1
|
Yin Law JH, Au CH, Leung AWS, Leung HCM, Wong EYL, Ip BBK, Ho DNY, Ma SY, Chan HMH, Chiu EKW, Chim JCS, Liang RHS, Wan TSK, Ma ESK. A multi-modal molecular characterization of the Philadelphia translocation featuring long read sequencing. Gene 2025; 950:149370. [PMID: 40024301 DOI: 10.1016/j.gene.2025.149370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025]
Abstract
OBJECTIVE Clinically significant structural variation (SV), notably chromosomal translocation, results in the formation of fusion genes that drive leukaemogenesis. Detection of SVs is vital in clinical diagnosis, prognosis and therapy of haematological malignancies. Current methods for SV identification are low in sensitivity for cryptic cases and time-consuming for complex cases. This study investigated the feasibility of long read sequencing as an approach for SV detection and precise breakpoint characterization. METHODS Six archival samples, including 4 bone marrow blood samples (F/66 B-ALL, F/25 B-ALL, M/53 CML, F/34 B-ALL) and 1 cytogenetic cell pellet each in cell culture medium (M/52 CML) or Carnoy's fixative (M/44 CML) with known and previously characterized BCR::ABL1 fusion transcript were selected for study. The genomic DNA was extracted from each case for further breakpoint characterization by long read sequencing (MinION R9.4.1 flow cell, Oxford Nanopore Technologies, UK). RESULTS All the genomic breakpoints were concordant with the RNA fusion transcript breakpoints. Three typical (e1a2, e13a2, and e14a2) and 3 variant (e23a2Ins52, e8a2, and e13a2ins74) BCR breakpoints were identified. CONCLUSION Using the Ph translocation as an example, long read sequencing is a promising alternative method to detect SV, revolutionizing detection of chromosomal translocation to a higher precision. A more comprehensive spectrum of SV can be resolved along with cytogenetic results, enabling precise diagnosis and personalized monitoring of haematological malignancies.
Collapse
Affiliation(s)
- Janet Hei Yin Law
- Division of Molecular Pathology, Department of Pathology, Hong Kong Sanatorium & Hospital, Hong Kong, China
| | - Chun Hang Au
- Division of Molecular Pathology, Department of Pathology, Hong Kong Sanatorium & Hospital, Hong Kong, China
| | - Amy Wing-Sze Leung
- Division of Molecular Pathology, Department of Pathology, Hong Kong Sanatorium & Hospital, Hong Kong, China
| | - Henry C M Leung
- Division of Molecular Pathology, Department of Pathology, Hong Kong Sanatorium & Hospital, Hong Kong, China
| | - Elaine Y L Wong
- Division of Molecular Pathology, Department of Pathology, Hong Kong Sanatorium & Hospital, Hong Kong, China
| | - Beca B K Ip
- Division of Molecular Pathology, Department of Pathology, Hong Kong Sanatorium & Hospital, Hong Kong, China
| | - Dona N Y Ho
- Division of Molecular Pathology, Department of Pathology, Hong Kong Sanatorium & Hospital, Hong Kong, China
| | - Shing Yan Ma
- Specialist in Haematology & Haematological Oncology, Hong Kong, China
| | - Helen M H Chan
- Specialist in Haematology & Haematological Oncology, Hong Kong, China
| | - Edmond K W Chiu
- Specialist in Haematology & Haematological Oncology, Hong Kong, China
| | - James C S Chim
- Department of Medicine and Comprehensive Oncology Centre, Hong Kong Sanatorium & Hospital, Hong Kong, China
| | - Raymond H S Liang
- Department of Medicine and Comprehensive Oncology Centre, Hong Kong Sanatorium & Hospital, Hong Kong, China
| | - Thomas S K Wan
- Division of Molecular Pathology, Department of Pathology, Hong Kong Sanatorium & Hospital, Hong Kong, China
| | - Edmond S K Ma
- Division of Molecular Pathology, Department of Pathology, Hong Kong Sanatorium & Hospital, Hong Kong, China.
| |
Collapse
|
2
|
Abdallah SB, Olfson E, Cappi C, Greenspun S, Zai G, Rosário MC, Willsey AJ, Shavitt RG, Miguel EC, Kennedy JL, Richter MA, Fernandez TV. Characterizing Rare DNA Copy-Number Variants in Pediatric Obsessive-Compulsive Disorder. J Am Acad Child Adolesc Psychiatry 2025:S0890-8567(25)00160-1. [PMID: 40122455 DOI: 10.1016/j.jaac.2025.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 01/21/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025]
Abstract
OBJECTIVE Pediatric obsessive-compulsive disorder (OCD) is a common neuropsychiatric disorder for which genetic factors play an important role. Recent studies have demonstrated an enrichment of rare de novo DNA single nucleotide variants in OCD cases compared to controls, and larger studies have examined copy-number variants (CNVs) using microarray data. Our study examines rare de novo CNVs using whole-exome sequencing (WES) data to provide additional insight into genetic factors and biological processes underlying OCD. METHOD We detected CNVs using whole-exome DNA sequencing (WES) data from 183 OCD trio families (unaffected parents and children with OCD) and 771 control families to test the hypothesis that rare de novo CNVs are enriched in OCD cases compared to controls. Our primary analysis used the eXome-Hidden Markov Model (XHMM) to identify CNVs in silico. We performed burden analyses comparing individuals with OCD vs. controls and downstream biological systems analyses of CNVs in probands with OCD. We then used a second algorithm (GATK-gCNV) to confirm our primary analysis. RESULTS Our findings demonstrate a higher rate of rare de novo CNVs detected by WES in individuals with OCD (0.07 CNVs per proband) compared to controls (0.005) (corrected rate ratio = 11.7 95% CI, 3.6-50.0, p = 4.00x10-6). We confirmed this enrichment using GATK-gCNV. The majority of these rare de novo CNVs in OCD cases are predicted to be pathogenic or likely pathogenic, and an examination of genes disrupted by rare de novo CNVs in OCD cases finds enrichment of several gene-ontology sets. CONCLUSION This study shows for the first time an enrichment of rare de novo CNVs detected by WES in OCD, complementing previous larger CNV studies and providing additional insight into genetic factors underlying OCD risk.
Collapse
Affiliation(s)
| | - Emily Olfson
- Yale University School of Medicine, New Haven, Connecticut
| | - Carolina Cappi
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | - Gwyneth Zai
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Instituteof Medical Science and Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | - A Jeremy Willsey
- UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California
| | - Roseli G Shavitt
- Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo,Brazil
| | | | - James L Kennedy
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Instituteof Medical Science and Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Margaret A Richter
- Instituteof Medical Science and Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Frederick W. Thompson Anxiety Disorders Centre, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | | |
Collapse
|
3
|
Yoon JG, Lee S, Park S, Jang SS, Cho J, Kim MJ, Kim SY, Kim WJ, Lee JS, Chae JH. Identification of a novel non-coding deletion in Allan-Herndon-Dudley syndrome by long-read HiFi genome sequencing. BMC Med Genomics 2025; 18:41. [PMID: 40033291 DOI: 10.1186/s12920-024-02058-4] [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: 03/26/2024] [Accepted: 11/27/2024] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Allan-Herndon-Dudley syndrome (AHDS) is an X-linked disorder caused by pathogenic variants in the SLC16A2 gene. Although most reported variants are found in protein-coding regions or adjacent junctions, structural variations (SVs) within non-coding regions have not been previously reported. METHODS We investigated two male siblings with severe neurodevelopmental disorders and spasticity, who had remained undiagnosed for over a decade and were negative from exome sequencing, utilizing long-read HiFi genome sequencing. We conducted a comprehensive analysis including short-tandem repeats (STRs) and SVs to identify the genetic cause in this familial case. RESULTS While coding variant and STR analyses yielded negative results, SV analysis revealed a novel hemizygous deletion in intron 1 of the SLC16A2 gene (chrX:74,460,691 - 74,463,566; 2,876 bp), inherited from their carrier mother and shared by the siblings. Determination of the breakpoints indicates that the deletion probably resulted from Alu/Alu-mediated rearrangements between homologous AluY pairs. The deleted region is predicted to include multiple transcription factor binding sites, such as Stat2, Zic1, Zic2, and FOXD3, which are crucial for the neurodevelopmental process, as well as a regulatory element including an eQTL (rs1263181) that is implicated in the tissue-specific regulation of SLC16A2 expression, notably in skeletal muscle and thyroid tissues. CONCLUSIONS This report, to our knowledge, is the first to describe a non-coding deletion associated with AHDS, demonstrating the potential utility of long-read sequencing for undiagnosed patients. Although interpreting variants in non-coding regions remains challenging, our study highlights this region as a high priority for future investigation and functional studies.
Collapse
Affiliation(s)
- Jihoon G Yoon
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Laboratory Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seungbok Lee
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Soojin Park
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Se Song Jang
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jaeso Cho
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Pediatrics, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Man Jin Kim
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Soo Yeon Kim
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Woo Joong Kim
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jin Sook Lee
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jong-Hee Chae
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
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
|
7
|
Chang K, Jian X, Wu C, Gao C, Li Y, Chen J, Xue B, Ding Y, Peng L, Wang B, He L, Xu Y, Li C, Li X, Wang Z, Zhao X, Pan D, Yang Q, Zhou J, Zhu Z, Liu Z, Xia D, Feng G, Zhang Q, Wen Y, Shi Y, Li Z. The Contribution of Mosaic Chromosomal Alterations to Schizophrenia. Biol Psychiatry 2025; 97:198-207. [PMID: 38942348 DOI: 10.1016/j.biopsych.2024.06.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 06/30/2024]
Abstract
BACKGROUND Mosaic chromosomal alterations are implicated in neuropsychiatric disorders, but the contribution to schizophrenia (SCZ) risk for somatic copy number variations (sCNVs) emerging in early developmental stages has not been fully established. METHODS We analyzed blood-derived genotype arrays from 9715 patients with SCZ and 28,822 control participants of Chinese descent using a computational tool (MoChA) based on long-range chromosomal information to detect mosaic chromosomal alterations. We focused on probable early developmental sCNVs through stringent filtering. We assessed the burden of sCNVs across varying cell fraction cutoffs, as well as the frequency with which genes were involved in sCNVs. We integrated this data with the PGC (Psychiatric Genomics Consortium) dataset, which comprises 12,834 SCZ cases and 11,648 controls of European descent, and complemented it with genotyping data from postmortem brain tissue of 936 participants (449 cases and 487 controls). RESULTS Patients with SCZ had a significantly higher somatic losses detection rate than control participants (1.00% vs. 0.52%; odds ratio = 1.91; 95% CI, 1.47-2.49; two-sided Fisher's exact test, p = 1.49 × 10-6). Further analysis indicated that the odds ratios escalated proportionately (from 1.91 to 2.78) with the increment in cell fraction cutoffs. Recurrent sCNVs associated with SCZ (odds ratio > 8; Fisher's exact test, p < .05) were identified, including notable regions at 10q21.1 (ZWINT), 3q26.1 (SLITRK3), 1q31.1 (BRINP3) and 12q21.31-21.32 (MGAT4C and NTS) in the Chinese cohort, and some regions were validated with PGC data. Cross-tissue validation pinpointed somatic losses at loci like 1p35.3-35.2 and 19p13.3-13.2. CONCLUSIONS The study highlights the significant impact of mosaic chromosomal alterations on SCZ, suggesting their pivotal role in the disorder's genetic etiology.
Collapse
Affiliation(s)
- Kaihui Chang
- The Affiliated Hospital of Qingdao University & The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China; School of Basic Medicine, Qingdao University, Qingdao, China; National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Xuemin Jian
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Chuanhong Wu
- The Affiliated Hospital of Qingdao University & The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China; School of Basic Medicine, Qingdao University, Qingdao, China
| | - Chengwen Gao
- The Affiliated Hospital of Qingdao University & The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China
| | - Yafang Li
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jianhua Chen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China; Shanghai Clinical Research Center for Mental Health, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Baiqiang Xue
- The Affiliated Hospital of Qingdao University & The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China; School of Public Health, Qingdao University, Qingdao, China
| | - Yonghe Ding
- The Affiliated Hospital of Qingdao University & The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China; School of Public Health, Qingdao University, Qingdao, China
| | - Lixia Peng
- The Affiliated Hospital of Qingdao University & The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China; School of Pharmacy, Qingdao University, Qingdao, China
| | - Baokun Wang
- The Affiliated Hospital of Qingdao University & The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China; School of Pharmacy, Qingdao University, Qingdao, China
| | - Lin He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Yifeng Xu
- Shanghai Clinical Research Center for Mental Health, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Changgui Li
- Shandong Provincial Key Laboratory of Metabolic Disease & the Metabolic Disease Institute of Qingdao University, Qingdao, China
| | - Xingwang Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Zhuo Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Xiangzhong Zhao
- The Affiliated Hospital of Qingdao University & The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China
| | - Dun Pan
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Qiangzhen Yang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Juan Zhou
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Zijia Zhu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Ze Liu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Disong Xia
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Guoyin Feng
- Shanghai Clinical Research Center for Mental Health, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Zhang
- The Affiliated Hospital of Qingdao University & The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China
| | - Yanqin Wen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Yongyong Shi
- The Affiliated Hospital of Qingdao University & The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China; School of Basic Medicine, Qingdao University, Qingdao, China; Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China; Shanghai Clinical Research Center for Mental Health, Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shandong Provincial Key Laboratory of Metabolic Disease & the Metabolic Disease Institute of Qingdao University, Qingdao, China; Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, China; Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China; Department of Psychiatry, First Teaching Hospital of Xinjiang Medical University, Urumqi, China; Changning Mental Health Center, Shanghai, China.
| | - Zhiqiang Li
- The Affiliated Hospital of Qingdao University & The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China; School of Basic Medicine, Qingdao University, Qingdao, China; Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China; School of Public Health, Qingdao University, Qingdao, China; School of Pharmacy, Qingdao University, Qingdao, China; Shandong Provincial Key Laboratory of Metabolic Disease & the Metabolic Disease Institute of Qingdao University, Qingdao, China; Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China.
| |
Collapse
|
8
|
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
|
9
|
Ahn JH, Yoon JG, Cho J, Lee S, Kim S, Kim MJ, Kim SY, Lee ST, Chu K, Lee SK, Kim HJ, Youn J, Jang JH, Chae JH, Moon J, Cho JW. Implementing genomic medicine in clinical practice for adults with undiagnosed rare diseases. NPJ Genom Med 2024; 9:63. [PMID: 39609445 PMCID: PMC11604660 DOI: 10.1038/s41525-024-00449-1] [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: 06/05/2024] [Accepted: 11/18/2024] [Indexed: 11/30/2024] Open
Abstract
The global burden of undiagnosed diseases, particularly in adults, is rising due to their significant socioeconomic impact. To address this, we enrolled 232 adult probands with undiagnosed conditions, utilizing bioinformatics tools for genetic analysis. Alongside exome and genome sequencing, repeat-primed PCR and Cas9-mediated nanopore sequencing were applied to suspected short tandem repeat disorders. Probands were classified into probable genetic (n = 128) or uncertain (n = 104) origins. The study found genetic causes in 66 individuals (28.4%) and non-genetic causes in 12 (5.2%), with a longer diagnostic journey for those in the probable genetic group or with pediatric symptom onset, emphasizing the need for increased efforts in these populations. Genetic diagnoses facilitated effective surveillance, cascade screening, drug repurposing, and pregnancy planning. This study demonstrates that integrating sequencing technologies improves diagnostic accuracy, may shorten the time to diagnosis, and enhances personalized management for adults with undiagnosed diseases.
Collapse
Affiliation(s)
- Jong Hyeon Ahn
- Department of Neurology, Samsung Medical Centre, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Jihoon G Yoon
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Laboratory Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jaeso Cho
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Pediatrics, Seoul National University Bundang Hospital, Seongnam-si, Republic of Korea
| | - Seungbok Lee
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Pediatrics, Seoul National University Children's Hospital, Seoul, Republic of Korea
| | - Sheehyun Kim
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Man Jin Kim
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Soo Yeon Kim
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Pediatrics, Seoul National University Children's Hospital, Seoul, Republic of Korea
| | - Soon-Tae Lee
- Department of Neurology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Kon Chu
- Department of Neurology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sang Kun Lee
- Department of Neurology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Han-Joon Kim
- Department of Neurology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jinyoung Youn
- Department of Neurology, Samsung Medical Centre, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Ja-Hyun Jang
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jong-Hee Chae
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Pediatrics, Seoul National University Children's Hospital, Seoul, Republic of Korea
| | - Jangsup Moon
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
- Department of Neurology, Seoul National University Hospital, Seoul, Republic of Korea.
| | - Jin Whan Cho
- Department of Neurology, Samsung Medical Centre, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea.
| |
Collapse
|
10
|
Su KJ, Qiu C, Greenbaum J, Zhang X, Liu A, Liu Y, Luo Z, Mungasavalli Gnanesh SS, Tian Q, Zhao LJ, Shen H, Deng HW. Genomic structural variations link multiple genes to bone mineral density in a multi-ethnic cohort study: Louisiana osteoporosis study. J Bone Miner Res 2024; 39:1474-1485. [PMID: 39167757 PMCID: PMC11425707 DOI: 10.1093/jbmr/zjae133] [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: 03/21/2024] [Revised: 07/11/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024]
Abstract
Osteoporosis, characterized by low BMD, is a highly heritable metabolic bone disorder. Although single nucleotide variations (SNVs) have been extensively studied, they explain only a fraction of BMD heritability. Although genomic structural variations (SVs) are large-scale genomic alterations that contribute to genetic diversity in shaping phenotypic variations, the role of SVs in osteoporosis susceptibility remains poorly understood. This study aims to identify and prioritize genes that harbor BMD-related SVs. We performed whole genome sequencing on 4982 subjects from the Louisiana Osteoporosis Study. To obtain high-confidence SVs, the detection of SVs was performed using an ensemble approach. The SVs were tested for association with BMD variation at the hip (HIP), femoral neck (FNK), and lumbar spine (SPN), respectively. Additionally, we conducted co-occurrence analysis using multi-omics approaches to prioritize the identified genes based on their functional importance. Stratification was employed to explore the sex- and ethnicity-specific effects. We identified significant SV-BMD associations: 125 for FNK-BMD, 99 for SPN-BMD, and 83 for HIP-BMD. We observed SVs that were commonly associated with both FNK and HIP BMDs in our combined and stratified analyses. These SVs explain 13.3% to 19.1% of BMD variation. Novel bone-related genes emerged, including LINC02370, ZNF family genes, and ZDHHC family genes. Additionally, FMN2, carrying BMD-related deletions, showed associations with FNK or HIP BMDs, with sex-specific effects. The co-occurrence analysis prioritized an RNA gene LINC00494 and ZNF family genes positively associated with BMDs at different skeletal sites. Two potential causal genes, IBSP and SPP1, for osteoporosis were also identified. Our study uncovers new insights into genetic factors influencing BMD through SV analysis. We highlight BMD-related SVs, revealing a mix of shared and specific genetic influences across skeletal sites and gender or ethnicity. These findings suggest potential roles in osteoporosis pathophysiology, opening avenues for further research and therapeutic targets.
Collapse
Affiliation(s)
- Kuan-Jui Su
- Deming Department of Medicine, School of Medicine, Tulane Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA 70112, United States
| | - Chuan Qiu
- Deming Department of Medicine, School of Medicine, Tulane Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA 70112, United States
| | - Jonathan Greenbaum
- Deming Department of Medicine, School of Medicine, Tulane Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA 70112, United States
| | - Xiao Zhang
- Deming Department of Medicine, School of Medicine, Tulane Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA 70112, United States
| | - Anqi Liu
- Deming Department of Medicine, School of Medicine, Tulane Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA 70112, United States
| | - Yong Liu
- Deming Department of Medicine, School of Medicine, Tulane Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA 70112, United States
| | - Zhe Luo
- Deming Department of Medicine, School of Medicine, Tulane Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA 70112, United States
| | - Shashank Sajjan Mungasavalli Gnanesh
- Deming Department of Medicine, School of Medicine, Tulane Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA 70112, United States
| | - Qing Tian
- Deming Department of Medicine, School of Medicine, Tulane Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA 70112, United States
| | - Lan-Juan Zhao
- Deming Department of Medicine, School of Medicine, Tulane Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA 70112, United States
| | - Hui Shen
- Deming Department of Medicine, School of Medicine, Tulane Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA 70112, United States
| | - Hong-Wen Deng
- Deming Department of Medicine, School of Medicine, Tulane Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA 70112, United States
| |
Collapse
|
11
|
Moura DS, López López D, di Lernia D, Martin-Ruiz M, Lopez-Alvarez M, Ramos R, Merino J, Dopazo J, Lopez-Guerrero J, Mondaza-Hernandez JL, Romero P, Hindi N, Garcia-Foncillas J, Martin-Broto J. Shared germline genomic variants in two patients with double primary gastrointestinal stromal tumours (GISTs). J Med Genet 2024; 61:927-934. [PMID: 39153853 DOI: 10.1136/jmg-2024-110109] [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: 05/07/2024] [Accepted: 07/31/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND Gastrointestinal stromal tumours (GISTs) are prevalent mesenchymal tumours of the gastrointestinal tract, commonly exhibiting structural variations in KIT and PDGFRA genes. While the mutational profiling of somatic tumours is well described, the genes behind the susceptibility to develop GIST are not yet fully discovered. This study explores the genomic landscape of two primary GIST cases, aiming to identify shared germline pathogenic variants and shed light on potential key players in tumourigenesis. METHODS Two patients with distinct genotypically and phenotypically GISTs underwent germline whole genome sequencing. CNV and single nucleotide variant (SNV) analyses were performed. RESULTS Both patients harbouring low-risk GISTs with different mutations (PDGFRA and KIT) shared homozygous germline pathogenic deletions in both CFHR1 and CFHR3 genes. CNV analysis revealed additional shared pathogenic deletions in other genes such as SLC25A24. No particular pathogenic SNV shared by both patients was detected. CONCLUSION Our study provides new insights into germline variants that can be associated with the development of GISTs, namely, CFHR1 and CFHR3 deep deletions. Further functional validation is warranted to elucidate the precise contributions of identified germline mutations in GIST development.
Collapse
Affiliation(s)
- David S Moura
- Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
| | - Daniel López López
- Computational Medicine Platform, Fundación progreso y salud (FPS), Hospital Virgen del Rocío, Seville, Spain
- Bioinformatics in Rare Diseases (BiER), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), FPS, Hospital Virgen del Rocio, Seville, Spain
| | - Davide di Lernia
- Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
| | - Marta Martin-Ruiz
- Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
| | | | - Rafael Ramos
- Pathology Department, University Hospital Son Espases, Mallorca, Spain
| | - Jose Merino
- Pathology Department, Fundación Jimenez Diaz University Hospital, Madrid, Spain
| | - Joaquin Dopazo
- Computational Medicine Platform, Fundación progreso y salud (FPS), Hospital Virgen del Rocío, Seville, Spain
- Bioinformatics in Rare Diseases (BiER), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), FPS, Hospital Virgen del Rocio, Seville, Spain
- Instituto de Biomedicina de Sevilla (IBiS; HUVR, CSIC, US), Sevilla, Spain
| | - Jose Lopez-Guerrero
- Laboratory of Molecular Biology, Fundación Instituto Valenciano de Oncologia, Valencia, Spain
| | - Jose L Mondaza-Hernandez
- Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
| | - Pablo Romero
- Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
| | - Nadia Hindi
- Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- Medical Oncology Department, Fundación Jimenez Diaz University Hospital, Madrid, Spain
- General de Villalba University Hospital, Madrid, Spain
| | - Jesus Garcia-Foncillas
- Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- Medical Oncology Department, Fundación Jimenez Diaz University Hospital, Madrid, Spain
| | - Javier Martin-Broto
- Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- Medical Oncology Department, Fundación Jimenez Diaz University Hospital, Madrid, Spain
- General de Villalba University Hospital, Madrid, Spain
| |
Collapse
|
12
|
Ormond C, Ryan NM, Byerley W, Heron EA, Corvin A. Investigating copy number variants in schizophrenia pedigrees using a new consensus pipeline called PECAN. Sci Rep 2024; 14:17518. [PMID: 39080331 PMCID: PMC11289470 DOI: 10.1038/s41598-024-66021-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 06/26/2024] [Indexed: 08/02/2024] Open
Abstract
Copy number variants (CNVs) have been implicated in many human diseases, including psychiatric disorders. Whole genome sequencing offers advantages in CNV calling compared to previous array-based methods. Here we present a robust and transparent CNV calling pipeline, PECAN (PEdigree Copy number vAriaNt calling), for short-read, whole genome sequencing data, comprised of a novel combination of four calling methods and structural variant genotyping. This method is scalable and can incorporate pedigree information to retain lower-confidence CNVs that would otherwise be discarded. We have robustly benchmarked PECAN using gold-standard CNV calls for two well-established evaluation samples, NA12878 and HG002, showing that PECAN performs with high precision and recall on both datasets, outperforming another pedigree-based CNV calling pipeline. As part of this work, we provide a list of high-confidence gold standard CNVs for the NA12878 reference sample, curated from multiple studies. We applied PECAN to a collection of pedigrees multiply affected with schizophrenia and identified a rare deletion that perfectly co-segregates with schizophrenia in one of the pedigrees. The CNV overlaps the gene PITRM1, which has been implicated in a complex phenotype including ataxia, developmental delay, and schizophrenia-like episodes in affected adults.
Collapse
Affiliation(s)
- Cathal Ormond
- Neuropsychiatric Genetics Research Group, Department of Psychiatry, Trinity Centre for Health Sciences, Trinity College Dublin, James' Street, Dublin 8, Ireland
| | - Niamh M Ryan
- Neuropsychiatric Genetics Research Group, Department of Psychiatry, Trinity Centre for Health Sciences, Trinity College Dublin, James' Street, Dublin 8, Ireland
| | - William Byerley
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, USA
| | - Elizabeth A Heron
- Neuropsychiatric Genetics Research Group, Department of Psychiatry, Trinity Centre for Health Sciences, Trinity College Dublin, James' Street, Dublin 8, Ireland
| | - Aiden Corvin
- Neuropsychiatric Genetics Research Group, Department of Psychiatry, Trinity Centre for Health Sciences, Trinity College Dublin, James' Street, Dublin 8, Ireland.
| |
Collapse
|
13
|
Wang C, Liu H, Li XY, Ma J, Gu Z, Feng X, Xie S, Tang BS, Chen S, Wang W, Wang J, Zhang J, Chan P. High-depth whole-genome sequencing identifies structure variants, copy number variants and short tandem repeats associated with Parkinson's disease. NPJ Parkinsons Dis 2024; 10:134. [PMID: 39043730 PMCID: PMC11266557 DOI: 10.1038/s41531-024-00722-1] [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: 01/07/2023] [Accepted: 05/10/2024] [Indexed: 07/25/2024] Open
Abstract
While numerous single nucleotide variants and small indels have been identified in Parkinson's disease (PD), the contribution of structural variants (SVs), copy number variants (CNVs), and short tandem repeats (STRs) remains poorly understood. Here we investigated the association using the high-depth whole-genome sequencing data from 466 Chinese PD patients and 513 controls. Totally, we identified 29,561 SVs, 32,153 CNVs, and 174,905 STRs, and found that CNV deletions were significantly enriched in the end-proportion of autosomal chromosomes in PD. After genome-wide association analysis and replication in an external cohort of 352 cases and 547 controls, we validated that the 1.6 kb-deletion neighboring MUC19, 12.4kb-deletion near RXFP1 and GGGAAA repeats in SLC2A13 were significantly associated with PD. Moreover, the MUC19 deletion and the SLC2A13 5-copy repeat reduced the penetrance of the LRRK2 G2385R variant. Moreover, genes with these variants were dosage-sensitive. These data provided novel insights into the genetic architecture of PD.
Collapse
Affiliation(s)
- Chaodong Wang
- Department of Neurology & Neurobiology, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Hankui Liu
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, 518083, China
| | - Xu-Ying Li
- Department of Neurology & Neurobiology, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Jinghong Ma
- Department of Neurology & Neurobiology, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Zhuqin Gu
- Department of Neurology & Neurobiology, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Xiuli Feng
- National Human Genome Center in Beijing, Beijing Economic-Technological Development Zone, Beijing, 100176, China
| | - Shu Xie
- National Human Genome Center in Beijing, Beijing Economic-Technological Development Zone, Beijing, 100176, China
| | - Bei-Sha Tang
- Department of Neurology, Xiangya Hospital, Central South University, State Key Laboratory of Medical Genetics, Changsha, China
| | - Shengdi Chen
- Department of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Wang
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, 518083, China
| | - Jian Wang
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, 518083, China
| | - Jianguo Zhang
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, 518083, China.
- Hebei Industrial Technology Research Institute of Genomics in Maternal & Child Health, Shijiazhuang, 050000, China.
| | - Piu Chan
- Department of Neurology & Neurobiology, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China.
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.
- Clinical Center for Parkinson's Disease, Capital Medical University, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory for Parkinson's Disease, Beijing, China.
- Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China.
| |
Collapse
|
14
|
Wiener E, Cottino L, Botha G, Nyangiri O, Noyes H, McLeod A, Jakubosky D, Adebamowo C, Awadalla P, Landouré G, Matshaba M, Matovu E, Ramsay M, Simo G, Simuunza M, Tiemessen C, Wonkam A, Sahibdeen V, Krause A, Lombard Z, Hazelhurst S. An assessment of the genomic structural variation landscape in Sub-Saharan African populations. RESEARCH SQUARE 2024:rs.3.rs-4485126. [PMID: 39041024 PMCID: PMC11261963 DOI: 10.21203/rs.3.rs-4485126/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Structural variants are responsible for a large part of genomic variation between individuals and play a role in both common and rare diseases. Databases cataloguing structural variants notably do not represent the full spectrum of global diversity, particularly missing information from most African populations. To address this representation gap, we analysed 1,091 high-coverage African genomes, 545 of which are public data sets, and 546 which have been analysed for structural variants for the first time. Variants were called using five different tools and datasets merged and jointly called using SURVIVOR. We identified 67,795 structural variants throughout the genome, with 10,421 genes having at least one variant. Using a conservative overlap in merged data, 6,414 of the structural variants (9.5%) are novel compared to the Database of Genomic Variants. This study contributes to knowledge of the landscape of structural variant diversity in Africa and presents a reliable dataset for potential applications in population genetics and health-related research.
Collapse
Affiliation(s)
- Emma Wiener
- Sydney Brenner Institute for Molecular Bioscience, University of the Witwatersrand, Johannesburg, South Africa
- Division of Human Genetics, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Laura Cottino
- Sydney Brenner Institute for Molecular Bioscience, University of the Witwatersrand, Johannesburg, South Africa
- Division of Human Genetics, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Gerrit Botha
- Computational Biology Unit, University of Cape Town, Cape Town, South Africa
| | - Oscar Nyangiri
- College of Veterinary Medicine, Animal Resources and Biosecurity Makerere University, Kampala, Uganda
| | - Harry Noyes
- Centre for Genomic Research, University of Liverpool, Liverpool, United Kingdom
| | - Annette McLeod
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, United Kingdom
| | - David Jakubosky
- Department of Biomedical Informatics, University of California, San Diego, United States of America
- Institute of Genomic Medicine, University of California, San Diego, United States of America
| | - Clement Adebamowo
- Department of Epidemiology and Public Health and Greenebaum Comprehensive Cancer Center University of Maryland School of Medicine, Baltimore, United States of America
| | - Phillip Awadalla
- Ontario Institute for Cancer Research, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Guida Landouré
- Faculty of Medicine and Odontostomatology University of Sciences, Techniques and Technology of Bamako, Bamako Mali
- Neurology Department Point ”G” University Hospital, Bamako, Mali
| | - Mogomotsi Matshaba
- Botswana-Baylor Children’s Clinical Center of Excellence, Gaborone, Botswana
- Baylor College of Medicine, Houston, United States
| | - Enock Matovu
- College of Veterinary Medicine, Animal Resources and Biosecurity Makerere University, Kampala, Uganda
| | - Michèle Ramsay
- Sydney Brenner Institute for Molecular Bioscience, University of the Witwatersrand, Johannesburg, South Africa
| | - Gustave Simo
- Molecular Parasitology and Entomology Unit, Department of Biochemistry University of Dschang, Dschang, Cameroon
| | - Martin Simuunza
- Department of Disease Control, School of Veterinary Medicine University of Zambia, Lusaka, Zambia
| | - Caroline Tiemessen
- Centre for HIV and STIs, National Institute for Communicable Diseases, National Health Laboratory Services and Faculty of Health Sciences University of the Witwatersrand, Johannesburg, South Africa
| | - Ambroise Wonkam
- McKusick-Nathans Institute and Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, United States of America
- Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Venesa Sahibdeen
- Division of Human Genetics, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Amanda Krause
- Division of Human Genetics, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Zané Lombard
- Division of Human Genetics, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Scott Hazelhurst
- Sydney Brenner Institute for Molecular Bioscience, University of the Witwatersrand, Johannesburg, South Africa
- School of Electrical & Information Engineering, University of the Witwatersrand, Johannesburg, South Africa
| | | |
Collapse
|
15
|
Kramer M, Goodwin S, Wappel R, Borio M, Offit K, Feldman DR, Stadler ZK, McCombie WR. Exploring the genetic and epigenetic underpinnings of early-onset cancers: Variant prioritization for long read whole genome sequencing from family cancer pedigrees. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.601096. [PMID: 39005350 PMCID: PMC11244929 DOI: 10.1101/2024.06.27.601096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Despite significant advances in our understanding of genetic cancer susceptibility, known inherited cancer predisposition syndromes explain at most 20% of early-onset cancers. As early-onset cancer prevalence continues to increase, the need to assess previously inaccessible areas of the human genome, harnessing a trio or quad family-based architecture for variant filtration, may reveal further insights into cancer susceptibility. To assess a broader spectrum of variation than can be ascertained by multi-gene panel sequencing, or even whole genome sequencing with short reads, we employed long read whole genome sequencing using an Oxford Nanopore Technology (ONT) PromethION of 3 families containing an early-onset cancer proband using a trio or quad family architecture. Analysis included 2 early-onset colorectal cancer family trios and one quad consisting of two siblings with testicular cancer, all with unaffected parents. Structural variants (SVs), epigenetic profiles and single nucleotide variants (SNVs) were determined for each individual, and a filtering strategy was employed to refine and prioritize candidate variants based on the family architecture. The family architecture enabled us to focus on inapposite variants while filtering variants shared with the unaffected parents, significantly decreasing background variation that can hamper identification of potentially disease causing differences. Candidate d e novo and compound heterozygous variants were identified in this way. Gene expression, in matched neoplastic and pre-neoplastic lesions, was assessed for one trio. Our study demonstrates the feasibility of a streamlined analysis of genomic variants from long read ONT whole genome sequencing and a way to prioritize key variants for further evaluation of pathogenicity, while revealing what may be missing from panel based analyses.
Collapse
|
16
|
Lin Y, Wang J, Xu R, Xu Z, Wang Y, Pan S, Zhang Y, Tao Q, Zhao Y, Yan C, Cao Z, Ji K. HiFi long-read amplicon sequencing for full-spectrum variants of human mtDNA. BMC Genomics 2024; 25:538. [PMID: 38822239 PMCID: PMC11141058 DOI: 10.1186/s12864-024-10433-9] [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: 02/26/2024] [Accepted: 05/20/2024] [Indexed: 06/02/2024] Open
Abstract
BACKGROUND Mitochondrial diseases (MDs) can be caused by single nucleotide variants (SNVs) and structural variants (SVs) in the mitochondrial genome (mtDNA). Presently, identifying deletions in small to medium-sized fragments and accurately detecting low-percentage variants remains challenging due to the limitations of next-generation sequencing (NGS). METHODS In this study, we integrated targeted long-range polymerase chain reaction (LR-PCR) and PacBio HiFi sequencing to analyze 34 participants, including 28 patients and 6 controls. Of these, 17 samples were subjected to both targeted LR-PCR and to compare the mtDNA variant detection efficacy. RESULTS Among the 28 patients tested by long-read sequencing (LRS), 2 patients were found positive for the m.3243 A > G hotspot variant, and 20 patients exhibited single or multiple deletion variants with a proportion exceeding 4%. Comparison between the results of LRS and NGS revealed that both methods exhibited similar efficacy in detecting SNVs exceeding 5%. However, LRS outperformed NGS in detecting SNVs with a ratio below 5%. As for SVs, LRS identified single or multiple deletions in 13 out of 17 cases, whereas NGS only detected single deletions in 8 cases. Furthermore, deletions identified by LRS were validated by Sanger sequencing and quantified in single muscle fibers using real-time PCR. Notably, LRS also effectively and accurately identified secondary mtDNA deletions in idiopathic inflammatory myopathies (IIMs). CONCLUSIONS LRS outperforms NGS in detecting various types of SNVs and SVs in mtDNA, including those with low frequencies. Our research is a significant advancement in medical comprehension and will provide profound insights into genetics.
Collapse
Affiliation(s)
- Yan Lin
- Research Institute of Neuromuscular and Neurodegenerative Diseases and Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Jiayin Wang
- Research Institute of Neuromuscular and Neurodegenerative Diseases and Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Ran Xu
- GrandOmics Biosciences, No.56 Zhichun Road, Haidian District, Beijing, 100098, China
| | - Zhe Xu
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Yifan Wang
- GrandOmics Biosciences, No.56 Zhichun Road, Haidian District, Beijing, 100098, China
| | - Shirang Pan
- GrandOmics Biosciences, No.56 Zhichun Road, Haidian District, Beijing, 100098, China
| | - Yan Zhang
- GrandOmics Biosciences, No.56 Zhichun Road, Haidian District, Beijing, 100098, China
| | - Qing Tao
- GrandOmics Biosciences, No.56 Zhichun Road, Haidian District, Beijing, 100098, China
| | - Yuying Zhao
- Research Institute of Neuromuscular and Neurodegenerative Diseases and Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Chuanzhu Yan
- Research Institute of Neuromuscular and Neurodegenerative Diseases and Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
- Mitochondrial Medicine Laboratory, Qilu Hospital (Qingdao), Shandong University, Qingdao, Shandong, 266035, China
- Brain Science Research Institute, Shandong University, Jinan, Shandong, 250012, China
| | - Zhenhua Cao
- GrandOmics Biosciences, No.56 Zhichun Road, Haidian District, Beijing, 100098, China.
| | - Kunqian Ji
- Research Institute of Neuromuscular and Neurodegenerative Diseases and Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
- Research Institute of Neuromuscular and Neurodegenerative Diseases, Department of Neurology, Qilu Hospital, Shandong University, No. 107 West Wenhua Road, Jinan, Shandong, 250012, China.
| |
Collapse
|
17
|
Fernández-Suárez E, González-Del Pozo M, Méndez-Vidal C, Martín-Sánchez M, Mena M, de la Morena-Barrio B, Corral J, Borrego S, Antiñolo G. Long-read sequencing improves the genetic diagnosis of retinitis pigmentosa by identifying an Alu retrotransposon insertion in the EYS gene. Mob DNA 2024; 15:9. [PMID: 38704576 PMCID: PMC11069205 DOI: 10.1186/s13100-024-00320-1] [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: 02/21/2024] [Accepted: 04/10/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Biallelic variants in EYS are the major cause of autosomal recessive retinitis pigmentosa (arRP) in certain populations, a clinically and genetically heterogeneous disease that may lead to legal blindness. EYS is one of the largest genes (~ 2 Mb) expressed in the retina, in which structural variants (SVs) represent a common cause of disease. However, their identification using short-read sequencing (SRS) is not always feasible. Here, we conducted targeted long-read sequencing (T-LRS) using adaptive sampling of EYS on the MinION sequencing platform (Oxford Nanopore Technologies) to definitively diagnose an arRP family, whose affected individuals (n = 3) carried the heterozygous pathogenic deletion of exons 32-33 in the EYS gene. As this was a recurrent variant identified in three additional families in our cohort, we also aimed to characterize the known deletion at the nucleotide level to assess a possible founder effect. RESULTS T-LRS in family A unveiled a heterozygous AluYa5 insertion in the coding exon 43 of EYS (chr6(GRCh37):g.64430524_64430525ins352), which segregated with the disease in compound heterozygosity with the previously identified deletion. Visual inspection of previous SRS alignments using IGV revealed several reads containing soft-clipped bases, accompanied by a slight drop in coverage at the Alu insertion site. This prompted us to develop a simplified program using grep command to investigate the recurrence of this variant in our cohort from SRS data. Moreover, LRS also allowed the characterization of the CNV as a ~ 56.4kb deletion spanning exons 32-33 of EYS (chr6(GRCh37):g.64764235_64820592del). The results of further characterization by Sanger sequencing and linkage analysis in the four families were consistent with a founder variant. CONCLUSIONS To our knowledge, this is the first report of a mobile element insertion into the coding sequence of EYS, as a likely cause of arRP in a family. Our study highlights the value of LRS technology in characterizing and identifying hidden pathogenic SVs, such as retrotransposon insertions, whose contribution to the etiopathogenesis of rare diseases may be underestimated.
Collapse
Affiliation(s)
- Elena Fernández-Suárez
- Department of Maternofetal Medicine, Genetics and Reproduction, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC, University of Seville, Seville, Spain
- Center for Biomedical Network Research On Rare Diseases (CIBERER), Seville, Spain
| | - María González-Del Pozo
- Department of Maternofetal Medicine, Genetics and Reproduction, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC, University of Seville, Seville, Spain
- Center for Biomedical Network Research On Rare Diseases (CIBERER), Seville, Spain
| | - Cristina Méndez-Vidal
- Department of Maternofetal Medicine, Genetics and Reproduction, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC, University of Seville, Seville, Spain
- Center for Biomedical Network Research On Rare Diseases (CIBERER), Seville, Spain
| | - Marta Martín-Sánchez
- Department of Maternofetal Medicine, Genetics and Reproduction, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC, University of Seville, Seville, Spain
- Center for Biomedical Network Research On Rare Diseases (CIBERER), Seville, Spain
| | - Marcela Mena
- Department of Maternofetal Medicine, Genetics and Reproduction, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC, University of Seville, Seville, Spain
- Center for Biomedical Network Research On Rare Diseases (CIBERER), Seville, Spain
| | - Belén de la Morena-Barrio
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Pascual Parrilla, CIBERER-ISCIII, Murcia, Spain
| | - Javier Corral
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Pascual Parrilla, CIBERER-ISCIII, Murcia, Spain
| | - Salud Borrego
- Department of Maternofetal Medicine, Genetics and Reproduction, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC, University of Seville, Seville, Spain.
- Center for Biomedical Network Research On Rare Diseases (CIBERER), Seville, Spain.
| | - Guillermo Antiñolo
- Department of Maternofetal Medicine, Genetics and Reproduction, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC, University of Seville, Seville, Spain.
- Center for Biomedical Network Research On Rare Diseases (CIBERER), Seville, Spain.
| |
Collapse
|
18
|
Bjørnstad PM, Aaløkken R, Åsheim J, Sundaram AYM, Felde CN, Østby GH, Dalland M, Sjursen W, Carrizosa C, Vigeland MD, Sorte HS, Sheng Y, Ariansen SL, Grindedal EM, Gilfillan GD. A 39 kb structural variant causing Lynch Syndrome detected by optical genome mapping and nanopore sequencing. Eur J Hum Genet 2024; 32:513-520. [PMID: 38030917 PMCID: PMC11061271 DOI: 10.1038/s41431-023-01494-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/19/2023] [Accepted: 11/06/2023] [Indexed: 12/01/2023] Open
Abstract
Lynch Syndrome (LS) is a hereditary cancer syndrome caused by pathogenic germline variants in one of the four mismatch repair (MMR) genes MLH1, MSH2, MSH6 and PMS2. It is characterized by a significantly increased risk of multiple cancer types, particularly colorectal and endometrial cancer, with autosomal dominant inheritance. Access to precise and sensitive methods for genetic testing is important, as early detection and prevention of cancer is possible when the variant is known. We present here two unrelated Norwegian families with family histories strongly suggestive of LS, where immunohistochemical and microsatellite instability analyses indicated presence of a pathogenic variant in MSH2, but targeted exon sequencing and multiplex ligation-dependent probe amplification (MLPA) were negative. Using Bionano optical genome mapping, we detected a 39 kb insertion in the MSH2 gene. Precise mapping of the insertion breakpoints and inserted sequence was performed by low-coverage whole-genome sequencing with an Oxford Nanopore MinION. The same variant was present in both families, and later found in other families from the same region of Norway, indicative of a founder event. To our knowledge, this is the first diagnosis of LS caused by a structural variant using these technologies. We suggest that structural variant detection be performed when LS is suspected but not confirmed with first-tier standard genetic testing.
Collapse
Affiliation(s)
- Pål Marius Bjørnstad
- Department Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Ragnhild Aaløkken
- Department Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - June Åsheim
- Department Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Arvind Y M Sundaram
- Department Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Caroline N Felde
- Department Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - G Henriette Østby
- Department Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Marianne Dalland
- Department Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Wenche Sjursen
- Department of Clinical & Molecular Medicine, NTNU and Department of Medical Genetics, St Olavs Hospital, Trondheim, Norway
| | - Christian Carrizosa
- Department Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Magnus D Vigeland
- Department Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
- Department of Forensic Sciences, Oslo University Hospital, 0372, Oslo, Norway
| | - Hanne S Sorte
- Department Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Ying Sheng
- Department Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Sarah L Ariansen
- Department Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Eli Marie Grindedal
- Department Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Gregor D Gilfillan
- Department Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway.
| |
Collapse
|
19
|
Wang J, Zhang C, Zhang L, Yao HJ, Liu X, Shi Y, Zhao J, Bo X, Chen H, Li L. Comparative study on genomic and epigenomic profiles of retinoblastoma or tuberous sclerosis complex via nanopore sequencing and a joint screening framework. Cancer Gene Ther 2024; 31:439-453. [PMID: 38146007 DOI: 10.1038/s41417-023-00714-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: 08/09/2023] [Revised: 11/23/2023] [Accepted: 11/29/2023] [Indexed: 12/27/2023]
Abstract
Recurrence and extraocular metastasis in advanced intraocular retinoblastoma (RB) are still major obstacles for successful treatment of Chinese children. Tuberous sclerosis complex (TSC) is a very rare, multisystemic genetic disorder characterized by hamartomatous growth. In this study, we aimed to compare genomic and epigenomic profiles with human RB or TSC using recently developed nanopore sequencing, and to identify disease-associated variations or genes. Peripheral blood samples were collected from either RB or RB/TSC patients plus their normal siblings, followed by nanopore sequencing and identification of disease-specific structural variations (SVs) and differentially methylated regions (DMRs) by a systematic biology strategy named as multiomics-based joint screening framework. In total, 316 RB- and 1295 TSC-unique SVs were identified, as well as 1072 RB- and 1114 TSC-associated DMRs, respectively. We eventually identified 6 key genes for RB for further functional validation. Knockdown of CDK19 with specific siRNAs significantly inhibited Y79 cellular proliferation and increased sensitivity to carboplatin, whereas downregulation of AHNAK2 promoted the cell growth as well as drug resistance. Those two genes might serve as potential diagnostic markers or therapeutic targets of RB. The systematic biology strategy combined with functional validation might be an effective approach for rare pediatric malignances with limited samples and challenging collection process.
Collapse
Affiliation(s)
- Junting Wang
- State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Biotechnology for Microbial Drugs, Department of Oncology, Institute of Medicinal Biotechnology (IMB), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), NO.1 Tiantan Xili, Beijing, 100050, China
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P.R. China
| | - Chengyue Zhang
- Department of Ophthalmology, Beijing Children's Hospital affiliated with Capital Medical University, National Center for Children's Health, Beijing, 100045, China.
| | - Li Zhang
- State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Biotechnology for Microbial Drugs, Department of Oncology, Institute of Medicinal Biotechnology (IMB), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), NO.1 Tiantan Xili, Beijing, 100050, China
| | - Hong-Juan Yao
- State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Biotechnology for Microbial Drugs, Department of Oncology, Institute of Medicinal Biotechnology (IMB), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), NO.1 Tiantan Xili, Beijing, 100050, China
| | - Xiaohong Liu
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, No.5 BeiXianGe St., Beijing, 100053, China
| | - Yuchen Shi
- Dongzhimen Hospital, Beijing University of Chinese Medicine, No.5 Haiyuncang, Beijing, 100700, China
| | - Junyang Zhao
- Department of Ophthalmology, Beijing Children's Hospital affiliated with Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Xiaochen Bo
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P.R. China
| | - Hebing Chen
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P.R. China.
| | - Liang Li
- State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Biotechnology for Microbial Drugs, Department of Oncology, Institute of Medicinal Biotechnology (IMB), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), NO.1 Tiantan Xili, Beijing, 100050, China.
| |
Collapse
|
20
|
Tanaka A, Ogawa M, Zhou Y, Namba K, Hendrickson RC, Miele MM, Li Z, Klimstra DS, Buckley PG, Gulcher J, Wang JY, Roehrl MHA. Proteogenomic characterization of primary colorectal cancer and metastatic progression identifies proteome-based subtypes and signatures. Cell Rep 2024; 43:113810. [PMID: 38377004 PMCID: PMC11288375 DOI: 10.1016/j.celrep.2024.113810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 10/26/2023] [Accepted: 02/01/2024] [Indexed: 02/22/2024] Open
Abstract
Metastatic progression of colorectal adenocarcinoma (CRC) remains poorly understood and poses significant challenges for treatment. To overcome these challenges, we performed multiomics analyses of primary CRC and liver metastases. Genomic alterations, such as structural variants or copy number alterations, were enriched in oncogenes and tumor suppressor genes and increased in metastases. Unsupervised mass spectrometry-based proteomics of 135 primary and 123 metastatic CRCs uncovered distinct proteomic subtypes, three each for primary and metastatic CRCs, respectively. Integrated analyses revealed that hypoxia, stemness, and immune signatures characterize these 6 subtypes. Hypoxic CRC harbors high epithelial-to-mesenchymal transition features and metabolic adaptation. CRC with a stemness signature shows high oncogenic pathway activation and alternative telomere lengthening (ALT) phenotype, especially in metastatic lesions. Tumor microenvironment analysis shows immune evasion via modulation of major histocompatibility complex (MHC) class I/II and antigen processing pathways. This study characterizes both primary and metastatic CRCs and provides a large proteogenomics dataset of metastatic progression.
Collapse
Affiliation(s)
- Atsushi Tanaka
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Makiko Ogawa
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yihua Zhou
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; ICU Department, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Kei Namba
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Ronald C Hendrickson
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Matthew M Miele
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zhuoning Li
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David S Klimstra
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Paige.AI, New York, NY, USA
| | | | | | | | - Michael H A Roehrl
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
21
|
Lee S, Kim J, Ohn JH. Exploring quantitative traits-associated copy number deletions through reanalysis of UK10K consortium whole genome sequencing cohorts. BMC Genomics 2023; 24:787. [PMID: 38110883 PMCID: PMC10729411 DOI: 10.1186/s12864-023-09903-3] [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: 08/01/2023] [Accepted: 12/13/2023] [Indexed: 12/20/2023] Open
Abstract
OBJECTIVES We performed comprehensive association analyses of common high-confidence gnomAD-reported copy number deletions (CNDs) with 60 quantitative traits from UK10K consortium WGS data. METHODS The study made use of data generated by the UK10K Consortium. UK10K consortium WGS data consist of TwinsUK (n = 1754, middle-aged females) and ALSPAC (n = 1867, birth to adolescence) cohorts. UK10K consortium called 18,739 CNDs (hg19) with GenomeSTRiP software. After filtering out variants with minor allele frequency < 0.05 or HWE P < 1.0 × 10- 6, 1222 (TwinsUK) and 1211 (ALSPAC) CNDs remained for association analyses with 60 normalized quantitative traits. RESULTS We identified 23 genome-wide significant associations at 13 loci, among which 2 associations reached experiment-wide significance. We found that two common deletions in chromosome 4, located between WDR1 and ZNF518B (23.3 kb, dbVar ID:nssv15888957, 4:10211262-10,234,569 and 9.8 kb, dbVar ID:nssv15888975, 4:10392422-10,402,191), were associated with uric acid levels (P = 5.23 × 10- 11 and 2.29 × 10- 8, respectively). We also discovered a novel deletion spanning chromosome 18 (823 bp, dbVar ID: nssv15841628, 8:74347187-74,348,010) associated with low HDL cholesterol levels (P = 4.15 × 10- 7). Additionally, we observed two red blood cell traits-associated loci with genome-wide significance, a 13.2 kb deletion in 7q22.1 (nssv15922542) and a 3.7 kb deletion in 12q24.12 (nssv15813226), both of which were located in regions previously reported to be associated with red blood cell traits. Two deletions in 11q11 (nssv15803200 and nssv15802240), where clusters of multiple olfactory receptor genes exist, and a deletion (nssv15929560) upstream to DOCK5 were associated with childhood obesity. Finally, when defining Trait-Associated copy number Deletions (TADs) as CNDs with phenotype associations at sub-threshold significance (P < 10- 3), we identified 157 (97.5%) out of 161 TADs in non-coding regions, with a mean size of 4 kb (range: 209 - 47,942 bp). CONCLUSION We conducted a reanalysis of the UK10K Whole Genome Sequencing cohort, which led to the identification of multiple high confidence copy number deletions associated with quantitative traits. These deletions have standard dbVar IDs and replicate previous findings, as well as reveal novel loci that require further replication studies.
Collapse
Affiliation(s)
- Sejoon Lee
- Precision Medicine Center, Future Innovation Research Division, Seoul National University Bundang Hospital, 173-82, Gumi-ro, Bundang-gu, Seongnam, Gyeonggi-do, 13620, South Korea
- Department of Pathology, Seoul National University Bundang Hospital, 173-82, Gumi-ro, Bundang-gu, Seongnam, Gyeonggi-do, 13620, South Korea
| | - Jinho Kim
- Precision Medicine Center, Future Innovation Research Division, Seoul National University Bundang Hospital, 173-82, Gumi-ro, Bundang-gu, Seongnam, Gyeonggi-do, 13620, South Korea
- Department of Laboratory Medicine, Seoul National University Bundang Hospital, 173-82, Gumi-ro, Bundang-gu, Seongnam, Gyeonggi-do, 13620, South Korea
| | - Jung Hun Ohn
- Precision Medicine Center, Future Innovation Research Division, Seoul National University Bundang Hospital, 173-82, Gumi-ro, Bundang-gu, Seongnam, Gyeonggi-do, 13620, South Korea.
- Department of Internal Medicine, Seoul National University Bundang Hospital, 173-82, Gumi-ro, Bundang-gu, Seongnam, Gyeonggi-do, 13620, South Korea.
- Department of Internal Medicine, College of Medicine, Seoul National University, 103, Daehak-ro, Jongno-gu, Seoul, 03080, South Korea.
| |
Collapse
|
22
|
Xu Z, Li Q, Marchionni L, Wang K. PhenoSV: interpretable phenotype-aware model for the prioritization of genes affected by structural variants. Nat Commun 2023; 14:7805. [PMID: 38016949 PMCID: PMC10684511 DOI: 10.1038/s41467-023-43651-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 11/15/2023] [Indexed: 11/30/2023] Open
Abstract
Structural variants (SVs) represent a major source of genetic variation associated with phenotypic diversity and disease susceptibility. While long-read sequencing can discover over 20,000 SVs per human genome, interpreting their functional consequences remains challenging. Existing methods for identifying disease-related SVs focus on deletion/duplication only and cannot prioritize individual genes affected by SVs, especially for noncoding SVs. Here, we introduce PhenoSV, a phenotype-aware machine-learning model that interprets all major types of SVs and genes affected. PhenoSV segments and annotates SVs with diverse genomic features and employs a transformer-based architecture to predict their impacts under a multiple-instance learning framework. With phenotype information, PhenoSV further utilizes gene-phenotype associations to prioritize phenotype-related SVs. Evaluation on extensive human SV datasets covering all SV types demonstrates PhenoSV's superior performance over competing methods. Applications in diseases suggest that PhenoSV can determine disease-related genes from SVs. A web server and a command-line tool for PhenoSV are available at https://phenosv.wglab.org .
Collapse
Affiliation(s)
- Zhuoran Xu
- Graduate Group in Genomics and Computational Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Quan Li
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, M5G2C1, Canada
| | - Luigi Marchionni
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Kai Wang
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
23
|
Martin-Broto J, Martinez-Garcia J, Moura DS, Redondo A, Gutierrez A, Lopez-Pousa A, Martinez-Trufero J, Sevilla I, Diaz-Beveridge R, Solis-Hernandez MP, Carnero A, Perez M, Marcilla D, Garcia-Foncillas J, Romero P, Fernandez-Jara J, Lopez-Lopez D, Arribas I, Hindi N. Phase II trial of CDK4/6 inhibitor palbociclib in advanced sarcoma based on mRNA expression of CDK4/ CDKN2A. Signal Transduct Target Ther 2023; 8:405. [PMID: 37875500 PMCID: PMC10598203 DOI: 10.1038/s41392-023-01661-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/06/2023] [Accepted: 09/16/2023] [Indexed: 10/26/2023] Open
Abstract
Cyclin-dependent kinases 4 and 6 (CDK4/6) inhibitors demonstrated activity in terms of progression-free survival (PFS) in advanced dedifferentiated liposarcoma (DD-LPS), a sarcoma with CDK4 amplification. CDK4 overexpression is by far more common than amplification in sarcomas and it might be a rational target for CDK inhibitors. Preclinical investigators of this study found that CDK4 overexpression, while not of CDKN2A, was the most consistent predictive factor for palbociclib efficacy in sarcomas. Advanced adult-type soft-tissue sarcoma, excluding DD-LPS, or bone sarcoma patients, progressing after at least one systemic line, whose tumors overexpressed CDK4, but not CDKN2A at baseline biopsy, were accrued in this single-arm phase II trial (EudraCT number: 2016-004039-19). With the main endpoint of a 6-month PFS rate, 40% was considered promising in this population. Palbociclib was administered orally at 125 mg/day for 21 days in 28-day cycles. A total of 214 patients with 236 CDK4/CDKN2A determinations were assessed for prescreening, archival material (141), and screening, baseline biopsy (95). There were 28 (29%) with favorable mRNA profiles from 95 screened patients at baseline. From 23 enrolled patients, 21 evaluable, the 6-month PFS rate was 29% (95% CI 9-48), and there were 6 patients out of 21 with a PFS longer than 6 months. The median PFS and overall survival were 4.2 (95% CI 3.6-4.8) and 12 (95% CI 8.7-15.4) months, respectively. Translational research showed a significant correlation between CDK4 mRNA and protein expression. Palbociclib was active in a variety of sarcoma subtypes, selected by CDK4/CDKN2A, and deserves further investigation in the sarcoma context.
Collapse
Affiliation(s)
- Javier Martin-Broto
- Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040, Madrid, Spain.
- Medical Oncology Department, Fundación Jimenez Diaz University Hospital, 28040, Madrid, Spain.
- General de Villalba University Hospital, 28400, Madrid, Spain.
| | | | - David S Moura
- Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040, Madrid, Spain
| | - Andres Redondo
- Department of Medical Oncology, Hospital Universitario La Paz-IdiPAZ, P. Castellana, 261, 28046, Madrid, Spain
| | - Antonio Gutierrez
- Hematology Department, University Hospital Son Espases, 07120, Mallorca, Spain
| | | | | | - Isabel Sevilla
- Investigación Clínica y Traslacional en Cáncer/ Instituto de Investigaciones Biomédicas de Malaga (IBIMA)/ Hospitales Universitarios Regional y Virgen de la Victoria de Malaga, Malaga, Spain
| | - Roberto Diaz-Beveridge
- Medical Oncology Department, Hospital Universitari i Politècnic La Fe, 46026, Valencia, Spain
| | | | - Amancio Carnero
- Instituto de Biomedicina de Sevilla (IBiS; HUVR, CSIC, US), 41013, Sevilla, Spain
| | - Marco Perez
- Instituto de Biomedicina de Sevilla (IBiS; HUVR, CSIC, US), 41013, Sevilla, Spain
- Pathology Department, Virgen del Rocio University Hospital, 41013, Sevilla, Spain
| | - David Marcilla
- Pathology Department, Virgen del Rocio University Hospital, 41013, Sevilla, Spain
| | - Jesus Garcia-Foncillas
- Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040, Madrid, Spain
- Medical Oncology Department, Fundación Jimenez Diaz University Hospital, 28040, Madrid, Spain
| | - Pablo Romero
- Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040, Madrid, Spain
| | - Javier Fernandez-Jara
- Radiology Department, Fundación Jimenez Diaz University Hospital, 28040, Madrid, Spain
| | - Daniel Lopez-Lopez
- Instituto de Biomedicina de Sevilla (IBiS; HUVR, CSIC, US), 41013, Sevilla, Spain
- Computational Medicine Platform, Fundación progreso y salud (FPS), Hospital Virgen del Rocío, 41013, Seville, Spain
- Bioinformatics in Rare Diseases (BiER). Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), FPS, Hospital Virgen del Rocio, Sevilla, Spain
| | - Ivan Arribas
- Universitat de València - ERI-CES, 46010, Valencia, Spain
| | - Nadia Hindi
- Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040, Madrid, Spain
- Medical Oncology Department, Fundación Jimenez Diaz University Hospital, 28040, Madrid, Spain
- General de Villalba University Hospital, 28400, Madrid, Spain
| |
Collapse
|
24
|
Abdi M, Aliyev E, Trost B, Kohailan M, Aamer W, Syed N, Shaath R, Gandhi GD, Engchuan W, Howe J, Thiruvahindrapuram B, Geng M, Whitney J, Syed A, Lakshmi J, Hussein S, Albashir N, Hussein A, Poggiolini I, Elhag SF, Palaniswamy S, Kambouris M, de Fatima Janjua M, Tahir MOE, Nazeer A, Shahwar D, Azeem MW, Mokrab Y, Aati NA, Akil A, Scherer SW, Kamal M, Fakhro KA. Genomic architecture of autism spectrum disorder in Qatar: The BARAKA-Qatar Study. Genome Med 2023; 15:81. [PMID: 37805537 PMCID: PMC10560429 DOI: 10.1186/s13073-023-01228-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 09/04/2023] [Indexed: 10/09/2023] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a neurodevelopmental condition characterized by impaired social and communication skills, restricted interests, and repetitive behaviors. The prevalence of ASD among children in Qatar was recently estimated to be 1.1%, though the genetic architecture underlying ASD both in Qatar and the greater Middle East has been largely unexplored. Here, we describe the first genomic data release from the BARAKA-Qatar Study-a nationwide program building a broadly consented biorepository of individuals with ASD and their families available for sample and data sharing and multi-omics research. METHODS In this first release, we present a comprehensive analysis of whole-genome sequencing (WGS) data of the first 100 families (372 individuals), investigating the genetic architecture, including single-nucleotide variants (SNVs), copy number variants (CNVs), tandem repeat expansions (TREs), as well as mitochondrial DNA variants (mtDNA) segregating with ASD in local families. RESULTS Overall, we identify potentially pathogenic variants in known genes or regions in 27 out of 100 families (27%), of which 11 variants (40.7%) were classified as pathogenic or likely-pathogenic based on American College of Medical Genetics (ACMG) guidelines. Dominant variants, including de novo and inherited, contributed to 15 (55.6%) of these families, consisting of SNVs/indels (66.7%), CNVs (13.3%), TREs (13.3%), and mtDNA variants (6.7%). Moreover, homozygous variants were found in 7 families (25.9%), with a sixfold increase in homozygous burden in consanguineous versus non-consanguineous families (13.6% and 1.8%, respectively). Furthermore, 28 novel ASD candidate genes were identified in 20 families, 23 of which had recurrent hits in MSSNG and SSC cohorts. CONCLUSIONS This study illustrates the value of ASD studies in under-represented populations and the importance of WGS as a comprehensive tool for establishing a molecular diagnosis for families with ASD. Moreover, it uncovers a significant role for recessive variation in ASD architecture in consanguineous settings and provides a unique resource of Middle Eastern genomes for future research to the global ASD community.
Collapse
Affiliation(s)
- Mona Abdi
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
- Department of Genetics, Sidra Medicine, Doha, Qatar
| | - Elbay Aliyev
- Department of Genetics, Sidra Medicine, Doha, Qatar
| | - Brett Trost
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON, Canada
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | | | - Waleed Aamer
- Department of Genetics, Sidra Medicine, Doha, Qatar
| | - Najeeb Syed
- Genomics Data Science Core, Sidra Medicine, Doha, Qatar
| | - Rulan Shaath
- Department of Genetics, Sidra Medicine, Doha, Qatar
| | | | - Worrawat Engchuan
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON, Canada
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Jennifer Howe
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON, Canada
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Bhooma Thiruvahindrapuram
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON, Canada
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Melissa Geng
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Joe Whitney
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON, Canada
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Amira Syed
- Department of Genetics, Sidra Medicine, Doha, Qatar
| | | | - Sura Hussein
- Department of Genetics, Sidra Medicine, Doha, Qatar
| | | | - Amal Hussein
- Department of Genetics, Sidra Medicine, Doha, Qatar
| | | | - Saba F Elhag
- Department of Genetics, Sidra Medicine, Doha, Qatar
- Hamad Medical Corporation, Doha, Qatar
| | | | - Marios Kambouris
- Pathology and Laboratory Medicine Department, Genetics Division, Sidra Medicine, Doha, Qatar
| | | | | | - Ahsan Nazeer
- Department of Psychiatry, Sidra Medicine, Doha, Qatar
- Weill Cornell Medicine, Doha, Qatar
| | - Durre Shahwar
- Department of Psychiatry, Sidra Medicine, Doha, Qatar
- Weill Cornell Medicine, Doha, Qatar
| | - Muhammad Waqar Azeem
- Department of Psychiatry, Sidra Medicine, Doha, Qatar
- Weill Cornell Medicine, Doha, Qatar
| | - Younes Mokrab
- Department of Genetics, Sidra Medicine, Doha, Qatar
- Department of Genetic Medicine, Weill Cornell Medicine, Doha, Qatar
- Qatar University, Doha, Qatar
| | | | - Ammira Akil
- Department of Genetics, Sidra Medicine, Doha, Qatar
| | - Stephen W Scherer
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON, Canada
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- McLaughlin Centre, University of Toronto, Toronto, ON, Canada
| | - Madeeha Kamal
- Department of Pediatrics, Sidra Medicine, Doha, Qatar
| | - Khalid A Fakhro
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar.
- Department of Genetics, Sidra Medicine, Doha, Qatar.
- Department of Genetic Medicine, Weill Cornell Medicine, Doha, Qatar.
| |
Collapse
|
25
|
Antinucci M, Comas D, Calafell F. Population history modulates the fitness effects of Copy Number Variation in the Roma. Hum Genet 2023; 142:1327-1343. [PMID: 37311904 PMCID: PMC10449987 DOI: 10.1007/s00439-023-02579-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/02/2023] [Indexed: 06/15/2023]
Abstract
We provide the first whole genome Copy Number Variant (CNV) study addressing Roma, along with reference populations from South Asia, the Middle East and Europe. Using CNV calling software for short-read sequence data, we identified 3171 deletions and 489 duplications. Taking into account the known population history of the Roma, as inferred from whole genome nucleotide variation, we could discern how this history has shaped CNV variation. As expected, patterns of deletion variation, but not duplication, in the Roma followed those obtained from single nucleotide polymorphisms (SNPs). Reduced effective population size resulting in slightly relaxed natural selection may explain our observation of an increase in intronic (but not exonic) deletions within Loss of Function (LoF)-intolerant genes. Over-representation analysis for LoF-intolerant gene sets hosting intronic deletions highlights a substantial accumulation of shared biological processes in Roma, intriguingly related to signaling, nervous system and development features, which may be related to the known profile of private disease in the population. Finally, we show the link between deletions and known trait-related SNPs reported in the genome-wide association study (GWAS) catalog, which exhibited even frequency distributions among the studied populations. This suggests that, in general human populations, the strong association between deletions and SNPs associated to biomedical conditions and traits could be widespread across continental populations, reflecting a common background of potentially disease/trait-related CNVs.
Collapse
Affiliation(s)
- Marco Antinucci
- Institute of Evolutionary Biology (UPF-CSIC), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - David Comas
- Institute of Evolutionary Biology (UPF-CSIC), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Francesc Calafell
- Institute of Evolutionary Biology (UPF-CSIC), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain.
| |
Collapse
|
26
|
Grossi A, Rusmini M, Cusano R, Massidda M, Santamaria G, Napoli F, Angelelli A, Fava D, Uva P, Ceccherini I, Maghnie M. Whole genome sequencing in ROHHAD trios proved inconclusive: what's beyond? Front Genet 2023; 14:1031074. [PMID: 37609037 PMCID: PMC10440434 DOI: 10.3389/fgene.2023.1031074] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 07/27/2023] [Indexed: 08/24/2023] Open
Abstract
Rapid-onset Obesity with Hypothalamic dysfunction, Hypoventilation and Autonomic Dysregulation (ROHHAD) is a rare, life-threatening, pediatric disorder of unknown etiology, whose diagnosis is made difficult by poor knowledge of clinical manifestation, and lack of any confirmatory tests. Children with ROHHAD usually present with rapid onset weight gain which may be followed, over months or years, by hypothalamic dysfunction, hypoventilation, autonomic dysfunction, including impaired bowel motility, and tumors of neural crest origin. Despite the lack of evidence of inheritance in ROHHAD, several studies have been conducted in recent years that have explored possible genetic origins, with unsuccessful results. In order to broaden the search for possible genetic risk factors, an attempt was made to analyse the non-coding variants in two trios (proband with parents), recruited in the Gaslini Children's Hospital in Genoa (Italy). Both patients were females, with a typical history of ROHHAD. Gene variants (single nucleotide variants, short insertions/deletions, splice variants or in tandem expansion of homopolymeric tracts) or altered genomic regions (copy number variations or structural variants) shared between the two probands were searched. Currently, we have not found any potentially pathogenic changes, consistent with the ROHHAD clinical phenotype, and involving genes, regions or pathways shared between the two trios. To definitively rule out the genetic etiology, third-generation sequencing technologies (e.g., long-reads sequencing, optical mapping) should be applied, as well as other pathways, including those associated with immunological and autoimmune disorders, should be explored, making use not only of genomics but also of different -omic datasets.
Collapse
Affiliation(s)
- A. Grossi
- Laboratory of Genetics and Genomics of Rare Diseases, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - M. Rusmini
- Laboratory of Genetics and Genomics of Rare Diseases, IRCCS Istituto Giannina Gaslini, Genova, Italy
- Clinical Bioinformatics, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - R. Cusano
- CRS4, Science and Technology Park Polaris, Pula, Italy
| | - M. Massidda
- CRS4, Science and Technology Park Polaris, Pula, Italy
| | - G. Santamaria
- Laboratory of Genetics and Genomics of Rare Diseases, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - F. Napoli
- Pediatric Clinic and Endocrinology, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - A. Angelelli
- D.I.N.O.G.M.I, Università degli Studi di Genova, Genova, Italy
| | - D. Fava
- D.I.N.O.G.M.I, Università degli Studi di Genova, Genova, Italy
| | - P. Uva
- Clinical Bioinformatics, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - I. Ceccherini
- Laboratory of Genetics and Genomics of Rare Diseases, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - M. Maghnie
- Pediatric Clinic and Endocrinology, IRCCS Istituto Giannina Gaslini, Genova, Italy
- D.I.N.O.G.M.I, Università degli Studi di Genova, Genova, Italy
| |
Collapse
|
27
|
Fernández-Suárez E, González-del Pozo M, García-Núñez A, Méndez-Vidal C, Martín-Sánchez M, Mejías-Carrasco JM, Ramos-Jiménez M, Morillo-Sánchez MJ, Rodríguez-de la Rúa E, Borrego S, Antiñolo G. Expanding the phenotype of THRB: a range of macular dystrophies as the major clinical manifestations in patients with a dominant splicing variant. Front Cell Dev Biol 2023; 11:1197744. [PMID: 37547476 PMCID: PMC10401274 DOI: 10.3389/fcell.2023.1197744] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 06/27/2023] [Indexed: 08/08/2023] Open
Abstract
Inherited retinal dystrophies (IRDs) are a clinically and genetically heterogeneous group of disorders that often severely impair vision. Some patients manifest poor central vision as the first symptom due to cone-dysfunction, which is consistent with cone dystrophy (COD), Stargardt disease (STGD), or macular dystrophy (MD) among others. Here, we aimed to identify the genetic cause of autosomal dominant COD in one family. WGS was performed in 3 affected and 1 unaffected individual using the TruSeq Nano DNA library kit and the NovaSeq 6,000 platform (Illumina). Data analysis identified a novel spliceogenic variant (c.283 + 1G>A) in the thyroid hormone receptor beta gene (THRB) as the candidate disease-associated variant. Further genetic analysis revealed the presence of the same heterozygous variant segregating in two additional unrelated dominant pedigrees including 9 affected individuals with a diagnosis of COD (1), STGD (4), MD (3) and unclear phenotype (1). THRB has been previously reported as a causal gene for autosomal dominant and recessive thyroid hormone resistance syndrome beta (RTHβ); however, none of the IRD patients exhibited RTHβ. Genotype-phenotype correlations showed that RTHβ can be caused by both truncating and missense variants, which are mainly located at the 3' (C-terminal/ligand-binding) region, which is common to both THRB isoforms (TRβ1 and TRβ2). In contrast, the c.283 + 1G>A variant is predicted to disrupt a splice site in the 5'-region of the gene that encodes the N-terminal domain of the TRβ1 isoform protein, leaving the TRβ2 isoform intact, which would explain the phenotypic variability observed between RTHβ and IRD patients. Interestingly, although monochromacy or cone response alterations have already been described in a few RTHβ patients, herein we report the first genetic association between a pathogenic variant in THRB and non-syndromic IRDs. We thereby expand the phenotype of THRB pathogenic variants including COD, STGD, or MD as the main clinical manifestation, which also reflects the extraordinary complexity of retinal functions mediated by the different THRB isoforms.
Collapse
Affiliation(s)
- Elena Fernández-Suárez
- Department of Maternofetal Medicine, Genetics and Reproduction, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/Spanish National Research Council (CSIC)/University of Seville, Seville, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Seville, Spain
| | - María González-del Pozo
- Department of Maternofetal Medicine, Genetics and Reproduction, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/Spanish National Research Council (CSIC)/University of Seville, Seville, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Seville, Spain
| | - Alejandro García-Núñez
- Department of Maternofetal Medicine, Genetics and Reproduction, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/Spanish National Research Council (CSIC)/University of Seville, Seville, Spain
| | - Cristina Méndez-Vidal
- Department of Maternofetal Medicine, Genetics and Reproduction, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/Spanish National Research Council (CSIC)/University of Seville, Seville, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Seville, Spain
| | - Marta Martín-Sánchez
- Department of Maternofetal Medicine, Genetics and Reproduction, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/Spanish National Research Council (CSIC)/University of Seville, Seville, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Seville, Spain
| | - José Manuel Mejías-Carrasco
- Department of Maternofetal Medicine, Genetics and Reproduction, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/Spanish National Research Council (CSIC)/University of Seville, Seville, Spain
| | - Manuel Ramos-Jiménez
- Department of Clinical Neurophysiology, University Hospital Virgen Macarena, Seville, Spain
| | | | - Enrique Rodríguez-de la Rúa
- Department of Ophthalmology, University Hospital Virgen Macarena, Seville, Spain
- RETICS Patología Ocular, OFTARED, Instituto de Salud Carlos III, Madrid, Spain
| | - Salud Borrego
- Department of Maternofetal Medicine, Genetics and Reproduction, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/Spanish National Research Council (CSIC)/University of Seville, Seville, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Seville, Spain
| | - Guillermo Antiñolo
- Department of Maternofetal Medicine, Genetics and Reproduction, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/Spanish National Research Council (CSIC)/University of Seville, Seville, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Seville, Spain
| |
Collapse
|
28
|
Liu X, Xu W, Leng F, Zhang P, Guo R, Zhang Y, Hao C, Ni X, Li W. NeuroCNVscore: a tissue-specific framework to prioritise the pathogenicity of CNVs in neurodevelopmental disorders. BMJ Paediatr Open 2023; 7:e001966. [PMID: 37407247 DOI: 10.1136/bmjpo-2023-001966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 06/15/2023] [Indexed: 07/07/2023] Open
Abstract
BACKGROUND Neurodevelopmental disorders (NDDs) are associated with altered development of the brain especially in childhood. Copy number variants (CNVs) play a crucial role in the genetic aetiology of NDDs by disturbing gene expression directly at linear sequence or remotely at three-dimensional genome level in a tissue-specific manner. Despite the substantial increase in NDD studies employing whole-genome sequencing, there is no specific tool for prioritising the pathogenicity of CNVs in the context of NDDs. METHODS Using an XGBoost classifier, we integrated 189 features that represent genomic sequences, gene information and functional/genomic segments for evaluating genome-wide CNVs in a neuro/brain-specific manner, to develop a new tool, neuroCNVscore. We used Human Phenotype Ontology to construct an independent NDD-related set. RESULTS Our neuroCNVscore framework (https://github.com/lxsbch/neuroCNVscore) achieved high predictive performance (precision recall=0.82; area under curve=0.85) and outperformed an existing reference method SVScore. Notably, the predicted pathogenic CNVs showed enrichment in known genes associated with autism. CONCLUSIONS NeuroCNVscore prioritises functional, deleterious and pathogenic CNVs in NDDs at whole genome-wide level, which is important for genetic studies and clinical genomic screening of NDDs as well as for providing novel biological insights into NDDs.
Collapse
Affiliation(s)
- Xuanshi Liu
- Beijing Children's Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, Beijing, China
- MOE Key Laboratory of Major Diseaseas in Children, Beijing, China
- Genetics and Birth Defects Control Centre, National Centre for Children's Health, Beijing, China
| | - Wenjian Xu
- Beijing Children's Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, Beijing, China
- MOE Key Laboratory of Major Diseaseas in Children, Beijing, China
- Genetics and Birth Defects Control Centre, National Centre for Children's Health, Beijing, China
| | - Fei Leng
- Beijing Children's Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, Beijing, China
- MOE Key Laboratory of Major Diseaseas in Children, Beijing, China
- Genetics and Birth Defects Control Centre, National Centre for Children's Health, Beijing, China
| | - Peng Zhang
- Beijing Children's Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, Beijing, China
- MOE Key Laboratory of Major Diseaseas in Children, Beijing, China
- Genetics and Birth Defects Control Centre, National Centre for Children's Health, Beijing, China
| | - Ruolan Guo
- Beijing Children's Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, Beijing, China
- MOE Key Laboratory of Major Diseaseas in Children, Beijing, China
- Genetics and Birth Defects Control Centre, National Centre for Children's Health, Beijing, China
| | - Yue Zhang
- Beijing Children's Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, Beijing, China
- MOE Key Laboratory of Major Diseaseas in Children, Beijing, China
- Genetics and Birth Defects Control Centre, National Centre for Children's Health, Beijing, China
| | - Chanjuan Hao
- Beijing Children's Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, Beijing, China
- MOE Key Laboratory of Major Diseaseas in Children, Beijing, China
- Genetics and Birth Defects Control Centre, National Centre for Children's Health, Beijing, China
| | - Xin Ni
- Beijing Children's Hospital, Capital Medical University, Beijing, China
- MOE Key Laboratory of Major Diseaseas in Children, Beijing, China
- National Centre for Children's Health, Beijing, China
| | - Wei Li
- Beijing Children's Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, Beijing, China
- MOE Key Laboratory of Major Diseaseas in Children, Beijing, China
- Genetics and Birth Defects Control Centre, National Centre for Children's Health, Beijing, China
| |
Collapse
|
29
|
Ravel JM, Renaud M, Muller J, Becker A, Renard É, Remen T, Lefort G, Dexheimer M, Jonveaux P, Leheup B, Bonnet C, Lambert L. Clinical utility of periodic reinterpretation of CNVs of uncertain significance: an 8-year retrospective study. Genome Med 2023; 15:39. [PMID: 37221613 DOI: 10.1186/s13073-023-01191-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 05/15/2023] [Indexed: 05/25/2023] Open
Abstract
BACKGROUND Array-CGH is the first-tier genetic test both in pre- and postnatal developmental disorders worldwide. Variants of uncertain significance (VUS) represent around 10~15% of reported copy number variants (CNVs). Even though VUS reanalysis has become usual in practice, no long-term study regarding CNV reinterpretation has been reported. METHODS This retrospective study examined 1641 CGH arrays performed over 8 years (2010-2017) to demonstrate the contribution of periodically re-analyzing CNVs of uncertain significance. CNVs were classified using AnnotSV on the one hand and manually curated on the other hand. The classification was based on the 2020 American College of Medical Genetics (ACMG) criteria. RESULTS Of the 1641 array-CGH analyzed, 259 (15.7%) showed at least one CNV initially reported as of uncertain significance. After reinterpretation, 106 of the 259 patients (40.9%) changed categories, and 12 of 259 (4.6%) had a VUS reclassified to likely pathogenic or pathogenic. Six were predisposing factors for neurodevelopmental disorder/autism spectrum disorder (ASD). CNV type (gain or loss) does not seem to impact the reclassification rate, unlike the length of the CNV: 75% of CNVs downgraded to benign or likely benign are less than 500 kb in size. CONCLUSIONS This study's high rate of reinterpretation suggests that CNV interpretation has rapidly evolved since 2010, thanks to the continuous enrichment of available databases. The reinterpreted CNV explained the phenotype for ten patients, leading to optimal genetic counseling. These findings suggest that CNVs should be reinterpreted at least every 2 years.
Collapse
Affiliation(s)
- Jean-Marie Ravel
- Service de génétique médicale, CHRU de Nancy, Nancy, France
- Laboratoire de génétique médicale, CHRU Nancy, Nancy, France
- Université de Lorraine, NGERE, F-54000Nancy, Inserm, France
| | - Mathilde Renaud
- Service de génétique médicale, CHRU de Nancy, Nancy, France
- Université de Lorraine, NGERE, F-54000Nancy, Inserm, France
| | - Jean Muller
- Laboratoires de Diagnostic Génétique, Institut de Génétique Médicale d'Alsace (IGMA), Hôpitaux Universitaires de Strasbourg, Strasbourg, France
- Laboratoire de Génétique Médicale, INSERM, UMRS_1112, Institut de Génétique Médicale d'Alsace (IGMA), Université de Strasbourg Faculté de Médecine de Strasbourg, 67000, Strasbourg, France
- Unité Fonctionnelle de Bioinformatique Médicale Appliquée au Diagnostic (UF7363), Hôpitaux Universitaires de Strasbourg, 67000, Strasbourg, France
| | - Aurélie Becker
- Laboratoire de génétique médicale, CHRU Nancy, Nancy, France
| | - Émeline Renard
- Department of pediatrics, Regional University Hospital of Nancy, Allée du Morvan, 54511, Vandoeuvre-Lès-Nancy, France
| | | | | | | | | | - Bruno Leheup
- Service de génétique médicale, CHRU de Nancy, Nancy, France
- Université de Lorraine, NGERE, F-54000Nancy, Inserm, France
| | - Céline Bonnet
- Laboratoire de génétique médicale, CHRU Nancy, Nancy, France.
- Université de Lorraine, NGERE, F-54000Nancy, Inserm, France.
| | - Laëtitia Lambert
- Service de génétique médicale, CHRU de Nancy, Nancy, France.
- Université de Lorraine, NGERE, F-54000Nancy, Inserm, France.
| |
Collapse
|
30
|
Geoffroy V, Lamouche JB, Guignard T, Nicaise S, Kress A, Scheidecker S, Le Béchec A, Muller J. The AnnotSV webserver in 2023: updated visualization and ranking. Nucleic Acids Res 2023:7175348. [PMID: 37216590 DOI: 10.1093/nar/gkad426] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 04/20/2023] [Accepted: 05/09/2023] [Indexed: 05/24/2023] Open
Abstract
Much of the human genetics variant repertoire is composed of single nucleotide variants (SNV) and small insertion/deletions (indel) but structural variants (SV) remain a major part of our modified DNA. SV detection has often been a complex question to answer either because of the necessity to use different technologies (array CGH, SNP array, Karyotype, Optical Genome Mapping…) to detect each category of SV or to get an appropriate resolution (Whole Genome Sequencing). Thanks to the deluge of pangenomic analysis, Human geneticists are accumulating SV and their interpretation remains time consuming and challenging. The AnnotSV webserver (https://www.lbgi.fr/AnnotSV/) aims at being an efficient tool to (i) annotate and interpret SV potential pathogenicity in the context of human diseases, (ii) recognize potential false positive variants from all the SV identified and (iii) visualize the patient variants repertoire. The most recent developments in the AnnotSV webserver are: (i) updated annotations sources and ranking, (ii) three novel output formats to allow diverse utilization (analysis, pipelines), as well as (iii) two novel user interfaces including an interactive circos view.
Collapse
Affiliation(s)
- Véronique Geoffroy
- Université de Brest, Inserm, EFS, UMR 1078, GGB, F-29200 Brest, France
- Laboratoire de Génétique Médicale, UMR 1112, INSERM, IGMA, Université de Strasbourg, Strasbourg, France
| | - Jean-Baptiste Lamouche
- Laboratoire de Génétique Médicale, UMR 1112, INSERM, IGMA, Université de Strasbourg, Strasbourg, France
- Unité Fonctionnelle de Bioinformatique Médicale appliquée au diagnostic (UF7363), Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | | | - Samuel Nicaise
- Unité Fonctionnelle de Bioinformatique Médicale appliquée au diagnostic (UF7363), Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Arnaud Kress
- Complex Systems and Translational Bioinformatics, ICube, UMR 7357, University of Strasbourg, CNRS, FMTS, Strasbourg, France
| | - Sophie Scheidecker
- Laboratoire de Génétique Médicale, UMR 1112, INSERM, IGMA, Université de Strasbourg, Strasbourg, France
- Laboratoires de Diagnostic Génétique, IGMA, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Antony Le Béchec
- Unité Fonctionnelle de Bioinformatique Médicale appliquée au diagnostic (UF7363), Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Jean Muller
- Laboratoire de Génétique Médicale, UMR 1112, INSERM, IGMA, Université de Strasbourg, Strasbourg, France
- Unité Fonctionnelle de Bioinformatique Médicale appliquée au diagnostic (UF7363), Hôpitaux Universitaires de Strasbourg, Strasbourg, France
- Laboratoires de Diagnostic Génétique, IGMA, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| |
Collapse
|
31
|
Spillmann RC, Tan QKG, Reuter C, Schoch K, Kohler J, Bonner D, Zastrow D, Alkelai A, Baugh E, Cope H, Marwaha S, Wheeler MT, Bernstein JA, Shashi V. A concurrent dual analysis of genomic data augments diagnoses: Experiences of 2 clinical sites in the Undiagnosed Diseases Network. Genet Med 2023; 25:100353. [PMID: 36481303 PMCID: PMC10506157 DOI: 10.1016/j.gim.2022.12.001] [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: 08/02/2022] [Revised: 11/28/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Next-generation sequencing (NGS) has revolutionized the diagnostic process for rare/ultrarare conditions. However, diagnosis rates differ between analytical pipelines. In the National Institutes of Health-Undiagnosed Diseases Network (UDN) study, each individual's NGS data are concurrently analyzed by the UDN sequencing core laboratory and the clinical sites. We examined the outcomes of this practice. METHODS A retrospective review was performed at 2 UDN clinical sites to compare the variants and diagnoses/candidate genes identified with the dual analyses of the NGS data. RESULTS In total, 95 individuals had 100 diagnoses/candidate genes. There was 59% concordance between the UDN sequencing core laboratories and the clinical sites in identifying diagnoses/candidate genes. The core laboratory provided more diagnoses, whereas the clinical sites prioritized more research variants/candidate genes (P < .001). The clinical sites solely identified 15% of the diagnoses/candidate genes. The differences between the 2 pipelines were more often because of variant prioritization disparities than variant detection. CONCLUSION The unique dual analysis of NGS data in the UDN synergistically enhances outcomes. The core laboratory provided a clinical analysis with more diagnoses and the clinical sites prioritized more research variants/candidate genes. Implementing such concurrent dual analyses in other genomic research studies and clinical settings can improve both variant detection and prioritization.
Collapse
Affiliation(s)
- Rebecca C Spillmann
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC
| | - Queenie K-G Tan
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC
| | - Chloe Reuter
- Stanford Center for Inherited Cardiovascular Disease, Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA; Stanford Center for Undiagnosed Diseases, Stanford University, and Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Kelly Schoch
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC
| | - Jennefer Kohler
- Stanford Center for Undiagnosed Diseases, Stanford University, and Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Devon Bonner
- Stanford Center for Undiagnosed Diseases, Stanford University, and Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Diane Zastrow
- Stanford Center for Undiagnosed Diseases, Stanford University, and Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Anna Alkelai
- Institute for Genome Medicine, Columbia University Medical Center, New York, NY
| | - Evan Baugh
- Institute for Genome Medicine, Columbia University Medical Center, New York, NY
| | - Heidi Cope
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC
| | - Shruti Marwaha
- Stanford Center for Undiagnosed Diseases, Stanford University, and Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Matthew T Wheeler
- Stanford Center for Inherited Cardiovascular Disease, Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA; Stanford Center for Undiagnosed Diseases, Stanford University, and Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Jonathan A Bernstein
- Stanford Center for Undiagnosed Diseases, Stanford University, and Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Vandana Shashi
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC.
| |
Collapse
|
32
|
Al-Kurbi AA, Aliyev E, AlSa’afin S, Aamer W, Palaniswamy S, Al-Maraghi A, Kilani H, Akil AAS, Stotland MA, Fakhro KA. A Complex Intrachromosomal Rearrangement Disrupting IRF6 in a Family with Popliteal Pterygium and Van der Woude Syndromes. Genes (Basel) 2023; 14:genes14040849. [PMID: 37107607 PMCID: PMC10137688 DOI: 10.3390/genes14040849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/26/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Clefts of the lip and/or palate (CL/P) are considered the most common form of congenital anomalies occurring either in isolation or in association with other clinical features. Van der woude syndrome (VWS) is associated with about 2% of all CL/P cases and is further characterized by having lower lip pits. Popliteal pterygium syndrome (PPS) is a more severe form of VWS, normally characterized by orofacial clefts, lower lip pits, skin webbing, skeletal anomalies and syndactyly of toes and fingers. Both syndromes are inherited in an autosomal dominant manner, usually caused by heterozygous mutations in the Interferon Regulatory Factor 6 (IRF6) gene. Here we report the case of a two-generation family where the index presented with popliteal pterygium syndrome while both the father and sister had clinical features of van der woude syndrome, but without any point mutations detected by re-sequencing of known gene panels or microarray testing. Using whole genome sequencing (WGS) followed by local de novo assembly, we discover and validate a copy-neutral, 429 kb complex intra-chromosomal rearrangement in the long arm of chromosome 1, disrupting the IRF6 gene. This variant is copy-neutral, novel against publicly available databases, and segregates in the family in an autosomal dominant pattern. This finding suggests that missing heritability in rare diseases may be due to complex genomic rearrangements that can be resolved by WGS and de novo assembly, helping deliver answers to patients where no genetic etiology was identified by other means.
Collapse
Affiliation(s)
- Alya A. Al-Kurbi
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha 34110, Qatar
- Department of Human Genetics, Sidra Medicine, Doha 26999, Qatar
| | - Elbay Aliyev
- Department of Human Genetics, Sidra Medicine, Doha 26999, Qatar
| | - Sana AlSa’afin
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha 34110, Qatar
| | - Waleed Aamer
- Department of Human Genetics, Sidra Medicine, Doha 26999, Qatar
| | | | | | - Houda Kilani
- Division of Plastic and Craniofacial Surgery, Sidra Medicine, Doha 26999, Qatar
| | | | - Mitchell A. Stotland
- Division of Plastic and Craniofacial Surgery, Sidra Medicine, Doha 26999, Qatar
- Department of Surgery, Weill Cornell Medical College, Doha 24144, Qatar
| | - Khalid A. Fakhro
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha 34110, Qatar
- Department of Human Genetics, Sidra Medicine, Doha 26999, Qatar
- Department of Genetic Medicine, Weill Cornell Medical College, Doha 24144, Qatar
- Correspondence:
| |
Collapse
|
33
|
Flerlage JE, Myers JR, Maciaszek JL, Oak N, Rashkin SR, Hui Y, Wang YD, Chen W, Wu G, Chang TC, Hamilton K, Tithi SS, Goldin LR, Rotunno M, Caporaso N, Vogt A, Flamish D, Wyatt K, Liu J, Tucker M, Hahn CN, Brown AL, Scott HS, Mullighan C, Nichols KE, Metzger ML, McMaster ML, Yang JJ, Rampersaud E. Discovery of novel predisposing coding and noncoding variants in familial Hodgkin lymphoma. Blood 2023; 141:1293-1307. [PMID: 35977101 PMCID: PMC10082357 DOI: 10.1182/blood.2022016056] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 07/12/2022] [Accepted: 08/02/2022] [Indexed: 11/20/2022] Open
Abstract
Familial aggregation of Hodgkin lymphoma (HL) has been demonstrated in large population studies, pointing to genetic predisposition to this hematological malignancy. To understand the genetic variants associated with the development of HL, we performed whole genome sequencing on 234 individuals with and without HL from 36 pedigrees that had 2 or more first-degree relatives with HL. Our pedigree selection criteria also required at least 1 affected individual aged <21 years, with the median age at diagnosis of 21.98 years (3-55 years). Family-based segregation analysis was performed for the identification of coding and noncoding variants using linkage and filtering approaches. Using our tiered variant prioritization algorithm, we identified 44 HL-risk variants in 28 pedigrees, of which 33 are coding and 11 are noncoding. The top 4 recurrent risk variants are a coding variant in KDR (rs56302315), a 5' untranslated region variant in KLHDC8B (rs387906223), a noncoding variant in an intron of PAX5 (rs147081110), and another noncoding variant in an intron of GATA3 (rs3824666). A newly identified splice variant in KDR (c.3849-2A>C) was observed for 1 pedigree, and high-confidence stop-gain variants affecting IRF7 (p.W238∗) and EEF2KMT (p.K116∗) were also observed. Multiple truncating variants in POLR1E were found in 3 independent pedigrees as well. Whereas KDR and KLHDC8B have previously been reported, PAX5, GATA3, IRF7, EEF2KMT, and POLR1E represent novel observations. Although there may be environmental factors influencing lymphomagenesis, we observed segregation of candidate germline variants likely to predispose HL in most of the pedigrees studied.
Collapse
Affiliation(s)
- Jamie E. Flerlage
- Department of Oncology, St. Jude Children’s Research Hospital and the University of Tennessee Health Sciences Center, Memphis, TN
| | - Jason R. Myers
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Jamie L. Maciaszek
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Ninad Oak
- Department of Oncology, St. Jude Children’s Research Hospital and the University of Tennessee Health Sciences Center, Memphis, TN
| | - Sara R. Rashkin
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Yawei Hui
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Yong-Dong Wang
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN
| | - Wenan Chen
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Gang Wu
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Ti-Cheng Chang
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Kayla Hamilton
- Department of Oncology, St. Jude Children’s Research Hospital and the University of Tennessee Health Sciences Center, Memphis, TN
| | - Saima S. Tithi
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Lynn R. Goldin
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Melissa Rotunno
- Division of Cancer Control and Population Sciences, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Neil Caporaso
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | | | | | | | - Jia Liu
- Leidos Biomedical, Inc, Frederick, MD
| | - Margaret Tucker
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Christopher N. Hahn
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Anna L. Brown
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Hamish S. Scott
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Charles Mullighan
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Kim E. Nichols
- Department of Oncology, St. Jude Children’s Research Hospital and the University of Tennessee Health Sciences Center, Memphis, TN
| | - Monika L. Metzger
- Department of Oncology, St. Jude Children’s Research Hospital and the University of Tennessee Health Sciences Center, Memphis, TN
- Department of Global Pediatric Medicine, St. Jude Children’s Research Hospital, Memphis, TN
| | - Mary L. McMaster
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN
| | - Jun J. Yang
- Department of Oncology, St. Jude Children’s Research Hospital and the University of Tennessee Health Sciences Center, Memphis, TN
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN
| | - Evadnie Rampersaud
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, TN
| |
Collapse
|
34
|
Privitera F, Trusso MA, Valentino F, Doddato G, Fallerini C, Brunelli G, D'Aurizio R, Furini S, Goracci A, Fagiolini A, Mari F, Renieri A, Ariani F. Heterozygosity for neuronal ceroid lipofuscinosis predisposes to bipolar disorder. REVISTA BRASILEIRA DE PSIQUIATRIA (SAO PAULO, BRAZIL : 1999) 2023; 45:11-19. [PMID: 35881528 PMCID: PMC9976914 DOI: 10.47626/1516-4446-2022-2650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 07/13/2022] [Indexed: 11/20/2022]
Abstract
OBJECTIVE Bipolar disorder is a heritable chronic mental disorder that causes psychosocial impairment through depressive/manic episodes. Familial transmission of bipolar disorder does not follow simple Mendelian patterns of inheritance. The aim of this study was to describe a large family with 12 members affected by bipolar disorder. Whole-exome sequencing was performed for eight members, three of whom were diagnosed with bipolar disorder, and another reported as "borderline." METHODS Whole-exome sequencing data allowed us to select variants that the affected members had in common, including and excluding the "borderline" individual with moderate anxiety and obsessive-compulsive traits. RESULTS The results favored designating certain genes as predispositional to bipolar disorder: a heterozygous missense variant in CLN6 resulted in a "borderline" phenotype that, if combined with a heterozygous missense variant in ZNF92, is responsible for the more severe bipolar disorder phenotype. Both rare missense changes are predicted to disrupt protein function. CONCLUSIONS Loss of both alleles in CLN6 causes neuronal ceroid lipofuscinosis, a severe progressive childhood neurological disorder. Our results indicate that heterozygous CLN6 carriers, previously reported as healthy, may be susceptible to bipolar disorder later in life if associated with additional variants in ZNF92.
Collapse
Affiliation(s)
- Flavia Privitera
- Medical Genetics, University of Siena, Italy. Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Italy
| | - Maria A Trusso
- Department of Molecular Medicine and Development, University of Siena, Siena, Italy
| | - Floriana Valentino
- Medical Genetics, University of Siena, Italy. Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Italy
| | - Gabriella Doddato
- Medical Genetics, University of Siena, Italy. Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Italy
| | - Chiara Fallerini
- Medical Genetics, University of Siena, Italy. Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Italy
| | - Giulia Brunelli
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Italy
| | - Romina D'Aurizio
- Institute of Informatics and Telematics, National Research Council, Pisa, Italy
| | - Simone Furini
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Italy
| | - Arianna Goracci
- Department of Molecular Medicine and Development, University of Siena, Siena, Italy. Department of Mental Health; Psychiatry Unit, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Andrea Fagiolini
- Department of Molecular Medicine and Development, University of Siena, Siena, Italy
| | - Francesca Mari
- Medical Genetics, University of Siena, Italy. Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Italy. Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Alessandra Renieri
- Medical Genetics, University of Siena, Italy. Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Italy. Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Francesca Ariani
- Medical Genetics, University of Siena, Italy. Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Italy. Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| |
Collapse
|
35
|
Fabian J, Dworschak GC, Waffenschmidt L, Schierbaum L, Bendixen C, Heilmann-Heimbach S, Sivalingam S, Buness A, Schwarzer N, Boemers TM, Schmiedeke E, Neser J, Leonhardt J, Kosch F, Weih S, Gielen HM, Hosie S, Kabs C, Palta M, Märzheuser S, Bode LM, Lacher M, Schäfer FM, Stehr M, Knorr C, Ure B, Kleine K, Rolle U, Zaniew M, Phillip G, Zwink N, Jenetzky E, Reutter H, Hilger AC. Genome-wide identification of disease-causing copy number variations in 450 individuals with anorectal malformations. Eur J Hum Genet 2023; 31:105-111. [PMID: 36319675 PMCID: PMC9822900 DOI: 10.1038/s41431-022-01216-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 07/03/2022] [Accepted: 10/06/2022] [Indexed: 01/08/2023] Open
Abstract
Anorectal malformations (ARM) represent a spectrum of rare malformations originating from a perturbated development of the embryonic hindgut. Approximately 60% occur as a part of a defined genetic syndrome or within the spectrum of additional congenital anomalies. Rare copy number variations (CNVs) have been associated with both syndromic and non-syndromic forms. The present study represents the largest study to date to explore the contribution of CNVs to the expression of ARMs. SNP-array-based molecular karyotyping was applied in 450 individuals with ARM and 4392 healthy controls. CNVs were identified from raw intensity data using PennCNV. Overlapping CNVs between cases and controls were discarded. Remaining CNVs were filtered using a stringent filter algorithm of nine filter steps. Prioritized CNVs were confirmed using qPCR. Filtering prioritized and qPCR confirmed four microscopic chromosomal anomalies and nine submicroscopic CNVs comprising seven microdeletions (del2p13.2, del4p16.2, del7q31.33, del9p24.1, del16q12.1, del18q32, del22q11.21) and two microduplications (dup2p13.2, dup17q12) in 14 individuals (12 singletons and one affected sib-pair). Within these CNVs, based on their embryonic expression data and function, we suggest FOXK2, LPP, and SALL3 as putative candidate genes. Overall, our CNV analysis identified putative microscopic and submicroscopic chromosomal rearrangements in 3% of cases. Functional characterization and re-sequencing of suggested candidate genes is warranted.
Collapse
Affiliation(s)
- Julia Fabian
- grid.10388.320000 0001 2240 3300Institute of Human Genetics, Medical Faculty of the University Bonn & University Hospital Bonn, Bonn, Germany
| | - Gabriel C. Dworschak
- grid.10388.320000 0001 2240 3300Institute of Human Genetics, Medical Faculty of the University Bonn & University Hospital Bonn, Bonn, Germany ,grid.15090.3d0000 0000 8786 803XDepartment of Neuropediatrics, University Hospital Bonn, Bonn, Germany ,grid.10388.320000 0001 2240 3300Institute of Anatomy, Medical Faculty, University of Bonn, Bonn, Germany
| | - Lea Waffenschmidt
- grid.10388.320000 0001 2240 3300Institute of Human Genetics, Medical Faculty of the University Bonn & University Hospital Bonn, Bonn, Germany
| | - Luca Schierbaum
- grid.10388.320000 0001 2240 3300Institute of Human Genetics, Medical Faculty of the University Bonn & University Hospital Bonn, Bonn, Germany
| | - Charlotte Bendixen
- grid.10388.320000 0001 2240 3300Institute of Human Genetics, Medical Faculty of the University Bonn & University Hospital Bonn, Bonn, Germany ,grid.15090.3d0000 0000 8786 803XUnit of Pediatric Surgery, Department of General, Visceral, Vascular and Thoracic Surgery, University Hospital Bonn, Bonn, Germany
| | - Stefanie Heilmann-Heimbach
- grid.10388.320000 0001 2240 3300Institute of Human Genetics, Medical Faculty of the University Bonn & University Hospital Bonn, Bonn, Germany
| | - Sugirthan Sivalingam
- grid.10388.320000 0001 2240 3300Institute for Medical Biometry, Informatics and Epidemiology, Medical Faculty, University of Bonn, Bonn, Germany ,grid.10388.320000 0001 2240 3300Institute for Genomic Statistics and Bioinformatics, Medical Faculty, University of Bonn, Bonn, Germany ,grid.10388.320000 0001 2240 3300Core Unit for Bioinformatics Data Analysis, Medical Faculty, University of Bonn, Bonn, Germany
| | - Andreas Buness
- grid.10388.320000 0001 2240 3300Institute for Medical Biometry, Informatics and Epidemiology, Medical Faculty, University of Bonn, Bonn, Germany ,grid.10388.320000 0001 2240 3300Institute for Genomic Statistics and Bioinformatics, Medical Faculty, University of Bonn, Bonn, Germany ,grid.10388.320000 0001 2240 3300Core Unit for Bioinformatics Data Analysis, Medical Faculty, University of Bonn, Bonn, Germany
| | - Nicole Schwarzer
- SoMA, The German Patient Support Organization for Anorectal Malformations and Hirschsprung Disease, Munich, Germany
| | - Thomas M. Boemers
- grid.411097.a0000 0000 8852 305XDepartment of Pediatric Surgery and Pediatric Urology, Children’s Hospital of Cologne Amsterdamer Strasse, Cologne, Germany
| | - Eberhard Schmiedeke
- grid.419807.30000 0004 0636 7065Clinic for Pediatric Surgery and Pediatric Urology, Klinikum Bremen Mitte, Bremen, Germany
| | - Jörg Neser
- Department of Pediatric Surgery, General Hospital, Chemnitz, Germany
| | - Johannes Leonhardt
- Department of Pediatric Surgery, Children’s Hospital Braunschweig, Braunschweig, Germany
| | - Ferdinand Kosch
- grid.419594.40000 0004 0391 0800Department of Pediatric Surgery, Städtisches Klinikum Karlsruhe, Karlsruhe, Germany
| | - Sandra Weih
- grid.5963.9Department of Pediatric Surgery, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Helen Maya Gielen
- Department of Pediatric Surgery, Asklepios Klinik Nord Heidberg, Hamburg, Deutschland
| | - Stuart Hosie
- grid.6936.a0000000123222966Muenchen Klinik gGmbH, Muenchen, Klinik Schwabing, Technische Universitaet Muenchen, Munich, Germany
| | - Carmen Kabs
- grid.6936.a0000000123222966Muenchen Klinik gGmbH, Muenchen, Klinik Schwabing, Technische Universitaet Muenchen, Munich, Germany
| | - Markus Palta
- grid.491593.30000 0004 0636 5983Department of Pediatric Surgery, Evangelisches Krankenhaus Hamm, Hamm, Germany
| | - Stefanie Märzheuser
- grid.413108.f0000 0000 9737 0454Department of Pediatric Surgery, Rostock University Medical Center, Rostock, Germany
| | - Lena Marie Bode
- grid.9647.c0000 0004 7669 9786Department of Pediatric Surgery, University of Leipzig, Leipzig, Germany
| | - Martin Lacher
- grid.9647.c0000 0004 7669 9786Department of Pediatric Surgery, University of Leipzig, Leipzig, Germany
| | - Frank-Mattias Schäfer
- grid.490647.8Department of Pediatric Surgery and Pediatric Urology, Cnopfsche Kinderklinik-Klinik Hallerwiese, Nürnberg, Germany
| | - Maximilian Stehr
- grid.490647.8Department of Pediatric Surgery and Pediatric Urology, Cnopfsche Kinderklinik-Klinik Hallerwiese, Nürnberg, Germany
| | - Christian Knorr
- Department of Pediatric Surgery and Orthopedics, University Children’s Hospital Regensburg (KUNO) at the Hospital St. Hedwig of the Order of St. John, Regensburg, Germany
| | - Benno Ure
- grid.10423.340000 0000 9529 9877Center of Pediatric Surgery Hannover, Hannover Medical School, Hannover, Germany
| | - Katharina Kleine
- grid.506180.a0000 0004 0560 0400Department of Pediatric Surgery, Evangelisches Krankenhaus Oberhausen, Oberhausen, Germany
| | - Udo Rolle
- grid.7839.50000 0004 1936 9721Department of Pediatric Surgery and Pediatric Urology, Goethe University Frankfurt, Frankfurt, Germany
| | - Marcin Zaniew
- grid.28048.360000 0001 0711 4236Department of Pediatrics, University of Zielona Góra, Zielona Góra, Poland
| | - Grote Phillip
- grid.7839.50000 0004 1936 9721Institute of Cardiovascular Regeneration, Center for Molecular Medicine, University of Frankfurt, Frankfurt am Main, Germany
| | - Nadine Zwink
- grid.410607.4Department of Child and Adolescent Psychiatry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Ekkehart Jenetzky
- grid.410607.4Department of Child and Adolescent Psychiatry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany ,grid.412581.b0000 0000 9024 6397Faculty of Health, School of Medicine, University of Witten/Herdecke, Witten, Germany
| | - Heiko Reutter
- grid.10388.320000 0001 2240 3300Institute of Human Genetics, Medical Faculty of the University Bonn & University Hospital Bonn, Bonn, Germany ,grid.5330.50000 0001 2107 3311Division of Neonatology and Pediatric Intensive Care, Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander University Nürnberg-Erlangen, Erlangen, Germany
| | - Alina C. Hilger
- grid.10388.320000 0001 2240 3300Institute of Human Genetics, Medical Faculty of the University Bonn & University Hospital Bonn, Bonn, Germany ,grid.5330.50000 0001 2107 3311Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander University Nürnberg-Erlangen, Erlangen, Germany ,grid.411668.c0000 0000 9935 6525Research Center On Rare Kidney Diseases (RECORD), University Hospital Erlangen, 91054 Erlangen, Germany
| |
Collapse
|
36
|
SCIP: software for efficient clinical interpretation of copy number variants detected by whole-genome sequencing. Hum Genet 2023; 142:201-216. [PMID: 36376761 PMCID: PMC9918589 DOI: 10.1007/s00439-022-02494-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 10/09/2022] [Indexed: 11/16/2022]
Abstract
Copy number variants (CNVs) represent major etiologic factors in rare genetic diseases. Current clinical CNV interpretation workflows require extensive back-and-forth with multiple tools and databases. This increases complexity and time burden, potentially resulting in missed genetic diagnoses. We present the Suite for CNV Interpretation and Prioritization (SCIP), a software package for the clinical interpretation of CNVs detected by whole-genome sequencing (WGS). The SCIP Visualization Module near-instantaneously displays all information necessary for CNV interpretation (variant quality, population frequency, inheritance pattern, and clinical relevance) on a single page-supported by modules providing variant filtration and prioritization. SCIP was comprehensively evaluated using WGS data from 1027 families with congenital cardiac disease and/or autism spectrum disorder, containing 187 pathogenic or likely pathogenic (P/LP) CNVs identified in previous curations. SCIP was efficient in filtration and prioritization: a median of just two CNVs per case were selected for review, yet it captured all P/LP findings (92.5% of which ranked 1st). SCIP was also able to identify one pathogenic CNV previously missed. SCIP was benchmarked against AnnotSV and a spreadsheet-based manual workflow and performed superiorly than both. In conclusion, SCIP is a novel software package for efficient clinical CNV interpretation, substantially faster and more accurate than previous tools (available at https://github.com/qd29/SCIP , a video tutorial series is available at https://bit.ly/SCIPVideos ).
Collapse
|
37
|
Talsania K, Shen TW, Chen X, Jaeger E, Li Z, Chen Z, Chen W, Tran B, Kusko R, Wang L, Pang AWC, Yang Z, Choudhari S, Colgan M, Fang LT, Carroll A, Shetty J, Kriga Y, German O, Smirnova T, Liu T, Li J, Kellman B, Hong K, Hastie AR, Natarajan A, Moshrefi A, Granat A, Truong T, Bombardi R, Mankinen V, Meerzaman D, Mason CE, Collins J, Stahlberg E, Xiao C, Wang C, Xiao W, Zhao Y. Structural variant analysis of a cancer reference cell line sample using multiple sequencing technologies. Genome Biol 2022; 23:255. [PMID: 36514120 PMCID: PMC9746098 DOI: 10.1186/s13059-022-02816-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 11/17/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The cancer genome is commonly altered with thousands of structural rearrangements including insertions, deletions, translocation, inversions, duplications, and copy number variations. Thus, structural variant (SV) characterization plays a paramount role in cancer target identification, oncology diagnostics, and personalized medicine. As part of the SEQC2 Consortium effort, the present study established and evaluated a consensus SV call set using a breast cancer reference cell line and matched normal control derived from the same donor, which were used in our companion benchmarking studies as reference samples. RESULTS We systematically investigated somatic SVs in the reference cancer cell line by comparing to a matched normal cell line using multiple NGS platforms including Illumina short-read, 10X Genomics linked reads, PacBio long reads, Oxford Nanopore long reads, and high-throughput chromosome conformation capture (Hi-C). We established a consensus SV call set of a total of 1788 SVs including 717 deletions, 230 duplications, 551 insertions, 133 inversions, 146 translocations, and 11 breakends for the reference cancer cell line. To independently evaluate and cross-validate the accuracy of our consensus SV call set, we used orthogonal methods including PCR-based validation, Affymetrix arrays, Bionano optical mapping, and identification of fusion genes detected from RNA-seq. We evaluated the strengths and weaknesses of each NGS technology for SV determination, and our findings provide an actionable guide to improve cancer genome SV detection sensitivity and accuracy. CONCLUSIONS A high-confidence consensus SV call set was established for the reference cancer cell line. A large subset of the variants identified was validated by multiple orthogonal methods.
Collapse
Affiliation(s)
- Keyur Talsania
- Sequencing Facility Bioinformatics Group, Advanced Biomedical and Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
- Bioinformatics and Computational Science Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Tsai-Wei Shen
- Sequencing Facility Bioinformatics Group, Advanced Biomedical and Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
- Bioinformatics and Computational Science Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Xiongfong Chen
- Sequencing Facility Bioinformatics Group, Advanced Biomedical and Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
- Bioinformatics and Computational Science Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | | | - Zhipan Li
- Sentieon Inc, Mountain View, CA, USA
| | - Zhong Chen
- Center for Genomics, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Wanqiu Chen
- Center for Genomics, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Bao Tran
- Sequencing Facility, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | | | - Limin Wang
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Zhaowei Yang
- Department of Allergy and Clinical Immunology, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Sulbha Choudhari
- Sequencing Facility Bioinformatics Group, Advanced Biomedical and Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
- Bioinformatics and Computational Science Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Michael Colgan
- Center for Drug Evaluation and Research, FDA, Silver Spring, MD, USA
| | - Li Tai Fang
- Bioinformatics Research & Early Development, Roche Sequencing Solutions Inc, 1301 Shoreway Road, Belmont, CA, 94002, USA
| | | | - Jyoti Shetty
- Sequencing Facility, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Yuliya Kriga
- Sequencing Facility, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Oksana German
- Sequencing Facility, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Tatyana Smirnova
- Sequencing Facility, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Tiantain Liu
- Center for Genomics, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Jing Li
- Department of Allergy and Clinical Immunology, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | | | - Karl Hong
- Bionano Genomics, San Diego, CA92121, USA
| | | | | | | | | | | | | | | | - Daoud Meerzaman
- Computational Genomics and Bioinformatics Branch, Center for Biomedical Informatics and Information Technology (CBIIT), National Cancer Institute, Rockville, MD, USA
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Jack Collins
- Sequencing Facility Bioinformatics Group, Advanced Biomedical and Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
- Bioinformatics and Computational Science Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Eric Stahlberg
- Bioinformatics and Computational Science Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Chunlin Xiao
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Charles Wang
- Center for Genomics, Loma Linda University School of Medicine, Loma Linda, CA, USA.
| | - Wenming Xiao
- Center for Drug Evaluation and Research, FDA, Silver Spring, MD, USA.
| | - Yongmei Zhao
- Sequencing Facility Bioinformatics Group, Advanced Biomedical and Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, USA.
- Bioinformatics and Computational Science Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA.
| |
Collapse
|
38
|
Kabiljo R, Bowles H, Marriott H, Jones AR, Bouton CR, Dobson RJ, Quinn JP, Al Khleifat A, Swanson CM, Al-Chalabi A, Iacoangeli A. RetroSnake: A modular pipeline to detect human endogenous retroviruses in genome sequencing data. iScience 2022; 25:105289. [PMID: 36339261 PMCID: PMC9626663 DOI: 10.1016/j.isci.2022.105289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 08/08/2022] [Accepted: 10/04/2022] [Indexed: 12/02/2022] Open
Abstract
Human endogenous retroviruses (HERVs) integrated into the human genome as a result of ancient exogenous infections and currently comprise ∼8% of our genome. The members of the most recently acquired HERV family, HERV-Ks, still retain the potential to produce viral molecules and have been linked to a wide range of diseases including cancer and neurodegeneration. Although a range of tools for HERV detection in NGS data exist, most of them lack wet lab validation and they do not cover all steps of the analysis. Here, we describe RetroSnake, an end-to-end, modular, computationally efficient, and customizable pipeline for the discovery of HERVs in short-read NGS data. RetroSnake is based on an extensively wet-lab validated protocol, it covers all steps of the analysis from raw data to the generation of annotated results presented as an interactive html file, and it is easy to use by life scientists without substantial computational training. Availability and implementation: The Pipeline and an extensive documentation are available on GitHub.
Collapse
Affiliation(s)
- Renata Kabiljo
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London SE5 8AF, UK
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9NU, UK
| | - Harry Bowles
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London SE5 8AF, UK
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9NU, UK
| | - Heather Marriott
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London SE5 8AF, UK
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9NU, UK
| | - Ashley R. Jones
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9NU, UK
| | - Clement R. Bouton
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, UK
| | - Richard J.B. Dobson
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London SE5 8AF, UK
- NIHR Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King’s College London, London, UK
- Institute of Health Informatics, University College London, London, UK
- NIHR Biomedical Research Centre at University College London Hospitals NHS Foundation Trust, London, UK
| | - John P. Quinn
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3BX, UK
| | - Ahmad Al Khleifat
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9NU, UK
| | - Chad M. Swanson
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, London, UK
| | - Ammar Al-Chalabi
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9NU, UK
| | - Alfredo Iacoangeli
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London SE5 8AF, UK
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9NU, UK
- NIHR Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King’s College London, London, UK
| |
Collapse
|
39
|
Piernik M, Brzezinski D, Sztromwasser P, Pacewicz K, Majer-Burman W, Gniot M, Sielski D, Bryzghalov O, Wozna A, Zawadzki P. DBFE: distribution-based feature extraction from structural variants in whole-genome data. Bioinformatics 2022; 38:4466-4473. [PMID: 35929780 DOI: 10.1093/bioinformatics/btac513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 07/12/2022] [Indexed: 12/24/2022] Open
Abstract
MOTIVATION Whole-genome sequencing has revolutionized biosciences by providing tools for constructing complete DNA sequences of individuals. With entire genomes at hand, scientists can pinpoint DNA fragments responsible for oncogenesis and predict patient responses to cancer treatments. Machine learning plays a paramount role in this process. However, the sheer volume of whole-genome data makes it difficult to encode the characteristics of genomic variants as features for learning algorithms. RESULTS In this article, we propose three feature extraction methods that facilitate classifier learning from sets of genomic variants. The core contributions of this work include: (i) strategies for determining features using variant length binning, clustering and density estimation; (ii) a programing library for automating distribution-based feature extraction in machine learning pipelines. The proposed methods have been validated on five real-world datasets using four different classification algorithms and a clustering approach. Experiments on genomes of 219 ovarian, 61 lung and 929 breast cancer patients show that the proposed approaches automatically identify genomic biomarkers associated with cancer subtypes and clinical response to oncological treatment. Finally, we show that the extracted features can be used alongside unsupervised learning methods to analyze genomic samples. AVAILABILITY AND IMPLEMENTATION The source code of the presented algorithms and reproducible experimental scripts are available on Github at https://github.com/MNMdiagnostics/dbfe. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Maciej Piernik
- Institute of Computing Science, Faculty of Computing and Telecommunications, Poznan University of Technology, 60-965 Poznan, Poland.,MNM Bioscience Inc., Cambridge, MA 02142, USA
| | - Dariusz Brzezinski
- Institute of Computing Science, Faculty of Computing and Telecommunications, Poznan University of Technology, 60-965 Poznan, Poland.,MNM Bioscience Inc., Cambridge, MA 02142, USA.,Institute of Bioorganic Chemistry of the Polish Academy of Sciences, 61-704 Poznan, Poland
| | | | | | | | - Michal Gniot
- MNM Bioscience Inc., Cambridge, MA 02142, USA.,Department of Hematology and Bone Marrow Transplantation, Poznan University of Medical Sciences, 60-569 Poznan, Poland
| | | | | | - Alicja Wozna
- MNM Bioscience Inc., Cambridge, MA 02142, USA.,Faculty of Physics, Adam Mickiewicz University, 61-614 Poznan, Poland
| | - Pawel Zawadzki
- MNM Bioscience Inc., Cambridge, MA 02142, USA.,Faculty of Physics, Adam Mickiewicz University, 61-614 Poznan, Poland
| |
Collapse
|
40
|
Quaio CRDAC, Coelho AVC, Moura LMS, Guedes RLM, Chen K, Ceroni JRM, Minillo RM, Caraciolo MP, Reis RDS, de Azevedo BMC, Nobrega MS, Teixeira ACB, Martinelli Lima M, da Mota TR, da Matta MC, Colichio GBC, Roncalho AL, Ferreira AFM, Campilongo GP, Perrone E, Virmond LDA, Moreno CA, Prota JRM, de França M, Cervato MC, de Almeida TF, de Oliveira Filho JB. Genomic study of nonsyndromic hearing loss in unaffected individuals: Frequency of pathogenic and likely pathogenic variants in a Brazilian cohort of 2,097 genomes. Front Genet 2022; 13:921324. [PMID: 36147510 PMCID: PMC9486813 DOI: 10.3389/fgene.2022.921324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
Hearing loss (HL) is a common sensory deficit in humans and represents an important clinical and social burden. We studied whole-genome sequencing data of a cohort of 2,097 individuals from the Brazilian Rare Genomes Project who were unaffected by hearing loss to investigate pathogenic and likely pathogenic variants associated with nonsyndromic hearing loss (NSHL). We found relevant frequencies of individuals harboring these alterations: 222 heterozygotes (10.59%) for sequence variants, 54 heterozygotes (2.58%) for copy-number variants (CNV), and four homozygotes (0.19%) for sequence variants. The top five most frequent genes and their corresponding combined allelic frequencies (AF) were GJB2 (AF = 1.57%), STRC (AF = 1%), OTOA (AF = 0.69%), TMPRSS3 (AF = 0.41%), and OTOF (AF = 0.29%). The most frequent sequence variant was GJB2:c.35del (AF = 0.72%), followed by OTOA:p. (Glu787Ter) (AF = 0.61%), while the most recurrent CNV was a microdeletion of 57.9 kb involving the STRC gene (AF = 0.91%). An important fraction of these individuals (n = 104; 4.96%) presented variants associated with autosomal dominant forms of NSHL, which may imply the development of some hearing impairment in the future. Using data from the heterozygous individuals for recessive forms and the Hardy–Weinberg equation, we estimated the population frequency of affected individuals with autosomal recessive NSHL to be 1:2,222. Considering that the overall prevalence of HL in adults ranges from 4–15% worldwide, our data indicate that an important fraction of this condition may be associated with a monogenic origin and dominant inheritance.
Collapse
Affiliation(s)
- Caio Robledo D’ Angioli Costa Quaio
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
- Instituto da Criança (Children’s Hospital), Hospital Das Clínicas HCFMUSP, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- *Correspondence: Caio Robledo D’ Angioli Costa Quaio, ; Joao Bosco de Oliveira Filho,
| | | | - Livia Maria Silva Moura
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
- VarsOmics, Sociedade Beneficente Israelita Brasileira Albert Einstein, São Paulo, SP, Brazil
| | - Rafael Lucas Muniz Guedes
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
- VarsOmics, Sociedade Beneficente Israelita Brasileira Albert Einstein, São Paulo, SP, Brazil
| | - Kelin Chen
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | | | | | - Marcel Pinheiro Caraciolo
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
- VarsOmics, Sociedade Beneficente Israelita Brasileira Albert Einstein, São Paulo, SP, Brazil
| | - Rodrigo de Souza Reis
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
- VarsOmics, Sociedade Beneficente Israelita Brasileira Albert Einstein, São Paulo, SP, Brazil
| | | | | | | | | | - Thamara Rayssa da Mota
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
- Programa de Pós Graduação em Tecnologias Energéticas e Nucleares (PROTEN), UFPE, Recife, Brazil
| | | | | | | | | | | | - Eduardo Perrone
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
- Departamento de Morfologia e Genética, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | | | - Carolina Araujo Moreno
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
- Departamento de Medicina Translacional, Área de Genética Médica e Medicina Genômica, Faculdade de Ciências Médicas, Universidade Estadual de Campinas, Campinas, SP, Brazil
| | - Joana Rosa Marques Prota
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
- Departamento de Medicina Translacional, Área de Genética Médica e Medicina Genômica, Faculdade de Ciências Médicas, Universidade Estadual de Campinas, Campinas, SP, Brazil
| | | | - Murilo Castro Cervato
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
- VarsOmics, Sociedade Beneficente Israelita Brasileira Albert Einstein, São Paulo, SP, Brazil
| | | | - Joao Bosco de Oliveira Filho
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
- *Correspondence: Caio Robledo D’ Angioli Costa Quaio, ; Joao Bosco de Oliveira Filho,
| |
Collapse
|
41
|
Jacobsen JOB, Kelly C, Cipriani V, Research Consortium GE, Mungall CJ, Reese J, Danis D, Robinson PN, Smedley D. Phenotype-driven approaches to enhance variant prioritization and diagnosis of rare disease. Hum Mutat 2022; 43:1071-1081. [PMID: 35391505 PMCID: PMC9288531 DOI: 10.1002/humu.24380] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 01/25/2022] [Accepted: 04/03/2022] [Indexed: 11/20/2022]
Abstract
Rare disease diagnostics and disease gene discovery have been revolutionized by whole-exome and genome sequencing but identifying the causative variant(s) from the millions in each individual remains challenging. The use of deep phenotyping of patients and reference genotype-phenotype knowledge, alongside variant data such as allele frequency, segregation, and predicted pathogenicity, has proved an effective strategy to tackle this issue. Here we review the numerous tools that have been developed to automate this approach and demonstrate the power of such an approach on several thousand diagnosed cases from the 100,000 Genomes Project. Finally, we discuss the challenges that need to be overcome if we are going to improve detection rates and help the majority of patients that still remain without a molecular diagnosis after state-of-the-art genomic interpretation.
Collapse
Affiliation(s)
- Julius O. B. Jacobsen
- William Harvey Research Institute, Charterhouse Square, Barts and the London School of Medicine and Dentistry QueenQueen Mary University of LondonLondonUK
| | - Catherine Kelly
- William Harvey Research Institute, Charterhouse Square, Barts and the London School of Medicine and Dentistry QueenQueen Mary University of LondonLondonUK
| | - Valentina Cipriani
- William Harvey Research Institute, Charterhouse Square, Barts and the London School of Medicine and Dentistry QueenQueen Mary University of LondonLondonUK
| | | | - Christopher J. Mungall
- Environmental Genomics and Systems Biology, Lawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| | - Justin Reese
- Environmental Genomics and Systems Biology, Lawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| | - Daniel Danis
- The Jackson Laboratory for Genomic MedicineFarmingtonConnecticutUSA
| | | | - Damian Smedley
- William Harvey Research Institute, Charterhouse Square, Barts and the London School of Medicine and Dentistry QueenQueen Mary University of LondonLondonUK
| |
Collapse
|
42
|
D’Aurizio R, Catona O, Pitasi M, Li YE, Ren B, Nicolis SK. Bridging between Mouse and Human Enhancer-Promoter Long-Range Interactions in Neural Stem Cells, to Understand Enhancer Function in Neurodevelopmental Disease. Int J Mol Sci 2022; 23:7964. [PMID: 35887306 PMCID: PMC9322198 DOI: 10.3390/ijms23147964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/08/2022] [Accepted: 07/14/2022] [Indexed: 11/16/2022] Open
Abstract
Non-coding variation in complex human disease has been well established by genome-wide association studies, and is thought to involve regulatory elements, such as enhancers, whose variation affects the expression of the gene responsible for the disease. The regulatory elements often lie far from the gene they regulate, or within introns of genes differing from the regulated gene, making it difficult to identify the gene whose function is affected by a given enhancer variation. Enhancers are connected to their target gene promoters via long-range physical interactions (loops). In our study, we re-mapped, onto the human genome, more than 10,000 enhancers connected to promoters via long-range interactions, that we had previously identified in mouse brain-derived neural stem cells by RNApolII-ChIA-PET analysis, coupled to ChIP-seq mapping of DNA/chromatin regions carrying epigenetic enhancer marks. These interactions are thought to be functionally relevant. We discovered, in the human genome, thousands of DNA regions syntenic with the interacting mouse DNA regions (enhancers and connected promoters). We further annotated these human regions regarding their overlap with sequence variants (single nucleotide polymorphisms, SNPs; copy number variants, CNVs), that were previously associated with neurodevelopmental disease in humans. We document various cases in which the genetic variant, associated in humans to neurodevelopmental disease, affects an enhancer involved in long-range interactions: SNPs, previously identified by genome-wide association studies to be associated with schizophrenia, bipolar disorder, and intelligence, are located within our human syntenic enhancers, and alter transcription factor recognition sites. Similarly, CNVs associated to autism spectrum disease and other neurodevelopmental disorders overlap with our human syntenic enhancers. Some of these enhancers are connected (in mice) to homologs of genes already associated to the human disease, strengthening the hypothesis that the gene is indeed involved in the disease. Other enhancers are connected to genes not previously associated with the disease, pointing to their possible pathogenetic involvement. Our observations provide a resource for further exploration of neural disease, in parallel with the now widespread genome-wide identification of DNA variants in patients with neural disease.
Collapse
Affiliation(s)
- Romina D’Aurizio
- Institute of Informatics and Telematics (IIT), National Research Council (CNR), 56124 Pisa, Italy;
| | - Orazio Catona
- Institute of Informatics and Telematics (IIT), National Research Council (CNR), 56124 Pisa, Italy;
| | - Mattia Pitasi
- Dipartimento di Biotecnologie e Bioscienze, University of Milano-Bicocca, 20126 Milano, Italy; (M.P.); (S.K.N.)
| | - Yang Eric Li
- University of California San Diego, La Jolla, CA 92093, USA; (Y.E.L.); (B.R.)
| | - Bing Ren
- University of California San Diego, La Jolla, CA 92093, USA; (Y.E.L.); (B.R.)
| | - Silvia Kirsten Nicolis
- Dipartimento di Biotecnologie e Bioscienze, University of Milano-Bicocca, 20126 Milano, Italy; (M.P.); (S.K.N.)
| |
Collapse
|
43
|
Lin D, Du H, Zhao S, Liu B, Song H, Wang G, Zhang W, Liang H, Liu P, Liu C, Han W, Li Z, Yang Y, Chen S, Zhao L, Li X, Wu Z, Qiu G, Wu Z, Zhang TJ, Wu N, Wang S, Liu J, Liu S, Zuo Y, Liu G, Yu C, Liu L, Shao J, Zhao S, Yan Z, Zhao H, Niu Y, Li X, Wang H, Ma C, Chen Z, Liu B, Cheng X, Lin J, Du H, Li Y, Song S, Tian W, Xie Z, Zhao Z, Zhao L, Zhao Z, Zheng Z, Huang Y, Sun N, Wu N. Phenotype expansion of variants affecting p38 MAPK signaling in hypospadias patients. Orphanet J Rare Dis 2022; 17:209. [PMID: 35606856 PMCID: PMC9128137 DOI: 10.1186/s13023-022-02334-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 04/26/2022] [Indexed: 11/10/2022] Open
Abstract
Background Hypospadias is a congenital anomaly of the male urogenital system. Genetics factors play an important role in its pathogenesis. To search for potential causal genes/variants for hypospadias, we performed exome sequencing in a pedigree with three patients across two generations and a cohort of 49 sporadic patients with hypospadias. Results A novel BRAF variant (NM_004333.6: c.362C > A) was found to co-segregate with the hypospadias phenotype in the disease pedigree. In cells overexpressing the BRAF mutant, the phosphorylation level of p38 MAPK was significantly increased as compared with the cells overexpressing the wild-type BRAF or RASopathy-related BRAF mutant. This variant further led to a reduced transcription level of the SRY gene, which is essential for the normal development of the male reproductive system. In the cohort of sporadic patients, we identified two additional variants in p38 MAPK signaling-related genes (TRIM67 and DAB2IP) potentially associated with hypospadias. Conclusion Our study expands the phenotypic spectrum of variants affecting p38 MAPK signaling toward the involvement of hypospadias.
Collapse
|
44
|
Danis D, Jacobsen JOB, Balachandran P, Zhu Q, Yilmaz F, Reese J, Haimel M, Lyon GJ, Helbig I, Mungall CJ, Beck CR, Lee C, Smedley D, Robinson PN. SvAnna: efficient and accurate pathogenicity prediction of coding and regulatory structural variants in long-read genome sequencing. Genome Med 2022; 14:44. [PMID: 35484572 PMCID: PMC9047340 DOI: 10.1186/s13073-022-01046-6] [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: 08/17/2021] [Accepted: 04/12/2022] [Indexed: 01/18/2023] Open
Abstract
Structural variants (SVs) are implicated in the etiology of Mendelian diseases but have been systematically underascertained owing to sequencing technology limitations. Long-read sequencing enables comprehensive detection of SVs, but approaches for prioritization of candidate SVs are needed. Structural variant Annotation and analysis (SvAnna) assesses all classes of SVs and their intersection with transcripts and regulatory sequences, relating predicted effects on gene function with clinical phenotype data. SvAnna places 87% of deleterious SVs in the top ten ranks. The interpretable prioritizations offered by SvAnna will facilitate the widespread adoption of long-read sequencing in diagnostic genomics. SvAnna is available at https://github.com/TheJacksonLaboratory/SvAnn a .
Collapse
Affiliation(s)
- Daniel Danis
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Julius O B Jacobsen
- William Harvey Research Institute, Charterhouse Square, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | | | - Qihui Zhu
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Feyza Yilmaz
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Justin Reese
- Division of Environmental Genomics and Systems Biology, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Matthias Haimel
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
- St. Anna Children's Cancer Research Institute, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Present address: Global Computational Biology and Digital Sciences, Boehringer Ingelheim Regional Center Vienna GmbH & Co KG, 1120, Vienna, Austria
| | - Gholson J Lyon
- Department of Human Genetics, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, USA
- Biology PhD Program, The Graduate Center, The City University of New York, New York, USA
| | - Ingo Helbig
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- The Epilepsy NeuroGenetics Initiative (ENGIN), Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Biomedical and Health Informatics (DBHi), Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher J Mungall
- Division of Environmental Genomics and Systems Biology, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Christine R Beck
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
- Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT, 06032, USA
- Institute for Systems Genomics, University of Connecticut, Storrs, CT, 06269, USA
| | - Charles Lee
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Damian Smedley
- William Harvey Research Institute, Charterhouse Square, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK.
| | - Peter N Robinson
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA.
- Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT, 06032, USA.
| |
Collapse
|
45
|
Cisarova K, Garavelli L, Caraffi SG, Peluso F, Valeri L, Gargano G, Gavioli S, Trimarchi G, Neri A, Campos-Xavier B, Superti-Furga A. A monoallelic SEC23A variant E599K associated with cranio-lenticulo-sutural dysplasia. Am J Med Genet A 2021; 188:319-325. [PMID: 34580982 PMCID: PMC9291540 DOI: 10.1002/ajmg.a.62506] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 11/06/2022]
Abstract
Cranio-lenticulo-sutural dysplasia (CLSD; MIM 607812) is a rare or underdiagnosed condition, as only two families have been reported. The original family (Boyadjiev et al., Human Genetics, 2003, 113, 1-9 and Boyadjiev et al., Nature Genetics, 2006, 38, 1192-1197) showed recessive inheritance of the condition with a biallelic SEC23A missense variant in affected individuals. In contrast, another child with sporadic CLSD had a monoallelic SEC23A variant inherited from the reportedly unaffected father (Boyadjiev et al., Clinical Genetics, 2011, 80, 169-176), raising questions on possible digenism. Here, we report a 2-month-old boy seen because of large fontanels with wide cranial sutures, a large forehead, hypertelorism, a thin nose, a high arched palate, and micrognathia. His mother was clinically unremarkable, while his father had a history of large fontanels in infancy who had closed only around age 10 years; he also had a large forehead, hypertelorism, a thin, beaked nose and was operated for bilateral glaucoma with exfoliation of the lens capsule. Trio genome sequencing and familial segregation revealed a monoallelic c.1795G > A transition in SEC23A that was de novo in the father and transmitted to the proband. The variant predicts a nonconservative substitution (p.E599K) in an ultra-conserved residue that is seen in 3D models of yeast SEC23 to be involved in direct binding between SEC23 and SAR1 subunits of the coat protein complex II coat. This observation confirms the link between SEC23A variants and CLSD but suggests that in addition to the recessive inheritance described in the original family, SEC23A variants may result in dominant inheritance of CLSD, possibly by a dominant-negative disruptive effect on the SEC23 multimer.
Collapse
Affiliation(s)
- Katarina Cisarova
- Division of Genetic Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Livia Garavelli
- Clinical Genetics Unit, Azienda USL-IRCCS of Reggio Emilia, Reggio Emilia, Italy
| | | | - Francesca Peluso
- Clinical Genetics Unit, Azienda USL-IRCCS of Reggio Emilia, Reggio Emilia, Italy
| | - Lara Valeri
- Clinical Genetics Unit, Azienda USL-IRCCS of Reggio Emilia, Reggio Emilia, Italy
| | - Giancarlo Gargano
- Neonatal Intensive Care Unit, Azienda USL-IRCCS of Reggio Emilia, Reggio Emilia, Italy
| | - Sara Gavioli
- Neonatal Intensive Care Unit, Azienda USL-IRCCS of Reggio Emilia, Reggio Emilia, Italy
| | - Gabriele Trimarchi
- Clinical Genetics Unit, Azienda USL-IRCCS of Reggio Emilia, Reggio Emilia, Italy
| | - Alberto Neri
- Ophthalmology Unit, Department of Surgery, Azienda USL-IRCCS of Reggio Emilia, Reggio Emilia, Italy
| | - Belinda Campos-Xavier
- Division of Genetic Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Andrea Superti-Furga
- Division of Genetic Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|