1
|
Nguyen TD, Winek MA, Rao MK, Dhyani SP, Lee MY. Nuclear envelope components in vascular mechanotransduction: emerging roles in vascular health and disease. Nucleus 2025; 16:2453752. [PMID: 39827403 DOI: 10.1080/19491034.2025.2453752] [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: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
The vascular network, uniquely sensitive to mechanical changes, translates biophysical forces into biochemical signals for vessel function. This process relies on the cell's architectural integrity, enabling uniform responses to physical stimuli. Recently, the nuclear envelope (NE) has emerged as a key regulator of vascular cell function. Studies implicate nucleoskeletal elements (e.g. nuclear lamina) and the linker of nucleoskeleton and cytoskeleton (LINC) complex in force transmission, emphasizing nucleo-cytoskeletal communication in mechanotransduction. The nuclear pore complex (NPC) and its component proteins (i.e. nucleoporins) also play roles in cardiovascular disease (CVD) progression. We herein summarize evidence on the roles of nuclear lamina proteins, LINC complex members, and nucleoporins in endothelial and vascular cell mechanotransduction. Numerous studies attribute NE components in cytoskeletal-related cellular behaviors to insinuate dysregulation of nucleocytoskeletal feedback and nucleocytoplasmic transport as a mechanism of endothelial and vascular dysfunction, and hence implications for aging and vascular pathophysiology.
Collapse
Affiliation(s)
- Tung D Nguyen
- Department of Physiology and Biophysics, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
- The Center for Cardiovascular Research, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
| | - Michael A Winek
- Department of Physiology and Biophysics, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
| | - Mihir K Rao
- Department of Physiology and Biophysics, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
| | - Shaiva P Dhyani
- Department of Physiology and Biophysics, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
| | - Monica Y Lee
- Department of Physiology and Biophysics, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
- The Center for Cardiovascular Research, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
| |
Collapse
|
2
|
Znaidi R, Massiani-Beaudoin O, Mailly P, Monnet H, Bonnifet T, Joshi RL, Fuchs J. Nuclear translocation of the LINE-1 encoded ORF1 protein alters nuclear envelope integrity in human neurons. Brain Res 2025; 1857:149579. [PMID: 40157412 DOI: 10.1016/j.brainres.2025.149579] [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: 03/06/2025] [Accepted: 03/17/2025] [Indexed: 04/01/2025]
Abstract
LINE-1 retrotransposons are increasingly implicated in aging and neurodegenerative diseases, yet the precise pathogenic mechanisms remain elusive. While the endonuclease and reverse transcriptase activities of LINE-1-encoded ORF2p can induce DNA damage and inflammation, a role of LINE-1 ORF1p in cellular dysfunctions stays unassigned. Here we demonstrate, using a neuronal cellular model, that ORF1p translocates into the nucleus upon arsenite-induced stress, directly interacting with nuclear import (KPNB1), nuclear pore complex (NUP153), and nuclear lamina (Lamin B1) proteins. Nuclear translocation of ORF1p disrupts nuclear integrity, nucleocytoplasmic transport, and heterochromatin structure, features linked to neurodegeneration and aging. Elevated nuclear ORF1p levels induced either by arsenite-induced stress, ORF1p overexpression, or as observed in Parkinson's disease post-mortem brain tissues correlate with impaired nuclear envelope (NE) morphology. Stress-induced nuclear alterations are mitigated by blocking ORF1p nuclear import or with the anti-aging drug remodelin. This study thus reveals a pathogenic action of nuclear ORF1p in human neurons driving NE alterations and thereby contributing to LINE-1-mediated cell toxicity.
Collapse
Affiliation(s)
- Rania Znaidi
- CIRB, Collège de France, Université PSL, CNRS, INSERM, 75005 Paris, France
| | | | - Philippe Mailly
- Orion Imaging Facility, CIRB, Collège de France, Université PSL, CNRS, INSERM, Labex Memolife, 75005 Paris, France
| | - Héloïse Monnet
- Orion Imaging Facility, CIRB, Collège de France, Université PSL, CNRS, INSERM, Labex Memolife, 75005 Paris, France
| | - Tom Bonnifet
- CIRB, Collège de France, Université PSL, CNRS, INSERM, 75005 Paris, France
| | - Rajiv L Joshi
- CIRB, Collège de France, Université PSL, CNRS, INSERM, 75005 Paris, France.
| | - Julia Fuchs
- CIRB, Collège de France, Université PSL, CNRS, INSERM, 75005 Paris, France.
| |
Collapse
|
3
|
Tao Y, Wang L, Chen E, Zhang S, Yang D, Chen W, He Y, Gu Y, Mao Y, Hu H. NAT10 promotes hepatocellular carcinoma progression by modulating the ac4C-DDIAS-PI3K-Akt axis. Sci Rep 2025; 15:17286. [PMID: 40389420 PMCID: PMC12089488 DOI: 10.1038/s41598-025-00707-x] [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: 05/24/2024] [Accepted: 04/30/2025] [Indexed: 05/21/2025] Open
Abstract
Primary liver cancer (PLC) is a prevalent tumor globally, ranking third in cancer-related mortality. The role of N4-acetylcysteine (ac4C) and N-acetyltransferase 10 (NAT10) in hepatocellular carcinoma (HCC) progression, migration, and invasion requires further elucidation. High NAT10 expression correlated with poor prognosis in HCC patients. Knockdown of NAT10 hindered HCC cell proliferation. AcRIP-seq screening revealed DDIAS as a significant downstream target of NAT10. Decreased NAT10 levels reduced DDIAS mRNA stability, leading to decreased proliferation, migration, and invasion of HCC cells upon DDIAS knockdown. Ectopic expression of DDIAS counteracted the effects of NAT10 knockdown by modulating the PI3K/AKT pathway. NAT10 was found to be elevated in HCC tissues compared to normal tissues, promoting HCC progression and correlating with shorter overall survival in patients. Mechanistically, NAT10 regulated HCC progression through the ac4C-DDIAS-PI3K-AKT axis.
Collapse
Affiliation(s)
- Yue Tao
- Wuxi Medical College, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
- Wuxi Ninth People's Hospital Affiliated to Soochow University, No.999 Liangxi Road, Binhu District, Wuxi, China
| | - Leisheng Wang
- Wuxi Medical College, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
| | - Enhong Chen
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Jiangnan University, 1000 Hefeng Rd,Binhu District, Wuxi, 214122, Jiangsu Province, China
| | - Shuo Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Jiangnan University, 1000 Hefeng Rd,Binhu District, Wuxi, 214122, Jiangsu Province, China
| | - Dongjie Yang
- Department of pathology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Rd,Binhu District, Wuxi, 214122, Jiangsu Province, China
| | - Wuqiang Chen
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Jiangnan University, 1000 Hefeng Rd,Binhu District, Wuxi, 214122, Jiangsu Province, China
| | - Youzhao He
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Jiangnan University, 1000 Hefeng Rd,Binhu District, Wuxi, 214122, Jiangsu Province, China
| | - Yuanlong Gu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Jiangnan University, 1000 Hefeng Rd,Binhu District, Wuxi, 214122, Jiangsu Province, China.
| | - Yong Mao
- Wuxi Medical College, Jiangnan University, Wuxi, 214122, Jiangsu Province, China.
- Department of cancer diagnosis and treatment center, Affiliated Hospital of Jiangnan University, 1000 Hefeng Rd,Binhu District, Wuxi, 214122, Jiangsu Province, China.
| | - Hao Hu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Jiangnan University, 1000 Hefeng Rd,Binhu District, Wuxi, 214122, Jiangsu Province, China.
- Wuxi Medical College, Jiangnan University, Wuxi, 214122, Jiangsu Province, China.
| |
Collapse
|
4
|
Bosmeny MS, Mamede JI, Gagnon KT. Resolving sequencing-based HIV-1 epitranscriptomics. Epigenomics 2025:1-12. [PMID: 40375799 DOI: 10.1080/17501911.2025.2504333] [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: 12/13/2024] [Accepted: 05/07/2025] [Indexed: 05/18/2025] Open
Abstract
The collection of HIV-1 RNA chemical modifications and their functional consequences in viral gene expression, host interactions, and the viral life cycle, referred to as HIV-1 epitranscriptomics, remain incompletely understood. While the field is evolving, diverse modification discovery methods, cell lines, HIV-1 sequences, and bioinformatics methods make a consensus view of the HIV-1 epitranscriptome difficult to resolve. Here, we review methods for identifying and interpreting N6-methyladenosine (m6A), 5-methylcytosine (m5C), pseudouridine (Ψ), 2´-O-methylation (Nm), and N4-acetylcytidine (ac4C) modifications in HIV-1, including antibody-based selection methods, chemical-treatment-based selection methods, and detection by nanopore direct RNA sequencing. We recommend the adoption of the latter as a standardized sequencing strategy to enable better benchmarking across diverse studies and help resolve HIV-1 epitranscriptomics.
Collapse
Affiliation(s)
- Michael S Bosmeny
- Department of Biochemistry, Wake Forest University, School of Medicine, Winston-Salem, NC, USA
| | - Joao I Mamede
- Department of Microbial Pathogens and Immunity, Rush University, Chicago, IL, USA
| | - Keith T Gagnon
- Department of Biochemistry, Wake Forest University, School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
5
|
Soto‐Ponce A, De Ita M, Castro‐Obregón S, Cortez D, Landesman Y, Magaña JJ, Gonzalo S, Zavaleta T, Soberano‐Nieto A, Unzueta J, Arrieta‐Cruz I, Nava P, Candelario‐Martínez A, García‐Aguirre I, Cisneros B. Targeting CRM1 for Progeria Syndrome Therapy. Aging Cell 2025; 24:e14495. [PMID: 39871520 PMCID: PMC12073922 DOI: 10.1111/acel.14495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 12/02/2024] [Accepted: 12/18/2024] [Indexed: 01/29/2025] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a premature aging disease caused by progerin, a mutant variant of lamin A. Progerin anchors aberrantly to the nuclear envelope disrupting a plethora of cellular processes, which in turn elicits senescence. We previously showed that the chromosomal region maintenance 1 (CRM1)-driven nuclear export pathway is abnormally enhanced in patient-derived fibroblasts, due to overexpression of CRM1. Interestingly, pharmacological inhibition of CRM1 using leptomycin B rescues the senescent phenotype of HGPS fibroblasts, delineating CRM1 as a potential therapeutic target against HGPS. As a proof of concept, we analyzed the beneficial effects of pharmacologically modulating CRM1 in dermal fibroblasts from HGPS patients and the LMNAG609G/G609G mouse, using the first-in-class selective inhibitor of CRM1 termed selinexor. Remarkably, treatment of HGPS fibroblasts with selinexor mitigated senescence and promoted progerin clearance via autophagy, while at the transcriptional level restored the expression of numerous differentially-expressed genes and rescued cellular processes linked to aging. In vivo, oral administration of selinexor to the progeric mouse resulted in decreased progerin immunostaining in the liver and aorta, decreased progerin levels in most liver, lung and kidney samples analyzed by immunoblotting, and improved aortic histopathology. Collectively our data indicate that selinexor exerts its geroprotective action by at least two mechanisms: normalizing the nucleocytoplasmic partition of proteins with a downstream effect on the aging-associated transcriptome and decreasing progerin levels. Further investigation of the overall effect of selinexor on LmnaG609G/G609G mouse physiology, with emphasis in cardiovascular function is warranted, to determine its therapeutic utility for HGPS and aging-associated disorders characterized by CRM1 overactivity.
Collapse
Affiliation(s)
- Adriana Soto‐Ponce
- Departamento de Genética y Biología MolecularCentro de Investigación y de Estudios AvanzadosCiudad de MéxicoMexico
| | - Marlon De Ita
- Departamento de Genética y Biología MolecularCentro de Investigación y de Estudios AvanzadosCiudad de MéxicoMexico
- Unidad de Investigación Médica en Genética Humana, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, IMSSSCiudad de MéxicoMexico
| | | | - Diego Cortez
- Centro de Ciencias Genómicas, UNAMCuernavacaMexico
| | | | - Jonathan J. Magaña
- Laboratorio de Medicina Genómica, Departamento de Genética (CENIAQ)Instituto Nacional de Rehabilitación‐Luis Guillermo Ibarra (INR‐LGII)Ciudad de MéxicoMexico
- Departamento de BioingenieríaEscuela de Ingeniería y Ciencias, Tecnologico de MonterreyCiudad de MéxicoMexico
| | - Susana Gonzalo
- Edward A. Doisy Department of Biochemistry and Molecular BiologySaint Louis University School of MedicineSt. LouisMissouriUSA
| | - Tania Zavaleta
- Departamento de Genética y Biología MolecularCentro de Investigación y de Estudios AvanzadosCiudad de MéxicoMexico
| | - Angelica Soberano‐Nieto
- Departamento de Genética y Biología MolecularCentro de Investigación y de Estudios AvanzadosCiudad de MéxicoMexico
| | - Juan Unzueta
- Unidad Iztapalapa, División de Ciencias Biológicas y de la SaludUniversidad Autónoma MetropolitanaCiudad de MéxicoMexico
| | - Isabel Arrieta‐Cruz
- Departamento de Investigación Básica, División de InvestigaciónInstituto Nacional de Geriatría, Secretaría de SaludCiudad de MéxicoMexico
| | - Porfirio Nava
- Departamento de Fisiología, Biofísica y NeurocienciasCentro de Investigación y de Estudios AvanzadosCiudad de MéxicoMexico
| | - Aurora Candelario‐Martínez
- Departamento de Fisiología, Biofísica y NeurocienciasCentro de Investigación y de Estudios AvanzadosCiudad de MéxicoMexico
| | - Ian García‐Aguirre
- Departamento de BioingenieríaEscuela de Ingeniería y Ciencias, Tecnologico de MonterreyCiudad de MéxicoMexico
| | - Bulmaro Cisneros
- Departamento de Genética y Biología MolecularCentro de Investigación y de Estudios AvanzadosCiudad de MéxicoMexico
| |
Collapse
|
6
|
Sun L, Li X, Xu F, Chen Y, Li X, Yang Z, Yang Y, Wang K, Ren T, Lin Z, Wang H, Wang X, Lu Y, Song Z, Cheng ZL, Wu D. A critical role of N 4-acetylation of cytidine in mRNA by NAT10 in T cell expansion and antiviral immunity. Nat Immunol 2025; 26:619-634. [PMID: 40045031 PMCID: PMC11957992 DOI: 10.1038/s41590-025-02100-2] [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: 02/11/2024] [Accepted: 01/26/2025] [Indexed: 04/02/2025]
Abstract
Following activation, naive T cells exit quiescence and require global translation for rapid expansion, yet the underlying mechanisms remain unclear. Here, we show that during T cell activation, cells upregulate the expression of N-acetyltransferase 10 (NAT10), an enzyme responsible for N4-acetylcytidine (ac4C) modification of mRNAs. ac4C-modified Myc mRNAs show higher translation efficiency, enabling rapid synthesis of MYC protein and supporting robust T cell expansion. Conditional deletion of Nat10 in mouse T cells causes severe cell cycle arrest and limitation of cell expansion due to MYC deficiency, ultimately exacerbating infection in an acute lymphocytic choriomeningitis virus model. Additionally, T cells from older individuals with lower NAT10 levels show proliferative defects, which may partially account for impaired antiviral responses in older individuals. This study reveals a mechanism governing T cell expansion, signal-dependent mRNA degradation induction and the potential in vivo biological significance of ac4C modification in T cell-mediated immune responses.
Collapse
Affiliation(s)
- Lu Sun
- Shanghai Key Laboratory of Lung Inflammation and Injury, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Medical Oncology, Shanghai Cancer Center & Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaoyan Li
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Geriatrics, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Feixiang Xu
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuwen Chen
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| | - Xushuo Li
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| | - Zhicheng Yang
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| | - Ying Yang
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| | - Ke Wang
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| | - Tianyi Ren
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zihao Lin
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiangdong Wang
- Shanghai Key Laboratory of Lung Inflammation and Injury, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yan Lu
- Institute of Metabolism and Regenerative Medicine, Digestive Endoscopic Center, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zhenju Song
- Shanghai Key Laboratory of Lung Inflammation and Injury, Zhongshan Hospital, Fudan University, Shanghai, China.
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Zhou-Li Cheng
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China.
| | - Duojiao Wu
- Shanghai Key Laboratory of Lung Inflammation and Injury, Zhongshan Hospital, Fudan University, Shanghai, China.
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
7
|
Hu M, Lv L, Lei Y, Chen M, Zhou S, Liu Z. NAT10 mediates TLR2 to promote podocyte senescence in adriamycin-induced nephropathy. Cell Death Dis 2025; 16:185. [PMID: 40108127 PMCID: PMC11923244 DOI: 10.1038/s41419-025-07515-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 02/17/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
N-acetyltransferase 10 (NAT10) is involved in regulating senescence. However, its role in glomerular diseases remains unclear. Therefore, this study aims to investigate the mechanisms by which NAT10 influences senescence and damage in an adriamycin (ADR)-induced nephropathy model. Senescence (p16 and p21) and DNA damage markers (γ-H2AX (ser139)) were assessed in ADR-induced nephropathy. NAT10 function was demonstrated using Remodelin or small interfering RNA (siRNA) interventions. Transcriptome sequencing was conducted to identify key downstream genes and pathways, while coimmunoprecipitation was performed to evaluate the relationship between NAT10 and toll-like receptor 2 (TLR2) expression. TLR2 overexpression or knockdown further validated its regulatory role in senescence. In ADR-treated mice, the expression levels of P53, P21, P16, γ-H2AX(S139) proteins were elevated, while those of WT-1 and nephrin were reduced. This effect was mitigated by Remodelin and siNAT10 administration. Transcriptome sequencing identified TLR2 as a key downstream gene, and coimmunoprecipitation, along with molecular docking models, confirmed its interaction with NAT10. TLR2 overexpression plasmid or siRNA was employed for recovery experiments. Together, the study findings suggest that NAT10 contributes to podocyte senescence and injury via interaction with TLR2. Further, it demonstrates that NAT10 alleviates ADR-induced podocyte senescence by interacting with TLR2, potentially through a P53-P21-dependent mechanism. Thus NAT10 could serve as a novel therapeutic target for treating podocyte senescence and proteinuric glomerulopathies.
Collapse
Affiliation(s)
- Mingyang Hu
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, PR China
- Henan Province Research Center for Kidney Diseases, Zhengzhou, PR China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, PR China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Linxiao Lv
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, PR China
- Henan Province Research Center for Kidney Diseases, Zhengzhou, PR China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, PR China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Yuqi Lei
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, PR China
- Henan Province Research Center for Kidney Diseases, Zhengzhou, PR China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, PR China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Min Chen
- Institute of Nephrology, Peking University, Beijing, PR China
| | - Sijie Zhou
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China.
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, PR China.
- Henan Province Research Center for Kidney Diseases, Zhengzhou, PR China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, PR China.
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, PR China.
| | - Zhangsuo Liu
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China.
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, PR China.
- Henan Province Research Center for Kidney Diseases, Zhengzhou, PR China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, PR China.
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, PR China.
| |
Collapse
|
8
|
Huang M, Li J, Bai J, Du X, Xu J. NAT10 induces mitochondrial dysfunction in lung epithelial cells by acetylating HMGB1 to exacerbate Pseudomonas aeruginosa-induced acute lung injury. Microb Pathog 2025; 200:107364. [PMID: 39909291 DOI: 10.1016/j.micpath.2025.107364] [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: 07/24/2024] [Revised: 01/25/2025] [Accepted: 02/01/2025] [Indexed: 02/07/2025]
Abstract
BACKGROUND Pseudomonas aeruginosa (PA) is a major pathogen that causes pneumonia and acute lung injury (ALI). Dysregulated NAT10 expression is associated with inflammatory and infectious diseases, but its role in PA-induced ALI remains unclear. METHODS A mouse pneumonia model was established by intratracheal injection of PA, and lentivirus-mediated NAT10 interference and HMGB1 overexpression vectors were administered via the tail vein. Lung mechanics, protein content, total cell counts, neutrophil counts, inflammatory factor levels in bronchoalveolar lavage fluid (BALF), and lung bacterial load were assessed 24 h after PA injection. HE staining was performed to evaluate lung structural damage. Intracellular oxidative stress levels in mouse lung epithelial cells (TC-1 cells) were measured by detecting ROS and MDA levels. Mitochondrial function was analyzed by testing the mitochondrial membrane potential, cytoplasmic accumulation of cytochrome C, mtDNA copy number, and ATP production. An N4-acetylcytidine (ac4C)-RNA immunoprecipitation assay was conducted to assess the ac4C level of HMGB1 mRNA. RESULTS NAT10 deficiency hindered PA infection-induced increases in immune cell infiltration, inflammatory factor levels, bacterial load, and ultimately lung structural and functional damage. However, upregulation of HMGB1 effectively antagonized the protective effects of NAT10 silencing in vivo. NAT10 knockdown suppressed PA-induced oxidative stress, mitochondrial dysfunction, and apoptosis in vitro. Whereas, HMGB1 overexpression reversed the inhibitory effects of NAT10 downregulation on PA-induced TC-1 cell injury. Mechanistically, as an acetyltransferase, NAT10 enhanced HMGB1 mRNA stability and protein expression by promoting HMGB1 mRNA ac4C modification. CONCLUSION NAT10 facilitated mitochondrial dysfunction in lung epithelial cells and exacerbated PA-induced ALI by promoting the N4-acetylcytidine of HMGB1 mRNA.
Collapse
Affiliation(s)
- Miaoyi Huang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China; Department of Respiratory Medicine, Xi'an Central Hospital, Xi'an, China
| | - Jianying Li
- Department of Respiratory Medicine, Xi'an Chest Hospital, Xi'an, China
| | - Jie Bai
- Department of Emergency Medicine, Xi'an Central Hospital, Xi'an, China
| | - Xusheng Du
- Department of Geratology Medicine, Xi'an Central Hospital, Xi'an, China
| | - Jiru Xu
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
9
|
Breusegem SY, Houghton J, Romero-Bueno R, Fragoso-Luna A, Kentistou KA, Ong KK, Janssen AFJ, Bright NA, Riedel CG, Perry JRB, Askjaer P, Larrieu D. A multiparametric anti-aging CRISPR screen uncovers a role for BAF in protein synthesis regulation. Nat Commun 2025; 16:1681. [PMID: 39956852 PMCID: PMC11830792 DOI: 10.1038/s41467-025-56916-5] [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: 07/31/2023] [Accepted: 01/28/2025] [Indexed: 02/18/2025] Open
Abstract
Progeria syndromes are very rare, incurable premature aging conditions recapitulating most aging features. Here, we report a whole genome, multiparametric CRISPR screen, identifying 43 genes that can rescue multiple cellular phenotypes associated with progeria. We implement the screen in fibroblasts from Néstor-Guillermo Progeria Syndrome male patients, carrying a homozygous A12T mutation in BAF. The hits are enriched for genes involved in protein synthesis, protein and RNA transport and osteoclast formation and are validated in a whole-organism Caenorhabditis elegans model. We further confirm that BAF A12T can disrupt protein synthesis rate and fidelity, which could contribute to premature aging in patients. This work highlights the power of multiparametric genome-wide suppressor screens to identify genes enhancing cellular resilience in premature aging and provide insights into the biology underlying progeria-associated cellular dysfunction.
Collapse
Affiliation(s)
- Sophia Y Breusegem
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Keith Peters Building, Hills Road, Cambridge, UK
- Sophia Y. Breusegem: MRC toxicology Unit, University of Cambridge, Tennis Court Road, Cambridge, UK
| | - Jack Houghton
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Keith Peters Building, Hills Road, Cambridge, UK
- Jack Houghton: Imperial College London, Exhibition Road, South Kensington, London, UK
| | - Raquel Romero-Bueno
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas-Universidad Pablo de Olavide-Junta de Andalucía, Seville, Spain
| | - Adrián Fragoso-Luna
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas-Universidad Pablo de Olavide-Junta de Andalucía, Seville, Spain
| | - Katherine A Kentistou
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Ken K Ong
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Anne F J Janssen
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Keith Peters Building, Hills Road, Cambridge, UK
- Anne F. J. Janssen: Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, Nijmegen, The Netherlands
| | - Nicholas A Bright
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Keith Peters Building, Hills Road, Cambridge, UK
| | | | - John R B Perry
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
- Metabolic Research Laboratory, Wellcome-MRC Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Peter Askjaer
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas-Universidad Pablo de Olavide-Junta de Andalucía, Seville, Spain
| | - Delphine Larrieu
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Keith Peters Building, Hills Road, Cambridge, UK.
- Delphine Larrieu: Altos Labs, Cambridge Institute of Science, Cambridge, UK.
| |
Collapse
|
10
|
Luo R, Zhang Y, Kumata K, Xie L, Kurihara Y, Ogawa M, Kokufuta T, Nengaki N, Wang F, Zhang MRR. The N-acetyltransferase 10 inhibitor [ 11C]remodelin: synthesis and preliminary positron emission tomography study in mice. EJNMMI Radiopharm Chem 2025; 10:6. [PMID: 39888477 PMCID: PMC11785860 DOI: 10.1186/s41181-025-00330-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 01/23/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND 4-(4-Cyanophenyl)-2-(2-cyclopentylidenehydrazinyl)thiazole (remodelin) is a potent N-acetyltransferase 10 (NAT10) inhibitor. This compound inhibits tumors and weakens tumor resistance to antitumor drugs. Moreover, remodelin has been found to enhance healthspan in an animal model of the human accelerated ageing syndrome. In this study, we synthesized C-11-labelled remodelin ([11C]remodelin) for the first time as a positron emission tomography (PET) probe and assessed its biodistribution in mice using PET. RESULTS [11C]Remodelin was synthesized by the reaction of a boron ester precursor (1) with hydrogen [11C]cyanide, which was prepared from the cyclotron-produced [11C]carbon dioxide via [11C]methane. The decay-corrected radiochemical yield of [11C]remodelin was 6.2 ± 2.3% (n = 20, based on [11C]carbon dioxide) with a synthesis time of 45 min and radiochemical purity of > 90%. A PET study with [11C]remodelin showed high uptake of radioactivity in the heart, liver, and small intestine of mice. The metabolite analysis indicated moderate metabolism of [11C]remodelin in the heart. CONCLUSIONS In the present study, we successfully synthesized [11C]remodelin and assessed its biodistribution of radioactivity in the mouse organs and tissues with PET. We are planning to prepare tumor and inflammatory models in which overexpression of NAT10 is possibly induced and conduct PET imaging for these animal models with [11C]remodelin to elucidate the relationship between NAT10 and diseases.
Collapse
Affiliation(s)
- Rui Luo
- Department of Advanced Nuclear Medicine Sciences, Institute of Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
- Department of Nuclear Medicine, Nanjing First Hospital, Affiliated to Nanjing Medical University, 68 Chanle Road, Nanjing, 210006, China
| | - Yiding Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute of Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| | - Katsushi Kumata
- Department of Advanced Nuclear Medicine Sciences, Institute of Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| | - Lin Xie
- Department of Advanced Nuclear Medicine Sciences, Institute of Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan.
| | - Yusuke Kurihara
- Department of Advanced Nuclear Medicine Sciences, Institute of Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
- SHI Accelerator Service, Ltd, 7-1-1 Nishigotanda, Shinagawa-ku, Tokyo, 141-0031, Japan
| | - Masanao Ogawa
- Department of Advanced Nuclear Medicine Sciences, Institute of Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
- SHI Accelerator Service, Ltd, 7-1-1 Nishigotanda, Shinagawa-ku, Tokyo, 141-0031, Japan
| | - Tomomi Kokufuta
- Department of Advanced Nuclear Medicine Sciences, Institute of Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| | - Nobuki Nengaki
- Department of Advanced Nuclear Medicine Sciences, Institute of Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
- SHI Accelerator Service, Ltd, 7-1-1 Nishigotanda, Shinagawa-ku, Tokyo, 141-0031, Japan
| | - Feng Wang
- Department of Advanced Nuclear Medicine Sciences, Institute of Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan.
- Department of Nuclear Medicine, Nanjing First Hospital, Affiliated to Nanjing Medical University, 68 Chanle Road, Nanjing, 210006, China.
| | - Ming-Rong R Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute of Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan.
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
11
|
Zhang W, Zhang J, Zhang Y, Zhai J, Sun B, Guo Y, Wang F. The up-regulation of RIPK3 mediated by ac4C modification promotes oxidative stress-induced granulosa cell senescence by inhibiting the Nrf2/HO-1 pathway. IUBMB Life 2025; 77:e2944. [PMID: 39865380 DOI: 10.1002/iub.2944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 12/31/2024] [Indexed: 01/30/2025]
Abstract
Abnormality of granulosa cells (GCs) is the critical cause of follicular atresia in premature ovarian failure (POF). RIPK3 is highly expressed in GCs derived from atretic follicles. We focus on uncovering how RIPK3 contributes to ovarian GC senescence. Primary GCs were treated with H₂O₂ to induce senescence. ROS was detected via DCFH-DA staining. Levels of senescence-related molecules and SA-β-Gal activity were examined. Cyclophosphamide was administered to mice to induce POF. The impact of RIPK3 on atretic follicles and sex hormones was evaluated through HE staining and ELISA, respectively. The acRIP-qPCR analysis of RIPK3 ac4C levels, RIP detection for interaction between RIPK3 and NAT10, and actinomycin D treatment to detect RIPK3 degradation were conducted. In H2O2-treated GCs and POF mouse ovaries, levels of RIPK3, ROS, senescence-related molecules, as well as SA-β-Gal activity, were all up-regulated, and this effect was suppressed by RIPK3 inhibition. RIPK3 interference reduced atretic follicles and FSH levels while increasing AMH and E2 levels. Nrf2 and HO-1 content were diminished in the models, whereas si-RIPK3 facilitated their expression. The effect of si-RIPK3 on decreased levels of ROS and senescence-related molecules was reversed by ML385. H2O2 decreased RIPK3 mRNA degradation and increased its ac4C modification. The ac4C modifying enzyme NAT10 was up-regulated in the models, and NAT10 enhanced RIPK3 mRNA stability through ac4C modification. NAT10 knockdown mitigated ovarian GC senescence by inhibiting RIPK3 expression. The promotion of RIPK3 mRNA stability through ac4C modification by NAT10, in turn, affects the Nrf2/HO-1 pathway and promotes ovarian GC senescence.
Collapse
Affiliation(s)
- Wanjun Zhang
- Department of Reproductive Medical Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jiahao Zhang
- Department of Reproductive Medical Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yile Zhang
- Department of Reproductive Medical Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jun Zhai
- Department of Reproductive Medical Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Bo Sun
- Department of Reproductive Medical Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yihong Guo
- Department of Reproductive Medical Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Fang Wang
- Department of Reproductive Medical Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| |
Collapse
|
12
|
Xiao B, Wu S, Tian Y, Huang W, Chen G, Luo D, Cai Y, Chen M, Zhang Y, Liu C, Zhao J, Li L. Advances of NAT10 in diseases: insights from dual properties as protein and RNA acetyltransferase. Cell Biol Toxicol 2024; 41:17. [PMID: 39725720 PMCID: PMC11671434 DOI: 10.1007/s10565-024-09962-6] [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: 07/14/2024] [Accepted: 11/29/2024] [Indexed: 12/28/2024]
Abstract
N-acetyltransferase 10 (NAT10) is a member of the Gcn5-related N-acetyltransferase (GNAT) family and it plays a crucial role in various cellular processes, such as regulation of cell mitosis, post-DNA damage response, autophagy and apoptosis regulation, ribosome biogenesis, RNA modification, and other related pathways through its intrinsic protein acetyltransferase and RNA acetyltransferase activities. Moreover, NAT10 is closely associated with the pathogenesis of tumors, Hutchinson-Gilford progeria syndrome (HGPS), systemic lupus erythematosus, pulmonary fibrosis, depression and host-pathogen interactions. In recent years, mRNA acetylation has emerged as a prominent focus of research due to its pivotal role in regulating RNA stability and translation. NAT10 stands out as the sole identified modification enzyme responsible for RNA acetylation. There remains some ambiguity regarding the similarities and differences in NAT10's actions on protein and RNA substrates. While NAT10 involves acetylation modification in both cases, which is a crucial molecular mechanism in epigenetic regulation, there are significant disparities in the catalytic mechanisms, regulatory pathways, and biological processes involved. Therefore, this review aims to offer a comprehensive overview of NAT10 as a protein and RNA acetyltransferase, covering its basic catalytic features, biological functions, and roles in related diseases.
Collapse
Affiliation(s)
- Bin Xiao
- Department of Laboratory Medicine, Affiliated Qingyuan Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, Guangdong, China.
- Department of Laboratory Medicine, Guangdong Provincial Second Hospital of Traditional Chinese Medicine, Guangdong Provincial Engineering Technology Research Institute of Traditional Chinese Medicine, The Fifth Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510095, Guangdong, China.
| | - Shunhong Wu
- Department of Laboratory Medicine, Affiliated Qingyuan Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, Guangdong, China
| | - Yan Tian
- Affiliated Qingyuan Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, Guangdong, China
| | - Weikai Huang
- Affiliated Qingyuan Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, Guangdong, China
| | - Guangzhan Chen
- Affiliated Qingyuan Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, Guangdong, China
| | - Dongxin Luo
- Affiliated Qingyuan Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, Guangdong, China
| | - Yishen Cai
- Department of Laboratory Medicine, Affiliated Qingyuan Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, Guangdong, China
| | - Ming Chen
- Department of Laboratory Medicine, Affiliated Qingyuan Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, Guangdong, China
| | - Yuqian Zhang
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Chuyan Liu
- Affiliated Qingyuan Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, Guangdong, China
| | - Junxiu Zhao
- College of Public Health, Dali University, Dali, 671003, Yunnan, China
| | - Linhai Li
- Department of Laboratory Medicine, Affiliated Qingyuan Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, Guangdong, China.
| |
Collapse
|
13
|
Lin J, Sumara I. Cytoplasmic nucleoporin assemblage: the cellular artwork in physiology and disease. Nucleus 2024; 15:2387534. [PMID: 39135336 PMCID: PMC11323873 DOI: 10.1080/19491034.2024.2387534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/29/2024] [Accepted: 07/29/2024] [Indexed: 08/16/2024] Open
Abstract
Nucleoporins, essential proteins building the nuclear pore, are pivotal for ensuring nucleocytoplasmic transport. While traditionally confined to the nuclear envelope, emerging evidence indicates their presence in various cytoplasmic structures, suggesting potential non-transport-related roles. This review consolidates findings on cytoplasmic nucleoporin assemblies across different states, including normal physiological conditions, stress, and pathology, exploring their structural organization, formation dynamics, and functional implications. We summarize the current knowledge and the latest concepts on the regulation of nucleoporin homeostasis, aiming to enhance our understanding of their unexpected roles in physiological and pathological processes.
Collapse
Affiliation(s)
- Junyan Lin
- Department of Development and Stem Cells, Institute of Genetics and Molecular and Cellular Biology (IGBMC), Illkirch, France
- Centre National de la Recherche Scientifique UMR 7104, Strasbourg, France
- Institut National de la Santé et de la Recherche Médicale U964, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
| | - Izabela Sumara
- Department of Development and Stem Cells, Institute of Genetics and Molecular and Cellular Biology (IGBMC), Illkirch, France
- Centre National de la Recherche Scientifique UMR 7104, Strasbourg, France
- Institut National de la Santé et de la Recherche Médicale U964, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
| |
Collapse
|
14
|
Kieronska-Rudek A, Kij A, Bar A, Kurpinska A, Mohaissen T, Grosicki M, Stojak M, Sternak M, Buczek E, Proniewski B, Kuś K, Suraj-Prazmowska J, Panek A, Pietrowska M, Zapotoczny S, Shanahan CM, Szabo C, Chlopicki S. Phylloquinone improves endothelial function, inhibits cellular senescence, and vascular inflammation. GeroScience 2024; 46:4909-4935. [PMID: 38980631 PMCID: PMC11336140 DOI: 10.1007/s11357-024-01225-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 05/24/2024] [Indexed: 07/10/2024] Open
Abstract
Phylloquinon (PK) and menaquinones (MK) are both naturally occurring compounds belonging to vitamin K group. Present study aimed to comprehensively analyze the influence of PK in several models of vascular dysfunction to determine whether PK has vasoprotective properties, similar to those previously described for MK. Effects of PK and MK on endothelial dysfunction were studied in ApoE/LDLR-/- mice in vivo, in the isolated aorta incubated with TNF, and in vascular cells as regard inflammation and cell senescence (including replicative and stress-induced models of senescence). Moreover, the vascular conversion of exogenous vitamins to endogenous MK-4 was analyzed. PK, as well as MK, given for 8 weeks in diet (10 mg/kg) resulted in comparable improvement in endothelial function in the ApoE/LDLR-/- mice. Similarly, PK and MK prevented TNF-induced impairment of endothelium-dependent vasorelaxation in the isolated aorta. In in vitro studies in endothelial and vascular smooth muscle cells, we identified that both PK and MK displayed anti-senescence effects via decreasing DNA damage while in endothelial cells anti-inflammatory activity was ascribed to the modulation of NFκB activation. The activity of PK and MK was comparable in terms of their effect on senescence and inflammation. Presence of endogenous synthesis of MK-4 from PK in aorta and endothelial and smooth muscle cells suggests a possible involvement of MK in vascular effects of PK. In conclusion, PK and MK display comparable vasoprotective effects, which may be ascribed, at least in part, to the inhibition of cell senescence and inflammation. The vasoprotective effect of PK in the vessel wall can be related to the direct effects of PK, as well as to the action of MK formed from PK in the vascular wall.
Collapse
Affiliation(s)
- Anna Kieronska-Rudek
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
- Chair of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
- Chair of Pharmacology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Agnieszka Kij
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Anna Bar
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Anna Kurpinska
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Tasnim Mohaissen
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Marek Grosicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Marta Stojak
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Magdalena Sternak
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Elżbieta Buczek
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Bartosz Proniewski
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Kamil Kuś
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Joanna Suraj-Prazmowska
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Agnieszka Panek
- Institute of Nuclear Physics Polish Academy of Sciences, Krakow, Poland
| | - Monika Pietrowska
- Centre for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
| | - Szczepan Zapotoczny
- Department of Physical Chemistry and Electrochemistry, Faculty of Chemistry, Jagiellonian University, Krakow, Poland
| | - Catherine M Shanahan
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, King's College London, London, UK
| | - Csaba Szabo
- Chair of Pharmacology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland.
- Chair of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland.
| |
Collapse
|
15
|
Wang Y, Su K, Wang C, Deng T, Liu X, Sun S, Jiang Y, Zhang C, Xing B, Du X. Chemotherapy-induced acetylation of ACLY by NAT10 promotes its nuclear accumulation and acetyl-CoA production to drive chemoresistance in hepatocellular carcinoma. Cell Death Dis 2024; 15:545. [PMID: 39085201 PMCID: PMC11291975 DOI: 10.1038/s41419-024-06951-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/21/2024] [Accepted: 07/24/2024] [Indexed: 08/02/2024]
Abstract
Chemotherapeutic efficacy is seriously impeded by chemoresistance in more than half of hepatocellular carcinoma (HCC) patients. However, the mechanisms involved in chemotherapy-induced upregulation of chemoresistant genes are not fully understood. Here, this study unravels a novel mechanism controlling nuclear acetyl-CoA production to activate the transcription of chemoresistant genes in HCC. NAT10 is upregulated in HCC tissues and its upregulation is correlated with poor prognosis of HCC patients. NAT10 is also upregulated in chemoresistant HCC cells. Targeting NAT10 increases the cytotoxicity of chemotherapy in HCC cells and mouse xenografts. Upon chemotherapy, NAT10 translocates from the nucleolus to the nucleus to activate the transcription of CYP2C9 and PIK3R1. Additionally, nuclear acetyl-CoA is specifically upregulated by NAT10. Mechanistically, NAT10 binds with ACLY in the nucleus and acetylates ACLY at K468 to counteract the SQSTM1-mediated degradation upon chemotherapy. ACLY K468-Ac specifically accumulates in the nucleus and increases nuclear acetyl-CoA production to activate the transcription of CYP2C9 and PIK3R1 through enhancing H3K27ac. Importantly, K468 is required for nuclear localization of ACLY. Significantly, ACLY K468-Ac is upregulated in HCC tissues, and ablation of ACLY K468-Ac sensitizes HCC cells and mouse xenografts to chemotherapy. Collectively, these findings identify NAT10 as a novel chemoresistant driver and the blockage of NAT10-mediated ACLY K468-Ac possesses the potential to attenuate HCC chemoresistance.
Collapse
MESH Headings
- Humans
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/drug therapy
- Liver Neoplasms/genetics
- Liver Neoplasms/pathology
- Acetyl Coenzyme A/metabolism
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/drug effects
- Animals
- Acetylation
- Mice
- Cell Nucleus/metabolism
- Cell Line, Tumor
- Mice, Nude
- Coenzyme A Ligases/metabolism
- Coenzyme A Ligases/genetics
- Gene Expression Regulation, Neoplastic/drug effects
- N-Terminal Acetyltransferases/metabolism
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Mice, Inbred BALB C
- Male
Collapse
Affiliation(s)
- Yuying Wang
- Department of Cell Biology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Kunqi Su
- Department of Cell Biology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Chang Wang
- Department of Cell Biology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Tao Deng
- Department of Cell Biology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xiaofeng Liu
- Hepatopancreatobiliary Surgery Department I, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Shiqi Sun
- Department of Cell Biology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yang Jiang
- Department of Cell Biology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Chunfeng Zhang
- Department of Medical Genetics, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Baocai Xing
- Hepatopancreatobiliary Surgery Department I, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China.
| | - Xiaojuan Du
- Department of Cell Biology, School of Basic Medical Sciences, Peking University, Beijing, China.
| |
Collapse
|
16
|
Wang M, Zhang J, Qiu J, Ma X, Xu C, Wu Q, Xing S, Chen X, Liu B. Doxycycline decelerates aging in progeria mice. Aging Cell 2024; 23:e14188. [PMID: 38686927 PMCID: PMC11258430 DOI: 10.1111/acel.14188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 03/26/2024] [Accepted: 04/18/2024] [Indexed: 05/02/2024] Open
Abstract
Beyond the antimicrobial activity, doxycycline (DOX) exhibits longevity-promoting effect in nematodes, while its effect on mammals is unclear. Here, we applied a mouse model of Hutchinson-Gilford progeria syndrome (HGPS), Zmpste24 knockout (KO) mice, and analyzed the antiaging effect of DOX. We found that the DOX treatment prolongs lifespan and ameliorates progeroid features of Zmpste24 KO mice, including the decline of body and tissue weight, exercise capacity and cortical bone density, and the shortened colon length. DOX treatment alleviates the abnormal nuclear envelope in multiple tissues, and attenuates cellular senescence and cell death of Zmpste24 KO and HGPS fibroblasts. DOX downregulates the level of proinflammatory IL6 in both serum and tissues. Moreover, the elevated α-tubulin (K40) acetylation mediated by NAT10 in progeria, is rescued by DOX treatment in the aorta tissues in Zmpste24 KO mice and fibroblasts. Collectively, our study uncovers that DOX can decelerate aging in progeria mice via counteracting IL6 expression and NAT10-mediated acetylation of α-tubulin.
Collapse
Affiliation(s)
- Ming Wang
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL‐SAI), Marshall Laboratory of Biomedical Engineering, International Cancer Center, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Jie Zhang
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL‐SAI), Marshall Laboratory of Biomedical Engineering, International Cancer Center, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Jiangping Qiu
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL‐SAI), Marshall Laboratory of Biomedical Engineering, International Cancer Center, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Xuan Ma
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL‐SAI), Marshall Laboratory of Biomedical Engineering, International Cancer Center, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Chenzhong Xu
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL‐SAI), Marshall Laboratory of Biomedical Engineering, International Cancer Center, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Qiuhuan Wu
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL‐SAI), Marshall Laboratory of Biomedical Engineering, International Cancer Center, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Shaojun Xing
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Xinchun Chen
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Baohua Liu
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL‐SAI), Marshall Laboratory of Biomedical Engineering, International Cancer Center, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| |
Collapse
|
17
|
Guo Q, Yu W, Tan J, Zhang J, Chen J, Rao S, Guo X, Cai K. Remodelin delays non-small cell lung cancer progression by inhibiting NAT10 via the EMT pathway. Cancer Med 2024; 13:e7283. [PMID: 38826095 PMCID: PMC11145023 DOI: 10.1002/cam4.7283] [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: 01/16/2024] [Revised: 04/22/2024] [Accepted: 04/28/2024] [Indexed: 06/04/2024] Open
Abstract
BACKGROUND Lung cancer remains the foremost reason of cancer-related mortality, with invasion and metastasis profoundly influencing patient prognosis. N-acetyltransferase 10 (NAT10) catalyzes the exclusive N (4)-acetylcytidine (ac4C) modification in eukaryotic RNA. NAT10 dysregulation is linked to various diseases, yet its role in non-small cell lung cancer (NSCLC) invasion and metastasis remains unclear. Our study delves into the clinical significance and functional aspects of NAT10 in NSCLC. METHODS We investigated NAT10's clinical relevance using The Cancer Genome Atlas (TCGA) and a group of 98 NSCLC patients. Employing WB, qRT-PCR, and IHC analyses, we assessed NAT10 expression in NSCLC tissues, bronchial epithelial cells (BECs), NSCLC cell lines, and mouse xenografts. Further, knockdown and overexpression techniques (siRNA, shRNA, and plasmid) were employed to evaluate NAT10's effects. A series of assays were carried out, including CCK-8, colony formation, wound healing, and transwell assays, to elucidate NAT10's role in proliferation, invasion, and metastasis. Additionally, we utilized lung cancer patient-derived 3D organoids, mouse xenograft models, and Remodelin (NAT10 inhibitor) to corroborate these findings. RESULTS Our investigations revealed high NAT10 expression in NSCLC tissues, cell lines and mouse xenograft models. High NAT10 level correlated with advanced T stage, lymph node metastasis and poor overall survive. NAT10 knockdown curtailed proliferation, invasion, and migration, whereas NAT10 overexpression yielded contrary effects. Furthermore, diminished NAT10 levels correlated with increased E-cadherin level whereas decreased N-cadherin and vimentin expressions, while heightened NAT10 expression displayed contrasting results. Notably, Remodelin efficiently attenuated NSCLC proliferation, invasion, and migration by inhibiting NAT10 through the epithelial-mesenchymal transition (EMT) pathway. CONCLUSIONS Our data underscore NAT10 as a potential therapeutic target for NSCLC, presenting avenues for targeted intervention against lung cancer through NAT10 inhibition.
Collapse
Affiliation(s)
- Quanwei Guo
- The First School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
- Department of Thoracic Surgery, Shenzhen HospitalSouthern Medical UniversityShenzhenChina
| | - Weijun Yu
- Bao'an District Hospital for Chronic Diseases Prevention and CureShenzhenChina
| | - Jianfeng Tan
- Department of Thoracic Surgery, Shenzhen HospitalSouthern Medical UniversityShenzhenChina
| | - Jianhua Zhang
- Department of Thoracic Surgery, Shenzhen HospitalSouthern Medical UniversityShenzhenChina
| | - Jin Chen
- Science and Education Department, Shenzhen HospitalSouthern Medical UniversityShenzhenChina
| | - Shuan Rao
- Department of Thoracic Surgery, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Xia Guo
- Center for Clinical Research and Innovation, Shenzhen HospitalSouthern Medical UniversityShenzhenChina
- Shenzhen Key Laboratory of Viral OncologyShenzhenChina
| | - Kaican Cai
- Department of Thoracic Surgery, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
18
|
Foo MXR, Ong PF, Yap ZX, Maric M, Bong CJS, Dröge P, Burke B, Dreesen O. Genetic and pharmacological modulation of lamin A farnesylation determines its function and turnover. Aging Cell 2024; 23:e14105. [PMID: 38504487 PMCID: PMC11113360 DOI: 10.1111/acel.14105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 01/10/2024] [Accepted: 01/29/2024] [Indexed: 03/21/2024] Open
Abstract
Hutchinson-Gilford Progeria syndrome (HGPS) is a severe premature ageing disorder caused by a 50 amino acid truncated (Δ50AA) and permanently farnesylated lamin A (LA) mutant called progerin. On a cellular level, progerin expression leads to heterochromatin loss, impaired nucleocytoplasmic transport, telomeric DNA damage and a permanent growth arrest called cellular senescence. Although the genetic basis for HGPS has been elucidated 20 years ago, the question whether the Δ50AA or the permanent farnesylation causes cellular defects has not been addressed. Moreover, we currently lack mechanistic insight into how the only FDA-approved progeria drug Lonafarnib, a farnesyltransferase inhibitor (FTI), ameliorates HGPS phenotypes. By expressing a variety of LA mutants using a doxycycline-inducible system, and in conjunction with FTI, we demonstrate that the permanent farnesylation, and not the Δ50AA, is solely responsible for progerin-induced cellular defects, as well as its rapid accumulation and slow clearance. Importantly, FTI does not affect clearance of progerin post-farnesylation and we demonstrate that early, but not late FTI treatment prevents HGPS phenotypes. Collectively, our study unravels the precise contributions of progerin's permanent farnesylation to its turnover and HGPS cellular phenotypes, and how FTI treatment ameliorates these. These findings are applicable to other diseases associated with permanently farnesylated proteins, such as adult-onset autosomal dominant leukodystrophy.
Collapse
Affiliation(s)
- Mattheus Xing Rong Foo
- A*STAR Skin Research Labs, Cell Ageing Laboratory, Skin Research Institute of Singapore, Singapore, Singapore
| | - Peh Fern Ong
- A*STAR Skin Research Labs, Cell Ageing Laboratory, Skin Research Institute of Singapore, Singapore, Singapore
| | - Zi Xuan Yap
- A*STAR Skin Research Labs, Cell Ageing Laboratory, Skin Research Institute of Singapore, Singapore, Singapore
| | - Martina Maric
- A*STAR Skin Research Labs, Cell Ageing Laboratory, Skin Research Institute of Singapore, Singapore, Singapore
| | - Christopher Jue Shi Bong
- A*STAR Skin Research Labs, Cell Ageing Laboratory, Skin Research Institute of Singapore, Singapore, Singapore
| | - Peter Dröge
- LambdaGen Pte. Ltd., Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Brian Burke
- A*STAR Skin Research Labs, Cell Ageing Laboratory, Skin Research Institute of Singapore, Singapore, Singapore
| | - Oliver Dreesen
- A*STAR Skin Research Labs, Cell Ageing Laboratory, Skin Research Institute of Singapore, Singapore, Singapore
| |
Collapse
|
19
|
Kudrin P, Singh A, Meierhofer D, Kuśnierczyk A, Ørom UAV. N4-acetylcytidine (ac4C) promotes mRNA localization to stress granules. EMBO Rep 2024; 25:1814-1834. [PMID: 38413733 PMCID: PMC11014937 DOI: 10.1038/s44319-024-00098-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 02/06/2024] [Accepted: 02/07/2024] [Indexed: 02/29/2024] Open
Abstract
Stress granules are an integral part of the stress response that are formed from non-translating mRNAs aggregated with proteins. While much is known about stress granules, the factors that drive their mRNA localization are incompletely described. Modification of mRNA can alter the properties of the nucleobases and affect processes such as translation, splicing and localization of individual transcripts. Here, we show that the RNA modification N4-acetylcytidine (ac4C) on mRNA associates with transcripts enriched in stress granules and that stress granule localized transcripts with ac4C are specifically translationally regulated. We also show that ac4C on mRNA can mediate localization of the protein NOP58 to stress granules. Our results suggest that acetylation of mRNA regulates localization of both stress-sensitive transcripts and RNA-binding proteins to stress granules and adds to our understanding of the molecular mechanisms responsible for stress granule formation.
Collapse
Affiliation(s)
- Pavel Kudrin
- Institute of Molecular Biology and Genetics, Aarhus University, 8000, Aarhus, Denmark
- Institute of Biomedicine and Translational Medicine, University of Tartu, 50411, Tartu, Estonia
| | - Ankita Singh
- Institute of Biomedicine, Aarhus University, 8000, Aarhus, Denmark
| | - David Meierhofer
- Max Planck Institute for Molecular Genetics, 14195, Berlin, Germany
| | - Anna Kuśnierczyk
- Proteomics and Modomics Experimental Core (PROMEC), Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology and the Central Norway Regional Health Authority, 7030, Trondheim, Norway
| | | |
Collapse
|
20
|
Zhou M, Gamage ST, Tran KA, Bartee D, Wei X, Yin B, Berger S, Meier JL, Marmorstein R. Molecular Basis for RNA Cytidine Acetylation by NAT10. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.27.587050. [PMID: 38585770 PMCID: PMC10996708 DOI: 10.1101/2024.03.27.587050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Human NAT10 acetylates the N4 position of cytidine in RNA, predominantly on rRNA and tRNA, to facilitate ribosome biogenesis and protein translation. NAT10 has been proposed as a therapeutic target in cancers as well as aging-associated pathologies such as Hutchinson-Gilford Progeria Syndrome (HGPS). The ∼120 kDa NAT10 protein uses its acetyl-CoA-dependent acetyltransferase, ATP-dependent helicase, and RNA binding domains in concert to mediate RNA-specific N4-cytidine acetylation. While the biochemical activity of NAT10 is well known, the molecular basis for catalysis of eukaryotic RNA acetylation remains relatively undefined. To provide molecular insights into the RNA-specific acetylation by NAT10, we determined the single particle cryo-EM structures of Chaetomium thermophilum NAT10 ( Ct NAT10) bound to a bisubstrate cytidine-CoA probe with and without ADP. The structures reveal that NAT10 forms a symmetrical heart-shaped dimer with conserved functional domains surrounding the acetyltransferase active sites harboring the cytidine-CoA probe. Structure-based mutagenesis with analysis of mutants in vitro supports the catalytic role of two conserved active site residues (His548 and Tyr549 in Ct NAT10), and two basic patches, both proximal and distal to the active site for RNA-specific acetylation. Yeast complementation analyses and senescence assays in human cells also implicates NAT10 catalytic activity in yeast thermoadaptation and cellular senescence. Comparison of the NAT10 structure to protein lysine and N-terminal acetyltransferase enzymes reveals an unusually open active site suggesting that these enzymes have been evolutionarily tailored for RNA recognition and cytidine-specific acetylation.
Collapse
|
21
|
Tiwari V, Alam MJ, Bhatia M, Navya M, Banerjee SK. The structure and function of lamin A/C: Special focus on cardiomyopathy and therapeutic interventions. Life Sci 2024; 341:122489. [PMID: 38340979 DOI: 10.1016/j.lfs.2024.122489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 01/21/2024] [Accepted: 02/03/2024] [Indexed: 02/12/2024]
Abstract
Lamins are inner nuclear membrane proteins that belong to the intermediate filament family. Lamin A/C lie adjacent to the heterochromatin structure in polymer form, providing skeletal to the nucleus. Based on the localization, lamin A/C provides nuclear stability and cytoskeleton to the nucleus and modulates chromatin organization and gene expression. Besides being the structural protein making the inner nuclear membrane in polymer form, lamin A/C functions as a signalling molecule involved in gene expression as an enhancer inside the nucleus. Lamin A/C regulates various cellular pathways like autophagy and energy balance in the cytoplasm. Its expression is highly variable in differentiated tissues, higher in hard tissues like bone and muscle cells, and lower in soft tissues like the liver and brain. In muscle cells, including the heart, lamin A/C must be expressed in a balanced state. Lamin A/C mutation is linked with various diseases, such as muscular dystrophy, lipodystrophy, and cardiomyopathies. It has been observed that a good number of mutations in the LMNA gene impact cardiac activity and its function. Although several works have been published, there are still several unexplored areas left regarding the lamin A/C function and structure in the cardiovascular system and its pathological state. In this review, we focus on the structural organization, expression pattern, and function of lamin A/C, its interacting partners, and the pathophysiology associated with mutations in the lamin A/C gene, with special emphasis on cardiovascular diseases. With the recent finding on lamin A/C, we have summarized the possible therapeutic interventions to treat cardiovascular symptoms and reverse the molecular changes.
Collapse
Affiliation(s)
- Vikas Tiwari
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Guwahati 781101, Assam, India
| | - Md Jahangir Alam
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Guwahati 781101, Assam, India; Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Madhavi Bhatia
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Guwahati 781101, Assam, India
| | - Malladi Navya
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Guwahati 781101, Assam, India
| | - Sanjay K Banerjee
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Guwahati 781101, Assam, India.
| |
Collapse
|
22
|
Son SM, Park SJ, Breusegem SY, Larrieu D, Rubinsztein DC. p300 nucleocytoplasmic shuttling underlies mTORC1 hyperactivation in Hutchinson-Gilford progeria syndrome. Nat Cell Biol 2024; 26:235-249. [PMID: 38267537 PMCID: PMC10866696 DOI: 10.1038/s41556-023-01338-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: 05/12/2023] [Accepted: 12/14/2023] [Indexed: 01/26/2024]
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) is a master regulator of cell growth, metabolism and autophagy. Multiple pathways modulate mTORC1 in response to nutrients. Here we describe that nucleus-cytoplasmic shuttling of p300/EP300 regulates mTORC1 activity in response to amino acid or glucose levels. Depletion of these nutrients causes cytoplasm-to-nucleus relocalization of p300 that decreases acetylation of the mTORC1 component raptor, thereby reducing mTORC1 activity and activating autophagy. This is mediated by AMP-activated protein kinase-dependent phosphorylation of p300 at serine 89. Nutrient addition to starved cells results in protein phosphatase 2A-dependent dephosphorylation of nuclear p300, enabling its CRM1-dependent export to the cytoplasm to mediate mTORC1 reactivation. p300 shuttling regulates mTORC1 in most cell types and occurs in response to altered nutrients in diverse mouse tissues. Interestingly, p300 cytoplasm-nucleus shuttling is altered in cells from patients with Hutchinson-Gilford progeria syndrome. p300 mislocalization by the disease-causing protein, progerin, activates mTORC1 and inhibits autophagy, phenotypes that are normalized by modulating p300 shuttling. These results reveal how nutrients regulate mTORC1, a cytoplasmic complex, by shuttling its positive regulator p300 in and out of the nucleus, and how this pathway is misregulated in Hutchinson-Gilford progeria syndrome, causing mTORC1 hyperactivation and defective autophagy.
Collapse
Affiliation(s)
- Sung Min Son
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - So Jung Park
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Sophia Y Breusegem
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Delphine Larrieu
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - David C Rubinsztein
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK.
- UK Dementia Research Institute, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK.
| |
Collapse
|
23
|
Hu Z, Lu Y, Cao J, Lin L, Chen X, Zhou Z, Pu J, Chen G, Ma X, Deng Q, Jin Y, Jiang L, Li Y, Li T, Liu J, Zhu S. N-acetyltransferase NAT10 controls cell fates via connecting mRNA cytidine acetylation to chromatin signaling. SCIENCE ADVANCES 2024; 10:eadh9871. [PMID: 38215194 PMCID: PMC10786415 DOI: 10.1126/sciadv.adh9871] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 12/14/2023] [Indexed: 01/14/2024]
Abstract
Cell fate transition involves dynamic changes of gene regulatory network and chromatin landscape, requiring multiple levels of regulation, yet the cross-talk between epitranscriptomic modification and chromatin signaling remains largely unknown. Here, we uncover that suppression of N-acetyltransferase 10 (NAT10), the writer for mRNA N4-acetylcytidine (ac4C) modification, can notably affect human embryonic stem cell (hESC) lineage differentiation and pluripotent reprogramming. With integrative analysis, we identify that NAT10-mediated ac4C modification regulates the protein levels of a subset of its targets, which are strongly enriched for fate-instructive chromatin regulators, and among them, histone chaperone ANP32B is experimentally verified and functionally relevant. Furthermore, NAT10-ac4C-ANP32B axis can modulate the chromatin landscape of their downstream genes (e.g., key regulators of Wnt and TGFβ pathways). Collectively, we show that NAT10 is an essential regulator of cellular plasticity, and its catalyzed mRNA cytidine acetylation represents a critical layer of epitranscriptomic modulation and uncover a previously unrecognized, direct cross-talk between epitranscriptomic modification and chromatin signaling during cell fate transitions.
Collapse
Affiliation(s)
- Zhensheng Hu
- Life Sciences Institute, The Second Affiliated Hospital and School of Medicine, The MOE Key Laboratory of Biosystems Homeostasis and Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yunkun Lu
- Life Sciences Institute, The Second Affiliated Hospital and School of Medicine, The MOE Key Laboratory of Biosystems Homeostasis and Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Jie Cao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, China
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Lianyu Lin
- Life Sciences Institute, The Second Affiliated Hospital and School of Medicine, The MOE Key Laboratory of Biosystems Homeostasis and Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xi Chen
- Life Sciences Institute, The Second Affiliated Hospital and School of Medicine, The MOE Key Laboratory of Biosystems Homeostasis and Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Ziyu Zhou
- Life Sciences Institute, The Second Affiliated Hospital and School of Medicine, The MOE Key Laboratory of Biosystems Homeostasis and Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Jiaqi Pu
- Life Sciences Institute, The Second Affiliated Hospital and School of Medicine, The MOE Key Laboratory of Biosystems Homeostasis and Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Zhejiang University, Hangzhou, Zhejiang 310058, China
- The Children’s Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Guo Chen
- Life Sciences Institute, The Second Affiliated Hospital and School of Medicine, The MOE Key Laboratory of Biosystems Homeostasis and Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xiaojie Ma
- Life Sciences Institute, The Second Affiliated Hospital and School of Medicine, The MOE Key Laboratory of Biosystems Homeostasis and Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Qian Deng
- Life Sciences Institute, The Second Affiliated Hospital and School of Medicine, The MOE Key Laboratory of Biosystems Homeostasis and Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yan Jin
- Life Sciences Institute, The Second Affiliated Hospital and School of Medicine, The MOE Key Laboratory of Biosystems Homeostasis and Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Liling Jiang
- Life Sciences Institute, The Second Affiliated Hospital and School of Medicine, The MOE Key Laboratory of Biosystems Homeostasis and Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yuhan Li
- Life Sciences Institute, The Second Affiliated Hospital and School of Medicine, The MOE Key Laboratory of Biosystems Homeostasis and Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Tengwei Li
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, China
| | - Jianzhao Liu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, China
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Saiyong Zhu
- Life Sciences Institute, The Second Affiliated Hospital and School of Medicine, The MOE Key Laboratory of Biosystems Homeostasis and Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Zhejiang University, Hangzhou, Zhejiang 310058, China
| |
Collapse
|
24
|
Zhang Y, Lei Y, Dong Y, Chen S, Sun S, Zhou F, Zhao Z, Chen B, Wei L, Chen J, Meng Z. Emerging roles of RNA ac4C modification and NAT10 in mammalian development and human diseases. Pharmacol Ther 2024; 253:108576. [PMID: 38065232 DOI: 10.1016/j.pharmthera.2023.108576] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 11/29/2023] [Accepted: 12/01/2023] [Indexed: 01/13/2024]
Abstract
RNA ac4C modification is a novel and rare chemical modification observed in mRNA. Traditional biochemical studies had primarily associated ac4C modification with tRNA and rRNA until in 2018, Arango D et al. first reported the presence of ac4C modification on mRNA and demonstrated its critical role in mRNA stability and translation regulation. Furthermore, they established that the ac4C modification on mRNA is mediated by the classical N-acetyltransferase NAT10. Subsequent studies have underscored the essential implications of NAT10 and mRNA ac4C modification across both physiological and pathological regulatory processes. In this review, we aimed to explore the discovery history of RNA ac4C modification, its detection methods, and its regulatory mechanisms in disease and physiological development. We offer a forward-looking examination and discourse concerning the employment of RNA ac4C modification as a prospective therapeutic strategy across diverse diseases. Furthermore, we comprehensively summarize the functions and mechanisms of NAT10 in gene expression regulation and pathogenesis independent of RNA ac4C modification.
Collapse
Affiliation(s)
- Yigan Zhang
- Institute of Biomedical Research, Department of Infectious Diseases, Regulatory Mechanism and Targeted Therapy for Liver Cancer Shiyan Key Laboratory, Hubei rovincial Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Yumei Lei
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yanbin Dong
- Institute of Biophysics, Chinese Academy of Sciences, Key Laboratory of Nucleic Acid Biology, Chinese Academy of Sciences, Beijing, China
| | - Shuwen Chen
- School of Biomedical Engineering, Hubei University of Medicine, Shiyan, China
| | - Siyuan Sun
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Fange Zhou
- The First Clinical School of Hubei University of Medicine, Shiyan, China
| | - Zhiwen Zhao
- Department of Emergency Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Bonan Chen
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Lv Wei
- Institute of Biophysics, Chinese Academy of Sciences, Key Laboratory of Nucleic Acid Biology, Chinese Academy of Sciences, Beijing, China.
| | - Juan Chen
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China.
| | - Zhongji Meng
- Institute of Biomedical Research, Department of Infectious Diseases, Regulatory Mechanism and Targeted Therapy for Liver Cancer Shiyan Key Laboratory, Hubei rovincial Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, China.
| |
Collapse
|
25
|
Zheng J, Lu Y, Lin Y, Si S, Guo B, Zhao X, Cui L. Epitranscriptomic modifications in mesenchymal stem cell differentiation: advances, mechanistic insights, and beyond. Cell Death Differ 2024; 31:9-27. [PMID: 37985811 PMCID: PMC10782030 DOI: 10.1038/s41418-023-01238-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/24/2023] [Accepted: 11/06/2023] [Indexed: 11/22/2023] Open
Abstract
RNA modifications, known as the "epitranscriptome", represent a key layer of regulation that influences a wide array of biological processes in mesenchymal stem cells (MSCs). These modifications, catalyzed by specific enzymes, often termed "writers", "readers", and "erasers", can dynamically alter the MSCs' transcriptomic landscape, thereby modulating cell differentiation, proliferation, and responses to environmental cues. These enzymes include members of the classes METTL, IGF2BP, WTAP, YTHD, FTO, NAT, and others. Many of these RNA-modifying agents are active during MSC lineage differentiation. This review provides a comprehensive overview of the current understanding of different RNA modifications in MSCs, their roles in regulating stem cell behavior, and their implications in MSC-based therapies. It delves into how RNA modifications impact MSC biology, the functional significance of individual modifications, and the complex interplay among these modifications. We further discuss how these intricate regulatory mechanisms contribute to the functional diversity of MSCs, and how they might be harnessed for therapeutic applications. The review also highlights current challenges and potential future directions in the study of RNA modifications in MSCs, emphasizing the need for innovative tools to precisely map these modifications and decipher their context-specific effects. Collectively, this work paves the way for a deeper understanding of the role of the epitranscriptome in MSC biology, potentially advancing therapeutic strategies in regenerative medicine and MSC-based therapies.
Collapse
Affiliation(s)
- Jiarong Zheng
- Department of Dentistry, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Ye Lu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Yunfan Lin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Shanshan Si
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Bing Guo
- Department of Dentistry, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China.
| | - Xinyuan Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China.
| | - Li Cui
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China.
- Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, 90095, CA, USA.
| |
Collapse
|
26
|
Schertzer M, Jullien L, Pinto AL, Calado RT, Revy P, Londoño-Vallejo A. Human RTEL1 Interacts with KPNB1 (Importin β) and NUP153 and Connects Nuclear Import to Nuclear Envelope Stability in S-Phase. Cells 2023; 12:2798. [PMID: 38132118 PMCID: PMC10741959 DOI: 10.3390/cells12242798] [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: 11/07/2023] [Revised: 11/29/2023] [Accepted: 12/03/2023] [Indexed: 12/23/2023] Open
Abstract
Regulator of TElomere Length Helicase 1 (RTEL1) is a helicase required for telomere maintenance and genome replication and repair. RTEL1 has been previously shown to participate in the nuclear export of small nuclear RNAs. Here we show that RTEL1 deficiency leads to a nuclear envelope destabilization exclusively in cells entering S-phase and in direct connection to origin firing. We discovered that inhibiting protein import also leads to similar, albeit non-cell cycle-related, nuclear envelope disruptions. Remarkably, overexpression of wild-type RTEL1, or of its C-terminal part lacking the helicase domain, protects cells against nuclear envelope anomalies mediated by protein import inhibition. We identified distinct domains in the C-terminus of RTEL1 essential for the interaction with KPNB1 (importin β) and NUP153, respectively, and we demonstrated that, on its own, the latter domain can promote the dynamic nuclear internalization of peptides that freely diffuse through the nuclear pore. Consistent with putative functions exerted in protein import, RTEL1 can be visualized on both sides of the nuclear pore using high-resolution microscopy. In all, our work points to an unanticipated, helicase-independent, role of RTEL1 in connecting both nucleocytoplasmic trafficking and nuclear envelope integrity to genome replication initiation in S-phase.
Collapse
Affiliation(s)
- Michael Schertzer
- Institut Curie, PSL Research University, CNRS, UMR3244, F-75005 Paris, France;
- Sorbonne Universités, CNRS, UMR3244, F-75005 Paris, France
| | - Laurent Jullien
- INSERM UMR 1163, Laboratory of Genome Dynamics in the Immune System, Equipe Labellisée Ligue Contre le Cancer, F-75006 Paris, France; (L.J.); (P.R.)
- Paris Descartes–Sorbonne Paris Cité University, Imagine Institute, F-75015 Paris, France
| | - André L. Pinto
- Department of Medical Imaging, Hematology, and Oncology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14049-900, Brazil; (A.L.P.); (R.T.C.)
| | - Rodrigo T. Calado
- Department of Medical Imaging, Hematology, and Oncology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14049-900, Brazil; (A.L.P.); (R.T.C.)
| | - Patrick Revy
- INSERM UMR 1163, Laboratory of Genome Dynamics in the Immune System, Equipe Labellisée Ligue Contre le Cancer, F-75006 Paris, France; (L.J.); (P.R.)
- Paris Descartes–Sorbonne Paris Cité University, Imagine Institute, F-75015 Paris, France
| | - Arturo Londoño-Vallejo
- Institut Curie, PSL Research University, CNRS, UMR3244, F-75005 Paris, France;
- Sorbonne Universités, CNRS, UMR3244, F-75005 Paris, France
| |
Collapse
|
27
|
Kurian L, Brandes RP. RNA Modification That Breaks the Heart: RNA Acetylase Nat10 Promotes Fibrosis. Circ Res 2023; 133:1003-1005. [PMID: 38060683 DOI: 10.1161/circresaha.123.323866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Affiliation(s)
- Leo Kurian
- Institute for Cardiovascular Physiology, Goethe University Frankfurt, Germany (L.K., R.P.B.)
| | - Ralf P Brandes
- Institute for Cardiovascular Physiology, Goethe University Frankfurt, Germany (L.K., R.P.B.)
- German Center for Cardiovascular Research (DZHK), Germany (R.P.B.)
| |
Collapse
|
28
|
Liu Y, Huang H, Zhang C, Fan H. N-acetyltransferase 10 promotes the progression of oral squamous cell carcinoma through N4-acetylcytidine RNA acetylation of MMP1 mRNA. Cancer Sci 2023; 114:4202-4215. [PMID: 37705232 PMCID: PMC10637085 DOI: 10.1111/cas.15946] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/20/2023] [Accepted: 07/26/2023] [Indexed: 09/15/2023] Open
Abstract
The pathogenesis of oral squamous cell carcinoma (OSCC) remains unclear. Therefore, clarifying its pathogenesis and molecular-level development mechanism has become the focus of OSCC research. N-acetyltransferase 10 (NAT10) is a crucial enzyme involved in mRNA acetylation, regulating target gene expression and biological functions of various diseases through mediating N4-acetylcytidine (ac4C) acetylation. However, its role in OSCC progression is not well understood. In this study, we showed that NAT10 was significantly upregulated in OSCC tissues compared to normal oral tissues. Moreover, lentivirus-mediated NAT10 knockdown markedly suppressed cell proliferation, migration, and invasion in two OSCC cell lines (SCC-9 and SCC-15). Interestingly, MMP1 was found to be significantly upregulated in OSCC tissues and was a potential target of NAT10. N-acetyltransferase 10 knockdown significantly reduced both the total and ac4C acetylated levels of MMP1 mRNA and decreased its mRNA stability. Xenograft experiments further confirmed the inhibitory effect of NAT10 knockdown on the tumorigenesis and metastasis ability of OSCC cells and decreased MMP1 expression in vivo. Additionally, NAT10 knockdown impaired the proliferation, migration, and invasion abilities in OSCC cell lines in an MMP1-dependent manner. Our results suggest that NAT10 acts as an oncogene in OSCC, and targeting ac4C acetylation could be a promising therapeutic strategy for OSCC treatment.
Collapse
Affiliation(s)
- Yi Liu
- Department of StomatologyThe First Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Hui Huang
- Department of StomatologyThe First Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Cun‐bao Zhang
- Department of Stomatology, Affiliated Hangzhou First People's HospitalZhejiang University School of MedicineZhejiangChina
| | - Hua‐nan Fan
- Department of StomatologyThe First Affiliated Hospital of Nanchang UniversityNanchangChina
| |
Collapse
|
29
|
Wei W, Zhang S, Han H, Wang X, Zheng S, Wang Z, Yang C, Wang L, Ma J, Guo S, Wang J, Liu L, Choe J, Lin S. NAT10-mediated ac4C tRNA modification promotes EGFR mRNA translation and gefitinib resistance in cancer. Cell Rep 2023; 42:112810. [PMID: 37463108 DOI: 10.1016/j.celrep.2023.112810] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 05/22/2023] [Accepted: 06/28/2023] [Indexed: 07/20/2023] Open
Abstract
Aberrant RNA modifications are frequently associated with cancers, while the underlying mechanisms and clinical significance remain poorly understood. Here, we find that the ac4C RNA acetyltransferase NAT10 is significantly upregulated in esophageal cancers (ESCAs) and associated with poor ESCA prognosis. In addition, using ESCA cell lines and mouse models, we confirm the critical functions of NAT10 in promoting ESCA tumorigenesis and progression in vitro and in vivo. Mechanistically, NAT10 depletion reduces the abundance of ac4C-modified tRNAs and decreases the translation efficiencies of mRNAs enriched for ac4C-modified tRNA-decoded codons. We further identify EGFR as a key downstream target that facilitates NAT10's oncogenic functions. In terms of clinical significance, we demonstrate that NAT10 depletion and gefitinib treatment synergistically inhibit ESCA progression in vitro and in vivo. Our data indicate the mechanisms underlying ESCA progression at the layer of mRNA translation control and provide molecular insights for the development of effective cancer therapeutic strategies.
Collapse
Affiliation(s)
- Wei Wei
- Department of Otolaryngology, Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Shuishen Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Hui Han
- Department of Otolaryngology, Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Xiaochen Wang
- Department of Otolaryngology, Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Siyi Zheng
- Department of Otolaryngology, Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Zhaoyu Wang
- Department of Otolaryngology, Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Chunlong Yang
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Lu Wang
- Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Jieyi Ma
- Department of Otolaryngology, Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Siyao Guo
- Department of Otolaryngology, Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Juan Wang
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Lianlian Liu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Junho Choe
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 04763, Republic of Korea; Research Institute for Natural Sciences, Hanyang University, Seoul 04763, Republic of Korea; Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul 04763, Republic of Korea; Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul 04763, Republic of Korea
| | - Shuibin Lin
- Department of Otolaryngology, Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
30
|
Luo J, Cao J, Chen C, Xie H. Emerging role of RNA acetylation modification ac4C in diseases: Current advances and future challenges. Biochem Pharmacol 2023; 213:115628. [PMID: 37247745 DOI: 10.1016/j.bcp.2023.115628] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/20/2023] [Accepted: 05/23/2023] [Indexed: 05/31/2023]
Abstract
The oldest known highly conserved modification of RNA, N4-acetylcytidine, is widely distributed from archaea to eukaryotes and acts as a posttranscriptional chemical modification of RNA, contributing to the correct reading of specific nucleotide sequences during translation, stabilising mRNA and improving transcription efficiency. Yeast Kre33 and human NAT10, the only known authors of ac4C, modify tRNA with the help of the Tan1/THUMPD1 adapter to stabilise its structure. Currently, the mRNA for N4-acetylcytidine (ac4C), catalysed by NAT10 (N-acetyltransferase 10), has been implicated in a variety of human diseases, particularly cancer. This article reviews advances in the study of ac4C modification of RNA and the ac4C-related gene NAT10 in normal physiological cell development, cancer, premature disease and viral infection and discusses its therapeutic promise and future research challenges.
Collapse
Affiliation(s)
- Jie Luo
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Jingsong Cao
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Institute of Clinical Medicine, University of South China, Hengyang 421001, China
| | - Cong Chen
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Haitao Xie
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
31
|
Cisneros B, García-Aguirre I, De Ita M, Arrieta-Cruz I, Rosas-Vargas H. Hutchinson-Gilford Progeria Syndrome: Cellular Mechanisms and Therapeutic Perspectives. Arch Med Res 2023; 54:102837. [PMID: 37390702 DOI: 10.1016/j.arcmed.2023.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 05/26/2023] [Accepted: 06/14/2023] [Indexed: 07/02/2023]
Abstract
In humans, aging is characterized by a gradual decline of physical and psychological functions, with the concomitant onset of chronic-degenerative diseases, which ultimately lead to death. The study of Hutchinson-Gilford progeria syndrome (HGPS), a premature aging disorder that recapitulates several features of natural aging, has provided important insights into deciphering the aging process. The genetic origin of HGPS is a de novo point mutation in the LMNA gene that drives the synthesis of progerin, mutant version of lamin A. Progerin is aberrantly anchored to the nuclear envelope disrupting a plethora of molecular processes; nonetheless, how progerin exerts a cascade of deleterious alterations at the cellular and systemic levels is not fully understood. Over the past decade, the use of different cellular and animal models for HGPS has allowed the identification of the molecular mechanisms underlying HGPS, paving the way towards the development of therapeutic treatments against the disease. In this review, we present an updated overview of the biology of HGPS, including its clinical features, description of key cellular processes affected by progerin (nuclear morphology and function, nucleolar activity, mitochondrial function, protein nucleocytoplasmic trafficking and telomere homeostasis), as well as discussion of the therapeutic strategies under development.
Collapse
Affiliation(s)
- Bulmaro Cisneros
- Genetics and Molecular Biology Department, Research and Advanced Studies Center, National Polytechnical Institute, Mexico City, Mexico
| | - Ian García-Aguirre
- Genetics and Molecular Biology Department, Research and Advanced Studies Center, National Polytechnical Institute, Mexico City, Mexico; Bioengineering Department, School of Engineering and Sciences, Tecnológico de Monterrey, Mexico City, Mexico
| | - Marlon De Ita
- Genetics and Molecular Biology Department, Research and Advanced Studies Center, National Polytechnical Institute, Mexico City, Mexico; Medical Research Unit in Human Genetics, Pediatrics Hospital, 21st Century National Medical Center, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Isabel Arrieta-Cruz
- Basic Research Department, Research Direction, National Institute of Geriatrics, Ministry of Health, Mexico City, Mexico
| | - Haydeé Rosas-Vargas
- Medical Research Unit in Human Genetics, Pediatrics Hospital, 21st Century National Medical Center, Instituto Mexicano del Seguro Social, Mexico City, Mexico.
| |
Collapse
|
32
|
Fragoso-Luna A, Askjaer P. The Nuclear Envelope in Ageing and Progeria. Subcell Biochem 2023; 102:53-75. [PMID: 36600129 DOI: 10.1007/978-3-031-21410-3_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Development from embryo to adult, organismal homeostasis and ageing are consecutive processes that rely on several functions of the nuclear envelope (NE). The NE compartmentalises the eukaryotic cells and provides physical stability to the genetic material in the nucleus. It provides spatiotemporal regulation of gene expression by controlling nuclear import and hence access of transcription factors to target genes as well as organisation of the genome into open and closed compartments. In addition, positioning of chromatin relative to the NE is important for DNA replication and repair and thereby also for genome stability. We discuss here the relevance of the NE in two classes of age-related human diseases. Firstly, we focus on the progeria syndromes Hutchinson-Gilford (HGPS) and Nestor-Guillermo (NGPS), which are caused by mutations in the LMNA and BANF1 genes, respectively. Both genes encode ubiquitously expressed components of the nuclear lamina that underlines the nuclear membranes. HGPS and NGPS patients manifest symptoms of accelerated ageing and cells from affected individuals show similar defects as cells from healthy old donors, including signs of increased DNA damage and epigenetic alternations. Secondly, we describe how several age-related neurodegenerative diseases, such as amyotrophic lateral sclerosis and Huntington's disease, are related with defects in nucleocytoplasmic transport. A common feature of this class of diseases is the accumulation of nuclear pore proteins and other transport factors in inclusions. Importantly, genetic manipulations of the nucleocytoplasmic transport machinery can alleviate disease-related phenotypes in cell and animal models, paving the way for potential therapeutic interventions.
Collapse
Affiliation(s)
- Adrián Fragoso-Luna
- Andalusian Centre for Developmental Biology, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Pablo de Olavide, Sevilla, Spain
| | - Peter Askjaer
- Andalusian Centre for Developmental Biology, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Pablo de Olavide, Sevilla, Spain.
| |
Collapse
|
33
|
The chaperone DNAJB6 surveils FG-nucleoporins and is required for interphase nuclear pore complex biogenesis. Nat Cell Biol 2022; 24:1584-1594. [PMID: 36302971 DOI: 10.1038/s41556-022-01010-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 09/12/2022] [Indexed: 01/18/2023]
Abstract
Biogenesis of nuclear pore complexes (NPCs) includes the formation of the permeability barrier composed of phenylalanine-glycine-rich nucleoporins (FG-Nups) that regulate the selective passage of biomolecules across the nuclear envelope. The FG-Nups are intrinsically disordered and prone to liquid-liquid phase separation and aggregation when isolated. How FG-Nups are protected from making inappropriate interactions during NPC biogenesis is not fully understood. Here we find that DNAJB6, a molecular chaperone of the heat shock protein network, forms foci in close proximity to NPCs. The number of these foci decreases upon removal of proteins involved in the early steps of interphase NPC biogenesis. Conversely, when this process is stalled in the last steps, the number of DNAJB6-containing foci increases and these foci are identified as herniations at the nuclear envelope. Immunoelectron tomography shows that DNAJB6 localizes inside the lumen of the herniations arising at NPC biogenesis intermediates. Loss of DNAJB6 results in the accumulation of cytosolic annulate lamellae, which are structures containing partly assembled NPCs, a feature associated with disturbances in NPC biogenesis. We find that DNAJB6 binds to FG-Nups and can prevent the aggregation of the FG region of several FG-Nups in cells and in vitro. Together, our data show that the molecular chaperone DNAJB6 provides quality control during NPC biogenesis and is involved in the surveillance of native intrinsically disordered FG-Nups.
Collapse
|
34
|
Zheng X, Wang Q, Zhou Y, Zhang D, Geng Y, Hu W, Wu C, Shi Y, Jiang J. N-acetyltransferase 10 promotes colon cancer progression by inhibiting ferroptosis through N4-acetylation and stabilization of ferroptosis suppressor protein 1 (FSP1) mRNA. CANCER COMMUNICATIONS (LONDON, ENGLAND) 2022; 42:1347-1366. [PMID: 36209353 PMCID: PMC9759759 DOI: 10.1002/cac2.12363] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 05/18/2022] [Accepted: 09/15/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND N-acetyltransferase 10 (NAT10) is the only enzyme known to mediate the N4-acetylcytidine (ac4C) modification of mRNA and is crucial for mRNA stability and translation efficiency. However, its role in cancer development and prognosis has not yet been explored. This study aimed to examine the possible role of NAT10 in colon cancer. METHODS The expression levels of NAT10 were evaluated by immunohistochemical analyses with a colon cancer tissue microarray, and its prognostic value in patients was further analyzed. Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting were performed to analyze NAT10 expression in harvested colon cancer tissues and cell lines. Stable NAT10-knockdown and NAT10-overexpressing colon cancer cell lines were constructed using lentivirus. The biological functions of NAT10 in colon cancer cell lines were analyzed in vitro by Cell Counting Kit-8 (CCK-8), wound healing, Transwell, cell cycle, and ferroptosis assays. Xenograft models were used to analyze the effect of NAT10 on the tumorigenesis and metastasis of colon cancer cells in vivo. Dot blotting, acetylated RNA immunoprecipitation-qPCR, and RNA stability analyses were performed to explore the mechanism by which NAT10 functions in colon cancer progression. RESULTS NAT10 was upregulated in colon cancer tissues and various colon cancer cell lines. This increased NAT10 expression was associated with shorter patient survival. Knockdown of NAT10 in two colon cancer cell lines (HT-29 and LoVo) impaired the proliferation, migration, invasion, tumor formation and metastasis of these cells, whereas overexpression of NAT10 promoted these abilities. Further analysis revealed that NAT10 exerted a strong effect on the mRNA stability and expression of ferroptosis suppressor protein 1 (FSP1) in HT-29 and LoVo cells. In these cells, FSP1 mRNA was found to be modified by ac4C acetylation, and this epigenetic modification was associated with the inhibition of ferroptosis. CONCLUSIONS Our study revealed that NAT10 plays a critical role in colon cancer development by affecting FSP1 mRNA stability and ferroptosis, suggesting that NAT10 could be a novel prognostic and therapeutic target in colon cancer.
Collapse
Affiliation(s)
- Xiao Zheng
- Department of Tumor Biological Treatmentthe Third Affiliated Hospital of Soochow UniversityChangzhouJiangsu213003P. R. China,Jiangsu Engineering Research Center for Tumor ImmunotherapyChangzhouJiangsu213003P. R. China,Institute for Cell Therapy of Soochow UniversityChangzhouJiangsu213003P. R. China
| | - Qi Wang
- Department of Tumor Biological Treatmentthe Third Affiliated Hospital of Soochow UniversityChangzhouJiangsu213003P. R. China,Jiangsu Engineering Research Center for Tumor ImmunotherapyChangzhouJiangsu213003P. R. China,Institute for Cell Therapy of Soochow UniversityChangzhouJiangsu213003P. R. China
| | - You Zhou
- Department of Tumor Biological Treatmentthe Third Affiliated Hospital of Soochow UniversityChangzhouJiangsu213003P. R. China,Jiangsu Engineering Research Center for Tumor ImmunotherapyChangzhouJiangsu213003P. R. China,Institute for Cell Therapy of Soochow UniversityChangzhouJiangsu213003P. R. China
| | - Dachuan Zhang
- Department of Pathologythe Third Affiliated Hospital of Soochow UniversityChangzhouJiangsu213003P. R. China
| | - Yiting Geng
- Jiangsu Engineering Research Center for Tumor ImmunotherapyChangzhouJiangsu213003P. R. China,Department of Oncologythe Third Affiliated Hospital of Soochow UniversityChangzhouJiangsu213003P. R. China
| | - Wenwei Hu
- Jiangsu Engineering Research Center for Tumor ImmunotherapyChangzhouJiangsu213003P. R. China,Department of Oncologythe Third Affiliated Hospital of Soochow UniversityChangzhouJiangsu213003P. R. China
| | - Changping Wu
- Department of Tumor Biological Treatmentthe Third Affiliated Hospital of Soochow UniversityChangzhouJiangsu213003P. R. China,Jiangsu Engineering Research Center for Tumor ImmunotherapyChangzhouJiangsu213003P. R. China,Department of Oncologythe Third Affiliated Hospital of Soochow UniversityChangzhouJiangsu213003P. R. China
| | - Yufang Shi
- Department of Tumor Biological Treatmentthe Third Affiliated Hospital of Soochow UniversityChangzhouJiangsu213003P. R. China,Jiangsu Engineering Research Center for Tumor ImmunotherapyChangzhouJiangsu213003P. R. China,Institute for Translational Medicine of Soochow UniversitySuzhouJiangsu215000P. R. China,CAS Key Laboratory of Tissue Microenvironment and TumorShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031P. R. China
| | - Jingting Jiang
- Department of Tumor Biological Treatmentthe Third Affiliated Hospital of Soochow UniversityChangzhouJiangsu213003P. R. China,Jiangsu Engineering Research Center for Tumor ImmunotherapyChangzhouJiangsu213003P. R. China,Institute for Cell Therapy of Soochow UniversityChangzhouJiangsu213003P. R. China,State Key Laboratory of Pharmaceutical BiotechnologyNanjing UniversityNanjingJiangsu210023P. R. China
| |
Collapse
|
35
|
NAT10 regulates neutrophil pyroptosis in sepsis via acetylating ULK1 RNA and activating STING pathway. Commun Biol 2022; 5:916. [PMID: 36068299 PMCID: PMC9448771 DOI: 10.1038/s42003-022-03868-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 08/22/2022] [Indexed: 11/28/2022] Open
Abstract
Emerging evidence suggests that pyroptosis is involved in sepsis. However, the role of neutrophil pyroptosis in sepsis and the mechanisms remains elusive. We find that N-acetyltransferase 10 (NAT10), an acetyltransferase responsible for the N4-acetylation of Cytidine (ac4C) in mRNA, is significantly downregulated in neutrophils from septic mice. Neutrophil-specific over-expression of NAT10 improves the survival and ameliorates lung injury in septic mice by inhibiting neutrophil pyroptosis. Notably, UNC-52-like kinase 1 (ULK1) is identified as the target of NAT10 in neutrophils. The decreased expression of NAT10 resultes in the decay of ULK1 transcripts and therefore the reduced expression of ULK1. As a regulator of STING phosphorylation, the loss of ULK1 enhances the activation of STING-IRF3 signaling and subsequently the elevated pyroptosis-inducing NLRP3 inflammasome in neutrophils. While over-expression of NAT10 restrains pyroptosis in neutrophils as well as septic lethality in mice by reversing the ULK1-STING-NLRP3 axis. The decreased expression of NAT10 are also observed in sepsis patients and its correlation with clinical severity is found. Collectively, our findings disclose that NAT10 is a negative regulator of neutrophil pyroptosis and its downregulation contributes to the progress of sepsis by exacerbating pyroptosis via the ULK1-STING-NLRP3 axis, therefore revealing a potential therapeutic target for sepsis. The enzyme N-acetyltransferase NAT10 is a negative regulator of neutrophil pyroptosis and its downregulation contributes to the progress of sepsis by exacerbating pyroptosis via the ULK1-STING-NLRP3 pathway.
Collapse
|
36
|
Gunkel P, Cordes VC. ZC3HC1 is a structural element of the nuclear basket effecting interlinkage of TPR polypeptides. Mol Biol Cell 2022; 33:ar82. [PMID: 35609216 DOI: 10.1091/mbc.e22-02-0037] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The nuclear basket (NB), anchored to the nuclear pore complex (NPC), is commonly looked upon as a structure built solely of protein TPR polypeptides, the latter thus regarded as the NB's only scaffold-forming components. In the current study, we report ZC3HC1 as a second structural element of the NB. Recently described as an NB-appended protein omnipresent in vertebrates, we now show that ZC3HC1, both in vivo and in vitro, enables in a stepwise manner the recruitment of TPR subpopulations to the NB and their linkage to already NPC-anchored TPR polypeptides. We further demonstrate that the degron-mediated rapid elimination of ZC3HC1 results in the prompt detachment of the ZC3HC1-appended TPR polypeptides from the NB and their release into the nucleoplasm, underscoring the role of ZC3HC1 as a natural structural element of the NB. Finally, we show that ZC3HC1 can keep TPR polypeptides positioned and linked to each other even at sites remote from the NB, in line with ZC3HC1 functioning as a protein connecting TPR polypeptides.
Collapse
Affiliation(s)
- Philip Gunkel
- Max Planck Institute for Multidisciplinary Sciences, 37077 Göttingen, Germany
| | - Volker C Cordes
- Max Planck Institute for Multidisciplinary Sciences, 37077 Göttingen, Germany
| |
Collapse
|
37
|
Buchinskaya NV, Akhenbekova AZ, Bugybay AA, Kostik MM. Progeria (Hutchinson-Gilford Syndrome): Literature Review and Clinical Case. CURRENT PEDIATRICS 2022; 21:253-264. [DOI: 10.15690/vsp.v21i3.2431] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Progeria, or Hutchinson-Gilford Syndrome is a rare disease from the group of laminopathies characterized by premature aging with skin, bones and cardiovascular system lesions. Pathogenesis is based on pathogenic variants in the LMNA gene leading to anomalies in the nuclear membrane morphology, gene expression disruption, chromatin structure changes, mitochondrial dysfunction, DNA repair and alternative splicing defects, and telomere shortening acceleration. Major manifestations of the disease are: skin lesions (scleroderma-like syndrome and pigmented lesions), lipodystrophy, late teeth eruption, teeth crowding, alopecia, nail dystrophy, osteolysis of distal phalanges, hip joints valgus deformation, joints contractures, atherosclerosis, hearing loss, early heart attacks and strokes. Scleroderma-like skin changes, osteoporosis, flexion contractures of hands’ interphalangeal joints, and hip joints osteoarthritis require differential diagnosis with rheumatic diseases. The basic strategy in management of patients with progeria is the prevention and treatment of its cardiovascular manifestations (early strokes and heart attacks, arterial hypertension, and atherosclerosis), as well as the increase of patients’ quality of life and daily activity. The efficacy of therapy in patients with progeria via the use of farnesyltransferase inhibitors (monotherapy; combination with bisphosphonates or statins), retinoids, and 1,25(OH)2 — vitamin D3 is studied. This literature review is updated with clinical case description of a girl with progeria. The diagnosis was confirmed by sequencing of the LMNA gene (Sanger), and previously described pathogenic variant in exon 11 (c.1824C>T, rs58596362) in the heterozygous state (p.Gly608Gly, NM_170707.3) was revealed.
Collapse
Affiliation(s)
| | | | | | - Mikhail M. Kostik
- Saint-Petersburg State Pediatric Medical University; Almazov National Medical Research Centre
| |
Collapse
|
38
|
Stiekema M, Houben F, Verheyen F, Borgers M, Menzel J, Meschkat M, van Zandvoort MAMJ, Ramaekers FCS, Broers JLV. The Role of Lamins in the Nucleoplasmic Reticulum, a Pleiomorphic Organelle That Enhances Nucleo-Cytoplasmic Interplay. Front Cell Dev Biol 2022; 10:914286. [PMID: 35784476 PMCID: PMC9243388 DOI: 10.3389/fcell.2022.914286] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/24/2022] [Indexed: 12/15/2022] Open
Abstract
Invaginations of the nuclear membrane occur in different shapes, sizes, and compositions. Part of these pleiomorphic invaginations make up the nucleoplasmic reticulum (NR), while others are merely nuclear folds. We define the NR as tubular invaginations consisting of either both the inner and outer nuclear membrane, or only the inner nuclear membrane. Specifically, invaginations of both the inner and outer nuclear membrane are also called type II NR, while those of only the inner nuclear membrane are defined as type I NR. The formation and structure of the NR is determined by proteins associated to the nuclear membrane, which induce a high membrane curvature leading to tubular invaginations. Here we review and discuss the current knowledge of nuclear invaginations and the NR in particular. An increase in tubular invaginations of the nuclear envelope is associated with several pathologies, such as laminopathies, cancer, (reversible) heart failure, and Alzheimer’s disease. Furthermore, viruses can induce both type I and II NR. In laminopathies, the amount of A-type lamins throughout the nucleus is generally decreased or the organization of lamins or lamin-associated proteins is disturbed. Also, lamin overexpression or modulation of lamin farnesylation status impacts NR formation, confirming the importance of lamin processing in NR formation. Virus infections reorganize the nuclear lamina via (de)phosphorylation of lamins, leading to an uneven thickness of the nuclear lamina and in turn lobulation of the nuclear membrane and the formation of invaginations of the inner nuclear membrane. Since most studies on the NR have been performed with cell cultures, we present additional proof for the existence of these structures in vivo, focusing on a variety of differentiated cardiovascular and hematopoietic cells. Furthermore, we substantiate the knowledge of the lamin composition of the NR by super-resolution images of the lamin A/C and B1 organization. Finally, we further highlight the essential role of lamins in NR formation by demonstrating that (over)expression of lamins can induce aberrant NR structures.
Collapse
Affiliation(s)
- Merel Stiekema
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands
- GROW-School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Frederik Houben
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands
- Department of Healthcare, PXL University College, Hasselt, Belgium
| | - Fons Verheyen
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Marcel Borgers
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | | | | | - Marc A. M. J. van Zandvoort
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands
- GROW-School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, Netherlands
- CARIM-School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, Netherlands
- Institute for Molecular Cardiovascular Research IMCAR, RWTH Aachen University, Aachen, Germany
| | - Frans C. S. Ramaekers
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands
- GROW-School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Jos L. V. Broers
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands
- GROW-School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, Netherlands
- CARIM-School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, Netherlands
- *Correspondence: Jos L. V. Broers,
| |
Collapse
|
39
|
Meinema AC, Marzelliusardottir A, Mirkovic M, Aspert T, Lee SS, Charvin G, Barral Y. DNA circles promote yeast ageing in part through stimulating the reorganization of nuclear pore complexes. eLife 2022; 11:71196. [PMID: 35373738 PMCID: PMC9020822 DOI: 10.7554/elife.71196] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 04/03/2022] [Indexed: 11/13/2022] Open
Abstract
The nuclear pore complex (NPC) mediates nearly all exchanges between nucleus and cytoplasm, and in many species it changes composition as the organism ages. However, how these changes arise and whether they contribute themselves to ageing is poorly understood. We show that SAGA-dependent attachment of DNA circles to NPCs in replicatively ageing yeast cells causes NPCs to lose their nuclear basket and cytoplasmic complexes. These NPCs were not recognized as defective by the NPC quality control machinery (SINC) and not targeted by ESCRTs. They interacted normally or more effectively with protein import and export factors but specifically lost mRNA export factors. Acetylation of Nup60 drove the displacement of basket and cytoplasmic complexes from circle-bound NPCs. Mutations preventing this remodeling extended the replicative lifespan of the cells. Thus, our data suggest that the anchorage of accumulating circles locks NPCs in a specialized state and that this process is intrinsically linked to the mechanisms by which ERCs promote ageing.
Collapse
Affiliation(s)
| | | | | | - Théo Aspert
- Department of Developmental Biology and Stem Cells, Institute of Genetics and Molecular and Cellular Biology, Illkirch, France
| | - Sung Sik Lee
- Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Gilles Charvin
- Department of Developmental Biology and Stem Cells, Institute of Genetics and Molecular and Cellular Biology, Illkirch, France
| | - Yves Barral
- Department of Biology, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
40
|
Sex-Specific Expression of Non-Coding RNA Fragments in Frontal Cortex, Hippocampus and Cerebellum of Rats. EPIGENOMES 2022; 6:epigenomes6020011. [PMID: 35466186 PMCID: PMC9036230 DOI: 10.3390/epigenomes6020011] [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: 02/02/2022] [Revised: 03/26/2022] [Accepted: 03/28/2022] [Indexed: 12/04/2022] Open
Abstract
Non-coding RNA fragments (ncRFs) are processed from various non-coding RNAs (ncRNAs), with the most abundant being those produced from tRNAs. ncRFs were reported in many animal and plant species. Many ncRFs exhibit tissue specificity or/and are affected by stress. There is, however, only a handful of reports that describe differential expression of ncRFs in the brain regions. In this work, we analyzed the abundance of ncRFs processed from four major ncRNAs, including tRNA (tRFs), snoRNA (snoRFs), snRNA (snRFs), and rRNA (rRFs) in the frontal cortex (FC), hippocampus (HIP), and cerebellum (CER) of male and female rats. We found brain-specific and sex-specific differences. Reads mapping to lincRNAs were significantly larger in CER as compared to HIP and CER, while those mapping to snRNAs and tRNA were smaller in HIP than in FC and CER. tRF reads were the most abundant among all ncRF reads, and FC had more reads than HIP and CER. Reads mapping to antisense ncRNAs were significantly larger in females than in males in FC. Additionally, males consistently had more tRF, snRF, and snoRF reads in all brain regions. rRFs were more abundant in males in FC and females in HIP. Several tRFs were significantly underrepresented, including tRF-ValCAC, tRF-ValACC, and tRF-LysCTT in all brain regions. We also found brain- and sex-specific differences in the number of brain function-related mRNA targets. To summarize, we found sex-specific differences in the expression of several ncRNA fragments in various brain regions of healthy rats.
Collapse
|
41
|
NAT10 regulates mitotic cell fate by acetylating Eg5 to control bipolar spindle assembly and chromosome segregation. Cell Death Differ 2022; 29:846-860. [PMID: 35210604 PMCID: PMC8989979 DOI: 10.1038/s41418-021-00899-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 11/07/2021] [Accepted: 11/09/2021] [Indexed: 02/07/2023] Open
Abstract
Cell fate of mitotic cell is controlled by spindle assembly. Deficient spindle assembly results in mitotic catastrophe leading to cell death to maintain cellular homeostasis. Therefore, inducing mitotic catastrophe provides a strategy for tumor therapy. Nucleolar acetyltransferase NAT10 has been found to regulate various cellular processes to maintain cell homeostasis. Here we report that NAT10 regulates mitotic cell fate by acetylating Eg5. NAT10 depletion results in multinuclear giant cells, which is the hallmark of mitotic catastrophe. Live-cell imaging showed that knockdown of NAT10 dramatically prolongs the mitotic time and induces defective chromosome segregation including chromosome misalignment, bridge and lagging. NAT10 binds and co-localizes with Eg5 in the centrosome during mitosis. Depletion of NAT10 reduces the centrosome loading of Eg5 and impairs the poleward movement of centrosome, leading to monopolar and asymmetrical spindle formation. Furthermore, NAT10 stabilizes Eg5 through its acetyltransferase function. NAT10 acetylates Eg5 at K771 to control Eg5 stabilization. We generated K771-Ac specific antibody and showed that Eg5 K771-Ac specifically localizes in the centrosome during mitosis. Additionally, K771 acetylation is required for the motor function of Eg5. The hyper-acetylation mimic Flag-Eg5 K771Q but not Flag-Eg5 rescued the NAT10 depletion-induced defective spindle formation and mitotic catastrophe, demonstrating that NAT10 controls mitosis through acetylating Eg5 K771. Collectively, we identify Eg5 as an important substrate of NAT10 in the control of mitosis and provide K771 as an essential acetylation site in the stabilization and motor function of Eg5. Our findings reveal that targeting the NAT10-mediated Eg5 K771 acetylation provides a potential strategy for tumor therapy.
Collapse
|
42
|
Kawakami S, Yoshitane H, Morimura T, Kimura W, Fukada Y. Diurnal shift of mouse activity by the deficiency of an aging-related gene Lmna. J Biochem 2022; 171:509-518. [PMID: 35137145 DOI: 10.1093/jb/mvac015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 02/02/2022] [Indexed: 11/13/2022] Open
Abstract
Nuclear lamina is a fundamental structure of the cell nucleus and regulates a wide range of molecular pathways. Defects of components of the nuclear lamina cause aging-like physiological disorders, called laminopathy. Generally, aging and diseases are often associated with perturbation of various time-of-day-dependent regulations, but it remains still elusive whether laminopathy induces any changes of the circadian clock and physiological rhythms. Here we demonstrated that deficiency of Lmna gene in mice caused an obvious shift of locomotor activities to the daytime. The abnormal activity profile was accompanied by a remarkable change in phase-angle between the central clock in the suprachiasmatic nucleus (SCN) and lung peripheral clocks, leaving the phase of the SCN clock unaffected by the mutation. These observations suggest that Lmna deficiency causes a change of the habitat from nocturnal to diurnal behaviors. On the other hand, molecular oscillation and its phase resetting mechanism were intact in both the Lmna-deficient cells and progeria-mimicking cells. Intriguingly, high-fat diet feeding extended the short lifespan and ameliorated the abnormalities of the behaviors and the phase of the peripheral clock in the Lmna-deficient mice. The present study supports the important contribution of the energy conditions to a shift between the diurnal and nocturnal activities.
Collapse
Affiliation(s)
- Satoshi Kawakami
- Department of Biological Sciences, School of Science, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan.,Circadian Clock Project, Tokyo Metropolitan Institute of Medical Science, Kamikitazawa 2-1-6, Setagaya-ku, Tokyo 156-8506, Japan
| | - Hikari Yoshitane
- Department of Biological Sciences, School of Science, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan.,Circadian Clock Project, Tokyo Metropolitan Institute of Medical Science, Kamikitazawa 2-1-6, Setagaya-ku, Tokyo 156-8506, Japan
| | - Taiki Morimura
- Department of Biological Sciences, School of Science, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan.,Circadian Clock Project, Tokyo Metropolitan Institute of Medical Science, Kamikitazawa 2-1-6, Setagaya-ku, Tokyo 156-8506, Japan
| | - Wataru Kimura
- RIKEN Center for Biosystems Dynamics Research, Minatojima-minamimachi 2-2-3, Chuo-ku, Kobe, Hyogo 650-0043, Japan
| | - Yoshitaka Fukada
- Department of Biological Sciences, School of Science, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan.,Circadian Clock Project, Tokyo Metropolitan Institute of Medical Science, Kamikitazawa 2-1-6, Setagaya-ku, Tokyo 156-8506, Japan.,Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
43
|
Choi J, Richards EJ. The edge of the nucleus: Variations on a theme. Dev Cell 2022; 57:3-4. [PMID: 35016004 DOI: 10.1016/j.devcel.2021.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The plant nuclear lamina utilizes distinct and highly divergent proteins to mediate chromatin interactions at the nuclear edge. In this issue of Developmental Cell, Tang et al. show that members of PNET2, a family of inner nuclear membrane proteins in Arabidopsis, are capable of binding histones and are involved in large-scale genome organization.
Collapse
|
44
|
Goodman LD, Cope H, Nil Z, Ravenscroft TA, Charng WL, Lu S, Tien AC, Pfundt R, Koolen DA, Haaxma CA, Veenstra-Knol HE, Wassink-Ruiter JSK, Wevers MR, Jones M, Walsh LE, Klee VH, Theunis M, Legius E, Steel D, Barwick KES, Kurian MA, Mohammad SS, Dale RC, Terhal PA, van Binsbergen E, Kirmse B, Robinette B, Cogné B, Isidor B, Grebe TA, Kulch P, Hainline BE, Sapp K, Morava E, Klee EW, Macke EL, Trapane P, Spencer C, Si Y, Begtrup A, Moulton MJ, Dutta D, Kanca O, Wangler MF, Yamamoto S, Bellen HJ, Tan QKG. TNPO2 variants associate with human developmental delays, neurologic deficits, and dysmorphic features and alter TNPO2 activity in Drosophila. Am J Hum Genet 2021; 108:1669-1691. [PMID: 34314705 PMCID: PMC8456166 DOI: 10.1016/j.ajhg.2021.06.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 06/27/2021] [Indexed: 12/11/2022] Open
Abstract
Transportin-2 (TNPO2) mediates multiple pathways including non-classical nucleocytoplasmic shuttling of >60 cargoes, such as developmental and neuronal proteins. We identified 15 individuals carrying de novo coding variants in TNPO2 who presented with global developmental delay (GDD), dysmorphic features, ophthalmologic abnormalities, and neurological features. To assess the nature of these variants, functional studies were performed in Drosophila. We found that fly dTnpo (orthologous to TNPO2) is expressed in a subset of neurons. dTnpo is critical for neuronal maintenance and function as downregulating dTnpo in mature neurons using RNAi disrupts neuronal activity and survival. Altering the activity and expression of dTnpo using mutant alleles or RNAi causes developmental defects, including eye and wing deformities and lethality. These effects are dosage dependent as more severe phenotypes are associated with stronger dTnpo loss. Interestingly, similar phenotypes are observed with dTnpo upregulation and ectopic expression of TNPO2, showing that loss and gain of Transportin activity causes developmental defects. Further, proband-associated variants can cause more or less severe developmental abnormalities compared to wild-type TNPO2 when ectopically expressed. The impact of the variants tested seems to correlate with their position within the protein. Specifically, those that fall within the RAN binding domain cause more severe toxicity and those in the acidic loop are less toxic. Variants within the cargo binding domain show tissue-dependent effects. In summary, dTnpo is an essential gene in flies during development and in neurons. Further, proband-associated de novo variants within TNPO2 disrupt the function of the encoded protein. Hence, TNPO2 variants are causative for neurodevelopmental abnormalities.
Collapse
Affiliation(s)
- Lindsey D Goodman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Heidi Cope
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
| | - Zelha Nil
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Thomas A Ravenscroft
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Wu-Lin Charng
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Shenzhao Lu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - An-Chi Tien
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Rolph Pfundt
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, PO Box 9101, Nijmegen, the Netherlands
| | - David A Koolen
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, PO Box 9101, Nijmegen, the Netherlands
| | - Charlotte A Haaxma
- Department of Pediatric Neurology, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, Geert Grooteplein Zuid 10, 6525 GA, PO Box 9101, the Netherlands
| | - Hermine E Veenstra-Knol
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, the Netherlands
| | - Jolien S Klein Wassink-Ruiter
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, the Netherlands
| | - Marijke R Wevers
- Department of Genetics, Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, the Netherlands
| | - Melissa Jones
- Houston Area Pediatric Neurology, 24514 Kingsland Blvd, Katy, TX 77494, USA
| | - Laurence E Walsh
- Department of Pediatric Neurology, Riley Hospital for Children, Indianapolis, IN 46202, USA
| | - Victoria H Klee
- Department of Pediatric Neurology, Riley Hospital for Children, Indianapolis, IN 46202, USA
| | - Miel Theunis
- Center for Human Genetics, University Hospital Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Eric Legius
- Department of Human Genetics, University of Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Dora Steel
- Molecular Neurosciences, Developmental Neurosciences, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK; Department of Neurology, Great Ormond Street Hospital, London WC1N 3JH, UK
| | - Katy E S Barwick
- Molecular Neurosciences, Developmental Neurosciences, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
| | - Manju A Kurian
- Molecular Neurosciences, Developmental Neurosciences, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK; Department of Neurology, Great Ormond Street Hospital, London WC1N 3JH, UK
| | - Shekeeb S Mohammad
- T.Y. Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, Westmead, NSW 2145, Australia; Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Sydney, Westmead, NSW 2145, Australia
| | - Russell C Dale
- T.Y. Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, Westmead, NSW 2145, Australia; Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Sydney, Westmead, NSW 2145, Australia
| | - Paulien A Terhal
- Department of Genetics, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Ellen van Binsbergen
- Department of Genetics, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Brian Kirmse
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Bethany Robinette
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Benjamin Cogné
- Centre hospitalier universitaire (CHU) de Nantes, Service de Génétique Médicale, 9 quai Moncousu, 44093 Nantes, France; INSERM, CNRS, UNIV Nantes, Centre hospitalier universitaire (CHU) de Nantes, l'institut du thorax, 44007 Nantes, France
| | - Bertrand Isidor
- Centre hospitalier universitaire (CHU) de Nantes, Service de Génétique Médicale, 9 quai Moncousu, 44093 Nantes, France; INSERM, CNRS, UNIV Nantes, Centre hospitalier universitaire (CHU) de Nantes, l'institut du thorax, 44007 Nantes, France
| | - Theresa A Grebe
- Phoenix Children's Hospital, Phoenix, AZ 85016, USA; Department of Child Health, University of Arizona College of Medicine Phoenix, Phoenix, AZ 85004, USA
| | - Peggy Kulch
- Phoenix Children's Hospital, Phoenix, AZ 85016, USA
| | - Bryan E Hainline
- Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Katherine Sapp
- Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Eva Morava
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA; Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA
| | - Eric W Klee
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA; Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA
| | - Erica L Macke
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Pamela Trapane
- University of Florida, College of Medicine, Jacksonville, Jacksonville, FL 32209, USA
| | - Christopher Spencer
- University of Florida, College of Medicine, Jacksonville, Jacksonville, FL 32209, USA
| | - Yue Si
- GeneDx, Gaithersburg, MD 20877, USA
| | | | - Matthew J Moulton
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Debdeep Dutta
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Oguz Kanca
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Michael F Wangler
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Shinya Yamamoto
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Queenie K-G Tan
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
45
|
Shrimp J, Jing Y, Gamage ST, Nelson KM, Han J, Bryson KM, Montgomery DC, Thomas JM, Nance KD, Sharma S, Fox SD, Andressen T, Sinclair WR, Wu H, Allali-Hassani A, Senisterra G, Vedadi M, Lafontaine D, Dahlin JL, Marmorstein R, Walters MA, Meier JL. Remodelin Is a Cryptic Assay Interference Chemotype That Does Not Inhibit NAT10-Dependent Cytidine Acetylation. ACS Med Chem Lett 2021; 12:887-892. [PMID: 34141066 PMCID: PMC8201477 DOI: 10.1021/acsmedchemlett.0c00193] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/21/2020] [Indexed: 12/21/2022] Open
Abstract
Remodelin is a putative small molecule inhibitor of the RNA acetyltransferase NAT10 which has shown preclinical efficacy in models of the premature aging disease Hutchinson-Gilford Progeria Syndrome (HGPS). Here we evaluate remodelin's assay interference characteristics and effects on NAT10-catalyzed RNA cytidine acetylation. We find the remodelin chemotype constitutes a cryptic assay interference compound, which does not react with small molecule thiols but demonstrates protein reactivity in ALARM NMR and proteome-wide affinity profiling assays. Biophysical analyses find no direct evidence for interaction of remodelin with the NAT10 acetyltransferase active site. Cellular studies verify that N4-acetylcytidine (ac4C) is a nonredundant target of NAT10 activity in human cell lines and find that this RNA modification is not affected by remodelin treatment in several orthogonal assays. These studies display the potential for remodelin's chemotype to interact with multiple protein targets in cells and indicate remodelin should not be applied as a specific chemical inhibitor of NAT10-catalyzed RNA acetylation.
Collapse
Affiliation(s)
- Jonathan
H. Shrimp
- Chemical
Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Yihang Jing
- Chemical
Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Supuni Thalalla Gamage
- Chemical
Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Kathryn M. Nelson
- University
of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Joseph Han
- Department
of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Keri M. Bryson
- Chemical
Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| | - David C. Montgomery
- Chemical
Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Justin M. Thomas
- Chemical
Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Kellie D. Nance
- Chemical
Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Sunny Sharma
- Department
of Cell Biology and Neuroscience, Rutgers
University, Piscataway, New Jersey 08854, United States
| | - Stephen D. Fox
- Protein
Characterization Laboratory, Cancer Research Technology Program, Frederick
National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland 21702, United States
| | - Thorkell Andressen
- Protein
Characterization Laboratory, Cancer Research Technology Program, Frederick
National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland 21702, United States
| | - Wilson R. Sinclair
- Chemical
Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Hong Wu
- Structural
Genomics Consortium, Toronto, Ontario M5G 1L7, Canada
| | | | | | - Masoud Vedadi
- Structural
Genomics Consortium, Toronto, Ontario M5G 1L7, Canada
| | - Denis Lafontaine
- RNA
Molecular Biology, Université Libre
de Bruxelles, Fonds de la Recherche Scientifique
(F.R.S./FNRS), 6041 Gosselies, Belgium
| | - Jayme L. Dahlin
- Department
of Pathology, Brigham and Women’s
Hospital and Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Ronen Marmorstein
- Department
of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department
of Biochemistry and Biophysics and Abramson Family Cancer Research
Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | | | - Jordan L. Meier
- Chemical
Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| |
Collapse
|
46
|
Hintze M, Katsanos D, Shahrezaei V, Barkoulas M. Phenotypic Robustness of Epidermal Stem Cell Number in C. elegans Is Modulated by the Activity of the Conserved N-acetyltransferase nath-10/NAT10. Front Cell Dev Biol 2021; 9:640856. [PMID: 34084768 PMCID: PMC8168469 DOI: 10.3389/fcell.2021.640856] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 04/19/2021] [Indexed: 12/14/2022] Open
Abstract
Individual cells and organisms experience perturbations from internal and external sources, yet manage to buffer these to produce consistent phenotypes, a property known as robustness. While phenotypic robustness has often been examined in unicellular organisms, it has not been sufficiently studied in multicellular animals. Here, we investigate phenotypic robustness in Caenorhabditis elegans seam cells. Seam cells are stem cell-like epithelial cells along the lateral edges of the animal, which go through asymmetric and symmetric divisions contributing cells to the hypodermis and neurons, while replenishing the stem cell reservoir. The terminal number of seam cells is almost invariant in the wild-type population, allowing the investigation of how developmental precision is achieved. We report here that a loss-of-function mutation in the highly conserved N-acetyltransferase nath-10/NAT10 increases seam cell number variance in the isogenic population. RNA-seq analysis revealed increased levels of mRNA transcript variability in nath-10 mutant populations, which may have an impact on the phenotypic variability observed. Furthermore, we found disruption of Wnt signaling upon perturbing nath-10 function, as evidenced by changes in POP-1/TCF nuclear distribution and ectopic activation of its GATA transcription factor target egl-18. These results highlight that NATH-10/NAT-10 can influence phenotypic variability partly through modulation of the Wnt signaling pathway.
Collapse
Affiliation(s)
- Mark Hintze
- Department of Life Sciences, Imperial College, London, United Kingdom
| | - Dimitris Katsanos
- Department of Life Sciences, Imperial College, London, United Kingdom
| | - Vahid Shahrezaei
- Department of Mathematics, Imperial College, London, United Kingdom
| | | |
Collapse
|
47
|
Dalhat MH, Altayb HN, Khan MI, Choudhry H. Structural insights of human N-acetyltransferase 10 and identification of its potential novel inhibitors. Sci Rep 2021; 11:6051. [PMID: 33723305 PMCID: PMC7960695 DOI: 10.1038/s41598-021-84908-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 02/19/2021] [Indexed: 01/31/2023] Open
Abstract
N-acetyltransferase 10 (NAT10), is an acetyltransferase that regulates RNA stability and translation processes. Association of NAT10 with several diseases including cancer, makes it a promising therapeutic target. Remodelin is the only known NAT10 inhibitor, but the structural information related to its binding with NAT10 is still obscure. Here, we predicted the human NAT10 structure using homology modeling that was not available previously and used human NAT10 to identify the novel binding site(s) of Remodelin. The alignment of the modeled human NAT10 showed 24% identity and 37% positivity with crystal structure of tRNA (Met) cytidine acetyltransferase. Molecular docking showed binding of Remodelin with NAT10 in acetyl-CoA binding pocket. Additionally, we screened a library of FDA-approved drugs for the identification of novel inhibitors of NAT10 activity. Binding score showed that four drugs namely, Fosaprepitant (- 11.709), Leucal (- 10.46), Fludarabine (- 10.347) and Dantrolene (- 9.875) bind to NAT10 and have better binding capability when compared with Acetyl-CoA (- 5.691) and Remodelin (- 5.3). Acetyl-CoA, Remodelin, and others exhibit hits for hydrophobic, hydrophilic and hydrogen interactions. Interestingly, Remodelin and others interact with the amino acid residues ILE629, GLY639, GLY641, LEU719, and PHE722 in the Acetyl-CoA binding pocket of NAT10 similar to Acetyl-CoA. Our findings revealed that Fosaprepitant, Leucal, Fludarabine, and Dantrolene are promising molecules that can be tested and developed as potential inhibitors of NAT10 acetyltransferase activity.
Collapse
Affiliation(s)
- Mahmood Hassan Dalhat
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
- Centre for Artificial Intelligence in Precision Medicine, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
- Cancer and Mutagenesis Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Hisham N Altayb
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
- Centre for Artificial Intelligence in Precision Medicine, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
- Cancer and Mutagenesis Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Mohammad Imran Khan
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
- Centre for Artificial Intelligence in Precision Medicine, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
- Cancer and Mutagenesis Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Hani Choudhry
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia.
- Centre for Artificial Intelligence in Precision Medicine, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia.
- Cancer and Mutagenesis Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia.
| |
Collapse
|
48
|
Röhrl JM, Arnold R, Djabali K. Nuclear Pore Complexes Cluster in Dysmorphic Nuclei of Normal and Progeria Cells during Replicative Senescence. Cells 2021; 10:cells10010153. [PMID: 33466669 PMCID: PMC7828780 DOI: 10.3390/cells10010153] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/10/2021] [Accepted: 01/11/2021] [Indexed: 01/10/2023] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare premature aging disease caused by a mutation in LMNA. A G608G mutation in exon 11 of LMNA is responsible for most HGPS cases, generating a truncated protein called “progerin”. Progerin is permanently farnesylated and accumulates in HGPS cells, causing multiple cellular defects such as nuclear dysmorphism, a thickened lamina, loss of heterochromatin, premature senescence, and clustering of Nuclear Pore Complexes (NPC). To identify the mechanism of NPC clustering in HGPS cells, we evaluated post-mitotic NPC assembly in control and HGPS cells and found no defects. Next, we examined the occurrence of NPC clustering in control and HGPS cells during replicative senescence. We reported that NPC clustering occurs solely in the dysmorphic nuclei of control and HGPS cells. Hence, NPC clustering occurred at a higher frequency in HGPS cells compared to control cells at early passages; however, in late cultures with similar senescence index, NPCs clustering occurred at a similar rate in both control and HGPS. Our results show that progerin does not disrupt post-mitotic reassembly of NPCs. However, NPCs frequently cluster in dysmorphic nuclei with a high progerin content. Additionally, nuclear envelope defects that arise during replicative senescence cause NPC clustering in senescent cells with dysmorphic nuclei.
Collapse
|
49
|
Squarzoni S, Schena E, Sabatelli P, Mattioli E, Capanni C, Cenni V, D'Apice MR, Andrenacci D, Sarli G, Pellegrino V, Festa A, Baruffaldi F, Storci G, Bonafè M, Barboni C, Sanapo M, Zaghini A, Lattanzi G. Interleukin-6 neutralization ameliorates symptoms in prematurely aged mice. Aging Cell 2021; 20:e13285. [PMID: 33393189 PMCID: PMC7811841 DOI: 10.1111/acel.13285] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/23/2020] [Accepted: 11/14/2020] [Indexed: 12/17/2022] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) causes premature aging in children, with adipose tissue, skin and bone deterioration, and cardiovascular impairment. In HGPS cells and mouse models, high levels of interleukin-6, an inflammatory cytokine linked to aging processes, have been detected. Here, we show that inhibition of interleukin-6 activity by tocilizumab, a neutralizing antibody raised against interleukin-6 receptors, counteracts progeroid features in both HGPS fibroblasts and LmnaG609G / G609G progeroid mice. Tocilizumab treatment limits the accumulation of progerin, the toxic protein produced in HGPS cells, rescues nuclear envelope and chromatin abnormalities, and attenuates the hyperactivated DNA damage response. In vivo administration of tocilizumab reduces aortic lesions and adipose tissue dystrophy, delays the onset of lipodystrophy and kyphosis, avoids motor impairment, and preserves a good quality of life in progeroid mice. This work identifies tocilizumab as a valuable tool in HGPS therapy and, speculatively, in the treatment of a variety of aging-related disorders.
Collapse
Affiliation(s)
- Stefano Squarzoni
- CNR Institute of Molecular Genetics “Luigi Luca Cavalli‐Sforza” Unit of Bologna Bologna Italy
- IRCCS Istituto Ortopedico Rizzoli Bologna Italy
| | - Elisa Schena
- CNR Institute of Molecular Genetics “Luigi Luca Cavalli‐Sforza” Unit of Bologna Bologna Italy
- IRCCS Istituto Ortopedico Rizzoli Bologna Italy
| | - Patrizia Sabatelli
- CNR Institute of Molecular Genetics “Luigi Luca Cavalli‐Sforza” Unit of Bologna Bologna Italy
- IRCCS Istituto Ortopedico Rizzoli Bologna Italy
| | - Elisabetta Mattioli
- CNR Institute of Molecular Genetics “Luigi Luca Cavalli‐Sforza” Unit of Bologna Bologna Italy
- IRCCS Istituto Ortopedico Rizzoli Bologna Italy
| | - Cristina Capanni
- CNR Institute of Molecular Genetics “Luigi Luca Cavalli‐Sforza” Unit of Bologna Bologna Italy
- IRCCS Istituto Ortopedico Rizzoli Bologna Italy
| | - Vittoria Cenni
- CNR Institute of Molecular Genetics “Luigi Luca Cavalli‐Sforza” Unit of Bologna Bologna Italy
- IRCCS Istituto Ortopedico Rizzoli Bologna Italy
| | | | - Davide Andrenacci
- CNR Institute of Molecular Genetics “Luigi Luca Cavalli‐Sforza” Unit of Bologna Bologna Italy
- IRCCS Istituto Ortopedico Rizzoli Bologna Italy
| | - Giuseppe Sarli
- Department of Veterinary Medical Sciences University of Bologna Bologna Italy
| | - Valeria Pellegrino
- Department of Veterinary Medical Sciences University of Bologna Bologna Italy
| | - Anna Festa
- Laboratory of Medical Technology IRCCS Istituto Ortopedico Rizzoli Bologna Italy
| | - Fabio Baruffaldi
- Laboratory of Medical Technology IRCCS Istituto Ortopedico Rizzoli Bologna Italy
| | - Gianluca Storci
- Department of Experimental, Diagnostic and Specialty Medicine University of Bologna Bologna Italy
| | - Massimiliano Bonafè
- Department of Experimental, Diagnostic and Specialty Medicine University of Bologna Bologna Italy
| | - Catia Barboni
- Department of Veterinary Medical Sciences University of Bologna Bologna Italy
| | - Mara Sanapo
- Department of Veterinary Medical Sciences University of Bologna Bologna Italy
| | - Anna Zaghini
- Department of Veterinary Medical Sciences University of Bologna Bologna Italy
| | - Giovanna Lattanzi
- CNR Institute of Molecular Genetics “Luigi Luca Cavalli‐Sforza” Unit of Bologna Bologna Italy
- IRCCS Istituto Ortopedico Rizzoli Bologna Italy
| |
Collapse
|
50
|
Lee J, Park J, Kim JH, Lee G, Park TE, Yoon KJ, Kim YK, Lim C. LSM12-EPAC1 defines a neuroprotective pathway that sustains the nucleocytoplasmic RAN gradient. PLoS Biol 2020; 18:e3001002. [PMID: 33362237 PMCID: PMC7757817 DOI: 10.1371/journal.pbio.3001002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 11/19/2020] [Indexed: 02/07/2023] Open
Abstract
Nucleocytoplasmic transport (NCT) defects have been implicated in neurodegenerative diseases such as C9ORF72-associated amyotrophic lateral sclerosis and frontotemporal dementia (C9-ALS/FTD). Here, we identify a neuroprotective pathway of like-Sm protein 12 (LSM12) and exchange protein directly activated by cyclic AMP 1 (EPAC1) that sustains the nucleocytoplasmic RAN gradient and thereby suppresses NCT dysfunction by the C9ORF72-derived poly(glycine-arginine) protein. LSM12 depletion in human neuroblastoma cells aggravated poly(GR)-induced impairment of NCT and nuclear integrity while promoting the nuclear accumulation of poly(GR) granules. In fact, LSM12 posttranscriptionally up-regulated EPAC1 expression, whereas EPAC1 overexpression rescued the RAN gradient and NCT defects in LSM12-deleted cells. C9-ALS patient-derived neurons differentiated from induced pluripotent stem cells (C9-ALS iPSNs) displayed low expression of LSM12 and EPAC1. Lentiviral overexpression of LSM12 or EPAC1 indeed restored the RAN gradient, mitigated the pathogenic mislocalization of TDP-43, and suppressed caspase-3 activation for apoptosis in C9-ALS iPSNs. EPAC1 depletion biochemically dissociated RAN-importin β1 from the cytoplasmic nuclear pore complex, thereby dissipating the nucleocytoplasmic RAN gradient essential for NCT. These findings define the LSM12-EPAC1 pathway as an important suppressor of the NCT-related pathologies in C9-ALS/FTD. A post-transcriptional circuit comprising LSM12 and EPAC1 suppresses neurodegenerative pathologies in C9ORF72-associated amyotrophic lateral sclerosis by establishing the RAN gradient and sustaining nucleocytoplasmic transport.
Collapse
Affiliation(s)
- Jongbo Lee
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Jumin Park
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Ji-hyung Kim
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Giwook Lee
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Tae-Eun Park
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Ki-Jun Yoon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Yoon Ki Kim
- Creative Research Initiatives Center for Molecular Biology of Translation, Korea University, Seoul, Republic of Korea
- Division of Life Sciences, Korea University, Seoul, Republic of Korea
| | - Chunghun Lim
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
- * E-mail:
| |
Collapse
|