1
|
Lee HK, Chen J, Philips RL, Lee SG, Feng X, Wu Z, Liu C, Schultz AB, Dalzell M, Meggendorfer M, Haferlach C, Birnbaum F, Sexton JA, Keating AE, O'Shea JJ, Young NS, Villarino AV, Furth PA, Hennighausen L. STAT5B leukemic mutations, altering SH2 tyrosine 665, have opposing impacts on immune gene programs. Life Sci Alliance 2025; 8:e202503222. [PMID: 40228864 PMCID: PMC11999048 DOI: 10.26508/lsa.202503222] [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: 01/14/2025] [Revised: 03/31/2025] [Accepted: 03/31/2025] [Indexed: 04/16/2025] Open
Abstract
STAT5B is a vital transcription factor for lymphocytes. Here, the function of two STAT5B mutations from human T-cell leukemias: one substituting tyrosine 665 with phenylalanine (STAT5BY665F) and the other with histidine (STAT5BY665H), was interrogated. In silico modeling predicted divergent energetic effects on homodimerization with a range of pathogenicity. In primary T cells in vitro, STAT5BY665F showed gain-of-function, whereas STAT5BY665H demonstrated loss-of-function. Introducing the mutation into the mouse genome illustrated that the gain-of-function Stat5b Y665F mutation resulted in accumulation of CD8+ effector and memory and CD4+ regulatory T cells, altering CD8+/CD4+ ratios. In contrast, STAT5BY665H "knock-in" mice showed diminished CD8+ effector and memory and CD4+ regulatory T cells. In contrast to WT STAT5B, the STAT5BY665F variant displayed greater STAT5 phosphorylation, DNA binding, and transcriptional activity after cytokine activation, whereas the STAT5BY665H variant resembled a null. The work exemplifies how joining in silico and in vivo studies of single nucleotides deepens our understanding of disease-associated variants, revealing structural determinants of altered function, defining mechanistic roles, and, specifically here, identifying a gain-of-function variant that does not directly induce hematopoietic malignancy.
Collapse
Affiliation(s)
- Hye Kyung Lee
- Laboratory of Genetics and Physiology, National Institute of Diabetes and Digestive and Kidney Diseases, US National Institutes of Health, Bethesda, MD, USA
| | - Jichun Chen
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Rachael L Philips
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sung-Gwon Lee
- Laboratory of Genetics and Physiology, National Institute of Diabetes and Digestive and Kidney Diseases, US National Institutes of Health, Bethesda, MD, USA
| | - Xingmin Feng
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Zhijie Wu
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Chengyu Liu
- Transgenic Core, National Heart, Lung, and Blood Institute, US National Institutes of Health, Bethesda, MD, USA
| | - Aaron B Schultz
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Molly Dalzell
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | | | - Claudia Haferlach
- Munich Leukemia Laboratory (MLL) Max-Lebsche-Platz 31, München, Germany
| | - Foster Birnbaum
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Joel A Sexton
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Amy E Keating
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - John J O'Shea
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Neal S Young
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alejandro V Villarino
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Priscilla A Furth
- Laboratory of Genetics and Physiology, National Institute of Diabetes and Digestive and Kidney Diseases, US National Institutes of Health, Bethesda, MD, USA
| | - Lothar Hennighausen
- Laboratory of Genetics and Physiology, National Institute of Diabetes and Digestive and Kidney Diseases, US National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
2
|
Yan F, Sun Y, Zhang S, Jia Y, Zhang J, Huang L, Xu Q, Zhang Y, Chen S, Wu X, Li R. Computer aided design of CGA-N9 derived peptides based on oligopeptide transporters and their antifungal evaluations. Bioorg Chem 2025; 160:108485. [PMID: 40267776 DOI: 10.1016/j.bioorg.2025.108485] [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: 01/24/2025] [Revised: 04/12/2025] [Accepted: 04/15/2025] [Indexed: 04/25/2025]
Abstract
CGA-N9 is an antifungal peptide that primarily targets Candida spp. with a mild activity. Our preceding research confirmed that the CGA-N9 crosses cell membrane with the assistance of C. tropicalis oligopeptide transporter (CtOPT) -1 and - 9. In this study, CGA-N9-derived peptides were designed following the molecular docking results with CtOPT-1 and -9. Compared with CGAN9, they exhibit higher transmembrane efficiency with the assistance of CtOPT-1 during the early phase of transmembrane processes and CtOPT-9 in the late phase. And they displayed significantly enhanced antifungal activity, with lower minimum inhibitory concentrations (MICs) against C. tropicalis, C. albicans, and C. parapsilosis, as well as improved biosafety. Among them, CGAN93 was the most optimizing, with a therapeutic index of 145.33. Furthermore, in a mouse model of systemic candidiasis, CGAN93 demonstrated a therapeutic effect comparable to fluconazole, significantly improving the survival rate of mice, attenuating organ damage, and enhancing the immune organ index. In conclusion, OPTs-based computer aided design is an effective strategy for enhancing the activities of antimicrobial peptides (AMPs) by improving transmembrane transport efficiency. CGAN93 is a promising drug candidate for treating Candidiasis.
Collapse
Affiliation(s)
- Fu Yan
- Zhengzhou Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, 450001 Zhengzhou, Henan, PR China; School of Biological Engineering, Henan University of Technology, 450001 Zhengzhou, Henan, PR China
| | - Yiqing Sun
- Zhengzhou Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, 450001 Zhengzhou, Henan, PR China; School of Biological Engineering, Henan University of Technology, 450001 Zhengzhou, Henan, PR China
| | - Shaojie Zhang
- Zhengzhou Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, 450001 Zhengzhou, Henan, PR China; School of Biological Engineering, Henan University of Technology, 450001 Zhengzhou, Henan, PR China
| | - Yifan Jia
- Zhengzhou Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, 450001 Zhengzhou, Henan, PR China; School of Biological Engineering, Henan University of Technology, 450001 Zhengzhou, Henan, PR China
| | - Jinhua Zhang
- Zhengzhou Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, 450001 Zhengzhou, Henan, PR China; School of Biological Engineering, Henan University of Technology, 450001 Zhengzhou, Henan, PR China
| | - Liang Huang
- School of Biological Engineering, Henan University of Technology, 450001 Zhengzhou, Henan, PR China
| | - Qiang Xu
- Zhengzhou Giant Biochemical Group Co., Ltd, 450001 Zhengzhou, Henan, PR China
| | - Yinzhi Zhang
- Zhengzhou Giant Biochemical Group Co., Ltd, 450001 Zhengzhou, Henan, PR China
| | - Shihua Chen
- School of Biological Engineering, Henan University of Technology, 450001 Zhengzhou, Henan, PR China
| | - Xingquan Wu
- School of Biological Engineering, Henan University of Technology, 450001 Zhengzhou, Henan, PR China
| | - Ruifang Li
- Zhengzhou Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, 450001 Zhengzhou, Henan, PR China; School of Biological Engineering, Henan University of Technology, 450001 Zhengzhou, Henan, PR China.
| |
Collapse
|
3
|
Dumas N, Portelli G, Ji Y, Dupont F, Jendoubi M, Lalli E. Detection of protein structural hotspots using AI distillation and explainability: application to the DAX-1 protein. NAR Genom Bioinform 2025; 7:lqaf047. [PMID: 40264682 PMCID: PMC12012785 DOI: 10.1093/nargab/lqaf047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 03/26/2025] [Accepted: 04/10/2025] [Indexed: 04/24/2025] Open
Abstract
AlphaMissense is a valuable resource for discerning important functional regions within proteins, providing pathogenicity heatmaps that highlight the pathogenic risk of specific mutations along the protein sequence. However, due to protein folding and long-range interactions, the actual structural alterations with functional implications may be occurring at a distance from the mutation site. As a result, the identification of the most sensitive structural regions for protein function may be hampered by the presence of mutations that indirectly affect the critical regions from a distance. In this study, we illustrate how the use of AlphaMissense predictions to train an XGBoost regression model on structural features extracted from the structures of protein variants predicted by OmegaFold enables the definition of a new explainability metric: a residue-based importance score that highlights the most critical structural domains within a protein sequence. To verify the accuracy of our approach, we applied it to the extensively studied protein DAX-1 and successfully identified critical structural domains. Notably, as this score only requires knowledge of the protein's amino acid sequence, it is valuable in guiding experimental investigations aimed at discovering functionally crucial regions in proteins that have been poorly characterized.
Collapse
Affiliation(s)
- Noé Dumas
- Thales SA, Thales Services Numériques, 06560 Valbonne—Sophia Antipolis, France
| | - Geoffrey Portelli
- Thales SA, Thales Services Numériques, 06560 Valbonne—Sophia Antipolis, France
| | - Yang Ji
- Thales SA, Thales Services Numériques, 06560 Valbonne—Sophia Antipolis, France
| | - Florent Dupont
- Thales SA, Thales Services Numériques, 06560 Valbonne—Sophia Antipolis, France
| | - Mehdi Jendoubi
- Thales SA, Thales Services Numériques, 06560 Valbonne—Sophia Antipolis, France
| | - Enzo Lalli
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne—Sophia Antipolis, France
- Institut national de la santé et de la recherche médicale, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne—Sophia Antipolis, France
- Université Côte d’Azur, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne—Sophia Antipolis, France
| |
Collapse
|
4
|
Merdler-Rabinowicz R, Dadush A, Patiyal S, Rajagopal PS, Daya G, Ben-Aroya S, Schäffer A, Eisenberg E, Ruppin E, Levanon E. A systematic evaluation of the therapeutic potential of endogenous-ADAR editors in cancer prevention and treatment. NAR Cancer 2025; 7:zcaf016. [PMID: 40330550 PMCID: PMC12053386 DOI: 10.1093/narcan/zcaf016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/10/2025] [Accepted: 05/05/2025] [Indexed: 05/08/2025] Open
Abstract
Adenosine deaminases acting on RNA (ADAR) enzymes constitute a natural cellular mechanism that induces A-to-I(G) editing, introducing genetic changes at the RNA level. Recently, interest in the endogenous-ADAR editor has emerged for correcting genetic mutations, consisting of a programmed oligonucleotide that attracts the native ADAR, thereby offering opportunities for medical therapy. Here, we systematically chart the scope of cancer mutations that endogenous-ADAR can correct. First, analyzing germline single nucleotide variants in cancer predisposition genes, we find that endogenous-ADAR can revert a fifth of them, reducing the risk of cancer development later in life. Second, examining somatic mutations across various cancer types, we find that it has the potential to correct at least one driver mutation in over a third of the samples, suggesting a promising future treatment strategy. We also highlight key driver mutations that are amenable to endogenous-ADAR, and are thus of special clinical interest. As using endogenous-ADAR entails delivering relatively small payloads, the prospects of delivering endogenous-ADAR to various cancers seem promising. We expect that the large scope of correctable mutations that are systematically charted here for the first time will pave the way for a new era of cancer treatment options.
Collapse
Affiliation(s)
- Rona Merdler-Rabinowicz
- Cancer Data Science Lab, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, United States
- Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, 5290002, Israel
- The Institute of Nanotechnology and Advanced Materials, Bar‐Ilan University, Ramat Gan, 5290002, Israel
| | - Ariel Dadush
- Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, 5290002, Israel
- The Institute of Nanotechnology and Advanced Materials, Bar‐Ilan University, Ramat Gan, 5290002, Israel
| | - Sumeet Patiyal
- Cancer Data Science Lab, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, United States
| | - Padma Sheila Rajagopal
- Cancer Data Science Lab, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, United States
| | - Gulzar N Daya
- Cancer Data Science Lab, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, United States
| | - Shay Ben-Aroya
- Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, 5290002, Israel
| | - Alejandro A Schäffer
- Cancer Data Science Lab, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, United States
| | - Eli Eisenberg
- Raymond and Beverly Sackler School of Physics and Astronomy, Tel-Aviv University, Tel Aviv, 6997801, Israel
| | - Eytan Ruppin
- Cancer Data Science Lab, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, United States
| | - Erez Y Levanon
- Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, 5290002, Israel
- The Institute of Nanotechnology and Advanced Materials, Bar‐Ilan University, Ramat Gan, 5290002, Israel
| |
Collapse
|
5
|
Kumar A, Sun YJ, Rasmussen DK, Hargrave A, Phillips C, Vu JT, Costa MG, Leung LSB, Yu C, Dubra A, Mahajan VB. Enhanced genotype-phenotype analysis using multimodal adaptive optics and 3D protein structure in Bietti Crystalline Dystrophy. Am J Ophthalmol Case Rep 2025; 38:102312. [PMID: 40236510 PMCID: PMC11997262 DOI: 10.1016/j.ajoc.2025.102312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 03/06/2025] [Accepted: 03/17/2025] [Indexed: 04/17/2025] Open
Abstract
Purpose Deep phenotyping of genetic retinal disease using multimodal adaptive optics ophthalmoscopy and protein structure variant analysis. Observations In a patient with extensive atrophy of the retinal pigment epithelium and yellow deposits in the retina, genetic testing identified two CYP4V2 variants: c.802-8_810delinsGC and c.1169G > A, p.Arg390His. AI-generated protein structures indicated loss of CYP4V2 function. Reflectance confocal and multiple-scattering Adaptive Optics Scanning Light Ophthalmoscopy (AOSLO) captured crystalline deposits throughout the retina as well as previously unreported cyst-like structures that were mainly independent from the crystalline deposits. Sequential AOSLO imaging was conducted and revealed anatomical and morphological changes in the cysts and surrounding cellular structures. Conclusions and importance Cyst-like changes may represent a new BCD degenerative feature. Characterizing retinal genetic disease variants with protein structural modeling and phenotyping with AOSLO represents an advanced approach for clinical diagnosis and may serve as a biomarker of disease progression.
Collapse
Affiliation(s)
- Aarushi Kumar
- Molecular Surgery Laboratory, Stanford University, Palo Alto, CA, USA
- Department of Ophthalmology, Byers Eye Institute, Stanford University, Palo Alto, CA, USA
| | - Young Joo Sun
- Molecular Surgery Laboratory, Stanford University, Palo Alto, CA, USA
- Department of Ophthalmology, Byers Eye Institute, Stanford University, Palo Alto, CA, USA
| | - Ditte K. Rasmussen
- Molecular Surgery Laboratory, Stanford University, Palo Alto, CA, USA
- Department of Ophthalmology, Byers Eye Institute, Stanford University, Palo Alto, CA, USA
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Aubrey Hargrave
- Department of Ophthalmology, Byers Eye Institute, Stanford University, Palo Alto, CA, USA
| | - Claudia Phillips
- Molecular Surgery Laboratory, Stanford University, Palo Alto, CA, USA
- Department of Ophthalmology, Byers Eye Institute, Stanford University, Palo Alto, CA, USA
| | - Jennifer T. Vu
- Molecular Surgery Laboratory, Stanford University, Palo Alto, CA, USA
- Department of Ophthalmology, Byers Eye Institute, Stanford University, Palo Alto, CA, USA
| | - Mauricio G.S. Costa
- Programa de Computação Científica, Vice-Presidência de Educação Informação e Comunicação, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Loh-Shan B. Leung
- Department of Ophthalmology, Byers Eye Institute, Stanford University, Palo Alto, CA, USA
| | - Charles Yu
- Department of Ophthalmology, Byers Eye Institute, Stanford University, Palo Alto, CA, USA
| | - Alfredo Dubra
- Department of Ophthalmology, Byers Eye Institute, Stanford University, Palo Alto, CA, USA
| | - Vinit B. Mahajan
- Molecular Surgery Laboratory, Stanford University, Palo Alto, CA, USA
- Department of Ophthalmology, Byers Eye Institute, Stanford University, Palo Alto, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| |
Collapse
|
6
|
Krenn M, Wagner M, Trimmel K, Bonelli S, Rath J, Jud J, Schwarz M, Milenkovic I, Weng R, Koren J, Baumgartner C, Brugger M, Brunet T, Graf E, Winkelmann J, Aull-Watschinger S, Zimprich F, Pataraia E. Holistic Exome-Based Genetic Testing in Adults With Epilepsy. Neurol Genet 2025; 11:e200260. [PMID: 40343077 PMCID: PMC12060788 DOI: 10.1212/nxg.0000000000200260] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 02/11/2025] [Indexed: 05/11/2025]
Abstract
Background and Objectives Exome sequencing (ES) is increasingly used in the diagnostic workup of epilepsies. While its utility has been extensively demonstrated in children, its role in adults remains to be defined. In this study, we evaluate the outcomes of a holistic exome-based approach in adults with epilepsy. Methods We included 106 adults with epilepsy and a presumed genetic etiology between January 2015 and December 2023 at the Medical University of Vienna, Austria. Diagnostic ES, including copy number variation (CNV) and mitochondrial analyses, was performed. We report on diagnostic outcomes, phenotype expansions, and research findings. Furthermore, we compared the diagnostic outcomes with 3 comprehensive gene panels. Results In our cohort, the diagnostic yield was 30.2%, outperforming all 3 simulated gene panels. A developmental and epileptic encephalopathy phenotype was associated with receiving a genetic diagnosis. Overall, 27 distinct molecular etiologies were identified. Eight patients had pathogenic CNVs, and 2 had mitochondrial DNA variants. Molecular diagnoses had potential clinical implications in 8 of 32 solved cases (25%), which were eventually exerted in 5 patients (15.6%). Tailored treatment changes were successfully applied in SCN1A-related epilepsy (discontinuation of sodium channel blockers) and GLUT1 deficiency (ketogenic diet). Three patients with mitochondrial diseases were referred for preventive screening investigations after the genetic diagnosis. Our findings expand the clinical spectrum of 3 known epilepsy genes. In addition, explorative variant prioritization identified heterozygous truncating variants in CLASP1 in 2 unrelated patients with focal epilepsy, suggesting it as a candidate gene. Discussion Our study strongly supports the use of holistic genetic approaches, encompassing CNV and mitochondrial analyses, in adults with epilepsy. Similar to pediatric cohorts, results may inform clinical care. Moreover, we report on phenotype expansions and a candidate gene discovery.
Collapse
Affiliation(s)
- Martin Krenn
- Department of Neurology, Medical University of Vienna, Austria
- Comprehensive Center for Clinical Neurosciences & Mental Health, Medical University of Vienna, Austria
| | - Matias Wagner
- Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Germany
- Institute of Neurogenomics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Karin Trimmel
- Department of Neurology, Medical University of Vienna, Austria
- Comprehensive Center for Clinical Neurosciences & Mental Health, Medical University of Vienna, Austria
| | - Silvia Bonelli
- Department of Neurology, Medical University of Vienna, Austria
- Comprehensive Center for Clinical Neurosciences & Mental Health, Medical University of Vienna, Austria
| | - Jakob Rath
- Department of Neurology, Medical University of Vienna, Austria
- Comprehensive Center for Clinical Neurosciences & Mental Health, Medical University of Vienna, Austria
| | - Judith Jud
- Department of Neurology, Medical University of Vienna, Austria
- Comprehensive Center for Clinical Neurosciences & Mental Health, Medical University of Vienna, Austria
| | - Michelle Schwarz
- Department of Neurology, Medical University of Vienna, Austria
- Comprehensive Center for Clinical Neurosciences & Mental Health, Medical University of Vienna, Austria
| | - Ivan Milenkovic
- Department of Neurology, Medical University of Vienna, Austria
- Comprehensive Center for Clinical Neurosciences & Mental Health, Medical University of Vienna, Austria
| | - Rosa Weng
- Department of Neurology, Medical University of Vienna, Austria
- Comprehensive Center for Clinical Neurosciences & Mental Health, Medical University of Vienna, Austria
| | - Johannes Koren
- Karl Landsteiner Institute for Clinical Epilepsy Research and Cognitive Neurology, Vienna, Austria
- Department of Neurology, Clinic Hietzing, Vienna, Austria
| | - Christoph Baumgartner
- Karl Landsteiner Institute for Clinical Epilepsy Research and Cognitive Neurology, Vienna, Austria
- Department of Neurology, Clinic Hietzing, Vienna, Austria
| | - Melanie Brugger
- Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Germany
- Department of Obstetrics and Gynecology, Klinikum Rechts der Isar, Technical University of Munich, Germany; and
| | - Theresa Brunet
- Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Germany
- Department of Pediatric Neurology and Developmental Medicine and Ludwig Maximilians University Center for Children with Medical Complexity, Dr. von Hauner Children's Hospital, Ludwig Maximilians University Hospital, Ludwig Maximilians University, Munich, Germany
| | - Elisabeth Graf
- Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Germany
| | - Juliane Winkelmann
- Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Germany
- Institute of Neurogenomics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Susanne Aull-Watschinger
- Department of Neurology, Medical University of Vienna, Austria
- Comprehensive Center for Clinical Neurosciences & Mental Health, Medical University of Vienna, Austria
| | - Fritz Zimprich
- Department of Neurology, Medical University of Vienna, Austria
- Comprehensive Center for Clinical Neurosciences & Mental Health, Medical University of Vienna, Austria
| | - Ekaterina Pataraia
- Department of Neurology, Medical University of Vienna, Austria
- Comprehensive Center for Clinical Neurosciences & Mental Health, Medical University of Vienna, Austria
| |
Collapse
|
7
|
Kurz NS, Kornrumpf K, Tucholski T, Drofenik K, König A, Beißbarth T, Dönitz J. Onkopus: precise interpretation and prioritization of sequence variants for biomedical research and precision medicine. Nucleic Acids Res 2025:gkaf376. [PMID: 40377094 DOI: 10.1093/nar/gkaf376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 04/14/2025] [Accepted: 04/25/2025] [Indexed: 05/18/2025] Open
Abstract
One of the major challenges in precision oncology is the identification of pathogenic, actionable variants and the selection of personalized treatments. We present Onkopus, a variant interpretation framework based on a modular architecture, for interpreting and prioritizing genetic alterations in cancer patients. A multitude of tools and databases are integrated into Onkopus to provide a comprehensive overview about the consequences of a variant, each with its own semantic, including pathogenicity predictions, allele frequency, biochemical and protein features, and therapeutic options. We present the characteristics of variants and personalized therapies in a clear and concise form, supported by interactive plots. To support the interpretation of variants of unknown significance (VUS), we present a protein analysis based on protein structures, which allows variants to be analyzed within the context of the entire protein, thereby serving as a starting point for understanding the underlying causes of variant pathogenicity. Onkopus has the potential to significantly enhance variant interpretation and the selection of actionable variants for identifying new targets, drug screens, drug testing using organoids, or personalized treatments in molecular tumor boards. We provide a free public instance of Onkopus at https://mtb.bioinf.med.uni-goettingen.de/onkopus.
Collapse
Affiliation(s)
- Nadine S Kurz
- Department of Medical Bioinformatics, University Medical Center Göttingen, 37077 Göttingen, Germany
- Göttingen Comprehensive Cancer Center (G-CCC), 37075 Göttingen, Germany
| | - Kevin Kornrumpf
- Department of Medical Bioinformatics, University Medical Center Göttingen, 37077 Göttingen, Germany
| | - Tim Tucholski
- Department of Medical Bioinformatics, University Medical Center Göttingen, 37077 Göttingen, Germany
- Institute of Pathology, University Medical Center Göttingen , 37075 Göttingen, Germany
| | - Klara Drofenik
- Department of Medical Bioinformatics, University Medical Center Göttingen, 37077 Göttingen, Germany
- Göttingen Comprehensive Cancer Center (G-CCC), 37075 Göttingen, Germany
| | - Alexander König
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Tim Beißbarth
- Department of Medical Bioinformatics, University Medical Center Göttingen, 37077 Göttingen, Germany
- Göttingen Comprehensive Cancer Center (G-CCC), 37075 Göttingen, Germany
- Campus Institute Data Science (CIDAS), Section Medical Data Science (MeDaS), 37077 Göttingen, Germany
| | - Jürgen Dönitz
- Department of Medical Bioinformatics, University Medical Center Göttingen, 37077 Göttingen, Germany
- Göttingen Comprehensive Cancer Center (G-CCC), 37075 Göttingen, Germany
- Campus Institute Data Science (CIDAS), Section Medical Data Science (MeDaS), 37077 Göttingen, Germany
| |
Collapse
|
8
|
Solomon BD, Cheatham M, de Guimarães TAC, Duong D, Haendel MA, Hsieh TC, Javanmardi B, Johnson B, Krawitz P, Kruszka P, Laurent T, Lee NC, McWalter K, Michaelides M, Mohnike K, Pontikos N, Guillen Sacoto MJ, Shwetar YJ, Ustach VD, Waikel RL, Woof W. Perspectives on the Current and Future State of Artificial Intelligence in Medical Genetics. Am J Med Genet A 2025:e64118. [PMID: 40375359 DOI: 10.1002/ajmg.a.64118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/14/2025] [Accepted: 05/02/2025] [Indexed: 05/18/2025]
Abstract
Artificial intelligence (AI) is rapidly transforming numerous aspects of daily life, including clinical practice and biomedical research. In light of this rapid transformation, and in the context of medical genetics, we assembled a group of leaders in the field to respond to the question about how AI is affecting, and especially how AI will affect, medical genetics. The authors who contributed to this collection of essays intentionally represent different areas of expertise, career stages, and geographies, and include diverse types of clinicians, computer scientists, and researchers. The individual pieces cover a wide range of areas related to medical genetics; we expect that these pieces may provide helpful windows into the ways in which AI is being actively studied, used, and considered in medical genetics.
Collapse
Affiliation(s)
- Benjamin D Solomon
- Medical Genomics Unit, National Human Genome Research Institute, Bethesda, Maryland, USA
| | - Morgan Cheatham
- Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Thales A C de Guimarães
- Moorfields Eye Hospital National Health Service Foundation Trust, London, UK
- University College London Institute of Ophthalmology, London, UK
- National Institute for Health and Care Research Moorfields Biomedical Research Centre, London, UK
| | - Dat Duong
- Medical Genomics Unit, National Human Genome Research Institute, Bethesda, Maryland, USA
| | - Melissa A Haendel
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Tzung-Chien Hsieh
- Institute for Genomic Statistics and Bioinformatics, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Behnam Javanmardi
- Institute for Genomic Statistics and Bioinformatics, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | | | - Peter Krawitz
- Institute for Genomic Statistics and Bioinformatics, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | | | | | - Ni-Chung Lee
- Department of Pediatrics and Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
| | | | - Michel Michaelides
- Moorfields Eye Hospital National Health Service Foundation Trust, London, UK
- University College London Institute of Ophthalmology, London, UK
- National Institute for Health and Care Research Moorfields Biomedical Research Centre, London, UK
| | - Klaus Mohnike
- Children's Hospital, Otto-von-Guericke-University, Magdeburg, Germany
| | - Nikolas Pontikos
- Moorfields Eye Hospital National Health Service Foundation Trust, London, UK
- University College London Institute of Ophthalmology, London, UK
- National Institute for Health and Care Research Moorfields Biomedical Research Centre, London, UK
| | | | - Yousif J Shwetar
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - Rebekah L Waikel
- Medical Genomics Unit, National Human Genome Research Institute, Bethesda, Maryland, USA
| | - William Woof
- University College London Institute of Ophthalmology, London, UK
- National Institute for Health and Care Research Moorfields Biomedical Research Centre, London, UK
| |
Collapse
|
9
|
Liu Z, Cao X, Wu M, Huang W, Dong X, Chen X, Zhang C. Mechanisms of PFBA toxicity in Chlorella vulgaris: Photosynthesis, oxidative stress, and antioxidant impairment. ENVIRONMENTAL RESEARCH 2025; 273:121228. [PMID: 40015437 DOI: 10.1016/j.envres.2025.121228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/12/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025]
Abstract
Perfluorobutanoic acid (PFBA), an emerging alternative to perfluorooctanoic acid (PFOA), has become increasingly prevalent in aquatic ecosystems, yet its ecotoxicological impacts remain poorly understood. This study investigated the aquatic toxicity of PFBA using the freshwater algae Chlorella vulgaris (C. vulgaris) as a model organism, employing a 96h pre-exposure assay to determine the median effective concentration followed by acute toxicity experiments analyzing multiple endpoints including growth, photosynthetic parameters, oxidative stress markers, and antioxidant enzyme activities. Computer simulation techniques were utilized to illustrate the underlying molecular mechanisms of PFBA toxicity. The results showed that the 96h-EC50 value of PFBA was 154.88 mg/L, which is comparable to conventional per- and polyfluoroalkyl substances (PFAS). Acute toxicity experiments revealed a biphasic dose-response relationship to the algal growth with the hormetic effects at the lower concentrations (30.97-92.93 mg/L) but inhibition at the higher levels (123.91-185.86 mg/L) of PFBA. High dosages of PFBA significantly decreased the maximum photosynthetic yield (Fv/Fm) and relative electron transfer rate (rETR), while inducing oxidative stress and inhibiting superoxide dismutase (SOD) and catalase (CAT) activities. Future AlphaFold2 modeling and molecular docking simulations demonstrated the potential binding of PFBA to photosystem II D1 C-terminal processing protease (PSII D1 protein), SOD, and CAT. These findings reveal a complex toxicity mechanism of PFBA on C. vulgaris involving photosynthetic disruption, oxidative stress, and antioxidant system impairment, contributing to the understanding of short-chain PFAS alternative ecotoxicity in aquatic ecosystems.
Collapse
Affiliation(s)
- Zeliang Liu
- Shaanxi Key Laboratory of Environmental Engineering, Xi'an University of Architecture and Technology, Xi'an, 710055, People's Republic of China; Key Laboratory of Northwest Water Resource, Environment and Ecology, MOE, Xi'an University of Architecture and Technology, Xi'an, 710055, People's Republic of China; Shaanxi province Higher Education Key Laboratory for Soil Pollution Remediation and Solid Waste Resource Utilization, Xi'an University of Architecture and Technology, Xi'an, 710055, People's Republic of China
| | - Xuanlin Cao
- Shaanxi province Higher Education Key Laboratory for Soil Pollution Remediation and Solid Waste Resource Utilization, Xi'an University of Architecture and Technology, Xi'an, 710055, People's Republic of China
| | - Manli Wu
- Shaanxi Key Laboratory of Environmental Engineering, Xi'an University of Architecture and Technology, Xi'an, 710055, People's Republic of China; Key Laboratory of Northwest Water Resource, Environment and Ecology, MOE, Xi'an University of Architecture and Technology, Xi'an, 710055, People's Republic of China; Shaanxi province Higher Education Key Laboratory for Soil Pollution Remediation and Solid Waste Resource Utilization, Xi'an University of Architecture and Technology, Xi'an, 710055, People's Republic of China.
| | - Wenjie Huang
- Shaanxi Key Laboratory of Environmental Engineering, Xi'an University of Architecture and Technology, Xi'an, 710055, People's Republic of China; Key Laboratory of Northwest Water Resource, Environment and Ecology, MOE, Xi'an University of Architecture and Technology, Xi'an, 710055, People's Republic of China
| | - Xia Dong
- Shaanxi Key Laboratory of Environmental Engineering, Xi'an University of Architecture and Technology, Xi'an, 710055, People's Republic of China; Key Laboratory of Northwest Water Resource, Environment and Ecology, MOE, Xi'an University of Architecture and Technology, Xi'an, 710055, People's Republic of China; Shaanxi province Higher Education Key Laboratory for Soil Pollution Remediation and Solid Waste Resource Utilization, Xi'an University of Architecture and Technology, Xi'an, 710055, People's Republic of China
| | - Xing Chen
- Dublin City University, School of Electronic Engineering, Ireland
| | - Chun Zhang
- Shaanxi Key Laboratory of Environmental Monitoring and Forewarning of Trace Pollutants, People's Republic of China.
| |
Collapse
|
10
|
Wang J, Zhang L, Wang S, Wang X, Li S, Gong P, Bai M, Paul A, Tvedt N, Ren H, Yang M, Zhang Z, Zhou S, Sun J, Wu X, Kuang H, Du Z, Dong Y, Shi X, Li M, Shukla D, Yan L, Guan Y. AlphaFold-Guided Bespoke Gene Editing Enhances Field-Grown Soybean Oil Contents. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2500290. [PMID: 40365797 DOI: 10.1002/advs.202500290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/19/2025] [Indexed: 05/15/2025]
Abstract
Enhancing the oil or protein content of soybean, a major crop for oil and protein production is highly desirable. GmSWEET10a encodes a sugar transporter that is strongly selected during domestication and breeding, increasing seed size and oil content. GmSWEET10b is functionally similar to GmSWEET10a, yet has not been artificially selected. Here, AlphaFold is used to find that C-terminal variants of GmSWEET10a can endow enhanced or reduced transport activity. Guided by AlphaFold, the functionality is improved for GmSWEET10a in terms of oil content through gene editing. Furthermore, novel GmSWEET10b haplotypes possessing strengthened or weakened sugar-transport capabilities that are absent in nature are engineered. Consequently, soybean oil content or protein content in independent GmSWEET10b gene-edited lines during multi-year and multi-site field trials is consistently increased, without negatively affecting yield. The study demonstrates that the combination of AlphaFold-guided protein design and gene editing has the potential to generate novel beneficial alleles, which can optimize protein function in the context of crop breeding.
Collapse
Affiliation(s)
- Jie Wang
- College of Resources and Environment, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Li Zhang
- Guangdong Provincial Key Laboratory of Plant Adaptation and Molecular Design, Innovative Center of Molecular Genetics and Evolution, School of Life Sciences, Guangzhou University, Guangzhou, 510006, China
| | - Shoudong Wang
- Key Laboratory of Soybean Molecular Design Breeding, State Key Laboratory of Black Soils Conservation and Utilization, Northeast Institute of Geography and Agroecology, Chinese Academy of Sciences, Changchun, 130102, China
| | - Xin Wang
- Guangdong Provincial Key Laboratory of Plant Adaptation and Molecular Design, Innovative Center of Molecular Genetics and Evolution, School of Life Sciences, Guangzhou University, Guangzhou, 510006, China
| | - Suning Li
- Jiangxi Province Key Laboratory of Oil Crops Genetic Improvement, Crop Institute, Jiangxi Academy of Agricultural Sciences, Nanchang, 330200, China
| | - Pingping Gong
- Fujian Provincial Key Laboratory of Haixia Applied Plant Systems Biology, Haixia Institute of Science and Technology and School of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Mengyan Bai
- Guangdong Provincial Key Laboratory of Plant Adaptation and Molecular Design, Innovative Center of Molecular Genetics and Evolution, School of Life Sciences, Guangzhou University, Guangzhou, 510006, China
| | - Arnav Paul
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Nathan Tvedt
- Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Hengrui Ren
- Guangdong Provincial Key Laboratory of Plant Adaptation and Molecular Design, Innovative Center of Molecular Genetics and Evolution, School of Life Sciences, Guangzhou University, Guangzhou, 510006, China
| | - Maoxiang Yang
- College of Resources and Environment, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Zhihui Zhang
- College of Resources and Environment, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Shaodong Zhou
- College of Resources and Environment, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Jiayi Sun
- Key Laboratory of Soybean Molecular Design Breeding, State Key Laboratory of Black Soils Conservation and Utilization, Northeast Institute of Geography and Agroecology, Chinese Academy of Sciences, Changchun, 130102, China
| | - Xianjin Wu
- College of Resources and Environment, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Huaqin Kuang
- Guangdong Provincial Key Laboratory of Plant Adaptation and Molecular Design, Innovative Center of Molecular Genetics and Evolution, School of Life Sciences, Guangzhou University, Guangzhou, 510006, China
| | - Zhenghua Du
- Fujian Provincial Key Laboratory of Haixia Applied Plant Systems Biology, Haixia Institute of Science and Technology and School of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Yonghui Dong
- Fujian Provincial Key Laboratory of Haixia Applied Plant Systems Biology, Haixia Institute of Science and Technology and School of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Xiaolei Shi
- Hebei Laboratory of Crop Genetics and Breeding, Institute of Cereal and Oil Crops, Hebei Academy of Agricultural and Forestry Sciences, Shijiazhuang, 050035, China
| | - Meina Li
- Guangdong Provincial Key Laboratory of Plant Adaptation and Molecular Design, Innovative Center of Molecular Genetics and Evolution, School of Life Sciences, Guangzhou University, Guangzhou, 510006, China
| | - Diwakar Shukla
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
- Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Long Yan
- Hebei Laboratory of Crop Genetics and Breeding, Institute of Cereal and Oil Crops, Hebei Academy of Agricultural and Forestry Sciences, Shijiazhuang, 050035, China
| | - Yuefeng Guan
- Guangdong Provincial Key Laboratory of Plant Adaptation and Molecular Design, Innovative Center of Molecular Genetics and Evolution, School of Life Sciences, Guangzhou University, Guangzhou, 510006, China
| |
Collapse
|
11
|
Ramasamy R, Raveendran M, Harris RA, Le HD, Mure LS, Benegiamo G, Dkhissi-Benyahya O, Cooper H, Rogers J, Panda S. Genome-wide allele-specific expression in multi-tissue samples from healthy male baboons reveals the transcriptional complexity of mammals. CELL GENOMICS 2025; 5:100823. [PMID: 40187355 DOI: 10.1016/j.xgen.2025.100823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 12/13/2024] [Accepted: 03/06/2025] [Indexed: 04/07/2025]
Abstract
Allele-specific expression (ASE) is pivotal in understanding the genetic underpinnings of phenotypic variation within species, differences in disease susceptibility, and responses to environmental factors. We processed 11 different tissue types collected from 12 age-matched healthy olive baboons (Papio anubis) for genome-wide ASE analysis. By sequencing their genomes at a minimum depth of 30×, we identified over 16 million single-nucleotide variants (SNVs). We also generated long-read sequencing data, enabling the phasing of all variants present within the coding regions of 96.5% of assayable protein-coding genes as a single haplotype block. Given the extensive heterozygosity of baboons relative to humans, we could quantify ASE across 72% of the total annotated protein-coding gene set. We identified genes that exhibit ASE and affect specific tissues and genotypes. We discovered ASE SNVs that also exist in human populations with identical alleles and that are designated as pathogenic by both the PrimateAI-3D and AlphaMissense models.
Collapse
Affiliation(s)
- Ramesh Ramasamy
- Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Muthuswamy Raveendran
- Human Genome Sequencing Center and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - R Alan Harris
- Human Genome Sequencing Center and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hiep D Le
- Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Ludovic S Mure
- Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Giorgia Benegiamo
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Ouria Dkhissi-Benyahya
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM, Stem Cell and Brain Research Institute U1208, 69500 Bron, France
| | - Howard Cooper
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM, Stem Cell and Brain Research Institute U1208, 69500 Bron, France
| | - Jeffrey Rogers
- Human Genome Sequencing Center and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Satchidananda Panda
- Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
12
|
Khalil A, Supek F. DiffInvex identifies evolutionary shifts in driver gene repertoires during tumorigenesis and chemotherapy. Nat Commun 2025; 16:4209. [PMID: 40360478 PMCID: PMC12075687 DOI: 10.1038/s41467-025-59397-8] [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: 08/07/2024] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Somatic cells can transform into tumors due to mutations, and the tumors further evolve towards increased aggressiveness and therapy resistance. We develop DiffInvex, a framework for identifying changes in selection acting on individual genes in somatic genomes, drawing on an empirical mutation rate baseline derived from non-coding DNA that accounts for shifts in neutral mutagenesis during cancer evolution. We apply DiffInvex to >11,000 somatic whole-genome sequences from ~30 cancer types or healthy tissues, identifying genes where point mutations are under conditional positive or negative selection during exposure to specific chemotherapeutics, suggesting drug resistance mechanisms occurring via point mutation. DiffInvex identifies 11 genes exhibiting treatment-associated selection for different classes of chemotherapies, linking selected mutations in PIK3CA, APC, MAP2K4, SMAD4, STK11 and MAP3K1 with drug exposure. Various gene-chemotherapy associations are further supported by differential functional impact of mutations pre- versus post-therapy, and are also replicated in independent studies. In addition to nominating drug resistance genes, we contrast the genomes of healthy versus cancerous cells of matched human tissues. We identify noncancerous expansion-specific drivers, including NOTCH1 and ARID1A. DiffInvex can also be applied to diverse analyses in cancer evolution to identify changes in driver gene repertoires across time or space.
Collapse
Affiliation(s)
- Ahmed Khalil
- Institute for Research in Biomedicine (IRB Barcelona), 08028, Barcelona, Spain
| | - Fran Supek
- Institute for Research in Biomedicine (IRB Barcelona), 08028, Barcelona, Spain.
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark.
- Catalan Institution for Research and Advanced Studies (ICREA), 08010, Barcelona, Spain.
| |
Collapse
|
13
|
Boone PM, Buenaventura T, King JWD, Merkenschlager M. X-linked competition - implications for human development and disease. Nat Rev Genet 2025:10.1038/s41576-025-00840-3. [PMID: 40355603 DOI: 10.1038/s41576-025-00840-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2025] [Indexed: 05/14/2025]
Abstract
During early mammalian female development, X chromosome inactivation leads to random transcriptional silencing of one of the two X chromosomes. This inactivation is maintained through subsequent cell divisions, leading to intra-individual diversity, whereby cells express either the maternal or paternal X chromosome. Differences in X chromosome sequence content can trigger competitive interactions between clones that may alter organismal development and skew the representation of X-linked sequence variants in a cell-type-specific manner - a recently described phenomenon termed X-linked competition in analogy to existing cell competition paradigms. Skewed representation can define the phenotypic impact of X-linked variants, for example, the manifestation of disease in female carriers of X-linked disease alleles. Here, we review what is currently known about X-linked competition, reflect on what remains to be learnt and map out the implications for X-linked human disease.
Collapse
Affiliation(s)
- Philip M Boone
- Cornelia de Lange Syndrome and Related Disorders Clinic, Boston Children's Hospital, Boston, MA, USA
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Teresa Buenaventura
- MRC Laboratory of Medical Sciences, London, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - James W D King
- MRC Laboratory of Medical Sciences, London, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Matthias Merkenschlager
- MRC Laboratory of Medical Sciences, London, UK.
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK.
| |
Collapse
|
14
|
Sangeet S, Sinha A, Nair MB, Mahata A, Sarkar R, Roy S. EVOLVE: A Web Platform for AI-Based Protein Mutation Prediction and Evolutionary Phase Exploration. J Chem Inf Model 2025; 65:4293-4310. [PMID: 40309917 DOI: 10.1021/acs.jcim.5c00026] [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: 05/02/2025]
Abstract
While predicting structure-function relationships from sequence data is fundamental in biophysical chemistry, identifying prospective single-point and collective mutation sites in proteins can help us stay ahead in understanding their potential effects on protein structure and function. Addressing the challenges of large sequence-space analysis, we present EVOLVE, a web tool enabling researchers to explore prospective mutation sites and their collective behavior. EVOLVE integrates a statistical mechanics-guided machine learning algorithms to predict probable mutational sites, with statistical mechanics calculating mutational entropy to accurately identify mutational hotspots. Validation against a number of viral protein sequences confirms its ability to predict mutations and their functional consequences. By leveraging statistical mechanics of phase transition concept, EVOLVE also quantifies mutational entropy fluctuations, offering a quantitative foundation for identifying Variants of Concern (VOC) or Variants under Monitoring (VUM) as per World Health Organization (WHO) guidelines. EVOLVE streamlines data upload and analysis with a user-friendly interface and comprehensive tutorials. Access EVOLVE free at https://evolve-iiserkol.com.
Collapse
Affiliation(s)
- Satyam Sangeet
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, West Bengal 741246, India
- School of Physics, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Anushree Sinha
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, West Bengal 741246, India
| | - Madhav B Nair
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, West Bengal 741246, India
| | - Arpita Mahata
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, West Bengal 741246, India
| | - Raju Sarkar
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, West Bengal 741246, India
| | - Susmita Roy
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, West Bengal 741246, India
| |
Collapse
|
15
|
Mondal S, Shrivastava P, Mehra R. Computing pathogenicity of mutations in human cytochrome P450 superfamily. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2025; 1873:141078. [PMID: 40349948 DOI: 10.1016/j.bbapap.2025.141078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/22/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
Cytochrome P450 (CYPs) are crucial heme-containing enzymes that metabolize drugs and endogenous compounds. In humans, 57 CYP isoforms have been identified, with over 200 mutations linked to severe disorders. Our comprehensive computational study assessed the reason for the pathogenicity of mutations by comparing pathogenic and non-pathogenic variants. We analyzed 25,94,151 mutations across 26 CYP structures using structure- and sequence-based methods, revealing a meaningful stability pattern: non-pathogenic > all > pathogenic mutation datasets. Notably, pathogenic mutations were predominantly buried within CYP structures, indicating a higher potential for pathogenesis. We identified three key amino acid properties affected by mutations: Gibbs free energy, isoelectric point, and volume. Furthermore, diseased mutations significantly reduced positive residue content, particularly due to arginine mutations, which directly influenced the isoelectric point. Our findings indicate a greater likelihood of pathogenic mutations occurring at conserved sites, disrupting CYP function. A higher frequency of pathogenic mutations was observed in heme sites, primarily involving arginine, which may interfere with arginine-heme interactions. Molecular docking revealed a differential binding of heme in wild-type and pathogenic CYPs. This study provides a foundational analysis of mutation effects across multiple CYPs. It models the chemical basis of CYP-related pathogenicity, facilitating the development of a semi-quantitative disease prediction model.
Collapse
Affiliation(s)
- Somnath Mondal
- Department of Chemistry, Indian Institute of Technology Bhilai, Durg 491002, Chhattisgarh, India
| | - Pranchal Shrivastava
- Department of Chemistry, Indian Institute of Technology Bhilai, Durg 491002, Chhattisgarh, India
| | - Rukmankesh Mehra
- Department of Chemistry, Indian Institute of Technology Bhilai, Durg 491002, Chhattisgarh, India; Department of Bioscience and Biomedical Engineering, Indian Institute of Technology Bhilai, Durg 491002, Chhattisgarh, India.
| |
Collapse
|
16
|
Jing X, Liu Z, Li W, Ma K, Zhang J, Yan Z, Zhang S, Lin J, Zhao J, Ong KK, Perry JRB, Zhao Y. Protein-truncating variants in UQCRC1 are associated with Parkinson's disease: evidence from half-million people. NPJ Parkinsons Dis 2025; 11:120. [PMID: 40346065 PMCID: PMC12064775 DOI: 10.1038/s41531-025-00987-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/30/2025] [Indexed: 05/11/2025] Open
Abstract
Recent studies have suggested a potential but inconsistent link between UQCRC1 and Parkinson's disease (PD). For the first time, we systematically investigated the association between non-synonymous variants in UQCRC1 and PD risk using data from the UK Biobank with half-million participants, which provide evidence supporting the role of UQCRC1 Protein-truncating variants (PTVs) in PD (P = 1.20 × 10-6, OR = 6.59) and highlight the importance of large-scale population studies in identifying rare genetic risk factors.
Collapse
Affiliation(s)
- Xiaoxi Jing
- Changping Laboratory, Yard-28, Science Park Road, Changping District, Beijing, P. R. China
| | - Zongzhi Liu
- Changping Laboratory, Yard-28, Science Park Road, Changping District, Beijing, P. R. China
| | - Wenwen Li
- Innovation Center for Neurological Disorders and Department of Neurology, National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Kaiyan Ma
- Changping Laboratory, Yard-28, Science Park Road, Changping District, Beijing, P. R. China
| | - Jiaxiang Zhang
- Changping Laboratory, Yard-28, Science Park Road, Changping District, Beijing, P. R. China
| | - Zeqi Yan
- Changping Laboratory, Yard-28, Science Park Road, Changping District, Beijing, P. R. China
| | - Shuo Zhang
- Changping Laboratory, Yard-28, Science Park Road, Changping District, Beijing, P. R. China
| | - Jiecong Lin
- Changping Laboratory, Yard-28, Science Park Road, Changping District, Beijing, P. R. China
| | - Junpeng Zhao
- Changping Laboratory, Yard-28, Science Park Road, Changping District, Beijing, P. R. China
| | - Ken K Ong
- Innovation Center for Neurological Disorders and Department of Neurology, National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - John R B Perry
- MRC Epidemiology Unit and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
- Metabolic Research Laboratories, MRC Metabolic Diseases Unit and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Yajie Zhao
- Changping Laboratory, Yard-28, Science Park Road, Changping District, Beijing, P. R. China.
| |
Collapse
|
17
|
Liu Z, Curtis D. Analysis of Rare Coding Variants in 470,000 UK Biobank Participants Reveals Genetic Associations With Childhood Asthma Predisposition. Int J Immunogenet 2025. [PMID: 40342259 DOI: 10.1111/iji.12714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Previous studies of genetic contributions to risk of childhood asthma have implicated common variants with small effect sizes. Some studies using exome sequence data have reported associations with rare coding variants having larger effects on risk, notably an analysis of 145,000 subjects which found association with loss of function (LOF) variants in FLG, the gene coding for filaggrin. Here, we report the results of an analysis of rare nonsynonymous and LOF variants, carried out on the full UK Biobank cohort of 470,000 exome-sequenced participants. The phenotype of childhood asthma was defined as reporting asthma with onset before 18. Regression analysis was applied to gene-wise tests for association of LOF and nonsynonymous variants. Forty-five tests using different pathogenicity predictors were applied to the first cohort of 200,000 participants. Subsequently, the 100 genes showing strongest evidence for association were analysed in the second cohort of 270,000 participants, using only the best-performing predictor for each gene. For FLG, separate analyses were carried out for participants with atopic dermatitis. Three genes achieved statistical significance after correction for testing these 100 genes: FLG, IL33 and PRKCQ. The effects on asthma risk and frequencies of variants in different functional categories were characterised for these genes. Damaging coding variants were associated with increased risk of asthma in FLG and IL33 but reduced risk in PRKCQ. FLG LOF variants were also associated with the risk of atopic dermatitis, and their effect on asthma risk was higher in people who reported a diagnosis of atopic dermatitis. Rare coding variants in a small number of genes have important effects on asthma risk. Further study of individual variant effects might elucidate mechanisms of pathogenesis. This research has been conducted using the UK Biobank Resource.
Collapse
Affiliation(s)
- Zhenzhen Liu
- UCL Genetics Institute, University College London, London, UK
| | - David Curtis
- UCL Genetics Institute, University College London, London, UK
| |
Collapse
|
18
|
Chhibbar P, Das J. Machine learning approaches enable the discovery of therapeutics across domains. Mol Ther 2025; 33:2269-2278. [PMID: 40186352 DOI: 10.1016/j.ymthe.2025.04.001] [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: 02/13/2025] [Revised: 03/21/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025] Open
Abstract
Multi-modal datasets have grown exponentially in the last decade. This has created an enormous demand for machine learning models that can predict complex outcomes by leveraging cellular, molecular, and humoral profiles. Corresponding inference of mechanisms can help to uncover new therapeutic targets. Here, we discuss how biological principles guide the design of predictive models and how interpretable machine learning can lead to novel mechanistic insights. We provide descriptions of multiple learning techniques and how suited they are to domain adaptations. Finally, we talk about broad learning capabilities of foundation models on large datasets and whether they can be used to provide meaningful inference about biological datasets.
Collapse
Affiliation(s)
- Prabal Chhibbar
- Centre for Systems Immunology, Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; Integrative Systems Biology PhD Program, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| | - Jishnu Das
- Centre for Systems Immunology, Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
19
|
Banerjee A, Bogetti AT, Bahar I. Accurate identification and mechanistic evaluation of pathogenic missense variants with Rhapsody-2. Proc Natl Acad Sci U S A 2025; 122:e2418100122. [PMID: 40314982 PMCID: PMC12067267 DOI: 10.1073/pnas.2418100122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 04/06/2025] [Indexed: 05/03/2025] Open
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, Stony Brook, NY11794
- Department of Biochemistry and Cell Biology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY11794
| | - Anthony T. Bogetti
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY11794
- Department of Biochemistry and Cell Biology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY11794
| | - Ivet Bahar
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY11794
- Department of Biochemistry and Cell Biology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY11794
| |
Collapse
|
20
|
Sharma I, Hall K, Moonah S. CRISPR genome editing using a combined positive and negative selection system. PLoS One 2025; 20:e0321881. [PMID: 40327602 PMCID: PMC12054870 DOI: 10.1371/journal.pone.0321881] [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: 12/17/2024] [Accepted: 03/11/2025] [Indexed: 05/08/2025] Open
Abstract
The clustered regularly interspaced short palindromic repeats (CRISPR)/Cas system is a powerful genome editing tool that has revolutionized research. Single nucleotide polymorphisms (SNPs) are the most common form of genetic variation in humans. Only a subset of these SNPs has been shown to be linked to genetic diseases, while the biological relevance of the majority remains unclear. Investigating these variants of unknown significance could provide valuable insights into their roles in biological processes, disease susceptibility, and treatment responses. While CRISPR/Cas has emerged as a transformative technology, its ability to make single nucleotide substitutions remains a significant limitation. Other techniques in single nucleotide editing, such as base editing and prime editing, offer promising possibilities to complement CRISPR/Cas systems, though they also have their own limitations. Hence, alternative approaches are necessary to overcome the limitations of CRISPR. Here, to improve the feasibility of generating single base edits in the genome, we provide a protocol that introduces a multiple expression and dual selection (MEDS) system, which, alongside CRISPR, utilizes the opposing roles of cytosine deaminase/uracil phosphoribosyltransferase (CD/UPRT) for negative selection and neomycin phosphotransferase II (NPT II) for positive selection. As a proof of concept and to demonstrate feasibility of the method, we used MEDS, along with traditional CRISPR-Cas9, to generate sickle hemoglobin by introducing a point mutation (A → T) in the sixth codon of the hemoglobin beta gene.
Collapse
Affiliation(s)
- Ishrya Sharma
- Department of Medicine, University of Florida, GainesvilleFlorida, United States of America
| | - Kerisa Hall
- Department of Medicine, University of Florida, GainesvilleFlorida, United States of America
| | - Shannon Moonah
- Department of Medicine, University of Florida, GainesvilleFlorida, United States of America
- Department of Molecular Genetics and Microbiology, University of Florida, GainesvilleFlorida, United States of America
| |
Collapse
|
21
|
Tekpinar M, David L, Henry T, Carbone A. PRESCOTT: a population aware, epistatic, and structural model accurately predicts missense effects. Genome Biol 2025; 26:113. [PMID: 40329382 PMCID: PMC12054230 DOI: 10.1186/s13059-025-03581-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 04/17/2025] [Indexed: 05/08/2025] Open
Abstract
Predicting the functional impact of point mutations is a critical challenge in genomics. PRESCOTT reconstructs complete mutational landscapes, identifies mutation-sensitive regions, and categorizes missense variants as benign, pathogenic, or variants of uncertain significance. Leveraging protein sequences, structural models, and population-specific allele frequencies, PRESCOTT surpasses existing methods in classifying ClinVar variants, the ACMG dataset, and over 1800 proteins from the Human Protein Dataset. Its online server facilitates mutation effect predictions for any protein and variant, and includes a database of over 19,000 human proteins, ready for population-specific analyses. Open access to residue-specific scores offers transparency and valuable insights for genomic medicine.
Collapse
Affiliation(s)
- Mustafa Tekpinar
- Department of Computational, Quantitative and Synthetic Biology (CQSB), Sorbonne Université, CNRS, IBPS, UMR 7238, Paris, 75005, France
| | - Laurent David
- Department of Computational, Quantitative and Synthetic Biology (CQSB), Sorbonne Université, CNRS, IBPS, UMR 7238, Paris, 75005, France
| | - Thomas Henry
- Centre International de Recherche en Infectiologie (CIRI), Inserm U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Univ Lyon, Lyon, 69007, France
| | - Alessandra Carbone
- Department of Computational, Quantitative and Synthetic Biology (CQSB), Sorbonne Université, CNRS, IBPS, UMR 7238, Paris, 75005, France.
- Institut Universitaire de France (IUF), Paris, France.
| |
Collapse
|
22
|
Zhou K, Gheybi K, Soh PXY, Hayes VM. Evaluating variant pathogenicity prediction tools to establish African inclusive guidelines for germline genetic testing. COMMUNICATIONS MEDICINE 2025; 5:157. [PMID: 40328947 PMCID: PMC12056225 DOI: 10.1038/s43856-025-00883-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 04/24/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Genetic germline testing is restricted for African patients. Lack of ancestrally relevant genomic data perpetuated by African diversity has resulted in European-biased curated clinical variant databases and pathogenic prediction guidelines. While numerous variant pathogenicity prediction tools (VPPTs) exist, their performance has yet to be established within the context of African diversity. METHODS To address this limitation, we assessed 54 VPPTs for predictive performance (sensitivity, specificity, false positive and negative rates) across 145,291 known pathogenic or benign variants derived from 50 Southern African and 50 European men matched for advanced prostate cancer. Prioritising VPPTs for optimal ancestral performance, we screened 5.3 million variants of unknown significance for predicted functional and oncogenic potential. RESULTS We observe a 2.1- and 4.1-fold increase in the number of known and predicted rare pathogenic or benign variants, respectively, against a 1.6-fold decrease in the number of available interrogated variants in our European over African data. Although sensitivity was significantly lower for our African data overall (0.66 vs 0.71, p = 9.86E-06), MetaSVM, CADD, Eigen-raw, BayesDel-noAF, phyloP100way-vertebrate and MVP outperformed irrespective of ancestry. Conversely, MutationTaster, DANN, LRT and GERP-RS were African-specific top performers, while MutationAssessor, PROVEAN, LIST-S2 and REVEL are European-specific. Using these pathogenic prediction workflows, we narrow the ancestral gap for potentially deleterious and oncogenic variant prediction in favour of our African data by 1.15- and 1.1-fold, respectively. CONCLUSION Although VPPT sensitivity favours European data, our findings provide guidelines for VPPT selection to maximise rare pathogenic variant prediction for African disease studies.
Collapse
Affiliation(s)
- Kangping Zhou
- Ancestry and Health Genomics Laboratory, Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW, Australia
| | - Kazzem Gheybi
- Ancestry and Health Genomics Laboratory, Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW, Australia
| | - Pamela X Y Soh
- Ancestry and Health Genomics Laboratory, Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW, Australia
| | - Vanessa M Hayes
- Ancestry and Health Genomics Laboratory, Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW, Australia.
- Manchester Cancer Research Centre, University of Manchester, Manchester, UK.
- School of Health Systems and Public Health, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa.
| |
Collapse
|
23
|
Schmitz EG, Griffith M, Griffith OL, Cooper MA. Identifying genetic errors of immunity due to mosaicism. J Exp Med 2025; 222:e20241045. [PMID: 40232243 PMCID: PMC11998702 DOI: 10.1084/jem.20241045] [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: 01/08/2025] [Revised: 02/24/2025] [Accepted: 03/24/2025] [Indexed: 04/16/2025] Open
Abstract
Inborn errors of immunity are monogenic disorders of the immune system that lead to immune deficiency and/or dysregulation in patients. Identification of precise genetic causes of disease aids diagnosis and advances our understanding of the human immune system; however, a significant portion of patients lack a molecular diagnosis. Somatic mosaicism, genetic changes in a subset of cells, is emerging as an important mechanism of immune disease in both young and older patients. Here, we review the current landscape of somatic genetic errors of immunity and methods for the detection and validation of somatic variants.
Collapse
Affiliation(s)
- Elizabeth G. Schmitz
- Division of Rheumatology/Immunology, Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, USA
| | - Malachi Griffith
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Obi L. Griffith
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Megan A. Cooper
- Division of Rheumatology/Immunology, Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
24
|
Lucas MC, Keßler T, Scharf F, Steinke-Lange V, Klink B, Laner A, Holinski-Feder E. A series of reviews in familial cancer: genetic cancer risk in context variants of uncertain significance in MMR genes: which procedures should be followed? Fam Cancer 2025; 24:42. [PMID: 40317406 DOI: 10.1007/s10689-025-00470-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 04/18/2025] [Indexed: 05/07/2025]
Abstract
Interpreting variants of uncertain significance (VUS) in mismatch repair (MMR) genes remains a major challenge in managing Lynch syndrome and other hereditary cancer syndromes. This review outlines recommended VUS classification procedures, encompassing foundational and specialized methodologies tailored for MMR genes by expert organizations, including InSiGHT and ClinGen's Hereditary Colorectal Cancer/Polyposis Variant Curation Expert Panel (VCEP). Key approaches include: (1) functional data, encompassing direct assays measuring MMR proficiency such as in vitro MMR assays, deep mutational scanning, and MMR cell-based assays, as well as techniques like methylation-tolerant assays, proteomic-based approaches, and RNA sequencing, all of which provide critical functional evidence supporting variant pathogenicity; (2) computational data/tools, including in silico meta-predictors and models, which contribute to robust VUS classification when integrated with experimental evidence; and (3) enhanced variant detection to identify the actual causal variant through whole-genome sequencing and long-read sequencing to detect pathogenic variants missed by traditional methods. These strategies improve diagnostic precision, support clinical decision-making for Lynch syndrome, and establish a flexible framework that can be applied to other OMIM-listed genes.
Collapse
Affiliation(s)
- Morghan C Lucas
- MGZ- Medical Genetics Center, Munich, Germany.
- Medizinische Klinik und Poliklinik IV- Campus Innenstadt, Klinikum der Universität München, Munich, Germany.
| | | | | | - Verena Steinke-Lange
- MGZ- Medical Genetics Center, Munich, Germany
- Medizinische Klinik und Poliklinik IV- Campus Innenstadt, Klinikum der Universität München, Munich, Germany
- Genturis European Reference Network (ERN) Genetic Tumor Risk (GENTURIS), Nijmegen, Netherlands
| | - Barbara Klink
- MGZ- Medical Genetics Center, Munich, Germany
- Genturis European Reference Network (ERN) Genetic Tumor Risk (GENTURIS), Nijmegen, Netherlands
| | | | - Elke Holinski-Feder
- MGZ- Medical Genetics Center, Munich, Germany
- Medizinische Klinik und Poliklinik IV- Campus Innenstadt, Klinikum der Universität München, Munich, Germany
- Genturis European Reference Network (ERN) Genetic Tumor Risk (GENTURIS), Nijmegen, Netherlands
| |
Collapse
|
25
|
Erdogan EN, Cheng CV, Caraffi SG, Ivanovski I, Piatelli G, Errichiello E, Papavasiliou AS, Vasileiou G, Reis A, Prince B, Hickey SE, Koboldt DC, Schneider MC, Porrmann J, Di Donato N, Leis T, Perry MS, Humberson J, Rotenberg J, Bakhtiari S, Magee H, Kheradmand S, Kruer MC, Swale A, Weber A, Landes C, Zuffardi O, Garavelli L, van Haeringen A, Ruivenkamp CAL, Pauly M, Au PYB, Dobyns WB, Aldinger KA. Further Delineation of the AUTS2 HX Repeat Domain-Related Phenotype. Am J Med Genet A 2025:e64093. [PMID: 40317680 DOI: 10.1002/ajmg.a.64093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/11/2025] [Accepted: 04/08/2025] [Indexed: 05/07/2025]
Abstract
Haploinsufficiency of AUTS2 is associated with a neurodevelopmental disorder characterized by intellectual disability, autistic features, and spasticity. AUTS2 protein interacts with p300, encoded by EP300, through the HX repeat domain of AUTS2, thereby activating transcription. We previously reported two de novo variants in the HX repeat domain of AUTS2. These variants disrupt the AUTS2-P300 interaction, resulting in a phenotype resembling Rubinstein-Taybi Syndrome (RSTS) associated with variants in EP300/CREBBP. Here, we expand beyond the initial clinical description to delineate the HX domain-associated phenotype and compare it to the AUTS2-haploinsufficient phenotype. We reviewed clinical data, photographs, and neuroimaging studies to examine genotype-phenotype relationships. Our review of 80 individuals included 14 individuals we present here and 66 individuals with AUTS2 variants presented in the literature. The clinical features for individuals with variants in the HX repeat domain include severe intellectual disability, severe language disability, distinct craniofacial and skeletal dysmorphic features, and neuroimaging findings. Facial dysmorphisms include wide and prominent nasal bridges with complex nasal shapes and dysmorphic eyebrows. Dysmorphisms include digit anomalies: Symphalangism and hypoplasia of distal phalanges, exclusive to the HX domain variant group. Cerebellar anomalies not seen with other AUTS2 variants are seen within this group. Our report delineates a distinct and severe clinical phenotype associated with variants in the AUTS2 HX domain, including an in-depth comparison with the AUTS2 haploinsufficiency phenotype features.
Collapse
Affiliation(s)
- Esin Nur Erdogan
- Norcliffe Foundation Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Chi Vicky Cheng
- Norcliffe Foundation Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Stefano G Caraffi
- Medical Genetics Unit, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Ivan Ivanovski
- Medical Genetics Unit, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Gianluca Piatelli
- Dipartimento Integrato Neuroscienze Mediche e Chirurgiche e Riabilitazione-Continuità Cure; U.O.C. Neurochirurgia, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Edoardo Errichiello
- Unit of Medical Genetics, Department of Molecular Medicine, University of Pavia, Pavia, Italy and Neurogenetics Research Center, IRCCS Mondino Foundation, Pavia, Italy
| | | | - Georgia Vasileiou
- Institute of Human Genetics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - André Reis
- Institute of Human Genetics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Bradley Prince
- Department of Medical Genetics, University of Calgary, Calgary, Canada
| | - Scott E Hickey
- Division of Genetics and Genomics, Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Pediatrics, The Ohio State College of Medicine, Columbus, Ohio, USA
| | - Daniel C Koboldt
- Department of Pediatrics, The Ohio State College of Medicine, Columbus, Ohio, USA
- Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Michael C Schneider
- Carle Physicians Group, Section of Neurology, St. Christopher's Hospital for Children, Urbana, Illinois, USA
| | - Joseph Porrmann
- Institute for Clinical Genetics, University Hospital TU Dresden, Dresden, Germany
| | - Nataliya Di Donato
- Institute for Clinical Genetics, University Hospital TU Dresden, Dresden, Germany
| | - Thomas Leis
- Department of Pediatrics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - M Scott Perry
- Jane and John Justin Institute for Mind Health, Fort Worth, Texas, USA
- Genetic Epilepsy Clinic, Cook Children's Medical Center, Fort Worth, Texas, USA
| | - Jennifer Humberson
- Genetics, University of Virginia Community Health Pediatric Specialty Care, Charlottesville, Virginia, USA
| | - Joshua Rotenberg
- Memorial Hermann Memorial City Medical Center, Houston, Texas, USA
| | - Somayeh Bakhtiari
- Pediatric Movement Disorders Program, Division of Pediatric Neurology, Barrow Neurological Institute, Phoenix Children's Hospital, Phoenix, Arizona, USA
- Departments of Child Health, Neurology, and Cellular & Molecular Medicine, and Program in Genetics, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, USA
| | - Helen Magee
- Pediatric Movement Disorders Program, Division of Pediatric Neurology, Barrow Neurological Institute, Phoenix Children's Hospital, Phoenix, Arizona, USA
- Departments of Child Health, Neurology, and Cellular & Molecular Medicine, and Program in Genetics, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, USA
| | | | - Michael C Kruer
- Pediatric Movement Disorders Program, Division of Pediatric Neurology, Barrow Neurological Institute, Phoenix Children's Hospital, Phoenix, Arizona, USA
- Departments of Child Health, Neurology, and Cellular & Molecular Medicine, and Program in Genetics, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, USA
| | - Andrew Swale
- North West Genomic Laboratory Hub (Liverpool), Manchester Centre for Genomic Medicine, Liverpool Women's Hospital, Liverpool, UK
| | | | - Caren Landes
- Alder hey Children's NHS Foundation Trust, Liverpool, UK
| | - Orsetta Zuffardi
- Unit of Medical Genetics, Department of Molecular Medicine, University of Pavia, Pavia, Italy and Neurogenetics Research Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Livia Garavelli
- Medical Genetics Unit, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Arie van Haeringen
- Department of Clinical Genetics, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Claudia A L Ruivenkamp
- Department of Clinical Genetics, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Melissa Pauly
- Institute of Human Genetics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ping Yee Billie Au
- Department of Medical Genetics, University of Calgary, Calgary, Canada
- Alberta Children's Hospital Research Institute, Calgary, Canada
| | - William B Dobyns
- Department of Pediatrics (Genetics), University of Minnesota, Minneapolis, Minnesota, USA
| | - Kimberly A Aldinger
- Norcliffe Foundation Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
- Department of Neurology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
26
|
Buckley RM, Bilgen N, Harris AC, Savolainen P, Tepeli C, Erdoğan M, Serres Armero A, Dreger DL, van Steenbeek FG, Hytönen MK, Parker HG, Hale J, Lohi H, Çınar Kul B, Boyko AR, Ostrander EA. Analysis of canine gene constraint identifies new variants for orofacial clefts and stature. Genome Res 2025; 35:1080-1093. [PMID: 40127928 PMCID: PMC12047267 DOI: 10.1101/gr.280092.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 03/10/2025] [Indexed: 03/26/2025]
Abstract
Dog breeding promotes within-group homogeneity through conformation to strict breed standards, while simultaneously driving between-group heterogeneity. There are over 350 recognized dog breeds that provide the foundation for investigating the genetic basis of phenotypic diversity. Typically, breed standard phenotypes such as stature, pelage, and craniofacial structure are analyzed through genetic association studies. However, such analyses are limited to assayed phenotypes only, leaving difficult-to-measure phenotypic subtleties easily overlooked. We investigated coding variation from over 2000 dogs, leading to discoveries of variants related to craniofacial morphology and stature. Breed-enriched variants were prioritized according to gene constraint, which was calculated using a mutation model derived from trinucleotide substitution probabilities. Among the newly found variants is a splice-acceptor variant in PDGFRA associated with bifid nose, a characteristic trait of Çatalburun dogs, implicating the gene's role in midline closure. Two additional LCORL variants, both associated with canine body size are also discovered: a frameshift that causes a premature stop in large breeds (>25 kg) and an intronic substitution found in small breeds (<10 kg), thus highlighting the importance of allelic heterogeneity in selection for breed traits. Most variants prioritized in this analysis are not associated with genomic signatures for breed differentiation, as these regions are enriched for constrained genes intolerant to nonsynonymous variation. This indicates trait selection in dogs is likely a balancing act between preserving essential gene functions and maximizing regulatory variation to drive phenotypic extremes.
Collapse
Affiliation(s)
- Reuben M Buckley
- National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Nüket Bilgen
- Department of Animal Genetics, Faculty of Veterinary Medicine, University of Ankara, Ankara 06110, Türkiye
| | - Alexander C Harris
- National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Peter Savolainen
- KTH Royal Institute of Technology, School of Chemistry, Biotechnology and Health, Science for Life Laboratory, SE-100 44 Stockholm, Sweden
| | - Cafer Tepeli
- Department of Animal Science, Faculty of Veterinary Medicine, University of Selcuk, Konya 42100, Türkiye
| | - Metin Erdoğan
- Department of Veterinary Biology and Genetics, Faculty of Veterinary Medicine, Afyon Kocatepe University, Afyonkarahisar 03200, Türkiye
| | - Aitor Serres Armero
- National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Dayna L Dreger
- National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Frank G van Steenbeek
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands
| | - Marjo K Hytönen
- Department of Medical and Clinical Genetics, University of Helsinki, 00014 Helsinki, Finland
- Department of Veterinary Biosciences, University of Helsinki, 00014 Helsinki, Finland
- Folkhälsan Research Center, 00290 Helsinki, Finland
| | - Heidi G Parker
- National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Jessica Hale
- National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Hannes Lohi
- Department of Medical and Clinical Genetics, University of Helsinki, 00014 Helsinki, Finland
- Department of Veterinary Biosciences, University of Helsinki, 00014 Helsinki, Finland
- Folkhälsan Research Center, 00290 Helsinki, Finland
| | - Bengi Çınar Kul
- Department of Animal Genetics, Faculty of Veterinary Medicine, University of Ankara, Ankara 06110, Türkiye
| | - Adam R Boyko
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York 14853, USA
- Embark Veterinary, Inc., Boston, Massachusetts 02210, USA
| | - Elaine A Ostrander
- National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892, USA;
| |
Collapse
|
27
|
Ward KS, Ptak CP, Pashkova N, Grider T, Peterson TA, Pareyson D, Pisciotta C, Saveri P, Moroni I, Laura M, Burns J, Menezes MP, Cornett K, Finkel R, Mukherjee-Clavin B, Sumner CJ, Greene M, Hamid OA, Herrmann D, Sadjadi R, Walk D, Züchner S, Reilly MM, Scherer SS, Piper RC, Shy ME. Charcot-Marie-Tooth disease type 1E: Clinical Natural History and Molecular Impact of PMP22 Variants. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.05.01.25326605. [PMID: 40343019 PMCID: PMC12060940 DOI: 10.1101/2025.05.01.25326605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Charcot-Marie-Tooth disease type 1E (CMT1E) is a rare, autosomal dominant peripheral neuropathy caused by missense variants, deletions, and truncations within the peripheral myelin protein-22 (PMP22) gene. CMT1E phenotypes vary depending on the specific variant, ranging from mild to severe, and there is little natural history and phenotypic progression data on individuals with CMT1E. Patients with CMT1E were evaluated during initial and follow-up visits at sites within the Inherited Neuropathy Consortium. Clinical characteristics were obtained from history, neurological exams, and nerve conduction studies. Clinical outcome measures were used to quantify baseline and longitudinal changes, including the Rasch-modified CMT Examination Score version 2 (CMTESv2-R) and the CMT Pediatric Scale (CMTPedS). The trafficking of PMP22 variants in transfected cells was correlated to disease severity. Twenty-four, presumed disease-causing PMP22 variants were identified in 50 individuals from 35 families, including 19 missense variants, three in-frame deletions, and two truncations. Twenty-nine patients presented with delayed walking during childhood. At their baseline evaluation, the mean CMTESv2-R in 46 patients was 16 ± 7.72 (out of 32), and the mean CMTPedS from 17 patients was 28 ± 6.35 (out of 44). Six individuals presented with hearing loss, eleven with scoliosis, three with hip dysplasia, and one with both scoliosis and hip dysplasia. Twenty variants were localized within in transmembrane domains; 31 of 35 individuals with these variants had moderate to severe phenotypes. Three variants were found in the extracellular domain and were associated with milder phenotypes. Reduced expression of PMP22 at the cell surface, and the location of missense variants within in the transmembrane domain correlated with disease severity. Pathogenic PMP22 variants located within the transmembrane regions usually cause a moderate to severe clinical phenotype, beginning in early childhood, and have impaired trafficking to the plasma membrane.
Collapse
Affiliation(s)
- Kailee S. Ward
- Department of Neurology, University of Iowa Health Care Medical Center, Iowa City, IA 52242, USA
| | - Christopher P. Ptak
- Biomolecular Nuclear Magnetic Resonance Facility, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Natalya Pashkova
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Tiffany Grider
- Department of Neurology, University of Iowa Health Care Medical Center, Iowa City, IA 52242, USA
| | - Tabitha A. Peterson
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Davide Pareyson
- Department of Clinical Neurosciences, Fondazione IRCCS Instituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Chiara Pisciotta
- Department of Clinical Neurosciences, Fondazione IRCCS Instituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Paola Saveri
- Department of Clinical Neurosciences, Fondazione IRCCS Instituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Isabella Moroni
- Department of Pediatric Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Matilde Laura
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Joshua Burns
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Manoj P. Menezes
- University of Sydney School of Health Sciences, Faculty of Medicine and Health; Paediatric Gait Analysis Service of New South Wales, Sydney Children’s Hospital Network, Sydney, 2145 Australia
| | - Kayla Cornett
- University of Sydney School of Health Sciences, Faculty of Medicine and Health; Paediatric Gait Analysis Service of New South Wales, Sydney Children’s Hospital Network, Sydney, 2145 Australia
| | - Richard Finkel
- Center for Experimental Neurotherapies, St. Jude Children’s Research Hospital, Memphis, TN USA
| | | | - Charlotte J. Sumner
- Department of Neurology, John Hopkins University School of Medicine, Baltimore, MD 21205
| | - Maxwell Greene
- Department of Neurology, Stanford University, Stanford, CA 94304, USA
| | - Omer Abdul Hamid
- Department of Neurology, Nemours Children’s Hospital, Orland, FL 32827, USA
| | - David Herrmann
- Department of Neurology, University of Rochester, Rochester, NY 14618, USA
| | - Reza Sadjadi
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - David Walk
- Department of Neurology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Stephan Züchner
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- John P. Hussman Institutue for Human Genetics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Mary M. Reilly
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Steven S. Scherer
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Robert C. Piper
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Michael E. Shy
- Department of Neurology, University of Iowa Health Care Medical Center, Iowa City, IA 52242, USA
| |
Collapse
|
28
|
Zhu H, Zhang J, Rao S, Durbin MD, Li Y, Lang R, Liu J, Xiao B, Shan H, Meng Z, Wang J, Tang X, Shi Z, Cox LL, Zhao S, Ware SM, Tan TY, de Silva M, Gallacher L, Liu T, Mi J, Zeng C, Zheng HF, Zhang Q, Antonarakis SE, Cox TC, Zhang YB. Common cis-regulatory variation modifies the penetrance of pathogenic SHROOM3 variants in craniofacial microsomia. Genome Res 2025; 35:1065-1079. [PMID: 40234029 PMCID: PMC12047249 DOI: 10.1101/gr.280047.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 03/10/2025] [Indexed: 04/17/2025]
Abstract
Pathogenic coding variants have been identified in thousands of genes, yet the mechanisms underlying the incomplete penetrance in individuals carrying these variants are poorly understood. In this study, in a cohort of 2009 craniofacial microsomia (CFM) patients of Chinese ancestry and 2625 Han Chinese controls, we identified multiple predicted pathogenic coding variants in SHROOM3 in both CFM patients and healthy individuals. We found that the penetrance of CFM correlates with specific haplotype combinations containing likely pathogenic-coding SHROOM3 variants and CFM-associated expression quantitative trait loci (eQTLs) of SHROOM3 expression. Further investigations implicate specific eQTL combinations, such as rs1001322 or rs344131, in combination with other significant CFM-associated eQTLs, which we term combined eQTL phenotype modifiers (CePMods). We additionally show that rs344131, located within a regulatory enhancer region of SHROOM3, demonstrates allele-specific effects on enhancer activity and thus impacts expression levels of the associated SHROOM3 allele harboring any rare coding variant. Our findings also suggest that CePMods may serve as pathogenic determinants, even in the absence of rare deleterious coding variants in SHROOM3 This highlights the critical role of allelic expression in determining the penetrance and severity of craniofacial abnormalities, including microtia and facial asymmetry. Additionally, using quantitative phenotyping, we demonstrate that both microtia and facial asymmetry are present in two separate Shroom3 mouse models, the severity of which is dependent on gene dosage. Our study establishes SHROOM3 as a likely pathogenic gene for CFM and demonstrates eQTLs as determinants of modified penetrance in the manifestation of the disease in individuals carrying likely pathogenic rare coding variants.
Collapse
Affiliation(s)
- Hao Zhu
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Jiao Zhang
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Beijing 100144, China
| | - Soumya Rao
- Department of Oral & Craniofacial Sciences, University of Missouri-Kansas City, Kansas City, Missouri 64108, USA
| | - Matthew D Durbin
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | - Ying Li
- Department of Otolaryngology-Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing 100051, China
| | - Ruirui Lang
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Jiqiang Liu
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Baichuan Xiao
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Hailin Shan
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Ziqiu Meng
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Jinmo Wang
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Xiaokai Tang
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Zhenni Shi
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Liza L Cox
- Department of Oral & Craniofacial Sciences, University of Missouri-Kansas City, Kansas City, Missouri 64108, USA
| | - Shouqin Zhao
- Department of Otolaryngology-Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing 100051, China
| | - Stephanie M Ware
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | - Tiong Y Tan
- Victorian Clinical Genetics Service, Royal Children's Hospital and Department of Pediatrics, University of Melbourne, Victoria 3052, Australia
| | - Michelle de Silva
- Victorian Clinical Genetics Service, Royal Children's Hospital and Department of Pediatrics, University of Melbourne, Victoria 3052, Australia
| | - Lyndon Gallacher
- Victorian Clinical Genetics Service, Royal Children's Hospital and Department of Pediatrics, University of Melbourne, Victoria 3052, Australia
| | - Ting Liu
- Department of Ophthalmology, Daping Hospital, Army Medical University, Chongqing 400000, China
| | - Jie Mi
- Center for Non-Communicable Disease Management, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Changqing Zeng
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Hou-Feng Zheng
- Center for Health and Data Science (CHDS), the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
- Diseases & Population (DaP) Geninfo Laboratory, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
| | - Qingguo Zhang
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Beijing 100144, China
| | - Stylianos E Antonarakis
- Department of Genetic Medicine and Development, University of Geneva Medical Faculty, Geneva 1211, Switzerland;
- Medigenome, Swiss Institute of Genomic Medicine, 1207 Geneva, Switzerland
- iGE3 Institute of Genetics and Genomes in Geneva, Geneva 1211, Switzerland
| | - Timothy C Cox
- Department of Oral & Craniofacial Sciences, University of Missouri-Kansas City, Kansas City, Missouri 64108, USA;
- Department of Pediatrics, University of Missouri-Kansas City, Kansas City, Missouri 64108, USA
| | - Yong-Biao Zhang
- School of Engineering Medicine, Beihang University, Beijing 100191, China;
- Key Laboratory of Big Data-Based Precision Medicine and Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing 100191, China
- National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), Beihang University, Beijing 100083, China
| |
Collapse
|
29
|
Ramadane-Morchadi L, Rotenberg N, Esteban-Sánchez A, Fortuno C, Gómez-Sanz A, Varga MJ, Chamberlin A, Richardson ME, Michailidou K, Pérez-Segura P, Spurdle AB, de la Hoya M. ACMG/AMP interpretation of BRCA1 missense variants: Structure-informed scores add evidence strength granularity to the PP3/BP4 computational evidence. Am J Hum Genet 2025; 112:993-1002. [PMID: 40233743 DOI: 10.1016/j.ajhg.2024.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 12/09/2024] [Accepted: 12/12/2024] [Indexed: 04/17/2025] Open
Abstract
Classification of missense variants is challenging. Lacking compelling clinical and/or functional data, ACMG/AMP lines of evidence are restricted to PM2 (rarity code applied at supporting level) and PP3/BP4 (computational evidence based mostly on multiple-sequence-alignment conservation tools). Currently, the ClinGen ENIGMA BRCA1/2 Variant Curation Expert Panel uses BayesDel to apply PP3/BP4 to missense variants located in the BRCA1 RING/BRCT domains. The ACMG/AMP framework does not refer explicitly to protein structure as a putative source of pathogenic/benign evidence. Here, we tested the value of incorporating structure-based evidence such as relative solvent accessibility (RSA), folding stability (ΔΔG), and/or AlphaMissense pathogenicity to the classification of BRCA1 missense variants. We used MAVE functional scores as proxies for pathogenicity/benignity. We computed RSA and FoldX5.0 ΔΔG predictions using as alternative input templates for either PDB files or AlphaFold2 models, and we retrieved pre-computed AlphaMissense and BayesDel scores. We calculated likelihood ratios toward pathogenicity/benignity provided by the tools (individually or combined). We performed a clinical validation of major findings using the large-scale BRIDGES case-control dataset. AlphaMissense outperforms ΔΔG and BayesDel, providing similar PP3/BP4 evidence strengths with lower rate of variants in the uninformative score range. AlphaMissense combined with ΔΔG increases evidence strength granularity. AlphaFold2 models perform well as input templates for ΔΔG predictions. Regardless of the tool, BP4 (but not PP3) is highly dependent on RSA, with benignity evidence provided only to variants targeting buried or partially buried residues (RSA ≤ 60%). Stratification by functional domain did not reveal major differences. In brief, structure-based analysis improves PP3/BP4 assessment, uncovering a relevant role for RSA.
Collapse
Affiliation(s)
- Lobna Ramadane-Morchadi
- Molecular Oncology Laboratory, Hospital Clínico San Carlos, IdISSC (Instituto de Investigación Sanitaria del Hospital Clínico San Carlos), 28040 Madrid, Spain
| | - Nitsan Rotenberg
- University of Queensland, Brisbane, QLD, Australia; Molecular Cancer Epidemiology Laboratory, QIMR Berghofer MRI, Herston, QLD 4006, Australia
| | - Ada Esteban-Sánchez
- Molecular Oncology Laboratory, Hospital Clínico San Carlos, IdISSC (Instituto de Investigación Sanitaria del Hospital Clínico San Carlos), 28040 Madrid, Spain
| | - Cristina Fortuno
- Molecular Cancer Epidemiology Laboratory, QIMR Berghofer MRI, Herston, QLD 4006, Australia
| | - Alicia Gómez-Sanz
- Molecular Oncology Laboratory, Hospital Clínico San Carlos, IdISSC (Instituto de Investigación Sanitaria del Hospital Clínico San Carlos), 28040 Madrid, Spain
| | | | | | | | - Kyriaki Michailidou
- Biostatistics Unit, The Cyprus Institute of Neurology & Genetics, 2371 Nicosia, Cyprus
| | - Pedro Pérez-Segura
- Molecular Oncology Laboratory, Hospital Clínico San Carlos, IdISSC (Instituto de Investigación Sanitaria del Hospital Clínico San Carlos), 28040 Madrid, Spain
| | - Amanda B Spurdle
- University of Queensland, Brisbane, QLD, Australia; Molecular Cancer Epidemiology Laboratory, QIMR Berghofer MRI, Herston, QLD 4006, Australia
| | - Miguel de la Hoya
- Molecular Oncology Laboratory, Hospital Clínico San Carlos, IdISSC (Instituto de Investigación Sanitaria del Hospital Clínico San Carlos), 28040 Madrid, Spain.
| |
Collapse
|
30
|
Taylor Gonzalez DJ, Djulbegovic MB, Sharma M, Antonietti M, Kim CK, Uversky VN, Karp CL, Shields CL, Wilson MW. AlphaMissense Predictions and ClinVar Annotations: A Deep Learning Approach to Uveal Melanoma. OPHTHALMOLOGY SCIENCE 2025; 5:100673. [PMID: 40114711 PMCID: PMC11925568 DOI: 10.1016/j.xops.2024.100673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/09/2024] [Accepted: 12/03/2024] [Indexed: 03/22/2025]
Abstract
Objective Uveal melanoma (UM) poses significant diagnostic and prognostic challenges due to its variable genetic landscape. We explore the use of a novel deep learning tool to assess the functional impact of genetic mutations in UM. Design A cross-sectional bioinformatics exploratory data analysis of genetic mutations from UM cases. Subjects Genetic data from patients diagnosed with UM were analyzed, explicitly focusing on missense mutations sourced from the Catalogue of Somatic Mutations in Cancer (COSMIC) database. Methods We identified missense mutations frequently observed in UM using the COSMIC database, assessed their potential pathogenicity using AlphaMissense, and visualized mutations using AlphaFold. Clinical significance was cross-validated with entries in the ClinVar database. Main Outcome Measures The primary outcomes measured were the agreement rates between AlphaMissense predictions and ClinVar annotations regarding the pathogenicity of mutations in critical genes associated with UM, such as GNAQ, GNA11, SF3B1, EIF1AX, and BAP1. Results Missense substitutions comprised 91.35% (n = 1310) of mutations in UM found on COSMIC. Of the 151 unique missense mutations analyzed in the most frequently mutated genes, only 40.4% (n = 61) had corresponding data in ClinVar. Notably, AlphaMissense provided definitive classifications for 27.2% (n = 41) of the mutations, which were labeled as "unknown significance" in ClinVar, underscoring its potential to offer more clarity in ambiguous cases. When excluding these mutations of uncertain significance, AlphaMissense showed perfect agreement (100%) with ClinVar across all analyzed genes, demonstrating no discrepancies where a mutation predicted as "pathogenic" was classified as "benign" or vice versa. Conclusions Integrating deep learning through AlphaMissense offers a promising approach to understanding the mutational landscape of UM. Our methodology holds the potential to improve genomic diagnostics and inform the development of personalized treatment strategies for UM. Financial Disclosures Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
| | - Mak B Djulbegovic
- Wills Eye Hospital, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Meghan Sharma
- Bascom Palmer Eye Institute, University of Miami, Miami, Florida
| | | | - Colin K Kim
- Bascom Palmer Eye Institute, University of Miami, Miami, Florida
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Carol L Karp
- Bascom Palmer Eye Institute, University of Miami, Miami, Florida
| | - Carol L Shields
- Ocular Oncology Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Matthew W Wilson
- Hamilton Eye Institute, University of Tennessee Science Center, Memphis, Tennessee
| |
Collapse
|
31
|
Bastarache L, Tinker RJ, Schuler BA, Richter L, Phillips JA, Stead WW, Hooker GW, Peterson JF, Ruderfer DM. Characterizing trends in clinical genetic testing: A single-center analysis of EHR data from 1.8 million patients over two decades. Am J Hum Genet 2025; 112:1029-1038. [PMID: 40245861 DOI: 10.1016/j.ajhg.2025.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 04/19/2025] Open
Abstract
A lack of structural data in electronic health records (EHRs) makes assessing the impact of genetic testing on clinical practice challenging. We extracted clinical genetic tests from the EHRs of more than 1.8 million patients seen at Vanderbilt University Medical Center from 2002 to 2022. With these data, we quantified the use of clinical genetic testing in healthcare and described how testing patterns and results changed over time. We assessed trends in types of genetic tests, tracked usage across medical specialties, and introduced a new measure, the genetically attributable fraction (GAF), to quantify the proportion of observed phenotypes attributable to a genetic diagnosis over time. We identified 104,392 tests and 19,032 molecularly confirmed diagnoses. The proportion of patients with genetic testing in their EHRs increased from 1.0% in 2002 to 6.1% in 2022, and testing became more comprehensive with the growing use of multi-gene panels. The number of unique diseases diagnosed with genetic testing increased from 51 in 2002 to 509 in 2022, and there was a rise in the number of variants of uncertain significance. The phenome-wide GAF for 6,505,620 diagnoses made in 2022 was 0.46%, and the GAF was greater than 5% for 74 phenotypes, including pancreatic insufficiency (67%), chorea (64%), atrial septal defect (24%), microcephaly (17%), paraganglioma (17%), and ovarian cancer (6.8%). Our study provides a comprehensive quantification of the increasing role of genetic testing at a major academic medical institution and demonstrates its growing utility in explaining the observed medical phenome.
Collapse
Affiliation(s)
- Lisa Bastarache
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA; Center for Digital Genomic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Rory J Tinker
- Division of Medical Genetics and Genomic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bryce A Schuler
- Center for Digital Genomic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Division of Medical Genetics and Genomic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lucas Richter
- Center for Digital Genomic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John A Phillips
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - William W Stead
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Gillian W Hooker
- Division of Medical Genetics and Genomic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Concert Genetics, Nashville, TN, USA
| | - Josh F Peterson
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Douglas M Ruderfer
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA; Center for Digital Genomic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
32
|
Feng M, Liu L, Xian ZN, Wei X, Li K, Yan W, Lu Q, Shi Y, He G. PSTP: accurate residue-level phase separation prediction using protein conformational and language model embeddings. Brief Bioinform 2025; 26:bbaf171. [PMID: 40315433 PMCID: PMC12047702 DOI: 10.1093/bib/bbaf171] [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: 01/18/2025] [Revised: 03/07/2025] [Accepted: 03/19/2025] [Indexed: 05/04/2025] Open
Abstract
Phase separation (PS) is essential in cellular processes and disease mechanisms, highlighting the need for predictive algorithms to analyze uncharacterized sequences and accelerate experimental validation. Current high-accuracy methods often rely on extensive annotations or handcrafted features, limiting their generalizability to sequences lacking such annotations and making it difficult to identify key protein regions involved in PS. We introduce Phase Separation's Transfer-learning Prediction (PSTP), which combines conformational embeddings with large language model embeddings, enabling state-of-the-art PS predictions from protein sequences alone. PSTP performs well across various prediction scenarios and shows potential for predicting novel-designed artificial proteins. Additionally, PSTP provides residue-level predictions that are highly correlated with experimentally validated PS regions. By analyzing 160 000+ variants, PSTP characterizes the strong link between the incidence of pathogenic variants and residue-level PS propensities in unconserved intrinsically disordered regions, offering insights into underexplored mutation effects. PSTP's sliding-window optimization reduces its memory usage to a few hundred megabytes, facilitating rapid execution on typical CPUs and GPUs. Offered via both a web server and an installable Python package, PSTP provides a versatile tool for decoding protein PS behavior and supporting disease-focused research.
Collapse
Affiliation(s)
- Mofan Feng
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, No. 1954 Huashan Road, Xuhui District, Shanghai 200030, China
- Shanghai Institute of Medical Genetics, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, No. 24 Lane 1400 West Beijing Road, Jing’an District, Shanghai 200040, China
| | - Liangjie Liu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, No. 1954 Huashan Road, Xuhui District, Shanghai 200030, China
- Shanghai Institute of Medical Genetics, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, No. 24 Lane 1400 West Beijing Road, Jing’an District, Shanghai 200040, China
| | - Zhuo-Ning Xian
- School of Environmental Science & Engineering, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Minhang District, Shanghai 200240, China
| | - Xiaoxi Wei
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, No. 1954 Huashan Road, Xuhui District, Shanghai 200030, China
| | - Keyi Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, No. 1954 Huashan Road, Xuhui District, Shanghai 200030, China
- Shanghai Institute of Medical Genetics, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, No. 24 Lane 1400 West Beijing Road, Jing’an District, Shanghai 200040, China
| | - Wenqian Yan
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, No. 1954 Huashan Road, Xuhui District, Shanghai 200030, China
- Shanghai Institute of Medical Genetics, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, No. 24 Lane 1400 West Beijing Road, Jing’an District, Shanghai 200040, China
| | - Qing Lu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, No. 1954 Huashan Road, Xuhui District, Shanghai 200030, China
| | - Yi Shi
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, No. 1954 Huashan Road, Xuhui District, Shanghai 200030, China
- Shanghai Institute of Medical Genetics, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, No. 24 Lane 1400 West Beijing Road, Jing’an District, Shanghai 200040, China
| | - Guang He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, No. 1954 Huashan Road, Xuhui District, Shanghai 200030, China
- Shanghai Institute of Medical Genetics, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, No. 24 Lane 1400 West Beijing Road, Jing’an District, Shanghai 200040, China
| |
Collapse
|
33
|
Zorn S, de Groot CJ, Brandt-Heunemann S, von Schnurbein J, Abawi O, Bounds R, Ruck L, Guijo B, Martos-Moreno GÁ, Nicaise C, Courbage S, Klehr-Martinelli M, Siebert R, Dubern B, Poitou C, Clément K, Argente J, Kühnen P, Farooqi IS, Wabitsch M, van den Akker E. Early childhood height, weight, and BMI development in children with monogenic obesity: a European multicentre, retrospective, observational study. THE LANCET. CHILD & ADOLESCENT HEALTH 2025; 9:297-305. [PMID: 40246357 DOI: 10.1016/s2352-4642(25)00065-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 04/19/2025]
Abstract
BACKGROUND Monogenic defects in the leptin-melanocortin pathway are associated with hyperphagia and severe, early-onset obesity. Early childhood growth patterns in height, weight, and BMI, might serve as phenotypic markers for specific genetic disorders; however, reliable data are scarce. This study aimed to evaluate the natural history of height, weight, and BMI in early childhood in a large European group of individuals with monogenic obesity. METHODS This multicentre observational study analysed height, weight, and BMI from birth to age 5 years in individuals diagnosed with biallelic (likely) pathogenic LEP, LEPR, POMC, PCSK1, or MC4R variants or monoallelic (likely) pathogenic MC4R variants from six European centres (Berlin and Ulm, Germany; Cambridge, UK; Madrid, Spain; Paris, France; Rotterdam, Netherlands). All patient data up to May 31, 2022 were included in this analysis. All individuals had at least two height or weight measurements between birth and age 5 years. Early childhood growth trajectories were compared with those of control children with obesity without a known genetic cause, following a negative next-generation sequencing panel. Diagnostic performance of BMI as a predictor test for monogenic obesity was also evaluated. FINDINGS We included 147 individuals with monogenic obesity. From the age of 6 months onwards, children with biallelic variants (n=88, 55% female vs 45% male) had substantially higher BMIs than those with monoallelic MC4R variants (n=59, 53% female vs 47% male) and control children (n=113, 59% female vs 41% male). Children with biallelic LEP, LEPR, and MC4R variants showed a steep BMI increase during the first year of life, followed by a plateau until age 5 years, whereas those with biallelic POMC variants did not plateau. Accelerated linear growth was only observed in children with biallelic MC4R variants starting from age 1 year. The optimal BMI cut-off for distinguishing individuals with biallelic variants from control individuals was identified at age 2 years, with a test positivity cutoff of 24·0 kg/m2 (sensitivity: 0·96 [95% CI 0·89-1·00], specificity: 0·83 [0·74-0·90], AUC: 0·96 [0·91-0·99], p<0·0001). However, BMI had poor diagnostic performance for monoallelic MC4R variants. INTERPRETATION This study identified characteristic early childhood BMI trajectories for different forms of monogenic obesity. From age 6 months onwards, individuals with biallelic variants can be distinguished from those with monoallelic variants and common obesity. A BMI ≥24 kg/m2 at age 2 years had good diagnostic performance for biallelic variants, informing future recommendations for genetic screening for monogenic obesity. FUNDING Federal Ministry of Education and Research as part of the German Center for Child and Adolescent Health, German Research Foundation, Spanish Ministry of Health, The Wellcome Trust, Botnar Fondation, Leducq Foundation, National Institute for Health Research (NIHR) Cambridge Biomedical Research Centre, and NIHR Senior Investigator Award.
Collapse
Affiliation(s)
- Stefanie Zorn
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany; German Center for Child and Adolescent Health (DZKJ), partner site Ulm, Ulm, Germany
| | - Cornelis Jan de Groot
- Division for Paediatric Endocrinology and Obesity Center, Department of Paediatrics, Erasmus MC-Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, Netherlands; Department of Paediatrics, IJsselland Hospital, Capelle aan den Ijssel, Netherlands
| | - Stephanie Brandt-Heunemann
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany; German Center for Child and Adolescent Health (DZKJ), partner site Ulm, Ulm, Germany
| | - Julia von Schnurbein
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Ozair Abawi
- Division for Paediatric Endocrinology and Obesity Center, Department of Paediatrics, Erasmus MC-Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Rebecca Bounds
- Wellcome-MRC Institute of Metabolic Science and NIHR Cambridge Biomedical Research Centre, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Lisa Ruck
- Department of Paediatric Endocrinology and Diabetology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Blanca Guijo
- Departments of Pediatrics & Pediatric Endocrinology, Research Institute "La Princesa", Hospital Infantil Universitario Niño Jesús, Universidad Autónoma de Madrid, Madrid, Spain
| | - Gabriel Á Martos-Moreno
- Departments of Pediatrics & Pediatric Endocrinology, Research Institute "La Princesa", Hospital Infantil Universitario Niño Jesús, Universidad Autónoma de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y NutriciÓn, Instituto de Salud Carlos III, Madrid, Spain
| | - Clarisse Nicaise
- Sorbonne University, Reference Centre for Rare Diseases, Pediatric Nutrition and Gastroenterology Department, Assistance Publique-Hôpitaux de Paris, Trousseau Hospital, Paris, France
| | - Sophie Courbage
- Sorbonne University, Reference Centre for Rare Diseases, Pediatric Nutrition and Gastroenterology Department, Assistance Publique-Hôpitaux de Paris, Trousseau Hospital, Paris, France
| | | | - Reiner Siebert
- German Center for Child and Adolescent Health (DZKJ), partner site Ulm, Ulm, Germany; Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany
| | - Béatrice Dubern
- Sorbonne University, Reference Centre for Rare Diseases, Pediatric Nutrition and Gastroenterology Department, Assistance Publique-Hôpitaux de Paris, Trousseau Hospital, Paris, France; Sorbonne University INSERM Nutrition and Obesity, Systemic Approaches NutriOmics, Paris, France
| | - Christine Poitou
- Sorbonne University INSERM Nutrition and Obesity, Systemic Approaches NutriOmics, Paris, France; Reference Centre for Rare Diseases, Nutrition Department, Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Paris, France
| | - Karine Clément
- Sorbonne University INSERM Nutrition and Obesity, Systemic Approaches NutriOmics, Paris, France; Reference Centre for Rare Diseases, Nutrition Department, Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Paris, France
| | - Jesús Argente
- Departments of Pediatrics & Pediatric Endocrinology, Research Institute "La Princesa", Hospital Infantil Universitario Niño Jesús, Universidad Autónoma de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y NutriciÓn, Instituto de Salud Carlos III, Madrid, Spain; IMDEA Food Institute, Madrid, Spain
| | - Peter Kühnen
- Department of Paediatric Endocrinology and Diabetology, Charité Universitätsmedizin Berlin, Berlin, Germany; German Center for Child and Adolescent Health (DZKJ), partner site Berlin, Berlin, Germany
| | - Ismaa Sadaf Farooqi
- Wellcome-MRC Institute of Metabolic Science and NIHR Cambridge Biomedical Research Centre, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Martin Wabitsch
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany; German Center for Child and Adolescent Health (DZKJ), partner site Ulm, Ulm, Germany.
| | - Erica van den Akker
- Division for Paediatric Endocrinology and Obesity Center, Department of Paediatrics, Erasmus MC-Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
34
|
O'Leary TJ, O'Leary BJ, O'Leary DP. A Perspective on Artificial Intelligence for Molecular Pathologists. J Mol Diagn 2025; 27:323-335. [PMID: 39954999 DOI: 10.1016/j.jmoldx.2025.01.005] [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: 06/19/2024] [Revised: 01/06/2025] [Accepted: 01/17/2025] [Indexed: 02/17/2025] Open
Abstract
The widespread adoption of next-generation sequencing technology in molecular pathology has enabled us to interrogate the genome as never before. The huge quantities of data generated by sequencing, the enormous complexity of human and microbial genetics, and the need for fast answers demand increasing use of automation as we diagnose disease and guide patient treatment. Much of this automation is based on tools that fall under umbrellas that have come to be known as machine learning and artificial intelligence. This review outlines some of the broad ideas that underpin these complex computational methods. It discusses the roles of pathologists and data scientists in generating new tools and factors to keep in mind when adopting these systems for use in molecular pathology. It pays special attention to regulatory and professional society guidance for validating them in individual institutions and to possible sources of bias. Finally, it briefly discusses ongoing efforts in computer science that may dramatically impact artificial intelligence in the future.
Collapse
Affiliation(s)
- Timothy J O'Leary
- Office of Research and Development, Veterans Health Administration, Washington, District of Columbia
| | | | - Dianne P O'Leary
- Department of Computer Science and Institute for Advanced Computing Studies, University of Maryland, College Park, Maryland.
| |
Collapse
|
35
|
Fiorenzani C, Mossa A, De Rubeis S. DEAD/DEAH-box RNA helicases shape the risk of neurodevelopmental disorders. Trends Genet 2025; 41:437-449. [PMID: 39828505 PMCID: PMC12055483 DOI: 10.1016/j.tig.2024.12.006] [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: 10/03/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 01/22/2025]
Abstract
The DEAD/DEAH-box family of RNA helicases (RHs) is among the most abundant and conserved in eukaryotes. These proteins catalyze the remodeling of RNAs to regulate their splicing, stability, localization, and translation. Rare genetic variants in DEAD/DEAH-box proteins have recently emerged as being associated with neurodevelopmental disorders (NDDs). Analyses in cellular and animal models have uncovered fundamental roles for these proteins during brain development. We discuss the genetic and functional evidence that implicates DEAD/DEAH-box proteins in brain development and NDDs, with a focus on how structural insights from paralogous genes can be leveraged to advance our understanding of the pathogenic mechanisms at play.
Collapse
Affiliation(s)
- Chiara Fiorenzani
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Alper Center for Neural Development and Regeneration, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Adele Mossa
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Alper Center for Neural Development and Regeneration, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Silvia De Rubeis
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Alper Center for Neural Development and Regeneration, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
36
|
Saparov A, Zech M. Big data and transformative bioinformatics in genomic diagnostics and beyond. Parkinsonism Relat Disord 2025; 134:107311. [PMID: 39924354 DOI: 10.1016/j.parkreldis.2025.107311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/23/2025] [Accepted: 01/25/2025] [Indexed: 02/11/2025]
Abstract
The current era of high-throughput analysis-driven research offers invaluable insights into disease etiologies, accurate diagnostics, pathogenesis, and personalized therapy. In the field of movement disorders, investigators are facing an increasing growth in the volume of produced patient-derived datasets, providing substantial opportunities for precision medicine approaches based on extensive information accessibility and advanced annotation practices. Integrating data from multiple sources, including phenomics, genomics, and multi-omics, is crucial for comprehensively understanding different types of movement disorders. Here, we explore formats and analytics of big data generated for patients with movement disorders, including strategies to meaningfully share the data for optimized patient benefit. We review computational methods that are essential to accelerate the process of evaluating the increasing amounts of specialized data collected. Based on concrete examples, we highlight how bioinformatic approaches facilitate the translation of multidimensional biological information into clinically relevant knowledge. Moreover, we outline the feasibility of computer-aided therapeutic target evaluation, and we discuss the importance of expanding the focus of big data research to understudied phenotypes such as dystonia.
Collapse
Affiliation(s)
- Alice Saparov
- Institute of Human Genetics, Technical University of Munich, School of Medicine and Health, Munich, Germany; Institute of Neurogenomics, Helmholtz Munich, Neuherberg, Germany; Institute for Advanced Study, Technical University of Munich, Garching, Germany
| | - Michael Zech
- Institute of Human Genetics, Technical University of Munich, School of Medicine and Health, Munich, Germany; Institute of Neurogenomics, Helmholtz Munich, Neuherberg, Germany; Institute for Advanced Study, Technical University of Munich, Garching, Germany.
| |
Collapse
|
37
|
Holleman AM, Deaton AM, Hoffing RA, Krohn L, LoGerfo P, Nioi P, Plekan ME, Akle Serrano S, Ticau S, Walshe TE, Borodovsky A, Ward LD. Rare predicted loss-of-function and damaging missense variants in CFHR5 associate with protection from age-related macular degeneration. Am J Hum Genet 2025; 112:1062-1080. [PMID: 40250423 DOI: 10.1016/j.ajhg.2025.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 03/19/2025] [Accepted: 03/21/2025] [Indexed: 04/20/2025] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of blindness among older adults worldwide, but treatment options are limited. Genetics studies have implicated the CFH locus, containing CFH and five CFHR genes, CFHR1-5, in AMD. While CFH has been robustly linked with AMD risk, potential additional roles for the CFHR genes remain unclear, obscured by strong linkage disequilibrium across the locus. Investigating rare coding variants can help to identify causal genes in such regions. We used whole-exome sequencing data from 406,952 UK Biobank participants to examine AMD associations with genes at the CFH locus. For each gene, we used burden testing to examine associations of rare (minor-allele frequency [MAF] < 1%) predicted loss-of-function (pLoF) and predicted damaging missense variants with AMD. We considered "broadly defined AMD" (ICD-10 35.3; ncases = 10,700) and "strictly defined AMD" (dry or wet AMD; ncases = 346). Adjusting for CFH-region variants known to independently associate with AMD, we find that CFHR5 rare variant burden significantly associates with a decreased risk of broadly defined AMD (odds ratio [OR] = 0.75, p = 7 × 10-4), with this association primarily driven by pLoF variants. Furthermore, the association of CFHR5 rare variants with AMD protection is estimated to be stronger for individuals with the CFH rs1061170 AMD risk allele (p.Tyr402His [p.Y402H]; interaction p = 0.04). Corresponding analyses of strict AMD were underpowered. However, we observe that thinning of the photoreceptor layer outer segment strongly predicts strict AMD and find that CFHR5 rare variant burden is significantly associated with increased thickness of this retinal layer (+0.34 SD, p = 4 × 10-4, n = 45,365). These findings suggest CFHR5 inhibition as a potential therapeutic approach for AMD.
Collapse
Affiliation(s)
| | | | | | - Lynne Krohn
- Alnylam Pharmaceuticals, Cambridge, MA 02142, USA
| | | | - Paul Nioi
- Alnylam Pharmaceuticals, Cambridge, MA 02142, USA
| | | | | | - Simina Ticau
- Alnylam Pharmaceuticals, Cambridge, MA 02142, USA
| | | | | | - Lucas D Ward
- Alnylam Pharmaceuticals, Cambridge, MA 02142, USA
| |
Collapse
|
38
|
Varga MJ, Richardson ME, Chamberlin A. Structural biology in variant interpretation: Perspectives and practices from two studies. Am J Hum Genet 2025; 112:984-992. [PMID: 40233741 DOI: 10.1016/j.ajhg.2025.03.010] [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: 10/28/2024] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 04/17/2025] Open
Abstract
Structural biology offers a powerful lens through which to assess genetic variants by providing insights into their impact on clinically relevant protein structure and function. Due to the availability of new, user-friendly, web-based tools, structural analyses by wider audiences have become more mainstream. These new tools, including AlphaMissense and AlphaFold, have recently been in the limelight due to their initial success and projected future promise; however, the intricacies and limitations of using these tools still need to be disseminated to the more general audience that is likely to use them in variant analysis. Here, we expound on frameworks applying structural biology to variant interpretation by examining two accompanying articles. To this end, we explore the nuances of choosing the correct protein model, compare and contrast various structural approaches, and highlight both the advantages and limitations of employing structural biology in variant interpretation. Using two articles published in this issue of The American Journal of Human Genetics as a baseline, we focus on case studies in TP53 and BRCA1 to illuminate gene-specific differences in the applications of structural information, which illustrate the complexities inherent in this field. Additionally, we discuss the implications of recent advancements, such as AlphaFold, and provide practical guidance for researchers navigating variant interpretation using structural biology.
Collapse
|
39
|
Moyer AM, Black JL. Pharmacogenomic Testing in the Clinical Laboratory: Historical Progress and Future Opportunities. Ann Lab Med 2025; 45:247-258. [PMID: 40170583 PMCID: PMC11996682 DOI: 10.3343/alm.2024.0652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/02/2025] [Accepted: 03/04/2025] [Indexed: 04/03/2025] Open
Abstract
Pharmacogenomics is a rapidly evolving field with a strong foundation in basic science dating back to 1960. Pharmacogenomic findings have been translated into clinical care through collaborative efforts of clinical practitioners, pharmacists, clinical laboratories, and research groups. The methods used have transitioned from targeted genotyping of relatively few variants in individual genes to multiplexed multi-gene panels, and sequencingbased methods are likely on the horizon; however, no system exists for classifying and reporting rare variants identified via sequencing-based approaches. Laboratory testing in pharmacogenomics is complex for several genes, including cytochrome P450 2D6 (CYP2D6), HLA-A, and HLA-B , owing to a high degree of polymorphisms, homology with other genes, and copy-number variation. These loci require specialized methods and familiarity with each gene, which may persist during the transition to next-generation sequencing. Increasing implementation across laboratories and clinical facilities has required cooperative efforts to develop standard testing targets, nomenclature, and reporting practices and guidelines for applying the results clinically. Beyond standardization, harmonization between pharmacogenomics and the broader field of genomic medicine may be essential for facilitating further adoption and realizing the full potential of personalized medicine. In this review, we describe the evolution of clinical laboratory testing for pharmacogenomics, including standardization efforts and the anticipated transition from targeted genotyping to sequencing-based pharmacogenomics. We speculate on potential upcoming developments, including pharmacoepigenetics, improved understanding of the impact of non-coding variants, use of large-scale functional genomics to characterize rare variants, and a renewed interest in polygenic risk or combinatorial approaches, which will drive the progression of the field.
Collapse
Affiliation(s)
- Ann M. Moyer
- Division of Laboratory Genetics and Genomics, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - John L. Black
- Division of Laboratory Genetics and Genomics, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
40
|
Hacisuleyman A, Gul A, Erman B. Role of Mutual Information Profile Shifts in Assessing the Pathogenicity of Mutations on Protein Functions: The Case of Pyrin Variants Associated With Familial Mediterranean Fever. Proteins 2025; 93:1035-1053. [PMID: 39739522 DOI: 10.1002/prot.26795] [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: 08/14/2024] [Revised: 12/16/2024] [Accepted: 12/19/2024] [Indexed: 01/02/2025]
Abstract
This study presents a novel method to assess the pathogenicity of pyrin protein mutations by using mutual information (MI) as a measure to quantify the correlation between residue motions or fluctuations and associated changes affecting the phenotype. The concept of MI profile shift is presented to quantify changes in MI upon mutation, revealing insights into residue-residue interactions at critical positions. We apply this method to the pyrin protein variants, which are associated with an autosomal recessively inherited disease called familial Mediterranean fever (FMF) since the available tools do not help predict the pathogenicity of the most penetrant variants. We demonstrate the utility of MI profile shifts in assessing the effects of mutations on protein stability, function, and disease phenotype. The importance of MI shifts, particularly the negative shifts observed in the pyrin example, as indicators of severe functional effects is emphasized. Additionally, the exploration of potential compensatory mechanisms suggested by positive MI shifts, which are otherwise random and inconsequential, is highlighted. The study also discusses challenges in relating MI profile changes to disease severity and advocates for comprehensive analysis considering genetic, environmental, and stochastic factors. Overall, this study provides insights into the molecular mechanisms underlying the pathogenesis of FMF and offers a framework for identifying potential therapeutic targets based on MI profile changes induced by mutations.
Collapse
Affiliation(s)
- Aysima Hacisuleyman
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Chemical and Biological Engineering, Koc University, Istanbul, Turkey
| | - Ahmet Gul
- Division of Rheumatology, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Burak Erman
- Chemical and Biological Engineering, Koc University, Istanbul, Turkey
| |
Collapse
|
41
|
Rotenberg N, Fortuno C, Varga MJ, Chamberlin AC, Ramadane-Morchadi L, Feng BJ, de la Hoya M, Richardson ME, Spurdle AB. Integration of protein stability and AlphaMissense scores improves bioinformatic impact prediction for p53 missense and in-frame amino acid deletion variants. Am J Hum Genet 2025; 112:1003-1014. [PMID: 40233742 DOI: 10.1016/j.ajhg.2025.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 01/05/2025] [Accepted: 01/13/2025] [Indexed: 04/17/2025] Open
Abstract
The clinical classification of germline missense variants and single-amino-acid deletions is challenging. The BayesDel and Align-GVGD bioinformatic prediction tools currently used for ClinGen TP53 variant curation expert panel (VCEP) classification do not directly capture changes in protein folding stability, measured using computed destabilization energies (ΔΔG scores). The AlphaMissense tool recently developed by Google DeepMind to predict pathogenicity for all human proteome missense variants is trained in part using AlphaFold2 architecture. Our study investigated whether protein folding stability and/or AlphaMissense scores could improve impact prediction for p53 missense and single-amino-acid deletion variants. ΔΔG scores were calculated for missense variants using FoldX and for single-amino-acid deletions using an AlphaFold2/RosettaRelax protocol. Residue surface exposure was categorized using relative solvent accessibility (RSA) measures. The predictive values of ΔΔG scores, AlphaMissense, BayesDel, and Align-GVGD were examined using Boruta and binary logistic regression based on functionally defined reference sets. The likelihood ratio (LR) toward pathogenicity was estimated and used to refine optimal categories for predicting variant pathogenicity for different RSA values. We showed that current VCEP predictive approaches for missense variants were improved by integrating ΔΔG scores ≥2.5 kcal/mol for partially buried and buried residues, but better performance was achieved using AlphaMissense with ΔΔG and RSA. For deletion variants, ΔΔG scores ≥4.8 Rosetta energy unit (REU) in buried residues outperformed currently used predictive approaches. Future TP53 VCEP specifications for p53 missense impact prediction may consider AlphaMissense, ΔΔG score, and RSA combined for substitution variants and ΔΔG score alone for deletion variants.
Collapse
Affiliation(s)
- Nitsan Rotenberg
- Molecular Cancer Epidemiology Laboratory, QIMR Berghofer MRI, Herston, QLD 4006, Australia; University of Queensland, Brisbane, QLD, Australia
| | - Cristina Fortuno
- Molecular Cancer Epidemiology Laboratory, QIMR Berghofer MRI, Herston, QLD 4006, Australia
| | | | | | - Lobna Ramadane-Morchadi
- Molecular Oncology Laboratory, Hospital Clínico San Carlos, IdISSC (Instituto de Investigación Sanitaria del Hospital Clínico San Carlos), 28040 Madrid, Spain
| | - Bing-Jian Feng
- University of Utah Department of Dermatology, Salt Lake City, UT, USA; University of Utah Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Miguel de la Hoya
- Molecular Oncology Laboratory, Hospital Clínico San Carlos, IdISSC (Instituto de Investigación Sanitaria del Hospital Clínico San Carlos), 28040 Madrid, Spain
| | | | - Amanda B Spurdle
- Molecular Cancer Epidemiology Laboratory, QIMR Berghofer MRI, Herston, QLD 4006, Australia; University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
42
|
Lopes-Pacheco M, Winters AG, Jackson JJ, Olson Rd JA, Kim M, Ledwitch KV, Tedman A, Jhangiani AR, Schlebach JP, Meiler J, Plate L, Oliver KE. Recent developments in cystic fibrosis drug discovery: where are we today? Expert Opin Drug Discov 2025; 20:659-682. [PMID: 40202089 DOI: 10.1080/17460441.2025.2490250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 03/17/2025] [Accepted: 04/03/2025] [Indexed: 04/10/2025]
Abstract
INTRODUCTION The advent of variant-specific disease-modifying drugs into clinical practice has provided remarkable benefits for people with cystic fibrosis (PwCF), a multi-organ life-limiting inherited disease. However, further efforts are needed to maximize therapeutic benefits as well as to increase the number of PwCF taking CFTR modulators. AREA COVERED The authors discuss some of the key limitations of the currently available CFTR modulator therapies (e.g. adverse reactions) and strategies in development to increase the number of available therapeutics for CF. These include novel methods to accelerate theratyping and identification of novel small molecules and cellular targets representing alternative or complementary therapies for CF. EXPERT OPINION While the CF therapy development pipeline continues to grow, there is a critical need to optimize strategies that will accelerate testing and approval of effective therapies for (ultra)rare CFTR variants as traditional assays and trials are not suitable to address such issues. Another major barrier that needs to be solved is the restricted access to currently available modulator therapies, which remains a significant burden for PwCF who are from racial and ethnic minorities and/or living in underprivileged regions.
Collapse
Affiliation(s)
- Miquéias Lopes-Pacheco
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Center for Cystic Fibrosis & Airways Disease Research, Emory University & Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Ashlyn G Winters
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Center for Cystic Fibrosis & Airways Disease Research, Emory University & Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - JaNise J Jackson
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Center for Cystic Fibrosis & Airways Disease Research, Emory University & Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - John A Olson Rd
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
- Program in Chemical and Physical Biology, Vanderbilt University, Nashville, TN, USA
| | - Minsoo Kim
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
- Program in Chemical and Physical Biology, Vanderbilt University, Nashville, TN, USA
| | - Kaitlyn V Ledwitch
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
- Center for Structural Biology, Vanderbilt University, Nashville, TN, USA
| | - Austin Tedman
- The James Tarpo Junior & Margaret Tarpo Department of Chemistry, Purdue University, West Lafayette, IN, USA
| | - Ashish R Jhangiani
- The James Tarpo Junior & Margaret Tarpo Department of Chemistry, Purdue University, West Lafayette, IN, USA
| | - Jonathan P Schlebach
- The James Tarpo Junior & Margaret Tarpo Department of Chemistry, Purdue University, West Lafayette, IN, USA
| | - Jens Meiler
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
- Center for Structural Biology, Vanderbilt University, Nashville, TN, USA
| | - Lars Plate
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Kathryn E Oliver
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Center for Cystic Fibrosis & Airways Disease Research, Emory University & Children's Healthcare of Atlanta, Atlanta, GA, USA
| |
Collapse
|
43
|
Merepa SS, Reis LM, Damián A, Bardakjian T, Schneider A, Trujillo-Tiebas MJ, Ayuso C, Galarza LC, Saez Villaverde R, Ortiz-Cabrera NV, Bax DA, Holt R, Ceroni F, Edery P, Grelet M, Riccardi F, Maillard L, Costakos D, Plaisancié J, Chassaing N, Corton M, Semina EV, Ragge NK. GJA8-associated developmental eye disorders: a new multicentre study highlights mutational hotspots and genotype-phenotype correlations. Eur J Hum Genet 2025:10.1038/s41431-025-01843-8. [PMID: 40301690 DOI: 10.1038/s41431-025-01843-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/20/2025] [Accepted: 03/25/2025] [Indexed: 05/01/2025] Open
Abstract
Variants in gap junction protein alpha 8 (GJA8), the gene encoding connexin 50 (Cx50), are primarily associated with developmental cataract, although some are associated with severe structural eye anomalies, such as aphakia (absent lens), microphthalmia (small eyes), and sclerocornea. To further define the relationship of GJA8 variants to ocular developmental disorders, we screened four large international cohorts with structural eye anomalies, including anophthalmia, microphthalmia, and coloboma (AMC) or cataracts. We identified 15 new families carrying 14 different heterozygous GJA8 variants (12 missense variants and two 1q21 microdeletions). The missense variants comprised 10 previously reported alterations in cases with eye anomalies [p.(Gly22Ser), p.(Val44Met), p.(Asp67Gly), p.(Arg76Cys), p.(Pro88Leu), p.(Gly94Glu), p.(Gly94Arg), p.(His98Arg), p.(Pro189Ser), and p.(Arg198Trp)] and two not yet linked with disease [p.(Thr39Met) and p.(Tyr66Asp)]. Their associated phenotypes ranged from isolated cataracts to a combination of microphthalmia and cataract with/without sclerocornea. Our study confirms GJA8 variants as an important source of genetic diagnoses for families with structural eye anomalies in addition to cataract and highlights specific mutational hotspots. Furthermore, we confirm an important genotype-phenotype correlation between sclerocornea and the p.(Gly94Arg) variant, and detail intra- and inter-familial phenotypic variability, which is important for clinical assessment and genetic counselling.
Collapse
Affiliation(s)
- Solomon S Merepa
- Faculty of Health, Science and Technology, School of Biological and Medical Sciences, Oxford Brookes University, Headington Campus, Gipsy Lane, Oxford, UK
| | - Linda M Reis
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Alejandra Damián
- Department of Genetics & Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital - Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- U704 Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | | | - Adele Schneider
- Einstein Medical Center Philadelphia, Philadelphia, PA, USA
- Wills Eye Hospital, Philadelphia, PA, USA
| | - María Jose Trujillo-Tiebas
- Department of Genetics & Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital - Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- U704 Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Carmen Ayuso
- Department of Genetics & Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital - Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- U704 Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | | | | | | | - Dorine A Bax
- Faculty of Health, Science and Technology, School of Biological and Medical Sciences, Oxford Brookes University, Headington Campus, Gipsy Lane, Oxford, UK
| | - Richard Holt
- Faculty of Health, Science and Technology, School of Biological and Medical Sciences, Oxford Brookes University, Headington Campus, Gipsy Lane, Oxford, UK
| | - Fabiola Ceroni
- Faculty of Health, Science and Technology, School of Biological and Medical Sciences, Oxford Brookes University, Headington Campus, Gipsy Lane, Oxford, UK
| | - Patrick Edery
- Université Claude Bernard Lyon 1, INSERM, CNRS, Centre de Recherche en Neurosciences de Lyon CRNL U1028 UMR5292, Genetics of Neurodevelopment Team, Bron, France
- Department of Genetics, Clinical Genetics Unit, Centre de Référence Maladies Rares des Anomalies du Développement, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, Bron, France
| | - Maude Grelet
- Centre Hospitalier Intercommunal de Toulon- La Seyne sur mer, Service de Génétique Médicale, Toulon, France
| | - Florence Riccardi
- Centre Hospitalier Intercommunal de Toulon- La Seyne sur mer, Service de Génétique Médicale, Toulon, France
| | - Lauriane Maillard
- Service d'Opthalmologie, Hôpital Purpan, CHU Toulouse, Toulouse, France
| | - Deborah Costakos
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Julie Plaisancié
- Laboratoire de Référence (LBMR) des anomalies malformatives de l'œil, Institut Fédératif de Biologie (IFB), CHU Toulouse, Toulouse, France
- Centre de Référence des Affections Rares en Génétique Ophtalmologique CARGO, Site Constitutif, CHU Toulouse, Toulouse, France
- Molecular, Cellular and Developmental Biology Unit (MCD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Nicolas Chassaing
- Laboratoire de Référence (LBMR) des anomalies malformatives de l'œil, Institut Fédératif de Biologie (IFB), CHU Toulouse, Toulouse, France
- Centre de Référence des Affections Rares en Génétique Ophtalmologique CARGO, Site Constitutif, CHU Toulouse, Toulouse, France
| | - Marta Corton
- Department of Genetics & Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital - Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- U704 Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Elena V Semina
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Nicola K Ragge
- Faculty of Health, Science and Technology, School of Biological and Medical Sciences, Oxford Brookes University, Headington Campus, Gipsy Lane, Oxford, UK.
- West Midlands Regional Clinical Genetics Service and Birmingham Health Partners, Birmingham Women's and Children's Foundation Trust, Birmingham, UK.
| |
Collapse
|
44
|
Qiu X, Wang S, Li C, Wang Y. Expression and immunological role of FUNDC2 in pan-cancer. PLoS One 2025; 20:e0319343. [PMID: 40294153 PMCID: PMC12036908 DOI: 10.1371/journal.pone.0319343] [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: 11/17/2024] [Accepted: 01/30/2025] [Indexed: 04/30/2025] Open
Abstract
FUNDC2 is a novel mitochondrial protein and is highly involved in various cancers. However, expression pattern and possible role and mechanism of FUNDC2 in pan-cancer remain to be investigated. TIMER 2.0 was used to investigate the expression patterns and immune infiltration of FUNDC2. GEPIA was applied to study the relationship between level of FUNDC2 and prognosis of the patients with pan-cancer. STRING was employed to analyze the potential interacting proteins of FUNDC2. The phosphorylation sites were predicted by cBioPortal and PhosphoNet. Furthermore, variations of FUNDC2 in cancers were investigated by cBioPortal. Finally, AlphaFold was used to predict the structure of FUNDC2. The data show that there were significant differences in the expression levels of FUNDC2 between cancer tissues and controls. Specifically, the levels of FUNDC2 in 8 cancers were significantly lower than the respective controls. The survival time of the cancer patients with higher levels of FUNDC2 was longer than that of lower FUNDC2 in most different types of cancers. The pattern of FUNDC2 was significantly related to immune infiltration of B cells of cancer patients. STRING analysis revealed that FUNDC2 can interact with FUNDC1, et al. Fifteen phosphorylation sites were predicted by PhosphoNet and cBioPortal, of which the S167 also overlapped with the mutation sites of FUNDC2. These data collectively show that the mitochondrial protein FUNDC2 may serve as a possible prognostic biomarker across various cancers and the mechanism may include immune infiltration.
Collapse
Affiliation(s)
- Xirong Qiu
- Department of Pharmacology, School of Medicine, Lijiang Culture and Tourism College, Lijiang, Yunnan, China
| | - Shuyu Wang
- Department of Pharmacology, School of Medicine, Lijiang Culture and Tourism College, Lijiang, Yunnan, China
| | - Chenlu Li
- Department of Pharmacology, School of Medicine, Lijiang Culture and Tourism College, Lijiang, Yunnan, China
| | - Yinan Wang
- Department of Pharmacology, School of Medicine, Lijiang Culture and Tourism College, Lijiang, Yunnan, China
| |
Collapse
|
45
|
Visibelli A, Finetti R, Niccolai P, Trezza A, Spiga O, Santucci A, Niccolai N. Profiling of Protein-Coding Missense Mutations in Mendelian Rare Diseases: Clues from Structural Bioinformatics. Int J Mol Sci 2025; 26:4072. [PMID: 40362311 PMCID: PMC12071383 DOI: 10.3390/ijms26094072] [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: 04/03/2025] [Revised: 04/23/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
The growing availability of protein structural data from experimental methods and accurate predictive models provides the opportunity to investigate the molecular origins of rare diseases (RDs) reviewed in the Orpha.net database. In this study, we analyzed the topology of 5728 missense mutation sites involved in Mendelian RDs (MRDs), forming the basis of our structural bioinformatics investigation. Each mutation site was characterized by side-chain position within the overall 3D protein structure and side-chain orientation. Atom depth quantitation, achieved by using SADIC v2.0, allowed the classification of all the mutation sites listed in our database. Particular attention was given to mutations where smaller amino acids replaced bulky, outward-oriented residues in the outer structural layers. Our findings reveal that structural features that could lead to the formation of void spaces in the outer protein region are very frequent. Notably, we identified 722 cases where MRD-associated mutations could generate new surface pockets with the potential to accommodate pharmaceutical ligands. Molecular dynamics (MD) simulations further supported the prevalence of cryptic pocket formation in a subset of drug-binding protein candidates, underscoring their potential for structure-based drug discovery in RDs.
Collapse
Affiliation(s)
- Anna Visibelli
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy; (A.V.); (R.F.); (A.T.); (O.S.); (N.N.)
| | - Rebecca Finetti
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy; (A.V.); (R.F.); (A.T.); (O.S.); (N.N.)
| | - Piero Niccolai
- Le Ricerche del BarLume Free Association, Ville di Corsano, Monteroni d’Arbia, 53014 Siena, Italy;
| | - Alfonso Trezza
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy; (A.V.); (R.F.); (A.T.); (O.S.); (N.N.)
| | - Ottavia Spiga
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy; (A.V.); (R.F.); (A.T.); (O.S.); (N.N.)
- Industry 4.0 Competence Center ARTES 4.0 Viale Rinaldo Piaggio, 56025 Pontedera, Italy
| | - Annalisa Santucci
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy; (A.V.); (R.F.); (A.T.); (O.S.); (N.N.)
- Industry 4.0 Competence Center ARTES 4.0 Viale Rinaldo Piaggio, 56025 Pontedera, Italy
| | - Neri Niccolai
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy; (A.V.); (R.F.); (A.T.); (O.S.); (N.N.)
- Le Ricerche del BarLume Free Association, Ville di Corsano, Monteroni d’Arbia, 53014 Siena, Italy;
| |
Collapse
|
46
|
Harrer P, Krygier M, Krenn M, Kittke V, Danis M, Krastev G, Saparov A, Pichon V, Malbos M, Scherer C, Dzinovic I, Skorvanek M, Kopajtich R, Prokisch H, Silvaieh S, Grisold A, Mazurkiewicz-Bełdzińska M, de Sainte Agathe JM, Winkelmann J, Necpal J, Jech R, Zech M. Expanding the Allelic and Clinical Heterogeneity of Movement Disorders Linked to Defects of Mitochondrial Adenosine Triphosphate Synthase. Mov Disord 2025. [PMID: 40276935 DOI: 10.1002/mds.30209] [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: 01/15/2025] [Revised: 03/13/2025] [Accepted: 03/17/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Defects of mitochondrial ATP synthase (ATPase) represent an emerging, yet incompletely understood group of neurodevelopmental diseases with abnormal movements. OBJECTIVE The aim of this study was to redefine the phenotypic and mutational spectrum of movement disorders linked to the ATPase subunit-encoding genes ATP5F1A and ATP5F1B. METHODS We recruited regionally distant patients who had been genome or exome sequenced. Fibroblast cultures from two patients were established to perform RNA sequencing, immunoblotting, mass spectrometry-based high-throughput quantitative proteomics, and ATPase activity assays. In silico three-dimensional missense variant modeling was performed. RESULTS We identified a patient with developmental delay, myoclonic dystonia, and spasticity who carried a heterozygous frameshift c.1404del (p.Glu469Serfs*3) variant in ATP5F1A. The patient's cells exhibited significant reductions in ATP5F1A mRNA, underexpression of the α-subunit of ATPase in association with other aberrantly expressed ATPase components, and compromised ATPase activity. In addition, a novel deleterious heterozygous ATP5F1A missense c.1252G>A (p.Gly418Arg) variant was discovered, shared by three patients from two families with hereditary spastic paraplegia (HSP). This variant mapped to a functionally important intersubunit communication site. A third heterozygous variant, c.1074+1G>T, affected a canonical donor splice site of ATP5F1B and resulted in exon skipping with significantly diminished ATP5F1B mRNA levels, as well as impaired ATPase activity. The associated phenotype consisted of cerebral palsy (CP) with prominent generalized dystonia. CONCLUSIONS Our data confirm and expand the role of dominant ATP5F1A and ATP5F1B variants in neurodevelopmental movement disorders. ATP5F1A/ATP5F1B-related ATPase diseases should be considered as a cause of dystonia, HSP, and CP. © 2025 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Philip Harrer
- Institute of Human Genetics, School of Medicine and Health, Technical University of Munich, Munich, Germany
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
| | - Magdalena Krygier
- Department of Developmental Neurology, Medical University of Gdansk, Gdansk, Poland
| | - Martin Krenn
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences & Mental Health, Medical University of Vienna, Vienna, Austria
| | - Volker Kittke
- Institute of Human Genetics, School of Medicine and Health, Technical University of Munich, Munich, Germany
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
| | - Martin Danis
- Neurological Clinic of Faculty Hospital Trnava and Slovak Health University Bratislava, Bratislava, Slovakia
| | - Georgi Krastev
- Neurological Clinic of Faculty Hospital Trnava and Slovak Health University Bratislava, Bratislava, Slovakia
| | - Alice Saparov
- Institute of Human Genetics, School of Medicine and Health, Technical University of Munich, Munich, Germany
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
- Institute for Advanced Study, Technical University of Munich, Garching, Germany
| | - Virginie Pichon
- CRMR Neurogenetique, Service de Neurologie, Centre Hospitalier, Universitaire d'Angers, Angers, France
| | - Marlène Malbos
- CRMRs "Anomalies du Développement et syndromes malformatifs" et "Déficiences Intellectuelles de causes rares," FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
- Laboratoire de Génomique Médicale, UF Innovation en diagnostic génomique des maladies rares, CHU Dijon Bourgogne, Dijon, France
| | - Clarisse Scherer
- CRMR Neurogenetique, Service de Neurologie, Centre Hospitalier, Universitaire d'Angers, Angers, France
| | - Ivana Dzinovic
- Institute of Human Genetics, School of Medicine and Health, Technical University of Munich, Munich, Germany
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
| | - Matej Skorvanek
- Department of Neurology, P.J. Safarik University, Kosice, Slovakia
- Department of Neurology, University Hospital of L. Pasteur, Kosice, Slovakia
| | - Robert Kopajtich
- Institute of Human Genetics, School of Medicine and Health, Technical University of Munich, Munich, Germany
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
| | - Holger Prokisch
- Institute of Human Genetics, School of Medicine and Health, Technical University of Munich, Munich, Germany
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
| | - Sara Silvaieh
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences & Mental Health, Medical University of Vienna, Vienna, Austria
| | - Anna Grisold
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences & Mental Health, Medical University of Vienna, Vienna, Austria
| | | | - Jean-Madeleine de Sainte Agathe
- Department of Medical Genetics, Sorbonne Université, AP-HP Sorbonne Université, Paris, France
- Laboratoire de Biologie Médicale Multi-Site SeqOIA, Sorbonne Université, Paris, France
| | - Juliane Winkelmann
- Institute of Human Genetics, School of Medicine and Health, Technical University of Munich, Munich, Germany
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
- DZPG (German Center for Mental Health), Munich, Germany
- Munich Cluster for Systems Neurology, SyNergy, Munich, Germany
| | - Jan Necpal
- Department of Neurology, Zvolen Hospital, Zvolen, Slovakia
- Parkinsonism and Movement Disorders Treatment Center, Zvolen Hospital, Zvolen, Slovakia
| | - Robert Jech
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Michael Zech
- Institute of Human Genetics, School of Medicine and Health, Technical University of Munich, Munich, Germany
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
- Institute for Advanced Study, Technical University of Munich, Garching, Germany
| |
Collapse
|
47
|
Brumage L, Best S, Hippe DS, Grunblatt E, Chanana P, Wu F, Lee MC, Ying Z, Ibrahim A, Chung JH, Vigil A, Fatherree J, Beronja S, Paddison P, Sullivan L, Nabet B, MacPherson D. In vivo functional screens reveal KEAP1 loss as a driver of chemoresistance in small cell lung cancer. SCIENCE ADVANCES 2025; 11:eadq7084. [PMID: 40267200 PMCID: PMC12017300 DOI: 10.1126/sciadv.adq7084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 03/18/2025] [Indexed: 04/25/2025]
Abstract
Exquisitely chemosensitive initially, small cell lung cancer (SCLC) exhibits dismal outcomes owing to rapid transition to chemoresistance. Elucidating the genetic underpinnings has been challenging owing to limitations with cellular models. As SCLC patient-derived xenograft (PDX) models mimic therapeutic responses, we perform genetic screens in chemosensitive PDX models to identify drivers of chemoresistance. cDNA overexpression screens identify MYC, MYCN, and MYCL, while CRISPR deletion screens identify KEAP1 loss as driving chemoresistance. Deletion of KEAP1 switched a chemosensitive SCLC PDX model to become chemoresistant and resulted in sensitivity to inhibition of glutamine metabolism. Data from the IMpower133 clinical trial revealed ~6% of patients with extensive-stage SCLC exhibit KEAP1 genetic alterations, with activation of a KEAP1/NRF2 transcriptional signature associated with reduced survival upon chemotherapy treatment. While roles for KEAP1/NRF2 have been unappreciated in SCLC, our genetic screens revealed KEAP1 loss as a driver of chemoresistance, while patient genomic analyses demonstrate clinical importance.
Collapse
Affiliation(s)
- Lauren Brumage
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Molecular and Cellular Biology Program, University of Washington Seattle, Seattle, WA, USA
| | - Scott Best
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Molecular and Cellular Biology Program, University of Washington Seattle, Seattle, WA, USA
| | - Daniel S. Hippe
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Eli Grunblatt
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Pritha Chanana
- Genomics and Bioinformatics Shared Resource, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Feinan Wu
- Genomics and Bioinformatics Shared Resource, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Zhe Ying
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ali Ibrahim
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jae Heun Chung
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Anna Vigil
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jackson Fatherree
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Slobodan Beronja
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Patrick Paddison
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Lucas Sullivan
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - David MacPherson
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| |
Collapse
|
48
|
Thorvaldsdottir B, Mansouri L, Sutton LA, Nadeu F, Meggendorfer M, Parker H, Brieghel C, Laidou S, Moia R, Rossi D, Kotaskova J, Delgado J, Rodríguez-Vicente AE, Benito R, Rigolin GM, Bonfiglio S, Scarfò L, Mattsson M, Davis Z, Baliakas P, Rapado I, Miras F, Martinez-Lopez J, de la Serna J, Hernández Rivas JM, Larráyoz MJ, Calasanz MJ, Smedby KE, Espinet B, Puiggros A, Bullinger L, Bosch F, Tazón-Vega B, Baran-Marszak F, Oscier D, Nguyen-Khac F, Zenz T, Terol MJ, Cuneo A, Hernández-Sánchez M, Pospisilova S, Gaidano G, Niemann CU, Campo E, Strefford JC, Ghia P, Stamatopoulos K, Rosenquist R. ATM aberrations in chronic lymphocytic leukemia: del(11q) rather than ATM mutations is an adverse-prognostic biomarker. Leukemia 2025:10.1038/s41375-025-02615-5. [PMID: 40275070 DOI: 10.1038/s41375-025-02615-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 04/01/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025]
Abstract
Despite the well-established adverse impact of del(11q) in chronic lymphocytic leukemia (CLL), the prognostic significance of somatic ATM mutations remains uncertain. We evaluated the effects of ATM aberrations (del(11q) and/or ATM mutations) on time-to-first-treatment (TTFT) in 3631 untreated patients with CLL, in the context of IGHV gene mutational status and mutations in nine CLL-related genes. ATM mutations were present in 246 cases (6.8%), frequently co-occurring with del(11q) (112/246 cases, 45.5%). ATM-mutated patients displayed a different spectrum of genetic abnormalities when comparing IGHV-mutated (M-CLL) and unmutated (U-CLL) cases: M-CLL was enriched for SF3B1 and NFKBIE mutations, whereas U-CLL showed mutual exclusivity with trisomy 12 and TP53 mutations. Isolated ATM mutations were rare, affecting 1.2% of Binet A patients and <1% of M-CLL cases. While univariable analysis revealed shorter TTFT for Binet A patients with any ATM aberration compared to ATM-wildtype, multivariable analysis identified only del(11q), trisomy 12, SF3B1, and EGR2 mutations as independent prognosticators of shorter TTFT among Binet A patients and within M-CLL and U-CLL subgroups. These findings highlight del(11q), and not ATM mutations, as a key biomarker of increased risk of early progression and need for therapy, particularly in otherwise indolent M-CLL, providing insights into risk-stratification and therapeutic decision-making.
Collapse
Affiliation(s)
- Birna Thorvaldsdottir
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Larry Mansouri
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Lesley-Ann Sutton
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Ferran Nadeu
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | | | - Helen Parker
- Cancer Genomics, School for Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Christian Brieghel
- Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Hematology, Danish Cancer Institute, Copenhagen, Denmark
| | - Stamatia Laidou
- Centre for Research and Technology Hellas, Institute of Applied Biosciences, Thessaloniki, Greece
| | - Riccardo Moia
- Division of Hematology, Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| | - Davide Rossi
- Clinic of Hematology, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Laboratory of Experimental Hematology, Institute of Oncology Research, Universita' della Svizzera Italiana, Bellinzona, Switzerland
| | - Jana Kotaskova
- Department of Internal Medicine, Hematology & Oncology, University Hospital Brno, Brno, Czech Republic
- Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Julio Delgado
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Ana E Rodríguez-Vicente
- Cancer Research Center (IBMCC) CSIC-University of Salamanca, Salamanca, Spain
- Instituto de Investigación Biomédica (IBSAL), Salamanca, Spain
- Department of Hematology, University Hospital of Salamanca, Salamanca, Spain
| | - Rocío Benito
- Cancer Research Center (IBMCC) CSIC-University of Salamanca, Salamanca, Spain
- Instituto de Investigación Biomédica (IBSAL), Salamanca, Spain
- Department of Hematology, University Hospital of Salamanca, Salamanca, Spain
| | - Gian Matteo Rigolin
- Hematology - Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | | | - Lydia Scarfò
- IRCCS Ospedale San Raffaele, Milano, Italy
- Università Vita-Salute San Raffaele, Milano, Italy
| | - Mattias Mattsson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Zadie Davis
- Molecular Pathology Department, University Hospitals Dorset, Bournemouth, UK
| | - Panagiotis Baliakas
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Inmaculada Rapado
- Hospital Universitario 12 Octubre, Madrid, Spain
- Spanish National Cancer Research (CNIO), Madrid, Spain
| | - Fatima Miras
- Hospital Universitario 12 Octubre, Madrid, Spain
| | - Joaquín Martinez-Lopez
- Hospital Universitario 12 Octubre, Madrid, Spain
- Spanish National Cancer Research (CNIO), Madrid, Spain
| | - Javier de la Serna
- Hospital Universitario 12 Octubre, Madrid, Spain
- Spanish National Cancer Research (CNIO), Madrid, Spain
| | - Jesús María Hernández Rivas
- Cancer Research Center (IBMCC) CSIC-University of Salamanca, Salamanca, Spain
- Instituto de Investigación Biomédica (IBSAL), Salamanca, Spain
- Department of Hematology, University Hospital of Salamanca, Salamanca, Spain
| | - María José Larráyoz
- Hematological Diseases Laboratory, CIMA LAB Diagnostics, University of Navarra, 31008 Pamplona, Spain, IdiSNA, Navarra Institute for Health Research, 31008, Pamplona, Spain
| | - María José Calasanz
- Hematological Diseases Laboratory, CIMA LAB Diagnostics, University of Navarra, 31008 Pamplona, Spain, IdiSNA, Navarra Institute for Health Research, 31008, Pamplona, Spain
| | - Karin E Smedby
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Blanca Espinet
- Molecular Cytogenetics Laboratory, Pathology Department, Hospital del Mar and Translational Research on Hematological Neoplasms Group, Hospital del Mar Research Institute (IMIM), Barcelona, Spain
| | - Anna Puiggros
- Molecular Cytogenetics Laboratory, Pathology Department, Hospital del Mar and Translational Research on Hematological Neoplasms Group, Hospital del Mar Research Institute (IMIM), Barcelona, Spain
| | - Lars Bullinger
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Hum-boldt-Universität zu Berlin, Berlin, Germany
| | - Francesc Bosch
- Department of Hematology, Hospital Universitari Vall d'Hebron (HUVH), Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Bárbara Tazón-Vega
- Department of Hematology, Hospital Universitari Vall d'Hebron (HUVH), Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Fanny Baran-Marszak
- Service d'hématologie biologique Hôpital Avicenne Assistance Publique des Hôpitaux de Paris Bobigny France, Bobigny, France
| | - David Oscier
- Molecular Pathology Department, University Hospitals Dorset, Bournemouth, UK
| | - Florence Nguyen-Khac
- Sorbonne Université, Service d'Hématologie Biologique, Hôpital Pitié-Salpêtrière, APHP, Paris, France
| | - Thorsten Zenz
- Department of Oncology and Haematology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Maria Jose Terol
- Department of Hematology, INCLIVA Research Insitute, University of Valencia, Valencia, Spain
| | - Antonio Cuneo
- Hematology - Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - María Hernández-Sánchez
- Cancer Research Center (IBMCC) CSIC-University of Salamanca, Salamanca, Spain
- Instituto de Investigación Biomédica (IBSAL), Salamanca, Spain
- Department of Hematology, University Hospital of Salamanca, Salamanca, Spain
- Department of Biochemistry and Molecular Biology, Pharmacy School, Universidad Complutense de Madrid, Madrid, Spain
| | - Sarka Pospisilova
- Department of Internal Medicine, Hematology & Oncology, University Hospital Brno, Brno, Czech Republic
- Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| | - Carsten U Niemann
- Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Hematology, Danish Cancer Institute, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Elias Campo
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Hospital Clínic of Barcelona, Barcelona, Spain
- Universitat de Barcelona, Barcelona, Spain
| | - Jonathan C Strefford
- Cancer Genomics, School for Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Paolo Ghia
- IRCCS Ospedale San Raffaele, Milano, Italy
- Università Vita-Salute San Raffaele, Milano, Italy
| | - Kostas Stamatopoulos
- Centre for Research and Technology Hellas, Institute of Applied Biosciences, Thessaloniki, Greece
| | - Richard Rosenquist
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.
- Clinical Genetics and Genomics, Karolinska University Hospital, Solna, Sweden.
| |
Collapse
|
49
|
Van Laer C, Lavend'homme R, Baert S, De Wispelaere K, Thys C, Kint C, Noppen S, Peerlinck K, Van Geet C, Schols D, Vanassche T, Labarque V, Verhamme P, Jacquemin M, Freson K. Functional assessment of genetic variants in thrombomodulin detected in patients with bleeding and thrombosis. Blood 2025; 145:1929-1942. [PMID: 39841007 DOI: 10.1182/blood.2024026454] [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: 08/06/2024] [Revised: 11/08/2024] [Accepted: 11/29/2024] [Indexed: 01/23/2025] Open
Abstract
ABSTRACT Thrombomodulin (TM) expressed on endothelial cells regulates coagulation. Specific nonsense variants in the TM gene, THBD, result in high soluble TM levels causing rare bleeding disorders. In contrast, although THBD variants have been associated with venous thromboembolism, this association remains controversial. A multigene panel was used to diagnose 601 patients with inherited bleeding or thrombotic disorders. This resulted in the identification of 8 THBD variants for 6 patients with a thrombotic (C175S, A282P, L433P, P501L, G502R, and P508L) and 2 patients with a bleeding (P260A and T478I) phenotype. These were all classified as variants of uncertain significance, and we here aimed to assess their functional role in coagulation. For this purpose, soluble and cell membrane-bound recombinant TM were produced in Expi293F cells. L433P TM showed a marked decrease in the inhibition of thrombin generation and complete inhibition of protein C and thrombin activatable fibrinolysis inhibitor (TAFI) activation. Soluble C175S TM showed decreased inhibition of thrombin generation and protein C activation, whereas no effect was observed for cell membrane-bound recombinant TM. For the other TM variants, no effect on thrombin generation, protein C, or TAFI activation could be observed. Surface plasmon resonance analysis showed no thrombin-TM binding in the presence of L433P because this residue is located at their interaction site. In conclusion, our study shows the functional effects of L433P TM and potentially C175S TM, which are compatible with an increased thrombosis risk. THBD variants are rare but can be relevant to both bleeding and thrombosis. Functional assays for these variants are critical to understand their roles.
Collapse
Affiliation(s)
- Christine Van Laer
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium
- Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Renaud Lavend'homme
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium
| | - Sarissa Baert
- Center for Human Genetics, University Hospitals Leuven, Leuven, Belgium
| | - Koenraad De Wispelaere
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium
| | - Chantal Thys
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium
| | - Cyrielle Kint
- Center for Human Genetics, University Hospitals Leuven, Leuven, Belgium
| | - Sam Noppen
- Department of Microbiology, Immunology and Transplantation, Laboratory of Virology and Chemotherapy, Rega Institute, University of Leuven, Leuven, Belgium
| | - Kathelijne Peerlinck
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium
| | - Chris Van Geet
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium
- Department of Pediatric Hemato-Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Dominique Schols
- Department of Microbiology, Immunology and Transplantation, Laboratory of Virology and Chemotherapy, Rega Institute, University of Leuven, Leuven, Belgium
| | - Thomas Vanassche
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium
- Department of Cardiovascular Diseases, Thrombosis, Haemostasis, and Vascular Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Veerle Labarque
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium
- Department of Pediatric Hemato-Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Peter Verhamme
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium
- Department of Cardiovascular Diseases, Thrombosis, Haemostasis, and Vascular Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Marc Jacquemin
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium
- Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Kathleen Freson
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium
| |
Collapse
|
50
|
Nitoiu A, Zhang Q, Tavares E, Li JM, Ahmed K, Green-Sanderson K, Rashid M, Morcos SM, Maynes JT, Campos EI, Sheffield VC, Vincent A, Héon E. Defective IFT57 underlies a novel cause of Bardet-Biedl syndrome. Hum Mol Genet 2025:ddaf058. [PMID: 40273360 DOI: 10.1093/hmg/ddaf058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 04/02/2025] [Indexed: 04/26/2025] Open
Abstract
A 29-year-old male presented with rod-cone degeneration leading to legal blindness, post-axial polydactyly, obesity, cognitive impairment, and fatty liver, features suggestive of a clinical diagnosis of Bardet-Biedl Syndrome (BBS). Following negative clinical genetic testing, genome analysis identified biallelic variants in IFT57: p.(Val397Glu) and p.(Lys225Asnfs*17). IFT57 is part of complex B of the intraflagellar transport (IFT) proteins, which is an adaptor to the anterograde transport of proteins, bringing cargo from the base of the primary cilia to the tip. Variants in IFT57 have not yet been associated with BBS or human retinal degeneration, but biallelic splicing variants were associated with a distinct ciliopathy: oral-facial-digital syndrome. Using patient-derived fibroblasts, IFT57-knockouts (KO) of RPE1, and mIMCD3 cells, we showed that p.(Lys225Asnfs*17) is subjected to non-sense mediated decay, and that p.(Val397Glu) is the predominant variant which leads to cilia defects. Exogenous expression of the p.(Val397Glu) variant partially restored structural and functional primary cilia defects, and of the anterograde transport in Ift57-KO mIMCD3 cells but it did not rescue primary cilia in retinal IFT57-KO-RPE1 cells. The cell autonomous effect, likely explains the retinal dystrophy in our proband with BBS.
Collapse
Affiliation(s)
- Alexandra Nitoiu
- Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, 1 King's College Circle, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Qihong Zhang
- Department of Pediatrics, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, Iowa 52242, United States
| | - Erika Tavares
- Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
| | - Janice Min Li
- Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
| | - Kashif Ahmed
- Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
| | - Kit Green-Sanderson
- Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
| | - Mahnoor Rashid
- Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
| | - Shahir M Morcos
- Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
- Department of Molecular Genetics, Medical Sciences Building, 1 King's College, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Jayson T Maynes
- Department of Anesthesia and Pain Medicine, Peter Gilgan Centre for Research and Learning, 686 Bay Street, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
- Program in Molecular Medicine, Peter Gilgan Centre for Research and Learning, 686 Bay Street, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
| | - Eric I Campos
- Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
- Department of Molecular Genetics, Medical Sciences Building, 1 King's College, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Val C Sheffield
- Department of Pediatrics, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, Iowa 52242, United States
| | - Ajoy Vincent
- Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, 1 King's College Circle, University of Toronto, Toronto, Ontario M5S 1A8, Canada
- Ophthalmology and Vision Sciences, 555 University Avenue, Hospital for Sick Children, University of Toronto, Toronto, Ontario M5G 1X8, Canada
| | - Elise Héon
- Genetics and Genome Biology, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, 1 King's College Circle, University of Toronto, Toronto, Ontario M5S 1A8, Canada
- Ophthalmology and Vision Sciences, 555 University Avenue, Hospital for Sick Children, University of Toronto, Toronto, Ontario M5G 1X8, Canada
| |
Collapse
|