1
|
Su T, Xia Y. A quantitative comparison of the deleteriousness of missense and nonsense mutations using the structurally resolved human protein interactome. Protein Sci 2025; 34:e70155. [PMID: 40384578 PMCID: PMC12086521 DOI: 10.1002/pro.70155] [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: 01/08/2025] [Revised: 04/02/2025] [Accepted: 04/22/2025] [Indexed: 05/20/2025]
Abstract
The complex genotype-to-phenotype relationships in Mendelian diseases can be elucidated by mutation-induced disturbances to the networks of molecular interactions (interactomes) in human cells. Missense and nonsense mutations cause distinct perturbations within the human protein interactome, leading to functional and phenotypic effects with varying degrees of severity. Here, we structurally resolve the human protein interactome at atomic-level resolutions and perform structural and thermodynamic calculations to assess the biophysical implications of these mutations. We focus on a specific type of missense mutation, known as "quasi-null" mutations, which destabilize proteins and cause similar functional consequences (node removal) to nonsense mutations. We propose a "fold difference" quantification of deleteriousness, which measures the ratio between the fractions of node-removal mutations in datasets of Mendelian disease-causing and non-pathogenic mutations. We estimate the fold differences of node-removal mutations to range from 3 (for quasi-null mutations with folding ΔΔG ≥2 kcal/mol) to 20 (for nonsense mutations). We observe a strong positive correlation between biophysical destabilization and phenotypic deleteriousness, demonstrating that the deleteriousness of quasi-null mutations spans a continuous spectrum, with nonsense mutations at the extreme (highly deleterious) end. Our findings substantiate the disparity in phenotypic severity between missense and nonsense mutations and suggest that mutation-induced protein destabilization is indicative of the phenotypic outcomes of missense mutations. Our analyses of node-removal mutations allow for the potential identification of proteins whose removal or destabilization lead to harmful phenotypes, enabling the development of targeted therapeutic approaches, and enhancing comprehension of the intricate mechanisms governing genotype-to-phenotype relationships in clinically relevant diseases.
Collapse
Affiliation(s)
- Ting‐Yi Su
- Graduate Program in Quantitative Life SciencesMcGill UniversityMontréalQuébecCanada
| | - Yu Xia
- Graduate Program in Quantitative Life SciencesMcGill UniversityMontréalQuébecCanada
- Department of BioengineeringMcGill UniversityMontréalQuébecCanada
| |
Collapse
|
2
|
Li Y, Man W, Li X, Wu X, Cui Y, Chen S, Li X, Lin Y, Jiang L, Wang Y. Plasmolipin deficiency is essential for HUVECs survival under hypoxic conditions. Cell Death Discov 2025; 11:239. [PMID: 40379643 DOI: 10.1038/s41420-025-02526-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 04/29/2025] [Accepted: 05/07/2025] [Indexed: 05/19/2025] Open
Abstract
This study aims to explore the molecules that affect the survival of Human Umbilical Vein Endothelial Cells (HUVECs) under hypoxia and their mechanisms of action. In hypoxia, plasmolipin (PLLP) was identified through the screening of CRISPR/Cas9 and small guide RNA (sgRNA) library. Functionally, PLLP knockout led to increase cell proliferation, cellular metabolism, tight junction formation, angiogenesis ability, migration and invasion in hypoxic HUVECs. Furthermore, PLLP knockout countered the inhibitory effects of bevacizumab on HUVECs angiogenesis and cell survival in hypoxic conditions. PLLP knockout was found to modulate the survival of HUVECs in hypoxia by enhancing the phosphorylation of AKT and ERK1/2 proteins. In conclusion, inhibiting the expression of PLLP in HUVECs promotes cell survival and maintenance of cellular functions under hypoxic condition. PLLP plays a crucial role in regulating cell survival in hypoxia through the activation of AKT and ERK1/2 pathways. This study identifies novel molecules that affect HUVECs survival under hypoxic conditions and provides a new possibility for future studies on cell survival under hypoxic conditions.
Collapse
Affiliation(s)
- Yanghua Li
- Medical College, Guangxi University, Nanning, China
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| | - Weiling Man
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| | - Xiang Li
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| | - Xiaojie Wu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| | - Yumeng Cui
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| | - Shiyun Chen
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| | - Xianhong Li
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| | - Yanli Lin
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China.
| | - Lihe Jiang
- Medical College, Guangxi University, Nanning, China.
- School of Basic Medical Sciences, Youjiang Medical University for Nationalities, Baise, China.
- Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China.
| | - Youliang Wang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China.
| |
Collapse
|
3
|
Kolakada D, Fu R, Biziaev N, Shuvalov A, Lore M, Campbell AE, Cortázar MA, Sajek MP, Hesselberth JR, Mukherjee N, Alkalaeva E, Coban-Akdemir ZH, Jagannathan S. Systematic analysis of nonsense variants uncovers peptide release rate as a novel modifier of nonsense-mediated mRNA decay. CELL GENOMICS 2025:100882. [PMID: 40393459 DOI: 10.1016/j.xgen.2025.100882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 03/11/2025] [Accepted: 04/23/2025] [Indexed: 05/22/2025]
Abstract
The phenotypic impact of nonsense variants is determined by nonsense-mediated mRNA decay (NMD), which degrades transcripts with premature termination codons (PTCs). Despite the clinical importance of nonsense variants, transcript-specific and context-dependent variations in NMD activity remain poorly understood. Here, we show that the amino acid preceding the PTC strongly influences NMD activity. Glycine codons promote robust NMD efficiency and show striking enrichment before PTCs but are depleted before normal termination codons. Glycine-PTC enrichment is particularly pronounced in genes tolerant to loss-of-function variants, suggesting efficient elimination of truncated proteins from nonessential genes. We further demonstrate that the peptide release rate during translation termination is an important determinant of NMD activity. We propose a "window of opportunity" model where translation termination kinetics modulate NMD activity. By revealing how sequence context shapes NMD activity through translation termination dynamics, our findings provide a mechanistic framework for improved clinical interpretation of nonsense variants.
Collapse
Affiliation(s)
- Divya Kolakada
- Department of Biochemistry and Molecular Genetics, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; Molecular Biology Graduate Program, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Rui Fu
- RNA Bioscience Initiative, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Nikita Biziaev
- Engelhardt Institute of Molecular Biology, The Russian Academy of Sciences, 119991 Moscow, Russia
| | - Alexey Shuvalov
- Engelhardt Institute of Molecular Biology, The Russian Academy of Sciences, 119991 Moscow, Russia
| | - Mlana Lore
- Molecular Biology Graduate Program, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Amy E Campbell
- Department of Biochemistry and Molecular Genetics, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Michael A Cortázar
- Department of Biochemistry and Molecular Genetics, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Marcin P Sajek
- Department of Biochemistry and Molecular Genetics, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; RNA Bioscience Initiative, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | - Jay R Hesselberth
- Department of Biochemistry and Molecular Genetics, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; RNA Bioscience Initiative, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Neelanjan Mukherjee
- Department of Biochemistry and Molecular Genetics, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; RNA Bioscience Initiative, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Elena Alkalaeva
- Engelhardt Institute of Molecular Biology, The Russian Academy of Sciences, 119991 Moscow, Russia
| | - Zeynep H Coban-Akdemir
- University of Texas Health Science Center at Houston, Houston, TX, USA, TOPMed Fellowship
| | - Sujatha Jagannathan
- Department of Biochemistry and Molecular Genetics, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; RNA Bioscience Initiative, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
4
|
Han L, Liu J, Zhang R, Cheng Y, Dong L, Wei L, Liu J, Wang K, Yu J. Insights From Nonsense-Mediated mRNA Decay for Prognosis in Homologous Recombination-Deficient Ovarian Cancer. Cancer Sci 2025; 116:1449-1463. [PMID: 40022542 PMCID: PMC12044663 DOI: 10.1111/cas.70034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 03/03/2025] Open
Abstract
Not all ovarian cancer patients with homologous recombination deficiency, especially those with germline BRCA mutations, can benefit from platinum-based and targeted therapy. Our study aimed to determine the value of nonsense-mediated mRNA decay, which targeted these mutations. The retrospective analysis of 797 ovarian cancer patients was performed using two public cohorts and one in-house cohort. We developed a prediction algorithm for nonsense-mediated mRNA decay to discriminate between trigger and escape status, finding that escape status indicated a better prognosis. Subsequently, we analyzed differential gene expression and functional pathways between the two statuses and filtered 8 genes associated with the cell cycle. Then the optimized key gene model was built using integrated machine learning algorithms (mean AUC > 0.89), which had a higher independent prognostic value for ovarian cancer with germline BRCA variants or homologous recombination deficiency than the nonsense-mediated mRNA decay algorithm. Furthermore, we classified patients into high- and low-risk groups by the machine learning model and found that the low-risk group had a better prognosis with higher drug response and immune levels of activated dendritic cells than the high-risk controls. Our findings provide a perspective based on nonsense-mediated mRNA decay and cell cycle pathways to distinguish subtypes of germline BRCA or homologous recombination deficiency.
Collapse
Affiliation(s)
- Lei Han
- Cancer Molecular Diagnostics CoreTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for CancerTianjinChina
| | - Jialing Liu
- Cancer Molecular Diagnostics CoreTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for CancerTianjinChina
| | - Runjiao Zhang
- Cancer Molecular Diagnostics CoreTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for CancerTianjinChina
| | - Yanan Cheng
- Cancer Molecular Diagnostics CoreTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for CancerTianjinChina
| | - Li Dong
- Cancer Molecular Diagnostics CoreTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for CancerTianjinChina
| | - Lijuan Wei
- Cancer Prevention CenterTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjinChina
| | - Juntian Liu
- Cancer Prevention CenterTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjinChina
| | - Ke Wang
- Department of Gynecologic OncologyTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjinChina
| | - Jinpu Yu
- Cancer Molecular Diagnostics CoreTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for CancerTianjinChina
| |
Collapse
|
5
|
Ramakrishna NB, Mohamad Sahari UB, Johmura Y, Ali NA, Alghamdi M, Bauer P, Khan S, Ordoñez N, Ferreira M, Pinto Basto J, Alkuraya FS, Faqeih EA, Mori M, Almontashiri NAM, Al Shamsi A, ElGhazali G, Abu Subieh H, Al Ojaimi M, El-Hattab AW, Said Al-Kindi SA, Alhashmi N, Alhabshan F, Al Saman A, Tfayli H, Arabi M, Khalifeh S, Taylor A, Alfadhel M, Jain R, Sinha S, Shenbagam S, Ramachandran R, Altunoğlu U, Jacob A, Thalange N, El Bejjani M, Perrin A, Shin JW, Al-Maawali A, Al-Shidhani A, Al-Futaisi A, Rabea F, Chekroun I, Almarri MA, Ohta T, Nakanishi M, Alsheikh-Ali A, Ali FR, Bertoli-Avella AM, Reversade B, Abou Tayoun A. FBXO22 deficiency defines a pleiotropic syndrome of growth restriction and multi-system anomalies associated with a unique epigenetic signature. Am J Hum Genet 2025; 112:1233-1246. [PMID: 40215970 DOI: 10.1016/j.ajhg.2025.03.013] [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: 10/08/2024] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 05/04/2025] Open
Abstract
FBXO22 encodes an F-box protein, which acts as a substrate-recognition component of the SKP1-CUL1-F-box (SCF) E3 ubiquitin ligase complex. Despite its known roles in the post-translational ubiquitination and degradation of specific substrates, including histone demethylases, the impact of FBXO22 on human development remains unknown. Here, we characterize a pleiotropic syndrome with prominent prenatal onset growth restriction and notable neurodevelopmental delay across 16 cases from 14 families. Through exome and genome sequencing, we identify four distinct homozygous FBXO22 variants with loss-of-function effects segregating with the disease: three predicted to lead to premature translation termination due to frameshift effects and a single-amino-acid-deletion variant, which, we show, impacts protein stability in vitro. We confirm that affected primary fibroblasts with a frameshift mutation are bereft of endogenous FBXO22 and show increased levels of the known substrate histone H3K9 demethylase KDM4B. Accordingly, we delineate a unique epigenetic signature for this disease in peripheral blood via long-read sequencing. Altogether, we identify and demonstrate that FBXO22 deficiency leads to a pleiotropic syndrome in humans, encompassing growth restriction and neurodevelopmental delay, the pathogenesis of which may be explained by broad chromatin alterations.
Collapse
Affiliation(s)
- Navin B Ramakrishna
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
| | - Umar Bin Mohamad Sahari
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore; Department of Biochemistry, National University of Singapore, Singapore 119260, Singapore
| | - Yoshikazu Johmura
- Division of Cancer and Senescence Biology, Cancer Research Institute, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Japan
| | - Nur Ain Ali
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
| | - Malak Alghamdi
- Unit of Medical Genetics, Department of Pediatrics, College of Medicine, King Saud University Medical City, King Saud University, Riyadh, Saudi Arabia
| | | | | | | | | | | | - Fowzan S Alkuraya
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Eissa Ali Faqeih
- Section of Medical Genetics, King Fahad Medical City, Children's Specialist Hospital, Riyadh, Saudi Arabia
| | - Mari Mori
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA; Genetic and Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA
| | - Naif A M Almontashiri
- Center for Genetics and Inherited Diseases, Taibah University, 7534 Abdul Muhsin Ibn Abdul Aziz, Al Ihn, Al-Madinah al-Munawwarah 42318, Saudi Arabia; Faculty of Applied Medical Sciences, Taibah University, Janadah Bin Umayyah Road, Tayba, Al-Madinah al-Munawwarah 42353, Saudi Arabia
| | - Aisha Al Shamsi
- Paediatrics Department, Tawam Hospital, Al-Ain, United Arab Emirates
| | - Gehad ElGhazali
- HQ Medical Operations Division, Union 71, Abu Dhabi, United Arab Emirates
| | - Hala Abu Subieh
- Maternal Fetal Medicine Department, Kanad Hospital, Al Ain, United Arab Emirates
| | - Mode Al Ojaimi
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Ayman W El-Hattab
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | | | | | - Fahad Alhabshan
- Department of Cardiac Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Abdulaziz Al Saman
- Pediatric Neurology Department, National Neuroscience Institute, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Hala Tfayli
- Pediatric Endocrinology and Diabetes, American University of Beirut Medical Center (AUBMC), Beirut, Lebanon
| | - Mariam Arabi
- Department of Pediatrics and Adolescent Medicine, Pediatric Cardiology Division, Children's Heart Center, American University of Beirut Medical Center, Beirut, Lebanon
| | - Simone Khalifeh
- Pediatric Neurology Division, American University of Beirut Medical Center, Beirut, Lebanon
| | - Alan Taylor
- Dubai Health Genomic Medicine Center, Dubai Health, Dubai, United Arab Emirates
| | - Majid Alfadhel
- Genetics and Precision Medicine Department (GPM), King Abdullah Specialized Children's Hospital (KASCH), King Abdulaziz Medical City, Ministry of National Guard Health Affairs (MNG-HA), Riyadh, Saudi Arabia; Medical Genomic Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (MNG-HA), Riyadh, Saudi Arabia
| | - Ruchi Jain
- Dubai Health Genomic Medicine Center, Dubai Health, Dubai, United Arab Emirates
| | - Shruti Sinha
- Dubai Health Genomic Medicine Center, Dubai Health, Dubai, United Arab Emirates
| | - Shruti Shenbagam
- Dubai Health Genomic Medicine Center, Dubai Health, Dubai, United Arab Emirates
| | - Revathy Ramachandran
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Healthcare City, Dubai, United Arab Emirates; Center for Genomic Discovery, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai, United Arab Emirates
| | - Umut Altunoğlu
- Medical Genetics Department, Koç University School of Medicine (KUSOM), Istanbul 34010, Turkey
| | - Anju Jacob
- Al Jalila Children's Specialty Hospital, Dubai, United Arab Emirates; Dubai Health, Dubai, United Arab Emirates
| | - Nandu Thalange
- Al Jalila Children's Specialty Hospital, Dubai, United Arab Emirates; Dubai Health, Dubai, United Arab Emirates
| | - Mireille El Bejjani
- Al Jalila Children's Specialty Hospital, Dubai, United Arab Emirates; Dubai Health, Dubai, United Arab Emirates
| | - Arnaud Perrin
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
| | - Jay W Shin
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore; Department of Biochemistry, National University of Singapore, Singapore 119260, Singapore
| | - Almundher Al-Maawali
- Child Health Department, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Azza Al-Shidhani
- Child Health Department, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Amna Al-Futaisi
- Child Health Department, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Fatma Rabea
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Healthcare City, Dubai, United Arab Emirates
| | - Ikram Chekroun
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Healthcare City, Dubai, United Arab Emirates
| | - Mohamed A Almarri
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Healthcare City, Dubai, United Arab Emirates; Genome Center, Dubai Police GHQ, Dubai, United Arab Emirates
| | - Tomohiko Ohta
- Department of Translational Oncology, St. Marianna University Graduate School of Medicine, Kawasaki, Japan
| | - Makoto Nakanishi
- Division of Cancer Cell Biology, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Alawi Alsheikh-Ali
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Healthcare City, Dubai, United Arab Emirates; Dubai Health, Dubai, United Arab Emirates
| | - Fahad R Ali
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Healthcare City, Dubai, United Arab Emirates; Center for Genomic Discovery, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai, United Arab Emirates
| | | | - Bruno Reversade
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore; Medical Genetics Department, Koç University School of Medicine (KUSOM), Istanbul 34010, Turkey; NUS Cardiovascular-Metabolic Disease Translational Research Programme (CVMD-TRP), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; Laboratory of Human Genetics & Therapeutics, Biological and Environmental Sciences and Engineering Division (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia.
| | - Ahmad Abou Tayoun
- Dubai Health Genomic Medicine Center, Dubai Health, Dubai, United Arab Emirates; Center for Genomic Discovery, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai, United Arab Emirates.
| |
Collapse
|
6
|
Martins Rodrigues F, Terekhanova NV, Imbach KJ, Clauser KR, Esai Selvan M, Mendizabal I, Geffen Y, Akiyama Y, Maynard M, Yaron TM, Li Y, Cao S, Storrs EP, Gonda OS, Gaite-Reguero A, Govindan A, Kawaler EA, Wyczalkowski MA, Klein RJ, Turhan B, Krug K, Mani DR, Leprevost FDV, Nesvizhskii AI, Carr SA, Fenyö D, Gillette MA, Colaprico A, Iavarone A, Robles AI, Huang KL, Kumar-Sinha C, Aguet F, Lazar AJ, Cantley LC, Marigorta UM, Gümüş ZH, Bailey MH, Getz G, Porta-Pardo E, Ding L. Precision proteogenomics reveals pan-cancer impact of germline variants. Cell 2025; 188:2312-2335.e26. [PMID: 40233739 DOI: 10.1016/j.cell.2025.03.026] [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: 10/09/2023] [Revised: 04/29/2024] [Accepted: 03/13/2025] [Indexed: 04/17/2025]
Abstract
We investigate the impact of germline variants on cancer patients' proteomes, encompassing 1,064 individuals across 10 cancer types. We introduced an approach, "precision peptidomics," mapping 337,469 coding germline variants onto peptides from patients' mass spectrometry data, revealing their potential impact on post-translational modifications, protein stability, allele-specific expression, and protein structure by leveraging the relevant protein databases. We identified rare pathogenic and common germline variants in cancer genes potentially affecting proteomic features, including variants altering protein abundance and structure and variants in kinases (ERBB2 and MAP2K2) impacting phosphorylation. Precision peptidome analysis predicted destabilizing events in signal-regulatory protein alpha (SIRPA) and glial fibrillary acid protein (GFAP), relevant to immunomodulation and glioblastoma diagnostics, respectively. Genome-wide association studies identified quantitative trait loci for gene expression and protein levels, spanning millions of SNPs and thousands of proteins. Polygenic risk scores correlated with distal effects from risk variants. Our findings emphasize the contribution of germline genetics to cancer heterogeneity and high-throughput precision peptidomics.
Collapse
Affiliation(s)
- Fernanda Martins Rodrigues
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Nadezhda V Terekhanova
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Kathleen J Imbach
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain; Universitat Autonoma de Barcelona, Barcelona, Spain
| | | | - Myvizhi Esai Selvan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Thoracic Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Isabel Mendizabal
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain; Translational Prostate Cancer Research Lab, CIC bioGUNE-Basurto, Biocruces Bizkaia Health Research Institute, Derio, Spain
| | - Yifat Geffen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Cancer Center and Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Yo Akiyama
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Tomer M Yaron
- Meyer Cancer Center, Department of Medicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Yize Li
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Song Cao
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Erik P Storrs
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Olivia S Gonda
- Department of Biology, Brigham Young University, Salt Lake City, UT, USA
| | - Adrian Gaite-Reguero
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
| | - Akshay Govindan
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Emily A Kawaler
- Applied Bioinformatics Laboratories, New York University Langone Health, New York City, NY, USA
| | - Matthew A Wyczalkowski
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Robert J Klein
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Berk Turhan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Karsten Krug
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - D R Mani
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Alexey I Nesvizhskii
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Steven A Carr
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - David Fenyö
- Institute for Systems Genetics, NYU Grossman School of Medicine, New York, NY, USA
| | | | - Antonio Colaprico
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Antonio Iavarone
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Neurological Surgery, Department of Biochemistry and Molecular Biology, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Ana I Robles
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Rockville, MD, USA
| | - Kuan-Lin Huang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Transformative Disease Modeling, Tisch Cancer Institute, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chandan Kumar-Sinha
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA; Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | | | - Alexander J Lazar
- Departments of Pathology and Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Urko M Marigorta
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Zeynep H Gümüş
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Thoracic Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Matthew H Bailey
- Department of Biology, Brigham Young University, Salt Lake City, UT, USA.
| | - Gad Getz
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Cancer Center and Department of Pathology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| | - Eduard Porta-Pardo
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain; Barcelona Supercomputing Center (BSC), Barcelona, Spain.
| | - Li Ding
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA; Siteman Cancer Center, Washington University in St. Louis, Saint Louis, MO, USA.
| |
Collapse
|
7
|
Torices L, Nunes‐Xavier CE, Pulido R. Therapeutic Potential of Translational Readthrough at Disease-Associated Premature Termination Codons From Tumor Suppressor Genes. IUBMB Life 2025; 77:e70018. [PMID: 40317855 PMCID: PMC12046619 DOI: 10.1002/iub.70018] [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: 02/03/2025] [Revised: 03/27/2025] [Accepted: 04/14/2025] [Indexed: 05/07/2025]
Abstract
Tumor suppressor genes are frequently targeted by mutations introducing premature termination codons (PTC) in the protein coding sequence, both in sporadic cancers and in the germline of patients with cancer predisposition syndromes. These mutations have a high pathogenic impact since they generate C-terminal truncated proteins with altered stability and function. In addition, PTC mutations trigger transcript degradation by nonsense-mediated mRNA decay. Suppression of PTC by translational readthrough restores protein biosynthesis and stabilizes the PTC-targeted mRNA, making a suitable therapeutic approach the reconstitution of active full-length tumor suppressor proteins by pharmacologically-induced translational readthrough. Here, we review the recent advances in small molecule pharmacological induction of translational readthrough of disease-associated PTC from tumor suppressor genes, and discuss the therapeutic potential of translational readthrough in specific groups of patients with hereditary syndromic cancers.
Collapse
Affiliation(s)
| | - Caroline E. Nunes‐Xavier
- Biobizkaia Health Research InstituteBarakaldoSpain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERERISCIIISpain
- Institute for Cancer ResearchOslo University HospitalOsloNorway
| | - Rafael Pulido
- Biobizkaia Health Research InstituteBarakaldoSpain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERERISCIIISpain
- IkerbasqueThe Basque Foundation for ScienceBilbaoSpain
| |
Collapse
|
8
|
Chen T, Wei Y, Kang J, Zhang D, Ye J, Sun X, Hong M, Zhang W, Wu H, Ding Z, Fei G. ADAR1-HNRNPL-Mediated CircCANX Decline Promotes Autophagy in Chronic Obstructive Pulmonary Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2414211. [PMID: 40091520 PMCID: PMC12079403 DOI: 10.1002/advs.202414211] [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] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 03/05/2025] [Indexed: 03/19/2025]
Abstract
Chronic obstructive pulmonary disease (COPD) is a characteristic chronic airway inflammatory disease that worsens over time, however, there are currently limited clinical therapeutics to suspend its progression. Circular RNAs (circRNAs), which have emerged as functional regulators in various diseases, including COPD, may server as new pharmacological targets in COPD. Here, it is identified a nuclear circRNA, circCANX, that is preferentially decreased in COPD. The linear splicing of CANX pre-mRNA, enhanced by the ADAR1-HNRNPL interaction, is responsible for the circCANX decline. Clinically, the higher circCANX expression is associated with a worse lung function index of FEV1/FVC among patients with COPD. CircCANX suppresses autophagy and stress granule (SG) formation to strengthen inflammation of COPD in vivo and in vitro. Mechanistically, circCANX recruits the tumor suppressor protein P53 (P53) mRNA and RNA helicase upstream frameshift 1 (UPF1) to form a ternary complex, which mediates P53 mRNA degradation through nonsense-mediated mRNA decay (NMD) process. Together, this study reveals an important circCANX-mediated regulatory mechanism in COPD, and provides new insights into the potential of circRNA-based drug and biomarker development for COPD.
Collapse
Affiliation(s)
- Ting‐Ting Chen
- Department of Respiratory and Critical Care MedicineFirst Affiliated Hospital of Anhui Medical UniversityHefeiAnhui Province230022China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui ProvinceHefeiAnhui Province230022China
| | - Yuan‐Yuan Wei
- Department of Respiratory and Critical Care MedicineFirst Affiliated Hospital of Anhui Medical UniversityHefeiAnhui Province230022China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui ProvinceHefeiAnhui Province230022China
| | - Jia‐Ying Kang
- Department of Respiratory and Critical Care MedicineFirst Affiliated Hospital of Anhui Medical UniversityHefeiAnhui Province230022China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui ProvinceHefeiAnhui Province230022China
| | - Da‐Wei Zhang
- Department of Respiratory and Critical Care MedicineFirst Affiliated Hospital of Anhui Medical UniversityHefeiAnhui Province230022China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui ProvinceHefeiAnhui Province230022China
| | - Jing‐Jing Ye
- Department of Respiratory and Critical Care MedicineFirst Affiliated Hospital of Anhui Medical UniversityHefeiAnhui Province230022China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui ProvinceHefeiAnhui Province230022China
| | - Xi‐Shi Sun
- Emergency Medicine CenterAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdong Province524000China
| | - Mei Hong
- Department of Respiratory and Critical Care MedicineFirst Affiliated Hospital of Anhui Medical UniversityHefeiAnhui Province230022China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui ProvinceHefeiAnhui Province230022China
| | - Wen‐Ting Zhang
- Department of Respiratory and Critical Care MedicineFirst Affiliated Hospital of Anhui Medical UniversityHefeiAnhui Province230022China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui ProvinceHefeiAnhui Province230022China
| | - Hui‐Mei Wu
- Department of Geriatric Respiratory and Critical Care MedicineFirst Affiliated Hospital of Anhui Medical UniversityHefeiAnhui Province230022China
| | - Zhen‐Xing Ding
- Department of Emergency MedicineFirst Affiliated Hospital of Anhui Medical UniversityHefeiAnhui Province230022China
| | - Guang‐He Fei
- Department of Respiratory and Critical Care MedicineFirst Affiliated Hospital of Anhui Medical UniversityHefeiAnhui Province230022China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui ProvinceHefeiAnhui Province230022China
| |
Collapse
|
9
|
Akamandisa MP, Boddicker NJ, Yadav S, Hu C, Hart SN, Ambrosone C, Anton-Culver H, Auer PL, Bodelon C, Burnside ES, Chen F, Eliassen AH, Goldgar DE, Haiman C, Hodge JM, Huang H, John EM, Karam R, Lacey JV, Lindstroem S, Martinez E, Na J, Neuhausen SL, O'Brien KM, Olson JE, Pal T, Palmer JR, Patel AV, Pesaran T, Polley EC, Richardson ME, Ruddy K, Sandler DP, Teras LR, Trentham-Dietz A, Vachon CM, Weinberg C, Winham SJ, Yao S, Zirpoli G, Kraft P, Weitzel JN, Domchek SM, Couch FJ, Nathanson KL. Association of Gene Variant Type and Location with Breast Cancer Risk in the General Population. Ann Oncol 2025:S0923-7534(25)00170-X. [PMID: 40288678 DOI: 10.1016/j.annonc.2025.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/18/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Pathogenic variants (PVs) in ATM, BRCA1, BRCA2, CHEK2, and PALB2 are associated with increased breast cancer risk. However, it is unknown whether this risk differs by PV type or location in carriers ascertained from the general population. PATIENTS AND METHODS To evaluate breast cancer risks associated with PV type and location in ATM, BRCA1, BRCA2, CHEK2, and PALB2, we performed age adjusted case-control association analysis in 32,247 women with and 32,544 age-matched women without breast cancer from the CARRIERS Consortium. PVs were grouped by type and location within genes and assessed for risks of breast cancer (odds ratios (OR), 95% confidence intervals (CI), and P-values) using logistic regression. RESULTS Compared to women carrying BRCA2 exon 11 protein truncating variants (PTVs) in the CARRIERS population-based study, women with BRCA2 ex1-10 PTVs (OR=13.5, 95%CI 6.0-38.7, P<0.001) and ex13-27 PTVs (OR=9.0, 95%CI 4.9-18.5, P<0.001) had higher breast cancer risks, lower rates of ER-negative breast cancer (ex13-27 OR=0.5, 95%CI 0.2-0.9, P=0.035; ex1-10 OR=0.5, 95%CI 0.1-1.0, P=0.065), and earlier age at breast cancer diagnosis (ex13-27 5.5 years, P<0.001; ex1-10 2.4 years, P=0.169). These associations with ER-negative breast cancer and age replicated in a high-risk clinical cohort from Ambry Genetics and the population-based UK Biobank cohort. No differences in risk by gene region were observed for PTVs in other predisposition genes. CONCLUSION Population-based and clinical high-risk cohorts establish that PTVs in exon 11 of BRCA2 are associated with reduced breast cancer risk, later age at diagnosis, and greater risk of ER-negative disease. These differential risks may improve individualized risk prediction and clinical management for women carrying BRCA2 PTVs.
Collapse
Affiliation(s)
- M P Akamandisa
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - N J Boddicker
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - S Yadav
- Department of Oncology, Mayo Clinic, Rochester, MN
| | - C Hu
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - S N Hart
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - C Ambrosone
- Department of Cancer Prevention and Control, Roswell Park Cancer Center, Buffalo, NY
| | | | - P L Auer
- Division of Biostatistics, Institute for Health & Equity, and Cancer Center, Medical College of Wisconsin, Milwaukee, WI
| | - C Bodelon
- Department of Population Science, American Cancer Society, Atlanta, GA
| | - E S Burnside
- Department of Radiology, University of Wisconsin, Madison, WI
| | - F Chen
- Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - A H Eliassen
- Harvard TH Chan School of Public Health, Harvard University, Cambridge, MA
| | | | - C Haiman
- Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - J M Hodge
- Department of Population Science, American Cancer Society, Atlanta, GA
| | - H Huang
- Harvard TH Chan School of Public Health, Harvard University, Cambridge, MA
| | - E M John
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Palo Alto, CA
| | - R Karam
- Ambry Genetics, Aliso Viejo, CA
| | - J V Lacey
- Beckman Research Institute, City of Hope Cancer Center, Duarte, CA
| | - S Lindstroem
- Department of Epidemiology, University of Washington, Seattle, WA
| | - E Martinez
- Department of Family Medicine and Public Health, University of California San Diego, San Diego, CA
| | - J Na
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - S L Neuhausen
- Beckman Research Institute, City of Hope Cancer Center, Duarte, CA
| | - K M O'Brien
- National Institute of Environmental Health Sciences, Durham, NC
| | - J E Olson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - T Pal
- Division of Genetic Medicine in the Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - J R Palmer
- Slone Epidemiology Center, Boston University, Boston, MA
| | - A V Patel
- Department of Population Science, American Cancer Society, Atlanta, GA
| | | | - E C Polley
- Department of Public Health Sciences, University of Chicago, Chicago, IL
| | | | - K Ruddy
- Department of Oncology, Mayo Clinic, Rochester, MN
| | - D P Sandler
- National Institute of Environmental Health Sciences, Durham, NC
| | - L R Teras
- Department of Population Science, American Cancer Society, Atlanta, GA
| | - A Trentham-Dietz
- University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
| | - C M Vachon
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - C Weinberg
- National Institute of Environmental Health Sciences, Durham, NC
| | - S J Winham
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - S Yao
- Department of Cancer Prevention and Control, Roswell Park Cancer Center, Buffalo, NY
| | - G Zirpoli
- Slone Epidemiology Center, Boston University, Boston, MA
| | - P Kraft
- Trans-Divisional Research Program, National Cancer Institute, Rockville, MD
| | - J N Weitzel
- The University of Kansas Cancer Center, Kansas City, KS
| | - S M Domchek
- Basser Center for BRCA, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA; Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - F J Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - K L Nathanson
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA; Basser Center for BRCA, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA.
| |
Collapse
|
10
|
Hu J, Wang X, Ge C, Qi W, Li Z, Wang Y, Lai W, Ji W, Xu H. TSP-1-CD47-integrin α4β1 axis drives T cell infiltration and synovial inflammation in rheumatoid arthritis. Front Immunol 2025; 16:1524304. [PMID: 40308591 PMCID: PMC12040643 DOI: 10.3389/fimmu.2025.1524304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 03/20/2025] [Indexed: 05/02/2025] Open
Abstract
Background Immune cell infiltration into joint synovial tissue and promotion of the inflammatory response are important processes in rheumatoid arthritis (RA). This article delves into the crucial role of CD47 in these processes, as well as the mechanisms at both cellular and molecular levels. Methods CD47, its ligand TSP-1, and related integrins' expression was analyzed in RA patients' synovial and blood samples vs. normals using GEO data. Additionally, a collagen-induced arthritis (CIA) model using Cd47 knockout rats was employed to explore the significant role of CD47 in the arthritic process. This was further validated in wild-type rat CIA model using CD47 antibodies and inhibitors targeting key enzymes in the CD47-activated integrin α4β1 signaling pathway. The crucial role of CD47 in the CIA model and its way of function were investigated at the animal whole-body level, through various joint section analyses, and at the cellular and molecular level. Results Analysis of synovial tissue samples (230 cases) and blood samples (1238 cases) from RA patients in the GEO database showed that the CD47, its ligand TSP-1 and related integrins were significantly overexpressed in RA patients. When Cd47 was knocked out in a rat CIA model, the disease severity of arthritis was significantly alleviated, and the T cell infiltration into rat synovial tissue was remarkably reduced, while the number of B cells, macrophages, and neutrophils did not noticeably change. Mechanistic studies indicated that CD47 on T cells interacts with TSP-1 on vascular endothelial cells in arthritic synovium, activating T cell integrin α4β1. The activated α4β1 binds to VCAM-1, promoting T cell infiltration and inflammatory factor secretion, thereby exacerbating synovial inflammation. The present study also showed that inhibiting the activities of key kinases PKA and Src, through which CD47 mediated integrin α4β1 activation, alleviated arthritis syndromes in CIA rats. Conclusion The three-molecule model of "TSP-1, CD47 and integrin α4β1" confirmed that CD47 plays an important role in the occurrence and progression of collagen-induced arthritis, a typical animal model of rheumatoid arthritis. Blocking the TSP-1-CD47 interaction or inhibiting CD47-activated integrin α4β1 on T cells could be a potential therapeutic strategy for rheumatoid arthritis.
Collapse
Affiliation(s)
- Jialiang Hu
- State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing, China
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation of Jiangsu Province, China Pharmaceutical University, Nanjing, China
| | - Xinmin Wang
- State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing, China
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation of Jiangsu Province, China Pharmaceutical University, Nanjing, China
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou, China
| | - Chuang Ge
- State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing, China
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation of Jiangsu Province, China Pharmaceutical University, Nanjing, China
| | - Weiyan Qi
- State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing, China
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation of Jiangsu Province, China Pharmaceutical University, Nanjing, China
| | - Zeqing Li
- State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing, China
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation of Jiangsu Province, China Pharmaceutical University, Nanjing, China
| | - Yaoyao Wang
- State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing, China
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation of Jiangsu Province, China Pharmaceutical University, Nanjing, China
| | - Wenting Lai
- State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing, China
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation of Jiangsu Province, China Pharmaceutical University, Nanjing, China
| | - Wei Ji
- Department of Rheumatology and Immunology, Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Nanjing, China
| | - Hanmei Xu
- State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing, China
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation of Jiangsu Province, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
11
|
Greene D, De Wispelaere K, Lees J, Codina-Solà M, Jensson BO, Hales E, Katrinecz A, Nieto Molina E, Pascoal S, Pfundt R, Schot R, Sevilla Porras M, Sleutels F, Valenzuela I, Wijngaard R, Arroyo Carrera I, Atton G, Casas-Alba D, Donnelly D, Duat Rodríguez A, Fernández Garoz B, Foulds N, García-Navas Núñez D, González Alguacil E, Jarvis J, Kant SG, Madrigal Bajo I, Martinez-Monseny AF, McKee S, Ortiz Cabrera NV, Rodríguez-Revenga Bodi L, Sariego Jamardo A, Stefansson K, Sulem P, Suri M, Van Karnebeek C, Vasudevan P, Vega Pajares AI, Carracedo Á, Engelen M, Lapunzina P, Morgan NP, Morte B, Rump P, Stirrups K, Tizzano EF, Barakat TS, O'Donoghue M, Pérez-Jurado LA, Freson K, Mumford AD, Turro E. Mutations in the small nuclear RNA gene RNU2-2 cause a severe neurodevelopmental disorder with prominent epilepsy. Nat Genet 2025:10.1038/s41588-025-02159-5. [PMID: 40210679 DOI: 10.1038/s41588-025-02159-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 03/10/2025] [Indexed: 04/12/2025]
Abstract
The major spliceosome includes five small nuclear RNA (snRNAs), U1, U2, U4, U5 and U6, each of which is encoded by multiple genes. We recently showed that mutations in RNU4-2, the gene that encodes the U4-2 snRNA, cause one of the most prevalent monogenic neurodevelopmental disorders. Here, we report that recurrent germline mutations in RNU2-2 (previously known as pseudogene RNU2-2P), a 191-bp gene that encodes the U2-2 snRNA, are responsible for a related disorder. By genetic association, we identified recurrent de novo single-nucleotide mutations at nucleotide positions 4 and 35 of RNU2-2 in nine cases. We replicated this finding in 16 additional cases, bringing the total to 25. We estimate that RNU2-2 syndrome has a prevalence of ~20% that of RNU4-2 syndrome. The disorder is characterized by intellectual disability, autistic behavior, microcephaly, hypotonia, epilepsy and hyperventilation. All cases display a severe and complex seizure phenotype. We found that U2-2 and canonical U2-1 were similarly expressed in blood. Despite mutant U2-2 being expressed in patient blood samples, we found no evidence of missplicing. Our findings cement the role of major spliceosomal snRNAs in the etiologies of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Daniel Greene
- Department of Medicine, University of Cambridge, Cambridge, UK
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Koenraad De Wispelaere
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium
| | - Jon Lees
- Bristol Medical School, University of Bristol, Bristol, UK
| | - Marta Codina-Solà
- Department of Clinical and Molecular Genetics, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Medicine Genetics Group Vall d'Hebron Research Institute, Barcelona, Spain
| | | | - Emma Hales
- NIHR BioResource, Cambridge University Hospitals, Cambridge, UK
- Department of Haematology, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Andrea Katrinecz
- NIHR BioResource, Cambridge University Hospitals, Cambridge, UK
- Department of Haematology, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Esther Nieto Molina
- Andalusian Platform for Computational Medicine, Andalusian Public Foundation Progress and Health-FPS, Seville, Spain
| | - Sonia Pascoal
- NIHR BioResource, Cambridge University Hospitals, Cambridge, UK
- Department of Haematology, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Rolph Pfundt
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Rachel Schot
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Marta Sevilla Porras
- CIBER-ER (Biomedical Network Research Center for Rare Diseases), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Frank Sleutels
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Irene Valenzuela
- Department of Clinical and Molecular Genetics, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Medicine Genetics Group Vall d'Hebron Research Institute, Barcelona, Spain
| | - Robin Wijngaard
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Giles Atton
- Wessex Clinical Genetics Service, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Didac Casas-Alba
- Clinical Genetics Department and Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Deirdre Donnelly
- Department of Medical Genetics, Belfast City Hospital, Belfast, UK
| | - Anna Duat Rodríguez
- CIBER-ER (Biomedical Network Research Center for Rare Diseases), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Genetics Department, Hospital Niño Jesús, Madrid, Spain
| | | | - Nicola Foulds
- Wessex Clinical Genetics Service, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | | | - Elena González Alguacil
- CIBER-ER (Biomedical Network Research Center for Rare Diseases), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Genetics Department, Hospital Niño Jesús, Madrid, Spain
| | - Joanna Jarvis
- Clinical Genetics Unit, Birmingham Women's Hospital, Birmingham, UK
| | - Sarina G Kant
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Irene Madrigal Bajo
- CIBER-ER (Biomedical Network Research Center for Rare Diseases), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Biochemistry and Molecular Genetics Department, Hospital Clinic of Barcelona and Institut de Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Antonio F Martinez-Monseny
- Clinical Genetics Department and Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Shane McKee
- Department of Medical Genetics, Belfast City Hospital, Belfast, UK
| | - Nelmar Valentina Ortiz Cabrera
- CIBER-ER (Biomedical Network Research Center for Rare Diseases), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Genetics Department, Hospital Niño Jesús, Madrid, Spain
| | - Laia Rodríguez-Revenga Bodi
- CIBER-ER (Biomedical Network Research Center for Rare Diseases), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Biochemistry and Molecular Genetics Department, Hospital Clinic of Barcelona and Institut de Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Andrea Sariego Jamardo
- Neuropediatric Department, Pediatric Service, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - Kari Stefansson
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | | | - Mohnish Suri
- Clinical Genetics, Nottingham University Hospital NHS Trust, Nottingham, UK
| | - Clara Van Karnebeek
- Departments of Pediatrics and Human Genetics, Emma Center for Personalized Medicine, Amsterdam Gastro-Enterology Endocrinology Metabolism, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Pradeep Vasudevan
- Clinical Genetics, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Ana Isabel Vega Pajares
- Genetics Department, Hospital Universitario Marqués de Valdecilla, Instituto de Investigación Valdecilla (IDIVAL), Santander, Spain
| | - Ángel Carracedo
- CIBER-ER (Biomedical Network Research Center for Rare Diseases), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Genomic Medicine Group, Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela, Spain
- Galician Foundation of Genomic Medicine, IDIS, Galician Service of Health, Santiago de Compostela, Spain
| | - Marc Engelen
- Department of Pediatric Neurology, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Pablo Lapunzina
- CIBER-ER (Biomedical Network Research Center for Rare Diseases), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Institute for Medical and Molecular Genetics (INGEMM), IdiPAZ, Madrid, Spain
| | - Natasha P Morgan
- NIHR BioResource, Cambridge University Hospitals, Cambridge, UK
- Department of Haematology, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Beatriz Morte
- CIBER-ER (Biomedical Network Research Center for Rare Diseases), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Patrick Rump
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Kathy Stirrups
- NIHR BioResource, Cambridge University Hospitals, Cambridge, UK
- Department of Haematology, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Eduardo F Tizzano
- Department of Clinical and Molecular Genetics, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Medicine Genetics Group Vall d'Hebron Research Institute, Barcelona, Spain
| | - Tahsin Stefan Barakat
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | | | - Luis Alberto Pérez-Jurado
- CIBER-ER (Biomedical Network Research Center for Rare Diseases), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Genetics Service, Hospital del Mar and Hospital del Mar Research Institute, Barcelona, Spain
| | - Kathleen Freson
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium
| | - Andrew D Mumford
- Bristol Medical School, University of Bristol, Bristol, UK
- NHS South West Genomic Medicine Service Alliance, Bristol, UK
| | - Ernest Turro
- Department of Medicine, University of Cambridge, Cambridge, UK.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
12
|
Han B, Chen D, Chen Z, Wang T, Zi K, Feng R, Yang X, Li L, Liang J, Gao X. The mouse resource at National Resource Center for Mutant Mice of China. Mamm Genome 2025:10.1007/s00335-025-10118-y. [PMID: 40146303 DOI: 10.1007/s00335-025-10118-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 02/22/2025] [Indexed: 03/28/2025]
Abstract
Mouse models serve as the most important laboratory resource for both biomedical research and preclinical study of drug development. National Resource Center of Mutant Mice (NRCMM) of China was initiated in 2001 and became one of the 31 members of National Science and Technology Resource Sharing Platform in 2019. Currently, NRCMM is co-managed by Model Animal Research Center of Nanjing University and Gempharmatech (GPT, a Shanghai Exchange enlisted public company). Dedicated to produce and collect genetic edited mouse models, NRCMM holds more than 22,000 mouse strains in 2024, compared with 18,500 strains reported in 2022. This review provides an update on our Knock-Out All Project (KOAP) and highlights resources available for immune system reconstitution models, disease models, and chromosome substitution strains at NRCMM.
Collapse
Affiliation(s)
- Bingzhou Han
- National Resource Center for Mutant Mice, GemPharmatech Co., Ltd, Nanjing, China
| | - Dongshen Chen
- National Resource Center for Mutant Mice, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Zhong Chen
- National Resource Center for Mutant Mice, GemPharmatech Co., Ltd, Nanjing, China
| | - Ting Wang
- National Resource Center for Mutant Mice, GemPharmatech Co., Ltd, Nanjing, China
| | - Kaiyuan Zi
- National Resource Center for Mutant Mice, GemPharmatech Co., Ltd, Nanjing, China
| | - Rui Feng
- National Resource Center for Mutant Mice, GemPharmatech Co., Ltd, Nanjing, China
| | - Xiaoliu Yang
- National Resource Center for Mutant Mice, GemPharmatech Co., Ltd, Nanjing, China
| | - Ling'en Li
- National Resource Center for Mutant Mice, GemPharmatech Co., Ltd, Nanjing, China
| | - Juan Liang
- National Resource Center for Mutant Mice, GemPharmatech Co., Ltd, Nanjing, China.
| | - Xiang Gao
- National Resource Center for Mutant Mice, GemPharmatech Co., Ltd, Nanjing, China.
- National Resource Center for Mutant Mice, Model Animal Research Center of Nanjing University, Nanjing, China.
- GemPharmatech Co., Ltd., Guangdong, China.
| |
Collapse
|
13
|
Cooper GW, Lee BP, Kim WJ, Su Y, Chen VZ, Salas E, Yang X, Lintner RE, Piccioni F, Giacomelli AO, Howard TP, Bagchi P, Conneely KN, Root DE, Liang B, Hahn WC, Gorkin DU, Biegel JA, Chi SN, Hong AL. SMARCB1 missense mutants disrupt SWI/SNF complex stability and remodeling activity. RESEARCH SQUARE 2025:rs.3.rs-6018128. [PMID: 40196006 PMCID: PMC11975040 DOI: 10.21203/rs.3.rs-6018128/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Chromatin remodeling complexes, such as the SWItch/Sucrose Non-Fermentable (SWI/SNF) complex, play key roles in regulating gene expression by modulating nucleosome positioning. The core subunit SMARCB1 is essential for these functions, as it anchors the complex to the nucleosome acidic patch, enabling effective chromatin remodeling. While biallelic inactivation of SMARCB1 is a hallmark of several aggressive pediatric malignancies, the functional implication of missense mutations is not fully understood. Current diagnostic approaches focus on detecting the presence or absence of SMARCB1 by immunohistochemistry (IHC) often without consideration of mutation status as such data is lacking. Here, we present the first comprehensive deep mutational scanning (DMS) of SMARCB1, encompassing 8,418 amino acid substitutions, to systematically assess their functional impact. We show that missense mutations in the RPT2 domain of SMARCB1 disrupt SMARCB1 tumor suppressor function by destabilizing the SWI/SNF complex. Notably, we identify mutations in RPT2 that impair chromatin remodeling and transcriptional regulation to an extent comparable to nonsense mutations, despite maintaining detectable protein expression, thus challenging the conventional diagnostic reliance on IHC. Importantly, these mutations seem to act by disrupting winged-helix domain flexibility. These findings provide a deeper understanding of the role of SMARCB1 in chromatin remodeling and cancer biology, highlighting the limitations of current mutation classification approaches. By establishing a high-throughput functional framework, this study offers a critical resource for elucidating SMARCB1's mutational landscape and its implications for cancer diagnostics.
Collapse
Affiliation(s)
- Garrett W Cooper
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Aflac Cancer and Blood Disorders Center - Children’s Healthcare of Atlanta, Atlanta, GA, USA
| | - Benjamin P Lee
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Aflac Cancer and Blood Disorders Center - Children’s Healthcare of Atlanta, Atlanta, GA, USA
| | - Won Jun Kim
- Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yongdong Su
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Aflac Cancer and Blood Disorders Center - Children’s Healthcare of Atlanta, Atlanta, GA, USA
| | - Victor Z Chen
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Aflac Cancer and Blood Disorders Center - Children’s Healthcare of Atlanta, Atlanta, GA, USA
| | - Eliseo Salas
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Xiaoping Yang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | | | | | - Pritha Bagchi
- Emory Integrated Proteomics Core, Emory University, Atlanta, GA, USA
| | - Karen N Conneely
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - David E Root
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Bo Liang
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - William C Hahn
- Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - David U Gorkin
- Department of Biology, Emory University, Atlanta, GA, USA
| | - Jaclyn A Biegel
- Department of Pathology, Children’s Hospital Los Angeles and Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Susan N Chi
- Dana-Farber Cancer Institute, Boston, MA, USA
- Boston Children’s Hospital, Boston, MA, USA
| | - Andrew L Hong
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Aflac Cancer and Blood Disorders Center - Children’s Healthcare of Atlanta, Atlanta, GA, USA
- Winship Cancer Institute, Atlanta, GA, USA
| |
Collapse
|
14
|
Alalam H, Šafhauzer M, Sunnerhagen P. New reporters for monitoring cellular NMD. RNA (NEW YORK, N.Y.) 2025; 31:600-611. [PMID: 39880586 PMCID: PMC11912909 DOI: 10.1261/rna.080272.124] [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: 10/16/2024] [Accepted: 01/20/2025] [Indexed: 01/31/2025]
Abstract
Nonsense-mediated decay (NMD) is a eukaryotic surveillance pathway that controls degradation of cytoplasmic transcripts with aberrant features. NMD-controlled RNA degradation acts to regulate a large fraction of the mRNA population. It has been implicated in cellular responses to infections and environmental stress, as well as in deregulation of tumor-promoting genes. NMD is executed by a set of three core factors conserved in evolution, UPF1-3, as well as additional influencing proteins such as kinases. Monitoring NMD activity is challenging due to the difficulties in quantitating RNA decay rates in vivo, and consequently, it has also been problematic to identify new factors influencing NMD. Here, we developed a genetic selection system in yeast to capture new components affecting NMD status. The reporter constructs link NMD target sequences with nutrient-selectable genetic markers. By crossing these reporters into a genome-wide library of deletion mutants and quantitating colony growth on a selective medium, we robustly detect previously known NMD components in a high-throughput fashion. In addition, we identify novel mutations influencing NMD status and implicate ribosome recycling as important for NMD. By using our constructed combinations of promoters, NMD target sequences, and selectable markers, the system can also efficiently detect mutations with a minor effect, or in special environments. Furthermore, it can be used to explore how NMD acts on targets of different structures.
Collapse
Affiliation(s)
- Hanna Alalam
- Department of Chemistry and Molecular Biology, University of Gothenburg, 413 90 Göteborg, Sweden
| | - Monika Šafhauzer
- Department of Chemistry and Molecular Biology, University of Gothenburg, 413 90 Göteborg, Sweden
| | - Per Sunnerhagen
- Department of Chemistry and Molecular Biology, University of Gothenburg, 413 90 Göteborg, Sweden
| |
Collapse
|
15
|
Mabin JW, Vock IW, Machyna M, Haque N, Thakran P, Zhang A, Rai G, Leibler INM, Inglese J, Simon MD, Hogg JR. Uncovering the isoform-resolution kinetic landscape of nonsense-mediated mRNA decay with EZbakR. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.12.642874. [PMID: 40161772 PMCID: PMC11952489 DOI: 10.1101/2025.03.12.642874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Cellular RNA levels are a product of synthesis and degradation kinetics, which can differ among transcripts of the same gene. An important cause of isoform-specific decay is the nonsense-mediated mRNA decay (NMD) pathway, which degrades transcripts with premature termination codons (PTCs) and other features. Understanding NMD functions requires strategies to quantify isoform kinetics; however, current approaches remain limited. Methods like nucleotide-recoding RNA-seq (NR-seq) enable insights into RNA kinetics, but existing bioinformatic tools do not provide robust, isoform-specific degradation rate constant estimates. We extend the EZbakR-suite by implementing a strategy to infer isoform-level kinetics from short-read NR-seq data. This approach uncovers unexpected variability in NMD efficiency among transcripts with conserved PTC-containing exons and rapid decay of a subset of mRNAs lacking PTCs. Our findings highlight the effects of competition between NMD and other decay pathways, provide mechanistic insights into established NMD efficiency correlates, and identify transcript features promoting efficient decay.
Collapse
Affiliation(s)
- Justin W. Mabin
- Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Isaac W. Vock
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06520, USA
- Institute of Biomolecular Design and Discovery, Yale University, West Haven, Connecticut 06516, USA
| | - Martin Machyna
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06520, USA
- Institute of Biomolecular Design and Discovery, Yale University, West Haven, Connecticut 06516, USA
- Present address: Paul-Ehrlich-Institut, Host-Pathogen-Interactions, 63225 Langen, Germany
| | - Nazmul Haque
- Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
- Present address: Ultragenyx, 7000 Shoreline Ct, South San Francisco, CA 94080
| | - Poonam Thakran
- Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Alexandra Zhang
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06520, USA
- Institute of Biomolecular Design and Discovery, Yale University, West Haven, Connecticut 06516, USA
| | - Ganesha Rai
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, 20850 Maryland, USA
| | | | - James Inglese
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, 20850 Maryland, USA
- Metabolic Medicine Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Matthew D. Simon
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06520, USA
- Institute of Biomolecular Design and Discovery, Yale University, West Haven, Connecticut 06516, USA
| | - J. Robert Hogg
- Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
16
|
Kilgore HR, Chinn I, Mikhael PG, Mitnikov I, Van Dongen C, Zylberberg G, Afeyan L, Banani S, Wilson-Hawken S, Lee TI, Barzilay R, Young RA. Protein codes promote selective subcellular compartmentalization. Science 2025; 387:1095-1101. [PMID: 39913643 PMCID: PMC12034300 DOI: 10.1126/science.adq2634] [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: 05/05/2024] [Revised: 11/07/2024] [Accepted: 01/28/2025] [Indexed: 02/12/2025]
Abstract
Cells have evolved mechanisms to distribute ~10 billion protein molecules to subcellular compartments where diverse proteins involved in shared functions must assemble. In this study, we demonstrate that proteins with shared functions share amino acid sequence codes that guide them to compartment destinations. We developed a protein language model, ProtGPS, that predicts with high performance the compartment localization of human proteins excluded from the training set. ProtGPS successfully guided generation of novel protein sequences that selectively assemble in the nucleolus. ProtGPS identified pathological mutations that change this code and lead to altered subcellular localization of proteins. Our results indicate that protein sequences contain not only a folding code but also a previously unrecognized code governing their distribution to diverse subcellular compartments.
Collapse
Affiliation(s)
- Henry R. Kilgore
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Itamar Chinn
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Peter G. Mikhael
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ilan Mitnikov
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Guy Zylberberg
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Lena Afeyan
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Salman Banani
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Susana Wilson-Hawken
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
- Program of Computational & Systems Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Tong Ihn Lee
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Regina Barzilay
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Richard A. Young
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
17
|
Moresco G, Rondinone O, Mauri A, Gorgoglione R, Graziani DMG, Dziuback M, Miozzo MR, Sirchia SM, Pietrogrande L, Peron A, Fontana L. A novel frameshift TBX4 variant in a family with ischio-coxo-podo-patellar syndrome and variable severity. Genes Genomics 2025; 47:341-349. [PMID: 39467966 PMCID: PMC11906559 DOI: 10.1007/s13258-024-01589-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 10/16/2024] [Indexed: 10/30/2024]
Abstract
BACKGROUND Congenital anomalies of the knee are a spectrum of rare disorders with wide clinical and genetic variability, which are mainly due to the complex processes underlying knee development. Despite progresses in understanding pathomechanisms and associated genes, many patients remain undiagnosed. OBJECTIVE To uncover the genetic bases of a congenital patellar dislocation affecting multiple family members with variable severity. METHODS We performed ES in the proband and his father, both showing bilateral patellar dislocation, his sister with a milder similar condition, and his unaffected mother. Sanger sequencing was then performed in the proband's brother and paternal aunt, both affected as well. RESULTS ES and Sanger sequencing identified the presence of the novel heterozygous frameshift mutation c.735delT in the TBX4 gene in all affected family members. TBX4 is associated with autosomal dominant ischio-coxo-podo-patellar syndrome with/without pulmonary arterial hypertension (ICPPS, #147891), reaching a diagnosis in the family. Intrafamilial clinical heterogeneity suggests that other factors might be involved, such as additional variants in TBX4 or in other modifier genes. Interestingly, we identified three additional variants in the TBX4 gene in the proband only, whose phenotype is more severe. Despite being classified as benign, one of these variants is predicted to disrupt a splicing protein binding site, and may therefore affect TBX4 alternative splicing, accounting for the more severe phenotype of the proband. CONCLUSION We expand and further delineate the genotypic and phenotypic spectrum of ICPPS. Further studies are necessary to shed light on the potential effect of this variant and on the variable phenotypic expressivity of TBX4-related phenotypes.
Collapse
Affiliation(s)
- Giada Moresco
- Medical Genetics, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Ornella Rondinone
- Medical Genetics, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Alessia Mauri
- Research Laboratories Coordination Unit, Fondazione IRCCS Ca Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Biomedical and Clinical Sciences, Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", University of Milan, Milan, Italy
| | | | - Daniela Maria Grazia Graziani
- Medical Genetics, ASST Santi Paolo e Carlo, Milan, Italy
- Medical Genetics Laboratory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Michal Dziuback
- Orthopedics and Traumatology Unit, ASST Santi Paolo e Carlo, Milan, Italy
| | - Monica Rosa Miozzo
- Medical Genetics, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
- Medical Genetics, ASST Santi Paolo e Carlo, Milan, Italy
| | - Silvia Maria Sirchia
- Medical Genetics, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Luca Pietrogrande
- Orthopedics and Traumatology Unit, ASST Santi Paolo e Carlo, Milan, Italy
- Orthopedics and Traumatology, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Angela Peron
- Medical Genetics, ASST Santi Paolo e Carlo, Milan, Italy.
- Division of Medical Genetics, Meyer Children's Hospital IRCCS, Florence, Italy.
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università degli Studi di Firenze, Florence, Italy.
| | - Laura Fontana
- Medical Genetics, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
- Medical Genetics, ASST Santi Paolo e Carlo, Milan, Italy
| |
Collapse
|
18
|
Elkhateeb N, Crookes R, Spiller M, Pavinato L, Palermo F, Brusco A, Parker M, Park SM, Mendes AC, Saraiva JM, Hammer TB, Nazaryan-Petersen L, Barakat TS, Wilke M, Bhoj E, Ahrens-Nicklas RC, Li D, Nomakuchi T, Brilstra EH, Hunt D, Johnson D, Mansour S, Oprych K, Mehta SG, Platzer K, Schnabel F, Kiep H, Faust H, Prinzing G, Wiltrout K, Radley JA, Serrano Russi AH, Atallah I, Campos-Xavier B, Amor DJ, Morgan AT, Fagerberg C, Andersen UA, Andersen CB, Bijlsma EK, Bird LM, Mullegama SV, Green A, Isidor B, Cogné B, Kenny J, Lynch SA, Quin S, Low K, Herget T, Kortüm F, Levy RJ, Morrison JL, Wheeler PG, Narumanch T, Peron K, Matthews N, Uhlman J, Bell L, Pang L, Scurr I, Belles RS, Salbert BA, Schaefer GB, Green S, Ros A, Rodríguez-Palmero A, Višnjar T, Writzl K, Vasudevan PC, Balasubramanian M. Expanding the phenotype and genotype spectrum of TAOK1 neurodevelopmental disorder and delineating TAOK2 neurodevelopmental disorder. Genet Med 2025; 27:101348. [PMID: 39737487 DOI: 10.1016/j.gim.2024.101348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 01/01/2025] Open
Abstract
PURPOSE The thousand and one kinase (TAOK) proteins are a group of serine/threonine-protein kinases involved in signaling pathways, cytoskeleton regulation, and neuronal development. TAOK1 variants are associated with a neurodevelopmental disorder (NDD) characterized by distinctive facial features, hypotonia, and feeding difficulties. TAOK2 variants have been reported to be associated with autism and early-onset obesity. However, a distinct TAOK2-NDD has not yet been delineated. METHODS We retrospectively studied the clinical and genetic data of individuals recruited from several centers with TAOK1 and TAOK2 variants that were detected through exome and genome sequencing. RESULTS We report 50 individuals with TAOK1 variants with associated phenotypes, including neurodevelopmental abnormalities (100%), macrocephaly (83%), and hypotonia (58%). We report male genital anomalies and hypoglycemia as novel phenotypes. Thirty-seven unique TAOK1 variants were identified. Most of the missense variants clustered in the protein kinase domain at residues that are intolerant to missense variation. We report 10 individuals with TAOK2 variants with associated phenotypes, including neurodevelopmental abnormalities (100%), macrocephaly (75%), autism (75%), and obesity (70%). CONCLUSION We describe the largest cohort of TAOK1-NDD to date, to our knowledge, expanding its phenotype and genotype spectrum with 30 novel variants. We delineated the phenotype of a novel TAOK2-NDD associated with neurodevelopmental abnormalities, autism, macrocephaly, and obesity.
Collapse
Affiliation(s)
- Nour Elkhateeb
- Department of Clinical Genetics, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom.
| | - Renarta Crookes
- Sheffield Diagnostic Genetics Service, Sheffield Children's Hospital NHS Foundation Trust, Sheffield, United Kingdom
| | - Michael Spiller
- Sheffield Diagnostic Genetics Service, Sheffield Children's Hospital NHS Foundation Trust, Sheffield, United Kingdom
| | - Lisa Pavinato
- Department of Medical Sciences, University of Turin, Turin, Italy; Institute of Oncology Research (IOR), Bellinzona Institutes of Science (BIOS(+)), Bellinzona, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Flavia Palermo
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Alfredo Brusco
- Department of Neurosciences Rita Levi-Montalcini, University of Turin, Turin, Italy; Medical Genetics Unit, Città della Salute e della Scienza University Hospital, Turin, Italy
| | - Michael Parker
- Sheffield Clinical Genetics Service, Sheffield Children's NHS Foundation Trust, Sheffield, United Kingdom
| | - Soo-Mi Park
- Department of Clinical Genetics, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Ariana Costa Mendes
- Medical Genetics Department, Hospital Pediátrico de Coimbra, Unidade Local de Saúde de Coimbra, Coimbra, Portugal
| | - Jorge M Saraiva
- Medical Genetics Department, Hospital Pediátrico de Coimbra, Unidade Local de Saúde de Coimbra, Coimbra, Portugal; University Clinic of Pediatrics, Faculty of Medicine, University of Coimbra, Portugal; Clinical Academic Center of Coimbra, Hospital Pediátrico de Coimbra, Unidade Local de Saúde de Coimbra, Coimbra, Portugal
| | - Trine Bjørg Hammer
- Department of Epilepsy Genetics and Personalized Medicine, Danish Epilepsy Centre, Dianalund, Denmark; Department of Genetics, Rigshospitalet, Copenhagen, Denmark
| | - Lusine Nazaryan-Petersen
- Department of Genetics, Rigshospitalet, Copenhagen, Denmark; Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tahsin Stefan Barakat
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands; Discovery Unit, Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands; ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Martina Wilke
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Elizabeth Bhoj
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | | | - Dong Li
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Tomoki Nomakuchi
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Eva H Brilstra
- Department of Genetics and Brain Center, University Medical Center Utrecht, The Netherlands
| | - David Hunt
- Wessex Clinical Genetics Service, Princess Anne Hospital, University Hospital Southampton NHS Trust, Southampton, United Kingdom
| | - Diana Johnson
- Sheffield Clinical Genetics Service, Sheffield Children's NHS Foundation Trust, Sheffield, United Kingdom
| | - Sahar Mansour
- South West Thames Regional Genetics Service, St George's University Hospitals NHS Foundation Trust, London, United Kingdom; School of Biological and Molecular Sciences, St George's University of London, London, United Kingdom
| | - Kathryn Oprych
- South West Thames Regional Genetics Service, St George's University Hospitals NHS Foundation Trust, London, United Kingdom
| | - Sarju G Mehta
- Department of Clinical Genetics, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Konrad Platzer
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany; Division of Neuropaediatrics, Hospital for Children and Adolescents, University Hospital Leipzig, Leipzig, Germany
| | - Franziska Schnabel
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany; Division of Neuropaediatrics, Hospital for Children and Adolescents, University Hospital Leipzig, Leipzig, Germany
| | - Henriette Kiep
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany; Division of Neuropaediatrics, Hospital for Children and Adolescents, University Hospital Leipzig, Leipzig, Germany
| | - Helene Faust
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany; Division of Neuropaediatrics, Hospital for Children and Adolescents, University Hospital Leipzig, Leipzig, Germany
| | | | | | - Jessica A Radley
- North West Thames Regional Genomics Service, London North West University Healthcare NHS Trust, Northwick Park Hospital, United Kingdom
| | - Alvaro H Serrano Russi
- Division of Genetics, Department of Pediatrics, East Tennessee State University (ETSU), Quillen College of Medicine, TN
| | - Isis Atallah
- Division of Genetic Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Belinda Campos-Xavier
- Division of Genetic Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - David J Amor
- Speech and Language, Murdoch Children's Research Institute, Melbourne, Victoria, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Angela T Morgan
- Speech and Language, Murdoch Children's Research Institute, Melbourne, Victoria, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Christina Fagerberg
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark; Department of Clinical Genetics, Lillebaelt Hospital, location Vejle Hospital, Vejle, Denmark
| | - Ulla A Andersen
- Department of Mental Health, Odense University Hospital, Odense, Denmark
| | | | - Emilia K Bijlsma
- Department of Clinical Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Lynne M Bird
- Division of Genetics and Dysmorphology, Department of Pediatrics, University of California San Diego, Rady Children's Hospital, San Diego, CA
| | | | - Andrew Green
- Department of Clinical Genetics, Children's Health Ireland at Crumlin, Dublin, Ireland; University College Dublin School of Medicine and Medical Science, Dublin, Ireland
| | - Bertrand Isidor
- Nantes Université, CNRS, INSERM, l'institut du thorax, Nantes, France; CHU Nantes, Service de Génétique Médicale, Nantes Université, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Benjamin Cogné
- Nantes Université, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Janna Kenny
- Department of Clinical Genetics, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Sally A Lynch
- Department of Clinical Genetics, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Shauna Quin
- Department of Clinical Genetics, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Karen Low
- Department of Clinical Genetics, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, United Kingdom; Bristol Medical School, University of Bristol, United Kingdom
| | - Theresia Herget
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fanny Kortüm
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Rebecca J Levy
- Neurology and Neurological Sciences, Division of Child Neurology, Stanford University and Lucile Packard Children's Hospital, Palo Alto, CA
| | | | | | - TaraChandra Narumanch
- Division of Genetics, Department of Pediatrics, West Virginia University, Morgantown, WV
| | - Kristina Peron
- Division of Genetics, Department of Pediatrics, West Virginia University, Morgantown, WV
| | - Nicole Matthews
- Division of Genetics, Department of Pediatrics, West Virginia University, Morgantown, WV
| | | | - Lauren Bell
- University of Illinois College of Medicine, Peoria, IL
| | - Lewis Pang
- Exeter Genomics Laboratory, RILD Building, Royal Devon and Exeter NHS Foundation Trust, Exeter, United Kingdom
| | - Ingrid Scurr
- Department of Clinical Genetics, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, United Kingdom
| | | | | | | | - Sarah Green
- University of Arkansas for Medical Sciences, Little Rock, AR
| | - Andrea Ros
- Department of Genetics, Hospital Universitari Germans Trias i Pujol, Catalonia, Spain
| | - Agustí Rodríguez-Palmero
- Pediatric Neurology Unit, Department of Pediatrics, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, Catalonia, Spain; Germans Trias i Pujol Research Institute (IGTP), Badalona, Barcelona, Spain
| | - Tanja Višnjar
- Clinical Institute of Genomic Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Karin Writzl
- Clinical Institute of Genomic Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia; Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Pradeep C Vasudevan
- Department of Clinical Genetics, Leicester Royal Infirmary, University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
| | - Meena Balasubramanian
- Sheffield Clinical Genetics Service, Sheffield Children's NHS Foundation Trust, Sheffield, United Kingdom; Division of Clinical Medicine, University of Sheffield, Sheffield, United Kingdom.
| |
Collapse
|
19
|
Peron A, D'Arco F, Aldinger KA, Smith-Hicks C, Zweier C, Gradek GA, Bradbury K, Accogli A, Andersen EF, Au PYB, Battini R, Beleford D, Bird LM, Bouman A, Bruel AL, Busk ØL, Campeau PM, Capra V, Carlston C, Carmichael J, Chassevent A, Clayton-Smith J, Bamshad MJ, Earl DL, Faivre L, Philippe C, Ferreira P, Graul-Neumann L, Green MJ, Haffner D, Haldipur P, Hanna S, Houge G, Jones WD, Kraus C, Kristiansen BE, Lespinasse J, Low KJ, Lynch SA, Maia S, Mao R, Kalinauskiene R, Melver C, McDonald K, Montgomery T, Morleo M, Motter C, Openshaw AS, Palumbos JC, Parikh AS, Perilla-Young Y, Powell CM, Person R, Desai M, Piard J, Pfundt R, Scala M, Serey-Gaut M, Shears D, Slavotinek A, Suri M, Turner C, Tvrdik T, Weiss K, Wentzensen IM, Zollino M, Hsieh TC, de Vries BBA, Guillemot F, Dobyns WB, Viskochil D, Dias C. BCL11A intellectual developmental disorder: defining the clinical spectrum and genotype-phenotype correlations. Eur J Hum Genet 2025; 33:312-324. [PMID: 39448799 PMCID: PMC11893779 DOI: 10.1038/s41431-024-01701-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/27/2024] [Accepted: 09/23/2024] [Indexed: 10/26/2024] Open
Abstract
An increasing number of individuals with intellectual developmental disorder (IDD) and heterozygous variants in BCL11A are identified, yet our knowledge of manifestations and mutational spectrum is lacking. To address this, we performed detailed analysis of 42 individuals with BCL11A-related IDD (BCL11A-IDD, a.k.a. Dias-Logan syndrome) ascertained through an international collaborative network, and reviewed 35 additional previously reported patients. Analysis of 77 affected individuals identified 60 unique disease-causing variants (30 frameshift, 7 missense, 6 splice-site, 17 stop-gain) and 8 unique BCL11A microdeletions. We define the most prevalent features of BCL11A-IDD: IDD, postnatal-onset microcephaly, hypotonia, behavioral abnormalities, autism spectrum disorder, and persistence of fetal hemoglobin (HbF), and identify autonomic dysregulation as new feature. BCL11A-IDD is distinguished from 2p16 microdeletion syndrome, which has a higher incidence of congenital anomalies. Our results underscore BCL11A as an important transcription factor in human hindbrain development, identifying a previously underrecognized phenotype of a small brainstem with a reduced pons/medulla ratio. Genotype-phenotype correlation revealed an isoform-dependent trend in severity of truncating variants: those affecting all isoforms are associated with higher frequency of hypotonia, and those affecting the long (BCL11A-L) and extra-long (-XL) isoforms, sparing the short (-S), are associated with higher frequency of postnatal microcephaly. With the largest international cohort to date, this study highlights persistence of fetal hemoglobin as a consistent biomarker and hindbrain abnormalities as a common feature. It contributes significantly to our understanding of BCL11A-IDD through an extensive unbiased multi-center assessment, providing valuable insights for diagnosis, management and counselling, and into BCL11A's role in brain development.
Collapse
Affiliation(s)
- Angela Peron
- Division of Medical Genetics, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT, USA.
- Medical Genetics, ASST Santi Paolo e Carlo, San Paolo Hospital, Milano, Italy.
- Department of Experimental and Clinical Biomedical Sciences, Università degli Studi di Firenze, Firenze, Italy.
- Medical Genetics, Meyer Children's Hospital IRCCS, Firenze, Italy.
| | - Felice D'Arco
- Department of Radiology, Great Ormond Street Hospital for Children, London, UK
| | - Kimberly A Aldinger
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Constance Smith-Hicks
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurogenetics, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Christiane Zweier
- Institute of Human Genetics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Human Genetics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Gyri A Gradek
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Kimberley Bradbury
- Department of Medical Genetics, Guy's and St. Thomas' NHS Foundation Trust, London, UK
- Wessex Regional Genetics Service, Princess Anne Hospital, Southampton, UK
| | - Andrea Accogli
- Genomics and Clinical Genetics, IRCCS Istituto Giannina Gaslini, Genova, Italy
- U.O.C. Genetica Medica, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Erica F Andersen
- ARUP Laboratories, Cytogenetics and Genomic Microarray, Salt Lake City, UT, USA
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Ping Yee Billie Au
- Department of Pediatrics, Division of Medical Genetics, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Roberta Battini
- IRCCS Fondazione Stella Maris, Pisa, Italy
- Dipartimento di Medicina Clinica e Sperimentale, University of Pisa, Pisa, Italy
| | - Daniah Beleford
- Division of Medical Genetics, Department of Pediatrics, Benioff Children's Hospital, University of California, San Francisco, CA, USA
- Department of Pediatrics and Physiology & Membrane Biology, University of California, Davis, CA, USA
| | - Lynne M Bird
- Department of Pediatrics, University of California San Diego, San Diego, CA, USA
- Division of Genetics/Dysmorphology, Rady Children's Hospital San Diego, San Diego, CA, USA
| | - Arjan Bouman
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Ange-Line Bruel
- INSERM UMR 1231 Equipe GAD, Université de Bourgogne, Dijon, France
- Unité Fonctionnelle d'Innovation diagnostique des maladies rares, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
| | - Øyvind Løvold Busk
- Department of Medical Genetics, Telemark Hospital Trust, 3710, Skien, Norway
| | - Philippe M Campeau
- Department of Pediatrics, CHU Sainte-Justine and University of Montreal, Montreal, QC, Canada
| | - Valeria Capra
- Genomics and Clinical Genetics, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Colleen Carlston
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
| | - Jenny Carmichael
- Department of Clinical Genetics, Addenbrooke's Hospital, Cambridge, UK
| | - Anna Chassevent
- Department of Neurogenetics, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Jill Clayton-Smith
- Division of Evolution and Genomic Sciences School of Biological Sciences University of Manchester, Manchester, UK
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Michael J Bamshad
- Division of Genetic Medicine, Seattle Children's Hospital, Seattle, WA, USA
- University of Washington, Seattle, WA, USA
| | - Dawn L Earl
- Division of Genetic Medicine, Seattle Children's Hospital, Seattle, WA, USA
- University of Washington, Seattle, WA, USA
| | - Laurence Faivre
- INSERM UMR 1231 Equipe GAD, Université de Bourgogne, Dijon, France
- Centre de Référence Maladies Rares Anomalies du développement et syndromes malformatifs, Centre de Génétique, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
| | - Christophe Philippe
- INSERM UMR 1231 Equipe GAD, Université de Bourgogne, Dijon, France
- Unité Fonctionnelle d'Innovation diagnostique des maladies rares, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
| | - Patrick Ferreira
- Department of Pediatrics, Division of Medical Genetics, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Luitgard Graul-Neumann
- Universitätsmedizin Berlin, Institut für Medizinische Genetik und Humangenetik, Berlin, Germany
| | - Mary J Green
- Experimental Histopathology Laboratory, The Francis Crick Institute, London, UK
| | - Darrah Haffner
- Department of Pediatrics, Division of Pediatric Neurology, Nationwide Children's Hospital and Ohio State University, Columbus, OH, USA
| | - Parthiv Haldipur
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Suhair Hanna
- Department of Pediatric Immunology, Rappaport Children's Hospital, Rambam Health Care Campus, Haifa, Israel
- Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Gunnar Houge
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Wendy D Jones
- North East Thames Regional Genetics Service, Great Ormond Street Hospital for Children, Great Ormond Street, London, UK
| | - Cornelia Kraus
- Institute of Human Genetics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | | | - James Lespinasse
- HDR - Service de Génétique Médicale, Centre Hospitalier Métropole Savoie, Chambery, France
| | - Karen J Low
- Clinical Genetics Service, University Hospitals Bristol and Weston NHS trust, Bristol, UK
| | - Sally Ann Lynch
- Department of Clinical Genetics, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Sofia Maia
- Medical Genetics Unit, Hospital Pediátrico, Centro Hospitalar Universidade de Coimbra, Coimbra, Portugal
| | - Rong Mao
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Ruta Kalinauskiene
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
- Department of Medical Genetics, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Catherine Melver
- Division of Medical Genetics, Akron Children's Hospital, Akron, OH, USA
| | | | - Tara Montgomery
- Northern Genetics Service, Institute of Genetic Medicine, Newcastle upon Tyne NHS Foundation Trust, Newcastle, UK
| | - Manuela Morleo
- Telethon Institute of Genetics and Medicine, Pozzuoli, Napoli, Italy
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Napoli, Italy
| | - Constance Motter
- Division of Medical Genetics, Akron Children's Hospital, Akron, OH, USA
| | - Amanda S Openshaw
- ARUP Laboratories, Cytogenetics and Genomic Microarray, Salt Lake City, UT, USA
| | - Janice Cox Palumbos
- Division of Medical Genetics, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Aditi Shah Parikh
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
- Center for Human Genetics, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Yezmin Perilla-Young
- Division of Pediatric Genetics and Metabolism, University of North Carolina, Chapel Hill, NC, USA
| | - Cynthia M Powell
- Division of Pediatric Genetics and Metabolism, University of North Carolina, Chapel Hill, NC, USA
| | | | | | - Juliette Piard
- Centre de Génétique Humaine, Université de Franche-Comté, CHU, Besançon, France
| | - Rolph Pfundt
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marcello Scala
- U.O.C. Genetica Medica, IRCCS Istituto Giannina Gaslini, Genova, Italy
- Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Margaux Serey-Gaut
- Centre de Génétique Humaine, Université de Franche-Comté, CHU, Besançon, France
- Centre de Recherche en Audiologie, Hôpital Necker, AP-HP. CUP, Paris, France
| | - Deborah Shears
- Oxford Centre for Genomic Medicine, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Anne Slavotinek
- Division of Medical Genetics, Department of Pediatrics, Benioff Children's Hospital, University of California, San Francisco, CA, USA
- Division of Human Genetics, Cincinnati Children's Hospital, and Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Mohnish Suri
- Nottingham Clinical Genetics Service; Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Claire Turner
- Clinical Genetics, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - Tatiana Tvrdik
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Karin Weiss
- Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
- Genetics Institute, Rambam Health Care Campus, Haifa, Israel
| | | | - Marcella Zollino
- Dipartimento Universitario Scienze della Vita e Sanità Pubblica, Sezione di Medicina Genomica, Università Cattolica Sacro Cuore, Roma, Italy
- Genetica Medica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Tzung-Chien Hsieh
- Institute for Genomic Statistics and Bioinformatics, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Bert B A de Vries
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Francois Guillemot
- Neural Stem Cell Biology Laboratory, The Francis Crick Institute, London, UK
| | - William B Dobyns
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
- Division of Genetics and Metabolism, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - David Viskochil
- Division of Medical Genetics, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Cristina Dias
- Department of Medical Genetics, Guy's and St. Thomas' NHS Foundation Trust, London, UK.
- North East Thames Regional Genetics Service, Great Ormond Street Hospital for Children, Great Ormond Street, London, UK.
- Neural Stem Cell Biology Laboratory, The Francis Crick Institute, London, UK.
- Department of Medical & Molecular Genetics, School of Basic and Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK.
| |
Collapse
|
20
|
Ruiz-Gutierrez N, Dupas J, Auquier E, Barbarin-Bocahu I, Gaudon-Plesse C, Saveanu C, Graille M, Le Hir H. RNA anchoring of Upf1 facilitates recruitment of Dcp2 in the NMD decapping complex. Nucleic Acids Res 2025; 53:gkaf160. [PMID: 40071934 PMCID: PMC11897886 DOI: 10.1093/nar/gkaf160] [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: 10/29/2024] [Revised: 02/06/2025] [Accepted: 02/18/2025] [Indexed: 03/15/2025] Open
Abstract
Upf1 RNA helicase is a pivotal factor in the conserved nonsense-mediated mRNA decay (NMD) process. Upf1 is responsible for coordinating the recognition of premature termination codons (PTCs) in a translation-dependent manner and subsequently triggering mRNA degradation. Multiple factors assist Upf1 during these two consecutive steps. In Saccharomyces cerevisiae, Upf2 and Upf3 associated with Upf1 (Upf1-2/3) contribute to PTC recognition but are absent from the Upf1-decapping complex that includes Nmd4, Ebs1, Dcp1, and Dcp2. Despite their importance for NMD, the organization and dynamics of these Upf1-containing complexes remain unclear. Using recombinant proteins, here we show how distinct domains of Upf1 make direct contacts with Dcp1/Dcp2, Nmd4, and Ebs1. These proteins also bind to each other, forming an extended network of interactions within the Upf1-decapping complex. Dcp2 and Upf2 compete for the same binding site on the N-terminal CH domain of Upf1, which explains the presence of two mutually exclusive Upf1-containing complexes in cells. Our data demonstrate that Nmd4-assisted recruitment of Upf1 promotes anchoring of the decapping enzyme to NMD targets.
Collapse
Affiliation(s)
- Nadia Ruiz-Gutierrez
- Institut de Biologie de l’Ecole Normale Supérieure (IBENS), Ecole normale supérieure, CNRS, INSERM, PSL Research University, 46 rue d’Ulm, 75005 Paris, France
| | - Jeanne Dupas
- Institut de Biologie de l’Ecole Normale Supérieure (IBENS), Ecole normale supérieure, CNRS, INSERM, PSL Research University, 46 rue d’Ulm, 75005 Paris, France
| | - Elvire Auquier
- Institut de Biologie de l’Ecole Normale Supérieure (IBENS), Ecole normale supérieure, CNRS, INSERM, PSL Research University, 46 rue d’Ulm, 75005 Paris, France
| | - Irène Barbarin-Bocahu
- Laboratoire de Biologie Structurale de la Cellule (BIOC), CNRS, Ecole polytechnique, Institut Polytechnique de Paris, 91120 Palaiseau, France
| | - Claudine Gaudon-Plesse
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR 7104, INSERM, U1258, Université de Strasbourg, Illkirch, France
| | - Cosmin Saveanu
- Institut Pasteur, Université Paris Cité, Unité de Biologie des ARN des Pathogènes Fongiques, 75015 Paris, France
| | - Marc Graille
- Laboratoire de Biologie Structurale de la Cellule (BIOC), CNRS, Ecole polytechnique, Institut Polytechnique de Paris, 91120 Palaiseau, France
| | - Hervé Le Hir
- Institut de Biologie de l’Ecole Normale Supérieure (IBENS), Ecole normale supérieure, CNRS, INSERM, PSL Research University, 46 rue d’Ulm, 75005 Paris, France
| |
Collapse
|
21
|
Cook AL, Sur S, Dobbyn L, Watson E, Cohen JD, Ptak B, Lee BS, Paul S, Hsiue E, Popoli M, Vogelstein B, Papadopoulos N, Bettegowda C, Gabrielson K, Zhou S, Kinzler KW, Wyhs N. Identification of nonsense-mediated decay inhibitors that alter the tumor immune landscape. eLife 2025; 13:RP95952. [PMID: 39960487 PMCID: PMC11832170 DOI: 10.7554/elife.95952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025] Open
Abstract
Despite exciting developments in cancer immunotherapy, its broad application is limited by the paucity of targetable antigens on the tumor cell surface. As an intrinsic cellular pathway, nonsense-mediated decay (NMD) conceals neoantigens through the destruction of the RNA products from genes harboring truncating mutations. We developed and conducted a high-throughput screen, based on the ratiometric analysis of transcripts, to identify critical mediators of NMD in human cells. This screen implicated disruption of kinase SMG1's phosphorylation of UPF1 as a potential disruptor of NMD. This led us to design a novel SMG1 inhibitor, KVS0001, that elevates the expression of transcripts and proteins resulting from human and murine truncating mutations in vitro and murine cells in vivo. Most importantly, KVS0001 concomitantly increased the presentation of immune-targetable human leukocyte antigens (HLA) class I-associated peptides from NMD-downregulated proteins on the surface of human cancer cells. KVS0001 provides new opportunities for studying NMD and the diseases in which NMD plays a role, including cancer and inherited diseases.
Collapse
Affiliation(s)
- Ashley L Cook
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
- Cellular and Molecular Medicine Graduate Program, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Surojit Sur
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
- Sidney Kimmel Cancer Center, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Laura Dobbyn
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
| | - Evangeline Watson
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Joshua D Cohen
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
- Sidney Kimmel Cancer Center, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Blair Ptak
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
| | - Bum Seok Lee
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
| | - Suman Paul
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
- Sidney Kimmel Cancer Center, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Emily Hsiue
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Maria Popoli
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Bert Vogelstein
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
- Cellular and Molecular Medicine Graduate Program, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
- Sidney Kimmel Cancer Center, Johns Hopkins University School of MedicineBaltimoreUnited States
- Howard Hughes Medical Institute, Johns Hopkins University School of MedicineBaltimoreUnited States
- Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Nickolas Papadopoulos
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
- Sidney Kimmel Cancer Center, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Chetan Bettegowda
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
- Cellular and Molecular Medicine Graduate Program, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
- Sidney Kimmel Cancer Center, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Neurosurgery, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Kathy Gabrielson
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
- Sidney Kimmel Cancer Center, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Shibin Zhou
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
- Sidney Kimmel Cancer Center, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Kenneth W Kinzler
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
- Cellular and Molecular Medicine Graduate Program, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
- Sidney Kimmel Cancer Center, Johns Hopkins University School of MedicineBaltimoreUnited States
- Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Nicolas Wyhs
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
- Sidney Kimmel Cancer Center, Johns Hopkins University School of MedicineBaltimoreUnited States
| |
Collapse
|
22
|
Das R, Panigrahi GK. Messenger RNA Surveillance: Current Understanding, Regulatory Mechanisms, and Future Implications. Mol Biotechnol 2025; 67:393-409. [PMID: 38411790 DOI: 10.1007/s12033-024-01062-4] [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: 10/10/2023] [Accepted: 01/02/2024] [Indexed: 02/28/2024]
Abstract
Nonsense-mediated mRNA decay (NMD) is an evolutionarily conserved surveillance mechanism in eukaryotes primarily deployed to ensure RNA quality control by eliminating aberrant transcripts and also involved in modulating the expression of several physiological transcripts. NMD, the mRNA surveillance pathway, is a major form of gene regulation in eukaryotes. NMD serves as one of the most significant quality control mechanisms as it primarily scans the newly synthesized transcripts and differentiates the aberrant and non-aberrant transcripts. The synthesis of truncated proteins is restricted, which would otherwise lead to cellular dysfunctions. The up-frameshift factors (UPFs) play a central role in executing the NMD event, largely by recognizing and recruiting multiple protein factors that result in the decay of non-physiological mRNAs. NMD exhibits astounding variability in its ability across eukaryotes in an array of pathological and physiological contexts. The detailed understanding of NMD and the underlying molecular mechanisms remains blurred. This review outlines our current understanding of NMD, in regulating multifaceted cellular events during development and disease. It also attempts to identify unanswered questions that deserve further investigation.
Collapse
Affiliation(s)
- Rutupurna Das
- Department of Zoology, School of Applied Sciences, Centurion University of Technology and Management, Jatni, Khordha, Odisha, India
| | - Gagan Kumar Panigrahi
- Department of Zoology, School of Applied Sciences, Centurion University of Technology and Management, Jatni, Khordha, Odisha, India.
| |
Collapse
|
23
|
Sadakierska-Chudy A, Szymanowski P, Szepieniec WK, Boniewska-Bernacka E, Pollak A. Whole Exome Sequencing Reveals Candidate Variants in Ion Channel Genes for Pelvic Muscle Dysfunction in Young Females with a Family History. Int Urogynecol J 2025; 36:457-468. [PMID: 39833541 DOI: 10.1007/s00192-025-06048-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 12/24/2024] [Indexed: 01/22/2025]
Abstract
INTRODUCTION AND HYPOTHESIS Pelvic floor dysfunction usually results in pelvic organ prolapse (POP) and/or urinary incontinence. In women, several factors, including pregnancy and vaginal delivery, can affect pelvic muscle conditions. The aim of the study was to perform a genetic analysis in young women with a family history of pelvic floor dysfunction to find potentially harmful variants or variants that increase the risk of developing pelvic floor disorders. METHODS We employed whole exome sequencing to test ten young women with pelvic floor muscle dysfunction (along with their parents) and a family history. The average age of symptoms was 29.1 (± 3.98) years old, soon after their first delivery. RESULTS In five out of ten patients, trio-based WES analysis revealed potentially pathogenic, causative nonsense variants in ion channel genes, including ATP1A4, CLCN1, GRIN2C, and ORAI1, as well as missense variants in PIEZO1 and RYR1. Additionally, some of these patients had variants in genes related to muscle function (MUSK) and connective tissue disorder (FKBP14, p.Glu122ArgfsTer7). The variants found in this study, such as CLCN1 (p.Arg894Ter) and MUSK (p.Val790Met), have already been associated with neuromuscular channelopathy and severe muscle weakness. CONCLUSIONS The identified candidate genes encode mainly proteins involved in electrical action potential and mechanical muscle contraction. The results suggest that the identified genetic variants may result in skeletal muscle ion channelopathies that affect muscle function, gradually leading to muscle hypotonia and weakness.
Collapse
Affiliation(s)
- Anna Sadakierska-Chudy
- Department of Genetics, Faculty of Medicine, Collegium Medicum, Andrzej Frycz Modrzewski Krakow University, Gustawa Herlinga-Grudzinskiego 1, 30-705, Krakow, Poland.
| | - Paweł Szymanowski
- Department of Gynecology and Urogynecology, Faculty of Medicine, Collegium Medicum, Andrzej Frycz Modrzewski Krakow University, Gustawa Herlinga-Grudzinskiego 1, 30-705, Krakow, Poland
| | - Wioletta Katarzyna Szepieniec
- Department of Gynecology and Urogynecology, Faculty of Medicine, Collegium Medicum, Andrzej Frycz Modrzewski Krakow University, Gustawa Herlinga-Grudzinskiego 1, 30-705, Krakow, Poland
| | - Ewa Boniewska-Bernacka
- Institute of Medical Sciences, Department of Biology and Genetics, Faculty of Medicine, University of Opole, Oleska 48, 45-052, Opole, Poland
| | - Agnieszka Pollak
- Department of Medical Genetics, Warsaw Medical University, Pawinskiego 3C, 02-106, Warsaw, Poland
| |
Collapse
|
24
|
Liu Y, Elmas A, Huang KL. Mutation impact on mRNA versus protein expression across human cancers. Gigascience 2025; 14:giae113. [PMID: 39775839 PMCID: PMC11702362 DOI: 10.1093/gigascience/giae113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/13/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Cancer mutations are often assumed to alter proteins, thus promoting tumorigenesis. However, how mutations affect protein expression-in addition to gene expression-has rarely been systematically investigated. This is significant as mRNA and protein levels frequently show only moderate correlation, driven by factors such as translation efficiency and protein degradation. Proteogenomic datasets from large tumor cohorts provide an opportunity to systematically analyze the effects of somatic mutations on mRNA and protein abundance and identify mutations with distinct impacts on these molecular levels. RESULTS We conduct a comprehensive analysis of mutation impacts on mRNA- and protein-level expressions of 953 cancer cases with paired genomics and global proteomic profiling across 6 cancer types. Protein-level impacts are validated for 47.2% of the somatic expression quantitative trait loci (seQTLs), including CDH1 and MSH3 truncations, as well as other mutations from likely "long-tail" driver genes. Devising a statistical pipeline for identifying somatic protein-specific QTLs (spsQTLs), we reveal several gene mutations, including NF1 and MAP2K4 truncations and TP53 missenses showing disproportional influence on protein abundance not readily explained by transcriptomics. Cross-validating with data from massively parallel assays of variant effects (MAVE), TP53 missenses associated with high tumor TP53 proteins are more likely to be experimentally confirmed as functional. CONCLUSION This study reveals that somatic mutations can exhibit distinct impacts on mRNA and protein levels, underscoring the necessity of integrating proteogenomic data to comprehensively identify functionally significant cancer mutations. These insights provide a framework for prioritizing mutations for further functional validation and therapeutic targeting.
Collapse
Affiliation(s)
- Yuqi Liu
- Department of Genetics and Genomic Sciences, Department of Artificial Intelligence and Human Health, Center for Transformative Disease Modeling, Tisch Cancer Institute, Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Abdulkadir Elmas
- Department of Genetics and Genomic Sciences, Department of Artificial Intelligence and Human Health, Center for Transformative Disease Modeling, Tisch Cancer Institute, Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kuan-lin Huang
- Department of Genetics and Genomic Sciences, Department of Artificial Intelligence and Human Health, Center for Transformative Disease Modeling, Tisch Cancer Institute, Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
25
|
Bhattarai D, Banday AZ, Sharda S, Patra PK, Walter JE, Sullivan KE. Novel Inherited N-terminus TAP1 Variants and Severe Clinical Manifestations- Are Genotype-Phenotype Correlations Emerging? J Clin Immunol 2025; 45:63. [PMID: 39751995 DOI: 10.1007/s10875-024-01856-w] [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: 08/20/2024] [Accepted: 12/26/2024] [Indexed: 01/04/2025]
Abstract
Major histocompatibility complex class I deficiency results from deleterious biallelic variants in TAP1, TAP2, TAPBP, and B2M genes. Only a few patients with variant-curated TAP1 deficiency (TAP1D) have been reported in the literature and the clinical phenotype has been variable with an emphasis on autoimmune and inflammatory complications. We report TAP1D in a Nepalese girl with a severe clinical phenotype with serious viral infections at a very young age. A novel frameshift termination variant near the protein's amino (N-) terminal was found. Variants in exon 1 of the TAP1 gene (as in our case) have not been reported previously. We also perform a brief review of TAP1D that hints at potential genotype-phenotype correlations. However, these findings need to be interpreted with due prudence given the small number of patients with TAP1D reported thus far.
Collapse
Affiliation(s)
- Dharmagat Bhattarai
- Advanced Center for Immunology and Rheumatology, Kathmandu, 44600, Nepal.
- Pediatric Clinical Immunology and Rheumatology, Division of Immunology & Pediatric Rheumatology, Advanced Centre for Immunology and Rheumatology, Kathmandu, Nepal.
| | - Aaqib Zaffar Banday
- Department of Pediatrics, Government Medical College (GMC) Srinagar, Srinagar, Kashmir, India
| | - Sheetal Sharda
- Department of Medical Genetics, Neuberg Center for Genomic Medicine, Ahmedabad, India
| | - Pratap Kumar Patra
- Department of Pediatrics, All India Institute of Medical Sciences Patna, Patna, Bihar, India
| | - Jolan E Walter
- Division of Allergy & Immunology, University of South Florida at Johns' Hopkins All Children's Hospital, St. Petersburg, 33701, USA
| | - Kathleen E Sullivan
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, 19104, USA
| |
Collapse
|
26
|
Embree CM, Stephanou A, Singh G. Direct and indirect effects of spliceosome disruption compromise gene regulation by Nonsense-Mediated mRNA Decay. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.27.630533. [PMID: 39763844 PMCID: PMC11703147 DOI: 10.1101/2024.12.27.630533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Pre-mRNA splicing, carried out in the nucleus by a large ribonucleoprotein machine known as the spliceosome, is functionally and physically coupled to the mRNA surveillance pathway in the cytoplasm called nonsense mediated mRNA decay (NMD). The NMD pathway monitors for premature translation termination signals, which can result from alternative splicing, by relying on the exon junction complex (EJC) deposited on exon-exon junctions by the spliceosome. Recently, multiple genetic screens in human cell lines have identified numerous spliceosome components as putative NMD factors. Using publicly available RNA-seq datasets from K562 and HepG2 cells depleted of 18 different spliceosome components, we find that natural NMD targeted mRNA isoforms are upregulated when members of the catalytic spliceosome are reduced. While some of this increase could be due to widespread pleiotropic effects of spliceosome dysfunction (e.g., reduced expression of NMD factors due to mis-splicing of their mRNAs), we identify that AQR, SF3B1, SF3B4 and CDC40 may have a more direct role in NMD. We also test the hypothesis that increased production of novel NMD substrates may overwhelm the pathway to find a direct correlation between the amount of novel NMD substrates detected and the degree of NMD inhibition observed. Finally, similar transcriptome alterations and NMD substrate upregulation are also observed in cells treated with spliceosome inhibitors and in cells derived from retinitis pigmentosa patients with mutations in PRPF8 and PRPF31. Overall, our results show that regardless of the cause, spliceosome disruption upregulates a broad set of NMD targets, which could contribute to cellular dysfunction in spliceosomopathies.
Collapse
Affiliation(s)
- Caleb M Embree
- Department of Molecular Genetics, Center for RNA Biology, The Ohio State University, Columbus, OH, 43210
| | - Andreas Stephanou
- Department of Molecular Genetics, Center for RNA Biology, The Ohio State University, Columbus, OH, 43210
| | - Guramrit Singh
- Department of Molecular Genetics, Center for RNA Biology, The Ohio State University, Columbus, OH, 43210
| |
Collapse
|
27
|
Boo SH, Shin MK, Hwang HJ, Hwang H, Chang S, Kim T, Baek D, Kim YK. Circular RNAs trigger nonsense-mediated mRNA decay. Mol Cell 2024; 84:4862-4877.e7. [PMID: 39667933 DOI: 10.1016/j.molcel.2024.11.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 08/13/2024] [Accepted: 11/15/2024] [Indexed: 12/14/2024]
Abstract
Circular RNAs (circRNAs) are covalently closed single-stranded RNAs produced predominantly through a back-splicing process. They play regulatory roles in various biological and physiological processes; however, the molecular mechanisms by which circRNAs operate remain unclear. Herein, we demonstrate that circRNAs facilitate rapid mRNA degradation through RNA-RNA interactions between circRNAs and the 3' untranslated regions (3' UTRs) of mRNAs. This interaction positions the exon-junction complexes (EJCs), deposited onto circRNAs by back-splicing, near the 3' UTRs of the mRNAs, thereby leading to EJC-dependent nonsense-mediated mRNA decay (NMD), a process we describe as circRNA-induced NMD (circNMD). Our transcriptomic analysis reveals hundreds of potential circNMD candidates, and the biological importance of circNMD in cellular apoptosis is validated. We also demonstrate that exogenously expressed circRNAs designed to interact with the 3' UTRs of endogenous mRNAs significantly downregulate the mRNA levels. Collectively, our observations provide compelling molecular evidence for circNMD and its potential therapeutic application in selective mRNA downregulation.
Collapse
Affiliation(s)
- Sung Ho Boo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Min-Kyung Shin
- Division of Life Sciences, Korea University, Seoul 02841, Republic of Korea
| | - Hyun Jung Hwang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Hyeonseo Hwang
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Sunwoo Chang
- Department of Life Science and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 03760, Republic of Korea
| | - TaeSoo Kim
- Department of Life Science and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Daehyun Baek
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Yoon Ki Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea.
| |
Collapse
|
28
|
Dawood M, Heavner B, Wheeler MM, Ungar RA, LoTempio J, Wiel L, Berger S, Bernstein JA, Chong JX, Délot EC, Eichler EE, Gibbs RA, Lupski JR, Shojaie A, Talkowski ME, Wagner AH, Wei CL, Wellington C, Wheeler MT, Carvalho CMB, Gifford CA, May S, Miller DE, Rehm HL, Sedlazeck FJ, Vilain E, O'Donnell-Luria A, Posey JE, Chadwick LH, Bamshad MJ, Montgomery SB. GREGoR: Accelerating Genomics for Rare Diseases. ARXIV 2024:arXiv:2412.14338v1. [PMID: 39764392 PMCID: PMC11702807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Rare diseases are collectively common, affecting approximately one in twenty individuals worldwide. In recent years, rapid progress has been made in rare disease diagnostics due to advances in DNA sequencing, development of new computational and experimental approaches to prioritize genes and genetic variants, and increased global exchange of clinical and genetic data. However, more than half of individuals suspected to have a rare disease lack a genetic diagnosis. The Genomics Research to Elucidate the Genetics of Rare Diseases (GREGoR) Consortium was initiated to study thousands of challenging rare disease cases and families and apply, standardize, and evaluate emerging genomics technologies and analytics to accelerate their adoption in clinical practice. Further, all data generated, currently representing ~7500 individuals from ~3000 families, is rapidly made available to researchers worldwide via the Genomic Data Science Analysis, Visualization, and Informatics Lab-space (AnVIL) to catalyze global efforts to develop approaches for genetic diagnoses in rare diseases (https://gregorconsortium.org/data). The majority of these families have undergone prior clinical genetic testing but remained unsolved, with most being exome-negative. Here, we describe the collaborative research framework, datasets, and discoveries comprising GREGoR that will provide foundational resources and substrates for the future of rare disease genomics.
Collapse
Affiliation(s)
- Moez Dawood
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA
| | - Ben Heavner
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Marsha M Wheeler
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Rachel A Ungar
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA, USA
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA, USA
- Stanford Center for Biomedical Ethics, School of Medicine, Stanford University, Stanford, CA, USA
| | - Jonathan LoTempio
- Institute for Clinical and Translational Science, University of California, Irvine, CA, USA
| | - Laurens Wiel
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA, USA
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA, USA
- Division of Cardiovascular Medicine, School of Medicine, Stanford University, Stanford, CA, USA
| | - Seth Berger
- Division of Genetics and Metabolism, Children's National Rare Disease Institute, Washington, DC, USA
- Center for Genetic Medicine Research, Children's National Rare Disease Institute, Washington, DC, USA
- Department of Genomics and Precision Medicine, George Washington University, Washington, DC, USA
| | - Jonathan A Bernstein
- Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA, USA
| | - Jessica X Chong
- Department of Pediatrics, Dvision of Genetic Medicine, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, WA, USA
| | - Emmanuèle C Délot
- Institute for Clinical and Translational Science, University of California, Irvine, CA, USA
| | - Evan E Eichler
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | - Richard A Gibbs
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - James R Lupski
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Ali Shojaie
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Michael E Talkowski
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Bioinformatics and Integrative Genomics, Harvard Medical School, Boston, MA, USA
| | - Alex H Wagner
- Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
- Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Chia-Lin Wei
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Christopher Wellington
- Office of Genomic Data Science, National Human Genome Research Institute, Bethesda, MD, USA
| | - Matthew T Wheeler
- Division of Cardiovascular Medicine, School of Medicine, Stanford University, Stanford, CA, USA
| | | | - Casey A Gifford
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA, USA
- Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA, USA
- Basic Science and Engineering Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Stanford, CA, USA
| | - Susanne May
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Danny E Miller
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Heidi L Rehm
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Fritz J Sedlazeck
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
- Department of Computer Science, Rice University, Houston, TX, USA
| | - Eric Vilain
- Institute for Clinical and Translational Science, University of California, Irvine, CA, USA
| | - Anne O'Donnell-Luria
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, MA, USA
- Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jennifer E Posey
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Lisa H Chadwick
- Division of Genome Sciences, National Human Genome Research Institute, Bethesda, MD, USA
| | - Michael J Bamshad
- Department of Pediatrics, Dvision of Genetic Medicine, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, WA, USA
- Department of Pediatrics, Division of Genetic Medicine, Seattle Children's Hospital, Seattle, WA, USA
| | - Stephen B Montgomery
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA, USA
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| |
Collapse
|
29
|
Taki T, Morimoto K, Mizuno S, Kuno A. KOnezumi-AID: Automation Software for Efficient Multiplex Gene Knockout Using Target-AID. Int J Mol Sci 2024; 25:13500. [PMID: 39769261 PMCID: PMC11679502 DOI: 10.3390/ijms252413500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/13/2024] [Accepted: 12/15/2024] [Indexed: 01/11/2025] Open
Abstract
With the groundbreaking advancements in genome editing technologies, particularly CRISPR-Cas9, creating knockout mutants has become highly efficient. However, the CRISPR-Cas9 system introduces DNA double-strand breaks, increasing the risk of chromosomal rearrangements and posing a major obstacle to simultaneous multiple gene knockout. Base-editing systems, such as Target-AID, are safe alternatives for precise base modifications without requiring DNA double-strand breaks, serving as promising solutions for existing challenges. Nevertheless, the absence of adequate tools to support Target-AID-based gene knockout highlights the need for a comprehensive system to design guide RNAs (gRNAs) for the simultaneous knockout of multiple genes. Here, we aimed to develop KOnezumi-AID, a command-line tool for gRNA design for Target-AID-mediated genome editing. KOnezumi-AID facilitates gene knockout by inducing the premature termination codons or promoting exon skipping, thereby generating experiment-ready gRNA designs for mouse and human genomes. Additionally, KOnezumi-AID exhibits batch processing capacity, enabling rapid and precise gRNA design for large-scale genome editing, including CRISPR screening. In summary, KOnezumi-AID is an efficient and user-friendly tool for gRNA design, streamlining genome editing workflows and advancing gene knockout research.
Collapse
Affiliation(s)
- Taito Taki
- College of Biological Sciences, University of Tsukuba, Tsukuba 305-8572, Japan;
| | - Kento Morimoto
- Doctoral Program in Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba 305-8575, Japan;
| | - Seiya Mizuno
- Laboratory Animal Resource Center, Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan
| | - Akihiro Kuno
- Laboratory Animal Resource Center, Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan
- Department of Anatomy and Embryology, Institute of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan
| |
Collapse
|
30
|
Dawood M, Fayer S, Pendyala S, Post M, Kalra D, Patterson K, Venner E, Muffley LA, Fowler DM, Rubin AF, Posey JE, Plon SE, Lupski JR, Gibbs RA, Starita LM, Robles-Espinoza CD, Coyote-Maestas W, Gallego Romero I. Using multiplexed functional data to reduce variant classification inequities in underrepresented populations. Genome Med 2024; 16:143. [PMID: 39627863 PMCID: PMC11616159 DOI: 10.1186/s13073-024-01392-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 10/03/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Multiplexed Assays of Variant Effects (MAVEs) can test all possible single variants in a gene of interest. The resulting saturation-style functional data may help resolve variant classification disparities between populations, especially for Variants of Uncertain Significance (VUS). METHODS We analyzed clinical significance classifications in 213,663 individuals of European-like genetic ancestry versus 206,975 individuals of non-European-like genetic ancestry from All of Us and the Genome Aggregation Database. Then, we incorporated clinically calibrated MAVE data into the Clinical Genome Resource's Variant Curation Expert Panel rules to automate VUS reclassification for BRCA1, TP53, and PTEN. RESULTS Using two orthogonal statistical approaches, we show a higher prevalence (p ≤ 5.95e - 06) of VUS in individuals of non-European-like genetic ancestry across all medical specialties assessed in all three databases. Further, in the non-European-like genetic ancestry group, higher rates of Benign or Likely Benign and variants with no clinical designation (p ≤ 2.5e - 05) were found across many medical specialties, whereas Pathogenic or Likely Pathogenic assignments were increased in individuals of European-like genetic ancestry (p ≤ 2.5e - 05). Using MAVE data, we reclassified VUS in individuals of non-European-like genetic ancestry at a significantly higher rate in comparison to reclassified VUS from European-like genetic ancestry (p = 9.1e - 03) effectively compensating for the VUS disparity. Further, essential code analysis showed equitable impact of MAVE evidence codes but inequitable impact of allele frequency (p = 7.47e - 06) and computational predictor (p = 6.92e - 05) evidence codes for individuals of non-European-like genetic ancestry. CONCLUSIONS Generation of saturation-style MAVE data should be a priority to reduce VUS disparities and produce equitable training data for future computational predictors.
Collapse
Affiliation(s)
- Moez Dawood
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA.
| | - Shawn Fayer
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Sriram Pendyala
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA, USA
| | - Mason Post
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Divya Kalra
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Karynne Patterson
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Eric Venner
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Lara A Muffley
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Douglas M Fowler
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Alan F Rubin
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Jennifer E Posey
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Sharon E Plon
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - James R Lupski
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Texas Children's Hospital, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Richard A Gibbs
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Lea M Starita
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Carla Daniela Robles-Espinoza
- Laboratorio Internacional de Investigación Sobre El Genoma Humano, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, Qro, Mexico
- CASM, Wellcome Sanger Institute, Hinxton, UK
| | - Willow Coyote-Maestas
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, USA.
- Quantitative Biosciences Institute, University of California, San Francisco, USA.
| | - Irene Gallego Romero
- Human Genomics and Evolution, St Vincent's Institute of Medical Research, Fitzroy, 3065, Australia.
- School of BioSciences and Melbourne Integrative Genomics, The University of Melbourne, Royal Parade, Parkville, 3010, Australia.
- Center for Genomics, Evolution and Medicine, Institute of Genomics, University of Tartu, Riia 23B, 51010, Tartu, Estonia.
- Mary MacKillop Institute for Health Research, Australian Catholic University, Fitzroy, Australia.
| |
Collapse
|
31
|
Patro AK, Panigrahi GK, Majumder S, Das R, Sahoo A. Nonsense-mediated mRNA decay: Physiological significance, mechanistic insights and future implications. Pathol Res Pract 2024; 264:155677. [PMID: 39486251 DOI: 10.1016/j.prp.2024.155677] [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: 09/13/2024] [Revised: 10/20/2024] [Accepted: 10/25/2024] [Indexed: 11/04/2024]
Abstract
Nonsense-mediated mRNA decay (NMD) is a quality control mechanism that detects and degrades premature aberrant transcripts and importantly, it also takes part in gene expression regulation by regulating the endogenous transcripts. NMD distinguishes aberrant and non-aberrant transcript by looking after the NMD signatures such as long 3' UTR. NMD modulates cellular surveillance and eliminates the plausible synthesis of truncated proteins as because if the aberrant mRNA escapes the surveillance pathway it can lead to potential negative phenotype resulting in genetic diseases. NMD involves multiple proteins and any alteration or mutation within these proteins results in various pathophysiological consequences. NMD plays a complex role in cancer, it can either aggravate or downregulates the tumour. Some tumours agitate NMD to deteriorate mRNAs encoding tumour suppressor proteins, stress response proteins and neoantigens. In other case, tumours suppress the NMD to encourage the expression of oncoproteins for tumour growth and survival. In this review, we have shed light on the core and associated proteins of NMD, further summarized the mechanism of the NMD pathway and also described the implications of mutations in NMD factors resulting in severe pathological conditions including neurodevelopmental disorder, effects on male sterility and cancer. Understanding the complexities of NMD regulation and its interaction with other cellular processes can lead to the development of new interventions for various diseases. This review summarizes the current understanding of NMD and its role in controlling various cellular processes in both development and disease.
Collapse
Affiliation(s)
- Asish Kumar Patro
- Department of Zoology, School of Applied Sciences, Centurion University of Technology and Management, Khordha, Odisha, India
| | - Gagan Kumar Panigrahi
- Department of Zoology, School of Applied Sciences, Centurion University of Technology and Management, Khordha, Odisha, India.
| | - Sanjoy Majumder
- Department of Zoology, School of Applied Sciences, Centurion University of Technology and Management, Khordha, Odisha, India
| | - Rutupurna Das
- Department of Zoology, School of Applied Sciences, Centurion University of Technology and Management, Khordha, Odisha, India
| | - Annapurna Sahoo
- Department of Zoology, School of Applied Sciences, Centurion University of Technology and Management, Khordha, Odisha, India.
| |
Collapse
|
32
|
Huang C, Huang Y, Pan L, Li L, Ling X, Wang C, Xiao Q, Zhai N, Long Y, Mo W, Lin F, Huang Y. A novel duplication mutation of SLC2A1 gene causing glucose transporter 1 deficiency syndrome. Gene 2024; 928:148762. [PMID: 39009233 DOI: 10.1016/j.gene.2024.148762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 07/05/2024] [Accepted: 07/10/2024] [Indexed: 07/17/2024]
Affiliation(s)
- Chaoyu Huang
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Nanning 530021, Guangxi, China
| | - Yunhua Huang
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Nanning 530021, Guangxi, China
| | - Liqiu Pan
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Nanning 530021, Guangxi, China
| | - Linlin Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Nanning 530021, Guangxi, China
| | - Xiaoting Ling
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Nanning 530021, Guangxi, China
| | - Chenghan Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Nanning 530021, Guangxi, China
| | - Qingxing Xiao
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Nanning 530021, Guangxi, China
| | - Ningneng Zhai
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Nanning 530021, Guangxi, China
| | - Yan Long
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Nanning 530021, Guangxi, China
| | - Wuning Mo
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Nanning 530021, Guangxi, China.
| | - Faquan Lin
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Nanning 530021, Guangxi, China.
| | - Yifang Huang
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Nanning 530021, Guangxi, China.
| |
Collapse
|
33
|
Sidoti D, Margotta V, Calosci D, Fiorentini E, Bacci C, Gensini F, Papi L, Montini M. Alu-Mediated Duplication and Deletion of Exon 11 Are Frequent Mechanisms of PALB2 Inactivation, Predisposing Individuals to Hereditary Breast-Ovarian Cancer Syndrome. Cancers (Basel) 2024; 16:4022. [PMID: 39682206 DOI: 10.3390/cancers16234022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVE Large genomic rearrangements of PALB2 gene, particularly deletions and duplications, have been linked to hereditary breast-ovarian cancer. Our research specifically focuses on delineating the intronic breakpoints associated with rearrangements of PALB2 exon 11, which is crucial for understanding the mechanisms underlying these genomic changes in patients with hereditary breast and ovarian syndrome. METHODS By using next-generation sequencing, we identified one duplication and three deletions of PALB2 exon 11, confirmed by Multiplex Ligation-Dependent Probe Amplification analysis. To assess the impact on transcription and potential splicing issues, reverse-transcription PCR was performed on patients' RNA. For the detailed characterization of intronic breakpoints, the primer walking approach and long-range PCR were implemented, followed by Sanger sequencing. RESULTS Our analysis revealed a tandem duplication of 5134 base pairs (bp) mediated by AluY repeats located in introns 10 and 11, respectively. Moreover, identical deletions were identified in three unrelated patients, encompassing an approximate 8050 bp region mediated by AluSx elements. Both genomic alterations resulted in a truncated PALB2 protein due to the introduction of a premature stop codon. CONCLUSIONS This study underscores the remarkable instability of intronic regions flanking exon 11 of PALB2 and identifies a previously unreported hotspot involving Alu repeats with very high sequence homology in introns 10 and 11 of the gene. Our findings suggest avenues for further research, such as investigating the prevalence of similar genomic rearrangements in larger cohorts and exploring functional studies to understand how these alterations contribute to hereditary breast cancer pathogenesis.
Collapse
Affiliation(s)
- Diletta Sidoti
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, 50139 Florence, Italy
| | - Valeria Margotta
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, 50139 Florence, Italy
| | - Diletta Calosci
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, 50139 Florence, Italy
| | - Erika Fiorentini
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, 50139 Florence, Italy
| | - Costanza Bacci
- Medical Genetics Unit, SOS Day-Service Palagi, USL Toscana Centro, 50122 Florence, Italy
| | - Francesca Gensini
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, 50139 Florence, Italy
| | - Laura Papi
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, 50139 Florence, Italy
| | - Marco Montini
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, 50139 Florence, Italy
| |
Collapse
|
34
|
Tilk S, Frydman J, Curtis C, Petrov DA. Cancers adapt to their mutational load by buffering protein misfolding stress. eLife 2024; 12:RP87301. [PMID: 39585785 PMCID: PMC11588338 DOI: 10.7554/elife.87301] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2024] Open
Abstract
In asexual populations that don't undergo recombination, such as cancer, deleterious mutations are expected to accrue readily due to genome-wide linkage between mutations. Despite this mutational load of often thousands of deleterious mutations, many tumors thrive. How tumors survive the damaging consequences of this mutational load is not well understood. Here, we investigate the functional consequences of mutational load in 10,295 human tumors by quantifying their phenotypic response through changes in gene expression. Using a generalized linear mixed model (GLMM), we find that high mutational load tumors up-regulate proteostasis machinery related to the mitigation and prevention of protein misfolding. We replicate these expression responses in cancer cell lines and show that the viability in high mutational load cancer cells is strongly dependent on complexes that degrade and refold proteins. This indicates that the upregulation of proteostasis machinery is causally important for high mutational burden tumors and uncovers new therapeutic vulnerabilities.
Collapse
Affiliation(s)
- Susanne Tilk
- Department of Biology, Stanford UniversityStanfordUnited States
| | - Judith Frydman
- Department of Biology, Stanford UniversityStanfordUnited States
| | - Christina Curtis
- Department of Medicine, Division of Oncology, Stanford University School of MedicineStanfordUnited States
- Department of Genetics, Stanford University School of MedicineStanfordUnited States
- Stanford Cancer Institute, Stanford University School of MedicineStanfordUnited States
| | - Dmitri A Petrov
- Department of Biology, Stanford UniversityStanfordUnited States
- Stanford Cancer Institute, Stanford University School of MedicineStanfordUnited States
| |
Collapse
|
35
|
Xiong X, Cesarato N, Gossmann Y, Wehner M, Kumar S, Thiele H, Demuth S, Oji V, Geyer M, Hamm H, Basmanav FB, Betz RC. A nonsense variant in KRT31 is associated with autosomal dominant monilethrix. Br J Dermatol 2024; 191:979-987. [PMID: 39026424 DOI: 10.1093/bjd/ljae298] [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: 03/27/2024] [Revised: 06/26/2024] [Accepted: 07/13/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND Monilethrix is a rare hereditary hair disorder that is characterized by a beaded hair shaft structure and increased hair fragility. Patients may also present with keratosis pilaris and nail changes. Research has identified three genes responsible for autosomal dominant monilethrix (KRT81, KRT83, KRT86) and one responsible for the autosomal recessive form (DSG4). OBJECTIVES To investigate the genetic basis of autosomal dominant monilethrix in families with no pathogenic variants in any of the known monilethrix genes, and to understand the mechanistic basis of variant pathogenicity using a cellular model. METHODS Nine affected individuals from four unrelated families were included. A clinical diagnosis of monilethrix was assigned based on clinical examination and/or trichoscopy. Exome sequencing was performed in six individuals to identify pathogenic variants; Sanger sequencing was used for co-segregation and haplotype analyses. Cell culture experiments [immunoblotting, immunofluorescence and reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR) analyses] were used to confirm variant pathogenicity, to determine the expression and subcellular localization of proteins, and to identify possible nonsense-mediated mRNA decay. RESULTS In six affected individuals with clinically suggested monilethrix, exome sequencing led to the identification of the nonsense variant c.1081G>T; p.(Glu361*) in KRT31, which was subsequently identified in other affected members of these families by Sanger sequencing. This variant led to the abolition of both the last three amino acids of the 2B subdomain and the complete C-terminal tail domain of keratin 31. Immunoblotting demonstrated that when co-expressed with its binding partner keratin 85, the truncated keratin 31 was still expressed, albeit less abundantly than the wildtype protein. Immunofluorescence revealed that p.(Glu361*) keratin 31 had an altered cytoskeletal localization and formed vesicular-like structures in the cell cytoplasm near the cell membrane. RT-qPCR analysis did not generate evidence for nonsense-mediated decay of the mutant transcript. CONCLUSIONS This study is the first to identify pathogenic variants in KRT31 as a cause of autosomal dominant monilethrix. This highlights the importance of hair keratin proteins in hair biology, and will increase the molecular diagnostic yield for rare ectodermal phenotypes of hair and nail tissues.
Collapse
Affiliation(s)
- Xing Xiong
- Institute of Human Genetics, University of Bonn, Medical Faculty and University Hospital Bonn, Bonn, Germany
| | - Nicole Cesarato
- Institute of Human Genetics, University of Bonn, Medical Faculty and University Hospital Bonn, Bonn, Germany
| | - Yasmina Gossmann
- Institute of Human Genetics, University of Bonn, Medical Faculty and University Hospital Bonn, Bonn, Germany
| | - Maria Wehner
- Institute of Human Genetics, University of Bonn, Medical Faculty and University Hospital Bonn, Bonn, Germany
| | - Sheetal Kumar
- Institute of Human Genetics, University of Bonn, Medical Faculty and University Hospital Bonn, Bonn, Germany
| | - Holger Thiele
- Cologne Center for Genomics, University of Cologne, Cologne, Germany
| | | | - Vinzenz Oji
- Department of Dermatology, University of Münster, Münster, Germany
| | - Matthias Geyer
- Institute of Structural Biology, University of Bonn, Medical Faculty and University Hospital Bonn, Bonn, Germany
| | - Henning Hamm
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - F Buket Basmanav
- Institute of Human Genetics, University of Bonn, Medical Faculty and University Hospital Bonn, Bonn, Germany
| | - Regina C Betz
- Institute of Human Genetics, University of Bonn, Medical Faculty and University Hospital Bonn, Bonn, Germany
| |
Collapse
|
36
|
Hu H, Tang J, Wang H, Guo X, Tu C, Li Z. The crosstalk between alternative splicing and circular RNA in cancer: pathogenic insights and therapeutic implications. Cell Mol Biol Lett 2024; 29:142. [PMID: 39550559 PMCID: PMC11568689 DOI: 10.1186/s11658-024-00662-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 11/05/2024] [Indexed: 11/18/2024] Open
Abstract
RNA splicing is a fundamental step of gene expression. While constitutive splicing removes introns and joins exons unbiasedly, alternative splicing (AS) selectively determines the assembly of exons and introns to generate RNA variants corresponding to the same transcript. The biogenesis of circular RNAs (circRNAs) is inextricably associated with AS. Back-splicing, the biogenic process of circRNA, is a special form of AS. In cancer, both AS and circRNA deviate from the original track. In the present review, we delve into the intricate interplay between AS and circRNAs in the context of cancer. The relationship between AS and circRNAs is intricate, where AS modulates the biogenesis of circRNAs and circRNAs in return regulate AS events. Beyond that, epigenetic and posttranscriptional modifications concurrently regulate AS and circRNAs. On the basis of this modality, we summarize current knowledge on how splicing factors and other RNA binding proteins regulate circRNA biogenesis, and how circRNAs interact with splicing factors to influence AS events. Specifically, the feedback loop regulation between circRNAs and AS events contributes greatly to oncogenesis and cancer progression. In summary, resolving the crosstalk between AS and circRNA will not only provide better insight into cancer biology but also provoke novel strategies to combat cancer.
Collapse
Affiliation(s)
- Hongkun Hu
- Department of Orthopaedics, Hunan Key Laboratory of Tumor Models and Individualized Medicine, Hunan Engineering Research Center of Artificial Intelligence-Based Medical Equipment, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Jinxin Tang
- Department of Orthopaedics, Hunan Key Laboratory of Tumor Models and Individualized Medicine, Hunan Engineering Research Center of Artificial Intelligence-Based Medical Equipment, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Hua Wang
- Department of Orthopaedics, Hunan Key Laboratory of Tumor Models and Individualized Medicine, Hunan Engineering Research Center of Artificial Intelligence-Based Medical Equipment, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Xiaoning Guo
- Department of Orthopaedics, Hunan Key Laboratory of Tumor Models and Individualized Medicine, Hunan Engineering Research Center of Artificial Intelligence-Based Medical Equipment, The Second Xiangya Hospital of Central South University, Changsha, 410011, China.
| | - Chao Tu
- Department of Orthopaedics, Hunan Key Laboratory of Tumor Models and Individualized Medicine, Hunan Engineering Research Center of Artificial Intelligence-Based Medical Equipment, The Second Xiangya Hospital of Central South University, Changsha, 410011, China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, 410011, China.
- Hunan Engineering Research Center of Artificial Intelligence-Based Medical Equipment, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China.
| | - Zhihong Li
- Department of Orthopaedics, Hunan Key Laboratory of Tumor Models and Individualized Medicine, Hunan Engineering Research Center of Artificial Intelligence-Based Medical Equipment, The Second Xiangya Hospital of Central South University, Changsha, 410011, China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, 410011, China.
- Hunan Engineering Research Center of Artificial Intelligence-Based Medical Equipment, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
37
|
Ma K, Huang S, Ng KK, Lake NJ, Joseph S, Xu J, Lek A, Ge L, Woodman KG, Koczwara KE, Cohen J, Ho V, O'Connor CL, Brindley MA, Campbell KP, Lek M. Saturation mutagenesis-reinforced functional assays for disease-related genes. Cell 2024; 187:6707-6724.e22. [PMID: 39326416 PMCID: PMC11568926 DOI: 10.1016/j.cell.2024.08.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 07/29/2024] [Accepted: 08/23/2024] [Indexed: 09/28/2024]
Abstract
Interpretation of disease-causing genetic variants remains a challenge in human genetics. Current costs and complexity of deep mutational scanning methods are obstacles for achieving genome-wide resolution of variants in disease-related genes. Our framework, saturation mutagenesis-reinforced functional assays (SMuRF), offers simple and cost-effective saturation mutagenesis paired with streamlined functional assays to enhance the interpretation of unresolved variants. Applying SMuRF to neuromuscular disease genes FKRP and LARGE1, we generated functional scores for all possible coding single-nucleotide variants, which aid in resolving clinically reported variants of uncertain significance. SMuRF also demonstrates utility in predicting disease severity, resolving critical structural regions, and providing training datasets for the development of computational predictors. Overall, our approach enables variant-to-function insights for disease genes in a cost-effective manner that can be broadly implemented by standard research laboratories.
Collapse
Affiliation(s)
- Kaiyue Ma
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA; Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China.
| | - Shushu Huang
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Kenneth K Ng
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Nicole J Lake
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Soumya Joseph
- Howard Hughes Medical Institute, Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, Department of Molecular Physiology and Biophysics and Department of Neurology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Jenny Xu
- Yale University, New Haven, CT, USA
| | - Angela Lek
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA; Muscular Dystrophy Association, Chicago, IL, USA
| | - Lin Ge
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA; Department of Neurology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Keryn G Woodman
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | | | - Justin Cohen
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Vincent Ho
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | | | - Melinda A Brindley
- Department of Infectious Diseases, Department of Population Health, University of Georgia, Athens, GA, USA
| | - Kevin P Campbell
- Howard Hughes Medical Institute, Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, Department of Molecular Physiology and Biophysics and Department of Neurology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Monkol Lek
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
38
|
Kim YG, Kang H, Lee B, Jang HJ, Park JH, Ha C, Park H, Kim JW. A spectrum of nonsense-mediated mRNA decay efficiency along the degree of mutational constraint. Commun Biol 2024; 7:1461. [PMID: 39511375 PMCID: PMC11544006 DOI: 10.1038/s42003-024-07136-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 10/24/2024] [Indexed: 11/15/2024] Open
Abstract
Despite its importance for regulating gene expression, nonsense-mediated mRNA decay (NMD) remains poorly understood. Here, we extend the findings of a previous landmark study that proposed several factors associated with NMD efficiency using matched genome and transcriptome data from The Cancer Genome Atlas Program (TCGA) by incorporating additional data including Genotype-Tissue Expression (GTEx), gnomAD, and metrics for mutational constraints. Factors affecting NMD efficiency are analyzed using an allele-specific expression (ASE)-based measure to reduce noise caused by random variations. Additionally, by combining our data with the updated allele frequency database of gnomAD, we demonstrate the spectrum of NMD efficiency according to the degree of gene-level mutational constraints (degree of a gene-tolerating loss-of-function variants). The NMD prediction model, trained on TCGA data, shows that gene-level mutational constraint is an important predictor of NMD efficiency. Findings of this study suggest the potential role of NMD on shaping the landscape of mutational constraints.
Collapse
Affiliation(s)
- Young-Gon Kim
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyunju Kang
- Department of Artificial Intelligence, Sungkyunkwan University College of Computing and Informatics, Suwon, Republic of Korea
| | - Beomki Lee
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Hyeok-Jae Jang
- Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Seoul, Republic of Korea
| | - Jong-Ho Park
- Clinical Genomics Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Changhee Ha
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hogun Park
- Department of Artificial Intelligence, Sungkyunkwan University College of Computing and Informatics, Suwon, Republic of Korea.
| | - Jong-Won Kim
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
39
|
Pal T, Mundt E, Richardson ME, Chao E, Pesaran T, Slavin TP, Couch FJ, Monteiro ANA. Reduced penetrance BRCA1 and BRCA2 pathogenic variants in clinical germline genetic testing. NPJ Precis Oncol 2024; 8:247. [PMID: 39488595 PMCID: PMC11531542 DOI: 10.1038/s41698-024-00741-4] [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: 03/12/2024] [Accepted: 10/19/2024] [Indexed: 11/04/2024] Open
Abstract
Prior studies have suggested the existence of reduced penetrance pathogenic variants (RPPVs) in BRCA1 and BRCA2 (BRCA) which pose challenges for patient counseling and care. Here, we sought to establish RPPVs as a new category of variants. Candidate BRCA RPPVs provided by two large clinical diagnostic laboratories were compiled to identify those with the highest likelihood of being a RPPV, based on concordant interpretations. Sixteen concordant candidate BRCA RPPVs across both laboratories were systematically assessed. RPPVs included missense, splice site, and frameshift variants. Our study establishes RPPVs as a new class of variants imparting a moderately increased risk of breast cancer, which impacts risk-informed cancer prevention strategies, and provides a framework to standardize interpretation and reporting of BRCA RPPVs. Further work to define clinically meaningful risk thresholds and categories for reporting BRCA RPPVs is needed to personalize cancer risks in conjunction with other risk factors.
Collapse
Affiliation(s)
- Tuya Pal
- Department of Medicine, Vanderbilt-Ingram Cancer Center, University Medical Center, Vanderbilt University, Nashville, TN, USA.
| | - Erin Mundt
- Myriad Genetics, Salt Lake City, UT, USA
| | | | | | | | | | - Fergus J Couch
- Department of Laboratory Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Alvaro N A Monteiro
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA.
| |
Collapse
|
40
|
Patro I, Sahoo A, Nayak BR, Das R, Majumder S, Panigrahi GK. Nonsense-Mediated mRNA Decay: Mechanistic Insights and Physiological Significance. Mol Biotechnol 2024; 66:3077-3091. [PMID: 37930508 DOI: 10.1007/s12033-023-00927-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 09/28/2023] [Indexed: 11/07/2023]
Abstract
Nonsense-mediated mRNA decay (NMD) is an evolutionarily conserved surveillance mechanism across eukaryotes and also regulates the expression of physiological transcripts, thus involved in gene regulation. It essentially ensures recognition and removal of aberrant transcripts. Therefore, the NMD protects the cellular system by restricting the synthesis of truncated proteins, potentially by eliminating the faulty mRNAs. NMD is an evolutionarily conserved surveillance mechanism across eukaryotes and also regulates the expression of physiological transcripts, thus involved in gene regulation as well. Primarily, the NMD machinery scans and differentiates the aberrant and non-aberrant transcripts. A myriad of cellular dysfunctions arise due to production of truncated proteins, so the NMD core proteins, the up-frameshift factors (UPFs) recognizes the faulty mRNAs and further recruits factors resulting in the mRNA degradation. NMD exhibits astounding variability in its ability in regulating cellular mechanisms including both pathological and physiological events. But, the detailed underlying molecular mechanisms in NMD remains blurred and require extensive investigation to gain insights on cellular homeostasis. The complexity in understanding of NMD pathway arises due to the involvement of numerous proteins, molecular interactions and their functioning in different steps of this process. Moreover methods such as alternative splicing generates numerous isoforms of mRNA, so it makes difficulties in understanding the impact of alternative splicing on the efficiency of NMD functioning. Role of NMD in cancer development is very complex. Studies have shown that in some cases cancer cells use NMD pathway as a tool to exploit the NMD mechanism to maintain tumor microenvironment. A greater level of understanding about the intricate mechanism of how tumor used NMD pathway for their benefits, a strategy can be developed for targeting and inhibiting NMD factors involved in pro-tumor activity. There are very little amount of information available about the NMD pathway, how it discriminate mRNAs that are targeted by NMD from those that are not. This review highlights our current understanding of NMD, specifically the regulatory mechanisms and attempts to outline less explored questions that warrant further investigations. Taken as a whole, a detailed molecular understanding of the NMD mechanism could lead to wide-ranging applications for improving cellular homeostasis and paving out strategies in combating pathological disorders leaping forward toward achieving United Nations sustainable development goals (SDG 3: Good health and well-being).
Collapse
Affiliation(s)
- Ipsita Patro
- School of Applied Sciences, Centurion University of Technology and Management, Bhubaneswar, Odisha, India
| | - Annapurna Sahoo
- School of Applied Sciences, Centurion University of Technology and Management, Bhubaneswar, Odisha, India.
| | - Bilash Ranjan Nayak
- School of Applied Sciences, Centurion University of Technology and Management, Bhubaneswar, Odisha, India
| | - Rutupurna Das
- School of Applied Sciences, Centurion University of Technology and Management, Bhubaneswar, Odisha, India
| | - Sanjoy Majumder
- School of Applied Sciences, Centurion University of Technology and Management, Bhubaneswar, Odisha, India
| | - Gagan Kumar Panigrahi
- School of Applied Sciences, Centurion University of Technology and Management, Bhubaneswar, Odisha, India.
| |
Collapse
|
41
|
Wang H, Wu Y, Bassetti JA, Wang Z, Oza VS, Rangu SA, McGivern B, Peng S, Liang L, Huang S, Gong Z, Xu Z, Lin Z. A gain-of-function variant in SREBF1 causes generalized skin hyperpigmentation with congenital cataracts. Br J Dermatol 2024; 191:805-815. [PMID: 39005171 DOI: 10.1093/bjd/ljae291] [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: 05/07/2024] [Revised: 07/04/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND Lipid metabolism has essential roles in skin barrier formation and the regulation of skin inflammation. Lipid homeostasis regulates skin melanogenesis, although the underlying mechanism remains largely unknown. Sterol regulatory element binding protein 1 (SREBP-1) is a key transcription factor essential for cellular lipid metabolism. Loss-of-function variants in SREBF1 are responsible for autosomal-dominant ichthyosis follicularis, alopecia and photophobia syndrome, emphasizing the significance of lipid homeostasis in skin keratinization. OBJECTIVES To identify the genetic basis of a new entity featuring diffuse skin hyperpigmentation with congenital cataracts, and to unravel the underlying mechanism for the pathogenesis of the SREBF1 variant. METHODS Whole-exome sequencing was performed to identify underlying genetic variants. Quantitative polymerase chain reaction, Western blot and immunofluorescence staining were used to assess the expression and the subcellular localization of the SREBF1 variant. The transcriptional activity of mutant SREBP-1 was determined by a luciferase reporter assay. A transgenic zebrafish model was constructed. RESULTS Two unrelated patients presented with generalized skin hyperpigmentation with skin xerosis, congenital cataracts and extracutaneous symptoms. We identified a de novo nonsense variant c.1289C>A (p.Ser430*) in SREBF1 in both patients. The variant encoded a truncated protein that showed preferential nucleus localization, in contrast to wild-type SREBP-1 which - in sterol-sufficient conditions - is mainly localized in the cytoplasm. The luciferase reporter assay revealed that the p.Ser430* mutant exhibited enhanced transcriptional activity. Cultured patient primary melanocytes showed increased melanin synthesis vs. those from healthy controls. At 35 days postfertilization, the p.Ser430* transgenic zebrafish model exhibited more black spots, along with upregulated expression of melanogenic genes. CONCLUSIONS We demonstrated that a gain-of-function variant of SREBF1 causes a previously undescribed disorder characterized by generalized skin hyperpigmentation and congenital cataracts. Our study reveals the involvement of SREBP-1 in melanogenesis and lens development, and paves the way for the development of novel therapeutic targets for skin dyspigmentation or cataracts.
Collapse
Affiliation(s)
- Huijun Wang
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Yuan Wu
- Department of Ophthalmology, Peking University First Hospital, Beijing, China
| | | | - Zhaoyang Wang
- Department of Dermatology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Vikash S Oza
- Department of Dermatology and Pediatrics, The Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
| | - Sneha A Rangu
- Albert Einstein College of Medicine, New York, NY, USA
| | | | - Sha Peng
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Lina Liang
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Shimiao Huang
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Zhuoqing Gong
- Department of Dermatology, Peking University First Hospital, Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, National Clinical Research Center for Skin and Immune Diseases, Beijing, China
| | - Zigang Xu
- Department of Dermatology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Zhimiao Lin
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
42
|
Akamandisa MP, Boddicker NJ, Yadav S, Hu C, Hart SN, Ambrosone C, Anton-Culver H, Auer PL, Bodelon C, Burnside ES, Chen F, Eliassen HA, Goldgar DE, Haiman C, Hodge JM, Huang H, John EM, Karam R, Lacey JV, Lindstroem S, Martinez E, Na J, Neuhausen SL, O'Brien KM, Olson JE, Pal T, Palmer JR, Patel AV, Pesaran T, Polley EC, Richardson ME, Ruddy K, Sandler DP, Teras LR, Trentham-Dietz A, Vachon CM, Weinberg C, Winham SJ, Yao S, Zirpoli G, Kraft P, Weitzel JN, Domchek SM, Couch FJ, Nathanson KL. Association of Gene Variant Type and Location with Breast Cancer Risk in the General Population. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.11.24315237. [PMID: 39417132 PMCID: PMC11482981 DOI: 10.1101/2024.10.11.24315237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Importance Pathogenic variants (PVs) in ATM, BRCA1, BRCA2, CHEK2 , and PALB2 are associated with increased breast cancer risk. However, it is unknown whether breast cancer risk differs by PV type or location in carriers ascertained from the general population. Objective To evaluate breast cancer risks associated with PV type and location in ATM, BRCA1, BRCA2, CHEK2 , and PALB2 . Design Age adjusted case-control association analysis for all participants, subsets of PV carriers, and women with no breast cancer family history in population-based and clinical testing cohorts. Setting Twelve US population-based studies within the Cancer Risk Estimates Related to Susceptibility (CARRIERS) Consortium, and breast cancer cases from the UK-Biobank and an Ambry Genetics clinical testing cohort. Participants 32,247 women with and 32,544 age-matched women without a breast cancer diagnosis from CARRIERS; 237 and 1351 women with BRCA2 PVs and breast cancer from the UKBB and Ambry Genetics, respectively. Exposures PVs in ATM, BRCA1, BRCA2, CHEK2, and PALB2. Main Outcomes and Measures PVs were grouped by type and location within genes and assessed for risks of breast cancer (odds ratios (OR), 95% confidence intervals (CI), and p-values) using logistic regression. Mean ages at diagnosis were compared using linear regression. Results Compared to women carrying BRCA2 exon 11 protein truncating variants (PTVs) in the CARRIERS population-based study, women with BRCA2 ex13-27 PTVs (OR=2.7, 95%CI 1.1-7.9) and ex1-10 PTVs (OR=1.6, 95%CI 0.8-3.5) had higher breast cancer risks, lower rates of ER-negative breast cancer (ex13-27 OR=0.5, 95%CI 0.2-0.9; ex1-10 OR=0.5, 95%CI 0.1-1.0), and earlier age of breast cancer diagnosis (ex13-27 5.5 years, p<0.001; ex1-10 2.4 years, p=0.17). These associations with ER-negative breast cancer and age replicated in a high-risk clinical cohort and the population-based UK Biobank cohort. No differences in risk or age at diagnosis by gene region were observed for PTVs in other predisposition genes. Conclusions and Relevance Population-based and clinical high-risk cohorts establish that PTVs in exon 11 of BRCA2 are associated with reduced risk of breast cancer, later age at diagnosis, and greater risk of ER-negative disease. These differential risks may improve individualized risk prediction and clinical management for women carrying BRCA2 PTVs. Key Points Question: Does ATM , BRCA1 , BRCA2 , CHEK2 and PALB2 pathogenic variant type and location influence breast cancer risk in population-based studies? Findings: Breast cancer risk and estrogen receptor status differ based on the type and location of pathogenic variants in BRCA2 . Women carrying protein truncating variants in exon 11 have a lower breast cancer risk in the population-based cohorts, older age at diagnosis and higher rates of estrogen receptor negative breast cancer than women with exon 1-10 or exon 13-27 truncation variants in population-based and clinical testing cohorts. Meaning: Incorporating pathogenic variant type and location in cancer risk models may improve individualized risk prediction.
Collapse
|
43
|
Pelleter M, Desaintjean C, Gyapay R, Massenavette B, Baudin F, Couque N, Tamisier R, Dudoignon B, Franco P, Mougenel-Chantereau A, Coutier L. A new nonsense pathogenic variant in exon 1 of PHOX2B leads to the diagnosis of congenital central hypoventilation syndrome with intra-familial variability. Arch Pediatr 2024; 31:470-472. [PMID: 39261201 DOI: 10.1016/j.arcped.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/20/2024] [Accepted: 06/30/2024] [Indexed: 09/13/2024]
Abstract
Congenital central hypoventilation syndrome (CCHS) is a rare genetic disorder of the autonomic nervous system resulting in decreased brain sensitivity to hypercapnia and hypoxia characterized by a genetic abnormality in the pair-like homeobox 2B (PHOX2B) gene. Most patients have a heterozygous expansion of the polyalanine repeat in exon 3 (PARM), while 10 % of patients have non-PARM (NPARM) mutations that can span the entire gene. The majority of pathogenic variants are de novo, but variants with incomplete penetrance can be identified in the heterozygous state. In the present report, CCHS was diagnosed in a symptomatic 3-month-old infant with neonatal respiratory distress. Genetic analysis revealed a new mutation in exon 1 of the PHOX2B gene - p.Ser28* (c.83C>G) - which was further identified in two family members, one minimally symptomatic and one asymptomatic. The identification of this new mutation supports the importance of sequencing the entire gene even when the classic PARM mutation is not found and highlights the phenotypic variability of CCHS.
Collapse
Affiliation(s)
- Morgane Pelleter
- Service de pneumologie, allergologie, mucoviscidose, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, 69500 Bron, France
| | - Charlène Desaintjean
- Service de pneumologie, allergologie, mucoviscidose, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, 69500 Bron, France
| | - Romane Gyapay
- Service de pneumologie, allergologie, mucoviscidose, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, 69500 Bron, France
| | - Bruno Massenavette
- Service de pneumologie, allergologie, mucoviscidose, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, 69500 Bron, France; Service d'Épileptologie Clinique, des Troubles du Sommeil et de Neurologie Fonctionnelle de l'Enfant, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, 69500 Bron, France
| | - Florent Baudin
- Service de réanimation pédiatrique, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, 69500 Bron, France; Agressions Pulmonaires et Circulatoires dans le Sepsis (APCSe), VetAgro Sup, Universités de Lyon, Marcy, l'Etoile, France
| | - Nathalie Couque
- Département de Génétique, Hôpital Robert Debré, Assistance Publique - Hôpitaux de Paris, 75000 Paris, France
| | - Renaud Tamisier
- Grenoble Alpes University, HP2 Laboratory, INSERM, 38043 Grenoble, France; Pôle Thorax et Vaisseaux, Grenoble Alpes University Hospital, 38043 Grenoble, France
| | - Benjamin Dudoignon
- Université de Paris, AP-HP, Hôpital Robert Debré, Service de Physiologie Pédiatrique Centre du Sommeil-CRMR Hypoventilations alvéolaires rares, INSERM NeuroDiderot, F-75019 Paris, France
| | - Patricia Franco
- Service d'Épileptologie Clinique, des Troubles du Sommeil et de Neurologie Fonctionnelle de l'Enfant, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, 69500 Bron, France; Unité INSERM U1028 CNRS UMR 5292, Université Lyon 1, Lyon, France
| | - Antoine Mougenel-Chantereau
- Service de pneumologie, allergologie, mucoviscidose, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, 69500 Bron, France
| | - Laurianne Coutier
- Service de pneumologie, allergologie, mucoviscidose, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, 69500 Bron, France; Service d'Épileptologie Clinique, des Troubles du Sommeil et de Neurologie Fonctionnelle de l'Enfant, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, 69500 Bron, France; Unité INSERM U1028 CNRS UMR 5292, Université Lyon 1, Lyon, France.
| |
Collapse
|
44
|
Azeem A, Ahmed AN, Khan N, Voutsina N, Ullah I, Ubeyratna N, Yasin M, Baple EL, Crosby AH, Rawlins LE, Saleha S. Investigating the genetic basis of hereditary spastic paraplegia and cerebellar Ataxia in Pakistani families. BMC Neurol 2024; 24:354. [PMID: 39304850 DOI: 10.1186/s12883-024-03855-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 09/06/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Hereditary Spastic Paraplegias (HSPs) and Hereditary Cerebellar Ataxias (HCAs) are progressive neurodegenerative disorders encompassing a spectrum of neurogenetic conditions with significant overlaps of clinical features. Spastic ataxias are a group of conditions that have features of both cerebellar ataxia and spasticity, and these conditions are frequently clinically challenging to distinguish. Accurate genetic diagnosis is crucial but challenging, particularly in resource-limited settings. This study aims to investigate the genetic basis of HSPs and HCAs in Pakistani families. METHODS Families from Khyber Pakhtunkhwa with at least two members showing HSP or HCA phenotypes, and who had not previously been analyzed genetically, were included. Families were referred for genetic analysis by local neurologists based on the proband's clinical features and signs of a potential genetic neurodegenerative disorder. Whole Exome Sequencing (WES) and Sanger sequencing were then used to identify and validate genetic variants, and to analyze variant segregation within families to determine inheritance patterns. The mean age of onset and standard deviation were calculated to assess variability among affected individuals, and the success rate was compared with literature reports using differences in proportions and Cohen's h. RESULTS Pathogenic variants associated with these conditions were identified in five of eight families, segregating according to autosomal recessive inheritance. These variants included previously reported SACS c.2182 C > T, p.(Arg728*), FA2H c.159_176del, p.(Arg53_Ile58del) and SPG11 c.2146 C > T, p.(Gln716*) variants, and two previously unreported variants in SACS c.2229del, p.(Phe743Leufs*8) and ZFYVE26 c.1926_1941del, p.(Tyr643Metfs*2). Additionally, FA2H and SPG11 variants were found to have recurrent occurrences, suggesting a potential founder effect within the Pakistani population. Onset age among affected individuals ranged from 1 to 14 years (M = 6.23, SD = 3.96). The diagnostic success rate was 62.5%, with moderate effect sizes compared to previous studies. CONCLUSIONS The findings of this study expand the genotypic and phenotypic spectrum of HSPs and HCAs in Pakistan and emphasize the importance of utilizing exome/genome sequencing for accurate diagnosis or support accurate differential diagnosis. This approach can improve genetic counseling and clinical management, addressing the challenges of diagnosing neurodegenerative disorders in resource-limited settings.
Collapse
Affiliation(s)
- Arfa Azeem
- Department of Biotechnology and Genetic Engineering, Kohat University of Science and Technology, Kohat, 26000, Khyber Pakhtunkhwa, Pakistan
| | - Asif Naveed Ahmed
- Department of Biotechnology and Genetic Engineering, Kohat University of Science and Technology, Kohat, 26000, Khyber Pakhtunkhwa, Pakistan
| | - Niamat Khan
- Department of Biotechnology and Genetic Engineering, Kohat University of Science and Technology, Kohat, 26000, Khyber Pakhtunkhwa, Pakistan
| | - Nikol Voutsina
- 2Medical Research, RILD Wellcome Wolfson Centre (Level 4), Royal Devon and Exeter NHS Foundation Trust, Exeter, Devon, EX2 5DW, UK
| | - Irfan Ullah
- Department of Neurology, Khyber Teaching Hospital, Peshawar, 25000, Khyber Pakhtunkhwa, Pakistan
| | - Nishanka Ubeyratna
- 2Medical Research, RILD Wellcome Wolfson Centre (Level 4), Royal Devon and Exeter NHS Foundation Trust, Exeter, Devon, EX2 5DW, UK
| | - Muhammad Yasin
- Department of Biotechnology and Genetic Engineering, Kohat University of Science and Technology, Kohat, 26000, Khyber Pakhtunkhwa, Pakistan
| | - Emma L Baple
- 2Medical Research, RILD Wellcome Wolfson Centre (Level 4), Royal Devon and Exeter NHS Foundation Trust, Exeter, Devon, EX2 5DW, UK
| | - Andrew H Crosby
- 2Medical Research, RILD Wellcome Wolfson Centre (Level 4), Royal Devon and Exeter NHS Foundation Trust, Exeter, Devon, EX2 5DW, UK
| | - Lettie E Rawlins
- 2Medical Research, RILD Wellcome Wolfson Centre (Level 4), Royal Devon and Exeter NHS Foundation Trust, Exeter, Devon, EX2 5DW, UK.
- Peninsula Clinical Genetics Service, Royal Devon & Exeter Hospital (Heavitree), Exeter, UK.
| | - Shamim Saleha
- Department of Biotechnology and Genetic Engineering, Kohat University of Science and Technology, Kohat, 26000, Khyber Pakhtunkhwa, Pakistan.
| |
Collapse
|
45
|
Britto-Borges T, Gehring NH, Boehm V, Dieterich C. NMDtxDB: data-driven identification and annotation of human NMD target transcripts. RNA (NEW YORK, N.Y.) 2024; 30:1277-1291. [PMID: 39095083 PMCID: PMC11404449 DOI: 10.1261/rna.080066.124] [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: 04/19/2024] [Accepted: 07/11/2024] [Indexed: 08/04/2024]
Abstract
The nonsense-mediated RNA decay (NMD) pathway is a crucial mechanism of mRNA quality control. Current annotations of NMD substrate RNAs are rarely data-driven, but use generally established rules. We present a data set with four cell lines and combinations for SMG5, SMG6, and SMG7 knockdowns or SMG7 knockout. Based on this data set, we implemented a workflow that combines Nanopore and Illumina sequencing to assemble a transcriptome, which is enriched for NMD target transcripts. Moreover, we use coding sequence information (CDS) from Ensembl, Gencode consensus Ribo-seq ORFs, and OpenProt to enhance the CDS annotation of novel transcript isoforms. In summary, 302,889 transcripts were obtained from the transcriptome assembly process, out of which 24% are absent from Ensembl database annotations, 48,213 contain a premature stop codon, and 6433 are significantly upregulated in three or more comparisons of NMD active versus deficient cell lines. We present an in-depth view of these results through the NMDtxDB database, which is available at https://shiny.dieterichlab.org/app/NMDtxDB, and supports the study of NMD-sensitive transcripts. We open sourced our implementation of the respective web-application and analysis workflow at https://github.com/dieterich-lab/NMDtxDB and https://github.com/dieterich-lab/nmd-wf.
Collapse
Affiliation(s)
- Thiago Britto-Borges
- Section of Bioinformatics and Systems Cardiology, Department of Internal Medicine III and Klaus Tschira Institute for Integrative Computational Cardiology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Niels H Gehring
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50674 Cologne, Germany
| | - Volker Boehm
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50674 Cologne, Germany
| | - Christoph Dieterich
- Section of Bioinformatics and Systems Cardiology, Department of Internal Medicine III and Klaus Tschira Institute for Integrative Computational Cardiology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| |
Collapse
|
46
|
Yang W, Wang S, Huo X, Yang K, Guo Z, Li Y, Ji X, Hao B, Liao S. Novel autosomal recessive SINO syndrome-associated KIDINS220 variants provide insight into the genotype-phenotype correlation. Heliyon 2024; 10:e37355. [PMID: 39296002 PMCID: PMC11408833 DOI: 10.1016/j.heliyon.2024.e37355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/21/2024] Open
Abstract
Background KIDINS220 encodes a transmembrane scaffold protein, kinase D-interacting substrate of 220 kDa, that regulates neurotrophin signaling. Variants in KIDINS220 have been linked to spastic paraplegia, intellectual disability, nystagmus, and obesity (SINO) syndrome or prenatal fatal cerebral ventriculomegaly and arthrogryposis (VENARG). This study aimed to investigate the genotype-phenotype correlation of pathogenic KIDINS220 variants. Methods We performed whole-exome sequencing on a patient with SINO syndrome and epilepsy. Identified pathogenic variants were confirmed using Sanger sequencing and evaluated with in silico tools. A comprehensive literature review was conducted to analyze the genetic and phenotypic data of both the newly diagnosed patient and previously reported cases with KIDINS220 variants. Results We identified novel compound heterozygous variants in KIDINS220, c.1556C > T (p.Thr519Met) and c.2374C > T (p.Arg792*), in the patient. Our analysis revealed that biallelic loss-of-function variants in KIDINS220 are associated with VENARG or autosomal recessive SINO (AR-SINO), whereas carboxy-terminal truncated variants that escape nonsense-mediated mRNA decay and lack amino acid residues 1507-1529 are linked to autosomal dominant SINO (AD-SINO). Patients with AR-SINO exhibit more severe clinical features compared to those with AD-SINO. Conclusions Our study expands the spectrum of KIDINS220 variants associated with AR-SINO and provides a valuable genotype-phenotype correlation for pathogenic KIDINS220 variants.
Collapse
Affiliation(s)
- Wenke Yang
- Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, China
- National Health Commission Key Laboratory of Birth Defects Prevention, Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, Zhengzhou, China
- School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Shuyue Wang
- Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, China
- Central Hospital of Wuhan, Wuhan, China
| | - Xiaodong Huo
- Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Ke Yang
- Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenglong Guo
- Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, China
- National Health Commission Key Laboratory of Birth Defects Prevention, Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, Zhengzhou, China
| | - Yanjun Li
- Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Xinying Ji
- School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Bingtao Hao
- Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, China
- National Health Commission Key Laboratory of Birth Defects Prevention, Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, Zhengzhou, China
| | - Shixiu Liao
- Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, China
- National Health Commission Key Laboratory of Birth Defects Prevention, Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, Zhengzhou, China
| |
Collapse
|
47
|
Behera A, Panigrahi GK, Sahoo A. Nonsense-Mediated mRNA Decay in Human Health and Diseases: Current Understanding, Regulatory Mechanisms and Future Perspectives. Mol Biotechnol 2024:10.1007/s12033-024-01267-7. [PMID: 39264527 DOI: 10.1007/s12033-024-01267-7] [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: 03/04/2024] [Accepted: 08/24/2024] [Indexed: 09/13/2024]
Abstract
Nonsense-mediated mRNA decay (NMD) is a surveillance mechanism that is conserved across all eukaryotes ensuring the quality of transcripts by targeting messenger RNA (mRNA) harbouring premature stop codons. It regulates the gene expression by targeting aberrant mRNA carrying pre-termination codons (PTCs) and eliminates C-terminal truncated proteins. NMD distinguishes aberrant and non-aberrant transcript by looking after long 3' UTRs and exon-junction complex (EJC) downstream of stop codon that indicate the presence of PTC. Therefore, NMD modulates cellular surveillance and eliminates the truncated proteins but if the PTC escapes the surveillance pathway it can lead to potential negative phenotype resulting in genetic diseases. The alternative splicing also contributes in formation of NMD-sensitive isoforms by introducing PTC. NMD plays a complex role in cancer, it can either aggravate or downregulates the tumour. Some tumours agitate NMD to deteriorate mRNAs encoding tumour suppressor proteins, stress response proteins and neoantigens. In other case, tumours suppress the NMD to encourage the expression of oncoproteins for tumour growth and survival. This mechanism augmented in the development of new therapeutics by PTC read-through mechanism and personalized medicine. Detailed studies on NMD surveillance will possibly lead towards development of strategies for improving human health aligning with United Nations sustainable development goals (SDG 3: Good health and well-being). The potential therapeutic applications of NMD pose a challenge in terms of safe and effective modulation. Understanding the complexities of NMD regulation and its interaction with other cellular processes can lead to the development of new interventions for various diseases.
Collapse
Affiliation(s)
- Amrita Behera
- Department of Zoology, School of Applied Sciences, Centurion University of Technology and Management, Jatni, Khordha, Odisha, India
| | - Gagan Kumar Panigrahi
- Department of Zoology, School of Applied Sciences, Centurion University of Technology and Management, Jatni, Khordha, Odisha, India.
| | - Annapurna Sahoo
- Department of Zoology, School of Applied Sciences, Centurion University of Technology and Management, Jatni, Khordha, Odisha, India.
| |
Collapse
|
48
|
Li F, Wang X, Zhang J, Zhang J, Jing X, Jiang Q, Zhou J, Cao L, Peng H, Tong D, Huang C. RBM8A, a new target of TEAD4, promotes breast cancer progression by regulating IGF1R and IRS-2. J Transl Med 2024; 22:823. [PMID: 39232805 PMCID: PMC11373126 DOI: 10.1186/s12967-024-05639-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 08/27/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND Breast cancer (BC) is the most common malignant tumor in women worldwide, and further elucidation of the molecular mechanisms involved in BC pathogenesis is essential to improve the prognosis of BC patients. RNA Binding Motif Protein 8 A (RBM8A), with high affinity to a myriad of RNA transcripts, has been shown to play a crucial role in genesis and progression of multiple cancers. We attempted to explore its functional significance and molecular mechanisms in BC. METHODS Bioinformatics analysis was performed on publicly available BC datasets. qRT-PCR was used to determine the expression of RBM8A in BC tissues. MTT assay, clone formation assay and flow cytometry were employed to examine BC cell proliferation and apoptosis in vitro. RNA immunoprecipitation (RIP) and RIP-seq were used to investigate the binding of RBM8A/EIF4A3 to the mRNA of IGF1R/IRS-2. RBM8A and EIF4A3 interactions were determined by co-immunoprecipitation (Co-IP) and immunofluorescence. Chromatin immunoprecipitation (Ch-IP) and dual-luciferase reporter assay were carried out to investigate the transcriptional regulation of RBM8A by TEAD4. Xenograft model was used to explore the effects of RBM8A and TEAD4 on BC cell growth in vivo. RESULTS In this study, we showed that RBM8A is abnormally highly expressed in BC and knockdown of RBM8A inhibits BC cell proliferation and induces apoptosis in vitro. EIF4A3, which phenocopy RBM8A in BC, forms a complex with RBM8A in BC. Moreover, EIF4A3 and RBM8A complex regulate the expression of IGF1R and IRS-2 to activate the PI3K/AKT signaling pathway, thereby promoting BC progression. In addition, we identified TEAD4 as a transcriptional activator of RBM8A by Ch-IP, dual luciferase reporter gene and a series of functional rescue assays. Furthermore, we demonstrated the in vivo pro-carcinogenic effects of TEAD4 and RBM8A by xenograft tumor experiments in nude mice. CONCLUSION Collectively, these findings suggest that TEAD4 novel transcriptional target RBM8A interacts with EIF4A3 to increase IGF1R and IRS-2 expression and activate PI3K/AKT signaling pathway, thereby further promoting the malignant phenotype of BC cells.
Collapse
Affiliation(s)
- Fang Li
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong, University School of Health Science Center, Xi'an, 710301, Shaanxi, China
| | - Xiaofei Wang
- Biomedical Experimental Center, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Jing Zhang
- Institute of Basic Medical Sciences, Xi'an Medical University, No. 1 XinWang Road, Weiyang District, Xi'an, 710021, Shaanxi, China
- Medical Research and Experimental Center, The Second Affiliated Hospital of Xi 'an Medical University, Xi'an, 710077, Shaanxi Province, China
| | - Jinyuan Zhang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong, University School of Health Science Center, Xi'an, 710301, Shaanxi, China
| | - Xintao Jing
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong, University School of Health Science Center, Xi'an, 710301, Shaanxi, China
| | - Qiuyu Jiang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong, University School of Health Science Center, Xi'an, 710301, Shaanxi, China
| | - Jing Zhou
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong, University School of Health Science Center, Xi'an, 710301, Shaanxi, China
| | - Li Cao
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong, University School of Health Science Center, Xi'an, 710301, Shaanxi, China
| | - Hang Peng
- Second Department of General Surgery, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Dongdong Tong
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong, University School of Health Science Center, Xi'an, 710301, Shaanxi, China.
| | - Chen Huang
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710000, Shaanxi, China.
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong, University School of Health Science Center, Xi'an, 710301, Shaanxi, China.
- Biomedical Experimental Center, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
49
|
Greene D, De Wispelaere K, Lees J, Katrinecz A, Pascoal S, Hales E, Codina-Solà M, Valenzuela I, Tizzano EF, Atton G, Donnelly D, Foulds N, Jarvis J, McKee S, O'Donoghue M, Suri M, Vasudevan P, Stirrups K, Morgan NP, Freson K, Mumford AD, Turro E. Mutations in the U2 snRNA gene RNU2-2P cause a severe neurodevelopmental disorder with prominent epilepsy. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.03.24312863. [PMID: 39281759 PMCID: PMC11398430 DOI: 10.1101/2024.09.03.24312863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
The major spliceosome comprises the five snRNAs U1, U2, U4, U5 and U6. We recently showed that mutations in RNU4- 2, which encodes U4 snRNA, cause one of the most prevalent monogenic neurodevelopmental disorders. Here, we report that recurrent germline mutations in RNU2-2P , a 191bp gene encoding U2 snRNA, are responsible for a related disorder. By genetic association, we implicated recurrent de novo single nucleotide mutations at nucleotide positions 4 and 35 of RNU2-2P among nine cases. We replicated this finding in six additional cases, bringing the total to 15. The disorder is characterized by intellectual disability, neurodevelopmental delay, autistic behavior, microcephaly, hypotonia, epilepsy and hyperventilation. All cases display a severe and complex seizure phenotype. Our findings cement the role of major spliceosomal snRNAs in the etiologies of neurodevelopmental disorders.
Collapse
|
50
|
Arana ÁJ, Sánchez L. Knockout, Knockdown, and the Schrödinger Paradox: Genetic Immunity to Phenotypic Recapitulation in Zebrafish. Genes (Basel) 2024; 15:1164. [PMID: 39336755 PMCID: PMC11431394 DOI: 10.3390/genes15091164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/27/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024] Open
Abstract
Previous research has highlighted significant phenotypic discrepancies between knockout and knockdown approaches in zebrafish, raising concerns about the reliability of these methods. However, our study suggests that these differences are not as pronounced as was once believed. By carefully examining the roles of maternal and zygotic gene contributions, we demonstrate that these factors significantly influence phenotypic outcomes, often accounting for the observed discrepancies. Our findings emphasize that morpholinos, despite their potential off-target effects, can be effective tools when used with rigorous controls. We introduce the concept of graded maternal contribution, which explains how the uneven distribution of maternal mRNA and proteins during gametogenesis impacts phenotypic variability. Our research categorizes genes into three types-susceptible, immune, and "Schrödinger" (conditional)-based on their phenotypic expression and interaction with genetic compensation mechanisms. This distinction provides new insights into the paradoxical outcomes observed in genetic studies. Ultimately, our work underscores the importance of considering both maternal and zygotic contributions, alongside rigorous experimental controls, to accurately interpret gene function and the mechanisms underlying disease. This study advocates for the continued use of morpholinos in conjunction with advanced genetic tools like CRISPR/Cas9, stressing the need for a meticulous experimental design to optimize the utility of zebrafish in genetic research and therapeutic development.
Collapse
|