1
|
Muffels IJ, Carter T, Rehmann H, Vastert SJ, Verrijn Stuart AA, Blank AC, Garde A, van der Zwaag B, De Lange IM, Giltay JC, van Gassen KL, Koop K, Asensio CS, van Hasselt PM. LIMK1 variants are associated with divergent endocrinological phenotypes and altered exocytosis dynamics. iScience 2025; 28:112585. [PMID: 40491492 PMCID: PMC12146536 DOI: 10.1016/j.isci.2025.112585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 10/11/2024] [Accepted: 04/30/2025] [Indexed: 06/11/2025] Open
Abstract
LIM kinase 1 (LIMK1) plays a pivotal role in dynamic actin remodeling through phosphorylation of cofilin, thereby regulating exocytosis. We report two individuals harboring LIMK1 de novo variants with dissimilar phenotypes: one exhibited epileptic encephalopathy and developmental delay, while the other showed common variable immune deficiency and glucose dysregulation. We suspected that the divergent phenotypic features arose from opposing effects on LIMK1 activity. Indeed, actin polymerization was significantly decreased in individual 1, whereas it was increased in individual 2. Insulin-secreting cell lines expressing the LIMK1 variant of individual 1 exhibited significantly slower exocytosis, contrasting the rapid and uncontrolled exocytosis in individual 2. Intriguingly, both variants led to increased overall insulin secretion. This first report of two individuals with LIMK1 variants with divergent effects on cofilin phosphorylation and actin polymerization, reveals that LIMK1 has an important role in tuned insulin exocytosis. These distinct exocytosis defects may underlie the glucose dysregulation observed.
Collapse
Affiliation(s)
- Irena J.J. Muffels
- Department of Metabolic Diseases, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Theodore Carter
- Department of Biological Sciences, College of Natural Sciences and Mathematics, Denver, CO, USA
| | - Holger Rehmann
- Department of Energy and Biotechnology, Flensburg University of Applied Sciences, Flensburg, Germany
| | - Sebastiaan J. Vastert
- Department of Pediatric Rheumatology and Immunology and Center for Translational Immunology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Annemarie A. Verrijn Stuart
- Department of Pediatric Endocrinology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Andreas C. Blank
- Department of Pediatric Cardiology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Aurore Garde
- Centre de Génétique et Centre de Référence Maladies Rares, Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (FHU TRANSLAD), Hôpital d'Enfants, Centre Hospitalier Universitaire de Dijon, Dijon, France
| | - Bert van der Zwaag
- Department of Medical Genetics, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Iris M. De Lange
- Department of Medical Genetics, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jacques C. Giltay
- Department of Medical Genetics, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Koen L.I. van Gassen
- Department of Medical Genetics, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Klaas Koop
- Department of Metabolic Diseases, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Cedric S. Asensio
- Department of Biological Sciences, College of Natural Sciences and Mathematics, Denver, CO, USA
| | - Peter M. van Hasselt
- Department of Metabolic Diseases, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
2
|
Jeon H, Lee JL, Shim H, Joe S, Byeon I, Kim CW, Lim SB, Park IJ, Yoon YS, Chu HBK, Kim YJ, Yu CS, Yang JO. Genetic variations and recurrence in stage III Korean colorectal cancer: Insights from tumor-only mutation analysis. PLoS One 2025; 20:e0323302. [PMID: 40408428 PMCID: PMC12101642 DOI: 10.1371/journal.pone.0323302] [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: 12/16/2024] [Accepted: 04/06/2025] [Indexed: 05/25/2025] Open
Abstract
Colorectal cancer (CRC) has the second highest incidence rate among all cancers in Korea, with approximately 30% of patients with regional CRC experiencing recurrence. Understanding the genetic drivers of recurrence is essential for early detection and targeted treatment. Therefore, many studies have focused on genetic analysis using tumor-normal matched samples, as this approach provides more comprehensive insights. However, tumor-only samples are far more common in clinical practice because of the difficulty in obtaining normal tissues, making developing robust methods for analyzing tumor-only data a pressing need. This study aimed to investigate the genetic variations associated with CRC recurrence using tumor-only whole-exome sequencing data from 200 Korean patients with stage III CRC. By applying stringent filtering using public databases including Genome Aggregation Database (gnomAD), Exome Aggregation Consortium (ExAC), Single Nucleotide Polymorphism Database (dbSNP), 1000 Genomes Project (1000G), Korean Variant Archive 2 (KOVA2), and Korean Reference Genome Database (KRGDB), we identified 221 statistically significant mutations across 195 genes with distinct distributions between the recurrence and non-recurrence groups. Furthermore, statistical analysis of the clinical data revealed that the T-category, N-category, and preoperative carcinoembryonic antigen levels were correlated with CRC recurrence. Moreover, we identified nine networks through protein-protein interaction analysis and identified networks with high feature importance. We also developed a CRC recurrence prediction model using PyCaret, which achieved an area under the curve (AUC) of 0.77. Our findings highlight the importance of robust variant filtering in tumor-only sample analyses and provide insights into the genetic landscape of CRC recurrence in the Korean population.
Collapse
Affiliation(s)
- Hajin Jeon
- Korea Bioinformation Center (KOBIC), Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Jong Lyul Lee
- Department of Surgery, Division of Colon and Rectal Surgery, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Republic of Korea
| | - Hyeran Shim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Soobok Joe
- Korea Bioinformation Center (KOBIC), Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Iksu Byeon
- Korea Bioinformation Center (KOBIC), Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Chan Wook Kim
- Department of Surgery, Division of Colon and Rectal Surgery, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Republic of Korea
| | - Seok-Byung Lim
- Department of Surgery, Division of Colon and Rectal Surgery, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Republic of Korea
| | - In Ja Park
- Department of Surgery, Division of Colon and Rectal Surgery, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Republic of Korea
| | - Yong Sik Yoon
- Department of Surgery, Division of Colon and Rectal Surgery, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Republic of Korea
| | - Hoang Bao Khanh Chu
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Young-Joon Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Chang Sik Yu
- Department of Surgery, Division of Colon and Rectal Surgery, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Republic of Korea
| | - Jin Ok Yang
- Korea Bioinformation Center (KOBIC), Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon, Republic of Korea
| |
Collapse
|
3
|
Kadri S, Craven KE, Fussell AM, Gee EPS, Jordan D, Klee EW, Krumm N, Temple-Smolkin RL, Zehir A, Zhang W, Sboner A. Clinical Bioinformatician Body of Knowledge-Bioinformatics and Software Core: A Report of the Association for Molecular Pathology. J Mol Diagn 2025:S1525-1578(25)00118-7. [PMID: 40398560 DOI: 10.1016/j.jmoldx.2025.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 03/07/2025] [Accepted: 04/10/2025] [Indexed: 05/23/2025] Open
Abstract
With the evolution of next-generation sequencing-based testing in molecular diagnostics laboratories, the clinical role of bioinformaticians has also evolved. The Association for Molecular Pathology's Clinical Bioinformatician Body of Knowledge aims to define the various roles the clinical bioinformatician operates individually or within a clinical bioinformatics team, along with proficiencies and skill sets that may be required or desirable across these roles. One of the most common professional responsibilities of a clinical bioinformatician is to implement bioinformatics pipelines, either vendor supplied or custom built for the assays in the molecular diagnostics laboratory, along with analysis and quality control of clinical genomics data. This second article in the series describes the various stages in the life cycle of a clinical bioinformatics pipeline and the considerations, areas of expertise, and skill sets required in each stage. This information may help laboratory professionals to better work with clinical bioinformaticians and laboratory directors to hire the appropriate expertise based on the specific needs of the laboratory.
Collapse
Affiliation(s)
- Sabah Kadri
- AMP Clinical Bioinformatician Body of Knowledge Bioinformatics and Software Knowledge Core Working Group of the Informatics Subdivision, Association for Molecular Pathology, Rockville, Maryland; Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois; AbbVie, North Chicago, Illinois.
| | - Kelly E Craven
- AMP Clinical Bioinformatician Body of Knowledge Bioinformatics and Software Knowledge Core Working Group of the Informatics Subdivision, Association for Molecular Pathology, Rockville, Maryland; Johns Hopkins University School of Medicine, Baltimore, Maryland; Memorial Sloan Kettering Cancer Center, New York, New York
| | - Amber M Fussell
- The Association for Molecular Pathology, Rockville, Maryland
| | - Elaine P S Gee
- AMP Clinical Bioinformatician Body of Knowledge Bioinformatics and Software Knowledge Core Working Group of the Informatics Subdivision, Association for Molecular Pathology, Rockville, Maryland; BigHead Analytics Group, Windsor, California; Mirvie, Inc., South San Francisco, California
| | - Danielle Jordan
- The Association for Molecular Pathology, Rockville, Maryland
| | - Eric W Klee
- AMP Clinical Bioinformatician Body of Knowledge Bioinformatics and Software Knowledge Core Working Group of the Informatics Subdivision, Association for Molecular Pathology, Rockville, Maryland; Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Niklas Krumm
- AMP Clinical Bioinformatician Body of Knowledge Bioinformatics and Software Knowledge Core Working Group of the Informatics Subdivision, Association for Molecular Pathology, Rockville, Maryland; University of Washington, Seattle, Washington
| | | | - Ahmet Zehir
- AMP Clinical Bioinformatician Body of Knowledge Bioinformatics and Software Knowledge Core Working Group of the Informatics Subdivision, Association for Molecular Pathology, Rockville, Maryland; Memorial Sloan Kettering Cancer Center, New York, New York
| | - Weiwei Zhang
- AMP Clinical Bioinformatician Body of Knowledge Bioinformatics and Software Knowledge Core Working Group of the Informatics Subdivision, Association for Molecular Pathology, Rockville, Maryland; Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Andrea Sboner
- AMP Clinical Bioinformatician Body of Knowledge Bioinformatics and Software Knowledge Core Working Group of the Informatics Subdivision, Association for Molecular Pathology, Rockville, Maryland; Department of Pathology and Laboratory Medicine, Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Meyer Cancer Center, Weill Cornell Medicine, New York, New York
| |
Collapse
|
4
|
Wang X, Sun Y, Guan XW, Wang YY, Hong DY, Zhang ZL, Li YH, Yang PY, Jiang T, Xu ZF. Effect of newborn genomic screening for lysosomal storage disorders: a cohort study in China. Genome Med 2025; 17:52. [PMID: 40355959 PMCID: PMC12070756 DOI: 10.1186/s13073-025-01483-z] [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: 08/08/2024] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Lysosomal storage disorders (LSDs) have a relatively high incidence among rare diseases and can lead to severe consequences if not treated promptly. However, many countries and regions have not included these disorders in their newborn screening programs, resulting in missed early detection, underdiagnosis, and delayed treatment. Newborn genomic screening (NBGS) has shown good screening effectiveness for traditional biochemical screening diseases; however, its effectiveness for LSDs has not yet been evaluated in the general newborn population. METHODS To evaluate the outcome of NBGS for LSDs, a cohort study was conducted involving newborns recruited from Nanjing Women and Children's Healthcare Hospital in China from March 18, 2022, to September 21, 2023. All participants underwent NBGS of 15 LSDs (18 genes) via dried blood spots, followed by enzyme activity testing for NBGS-positive individuals. The study calculated the incidence and carrier rates for each LSD though NBGS, as well as the positive screening rate, the false positive rate and the positive predictive value of the screening process. RESULTS Among 22,687 newborns (11,996 males [52.88%]), 1344 (6.0%) were identified as carriers, and 30 (0.13%) were initially positive for LSDs. Of these, 4 were excluded, 15 were diagnosed as LSD-presymptomatic individuals based on enzyme deficiency and pathogenic variants conforming to inheritance patterns, and 11 remain under follow-up. The estimated combined birth incidence of LSDs in Nanjing was 1/1512, primarily including Fabry disease, Krabbe disease, glycogen storage disease type II, Niemann-Pick disease, and mucopolysaccharidosis type II. Rather than directly comparing NBGS and enzyme activity screening, this study evaluated two sequential screening strategies: (1) NBGS-first with reflex enzyme testing and (2) enzyme activity-first with reflex genomic testing. The NBGS-first strategy demonstrated higher sensitivity and specificity, with a significantly lower false positive rate and higher positive predictive values compared to the enzyme-first strategy (P < 0.05). CONCLUSIONS This study highlights the potential of NBGS to enhance early detection of presymptomatic LSD individuals, enabling timely interventions and improving newborn health outcomes. Integrating NBGS into routine newborn screening programs could provide an effective and proactive approach for LSD identification and management.
Collapse
Affiliation(s)
- Xin Wang
- Genetic Medicine Center, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Qinhuai District, 123 Tianfei Lane, Mochou Road, Nanjing, Jiangsu Province, 210004, China
| | - Yun Sun
- Genetic Medicine Center, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Qinhuai District, 123 Tianfei Lane, Mochou Road, Nanjing, Jiangsu Province, 210004, China
| | - Xian-Wei Guan
- Genetic Medicine Center, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Qinhuai District, 123 Tianfei Lane, Mochou Road, Nanjing, Jiangsu Province, 210004, China
| | - Yan-Yun Wang
- Genetic Medicine Center, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Qinhuai District, 123 Tianfei Lane, Mochou Road, Nanjing, Jiangsu Province, 210004, China
| | - Dong-Yang Hong
- Genetic Medicine Center, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Qinhuai District, 123 Tianfei Lane, Mochou Road, Nanjing, Jiangsu Province, 210004, China
| | - Zhi-Lei Zhang
- Genetic Medicine Center, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Qinhuai District, 123 Tianfei Lane, Mochou Road, Nanjing, Jiangsu Province, 210004, China
| | - Ya-Hong Li
- Genetic Medicine Center, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Qinhuai District, 123 Tianfei Lane, Mochou Road, Nanjing, Jiangsu Province, 210004, China
| | - Pei-Ying Yang
- Genetic Medicine Center, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Qinhuai District, 123 Tianfei Lane, Mochou Road, Nanjing, Jiangsu Province, 210004, China
| | - Tao Jiang
- Genetic Medicine Center, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Qinhuai District, 123 Tianfei Lane, Mochou Road, Nanjing, Jiangsu Province, 210004, China.
| | - Zheng-Feng Xu
- Genetic Medicine Center, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Qinhuai District, 123 Tianfei Lane, Mochou Road, Nanjing, Jiangsu Province, 210004, China.
| |
Collapse
|
5
|
Shear MA, Wiita AP, Yu J, Wayman B, Sparks TN, Norton ME, Swanson K. Reporting Criteria for Prenatally Identified Variants of Uncertain Significance Differs Among Cytogenetics Laboratories in North America. Prenat Diagn 2025. [PMID: 40326825 DOI: 10.1002/pd.6785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 02/09/2025] [Accepted: 03/17/2025] [Indexed: 05/07/2025]
Abstract
OBJECTIVE Current technical standards for chromosomal microarray (CMA) interpretation are not prescriptive for reporting variants of uncertain significance (VUS) identified prenatally. We sought to compare prenatal CMA reporting among cytogenetic labs and identify potential drivers of practice variation. METHODS We conducted an electronic cross-sectional survey of cytogeneticists in the United States and Canada from July-December 2023. Participants were identified through the American Cytogenetics Forum List. RESULTS Labs reported differences in their size threshold used when reporting CNVs lacking OMIM annotated genes as a VUS, variable use of clinical data such as ultrasound or family history when deciding to report a VUS, and differences in opinion regarding the underlying pathogenicity of certain CNVs. Many cytogeneticists reported concerns about legal liability related to prenatal CMA reporting, and many shared concerns that a patient may terminate a pregnancy based on a VUS. CONCLUSION Reporting criteria for prenatally identified variants of uncertain significance differs among cytogenetic laboratories in North America. Many possible drivers of this practice variation were identified, including a lack of national guidelines that comprehensively address the unique considerations for prenatal CMA reporting.
Collapse
Affiliation(s)
- Matthew A Shear
- Division of Maternal-Fetal Medicine and Reproductive Genetics, University of California, San Francisco, California, USA
- Division of Medical Genetics and Genomics, University of California, San Francisco, California, USA
| | - Arun P Wiita
- Department of Laboratory Medicine, University of California, San Francisco, California, USA
| | - Jingwei Yu
- Department of Laboratory Medicine, University of California, San Francisco, California, USA
| | - Brette Wayman
- Department of Laboratory Medicine, University of California, San Francisco, California, USA
| | - Teresa N Sparks
- Division of Maternal-Fetal Medicine and Reproductive Genetics, University of California, San Francisco, California, USA
| | - Mary E Norton
- Division of Maternal-Fetal Medicine and Reproductive Genetics, University of California, San Francisco, California, USA
| | - Kate Swanson
- Division of Maternal-Fetal Medicine and Reproductive Genetics, University of California, San Francisco, California, USA
- Division of Medical Genetics and Genomics, University of California, San Francisco, California, USA
| |
Collapse
|
6
|
Arenillas C, Celada L, Ruiz-Cantador J, Calsina B, Datta D, García-Galea E, Fasani R, Moreno-Cárdenas AB, Alba-Linares JJ, Miranda-Barrio B, Martínez-Montes ÁM, Alvarez-Escola C, Lecumberri B, González García A, K. Flores S, Esquivel E, Ding Y, Peitzsch M, Robles-Guirado JÁ, Regojo Zapata RM, Pozo-Kreilinger JJ, Iglesias C, Dwight T, Muir CA, Oleaga A, Garrido-Lestache Rodríguez-Monte ME, Del Cerro MJ, Martínez-Bendayán I, Álvarez-González E, Cubiella T, Lourenço DM, A. Pereira MA, Burnichon N, Buffet A, Broberg C, Dickson PV, Fraga MF, Llorente Pendás JL, Rueda Soriano J, Buendía Fuentes F, Toledo SP, Clifton-Bligh R, Dienstmann R, Villanueva J, Capdevila J, Gimenez-Roqueplo AP, Favier J, Nuciforo P, Young WF, Bechmann N, Opotowsky AR, Vaidya A, Bancos I, Weghorn D, Robledo M, Casteràs A, Dos-Subirà L, Adameyko I, Chiara MD, Dahia PL, Toledo RA. Convergent Genetic Adaptation in Human Tumors Developed Under Systemic Hypoxia and in Populations Living at High Altitudes. Cancer Discov 2025; 15:1037-1062. [PMID: 40199338 PMCID: PMC12046333 DOI: 10.1158/2159-8290.cd-24-0943] [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: 07/04/2024] [Revised: 11/21/2024] [Accepted: 01/27/2025] [Indexed: 04/10/2025]
Abstract
SIGNIFICANCE This study reveals a broad convergence in genetic adaptation to hypoxia between natural populations and tumors, suggesting that insights from natural populations could enhance our understanding of cancer biology and identify novel therapeutic targets. See related commentary by Lee, p. 875.
Collapse
Affiliation(s)
- Carlota Arenillas
- Biomarkers and Clonal Dynamics Group, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Lucía Celada
- Health Research Institute of Asturias (ISPA), University of Oviedo, Oviedo, Spain
- Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain
| | - José Ruiz-Cantador
- Adult Congenital Heart Disease Unit, Department of Cardiology, Hospital Universitario La Paz, Madrid, Spain
| | - Bruna Calsina
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Debayan Datta
- Biomarkers and Clonal Dynamics Group, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Eduardo García-Galea
- Oncology Data Science (ODysSey) Group, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Roberta Fasani
- Molecular Oncology Group, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Ana Belén Moreno-Cárdenas
- Biomarkers and Clonal Dynamics Group, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Juan José Alba-Linares
- Health Research Institute of Asturias (ISPA), University of Oviedo, Oviedo, Spain
- Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain
- Cancer Epigenetics and Nanomedicine Laboratory, Nanomaterials and Nanotechnology Research Center (CINN-CSIC), University of Oviedo, Oviedo, Spain
- Department of Organisms and Systems Biology (B.O.S.), University of Oviedo, Oviedo, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Berta Miranda-Barrio
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Integrated Adult Congenital Heart Disease Unit, Department of Cardiology, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- European Reference Network for Rare, Low-Prevalence, or Complex Diseases of the Heart (ERN GUARD-Heart), Amsterdam, the Netherlands
| | - Ángel M. Martínez-Montes
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | - Beatriz Lecumberri
- Department of Endocrinology and Nutrition, Hospital Universitario La Paz, Madrid, Spain
| | - Ana González García
- Adult Congenital Heart Disease Unit, Department of Cardiology, Hospital Universitario La Paz, Madrid, Spain
| | - Shahida K. Flores
- Division of Hematology and Medical Oncology, Department of Medicine, Mays Cancer Center, University of Texas Health Science Center, San Antonio, Texas
| | - Emmanuel Esquivel
- Division of Hematology and Medical Oncology, Department of Medicine, Mays Cancer Center, University of Texas Health Science Center, San Antonio, Texas
| | - Yanli Ding
- Department of Pathology, University of Texas Health Science Center, San Antonio, Texas
| | - Mirko Peitzsch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - José-Ángel Robles-Guirado
- Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | | | | | - Carmela Iglesias
- Department of Pathology, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Trisha Dwight
- Cancer Genetics, Kolling Institute, Royal North Shore Hospital, Sydney, Australia
- The University of Sydney, Sydney, Australia
| | - Christopher A. Muir
- Department of Endocrinology, St. Vincent’s Hospital, Sydney, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| | - Amelia Oleaga
- Department of Endocrinology and Nutrition, Hospital Universitario de Basurto, Bilbao, Spain
| | | | - Maria Jesús Del Cerro
- Department of Pediatric Cardiology, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Isaac Martínez-Bendayán
- Department of Pediatric Cardiology, Instituto de Investigación Biomédica (Cardiopatía Estructural y Congénita) and Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
| | - Enol Álvarez-González
- Health Research Institute of Asturias (ISPA), University of Oviedo, Oviedo, Spain
- Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain
- Department of Functional Biology, Genetic Area, University of Oviedo, Oviedo, Spain
| | - Tamara Cubiella
- Health Research Institute of Asturias (ISPA), University of Oviedo, Oviedo, Spain
- Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain
| | - Delmar Muniz Lourenço
- Endocrinology Division, Hospital das Clínicas, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Maria Adelaide A. Pereira
- Endocrinology Division, Hospital das Clínicas, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Nelly Burnichon
- AP-HP, Hôpital Européen Georges Pompidou, Département de Médecine Génomique des Tumeurs et des Cancers, Paris, France
- Inserm, Centre de recherche des Cordeliers, Université Paris-Cité, Sorbonne Université, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Alexandre Buffet
- AP-HP, Hôpital Européen Georges Pompidou, Département de Médecine Génomique des Tumeurs et des Cancers, Paris, France
- Inserm, Centre de recherche des Cordeliers, Université Paris-Cité, Sorbonne Université, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Craig Broberg
- Adult Congenital Heart Program, Division of Cardiology, Oregon Health and Science University, Portland, Oregon
| | - Paxton V. Dickson
- Department of Surgery, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Mario F. Fraga
- Health Research Institute of Asturias (ISPA), University of Oviedo, Oviedo, Spain
- Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain
- Cancer Epigenetics and Nanomedicine Laboratory, Nanomaterials and Nanotechnology Research Center (CINN-CSIC), University of Oviedo, Oviedo, Spain
- Department of Organisms and Systems Biology (B.O.S.), University of Oviedo, Oviedo, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - José Luis Llorente Pendás
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Health Research Institute of Asturias (ISPA), University of Oviedo, Oviedo, Spain
- Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Joaquín Rueda Soriano
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Adult Congenital Heart Disease Unit, Department of Cardiology, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Francisco Buendía Fuentes
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Adult Congenital Heart Disease Unit, Department of Cardiology, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | | | - Roderick Clifton-Bligh
- Department of Endocrinology and Cancer Genetics Unit, Kolling Institute, Royal North Shore Hospital, Sydney, Australia
| | - Rodrigo Dienstmann
- Oncology Data Science (ODysSey) Group, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- University of Vic – Central University of Catalonia, Vic, Spain
| | - Josep Villanueva
- Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Jaume Capdevila
- Neuroendocrine and Endocrine Tumor Translational Research Program (NET-VHIO), Vall Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Department of Medical Oncology, Gastrointestinal and Endocrine Tumor Unit, Vall d’Hebron Hospital Universitari, Vall d’Hebron Hospital Campus, Barcelona, Spain
| | - Anne-Paule Gimenez-Roqueplo
- AP-HP, Hôpital Européen Georges Pompidou, Département de Médecine Génomique des Tumeurs et des Cancers, Paris, France
- Inserm, Centre de recherche des Cordeliers, Université Paris-Cité, Sorbonne Université, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Judith Favier
- Inserm, Centre de recherche des Cordeliers, Université Paris-Cité, Sorbonne Université, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Paolo Nuciforo
- Molecular Oncology Group, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | | | - Nicole Bechmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Alexander R. Opotowsky
- Cincinnati Adult Congenital Heart Disease Program, Heart Institute, Cincinnati Children’s Hospital, University of Cincinnati, Cincinnati, Ohio
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Anand Vaidya
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Irina Bancos
- Division of Endocrinology, Mayo Clinic, Rochester, Minnesota
| | | | - Mercedes Robledo
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Anna Casteràs
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Department of Endocrinology and Nutrition, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Laura Dos-Subirà
- Integrated Adult Congenital Heart Disease Unit, Department of Cardiology, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- European Reference Network for Rare, Low-Prevalence, or Complex Diseases of the Heart (ERN GUARD-Heart), Amsterdam, the Netherlands
| | - Igor Adameyko
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
- Department of Physiology and Pharmacology, Karolinska Institutet Stockholm, Sweden
| | - María-Dolores Chiara
- Health Research Institute of Asturias (ISPA), University of Oviedo, Oviedo, Spain
- Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain
| | - Patricia L.M. Dahia
- Division of Hematology and Medical Oncology, Department of Medicine, Mays Cancer Center, University of Texas Health Science Center, San Antonio, Texas
| | - Rodrigo A. Toledo
- Biomarkers and Clonal Dynamics Group, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
7
|
Nkya S, Nzunda C, Saukiwa E, Kaywanga F, Buchard E, Solomon D, Christopher H, Ngowi D, Johansen J, Urio F, Mgaya J, Kindole C, Yonazi M, Karim S, Alimohamed MZ, Sangeda RZ, Chamba C, Dandara C, Novelli E, Chimusa ER, Makani J. Exploring pharmacogenetic factors influencing hydroxyurea response in tanzanian sickle cell disease patients: a genomic medicine approach. THE PHARMACOGENOMICS JOURNAL 2025; 25:11. [PMID: 40268903 DOI: 10.1038/s41397-025-00372-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 04/02/2025] [Accepted: 04/17/2025] [Indexed: 04/25/2025]
Abstract
In sub-Saharan Africa, sickle cell disease (SCD) remains a significant public health challenge. Despite the discovery of SCD over a century ago, progress in developing and accessing effective treatments has been limited. Hydroxyurea is the primary drug used for managing SCD and associated with improving clinical outcomes. However, up to 30% of patients do not respond to hydroxyurea, likely due to genetic factors. This study involved 148 individuals with SCD investigated the association of hydroxyurea response with genetic variants across 13 loci associated with HbF synthesis and drug metabolism, focusing on MYB, HBB, HBG1, HBG2, BCL11A, KLF10, HAO2, NOS1, ARG2, SAR1A, CYP2C9, and CYP2E1. Significant associations with hydroxyurea response were identified in CYP2C9, CYP2E1, KLF10, BCL11A, ARG2, HBG1, SAR1A, MYB, and NOS1 loci. Furthermore, pathway enrichment and gene-gene interaction analyses provide deeper insights into the genetic mechanisms underlying hydroxyurea treatment response, highlighting potential avenues for personalized therapy in SCD management.
Collapse
Affiliation(s)
- Siana Nkya
- Department of Haematology and Blood Transfusion,, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
- Department of Biochemistry and Molecular Biology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
- Tanzania Human Genetics Organisation, Dar es Salaam, Tanzania
- Sickle Cell Program, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Collin Nzunda
- Department of Haematology and Blood Transfusion,, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania.
| | - Emmanuel Saukiwa
- Department of Biochemistry and Molecular Biology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Frida Kaywanga
- Department of Haematology and Blood Transfusion,, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
- Tanzania Human Genetics Organisation, Dar es Salaam, Tanzania
- Sickle Cell Program, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Eliud Buchard
- Department of Haematology and Blood Transfusion,, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - David Solomon
- Department of Haematology and Blood Transfusion,, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Heavenlight Christopher
- Department of Haematology and Blood Transfusion,, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Doreen Ngowi
- Department of Haematology and Blood Transfusion,, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Julieth Johansen
- Department of Haematology and Blood Transfusion,, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Florence Urio
- Department of Biochemistry and Molecular Biology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Josephine Mgaya
- Sickle Cell Program, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | | | - Mbonea Yonazi
- Muhimbili National Hospital, Dar es Salaam, Tanzania
| | - Salman Karim
- Department of Biochemistry and Molecular Biology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Mohamed Zahir Alimohamed
- Department of Biochemistry and Molecular Biology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
- Tanzania Human Genetics Organisation, Dar es Salaam, Tanzania
- Sickle Cell Program, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Raphael Z Sangeda
- Tanzania Human Genetics Organisation, Dar es Salaam, Tanzania
- Sickle Cell Program, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
- Department Pharmaceutical Microbiology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Clara Chamba
- Department of Haematology and Blood Transfusion,, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Collet Dandara
- Division of Human Genetics, Department of Pathology & Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
- Pharmacogenomics Research and Translation Unit, South African Medical Research Council, Cape Town, South Africa
| | - Enrico Novelli
- Vascular Medicine Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Emile R Chimusa
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle, Tyne and Wear, UK
| | - Julie Makani
- Department of Haematology and Blood Transfusion,, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
- Tanzania Human Genetics Organisation, Dar es Salaam, Tanzania
- Sickle Cell Program, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
- SickleInAfrica, Cape Town, South Africa
- Imperial College London, London, UK
| |
Collapse
|
8
|
Chagas PS, Garcia CB, Leopoldino AM. Genomic Insights into Oral Cancer Highlight Mutant SIGMAR1 as a Critical Target to Overcome Chemoresistance. Biochem Genet 2025:10.1007/s10528-025-11108-0. [PMID: 40257692 DOI: 10.1007/s10528-025-11108-0] [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: 01/30/2025] [Accepted: 04/13/2025] [Indexed: 04/22/2025]
Abstract
Oral cancer (OC) is a highly aggressive malignancy characterized by uncontrolled cell proliferation in the oral cavity. Recent studies have highlighted the role of Sigma-1 receptor (SIGMAR1) mutations in cancer progression, disrupting cellular homeostasis, altering gene and protein expression, and promoting drug resistance. However, its role in OC remains scarce. This study investigated SIGMAR1 mutations, expression profiles, and their potential link to drug resistance in OC. Using 2008 OC samples from the TCGA Pan-Cancer Atlas, we identified SIGMAR1 genetic alterations in 4% of cases, including missense mutations, deletions, and amplifications. In the HN13 OC cell line, Sanger sequencing revealed a novel heterozygous Asp-to-Gly (c.585C > G) missense mutation. Quantitative RT-PCR and Western blot analyses showed SIGMAR1 overexpression in HN13 cells compared to non-tumor oral keratinocytes (NOK-SI). Silencing SIGMAR1 increased HN13 cell sensitivity to cisplatin, indicating its role in drug resistance. This study is the first to report the c.585C > G mutation in SIGMAR1 and demonstrate its contribution to cisplatin resistance, a major chemotherapy challenge to OC treatment. These findings highlight SIGMAR1's critical role in OC pathogenesis and its potential as a therapeutic target to overcome chemoresistance. The results also pave the way for future research into RNA-based therapies and precision oncology interventions.
Collapse
Affiliation(s)
- Pablo Shimaoka Chagas
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. Do Café S/N, Ribeirão Preto, SP, 14040-903, Brazil.
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, 30912, USA.
| | - Cristiana Bernadelli Garcia
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. Do Café S/N, Ribeirão Preto, SP, 14040-903, Brazil
| | - Andréia Machado Leopoldino
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. Do Café S/N, Ribeirão Preto, SP, 14040-903, Brazil
| |
Collapse
|
9
|
Trasviña-Arenas CH, Dissanayake UC, Tamayo N, Hashemian M, Lin WJ, Demir M, Hoyos-Gonzalez N, Fisher AJ, Cisneros GA, Horvath MP, David SS. Structure of human MUTYH and functional profiling of cancer-associated variants reveal an allosteric network between its [4Fe-4S] cluster cofactor and active site required for DNA repair. Nat Commun 2025; 16:3596. [PMID: 40234396 PMCID: PMC12000561 DOI: 10.1038/s41467-025-58361-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 03/20/2025] [Indexed: 04/17/2025] Open
Abstract
MUTYH is a clinically important DNA glycosylase that thwarts mutations by initiating base-excision repair at 8-oxoguanine (OG):A lesions. The roles for its [4Fe-4S] cofactor in DNA repair remain enigmatic. Functional profiling of cancer-associated variants near the [4Fe-4S] cofactor reveals that most variations abrogate both retention of the cofactor and enzyme activity. Surprisingly, R241Q and N238S retained the metal cluster and bound substrate DNA tightly, but were completely inactive. We determine the crystal structure of human MUTYH bound to a transition state mimic and this shows that Arg241 and Asn238 build an H-bond network connecting the [4Fe-4S] cluster to the catalytic Asp236 that mediates base excision. The structure of the bacterial MutY variant R149Q, along with molecular dynamics simulations of the human enzyme, support a model in which the cofactor functions to position and activate the catalytic Asp. These results suggest that allosteric cross-talk between the DNA binding [4Fe-4S] cofactor and the base excision site of MUTYH regulate its DNA repair function.
Collapse
Affiliation(s)
- Carlos H Trasviña-Arenas
- Department of Chemistry, University of California, Davis, CA, USA
- Research Center on Aging, Center for Research and Advanced Studies (CINVESTAV), Mexico City, Mexico
| | - Upeksha C Dissanayake
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, TX, USA
| | - Nikole Tamayo
- Department of Chemistry, University of California, Davis, CA, USA
- Chemistry and Chemical Biology Graduate Program, University of California, Davis, CA, USA
| | - Mohammad Hashemian
- Department of Chemistry, University of California, Davis, CA, USA
- Chemistry and Chemical Biology Graduate Program, University of California, Davis, CA, USA
| | - Wen-Jen Lin
- Department of Chemistry, University of California, Davis, CA, USA
- Chemistry and Chemical Biology Graduate Program, University of California, Davis, CA, USA
| | - Merve Demir
- Department of Chemistry, University of California, Davis, CA, USA
- Chemistry and Chemical Biology Graduate Program, University of California, Davis, CA, USA
| | | | - Andrew J Fisher
- Department of Chemistry, University of California, Davis, CA, USA
- Chemistry and Chemical Biology Graduate Program, University of California, Davis, CA, USA
- Department of Molecular and Cellular Biology, University of California, Davis, CA, USA
| | - G Andrés Cisneros
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, TX, USA.
- Department of Physics, University of Texas at Dallas, Richardson, TX, USA.
| | - Martin P Horvath
- School of Biological Sciences, University of Utah, Salt Lake City, UT, USA.
| | - Sheila S David
- Department of Chemistry, University of California, Davis, CA, USA.
- Chemistry and Chemical Biology Graduate Program, University of California, Davis, CA, USA.
| |
Collapse
|
10
|
Bao J, Wen J, Chang C, Mu S, Chen J, Shivakumar M, Cui Y, Erus G, Yang Z, Yang S, Wen Z, Zhao Y, Kim D, Duong-Tran D, Saykin AJ, Zhao B, Davatzikos C, Long Q, Shen L. A genetically informed brain atlas for enhancing brain imaging genomics. Nat Commun 2025; 16:3524. [PMID: 40229250 PMCID: PMC11997130 DOI: 10.1038/s41467-025-57636-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 02/24/2025] [Indexed: 04/16/2025] Open
Abstract
Brain imaging genomics has manifested considerable potential in illuminating the genetic determinants of human brain structure and function. This has propelled us to develop the GIANT (Genetically Informed brAiN aTlas) that accounts for genetic and neuroanatomical variations simultaneously. Integrating voxel-wise heritability and spatial proximity, GIANT clusters brain voxels into genetically informed regions, while retaining fundamental anatomical knowledge. Compared to conventional (non-genetics) brain atlases, GIANT exhibits smaller intra-region variations and larger inter-region variations in terms of voxel-wise heritability. As a result, GIANT yields increased regional SNP heritability, enhanced polygenicity, and its polygenic risk score explains more brain volumetric variation than traditional neuroanatomical brain atlases. We provide extensive validation to GIANT and demonstrate its neuroanatomical validity, confirming its generalizability across populations with diverse genetic ancestries and various brain conditions. Furthermore, we present a comprehensive genetic architecture of the GIANT regions, covering their functional annotation at the molecular levels, their associations with other complex traits/diseases, and the genetic and phenotypic correlations among GIANT-defined imaging endophenotypes. In summary, GIANT constitutes a brain atlas that captures the complexity of genetic and neuroanatomical heterogeneity, thereby enhancing the discovery power and applicability of imaging genomics investigations in biomedical science.
Collapse
Affiliation(s)
- Jingxuan Bao
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Graduate Group in Genomics and Computational Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Junhao Wen
- Laboratory of AI and Biomedical Science (LABS), Columbia University, New York, NY, USA
- Center for Innovation in Imaging Biomarkers and Integrated Diagnostics (CIMBID), Department of Radiology, Columbia University, New York, NY, USA
- New York Genome Center (NYGC), New York, NY, USA
| | - Changgee Chang
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Shizhuo Mu
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jiong Chen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Graduate Group in Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Manu Shivakumar
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Graduate Group in Genomics and Computational Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Yuhan Cui
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Guray Erus
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Zhijian Yang
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Graduate Group in Applied Mathematics and Computational Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Shu Yang
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Zixuan Wen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Graduate Group in Applied Mathematics and Computational Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Yize Zhao
- Department of Biostatistics, Yale University School of Public Health, New Haven, CT, USA
| | - Dokyoon Kim
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Duy Duong-Tran
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Mathematics, United States Naval Academy, Annapolis, MD, USA
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Indiana University, Indianapolis, IN, USA
| | - Bingxin Zhao
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- Center for AI and Data Science for Integrated Diagnostics (AI2D), Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Qi Long
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
11
|
Gomes DHF, Medeiros IG, Petta TB, Stransky B, de Souza JES. DTreePred: an online viewer based on machine learning for pathogenicity prediction of genomic variants. BMC Bioinformatics 2025; 26:101. [PMID: 40205335 PMCID: PMC11983909 DOI: 10.1186/s12859-025-06113-4] [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: 10/18/2024] [Accepted: 03/12/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND A significant challenge in precision medicine is confidently identifying mutations detected in sequencing processes that play roles in disease treatment or diagnosis. Furthermore, the lack of representativeness of single nucleotide variants in public databases and low sequencing rates in underrepresented populations pose defies, with many pathogenic mutations still awaiting discovery. Mutational pathogenicity predictors have gained relevance as supportive tools in medical decision-making. However, significant disagreement among different tools regarding pathogenicity identification is rooted, necessitating manual verification to confirm mutation effects accurately. RESULTS This article presents a cross-platform mobile application, DTreePred, an online visualization tool for assessing the pathogenicity of nucleotide variants. DTreePred utilizes a machine learning-based pathogenicity model, including a decision tree algorithm and 15 machine learning classifiers alongside classical predictors. Connecting public databases with diverse prediction algorithms streamlines variant analysis, whereas the decision tree algorithm enhances the accuracy and reliability of variant pathogenicity data. This integration of information from various sources and prediction techniques aims to serve as a functional guide for decision-making in clinical practice. In addition, we tested DTreePred in a case study involving a cohort from Rio Grande do Norte, Brazil. By categorizing nucleotide variants from the list of oncogenes and suppressor genes classified in ClinVar as inexact data, DTreePred successfully revealed the pathogenicity of more than 95% of the nucleotide variants. Furthermore, an integrity test with 200 known mutations yielded an accuracy of 97%, surpassing rates expected from previous models. CONCLUSIONS DTreePred offers a robust solution for reducing uncertainty in clinical decision-making regarding pathogenic variants. Improving the accuracy of pathogenicity assessments has the potential to significantly increase the precision of medical diagnoses and treatments, particularly for underrepresented populations.
Collapse
Affiliation(s)
- Daniel Henrique Ferreira Gomes
- Bioinformatics Postgraduate Program, Metrópole Digital Institute, Federal University of Rio Grande Do Norte, Natal, Rio Grande Do Norte, 59078-400, Brazil
- Bioinformatics Multidisciplinary Environment (BioME), Metrópole Digital Institute, Federal University of Rio Grande Do Norte, Natal, Rio Grande Do Norte, 59078-400, Brazil
| | | | - Tirzah Braz Petta
- Bioinformatics Postgraduate Program, Metrópole Digital Institute, Federal University of Rio Grande Do Norte, Natal, Rio Grande Do Norte, 59078-400, Brazil
- Keck School of Medicine, Department of Translational Genomics, University of Southern California, 1450 Biggy St., Los Angeles, CA, 90089, USA
| | - Beatriz Stransky
- Bioinformatics Postgraduate Program, Metrópole Digital Institute, Federal University of Rio Grande Do Norte, Natal, Rio Grande Do Norte, 59078-400, Brazil
- Bioinformatics Multidisciplinary Environment (BioME), Metrópole Digital Institute, Federal University of Rio Grande Do Norte, Natal, Rio Grande Do Norte, 59078-400, Brazil
| | - Jorge Estefano Santana de Souza
- Bioinformatics Postgraduate Program, Metrópole Digital Institute, Federal University of Rio Grande Do Norte, Natal, Rio Grande Do Norte, 59078-400, Brazil.
- Bioinformatics Multidisciplinary Environment (BioME), Metrópole Digital Institute, Federal University of Rio Grande Do Norte, Natal, Rio Grande Do Norte, 59078-400, Brazil.
| |
Collapse
|
12
|
Patel M, Pottier C, Fan KH, Cetin A, Johnson M, Ali M, Liu M, Gorijala P, Budde J, Shi R, Cohen AD, Becker JT, Snitz BE, Aizenstein H, Lopez OL, Morris JC, Kamboh MI, Cruchaga C. Whole-genome sequencing reveals the impact of lipid pathway and APOE genotype on brain amyloidosis. Hum Mol Genet 2025; 34:739-748. [PMID: 39927718 PMCID: PMC11973900 DOI: 10.1093/hmg/ddaf017] [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: 08/08/2024] [Revised: 10/11/2024] [Accepted: 01/29/2025] [Indexed: 02/11/2025] Open
Abstract
Amyloid-PET imaging tracks the accumulation of amyloid beta (Aβ) deposits in the brain. Amyloid plaques accumulation may begin 10 to 20 years before the individual experiences clinical symptoms associated with Alzheimer's diseases (ad). Recent large-scale genome-wide association studies reported common risk factors associated with brain amyloidosis, suggesting that this endophenotype is driven by genetic variants. However, these loci pinpoint to large genomic regions and the functional variants remain to be identified. To identify new risk factors associated with brain amyloid deposition, we performed whole-genome sequencing on a large cohort of European descent individuals with amyloid PET imaging data (n = 1,888). Gene-based analysis for coding variants was performed using SKAT-O for amyloid PET as a quantitative endophenotype that identified genome-wide significant association for APOE (P = 2.45 × 10-10), and 26 new candidate genes with suggestive significance association (P < 5. 0 × 10-03) including SCN7A (P = 7.31 × 10-05), SH3GL1 (P = 7.56 × 10-04), and MFSD12 (P = 8.51 × 10-04). Enrichment analysis highlighted the lipid binding pathways as associated with Aβ deposition in brain driven by PITPNM3 (P = 4.27 × 10-03), APOE (P = 2.45 × 10-10), AP2A2 (P = 1.06 × 10-03), and SH3GL1 (P = 7.56 × 10-04). Overall, our data strongly support a connection between lipid metabolism and the deposition of Aβ in the brain. Our study illuminates promising avenues for therapeutic interventions targeting lipid metabolism to address brain amyloidosis.
Collapse
Affiliation(s)
- Maulikkumar Patel
- Department of Psychiatry, Neurogenomics and Informatics, Washington University School of Medicine, 4444 Forest Park Ave, St. Louis, MO 63108, United States
| | - Cyril Pottier
- Department of Psychiatry, Neurogenomics and Informatics, Department of Neurology, Washington University School of Medicine, 4444 Forest Park Ave, St. Louis, MO 63108, United States
| | - Kang-Hsien Fan
- Department of Human Genetics, University of Pittsburgh, 130 De Soto St, Pittsburgh, PA 15261, United States
| | - Arda Cetin
- Department of Psychiatry, Neurogenomics and Informatics, Washington University School of Medicine, 4444 Forest Park Ave, St. Louis, MO 63108, United States
| | - Matthew Johnson
- Department of Psychiatry, Neurogenomics and Informatics, Washington University School of Medicine, 4444 Forest Park Ave, St. Louis, MO 63108, United States
| | - Muhammad Ali
- Department of Psychiatry, Neurogenomics and Informatics, Washington University School of Medicine, 4444 Forest Park Ave, St. Louis, MO 63108, United States
| | - Menghan Liu
- Department of Psychiatry, Neurogenomics and Informatics, Washington University School of Medicine, 4444 Forest Park Ave, St. Louis, MO 63108, United States
| | - Priyanka Gorijala
- Department of Psychiatry, Neurogenomics and Informatics, Washington University School of Medicine, 4444 Forest Park Ave, St. Louis, MO 63108, United States
| | - John Budde
- Department of Psychiatry, Neurogenomics and Informatics, Washington University School of Medicine, 4444 Forest Park Ave, St. Louis, MO 63108, United States
| | - Ruyu Shi
- Department of Human Genetics, University of Pittsburgh, 130 De Soto St, Pittsburgh, PA 15261, United States
| | - Ann D Cohen
- Department of Psychiatry, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA 15213, United States
| | - James T Becker
- Department of Neurology, University of Pittsburgh, 3471 Fifth Avenue, Pittsburgh, PA 15213, United States
| | - Beth E Snitz
- Department of Neurology, University of Pittsburgh, 3471 Fifth Avenue, Pittsburgh, PA 15213, United States
| | - Howard Aizenstein
- Department of Human Genetics, University of Pittsburgh, 130 De Soto St, Pittsburgh, PA 15261, United States
| | - Oscar L Lopez
- Department of Neurology, University of Pittsburgh, 3471 Fifth Avenue, Pittsburgh, PA 15213, United States
| | - John C Morris
- Department of Neurology, Hope Center for Neurologic Diseases, Section on Aging & Dementia, Institute of Clinical and Translational Sciences, Knight Alzheimer Disease Research Center Washington University School of Medicine, 4901 Forest Park Ave 4th floor, St. Louis, MO 63108, United States
| | - M Ilyas Kamboh
- Department of Human Genetics, Department of Psychiatry University of Pittsburgh, 130 De Soto St, Pittsburgh, PA 15261, United States
| | - Carlos Cruchaga
- Department of Psychiatry, Neurogenomics and Informatics, Department of Neurology, Hope Center for Neurologic Diseases, Knight Alzheimer Disease Research Center, Washington University School of Medicine, 4444 Forest Park Ave, St. Louis, MO 63108, United States
| |
Collapse
|
13
|
Pilalis E, Zisis D, Andrinopoulou C, Karamanidou T, Antonara M, Stavropoulos TG, Chatziioannou A. Genome-wide functional annotation of variants: a systematic review of state-of-the-art tools, techniques and resources. Front Pharmacol 2025; 16:1474026. [PMID: 40098614 PMCID: PMC11911558 DOI: 10.3389/fphar.2025.1474026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 02/03/2025] [Indexed: 03/19/2025] Open
Abstract
The recent advancement of sequencing technologies marks a significant shift in the character and complexity of the digital genomic data universe, encompassing diverse types of molecular data, screened through manifold technological platforms. As a result, a plethora of fully assembled genomes are generated that span vertically the evolutionary scale. Notwithstanding the tsunami of thriving innovations that accomplish unprecedented, nucleotide-level, structural and functional annotation, an exhaustive, systemic, massive genome-wide functional annotation remains elusive, particularly when the criterion is automation and efficiency in data-agnostic interpretation. The latter is of paramount importance for the elaboration of strategies for sophisticated, data-driven genome-wide annotation, which aim to impart a sustainable and comprehensive systemic approach to addressing whole genome variation. Therefore, it is essential to develop methods and tools that promote systematic functional genomic annotation, with emphasis on mechanistic information exceeding the limits of coding regions, and exploiting the chunks of pertinent information residing in non-coding regions, including promoter and enhancer sequences, non-coding RNAs, DNA methylation sites, transcription factor binding sites, transposable elements and more. This review provides an overview of the current state-of-the-art in genome-wide functional annotation of genetic variation, including existing bioinformatic tools, resources, databases and platforms currently available or reported in the literature. Particular emphasis is placed on the functional annotation of variants that lie outside protein-coding genomic regions (intronic or intergenic), their potential co-localization with regulatory element areas, such as putative non-coding RNA regions, and the assessment of their functional impact on the investigated phenotype. In addition, state-of-the-art tools that leverage data obtained from WGS and GWAS-based analyses are discussed, along with future bioinformatics directions and developments. These future directions emphasize efficient, comprehensive, and largely automated functional annotation of both coding and non-coding genomic variants, as well as their optimal evaluation.
Collapse
Affiliation(s)
| | | | | | | | - Maria Antonara
- Pfizer Center for Digital Innovation, Thessaloniki, Greece
| | | | - Aristotelis Chatziioannou
- e-NIOS Applications PC, Kallithea, Greece
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| |
Collapse
|
14
|
Lee J, Kim Y, Lee C, Jeon SS, Seo H, Lee J, Choi J, Kang M, Kim E, Shin K. Generation of prostate cancer assembloids modeling the patient-specific tumor microenvironment. PLoS Genet 2025; 21:e1011652. [PMID: 40163511 PMCID: PMC12002641 DOI: 10.1371/journal.pgen.1011652] [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: 08/02/2024] [Revised: 04/16/2025] [Accepted: 03/09/2025] [Indexed: 04/02/2025] Open
Abstract
Prostate cancer (PC) is the most frequently diagnosed malignancy among men and contributes significantly to cancer-related mortality. While recent advances in in vitro PC modeling systems have been made, there remains a lack of robust preclinical models that faithfully recapitulate the genetic and phenotypic characteristics across various PC subtypes-from localized PC (LPC) to castration-resistant PC (CRPC)-along with associated stromal cells. Here, we established human PC assembloids from LPC and CRPC tissues by reconstituting tumor organoids with corresponding cancer-associated fibroblasts (CAFs), thereby incorporating aspects of the tumor microenvironment (TME). Established PC organoids exhibited high concordance in genomic landscape with parental tumors, and the tumor assembloids showed a higher degree of phenotypic similarity to parental tumors compared to tumor organoids without CAFs. PC assembloids displayed increased proliferation and reduced sensitivity to anti-cancer treatments, indicating that PC assembloids are potent tools for understanding PC biology, investigating the interaction between tumor and CAFs, and identifying personalized therapeutic targets.
Collapse
Affiliation(s)
- Juhee Lee
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
| | - Yunhee Kim
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
| | - Cheol Lee
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seong Soo Jeon
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hae Seo
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jongwon Lee
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jungmin Choi
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Minyong Kang
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Health Sciences and Technology, The Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
| | - Eunjee Kim
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
| | - Kunyoo Shin
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
15
|
DeSpenza T, Kiziltug E, Allington G, Barson DG, McGee S, O'Connor D, Robert SM, Mekbib KY, Nanda P, Greenberg ABW, Singh A, Duy PQ, Mandino F, Zhao S, Lynn A, Reeves BC, Marlier A, Getz SA, Nelson-Williams C, Shimelis H, Walsh LK, Zhang J, Wang W, Prina ML, OuYang A, Abdulkareem AF, Smith H, Shohfi J, Mehta NH, Dennis E, Reduron LR, Hong J, Butler W, Carter BS, Deniz E, Lake EMR, Constable RT, Sahin M, Srivastava S, Winden K, Hoffman EJ, Carlson M, Gunel M, Lifton RP, Alper SL, Jin SC, Crair MC, Moreno-De-Luca A, Luikart BW, Kahle KT. PTEN mutations impair CSF dynamics and cortical networks by dysregulating periventricular neural progenitors. Nat Neurosci 2025; 28:536-557. [PMID: 39994410 PMCID: PMC12038823 DOI: 10.1038/s41593-024-01865-3] [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: 07/06/2023] [Accepted: 12/05/2024] [Indexed: 02/26/2025]
Abstract
Enlargement of the cerebrospinal fluid (CSF)-filled brain ventricles (ventriculomegaly) is a defining feature of congenital hydrocephalus (CH) and an under-recognized concomitant of autism. Here, we show that de novo mutations in the autism risk gene PTEN are among the most frequent monogenic causes of CH and primary ventriculomegaly. Mouse Pten-mutant ventriculomegaly results from aqueductal stenosis due to hyperproliferation of periventricular Nkx2.1+ neural progenitor cells (NPCs) and increased CSF production from hyperplastic choroid plexus. Pten-mutant ventriculomegalic cortices exhibit network dysfunction from increased activity of Nkx2.1+ NPC-derived inhibitory interneurons. Raptor deletion or postnatal everolimus treatment corrects ventriculomegaly, rescues cortical deficits and increases survival by antagonizing mTORC1-dependent Nkx2.1+ NPC pathology. Thus, PTEN mutations concurrently alter CSF dynamics and cortical networks by dysregulating Nkx2.1+ NPCs. These results implicate a nonsurgical treatment for CH, demonstrate a genetic association of ventriculomegaly and ASD, and help explain neurodevelopmental phenotypes refractory to CSF shunting in select individuals with CH.
Collapse
Affiliation(s)
- Tyrone DeSpenza
- Interdepartmental Neuroscience Program, Yale School of Medicine, Yale University, New Haven, CT, USA
- Medical Scientist Training Program, Yale School of Medicine, Yale University, New Haven, CT, USA
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
- Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Emre Kiziltug
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Garrett Allington
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, CT, USA
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons and New York Presbyterian Hospital, New York, NY, USA
| | - Daniel G Barson
- Interdepartmental Neuroscience Program, Yale School of Medicine, Yale University, New Haven, CT, USA
- Medical Scientist Training Program, Yale School of Medicine, Yale University, New Haven, CT, USA
| | | | - David O'Connor
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Stephanie M Robert
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Kedous Y Mekbib
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Pranav Nanda
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ana B W Greenberg
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Amrita Singh
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Phan Q Duy
- Interdepartmental Neuroscience Program, Yale School of Medicine, Yale University, New Haven, CT, USA
- Medical Scientist Training Program, Yale School of Medicine, Yale University, New Haven, CT, USA
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Francesca Mandino
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Shujuan Zhao
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Anna Lynn
- Medical Scientist Training Program, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Benjamin C Reeves
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Arnaud Marlier
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Stephanie A Getz
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Carol Nelson-Williams
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Hermela Shimelis
- Autism & Developmental Medicine Institute, Geisinger, Lewisburg, PA, USA
| | - Lauren K Walsh
- Autism & Developmental Medicine Institute, Geisinger, Lewisburg, PA, USA
| | - Junhui Zhang
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Wei Wang
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Mackenzi L Prina
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Department of Neurobiology, UAB Heersink School of Medicine, Birmingham, AL, USA
| | - Annaliese OuYang
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Asan F Abdulkareem
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Department of Neurobiology, UAB Heersink School of Medicine, Birmingham, AL, USA
| | - Hannah Smith
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - John Shohfi
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Neel H Mehta
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Evan Dennis
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Laetitia R Reduron
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Jennifer Hong
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - William Butler
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Bob S Carter
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Engin Deniz
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Evelyn M R Lake
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - R Todd Constable
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Mustafa Sahin
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Siddharth Srivastava
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kellen Winden
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ellen J Hoffman
- Child Study Center, Yale School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Marina Carlson
- Interdepartmental Neuroscience Program, Yale School of Medicine, Yale University, New Haven, CT, USA
- Child Study Center, Yale School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Murat Gunel
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Richard P Lifton
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY, USA
| | - Seth L Alper
- Division of Nephrology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, and Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Diagnostic Medicine Institute, Geisinger, Danville, PA, USA
| | - Sheng Chih Jin
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael C Crair
- Interdepartmental Neuroscience Program, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Andres Moreno-De-Luca
- Autism & Developmental Medicine Institute, Geisinger, Lewisburg, PA, USA
- Department of Radiology, Diagnostic Medicine Institute, Geisinger, Danville, PA, USA
| | - Bryan W Luikart
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA.
- Department of Neurobiology, UAB Heersink School of Medicine, Birmingham, AL, USA.
| | - Kristopher T Kahle
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA.
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
16
|
Xue Y, Liu M, Chen K, Xu Y, Yang J. RAB32 Ser71Arg in Chinese patients with Parkinson's disease. Neurol Sci 2025; 46:1379-1381. [PMID: 39652162 DOI: 10.1007/s10072-024-07910-0] [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: 10/24/2024] [Accepted: 11/25/2024] [Indexed: 04/29/2025]
Abstract
BACKGROUND The RAB32 Ser71Arg variant has been identified as a novel risk locus for Parkinson's disease (PD) in North American, European and North African populations. However, its pathogenicity in Asian populations remains unclear. METHOD To investigate this, we screened for the RAB32 c.213C > G (Ser71Arg) variant using Sanger sequencing in 1,099 PD patients and 1,549 controls. And we search for the RAB32 Ser71Arg variant in public databases to identified its mutant frequency. RESULTS Our results show that no individuals carrying the RAB32 Ser71Arg variant were identified in our cohort. Additionally, this variant rarely appears in Asian population databases. CONCLUSION Our findings suggest that the RAB32 Ser71Arg variant is unlikely to be a risk locus for PD in Chinese patients, which is potentially attributed to racial or ethnic differences.
Collapse
Affiliation(s)
- Yinge Xue
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jianshe Road, Zhengzhou, 450052, Henan, China
| | - Minglei Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jianshe Road, Zhengzhou, 450052, Henan, China
| | - Kaixin Chen
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jianshe Road, Zhengzhou, 450052, Henan, China
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jianshe Road, Zhengzhou, 450052, Henan, China
| | - Jing Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jianshe Road, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
17
|
Huang T, Liu YN, Ding DT, Wang Q, Xie QL, Miao XC, Qin C, Huang XF, Li J. Identification of a novel single nucleotide deletion in the NHS causing Nance-Horan syndrome. BMC Ophthalmol 2025; 25:92. [PMID: 39994540 PMCID: PMC11854407 DOI: 10.1186/s12886-025-03933-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 02/18/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Nance-Horan syndrome (NHS) is a rare X-linked dominant disorder caused by pathogenic variants in the NHS gene on chromosome Xp22.2-Xp22.13. Clinical manifestations consist of congenital cataracts, along with dysmorphic facial features and dental anomalies and, in certain instances, intellectual disability. This study aimed to identify the genetic cause responsible for NHS in a Chinese family with four individuals primarily presenting with congenital cataracts. METHODS Genomic DNA was collected from six family members, including four affected individuals (three females and one male) from a two-generation family. The family history and clinical data were documented. Whole-exome sequencing was performed on the proband, and candidate pathogenic variants were filtered through a series of screening steps and validated by Sanger sequencing. Co-segregation analysis was conducted to confirm the pathogenicity of the identified variant. RESULTS Genetic analysis revealed a novel frameshift pathogenic variant in NHS gene (c.1735delA: p.R579Gfs*91) present in all four affected members. All affected members exhibited congenital cataracts, congenital ptosis, strabismus, high myopia as well as dental and facial anomalies, and more severe characteristic features observed in the male patient. These clinical manifestations were consistent with the phenotype of NHS. CONCLUSION This study identified a novel NHS pathogenic variant in a Chinese family, expanding the mutational spectrum of NHS. Contrary to previous reports of female carriers exhibiting mild symptoms, we demonstrated severe ocular phenotypes in three affected females. These findings will assist in providing genetic counseling for NHS patients.
Collapse
Affiliation(s)
- Teng Huang
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Ya-Nan Liu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Dan-Tong Ding
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Qiao Wang
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Qiu-Ling Xie
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xue-Chuan Miao
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Chuan Qin
- Institute of PSI Genomics Co., Ltd, Wenzhou, China
| | - Xiu-Feng Huang
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, Wenzhou, 325027, China.
| | - Jin Li
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China.
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China.
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
18
|
Salvador L, Valle JD, Dorca E, Chong AS, Chong AL, Camacho Valenzuela J, Munté E, Rioja C, Martí-Sánchez L, Salinas M, Darder E, Fabian MR, Brunet J, Salvador H, Lázaro C, Rivera B. DICER1 in pediatric and adult cancer predisposition populations: Prevalence, phenotypes, and mosaicism. Genet Med 2025; 27:101385. [PMID: 39976125 DOI: 10.1016/j.gim.2025.101385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/21/2025] Open
Abstract
PURPOSE DICER1 tumor predisposition syndrome (DTPS) is a hereditary condition affecting children and young adults. Identification of DICER1 carriers is key for prevention and actionability in families. However, DTPS diagnosis is hindered by its incomplete penetrance and broad phenotypic spectrum. METHODS We performed an analysis of DICER1 sequencing data from 92 children and 6108 adults with suspected cancer predisposition syndrome. Clinical and DICER1 somatic data from selected carriers and public data sets were studied. RESULTS The prevalence of germline DICER1 pathogenic variants was 1:30 in children and 1:3054 in adults. No adult referral phenotype was a known DTPS-associated tumor, although 3 of 5 carriers developed thyroid alterations. We provide functional evidence supporting the pathogenicity of a novel in-frame deletion. A 56-year-old woman with ovarian carcinoma and toxic diffuse thyroid hyperplasia was found to have a postzygotic hotspot missense variant. CONCLUSION The prevalence of DICER1 pathogenic variants in cancer predisposition populations was 5 to 6 times that reported in the general population. Pediatric-onset DTPS is well characterized, whereas adult carriers mainly present with thyroid abnormalities in the absence of DICER1-related family history, thus requiring accurate criteria for its identification when in constellation with other tumor types. Postzygotic hotspot missense variants may exist without the expected severe phenotype.
Collapse
Affiliation(s)
- Lluis Salvador
- Hereditary Cancer Program, Catalan Institute of Oncology (ICO), L'Hospitalet de Llobregat, Barcelona, Spain; Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Jesús Del Valle
- Hereditary Cancer Program, Catalan Institute of Oncology (ICO), L'Hospitalet de Llobregat, Barcelona, Spain; Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Eduard Dorca
- Pathology Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Anne-Sophie Chong
- Hereditary Cancer Program, Catalan Institute of Oncology (ICO), L'Hospitalet de Llobregat, Barcelona, Spain; Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Genetics Program, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Anne-Laure Chong
- Cancer Research Axis, Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada
| | - José Camacho Valenzuela
- Department of Human Genetics, McGill University, Montreal, QC, Canada; Cancer Research Program, Centre for Translational Biology, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Elisabet Munté
- Hereditary Cancer Program, Catalan Institute of Oncology (ICO), L'Hospitalet de Llobregat, Barcelona, Spain; Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Cristina Rioja
- Hereditary Cancer Program, Catalan Institute of Oncology (ICO), L'Hospitalet de Llobregat, Barcelona, Spain; Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Laura Martí-Sánchez
- Department of Clinical Biochemistry, Sant Joan de Déu Barcelona Children's Hospital, Esplugues de Llobregat, Barcelona, Spain
| | - Mónica Salinas
- Hereditary Cancer Program, Catalan Institute of Oncology (ICO), L'Hospitalet de Llobregat, Barcelona, Spain; Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Esther Darder
- Hereditary Cancer Program, Catalan Institute of Oncology (ICO) and Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Marc R Fabian
- Cancer Research Axis, Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada; Gerald Bronfman Department of Oncology, McGill University, Montreal, Quebec, Canada
| | - Joan Brunet
- Hereditary Cancer Program, Catalan Institute of Oncology (ICO), L'Hospitalet de Llobregat, Barcelona, Spain; Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Hereditary Cancer Program, Catalan Institute of Oncology (ICO) and Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Hector Salvador
- Department of Pediatric Oncology, Neurocutaneous Disorders and Cancer Predisposition Unit, Sant Joan de Déu Barcelona Children's Hospital, Esplugues de Llobregat, Barcelona, Spain
| | - Conxi Lázaro
- Hereditary Cancer Program, Catalan Institute of Oncology (ICO), L'Hospitalet de Llobregat, Barcelona, Spain; Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Barbara Rivera
- Hereditary Cancer Program, Catalan Institute of Oncology (ICO), L'Hospitalet de Llobregat, Barcelona, Spain; Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Cancer Research Axis, Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada; Gerald Bronfman Department of Oncology, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
19
|
Bunting EL, Donaldson J, Cumming SA, Olive J, Broom E, Miclăuș M, Hamilton J, Tegtmeyer M, Zhao HT, Brenton J, Lee WS, Handsaker RE, Li S, Ford B, Ryten M, McCarroll SA, Kordasiewicz HB, Monckton DG, Balmus G, Flower M, Tabrizi SJ. Antisense oligonucleotide-mediated MSH3 suppression reduces somatic CAG repeat expansion in Huntington's disease iPSC-derived striatal neurons. Sci Transl Med 2025; 17:eadn4600. [PMID: 39937881 DOI: 10.1126/scitranslmed.adn4600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 01/23/2025] [Indexed: 02/14/2025]
Abstract
Expanded CAG alleles in the huntingtin (HTT) gene that cause the neurodegenerative disorder Huntington's disease (HD) are genetically unstable and continue to expand somatically throughout life, driving HD onset and progression. MSH3, a DNA mismatch repair protein, modifies HD onset and progression by driving this somatic CAG repeat expansion process. MSH3 is relatively tolerant of loss-of-function variation in humans, making it a potential therapeutic target. Here, we show that an MSH3-targeting antisense oligonucleotide (ASO) effectively engaged with its RNA target in induced pluripotent stem cell (iPSC)-derived striatal neurons obtained from a patient with HD carrying 125 HTT CAG repeats (the 125 CAG iPSC line). ASO treatment led to a dose-dependent reduction of MSH3 and subsequent stalling of CAG repeat expansion in these striatal neurons. Bulk RNA sequencing revealed a safe profile for MSH3 reduction, even when reduced by >95%. Maximal knockdown of MSH3 also effectively slowed CAG repeat expansion in striatal neurons with an otherwise accelerated expansion rate, derived from the 125 CAG iPSC line where FAN1 was knocked out by CRISPR-Cas9 editing. Last, we created a knock-in mouse model expressing the human MSH3 gene and demonstrated effective in vivo reduction in human MSH3 after ASO treatment. Our study shows that ASO-mediated MSH3 reduction can prevent HTT CAG repeat expansion in HD 125 CAG iPSC-derived striatal neurons, highlighting the therapeutic potential of this approach.
Collapse
Affiliation(s)
- Emma L Bunting
- Huntington's Disease Centre and Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology and UK Dementia Research Institute, UCL, London, UK
| | - Jasmine Donaldson
- Huntington's Disease Centre and Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology and UK Dementia Research Institute, UCL, London, UK
| | - Sarah A Cumming
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Jessica Olive
- Huntington's Disease Centre and Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology and UK Dementia Research Institute, UCL, London, UK
| | - Elizabeth Broom
- Huntington's Disease Centre and Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology and UK Dementia Research Institute, UCL, London, UK
| | - Mihai Miclăuș
- UK Dementia Research Institute at University of Cambridge and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0AH, UK
- Department of Molecular Neuroscience, Transylvanian Institute of Neuroscience, 400191 Cluj-Napoca, Romania
| | - Joseph Hamilton
- Huntington's Disease Centre and Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology and UK Dementia Research Institute, UCL, London, UK
| | - Matthew Tegtmeyer
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Jonathan Brenton
- UK Dementia Research Institute at University of Cambridge and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0AH, UK
- Department of Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, London, UK
| | - Won-Seok Lee
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Robert E Handsaker
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Susan Li
- Ionis Pharmaceuticals, Carlsbad, CA, USA
| | | | - Mina Ryten
- UK Dementia Research Institute at University of Cambridge and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0AH, UK
- Department of Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, London, UK
| | - Steven A McCarroll
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | | | - Darren G Monckton
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Gabriel Balmus
- UK Dementia Research Institute at University of Cambridge and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0AH, UK
- Department of Molecular Neuroscience, Transylvanian Institute of Neuroscience, 400191 Cluj-Napoca, Romania
| | - Michael Flower
- Huntington's Disease Centre and Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology and UK Dementia Research Institute, UCL, London, UK
| | - Sarah J Tabrizi
- Huntington's Disease Centre and Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology and UK Dementia Research Institute, UCL, London, UK
| |
Collapse
|
20
|
Feng Q, Jiang L, Zhang S, He C, Mei L, Liu Y. A novel frameshift mutation in the DIAPH1 gene causes a Chinese family autosomal dominant nonsyndromic hearing loss: Mutation in DIAPH1 causes hearing loss. Gene 2025; 936:149088. [PMID: 39542281 DOI: 10.1016/j.gene.2024.149088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 10/08/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
OBJECTIVE This study reports a novel heterozygous, likely truncating mutation in the diaphanous homolog 1 (DIAPH1) gene associated with non-syndromic hearing loss. METHODS Family members underwent audiological and imaging assessments, whole-exome sequencing (WES), and Sanger sequencing. RESULTS Sensorineural hearing loss was observed in all five individuals, with severity ranging from mild to severe. None of the affected patients reported vestibular complaints, and routine blood tests showed normal platelet counts. Whole-exome sequencing (WES) revealed a novel frameshift variation, c.3555delA (p.Gln1185Hisfs*3), in exon 26 of the DIAPH1 gene. This variation co-segregated with the hearing-impaired phenotype in the family. The data collected support the classification of c.3555delA as a genetic etiology of hereditary hearing loss according to the American College of Medical Genetics and Genomics guidelines. CONCLUSION We identified a novel pathogenic mutation in the DIAPH1 gene, thereby expanding the mutation spectrum associated with hearing loss.
Collapse
Affiliation(s)
- Qi Feng
- Department of Otolaryngology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China; Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, Hunan 410008, China
| | - Lu Jiang
- Department of Otolaryngology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China; Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, Hunan 410008, China
| | - Shuai Zhang
- Department of Otolaryngology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China; Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, Hunan 410008, China
| | - Chufeng He
- Department of Otolaryngology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China; Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, Hunan 410008, China
| | - Lingyun Mei
- Department of Otolaryngology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China; Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, Hunan 410008, China.
| | - Yalan Liu
- Department of Otolaryngology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China; Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, Hunan 410008, China.
| |
Collapse
|
21
|
Qu HQ, Delfiner MS, Gangireddy C, Vaidya A, Nguyen K, Whitman IR, Wang J, Song J, Bristow MR, McTiernan CF, Gerhard GS, Hakonarson H, Feldman AM. Rare variants in cardiomyopathy genes predispose to cardiac injury in severe COVID-19 patients of African or Hispanic ancestry. J Mol Med (Berl) 2025; 103:175-185. [PMID: 39730912 PMCID: PMC11799050 DOI: 10.1007/s00109-024-02510-z] [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: 09/09/2024] [Revised: 12/16/2024] [Accepted: 12/19/2024] [Indexed: 12/29/2024]
Abstract
In one of the earliest reports from China during COVID-19, it was noted that over 20% of patients hospitalized with the disease had significant elevations of troponin, a marker of myocardial tissue damage, that put them at a higher risk. In a hypothesis-independent whole exome sequencing (WES) study in hospitalized COVID-19 patients of diverse ancestry, we observed putative enrichment in pathogenic variants in genes known to be involved in the pathogenesis of cardiomyopathy. This observation led us to hypothesize that the observed high morbidity and mortality in these patients might be due to the presence of rare genetic factors that had previously been silent but became relevant as a consequence of the severe stress inflicted by an infection with SARS-CoV-2. To test this hypothesis, we analyzed our WES data generated from a cohort of 325 patients sequentially admitted for COVID-19 infection. In this predominantly minority population (53.9% African ancestry and 37.9% Hispanic/Latin ancestry), our initial analysis screen identified 263 variants that were identified as highly deleterious (HD) from a total of 26,661 variants of interest that represented 215 genes. Of those, we identified 46 genes (in 58 patients) harboring rare HD coding variants that were previously implicated in dilated cardiomyopathy and were considered as disease initiators for the severe COVID-19 in this study. These findings offer valuable insights into the molecular mechanisms and genetic susceptibility to heart injury in severe COVID-19. KEY MESSAGES: COVID-19 may cause cardiac damage in some affected patients without a plausible biological explanation. Our study reveals an enrichment of highly deleterious variants linked to cardiomyopathy in severe COVID-19 patients. Genetic profiling unveils the molecular basis of severe COVID-19-related heart injury, potentially aiding in patient stratification.
Collapse
Affiliation(s)
- Hui-Qi Qu
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Matthew S Delfiner
- Department of Medicine, Division of Cardiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Chethan Gangireddy
- Department of Medicine, Division of Cardiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Anjali Vaidya
- Department of Medicine, Division of Cardiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Kenny Nguyen
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Isaac R Whitman
- Department of Medicine, Division of Cardiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - JuFang Wang
- Department of Medicine, Division of Cardiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Jianliang Song
- Department of Medicine, Division of Cardiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Michael R Bristow
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Charles F McTiernan
- Division of Cardiology, Heart and Vascular Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Glenn S Gerhard
- Department of Human Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Hakon Hakonarson
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Arthur M Feldman
- Department of Medicine, Division of Cardiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA.
| |
Collapse
|
22
|
Zhao W, Tao Y, Xiong J, Liu L, Wang Z, Shao C, Shang L, Hu Y, Xu Y, Su Y, Yu J, Feng T, Xie J, Xu H, Zhang Z, Peng J, Wu J, Zhang Y, Zhu S, Xia K, Tang B, Zhao G, Li J, Li B. GoFCards: an integrated database and analytic platform for gain of function variants in humans. Nucleic Acids Res 2025; 53:D976-D988. [PMID: 39578693 PMCID: PMC11701611 DOI: 10.1093/nar/gkae1079] [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: 08/15/2024] [Revised: 10/20/2024] [Accepted: 10/28/2024] [Indexed: 11/24/2024] Open
Abstract
Gain-of-function (GOF) variants, which introduce new or amplify protein functions, are essential for understanding disease mechanisms. Despite advances in genomics and functional research, identifying and analyzing pathogenic GOF variants remains challenging owing to fragmented data and database limitations, underscoring the difficulty in accessing critical genetic information. To address this challenge, we manually reviewed the literature, pinpointing 3089 single-nucleotide variants and 72 insertions and deletions in 579 genes associated with 1299 diseases from 2069 studies, and integrated these with the 3.5 million predicted GOF variants. Our approach is complemented by a proprietary scoring system that prioritizes GOF variants on the basis of the evidence supporting their GOF effects and provides predictive scores for variants that lack existing documentation. We then developed a database named GoFCards for general geneticists and clinicians to easily obtain GOF variants in humans (http://www.genemed.tech/gofcards). This database also contains data from >150 sources and offers comprehensive variant-level and gene-level annotations, with the aim of providing users with convenient access to detailed and relevant genetic information. Furthermore, GoFCards empowers users with limited bioinformatic skills to analyze and annotate genetic data, and prioritize GOF variants. GoFCards offers an efficient platform for interpreting GOF variants and thereby advancing genetic research.
Collapse
Affiliation(s)
- Wenjing Zhao
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital & Center for Medical Genetics, School of Life Sciences, Central South University, No. 87 Xiangya Road, Furong District, Changsha, Hunan 410008, China
- Department of Medical Genetics, NHC Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China, The First People's Hospital of Yunnan Province, No. 157 Jinbi Road, Xishan District, Kunming, Yunnan 650000, China
- School of Medicinie, Kunming University of Science and Technology, No. 727 Jingming South Road, Chenggong District, Kunming, Yunnan 650000, China
| | - Youfu Tao
- Xiangya School of Medicine, Central South University, No. 172 Tongzipo Road, Yuelu District, Changsha, Hunan 410008, China
| | - Jiayi Xiong
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital & Center for Medical Genetics, School of Life Sciences, Central South University, No. 87 Xiangya Road, Furong District, Changsha, Hunan 410008, China
| | - Lei Liu
- School of Life Science, Central South University, No. 172 Tongzipo Road, Yuelu District, Changsha, Hunan 410008, China
| | - Zhongqing Wang
- School of Medicinie, Kunming University of Science and Technology, No. 727 Jingming South Road, Chenggong District, Kunming, Yunnan 650000, China
| | - Chuhan Shao
- Xiangya School of Medicine, Central South University, No. 172 Tongzipo Road, Yuelu District, Changsha, Hunan 410008, China
| | - Ling Shang
- Xiangya School of Medicine, Central South University, No. 172 Tongzipo Road, Yuelu District, Changsha, Hunan 410008, China
| | - Yue Hu
- Xiangya School of Medicine, Central South University, No. 172 Tongzipo Road, Yuelu District, Changsha, Hunan 410008, China
| | - Yishu Xu
- Xiangya School of Medicine, Central South University, No. 172 Tongzipo Road, Yuelu District, Changsha, Hunan 410008, China
| | - Yingluo Su
- Xiangya School of Medicine, Central South University, No. 172 Tongzipo Road, Yuelu District, Changsha, Hunan 410008, China
| | - Jiahui Yu
- Xiangya School of Medicine, Central South University, No. 172 Tongzipo Road, Yuelu District, Changsha, Hunan 410008, China
| | - Tianyi Feng
- Xiangya School of Medicine, Central South University, No. 172 Tongzipo Road, Yuelu District, Changsha, Hunan 410008, China
| | - Junyi Xie
- School of Life Science, Central South University, No. 172 Tongzipo Road, Yuelu District, Changsha, Hunan 410008, China
| | - Huijuan Xu
- School of Life Science, Central South University, No. 172 Tongzipo Road, Yuelu District, Changsha, Hunan 410008, China
| | - Zijun Zhang
- School of Life Science, Central South University, No. 172 Tongzipo Road, Yuelu District, Changsha, Hunan 410008, China
| | - Jiayi Peng
- School of Life Science, Central South University, No. 172 Tongzipo Road, Yuelu District, Changsha, Hunan 410008, China
| | - Jianbin Wu
- School of Life Science, Central South University, No. 172 Tongzipo Road, Yuelu District, Changsha, Hunan 410008, China
| | - Yuchang Zhang
- School of Life Science, Central South University, No. 172 Tongzipo Road, Yuelu District, Changsha, Hunan 410008, China
| | - Shaobo Zhu
- School of Life Science, Central South University, No. 172 Tongzipo Road, Yuelu District, Changsha, Hunan 410008, China
| | - Kun Xia
- MOE Key Laboratory of Pediatric Rare Diseases & Hunan Key Laboratory of Medical Genetics, Central South University, No. 110 Xiangya Road, Furong District, Changsha, Hunan 410008, China
| | - Beisha Tang
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital & Center for Medical Genetics, School of Life Sciences, Central South University, No. 87 Xiangya Road, Furong District, Changsha, Hunan 410008, China
- Department of Neurology & Multi-omics Research Center for Brain Disorders, The First Affiliated Hospital University of South China, 69 Chuan Shan Road, Shi Gu District, Hengyang, Hunan 421000, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Department of Neurology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Furong District, Changsha,Hunan 410008, China
| | - Guihu Zhao
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital & Center for Medical Genetics, School of Life Sciences, Central South University, No. 87 Xiangya Road, Furong District, Changsha, Hunan 410008, China
| | - Jinchen Li
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital & Center for Medical Genetics, School of Life Sciences, Central South University, No. 87 Xiangya Road, Furong District, Changsha, Hunan 410008, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Department of Neurology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Furong District, Changsha,Hunan 410008, China
- Bioinformatics Center, Furong Laboratory & Xiangya Hospital, Central South University, No. 87 Xiangya Road, Furong District, Changsha, Hunan 410008, China
| | - Bin Li
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital & Center for Medical Genetics, School of Life Sciences, Central South University, No. 87 Xiangya Road, Furong District, Changsha, Hunan 410008, China
| |
Collapse
|
23
|
Lin J, Mu Y, Liu L, Meng Y, Chen T, Fan X, Yuan J, Shen M, Pan J, Ren Y, Yu S, Chen Y. Machine learning based on multiplatform tests assists in subtype classification of mature B-cell neoplasms. Br J Haematol 2025; 206:224-234. [PMID: 39627967 DOI: 10.1111/bjh.19934] [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: 06/21/2024] [Accepted: 11/19/2024] [Indexed: 01/19/2025]
Abstract
Mature B-cell neoplasms (MBNs) are clonal proliferative diseases encompassing over 40 subtypes. The WHO classification (morphology, immunology, cytogenetics and molecular biology) provides comprehensive diagnostic understandings. However, MBN subtyping relies heavily on the expertise of clinicians and pathologists, and differences in clinical experience can lead to variations in subtyping efficiency and consistency. Additionally, due to the diversity in genetic backgrounds, machine learning (ML) models constructed based on Western populations may not be suitable for Chinese MBN patients. To construct a highly accurate classification model suitable for Chinese MBN patients, we first developed an ML model based on next-generation sequencing (NGS) from Chinese MBN patients, with an accuracy of 0.719, which decreased to 0.707 after model feature selection. Another ML model based on NGS and tumour cell size had an accuracy of 0.715, which increased to 0.763 after model feature selection. Both models were more accurate than models constructed using Western MBN patient databases. Furthermore, by adding flow cytometry for CD5 and CD10, the accuracy reached 0.864, which further improved to 0.872 after model feature selection. These models are accessible via an open-access website. Overall, ML models incorporating multiplatform tests can serve as practical auxiliary tools for MBN subtype classification.
Collapse
MESH Headings
- Lymphoma, B-Cell/classification
- Lymphoma, B-Cell/diagnosis
- Lymphoma, B-Cell/genetics
- Lymphoma, B-Cell/pathology
- Lymphoma, Follicular/classification
- Lymphoma, Follicular/diagnosis
- Lymphoma, Follicular/genetics
- Lymphoma, Follicular/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/classification
- Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Hairy Cell/classification
- Leukemia, Hairy Cell/diagnosis
- Leukemia, Hairy Cell/genetics
- Leukemia, Hairy Cell/pathology
- Waldenstrom Macroglobulinemia/classification
- Waldenstrom Macroglobulinemia/diagnosis
- Waldenstrom Macroglobulinemia/genetics
- Waldenstrom Macroglobulinemia/pathology
- Lymphoma, Mantle-Cell/classification
- Lymphoma, Mantle-Cell/diagnosis
- Lymphoma, Mantle-Cell/genetics
- Lymphoma, Mantle-Cell/pathology
- Machine Learning
- High-Throughput Nucleotide Sequencing
- Flow Cytometry
- CD5 Antigens/analysis
- Neprilysin/analysis
- Humans
- Immunohistochemistry
- Bone Marrow/pathology
- Mutation
- East Asian People/genetics
- Retrospective Studies
Collapse
Affiliation(s)
- Junwei Lin
- Guangzhou Medical University, Guangzhou, China
- Guangzhou KingMed Transformative Medicine Institute Co., Ltd., Guangzhou, China
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yafei Mu
- Guangzhou Medical University, Guangzhou, China
- Guangzhou KingMed Transformative Medicine Institute Co., Ltd., Guangzhou, China
| | - Lingling Liu
- Department of Hematology, The Third Affiliated Hospital of Sun Yat-sen University and Sun Yat-sen Institute of Hematology, Guangzhou, China
| | - Yuhuan Meng
- Guangzhou Medical University, Guangzhou, China
- Guangzhou KingMed Transformative Medicine Institute Co., Ltd., Guangzhou, China
| | - Tao Chen
- Guangzhou Medical University, Guangzhou, China
- Guangzhou KingMed Transformative Medicine Institute Co., Ltd., Guangzhou, China
| | - Xijie Fan
- Guangzhou KingMed Transformative Medicine Institute Co., Ltd., Guangzhou, China
| | - Jiecheng Yuan
- Guangzhou Medical University, Guangzhou, China
- Guangzhou KingMed Transformative Medicine Institute Co., Ltd., Guangzhou, China
| | - Maoting Shen
- Guangzhou Medical University, Guangzhou, China
- Guangzhou KingMed Transformative Medicine Institute Co., Ltd., Guangzhou, China
| | - Jianhua Pan
- Guangzhou Medical University, Guangzhou, China
- Guangzhou KingMed Center for Clinical Laboratory Co., Ltd., Guangzhou, China
- Guangzhou KingMed Diagnostics Group Co., Ltd., Guangzhou, China
| | - Yuxia Ren
- Guangzhou KingMed Center for Clinical Laboratory Co., Ltd., Guangzhou, China
| | - Shihui Yu
- Guangzhou Medical University, Guangzhou, China
- Guangzhou KingMed Transformative Medicine Institute Co., Ltd., Guangzhou, China
- Guangzhou KingMed Center for Clinical Laboratory Co., Ltd., Guangzhou, China
- Guangzhou KingMed Diagnostics Group Co., Ltd., Guangzhou, China
| | - Yuxin Chen
- Guangzhou Medical University, Guangzhou, China
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
- Guangzhou KingMed Center for Clinical Laboratory Co., Ltd., Guangzhou, China
| |
Collapse
|
24
|
Piriyapongsa J, Chumnumwat S, Kaewprommal P, Triparn K, Suvichapanich S, Udomsinprasert W, Jittikoon J, Shaw PJ, Nakhonsri V, Ngamphiw C, Wangkumhang P, Pithukpakorn M, Roothumnong E, Wiboonthanasarn S, Kuptanon C, Jinawath N, Porntaveetus T, Suriyaphol P, Viprakasit V, Pisitkun P, Kantaputra P, Tim-Aroon T, Wattanasirichaigoon D, Sura T, Suphapeetiporn K, Sripichai O, Khongphatthanayothin A, Fucharoen S, Ngamphaiboon N, Shotelersuk V, Mahasirimongkol S, Tongsima S. Pharmacogenomic landscape of the Thai population from genome sequencing of 949 individuals. Sci Rep 2024; 14:30683. [PMID: 39730427 DOI: 10.1038/s41598-024-79018-6] [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: 05/30/2023] [Accepted: 11/04/2024] [Indexed: 12/29/2024] Open
Abstract
Inter-individual variability in drug responses is significantly influenced by genetic factors, underscoring the importance of population-specific pharmacogenomic studies to optimize clinical outcomes. In this study, we analyzed whole genome sequencing data from 949 unrelated Thai individuals and conducted an in-depth analysis of 3239 genes involved in drug pharmacokinetics, pharmacodynamics, or immune-mediated adverse drug reactions. We identified 43 single nucleotide polymorphisms (SNPs), 134 diplotypes, and 15 human leukocyte antigen (HLA) alleles, all with moderate to high clinical significance. On average, each Thai individual carried 14 SNPs, one to two HLA alleles, and six diplotypes with actionable phenotypic associations. Clinically important diplotypes were present in over 20% of individuals for seven genes (CYP2A6, CYP2B6, CYP2C19, CYP3A5, NAT2, SLCO1B1, and VKORC1). In addition, clinically significant SNPs with allele frequencies exceeding 20% were identified among 15 genes, including VKORC1, CYP4F2, and ABCG2. We also identified 21,211 potentially deleterious variants among 3239 genes. Of these variants, 3746 were novel. The comprehensive dataset from this study serves as a valuable resource of pharmacogenomic variants in the Thai population, which will facilitate the development of personalized drug therapies and enhance patient care in Thailand.
Collapse
Affiliation(s)
- Jittima Piriyapongsa
- National Biobank of Thailand, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Supatat Chumnumwat
- Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Pavita Kaewprommal
- National Biobank of Thailand, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Kwankom Triparn
- National Biobank of Thailand, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | | | | | - Jiraphun Jittikoon
- Department of Biochemistry, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Philip J Shaw
- Medical Molecular Biotechnology Research Unit, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Vorthunju Nakhonsri
- National Biobank of Thailand, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Chumpol Ngamphiw
- National Biobank of Thailand, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Pongsakorn Wangkumhang
- National Biobank of Thailand, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Manop Pithukpakorn
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Ekkapong Roothumnong
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Supakit Wiboonthanasarn
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Chulaluck Kuptanon
- Department of Pediatrics, Queen Sirikit National Institute of Child Health, Bangkok, Thailand
- Department of Pediatrics, College of Medicine, Rangsit University, Pathum Thani, Thailand
| | - Natini Jinawath
- Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
- Integrative Computational BioScience Center (ICBS), Mahidol University, Nakhon Pathom, Thailand
| | - Thantrira Porntaveetus
- Center of Excellence in Genomics and Precision Dentistry, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Prapat Suriyaphol
- Office for Research and Development, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Vip Viprakasit
- Division of Hematology & Oncology, Department of Pediatrics & Siriraj Thalassemia Center, Siriraj Research Hospital, Mahidol University, Bangkok, Thailand
| | - Prapaporn Pisitkun
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Piranit Kantaputra
- Division of Pediatric Dentistry, Department of Orthodontics and Pediatric Dentistry, Center of Excellence in Medical Genetics Research, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - Thipwimol Tim-Aroon
- Division of Medical Genetics, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Duangrurdee Wattanasirichaigoon
- Division of Medical Genetics, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Thanyachai Sura
- Medical Genetics and Molecular Medicine Unit, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Kanya Suphapeetiporn
- Center of Excellence for Medical Genomics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Orapan Sripichai
- National Institute of Health, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, Thailand
| | - Apichai Khongphatthanayothin
- Division of Cardiology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Arrhythmia Research Chulalongkorn University, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Suthat Fucharoen
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Nuttapong Ngamphaiboon
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Vorasuk Shotelersuk
- Center of Excellence for Medical Genomics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Surakameth Mahasirimongkol
- Information and Communication Technology Center, Office of Permanent Secretary, Ministry of Public Health, Nonthaburi, Thailand.
| | - Sissades Tongsima
- National Biobank of Thailand, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand.
| |
Collapse
|
25
|
Gao Z, Liu Q, Zeng W, Jiang R, Wong WH. EpiGePT: a pretrained transformer-based language model for context-specific human epigenomics. Genome Biol 2024; 25:310. [PMID: 39696471 PMCID: PMC11657395 DOI: 10.1186/s13059-024-03449-7] [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: 03/27/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
The inherent similarities between natural language and biological sequences have inspired the use of large language models in genomics, but current models struggle to incorporate chromatin interactions or predict in unseen cellular contexts. To address this, we propose EpiGePT, a transformer-based model designed for predicting context-specific human epigenomic signals. By incorporating transcription factor activities and 3D genome interactions, EpiGePT outperforms existing methods in epigenomic signal prediction tasks, especially in cell-type-specific long-range interaction predictions and genetic variant impacts, advancing our understanding of gene regulation. A free online prediction service is available at http://health.tsinghua.edu.cn/epigept .
Collapse
Affiliation(s)
- Zijing Gao
- Ministry of Education Key Laboratory of Bioinformatics, Bioinformatics Division at the Beijing National Research Center for Information Science and Technology, Center for Synthetic and Systems Biology, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Qiao Liu
- Department of Statistics, Stanford University, CA, Stanford, 94305, USA.
| | - Wanwen Zeng
- Department of Statistics, Stanford University, CA, Stanford, 94305, USA
| | - Rui Jiang
- Ministry of Education Key Laboratory of Bioinformatics, Bioinformatics Division at the Beijing National Research Center for Information Science and Technology, Center for Synthetic and Systems Biology, Department of Automation, Tsinghua University, Beijing, 100084, China.
| | - Wing Hung Wong
- Department of Statistics, Stanford University, CA, Stanford, 94305, USA.
- Department of Biomedical Data Science, Bio-X Program, Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
26
|
Gogate A, Kaur K, Khalil R, Bashtawi M, Morris MA, Goodspeed K, Evans P, Chahrour MH. The genetic landscape of autism spectrum disorder in an ancestrally diverse cohort. NPJ Genom Med 2024; 9:62. [PMID: 39632905 PMCID: PMC11618689 DOI: 10.1038/s41525-024-00444-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 10/28/2024] [Indexed: 12/07/2024] Open
Abstract
Autism spectrum disorder (ASD) comprises neurodevelopmental disorders with wide variability in genetic causes and phenotypes, making it challenging to pinpoint causal genes. We performed whole exome sequencing on a modest, ancestrally diverse cohort of 195 families, including 754 individuals (222 with ASD), and identified 38,834 novel private variants. In 68 individuals with ASD (~30%), we identified 92 potentially pathogenic variants in 73 known genes, including BCORL1, CDKL5, CHAMP1, KAT6A, MECP2, and SETD1B. Additionally, we identified 158 potentially pathogenic variants in 120 candidate genes, including DLG3, GABRQ, KALRN, KCTD16, and SLC8A3. We also found 34 copy number variants in 31 individuals overlapping known ASD loci. Our work expands the catalog of ASD genetics by identifying hundreds of variants across diverse ancestral backgrounds, highlighting convergence on nervous system development and signal transduction. These findings provide insights into the genetic underpinnings of ASD and inform molecular diagnosis and potential therapeutic targets.
Collapse
Affiliation(s)
- Ashlesha Gogate
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Kiran Kaur
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Raida Khalil
- Department of Biotechnology and Genetic Engineering, Faculty of Science, University of Philadelphia, Amman, Jordan
| | - Mahmoud Bashtawi
- Department of Psychiatry, Jordan University of Science and Technology, King Abdullah University Hospital, Ramtha, Jordan
| | - Mary Ann Morris
- UT Southwestern and Children's Health Center for Autism Care, Children's Medical Center Dallas, Dallas, TX, 75247, USA
| | - Kimberly Goodspeed
- UT Southwestern and Children's Health Center for Autism Care, Children's Medical Center Dallas, Dallas, TX, 75247, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Patricia Evans
- UT Southwestern and Children's Health Center for Autism Care, Children's Medical Center Dallas, Dallas, TX, 75247, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Maria H Chahrour
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
27
|
Shi Q, Dai M, Ma Y, Liu J, Liu X, Wang XJ. DRED: A Comprehensive Database of Genes Related to Repeat Expansion Diseases. GENOMICS, PROTEOMICS & BIOINFORMATICS 2024; 22:qzae068. [PMID: 39348154 PMCID: PMC11696699 DOI: 10.1093/gpbjnl/qzae068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 09/13/2024] [Accepted: 09/25/2024] [Indexed: 10/01/2024]
Abstract
Expansion of tandem repeats in genes often causes severe diseases, such as fragile X syndrome, Huntington's disease, and spinocerebellar ataxia. However, information on genes associated with repeat expansion diseases is scattered throughout the literature, systematic prediction of potential genes that may cause diseases via repeat expansion is also lacking. Here, we develop DRED, a Database of genes related to Repeat Expansion Diseases, as a manually-curated database that covers all known 61 genes related to repeat expansion diseases reported in PubMed and OMIM, along with the detailed repeat information for each gene. DRED also includes 516 genes with the potential to cause diseases via repeat expansion, which were predicted based on their repeat composition, genetic variations, genomic features, and disease associations. Various types of information on repeat expansion diseases and their corresponding genes/repeats are presented in DRED, together with links to external resources, such as NCBI and ClinVar. DRED provides user-friendly interfaces with comprehensive functions, and can serve as a central data resource for basic research and repeat expansion disease-related medical diagnosis. DRED is freely accessible at http://omicslab.genetics.ac.cn/dred, and will be frequently updated to include newly reported genes related to repeat expansion diseases.
Collapse
Affiliation(s)
- Qingqing Shi
- Key Laboratory of Genetic Network Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Min Dai
- Key Laboratory of Genetic Network Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yingke Ma
- Key Laboratory of Genetic Network Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jun Liu
- Key Laboratory of Genetic Network Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiuying Liu
- Key Laboratory of Genetic Network Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiu-Jie Wang
- Key Laboratory of Genetic Network Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
28
|
Murali H, Wang P, Liao EC, Wang K. Genetic variant classification by predicted protein structure: A case study on IRF6. Comput Struct Biotechnol J 2024; 23:892-904. [PMID: 38370976 PMCID: PMC10869248 DOI: 10.1016/j.csbj.2024.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/20/2024] Open
Abstract
Next-generation genome sequencing has revolutionized genetic testing, identifying numerous rare disease-associated gene variants. However, to impute pathogenicity, computational approaches remain inadequate and functional testing of gene variant is required to provide the highest level of evidence. The emergence of AlphaFold2 has transformed the field of protein structure determination, and here we outline a strategy that leverages predicted protein structure to enhance genetic variant classification. We used the gene IRF6 as a case study due to its clinical relevance, its critical role in cleft lip/palate malformation, and the availability of experimental data on the pathogenicity of IRF6 gene variants through phenotype rescue experiments in irf6-/- zebrafish. We compared results from over 30 pathogenicity prediction tools on 37 IRF6 missense variants. IRF6 lacks an experimentally derived structure, so we used predicted structures to explore associations between mutational clustering and pathogenicity. We found that among these variants, 19 of 37 were unanimously predicted as deleterious by computational tools. Comparing in silico predictions with experimental findings, 12 variants predicted as pathogenic were experimentally determined as benign. Even with the recently published AlphaMissense model, 15/18 (83%) of the predicted pathogenic variants were experimentally determined as benign. In comparison, mapping variants to the protein revealed deleterious mutation clusters around the protein binding domain, whereas N-terminal variants tend to be benign, suggesting the importance of structural information in determining pathogenicity of mutations in this gene. In conclusion, incorporating gene-specific structural features of known pathogenic/benign mutations may provide meaningful insights into pathogenicity predictions in a gene-specific manner and facilitate the interpretation of variant pathogenicity.
Collapse
Affiliation(s)
- Hemma Murali
- Graduate Program in Biochemistry and Molecular Biophysics, University of Pennsylvania, Philadelphia, PA 19104, United States
- Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - Peng Wang
- Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, United States
- Master of Biotechnology Program, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Eric C. Liao
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
- Center for Craniofacial Innovation, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - Kai Wang
- Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, United States
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| |
Collapse
|
29
|
Thompson MD, Reiner-Link D, Berghella A, Rana BK, Rovati GE, Capra V, Gorvin CM, Hauser AS. G protein-coupled receptor (GPCR) pharmacogenomics. Crit Rev Clin Lab Sci 2024; 61:641-684. [PMID: 39119983 DOI: 10.1080/10408363.2024.2358304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/03/2023] [Accepted: 05/18/2024] [Indexed: 08/10/2024]
Abstract
The field of pharmacogenetics, the investigation of the influence of one or more sequence variants on drug response phenotypes, is a special case of pharmacogenomics, a discipline that takes a genome-wide approach. Massively parallel, next generation sequencing (NGS), has allowed pharmacogenetics to be subsumed by pharmacogenomics with respect to the identification of variants associated with responders and non-responders, optimal drug response, and adverse drug reactions. A plethora of rare and common naturally-occurring GPCR variants must be considered in the context of signals from across the genome. Many fundamentals of pharmacogenetics were established for G protein-coupled receptor (GPCR) genes because they are primary targets for a large number of therapeutic drugs. Functional studies, demonstrating likely-pathogenic and pathogenic GPCR variants, have been integral to establishing models used for in silico analysis. Variants in GPCR genes include both coding and non-coding single nucleotide variants and insertion or deletions (indels) that affect cell surface expression (trafficking, dimerization, and desensitization/downregulation), ligand binding and G protein coupling, and variants that result in alternate splicing encoding isoforms/variable expression. As the breadth of data on the GPCR genome increases, we may expect an increase in the use of drug labels that note variants that significantly impact the clinical use of GPCR-targeting agents. We discuss the implications of GPCR pharmacogenomic data derived from the genomes available from individuals who have been well-phenotyped for receptor structure and function and receptor-ligand interactions, and the potential benefits to patients of optimized drug selection. Examples discussed include the renin-angiotensin system in SARS-CoV-2 (COVID-19) infection, the probable role of chemokine receptors in the cytokine storm, and potential protease activating receptor (PAR) interventions. Resources dedicated to GPCRs, including publicly available computational tools, are also discussed.
Collapse
Affiliation(s)
- Miles D Thompson
- Krembil Brain Institute, Toronto Western Hospital, Toronto, Ontario, Canada
| | - David Reiner-Link
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Alessandro Berghella
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Brinda K Rana
- Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| | - G Enrico Rovati
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Valerie Capra
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Caroline M Gorvin
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, United Kingdom
| | - Alexander S Hauser
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
30
|
Singh V, Katiyar A, Malik P, Kumar S, Mohan A, Singh H, Jain D. Identification of molecular biomarkers associated with non-small-cell lung carcinoma (NSCLC) using whole-exome sequencing. Cancer Biomark 2024; 41:CBM220211. [PMID: 37694353 DOI: 10.3233/cbm-220211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
ObjectivesSignificant progress has been made in the treatment of patients with pulmonary adenocarcinoma (ADCA) based on molecular profiling. However, no such molecular target exists for squamous cell carcinoma (SQCC). An exome sequence may provide new markers for personalized medicine for lung cancer patients of all subtypes. The current study aims to discover new genetic markers that can be used as universal biomarkers for non-small cell lung cancer (NSCLC).MethodsWES of 19 advanced NSCLC patients (10 ADCA and 9 SQCC) was performed using Illumina HiSeq 2000. Variant calling was performed using GATK HaplotypeCaller and then the impacts of variants on protein structure or function were predicted using SnpEff and ANNOVAR. The clinical impact of somatic variants in cancer was assessed using cancer archives. Somatic variants were further prioritized using a knowledge-driven variant interpretation approach. Sanger sequencing was used to validate functionally important variants.ResultsWe identified 24 rare single-nucleotide variants (SNVs) including 17 non-synonymous SNVs, and 7 INDELs in 18 genes possibly linked to lung carcinoma. Variants were classified as known somatic (n = 10), deleterious (n = 8), and variant of uncertain significance (n = 6). We found TBP and MPRIP genes exclusively associated with ADCA subtypes, FBOX6 with SQCC subtypes and GPRIN2, KCNJ18 and TEKT4 genes mutated in all the patients. The Sanger sequencing of 10 high-confidence somatic SNVs showed 100% concordance in 7 genes, and 80% concordance in the remaining 3 genes.ConclusionsOur bioinformatics analysis identified KCNJ18, GPRIN2, TEKT4, HRNR, FOLR3, ESSRA, CTBP2, MPRIP, TBP, and FBXO6 may contribute to progression in NSCLC and could be used as new biomarkers for the treatment. The mechanism by which GPRIN2, KCNJ12, and TEKT4 contribute to tumorigenesis is unclear, but our results suggest they may play an important role in NSCLC and it is worth investigating in future.
Collapse
Affiliation(s)
- Varsha Singh
- Department of Pathology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Amit Katiyar
- Bioinformatics Facility, Centralized Core Research Facility, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Prabhat Malik
- Department of Medical Oncology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Sunil Kumar
- Department of Surgical Oncology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Anant Mohan
- Department of Pulmonary Critical Care & Sleep Medicine, All India Institute of Medical Sciences, New Delhi, Ansari Nagar, India
| | - Harpreet Singh
- ICMR-AIIMS Computational Genomics Center, Division of Biomedical Informatics, Indian Council of Medical Research, Ansari Nagar, New Delhi, India
| | - Deepali Jain
- Department of Pathology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| |
Collapse
|
31
|
Lim JH, Shin HT, Park S, Ryu WK, Kim L, Lee KH, Ko SM, Lee SJ, Kim JS, Ryu JS. Bronchial washing fluid sequencing is useful in the diagnosis of lung cancer with necrotic tumor. Transl Oncol 2024; 50:102134. [PMID: 39353233 PMCID: PMC11472095 DOI: 10.1016/j.tranon.2024.102134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/27/2024] [Accepted: 09/19/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Early-stage lung cancers detected by low-dose computed tomography (CT) often require confirmation through invasive procedures due to the absence of endobronchial lesions. This study assesses the diagnostic utility of bronchial washing fluid (BW) sequencing, a less invasive alternative, aiming to identify patient characteristics most suited for this approach. METHODS From June 2017 to March 2018, we conducted a prospective cohort study by enrolling patients with incidental lung lesions suspected of early-stage lung cancer at two independent hospitals, and 114 were diagnosed with lung cancer while 50 were diagnosed with benign lesions. BW sequencing was performed using a targeted gene panel, and the clinical characteristics of patients detected with cancer through sequencing were identified. RESULTS Malignant cells were detected in 33 patients (28.9 %) through BW cytology. By applying specificity-focused mutation criteria, BW sequencing classified 42 patients (36.8 %) as having cancer. Among the cancer patients who were BW sequencing positive and BW cytology negative, 15 patients (75.0 %) showed necrosis on CT. The sensitivity of BW sequencing was particularly enhanced in patients with necrotic tumors, reaching 75 %. CONCLUSIONS BW sequencing presents a viable, non-invasive diagnostic option for early-stage lung cancer, especially valuable in patients with necrotic lesions. By potentially reducing the reliance on more invasive diagnostic procedures, this method could streamline clinical workflows, decrease patient burden, and improve overall diagnostic efficiency.
Collapse
Affiliation(s)
- Jun Hyeok Lim
- Department of Internal Medicine, Inha University Hospital, Inha University College of Medicine, Incheon, South Korea
| | - Hyun-Tae Shin
- Research Center for Controlling Intercellular Communication (RCIC), Inha University School of Medicine, Inha University, Incheon, South Korea; Department of Dermatology, Inha University Hospital, Inha University College of Medicine, Incheon, South Korea
| | - Sunmin Park
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Woo Kyung Ryu
- Department of Internal Medicine, Inha University Hospital, Inha University College of Medicine, Incheon, South Korea
| | - Lucia Kim
- Department of Pathology, Inha University Hospital, Inha University College of Medicine, Incheon, South Korea
| | - Kyung-Hee Lee
- Department of Radiology, Inha University Hospital, Inha University College of Medicine, Incheon, South Korea
| | - Sung Min Ko
- Department of Radiology, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | | | - Jung Soo Kim
- Department of Internal Medicine, Inha University Hospital, Inha University College of Medicine, Incheon, South Korea
| | - Jeong-Seon Ryu
- Department of Internal Medicine, Inha University Hospital, Inha University College of Medicine, Incheon, South Korea.
| |
Collapse
|
32
|
Vorsteveld EE, Van der Made CI, Smeekens SP, Schuurs-Hoeijmakers JH, Astuti G, Diepstra H, Gilissen C, Hoenselaar E, Janssen A, van Roozendaal K, Engelen JSV, Steyaert W, Weiss MM, Yntema HG, Mantere T, AlZahrani MS, van Aerde K, Derfalvi B, Faqeih EA, Henriet SSV, van Hoof E, Idressi E, Issekutz TB, Jongmans MCJ, Keski-Filppula R, Krapels I, Te Loo M, Mulders-Manders CM, Ten Oever J, Potjewijd J, Sarhan NT, Slot MC, Terhal PA, Thijs H, Vandersteen A, Vanhoutte EK, van de Veerdonk F, van Well G, Netea MG, Simons A, Hoischen A. Clinical exome sequencing data from patients with inborn errors of immunity: Cohort level diagnostic yield and the benefit of systematic reanalysis. Clin Immunol 2024; 268:110375. [PMID: 39369972 DOI: 10.1016/j.clim.2024.110375] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/08/2024]
Abstract
While next generation sequencing has expanded the scientific understanding of Inborn Errors of Immunity (IEI), the clinical use and re-use of exome sequencing is still emerging. We revisited clinical exome data from 1300 IEI patients using an updated in silico IEI gene panel. Variants were classified and curated through expert review. The molecular diagnostic yield after standard exome analysis was 11.8 %. Through systematic reanalysis, we identified variants of interest in 5.2 % of undiagnosed patients, with 76.7 % being (candidate) disease-causing, providing a (candidate) diagnosis in 15.2 % of our cohort. We find a 1.7 percentage point increase in conclusive molecular diagnoses. We find a high degree of actionability in patients with a genetic diagnosis (76.4 %). Despite the modest absolute diagnostic gain, these data support the benefit of iterative exome reanalysis in IEI patients, conveying the notion that our current understanding of genes and variants involved in IEI is by far not saturated.
Collapse
Affiliation(s)
- Emil E Vorsteveld
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands; RadboudUMC Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Caspar I Van der Made
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands; RadboudUMC Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Sanne P Smeekens
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Galuh Astuti
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands; RadboudUMC Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Heleen Diepstra
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Christian Gilissen
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands; RadboudUMC Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Evelien Hoenselaar
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Alice Janssen
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kees van Roozendaal
- Department of Clinical Genetics, Maastricht University Medical Center+, Maastricht, The Netherlands
| | | | - Wouter Steyaert
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands; RadboudUMC Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marjan M Weiss
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Helger G Yntema
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands; Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tuomo Mantere
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands; Laboratory of Cancer Genetics and Tumor Biology, Translational Medicine Research Unit and Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Mofareh S AlZahrani
- Department of Pediatrics, Children's specialist Hospital, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Koen van Aerde
- Department of Paediatrics, Amalia Children's Hospital, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Beata Derfalvi
- Division of Immunology, Department of Pediatrics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Eissa Ali Faqeih
- Section of Medical Genetics, Children's Specialist Hospital, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Stefanie S V Henriet
- RadboudUMC Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Pediatric Infectious Diseases and Immunology, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Elise van Hoof
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Eman Idressi
- Department of Pediatrics, Children's specialist Hospital, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Thomas B Issekutz
- Department of Pediatrics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Marjolijn C J Jongmans
- Princess Máxima Center for Pediatric Oncology and Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Riikka Keski-Filppula
- PEDEGO Research Unit, University of Oulu, Oulu, Finland; Department of Clinical Genetics, Oulu University Hospital, Oulu, Finland; Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Ingrid Krapels
- Department of Clinical Genetics, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Maroeska Te Loo
- Department of Pediatric Hematology, Amalia children's hospital, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Catharina M Mulders-Manders
- Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands; Radboud Expertise Center for Immunodeficiency and Autoinflammation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jaap Ten Oever
- RadboudUMC Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands; Radboud Centre for Infectious Diseases (RCI), Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Judith Potjewijd
- Department of Internal Medicine, Division of Experimental and Clinical Immunology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Nora Tarig Sarhan
- Section of Medical Genetics, Children's Specialist Hospital, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Marjan C Slot
- Department of Allergology and Clinical Immunology, Maastricht UMC+, Maastricht, The Netherlands
| | - Paulien A Terhal
- Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Herman Thijs
- Department of Pediatrics, Gelre Ziekenhuizen Zutphen, The Netherlands
| | - Anthony Vandersteen
- Division of Medical Genetics, Department of Pediatrics, Dalhousie University, Halifax, Nova Scotia, Canada; Maritime Medical Genetics Service, IWK Health Centre, Halifax, Nova Scotia, Canada
| | - Els K Vanhoutte
- Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Frank van de Veerdonk
- Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands; Radboud Centre for Infectious Diseases (RCI), Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Gijs van Well
- Department of Paediatrics, Maastricht University Medical Center, MosaKids Children's Hospital, Maastricht, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands; Radboud Centre for Infectious Diseases (RCI), Radboud University Medical Centre, Nijmegen, The Netherlands; Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Annet Simons
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Alexander Hoischen
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands; RadboudUMC Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands; Radboud Expertise Center for Immunodeficiency and Autoinflammation, Radboud University Medical Center, Nijmegen, The Netherlands; Radboud Centre for Infectious Diseases (RCI), Radboud University Medical Centre, Nijmegen, The Netherlands.
| |
Collapse
|
33
|
Fang J, Yang X, Tang M, Li S, Han F, Zhou L, Li M, Yang M, Cui L, Zhang S, Zhu Y, Yao M, Ni J. Rare RNF213 variants is related to early-onset intracranial atherosclerosis: A Chinese community-based study. J Stroke Cerebrovasc Dis 2024; 33:107982. [PMID: 39233284 DOI: 10.1016/j.jstrokecerebrovasdis.2024.107982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/06/2024] [Accepted: 08/27/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND The relationship between rare variants in Ring finger protein 213 (RNF213) and intracranial atherosclerosis (ICAS) remained unelucidated. Using whole-exome sequencing (WES) and high-resolution magnetic resonance imaging (HR-MRI), this study aimed at investigating the association between rare RNF213 variants and ICAS within a Chinese community-dwelling population. METHODS The present study included 821 participants from Shunyi cohort. Genetic data of rare RNF213 variants were acquired by WES and were categorized by functional domains. Intracranial and extracranial atherosclerosis were assessed by brain HR-MRI and carotid ultrasound, respectively. Logistic regression and generalized linear regression were applied to evaluate the effects of rare RNF213 variants on atherosclerosis. Stratification by age were conducted with 50 years old set as the cutoff value. RESULTS Ninety-five participants were identified as carriers of rare RNF213 variants. Carotid plaques were observed in 367 (44.7 %) participants, while ICAS was identified in 306 (37.3 %). Rare variants of RNF213 was not associated with ECAS. Employing HR-MRI, both the presence of rare variants (β = 0.150, P = 0.025) and numerical count of variants (β = 0.182, P = 0.003) were significantly correlated with ICAS within the group of age ≤50 years. Both variant existence (β = 0.154, P = 0.014) and variant count (β = 0.188, P = 0.003) were significantly associated with plaques in middle cerebral arteries within younger subgroup, rather than basilar arteries. Furthermore, a significant association was observed between variants that located outside the N-arm domain and ICAS in the younger subgroup (OR = 2.522, P = 0.030). Statistical results remained robust after adjusted for age, gender, and cardiovascular risk factors. CONCLUSIONS Rare variants of RNF213 is associated with age-related ICAS in general Chinese population, highlighting the potential role of RNF213 as a genetic contributor to early-onset ICAS.
Collapse
Affiliation(s)
- Jianxun Fang
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Xinzhuang Yang
- Center for bioinformatics, National Infrastructures for Translational Medicine, Institute of Clinical Medicine & Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Mingyu Tang
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Shengde Li
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Fei Han
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Lixin Zhou
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Mingli Li
- Department of Radiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Meng Yang
- Department of Ultrasound, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Liying Cui
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Shuyang Zhang
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Yicheng Zhu
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Ming Yao
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China.
| | - Jun Ni
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China.
| |
Collapse
|
34
|
Kim JH, Shin JY, Park SY, Seo SS, Kang S, Yoo CW, Park SY, Lim MC. The discrepancy of somatic BRCA1/2 pathogenic variants from two different platforms in epithelial ovarian, fallopian tube, and peritoneal cancer. Sci Rep 2024; 14:25879. [PMID: 39468117 PMCID: PMC11519512 DOI: 10.1038/s41598-024-75230-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/03/2024] [Indexed: 10/30/2024] Open
Abstract
The somatic BRCA1 or BRCA2 Pathogenic Variant (PV)/Likely PV (LPV) from Next Generation Sequencing (NGS) is the most important biomarker for PARP inhibitor use and maintenance-targeted therapies. A discrepancy in the detection rates of BRCA1 and BRCA2 PV/LPV was identified among the NGS platforms. The objective of this study was to compare the somatic BRCA results from two distinct platforms using the same cohort and to identify the causes of these differences. Patients with epithelial ovarian cancer who concurrently underwent tumor NGS using two different platforms between January 2022 and June 2023 were included in this study. The two platforms used were in-house tumor NGS (Illumina NextSeq 550Dx, SureSelectXT library kit, and datasets from 1000 Genomes, ESP6500, ExAC, and ClinVar) and GreenPlan homologous recombination deficiency (HRD) test (Illumina NextSeq 550Dx, customized Twist Bioscience library kit, and datasets from COSMIC and OncoKB). The results of somatic mutations in BRCA1 and BRCA2 were compared between the two platforms. Of the 118 patients, 11.9% (n = 14) exhibited a discordant interpretation of BRCA1 or BRCA2 between the two platforms. Eleven patients (9.3%) exhibited negative results in the in-house platform but positive results (eight seven as PV of BRCA1, one as PV of BRCA2, one as LPV of BRCA1, and two as LPV of BRCA2) in the GreenPlan HRD test, while three patients (2.6%) had positive BRCA pathogenic variants (two as PV of BRCA1 [c.3340G > T, c.5152 + 3 A > C], one as LPV of BRCA2 [c.8174G > T], and one as LPV of BRCA1 [c.5017_5019delCAC]) in the in-house platform but a negative result in the GreenPlan HRD test. The discordance rate of somatic BRCA1 or BRCA2 mutations from different platforms was approximately 12%. In the case of the strong implication of BRCA PV/LPV with a negative result with one genetic test, different platforms could be considered in limited cases. Careful interpretation and further studies for the cross-validation of gene analysis platforms are needed.
Collapse
Affiliation(s)
- Ji Hyun Kim
- Center for Gynecologic Cancer, National Cancer Center, Goyang, Korea
| | - Jun-Young Shin
- Department of Pathology, National Cancer Center Korea, Goyang, Korea
| | - Seog-Yun Park
- Department of Pathology, National Cancer Center Korea, Goyang, Korea
| | - Sang-Soo Seo
- Center for Gynecologic Cancer, National Cancer Center, Goyang, Korea
| | - Sokbom Kang
- Center for Gynecologic Cancer, National Cancer Center, Goyang, Korea
- Department of Pathology, National Cancer Center Korea, Goyang, Korea
- Department of Cancer Control and Policy, National Cancer Center Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
| | - Chong Woo Yoo
- Department of Pathology, National Cancer Center Korea, Goyang, Korea
| | - Sang-Yoon Park
- Center for Gynecologic Cancer, National Cancer Center, Goyang, Korea
| | - Myong Cheol Lim
- Center for Gynecologic Cancer, National Cancer Center, Goyang, Korea.
- Department of Cancer Control and Policy, National Cancer Center Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea.
- Division of Rare and Refractory Cancer, Research Institute, National Cancer Center, Goyang, Korea.
- Center for Clinical Trial, Hospital, National Cancer Center, Goyang, Korea.
| |
Collapse
|
35
|
Lee U, Szabova L, Collins VJ, Gordon M, Johnson K, Householder D, Jorgensen S, Lu L, Bassel L, Elloumi F, Peer CJ, Nelson AE, Varriano S, Varma S, Roberts RD, Ohler ZW, Figg WD, Sharan SK, Pommier Y, Heske CM. Treatment with novel topoisomerase inhibitors in Ewing sarcoma models reveals heterogeneity of tumor response. Front Cell Dev Biol 2024; 12:1462840. [PMID: 39512899 PMCID: PMC11542432 DOI: 10.3389/fcell.2024.1462840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 09/30/2024] [Indexed: 11/15/2024] Open
Abstract
Introduction The topoisomerase 1 (TOP1) inhibitor irinotecan is a standard-of-care agent for relapsed Ewing sarcoma (EWS), but its efficacy is limited by chemical instability, rapid clearance and reversibility, and dose-limiting toxicities, such as diarrhea. Indenoisoquinolines (IIQs) represent a new class of clinical TOP1 inhibitors designed to address these limitations. Methods In this study, we evaluated the preclinical efficacy of three IIQs (LMP400, LMP744, and LMP776) in relevant models of EWS. We characterized the pharmacokinetics of IIQs in orthotopic xenograft models of EWS, optimized the dosing regimen through tolerability studies, and tested the efficacy of IIQs in a panel of six molecularly heterogeneous EWS patient-derived xenograft (PDX) models. For each PDX, we conducted whole genome and RNA sequencing, and methylation analysis. Results We show that IIQs potently inhibit the proliferation of EWS cells in vitro, inducing complete cell growth inhibition at nanomolar concentrations via induction of DNA damage and apoptotic cell death. LMP400 treatment induced ≥30% tumor regression in two of six PDX models, with more durable regression compared to irinotecan treatment in one of these models. RNA sequencing of PDX models identified a candidate predictive biomarker gene signature for LMP400 response. These data, along with pharmacogenomic data on IIQs in sarcoma cell lines, are available at a new interactive public website: https://discover.nci.nih.gov/rsconnect/EwingSarcomaMinerCDB/. Discussion Our findings suggest that IIQs may be promising new agents for a subset of EWS patients.
Collapse
Affiliation(s)
- Unsun Lee
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Ludmila Szabova
- Center for Advanced Preclinical Research, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, United States
| | - Victor J. Collins
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Melanie Gordon
- Center for Advanced Preclinical Research, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, United States
| | - Kristine Johnson
- Center for Advanced Preclinical Research, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, United States
| | - Deborah Householder
- Center for Advanced Preclinical Research, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, United States
| | - Stephanie Jorgensen
- Center for Advanced Preclinical Research, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, United States
| | - Lucy Lu
- Center for Advanced Preclinical Research, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, United States
| | - Laura Bassel
- Center for Advanced Preclinical Research, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, United States
| | - Fathi Elloumi
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Cody J. Peer
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Ariana E. Nelson
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Sophia Varriano
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Sudhir Varma
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Ryan D. Roberts
- Center for Childhood Cancer Research, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH, United States
- Department of Hematology, Oncology and Bone Marrow Transplant, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Zoe Weaver Ohler
- Center for Advanced Preclinical Research, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, United States
| | - William D. Figg
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Shyam K. Sharan
- Center for Advanced Preclinical Research, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, United States
| | - Yves Pommier
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Christine M. Heske
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
36
|
García S A, Costa M, Perez A, Pastor O. CardioGraph: a platform to study variations associated with familiar cardiopathies. BMC Med Inform Decis Mak 2024; 23:303. [PMID: 39434095 PMCID: PMC11494761 DOI: 10.1186/s12911-024-02700-2] [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: 12/20/2022] [Accepted: 09/26/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND Familiar cardiopathies are genetic disorders that affect the heart. Cardiologists face a significant problem when treating patients suffering from these disorders: most DNA variations are novel (i.e., they have not been classified before). To facilitate the analysis of novel variations, we present CardioGraph, a platform specially designed to support the analysis of novel variations and help determine whether they are relevant for diagnosis. To do this, CardioGraph identifies and annotates the consequence of variations and provides contextual information regarding which heart structures, pathways, and biological processes are potentially affected by those variations. METHODS We conducted our work through three steps. First, we define a data model to support the representation of the heterogeneous information. Second, we instantiate this data model to integrate and represent all the genomics knowledge available for familiar cardiopathies. In this step, we consider genomic data sources and the scientific literature. Third, the design and implementation of the CardioGraph platform. A three-tier structure was used: the database, the backend, and the frontend. RESULTS Three main results were obtained: the data model, the knowledge base generated with the instantiation of the data model, and the platform itself. The platform code has been included as supplemental material in this manuscript. Besides, an instance is publicly available in the following link: https://genomics-hub.pros.dsic.upv.es:3090 . CONCLUSION CardioGraph is a platform that supports the analysis of novel variations. Future work will expand the body of knowledge about familiar cardiopathies and include new information about hotspots, functional studies, and previously reported variations.
Collapse
Affiliation(s)
- Alberto García S
- PROS Research Center, VRAIN Research Institute, Universitat Politècnica de València, Camino de Vera, Valencia, Spain.
| | - Mireia Costa
- PROS Research Center, VRAIN Research Institute, Universitat Politècnica de València, Camino de Vera, Valencia, Spain
| | - Ana Perez
- PROS Research Center, VRAIN Research Institute, Universitat Politècnica de València, Camino de Vera, Valencia, Spain
| | - Oscar Pastor
- PROS Research Center, VRAIN Research Institute, Universitat Politècnica de València, Camino de Vera, Valencia, Spain
| |
Collapse
|
37
|
Wong GYM, Li J, McKay M, Castaneda M, Bhimani N, Diakos C, Hugh TJ, Molloy MP. Proteogenomic Characterization of Early Intrahepatic Recurrence after Curative-Intent Treatment of Colorectal Liver Metastases. J Proteome Res 2024; 23:4523-4537. [PMID: 39264718 DOI: 10.1021/acs.jproteome.4c00440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2024]
Abstract
Clinical and pathological factors are insufficient to accurately identify patients at risk of early recurrence after curative-intent treatment of colorectal liver metastases (CRLM). This study aimed to identify candidate prognostic proteogenomic biomarkers for early intrahepatic recurrence after curative-intent resection of CRLM. Patients diagnosed with intrahepatic recurrence within 6 months of liver resection were categorized as the "early recurrence" group, while those who achieved a recurrence-free status for 10 years were designated as "durable remission". Comprehensive genomic and proteomic profiling of fresh frozen samples from these prognostically distinct groups was performed using the TruSight Oncology 500 assay and label-free data-dependent acquisition liquid chromatography-mass spectrometry. Genetic alterations were identified in 117 of the 523 profiled genes in patients with early recurrence. The most common somatic mutations linked to early recurrence were TP53 (88%), APC (71%), KRAS (38%), and SMAD4 (21%). SMAD4 alterations were absent in samples from patients with a durable remission. Calponin-2, versican core protein, glutathione peroxidase 3, fibulin-5, and amyloid-β precursor protein were upregulated more than 2-fold in early recurrence. Exploratory analysis of these proteogenomic biomarkers suggests that SMAD4, calponin-2, and glutathione peroxidase 3 may have the potential to predict early recurrence, enabling improved prognostication and precision oncology in CRLM.
Collapse
Affiliation(s)
- Geoffrey Yuet Mun Wong
- Department of Upper Gastrointestinal Surgery, Royal North Shore Hospital, Sydney, New South Wales 2065, Australia
- Northern Clinical School, The University of Sydney, Sydney, New South Wales 2065, Australia
- Bowel Cancer and Biomarker Research Laboratory, Kolling Institute, St Leonards, New South Wales 2065, Australia
| | - Jun Li
- Bowel Cancer and Biomarker Research Laboratory, Kolling Institute, St Leonards, New South Wales 2065, Australia
| | - Matthew McKay
- Bowel Cancer and Biomarker Research Laboratory, Kolling Institute, St Leonards, New South Wales 2065, Australia
| | - Miguel Castaneda
- Bowel Cancer and Biomarker Research Laboratory, Kolling Institute, St Leonards, New South Wales 2065, Australia
| | - Nazim Bhimani
- Department of Upper Gastrointestinal Surgery, Royal North Shore Hospital, Sydney, New South Wales 2065, Australia
- Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales 2050, Australia
| | - Connie Diakos
- Northern Clinical School, The University of Sydney, Sydney, New South Wales 2065, Australia
- Department of Medical Oncology, Royal North Shore Hospital, Sydney, New South Wales 2065, Australia
| | - Thomas J Hugh
- Department of Upper Gastrointestinal Surgery, Royal North Shore Hospital, Sydney, New South Wales 2065, Australia
- Northern Clinical School, The University of Sydney, Sydney, New South Wales 2065, Australia
| | - Mark P Molloy
- Bowel Cancer and Biomarker Research Laboratory, Kolling Institute, St Leonards, New South Wales 2065, Australia
| |
Collapse
|
38
|
Li YL, Lin J, Huang X, Zeng RH, Zhang G, Xu JN, Lin KJ, Chen XS, He MF, Qiao JD, Cheng X, Zhu D, Xiong ZQ, Chen WJ. Heterozygous Variants in KCNJ10 Cause Paroxysmal Kinesigenic Dyskinesia Via Haploinsufficiency. Ann Neurol 2024; 96:758-773. [PMID: 38979912 DOI: 10.1002/ana.27018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 06/07/2024] [Accepted: 06/12/2024] [Indexed: 07/10/2024]
Abstract
OBJECTIVE Most paroxysmal kinesigenic dyskinesia (PKD) cases are hereditary, yet approximately 60% of patients remain genetically undiagnosed. We undertook the present study to uncover the genetic basis for undiagnosed PKD patients. METHODS Whole-exome sequencing was performed for 106 PRRT2-negative PKD probands. The functional impact of the genetic variants was investigated in HEK293T cells and Drosophila. RESULTS Heterozygous variants in KCNJ10 were identified in 11 individuals from 8 unrelated families, which accounted for 7.5% (8/106) of the PRRT2-negative probands. Both co-segregation of the identified variants and the significantly higher frequency of rare KCNJ10 variants in PKD cases supported impacts from the detected KCNJ10 heterozygous variants on PKD pathogenesis. Moreover, a KCNJ10 mutation-carrying father from a typical EAST/SeSAME family was identified as a PKD patient. All patients manifested dystonia attacks triggered by sudden movement with a short episodic duration. Patch-clamp recordings in HEK293T cells revealed apparent reductions in K+ currents of the patient-derived variants, indicating a loss-of-function. In Drosophila, milder hyperexcitability phenotypes were observed in heterozygous Irk2 knock-in flies compared to homozygotes, supporting haploinsufficiency as the mechanism for the detected heterozygous variants. Electrophysiological recordings showed that excitatory neurons in Irk2 haploinsufficiency flies exhibited increased excitability, and glia-specific complementation with human Kir4.1 rescued the Irk2 mutant phenotypes. INTERPRETATION Our study established haploinsufficiency resulting from heterozygous variants in KCNJ10 can be understood as a previously unrecognized genetic cause for PKD and provided evidence of glial involvement in the pathophysiology of PKD. ANN NEUROL 2024;96:758-773.
Collapse
Affiliation(s)
- Yun-Lu Li
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian, China
| | - Jingjing Lin
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian, China
| | - Xuejing Huang
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian, China
| | - Rui-Huang Zeng
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian, China
| | - Guangyu Zhang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jie-Ni Xu
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian, China
| | - Kai-Jun Lin
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian, China
| | - Xin-Shuo Chen
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian, China
| | - Ming-Feng He
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jing-Da Qiao
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xuewen Cheng
- Lingang Laboratory, Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| | - Dengna Zhu
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhi-Qi Xiong
- Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience and State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Wan-Jin Chen
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
39
|
Richards T, Wilson P, Goggolidou P. Next generation sequencing identifies WNT signalling as a significant pathway in Autosomal Recessive Polycystic Kidney Disease (ARPKD) manifestation and may be linked to disease severity. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167309. [PMID: 38885798 DOI: 10.1016/j.bbadis.2024.167309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 05/28/2024] [Accepted: 06/11/2024] [Indexed: 06/20/2024]
Abstract
INTRODUCTION Autosomal Recessive Polycystic Kidney Disease (ARPKD) is a rare paediatric disease primarily caused by sequence variants in PKHD1. ARPKD presents with considerable clinical variability relating to the type of PKHD1 sequence variant, but not its position. Animal models of Polycystic Kidney Disease (PKD) suggest a complex genetic landscape, with genetic modifiers as a potential cause of disease variability. METHODS To investigate in an unbiased manner the molecular mechanisms of ARPKD and identify potential indicators of disease severity, Whole Exome Sequencing (WES) and RNA-Sequencing (RNA-Seq) were employed on human ARPKD kidneys and age-matched healthy controls. RESULTS WES confirmed the clinical diagnosis of ARPKD in our patient cohort consisting of ten ARPKD kidneys. Sequence variant type, nor position of PKHD1 sequence variants, was linked to disease severity. Sequence variants in genes associated with other ciliopathies were detected in the ARPKD cohort, but only PKD1 could be linked to disease severity. Transcriptomic analysis on a subset of four ARPKD kidneys representing severe and moderate ARPKD, identified a significant number of genes relating to WNT signalling, cellular metabolism and development. Increased expression of WNT signalling-related genes was validated by RT-qPCR in severe and moderate ARPKD kidneys. Two individuals in our cohort with the same PKHD1 sequence variants but different rates of kidney disease progression, with displayed transcriptomic differences in the expression of WNT signalling genes. CONCLUSION ARPKD kidney transcriptomics highlights changes in WNT signalling as potentially significant in ARPKD manifestation and severity, providing indicators for slowing down the progression of ARPKD.
Collapse
Affiliation(s)
- Taylor Richards
- School of Biomedical Science and Physiology, Faculty of Science and Engineering, University of Wolverhampton, Wulfruna Street, Wolverhampton WV1 1LY, UK
| | - Patricia Wilson
- Centre for Nephrology, UCL Medical School, Royal Free Campus, Rowland Hill, London NW3 2PF, UK
| | - Paraskevi Goggolidou
- School of Biomedical Science and Physiology, Faculty of Science and Engineering, University of Wolverhampton, Wulfruna Street, Wolverhampton WV1 1LY, UK.
| |
Collapse
|
40
|
Yang K, Zhang T, Niu R, Zhao L, Cheng Z, Li J, Wang L. Unveiling the role of IGF1R in autism spectrum disorder: a multi-omics approach to decipher common pathogenic mechanisms in the IGF signaling pathway. Front Genet 2024; 15:1483574. [PMID: 39376742 PMCID: PMC11456441 DOI: 10.3389/fgene.2024.1483574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 09/09/2024] [Indexed: 10/09/2024] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental condition marked by impairments in social interaction, communication, and repetitive behaviors. Emerging evidence suggests that the insulin-like growth factor (IGF) signaling pathway plays a critical role in ASD pathogenesis; however, the precise pathogenic mechanisms remain elusive. This study utilizes multi-omics approaches to investigate the pathogenic mechanisms of ASD susceptibility genes within the IGF pathway. Whole-exome sequencing (WES) revealed a significant enrichment of rare variants in key IGF signaling components, particularly the IGF receptor 1 (IGF1R), in a cohort of Chinese Han individuals diagnosed with ASD, as well as in ASD patients from the SFARI SPARK WES database. Subsequent single-cell RNA sequencing (scRNA-seq) of cortical tissues from children with ASD demonstrated elevated expression of IGF receptors in parvalbumin (PV) interneurons, suggesting a substantial impact on their development. Notably, IGF1R appears to mediate the effects of IGF2R on these neurons. Additionally, transcriptomic analysis of brain organoids derived from ASD patients indicated a significant association between IGF1R and ASD. Protein-protein interaction (PPI) and gene regulatory network (GRN) analyses further identified ASD susceptibility genes that interact with and regulate IGF1R expression. In conclusion, IGF1R emerges as a central node within the IGF signaling pathway, representing a potential common pathogenic mechanism and therapeutic target for ASD. These findings highlight the need for further investigation into the modulation of this pathway as a strategy for ASD intervention.
Collapse
Affiliation(s)
- Kang Yang
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, China
| | - Tian Zhang
- Affiliated Mental Health Center & Hangzhou Seventh People’s Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruize Niu
- Affiliated Mental Health Center of Kuming Medical University, Yunnan Psychiatric Hospital, Kunming, China
| | - Liyang Zhao
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, China
| | - Zhonghe Cheng
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, China
| | - Jun Li
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, China
| | - Lifang Wang
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, China
| |
Collapse
|
41
|
Diossy M, Tisza V, Li H, Sahgal P, Zhou J, Sztupinszki Z, Young D, Nousome D, Kuo C, Jiang J, Chen Y, Ebner R, Sesterhenn IA, Moncur JT, Chesnut GT, Petrovics G, Klus GT, Valcz G, Nuzzo PV, Ribli D, Börcsök J, Prosz A, Krzystanek M, Ried T, Szuts D, Rizwan K, Kaochar S, Pathania S, D'Andrea AD, Csabai I, Srivastava S, Freedman ML, Dobi A, Spisak S, Szallasi Z. Frequent CHD1 deletions in prostate cancers of African American men is associated with rapid disease progression. NPJ Precis Oncol 2024; 8:208. [PMID: 39294262 PMCID: PMC11411125 DOI: 10.1038/s41698-024-00705-8] [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: 02/27/2024] [Accepted: 09/11/2024] [Indexed: 09/20/2024] Open
Abstract
We analyzed genomic data from the prostate cancer of African- and European American men to identify differences contributing to racial disparity of outcome. We also performed FISH-based studies of Chromodomain helicase DNA-binding protein 1 (CHD1) loss on prostate cancer tissue microarrays. We created CHD1-deficient prostate cancer cell lines for genomic, drug sensitivity and functional homologous recombination (HR) activity analysis. Subclonal deletion of CHD1 was nearly three times as frequent in prostate tumors of African American than in European American men and it associates with rapid disease progression. CHD1 deletion was not associated with HR deficiency associated mutational signatures or HR deficiency as detected by RAD51 foci formation. This was consistent with the moderate increase of olaparib and talazoparib sensitivity with several CHD1 deficient cell lines showing talazoparib sensitivity in the clinically relevant concentration range. CHD1 loss may contribute to worse disease outcome in African American men.
Collapse
Affiliation(s)
- Miklos Diossy
- Danish Cancer Institute, Copenhagen, Denmark
- Computational Health Informatics Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Viktoria Tisza
- Computational Health Informatics Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Hua Li
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, MD, USA
| | - Pranshu Sahgal
- Computational Health Informatics Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- The Eli and Edythe L. Broad Institute, Cambridge, MA, USA
| | - Jia Zhou
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Zsofia Sztupinszki
- Danish Cancer Institute, Copenhagen, Denmark
- Computational Health Informatics Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Denise Young
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, MD, USA
| | - Darryl Nousome
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, MD, USA
| | - Claire Kuo
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, MD, USA
| | - Jiji Jiang
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, MD, USA
| | - Yongmei Chen
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, MD, USA
| | | | | | | | - Gregory T Chesnut
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Gyorgy Petrovics
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, MD, USA
| | - Gregory T Klus
- Computational Health Informatics Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Gabor Valcz
- MTA-SE Molecular Medicine Research Group, Hungarian Academy of Sciences, Budapest, 1051, Hungary
| | - Pier Vitale Nuzzo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Dezso Ribli
- Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary
| | | | - Aurel Prosz
- Danish Cancer Institute, Copenhagen, Denmark
| | | | - Thomas Ried
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - David Szuts
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
| | - Kinza Rizwan
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Salma Kaochar
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Shailja Pathania
- Center for Personalized Cancer Therapy, University of Massachusetts, Boston, MA, USA
- Department of Biology, University of Massachusetts, Boston, MA, USA
| | - Alan D D'Andrea
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - Istvan Csabai
- Department of Physics of Complex Systems, Eötvös Loránd University, Budapest, Hungary
| | - Shiv Srivastava
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Department of Biochemistry and Molecular & Cell Biology, Georgetown University School of Medicine, Washington, DC, USA
| | - Matthew L Freedman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
- The Eli and Edythe L. Broad Institute, Cambridge, MA, USA.
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - Albert Dobi
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, MD, USA.
| | - Sandor Spisak
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary.
| | - Zoltan Szallasi
- Danish Cancer Institute, Copenhagen, Denmark.
- Computational Health Informatics Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
- 2nd Department of Pathology and Department of Bioinformatics, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
42
|
Rui X, Zhang X, Jia X, Han J, Wang C, Cao Q, Zhong O, Ding J, Zhao C, Zhang J, Ling X, Li H, Ma X, Meng Q, Huo R. Variants in NLRP2 and ZFP36L2, non-core components of the human subcortical maternal complex, cause female infertility with embryonic development arrest. Mol Hum Reprod 2024; 30:gaae031. [PMID: 39178021 DOI: 10.1093/molehr/gaae031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 07/30/2024] [Indexed: 08/24/2024] Open
Abstract
The subcortical maternal complex (SCMC), which is vital in oocyte maturation and embryogenesis, consists of core proteins (NLRP5, TLE6, OOEP), non-core proteins (PADI6, KHDC3L, NLRP2, NLRP7), and other unknown proteins that are encoded by maternal effect genes. Some variants of SCMC genes have been linked to female infertility characterized by embryonic development arrest. However, so far, the candidate non-core SCMC components associated with embryonic development need further exploration and the pathogenic variants that have been identified are still limited. In this study, we discovered two novel variants [p.(Ala131Val) and p.(Met326Val)] of NLRP2 in patients with primary infertility displaying embryonic development arrest from large families. In vitro studies using 293T cells and mouse oocytes, respectively, showed that these variants significantly decreased protein expression and caused the phenotype of embryonic development arrest. Additionally, we combined the 'DevOmics' database with the whole exome sequence data of our cohort and screened out a new candidate non-core SCMC gene ZFP36L2. Its variants [p.(Ala241Pro) and p.(Pro291dup)] were found to be responsible for embryonic development arrest. Co-immunoprecipitation experiments in 293T cells, used to demonstrate the interaction between proteins, verified that ZFP36L2 is one of the human SCMC components, and microinjection of ZFP36L2 complementary RNA variants into mouse oocytes affected embryonic development. Furthermore, the ZFP36L2 variants were associated with disrupted stability of its target mRNAs, which resulted in aberrant H3K4me3 and H3K9me3 levels. These disruptions decreased oocyte quality and further developmental potential. Overall, this is the first report of ZFP36L2 as a non-core component of the human SCMC and we found four novel pathogenic variants in the NLRP2 and ZFP36L2 genes in 4 of 161 patients that caused human embryonic development arrest. These findings contribute to the genetic diagnosis of female infertility and provide new insights into the physiological function of SCMC in female reproduction.
Collapse
Affiliation(s)
- Ximan Rui
- Department of Histology and Embryology, State Key Laboratory of Reproductive Medicine and Offspring Health, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Xiaolan Zhang
- Department of Reproductive Medicine, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Xinru Jia
- Department of Histology and Embryology, State Key Laboratory of Reproductive Medicine and Offspring Health, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Jian Han
- Department of Histology and Embryology, State Key Laboratory of Reproductive Medicine and Offspring Health, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Congjing Wang
- Department of Histology and Embryology, State Key Laboratory of Reproductive Medicine and Offspring Health, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Qiqi Cao
- Department of Histology and Embryology, State Key Laboratory of Reproductive Medicine and Offspring Health, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Ou Zhong
- Department of Histology and Embryology, State Key Laboratory of Reproductive Medicine and Offspring Health, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Jie Ding
- Department of Histology and Embryology, State Key Laboratory of Reproductive Medicine and Offspring Health, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
- Reproductive Genetic Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Chun Zhao
- Department of Reproductive Medicine, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Junqiang Zhang
- Department of Reproductive Medicine, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Xiufeng Ling
- Department of Reproductive Medicine, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Hong Li
- Department of Histology and Embryology, State Key Laboratory of Reproductive Medicine and Offspring Health, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
- Reproductive Genetic Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Xiang Ma
- State Key Laboratory of Reproductive Medicine and Offspring Health, Clinical Center of Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qingxia Meng
- Department of Histology and Embryology, State Key Laboratory of Reproductive Medicine and Offspring Health, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
- Reproductive Genetic Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Ran Huo
- Department of Histology and Embryology, State Key Laboratory of Reproductive Medicine and Offspring Health, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
- Innovation Center of Suzhou, Nanjing Medical University, Suzhou, China
| |
Collapse
|
43
|
Marin V, Lebreton L, Guibet C, Mesli S, Redonnet-Vernhet I, Dexant M, Lamireau D, Roche S, Gaschignard M, Delmas J, Margot H, Bar C. Case report: Unveiling genetic and phenotypic variability in Nonketotic hyperglycinemia: an atypical early onset case associated with a novel GLRX5 variant. Front Genet 2024; 15:1432272. [PMID: 39323869 PMCID: PMC11422140 DOI: 10.3389/fgene.2024.1432272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/23/2024] [Indexed: 09/27/2024] Open
Abstract
Nonketotic hyperglycinemia (NKH) is a rare, autosomal recessive metabolic disorder usually associated with mutations in genes AMT, GLDC or GCSH involved in the glycine cleavage complex. Other genes have been linked with less severe NKH, associated with deficiency of lipoate cofactor such as GLRX5, LIAS, BOLA3. We identified a new case of GLRX5-mediated NKH who presented at 2-month with severe developmental delay and seizures. The initial suspicion was raised by the MRI and then confirmed by glycine measurements in cerebrospinal fluid and blood. Genetic analysis revealed a previously undescribed homozygous variant in the GLRX5 gene [NM_016417.3:c.367G>C; p. (Asp123His)]. Despite medication and supportive care, he died at the age of 4 months after a sudden neurological deterioration. It was decided to limit therapeutic interventions due to the severity of the prognosis. The case was more severe than the previous GLRX5-mediated NKH described, regarding the early age at onset and the severity. Moreover, the genetic variant was located at a potentially crucial site for glutathione binding in the GLRX5 protein. This report, thereby, expands our understanding of NKH's genetic underpinnings and phenotypic variability, highlighting the crucial role of GLRX5 and other related genes in variant NKH.
Collapse
Affiliation(s)
- Victor Marin
- Service de Biochimie, Groupe Hospitalier Pellegrin, CHU de Bordeaux, Bordeaux, France
| | - Louis Lebreton
- Service de Biochimie, Groupe Hospitalier Pellegrin, CHU de Bordeaux, Bordeaux, France
| | - Claire Guibet
- Service de Biochimie, Groupe Hospitalier Pellegrin, CHU de Bordeaux, Bordeaux, France
- INSERM BRIC U1312 Université de Bordeaux, Bordeaux, France
| | - Samir Mesli
- Service de Biochimie, Groupe Hospitalier Pellegrin, CHU de Bordeaux, Bordeaux, France
| | | | - Mathurin Dexant
- Service de Biochimie, Groupe Hospitalier Pellegrin, CHU de Bordeaux, Bordeaux, France
| | - Delphine Lamireau
- Hôpital Pédiatrique, Pôle Pédiatrique, CHU de Bordeaux, Bordeaux, France
| | - Sandrine Roche
- Hôpital Pédiatrique, Pôle Pédiatrique, CHU de Bordeaux, Bordeaux, France
| | | | - Jean Delmas
- Service d’Imagerie Anténatale, de l’Enfant et de la Femme, Groupe Hospitalier Pellegrin, CHU de Bordeaux, Bordeaux, France
| | - Henri Margot
- Department of Medical Genetics, University of Bordeaux, MRGM INSERM U1211, CHU de Bordeaux, Bordeaux, France
| | - Claire Bar
- Service de Neurologie Pédiatrique, CHU Bordeaux, University Bordeaux, CNRS, INCIA, UMR 5287, NRGen Team, Bordeaux, France
| |
Collapse
|
44
|
Claus LR, Ernst RF, Elferink MG, van Deutekom HW, van der Zwaag B, van Eerde AM. The Importance of Copy Number Variant Analysis in Patients with Monogenic Kidney Disease. Kidney Int Rep 2024; 9:2695-2704. [PMID: 39291214 PMCID: PMC11403095 DOI: 10.1016/j.ekir.2024.06.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 06/17/2024] [Indexed: 09/19/2024] Open
Abstract
Introduction Genetic testing can reveal monogenic causes of kidney diseases, offering diagnostic, therapeutic, and prognostic benefits. Although single nucleotide variants (SNVs) and copy number variants (CNVs) can result in kidney disease, CNV analysis is not always included in genetic testing. Methods We investigated the diagnostic value of CNV analysis in 2432 patients with kidney disease genetically tested at the University Medical Centre Utrecht between 2014 and May 2022. We combined previous diagnostic testing results, encompassing SNVs and CNVs, with newly acquired results based on retrospective CNV analysis. The reported yield considers both the American College of Medical Genetics and Genomics (ACMG) classification and whether the genotype actually results in disease. Results We report a diagnostic yield of at least 23% for our complete diagnostic cohort. The total diagnostic yield based solely on CNVs was 2.4%. The overall contribution of CNV analysis, defined as the proportion of positive genetic tests requiring CNV analysis, was 10.5% and varied among different disease subcategories, with the highest impact seen in congenital anomalies of the kidney and urinary tract (CAKUT) and chronic kidney disease at a young age. We highlight the efficiency of exome-based CNV calling, which reduces the need for additional diagnostic tests. Furthermore, a complex structural variant, likely a COL4A4 founder variant, was identified. Additional findings unrelated to kidney diseases were reported in a small percentage of cases. Conclusion In summary, this study demonstrates the substantial diagnostic value of CNV analysis, providing insights into its contribution to the diagnostic yield and advocating for its routine inclusion in genetic testing of patients with kidney disease.
Collapse
Affiliation(s)
- Laura R. Claus
- Department of Genetics, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Robert F. Ernst
- Department of Genetics, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Martin G. Elferink
- Department of Genetics, University Medical Centre Utrecht, Utrecht, the Netherlands
| | | | - Bert van der Zwaag
- Department of Genetics, University Medical Centre Utrecht, Utrecht, the Netherlands
| | | |
Collapse
|
45
|
Tsao WC, Yu RL, Li CT, Tsai WF, Chuang WL, Huang JF, Dai CY, Tan CH. Viral hepatitis moderates the impact of TGFB1 on neurocognitive impairment. Kaohsiung J Med Sci 2024; 40:852-861. [PMID: 38970443 DOI: 10.1002/kjm2.12872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 07/08/2024] Open
Abstract
Recent studies have identified a correlation between chronic viral hepatitis and cognitive impairment, yet the underlying mechanisms remain unclear. This study investigated the influence of TGFB1 genetic polymorphisms on cognitive function in individuals with and without hepatitis infections, hypothesizing that these polymorphisms and the viral hepatitis-induced inflammatory environment interact to affect cognitive abilities. Participants (173 with viral hepatitis and 258 healthy controls) were recruited. Genotyping of TGFB1 SNPs was performed using the C2-58 Axiom Genome-Wide TWB 2.0 Array Plate. Cognitive function was assessed using the MMSE and MoCA tests. Our results showed that healthy individuals carrying the C allele of rs2241715 displayed better performance in sentence writing (p = 0.020) and language tasks (p = 0.022). Notably, viral hepatitis was found to moderate the impact of the rs2241715 genotype on language function (p = 0.002). Similarly, those carrying the T allele of rs10417924 demonstrated superior orientation to time (p = 0.002), with viral hepatitis modifying the influence of the SNP on this particular cognitive function (p = 0.010). Our findings underscore the significant role of TGFβ1 in cognitive function and the moderating impact of viral hepatitis on TGFB1 SNP effects. These findings illuminate the potential of TGFB1 as a therapeutic target for cognitive impairment induced by viral hepatitis, thus broadening our understanding of TGFβ1 functionality in the pathogenesis of neurodegeneration.
Collapse
Affiliation(s)
- Wei-Chia Tsao
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Rwei-Ling Yu
- Institute of Behavioral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chi-Ting Li
- Department of Psychology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wei-Fang Tsai
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wan-Long Chuang
- Hepatobiliary Division, Department of Internal Medicine, and Hepatitis Center Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jee-Fu Huang
- Hepatobiliary Division, Department of Internal Medicine, and Hepatitis Center Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Yen Dai
- Hepatobiliary Division, Department of Internal Medicine, and Hepatitis Center Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chun-Hsiang Tan
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
46
|
Sun X, Jia Q, Li K, Tian C, Yi L, Yan L, Zheng J, Jia X, Gu M. Comparative genomic landscape of lower-grade glioma and glioblastoma. PLoS One 2024; 19:e0309536. [PMID: 39208202 PMCID: PMC11361568 DOI: 10.1371/journal.pone.0309536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
Biomarkers for classifying and grading gliomas have been extensively explored, whereas populations in public databases were mostly Western/European. Based on public databases cannot accurately represent Chinese population. To identify molecular characteristics associated with clinical outcomes of lower-grade glioma (LGG) and glioblastoma (GBM) in the Chinese population, we performed whole-exome sequencing (WES) in 16 LGG and 35 GBM tumor tissues. TP53 (36/51), TERT (31/51), ATRX (16/51), EFGLAM (14/51), and IDH1 (13/51) were the most common genes harboring mutations. IDH1 mutation (c.G395A; p.R132H) was significantly enriched in LGG, whereas PCDHGA10 mutation (c.A265G; p.I89V) in GBM. IDH1-wildtype and PCDHGA10 mutation were significantly related to poor prognosis. IDH1 is an important biomarker in gliomas, whereas PCDHGA10 mutation has not been reported to correlate with gliomas. Different copy number variations (CNVs) and oncogenic signaling pathways were identified between LGG and GBM. Differential genomic landscapes between LGG and GBM were revealed in the Chinese population, and PCDHGA10, for the first time, was identified as the prognostic factor of gliomas. Our results might provide a basis for molecular classification and identification of diagnostic biomarkers and even potential therapeutic targets for gliomas.
Collapse
Affiliation(s)
- Xinxin Sun
- Joint Laboratory for Translational Medicine Research, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| | - Qingbin Jia
- Department of Neurosurgery, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| | - Kun Li
- Joint Laboratory for Translational Medicine Research, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| | - Conghui Tian
- Joint Laboratory for Translational Medicine Research, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| | - Lili Yi
- Joint Laboratory for Translational Medicine Research, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| | - Lili Yan
- Joint Laboratory for Translational Medicine Research, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| | - Juan Zheng
- Joint Laboratory for Translational Medicine Research, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| | - Xiaodong Jia
- Joint Laboratory for Translational Medicine Research, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| | - Mingliang Gu
- Joint Laboratory for Translational Medicine Research, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| |
Collapse
|
47
|
Endrakanti M, Sharma J, Ethayathulla AS, Kaur P, Khan SA, Kabra M, Gupta N. ECEL1 related distal arthrogryposis 5D in an Indian cohort-Report of recognizable musculoskeletal phenotype and a possible founder variant. Am J Med Genet A 2024; 194:e63592. [PMID: 38568023 DOI: 10.1002/ajmg.a.63592] [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: 07/30/2023] [Revised: 01/24/2024] [Accepted: 03/02/2024] [Indexed: 07/05/2024]
Abstract
Distal arthrogryposis type 5D (DA5D) is clinically characterized by knee extension contractures, distal joint contractures, clubfoot, micrognathia, ptosis, and scoliosis. We report nine affected individuals from eight unrelated Indian families with DA5D. Although the overall musculoskeletal phenotype is not very distinct from other distal arthrogryposis, the presence of fixed knee extension contractures with or without scoliosis could be an important early pointer to DA5D. We also report a possible founder variant in ECEL1 along with four novel variants and further expand the genotypic spectrum of DA5D.
Collapse
Affiliation(s)
- Mounika Endrakanti
- Division of Genetics, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Jyoti Sharma
- Division of Genetics, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Abdul S Ethayathulla
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Punit Kaur
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Shah Alam Khan
- Department of Orthopedics, All India Institute of Medical Sciences, New Delhi, India
| | - Madhulika Kabra
- Division of Genetics, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Neerja Gupta
- Division of Genetics, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
48
|
Subramanian K, Chopra M, Kahali B. Landscape of genomic structural variations in Indian population-based cohorts: Deeper insights into their prevalence and clinical relevance. HGG ADVANCES 2024; 5:100285. [PMID: 38521976 PMCID: PMC11007539 DOI: 10.1016/j.xhgg.2024.100285] [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: 09/14/2023] [Revised: 03/13/2024] [Accepted: 03/20/2024] [Indexed: 03/25/2024] Open
Abstract
Structural variations (SV) are large (>50 base pairs) genomic rearrangements comprising deletions, duplications, insertions, inversions, and translocations. Studying SVs is important because they play active and critical roles in regulating gene expression, determining disease predispositions, and identifying population-specific differences among individuals of diverse ancestries. However, SV discoveries in the Indian population using whole-genome sequencing (WGS) have been limited. In this study, using short-read WGS having an average 42X depth of coverage, we identify and characterize 36,210 SVs from 529 individuals enrolled in population-based cohorts in India. These SVs include 24,574 deletions, 2,913 duplications, 8,710 insertions, and 13 inversions; 1.26% (456 out of 36,210) of the identified SVs can potentially impact the coding regions of genes. Furthermore, 56 of these SVs are highly intolerant to loss-of-function changes to the mapped genes, and five SVs impacting ADAMTS17, CCDC40, and RHCE are common in our study individuals. Seven rare SVs significantly impact dosage sensitivity of genes known to be associated with various clinical phenotypes. Most of the SVs in our study are rare and heterozygous. This fine-scale SV discovery in the underrepresented Indian population provides valuable insights that extend beyond Eurocentric human genetic studies.
Collapse
Affiliation(s)
- Krithika Subramanian
- Centre for Brain Research, Indian Institute of Science, Bangalore 560012, India; Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Mehak Chopra
- Centre for Brain Research, Indian Institute of Science, Bangalore 560012, India
| | - Bratati Kahali
- Centre for Brain Research, Indian Institute of Science, Bangalore 560012, India.
| |
Collapse
|
49
|
Stephenson JD, Totoo P, Burke D, Jänes J, Beltrao P, Martin M. ProtVar: mapping and contextualizing human missense variation. Nucleic Acids Res 2024; 52:W140-W147. [PMID: 38769064 PMCID: PMC11223857 DOI: 10.1093/nar/gkae413] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/26/2024] [Accepted: 05/03/2024] [Indexed: 05/22/2024] Open
Abstract
Genomic variation can impact normal biological function in complex ways and so understanding variant effects requires a broad range of data to be coherently assimilated. Whilst the volume of human variant data and relevant annotations has increased, the corresponding increase in the breadth of participating fields, standards and versioning mean that moving between genomic, coding, protein and structure positions is increasingly complex. In turn this makes investigating variants in diverse formats and assimilating annotations from different resources challenging. ProtVar addresses these issues to facilitate the contextualization and interpretation of human missense variation with unparalleled flexibility and ease of accessibility for use by the broadest range of researchers. By precalculating all possible variants in the human proteome it offers near instantaneous mapping between all relevant data types. It also combines data and analyses from a plethora of resources to bring together genomic, protein sequence and function annotations as well as structural insights and predictions to better understand the likely effect of missense variation in humans. It is offered as an intuitive web server https://www.ebi.ac.uk/protvar where data can be explored and downloaded, and can be accessed programmatically via an API.
Collapse
Affiliation(s)
| | - Prabhat Totoo
- EMBL-EBI, Wellcome Genome Campus, Hinxton CB10 1SD, Cambridgeshire, UK
| | | | - Jürgen Jänes
- Department of Biology, Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
| | - Pedro Beltrao
- Department of Biology, Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Maria J Martin
- EMBL-EBI, Wellcome Genome Campus, Hinxton CB10 1SD, Cambridgeshire, UK
| |
Collapse
|
50
|
Wang J, Wang M, Moshiri A, Harris RA, Raveendran M, Nguyen T, Kim S, Young L, Wang K, Wiseman R, O'Connor DH, Johnson Z, Martinez M, Montague MJ, Sayers K, Lyke M, Vallender E, Stout T, Li Y, Thomasy SM, Rogers J, Chen R. Genetic diversity of 1,845 rhesus macaques improves genetic variation interpretation and identifies disease models. Nat Commun 2024; 15:5658. [PMID: 38969634 PMCID: PMC11226599 DOI: 10.1038/s41467-024-49922-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 06/25/2024] [Indexed: 07/07/2024] Open
Abstract
Understanding and treating human diseases require valid animal models. Leveraging the genetic diversity in rhesus macaque populations across eight primate centers in the United States, we conduct targeted-sequencing on 1845 individuals for 374 genes linked to inherited human retinal and neurodevelopmental diseases. We identify over 47,000 single nucleotide variants, a substantial proportion of which are shared with human populations. By combining rhesus and human allele frequencies with established variant prediction methods, we develop a machine learning-based score that outperforms established methods in predicting missense variant pathogenicity. Remarkably, we find a marked number of loss-of-function variants and putative deleterious variants, which may lead to the development of rhesus disease models. Through phenotyping of macaques carrying a pathogenic OPA1:p.A8S variant, we identify a genetic model of autosomal dominant optic atrophy. Finally, we present a public website housing variant and genotype data from over two thousand rhesus macaques.
Collapse
Affiliation(s)
- Jun Wang
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Meng Wang
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Ala Moshiri
- Department of Ophthalmology & Vision Science, School of Medicine, UC Davis, Sacramento, California, USA
| | - R Alan Harris
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Muthuswamy Raveendran
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Tracy Nguyen
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California-Davis, Davis, California, USA
| | - Soohyun Kim
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California-Davis, Davis, California, USA
| | - Laura Young
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California-Davis, Davis, California, USA
| | - Keqing Wang
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Roger Wiseman
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - David H O'Connor
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Zach Johnson
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Melween Martinez
- Caribbean Primate Research Center, University of Puerto Rico, Punta Santiago, Humacao, Puerto Rico
| | - Michael J Montague
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ken Sayers
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Martha Lyke
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Eric Vallender
- Tulane National Primate Research Center, Tulane university, Covington, Louisiana, USA
| | - Tim Stout
- Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, Texas, USA
| | - Yumei Li
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Sara M Thomasy
- Department of Ophthalmology & Vision Science, School of Medicine, UC Davis, Sacramento, California, USA
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California-Davis, Davis, California, USA
- California National Primate Research Center, University of California-Davis, Davis, California, USA
| | - Jeffrey Rogers
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Rui Chen
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA.
| |
Collapse
|