lennart Johansson
- Department of Genetics, Genomics Coordination Center, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Mridul Johari
- Folkhälsan Research Centre and Medicum, University of Helsinki, Helsinki, Finland
| | - Erik-Jan Kamsteeg
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Melanie Kellner
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research (HIH), University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Tjitske Kleefstra
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Clinical Genetics, Erasmus MC, Rotterdam, the Netherlands
- Center of Excellence for Neuropsychiatry, Vincent van Gogh Institute for Psychiatry, Venray, the Netherlands
| | - Didier Lacombe
- MRGM, Maladies Rares: Génétique et Métabolisme, INSERM U1211, Université de Bordeaux, Bordeaux, France
- Service de Génétique Médicale, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Hanns Lochmüller
- Centro Nacional de Análisis Genómico (CNAG), Barcelona, Spain
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Ontario, Canada
- Department of Neuropediatrics and Muscle Disorders, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Division of Neurology, Department of Medicine, The Ottawa Hospital, Ottawa, Ontario, Canada
- Brain and Mind Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Estrella López-Martín
- Institute of Rare Diseases Research, Spanish Undiagnosed Rare Diseases Cases Program (SpainUDP) & Undiagnosed Diseases Network International (UDNI), Instituto de Salud Carlos III, Madrid, Spain
| | - Alfons Macaya
- Pediatric Neurology Research Group, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Anna Marcé-Grau
- Pediatric Neurology Research Group, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Aleš Maver
- Clinical Institute of Genomic Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Heba Morsy
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
- Department of Human Genetics, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Hospital, London, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - Francesco Musacchia
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania "Luigi Vanvitelli", Naples, Italy
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Isabelle Nelson
- Centre de Recherche en Myologie, Sorbonne Université, Inserm, Institut de Myologie, Paris, France
| | - Vincenzo Nigro
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania "Luigi Vanvitelli", Naples, Italy
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Catarina Olimpio
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- East Anglian Medical Genetics Service, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Carla Oliveira
- IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
- Institut du Cerveau, Sorbonne University, Paris, France
| | - Jaroslava Paulasová Schwabová
- Centre of Hereditary Ataxia, Department of Neurology, Charles University Prague-2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Martje G Pauly
- Institute of Systems Motor Science, University of Lübeck, Lübeck, Germany
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
- Department of Neurology, University Hospital Schleswig Holstein, Lübeck, Germany
| | - Borut Peterlin
- Clinical Institute of Genomic Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Sophia Peters
- Institute of Human Genetics, Medical Faculty, University of Bonn, Bonn, Germany
| | - Rolph Pfundt
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Giulio Piluso
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania "Luigi Vanvitelli", Naples, Italy
| | - Davide Piscia
- Centro Nacional de Análisis Genómico (CNAG), Barcelona, Spain
- Universitat de Barcelona (UB), Barcelona, Spain
| | - Manuel Posada
- Institute of Rare Diseases Research, Spanish Undiagnosed Rare Diseases Cases Program (SpainUDP) & Undiagnosed Diseases Network International (UDNI), Instituto de Salud Carlos III, Madrid, Spain
| | - Selina Reich
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research (HIH), University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Alessandra Renieri
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy
- Medical Genetics, University of Siena, Siena, Italy
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Lukas Ryba
- Department of Biology and Medical Genetics, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Karolis Šablauskas
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Institute of Data Science and Digital Technologies, Vilnius University, Vilnius, Lithuania
| | - Marco Savarese
- Folkhälsan Research Centre and Medicum, University of Helsinki, Helsinki, Finland
| | - Ludger Schöls
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research (HIH), University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Leon Schütz
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Verena Steinke-Lange
- Medizinische Klinik und Poliklinik IV - Campus Innenstadt, Klinikum der Universität München, Munich, Germany
- MGZ - Medical Genetics Center, Munich, Germany
| | | | - Volker Straub
- John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Marc Sturm
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Morris A Swertz
- Department of Genetics, Genomics Coordination Center, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Marco Tartaglia
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Iris B A W Te Paske
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Radboud Institute for Medical Innovation, Nijmegen, the Netherlands
| | - Rachel Thompson
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Annalaura Torella
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania "Luigi Vanvitelli", Naples, Italy
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Christina Trainor
- John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Bjarne Udd
- Folkhälsan Research Centre and Medicum, University of Helsinki, Helsinki, Finland
- Tampere Neuromuscular Center, Tampere, Finland
- Vasa Central Hospital, Vaasa, Finland
| | - Liedewei Van de Vondel
- Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- Laboratory of Neuromuscular Pathology, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
- Peripheral Neuropathy Research Group, University of Antwerp, Antwerp, Belgium
| | - Bart van de Warrenburg
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Neurology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jeroen van Reeuwijk
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jana Vandrovcova
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Antonio Vitobello
- University of Burgundy, Dijon, France
- Functional Unit for Diagnostic Innovation in Rare Diseases, Dijon Bourgogne University Hospital, Dijon, France
| | - Janet Vos
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Radboud Institute for Medical Innovation, Nijmegen, the Netherlands
| | - Emílie Vyhnálková
- Department of Biology and Medical Genetics, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Robin Wijngaard
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Radboud Institute for Medical Innovation, Nijmegen, the Netherlands
| | - Carlo Wilke
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research (HIH), University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Doreen William
- Institute of Clinical Genetics, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany
| | - Jishu Xu
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research (HIH), University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Burcu Yaldiz
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Luca Zalatnai
- Centro Nacional de Análisis Genómico (CNAG), Barcelona, Spain
- Universitat de Barcelona (UB), Barcelona, Spain
| | - Birte Zurek
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Anthony J Brookes
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Teresinha Evangelista
- Centre de Recherche en Myologie, Sorbonne Université, Inserm, Institut de Myologie, Paris, France
| | - Christian Gilissen
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Radboud Institute for Medical Innovation, Nijmegen, the Netherlands
| | - Holm Graessner
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
- Centre for Rare Diseases, University of Tübingen, Tübingen, Germany
| | - Nicoline Hoogerbrugge
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Radboud Institute for Medical Innovation, Nijmegen, the Netherlands
| | - Stephan Ossowski
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
- NGS Competence Center Tübingen (NCCT), University of Tübingen, Tübingen, Germany
| | - Olaf Riess
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
- Centre for Rare Diseases, University of Tübingen, Tübingen, Germany
| | - Rebecca Schüle
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research (HIH), University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Matthis Synofzik
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research (HIH), University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Alain Verloes
- Dept of Genetics, Assistance Publique-Hôpitaux de Paris, Université de Paris, Robert DEBRE University Hospital, Paris, France
- INSERM UMR 1141 "NeuroDiderot", Hôpital Robert DEBRE, Paris, France
| | - Leslie Matalonga
- Centro Nacional de Análisis Genómico (CNAG), Barcelona, Spain
- Universitat de Barcelona (UB), Barcelona, Spain
| | - Han G Brunner
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Clinical Genetics, Maastricht University Medical Centre and GROW School for Development and Oncology, University of Maastricht, Maastricht, the Netherlands
| | - Katja Lohmann
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Richarda M de Voer
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Radboud Institute for Medical Innovation, Nijmegen, the Netherlands
| | - Ana Töpf
- John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Lisenka E L M Vissers
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Sergi Beltran
- Centro Nacional de Análisis Genómico (CNAG), Barcelona, Spain.
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona (UB), Barcelona, Spain.
| | - Alexander Hoischen
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands.
- Radboud Institute for Medical Innovation, Nijmegen, the Netherlands.
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
29
|
Au-Yeung CCY, Cheung YT, Cheng JYT, Ip KWH, Lee SD, Yang VYT, Lau AYT, Lee CKC, Chong PKH, Lau KW, van Lunenburg JTJ, Zheng DFD, Ho BHM, Tik C, Ho KKK, Rajaby R, Au CH, Yu MHC, Sung WK. UniVar: A variant interpretation platform enhancing rare disease diagnosis through robust filtering and unified analysis of SNV, INDEL, CNV and SV. Comput Biol Med 2025; 185:109560. [PMID: 39700857 DOI: 10.1016/j.compbiomed.2024.109560] [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: 08/20/2024] [Revised: 11/24/2024] [Accepted: 12/08/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND Interpreting the pathogenicity of genetic variants associated with rare diseases is a laborious and time-consuming endeavour. To streamline the diagnostic process and lighten the burden of variant interpretation, it is crucial to automate variant annotation and prioritization. Unfortunately, currently available variant interpretation tools lack a unified and comprehensive workflow that can collectively assess the clinical significance of these types of variants together: small nucleotide variants (SNVs), small insertions/deletions (INDELs), copy number variants (CNVs) and structural variants (SVs). RESULTS The Unified Variant Interpretation Platform (UniVar) is a free web server tool that offers an automated and comprehensive workflow on annotation, filtering and prioritization for SNV, INDEL, CNV and SV collectively to identify disease-causing variants for rare diseases in one interface, ensuring accessibility for users even without programming expertise. To filter common CNVs/SVs, a diverse SV catalogue has been generated, that enables robust filtering of common SVs based on population allele frequency. Through benchmarking our SV catalogue, we showed that it is more complete and accurate than the state-of-the-art SV catalogues. Furthermore, to cope with those patients without detailed clinical information, we have developed a novel computational method that enables variant prioritization from gene panels. Our analysis shows that our approach could prioritize pathogenic variants as effective as using HPO terms assigned by clinicians, which adds value for cases without specific clinically assigned HPO terms. Lastly, through a practical case study of disease-causing compound heterozygous variants across SNV and SV, we demonstrated the uniqueness and effectiveness in variant interpretation of UniVar, edging over any existing interpretation tools. CONCLUSIONS UniVar is a unified and versatile platform that empowers researchers and clinicians to identify and interpret disease-causing variants in rare diseases efficiently through a single holistic interface and without a prerequisite for HPO terms. It is freely available without login and installation at https://univar.live/.
Collapse
Affiliation(s)
- Cherie C Y Au-Yeung
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Yuen-Ting Cheung
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Joshua Y T Cheng
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Ken W H Ip
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Sau-Dan Lee
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Victor Y T Yang
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Amy Y T Lau
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Chit K C Lee
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Peter K H Chong
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China
| | - King Wai Lau
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China
| | | | - Damon F D Zheng
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Brian H M Ho
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Crystal Tik
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Kingsley K K Ho
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Ramesh Rajaby
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China; Shibuya Laboratory, Division of Medical Data Informatics, Human Genome Center, University of Tokyo, Japan
| | - Chun-Hang Au
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Mullin H C Yu
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Wing-Kin Sung
- Hong Kong Genome Institute, Hong Kong Science Park, Shatin, Hong Kong, China; Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong, China; Laboratory of Computational Genomics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
30
|
Chiriatti L, Priolo M, Onesimo R, Carvetta M, Leoni C, Bruselles A, Radio FC, Cappelletti C, Ferilli M, Ricci D, Niceta M, Cordeddu V, Ciolfi A, Mancini C, Zampino G, Tartaglia M. The Arg99Gln Substitution in HNRNPC Is Associated with a Distinctive Clinical Phenotype Characterized by Facial Dysmorphism and Ocular and Cochlear Anomalies. Genes (Basel) 2025; 16:176. [PMID: 40004505 PMCID: PMC11854916 DOI: 10.3390/genes16020176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/23/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Heterozygous variants in the heterogeneous nuclear ribonucleoprotein C gene (HNRNPC) have recently been reported to cause intellectual developmental disorder-74 (MRD74), a neurodevelopmental disorder with no recurrent diagnostic handles. Affected individuals show variable, non-specific, and subtle dysmorphic features. The degree of developmental delay (DD)/intellectual disability (ID) is also wide, ranging from mild to severe. The mutational spectrum is relatively broad with exon deletions and splice site and frameshift variants distributed along the entire length of the gene leading to HNRNPC loss of function. Only two missense changes located within the RNA-binding motif (RBM) and adjacent linker region of the more abundant isoform (Arg64Trp and Arg99Gln) have been described. Notably, the Arg99Gln amino acid substitution was reported in a subject presenting with a more complex and unique clinical phenotype characterized by distinctive facial features, DD/ID, cochlear aplasia, and bilateral colobomatous microphthalmia, suggesting the possible occurrence of phenotypic heterogeneity. Results: Here, we report the second individual carrying the Arg99Gln change in HNRNPC and having clinical features with a significant overlap with the peculiar phenotype of the previously described subject, supporting the occurrence of a genotype-phenotype correlation. Conclusions: Due to the concomitant occurrence of ocular and cochlear involvement as recognizable diagnostic handles, we propose that the HNRNPCArg99Gln-related phenotype should be considered as a potential differential diagnosis in subjects with ID and major signs of CHARGE syndrome not fulfilling the minimum criteria for a clinical diagnosis.
Collapse
Affiliation(s)
- Luigi Chiriatti
- Molecular Genetics and Functional Genomics, Bambino Gesù Children’s Hospital, IRCCS, 00143 Rome, Italy; (L.C.); (M.C.); (F.C.R.); (C.C.); (M.F.); (M.N.); (A.C.); (C.M.); (M.T.)
| | - Manuela Priolo
- Medical and Molecular Genetics, AORN A. Cardarelli, 80131 Naples, Italy
| | - Roberta Onesimo
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (R.O.); (C.L.)
| | - Mattia Carvetta
- Molecular Genetics and Functional Genomics, Bambino Gesù Children’s Hospital, IRCCS, 00143 Rome, Italy; (L.C.); (M.C.); (F.C.R.); (C.C.); (M.F.); (M.N.); (A.C.); (C.M.); (M.T.)
- Department of Biochemical Sciences “Alessandro Rossi Fanelli”, Sapienza University, 00185 Rome, Italy
| | - Chiara Leoni
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (R.O.); (C.L.)
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Alessandro Bruselles
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.B.); (V.C.)
| | - Francesca Clementina Radio
- Molecular Genetics and Functional Genomics, Bambino Gesù Children’s Hospital, IRCCS, 00143 Rome, Italy; (L.C.); (M.C.); (F.C.R.); (C.C.); (M.F.); (M.N.); (A.C.); (C.M.); (M.T.)
| | - Camilla Cappelletti
- Molecular Genetics and Functional Genomics, Bambino Gesù Children’s Hospital, IRCCS, 00143 Rome, Italy; (L.C.); (M.C.); (F.C.R.); (C.C.); (M.F.); (M.N.); (A.C.); (C.M.); (M.T.)
- Department of Biomedicine and Prevention, University “Tor Vergata”, 00173 Rome, Italy
| | - Marco Ferilli
- Molecular Genetics and Functional Genomics, Bambino Gesù Children’s Hospital, IRCCS, 00143 Rome, Italy; (L.C.); (M.C.); (F.C.R.); (C.C.); (M.F.); (M.N.); (A.C.); (C.M.); (M.T.)
- Department of Computer, Control and Management Engineering, Sapienza University, 00185 Rome, Italy
| | - Daniela Ricci
- National Centre of Services and Research for Prevention of Blindness and Rehabilitation of Low Vision Patients, IAPB Italia Onlus, 00185 Rome, Italy;
- Pediatric Neuropsychiatric Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Marcello Niceta
- Molecular Genetics and Functional Genomics, Bambino Gesù Children’s Hospital, IRCCS, 00143 Rome, Italy; (L.C.); (M.C.); (F.C.R.); (C.C.); (M.F.); (M.N.); (A.C.); (C.M.); (M.T.)
| | - Viviana Cordeddu
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.B.); (V.C.)
| | - Andrea Ciolfi
- Molecular Genetics and Functional Genomics, Bambino Gesù Children’s Hospital, IRCCS, 00143 Rome, Italy; (L.C.); (M.C.); (F.C.R.); (C.C.); (M.F.); (M.N.); (A.C.); (C.M.); (M.T.)
| | - Cecilia Mancini
- Molecular Genetics and Functional Genomics, Bambino Gesù Children’s Hospital, IRCCS, 00143 Rome, Italy; (L.C.); (M.C.); (F.C.R.); (C.C.); (M.F.); (M.N.); (A.C.); (C.M.); (M.T.)
| | - Giuseppe Zampino
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (R.O.); (C.L.)
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Marco Tartaglia
- Molecular Genetics and Functional Genomics, Bambino Gesù Children’s Hospital, IRCCS, 00143 Rome, Italy; (L.C.); (M.C.); (F.C.R.); (C.C.); (M.F.); (M.N.); (A.C.); (C.M.); (M.T.)
| |
Collapse
|
31
|
Diallo M, Defay-Stinat A, Gindensperger V, Sequeira A, Trimouille A, Javerzat S, Bourgeade L, Plaisant C, Lasseaux E, Michaud V, Drumare I, Arveiler B. A 65 kilobase deletion of the upstream TYR gene region in a family with oculocutaneous albinism type 1. Gene 2025; 935:149079. [PMID: 39510327 DOI: 10.1016/j.gene.2024.149079] [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: 08/27/2024] [Revised: 10/01/2024] [Accepted: 11/04/2024] [Indexed: 11/15/2024]
Abstract
Oculocutaneous albinism type 1 is caused by variants in the TYR (tyrosinase) gene. We describe a family with two affected sibs who inherited the pathogenic missense TYR variant c.1146C > A;p.(Asn382Lys) from their mother and a deletion encompassing 65 kilobase pairs of the upstream region of the gene between hg38 coordinates chr11:89110944 and chr11:89175770, from their father. The deletion likely arose by non-homologous recombination since the regions including the two deletion breakpoints share no sequence homology. The deletion contains a single enhancer element that is homologous to a 5' Tyr core regulatory element in the mouse. A luciferase reporter assay showed that this element had a positive regulatory activity. This represents to our knowledge the first deletion solely restricted to non-coding upstream sequences of the TYR gene. It is assumed that the deletion down-regulates expression of the TYR gene and is therefore pathogenic, allowing to establish the diagnosis of OCA 1 in the patients. This study underscores the need to extend the search for pathogenic variants to regulatory regions either by whole genome sequencing or by targeted next generation sequencing of a panel including entire genes (exons, introns, flanking sequences) in order to improve the diagnostic rate in patients with albinism.
Collapse
Affiliation(s)
- Modibo Diallo
- Laboratoire Maladies Rares, Génétique et Métabolisme, Bordeaux University INSERM U1211, Bordeaux, France
| | - Alicia Defay-Stinat
- Laboratoire Maladies Rares, Génétique et Métabolisme, Bordeaux University INSERM U1211, Bordeaux, France
| | - Victor Gindensperger
- Laboratoire Maladies Rares, Génétique et Métabolisme, Bordeaux University INSERM U1211, Bordeaux, France
| | - Angèle Sequeira
- Laboratoire Maladies Rares, Génétique et Métabolisme, Bordeaux University INSERM U1211, Bordeaux, France
| | - Aurélien Trimouille
- Laboratoire Maladies Rares, Génétique et Métabolisme, Bordeaux University INSERM U1211, Bordeaux, France
| | - Sophie Javerzat
- Laboratoire Maladies Rares, Génétique et Métabolisme, Bordeaux University INSERM U1211, Bordeaux, France
| | - Laetitia Bourgeade
- Service de Génétique Médicale, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Claudio Plaisant
- Service de Génétique Médicale, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Eulalie Lasseaux
- Service de Génétique Médicale, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Vincent Michaud
- Laboratoire Maladies Rares, Génétique et Métabolisme, Bordeaux University INSERM U1211, Bordeaux, France; Service de Génétique Médicale, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Isabelle Drumare
- Service d'Exploration Fonctionnelle de la Vision et de Neuro-Ophtalmologie, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Benoit Arveiler
- Laboratoire Maladies Rares, Génétique et Métabolisme, Bordeaux University INSERM U1211, Bordeaux, France; Service de Génétique Médicale, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France.
| |
Collapse
|
32
|
Espejo Valle-Inclan J, De Noon S, Trevers K, Elrick H, van Belzen IAEM, Zumalave S, Sauer CM, Tanguy M, Butters T, Muyas F, Rust AG, Amary F, Tirabosco R, Giess A, Sosinsky A, Elgar G, Flanagan AM, Cortés-Ciriano I. Ongoing chromothripsis underpins osteosarcoma genome complexity and clonal evolution. Cell 2025; 188:352-370.e22. [PMID: 39814020 DOI: 10.1016/j.cell.2024.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 08/05/2024] [Accepted: 12/07/2024] [Indexed: 01/18/2025]
Abstract
Osteosarcoma is the most common primary cancer of the bone, with a peak incidence in children and young adults. Using multi-region whole-genome sequencing, we find that chromothripsis is an ongoing mutational process, occurring subclonally in 74% of osteosarcomas. Chromothripsis generates highly unstable derivative chromosomes, the ongoing evolution of which drives the acquisition of oncogenic mutations, clonal diversification, and intra-tumor heterogeneity across diverse sarcomas and carcinomas. In addition, we characterize a new mechanism, termed loss-translocation-amplification (LTA) chromothripsis, which mediates punctuated evolution in about half of pediatric and adult high-grade osteosarcomas. LTA chromothripsis occurs when a single double-strand break triggers concomitant TP53 inactivation and oncogene amplification through breakage-fusion-bridge cycles. It is particularly prevalent in osteosarcoma and is not detected in other cancers driven by TP53 mutation. Finally, we identify the level of genome-wide loss of heterozygosity as a strong prognostic indicator for high-grade osteosarcoma.
Collapse
Affiliation(s)
| | - Solange De Noon
- Research Department of Pathology, University College London Cancer Institute, London WC1E 6DD, UK; Department of Histopathology, Royal National Orthopaedic Hospital, Stanmore HA7 4LP, UK
| | - Katherine Trevers
- Research Department of Pathology, University College London Cancer Institute, London WC1E 6DD, UK; Department of Histopathology, Royal National Orthopaedic Hospital, Stanmore HA7 4LP, UK
| | - Hillary Elrick
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton CB10 1SA, UK
| | - Ianthe A E M van Belzen
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton CB10 1SA, UK
| | - Sonia Zumalave
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton CB10 1SA, UK
| | - Carolin M Sauer
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton CB10 1SA, UK
| | - Mélanie Tanguy
- Scientific Research and Development, Genomics England, One Canada Square, London E14 5AB, UK
| | - Thomas Butters
- Research Department of Pathology, University College London Cancer Institute, London WC1E 6DD, UK; Department of Histopathology, Royal National Orthopaedic Hospital, Stanmore HA7 4LP, UK
| | - Francesc Muyas
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton CB10 1SA, UK
| | - Alistair G Rust
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton CB10 1SA, UK
| | - Fernanda Amary
- Research Department of Pathology, University College London Cancer Institute, London WC1E 6DD, UK; Department of Histopathology, Royal National Orthopaedic Hospital, Stanmore HA7 4LP, UK
| | - Roberto Tirabosco
- Research Department of Pathology, University College London Cancer Institute, London WC1E 6DD, UK; Department of Histopathology, Royal National Orthopaedic Hospital, Stanmore HA7 4LP, UK
| | - Adam Giess
- Scientific Research and Development, Genomics England, One Canada Square, London E14 5AB, UK
| | | | - Greg Elgar
- Scientific Research and Development, Genomics England, One Canada Square, London E14 5AB, UK
| | - Adrienne M Flanagan
- Research Department of Pathology, University College London Cancer Institute, London WC1E 6DD, UK; Department of Histopathology, Royal National Orthopaedic Hospital, Stanmore HA7 4LP, UK.
| | - Isidro Cortés-Ciriano
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton CB10 1SA, UK.
| |
Collapse
|
33
|
Li R, Chu H, Gao K, Luo H, Jiang Y. SUMMER: an integrated nanopore sequencing pipeline for variants detection and clinical annotation on the human genome. Funct Integr Genomics 2025; 25:21. [PMID: 39836277 PMCID: PMC11750885 DOI: 10.1007/s10142-025-01534-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] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/09/2025] [Accepted: 01/11/2025] [Indexed: 01/22/2025]
Abstract
Long-read sequencing has emerged as a transformative technology in recent years, offering significant potential for the molecular diagnosis of unresolved genetic disorders. Despite its promise, the comprehensive detection and clinical annotation of genomic variants remain intricate and technically demanding. We present SUMMER, an integrated and structured workflow specifically designed to process raw Nanopore sequencing reads. SUMMER facilitates an in-depth analysis of multiple variant types, including SNV, SV, short tandem repeat and mobile element insertion. For clinical applications, SUMMER employs SvAnna to prioritize SV candidates based on phenotype relevance and utilizes Straglr to provide reference distributions of non-pathogenic unit counts for 55 known pathogenic short tandem repeats. By addressing critical challenges in variant detection and annotation, SUMMER seeks to advance the clinical utility of long-read sequencing in diagnostic genomics. SUMMER is available on the web at https://github.com/carolhuaxia/summer .
Collapse
Affiliation(s)
- Renqiuguo Li
- Children's Medical Center, Peking University First Hospital, No.5 Le Yuan Road, Daxing District, 100034, Beijing, China
| | - Hongyuan Chu
- Children's Medical Center, Peking University First Hospital, No.5 Le Yuan Road, Daxing District, 100034, Beijing, China
| | - Kai Gao
- Children's Medical Center, Peking University First Hospital, No.5 Le Yuan Road, Daxing District, 100034, Beijing, China
| | - Huaxia Luo
- Children's Medical Center, Peking University First Hospital, No.5 Le Yuan Road, Daxing District, 100034, Beijing, China.
| | - Yuwu Jiang
- Children's Medical Center, Peking University First Hospital, No.5 Le Yuan Road, Daxing District, 100034, Beijing, China.
| |
Collapse
|
34
|
Huang Y, Deng W, Huang H, Zhang X, Chen X, Ye J, Luo S, Yu T, Yao H, Du H, He X. Detection of inversion with breakpoints in ARSB causing MPS VI by whole-genome sequencing: lessons learned and best practices. Front Genet 2025; 15:1452498. [PMID: 39845185 PMCID: PMC11750778 DOI: 10.3389/fgene.2024.1452498] [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: 06/21/2024] [Accepted: 12/02/2024] [Indexed: 01/24/2025] Open
Abstract
Introduction Mucopolysaccharidosis type VI (MPSVI), an autosomal recessive lysosomal storage disorder caused by pathogenic variants in ARSB gene. Usually, whole exome sequencing (WES) can identify these variants, and if WES failed to detect causative variants, whole-genome sequencing (WGS) may be considered to investigate deep intronic variations and structural alterations in patients. Methods Whole-exome sequencing (WES) and whole genome sequencing (WGS) were performed in a Chinese family having a boy with suspected diagnosis of MPS with macrocephaly, coarse facial features, broad forehead, thick lips, frontal bossing, craniosynostosis, blue spots, frequent upper respiratory infections, inguinal hernia, and dysostosis multiplex. Lysosomal enzymatic assays for leucocytes were used to assess the activity of arylsulfatase B of the boy's leucocytes. Sanger sequencing and karyotyping analysis were used to validate the variants identified in the boy and his parents. Results This boy diagnosed with MPSVI based on clinical phenotypes and laboratory biochemical assays, and WES identified only a maternally inherited missense variant, c.908G>T (p.Gly303Val), in the ARSB gene. By performing WGS, we found a paracentric inversion involving chromosome 5q14.1q13.2 (78180730-138771424 inv), disrupting the ARSB gene on the proband and his father. The inversion was confirmed through karyotyping analysis, and the breakpoints were validated by agarose gel electrophoresis and Sanger sequencing. Disscussion This study reminds us that WGS should be done when WES failed to achieve a molecular diagonosis, and it also underscores the importance of WGS especially in cases of high clinical suspicion.
Collapse
Affiliation(s)
- Yufeng Huang
- Genetics and Precision Medical Center, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenyue Deng
- Department of Neurosurgery, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Huang
- Genetics and Precision Medical Center, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiankai Zhang
- Genetics and Precision Medical Center, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohong Chen
- Department of Endocrinology and Genetic Metabolism, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian Ye
- Genetics and Precision Medical Center, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sukun Luo
- Genetics and Precision Medical Center, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Yu
- Genetics and Precision Medical Center, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Yao
- Department of Endocrinology and Genetic Metabolism, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Du
- Department of Neurosurgery, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuelian He
- Genetics and Precision Medical Center, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
35
|
Shinawi M, Wegner DJ, Paul AJ, Buchser W, Schmidt R, Sharma J, Sardiello M, Sisco K, Manwaring L, Reynolds M, Fulton R, Fronick C, Shaver A, Huang TY, Carroll A, Roessler K, Halpern AL, Dickson PI, Wambach JA. Atypical free sialic acid storage disorder associated with tissue specific mosaicism of SLC17A5. Mol Genet Metab 2025; 144:109004. [PMID: 39742826 DOI: 10.1016/j.ymgme.2024.109004] [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: 08/14/2024] [Revised: 12/03/2024] [Accepted: 12/10/2024] [Indexed: 01/04/2025]
Abstract
Free sialic acid storage disorder (FSASD) is a rare autosomal recessive lysosomal storage disease caused by pathogenic SLC17A5 variants with variable disease severity. We performed a multidisciplinary evaluation of an adolescent female with suspected lysosomal storage disease and conducted comprehensive studies to uncover the molecular etiology. The proband exhibited intellectual disability, a storage disease gestalt, and mildly elevated urine free sialic acid levels. Skin electron micrographs showed prominent cytoplasmic vacuolation. Clinical exome and genome sequencing identified a maternally-inherited SLC17A5 variant: c.533delC;p.Thr178Asnfs*34. RNASeq of proband skin fibroblasts revealed exon 3 skipping, which was not detected in RNA from proband blood or parental fibroblasts. Targeted deep sequencing of proband fibroblast DNA revealed a 184 bp deletion in ∼15 % of reads, encompassing the 3' end of exon 3. Illumina Complete Long Read sequencing confirmed the deletion was in the paternally-inherited allele and found in a mosaic state in proband fibroblasts and muscle but not in blood or buccal cells. Functional studies, including SLC17A5 knockout cells and transient transfections of mutated SLC17A5 demonstrated pathogenicity of the identified variants. We report an adolescent female with atypical FSASD with tissue-specific mosaicism for an intragenic deletion in SLC17A5, explaining the atypical clinical course, mild biochemical abnormalities, and long diagnostic process.
Collapse
Affiliation(s)
- Marwan Shinawi
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Daniel J Wegner
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Alexander J Paul
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States of America
| | - William Buchser
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Robert Schmidt
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Jaiprakash Sharma
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Marco Sardiello
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Kathleen Sisco
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Linda Manwaring
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Margaret Reynolds
- Department of Ophthalmology, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Robert Fulton
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Catrina Fronick
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Andrew Shaver
- Illumina Inc, San Diego, CA, United States of America
| | - Tina Y Huang
- Illumina Inc, San Diego, CA, United States of America
| | | | | | | | - Patricia I Dickson
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States of America.
| | - Jennifer A Wambach
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States of America
| |
Collapse
|
36
|
Spinazzi M, Savarese M, Letournel F, Sagath L, Manero F, Guichet A, Hoischen A, Metay C, Gouju J, Udd B. Myotilin gene duplication causing late-onset myotilinopathy. Eur J Neurol 2025; 32:e70029. [PMID: 39757377 PMCID: PMC11702382 DOI: 10.1111/ene.70029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/12/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND myotilinopathy is a very rare inherited muscle disease that belongs to the group of myofibrillar myopathies. These diseases share a common alteration of the sarcomere organization at the level of the Z disk resulting in pathological protein aggregation, autophagic abnormalities, and ultimately muscle degeneration. Most reported cases are due to dominant missense mutations in the MYOT gene, two of which are largely recurrent. METHODS We describe the clinical, radiological, pathological, and molecular analysis including long-read sequencing of a family affected by late-onset dominant proximodistal myopathy and muscle hypertrophy. RESULTS We identified a duplication of the entire MYOT gene as the molecular cause of late-onset-myotilinopapthy with typical clinical and pathological features. CONCLUSIONS This study expands the molecular spectrum of myotilinopathy and highlights the use of long-read sequencing in the diagnosis of genetic neurological diseases caused by duplications and genomic structural variants. Myotilinopathy as well as other myofibrillar and distal myopathies should be considered in the differential diagnosis of patients affected by distal muscle weakness, even when presenting at an old age.
Collapse
Affiliation(s)
- Marco Spinazzi
- Neuromuscular Reference Center, Department of NeurologyCHU d'ANGERS/ INSERM U1083AngersFrance
- Neurobiology and Neuropathology Unit, Department of PathologyCHU d'AngersAngersFrance
| | | | - Franck Letournel
- Neurobiology and Neuropathology Unit, Department of PathologyCHU d'AngersAngersFrance
| | - Lydia Sagath
- Department of Human Genetics, Research Institute for Medical InnovationRadboud University Medical CenterNijmegenNetherlands
| | | | - Agnès Guichet
- Service de Génétique CHU—CNRS 6015/INSERM U1083—Université d'AngersAngersFrance
| | - Alexander Hoischen
- Department of Human Genetics, Research Institute for Medical InnovationRadboud University Medical CenterNijmegenNetherlands
- Department of Internal Medicine, Radboud Expertise Center for Immunodeficiency and Autoinflammation and Radboud Center for Infectious Disease (RCI)Radboud University Medical CenterNijmegenNetherlands
| | - Corinne Metay
- APHP, GH Pitie‐Salpetriere, Molecular and Cellular Cardiogenetics and Myogenetics UF, Chromosomic and Molecular Genetic Center and INSERM UMRS 974Institute of MyologyParisFrance
| | - Julien Gouju
- Neurobiology and Neuropathology Unit, Department of PathologyCHU d'AngersAngersFrance
| | - Bjarne Udd
- Folkhalsan Research CenterHelsinkiFinland
- Tampere Neuromuscular Reference CenterTampere University HospitalTampereFinland
| |
Collapse
|
37
|
Imran M, Agarwal D, Menon K, Scaria V, Sivasubbu S. Novel 327bp Alu element insertion in LDLR exon 17 causes alternative splicing and familial hypercholesterolemia. J Clin Lipidol 2025; 19:114-124. [PMID: 39843294 DOI: 10.1016/j.jacl.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/30/2024] [Accepted: 11/20/2024] [Indexed: 01/24/2025]
Abstract
BACKGROUND Homozygous familial hypercholesterolemia (HoFH) is a severe form of familial hypercholesterolemia (FH) characterized by high low-density lipoprotein cholesterol (LDL-C) levels and increased coronary artery disease risk. This study reports a novel Alu insertion in the LDLR gene in a consanguineous Indian family, causing FH. OBJECTIVE To identify and characterize the mutation causing HoFH in a proband and their family members. METHODS Clinical exome sequencing was conducted on the proband with subsequent bioinformatic analysis of single nucleotide variants, loss-of-function variants, structural variants, and mobile element insertions (MEI). Polymerase chain reaction (PCR) amplification and Sanger sequencing of exon 17 of the LDLR gene were performed to elucidate the sequence and length of the Alu insertion. Additionally, RNA analysis of the proband identified splice site events. RESULTS Bioinformatic analysis revealed a small sequence duplication followed by an Alu element insertion. PCR amplification and Sanger sequencing uncovered a 17 base pair (bp) duplication at the breakpoint, a "T" base insertion, followed by a 309 bp Alu Yb8 insertion. This led to a 70 bp deletion at the beginning of exon 17 due to alternative splicing, resulting in a frameshift and extended protein truncation. The proband and siblings were homozygous for the mutation, while the parents and 2 other family members were heterozygous. CONCLUSION Our study uncovers a novel AluYb8 element insertion in the LDLR gene, highlighting the need for MEI detection in genetic screening for FH. Reanalyzing FH cohorts for MEIs could significantly improve diagnostic accuracy and enhance our understanding of FH genetics.
Collapse
Affiliation(s)
- Mohamed Imran
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India (Dr Imran, Scaria and Sivasubbu); Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India (Dr Imran, Scaria and Sivasubbu)
| | - Divya Agarwal
- Apollo Genomics Institute, Indraprastha Apollo Hospital, New Delhi, India (Dr Agarwal and Menon)
| | - Kriti Menon
- Apollo Genomics Institute, Indraprastha Apollo Hospital, New Delhi, India (Dr Agarwal and Menon)
| | - Vinod Scaria
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India (Dr Imran, Scaria and Sivasubbu); Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India (Dr Imran, Scaria and Sivasubbu).
| | - Sridhar Sivasubbu
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India (Dr Imran, Scaria and Sivasubbu); Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India (Dr Imran, Scaria and Sivasubbu).
| |
Collapse
|
38
|
Wang Y, Ju R, Jiang J, Mao L, Li X, Deng M. Concomitant presence of a novel ARPP21 variant and CNVs in Chinese familial amyotrophic lateral sclerosis-frontotemporal dementia patients. Neurol Sci 2025; 46:195-205. [PMID: 39271636 DOI: 10.1007/s10072-024-07759-3] [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: 07/17/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disorder marked by the degeneration of motor neurons and progressive muscle weakness. Heredity plays an important part in the pathogenesis of ALS. Recently, with the emergence of the oligogenic pathogenic mechanism in ALS and the ongoing discovery of new mutated genes and genomic variants, there is an emerging need for larger-scale and more comprehensive genetic screenings in higher resolution. In this study, we performed whole-genome sequencing (WGS) on 34 familial ALS probands lacking the most common disease-causing mutations to explore the genetic landscape of Chinese ALS patients further. Among them, we identified a novel ARPP21 c.1231G > A (p.Glu411Lys) variant and two copy number variations (CNVs) affecting the PFN1 and RBCK1 genes in a patient with ALS-frontotemporal dementia (FTD). This marks the first report of an ARPP21 variant in Chinese ALS-FTD patients, providing fresh evidence for the association between ARPP21 and ALS. Our findings also underscore the potential role of CNVs in ALS-FTD, suggesting that the cumulative effect of multiple rare variants may contribute to disease onset. Furthermore, compared to the averages in our cohort and the reported Chinese ALS population, this patient displayed a shorter survival time and more rapid disease progression, suggesting the possibility of an oligogenic mechanism in disease pathogenesis. Further research will contribute to a deeper understanding of the rare mutations and their interactions, thus advancing our understanding of the genetic mechanisms underlying ALS and ALS-FTD.
Collapse
Affiliation(s)
- Yiying Wang
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, 100191, China
| | - Runqing Ju
- The Affiliated High School of Peking University Dalton Academy, Beijing, 100190, China
| | - Jingsi Jiang
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, 100191, China
| | - Le Mao
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, 100191, China
| | - Xiaogang Li
- Department of Neurology, Peking University Third Hospital, Beijing, 100191, China
| | - Min Deng
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
39
|
Krause J, Classen C, Dey D, Lausberg E, Kessler L, Eggermann T, Kurth I, Begemann M, Kraft F. CNVizard-a lightweight streamlit application for an interactive analysis of copy number variants. BMC Bioinformatics 2024; 25:376. [PMID: 39690401 DOI: 10.1186/s12859-024-06010-2] [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: 09/10/2024] [Accepted: 12/09/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND Methods to call, analyze and visualize copy number variations (CNVs) from massive parallel sequencing data have been widely adopted in clinical practice and genetic research. To enable a streamlined analysis of CNV data, comprehensive annotations and good visualizations are indispensable. The ability to detect single exon CNVs is another important feature for genetic testing. Nonetheless, most available open-source tools come with limitations in at least one of these areas. One additional drawback is that available tools deliver data in an unstructured and static format which requires subsequent visualization and formatting efforts. RESULTS Here we present CNVizard, an interactive Streamlit app allowing a comprehensive visualization of CNVkit data. Furthermore, combining CNVizard with the CNVand pipeline allows the annotation and visualization of CNV or SV VCF files from any CNV caller. CONCLUSION CNVizard, in combination with CNVand, enables the comprehensive and streamlined analysis of short- and long-read sequencing data and provide an intuitive webapp-like experience enabling an interactive visualization of CNV data.
Collapse
Affiliation(s)
- Jeremias Krause
- Medical Faculty, Institute for Human Genetics and Genomic Medicine, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, North-Rhine-Westphalia, Germany.
| | - Carlos Classen
- Medical Faculty, Institute for Human Genetics and Genomic Medicine, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, North-Rhine-Westphalia, Germany
| | - Daniela Dey
- Medical Faculty, Institute for Human Genetics and Genomic Medicine, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, North-Rhine-Westphalia, Germany
| | - Eva Lausberg
- Medical Faculty, Institute for Human Genetics and Genomic Medicine, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, North-Rhine-Westphalia, Germany
| | - Luise Kessler
- Medical Faculty, Institute for Human Genetics and Genomic Medicine, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, North-Rhine-Westphalia, Germany
| | - Thomas Eggermann
- Medical Faculty, Institute for Human Genetics and Genomic Medicine, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, North-Rhine-Westphalia, Germany
| | - Ingo Kurth
- Medical Faculty, Institute for Human Genetics and Genomic Medicine, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, North-Rhine-Westphalia, Germany
| | - Matthias Begemann
- Medical Faculty, Institute for Human Genetics and Genomic Medicine, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, North-Rhine-Westphalia, Germany
| | - Florian Kraft
- Medical Faculty, Institute for Human Genetics and Genomic Medicine, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, North-Rhine-Westphalia, Germany
| |
Collapse
|
40
|
Hsu JS, Wu DC, Shih SH, Liu JF, Tsai YC, Lee TL, Chen WA, Tseng YH, Lo YC, Lin HY, Chen YC, Chen JY, Chou TH, Chang DTH, Su MW, Guo WH, Mao HH, Chen CY, Chen PL. Complete genomic profiles of 1496 Taiwanese reveal curated medical insights. J Adv Res 2024; 66:197-207. [PMID: 38159844 PMCID: PMC11675050 DOI: 10.1016/j.jare.2023.12.018] [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: 08/14/2023] [Revised: 12/03/2023] [Accepted: 12/27/2023] [Indexed: 01/03/2024] Open
Abstract
INTRODUCTION The population of Taiwan has a long history of ethno-cultural evolution. The Taiwanese population was isolated from other large populations such as the European, Han Chinese, and Japanese population. The Taiwan Biobank (TWB) project has built a nationwide database, particularly for personal whole-genome sequence (WGS) to facilitate basic and clinical collaboration nationally and internationally, making it one of the most valuable public datasets of the East Asian population. OBJECTIVES This study provides comprehensive medical genomic findings from TWB WGS data, for better characterization of disease susceptibility and the choice of ideal treatment regimens in Taiwanese population. METHODS We reanalyzed 1496 WGS using a PrecisionFDA Truth challenge winner method Sentieon DNAscope. Single nucleotide variants (SNV) and small insertions/deletions (INDEL) were benchmarked. We also analyzed pharmacogenomic (PGx) drug-associated alleles, and copy number variants (CNV). Multiple practicing clinicians reviewed and curated the clinically significant variants. Variant annotations can be browsed at TaiwanGenomes (https://genomes.tw). RESULTS We found that each participant had an average of 6,870.7 globally novel variants and 75.3% (831/1103) of the participants harbored at least one PharmGKB-selected high evidence level human leukocyte antigen (HLA) risk allele. 54 PharmGKB-reported high-level instances of evidence of Cytochrome P450 variant-drug pairs, with a population frequency of over 13.2%. We also identified 23 variants in the ACMG secondary finding V3 gene list from 25 participants, suggesting that 1.67% (25/1496) of the population is harboring at least one medical actionable variant. Our carrier status analyses suggest that one in 25 couples (3.94%) would risk having offspring with at least one pathogenic variant, which is in line with rates found in Japan and Singapore. For pathogenic CNV, we detected 6.88% and 2.02% carrier rates for alpha thalassemia and spinal muscular atrophy, respectively. CONCLUSION Our study highlights the overall medical insights of a complete Taiwanese genomic profile.
Collapse
Affiliation(s)
- Jacob Shujui Hsu
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei 100025, Taiwan; Institute of Molecular Medicine, National Taiwan University College of Medicine, Taipei 100233, Taiwan
| | - Dung-Chi Wu
- Genome and Systems Biology Degree Program, National Taiwan University and Academia Sinica, Taipei 10617, Taiwan
| | - Shang-Hung Shih
- Genome and Systems Biology Degree Program, National Taiwan University and Academia Sinica, Taipei 10617, Taiwan
| | - Jen-Feng Liu
- Institute of Molecular Medicine, National Taiwan University College of Medicine, Taipei 100233, Taiwan
| | - Ya-Chen Tsai
- Department of Biomechatronics Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Tung-Lin Lee
- Department of Medical Genetics, National Taiwan University Hospital, Taipei 100226, Taiwan
| | - Wei-An Chen
- Department of Medical Genetics, National Taiwan University Hospital, Taipei 100226, Taiwan
| | - Yi-Hsuan Tseng
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei 100025, Taiwan
| | - Yi-Chung Lo
- Department of Electrical Engineering, National Cheng-Kung University, Tainan 701401, Taiwan
| | - Hong-Ye Lin
- Department of Biomechatronics Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Yi-Chieh Chen
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei 100025, Taiwan
| | - Jing-Yi Chen
- Department of Electrical Engineering, National Cheng-Kung University, Tainan 701401, Taiwan
| | - Ting-Hsuan Chou
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei 100025, Taiwan
| | - Darby Tien-Hao Chang
- Department of Electrical Engineering, National Cheng-Kung University, Tainan 701401, Taiwan; Digital Technology Division, SinoPac Holdings, Taiwan
| | - Ming Wei Su
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115201, Taiwan
| | - Wei-Hong Guo
- Department of Biomechatronics Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Hsin-Hsiang Mao
- Department of Biomechatronics Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Chien-Yu Chen
- Genome and Systems Biology Degree Program, National Taiwan University and Academia Sinica, Taipei 10617, Taiwan; Department of Biomechatronics Engineering, National Taiwan University, Taipei 10617, Taiwan.
| | - Pei-Lung Chen
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei 100025, Taiwan; Institute of Molecular Medicine, National Taiwan University College of Medicine, Taipei 100233, Taiwan; Genome and Systems Biology Degree Program, National Taiwan University and Academia Sinica, Taipei 10617, Taiwan; Department of Medical Genetics, National Taiwan University Hospital, Taipei 100226, Taiwan; Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei 100233, Taiwan.
| |
Collapse
|
41
|
Reshadmanesh A, Dehdahsi S, Ahangari F, Kahrizi K, Kariminejad A, Mahdavi SS, Talebi S, Najmabadi H. First Case of Macrocephaly, Dysmorphic Facies, and Psychomotor Retardation Harboring Co-inherited Variants in HERC1 and PMP22 Genes from Iran: Two Novel Variants. ARCHIVES OF IRANIAN MEDICINE 2024; 27:700-706. [PMID: 39891458 PMCID: PMC11786211 DOI: 10.34172/aim.31593] [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: 07/01/2024] [Accepted: 08/21/2024] [Indexed: 02/03/2025]
Abstract
Here, we report a case with concomitant variants: a novel homozygous HERC1 gene variant and a novel heterozygous PMP22 duplication. The 2-year-old male presented with seizures, developmental delay, macrocephaly, hypotonia, unilateral hypertrophy, thoracic scoliosis, normal brain MRI, and elevated homocysteine level which normalized after treatment. Whole exome sequencing (WES) revealed a co-occurrence of a homozygous novel likely pathogenic variant in the HERC1 gene (NM_003922.3:c.1280dup (p.ILe469Aspfs*33) and a novel heterozygous large duplication of exon 1-5 in the PMP22 gene, which has not been reported previously. The case underscores the challenges in understanding genotype-phenotype correlations and suggests a potential interplay between these genetic variants in shaping the current and future clinical phenotype of the patient. In the case of genetic diseases, this event may have important implications on family members' counseling, and concomitant variants in Charcot-Marie-Tooth (CMT) families should be considered when significant intra-familial clinical heterogeneity is observed.
Collapse
Affiliation(s)
- Azadeh Reshadmanesh
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Shima Dehdahsi
- Kariminejad-Najmabadi Pathology & Genetics Center, Tehran, Iran
| | | | - Kimia Kahrizi
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | | | | | - Saeed Talebi
- Department of Medical Genetics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Najmabadi
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
- Kariminejad-Najmabadi Pathology & Genetics Center, Tehran, Iran
| |
Collapse
|
42
|
Pal S, Kulshrestha S, Garg N, Gupta D, Gupta ND, Puri RD. Two-Compound Heterozygous Deletions Affecting TUBGCP6 in a Patient with Microcephaly and Ocular Abnormalities and in an Unborn Sibling with Abnormal Sulcation. Mol Syndromol 2024; 15:503-516. [PMID: 39634241 PMCID: PMC11614437 DOI: 10.1159/000539099] [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: 02/21/2024] [Accepted: 04/24/2024] [Indexed: 12/07/2024] Open
Abstract
Introduction TUBGCP6-related disorder is a known cause of autosomal recessive microcephaly and chorioretinopathy, which was originally recognized as a new syndrome based on unique ocular findings on a phenotypic overlap of microcephalic primordial short stature. Since the elucidation of its molecular mechanism, limited families have been published in literature and the disorder remains rare worldwide. Case Presentation We present the first Indian family with an affected child and sibling fetus with microcephaly, dysmorphism, and agyria/pachygyria complex on brain imaging in both and short stature, intellectual disability, and visual impairment in proband. As for many patients with long diagnostic odysseys, this child also underwent multiple genomic tests. Genome sequencing through the Indian Undiagnosed Disease Program (I-UDP) confirmed the diagnosis in both proband and sibling fetus. Compound heterozygous variants were identified in TUBGCP6 including an eleven base pair deletion (inherited from father) and 405 base pair large deletion (inherited from mother). Reverse phenotyping to confirm the ocular phenotype in proband confirmed TUBGCP6-related microcephaly and chorioretinopathy. We report third trimester microcephaly with ventriculomegaly and abnormal sulcation as part of the antenatal presentation for this condition. Conclusion This case represents an Indian family with a seemingly obvious clinical diagnosis compounded by a long diagnostic odyssey and the first ever structural variant to be identified via whole genome sequencing in TUBGCP6 in trans with an indel variant.
Collapse
Affiliation(s)
- Swasti Pal
- Institute of Medical Genetics and Genomics Ganga Ram Institute of Post Graduate Medical Education and Research Sir Ganga Ram Hospital, New Delhi, India
| | - Samarth Kulshrestha
- Institute of Medical Genetics and Genomics Ganga Ram Institute of Post Graduate Medical Education and Research Sir Ganga Ram Hospital, New Delhi, India
| | - Neha Garg
- Institute of Medical Genetics and Genomics Ganga Ram Institute of Post Graduate Medical Education and Research Sir Ganga Ram Hospital, New Delhi, India
| | - Deepti Gupta
- Institute of Medical Genetics and Genomics Ganga Ram Institute of Post Graduate Medical Education and Research Sir Ganga Ram Hospital, New Delhi, India
| | - Nandita Dimri Gupta
- Department of Fetal Medicine, Ganga Ram Institute of Post Graduate Medical Education and Research, Sir Ganga Ram Hospital, New Delhi, India
| | - Ratna Dua Puri
- Institute of Medical Genetics and Genomics Ganga Ram Institute of Post Graduate Medical Education and Research Sir Ganga Ram Hospital, New Delhi, India
| |
Collapse
|
43
|
Snelling T, Garnotel LO, Jeru I, Tusseau M, Cuisset L, Perlat A, Minard G, Benquey T, Maucourant Y, Wood NT, Cohen P, Ziegler A. Discovery and functional analysis of a novel ALPK1 variant in ROSAH syndrome. Open Biol 2024; 14:240260. [PMID: 39626775 PMCID: PMC11614533 DOI: 10.1098/rsob.240260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/29/2024] [Accepted: 10/29/2024] [Indexed: 12/06/2024] Open
Abstract
Retinal dystrophy, optic nerve oedema, splenomegaly, anhidrosis and migraine headache (ROSAH) syndrome is an autosomal dominant disorder and to date is known to be caused by either the Thr237Met or Tyr254Cys variant in the protein kinase ALPK1. Here, we identify a family in which ROSAH syndrome is caused by a novel variant in which Ser277 is changed to Phe. All six patients examined display ocular inflammation and optic nerve elevation, four have retinal degeneration and four are registered blind. In contrast to wild-type ALPK1, which is activated specifically by bacterial ADP-heptose, ALPK1[Ser277Phe] is also activated by the human metabolites UDP-mannose and ADP-ribose and more strongly than the most frequent ROSAH-causing variant (ALPK1[Thr237Met]) but, unlike ALPK1[Thr237Met], ALPK1[Ser277Phe] is also activated by GDP-mannose. These observations can explain why ALPK1 variants causing ROSAH syndrome display constitutive activity in human cells. The side chains of Ser277 and Tyr254 interact in the crystal structure of ALPK1, but mutational analysis established that it is not the loss of this hydrogen bond between Ser277 and Tyr254 that alters the specificity of the ADP-heptose-binding pocket in the Ser277Phe and Tyr254Cys variants. The characterization of ALPK1 variants that cause ROSAH syndrome suggests ways in which drugs that selectively inhibit these disease-causing variants may be developed.
Collapse
Affiliation(s)
- Tom Snelling
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Leo O. Garnotel
- Department of Ophthalmology, University Hospital of Reims, Reims, France
| | - Isabelle Jeru
- Department of Medical Genetics, Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France
| | - Maud Tusseau
- Centre International de Recherche en Infectiologie, Inserm, U1111, University Claude Bernard, Lyon 1, UMR5308, ENS de Lyon, Lyon, France
- Hospices Civils de Lyon, Department of Medical Genetics, University Hospital of Lyon, Lyon, France
| | - Laurence Cuisset
- Université Paris Cité, Service de Médecine Génomique des Maladies de Système et D'Organe, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Antoinette Perlat
- Department of Internal Medicine and Clinical Immunology, Pontchaillou Hospital, Rennes, France
| | - Geoffrey Minard
- Department of Internal Medicine and Clinical Immunology, University Hospital of Reims, Reims, France
| | | | - Yann Maucourant
- Department of Ophthalmology, Pontchaillou Hospital, Rennes, France
| | - Nicola T. Wood
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Philip Cohen
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Alban Ziegler
- Department of Medical Genetics, University Hospital of Reims, Reims, France
- Department of Medical Genetics, University Hospital of Toulouse, Toulouse, France
| |
Collapse
|
44
|
Buianova AA, Bazanova MV, Belova VA, Ilyina GA, Samitova AF, Shmitko AO, Balakina AV, Pavlova AS, Suchalko ON, Korostin DO, Machalov AS, Daikhes NA, Rebrikov DV. Heterogeneous Group of Genetically Determined Auditory Neuropathy Spectrum Disorders. Int J Mol Sci 2024; 25:12554. [PMID: 39684270 DOI: 10.3390/ijms252312554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/18/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
Auditory neuropathy spectrum disorder (ANSD) is often missed by standard hearing tests, accounting for up to 10% of hearing impairments (HI) and commonly linked to variants in 23 genes. We assessed 122 children with HI, including 102 with sensorineural hearing loss (SNHL) and 20 with ANSD. SNHL patients were genotyped for common GJB2 variants using qPCR, while ANSD patients underwent whole exome sequencing, with variants analyzed across 249 genes. Homozygous GJB2 variants were found in 54.9% of SNHL patients. In 60% of ANSD patients, variants were detected in OTOF (25%), CDH23, TMC1, COL11A1, PRPS1, TWNK, and HOMER2 genes, including eight novel variants. Transient evoked otoacoustic emissions testing revealed differences at 4000 Hz (p = 0.0084) between the ANSD and SNHL groups. The auditory steady-state response (ASSR) test showed significant differences at 500 Hz (p = 2.69 × 10-4) and 1000 Hz (p = 0.0255) compared to pure-tone audiometry (PTA) in ANSD patients. Our questionnaire shows that the parents of children with SNHL often report an improved quality of life with hearing aids or cochlear implants, while parents of children with ANSD frequently experience uncertainty about outcomes (p = 0.0026), leading to lower satisfaction.
Collapse
Affiliation(s)
- Anastasiia A Buianova
- The Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117513 Moscow, Russia
| | - Marina V Bazanova
- FSBI 'The National Medical Research Center for Otorhinolaryngology of the Federal Medico-Biological Agency of Russia', 123182 Moscow, Russia
| | - Vera A Belova
- The Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117513 Moscow, Russia
| | - Galit A Ilyina
- The Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117513 Moscow, Russia
| | - Alina F Samitova
- The Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117513 Moscow, Russia
| | - Anna O Shmitko
- The Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117513 Moscow, Russia
| | - Anna V Balakina
- FSBI 'The National Medical Research Center for Otorhinolaryngology of the Federal Medico-Biological Agency of Russia', 123182 Moscow, Russia
| | - Anna S Pavlova
- The Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117513 Moscow, Russia
| | - Oleg N Suchalko
- The Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117513 Moscow, Russia
| | - Dmitriy O Korostin
- The Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117513 Moscow, Russia
| | - Anton S Machalov
- FSBI 'The National Medical Research Center for Otorhinolaryngology of the Federal Medico-Biological Agency of Russia', 123182 Moscow, Russia
- Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Nikolai A Daikhes
- FSBI 'The National Medical Research Center for Otorhinolaryngology of the Federal Medico-Biological Agency of Russia', 123182 Moscow, Russia
- Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Denis V Rebrikov
- The Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117513 Moscow, Russia
- FSBI 'National Medical Research Center for Obstetrics, Gynecology, and Perinatology Named After Academician V.I. Kulakov', 117198 Moscow, Russia
| |
Collapse
|
45
|
Tan JHJ, Li Z, Porta MG, Rajaby R, Lim WK, Tan YA, Jimenez RT, Teo R, Hebrard M, Ow JL, Ang S, Jeyakani J, Chong YS, Lim TH, Goh LL, Tham YC, Leong KP, Chin CWL, Davila S, Karnani N, Cheng CY, Chambers J, Tai ES, Liu J, Sim X, Sung WK, Prabhakar S, Tan P, Bertin N. A Catalogue of Structural Variation across Ancestrally Diverse Asian Genomes. Nat Commun 2024; 15:9507. [PMID: 39496583 PMCID: PMC11535549 DOI: 10.1038/s41467-024-53620-8] [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: 09/22/2023] [Accepted: 10/14/2024] [Indexed: 11/06/2024] Open
Abstract
Structural variants (SVs) are significant contributors to inter-individual genetic variation associated with traits and diseases. Current SV studies using whole-genome sequencing (WGS) have a largely Eurocentric composition, with little known about SV diversity in other ancestries, particularly from Asia. Here, we present a WGS catalogue of 73,035 SVs from 8392 Singaporeans of East Asian, Southeast Asian and South Asian ancestries, of which ~65% (47,770 SVs) are novel. We show that Asian populations can be stratified by their global SV patterns and identified 42,239 novel SVs that are specific to Asian populations. 52% of these novel SVs are restricted to one of the three major ancestry groups studied (Indian, Chinese or Malay). We uncovered SVs affecting major clinically actionable loci. Lastly, by identifying SVs in linkage disequilibrium with single-nucleotide variants, we demonstrate the utility of our SV catalogue in the fine-mapping of Asian GWAS variants and identification of potential causative variants. These results augment our knowledge of structural variation across human populations, thereby reducing current ancestry biases in global references of genetic variation afflicting equity, diversity and inclusion in genetic research.
Collapse
Affiliation(s)
- Joanna Hui Juan Tan
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Zhihui Li
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Mar Gonzalez Porta
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
- Nalagenetics, Singapore, Singapore
| | - Ramesh Rajaby
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
- Human Genome Center, University of Tokyo, Bunkyō, Japan
| | - Weng Khong Lim
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
- SingHealth Duke-NUS Institute of Precision Medicine, Singapore Health Services, Duke-NUS Medical School, Singapore, Singapore
- SingHealth Duke-NUS Genomic Medicine Centre, Duke-NUS Medical School, Singapore, Singapore
| | - Ye An Tan
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
| | - Rodrigo Toro Jimenez
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Renyi Teo
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Maxime Hebrard
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Jack Ling Ow
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Shimin Ang
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Justin Jeyakani
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Yap Seng Chong
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Tock Han Lim
- NHG Eye Institute, Tan Tock Seng Hospital, National Healthcare Group, Singapore, Singapore
| | - Liuh Ling Goh
- Personalised Medicine Service, Tan Tock Seng Hospital, Singapore, Singapore
| | - Yih Chung Tham
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Centre for Innovation and Precision Eye Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Khai Pang Leong
- Personalised Medicine Service, Tan Tock Seng Hospital, Singapore, Singapore
| | - Calvin Woon Loong Chin
- Department of Cardiology, National Heart Centre Singapore, Singapore, Singapore
- Cardiovascular ACP, Duke-NUS Medical School, Singapore, Singapore
| | - Sonia Davila
- SingHealth Duke-NUS Genomic Medicine Centre, Duke-NUS Medical School, Singapore, Singapore
- SingHealth Duke-NUS Institute of Precision medicine, Singapore Health Services, Singapore, Singapore
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
- Translational Medicine, Sidra Medicine, Ar-Rayyan, Qatar
| | - Neerja Karnani
- Human Development, Singapore Institute for Clinical Sciences, Singapore, Singapore
- Clinical Data Engagement, Bioinformatics Institute, Agency for Science, Technology and Research, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ching-Yu Cheng
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Centre for Innovation and Precision Eye Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - John Chambers
- Population and Global Health, Nanyang Technological University, Lee Kong Chian School of Medicine, Singapore, Singapore
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Precision Health Research, Singapore, Singapore
| | - E Shyong Tai
- Duke-NUS Medical School, Singapore, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
- Precision Health Research, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jianjun Liu
- Laboratory of Human Genomics, Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xueling Sim
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
| | - Wing Kin Sung
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
- Hong Kong Genome Institute, Hong Kong, Hong Kong
- Department of Chemical Pathology, Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Shyam Prabhakar
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.
| | - Patrick Tan
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore.
- Duke-NUS Medical School, Singapore, Singapore.
- SingHealth Duke-NUS Institute of Precision Medicine, Singapore Health Services, Duke-NUS Medical School, Singapore, Singapore.
- Precision Health Research, Singapore, Singapore.
| | - Nicolas Bertin
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore.
| |
Collapse
|
46
|
Qiu JJ, Chang XY, Zhang N, Guo LP, Wang S, Gu WY, Yin YM, Shi ZW, Hua KQ. Genetic variation and molecular profiling of congenital malformations of the female genital tract based on whole-genome sequencing. World J Pediatr 2024; 20:1179-1195. [PMID: 39251565 DOI: 10.1007/s12519-024-00839-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 08/07/2024] [Indexed: 09/11/2024]
Abstract
BACKGROUND Congenital malformations of the female genital tract (CM-FGT) are characterized by abnormal development of the fallopian tubes, uterus, and vagina, often accompanied by malformations in the urinary system, bones and hearing. However, no definitive pathogenic genes and molecular genetic causes have been identified. METHODS We present the largest whole-genome sequencing study of CM-FGT to date, analyzing 590 individuals in China: 95 patients, 442 case-controls, and 53 familial controls. RESULTS Among the patients, 5.3% carried known CM-FGT-related variants. Pedigree and case-control analyses in two dimensions of coding and non-coding regulatory regions revealed seven novel de novo copy number variations, 12 rare single-nucleotide variations, and 10 rare 3' untranslated region (UTR) mutations in genes related to CM-FGT, particularly highlighting ASH1L as a pathogenic gene. Single-cell sequencing data showed that the majority of CM-FGT-related risk genes are spatiotemporally specifically expressed early in uterus development. CONCLUSIONS In conclusion, this study identified novel variants related to CM-FGT, particularly highlighting ASH1L as a pathogenic gene. The findings provide insights into the genetic variants underlying CM-FGT, with single-cell sequencing data revealing spatiotemporal specific expression patterns of key risk genes early in uterine development. This study significantly advances the understanding of CM-FGT etiology and genetic landscape, offering new opportunities for prenatal screening.
Collapse
Affiliation(s)
- Jun-Jun Qiu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, 413 Zhaozhou Road, Shanghai, 200011, China
| | - Xing-Yu Chang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, 413 Zhaozhou Road, Shanghai, 200011, China
| | - Ning Zhang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, 413 Zhaozhou Road, Shanghai, 200011, China
| | - Luo-Pei Guo
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, 413 Zhaozhou Road, Shanghai, 200011, China
| | - Shuai Wang
- Data and Analysis Center for Genetic Diseases, Chigene Translational Medicine Research Center, Beijing, 100032, China
| | - Wei-Yue Gu
- Data and Analysis Center for Genetic Diseases, Chigene Translational Medicine Research Center, Beijing, 100032, China
| | - Yi-Meng Yin
- Translational Research Institute of Brain and Brain-Like Intelligence, School of Medicine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, 200434, China.
| | - Zhi-Wen Shi
- Data and Analysis Center for Genetic Diseases, Chigene Translational Medicine Research Center, Beijing, 100032, China.
| | - Ke-Qin Hua
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China.
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, 413 Zhaozhou Road, Shanghai, 200011, China.
| |
Collapse
|
47
|
Chang S, Liu JJ, Zhao Y, Pang T, Zheng X, Song Z, Zhang A, Gao X, Luo L, Guo Y, Liu J, Yang L, Lu L. Whole-genome sequencing identifies novel genes for autism in Chinese trios. SCIENCE CHINA. LIFE SCIENCES 2024; 67:2368-2381. [PMID: 39126614 DOI: 10.1007/s11427-023-2564-8] [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: 11/30/2023] [Accepted: 03/16/2024] [Indexed: 08/12/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder with high genetic heritability but heterogeneity. Fully understanding its genetics requires whole-genome sequencing (WGS), but the ASD studies utilizing WGS data in Chinese population are limited. In this study, we present a WGS study for 334 individuals, including 112 ASD patients and their non-ASD parents. We identified 146 de novo variants in coding regions in 85 cases and 60 inherited variants in coding regions. By integrating these variants with an association model, we identified 33 potential risk genes (P<0.001) enriched in neuron and regulation related biological process. Besides the well-known ASD genes (SCN2A, NF1, SHANK3, CHD8 etc.), several high confidence genes were highlighted by a series of functional analyses, including CTNND1, DGKZ, LRP1, DDN, ZNF483, NR4A2, SMAD6, INTS1, and MRPL12, with more supported evidence from GO enrichment, expression and network analysis. We also integrated RNA-seq data to analyze the effect of the variants on the gene expression and found 12 genes in the individuals with the related variants had relatively biased expression. We further presented the clinical phenotypes of the proband carrying the risk genes in both our samples and Caucasian samples to show the effect of the risk genes on phenotype. Regarding variants in non-coding regions, a total of 74 de novo variants and 30 inherited variants were predicted as pathogenic with high confidence, which were mapped to specific genes or regulatory features. The number of de novo variants found in patient was significantly associated with the parents' ages at the birth of the child, and gender with trend. We also identified small de novo structural variants in ASD trios. The results in this study provided important evidence for understanding the genetic mechanism of ASD.
Collapse
Affiliation(s)
- Suhua Chang
- Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University Sixth Hospital, Beijing, 100191, China
- Chinese Academy of Medical Sciences Research Unit (No.2018RU006), Peking University, Beijing, 100191, China
| | - Jia Jia Liu
- Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University Sixth Hospital, Beijing, 100191, China
- School of Nursing, Peking University, Beijing, 100191, China
| | - Yilu Zhao
- Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University Sixth Hospital, Beijing, 100191, China
| | - Tao Pang
- Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University Sixth Hospital, Beijing, 100191, China
| | - Xiangyu Zheng
- Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University Sixth Hospital, Beijing, 100191, China
| | | | - Anyi Zhang
- Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University Sixth Hospital, Beijing, 100191, China
| | - Xuping Gao
- Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University Sixth Hospital, Beijing, 100191, China
| | - Lingxue Luo
- Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University Sixth Hospital, Beijing, 100191, China
| | - Yanqing Guo
- Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University Sixth Hospital, Beijing, 100191, China.
| | - Jing Liu
- Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University Sixth Hospital, Beijing, 100191, China.
| | - Li Yang
- Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University Sixth Hospital, Beijing, 100191, China.
| | - Lin Lu
- Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University Sixth Hospital, Beijing, 100191, China.
- Chinese Academy of Medical Sciences Research Unit (No.2018RU006), Peking University, Beijing, 100191, China.
- National Institute on Drug Dependence, Peking University, Beijing, 100191, China.
| |
Collapse
|
48
|
Demidov G, Yaldiz B, Garcia-Pelaez J, de Boer E, Schuermans N, Van de Vondel L, Paramonov I, Johansson LF, Musacchia F, Benetti E, Bullich G, Sablauskas K, Beltran S, Gilissen C, Hoischen A, Ossowski S, de Voer R, Lohmann K, Oliveira C, Topf A, Vissers LELM, Laurie S. Comprehensive reanalysis for CNVs in ES data from unsolved rare disease cases results in new diagnoses. NPJ Genom Med 2024; 9:49. [PMID: 39461972 PMCID: PMC11513043 DOI: 10.1038/s41525-024-00436-6] [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: 02/14/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
We report the results of a comprehensive copy number variant (CNV) reanalysis of 9171 exome sequencing datasets from 5757 families affected by a rare disease (RD). The data reanalysed was extremely heterogeneous, having been generated using 28 different enrichment kits by 42 different research groups across Europe partnering in the Solve-RD project. Each research group had previously undertaken their own analysis of the data but failed to identify disease-causing variants. We applied three CNV calling algorithms to maximise sensitivity, and rare CNVs overlapping genes of interest, provided by four partner European Reference Networks, were taken forward for interpretation by clinical experts. This reanalysis has resulted in a molecular diagnosis being provided to 51 families in this sample, with ClinCNV performing the best of the three algorithms. We also identified partially explanatory pathogenic CNVs in a further 34 individuals. This work illustrates the value of reanalysing ES cold cases for CNVs.
Collapse
Affiliation(s)
- German Demidov
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany.
- Institute for Bioinformatics and Medical Informatics (IBMI), University of Tübingen, Tübingen, Germany.
| | - Burcu Yaldiz
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - José Garcia-Pelaez
- i3S - Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
| | - Elke de Boer
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Nika Schuermans
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Liedewei Van de Vondel
- Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- Laboratory of Neuromuscular Pathology, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Ida Paramonov
- Centro Nacional de Análisis Genómico (CNAG), C/Baldiri Reixac 4, 08028, Barcelona, Spain
- Universitat de Barcelona (UB), Barcelona, Spain
| | - Lennart F Johansson
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Francesco Musacchia
- Center for Human Technologies, Italian Institute of Technology (IIT), Genova, Italy
- Telethon Institute for Genetics and Medicine, 80078, Pozzuoli (Napoli), Italy
| | - Elisa Benetti
- Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, 53100, Siena, Italy
| | - Gemma Bullich
- Centro Nacional de Análisis Genómico (CNAG), C/Baldiri Reixac 4, 08028, Barcelona, Spain
- Universitat de Barcelona (UB), Barcelona, Spain
| | - Karolis Sablauskas
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Institute of Data Science and Digital Technologies, Vilnius University, Vilnius, Lithuania
| | - Sergi Beltran
- Centro Nacional de Análisis Genómico (CNAG), C/Baldiri Reixac 4, 08028, Barcelona, Spain
- Universitat de Barcelona (UB), Barcelona, Spain
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona (UB), Barcelona, Spain
| | - Christian Gilissen
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Alexander Hoischen
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Stephan Ossowski
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
- Institute for Bioinformatics and Medical Informatics (IBMI), University of Tübingen, Tübingen, Germany
| | - Richarda de Voer
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Katja Lohmann
- Institute of Neurogenetics, University of Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany
| | - Carla Oliveira
- i3S - Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
| | - Ana Topf
- John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Lisenka E L M Vissers
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Steven Laurie
- Centro Nacional de Análisis Genómico (CNAG), C/Baldiri Reixac 4, 08028, Barcelona, Spain.
- Universitat de Barcelona (UB), Barcelona, Spain.
| |
Collapse
|
49
|
Sina M, Zarinfam S, Giliani SC, Poliani PL, Majidzadeh-A K. Germline testing of Iranian families suspected of Lynch syndrome: molecular characterization and current surveillance of families with pathogenic variants in MSH2, MSH6, and PMS2. Eur J Cancer Prev 2024:00008469-990000000-00180. [PMID: 39436407 DOI: 10.1097/cej.0000000000000916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
Lynch syndrome accounts for 3-5% of all colorectal and endometrial cancer cases, and suboptimal management of Lynch syndrome in the Middle East resulted in the underdiagnosis of mutation carriers. Probands from 24 unrelated Iranian families with a history of cancer(s) suggestive of Lynch syndrome underwent microsatellite instability analysis or immunohistochemistry, multigene panel testing, copy number variation detection, or multiplex ligation-dependent probe amplification. Pathogenic variants were identified in five patients (21%), including three in MSH2, one in MSH6, and one in PMS2. Microsatellite instability analysis showed the lengths of the CAT25 marker in tumor and normal samples were 149 and 148 bp, respectively. Among 21 family members with Lynch syndrome in the MSH2 gene, identified from the three families who previously underwent cascade screening, colorectal and endometrial cancers were the most frequent. While 66% of patients had insurance that included coverage for mutation carrier screening, only one insurance provider extended coverage for next-generation sequencing. Special attention to probands and telematic management of at-risk relatives to organize blood sample collection at their convenience enhanced cascade testing 20-fold per proband. In conclusion, the age of onset and segregation analysis indicated that PMS1 may not be a cancer susceptibility gene, and the tumor spectrum in MSH2 pathogenic carriers is similar to Western countries. Collecting blood samples at patients' convenience is a possible strategy to reduce the cost of identifying Lynch syndrome through cascade testing. The genetic analysis of patients for inherited cancers would optimize the current management of Lynch syndrome in Iran by omitting noncarriers from surveillance programs.
Collapse
Affiliation(s)
- Mohammad Sina
- A. Nocivelli Institute for Molecular Medicine, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Genetics Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Shiva Zarinfam
- Genetics Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Silvia Clara Giliani
- A. Nocivelli Institute for Molecular Medicine, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Section of Cytogenetics and Medical Genetics, Laboratory Department, ASST Spedali Civili
| | - Pietro Luigi Poliani
- Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Keivan Majidzadeh-A
- Genetics Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| |
Collapse
|
50
|
Rodilla C, Núñez-Moreno G, Benitez Y, Romero R, Fernández-Caballero L, Mínguez P, Corton M, Ayuso C. Cas9-targeted-based long-read sequencing for genetic screening of RPE65 locus. Front Genet 2024; 15:1439153. [PMID: 39469149 PMCID: PMC11513366 DOI: 10.3389/fgene.2024.1439153] [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: 05/27/2024] [Accepted: 08/06/2024] [Indexed: 10/30/2024] Open
Abstract
Introduction Long-read sequencing (LRS) enables accurate structural variant detection and variant phasing. When a molecular diagnosis is suspected, target enrichment can reduce the cost and duration of sequencing. Methods LRS was conducted in five inherited retinal dystrophy (IRD) patients harboring a monoallelic variant in RPE65 that remained uncharacterized after clinical exome sequencing (CES). CRISPR-Cas9 guide RNA probes were designed to target a 31 kb region, including the entire RPE65 locus. The DNA was sequenced on a MinION platform. Short-read ×30 whole-genome sequencing (WGS) was performed for five patients to validate nanopore results. Results The nanopore sequencing process yielded a median of 271 reads within the targeted region, with a mean depth of 109 and a median read size of 8 kb. All variants identified by CES have been detected using this approach, and no additional RPE65 gene causative variants were found. Nanopore variant detection demonstrated performance akin to short-read WGS at similar coverage levels, although exhibiting increased false positive calls at lower coverage. Discussion In this study, we explore the advantages of using a targeted approach together with long-read sequencing to identify variants associated with IRD. The results underscore the utility of targeted long reads for characterizing patients affected by rare diseases when first-tier diagnostic tests are non-conclusive.
Collapse
Affiliation(s)
- Cristina Rodilla
- Department of Genetics and Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Gonzalo Núñez-Moreno
- Department of Genetics and Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Bioinformatics Unit, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
| | - Yolanda Benitez
- Department of Genetics and Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- Bioinformatics Unit, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
| | - Raquel Romero
- Department of Genetics and Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Bioinformatics Unit, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
| | - Lidia Fernández-Caballero
- Department of Genetics and Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Pablo Mínguez
- Department of Genetics and Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Bioinformatics Unit, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
| | - Marta Corton
- Department of Genetics and Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Carmen Ayuso
- Department of Genetics and Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|