101
|
Claus I, Sivalingam S, Koller AC, Weiß A, Mathey CM, Sindermann L, Klein D, Henschel L, Ludwig KU, Hoffmann P, Heimbach A, Heilmann-Heimbach S, Vedder H, Kammerer-Ciernioch J, Stürmer T, Streit F, Maaser-Hecker A, Nenadić I, Baune BT, Hartmann AM, Konte B, Giegling I, Heilbronner U, Wagner M, Philipsen A, Schmidt B, Rujescu D, Buness A, Schulze TG, Rietschel M, Forstner AJ, Nöthen MM, Degenhardt F. Contribution of Rare and Potentially Functionally Relevant Sequence Variants in Schizophrenia Risk-Locus Xq28,distal. Am J Med Genet B Neuropsychiatr Genet 2025; 198:e33011. [PMID: 39473393 DOI: 10.1002/ajmg.b.33011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 09/01/2024] [Accepted: 09/17/2024] [Indexed: 03/04/2025]
Abstract
Duplications of the Xq28,distal locus have been described in male and female patients with schizophrenia (SCZ) or intellectual disability. The Xq28,distal locus spans eight protein-coding genes (F8, CMC4, MTCP1, BRCC3, VBP1, FUNDC2, CLIC2, and RAB39B) and is flanked by recurrent genomic breakpoints. Thus, the issue of which gene/s at this locus is/are relevant in terms of SCZ pathogenesis remains unclear. The aim of this study was to investigate the contribution of rare and potentially functionally relevant sequence variants within the Xq28,distal locus to SCZ risk using the single-molecule molecular inversion probes (smMIP) method. Targeted sequencing was performed in a cohort of 1935 patients with SCZ and 1905 controls of European ancestry. The consecutive statistical analysis addressed two main areas. On the level of the individual variants, allele counts in the patient and control cohort were systematically compared with a Fisher's exact test: (i) for the entire present study cohort; (ii) for patients and controls separated by sex; and (iii) in combination with data published by the Schizophrenia Exome Meta-Analysis (SCHEMA) consortium. On the gene-wise level, a burden analysis was performed using the X-chromosomal model of the Optimal Unified Sequence Kernel Association Test (SKAT-O), with adjustment for possible sex-specific effects. Targeted sequencing identified a total of 13 rare and potentially functional variants in four patients and 11 controls. However, neither at the level of individual rare and potentially functional variants nor at the level of the eight protein-coding genes at the Xq28,distal locus was a statistically significant enrichment in patients compared to controls observed. Although inconclusive, the present findings represent a step toward improved understanding of the contribution of X-chromosomal risk factors in neuropsychiatric disorder development, which is an underrepresented aspect of genetic studies in this field.
Collapse
Grants
- BONFOR Research Funding Program of the Faculty of Medicine, University of Bonn
- Dr. Lisa Oehler Foundation
- 01ZX1614K Federal Ministry of Education and Research; projects: IntegraMent and BipoLife
- 01EE1404H Federal Ministry of Education and Research; projects: IntegraMent and BipoLife
- 945151 European Union's Horizon 2020 Research and Innovation Programme; projects: PSY-PGx, GEPI-BIOPSY and MulioBio
- 01EW2005 European Union's Horizon 2020 Research and Innovation Programme; projects: PSY-PGx, GEPI-BIOPSY and MulioBio
- 01EW2009 European Union's Horizon 2020 Research and Innovation Programme; projects: PSY-PGx, GEPI-BIOPSY and MulioBio
- 514201724 German Research Foundation; Projects: PsyCourse, KFO 241 and Heidelberg Cohort Study of the Elderly
- STU 235/10-2 German Research Foundation; Projects: PsyCourse, KFO 241 and Heidelberg Cohort Study of the Elderly
- HE 2443/8-1 German Research Foundation; Projects: PsyCourse, KFO 241 and Heidelberg Cohort Study of the Elderly
- AM37/19-1 German Research Foundation; Projects: PsyCourse, KFO 241 and Heidelberg Cohort Study of the Elderly
- SCHU1603/4-1,5-1,7-1 German Research Foundation; Projects: PsyCourse, KFO 241 and Heidelberg Cohort Study of the Elderly
Collapse
Affiliation(s)
- I Claus
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - S Sivalingam
- Institute for Medical Biometry, Informatics and Epidemiology, Medical Faculty, University of Bonn, Bonn, Germany
- Institute for Genomic Statistics and Bioinformatics, Medical Faculty, University of Bonn, Bonn, Germany
- Core Unit for Bioinformatics Data Analysis, Medical Faculty, University of Bonn, Bonn, Germany
- Institute of Human Genetics, Medical Faculty, University Hospital of Düsseldorf, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - A C Koller
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - A Weiß
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - C M Mathey
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - L Sindermann
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - D Klein
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - L Henschel
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - K U Ludwig
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - P Hoffmann
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
- Division of Medical Genetics, University Hospital and Department of Biomedicine, University of Basel, Basel, Switzerland
| | - A Heimbach
- NGS Core Facility, Medical Faculty, University of Bonn, Bonn, Germany
| | - S Heilmann-Heimbach
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - H Vedder
- Psychiatric Center Nordbaden, Wiesloch, Germany
| | | | - T Stürmer
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - F Streit
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - A Maaser-Hecker
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
- Genetics and Aging Research Unit, Department of Neurology, Mass General Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - I Nenadić
- Department of Psychiatry und Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg, Marburg, Germany
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany
| | - B T Baune
- Department of Psychiatry, University of Münster, Münster, Germany
- Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Central Melbourne, Australia
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Central Melbourne, Australia
| | - A M Hartmann
- Department of Psychiatry and Psychotherapy, Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - B Konte
- Department of Psychiatry and Psychotherapy, Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - I Giegling
- Department of Psychiatry and Psychotherapy, Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - U Heilbronner
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, Germany
| | - M Wagner
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - A Philipsen
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - B Schmidt
- Institute for Medical Informatics, Biometry and Epidemiology (IMIBE), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - D Rujescu
- Department of Psychiatry and Psychotherapy, Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - A Buness
- Institute for Medical Biometry, Informatics and Epidemiology, Medical Faculty, University of Bonn, Bonn, Germany
- Institute for Genomic Statistics and Bioinformatics, Medical Faculty, University of Bonn, Bonn, Germany
- Core Unit for Bioinformatics Data Analysis, Medical Faculty, University of Bonn, Bonn, Germany
| | - T G Schulze
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, Germany
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, New York, USA
| | - M Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - A J Forstner
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
- Institute of Neuroscience and Medicine (INM-1), Research Center Jülich, Jülich, Germany
| | - M M Nöthen
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - F Degenhardt
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
102
|
Wang WA, Garofoli A, Ferrada E, Klimek C, Steurer B, Ingles-Prieto A, Osthushenrich T, MacNamara A, Malarstig A, Wiedmer T, Superti-Furga G. Human genetic variants in SLC39A8 impact uptake and steady-state metal levels within the cell. Life Sci Alliance 2025; 8:e202403028. [PMID: 39884836 PMCID: PMC11782468 DOI: 10.26508/lsa.202403028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 01/15/2025] [Accepted: 01/15/2025] [Indexed: 02/01/2025] Open
Abstract
The human SLC39A8 (hSLC39A8) gene encodes a plasma membrane protein SLC39A8 (ZIP8) that mediates the specific uptake of the metals Cd2+, Mn2+, Zn2+, Fe2+, Co2+, and Se4+ Pathogenic variants within hSLC39A8 are associated with congenital disorder of glycosylation type 2 (CDG type II) or Leigh-like syndrome. However, numerous mutations of uncertain significance are also linked to different conditions or benign traits. Our study characterized 21 hSLC39A8 variants and measured their impact on protein localization and intracellular levels of Cd2+, Zn2+, and Mn2+ We identified four variants that disrupt protein expression, five variants with high retention in the endoplasmic reticulum, and 12 variants with localization to the plasma membrane. From the 12 variants with plasma membrane localization, we identified three with complete loss of detectable ion uptake by the cell and five with differential uptake between metal ions. Further in silico analysis on protein stability identified variants that may affect the stability of homodimer interfaces. This study elucidates the variety of effects of hSLC39A8 variants on ZIP8 and on diseases involving disrupted metal ion homeostasis.
Collapse
Affiliation(s)
- Wen-An Wang
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Andrea Garofoli
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Evandro Ferrada
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Instituto de Sistemas Complejos de Valparaíso (ISCV), Valparaíso, Chile
| | - Christoph Klimek
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Barbara Steurer
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Alvaro Ingles-Prieto
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | | | | | - Anders Malarstig
- Pfizer Worldwide Research, Development and Medical, Stockholm, Sweden
| | - Tabea Wiedmer
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Giulio Superti-Furga
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
103
|
Tischer A, Moon-Tasson L, Auton M. Structure-resolved dynamics of type 2M von Willebrand disease. J Thromb Haemost 2025; 23:1215-1228. [PMID: 39756657 PMCID: PMC11972889 DOI: 10.1016/j.jtha.2024.12.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/22/2024] [Accepted: 12/20/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND Genetically determined amino acid substitutions in the platelet adhesive A1 domain alter von Willebrand factor's (VWF) platelet agglutination competence, resulting in both gain- (type 2B) and loss-of-function (type 2M) phenotypes of von Willebrand disease. Prior studies of variants in both phenotypes revealed defects in secondary structure that altered stability and folding of the domain. An intriguing observation was that loss of function arose from both misfolding of A1 and, in a few cases, hyperstabilization of the native structure. OBJECTIVES To fully understand the 2M phenotype, we thoroughly investigated the structure/function relationships of 15 additional type 2M variants and 2 polymorphisms in the A1 domain. METHODS These variants were characterized using circular dichroism, fluorescence, calorimetry, hydrogen-deuterium exchange mass spectrometry, surface plasmon resonance, and platelet adhesion under shear flow. RESULTS Six variants were natively folded, with 4 being hyperstabilized. Nine variants disordered A1, causing a loss in α-helical structure and unfolding enthalpy. GPIbα binding affinity and platelet adhesion dynamics were highly correlated to helical structure. Hydrogen-deuterium exchange resolved specific C-terminal secondary structure elements that differentially diminish the GPIbα binding affinity of A1. These localized structural perturbations were highly correlated to GPIbα binding affinity and shear-dependent platelet adhesion. CONCLUSION While hyperstabilized dynamics in A1 do impair stable platelet attachment to VWF under flow, variant-induced localized disorder in specific regions of the domain misfolds A1 and abrogates platelet adhesion. These 2 opposing conformational properties represent 2 structural classes of VWF that drive the loss-of-function phenotype that is type 2M von Willebrand disease.
Collapse
Affiliation(s)
- Alexander Tischer
- Division of Hematology, Departments of Internal Medicine and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Laurie Moon-Tasson
- Division of Hematology, Departments of Internal Medicine and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Matthew Auton
- Division of Hematology, Departments of Internal Medicine and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA.
| |
Collapse
|
104
|
Yuan Q, Hodgkinson C, Liu X, Barton B, Diazgranados N, Schwandt M, Morgan T, Bataller R, Liangpunsakul S, Nagy LE, Goldman D. Exome-wide association analysis identifies novel risk loci for alcohol-associated hepatitis. Hepatology 2025; 81:1304-1317. [PMID: 39058584 PMCID: PMC11902603 DOI: 10.1097/hep.0000000000001027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND AND AIMS Alcohol-associated hepatitis (AH) is a clinically severe, acute disease that afflicts only a fraction of patients with alcohol use disorder. Genomic studies of alcohol-associated cirrhosis (AC) have identified several genes of large effect, but the genetic and environmental factors that lead to AH and AC, and their degree of genetic overlap, remain largely unknown. This study aims to identify genes and genetic variations that contribute to the development of AH. APPROACH AND RESULTS Exome-sequencing of patients with AH (N=784) and heavy drinking controls (N=951) identified an exome-wide significant association for AH at patalin-like phospholipase domain containing 3, as previously observed for AC in genome-wide association study, although with a much lower effect size. Single nucleotide polymorphisms (SNPs) of large effect size at inducible T cell costimulatory ligand ( ICOSLG ) (Chr 21) and TOX4/RAB2B (Chr 14) were also exome-wide significant. ICOSLG encodes a co-stimulatory signal for T-cell proliferation and cytokine secretion and induces B-cell proliferation and differentiation. TOX high mobility group box family member 4 ( TOX4 ) was previously implicated in diabetes and immune system function. Other genes previously implicated in AC did not strongly contribute to AH, and the only prominently implicated (but not exome-wide significant) gene overlapping with alcohol use disorder was alcohol dehydrogenase 1B ( ADH1B ). Polygenic signals for AH were observed in both common and rare variant analysis and identified genes with roles associated with inflammation. CONCLUSIONS This study has identified 2 new genes of high effect size with a previously unknown contribution to alcohol-associated liver disease and highlights both the overlap in etiology between liver diseases and the unique origins of AH.
Collapse
Affiliation(s)
- Qiaoping Yuan
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - Colin Hodgkinson
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - Xiaochen Liu
- Department of Epidemiology and Biostatistics, University of California, Irvine, Irvine, California, USA
| | - Bruce Barton
- Department of Population & Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Nancy Diazgranados
- Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - Melanie Schwandt
- Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | | | - Timothy Morgan
- Department of Gastroenterology, Long Beach Veterans Healthcare System (VALVE), Long Beach, California, USA
- Department of Medicine, University of California, Irvine, CA, USA
| | - Ramon Bataller
- Liver Unit, Hospital Clínic de Barcelona, Barcelona, Spain
- Facultad de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Suthat Liangpunsakul
- Division of Gastroenterology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Roudebush Veterans Administration Medical Center, Indianapolis, Indiana, USA
| | - Laura E. Nagy
- Department of Inflammation & Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA
- Department of Molecular Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - David Goldman
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
- Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| |
Collapse
|
105
|
Soldà G, Asselta R. Applying artificial intelligence to uncover the genetic landscape of coagulation factors. J Thromb Haemost 2025; 23:1133-1145. [PMID: 39798926 DOI: 10.1016/j.jtha.2024.12.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/20/2024] [Accepted: 12/26/2024] [Indexed: 01/15/2025]
Abstract
Artificial intelligence (AI) is rapidly advancing our ability to identify and interpret genetic variants associated with coagulation factor deficiencies. This review introduces AI, with a specific focus on machine learning (ML) methods, and examines its applications in the field of coagulation genetics over the past decade. We observed a significant increase in AI-related publications, with a focus on hemophilia A and B. ML approaches have shown promise in predicting the functional impact of genetic variants and establishing genotype-phenotype correlations, exemplified by tools like "Hema-Class" for factor VIII variants. However, some challenges remain, including the need to expand variant selection beyond missense mutations (which is now the standard of most studies). For the future, the integration of AI in calling, detecting, and interpreting genetic variants can significantly improve our ability to process large-scale genomic data. In this frame, we discuss various AI/ML-based tools for genetic variant detection and interpretation, highlighting their strengths and limitations. As the field evolves, the synergistic application of multiple AI models, coupled with rigorous validation strategies, will be crucial in advancing our understanding of coagulation disorders and for personalizing treatment approaches.
Collapse
Affiliation(s)
- Giulia Soldà
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, Medical Genetics and RNA Biology Unit, Rozzano, Milan, Italy
| | - Rosanna Asselta
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, Medical Genetics and RNA Biology Unit, Rozzano, Milan, Italy.
| |
Collapse
|
106
|
Andriano A, Desantis V, Marasco C, Marzollo A, Bresolin S, Resta N, Di Marzo L, Pappagallo F, Mascolo A, Caradonna IC, D'Amore S, Vacca A, Solimando AG. Genomic profiling at a single center cracks the code in inborn errors of immunity. Intern Emerg Med 2025; 20:761-770. [PMID: 39873967 PMCID: PMC12009233 DOI: 10.1007/s11739-025-03871-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/08/2025] [Indexed: 01/30/2025]
Abstract
Inborn errors of immunity (IEI) entail a diverse group of disorders resulting from hereditary or de novo mutations in single genes, leading to immune dysregulation. This study explores the clinical utility of next-generation sequencing (NGS) techniques in diagnosing monogenic immune defects. Eight patients attending the immunodeficiency clinic and with unclassified antibody deficiency were included in the analysis. Clinical records, immune characteristics, and family histories were reviewed, and a target gene panel (TGP) sequencing was performed to identify pathogenic variants. TGPs identified seven variants in TNFRSF13B (TACI), CARMIL2, STAT1, STAT3, and ORAI1 genes. These findings provided definitive diagnoses and proper prognostic assessment. Patients exhibited a wide range of clinical manifestations, including recurrent infections, autoimmune cytopenias, and organ-specific complications. The genetic diversity observed highlights the importance of genetic testing in diagnosing IEIs and tailoring treatments. This study underscores the role of TGPs in diagnosing IEIs, revealing significant genetic heterogeneity and phenotypic variability. They offer a precise tool for identifying underlying genetic defects, facilitating personalized medicine approaches, and eventually improving patient outcomes. The findings emphasize the need for comprehensive genetic testing to uncover novel pathogenic variants, enhancing our understanding of immune system dysfunction. NGS is a critical tool for the management of IEI, enabling precise diagnosis and personalized treatment strategies. Despite resource limitations, the progressive affordability is likely to expand its clinical utility, ultimately improving patient care and advancing the field of immunology. In the meantime, accurate phenotypic assessment is essential for resource optimization and case prioritization.
Collapse
Affiliation(s)
- Alessandro Andriano
- Pharmacology Section, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro Medical School, Bari, Italy
| | - Vanessa Desantis
- Pharmacology Section, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro Medical School, Bari, Italy
| | - Carolina Marasco
- Unit of Internal Medicine and Clinical Oncology "G. Baccelli", Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro Medical School, Bari, Italy
| | - Antonio Marzollo
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Padua University Hospital, Padua, Italy
| | - Silvia Bresolin
- Onco-Hematology, Stem Cell Transplant and Gene Therapy, Istituto Di Ricerca Pediatrica Foundation - Città Della Speranza, Padua, Italy
| | - Nicoletta Resta
- Medical Genetic, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro, Bari, Italy
| | - Lucia Di Marzo
- Pharmacology Section, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro Medical School, Bari, Italy
| | - Fabrizio Pappagallo
- Unit of Internal Medicine and Clinical Oncology "G. Baccelli", Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro Medical School, Bari, Italy
| | - Antonella Mascolo
- Unit of Internal Medicine and Clinical Oncology "G. Baccelli", Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro Medical School, Bari, Italy
| | - Ingrid Catalina Caradonna
- Pharmacology Section, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro Medical School, Bari, Italy
| | - Simona D'Amore
- Unit of Internal Medicine and Clinical Oncology "G. Baccelli", Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro Medical School, Bari, Italy
| | - Angelo Vacca
- Unit of Internal Medicine and Clinical Oncology "G. Baccelli", Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro Medical School, Bari, Italy
| | - Antonio Giovanni Solimando
- Unit of Internal Medicine and Clinical Oncology "G. Baccelli", Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro Medical School, Bari, Italy.
| |
Collapse
|
107
|
Morimoto J, Pietras Z. Proteome Size Is Positively Correlated with Lifespan in Mammals but Negatively Correlated with Lifespan in Birds. Adv Biol (Weinh) 2025; 9:e2400633. [PMID: 39957468 PMCID: PMC12001000 DOI: 10.1002/adbi.202400633] [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: 10/21/2024] [Revised: 01/08/2025] [Indexed: 02/18/2025]
Abstract
The central dogma describes the unidirectional flow of genetic information from DNA to proteins, leading to an underappreciation of the potential for the information contained in proteomes (the full set of proteins in an organism) to reflect broader biological processes such as lifespan. Here, this is addressed by examining how the size and composition of 276 proteomes from four vertebrate classes are related to lifespan. After accounting for the relationship between body weight and lifespan, lifespan is negatively correlated with proteome size in birds and, to a weaker extent, in fish, and positively correlated with lifespan in mammals. Proteome composition varies amongst the four vertebrate classes, but there is no evidence that any specific amino acid correlated with lifespan. The findings in relation to the role of dietary amino acid restriction are discussed on lifespan extension and raise questions about evolutionary and structural forces shaping proteome composition across species.
Collapse
Affiliation(s)
- Juliano Morimoto
- Institute of MathematicsSchool of Natural and Computing SciencesUniversity of AberdeenFraser Noble BuildingAberdeenAB24 3UEUK
- Programa de Pós‐graduação em Ecologia e ConservaçãoUniversidade Federal do ParanáCuritiba82590‐300Brazil
| | - Zuzanna Pietras
- Department of PhysicsChemistry and Biology (IFM)Linköping UniversityLinköping581 83Sweden
| |
Collapse
|
108
|
Shen L, Ye X, Wang X, Song C, Yang B. Chinese Family With Knobloch Syndrome Associated With a Novel PAK2 Variant Leading to Reduced Phosphorylation Levels. Mol Genet Genomic Med 2025; 13:e70099. [PMID: 40247748 PMCID: PMC12006727 DOI: 10.1002/mgg3.70099] [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: 01/07/2025] [Revised: 03/28/2025] [Accepted: 04/08/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Biallelic variants of COL18A1 cause Knobloch syndrome (KNO), a rare genetic disorder, characterized by oculopathy and structural defects. Recently, several studies have suggested that novel de novo missense variants in PAK2 may be associated with KNO; however, there are few case reports. This study aimed to investigate a patient with KNO who initially presented with seizures and expand the PAK2 genotype and phenotype spectrum. METHODS This study included a Chinese family with a proband who primarily presented with epilepsy and developmental delay. Whole-exome sequencing and Sanger sequencing were performed to analyze potential variants. Structural modeling was performed to assess the impact of the variant on the protein structure. In vitro, a mutant plasmid was constructed and transfected into 293T cells to conduct phosphorylation assays, and phosphorylation levels at Ser141 of PAK2 were assessed. The PAK kinase inhibitor FRAX597 was used to confirm the specificity of the western blot results. RESULTS A de novo variant of PAK2 gene, NM_002577.4: c.1049G>A (p.Arg350Lys) was found in the patient but not in his parents or sister. This variant was found to be located in the kinase domain and may alter the hydrogen-bond network, potentially affecting kinase activity. In vitro functional experiments demonstrated that the variant may lead to reduced protein levels. Moreover, Western blot analysis showed a significant decrease in the phosphorylation level at Ser141 compared to the wild-type plasmid, indicating that the variant may lead to decreased PAK2 phosphorylation levels. CONCLUSION The clinical manifestations in this patient may be associated with a novel PAK2 variant, and the atypical presentation of KNO suggests that PAK2-related KNO may have a broader phenotypic spectrum.
Collapse
Affiliation(s)
- Liwei Shen
- Department of NeurologyAnhui Provincial Children's Hospital/Children's Hospital of Fudan University (Affiliated Anhui Branch)HefeiChina
| | - Xiaofei Ye
- Department of NeurologyAnhui Provincial Children's Hospital/Children's Hospital of Fudan University (Affiliated Anhui Branch)HefeiChina
| | - Xiaocui Wang
- Department of NeurologyAnhui Provincial Children's Hospital/Children's Hospital of Fudan University (Affiliated Anhui Branch)HefeiChina
| | - Conglei Song
- Department of NeurologyAnhui Provincial Children's Hospital/Children's Hospital of Fudan University (Affiliated Anhui Branch)HefeiChina
| | - Bin Yang
- Department of NeurologyAnhui Provincial Children's Hospital/Children's Hospital of Fudan University (Affiliated Anhui Branch)HefeiChina
| |
Collapse
|
109
|
Arany ES, Zocche D, Mellerio JE, Holder-Espinasse M, Cobben J. CHIME Syndrome in a Child With Homozygous PIGL p.Leu167Pro Variant. Am J Med Genet A 2025; 197:e63962. [PMID: 39641205 DOI: 10.1002/ajmg.a.63962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/07/2024] [Accepted: 11/24/2024] [Indexed: 12/07/2024]
Abstract
CHIME syndrome is a variable condition characterized by ichthyosiform dermatosis, accompanied by intellectual disability, ocular colobomas, ear anomalies, and heart defects. It is an autosomal recessive condition caused by biallelic pathogenic variants in the PIGL gene. Until now, all reports of individuals affected with CHIME syndrome showed the PIGL c.500T>C p.Leu167Pro DNA variant on one allele of the PIGL gene, in combination with another PIGL DNA variant on the other allele. This has led to the hypothesis that the p.Leu167Pro variant determines to a mild phenotypic effect only and that the core phenotype is determined by the second PIGL DNA variant. We report the first individual with CHIME syndrome, a 6-year-old girl, with homozygous PIGL p.Leu167Pro variants, defusing this hypothesis as she is not mildly affected. As CHIME is a very rare condition, it is expected that a significant proportion of cases will be due to homozygous gene variants, especially of founder DNA variants, and offspring of consanguineous parents. We speculate that the lack of homozygous p.Leu167Pro DNA variants so far has been due to chance and that other homozygous cases will be identified in future reports of affected individuals.
Collapse
Affiliation(s)
- Eszter Sara Arany
- Northwest Thames Regional Genetics Service, Northwick Park & St Mark's Hospitals, London, UK
- Imperial College London, London, UK
| | - David Zocche
- Northwest Thames Regional Genetics Service, Northwick Park & St Mark's Hospitals, London, UK
| | - Jemima E Mellerio
- St John's Institute of Dermatology, Guy's & St Thomas' NHS Foundation Trust, London, UK
| | | | - Jan Cobben
- Northwest Thames Regional Genetics Service, Northwick Park & St Mark's Hospitals, London, UK
- Imperial College London, London, UK
| |
Collapse
|
110
|
Lei Y, Tsang JS. Systems Human Immunology and AI: Immune Setpoint and Immune Health. Annu Rev Immunol 2025; 43:693-722. [PMID: 40279304 DOI: 10.1146/annurev-immunol-090122-042631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2025]
Abstract
The immune system, critical for human health and implicated in many diseases, defends against pathogens, monitors physiological stress, and maintains tissue and organismal homeostasis. It exhibits substantial variability both within and across individuals and populations. Recent technological and conceptual progress in systems human immunology has provided predictive insights that link personal immune states to intervention responses and disease susceptibilities. Artificial intelligence (AI), particularly machine learning (ML), has emerged as a powerful tool for analyzing complex immune data sets, revealing hidden patterns across biological scales, and enabling predictive models for individualistic immune responses and potentially personalized interventions. This review highlights recent advances in deciphering human immune variation and predicting outcomes, particularly through the concepts of immune setpoint, immune health, and use of the immune system as a window for measuring health. We also provide a brief history of AI; review ML modeling approaches, including their applications in systems human immunology; and explore the potential of AI to develop predictive models and personal immune state embeddings to detect early signs of disease, forecast responses to interventions, and guide personalized health strategies.
Collapse
Affiliation(s)
- Yona Lei
- Yale Center for Systems and Engineering Immunology and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA;
| | - John S Tsang
- Yale Center for Systems and Engineering Immunology and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA;
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
- Chan Zuckerberg Biohub NY, New Haven, Connecticut, USA
| |
Collapse
|
111
|
Liu X, Teng L, Sun J. Classification and prediction of variants associated with hearing loss using sequence information in the vicinity of mutation sites. Comput Biol Chem 2025; 115:108321. [PMID: 39675189 DOI: 10.1016/j.compbiolchem.2024.108321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/29/2024] [Accepted: 12/10/2024] [Indexed: 12/17/2024]
Abstract
Hearing impairment is a major global health problem, affecting more than 5 % of the world's population at various ages, from neonates to the elderly. Among the common genetic variations in humans, single nucleotide variations and small insertions or deletions predominate. The study of hearing loss resulting from these variations is proving invaluable in the analysis and diagnosis of hearing disorders. The identification of pathogenic mutations is frequently a lengthy and laborious process. Existing computational prediction tools have been developed primarily for common diseases and genome-wide analyses, with less focus on deafness. This study proposes a novel approach that focuses on the regions surrounding mutation sites. Mutation sites associated with deafness and their flanking regions of different lengths were extracted from relevant databases and combined into seven distinct segments of different lengths. The information-theoretic features of these segments were computed. Five machine learning algorithms were then used for training, resulting in the construction of a model capable of classifying and predicting deafness-related mutations. For fragments encompassing the 250 bp regions upstream and downstream of the mutations, the average AUC of the five classifiers on the independent test set is 0.89 and the average ACC is 0.85, indicating that the model has a high recognition rate of the pathogenic deafness mutation site. An ensemble approach was also applied to predict variants of uncertain significance (VUS) that may be associated with deafness. These variants were then scored and ranked to assess their likelihood of contributing to the condition.
Collapse
Affiliation(s)
- Xiao Liu
- School of Microelectronics and Communication Engineering, Chongqing University, Chongqing 401331, China.
| | - Li Teng
- School of Microelectronics and Communication Engineering, Chongqing University, Chongqing 401331, China
| | - Jing Sun
- School of Microelectronics and Communication Engineering, Chongqing University, Chongqing 401331, China
| |
Collapse
|
112
|
Dahlberg PM, Harris HK, Lloyd Holder J. Chronic Catatonia in an Individual With a De Novo Missense SHANK1 Variant. Am J Med Genet A 2025; 197:e63943. [PMID: 39569511 DOI: 10.1002/ajmg.a.63943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 10/31/2024] [Accepted: 11/08/2024] [Indexed: 11/22/2024]
Abstract
SHANK1 encodes a scaffolding protein of the SHANK family that includes SHANK1, SHANK2 and SHANK3. All of the SHANK proteins are enriched at the post-synaptic density of excitatory synapses. Here, we present an 11-year-old boy with a history of developmental delays and no family history of psychiatric disorders who developed catatonia. MRI of his brain and spine were negative as was a workup for autoimmune encephalitis. The proband's genetic testing revealed a de novo heterozygous SHANK1 missense variant. Although catatonia has been reported previously in individuals with SHANK3 loss-of-function mutations, this is the first time catatonia has been described in an individual with a SHANK1 variant.
Collapse
Affiliation(s)
- Paige M Dahlberg
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
| | - Holly K Harris
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - J Lloyd Holder
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
| |
Collapse
|
113
|
Iwama K, Kato M, Uchiyama Y, Sakamoto M, Miyamoto R, Izumi Y, Ohashi K, Hattori A, Yoshida N, Azuma Y, Watanabe A, Ikeda C, Shimizu-Motohashi Y, Kusabiraki S, Nakagawa E, Sasaki M, Sugai K, Ohori S, Tsuchida N, Hamanaka K, Koshimizu E, Fujita A, Nakashima M, Miyatake S, Sengoku T, Ogata K, Saitoh S, Saitsu H, Ito S, Mizuguchi T, Matsumoto N. Clinical and genetic spectrum of patients with IRF2BPL syndrome. J Hum Genet 2025; 70:181-188. [PMID: 39843638 DOI: 10.1038/s10038-025-01316-2] [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: 08/08/2024] [Revised: 12/25/2024] [Accepted: 01/10/2025] [Indexed: 01/24/2025]
Abstract
Interferon regulatory factor 2 binding protein-like (IRF2BPL) is a single-exon gene that is ubiquitously expressed in various tissues, including the brain. IRF2BPL encodes a transcription factor with two zinc-finger domains that potentially downregulate WNT signaling in the nervous system. Pathogenic IRF2BPL variants have been reported to cause developmental delay, seizures, myoclonus epilepsies, autistic spectrum disorder, and other neurodevelopmental disorders. Exome sequencing of 10 patients with developmental delay and/or epilepsy from nine families revealed nine pathogenic IRF2BPL variants, of which eight were novel: five missense, one in-frame indel, and three truncating variants. Using reported pathogenic and benign variants, we highlight here several regions of IRF2BPL that deviate in the frequency of pathogenic and benign variants. This study of detailed clinical and genetic information shows that IRF2BPL missense and in-frame indel variants are often associated with seizures and developmental delay.
Collapse
Affiliation(s)
- Kazuhiro Iwama
- Department of Human Genetics, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
- Department of Pediatrics, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Mitsuhiro Kato
- Department of Pediatrics, Showa University School of Medicine, Tokyo, Japan
| | - Yuri Uchiyama
- Department of Human Genetics, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
- Department of Rare Disease Genomics, Yokohama City University Hospital, Yokohama, Japan
| | - Masamune Sakamoto
- Department of Human Genetics, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
- Department of Pediatrics, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
- Department of Rare Disease Genomics, Yokohama City University Hospital, Yokohama, Japan
| | - Ryosuke Miyamoto
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Yuishin Izumi
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Kei Ohashi
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Ayako Hattori
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
- Department of Pediatrics, Nagoya City University East Medical Center, Nagoya, Japan
| | - Noboru Yoshida
- Department of Pediatrics, Juntendo University Nerima Hospital, Tokyo, Japan
| | - Yoshiteru Azuma
- Department of Pediatrics, Aichi Medical University, Nagakute, Japan
| | - Akito Watanabe
- Department of Pediatrics, Shizuoka Institute of Epilepsy and Neurological Disorders, National Epilepsy Center, Shizuoka, Japan
- Department of Pediatrics, Toyonaka Municipal Hospital, Toyonaka, Osaka, Japan
| | - Chizuru Ikeda
- Department of Pediatrics, Kumamoto Saishunso National Hospital, Koshi, Kumamoto, Japan
| | - Yuko Shimizu-Motohashi
- Department of Child Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Shohei Kusabiraki
- Department of Child Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Eiji Nakagawa
- Department of Child Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Masayuki Sasaki
- Department of Child Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
- Department of Pediatrics, Tokyo Children's Rehabilitation Hospital, Tokyo, Japan
| | - Kenji Sugai
- Department of Child Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
- Division of Pediatrics, Soleil Kawasaki Medical Center for the Severely Disabled, Kawasaki, Japan
| | - Sachiko Ohori
- Department of Laboratory Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Naomi Tsuchida
- Department of Human Genetics, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
- Department of Rare Disease Genomics, Yokohama City University Hospital, Yokohama, Japan
| | - Kohei Hamanaka
- Department of Human Genetics, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Eriko Koshimizu
- Department of Human Genetics, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Atsushi Fujita
- Department of Human Genetics, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Mitsuko Nakashima
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Satoko Miyatake
- Department of Human Genetics, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
- Department of Clinical Genetics, Yokohama City University Hospital, Yokohama City University, Yokohama, Japan
| | - Toru Sengoku
- Department of Biochemistry, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Kazuhiro Ogata
- Department of Biochemistry, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Shinji Saitoh
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hirotomo Saitsu
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Shuichi Ito
- Department of Pediatrics, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Takeshi Mizuguchi
- Department of Human Genetics, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Graduate School of Medicine, Yokohama City University, Yokohama, Japan.
- Department of Rare Disease Genomics, Yokohama City University Hospital, Yokohama, Japan.
- Department of Clinical Genetics, Yokohama City University Hospital, Yokohama City University, Yokohama, Japan.
| |
Collapse
|
114
|
Zhozhikov L, Vasilev F, Maksimova N. Protein-Variant-Phenotype Study of NBAS Using AlphaFold in the Aspect of SOPH Syndrome. Proteins 2025; 93:871-884. [PMID: 39641476 DOI: 10.1002/prot.26764] [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: 07/13/2024] [Revised: 10/04/2024] [Accepted: 11/01/2024] [Indexed: 12/07/2024]
Abstract
NBAS gene variants cause phenotypically distinct and nonoverlapping conditions, SOPH syndrome and ILFS2. NBAS is a so-called "moonlighting" protein responsible for retrograde membrane trafficking and nonsense-mediated decay. However, its three-dimensional model and the nature of its possible interactions with other proteins have remained elusive. Here, we used AlphaFold to predict protein-protein interaction (PPI) sites and mapped them to NBAS pathogenic variants. We repeated in silico milestone studies of the NBAS protein to explain the multisystem phenotype of its variants, with particular emphasis on the SOPH variant (p.R1914H). We revealed the putative binding sites for the main interaction partners of NBAS and assessed the implications of these binding sites for the subdomain architecture of the NBAS protein. Using AlphaFold, we disclosed the far-reaching impact of NBAS variants on the development of each phenotypic trait in patients with NBAS-related pathologies.
Collapse
Affiliation(s)
- Leonid Zhozhikov
- Research Laboratory of "Molecular Medicine and Human Genetics", Institute of Medicine, Ammosov North-Eastern Federal University, Yakutsk, Republic of Sakha (Yakutia), Russia
| | - Filipp Vasilev
- Research Laboratory of "Molecular Medicine and Human Genetics", Institute of Medicine, Ammosov North-Eastern Federal University, Yakutsk, Republic of Sakha (Yakutia), Russia
| | - Nadezhda Maksimova
- Research Laboratory of "Molecular Medicine and Human Genetics", Institute of Medicine, Ammosov North-Eastern Federal University, Yakutsk, Republic of Sakha (Yakutia), Russia
| |
Collapse
|
115
|
Försti A, Ambrozkiewicz F, Marciniak M, Lubinski J, Hemminki K. Search for germline gene variants in colorectal cancer families presenting with multiple primary colorectal cancers. Int J Cancer 2025; 156:1393-1403. [PMID: 39654522 PMCID: PMC11789446 DOI: 10.1002/ijc.35283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/15/2024] [Accepted: 11/22/2024] [Indexed: 02/04/2025]
Abstract
A double primary colorectal cancer (CRC) in a familial setting signals a high risk of CRC. In order to identify novel CRC susceptibility genes, we whole-exome sequenced germline DNA from nine persons with a double primary CRC and a family history of CRC. The detected variants were processed by bioinformatics filtering and prioritization, including STRING protein-protein interaction and pathway analysis. A total of 150 missense, 19 stop-gain, 22 frameshift and 13 canonical splice site variants fulfilled our filtering criteria. The STRING analysis identified 20 DNA repair/cell cycle proteins as the main cluster, related to genes CHEK2, EXO1, FAAP24, FANCI, MCPH1, POLL, PRC1, RECQL, RECQL5, RRM2, SHCBP1, SMC2, XRCC1, in addition to CDK18, ENDOV, ZW10 and the known mismatch repair genes. Another STRING network included extracellular matrix genes and TGFβ signaling genes. In the nine whole-exome sequenced patients, eight harbored at least two candidate DNA repair/cell cycle/TGFβ signaling gene variants. The number of families is too small to provide evidence for individual variants but, considering the known role of DNA repair/cell cycle genes in CRC, the clustering of multiple deleterious variants in the present families suggests that these, perhaps jointly, contributed to CRC development in these families.
Collapse
Affiliation(s)
- Asta Försti
- Hopp Children's Cancer Center (KiTZ)HeidelbergGermany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ)German Cancer Consortium (DKTK)HeidelbergGermany
| | - Filip Ambrozkiewicz
- Biomedical Center, Faculty of MedicineCharles University PilsenPilsenCzech Republic
| | - Magdalena Marciniak
- Department of Genetics and Pathology, International Hereditary Cancer CenterPomeranian Medical University in SzczecinSzczecinPoland
| | - Jan Lubinski
- Department of Genetics and Pathology, International Hereditary Cancer CenterPomeranian Medical University in SzczecinSzczecinPoland
| | - Kari Hemminki
- Biomedical Center, Faculty of MedicineCharles University PilsenPilsenCzech Republic
- Division of Cancer EpidemiologyGerman Cancer Research Center (DKFZ)HeidelbergGermany
| |
Collapse
|
116
|
Flores E, Acharya N, Castañeda CA, Sukenik S. Single-point mutations in disordered proteins: Linking sequence, ensemble, and function. Curr Opin Struct Biol 2025; 91:102987. [PMID: 39914051 DOI: 10.1016/j.sbi.2025.102987] [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: 11/13/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 03/08/2025]
Abstract
Mutations in genomic DNA often result in single-point missense mutations in proteins. For folded proteins, the functional effect of these missense mutations can often be understood by their impact on structure. However, missense mutations in intrinsically disordered protein regions (IDRs) remain poorly understood. In IDRs, function can depend on the structural ensemble- the collection of accessible, interchanging conformations that is encoded in their amino acid sequence. We argue that, analogously to folded proteins, single-point mutations in IDRs can alter their structural ensemble, and consequently alter their biological function. To make this argument, we first provide experimental evidence from the literature showcasing how single-point missense mutations in IDRs affect their ensemble dimensions. Then, we use genomic data from patients to show that disease-linked missense mutations occurring in IDRs can, in many cases, significantly alter IDR structural ensembles. We hope this analysis prompts further study of disease-linked, single-point mutations in IDRs.
Collapse
Affiliation(s)
- Eduardo Flores
- Department of Chemistry and Biochemistry, UC Merced, United States
| | | | - Carlos A Castañeda
- Department of Chemistry, Syracuse University, United States; Department of Biology, Syracuse University, United States; Bioinspired Institute, Syracuse University, United States.
| | - Shahar Sukenik
- Department of Chemistry and Biochemistry, UC Merced, United States; Department of Chemistry, Syracuse University, United States; Bioinspired Institute, Syracuse University, United States.
| |
Collapse
|
117
|
Uehara Y, Izumi H, Kobayashi IS, Matsumoto S, Hosomi Y, Okuno T, Sugisaka J, Takase N, Taima K, Sasaki S, Teranishi S, Miyamoto S, Mori M, Nakashima C, Asano S, Oi H, Sakai T, Shibata Y, Udagawa H, Sugiyama E, Nosaki K, Umemura S, Zenke Y, Yoh K, Ikeda S, Costa DB, Kobayashi SS, Goto K. Efficacy of EGFR tyrosine kinase inhibitors in patients with non-small cell lung cancer with EGFR exon 19 insertions: clinical-genomic, preclinical analysis through LC-SCRUM-Asia (multi-institutional genomic screening registry). Lung Cancer 2025; 202:108479. [PMID: 40088581 DOI: 10.1016/j.lungcan.2025.108479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/18/2025] [Accepted: 03/01/2025] [Indexed: 03/17/2025]
Abstract
BACKGROUND EGFR exon 19 insertions (EGFRex19ins) are rare EGFR mutations. Their clinical-genomic characteristics and outcomes with EGFR-tyrosine kinase inhibitors (TKIs) remain uncertain. METHODS We evaluated the clinical-genomic characteristics and outcomes of EGFR-TKIs for EGFRex19ins in the multi-institutional prospective lung cancer genomic screening project (LC-SCRUM-Asia). We also studied preclinical Ba/F3 models expressing EGFR-K745_E746insIPVAIK (Ba/F3-IPVAIK) to investigate their sensitivity to 1st-, 2nd-, 3rd-generation, and EGFR exon 20 insertion-active TKIs. RESULTS In LC-SCRUM-Asia, 16,204 NSCLC patients were enrolled from March 2015 to December 2023. EGFRex19ins were detected in 13 samples (0.1 % of NSCLC). The median age was 72 years (range, 38-80); most patients were female (77 %), had adenocarcinoma (92 %), and were never-smokers (62 %). Twelve patients (93 %) had EGFR-K745_E746insIPVAIK, while one (7 %) had EGFR-K745_E746insVPVAIK. The most frequent co-mutation was TP53 (62 %); no patients had other driver alterations. Six patients (46 %) tested positive for EGFR exon 19 deletions with PCR-based Cobas EGFR test, likely due to cross-reactivity arising from sequence homology. Twelve patients received EGFR-TKIs; five (42 %) experienced partial response. In the preclinical study, Ba/F3-IPVAIK showed the highest sensitivity to 2nd-generation EGFR-TKIs compared to other EGFR-TKIs. Structural studies supported these consistent results. When broken down by EGFR-TKI generations, response rates for 1st-, 2nd-, and 3rd-generation TKIs were 50 % (1/2), 80 % (4/5), and 0 % (0/5), respectively. The median PFS for 1st-, 2nd-, and 3rd-generation TKIs were 8.7 (95 % CI, 7.4-NR), 14.7 (95 % CI, 8.0-NR), and 4.4 (95 % CI, 3.4-NR) months, respectively. CONCLUSION Our preclinical, structural, and clinical findings indicate 2nd-generation EGFR-TKIs are more effective for EGFRex19ins compared to other TKIs.
Collapse
Affiliation(s)
- Yuji Uehara
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan; Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan; Department of Precision Cancer Medicine, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroki Izumi
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan.
| | - Ikei S Kobayashi
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Shingo Matsumoto
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yukio Hosomi
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Takae Okuno
- Division of Medical Oncology & Respiratory Medicine, Department of Internal Medicine, Shimane University Faculty of Medicine, Izumo, Japan
| | - Jun Sugisaka
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Naoto Takase
- Department of Medical Oncology, Takarazuka City Hospital, Takarazuka, Japan
| | - Kageaki Taima
- Department of Respiratory Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Shinichi Sasaki
- Department of Respiratory Medicine, Juntendo University Urayasu Hospital, Urayasu, Japan
| | - Shuhei Teranishi
- Respiratory Disease Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Shingo Miyamoto
- Department of Medical Oncology, Japanese Red Cross Medical Center, Tokyo, Japan
| | - Masahide Mori
- Department of Thoracic Oncology, NHO Osaka Toneyama Medical Center, Toyonaka, Japan
| | - Chiho Nakashima
- Division of Hematology, Respiratory Medicine and Oncology, Faculty of Medicine, Saga University, Saga, Japan
| | - Shuichi Asano
- Department of Respirology, Japan Community Health Care Organization Chukyo Hospital, Nagoya, Japan
| | - Hajime Oi
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Tetsuya Sakai
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yuji Shibata
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hibiki Udagawa
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Eri Sugiyama
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kaname Nosaki
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Shigeki Umemura
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yoshitaka Zenke
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kiyotaka Yoh
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Sadakatsu Ikeda
- Department of Precision Cancer Medicine, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University, Tokyo, Japan
| | - Daniel B Costa
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Susumu S Kobayashi
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States; Division of Translational Genomics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan.
| | - Koichi Goto
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| |
Collapse
|
118
|
Loughrey PB, Mothojakan NB, Iacovazzo D, Arni A, Aflorei ED, Arnaldi G, Barlier A, Beckers A, Bizzi MF, Chanson P, Dal J, Daly AF, Dang MN, David A, Andrade MDO, Else T, Elston MS, Evans A, Ferrau F, Fica S, Flanagan D, Gadelha MR, Grossman AB, Kapur S, Khoo B, Kumar AV, Kumar-Sinha C, Lechan RM, Ludman M, Metherell LA, Miljic D, Mourougavelou V, Musat M, Occhi G, Owens M, Pascanu I, Pinheiro SVB, Radian S, Ribeiro-Oliveira A, Schöfl C, Patel KA, Hernández-Ramírez LC, Korbonits M. Reassessing the role of the p.(Arg304Gln) missense AIP variant in pituitary tumorigenesis. Eur J Endocrinol 2025; 192:385-397. [PMID: 40070360 PMCID: PMC11962913 DOI: 10.1093/ejendo/lvaf044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/02/2024] [Accepted: 03/10/2025] [Indexed: 04/03/2025]
Abstract
OBJECTIVE Heterozygous germline loss-of-function variants in AIP are associated with young-onset growth hormone and/or prolactin-secreting pituitary tumours. However, the pathogenic role of the c.911G > A; p.(Arg304Gln) (R304Q) AIP variant has been controversial. Recent data from public exome/genome databases show this variant is not infrequent. The objective of this work was to reassess the pathogenicity of R304Q based on clinical, genomic, and functional assay data. DESIGN Data were collected on published R304Q pituitary neuroendocrine tumour cases and from International Familial Isolated Pituitary Adenoma Consortium R304Q cases (n = 38, R304Q cohort). Clinical features, population cohort frequency, computational analyses, prediction models, presence of loss-of-heterozygosity, and in vitro/in vivo functional studies were assessed and compared with data from pathogenic/likely pathogenic AIP variant patients (AIPmut cohort, n = 184). RESULTS Of 38 R304Q patients, 61% (23/38) had growth hormone excess, in contrast to 80% of AIPmut cohort (147/184, P < .001). R304Q cohort was older at disease onset and diagnosis than the AIPmut cohort (median [quartiles] onset: 25 y [16-35] vs 16 y [14-23], P < .001; median [quartiles] diagnosis: 36 y [24-44] vs 21 y [15-29], P < .001). R304Q is present in gnomADv2.1 (0.31%) and UK Biobank (0.16%), including three persons with homozygous R304Q. No loss-of-heterozygosity was detected in four R304Q pituitary neuroendocrine tumour samples. In silico predictions and experimental data were conflicting. CONCLUSIONS Evidence suggests that R304Q is not pathogenic for pituitary neuroendocrine tumour. We recommend changing this variant classification to likely benign and do not recommend pre-symptomatic genetic testing of family members or follow-up of already identified unaffected individuals with the R304Q variant.
Collapse
Affiliation(s)
- Paul Benjamin Loughrey
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, BT9 7AE, United Kingdom
| | - Nadira B Mothojakan
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Donato Iacovazzo
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Ankit Arni
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter, EX1 2HZ, United Kingdom
| | - Elena D Aflorei
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Giorgio Arnaldi
- Section of Endocrinology, PROMISE, University of Palermo, Palermo 90127, Italy
- Unità Operativa Complessa of Endocrine Diseases, A.O.U.P. Paolo Giaccone of Palermo, Palermo 90127, Italy
| | - Anne Barlier
- Aix Marseille Univ APHM, INSERM, UMR1251 MMG, Laboratory of Molecular Biology GEnOPé, Biogénopôle, Hôpital de la Timone, Marseille 13385, France
| | - Albert Beckers
- Department of Endocrinology, Centre Hospitalier Universitaire de Liège, University of Liège, Liège 4000, Belgium
| | - Mariana F Bizzi
- Department of Internal Medicine, Federal University of Minas Gerais, Belo Horizonte/Minas Gerais, 30130-100, Brazil
| | - Philippe Chanson
- Université Paris-Saclay, Inserm, Physiologie et Physiopathologie Endocriniennes, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Service d’Endocrinologie et des Maladies de la Reproduction, Centre de Référence des Maladies Rares de l’Hypophyse HYPO, Le Kremlin-Bicêtre, 94275, France
| | - Jakob Dal
- Department of Endocrinology, Aalborg University Hospital, Aalborg 9000, Denmark
| | - Adrian F Daly
- Department of Endocrinology, Centre Hospitalier Universitaire de Liège, University of Liège, Liège 4000, Belgium
| | - Mary N Dang
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Alessia David
- Centre for Bioinformatics, Department of Life Sciences, Imperial College London, London, SW7 2AZ, United Kingdom
| | - Matheus de Oliveira Andrade
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
- Faculty of Medicine, University of Brasilia, Brasilia 70910-900, Brazil
| | - Tobias Else
- MEND/Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI 48109, United States
| | - Marianne S Elston
- Waikato Clinical Campus, The University of Auckland, Hamilton 3216, New Zealand
| | - Amy Evans
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Francesco Ferrau
- Department of Human Pathology of Adulthood and Childhood ‘G. Barresi’, University of Messina, Messina 98125, Italy
| | - Simona Fica
- Endocrinology and Diabetes Department, Elias Hospital, University of Medicine and Pharmacy Carol Davila Bucharest, Bucharest 011461, Romania
| | - Daniel Flanagan
- Department of Endocrinology, University Hospitals Plymouth NHS Trust, Plymouth, PL6 8DH, United Kingdom
| | - Monica R Gadelha
- Endocrinology Unit, Department of Internal Medicine, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-853, Brazil
| | - Ashley B Grossman
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Sonal Kapur
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Bernard Khoo
- Division of Medicine, University College London, Royal Free Campus, London, NW3 2PS, United Kingdom
| | - Ajith V Kumar
- North East Thames Regional Genetics Service, Great Ormond Street Hospital for Children, London, WC1N 3BH, United Kingdom
| | - Chandan Kumar-Sinha
- Michigan Center for Translational Pathology, Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109-0940, United States
| | - Ronald M Lechan
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Tupper Research Institute, Tufts Medical Center, Boston, MA 02111, United States
| | - Mark Ludman
- Institute of Genetics, Meir Medical Center, Kfar Saba, 4428164, Israel
| | - Louise A Metherell
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Dragana Miljic
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Center of Serbia, Belgrade 11000, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade 11000, Serbia
| | - Vishnou Mourougavelou
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Madalina Musat
- National Institute of Endocrinology, University of Medicine and Pharmacy Carol Davila Bucharest, Bucharest 050474, Romania
| | - Gianluca Occhi
- Department of Biology, University of Padua, Padua 35128, Italy
| | - Martina Owens
- Exeter Genomics Laboratory, Royal Devon University Healthcare NHS Foundation Trust, Exeter, EX2 5DW, United Kingdom
| | - Ionela Pascanu
- Department of Endocrinology, George Emil Palade University of Medicine Pharmacy Science and Technology of Targu Mures, Targu Mures 540139, Romania
| | - Sergio V B Pinheiro
- Department of Pediatrics, Federal University of Minas Gerais, Belo Horizonte/Minas Gerais 30130-100, Brazil
| | - Serban Radian
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Antonio Ribeiro-Oliveira
- Department of Internal Medicine, Federal University of Minas Gerais, Belo Horizonte/Minas Gerais, 30130-100, Brazil
| | - Christof Schöfl
- Center of Endocrinology and Metabolism, Bamberg and Erlangen, Obstmarkt 1, Bamberg 96047, Germany
| | - Kashyap A Patel
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter, EX1 2HZ, United Kingdom
| | - Laura C Hernández-Ramírez
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
- Red de Apoyo a la Investigación, Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City 14080, Mexico
| | - Márta Korbonits
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| |
Collapse
|
119
|
Ly J, Tao YF, Di Bernardo M, Khalizeva E, Giuliano CJ, Lourido S, Fleming MD, Cheeseman IM. Alternative start codon selection shapes mitochondrial function during evolution, homeostasis, and disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.27.645657. [PMID: 40196624 PMCID: PMC11974929 DOI: 10.1101/2025.03.27.645657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Mitochondrial endosymbiosis was a pivotal event in eukaryotic evolution, requiring core proteins to adapt to function both within the mitochondria and in the host cell. Here, we systematically profile the localization of protein isoforms generated by alternate start codon selection during translation. We identify hundreds of pairs of differentially-localized protein isoforms, many of which affect mitochondrial targeting and are essential for mitochondrial function. The emergence of dual-localized mitochondrial protein isoforms coincides with mitochondrial acquisition during early eukaryotic evolution. We further reveal that eukaryotes use diverse mechanisms-such as leaky ribosome scanning, alternative transcription, and paralog duplication-to maintain the production of dual-localized isoforms. Finally, we identify multiple isoforms that are specifically dysregulated by rare disease patient mutations and demonstrate how these mutations can help explain unique clinical presentations. Together, our findings illuminate the evolutionary and pathological relevance of alternative translation initiation, offering new insights into the molecular underpinnings of mitochondrial biology.
Collapse
Affiliation(s)
- Jimmy Ly
- Whitehead Institute for Biomedical Research, Cambridge, United States
- Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| | - Yi Fei Tao
- Whitehead Institute for Biomedical Research, Cambridge, United States
- Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| | - Matteo Di Bernardo
- Whitehead Institute for Biomedical Research, Cambridge, United States
- Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| | - Ekaterina Khalizeva
- Whitehead Institute for Biomedical Research, Cambridge, United States
- Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| | - Christopher J. Giuliano
- Whitehead Institute for Biomedical Research, Cambridge, United States
- Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| | - Sebastian Lourido
- Whitehead Institute for Biomedical Research, Cambridge, United States
- Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| | - Mark D. Fleming
- Department of Pathology, Boston Children's Hospital, Boston, Massachusetts, United States
| | - Iain M. Cheeseman
- Whitehead Institute for Biomedical Research, Cambridge, United States
- Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| |
Collapse
|
120
|
Malik S, Jawad Ul Hasnain M, Zaib G, Saadia H, Malik A, Zahid A. Comprehensive structural and functional analyses of RAD50 nsSNPs: from prediction to impact assessment. FRONTIERS IN BIOINFORMATICS 2025; 5:1535482. [PMID: 40206634 PMCID: PMC11979129 DOI: 10.3389/fbinf.2025.1535482] [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: 11/27/2024] [Accepted: 02/24/2025] [Indexed: 04/11/2025] Open
Abstract
Background The RAD50 gene on chromosome 5q3.11 plays an important role in the MRN (Mre11-Rad50-Nbs1) complex. This complex orchestrates cellular responses to the DNA double-strand breaks (DSBs) through several pathways for genome stability. This study aims to investigate the functional impact of non-synonymous single-nucleotide polymorphisms (nsSNPs) in RAD50 (a breast cancer-associated gene) and focuses on their consequences on protein structure and interaction within the MRN complex. Methods A total of 1,806 nsSNPs were retrieved and subjected to variant analysis using a set of computational tools and ConSurf. Pathogenicity and protein stability criteria were established based on specific tools. Highly conserved damaging nsSNPs were prioritized for the structural analysis. GOR-IV was used for secondary structure prediction, whereas AlphaFold, RoseTTAFold, and I-TASSER were used for protein structure prediction. The docking of RAD50-Mre11A complexes was performed using HADDOCK to assess the impact of nsSNPs on protein-protein interactions. Molecular dynamic simulation was performed to verify the role of mutants in molecular docking analysis. Results A subset of pathogenic and disease-associated nsSNPs in the RAD50 gene altered the protein stability and interactions with the Mre11A protein. Substantial alterations in the interacting profiles of mutants (A73P, V117F, L518P, L1092R, N1144S, and A1209T) suggest potential implications for DNA repair mechanisms and genome stability. Conclusion The study discloses the normative impact of RAD50 mutations on the pathophysiology of breast cancer. It can provide the basis to treat RAD50 mutation-deficient cells.
Collapse
Affiliation(s)
- Samina Malik
- University College of Medicine and Dentistry, The University of Lahore, IMBB, UOL, Lahore, Pakistan
| | - Mirza Jawad Ul Hasnain
- Department of Biological Sciences, Virtual University of Pakistan, Islamabad, Pakistan
- Center for Bioinformatics and Computational Biology, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Gul Zaib
- School of Pain and Regenerative Medicine, The University of Lahore, IMBB, UOL, Lahore, Pakistan
| | - Haleema Saadia
- School of Pain and Regenerative Medicine, The University of Lahore, IMBB, UOL, Lahore, Pakistan
| | - Arif Malik
- School of Pain and Regenerative Medicine, The University of Lahore, IMBB, UOL, Lahore, Pakistan
| | - Ayesha Zahid
- School of Pain and Regenerative Medicine, The University of Lahore, IMBB, UOL, Lahore, Pakistan
| |
Collapse
|
121
|
Moth CW, Sheehan JH, Mamun AA, Sivley RM, Gulsevin A, Rinker D, Capra JA, Meiler J. VUStruct: a compute pipeline for high throughput and personalized structural biology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.06.606224. [PMID: 39149406 PMCID: PMC11326201 DOI: 10.1101/2024.08.06.606224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Effective diagnosis and treatment of rare genetic disorders requires the interpretation of a patient's genetic variants of unknown significance (VUSs). Today, clinical decision-making is primarily guided by gene-phenotype association databases and DNA-based scoring methods. Our web-accessible variant analysis pipeline, VUStruct, supplements these established approaches by deeply analyzing the downstream molecular impact of variation in context of 3D protein structure. VUStruct's growing impact is fueled by the co-proliferation of protein 3D structural models, gene sequencing, compute power, and artificial intelligence. Contextualizing VUSs in protein 3D structural models also illuminates longitudinal genomics studies and biochemical bench research focused on VUS, and we created VUStruct for clinicians and researchers alike. We now introduce VUStruct to the broad scientific community as a mature, web-facing, extensible, High-Performance Computing (HPC) software pipeline. VUStruct maps missense variants onto automatically selected protein structures and launches a broad range of analyses. These include energy-based assessments of protein folding and stability, pathogenicity prediction through spatial clustering analysis, and machine learning (ML) predictors of binding surface disruptions and nearby post-translational modification sites. The pipeline also considers the entire input set of VUS and identifies genes potentially involved in digenic disease. VUStruct's utility in clinical rare disease genome interpretation has been demonstrated through its analysis of over 175 Undiagnosed Disease Network (UDN) Patient cases. VUStruct-leveraged hypotheses have often informed clinicians in their consideration of additional patient testing, and we report here details from two cases where VUStruct was key to their solution. We also note successes with academic research collaborators, for whom VUStruct has informed research directions in both computational genomics and wet lab studies.
Collapse
Affiliation(s)
- Christopher W. Moth
- Departments of Chemistry, Pharmacology, and Biomedical Informatics; Center for Structural Biology and Institute of Chemical Biology; Vanderbilt Univ., Nashville, TN 37232, USA
| | - Jonathan H. Sheehan
- Division of Infection Diseases, Milliken Dept. of Internal Medicine, Washington Univ. of Medicine in St. Louis, MO 63110, USA
| | - Abdullah Al Mamun
- Departments of Chemistry, Pharmacology, and Biomedical Informatics; Center for Structural Biology and Institute of Chemical Biology; Vanderbilt Univ., Nashville, TN 37232, USA
| | | | - Alican Gulsevin
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Butler University, Indianapolis, IN 46208, USA
| | - David Rinker
- Department of Biological Sciences, Evolutionary Studies Initiative; Vanderbilt Univ., Nashville, TN 37232, USA
| | | | - John A. Capra
- Bakar Computational Health Science Institute and Department of Epidemiology and Biostatistics, Univ. of California San Francisco, CA 94143, USA
| | - Jens Meiler
- Departments of Chemistry, Pharmacology, and Biomedical Informatics; Center for Structural Biology and Institute of Chemical Biology; Vanderbilt Univ., Nashville, TN 37232, USA
- Leipzig University Medical School, Institute for Drug Discovery, Brüderstraße 34, 04103 Leipzig, Germany
| |
Collapse
|
122
|
Zhang J, Ali MY, Chong HB, Tien PC, Woods J, Noble C, Vornbäumen T, Ordulu Z, Possemato AP, Harry S, Fonticella JM, Fellah L, Harrison D, Ge M, Khandelwal N, Huang Y, Chauvin M, Bischof AT, Hambelton GM, Gohar MF, Zhang S, Choi M, Bouberhan S, Oliva E, Mino-Kenudson M, Pavlova NN, Lawrence M, Gainor JF, Beausoleil SA, Bardeesy N, Mostoslavsky R, Pépin D, Ott CJ, Liau B, Bar-Peled L. Oxidation of retromer complex controls mitochondrial translation. Nature 2025:10.1038/s41586-025-08756-y. [PMID: 40140582 DOI: 10.1038/s41586-025-08756-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 02/07/2025] [Indexed: 03/28/2025]
Abstract
Reactive oxygen species (ROS) underlie human pathologies including cancer and neurodegeneration1,2. However, the proteins that sense ROS levels and regulate their production through their cysteine residues remain ill defined. Here, using systematic base-editing and computational screens, we identify cysteines in VPS35, a member of the retromer trafficking complex3, that phenocopy inhibition of mitochondrial translation when mutated. We find that VPS35 underlies a reactive metabolite-sensing pathway that lowers mitochondrial translation to decrease ROS levels. Intracellular hydrogen peroxide oxidizes cysteine residues in VPS35, resulting in retromer dissociation from endosomal membranes and subsequent plasma membrane remodelling. We demonstrate that plasma membrane localization of the retromer substrate SLC7A1 is required to sustain mitochondrial translation. Furthermore, decreasing VPS35 levels or oxidation of its ROS-sensing cysteines confers resistance to ROS-generating chemotherapies, including cisplatin, in ovarian cancer models. Thus, we identify that intracellular ROS levels are communicated to the plasma membrane through VPS35 to regulate mitochondrial translation, connecting cytosolic ROS sensing to mitochondrial ROS production.
Collapse
Affiliation(s)
- Junbing Zhang
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA.
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Md Yousuf Ali
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Harrison Byron Chong
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Pei-Chieh Tien
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - James Woods
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Carolina Noble
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Tristan Vornbäumen
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Zehra Ordulu
- Brigham and Women's Hospital, Department of Pathology, Harvard Medical School, MA, USA
| | | | - Stefan Harry
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Jay Miguel Fonticella
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Lina Fellah
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Drew Harrison
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Maolin Ge
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Neha Khandelwal
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Yingfei Huang
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Maëva Chauvin
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA
| | - Anica Tamara Bischof
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | | | - Magdy Farag Gohar
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Siwen Zhang
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - MinGyu Choi
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sara Bouberhan
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Division of Hematology/Oncology, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Esther Oliva
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Mari Mino-Kenudson
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Natalya N Pavlova
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Michael Lawrence
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Justin F Gainor
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | | | - Nabeel Bardeesy
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Raul Mostoslavsky
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - David Pépin
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA
| | - Christopher J Ott
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Brian Liau
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Liron Bar-Peled
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA.
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
123
|
Wang SK, Li J, Nair S, Korasaju R, Chen Y, Zhang Y, Kundaje A, Liu Y, Wang N, Chang HY. Single-cell multiome and enhancer connectome of human retinal pigment epithelium and choroid nominate pathogenic variants in age-related macular degeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.21.644670. [PMID: 40196652 PMCID: PMC11974679 DOI: 10.1101/2025.03.21.644670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Age-related macular degeneration (AMD) is a leading cause of vision loss worldwide. Genome-wide association studies (GWAS) of AMD have identified dozens of risk loci that may house disease targets. However, variants at these loci are largely noncoding, making it difficult to assess their function and whether they are causal. Here, we present a single-cell gene expression and chromatin accessibility atlas of human retinal pigment epithelium (RPE) and choroid to systematically analyze both coding and noncoding variants implicated in AMD. We employ HiChIP and Activity-by-Contact modeling to map enhancers in these tissues and predict cell and gene targets of risk variants. We further perform allele-specific self-transcribing active regulatory region sequencing (STARR-seq) to functionally test variant activity in RPE cells, including in the context of complement activation. Our work nominates new pathogenic variants and mechanisms in AMD and offers a rich and accessible resource for studying diseases of the RPE and choroid.
Collapse
Affiliation(s)
- Sean K Wang
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA, USA
- Department of Ophthalmology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jiaying Li
- Department of Ophthalmology, Stanford University School of Medicine, Stanford, CA, USA
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Surag Nair
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Reshma Korasaju
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA, USA
| | - Yun Chen
- Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Yuanyuan Zhang
- Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Anshul Kundaje
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Yuwen Liu
- Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- Kunpeng Institute of Modern Agriculture at Foshan, Chinese Academy of Agricultural Sciences, Foshan, China
| | - Ningli Wang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Henan Academy of Innovations in Medical Science, Henan, China
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Current address: Amgen Research, South San Francisco, CA, USA
- Lead contact
| |
Collapse
|
124
|
Jin F, Cheng N, Wang L, Ye B, Xia J. FDPSM: Feature-Driven Prediction Modeling of Pathogenic Synonymous Mutations. J Chem Inf Model 2025; 65:3064-3076. [PMID: 40082068 DOI: 10.1021/acs.jcim.4c02139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Synonymous mutations, once considered to be biologically neutral, are now recognized to affect protein expression and function by altering the RNA splicing, stability, or translation efficiency. These effects can contribute to disease, making the prediction of the pathogenicity a crucial task. Computational methods have been developed to analyze the sequence features and biological functions of synonymous mutations, but existing methods face limitations, including scarcity of labeled data, reliance on other prediction tools, and insufficient representation of feature interrelationships. Here, we present FDPSM, a novel prediction method specifically designed to predict pathogenic synonymous mutations. FDPSM was trained on a robust data set of 4251 positive and negative training samples to enhance predictive accuracy. The method leveraged a comprehensive set of features, including genomic context, conservation, splicing effects, functional effects, and epigenomics, without relying on prediction scores from other mutation pathogenicity tools. Recognizing that original features alone may not fully capture the distinctions between pathogenic and benign synonymous mutations, we enhanced the feature set by extracting effective information from the interactions and distribution of these features. The experimental results showed that FDPSM significantly outperformed existing methods in predicting the pathogenicity of synonymous mutations, offering a more accurate and reliable tool for this important task. FDPSM is available at https://github.com/xialab-ahu/FDPSM.
Collapse
Affiliation(s)
- Fangfang Jin
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China
| | - Na Cheng
- School of Biomedical Engineering, Anhui Medical University, Hefei, Anhui 230032, China
| | - Lihua Wang
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China
- School of Information Engineering, Huangshan University, Huangshan, Anhui 245041, China
| | - Bin Ye
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China
- School of Computer Science and Technology, Anhui University, Hefei, Anhui 230601, China
| | - Junfeng Xia
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China
| |
Collapse
|
125
|
Radjasandirane R, Diharce J, Gelly JC, de Brevern AG. Insights for variant clinical interpretation based on a benchmark of 65 variant effect predictors. Genomics 2025; 117:111036. [PMID: 40127826 DOI: 10.1016/j.ygeno.2025.111036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/20/2025] [Accepted: 03/20/2025] [Indexed: 03/26/2025]
Abstract
Single amino acid substitutions in protein sequences are generally harmless, but a certain number of these changes can lead to disease. Accurately predicting the effect of genetic variants is crucial for clinicians as it accelerates the diagnosis of patients with missense variants associated with health problems. Many computational tools have been developed to predict the pathogenicity of genetic variants with various approaches. Analysing the performance of these different computational tools is crucial to provide guidance to both future users and especially clinicians. In this study, a large-scale investigation of 65 tools was conducted. Variants from both clinical and functional contexts were used, incorporating data from the ClinVar database and bibliographic sources. The analysis showed that AlphaMissense often performed very well and was in fact one of the best options among the existing tools. In addition, as expected, meta-predictors perform well on average. Tools using evolutionary information showed the best performance for functional variants. These results also highlighted some heterogeneity in the difficulty of predicting some specific variants while others are always well categorized. Strikingly, the majority of variants from the ClinVar database appear to be easy to predict, while variants from other sources of data are more challenging. This raises questions about the use of ClinVar and the dataset used to validate tools accuracy. In addition, these results show that this variant predictability can be divided into three distinct classes: easy, moderate and hard to predict. We analyzed the parameters leading to these differences and showed that the classes are related to structural and functional information.
Collapse
Affiliation(s)
- Ragousandirane Radjasandirane
- Université Paris Cité and Université de la Réunion, INSERM, EFS, BIGR U1134, DSIMB Bioinformatics team, F-75015 Paris, France
| | - Julien Diharce
- Université Paris Cité and Université de la Réunion, INSERM, EFS, BIGR U1134, DSIMB Bioinformatics team, F-75015 Paris, France
| | - Jean-Christophe Gelly
- Université Paris Cité and Université de la Réunion, INSERM, EFS, BIGR U1134, DSIMB Bioinformatics team, F-75015 Paris, France
| | - Alexandre G de Brevern
- Université Paris Cité and Université de la Réunion, INSERM, EFS, BIGR U1134, DSIMB Bioinformatics team, F-75015 Paris, France.
| |
Collapse
|
126
|
Weston T, Ng J, Gracia Carmona O, Gautel M, Fraternali F. TITINdb2-expanding annotation and structural information for protein variants in the giant sarcomeric protein titin. BIOINFORMATICS ADVANCES 2025; 5:vbaf062. [PMID: 40270927 PMCID: PMC12017618 DOI: 10.1093/bioadv/vbaf062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 02/28/2025] [Accepted: 03/20/2025] [Indexed: 04/25/2025]
Abstract
Summary We present TITINdb2, an update to the TITINdb database previously constructed to facilitate the identification of pathogenic missense variants in the giant protein titin, which are associated with a variety of skeletal and cardiac myopathies. The database and web portal have been substantially revised and include the following new features: (i) an increase in computational annotation from 4 to 20 variant impact predictors, available through a new custom data table dialogue; (ii) through structural coverage of single domains with AlphaFold2 predicted models; (iii) newly predicted domain-domain interface annotations; (iv) an expanded in silico saturation mutagenesis incorporating four variant impact predictors; (v) a comprehensive overhaul of available data, including population data sources and variants reported pathogenic in the literature; and (vi) a curated mapping of existing protein, transcript, and chromosomal sequence positions and a new variant conversion tool to translate variants in one format to any other format. Availability and implementation The database is accessible via titindb.kcl.ac.uk/TITINdb/.
Collapse
Affiliation(s)
- Timir Weston
- Randall Centre for Cell & Molecular Biophysics, School of Basic & Medical Biosciences, King’s College London, London SE1 1UL, United Kingdom
- Institute of Structural and Molecular Biology, University College London, London WC1E 6BT, United Kingdom
- Research Department of Structural and Molecular Biology, Division of Biosciences, University College London, London WC1E 6BT, United Kingdom
| | - Joseph Ng
- Institute of Structural and Molecular Biology, University College London, London WC1E 6BT, United Kingdom
- Research Department of Structural and Molecular Biology, Division of Biosciences, University College London, London WC1E 6BT, United Kingdom
- Department of Biological Sciences, Birkbeck, University of London, London WC1E 7HX, United Kingdom
| | - Oriol Gracia Carmona
- Randall Centre for Cell & Molecular Biophysics, School of Basic & Medical Biosciences, King’s College London, London SE1 1UL, United Kingdom
- Institute of Structural and Molecular Biology, University College London, London WC1E 6BT, United Kingdom
- Research Department of Structural and Molecular Biology, Division of Biosciences, University College London, London WC1E 6BT, United Kingdom
- Department of Biological Sciences, Birkbeck, University of London, London WC1E 7HX, United Kingdom
| | - Mathias Gautel
- Randall Centre for Cell & Molecular Biophysics, School of Basic & Medical Biosciences, King’s College London, London SE1 1UL, United Kingdom
| | - Franca Fraternali
- Institute of Structural and Molecular Biology, University College London, London WC1E 6BT, United Kingdom
- Research Department of Structural and Molecular Biology, Division of Biosciences, University College London, London WC1E 6BT, United Kingdom
- Department of Biological Sciences, Birkbeck, University of London, London WC1E 7HX, United Kingdom
| |
Collapse
|
127
|
Yamauchi K, Ku M, Mitchell DW, Shen A, Dauda K, Vanags L, Schmeckpeper J, Knollmann BC, O’Neill MJ, Kroncke BM. Structural Evaluation of RYR2-CPVT Missense Variants and Continuous Bayesian Estimates of their Penetrance. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.20.25324327. [PMID: 40166532 PMCID: PMC11957170 DOI: 10.1101/2025.03.20.25324327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Background Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT) is strongly associated with rare missense variants in RYR2, the gene encoding the intracellular calcium release channel RyR2. Precision medicine is complicated by incomplete penetrance, particularly in the case of RYR2-CPVT variants. Objective To improve structural understanding and clinical actionability of RYR2-CPVT incomplete penetrance. Methods We curated 179 manuscripts reviewed by three individuals to extrapolate RYR2-CPVT missense variant genotype-phenotype relationships. Purportedly neutral control variants were ascertained from RYR2 missense variants observed in gnomAD and ClinVar. We performed an RYR2-CPVT Bayesian penetrance analysis by conditioning a CPVT penetrance prior on variant-specific features (in silico and structural) calibrated by heterozygote phenotypes. We compared the calibration of our Bayesian penetrance estimates and our previous described structural density metric with in silico predictors REVEL, AlphaMissense and ClinVar annotations, using Spearman rank-order correlations, and Brier Scores. Penetrance estimates were superimposed upon a cryo-EM structure of RyR2 to investigate 'hot-spot' heterogeneity. Results From the literature and gnomAD, we identified 1,014 affected missense RYR2 heterozygotes (468 unique variants) among a total of 622,575 heterozygotes (5,181 unique variants). Among the predictors, our Bayesian prior score had the highest Spearman rank-order and lowest Brier scores, respectively (0.19; 0.0090), compared to ClinVar (0.083; 0.019), REVEL (0.16; 0.018), or AlphaMissense (0.18; 0.018). Penetrance estimates for all RYR2 missense variants are prospectively hosted at our Variant Browser website. Conclusions Bayesian penetrance scores outperform current tools in evaluating variant penetrance. We provide prospective CPVT penetrance values for 29,242 RYR2 missense variants at our online Variant Browser.
Collapse
Affiliation(s)
- Kohei Yamauchi
- Department of Cardiovascular Medicine, Shiga University of Medical Science, Otsu, Shiga Prefecture, Japan
| | - Matthew Ku
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Devyn W. Mitchell
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Alex Shen
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Kundivy Dauda
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Loren Vanags
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jeffrey Schmeckpeper
- Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Bjorn C. Knollmann
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Matthew J. O’Neill
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States
- Clinical Fellow, Harvard Medical School, Boston, MA, United States
| | - Brett M. Kroncke
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
128
|
Schmid EW, Walter JC. Predictomes, a classifier-curated database of AlphaFold-modeled protein-protein interactions. Mol Cell 2025; 85:1216-1232.e5. [PMID: 40015271 PMCID: PMC11931459 DOI: 10.1016/j.molcel.2025.01.034] [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: 04/03/2024] [Revised: 12/17/2024] [Accepted: 01/31/2025] [Indexed: 03/01/2025]
Abstract
Protein-protein interactions (PPIs) are ubiquitous in biology, yet a comprehensive structural characterization of the PPIs underlying cellular processes is lacking. AlphaFold-Multimer (AF-M) has the potential to fill this knowledge gap, but standard AF-M confidence metrics do not reliably separate relevant PPIs from an abundance of false positive predictions. To address this limitation, we used machine learning on curated datasets to train a structure prediction and omics-informed classifier (SPOC) that effectively separates true and false AF-M predictions of PPIs, including in proteome-wide screens. We applied SPOC to an all-by-all matrix of nearly 300 human genome maintenance proteins, generating ∼40,000 predictions that can be viewed at predictomes.org, where users can also score their own predictions with SPOC. High-confidence PPIs discovered using our approach enable hypothesis generation in genome maintenance. Our results provide a framework for interpreting large-scale AF-M screens and help lay the foundation for a proteome-wide structural interactome.
Collapse
Affiliation(s)
- Ernst W Schmid
- Department of Biological Chemistry & Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Johannes C Walter
- Department of Biological Chemistry & Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Boston, MA 02115, USA.
| |
Collapse
|
129
|
Bianco L, Navarro J, Michiels C, Sangermano R, Condroyer C, Antonio A, Antropoli A, Andrieu C, Place EM, Pierce EA, El Shamieh S, Smirnov V, Kalatzis V, Mansard L, Roux AF, Bocquet B, Sahel JA, Meunier I, Bujakowska KM, Audo I, Zeitz C. Identification of IDH3G, encoding the gamma subunit of mitochondrial isocitrate dehydrogenase, as a novel candidate gene for X-linked retinitis pigmentosa. Genet Med 2025; 27:101418. [PMID: 40119724 DOI: 10.1016/j.gim.2025.101418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 03/24/2025] Open
Abstract
PURPOSE Retinitis pigmentosa (RP) is a genetically heterogeneous group of retinal degenerative disorders characterized by the loss of rod and cone photoreceptors, leading to visual impairment and blindness. To date, to our knowledge, X-linked RP has been associated with variants in 3 genes (RPGR, RP2, and OFD1), whereas genetic defects at 3 loci (RP6, RP24, and RP34) are yet unidentified. The aim of this study was to identify a novel candidate gene underlying X-linked RP. METHODS Participants were identified from cohorts of genetically unsolved male individuals affected by RP, who underwent genome sequencing, exome sequencing, or candidate gene screening via direct Sanger sequencing at 3 referral centers. Specifically, 2 probands were identified at the National Reference Centre for Rare Retinal Diseases (Paris, France), 2 at the Massachusetts Eye and Ear Hospital (Boston, MA), and 1 at the National Reference Centre for Inherited Sensory Diseases (Montpellier, France). The pathogenicity of the identified variants was assessed using bioinformatic predictions, protein expression analyses, and mitochondrial function assays. RESULTS We identified 4 rare single-nucleotide variants in IDH3G (HGNC:5386), located at the RP34 locus on the X chromosome, and a complete gene deletion, in 5 unrelated male individuals affected with nonsyndromic RP. The variants segregated with the phenotype in all available family members. In all cases, the disease severity was intermediate. None had high myopia. IDH3G encodes the γ subunit of mitochondrial isocitrate dehydrogenase (IDH3), an enzyme involved in the citric acid cycle, which is expressed in the inner segments of photoreceptors. Variants in IDH3A and IDH3B, encoding the other subunits of IDH3, have already been associated with nonsyndromic autosomal recessive RP. Bioinformatic predictions and functional assays support a pathogenic role for the variants identified in this study, possibly through partial loss of enzymatic activity and mitochondrial function. CONCLUSION Our findings suggest that variants in IDH3G are a novel cause of X-linked RP.
Collapse
Affiliation(s)
- Lorenzo Bianco
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France; Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, Centre de Référence Maladies Rares REFERET and INSERM-DGOS CIC 1423, Paris, France; Department of Ophthalmology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Julien Navarro
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | | | - Riccardo Sangermano
- Ocular Genomics Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA
| | | | - Aline Antonio
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Alessio Antropoli
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France; Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, Centre de Référence Maladies Rares REFERET and INSERM-DGOS CIC 1423, Paris, France; Department of Ophthalmology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Camille Andrieu
- Department of Ophthalmology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Emily M Place
- Ocular Genomics Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA
| | - Eric A Pierce
- Ocular Genomics Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA
| | - Said El Shamieh
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France; Molecular Testing Laboratory, Department of Medical Laboratory Technology, Faculty of Health Sciences, Beirut Arab University, Beirut, Lebanon
| | - Vasily Smirnov
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France; Exploration de la Vision et Neuro-Ophtalmologie, CHU de Lille, Lille, France
| | - Vasiliki Kalatzis
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, Inserm, Montpellier, France
| | - Luke Mansard
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, Inserm, Montpellier, France
| | - Anne-Françoise Roux
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, Inserm, Montpellier, France
| | - Béatrice Bocquet
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, Inserm, Montpellier, France; National Reference Centre for Inherited Sensory Diseases, University of Montpellier, CHU, Montpellier, France
| | - José-Alain Sahel
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France; Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, Centre de Référence Maladies Rares REFERET and INSERM-DGOS CIC 1423, Paris, France; Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburg, PA
| | - Isabelle Meunier
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, Inserm, Montpellier, France; National Reference Centre for Inherited Sensory Diseases, University of Montpellier, CHU, Montpellier, France
| | - Kinga M Bujakowska
- Ocular Genomics Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA
| | - Isabelle Audo
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France; Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, Centre de Référence Maladies Rares REFERET and INSERM-DGOS CIC 1423, Paris, France.
| | - Christina Zeitz
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France.
| |
Collapse
|
130
|
Meisner JK, Renberg A, Smith ED, Tsan YC, Elder B, Bullard A, Merritt O, Zheng SL, Lakdawala N, Owens A, Ryan TD, Miller EM, Rossano J, Lin KY, Claggett B, Ashley E, Michels M, Lampert R, Stendahl JC, Abrams D, Semsarian C, Parikh VN, Wheeler M, Ingles J, Day SM, Saberi S, Russell MW, Previs M, Ho C, Ware JS, Helms AS. Low Penetrance Sarcomere Variants Contribute to Additive Risk in Hypertrophic Cardiomyopathy. Circulation 2025; 151:783-798. [PMID: 39633578 PMCID: PMC11913586 DOI: 10.1161/circulationaha.124.069398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 10/24/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Classically, hypertrophic cardiomyopathy (HCM) has been viewed as a single-gene (monogenic) disease caused by pathogenic variants in sarcomere genes. Pathogenic sarcomere variants are individually rare and convey high risk for developing HCM (highly penetrant). Recently, important polygenic contributions have also been characterized. Low penetrance sarcomere variants (LowSVs) at intermediate frequencies and effect sizes have not been systematically investigated. We hypothesize that LowSVs may be common in HCM with substantial influence on disease risk and severity. METHODS Among all sarcomere variants observed in the Sarcomeric Human Cardiomyopathy Registry (SHaRe), we identified putative LowSVs defined by (1) population frequency greater than expected for highly penetrant (monogenic) HCM (allele frequency >5×10-5 in the Genome Aggregation Database, gnomAD) and (2) moderate enrichment (>2×) in patients with HCM compared with gnomAD. LowSVs were examined for their association with disease severity and clinical outcomes. Functional effects of selected LowSVs were assessed using induced pluripotent stem cell-derived cardiomyocytes. Association of LowSVs with HCM-adjacent traits in the general population was tested using UK Biobank cardiac magnetic resonance imaging data. RESULTS Among 6045 patients and 1159 unique variants in sarcomere genes, 12 LowSVs were identified. LowSVs were collectively common in the general population (1:350) and moderately enriched in HCM (aggregate odds ratio, 14.9 [95% CI, 12.5-17.9]). Isolated LowSVs were associated with an older age of HCM diagnosis and fewer adverse events. However, LowSVs in combination with a pathogenic sarcomere variant conferred higher morbidity (eg, composite adverse event hazard ratio, 5.4 [95% CI, 3.0-9.8] versus single pathogenic sarcomere variant, 2.0 [95% CI, 1.8-2.2]; P<0.001). An intermediate functional impact was validated for 2 specific LowSVs-MYBPC3 c.442G>A (partial splice gain) and TNNT2 c.832C>T (intermediate effect on contractile mechanics). Cardiac magnetic resonance imaging analysis of the general population revealed 5 of 12 LowSVs were significantly associated with HCM-adjacent traits without overt HCM. CONCLUSIONS This study establishes a new class of low penetrance sarcomere variants that are relatively common in the population. When penetrant, isolated LowSVs cause mild HCM. In combination with pathogenic sarcomere variants, LowSVs markedly increase disease severity, supporting a clinically significant additive effect. Last, LowSVs also contribute to age-related remodeling even in the absence of overt HCM.
Collapse
Affiliation(s)
- Joshua K Meisner
- Department of Pediatrics, Division of Pediatric Cardiology, University of Michigan, Ann Arbor
| | - Aaron Renberg
- Cellular and Molecular Biology Program, Medical School, University of Michigan, Ann Arbor
| | - Eric D Smith
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor
| | - Yao-Chang Tsan
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor
| | - Brynn Elder
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor
| | - Abbey Bullard
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor
| | - Owen Merritt
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor
| | - Sean L Zheng
- National Heart and Lung Institute and MRC Laboratory of Medical Sciences, Imperial College London, United Kingdom
| | - Neal Lakdawala
- Cardiovascular Division, Brigham and Women’s Hospital, Boston, MA
| | - Anjali Owens
- Penn Center for Inherited Cardiovascular Disease, Hospital of the University of Pennsylvania & Perelman School of Medicine at the University of Pennsylvania, Philadelphia (A.O., S.M.D.)
| | - Thomas D Ryan
- Department of Pediatrics, University of Cincinnati College of Medicine, Heart Institute, Cincinnati Children’s Hospital Medical Center, OH
| | - Erin M Miller
- Department of Pediatrics, University of Cincinnati College of Medicine, Heart Institute, Cincinnati Children’s Hospital Medical Center, OH
| | - Joseph Rossano
- Department of Pediatrics, Children’s Hospital of Philadelphia, PA
| | - Kimberly Y Lin
- Department of Pediatrics, Children’s Hospital of Philadelphia, PA
| | - Brian Claggett
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA
| | - Euan Ashley
- Center for Inherited Cardiovascular Disease, Stanford Medicine, CA
| | - Michelle Michels
- Department of Cardiology, Thoraxcenter, Erasmus Medical Center Rotterdam, The Netherlands
| | - Rachel Lampert
- Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT
| | - John C Stendahl
- Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT
| | - Dominic Abrams
- Center for Cardiovascular Genetics, Boston Children’s Hospital, MA
| | - Christopher Semsarian
- Agnes Ginges Centre for Molecular Cardiology at Centenary Institute, Sydney Medical School Faculty of Medicine and Health, University of Sydney, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
| | | | - Matthew Wheeler
- Center for Inherited Cardiovascular Disease, Stanford Medicine, CA
| | - Jodie Ingles
- Genomics and Inherited Disease Program, Garvan Institute of Medical Research and University of New South Wales, Sydney, Australia
| | - Sharlene M Day
- Penn Center for Inherited Cardiovascular Disease, Hospital of the University of Pennsylvania & Perelman School of Medicine at the University of Pennsylvania, Philadelphia (A.O., S.M.D.)
| | - Sara Saberi
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor
| | - Mark W Russell
- Department of Pediatrics, Division of Pediatric Cardiology, University of Michigan, Ann Arbor
| | - Michael Previs
- Genomics and Inherited Disease Program, Garvan Institute of Medical Research and University of New South Wales, Sydney, Australia
| | - Carolyn Ho
- Cardiovascular Division, Brigham and Women’s Hospital, Boston, MA
| | - James S Ware
- National Heart and Lung Institute and MRC Laboratory of Medical Sciences, Imperial College London, United Kingdom
| | - Adam S Helms
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor
| |
Collapse
|
131
|
Ullanat V, Jing B, Sledzieski S, Berger B. Learning the language of protein-protein interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.09.642188. [PMID: 40166198 PMCID: PMC11956943 DOI: 10.1101/2025.03.09.642188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Protein Language Models (PLMs) trained on large databases of protein sequences have proven effective in modeling protein biology across a wide range of applications. However, while PLMs excel at capturing individual protein properties, they face challenges in natively representing protein-protein interactions (PPIs), which are crucial to understanding cellular processes and disease mechanisms. Here, we introduce MINT, a PLM specifically designed to model sets of interacting proteins in a contextual and scalable manner. Using unsupervised training on a large curated PPI dataset derived from the STRING database, MINT outperforms existing PLMs in diverse tasks relating to protein-protein interactions, including binding affinity prediction and estimation of mutational effects. Beyond these core capabilities, it excels at modeling interactions in complex protein assemblies and surpasses specialized models in antibody-antigen modeling and T cell receptor-epitope binding prediction. MINT's predictions of mutational impacts on oncogenic PPIs align with experimental studies, and it provides reliable estimates for the potential for cross-neutralization of antibodies against SARS-CoV-2 variants of concern. These findings position MINT as a powerful tool for elucidating complex protein interactions, with significant implications for biomedical research and therapeutic discovery.
Collapse
Affiliation(s)
- Varun Ullanat
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA
| | - Bowen Jing
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA
| | - Samuel Sledzieski
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA
- Center for Computational Biology, Flatiron Insitute, New York, NY
| | - Bonnie Berger
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA
- Department of Mathematics, Massachusetts Institute of Technology, MA
| |
Collapse
|
132
|
Alay MT. A novel seven-tier framework for the classification of MEFV missense variants using adaptive and rigid classifiers. Sci Rep 2025; 15:9054. [PMID: 40090944 PMCID: PMC11911402 DOI: 10.1038/s41598-025-94142-7] [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: 09/19/2024] [Accepted: 03/12/2025] [Indexed: 03/19/2025] Open
Abstract
There is a great discrepancy between the clinical categorization of MEFV gene variants and in silico tool predictions. In this study, we developed a seven-tier classification system for MEFV missense variants of unknown significance and recommended a generalized pipeline for other gene classifications. We extracted 12,017 human MEFV gene variants from the Ensembl database. After extraction, we detected 6034 missense variants. In the next step, we selected 42 in silico tools for our classification model. We determined the optimal value via the scores from three in silico tools. For the implementation of machine learning methods, we used two bagging methods and two boosting methods. After predicting known variants, we applied our model to 5507 variants of unknown significance. In the final stage, we applied the developed framework to the entire dataset to rigorously evaluate its classification performance and validate its potential clinical utility. The XGBoost model achieved the highest accuracy at 0.9882 (± 0.0295), followed by Extremely Randomized Trees (0.9835 ± 0.0335), Random Forest (0.9788 ± 0.0158), and AdaBoost (0.9671 ± 0.0815). Following the refinement of the dataset and the introduction of a novel classification and clustering methodology, the proportion of known variants increased from 6.9 to 29.4%, marking a 4.3-fold relative improvement. Furthermore, we identified two novel hotspot regions and one tolerant site, offering valuable insights into the functional structure of the pyrin protein. Rigid and adaptive classifiers offer an innovative framework for VOUS classification, integrating a grayscale interpretation system with cutting-edge in silico tools and machine learning algorithms. This approach not only improves the accuracy of MEFV gene variant classification but also identifies new hotspot regions for functional studies, paving the way for scalable applications to other genes and might contribute to advancing precision genomic medicine in the future.
Collapse
Affiliation(s)
- Mustafa Tarık Alay
- Department of Medical Genetics, Ankara Etlik City Hospital, Ankara, Turkey.
| |
Collapse
|
133
|
Zhang T, Zhao W, Wirth C, Díaz-Gay M, Yin J, Cecati M, Marchegiani F, Hoang PH, Leduc C, Baine MK, Travis WD, Sholl LM, Joubert P, Sang J, McElderry JP, Klein A, Khandekar A, Hartman C, Rosenbaum J, Colón-Matos FJ, Miraftab M, Saha M, Lee OW, Jones KM, Caporaso NE, Wong MP, Leung KC, Agnes Hsiung C, Chen CY, Edell ES, Martínez Santamaría J, Schabath MB, Yendamuri SS, Manczuk M, Lissowska J, Świątkowska B, Mukeria A, Shangina O, Zaridze D, Holcatova I, Mates D, Milosavljevic S, Savic M, Bossé Y, Gould Rothberg BE, Christiani DC, Gaborieau V, Brennan P, Liu G, Hofman P, Homer R, Yang SR, Pesatori AC, Consonni D, Yang L, Zhu B, Shi J, Brown K, Rothman N, Chanock SJ, Alexandrov LB, Choi J, Cardelli M, Lan Q, Nowak MA, Wedge DC, Landi MT. Deciphering lung adenocarcinoma evolution and the role of LINE-1 retrotransposition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.643063. [PMID: 40161734 PMCID: PMC11952568 DOI: 10.1101/2025.03.14.643063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Understanding lung cancer evolution can identify tools for intercepting its growth. In a landscape analysis of 1024 lung adenocarcinomas (LUAD) with deep whole-genome sequencing integrated with multiomic data, we identified 542 LUAD that displayed diverse clonal architecture. In this group, we observed an interplay between mobile elements, endogenous and exogenous mutational processes, distinct driver genes, and epidemiological features. Our results revealed divergent evolutionary trajectories based on tobacco smoking exposure, ancestry, and sex. LUAD from smokers showed an abundance of tobacco-related C:G>A:T driver mutations in KRAS plus short subclonal diversification. LUAD in never smokers showed early occurrence of copy number alterations and EGFR mutations associated with SBS5 and SBS40a mutational signatures. Tumors harboring EGFR mutations exhibited long latency, particularly in females of European-ancestry (EU_N). In EU_N, EGFR mutations preceded the occurrence of other driver genes, including TP53 and RBM10. Tumors from Asian never smokers showed a short clonal evolution and presented with heterogeneous repetitive patterns for the inferred mutational order. Importantly, we found that the mutational signature ID2 is a marker of a previously unrecognized mechanism for LUAD evolution. Tumors with ID2 showed short latency and high L1 retrotransposon activity linked to L1 promoter demethylation. These tumors exhibited an aggressive phenotype, characterized by increased genomic instability, elevated hypoxia scores, low burden of neoantigens, propensity to develop metastasis, and poor overall survival. Reactivated L1 retrotransposition-induced mutagenesis can contribute to the origin of the mutational signature ID2, including through the regulation of the transcriptional factor ZNF695, a member of the KZFP family. The complex nature of LUAD evolution creates both challenges and opportunities for screening and treatment plans.
Collapse
Affiliation(s)
- Tongwu Zhang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Wei Zhao
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Christopher Wirth
- Manchester Cancer Research Centre, The University of Manchester, Manchester, UK
| | - Marcos Díaz-Gay
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
- Digital Genomics Group, Structural Biology Program, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Jinhu Yin
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Monia Cecati
- Advanced Technology Center for Aging Research, IRCCS INRCA, Ancona, Italy
| | | | - Phuc H Hoang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Charles Leduc
- Department of Pathology, Centre Hospitalier de l'Université de Montréal, Montreal, Canada
| | - Marina K Baine
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - William D Travis
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lynette M Sholl
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Philippe Joubert
- Institut universitaire de cardiologie et de pneumologie de Québec, Laval University, Quebec City, Canada
| | - Jian Sang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - John P McElderry
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Alyssa Klein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Azhar Khandekar
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Caleb Hartman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | | | - Frank J Colón-Matos
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Mona Miraftab
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Monjoy Saha
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Olivia W Lee
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Kristine M Jones
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
- Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Neil E Caporaso
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
- Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Maria Pik Wong
- Queen Mary Hospital, The University of Hong Kong, Hong Kong, China
| | - Kin Chung Leung
- Department of Pathology, The University of Hong Kong, Hong Kong, China
| | - Chao Agnes Hsiung
- Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Taiwan
| | - Chih-Yi Chen
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Surgery, Division of Thoracic Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Eric S Edell
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Matthew B Schabath
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Sai S Yendamuri
- Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Marta Manczuk
- Department of Cancer Epidemiology and Primary Prevention, Maria Skłodowska-Curie National Research Institute of Oncology, Warshaw, Poland
| | - Jolanta Lissowska
- Department of Cancer Epidemiology and Primary Prevention, Maria Skłodowska-Curie National Research Institute of Oncology, Warshaw, Poland
| | - Beata Świątkowska
- Department of Environmental Epidemiology, Nofer Institute of Occupational Medicine, Łódź, Poland
| | - Anush Mukeria
- Department of Clinical Epidemiology, N.N. Blokhin National Medical Research Centre of Oncology, Moscow, Russia
| | - Oxana Shangina
- Department of Clinical Epidemiology, N.N. Blokhin National Medical Research Centre of Oncology, Moscow, Russia
| | - David Zaridze
- Department of Clinical Epidemiology, N.N. Blokhin National Medical Research Centre of Oncology, Moscow, Russia
| | - Ivana Holcatova
- Institute of Hygiene and Epidemiology, 1st Faculty of Medicine, Charles University, Prague, Czech Republic
- Department of Oncology, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Dana Mates
- Department of Occupational Health and Toxicology, National Center for Environmental Risk Monitoring, National Institute of Public Health, Bucharest, Romania
| | - Sasa Milosavljevic
- International Organisation for Cancer Prevention and Research (IOCPR), Belgrade, Serbia
| | - Milan Savic
- Department of Thoracic Surgery, Clinical Center of Serbia, Belgrade, Serbia
| | - Yohan Bossé
- Institut universitaire de cardiologie et de pneumologie de Québec, Laval University, Quebec City, Canada
| | - Bonnie E Gould Rothberg
- Sylvester Comprehensive Cancer Center, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - David C Christiani
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Valerie Gaborieau
- Genomic Epidemiology Branch, International Agency for Research on Cancer (IARC/WHO), Lyon, France
| | - Paul Brennan
- Genomic Epidemiology Branch, International Agency for Research on Cancer (IARC/WHO), Lyon, France
| | - Geoffrey Liu
- Princess Margaret Cancer Center, University of Toronto, Toronto, Ontario, Canada
| | - Paul Hofman
- IHU RespirERA, Biobank-BB-0033-0025, Côte d'Azur University, Nice, France
| | - Robert Homer
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Soo-Ryum Yang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Angela C Pesatori
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Dario Consonni
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Lixing Yang
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA
- Department of Human Genetics, The University of Chicago, Chicago, IL, USA
- The University of Chicago Medicine Comprehensive Cancer Center, The University of Chicago, Chicago, IL, USA
| | - Bin Zhu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Jianxin Shi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Kevin Brown
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Nathaniel Rothman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Ludmil B Alexandrov
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
- Sanford Stem Cell Institute, University of California San Diego, La Jolla, CA, USA
| | - Jiyeon Choi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Maurizio Cardelli
- Advanced Technology Center for Aging Research, IRCCS INRCA, Ancona, Italy
| | - Qing Lan
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Martin A Nowak
- Department of Mathematics, Harvard University, Cambridge, MA, USA
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - David C Wedge
- Manchester Cancer Research Centre, The University of Manchester, Manchester, UK
- Manchester NIHR Biomedical Research Centre, Manchester, UK
| | - Maria Teresa Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
134
|
Zheng Y, Yu K, Lin JF, Liang Z, Zhang Q, Li J, Wu QN, He CY, Lin M, Zhao Q, Zuo ZX, Ju HQ, Xu RH, Liu ZX. Deep learning prioritizes cancer mutations that alter protein nucleocytoplasmic shuttling to drive tumorigenesis. Nat Commun 2025; 16:2511. [PMID: 40087285 PMCID: PMC11909177 DOI: 10.1038/s41467-025-57858-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 03/05/2025] [Indexed: 03/17/2025] Open
Abstract
Genetic variants can affect protein function by driving aberrant subcellular localization. However, comprehensive analysis of how mutations promote tumor progression by influencing nuclear localization is currently lacking. Here, we systematically characterize potential shuttling-attacking mutations (SAMs) across cancers through developing the deep learning model pSAM for the ab initio decoding of the sequence determinants of nucleocytoplasmic shuttling. Leveraging cancer mutations across 11 cancer types, we find that SAMs enrich functional genetic variations and critical genes in cancer. We experimentally validate a dozen SAMs, among which R14M in PTEN, P255L in CHFR, etc. are identified to disrupt the nuclear localization signals through interfering their interactions with importins. Further studies confirm that the nucleocytoplasmic shuttling altered by SAMs in PTEN and CHFR rewire the downstream signaling and eliminate their function of tumor suppression. Thus, this study will help to understand the molecular traits of nucleocytoplasmic shuttling and their dysfunctions mediated by genetic variants.
Collapse
Grants
- This study was supported by the National Key R&D Program of China [2021YFA1302100], National Natural Science Foundation of China [32370698, 81972239], Program for Guangdong Introducing Innovative and Entrepreneurial Teams [2017ZT07S096], Tip-Top Scientific and Technical Innovative Youth Talents of Guangdong Special Support Program [2019TQ05Y351], Young Talents Program of Sun Yat-sen University Cancer Center [YTP-SYSUCC-0029], Science and Technology Program of Guangzhou [202206080011], Guangdong Basic and Applied Basic Research Foundation [2023B1515040030] and CAMS Innovation Fund for Medical Sciences (CIFMS) [2019-I2M-5-036].
- This study was supported by the Chih Kuang Scholarship for Outstanding Young Physician-Scientists of Sun Yat-sen University Cancer Center [CKS-SYSUCC-2024009] and the Postdoctoral Science Foundation of China [2024M763801, GZB20240907].
- This study was supported by the Noncommunicable Chronic Diseases-National Science and Technology Major Project [2023ZD0501600], National Natural Science Foundation of China [82321003, 82173128] and Cancer Innovative Research Program of Sun Yat-sen University Cancer Center [CIRP-SYSUCC-0004].
Collapse
Affiliation(s)
- Yongqiang Zheng
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Kai Yu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, 77030, USA
| | - Jin-Fei Lin
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Department of Clinical Laboratory, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Zhuoran Liang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Qingfeng Zhang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Junteng Li
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Qi-Nian Wu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Cai-Yun He
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Mei Lin
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Qi Zhao
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Zhi-Xiang Zuo
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Huai-Qiang Ju
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Rui-Hua Xu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, 510060, China.
| | - Ze-Xian Liu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
135
|
May M, Chuah A, Lehmann N, Goodall L, Cho V, Andrews TD. Functionally constrained human proteins are less prone to mutational instability from single amino acid substitutions. Nat Commun 2025; 16:2492. [PMID: 40082446 PMCID: PMC11906876 DOI: 10.1038/s41467-025-57757-y] [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: 12/13/2023] [Accepted: 02/27/2025] [Indexed: 03/16/2025] Open
Abstract
Missense mutations that disrupt protein structural stability are a common pathogenic mechanism in human genetic disease. Here, we quantify potential disruption of protein stability due to amino acid substitution and show that functionally constrained proteins are less susceptible to large mutational changes in stability. Mechanistically, this relates to greater intrinsic disorder among constrained proteins and to increased B-factors in the ordered regions of constrained proteins. This phenomenon means that constrained proteins exhibit smaller stability effects due to missense mutations, and partly explains why overtransmission of pathogenic missense variation is less prevalent in genetic disorders characterised by protein truncations. We show that the most functionally constrained proteins are depleted of both destabilising and overly-stabilising amino acid variation in disease-free populations. Despite this, amino acid substitutions with large stability effects in functionally constrained proteins are still highly prevalent among pathogenic human genetic variation. Importantly, we observe that there are approximately five times more missense variants with large stability effects than there are unambiguous loss-of-function mutations. Missense variants with disruption of stability effects recapitulate the per-gene patterns of functional constraint observed with protein truncating loss-of-function variation, yet their relative abundance abrogates difficulties encountered when estimating functional constraint for the shortest human genes.
Collapse
Affiliation(s)
- Maryam May
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Aaron Chuah
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
- National Cancer Centre Singapore, Singapore, Singapore
| | - Nicole Lehmann
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Llewelyn Goodall
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
- School of Computing, College of Engineering, Computer Science and Cybernetics, The Australian National University, Canberra, Australia
| | - Vicky Cho
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - T Daniel Andrews
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australia.
- School of Computing, College of Engineering, Computer Science and Cybernetics, The Australian National University, Canberra, Australia.
| |
Collapse
|
136
|
Phillips CL, So C, Gillis MF, Harrison J, Hsu CH, Armao D, Snider NT. The Kelch 3 motif on gigaxonin mediates the interaction with NUDCD3 and regulates vimentin filament morphology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.10.641328. [PMID: 40161598 PMCID: PMC11952450 DOI: 10.1101/2025.03.10.641328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Gigaxonin is an intermediate filament (IF)-interacting partner belonging to the Kelch-like (KLHL) protein family. Gigaxonin is encoded by the KLHL16 gene, which is mutated in Giant Axonal Neuropathy (GAN). The lack of functional gigaxonin in GAN patient cells impairs IF proteostasis, leading to focal abnormal accumulations of IFs and compromised neuronal function. We hypothesized that gigaxonin forms molecular interactions via specific sequence motifs to regulate IF proteostasis. The goal of this study was to examine how distinct Kelch motifs on gigaxonin regulate IF protein degradation and filament morphology. We analyzed vimentin IFs in HEK293 cells overexpressing wild type (WT) gigaxonin, or gigaxonin lacking each of the six individual Kelch motifs: K1 (aa274-326), K2 (aa327-374), K3 (aa376-421), K4 (aa422-468), K5 (aa470-522), and K6 (aa528-574). All six gigaxonin deletion mutants (ΔK1-ΔK6) promoted the degradation of soluble vimentin. The ΔK3 gigaxonin mutant exhibited soluble vimentin degradation and promoted the bundling of vimentin IFs relative to WT gigaxonin. Using mass spectrometry proteomic analysis we found that, relative to WT gigaxonin, ΔK3 gigaxonin had increased associations with ubiquitination-associated and mitochondrial proteins and lost the association with the NudC domain-containing protein 3 (NUDCD3), a molecular chaperone enriched in the nervous system. Collectively, our cell biological data show the induction of an abnormal GAN-like IF phenotype in cells expressing ΔK3-gigaxonin, while our mass spectrometry profiling links the loss of gigaxonin-NUDCD3 interactions with defective IF proteostasis, revealing NUDCD3 as a potential new target in GAN.
Collapse
Affiliation(s)
- Cassandra L. Phillips
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill
| | - Christina So
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill
| | - Meredith F. Gillis
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill
| | - Jonathan Harrison
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill
| | - Chih-Hsuan Hsu
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill
| | - Diane Armao
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill
- Department of Radiology, University of North Carolina at Chapel Hill
| | - Natasha T. Snider
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill
| |
Collapse
|
137
|
Collin-Chavagnac D, Saint-Martin C, Bedidi L, Lebreton L, Aslanzadeh V, Vigouroux C, Bellanné-Chantelot C, Semple RK, Lascols O, Jéru I. Insulin receptor variants: Extending the traditional Mendelian spectrum. Genet Med 2025; 27:101404. [PMID: 40094207 DOI: 10.1016/j.gim.2025.101404] [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: 10/07/2024] [Revised: 02/28/2025] [Accepted: 03/06/2025] [Indexed: 03/19/2025] Open
Abstract
PURPOSE INSR encodes the insulin receptor, the essential entrainer of growth and metabolism to nutritional cues. INSR variants cause a spectrum of monogenic insulin resistance (IR) syndromes, namely, type A insulin resistance, Rabson-Mendenhall, and Donohue syndromes. However, to our knowledge, no large cohort studies focused on variant classification and its diagnostic value have been described. METHODS This multicentric cohort study included 73 patients carrying INSR variants, referred for IR by 52 centers from 6 countries. Variants were classified using new bioinformatic tools relying on different prediction mechanisms and the American College of Medical Genetics and Genomics guidelines. RESULTS Besides expanding the INSR mutational spectrum, this study suggested a semidominant inheritance in several Donohue/Rabson-Mendenhall syndrome families. Questioning strictly Mendelian inheritance, heterozygous loss-of-function (LoF) variants were mostly found in overweight patients, with a higher LoF frequency in IR patients than in the general population (odds ratio 5.77). Diagnostic challenges arose when trying to refine classification criteria for variants of uncertain significance. Among the variant effect predictors assessed, MISTIC and AlphaMissense outperformed REVEL. CONCLUSION The spectrum of INSR-related disorders extends beyond traditional entities. Heterozygous INSR LoF variants may increase IR susceptibility. International collaboration and functional assays are needed to drive precision medicine forward.
Collapse
Affiliation(s)
- Delphine Collin-Chavagnac
- Department of Biochemistry and Molecular Biology, Reference Medical Biology Laboratory for Insulin Resistance and Metabolic Syndrome, Hospices civils de Lyon, Lyon, France; CarMeN Laboratory, Université Claude Bernard Lyon 1, Inserm, INRAE, Pierre Bénite, France
| | - Cécile Saint-Martin
- Department of Medical Genetics, Reference Medical Biology Laboratory for Insulin Resistance and Lipodystrophy, Pitié-Salpêtrière Hospital, Assistance-Publique Hôpitaux de Paris (AP-HP), Paris, France
| | - Lotfi Bedidi
- Department of Medical Genetics, Reference Medical Biology Laboratory for Insulin Resistance and Lipodystrophy, Pitié-Salpêtrière Hospital, Assistance-Publique Hôpitaux de Paris (AP-HP), Paris, France
| | - Louis Lebreton
- Laboratory of Biochemistry, University Hospital Centre Bordeaux, Bordeaux, France
| | - Vahid Aslanzadeh
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom; MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Corinne Vigouroux
- Department of Molecular Biology and Genetics, Saint-Antoine Hospital, AP-HP, Paris, France; Inserm UMR S938, Saint-Antoine Research Centre, Institute of Cardiometabolism and Nutrition, Sorbonne University, Paris, France; Department of Endocrinology, National Reference Center for Rare Diseases of Insulin Secretion and Insulin Sensitivity, Endo-ERN Center for Rare Endocrine Diseases, Saint-Antoine Hospital, AP-HP, Paris, France
| | - Christine Bellanné-Chantelot
- Department of Medical Genetics, Reference Medical Biology Laboratory for Insulin Resistance and Lipodystrophy, Pitié-Salpêtrière Hospital, Assistance-Publique Hôpitaux de Paris (AP-HP), Paris, France
| | - Robert K Semple
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom; MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Olivier Lascols
- Department of Molecular Biology and Genetics, Saint-Antoine Hospital, AP-HP, Paris, France; Inserm UMR S938, Saint-Antoine Research Centre, Institute of Cardiometabolism and Nutrition, Sorbonne University, Paris, France
| | - Isabelle Jéru
- Department of Medical Genetics, Reference Medical Biology Laboratory for Insulin Resistance and Lipodystrophy, Pitié-Salpêtrière Hospital, Assistance-Publique Hôpitaux de Paris (AP-HP), Paris, France; Department of Molecular Biology and Genetics, Saint-Antoine Hospital, AP-HP, Paris, France; Inserm UMR S938, Saint-Antoine Research Centre, Institute of Cardiometabolism and Nutrition, Sorbonne University, Paris, France.
| |
Collapse
|
138
|
Couzens A, Neerman-Arbez M. Congenital Fibrinogen Deficiencies: Not So Rare. Hamostaseologie 2025. [PMID: 40074015 DOI: 10.1055/a-2511-3314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2025] Open
Abstract
Congenital fibrinogen deficiencies (CFDs), traditionally considered rare monogenic disorders, are now recognized as more prevalent and genetically complex than previously thought. Indeed, the symptoms manifested in CFD patients, such as bleeding and thrombosis, are likely to result from variation in several genes rather than solely driven by variants in one of the three fibrinogen genes, FGB, FGA, and FGG. This review highlights recent advances in understanding the genetic causes of CFD and their variability, facilitated by the growing use and availability of next-generation sequencing data. Using gnomAD v4.1.0. data, which includes more than 800,000 individuals, we provide updated global prevalence estimates for CFDs based on frequencies of predicted deleterious variants in FGB, FGA, and FGG. Recessively inherited fibrinogen deficiencies (homozygous genotypes) could be present in around 29 individuals per million, while dominantly inherited deficiencies (heterozygous genotypes) may be present in up to 15,000 per million. These increased estimates can be attributed to the inclusion of broader, more diverse genetic datasets in the new version of gnomAD, thus capturing a greater range of rare variants and homozygous cases.
Collapse
Affiliation(s)
- Alexander Couzens
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva and Institute of Genetics and Genomics in Geneva (iGE3), Geneva, Switzerland
| | - Marguerite Neerman-Arbez
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva and Institute of Genetics and Genomics in Geneva (iGE3), Geneva, Switzerland
| |
Collapse
|
139
|
Laquatra C, Magro A, Guarra F, Lambrughi M, Ferrone L, Fracasso G, Bacchin M, La Spina M, Moroni E, Papaleo E, Colombo G, Rasola A. Point mutations of the mitochondrial chaperone TRAP1 affect its functions and pro-neoplastic activity. Cell Death Dis 2025; 16:172. [PMID: 40074754 PMCID: PMC11903959 DOI: 10.1038/s41419-025-07467-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 01/28/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025]
Abstract
The mitochondrial chaperone TRAP1 is a key regulator of cellular homeostasis and its activity has important implications in neurodegeneration, ischemia and cancer. Recent evidence has indicated that TRAP1 mutations are involved in several disorders, even though the structural basis for the impact of point mutations on TRAP1 functions has never been studied. By exploiting a modular structure-based framework and molecular dynamics simulations, we investigated the effect of five TRAP1 mutations on its structure and stability. Each mutation differentially impacts long-range interactions, intra and inter-protomer dynamics and ATPase activity. Changes in these parameters influence TRAP1 functions, as revealed by their effects on the activity of the TRAP1 interactor succinate dehydrogenase (SDH). In keeping with this, TRAP1 point mutations affect the growth and migration of aggressive sarcoma cells, and alter sensitivity to a selective TRAP1 inhibitor. Our work provides new insights on the structure-activity relationship of TRAP1, identifying crucial amino acid residues that regulate TRAP1 proteostatic functions and pro-neoplastic activity.
Collapse
Affiliation(s)
- Claudio Laquatra
- Department of Biomedical Sciences, University of Padova, Padova, Italy.
| | - Alessia Magro
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | | | - Matteo Lambrughi
- Cancer Structural Biology, Danish Cancer Institute, Copenhagen, Denmark
| | - Lavinia Ferrone
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Giulio Fracasso
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Melissa Bacchin
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Martina La Spina
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | | | - Elena Papaleo
- Cancer Structural Biology, Danish Cancer Institute, Copenhagen, Denmark
- Cancer System Biology, Section for Bioinformatics, Department of Health and Technology, Technical University of Denmark, Lyngby, Denmark
| | | | - Andrea Rasola
- Department of Biomedical Sciences, University of Padova, Padova, Italy.
| |
Collapse
|
140
|
Liu LH, Lei W, Zhang Z, Lai S, Xu B, Ge Q, Luo J, Yang M, Zhang Y, Chen J, Zhong Q, Wu YR, Jiang A. OMEGA-guided DNA polymerases enable random mutagenesis in a tunable window. Trends Biotechnol 2025:S0167-7799(25)00048-4. [PMID: 40074636 DOI: 10.1016/j.tibtech.2025.02.011] [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: 10/19/2024] [Revised: 02/07/2025] [Accepted: 02/11/2025] [Indexed: 03/14/2025]
Abstract
Targeted random mutagenesis is crucial for breeding, directed evolution, and gene function studies, yet efficient tools remain scarce. Here, we present obligate mobile element guided activity (OMEGA)-R, an innovative targeted random mutagenesis system that integrates SpyCatcher-enIscB and PolI3M-TBD-SpyTag, outperforming existing state-of-the-art technologies in key metrics, such as protein size, mutagenesis efficiency, window length, and continuity. OMEGA-R achieves a dramatic enhancement of on-target mutagenesis, reaching a rate of 1.4 × 10-5 base pairs (bp) per generation (bpg), with minimal off-target effects, in both Escherichia coli and Bacillus subtilis. The system also demonstrates exceptional compatibility with high-throughput screening (HTS) technologies, including fluorescence-activated droplet sorting (FADS) and phage-assisted continuous evolution (PACE). Utilizing OMEGA-R, we successfully identified a series of effective mutations within the T7 promoter (pT7), ribosome-binding site (RBS), superfolder GFP (sfGFP), and autocyclizing ribozyme (AR), which are invaluable for the development of high-performance biotechnology tools. These findings underscore the high efficiency and broad application potential of OMEGA-R.
Collapse
Affiliation(s)
- Li-Hua Liu
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd, Guangzhou Qianxiang Bioworks Co., Ltd, Guangzhou, Guangdong 510000, PR China; Biology Department and Institute of Marine Sciences, College of Science, Shantou University, Shantou 515063, PR China
| | - Wei Lei
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd, Guangzhou Qianxiang Bioworks Co., Ltd, Guangzhou, Guangdong 510000, PR China
| | - Zhiqian Zhang
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd, Guangzhou Qianxiang Bioworks Co., Ltd, Guangzhou, Guangdong 510000, PR China
| | - Shijing Lai
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd, Guangzhou Qianxiang Bioworks Co., Ltd, Guangzhou, Guangdong 510000, PR China
| | - Bo Xu
- School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning 437100, PR China
| | - Qijun Ge
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd, Guangzhou Qianxiang Bioworks Co., Ltd, Guangzhou, Guangdong 510000, PR China
| | - Jiawen Luo
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd, Guangzhou Qianxiang Bioworks Co., Ltd, Guangzhou, Guangdong 510000, PR China
| | - Min Yang
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd, Guangzhou Qianxiang Bioworks Co., Ltd, Guangzhou, Guangdong 510000, PR China
| | - Yang Zhang
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd, Guangzhou Qianxiang Bioworks Co., Ltd, Guangzhou, Guangdong 510000, PR China
| | - Jinde Chen
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd, Guangzhou Qianxiang Bioworks Co., Ltd, Guangzhou, Guangdong 510000, PR China
| | - Qiuru Zhong
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd, Guangzhou Qianxiang Bioworks Co., Ltd, Guangzhou, Guangdong 510000, PR China
| | - Yi-Rui Wu
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd, Guangzhou Qianxiang Bioworks Co., Ltd, Guangzhou, Guangdong 510000, PR China
| | - Ao Jiang
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd, Guangzhou Qianxiang Bioworks Co., Ltd, Guangzhou, Guangdong 510000, PR China.
| |
Collapse
|
141
|
Bergquist T, Stenton SL, Nadeau EAW, Byrne AB, Greenblatt MS, Harrison SM, Tavtigian SV, O'Donnell-Luria A, Biesecker LG, Radivojac P, Brenner SE, Pejaver V. Calibration of additional computational tools expands ClinGen recommendation options for variant classification with PP3/BP4 criteria. Genet Med 2025; 27:101402. [PMID: 40084623 DOI: 10.1016/j.gim.2025.101402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 02/26/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025] Open
Abstract
PURPOSE We previously developed an approach to calibrate computational tools for clinical variant classification, updating recommendations for the reliable use of variant impact predictors to provide evidence strength up to Strong. A new generation of tools using distinctive approaches has since been released, and these methods must be independently calibrated for clinical application. METHODS Using our local posterior probability-based calibration and our established data set of ClinVar pathogenic and benign variants, we determined the strength of evidence provided by 3 new tools (AlphaMissense, ESM1b, and VARITY) and calibrated scores meeting each evidence strength. RESULTS All 3 tools reached the Strong level of evidence for variant pathogenicity and Moderate for benignity, although sometimes for few variants. Compared with previously recommended tools, these yielded at best only modest improvements in the trade-offs between evidence strength and false-positive predictions. CONCLUSION At calibrated thresholds, 3 new computational predictors provided evidence for variant pathogenicity at similar strength to the 4 previously recommended predictors (and comparable with functional assays for some variants). This calibration broadens the scope of computational tools for application in clinical variant classification. Their new approaches offer promise for future advancement of the field.
Collapse
Affiliation(s)
- Timothy Bergquist
- Institute for Genomic Health, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Sarah L Stenton
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA; Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Emily A W Nadeau
- Department of Medicine and University of Vermont Cancer Center, University of Vermont, Larner College of Medicine, Burlington, VT
| | - Alicia B Byrne
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Marc S Greenblatt
- Department of Medicine and University of Vermont Cancer Center, University of Vermont, Larner College of Medicine, Burlington, VT
| | - Steven M Harrison
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA; Ambry Genetics, Aliso Viejo, CA
| | - Sean V Tavtigian
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT
| | - Anne O'Donnell-Luria
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA; Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Leslie G Biesecker
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Predrag Radivojac
- Khoury College of Computer Sciences, Northeastern University, Boston, MA
| | - Steven E Brenner
- Department of Plant and Microbial Biology and Center for Computational Biology, University of California, Berkeley, CA
| | - Vikas Pejaver
- Institute for Genomic Health, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
142
|
Nagarajan A, Amberg-Johnson K, Paull E, Huang K, Ghanakota P, Chandrasinghe A, Chief Elk J, Sampson JM, Wang L, Abel R, Albanese SK. Predicting Resistance to Small Molecule Kinase Inhibitors. J Chem Inf Model 2025; 65:2543-2557. [PMID: 39979081 DOI: 10.1021/acs.jcim.4c02313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Drug resistance is a critical challenge in treating diseases like cancer and infectious disease. This study presents a novel computational workflow for predicting on-target resistance mutations to small molecule inhibitors (SMIs). The approach integrates genetic models with alchemical free energy perturbation (FEP+) calculations to identify likely resistance mutations. Specifically, a genetic model, RECODE, leverages cancer-specific mutation patterns to prioritize probable amino acid changes. Physics-based calculations assess the impact of these mutations on protein stability, endogenous substrate binding, and inhibitor binding. We apply this approach retrospectively to gefitinib and osimertinib, two clinical epidermal growth factor receptor (EGFR) inhibitors used to treat nonsmall cell lung cancer (NSCLC). Among hundreds of possible mutations, the pipeline accurately predicted 4 out of 11 and 7 out of 19 known binding site mutations for gefitinib and osimertinib, respectively, including the clinically relevant T790M and C797S resistance mutations. This study demonstrates the potential of integrating genetic models and physics-based calculations to predict SMI resistance mutations. This approach can be applied to other kinases and target classes, potentially enabling the design of next-generation inhibitors with improved durability of response in patients.
Collapse
Affiliation(s)
- Anu Nagarajan
- Schrödinger, New York, New York 10036, United States
| | | | - Evan Paull
- Schrödinger, New York, New York 10036, United States
| | - Kunling Huang
- Schrödinger, New York, New York 10036, United States
| | | | | | | | | | - Lingle Wang
- Schrödinger, New York, New York 10036, United States
| | - Robert Abel
- Schrödinger, New York, New York 10036, United States
| | | |
Collapse
|
143
|
Marohl T, Atkins KA, Wang L, Janes KA. PCSK5 M452I is a recessive hypomorph exclusive to MCF10DCIS.com cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.03.641323. [PMID: 40093128 PMCID: PMC11908202 DOI: 10.1101/2025.03.03.641323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
The most widely used cell line for studying ductal carcinoma in situ (DCIS) premalignancy is the transformed breast epithelial cell line, MCF10DCIS.com. During its original clonal isolation and selection, MCF10DCIS.com acquired a heterozygous M452I mutation in the proprotein convertase PCSK5, which has never been reported in any human cancer. The mutation is noteworthy because PCSK5 matures GDF11, a TGFβ-superfamily ligand that suppresses progression of triple-negative breast cancer. We asked here whether PCSK5M452I and its activity toward GDF11 might contribute to the unique properties of MCF10DCIS.com. Using an optimized in-cell GDF11 maturation assay, we found that overexpressed PCSK5M452I was measurably active but at a fraction of the wildtype enzyme. In a PCSK5 -/- clone of MCF10DCIS.com reconstituted with different PCSK5 alleles, PCSK5M452I was mildly defective in anterograde transport. However, the multicellular organization of PCSK5M452I addback cells in 3D matrigel cultures was significantly less compact than wildtype and indistinguishable from a PCSK5T288P null allele. Growth of intraductal MCF10DCIS.com xenografts was similarly impaired along with the frequency of comedo necrosis and stromal activation. In no setting did PCSK5M452I exhibit gain-of-function activity, leading us to conclude that it is hypomorphic and thus compensated by the remaining wildtype allele in MCF10DCIS.com. Implications This work reassures that an exotic PCSK5 mutation is not responsible for the salient characteristics of the MCF10DCIS.com cell line.
Collapse
Affiliation(s)
- Taylor Marohl
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908
| | - Kristen A. Atkins
- Department of Pathology, University of Virginia, Charlottesville, VA 22908
| | - Lixin Wang
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908
| | - Kevin A. Janes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908
- Department of Biochemistry & Molecular Genetics, University of Virginia, Charlottesville, VA 22908
| |
Collapse
|
144
|
Hemker SL, Marsh A, Hernandez F, Glick E, Clark G, Bashir A, Jiang K, Kitzman JO. Saturation mapping of MUTYH variant effects using DNA repair reporters. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.01.640912. [PMID: 40093110 PMCID: PMC11908140 DOI: 10.1101/2025.03.01.640912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Variants of uncertain significance (VUS) limit the actionability of genetic testing. A prominent example is MUTYH, a base excision repair factor associated with polyposis and colorectal cancer, which has a pathogenic variant carrier rate approaching 1 in 50 individuals in some populations. To systematically interrogate variant function in MUTYH, we coupled deep mutational scanning with a DNA repair reporter containing its lesion substrate, 8OG:A. Our variant-to-function map covers >97% of all possible MUTYH point variants (n=10,941) and achieves 100% accuracy classifying the pathogenicity of known clinical variants (n=247). Leveraging a large clinical registry, we observe significant associations with colorectal polyps and cancer, with more severely impaired missense variants conferring greater risk. We recapitulate known functional differences between pathogenic founder alleles, and highlight sites of complete missense intolerance, including residues that intercalate DNA and coordinate essential Zn2+ or Fe-S clusters. This map provides a resource to resolve the 1,032 existing missense VUS and 90 variants with conflicting interpretations in MUTYH, and demonstrates a scalable strategy to interrogate other clinically relevant DNA repair factors.
Collapse
Affiliation(s)
- Shelby L. Hemker
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | | | | - Elena Glick
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Grace Clark
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Alyssa Bashir
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Krystal Jiang
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jacob O. Kitzman
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Gilbert S. Omenn Department of Computational Medicine & Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
145
|
Sándor M, Scheers I, Masamune A, Witt H, LaRusch J, Chen JM, Németh BC, Geisz A, Uc A, Sahin-Tóth M. AlphaMissense versus laboratory-based pathogenicity prediction of 13 novel missense CPA1 variants from pancreatitis cases. Gut 2025; 74:678-679. [PMID: 39256032 PMCID: PMC11885027 DOI: 10.1136/gutjnl-2024-333697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 08/24/2024] [Indexed: 09/12/2024]
Affiliation(s)
- Máté Sándor
- Department of Surgery, University of California Los Angeles, Los Angeles, California, USA
| | - Isabelle Scheers
- Department of Pediatric Gastroenterology and Hepatology, Cliniques universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Atsushi Masamune
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Heiko Witt
- Paediatric Nutritional Medicine, Else Kröner-Fresenius Zentrum für Ernährungsmedizin (EKFZ), Technische Universität München (TUM), Freising, Germany
| | | | - Jian-Min Chen
- Univ Brest, Inserm, EFS, UMR 1078, GGB, F-29200, Brest, France
| | - Balázs Csaba Németh
- Center for Gastroenterology, Department of Medicine, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Hungarian Centre of Excellence for Molecular Medicine, Translational Pancreatology Research Group, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Andrea Geisz
- Department of Surgery, Boston University, Boston, Massachusetts, USA
| | - Aliye Uc
- Stead Family Department of Pediatrics and Department of Radiation Oncology, University of Iowa, Iowa City, Iowa, USA
| | - Miklós Sahin-Tóth
- Department of Surgery, University of California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
146
|
Banerjee A, Bogetti A, Bahar I. Accurate Identification and Mechanistic Evaluation of Pathogenic Missense Variants with Rhapsody-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.17.638727. [PMID: 40027614 PMCID: PMC11870481 DOI: 10.1101/2025.02.17.638727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Understanding the effects of missense mutations or single amino acid variants (SAVs) on protein function is crucial for elucidating the molecular basis of diseases/disorders and designing rational therapies. We introduce here Rhapsody-2, a machine learning tool for discriminating pathogenic and neutral SAVs, significantly expanding on a precursor limited by the availability of structural data. With the advent of AlphaFold2 as a powerful tool for structure prediction, Rhapsody-2 is trained on a significantly expanded dataset of 117,525 SAVs corresponding to 12,094 human proteins reported in the ClinVar database. Adopting a broad set of descriptors composed of sequence evolutionary, structural, dynamic, and energetics features in the training algorithm, Rhapsody-2 achieved an AUROC of 0.94 in 10-fold cross-validation when all SAVs of a particular test protein (mutant) were excluded from the training set. Benchmarking against a variety of testing datasets demonstrated the high performance of Rhapsody-2. While sequence evolutionary descriptors play a dominant role in pathogenicity prediction, those based on structural dynamics provide a mechanistic interpretation. Notably, residues involved in allosteric communication, and those distinguished by pronounced fluctuations in the high frequency modes of motion or subject to spatial constraints in soft modes usually give rise to pathogenicity when mutated. Overall, Rhapsody-2 provides an efficient and transparent tool for accurately predicting the pathogenicity of SAVs and unraveling the mechanistic basis of the observed behavior, thus advancing our understanding of genotype-to-phenotype relations.
Collapse
Affiliation(s)
- Anupam Banerjee
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, New York 11794, USA
| | - Anthony Bogetti
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, New York 11794, USA
| | - Ivet Bahar
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, New York 11794, USA
- Department of Biochemistry and Cell Biology, Renaissance School of Medicine, Stony Brook University, New York 11794, USA
| |
Collapse
|
147
|
LaFlam TN, Billesbølle CB, Dinh T, Wolfreys FD, Lu E, Matteson T, An J, Xu Y, Singhal A, Brandes N, Ntranos V, Manglik A, Cyster JG, Ye CJ. Phenotypic pleiotropy of missense variants in human B cell-confinement receptor P2RY8. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.28.640567. [PMID: 40093123 PMCID: PMC11908195 DOI: 10.1101/2025.02.28.640567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Missense variants can have pleiotropic effects on protein function and predicting these effects can be difficult. We performed near-saturation deep mutational scanning of P2RY8, a G-protein-coupled receptor that promotes germinal center B cell confinement. We assayed the effect of each variant on surface expression, migration, and proliferation. We delineated variants that affected both expression and function, affected function independently of expression, and discrepantly affected migration and proliferation. We also used cryo-electron microscopy to determine the structure of activated, ligand-bound P2RY8, providing structural insights into the effects of variants on ligand binding and signal transmission. We applied the deep mutational scanning results to both improve computational variant effect predictions and to characterize the phenotype of germline variants and lymphoma-associated variants. Together, our results demonstrate the power of integrating deep mutational scanning, structure determination, and in silico prediction to advance the understanding of a receptor important in human health.
Collapse
Affiliation(s)
- Taylor N. LaFlam
- Division of Pediatric Rheumatology, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
| | - Christian B. Billesbølle
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Tuan Dinh
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Finn D. Wolfreys
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Erick Lu
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Tomas Matteson
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Jinping An
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Ying Xu
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Arushi Singhal
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
| | - Nadav Brandes
- Department of Biochemistry and Molecular Pharmacology, New York University, New York, NY, USA
| | - Vasilis Ntranos
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
- Diabetes Center, University of California, San Francisco, CA, USA
| | - Aashish Manglik
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Quantitative Biosciences Institute, San Francisco, CA, USA
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA
| | - Jason G. Cyster
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Chun Jimmie Ye
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA, USA
- Arc Institute, Palo Alto, CA, USA
| |
Collapse
|
148
|
Dwivedi A, Scalsky RJ, Harris DG, Stabler TC, Shrestha B, Joshi S, Gandhi C, Munro JB, Ifeonu OO, Ouedraogo A, Tiono AB, Coulibaly D, Ouattara A, Richie TL, Sim BKL, Plowe CV, Lyke KE, Takala-Harrison S, Hoffman SL, Thera MA, Sirima SB, Laurens MB, Silva JC. Protective targets of PfSPZ vaccines identified from whole-genome sieve analysis of isolates from malaria vaccine efficacy trials in West Africa. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.04.25323352. [PMID: 40093207 PMCID: PMC11908318 DOI: 10.1101/2025.03.04.25323352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Identification of antigens targeted by a protective response is a central quest in malaria vaccinology. Whole-genome sieve analysis (SAWG) in samples collected from placebo-controlled field trials of Plasmodium falciparum (Pf) sporozoite (SPZ) vaccines may enable identification of Pf pre-erythrocytic antigens. We applied SAWG to genomic data generated from Pf isolates collected during two field trials measuring the efficacy, in malaria-exposed African adults, of two PfSPZ vaccines. These randomized, double-blind, placebo-controlled trials were conducted in regions of Mali and Burkina Faso characterized by high seasonal transmission, where parasite genetic diversity is high. Genomic sites in which the vaccine allelic state was significantly underrepresented among breakthrough infections in vaccinees relative to placebo recipients were termed "target sites". Protein-coding loci containing target sites that changed amino acids were termed "target loci". The SAWG conducted on clinical trial samples from the Burkina Faso and Mali trials identified 138 and 80 single-copy protein-coding target loci in the Burkinabe and Malian data sets, respectively, with twelve common to both, a number significantly higher than expected (E = 3.9; 99%CI = [0, 9]). Among these was the thrombospondin-related anonymous protein locus, which encodes PfSSP2|TRAP, one of the most abundant and well-characterized pre-erythrocytic stage antigen as well as other genes encoding membrane-associated proteins of unknown function. These results identify SAWG as a potentially powerful tool for identifying protective vaccine antigens in recombining pathogens with large genome size and reveals potential new protective Pf antigens.
Collapse
Affiliation(s)
- Ankit Dwivedi
- Institute for Genome Sciences, University of Maryland School of Medicine; Baltimore, MD 21201, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | - Ryan J. Scalsky
- Institute for Genome Sciences, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | - David G. Harris
- Department of Computer Science, University of Maryland College Park; College Park, MD 20742, USA
| | | | - Biraj Shrestha
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | - Sudhaunshu Joshi
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | - Chakshu Gandhi
- Institute for Genome Sciences, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | - James B. Munro
- Institute for Genome Sciences, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | - Olukemi O. Ifeonu
- Institute for Genome Sciences, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | | | - Alfred B. Tiono
- Groupe de Recherche Action en Santé; Ouagadougou, Burkina Faso
| | - Drissa Coulibaly
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako; Bamako, Mali
| | - Amed Ouattara
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | | | | | - Christopher V. Plowe
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | - Kirsten E. Lyke
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | - Shannon Takala-Harrison
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | | | - Mahamadou A. Thera
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako; Bamako, Mali
| | | | - Matthew B. Laurens
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | - Joana C. Silva
- Institute for Genome Sciences, University of Maryland School of Medicine; Baltimore, MD 21201, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine; Baltimore, MD 21201, USA
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical (IHMT), Universidade NOVA de Lisboa (NOVA); 1349-008 Lisboa, Portugal
| |
Collapse
|
149
|
Yu H, He G, Wang W, Qin S, Wang Y, Bai M, Shu K, Pu D. A graph neural network approach for accurate prediction of pathogenicity in multi-type variants. Brief Bioinform 2025; 26:bbaf151. [PMID: 40251830 PMCID: PMC12008122 DOI: 10.1093/bib/bbaf151] [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: 01/22/2025] [Revised: 03/05/2025] [Accepted: 03/19/2025] [Indexed: 04/21/2025] Open
Abstract
Accurate prediction of pathogenic variants in human disease-associated genes would have a profound effect on clinical decision-making; however, it remains a significant challenge due to the overwhelming number of these variants. We propose graph neural network for multimodal annotation-based pathogenicity prediction (GNN-MAP), a novel deep learning framework that effectively integrates multimodal annotations and similarity relationships among variants to predict the pathogenicity of multi-type variants. Trained on the ClinVar dataset, GNN-MAP exhibits superior predictive performance in internal validation and orthogonal test datasets, accurately predicting variant pathogenicity. Notably, GNN-MAP enables accurate prediction of the pathogenicity of rare variants and highly imbalanced datasets. Furthermore, it achieves high performance in the pathogenicity prediction of inherited retinal disease-specific variants, highlighting its effectiveness in disease-specific variant prediction. These findings suggest that the robust capability of GNN-MAP to predict pathogenicity across multiple variant types and datasets holds significant potential for applications in research and clinical settings.
Collapse
Affiliation(s)
- Hongtao Yu
- College of Computer Science and Technology, Chongqing University of Posts and Telecommunications, No. 2 Chongwen Road, Nan'an District, Chongqing 400065, China
- Chongqing Key Laboratory of Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, No. 2 Chongwen Road, Nan'an District, Chongqing 400065, China
| | - Guojing He
- College of Computer Science and Technology, Chongqing University of Posts and Telecommunications, No. 2 Chongwen Road, Nan'an District, Chongqing 400065, China
- Chongqing Key Laboratory of Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, No. 2 Chongwen Road, Nan'an District, Chongqing 400065, China
| | - Wei Wang
- College of Computer Science and Technology, Chongqing University of Posts and Telecommunications, No. 2 Chongwen Road, Nan'an District, Chongqing 400065, China
- Chongqing Key Laboratory of Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, No. 2 Chongwen Road, Nan'an District, Chongqing 400065, China
| | - Senbiao Qin
- Chongqing Key Laboratory of Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, No. 2 Chongwen Road, Nan'an District, Chongqing 400065, China
| | - Yu Wang
- Chongqing Key Laboratory of Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, No. 2 Chongwen Road, Nan'an District, Chongqing 400065, China
| | - Mingze Bai
- Chongqing Key Laboratory of Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, No. 2 Chongwen Road, Nan'an District, Chongqing 400065, China
| | - Kunxian Shu
- Chongqing Key Laboratory of Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, No. 2 Chongwen Road, Nan'an District, Chongqing 400065, China
| | - Dan Pu
- Chongqing Key Laboratory of Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, No. 2 Chongwen Road, Nan'an District, Chongqing 400065, China
| |
Collapse
|
150
|
Krawczyk M, Fernandez-Fuentes N, Fidyt K, Winiarski T, Pepek M, Graczyk-Jarzynka A, Davis J, Bousquets-Muñoz P, Puente XS, Menendez P, Benard E, Wälchli S, Thomas-Tikhonenko A, Winiarska M. The costimulatory domain influences CD19 CAR-T cell resistance development in B-cell malignancies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.28.640707. [PMID: 40093096 PMCID: PMC11908201 DOI: 10.1101/2025.02.28.640707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
CD19-CAR-T-cells emerge as a major therapeutic option for relapsed/refractory B-cell-derived malignancies, however approximately half of patients eventually relapse. To identify resistance-driving factors, we repeatedly exposed B-cell lymphoma/B-cell acute lymphoblastic leukemia to 4-1BB/CD28-based CD19-CAR-T-cells in vitro. Generated models revealed costimulatory domain-dependent differences in CD19 loss. While CD19-4-1BB-CAR-T-cells induced combination epitope/total CD19 protein loss, CD19-CD28-CAR-T-cells did not drive antigen-escape. Consistent with observations in patients relapsing after CD19-4-1BB-CAR-T-cells, we identified CD19 frameshift/missense mutations affecting residues critical for FMC63 epitope recognition. Mathematical simulations revealed that differences between CD19-4-1BB- and CD19-CD28-CAR-T-cells activity against low-antigen-expressing tumor contribute to heterogeneous therapeutic responses. By integrating in vitro and in silico data, we propose a biological scenario where CD19-4-1BB-CAR-T-cells fail to eliminate low-antigen tumor cells, fostering CAR-resistance. These findings offer mechanistic insight into the observed clinical differences between axi-cel (CD28-based) and tisa-cel (4-1BB-based)-treated B-cell lymphoma patients and advance our understanding on CAR-T resistance. Furthermore, we underscore the need for specific FMC63 epitope detection to deliver information on antigen levels accessible for CD19-CAR-T-cells.
Collapse
Affiliation(s)
- Marta Krawczyk
- Department of Immunology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
- Doctoral School of Translational Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Narcis Fernandez-Fuentes
- Josep Carreras Leukemia Research Institute, Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain
| | - Klaudyna Fidyt
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
- Josep Carreras Leukemia Research Institute, Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain
| | - Tomasz Winiarski
- Warsaw University of Technology, Institute of Control and Computation Engineering, Warsaw, Poland
| | - Monika Pepek
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Agnieszka Graczyk-Jarzynka
- Department of Immunology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Jacinta Davis
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Pablo Bousquets-Muñoz
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Xose S Puente
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Pablo Menendez
- Josep Carreras Leukemia Research Institute, Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Red Española de Terapias Avanzadas (TERAV) - Instituto de Salud Carlos III (ISCII)
- Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Institut de Recerca Hospital Sant Joan de Déu-Pediatric Cancer Center Barcelona (SJD-PCCB), Barcelona, Spain
| | - Emmanuelle Benard
- Translational Research Unit, Section of Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Sébastien Wälchli
- Translational Research Unit, Section of Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Andrei Thomas-Tikhonenko
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology & Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Magdalena Winiarska
- Department of Immunology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|