1
|
Hammami NEH, Mérindol N, Plourde MB, Maisonnet T, Lebel S, Berthoux L. SUMO-3 promotes the ubiquitin-dependent turnover of TRIM55. Biochem Cell Biol 2024; 102:73-84. [PMID: 37703582 DOI: 10.1139/bcb-2023-0153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023] Open
Abstract
Human muscle-specific RING fingers (MURFs) are members of the tripartite motif (TRIM) family of proteins characterized by their C-terminal subgroup one signature domain. MURFs play a role in sarcomere formation and microtubule dynamics. It was previously established that some TRIMs undergo post-translational modification by small ubiquitin-like modifier (SUMO). In this study, we explored the putative SUMOylation of MURF proteins as well as their interactions with SUMO. MURF proteins (TRIM54, TRIM55, and TRIM63) were not found to be SUMOylated. However, TRIM55 turnover by proteasomal and lysosomal degradation was higher upon overexpression of SUMO-3 but not of SUMO-1. Furthermore, it is predicted that TRIM55 contains two potential SUMO-interacting motifs (SIMs). We found that SIM1- and SIM2-mutated TRIM55 were more stable than the wild-type (WT) protein partly due to decreased degradation. Consistently, SIM-mutated TRIM55 was less polyubiquitinated than the WT protein, despite similar monoubiquitination levels. Using IF microscopy, we observed that SIM motifs influenced TRIM55 subcellular localization. In conclusion, our results suggest that SUMO-3 or SUMO-3-modified proteins modulate the localization, stability, and RING ubiquitin ligase activity of TRIM55.
Collapse
Affiliation(s)
- Nour-El-Houda Hammami
- Department of medical biology, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada
| | - Natacha Mérindol
- Department of medical biology, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada
| | - Mélodie B Plourde
- Department of medical biology, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada
| | - Tara Maisonnet
- Department of medical biology, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada
| | - Sophie Lebel
- Department of medical biology, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada
| | - Lionel Berthoux
- Department of medical biology, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada
| |
Collapse
|
2
|
Zambrano-Carrasco J, Zou J, Wang W, Sun X, Li J, Su H. Emerging Roles of Cullin-RING Ubiquitin Ligases in Cardiac Development. Cells 2024; 13:235. [PMID: 38334627 PMCID: PMC10854628 DOI: 10.3390/cells13030235] [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: 12/23/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/10/2024] Open
Abstract
Heart development is a spatiotemporally regulated process that extends from the embryonic phase to postnatal stages. Disruption of this highly orchestrated process can lead to congenital heart disease or predispose the heart to cardiomyopathy or heart failure. Consequently, gaining an in-depth understanding of the molecular mechanisms governing cardiac development holds considerable promise for the development of innovative therapies for various cardiac ailments. While significant progress in uncovering novel transcriptional and epigenetic regulators of heart development has been made, the exploration of post-translational mechanisms that influence this process has lagged. Culling-RING E3 ubiquitin ligases (CRLs), the largest family of ubiquitin ligases, control the ubiquitination and degradation of ~20% of intracellular proteins. Emerging evidence has uncovered the critical roles of CRLs in the regulation of a wide range of cellular, physiological, and pathological processes. In this review, we summarize current findings on the versatile regulation of cardiac morphogenesis and maturation by CRLs and present future perspectives to advance our comprehensive understanding of how CRLs govern cardiac developmental processes.
Collapse
Affiliation(s)
- Josue Zambrano-Carrasco
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (J.Z.-C.); (J.Z.)
| | - Jianqiu Zou
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (J.Z.-C.); (J.Z.)
| | - Wenjuan Wang
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (J.Z.-C.); (J.Z.)
| | - Xinghui Sun
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, USA;
| | - Jie Li
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (J.Z.-C.); (J.Z.)
| | - Huabo Su
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (J.Z.-C.); (J.Z.)
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
3
|
Chen J, Feng X, Zhou X, Li Y. Role of the tripartite motif-containing (TRIM) family of proteins in insulin resistance and related disorders. Diabetes Obes Metab 2024; 26:3-15. [PMID: 37726973 DOI: 10.1111/dom.15294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/27/2023] [Accepted: 09/05/2023] [Indexed: 09/21/2023]
Abstract
Emerging evidence suggests that the ubiquitin-mediated degradation of insulin-signalling-related proteins may be involved in the development of insulin resistance and its related disorders. Tripartite motif-containing (TRIM) proteins, a superfamily belonging to the E3 ubiquitin ligases, are capable of controlling protein levels and function by ubiquitination, which is essential for the modulation of insulin sensitivity. Recent research has indicated that some of these TRIMs act as key regulatory factors of metabolic disorders such as type 2 diabetes mellitus, obesity, nonalcoholic fatty liver disease, and atherosclerosis. This review provides a comprehensive overview of the latest evidence linking TRIMs to the regulation of insulin resistance and its related disorders, their roles in regulating multiple signalling pathways or cellular processes, such as insulin signalling pathways, peroxisome proliferator-activated receptor signalling pathways, glucose and lipid metabolism, the inflammatory response, and cell cycle control, as well as recent advances in the development of TRIM-targeted drugs.
Collapse
Affiliation(s)
- Jianrong Chen
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Clinical Research Centre for Endocrine and Metabolic disease, Nanchang, China
- Jiangxi Branch of National Clinical Research Centre for Metabolic disease, Nanchang, China
| | - Xianjie Feng
- Evidence-based Medicine Research Centre, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Xu Zhou
- Evidence-based Medicine Research Centre, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Yong Li
- Department of Anaesthesiology, Medical Centre of Anaesthesiology and Pain, First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
4
|
Liao Y, Ke B, Long X, Xu J, Wu Y. Abnormalities in the SIRT1-SIRT3 axis promote myocardial ischemia-reperfusion injury through ferroptosis caused by silencing the PINK1/Parkin signaling pathway. BMC Cardiovasc Disord 2023; 23:582. [PMID: 38012584 PMCID: PMC10683361 DOI: 10.1186/s12872-023-03603-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 11/07/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND Myocardial ischemia-reperfusion injury (MIRI) is one of the main reasons for poor prognosis in patients with ischemic cardiomyopathy (ICM). To date, the mechanism remains unknown. As members of the silent information regulator 2 (SIR2) family, both SIRT1 and SIRT3 have been shown to play critical roles in protecting cardiomyocytes against MIRI, but their specific protective mechanism, their interact between the two and their relationship with ferroptosis are still unclear. Hence, in this study, we investigated the interact and specific mechanism of SIRT1 and SIRT3 in protecting cardiomyocytes against MIRI, as well as their association with ferroptosis. METHODS Bioinformatics analysis methods were used to explore the expression of SIRT1 and SIRT3 during MIRI, and then a cell hypoxia/reoxygenation injury model was constructed to verify the results. Then, Pearson correlation analysis was further used to explore the relationship between SIRT1 and SIRT3, whose roles in the regulation of ferroptosis were also analysed by gene knock down, Western Blotting and flow cytometry. Several biomarkers, such as Fe2+ concentration, lipid peroxidation marker MDA and mitochondrial membrane potential (MMP), were used to evaluate changes in ferroptosis. RESULTS The expression of SIRT1 and SIRT3 was abnormal during MIRI, and SIRT1 was significantly negatively correlated with SIRT3 in the SIRT1-SIRT3 axis. Further analysis revealed that the SIRT1-SIRT3 axis was closely correlated with ferroptosis, and its silencing effectively increase the incidence of ferroptosis. Furthermore, SIRT1-SIRT3 axis silencing was accompanied by changes in PINK1, Parkin, P62/SQSTM1 and LC3 expression. PINK1 silencing significantly increased the incidence of ferroptosis, while resveratrol (Res) and/or honokiol (HKL) effectively reversed the outcome. CONCLUSION Abnormalities in the SIRT1-SIRT3 axis promote MIRI through ferroptosis caused by silencing the PINK1/Parkin signaling pathway.
Collapse
Affiliation(s)
- Yunfei Liao
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- East China Digital Medical Engineering Research Institute, Shangrao, China
| | - Ben Ke
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaoyan Long
- East China Digital Medical Engineering Research Institute, Shangrao, China
| | - Jianjun Xu
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China.
| | - Yongbing Wu
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
5
|
Lu X, Yuan Y, Cai N, Rao D, Chen M, Chen X, Zhang B, Liang H, Zhang L. TRIM55 Promotes Proliferation of Hepatocellular Carcinoma Through Stabilizing TRIP6 to Activate Wnt/β-Catenin Signaling. J Hepatocell Carcinoma 2023; 10:1281-1293. [PMID: 37554583 PMCID: PMC10406114 DOI: 10.2147/jhc.s418049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/19/2023] [Indexed: 08/10/2023] Open
Abstract
PURPOSE Tripartite motif containing 55 (TRIM55) is a member of the TRIM family and functions as an E3 ubiquitin ligase. It acts as a cancer promoter or suppressor in the malignant processes of multiple cancers. However, its proliferative function in hepatocellular carcinoma (HCC) has been poorly studied, and its underlying molecular mechanism remains unclear. In the present study, we investigated the role of TRIM55 in HCC and its mechanism of promoting HCC proliferation. MATERIALS AND METHODS Protein expression levels of TRIM55 were measured in paired HCC and normal tissue samples using immunohistochemical (IHC) staining. The correlation between TRIM55 and clinical features was evaluated by statistical analysis. At the same time, overexpression and knockdown experiments, cycloheximide (CHX) interference experiments, ubiquitination, co-immunoprecipitation and immunofluorescence staining experiments, as well as animal experiments were used to evaluate the potential mechanism that TRIM55 promotes proliferation of hepatocellular carcinoma in vitro and in vivo. RESULTS TRIM55 expression in HCC specimens was higher compared with the corresponding non-tumor tissues. The overall survival and disease-free survival time of patients with high TRIM55 expression were shorter than those with low expression of TRIM55. Functionally, TRIM55 promoted the proliferation of HCC cells and accelerated the growth of HCC xenografts. Mechanistically, TRIM55 interacted with thyroid receptor interacting protein 6 (TRIP6) and regulate its stability by influencing the ubiquitination process, thereby affecting the Wnt signaling pathway. CONCLUSION Our results indicate that TRIM55 promotes HCC proliferation by activating Wnt signaling pathways by stabilizing TRIP6. Therefore, targeting TRIM55 may be an effective therapeutic strategy to inhibit HCC growth.
Collapse
Affiliation(s)
- Xun Lu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Clinical Medical Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yue Yuan
- Division of Gastroenterology, Department of Internal Medicine at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Ning Cai
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Clinical Medical Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Dean Rao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Clinical Medical Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Min Chen
- Division of Gastroenterology, Department of Internal Medicine at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Clinical Medical Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Clinical Medical Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Huifang Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Clinical Medical Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Lei Zhang
- Clinical Medical Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Key Laboratory of Hepatobiliary and Pancreatic Diseases of Shanxi Province (Preparatory), Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Shanxi Medical University; Shanxi Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Taiyuan, People’s Republic of China
| |
Collapse
|
6
|
McNamara JW, Parker BL, Voges HK, Mehdiabadi NR, Bolk F, Ahmad F, Chung JD, Charitakis N, Molendijk J, Zech ATL, Lal S, Ramialison M, Karavendzas K, Pointer HL, Syrris P, Lopes LR, Elliott PM, Lynch GS, Mills RJ, Hudson JE, Watt KI, Porrello ER, Elliott DA. Alpha kinase 3 signaling at the M-band maintains sarcomere integrity and proteostasis in striated muscle. NATURE CARDIOVASCULAR RESEARCH 2023; 2:159-173. [PMID: 39196058 PMCID: PMC11358020 DOI: 10.1038/s44161-023-00219-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 01/19/2023] [Indexed: 08/29/2024]
Abstract
Muscle contraction is driven by the molecular machinery of the sarcomere. As phosphorylation is a critical regulator of muscle function, the identification of regulatory kinases is important for understanding sarcomere biology. Pathogenic variants in alpha kinase 3 (ALPK3) cause cardiomyopathy and musculoskeletal disease, but little is known about this atypical kinase. Here we show that ALPK3 is an essential component of the M-band of the sarcomere and define the ALPK3-dependent phosphoproteome. ALPK3 deficiency impaired contractility both in human cardiac organoids and in the hearts of mice harboring a pathogenic truncating Alpk3 variant. ALPK3-dependent phosphopeptides were enriched for sarcomeric components of the M-band and the ubiquitin-binding protein sequestosome-1 (SQSTM1) (also known as p62). Analysis of the ALPK3 interactome confirmed binding to M-band proteins including SQSTM1. In human pluripotent stem cell-derived cardiomyocytes modeling cardiomyopathic ALPK3 mutations, sarcomeric organization and M-band localization of SQSTM1 were abnormal suggesting that this mechanism may underly disease pathogenesis.
Collapse
Affiliation(s)
- James W McNamara
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine, Royal Children's Hospital, Melbourne, Victoria, Australia
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
- Centre for Muscle Research, University of Melbourne, Melbourne, Victoria, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Benjamin L Parker
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
- Centre for Muscle Research, University of Melbourne, Melbourne, Victoria, Australia
| | - Holly K Voges
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine, Royal Children's Hospital, Melbourne, Victoria, Australia
- School of Biomedical Sciences and Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Neda R Mehdiabadi
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia
| | - Francesca Bolk
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Feroz Ahmad
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine, Royal Children's Hospital, Melbourne, Victoria, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Jin D Chung
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
- Centre for Muscle Research, University of Melbourne, Melbourne, Victoria, Australia
| | - Natalie Charitakis
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- School of Biomedical Sciences and Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Jeffrey Molendijk
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Antonia T L Zech
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine, Royal Children's Hospital, Melbourne, Victoria, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Sean Lal
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, New South Wales, Australia
| | - Mirana Ramialison
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- School of Biomedical Sciences and Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia
| | - Kathy Karavendzas
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Hayley L Pointer
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Petros Syrris
- Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, London, UK
- Barts Heart Centre, St. Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Luis R Lopes
- Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, London, UK
- Barts Heart Centre, St. Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Perry M Elliott
- Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, London, UK
- Barts Heart Centre, St. Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Gordon S Lynch
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
- Centre for Muscle Research, University of Melbourne, Melbourne, Victoria, Australia
| | - Richard J Mills
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Queensland Institute of Medical Research Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - James E Hudson
- Queensland Institute of Medical Research Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Kevin I Watt
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Enzo R Porrello
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia.
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine, Royal Children's Hospital, Melbourne, Victoria, Australia.
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia.
- Centre for Muscle Research, University of Melbourne, Melbourne, Victoria, Australia.
- Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, Victoria, Australia.
| | - David A Elliott
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia.
- Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine, Royal Children's Hospital, Melbourne, Victoria, Australia.
- Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, Victoria, Australia.
- School of Biomedical Sciences and Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia.
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
7
|
Yang X, Zhang Y, Xue Z, Hu Y, Zhou W, Xue Z, Liu X, Liu G, Li W, Liu X, Li X, Han M, Wang J. TRIM56 promotes malignant progression of glioblastoma by stabilizing cIAP1 protein. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:336. [PMID: 36471347 PMCID: PMC9724401 DOI: 10.1186/s13046-022-02534-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 11/07/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND The tripartite motif (TRIM) family of proteins plays a key role in the developmental growth and therapeutic resistance of many tumors. However, the regulatory mechanisms and biological functions of TRIM proteins in human glioblastoma (GBM) are not yet fully understood. In this study, we focused on TRIM56, which emerged as the most differentially expressed TRIM family member with increased expression in GBM. METHODS Western blot, real-time quantitative PCR (qRT-PCR), immunofluorescence (IF) and immunohistochemistry (IHC) were used to study the expression levels of TRIM56 and cIAP1 in GBM cell lines. Co-immunoprecipitation (co-IP) was used to explore the specific binding between target proteins and TRIM56. A xenograft animal model was used to verify the tumor promoting effect of TRIM56 on glioma in vivo. RESULTS We observed elevated expression of TRIM56 in malignant gliomas and revealed that TRIM56 promoted glioma progression in vitro and in a GBM xenograft model in nude mice. Analysis of the Human Ubiquitin Array and co-IPs showed that cIAP1 is a protein downstream of TRIM56. TRIM56 deubiquitinated cIAP1, mainly through the zinc finger domain (amino acids 21-205) of TRIM56, thereby reducing the degradation of cIAP1 and thus increasing its expression. TRIM56 also showed prognostic significance in overall survival of glioma patients. CONCLUSIONS TRIM56-regulated post-translational modifications may contribute to glioma development through stabilization of cIAP1. Furthermore, TRIM56 may serve as a novel prognostic indicator and therapeutic molecular target for GBM.
Collapse
Affiliation(s)
- Xu Yang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Xi Road, 250012 Jinan, Shandong People’s Republic of China ,Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250012 China ,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012 China
| | - Yan Zhang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Xi Road, 250012 Jinan, Shandong People’s Republic of China ,Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250012 China ,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012 China
| | - Zhiwei Xue
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Xi Road, 250012 Jinan, Shandong People’s Republic of China ,Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250012 China ,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012 China
| | - Yaotian Hu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Xi Road, 250012 Jinan, Shandong People’s Republic of China ,Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250012 China ,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012 China
| | - Wenjing Zhou
- grid.460018.b0000 0004 1769 9639Department of Blood Transfusion, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250022 China
| | - Zhiyi Xue
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Xi Road, 250012 Jinan, Shandong People’s Republic of China ,Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250012 China ,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012 China
| | - Xuemeng Liu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Xi Road, 250012 Jinan, Shandong People’s Republic of China ,Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250012 China ,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012 China
| | - Guowei Liu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Xi Road, 250012 Jinan, Shandong People’s Republic of China ,Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250012 China ,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012 China
| | - Wenjie Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Xi Road, 250012 Jinan, Shandong People’s Republic of China ,Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250012 China ,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012 China
| | - Xiaofei Liu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Xi Road, 250012 Jinan, Shandong People’s Republic of China ,Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250012 China ,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012 China
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Xi Road, 250012 Jinan, Shandong People’s Republic of China ,Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250012 China ,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012 China
| | - Mingzhi Han
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Xi Road, 250012 Jinan, Shandong People’s Republic of China ,Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250012 China ,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012 China ,grid.27255.370000 0004 1761 1174Medical Integration and Practice Center, Cheeloo College of Medicine, Shandong University, Jinan, 250012 China
| | - Jian Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Xi Road, 250012 Jinan, Shandong People’s Republic of China ,Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250012 China ,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012 China ,grid.7914.b0000 0004 1936 7443Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, 5009 Bergen, Norway
| |
Collapse
|
8
|
Agarwal R, Wakimoto H, Paulo JA, Zhang Q, Reichart D, Toepfer C, Sharma A, Tai AC, Lun M, Gorham J, DePalma SR, Gygi SP, Seidman J, Seidman CE. Pathogenesis of Cardiomyopathy Caused by Variants in ALPK3, an Essential Pseudokinase in the Cardiomyocyte Nucleus and Sarcomere. Circulation 2022; 146:1674-1693. [PMID: 36321451 PMCID: PMC9698156 DOI: 10.1161/circulationaha.122.059688] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND ALPK3 encodes α-kinase 3, a muscle-specific protein of unknown function. ALPK3 loss-of-function variants cause cardiomyopathy with distinctive clinical manifestations in both children and adults, but the molecular functions of ALPK3 remain poorly understood. METHODS We explored the putative kinase activity of ALPK3 and the consequences of damaging variants using isogenic human induced pluripotent stem cell-derived cardiomyocytes, mice, and human patient tissues. RESULTS Multiple sequence alignment of all human α-kinase domains and their orthologs revealed 4 conserved residues that were variant only in ALPK3, demonstrating evolutionary divergence of the ALPK3 α-kinase domain sequence. Phosphoproteomic evaluation of both ALPK3 kinase domain inhibition and overexpression failed to detect significant changes in catalytic activity, establishing ALPK3 as a pseudokinase. Investigations into alternative functions revealed that ALPK3 colocalized with myomesin proteins (MYOM1, MYOM2) at both the nuclear envelope and the sarcomere M-band. ALPK3 loss-of-function variants caused myomesin proteins to mislocalize and also dysregulated several additional M-band proteins involved in sarcomere protein turnover, which ultimately impaired cardiomyocyte structure and function. CONCLUSIONS ALPK3 is an essential cardiac pseudokinase that inserts in the nuclear envelope and the sarcomere M-band. Loss of ALPK3 causes mislocalization of myomesins, critical force-buffering proteins in cardiomyocytes, and also dysregulates M-band proteins necessary for sarcomere protein turnover. We conclude that ALPK3 cardiomyopathy induces ventricular dilatation caused by insufficient myomesin-mediated force buffering and hypertrophy by impairment of sarcomere proteostasis.
Collapse
Affiliation(s)
- Radhika Agarwal
- Department of Genetics (R.A., H.W., Q.Z., D.R., C.T., A.S., A.C.T., M.L., J.G., S.R.D., J.G.S., C.E.S.), Harvard Medical School, Boston, MA
| | - Hiroko Wakimoto
- Department of Genetics (R.A., H.W., Q.Z., D.R., C.T., A.S., A.C.T., M.L., J.G., S.R.D., J.G.S., C.E.S.), Harvard Medical School, Boston, MA
| | - Joao A. Paulo
- Department of Cell Biology (J.A.P., S.P.G.), Harvard Medical School, Boston, MA
| | - Qi Zhang
- Department of Genetics (R.A., H.W., Q.Z., D.R., C.T., A.S., A.C.T., M.L., J.G., S.R.D., J.G.S., C.E.S.), Harvard Medical School, Boston, MA
| | - Daniel Reichart
- Department of Genetics (R.A., H.W., Q.Z., D.R., C.T., A.S., A.C.T., M.L., J.G., S.R.D., J.G.S., C.E.S.), Harvard Medical School, Boston, MA
| | - Christopher Toepfer
- Department of Genetics (R.A., H.W., Q.Z., D.R., C.T., A.S., A.C.T., M.L., J.G., S.R.D., J.G.S., C.E.S.), Harvard Medical School, Boston, MA.,Radcliffe Department of Medicine (C.T.), University of Oxford, United Kingdom.,Wellcome Centre for Human Genetics (C.T.), University of Oxford, United Kingdom
| | - Arun Sharma
- Department of Genetics (R.A., H.W., Q.Z., D.R., C.T., A.S., A.C.T., M.L., J.G., S.R.D., J.G.S., C.E.S.), Harvard Medical School, Boston, MA.,Board of Governors Regenerative Medicine Institute (A.S.), Cedars-Sinai Medical Center, Los Angeles, CA.,Smidt Heart Institute (A.S.), Cedars-Sinai Medical Center, Los Angeles, CA.,Department of Biomedical Sciences (A.S.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Angela C. Tai
- Department of Genetics (R.A., H.W., Q.Z., D.R., C.T., A.S., A.C.T., M.L., J.G., S.R.D., J.G.S., C.E.S.), Harvard Medical School, Boston, MA
| | - Mingyue Lun
- Department of Genetics (R.A., H.W., Q.Z., D.R., C.T., A.S., A.C.T., M.L., J.G., S.R.D., J.G.S., C.E.S.), Harvard Medical School, Boston, MA
| | - Joshua Gorham
- Department of Genetics (R.A., H.W., Q.Z., D.R., C.T., A.S., A.C.T., M.L., J.G., S.R.D., J.G.S., C.E.S.), Harvard Medical School, Boston, MA
| | - Steven R. DePalma
- Department of Genetics (R.A., H.W., Q.Z., D.R., C.T., A.S., A.C.T., M.L., J.G., S.R.D., J.G.S., C.E.S.), Harvard Medical School, Boston, MA
| | - Steven P. Gygi
- Department of Cell Biology (J.A.P., S.P.G.), Harvard Medical School, Boston, MA
| | - J.G. Seidman
- Department of Genetics (R.A., H.W., Q.Z., D.R., C.T., A.S., A.C.T., M.L., J.G., S.R.D., J.G.S., C.E.S.), Harvard Medical School, Boston, MA
| | - Christine E. Seidman
- Department of Genetics (R.A., H.W., Q.Z., D.R., C.T., A.S., A.C.T., M.L., J.G., S.R.D., J.G.S., C.E.S.), Harvard Medical School, Boston, MA.,Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA (C.E.S.).,Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.)
| |
Collapse
|
9
|
Li WW, Yuan H, Kong S, Tian SB. E3 ubiquitin ligase TRIM55 promotes metastasis of gastric cancer cells by mediating epithelial-mesenchymal transition. World J Gastrointest Oncol 2022; 14:2183-2194. [PMID: 36438697 PMCID: PMC9694263 DOI: 10.4251/wjgo.v14.i11.2183] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 09/28/2022] [Accepted: 10/27/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Gastric cancer (GC) is considered a major global health problem. The role of TRIM55, a member of the three-domain protein family, in GC is unknown.
AIM To determine the expression of TRIM55 in GC tissues and its relationship with clinicopathological characteristics, and to investigate the effects of TRIM55 on the malignant biological behavior of GC cells.
METHODS Differential expression of TRIM55 in GC and para-cancer tissues was detected by immunohistochemistry, and the relationship between TRIM55 level and clinicopathological characteristics and prognosis was analyzed. Gain-of-function, loss-of-function, cell counting kit-8 assay, colony formation, transwell assay, wound healing assay, and western blot analysis were used to assess the potential role of TRIM55 in the development of GC.
RESULTS TRIM55 expression was significantly increased in GC tissues compared with adjacent normal tissues. High expression of TRIM55 was associated with advanced pathological stage and poor prognosis. Overexpression of TRIM55 promoted invasion and metastasis of GC cells in vitro by regulating epithelial-mesenchymal transition (EMT), whereas knockdown of TRIM55 had the opposite effect. Our data showed that TRIM55 is highly expressed in GC tissues, and is associated with poor prognosis. TRIM55 plays the role of an oncogene in GC, and it promotes metastasis of GC through the regulation of EMT.
CONCLUSION TRIM55 may be a possible target for the diagnosis and prognosis of GC patients.
Collapse
Affiliation(s)
- Wei-Wei Li
- Department of Pulmonary and Critical Care Medicine, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250021, Shandong Province, China
- Department of Critical Care Medicine, The 960th Hospital of the People's Liberation Army Joint Logistics Support Force, Jinan 250031, Shandong Province, China
| | - Hao Yuan
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong Province, China
| | - Shuai Kong
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong Province, China
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250021, Shandong Province, China
| | - Shu-Bo Tian
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong Province, China
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250021, Shandong Province, China
| |
Collapse
|
10
|
Kötter S, Krüger M. Protein Quality Control at the Sarcomere: Titin Protection and Turnover and Implications for Disease Development. Front Physiol 2022; 13:914296. [PMID: 35846001 PMCID: PMC9281568 DOI: 10.3389/fphys.2022.914296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 06/10/2022] [Indexed: 11/26/2022] Open
Abstract
Sarcomeres are mainly composed of filament and signaling proteins and are the smallest molecular units of muscle contraction and relaxation. The sarcomere protein titin serves as a molecular spring whose stiffness mediates myofilament extensibility in skeletal and cardiac muscle. Due to the enormous size of titin and its tight integration into the sarcomere, the incorporation and degradation of the titin filament is a highly complex task. The details of the molecular processes involved in titin turnover are not fully understood, but the involvement of different intracellular degradation mechanisms has recently been described. This review summarizes the current state of research with particular emphasis on the relationship between titin and protein quality control. We highlight the involvement of the proteasome, autophagy, heat shock proteins, and proteases in the protection and degradation of titin in heart and skeletal muscle. Because the fine-tuned balance of degradation and protein expression can be disrupted under pathological conditions, the review also provides an overview of previously known perturbations in protein quality control and discusses how these affect sarcomeric proteins, and titin in particular, in various disease states.
Collapse
|
11
|
Dittloff KT, Spanghero E, Solís C, Banach K, Russell B. Transthyretin deposition alters cardiomyocyte sarcomeric architecture, calcium transients, and contractile force. Physiol Rep 2022; 10:e15207. [PMID: 35262277 PMCID: PMC8906053 DOI: 10.14814/phy2.15207] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/20/2022] [Accepted: 02/06/2022] [Indexed: 06/14/2023] Open
Abstract
Age-related wild-type transthyretin amyloidosis (wtATTR) is characterized by systemic deposition of amyloidogenic fibrils of misfolded transthyretin (TTR) in the connective tissue of many organs. In the heart, this leads to age-related heart failure with preserved ejection fraction (HFpEF). The hypothesis tested is that TTR deposited in vitro disrupts cardiac myocyte cell-to-cell and cell-to-matrix adhesion complexes, resulting in altered calcium handling, force generation, and sarcomeric disorganization. Human iPSC-derived cardiomyocytes and neonatal rat ventricular myocytes (NRVMs), when grown on TTR-coated polymeric substrata mimicking the stiffness of the healthy human myocardium (10 kPa), had decreased contraction and relaxation velocities as well as decreased force production measured using traction force microscopy. Both NRVMs and adult mouse atrial cardiomyocytes had altered calcium kinetics with prolonged transients when cultured on TTR fibril-coated substrates. Furthermore, NRVMs grown on stiff (~GPa), flat or microgrooved substrates coated with TTR fibrils exhibited significantly decreased intercellular electrical coupling as shown by FRAP dynamics of cells loaded with the gap junction-permeable dye calcein-AM, along with decreased gap junction content as determined by quantitative connexin 43 staining. Significant sarcomeric disorganization and loss of sarcomere content, with increased ubiquitin localization to the sarcomere, were seen in NRVMs on various TTR fibril-coated substrata. TTR presence decreased intercellular mechanical junctions as evidenced by quantitative immunofluorescence staining of N-cadherin and vinculin. Current therapies for wtATTR are cost-prohibitive and only slow the disease progression; therefore, better understanding of cardiomyocyte maladaptation induced by TTR amyloid may identify novel therapeutic targets.
Collapse
Affiliation(s)
- Kyle T. Dittloff
- Department of Physiology and BiophysicsUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Emanuele Spanghero
- Department of Biomedical EngineeringUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Christopher Solís
- Department of Physiology and BiophysicsUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Kathrin Banach
- Department of Internal Medicine/CardiologyRush University Medical CenterChicagoIllinoisUSA
| | - Brenda Russell
- Department of Physiology and BiophysicsUniversity of Illinois at ChicagoChicagoIllinoisUSA
- Department of Biomedical EngineeringUniversity of Illinois at ChicagoChicagoIllinoisUSA
| |
Collapse
|
12
|
Guo T, Zhang Z, Zhu L, Chen W, Ding Y, Li W, Huang Y, Huang J, Pan X. TRIM55 suppresses malignant biological behavior of lung adenocarcinoma cells by increasing protein degradation of Snail1. Cancer Biol Ther 2022; 23:17-26. [PMID: 34974792 PMCID: PMC8812808 DOI: 10.1080/15384047.2021.2004835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Up until now, cancer refractoriness and distal organ metastatic disease remain as major obstacles for oncologists to achieve satisfactory therapeutic effects for lung adenocarcinoma patients. Previous studies indicated that TRIM55, which participates in the natural development of muscle and cardiovascular system, plays a protective role in hepatocellular carcinoma (HCC) pathogenesis. Therefore, in this study, we aimed to unveil the detailed molecular mechanism of TRIM55 and identify the potential target for lung adenocarcinoma patients. Surgical samples and lung cancer cell lines were collected to detect the TRIM55 expression for patients with or without lymph node/distal organ metastasis. Cellular functional assays including transwell assay, wound healing assay, cellular survivability assay, etc. as well as ubiquitination assay were performed to evaluate the impact of TRIM55/Snail1 regulatory network via the UPP pathway on lung cancer tumor cell migration and chemo-resistance. Lung cancer tissues and tumor cell lines exhibited significantly lower levels of TRIM55 expression. Functional study further indicated that TRIM55 inhibited chemo-resistance, migration, and cancer stem-cell like phenotype of tumor cells. Further detailed molecular experiments indicated that TRIM55 promoted degradation of Snail1 via the UPP pathway, which played an interesting role in the regulation of cancer cell malignancy. This study provided novel theory that TRIM55 acted as a potential tumor suppressor by inhibition of tumor cell malignancy through enhancement of Snail1 degradation via the UPP pathway. Our research will inspire further exploration on TRIM55 to promote therapeutic effects for lung adenocarcinoma patients.
Collapse
Affiliation(s)
- Tianxing Guo
- Department of Thoracic Surgery, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou China
| | - Zhenlong Zhang
- Department of Thoracic Surgery, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou China
| | - Lihuan Zhu
- Department of Thoracic Surgery, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou China
| | - Wenshu Chen
- Department of Thoracic Surgery, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou China
| | - Yun Ding
- Department of Thoracic Surgery, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou China
| | - Wujin Li
- Department of Thoracic Surgery, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou China
| | - Yangyun Huang
- Department of Thoracic Surgery, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou China
| | - Jianyuan Huang
- Department of Thoracic Surgery, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou China
| | - Xiaojie Pan
- Department of Thoracic Surgery, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou China
| |
Collapse
|
13
|
Zhu J, Wu Y, Lao S, Shen J, Yu Y, Fang C, Zhang N, Li Y, Zhang R. Targeting TRIM54/Axin1/β-Catenin Axis Prohibits Proliferation and Metastasis in Hepatocellular Carcinoma. Front Oncol 2021; 11:759842. [PMID: 34956880 PMCID: PMC8695909 DOI: 10.3389/fonc.2021.759842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/15/2021] [Indexed: 11/15/2022] Open
Abstract
Accumulating evidence demonstrates that dysregulation of ubiquitin-mediated degradation of oncogene or suppressors plays an important role in several diseases. However, the function and molecular mechanisms of ubiquitin ligases underlying hepatocellular carcinoma (HCC) remain elusive. In the current study, we show that overexpression of TRIM54 was associated with HCC progression. TRIM54 overexpression facilitates proliferation and lung metastasis; however, inhibition of TRIM54 significantly suppressed HCC progression both in vitro and in vivo. Mechanically, we demonstrated that TRIM54 directly interacts with Axis inhibition proteins 1 (Axin1) and induces E3 ligase-dependent proteasomal turnover of Axin1 and substantially induces sustained activation of wnt/β-catenin in HCC cell lines. Furthermore, we showed that inhibition of the wnt/β-catenin signaling pathway via small molecule inhibitors significantly suppressed TRIM54-induced proliferation. Our data suggest that TRIM54 might function as an oncogenic gene and targeting the TRIM54/Axin1/β-catenin axis signaling may be a promising prognostic factor and a valuable therapeutic target for HCC.
Collapse
Affiliation(s)
- Jinrong Zhu
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yongqi Wu
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Shaoxi Lao
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jianfei Shen
- Department of Cardiothoracic Surgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Yijian Yu
- Department of Cardiothoracic Surgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Chunqiang Fang
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Na Zhang
- Department of General Practice, Heyuan People's Hospital, Heyuan, Guangdong, China
| | - Yan Li
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Rongxin Zhang
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
14
|
Müller E, Salcan S, Bongardt S, Barbosa DM, Krüger M, Kötter S. E3-ligase knock down revealed differential titin degradation by autopagy and the ubiquitin proteasome system. Sci Rep 2021; 11:21134. [PMID: 34702928 PMCID: PMC8548520 DOI: 10.1038/s41598-021-00618-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 09/30/2021] [Indexed: 01/09/2023] Open
Abstract
The sarcomere protein titin is a major determinant of cardiomyocyte stiffness and ventricular distensibility. The constant mechanical stress on titin requires well-controlled protein quality control, the exact mechanisms of which have not yet been fully elucidated. Here, we analyzed E3-ligases potentially responsible for cardiac titin ubiquitination and specifically studied the involvement of the autophagosomal system in titin degradation. Pharmacological inhibition of autophagy and the proteasome in cultured primary rat cardiomyocytes significantly elevated titin ubiquitination and increased titin degradation. Using in-vitro pull down assays we identified binding of E3-ligases MuRF1-3, CHIP and Fbx32 to several titin domains. Immunofluorescence analysis showed sarcomeric localization of the E3-ligases. siRNA-mediated knock-down of the E3-ligases MuRF-1, -3 and a combination of CHIP/Fbx32 significantly reduced autophagy-related titin ubiquitination, whereas knock-down of MuRF-2 and -3 reduced proteasome-related titin ubiquitination. We demonstrated that the proteasomal and the autophagosomal-lysosomal system participate in degradation of the titin filament. We found that ubiquitination and degradation of titin are partially regulated by E3-ligases of the MuRF family. We further identified CHIP and Fbx32 as E3-ligases involved in titin ubiquitination.
Collapse
Affiliation(s)
- Erik Müller
- Department of Cardiovascular Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Universitätsstr. 1, 22.03 02, 40225, Düsseldorf, Germany
| | - Senem Salcan
- Department of Cardiovascular Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Universitätsstr. 1, 22.03 02, 40225, Düsseldorf, Germany
| | - Sabine Bongardt
- Department of Cardiovascular Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Universitätsstr. 1, 22.03 02, 40225, Düsseldorf, Germany
| | - David Monteiro Barbosa
- Department of Cardiovascular Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Universitätsstr. 1, 22.03 02, 40225, Düsseldorf, Germany
| | - Martina Krüger
- Department of Cardiovascular Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Universitätsstr. 1, 22.03 02, 40225, Düsseldorf, Germany
| | - Sebastian Kötter
- Department of Cardiovascular Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Universitätsstr. 1, 22.03 02, 40225, Düsseldorf, Germany.
| |
Collapse
|
15
|
Crocini C, Gotthardt M. Cardiac sarcomere mechanics in health and disease. Biophys Rev 2021; 13:637-652. [PMID: 34745372 PMCID: PMC8553709 DOI: 10.1007/s12551-021-00840-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 08/27/2021] [Indexed: 12/23/2022] Open
Abstract
The sarcomere is the fundamental structural and functional unit of striated muscle and is directly responsible for most of its mechanical properties. The sarcomere generates active or contractile forces and determines the passive or elastic properties of striated muscle. In the heart, mutations in sarcomeric proteins are responsible for the majority of genetically inherited cardiomyopathies. Here, we review the major determinants of cardiac sarcomere mechanics including the key structural components that contribute to active and passive tension. We dissect the molecular and structural basis of active force generation, including sarcomere composition, structure, activation, and relaxation. We then explore the giant sarcomere-resident protein titin, the major contributor to cardiac passive tension. We discuss sarcomere dynamics exemplified by the regulation of titin-based stiffness and the titin life cycle. Finally, we provide an overview of therapeutic strategies that target the sarcomere to improve cardiac contraction and filling.
Collapse
Affiliation(s)
- Claudia Crocini
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Neuromuscular and Cardiovascular Cell Biology, Berlin, Germany
- German Center for Cardiovascular Research (DZHK) Partner Site Berlin, Berlin, Germany
- BioFrontiers Institute & Department of Molecular and Cellular Development, University of Colorado Boulder, Boulder, USA
| | - Michael Gotthardt
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Neuromuscular and Cardiovascular Cell Biology, Berlin, Germany
- German Center for Cardiovascular Research (DZHK) Partner Site Berlin, Berlin, Germany
- Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| |
Collapse
|
16
|
Russell B, Solís C. Mechanosignaling pathways alter muscle structure and function by post-translational modification of existing sarcomeric proteins to optimize energy usage. J Muscle Res Cell Motil 2021; 42:367-380. [PMID: 33595762 DOI: 10.1007/s10974-021-09596-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 01/04/2021] [Indexed: 12/29/2022]
Abstract
A transduced mechanical signal arriving at its destination in muscle alters sarcomeric structure and function. A major question addressed is how muscle mass and tension generation are optimized to match actual performance demands so that little energy is wasted. Three cases for improved energy efficiency are examined: the troponin complex for tuning force production, control of the myosin heads in a resting state, and the Z-disc proteins for sarcomere assembly. On arrival, the regulation of protein complexes is often controlled by post-translational modification (PTM), of which the most common are phosphorylation by kinases, deacetylation by histone deacetylases and ubiquitination by E3 ligases. Another branch of signals acts not through peptide covalent bonding but via ligand interactions (e.g. Ca2+ and phosphoinositide binding). The myosin head and the regulation of its binding to actin by the troponin complex is the best and earliest example of signal destinations that modify myofibrillar contractility. PTMs in the troponin complex regulate both the efficiency of the contractile function to match physiologic demand for work, and muscle mass via protein degradation. The regulation of sarcomere assembly by integration of incoming signaling pathways causing the same PTMs or ligand binding are discussed in response to mechanical loading and unloading by the Z-disc proteins CapZ, α-actinin, telethonin, titin N-termini, and others. Many human mutations that lead to cardiomyopathy and heart disease occur in the proteins discussed above, which often occur at their PTM or ligand binding sites.
Collapse
Affiliation(s)
- Brenda Russell
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| | - Christopher Solís
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| |
Collapse
|
17
|
Zhang JR, Li XX, Hu WN, Li CY. Emerging Role of TRIM Family Proteins in Cardiovascular Disease. Cardiology 2020; 145:390-400. [PMID: 32305978 DOI: 10.1159/000506150] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 01/23/2020] [Indexed: 11/19/2022]
Abstract
Ubiquitination is one of the basic mechanisms of cell protein homeostasis and degradation and is accomplished by 3 enzymes, E1, E2, and E3. Tripartite motif-containing proteins (TRIMs) constitute the largest subfamily of RING E3 ligases, with >70 current members in humans and mice. These members are involved in multiple biological processes, including growth, differentiation, and apoptosis as well as disease and tumorigenesis. Accumulating evidence has shown that many TRIM proteins are associated with various cardiac processes and pathologies, such as heart development, signal transduction, protein degradation, autophagy mediation, ion channel regulation, congenital heart disease, and cardiomyopathies. In this review, we provide an overview of the TRIM family and discuss its involvement in the regulation of cardiac proteostasis and pathophysiology and its potential therapeutic implications.
Collapse
Affiliation(s)
- Jing-Rui Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xin-Xin Li
- Department of Respiratory Medicine, Tangshan People's Hospital, Tangshan, China
| | - Wan-Ning Hu
- Department of Cardiology, Laboratory of Molecular Biology, Tangshan Gongren Hospital, Tangshan, China,
| | - Chang-Yi Li
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Department of Cardiology, Laboratory of Molecular Biology, Tangshan Gongren Hospital, Tangshan, China
| |
Collapse
|
18
|
Heliste J, Chheda H, Paatero I, Salminen TA, Akimov Y, Paavola J, Elenius K, Aittokallio T. Genetic and functional implications of an exonic TRIM55 variant in heart failure. J Mol Cell Cardiol 2019; 138:222-233. [PMID: 31866377 DOI: 10.1016/j.yjmcc.2019.12.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 12/09/2019] [Accepted: 12/12/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND To tackle the missing heritability of sporadic heart failure, we screened for novel heart failure-associated genetic variants in the Finnish population and functionally characterized a novel variant in vitro and in vivo. METHODS AND RESULTS Heart failure-associated variants were screened in genotyping array data of the FINRISK study, consisting of 994 cases and 20,118 controls. Based on logistic regression analysis, a potentially damaging variant in TRIM55 (rs138811034), encoding an E140K variant, was selected for validations. In HL-1 cardiomyocytes, we used CRISPR/Cas9 technology to introduce the variant in the endogenous locus, and additionally TRIM55 wildtype or E140K was overexpressed from plasmid. Functional responses were profiled using whole-genome RNA sequencing, RT-PCR and Western analyses, cell viability and cell cycle assays and cell surface area measurements. In zebrafish embryos, cardiac contractility was measured using videomicroscopy after CRISPR-mediated knockout of trim55a or plasmid overexpression of TRIM55 WT or E140K. Genes related to muscle contraction and cardiac stress were highly regulated in Trim55 E140K/- cardiomyocytes. When compared to the WT/WT cells, the variant cells demonstrated reduced viability, significant hypertrophic response to isoproterenol, p21 protein overexpression and impaired cell cycle progression. In zebrafish embryos, the deletion of trim55a or overexpression of TRIM55 E140K reduced cardiac contractility as compared to embryos with wildtype genotype or overexpression of WT TRIM55, respectively. CONCLUSIONS A previously uncharacterized TRIM55 E140K variant demonstrated a number of functional implications for cardiomyocyte functions in vitro and in vivo. These findings suggest a novel role for TRIM55 polymorphism in predisposing to heart failure.
Collapse
Affiliation(s)
- Juho Heliste
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Biomedicum 2U, Tukholmankatu 8, FI-00290 Helsinki, Finland; Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, FI-20014 Turku, Finland; Turku Doctoral Programme of Molecular Medicine, University of Turku, Turku, Finland
| | - Himanshu Chheda
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Biomedicum 2U, Tukholmankatu 8, FI-00290 Helsinki, Finland
| | - Ilkka Paatero
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6, FI-20520 Turku, Finland
| | - Tiina A Salminen
- Structural Bioinformatics Laboratory, Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Tykistökatu 6, FI-20520 Turku, Finland
| | - Yevhen Akimov
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Biomedicum 2U, Tukholmankatu 8, FI-00290 Helsinki, Finland
| | - Jere Paavola
- Unit of Cardiovascular Research, Minerva Foundation Institute for Medical Research, Biomedicum 2U, Tukholmankatu 8, FI-00290 Helsinki, Finland
| | - Klaus Elenius
- Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, FI-20014 Turku, Finland; Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6, FI-20520 Turku, Finland; Medicity Research Laboratories, University of Turku, Tykistökatu 6, FI-20520 Turku, Finland; Department of Oncology, Turku University Hospital, PO Box 52, FI-20521 Turku, Finland
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Biomedicum 2U, Tukholmankatu 8, FI-00290 Helsinki, Finland; Department of Mathematics and Statistics, University of Turku, Vesilinnantie 5, FI-20014 Turku, Finland.
| |
Collapse
|
19
|
Skeletal Muscle-Specific Methyltransferase METTL21C Trimethylates p97 and Regulates Autophagy-Associated Protein Breakdown. Cell Rep 2019; 23:1342-1356. [PMID: 29719249 DOI: 10.1016/j.celrep.2018.03.136] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 03/02/2018] [Accepted: 03/29/2018] [Indexed: 01/04/2023] Open
Abstract
Protein aggregates and cytoplasmic vacuolization are major hallmarks of multisystem proteinopathies (MSPs) that lead to muscle weakness. Here, we identify METTL21C as a skeletal muscle-specific lysine methyltransferase. Insertion of a β-galactosidase cassette into the Mettl21c mouse locus revealed that METTL21C is specifically expressed in MYH7-positive skeletal muscle fibers. Ablation of the Mettl21c gene reduced endurance capacity and led to age-dependent accumulation of autophagic vacuoles in skeletal muscle. Denervation-induced muscle atrophy highlighted further impairments of autophagy-related proteins, including LC3, p62, and cathepsins, in Mettl21c-/- muscles. In addition, we demonstrate that METTL21C interacts with the ATPase p97 (VCP), which is mutated in various human MSP conditions. We reveal that METTL21C trimethylates p97 on the Lys315 residue and found that loss of this modification reduced p97 hexamer formation and ATPase activity in vivo. We conclude that the methyltransferase METTL21C is an important modulator of protein degradation in skeletal muscle under both normal and enhanced protein breakdown conditions.
Collapse
|
20
|
Multi-Staged Regulation of Lipid Signaling Mediators during Myogenesis by COX-1/2 Pathways. Int J Mol Sci 2019; 20:ijms20184326. [PMID: 31487817 PMCID: PMC6769623 DOI: 10.3390/ijms20184326] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 08/21/2019] [Indexed: 01/04/2023] Open
Abstract
Cyclooxygenases (COXs), including COX-1 and -2, are enzymes essential for lipid mediator (LMs) syntheses from arachidonic acid (AA), such as prostaglandins (PGs). Furthermore, COXs could interplay with other enzymes such as lipoxygenases (LOXs) and cytochrome P450s (CYPs) to regulate the signaling of LMs. In this study, to comprehensively analyze the function of COX-1 and -2 in regulating the signaling of bioactive LMs in skeletal muscle, mouse primary myoblasts and C2C12 cells were transfected with specific COX-1 and -2 siRNAs, followed by targeted lipidomic analysis and customized quantitative PCR gene array analysis. Knocking down COXs, particularly COX-1, significantly reduced the release of PGs from muscle cells, especially PGE2 and PGF2α, as well as oleoylethanolamide (OEA) and arachidonoylethanolamine (AEA). Moreover, COXs could interplay with LOXs to regulate the signaling of hydroxyeicosatetraenoic acids (HETEs). The changes in LMs are associated with the expression of genes, such as Itrp1 (calcium signaling) and Myh7 (myogenic differentiation), in skeletal muscle. In conclusion, both COX-1 and -2 contribute to LMs production during myogenesis in vitro, and COXs could interact with LOXs during this process. These interactions and the fine-tuning of the levels of these LMs are most likely important for skeletal muscle myogenesis, and potentially, muscle repair and regeneration.
Collapse
|
21
|
Silvestre JG, Baptista IL, Silva WJ, Cruz A, Silva MT, Miyabara EH, Labeit S, Moriscot AS. The E3 ligase MuRF2 plays a key role in the functional capacity of skeletal muscle fibroblasts. ACTA ACUST UNITED AC 2019; 52:e8551. [PMID: 31482977 PMCID: PMC6720025 DOI: 10.1590/1414-431x20198551] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 07/11/2019] [Indexed: 12/13/2022]
Abstract
Fibroblasts are a highly heterogeneous population of cells, being found in a large number of different tissues. These cells produce the extracellular matrix, which is essential to preserve structural integrity of connective tissues. Fibroblasts are frequently engaged in migration and remodeling, exerting traction forces in the extracellular matrix, which is crucial for matrix deposition and wound healing. In addition, previous studies performed on primary myoblasts suggest that the E3 ligase MuRF2 might function as a cytoskeleton adaptor. Here, we hypothesized that MuRF2 also plays a functional role in skeletal muscle fibroblasts. We found that skeletal muscle fibroblasts express MuRF2 and its siRNA knock-down promoted decreased fibroblast migration, cell border accumulation of polymerized actin, and down-regulation of the phospho-Akt expression. Our results indicated that MuRF2 was necessary to maintain the actin cytoskeleton functionality in skeletal muscle fibroblasts via Akt activity and exerted an important role in extracellular matrix remodeling in the skeletal muscle tissue.
Collapse
Affiliation(s)
- J G Silvestre
- Departamento de Anatomia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brasil
| | - I L Baptista
- Faculdade de Ciências Aplicadas, UNICAMP, Limeira, SP, Brasil
| | - W J Silva
- Departamento de Anatomia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brasil
| | - A Cruz
- Departamento de Anatomia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brasil
| | - M T Silva
- Departamento de Anatomia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brasil
| | - E H Miyabara
- Departamento de Anatomia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brasil
| | - S Labeit
- Institute for Integrative Pathophysiology, Mannheim Medical University, Faculty for Clinical Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - A S Moriscot
- Departamento de Anatomia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brasil
| |
Collapse
|
22
|
Sun X, Zhao D, Lu F, Peng S, Yu M, Liu N, Sun Y, Du H, Wang B, Chen J, Dong S, Lu F, Zhang W. Hydrogen sulfide regulates muscle RING finger-1 protein S-sulfhydration at Cys 44 to prevent cardiac structural damage in diabetic cardiomyopathy. Br J Pharmacol 2019; 177:836-856. [PMID: 30734268 DOI: 10.1111/bph.14601] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 12/04/2018] [Accepted: 12/19/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE Hydrogen sulfide (H2 S) plays important roles as a gasotransmitter in pathologies. Increased expression of the E3 ubiquitin ligase, muscle RING finger-1 (MuRF1), may be involved in diabetic cardiomyopathy. Here we have investigated whether and how exogenous H2 S alleviates cardiac muscle degradation through modifications of MuRF1 S-sulfhydration in db/db mice. EXPERIMENTAL APPROACH Neonatal rat cardiomyocytes were treated with high glucose (40 mM), oleate (100 μM), palmitate (400 μM), and NaHS (100 μM) for 72 hr. MuRF1 was silenced with siRNA technology and mutation at Cys44 . Endoplasmic reticulum stress markers, MuRF1 expression, and ubiquitination level were measured. db/db mice were injected with NaHS (39 μmol·kg-1 ) for 20 weeks. Echocardiography, cardiac ultrastructure, cystathionine-γ-lyase, cardiac structure proteins expression, and S-sulfhydration production were measured. KEY RESULTS H2 S levels and cystathionine-γ-lyase protein expression in myocardium were decreased in db/db mice. Exogenous H2 S reversed endoplasmic reticulum stress, including impairment of the function of cardiomyocytes and structural damage in db/db mice. Exogenous H2 S could suppress the levels of myosin heavy chain 6 and myosin light chain 2 ubiquitination in cardiac tissues of db/db mice, and MuRF1 was modified by S-sulfhydration, following treatment with exogenous H2 S, to reduce the interaction between MuRF1 and myosin heavy chain 6 and myosin light chain 2. CONCLUSIONS AND IMPLICATIONS Our findings suggest that H2 S regulates MuRF1 S-sulfhydration at Cys44 to prevent myocardial degradation in the cardiac tissues of db/db mice. LINKED ARTICLES This article is part of a themed section on Hydrogen Sulfide in Biology & Medicine. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.4/issuetoc.
Collapse
Affiliation(s)
- Xiaojiao Sun
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Dechao Zhao
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Fangping Lu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Shuo Peng
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Miao Yu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Ning Liu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Yu Sun
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Haining Du
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Bingzhu Wang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Jian Chen
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Shiyun Dong
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Fanghao Lu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Weihua Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin, China.,Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University Ministry of Education, Harbin, China
| |
Collapse
|
23
|
SPRED2 deficiency elicits cardiac arrhythmias and premature death via impaired autophagy. J Mol Cell Cardiol 2019; 129:13-26. [DOI: 10.1016/j.yjmcc.2019.01.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 01/25/2019] [Accepted: 01/25/2019] [Indexed: 01/20/2023]
|
24
|
Li X, Huang L, Gao W. Overexpression of Tripartite Motif Conaining 55 (TRIM55) Inhibits Migration and Invasion of Hepatocellular Carcinoma (HCC) Cells via Epithelial-Mesenchymal Transition and Matrix Metalloproteinase-2 (MMP2). Med Sci Monit 2019; 25:771-777. [PMID: 30685767 PMCID: PMC6360872 DOI: 10.12659/msm.910984] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Tripartite motif containing 55 (TRIM55) plays a regulatory role in assembly of sarcomeres, but few studies have assessed its function in hepatocellular carcinoma (HCC). MATERIAL AND METHODS Immunohistochemistry (IHC) was used to detect expression of TRIM55 in tissues samples of HCC patients. Transwell assay was used to study migration and invasion ability of HCC cells. Western blot and immunofluorescence (IF) were used to analyze mechanism of TRIM55 in cell migration and invasion. RESULTS We found TRIM55 was downregulated in HCC tissues and was associated with prognosis of HCC patients. Cox regression analysis showed that TRIM55 was an independent risk factor of prognosis of HCC patients. Overexpression of TRIM55 was associated with lower cell migration and invasion ability, and it led to high expression of E-cadherin and low expression of Vimentin and MMP2. CONCLUSIONS Our study found TRIM55 is an independent factor affecting the prognosis of HCC patients, and overexpression of TRIM55 inhibits migration and invasion of HCC cells through epithelial-mesenchymal transition and MMP2.
Collapse
Affiliation(s)
- Xinyu Li
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China (mainland)
| | - Lei Huang
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China (mainland)
| | - Weijie Gao
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China (mainland)
| |
Collapse
|
25
|
Han T, Guo M, Gan M, Yu B, Tian X, Wang JB. TRIM59 regulates autophagy through modulating both the transcription and the ubiquitination of BECN1. Autophagy 2018; 14:2035-2048. [PMID: 30231667 PMCID: PMC6984771 DOI: 10.1080/15548627.2018.1491493] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Macroautophagy/autophagy is a multistep cellular process that sequesters cytoplasmic components for lysosomal degradation. BECN1/Beclin1 is a central protein that assembles cofactors for the formation of a BECN1-PIK3C3-PIK3R4 complex to trigger the autophagy protein cascade. Discovering the regulators of BECN1 is important for understanding the mechanism of autophagy induction. Here, we demonstrate that TRIM59, a tripartite motif protein, plays an important role in autophagy regulation in non-small cell lung cancer (NSCLC). On the one hand, TRIM59 regulates the transcription of BECN1 through negatively modulating the NFKB pathway. On the other hand, TRIM59 regulates TRAF6 induced K63-linked ubiquitination of BECN1, thus affecting the formation of the BECN1-PIK3C3 complex. We further demonstrate that TRIM59 can mediate K48-linked ubiquitination of TRAF6 and promote the proteasomal degradation of TRAF6. Taken together, our findings reveal novel dual roles for TRIM59 in autophagy regulation by affecting both the transcription and the ubiquitination of BECN1. Abbreviations: ACTB: actin beta; BECN1: beclin 1; CHX: cycloheximide; CQ: chloroquine; GFP: green fluorescent protein; HA: haemagglutinin tag; His: polyhistidine tag; LC3B: microtubule associated protein 1 light chain 3 beta; NFKB: nuclear factor kappa B; NFKBIA: NFKB inhibitor alpha; NSCLC: non-small cell lung cancer; PIK3C3: phosphatidylinositol 3-kinase catalytic subunit type 3; RELA: RELA proto-oncogene, NF-kB subunit; SQSTM1: sequestosome 1; tGFP: Turbo green fluorescent protein; TRAF6: TNF receptor associated factor 6; TRIM59: tripartite motif containing 59; B: ubiquitin
Collapse
Affiliation(s)
- Tianyu Han
- a Institute of Translational Medicine , Nanchang University , Nanchang , Jiangxi , China.,b School of Life Sciences , Nanchang University , Nanchang , Jiangxi , China
| | - Meng Guo
- a Institute of Translational Medicine , Nanchang University , Nanchang , Jiangxi , China
| | - Mingxi Gan
- a Institute of Translational Medicine , Nanchang University , Nanchang , Jiangxi , China
| | - Bentong Yu
- c Department of Cardiovascular Surgery , The First Affiliated Hospital of Nanchang University , Nanchang , Jiangxi , China
| | - Xiaoli Tian
- b School of Life Sciences , Nanchang University , Nanchang , Jiangxi , China
| | - Jian-Bin Wang
- a Institute of Translational Medicine , Nanchang University , Nanchang , Jiangxi , China
| |
Collapse
|
26
|
Dunn-Fletcher CE, Muglia LM, Pavlicev M, Wolf G, Sun MA, Hu YC, Huffman E, Tumukuntala S, Thiele K, Mukherjee A, Zoubovsky S, Zhang X, Swaggart KA, Lamm KYB, Jones H, Macfarlan TS, Muglia LJ. Anthropoid primate-specific retroviral element THE1B controls expression of CRH in placenta and alters gestation length. PLoS Biol 2018; 16:e2006337. [PMID: 30231016 PMCID: PMC6166974 DOI: 10.1371/journal.pbio.2006337] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 10/01/2018] [Accepted: 09/10/2018] [Indexed: 01/22/2023] Open
Abstract
Pregnancy and parturition are intricately regulated to ensure successful reproductive outcomes. However, the factors that control gestational length in humans and other anthropoid primates remain poorly defined. Here, we show the endogenous retroviral long terminal repeat transposon-like human element 1B (THE1B) selectively controls placental expression of corticotropin-releasing hormone (CRH) that, in turn, influences gestational length and birth timing. Placental expression of CRH and subsequently prolonged gestational length were found in two independent strains of transgenic mice carrying a 180-kb human bacterial artificial chromosome (BAC) DNA that contained the full length of CRH and extended flanking regions, including THE1B. Restricted deletion of THE1B silenced placental CRH expression and normalized birth timing in these transgenic lines. Furthermore, we revealed an interaction at the 5′ insertion site of THE1B with distal-less homeobox 3 (DLX3), a transcription factor expressed in placenta. Together, these findings suggest that retroviral insertion of THE1B into the anthropoid primate genome may have initiated expression of CRH in placental syncytiotrophoblasts via DLX3 and that this placental CRH is sufficient to alter the timing of birth. The proper timing of delivery is critical during pregnancy; if too early or too late, the baby will be at risk of serious health problems and even death. Corticotropin-releasing hormone (CRH) is a protein that can be detected in maternal blood, and its concentration correlates with the timing of birth. In humans and other anthropoid primates, CRH is made by the placenta, whereas in other mammals, it is produced in a specialized region of the brain. To understand the regulation and evolution of this key protein, we inserted the human CRH gene and nearby regions into the mouse genome, which resulted in human CRH expression in the mouse placenta. Mouse litters that make CRH in their placentas are born later than control mice, showing that CRH can directly affect birth timing. Using our mouse model, we then selectively deleted a remnant of an ancient retrovirus that is normally found in the DNA of anthropoid primates and demonstrated that this specific region controls expression of CRH in the placenta. Deletion of this region also restored normal birth timing in the mice by eliminating CRH production from the placenta. We propose that retroviral regulation of CRH in the placenta may be a mechanism of controlling birth timing in humans and other anthropoid primates.
Collapse
Affiliation(s)
- Caitlin E. Dunn-Fletcher
- Division of Human Genetics, Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
- * E-mail: (CED); (LJM)
| | - Lisa M. Muglia
- Division of Human Genetics, Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Mihaela Pavlicev
- Division of Human Genetics, Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Gernot Wolf
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, The National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ming-An Sun
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, The National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yueh-Chiang Hu
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Elizabeth Huffman
- Division of Human Genetics, Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Shivani Tumukuntala
- Division of Human Genetics, Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Katri Thiele
- Division of Human Genetics, Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Amrita Mukherjee
- Division of Human Genetics, Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Sandra Zoubovsky
- Division of Human Genetics, Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Xuzhe Zhang
- Division of Human Genetics, Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Kayleigh A. Swaggart
- Division of Human Genetics, Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Katherine Y. Bezold Lamm
- Division of Human Genetics, Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Helen Jones
- Division of Pediatric Surgery, Cincinnati Children’s Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Todd S. Macfarlan
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, The National Institutes of Health, Bethesda, Maryland, United States of America
| | - Louis J. Muglia
- Division of Human Genetics, Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
- * E-mail: (CED); (LJM)
| |
Collapse
|
27
|
Borlepawar A, Frey N, Rangrez AY. A systematic view on E3 ligase Ring TRIMmers with a focus on cardiac function and disease. Trends Cardiovasc Med 2018; 29:1-8. [PMID: 29880235 DOI: 10.1016/j.tcm.2018.05.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 05/05/2018] [Accepted: 05/22/2018] [Indexed: 01/01/2023]
Abstract
Ubiquitination, a post-translational modification via ubiquitin-proteasome-system, is one of the vital cellular processes involved in intracellular signaling, cell death, transcriptional control, etc. Importantly, it prevents the aggregation of non-functional, misfolded or unfolded, potentially toxic proteins to maintain cellular protein homeostasis. Ubiquitination is accomplished by the concerted action of three enzymatic steps involving E1 activating enzymes, E2 conjugating enzymes, and E3 ligases. Tripartite motif-containing (TRIM) proteins are one of the integral members of E3 ubiquitin ligases in metazoans modulating essential cellular pathways. For long, MuRFs (Muscle ring finger proteins) were the most extensively studied TRIMs for their cardiac function. Recent research advances in the field and our analysis presented here, however, demonstrated broader and ever increasing involvement of additional TRIM E3 ligases in the pathophysiology of heart. In this review, we summarize the known cardiac E3 ligases and their targets, and discuss their role and importance in cardiac proteostasis, pathophysiology and potential therapeutic implications with specific focus on TRIM E3 ligases.
Collapse
Affiliation(s)
- Ankush Borlepawar
- Department of Internal Medicine III, University of Kiel, Arnold-Heller-Str. 3, 24105, Kiel, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany
| | - Norbert Frey
- Department of Internal Medicine III, University of Kiel, Arnold-Heller-Str. 3, 24105, Kiel, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany
| | - Ashraf Yusuf Rangrez
- Department of Internal Medicine III, University of Kiel, Arnold-Heller-Str. 3, 24105, Kiel, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany.
| |
Collapse
|
28
|
Fusco C, Mandriani B, Di Rienzo M, Micale L, Malerba N, Cocciadiferro D, Sjøttem E, Augello B, Squeo GM, Pellico MT, Jain A, Johansen T, Fimia GM, Merla G. TRIM50 regulates Beclin 1 proautophagic activity. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:908-919. [PMID: 29604308 DOI: 10.1016/j.bbamcr.2018.03.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 03/15/2018] [Accepted: 03/25/2018] [Indexed: 02/07/2023]
Abstract
Autophagy is a catabolic process needed for maintaining cell viability and homeostasis in response to numerous stress conditions. Emerging evidence indicates that the ubiquitin system has a major role in this process. TRIMs, an E3 ligase protein family, contribute to selective autophagy acting as receptors and regulators of the autophagy proteins recognizing endogenous or exogenous targets through intermediary autophagic tags, such as ubiquitin. Here we report that TRIM50 fosters the initiation phase of starvation-induced autophagy and associates with Beclin1, a central component of autophagy initiation complex. We show that TRIM50, via the RING domain, ubiquitinates Beclin 1 in a K63-dependent manner enhancing its binding with ULK1 and autophagy activity. Finally, we found that the Lys-372 residue of TRIM50, critical for its own acetylation, is necessary for its E3 ligase activity that governs Beclin1 ubiquitination. Our study expands the roles of TRIMs in regulating selective autophagy, revealing an acetylation-ubiquitination dependent control for autophagy modulation.
Collapse
Affiliation(s)
- Carmela Fusco
- Division of Medical Genetics, IRCCS Casa Sollievo Della Sofferenza, Viale Cappuccini, 71013 San Giovanni Rotondo, Italy
| | - Barbara Mandriani
- Division of Medical Genetics, IRCCS Casa Sollievo Della Sofferenza, Viale Cappuccini, 71013 San Giovanni Rotondo, Italy
| | - Martina Di Rienzo
- National Institute for Infectious Diseases IRCCS 'L. Spallanzani', Rome, Italy; Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Lucia Micale
- Division of Medical Genetics, IRCCS Casa Sollievo Della Sofferenza, Viale Cappuccini, 71013 San Giovanni Rotondo, Italy
| | - Natascia Malerba
- Division of Medical Genetics, IRCCS Casa Sollievo Della Sofferenza, Viale Cappuccini, 71013 San Giovanni Rotondo, Italy
| | - Dario Cocciadiferro
- Division of Medical Genetics, IRCCS Casa Sollievo Della Sofferenza, Viale Cappuccini, 71013 San Giovanni Rotondo, Italy; Ph.D Program in Experimental and Regenerative Medicine, University of Foggia, Italy
| | - Eva Sjøttem
- Molecular Cancer Research Group, Institute of Medical Biology, University of Tromsø-The Arctic University of Norway, 9037 Tromsø, Norway
| | - Bartolomeo Augello
- Division of Medical Genetics, IRCCS Casa Sollievo Della Sofferenza, Viale Cappuccini, 71013 San Giovanni Rotondo, Italy
| | - Gabriella Maria Squeo
- Division of Medical Genetics, IRCCS Casa Sollievo Della Sofferenza, Viale Cappuccini, 71013 San Giovanni Rotondo, Italy
| | - Maria Teresa Pellico
- Division of Medical Genetics, IRCCS Casa Sollievo Della Sofferenza, Viale Cappuccini, 71013 San Giovanni Rotondo, Italy
| | - Ashish Jain
- Molecular Cancer Research Group, Institute of Medical Biology, University of Tromsø-The Arctic University of Norway, 9037 Tromsø, Norway; Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Montebello, N-0379 Oslo, Norway
| | - Terje Johansen
- Molecular Cancer Research Group, Institute of Medical Biology, University of Tromsø-The Arctic University of Norway, 9037 Tromsø, Norway
| | - Gian Maria Fimia
- National Institute for Infectious Diseases IRCCS 'L. Spallanzani', Rome, Italy; Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce 73100, Italy
| | - Giuseppe Merla
- Division of Medical Genetics, IRCCS Casa Sollievo Della Sofferenza, Viale Cappuccini, 71013 San Giovanni Rotondo, Italy.
| |
Collapse
|
29
|
Wang L, Geist J, Grogan A, Hu LYR, Kontrogianni-Konstantopoulos A. Thick Filament Protein Network, Functions, and Disease Association. Compr Physiol 2018; 8:631-709. [PMID: 29687901 PMCID: PMC6404781 DOI: 10.1002/cphy.c170023] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Sarcomeres consist of highly ordered arrays of thick myosin and thin actin filaments along with accessory proteins. Thick filaments occupy the center of sarcomeres where they partially overlap with thin filaments. The sliding of thick filaments past thin filaments is a highly regulated process that occurs in an ATP-dependent manner driving muscle contraction. In addition to myosin that makes up the backbone of the thick filament, four other proteins which are intimately bound to the thick filament, myosin binding protein-C, titin, myomesin, and obscurin play important structural and regulatory roles. Consistent with this, mutations in the respective genes have been associated with idiopathic and congenital forms of skeletal and cardiac myopathies. In this review, we aim to summarize our current knowledge on the molecular structure, subcellular localization, interacting partners, function, modulation via posttranslational modifications, and disease involvement of these five major proteins that comprise the thick filament of striated muscle cells. © 2018 American Physiological Society. Compr Physiol 8:631-709, 2018.
Collapse
Affiliation(s)
- Li Wang
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | - Janelle Geist
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | - Alyssa Grogan
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | - Li-Yen R. Hu
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | | |
Collapse
|
30
|
Bian H, Gao S, Zhang D, Zhao Q, Li F, Li X, Sun S, Song S, Li T, Zhu Q, Ren W, Qin C, Qi J. The E3 ubiquitin ligase MuRF2 attenuates LPS-induced macrophage activation by inhibiting production of inflammatory cytokines and migration. FEBS Open Bio 2018; 8:234-243. [PMID: 29435413 PMCID: PMC5794466 DOI: 10.1002/2211-5463.12367] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 11/15/2017] [Accepted: 11/24/2017] [Indexed: 01/06/2023] Open
Abstract
Muscle RING-finger (MuRF) proteins are E3 ubiquitin ligases that are expressed in striated muscle. MuRF2 is an important member of this family, but whether it is expressed in tissues other than striated muscle has not been thoroughly elucidated to date. In this study, we determined that MuRF2 is also expressed in other vital organs, including liver, lung, brain, spleen and kidney. Moreover, we show that the level of MuRF2 expression is significantly decreased in hepatic mononuclear cells of mice with lipopolysaccharide (LPS)/d-galactosamine-induced hepatitis and negatively correlated with the serum levels of alanine aminotransferase and aspartate aminotransferase in these mice. Furthermore, the expression of MuRF2 was down-regulated in RAW264.7 cells activated with LPS but not in cells treated with polyinosinic-polycytidylic acid (Poly(I:C)) or with lipidosome plus Poly(I:C). We also found that MuRF2 was able to translocate from the cytoplasm to the nucleus in RAW264.7 cells activated with LPS but not in cells treated with Poly(I:C). In addition, we demonstrated that interleukin 6 and tumour necrosis factor α production and macrophage migration were inhibited after MuRF2 was overexpressed in RAW264.7 cells. We further verified that nuclear factor-κB p65 subunit level was greatly reduced in RAW264.7 macrophage nuclei by gain of function. Taken together, these findings indicate that MuRF2 may rescue LPS-induced macrophage activation by suppressing the production of proinflammatory cytokines and cell migration. We also identify a novel function of MuRF2 in non-muscle tissues and cells.
Collapse
Affiliation(s)
- Hongjun Bian
- Shandong Provincial Hospital Affiliated to Shandong University Jinan China.,Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control Jinan China
| | - Shanshan Gao
- Shandong Provincial Hospital Affiliated to Shandong University Jinan China.,Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control Jinan China
| | - Di Zhang
- Shandong Provincial Hospital Affiliated to Shandong University Jinan China.,Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control Jinan China
| | - Qi Zhao
- Shandong Provincial Hospital Affiliated to Shandong University Jinan China.,Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control Jinan China
| | - Feifei Li
- Shandong Provincial Hospital Affiliated to Shandong University Jinan China.,Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control Jinan China
| | - Xiao Li
- Shandong Provincial Hospital Affiliated to Shandong University Jinan China.,Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control Jinan China
| | - Shuohuan Sun
- Shandong Provincial Hospital Affiliated to Shandong University Jinan China.,Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control Jinan China
| | - Shouyang Song
- Shandong Provincial Hospital Affiliated to Shandong University Jinan China.,Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control Jinan China
| | - Tao Li
- Shandong Provincial Hospital Affiliated to Shandong University Jinan China.,Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control Jinan China
| | - Qiang Zhu
- Shandong Provincial Hospital Affiliated to Shandong University Jinan China.,Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control Jinan China
| | - Wanhua Ren
- Shandong Provincial Hospital Affiliated to Shandong University Jinan China.,Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control Jinan China
| | - Chengyong Qin
- Shandong Provincial Hospital Affiliated to Shandong University Jinan China.,Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control Jinan China
| | - Jianni Qi
- Shandong Provincial Hospital Affiliated to Shandong University Jinan China.,Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control Jinan China
| |
Collapse
|
31
|
Hatakeyama S. TRIM Family Proteins: Roles in Autophagy, Immunity, and Carcinogenesis. Trends Biochem Sci 2017; 42:297-311. [DOI: 10.1016/j.tibs.2017.01.002] [Citation(s) in RCA: 635] [Impact Index Per Article: 79.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 12/28/2016] [Accepted: 01/02/2017] [Indexed: 01/19/2023]
|
32
|
Li M, Andersson-Lendahl M, Sejersen T, Arner A. Knockdown of fast skeletal myosin-binding protein C in zebrafish results in a severe skeletal myopathy. ACTA ACUST UNITED AC 2016; 147:309-22. [PMID: 27022191 PMCID: PMC4810067 DOI: 10.1085/jgp.201511452] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 02/26/2016] [Indexed: 12/30/2022]
Abstract
MyBPC: A muscle protein for all seasons. Myosin-binding protein C (MyBPC) in the muscle sarcomere interacts with several contractile and structural proteins. Mutations in the cardiac isoform (MyBPC-3) in humans, or animal knockout, are associated with cardiomyopathy. Function of the fast skeletal isoform (MyBPC-2) in living muscles is less understood. This question was addressed using zebrafish models, combining gene expression data with functional analysis of contractility and small-angle x-ray diffraction measurements of filament structure. Fast skeletal MyBPC-2B, the major isoform, was knocked down by >50% using morpholino antisense nucleotides. These morphants exhibited a skeletal myopathy with elevated apoptosis and up-regulation of factors associated with muscle protein degradation. Morphant muscles had shorter sarcomeres with a broader length distribution, shorter actin filaments, and a wider interfilament spacing compared with controls, suggesting that fast skeletal MyBPC has a role in sarcomere assembly. Active force was reduced more than expected from the decrease in muscle size, suggesting that MyBPC-2 is required for optimal force generation at the cross-bridge level. The maximal shortening velocity was significantly increased in the MyBPC-2 morphants, but when related to the sarcomere length, the difference was smaller, reflecting that the decrease in MyBPC-2B content and the resulting myopathy were accompanied by only a minor influence on filament shortening kinetics. In the controls, equatorial patterns from small-angle x-ray scattering revealed that comparatively few cross-bridges are attached (as evaluated by the intensity ratio of the 11 and 10 equatorial reflections) during active contraction. X-ray scattering data from relaxed and contracting morphants were not significantly different from those in controls. However, the increase in the 11:10 intensity ratio in rigor was lower compared with that in controls, possibly reflecting effects of MyBPC on the cross-bridge interactions. In conclusion, lack of MyBPC-2 results in a severe skeletal myopathy with structural changes and muscle weakness.
Collapse
Affiliation(s)
- Mei Li
- Department of Physiology and Pharmacology, Department of Cell and Molecular Biology, and Department of Women's and Children's Health, Karolinska Institutet, SE 171 77 Stockholm, Sweden
| | - Monika Andersson-Lendahl
- Department of Physiology and Pharmacology, Department of Cell and Molecular Biology, and Department of Women's and Children's Health, Karolinska Institutet, SE 171 77 Stockholm, Sweden
| | - Thomas Sejersen
- Department of Physiology and Pharmacology, Department of Cell and Molecular Biology, and Department of Women's and Children's Health, Karolinska Institutet, SE 171 77 Stockholm, Sweden
| | - Anders Arner
- Department of Physiology and Pharmacology, Department of Cell and Molecular Biology, and Department of Women's and Children's Health, Karolinska Institutet, SE 171 77 Stockholm, Sweden
| |
Collapse
|
33
|
Skugor S, Jodaa Holm H, Bjelland AK, Pino J, Evensen Ø, Krasnov A, Wadsworth S. Nutrigenomic effects of glucosinolates on liver, muscle and distal kidney in parasite-free and salmon louse infected Atlantic salmon. Parasit Vectors 2016; 9:639. [PMID: 27955686 PMCID: PMC5153675 DOI: 10.1186/s13071-016-1921-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 11/28/2016] [Indexed: 12/22/2022] Open
Abstract
Background Reduction of Lepeophtheirus salmonis infection in Atlantic salmon achieved by glucosinolates (GLs) from Brassica plants was recently reported. However, wider application of functional feeds based on GLs requires better knowledge of their positive and adverse effects. Methods Liver, distal kidney and muscle transcriptomes of salmon exposed to the extreme dose of GLs were profiled by microarray, while qPCR analysis followed up selected hepatic and renal responses under the extreme and moderate GLs dose during the L. salmonis challenge. Transcriptional analysis were complemented with measurements of organ indices, liver steatosis and plasma profiling, including indicators of cytolysis and bilirubin. Finally, the third trial was performed to quantify the effect of lower GLs doses on growth. Results The extreme GLs dose caused a decrease in hepatic fat deposition and growth, in line with microarray findings, which suggested tissue remodeling and reduction of cellular proliferation in the skeletal muscle and liver. Lower GLs inclusion levels in a follow-up trial did not show negative effects on growth. Microarray analysis of the distal kidney pointed to activation of anti-fibrotic responses under the overexposure. However, analyses of ALT, CK and AST enzymes in plasma provided no evidence of increased cytolysis and organ damage. Prevalent activation of phase-2 detoxification genes that occurred in all three tissues could be considered part of beneficial effects caused by the extreme dose of GLs. In addition, transcriptomic evidence suggested GLs-mediated iron and heme withdrawal response, including increased heme degradation in muscle (upregulation of heme oxygenase-1), decrease of its synthesis in liver (downregulation of porphobilinogen deaminase) and increased iron sequestration from blood (hepatic induction of hepcidin-1 and renal induction of intracellular storage protein ferritin). This response could be advantageous for salmon upon encountering lice, which depend on the host for the provision of iron carrying heme. Most of the hepatic genes studied by qPCR showed similar expression levels in fish exposed to GLs, lice and their combination, while renal induction of leptin suggested heightened stress by the combination of extreme dose of GLs and lice. High expression of interferonγ (cytokine considered organ-protective in mammalian kidney) was detected at the moderate GLs level. This fish also showed highest plasma bilirubin levels (degradation product of heme), and had lowest number of attached lice, further supporting hypothesis that making heme unavailable to lice could be part of an effective anti-parasitic strategy. Conclusions Modulation of detoxification and iron metabolism in Atlantic salmon tissues could be beneficial prior and during lice infestations. Investigation of anti-lice functional feeds based on low and moderate GLs inclusion levels thus deserves further attention. Electronic supplementary material The online version of this article (doi:10.1186/s13071-016-1921-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Stanko Skugor
- Cargill Innovation Center, Sea Lice Research Centre, Oslo, Norway.
| | - Helle Jodaa Holm
- Norwegian University of Life Sciences, Faculty of Veterinary Medicine and Biosciences, Sea Lice Research Centre, Oslo, Norway
| | | | - Jorge Pino
- Cargill Innovation Center, Puerto Montt, Chile
| | - Øystein Evensen
- Norwegian University of Life Sciences, Faculty of Veterinary Medicine and Biosciences, Sea Lice Research Centre, Oslo, Norway
| | | | | |
Collapse
|
34
|
Parry TL, Willis MS. Cardiac ubiquitin ligases: Their role in cardiac metabolism, autophagy, cardioprotection and therapeutic potential. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1862:2259-2269. [PMID: 27421947 PMCID: PMC5159290 DOI: 10.1016/j.bbadis.2016.07.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 07/05/2016] [Accepted: 07/11/2016] [Indexed: 12/19/2022]
Abstract
Both the ubiquitin-proteasome system (UPS) and the lysosomal autophagy system have emerged as complementary key players responsible for the turnover of cellular proteins. The regulation of protein turnover is critical to cardiomyocytes as post-mitotic cells with very limited regenerative capacity. In this focused review, we describe the emerging interface between the UPS and autophagy, with E3's regulating autophagy at two critical points through multiple mechanisms. Moreover, we discuss recent insights in how both the UPS and autophagy can alter metabolism at various levels, to present new ways to think about therapeutically regulating autophagy in a focused manner to optimize disease-specific cardioprotection, without harming the overall homeostasis of protein quality control. This article is part of a Special Issue entitled: The role of post-translational protein modifications on heart and vascular metabolism edited by Jason R.B. Dyck & Jan F.C. Glatz.
Collapse
Affiliation(s)
- Traci L Parry
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA; Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Monte S Willis
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA; Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA; Department of Pharmacology, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
35
|
Abstract
Autophagy, a pathway for lysosomal-mediated cellular degradation, has recently been described as a regulator of cell migration. Although the molecular mechanisms underlying autophagy-dependent motility are only beginning to emerge, new work demonstrates that selective autophagy mediated by the autophagy cargo receptor, NBR1, specifically promotes the dynamic turnover of integrin-based focal adhesion sites during motility. Here, we discuss the detailed mechanisms through which NBR1-dependent selective autophagy supports focal adhesion remodeling, and we describe the interconnections between this pathway and other established regulators of focal adhesion turnover, such as microtubules. We also highlight studies that examine the contribution of autophagy to selective degradation of proteins that mediate cellular tension and to integrin trafficking; these findings hint at further roles for autophagy in supporting adhesion and migration. Given the recently appreciated importance of selective autophagy in diverse cellular processes, we propose that further investigation into autophagy-mediated focal adhesion turnover will not only shed light onto how focal adhesions are regulated but will also unveil new mechanisms regulating selective autophagy.
Collapse
Affiliation(s)
- Candia M Kenific
- Department of Pathology and Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143, USA Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA 94143, USA
| | - Torsten Wittmann
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jayanta Debnath
- Department of Pathology and Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
36
|
Lodka D, Pahuja A, Geers-Knörr C, Scheibe RJ, Nowak M, Hamati J, Köhncke C, Purfürst B, Kanashova T, Schmidt S, Glass DJ, Morano I, Heuser A, Kraft T, Bassel-Duby R, Olson EN, Dittmar G, Sommer T, Fielitz J. Muscle RING-finger 2 and 3 maintain striated-muscle structure and function. J Cachexia Sarcopenia Muscle 2016; 7:165-80. [PMID: 27493870 PMCID: PMC4863828 DOI: 10.1002/jcsm.12057] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 05/24/2015] [Accepted: 06/04/2015] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND The Muscle-specific RING-finger (MuRF) protein family of E3 ubiquitin ligases is important for maintenance of muscular structure and function. MuRF proteins mediate adaptation of striated muscles to stress. MuRF2 and MuRF3 bind to microtubules and are implicated in sarcomere formation with noticeable functional redundancy. However, if this redundancy is important for muscle function in vivo is unknown. Our objective was to investigate cooperative function of MuRF2 and MuRF3 in the skeletal muscle and the heart in vivo. METHODS MuRF2 and MuRF3 double knockout mice (DKO) were generated and phenotypically characterized. Skeletal muscle and the heart were investigated by morphological measurements, histological analyses, electron microscopy, immunoblotting, and real-time PCR. Isolated muscles were subjected to in vitro force measurements. Cardiac function was determined by echocardiography and working heart preparations. Function of cardiomyocytes was measured in vitro. Cell culture experiments and mass-spectrometry were used for mechanistic analyses. RESULTS DKO mice showed a protein aggregate myopathy in skeletal muscle. Maximal force development was reduced in DKO soleus and extensor digitorum longus. Additionally, a fibre type shift towards slow/type I fibres occurred in DKO soleus and extensor digitorum longus. MuRF2 and MuRF3-deficient hearts showed decreased systolic and diastolic function. Further analyses revealed an increased expression of the myosin heavy chain isoform beta/slow and disturbed calcium handling as potential causes for the phenotype in DKO hearts. CONCLUSIONS The redundant function of MuRF2 and MuRF3 is important for maintenance of skeletal muscle and cardiac structure and function in vivo.
Collapse
Affiliation(s)
- Dörte Lodka
- Department of Molecular Cardiology, Experimental and Clinical Research Center (ECRC) Max Delbrück Center for Molecular Medicine and Charité Universitätsmedizin Berlin, Campus Buch 13125 Berlin Germany
| | - Aanchal Pahuja
- Institute of Molecular and Cell Physiology Hannover Medical School 30625 Hannover Germany
| | - Cornelia Geers-Knörr
- Institute of Molecular and Cell Physiology Hannover Medical School 30625 Hannover Germany
| | - Renate J Scheibe
- Institute of Physiological Chemistry Hannover Medical School 30625 Hannover Germany
| | - Marcel Nowak
- Department of Molecular Cardiology, Experimental and Clinical Research Center (ECRC) Max Delbrück Center for Molecular Medicine and Charité Universitätsmedizin Berlin, Campus Buch 13125 Berlin Germany; Department of Intracellular Proteolysis Max Delbrück Center for Molecular Medicine 13125 Berlin Germany
| | - Jida Hamati
- Department of Molecular Cardiology, Experimental and Clinical Research Center (ECRC) Max Delbrück Center for Molecular Medicine and Charité Universitätsmedizin Berlin, Campus Buch 13125 Berlin Germany
| | - Clemens Köhncke
- Department of Molecular Muscle Physiology Max Delbrück Center for Molecular Medicine 13125 Berlin Germany
| | - Bettina Purfürst
- Department of Electron Microscopy Max Delbrück Center for Molecular Medicine 13125 Berlin Germany
| | - Tamara Kanashova
- Department of Mass Spectrometry Max Delbrück Center for Molecular Medicine 13125 Berlin Germany
| | - Sibylle Schmidt
- Department of Molecular Cardiology, Experimental and Clinical Research Center (ECRC) Max Delbrück Center for Molecular Medicine and Charité Universitätsmedizin Berlin, Campus Buch 13125 Berlin Germany
| | - David J Glass
- Novartis Institutes for Biomedical Research Cambridge Massachusetts 02139 USA
| | - Ingo Morano
- Department of Molecular Muscle Physiology Max Delbrück Center for Molecular Medicine 13125 Berlin Germany
| | - Arnd Heuser
- Department of Cardiovascular Molecular Genetics Max Delbrück Center for Molecular Medicine 13125 Berlin Germany
| | - Theresia Kraft
- Institute of Molecular and Cell Physiology Hannover Medical School 30625 Hannover Germany
| | - Rhonda Bassel-Duby
- Department of Molecular Biology University of Texas Southwestern Medical Center Dallas Texas 75390-9148 USA
| | - Eric N Olson
- Department of Molecular Biology University of Texas Southwestern Medical Center Dallas Texas 75390-9148 USA
| | - Gunnar Dittmar
- Department of Mass Spectrometry Max Delbrück Center for Molecular Medicine 13125 Berlin Germany
| | - Thomas Sommer
- Department of Intracellular Proteolysis Max Delbrück Center for Molecular Medicine 13125 Berlin Germany
| | - Jens Fielitz
- Department of Molecular Cardiology, Experimental and Clinical Research Center (ECRC) Max Delbrück Center for Molecular Medicine and Charité Universitätsmedizin Berlin, Campus Buch 13125 Berlin Germany; Department of Cardiology Charité Universitätsmedizin Berlin, Campus Virchow 13353 Berlin Germany
| |
Collapse
|
37
|
Kimura T, Mandell M, Deretic V. Precision autophagy directed by receptor regulators - emerging examples within the TRIM family. J Cell Sci 2016; 129:881-91. [PMID: 26906420 DOI: 10.1242/jcs.163758] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Selective autophagy entails cooperation between target recognition and assembly of the autophagic apparatus. Target recognition is conducted by receptors that often recognize tags, such as ubiquitin and galectins, although examples of selective autophagy independent of these tags are emerging. It is less known how receptors cooperate with the upstream autophagic regulators, beyond the well-characterized association of receptors with Atg8 or its homologs, such as LC3B (encoded by MAP1LC3B), on autophagic membranes. The molecular details of the emerging role in autophagy of the family of proteins called TRIMs shed light on the coordination between cargo recognition and the assembly and activation of the principal autophagy regulators. In their autophagy roles, TRIMs act both as receptors and as platforms ('receptor regulators') for the assembly of the core autophagy regulators, such as ULK1 and Beclin 1 in their activated state. As autophagic receptors, TRIMs can directly recognize endogenous or exogenous targets, obviating a need for intermediary autophagic tags, such as ubiquitin and galectins. The receptor and regulatory features embodied within the same entity allow TRIMs to govern cargo degradation in a highly exact process termed 'precision autophagy'.
Collapse
Affiliation(s)
- Tomonori Kimura
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Michael Mandell
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Vojo Deretic
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| |
Collapse
|
38
|
Gralinski LE, Ferris MT, Aylor DL, Whitmore AC, Green R, Frieman MB, Deming D, Menachery VD, Miller DR, Buus RJ, Bell TA, Churchill GA, Threadgill DW, Katze MG, McMillan L, Valdar W, Heise MT, Pardo-Manuel de Villena F, Baric RS. Genome Wide Identification of SARS-CoV Susceptibility Loci Using the Collaborative Cross. PLoS Genet 2015; 11:e1005504. [PMID: 26452100 PMCID: PMC4599853 DOI: 10.1371/journal.pgen.1005504] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 08/15/2015] [Indexed: 01/21/2023] Open
Abstract
New systems genetics approaches are needed to rapidly identify host genes and genetic networks that regulate complex disease outcomes. Using genetically diverse animals from incipient lines of the Collaborative Cross mouse panel, we demonstrate a greatly expanded range of phenotypes relative to classical mouse models of SARS-CoV infection including lung pathology, weight loss and viral titer. Genetic mapping revealed several loci contributing to differential disease responses, including an 8.5Mb locus associated with vascular cuffing on chromosome 3 that contained 23 genes and 13 noncoding RNAs. Integrating phenotypic and genetic data narrowed this region to a single gene, Trim55, an E3 ubiquitin ligase with a role in muscle fiber maintenance. Lung pathology and transcriptomic data from mice genetically deficient in Trim55 were used to validate its role in SARS-CoV-induced vascular cuffing and inflammation. These data establish the Collaborative Cross platform as a powerful genetic resource for uncovering genetic contributions of complex traits in microbial disease severity, inflammation and virus replication in models of outbred populations.
Collapse
Affiliation(s)
- Lisa E. Gralinski
- Department of Epidemiology, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Martin T. Ferris
- Department of Genetics, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - David L. Aylor
- Department of Genetics, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Alan C. Whitmore
- Department of Genetics, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Richard Green
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
| | - Matthew B. Frieman
- Department of Epidemiology, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Damon Deming
- Department of Epidemiology, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Vineet D. Menachery
- Department of Epidemiology, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Darla R. Miller
- Department of Genetics, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Ryan J. Buus
- Department of Genetics, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Timothy A. Bell
- Department of Genetics, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | | | - David W. Threadgill
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas, United States of America
| | - Michael G. Katze
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
| | - Leonard McMillan
- Department of Computer Science, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - William Valdar
- Department of Genetics, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Mark T. Heise
- Department of Genetics, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Fernando Pardo-Manuel de Villena
- Department of Genetics, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Ralph S. Baric
- Department of Epidemiology, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
39
|
He J, Quintana MT, Sullivan J, L Parry T, J Grevengoed T, Schisler JC, Hill JA, Yates CC, Mapanga RF, Essop MF, Stansfield WE, Bain JR, Newgard CB, Muehlbauer MJ, Han Y, Clarke BA, Willis MS. MuRF2 regulates PPARγ1 activity to protect against diabetic cardiomyopathy and enhance weight gain induced by a high fat diet. Cardiovasc Diabetol 2015; 14:97. [PMID: 26242235 PMCID: PMC4526192 DOI: 10.1186/s12933-015-0252-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 06/30/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND In diabetes mellitus the morbidity and mortality of cardiovascular disease is increased and represents an important independent mechanism by which heart disease is exacerbated. The pathogenesis of diabetic cardiomyopathy involves the enhanced activation of PPAR transcription factors, including PPARα, and to a lesser degree PPARβ and PPARγ1. How these transcription factors are regulated in the heart is largely unknown. Recent studies have described post-translational ubiquitination of PPARs as ways in which PPAR activity is inhibited in cancer. However, specific mechanisms in the heart have not previously been described. Recent studies have implicated the muscle-specific ubiquitin ligase muscle ring finger-2 (MuRF2) in inhibiting the nuclear transcription factor SRF. Initial studies of MuRF2-/- hearts revealed enhanced PPAR activity, leading to the hypothesis that MuRF2 regulates PPAR activity by post-translational ubiquitination. METHODS MuRF2-/- mice were challenged with a 26-week 60% fat diet designed to simulate obesity-mediated insulin resistance and diabetic cardiomyopathy. Mice were followed by conscious echocardiography, blood glucose, tissue triglyceride, glycogen levels, immunoblot analysis of intracellular signaling, heart and skeletal muscle morphometrics, and PPARα, PPARβ, and PPARγ1-regulated mRNA expression. RESULTS MuRF2 protein levels increase ~20% during the development of diabetic cardiomyopathy induced by high fat diet. Compared to littermate wildtype hearts, MuRF2-/- hearts exhibit an exaggerated diabetic cardiomyopathy, characterized by an early onset systolic dysfunction, larger left ventricular mass, and higher heart weight. MuRF2-/- hearts had significantly increased PPARα- and PPARγ1-regulated gene expression by RT-qPCR, consistent with MuRF2's regulation of these transcription factors in vivo. Mechanistically, MuRF2 mono-ubiquitinated PPARα and PPARγ1 in vitro, consistent with its non-degradatory role in diabetic cardiomyopathy. However, increasing MuRF2:PPARγ1 (>5:1) beyond physiological levels drove poly-ubiquitin-mediated degradation of PPARγ1 in vitro, indicating large MuRF2 increases may lead to PPAR degradation if found in other disease states. CONCLUSIONS Mutations in MuRF2 have been described to contribute to the severity of familial hypertrophic cardiomyopathy. The present study suggests that the lack of MuRF2, as found in these patients, can result in an exaggerated diabetic cardiomyopathy. These studies also identify MuRF2 as the first ubiquitin ligase to regulate cardiac PPARα and PPARγ1 activities in vivo via post-translational modification without degradation.
Collapse
Affiliation(s)
- Jun He
- Department of Pathology and Laboratory Medicine, University of North Carolina, 111 Mason Farm Road, MBRB 2340B, Chapel Hill, NC, USA. .,General Hospital of Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China.
| | - Megan T Quintana
- Department of Surgery, University of North Carolina, Chapel Hill, NC, USA.
| | - Jenyth Sullivan
- Department of Biology, University of North Carolina, Chapel Hill, NC, USA.
| | - Traci L Parry
- McAllister Heart Institute, University of North Carolina, 111 Mason Farm Road, MBRB 2340B, Chapel Hill, NC, USA.
| | - Trisha J Grevengoed
- Department of Nutrition, University of North Carolina, Chapel Hill, NC, USA.
| | - Jonathan C Schisler
- McAllister Heart Institute, University of North Carolina, 111 Mason Farm Road, MBRB 2340B, Chapel Hill, NC, USA. .,Department of Pharmacology, University of North Carolina, Chapel Hill, NC, USA.
| | - Joseph A Hill
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Cecelia C Yates
- Department of Health Promotions and Development, School of Nursing, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Rudo F Mapanga
- Cardio-Metabolic Research Group (CMRG), Department of Physiological Sciences, Stellenbosch University, Stellenbosch, 7600, South Africa.
| | - M Faadiel Essop
- Cardio-Metabolic Research Group (CMRG), Department of Physiological Sciences, Stellenbosch University, Stellenbosch, 7600, South Africa.
| | | | - James R Bain
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA. .,Division of Endocrinology, Metabolism, and Nutrition, Department of Medicine, Duke University Medical Center, Durham, NC, USA.
| | - Christopher B Newgard
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA. .,Division of Endocrinology, Metabolism, and Nutrition, Department of Medicine, Duke University Medical Center, Durham, NC, USA.
| | - Michael J Muehlbauer
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA.
| | - Yipin Han
- East Chapel Hill High School, Chapel Hill, NC, USA.
| | - Brian A Clarke
- Novartis, Novartis Institutes for BioMedical Research, Inc., 400 Technology Square, Boston, MA, 601-4214, USA.
| | - Monte S Willis
- Department of Pathology and Laboratory Medicine, University of North Carolina, 111 Mason Farm Road, MBRB 2340B, Chapel Hill, NC, USA. .,McAllister Heart Institute, University of North Carolina, 111 Mason Farm Road, MBRB 2340B, Chapel Hill, NC, USA.
| |
Collapse
|
40
|
Quintana MT, He J, Sullivan J, Grevengoed T, Schisler J, Han Y, Hill JA, Yates CC, Stansfield WE, Mapanga RF, Essop MF, Muehlbauer MJ, Newgard CB, Bain JR, Willis MS. Muscle ring finger-3 protects against diabetic cardiomyopathy induced by a high fat diet. BMC Endocr Disord 2015; 15:36. [PMID: 26215257 PMCID: PMC4515942 DOI: 10.1186/s12902-015-0028-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND The pathogenesis of diabetic cardiomyopathy (DCM) involves the enhanced activation of peroxisome proliferator activating receptor (PPAR) transcription factors, including the most prominent isoform in the heart, PPARα. In cancer cells and adipocytes, post-translational modification of PPARs have been identified, including ligand-dependent degradation of PPARs by specific ubiquitin ligases. However, the regulation of PPARs in cardiomyocytes and heart have not previously been identified. We recently identified that muscle ring finger-1 (MuRF1) and MuRF2 differentially inhibit PPAR activities by mono-ubiquitination, leading to the hypothesis that MuRF3 may regulate PPAR activity in vivo to regulate DCM. METHODS MuRF3-/- mice were challenged with 26 weeks 60% high fat diet to induce insulin resistance and DCM. Conscious echocardiography, blood glucose, tissue triglyceride, glycogen levels, immunoblot analysis of intracellular signaling, heart and skeletal muscle morphometrics, and PPARα, PPARβ, and PPARγ1 activities were assayed. RESULTS MuRF3-/- mice exhibited a premature systolic heart failure by 6 weeks high fat diet (vs. 12 weeks in MuRF3+/+). MuRF3-/- mice weighed significantly less than sibling-matched wildtype mice after 26 weeks HFD. These differences may be largely due to resistance to fat accumulation, as MRI analysis revealed MuRF3-/- mice had significantly less fat mass, but not lean body mass. In vitro ubiquitination assays identified MuRF3 mono-ubiquitinated PPARα and PPARγ1, but not PPARβ. CONCLUSIONS These findings suggest that MuRF3 helps stabilize cardiac PPARα and PPARγ1 in vivo to support resistance to the development of DCM. MuRF3 also plays an unexpected role in regulating fat storage despite being found only in striated muscle.
Collapse
Affiliation(s)
- Megan T Quintana
- Department of Surgery, University of North Carolina, Chapel Hill, NC, USA.
| | - Jun He
- Department of Pathology & Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA.
- General Hospital of Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China.
| | - Jenyth Sullivan
- Department of Biology, University of North Carolina, Chapel Hill, NC, USA.
| | - Trisha Grevengoed
- Department of Nutrition, University of North Carolina, Chapel Hill, NC, USA.
| | - Jonathan Schisler
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC, USA.
| | - Yipin Han
- North Carolina State University, Department of Engineering, Raleigh, NC, USA.
| | - Joseph A Hill
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Cecelia C Yates
- Department of Health Promotions and Development, School of Nursing, University of Pittsburgh, Pittsburgh, PA, USA.
| | | | - Rudo F Mapanga
- Cardio-Metabolic Research Group (CMRG), Department of Physiological Sciences, Stellenbosch University, Stellenbosch, 7600, South Africa.
| | - M Faadiel Essop
- Cardio-Metabolic Research Group (CMRG), Department of Physiological Sciences, Stellenbosch University, Stellenbosch, 7600, South Africa.
| | - Michael J Muehlbauer
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA.
| | - Christopher B Newgard
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA.
- Division of Endocrinology, Metabolism, and Nutrition, Department of Medicine, Duke University Medical Center, Durham, NC, USA.
| | - James R Bain
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA.
- Division of Endocrinology, Metabolism, and Nutrition, Department of Medicine, Duke University Medical Center, Durham, NC, USA.
| | - Monte S Willis
- Department of Pathology & Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA.
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
41
|
Abstract
The main function of the heart is to pump blood to the different parts of the organism, a task that is efficiently accomplished through proper electric and metabolic coupling between cardiac cells, ensured by gap junctions (GJ). Cardiomyocytes are the major cell population in the heart, and as cells with low mitotic activity, are highly dependent upon mechanisms of protein degradation. In the heart, both the ubiquitin-proteasome system (UPS) and autophagy participate in the fine-tune regulation of cardiac remodelling and function, either in physiological or pathological conditions. Indeed, besides controlling cardiac signalling pathways, UPS and autophagy have been implicated in the turnover of several myocardial proteins. Degradation of Cx43, the major ventricular GJ protein, has been associated to up-regulation of autophagy at the onset of heart ischemia and ischemia/reperfusion (I/R), which can have profound implications upon cardiac function. In this review, we present recent studies devoted to the involvement of autophagy and UPS in heart homoeostasis, with a particular focus on GJ.
Collapse
|
42
|
Mandell MA, Jain A, Arko-Mensah J, Chauhan S, Kimura T, Dinkins C, Silvestri G, Münch J, Kirchhoff F, Simonsen A, Wei Y, Levine B, Johansen T, Deretic V. TRIM proteins regulate autophagy and can target autophagic substrates by direct recognition. Dev Cell 2014; 30:394-409. [PMID: 25127057 DOI: 10.1016/j.devcel.2014.06.013] [Citation(s) in RCA: 262] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 05/11/2014] [Accepted: 06/17/2014] [Indexed: 12/22/2022]
Abstract
Autophagy, a homeostatic process whereby eukaryotic cells target cytoplasmic cargo for degradation, plays a broad role in health and disease states. Here we screened the TRIM family for roles in autophagy and found that half of TRIMs modulated autophagy. In mechanistic studies, we show that TRIMs associate with autophagy factors and act as platforms assembling ULK1 and Beclin 1 in their activated states. Furthermore, TRIM5α acts as a selective autophagy receptor. Based on direct sequence-specific recognition, TRIM5α delivered its cognate cytosolic target, a viral capsid protein, for autophagic degradation. Thus, our study establishes that TRIMs can function both as regulators of autophagy and as autophagic cargo receptors, and reveals a basis for selective autophagy in mammalian cells.
Collapse
Affiliation(s)
- Michael A Mandell
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, 915 Camino de Salud NE, Albuquerque, NM 87131, USA
| | - Ashish Jain
- Molecular Cancer Research Group, Institute of Medical Biology, University of Tromsø-The Arctic University of Norway, 9037 Tromsø, Norway
| | - John Arko-Mensah
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, 915 Camino de Salud NE, Albuquerque, NM 87131, USA
| | - Santosh Chauhan
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, 915 Camino de Salud NE, Albuquerque, NM 87131, USA
| | - Tomonori Kimura
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, 915 Camino de Salud NE, Albuquerque, NM 87131, USA
| | - Christina Dinkins
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, 915 Camino de Salud NE, Albuquerque, NM 87131, USA
| | - Guido Silvestri
- Yerkes National Primate Research Center, Emory University, 3014 Yerkes, 954 Gatewood Road NE, Atlanta, GA 30329, USA
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, Meyerhofstrasse 1, 89081 Ulm, Germany
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, Meyerhofstrasse 1, 89081 Ulm, Germany
| | - Anne Simonsen
- Department of Biochemistry, Institute of Basic Medical Sciences, University of Oslo, 0317 Oslo, Norway
| | - Yongjie Wei
- Center for Autophagy Research and Howard Hughes Medical Institute, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Beth Levine
- Center for Autophagy Research and Howard Hughes Medical Institute, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Terje Johansen
- Molecular Cancer Research Group, Institute of Medical Biology, University of Tromsø-The Arctic University of Norway, 9037 Tromsø, Norway
| | - Vojo Deretic
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, 915 Camino de Salud NE, Albuquerque, NM 87131, USA.
| |
Collapse
|
43
|
Iyer CC, McGovern VL, Wise DO, Glass DJ, Burghes AHM. Deletion of atrophy enhancing genes fails to ameliorate the phenotype in a mouse model of spinal muscular atrophy. Neuromuscul Disord 2014; 24:436-44. [PMID: 24656734 DOI: 10.1016/j.nmd.2014.02.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 01/16/2014] [Accepted: 02/11/2014] [Indexed: 11/25/2022]
Abstract
Spinal muscular atrophy (SMA) is an autosomal recessive disease causing degeneration of lower motor neurons and muscle atrophy. One therapeutic avenue for SMA is targeting signaling pathways in muscle to ameliorate atrophy. Muscle Atrophy F-box, MAFbx, and Muscle RING Finger 1, MuRF1, are muscle-specific ubiquitin ligases upregulated in skeletal and cardiac muscle during atrophy. Homozygous knock-out of MAFbx or MuRF1 causes muscle sparing in adult mice subjected to atrophy by denervation. We wished to determine whether blockage of the major muscle atrophy pathways by deletion of MAFbx or MuRF1 in a mouse model of SMA would improve the phenotype. Deletion of MAFbx in the Δ7 SMA mouse model had no effect on the weight and the survival of the mice while deletion of MuRF1 was deleterious. MAFbx(-/-)-SMA mice showed a significant alteration in fiber size distribution tending towards larger fibers. In skeletal and cardiac tissue MAFbx and MuRF1 transcripts were upregulated whereas MuRF2 and MuRF3 levels were unchanged in Δ7 SMA mice. We conclude that deletion of the muscle ubiquitin ligases does not improve the phenotype of a Δ7 SMA mouse. Furthermore, it seems unlikely that the beneficial effect of HDAC inhibitors is mediated through inhibition of MAFbx and MuRF1.
Collapse
Affiliation(s)
- Chitra C Iyer
- Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, OH, USA
| | - Vicki L McGovern
- Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, OH, USA
| | - Dawnne O Wise
- Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, OH, USA
| | - David J Glass
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Arthur H M Burghes
- Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, OH, USA; Department of Neurology, The Ohio State University, Columbus, OH, USA; Department of Molecular Genetics, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
44
|
Pizon V, Rybina S, Gerbal F, Delort F, Vicart P, Baldacci G, Karsenti E. MURF2B, a novel LC3-binding protein, participates with MURF2A in the switch between autophagy and ubiquitin proteasome system during differentiation of C2C12 muscle cells. PLoS One 2013; 8:e76140. [PMID: 24124537 PMCID: PMC3790703 DOI: 10.1371/journal.pone.0076140] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 08/24/2013] [Indexed: 01/08/2023] Open
Abstract
The ubiquitin proteasome system and macroautophagy are proteolytic pathways essential in the maintenance of cellular homeostasis during differentiation and remodelling of skeletal muscle. In both pathways, proteins to be degraded are tagged with polyubiquitin. In skeletal muscles, the MURF2 proteins display E3 ubiquitin ligase structure suggesting that they may covalently attach ubiquitin polypeptides to still unknown target proteins. So far only MURF2A isoforms were studied and shown to interact with p62/SQSTM1, a protein implicated in macroautophagic and ubiquitin proteasome system degradations. Here, we analyzed the MURF2B and MURF2A proteins and show that the ratio of the isoforms changes during differentiation of muscle C2C12 cells and that the shift of the isoforms expression follows the sequential activation of autophagic or proteasomal degradation. We also show that MURF2B has a functional domain needed for its interaction with LC3, a protein needed for autophagic vesicles formation. Using specific MURF2 RNAi cells we observed that MURF2A and MURF2B are both needed for the formation of autophagosomes and that in the absence of MURF2B, the cells expressing MURF2A display an activated ubiquitin proteasome system implicated in the degradation of p62/SQSTM1 by UPS. Altogether, our results indicate that MURF2A and MURF2B proteins could participate in the molecular switch between the two ubiquitin degradative pathways.
Collapse
Affiliation(s)
- Véronique Pizon
- University Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, CNRS EAC4413, Paris, France
- * E-mail:
| | - Sofia Rybina
- European Molecular Biology Laboratory, Heidelberg, Germany
| | - Fabien Gerbal
- Université Paris Diderot, Matière et Systèmes Complexes, CNRS UMR 7057, Paris, France
- Université Pierre et Marie Curie, Physics Department-UFR925, Paris, France
| | - Florence Delort
- University Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, CNRS EAC4413, Paris, France
| | - Patrick Vicart
- University Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, CNRS EAC4413, Paris, France
| | - Giuseppe Baldacci
- Université Paris Diderot, CNRS, Institut Jacques Monod, Paris, France
| | - Eric Karsenti
- European Molecular Biology Laboratory, Heidelberg, Germany
| |
Collapse
|
45
|
Weterman MAJ, Barth PG, van Spaendonck-Zwarts KY, Aronica E, Poll-The BT, Brouwer OF, van Tintelen JP, Qahar Z, Bradley EJ, de Wissel M, Salviati L, Angelini C, van den Heuvel L, Thomasse YEM, Backx AP, Nürnberg G, Nürnberg P, Baas F. Recessive MYL2 mutations cause infantile type I muscle fibre disease and cardiomyopathy. ACTA ACUST UNITED AC 2013; 136:282-93. [PMID: 23365102 DOI: 10.1093/brain/aws293] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
A cardioskeletal myopathy with onset and death in infancy, morphological features of muscle type I hypotrophy with myofibrillar disorganization and dilated cardiomyopathy was previously reported in three Dutch families. Here we report the genetic cause of this disorder. Multipoint parametric linkage analysis of six Dutch patients identified a homozygous region of 2.1 Mb on chromosome 12, which was shared between all Dutch patients, with a log of odds score of 10.82. Sequence analysis of the entire linkage region resulted in the identification of a homozygous mutation in the last acceptor splice site of the myosin regulatory light chain 2 gene (MYL2) as the genetic cause. MYL2 encodes a myosin regulatory light chain (MLC-2V). The myosin regulatory light chains bind, together with the essential light chains, to the flexible neck region of the myosin heavy chain in the hexameric myosin complex and have a structural and regulatory role in muscle contraction. The MYL2 mutation results in use of a cryptic splice site upstream of the last exon causing a frameshift and replacement of the last 32 codons by 20 different codons. Whole exome sequencing of an Italian patient with similar clinical features showed compound heterozygosity for two other mutations affecting the same exon of MYL2, also resulting in mutant proteins with altered C-terminal tails. As a consequence of these mutations, the second EF-hand domain is disrupted. EF-hands, assumed to function as calcium sensors, can undergo a conformational change upon binding of calcium that is critical for interactions with downstream targets. Immunohistochemical staining of skeletal muscle tissue of the Dutch patients showed a diffuse and weak expression of the mutant protein without clear fibre specificity, while normal protein was absent. Heterozygous missense mutations in MYL2 are known to cause dominant hypertrophic cardiomyopathy; however, none of the parents showed signs of cardiomyopathy. In conclusion, the mutations in the last exon of MYL2 are responsible for a novel autosomal recessive lethal myosinopathy due to defects changing the C-terminal tail of the ventricular form of the myosin regulatory light chain. We propose 'light chain myopathy' as a name for this MYL2-associated myopathy.
Collapse
Affiliation(s)
- Marian A J Weterman
- Department of Genome Analysis k2-213, Academic Medical Centre Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Cullup T, Kho AL, Dionisi-Vici C, Brandmeier B, Smith F, Urry Z, Simpson MA, Yau S, Bertini E, McClelland V, Al-Owain M, Koelker S, Koerner C, Hoffmann GF, Wijburg FA, Hoedt AET, Rogers C, Manchester D, Miyata R, Hayashi M, Said E, Soler D, Kroisel PM, Windpassinger C, Filloux FM, Al-Kaabi S, Hertecant J, Del Campo M, Buk S, Bodi I, Goebel HH, Sewry CA, Abbs S, Mohammed S, Josifova D, Gautel M, Jungbluth H. Recessive mutations in EPG5 cause Vici syndrome, a multisystem disorder with defective autophagy. Nat Genet 2013; 45:83-7. [PMID: 23222957 PMCID: PMC4012842 DOI: 10.1038/ng.2497] [Citation(s) in RCA: 201] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Accepted: 11/15/2012] [Indexed: 01/07/2023]
Abstract
Vici syndrome is a recessively inherited multisystem disorder characterized by callosal agenesis, cataracts, cardiomyopathy, combined immunodeficiency and hypopigmentation. To investigate the molecular basis of Vici syndrome, we carried out exome and Sanger sequence analysis in a cohort of 18 affected individuals. We identified recessive mutations in EPG5 (previously KIAA1632), indicating a causative role in Vici syndrome. EPG5 is the human homolog of the metazoan-specific autophagy gene epg-5, encoding a key autophagy regulator (ectopic P-granules autophagy protein 5) implicated in the formation of autolysosomes. Further studies showed a severe block in autophagosomal clearance in muscle and fibroblasts from individuals with mutant EPG5, resulting in the accumulation of autophagic cargo in autophagosomes. These findings position Vici syndrome as a paradigm of human multisystem disorders associated with defective autophagy and suggest a fundamental role of the autophagy pathway in the immune system and the anatomical and functional formation of organs such as the brain and heart.
Collapse
Affiliation(s)
- Thomas Cullup
- DNA Laboratory, Guy’s and St. Thomas’ Serco Pathology, Guy’s Hospital, London, UK
| | - Ay L. Kho
- Randall Division of Cell and Molecular Biophysics, King’s College, London, UK
- Cardiovascular Division, King’s College London BHF Centre of Research Excellence, London, UK
| | - Carlo Dionisi-Vici
- Division of Metabolism, Bambino Gesu Children’s Hospital, Istituto di Ricovero e Cure a Carattere Scientifico, Rome, Italy
- Laboratory of Molecular Medicine, Bambino Gesu Children’s Hospital, Istituto di Ricovero e Cure a Carattere Scientifico, Rome, Italy
| | - Birgit Brandmeier
- Randall Division of Cell and Molecular Biophysics, King’s College, London, UK
- Cardiovascular Division, King’s College London BHF Centre of Research Excellence, London, UK
| | - Frances Smith
- DNA Laboratory, Guy’s and St. Thomas’ Serco Pathology, Guy’s Hospital, London, UK
| | - Zoe Urry
- Division of Genetics and Molecular Medicine, King’s College London School of Medicine, Guy’s Hospital, London, UK
| | - Michael A. Simpson
- Division of Genetics and Molecular Medicine, King’s College London School of Medicine, Guy’s Hospital, London, UK
| | - Shu Yau
- DNA Laboratory, Guy’s and St. Thomas’ Serco Pathology, Guy’s Hospital, London, UK
| | - Enrico Bertini
- Laboratory of Molecular Medicine, Bambino Gesu Children’s Hospital, Istituto di Ricovero e Cure a Carattere Scientifico, Rome, Italy
| | - Verity McClelland
- Department of Paediatric Neurology, Evelina Children’s Hospital, Guy’s and St. Thomas’ NHS Foundation Trust, London, UK
| | - Mohammed Al-Owain
- Department of Medical Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
- Alfaisal University, Riyadh, Saudi Arabia
| | - Stefan Koelker
- Division of Inherited Metabolic Diseases, University Children’s Hospital, Heidelberg, Germany
| | - Christian Koerner
- Division of Inherited Metabolic Diseases, University Children’s Hospital, Heidelberg, Germany
| | - Georg F. Hoffmann
- Division of Inherited Metabolic Diseases, University Children’s Hospital, Heidelberg, Germany
| | - Frits A. Wijburg
- Department of Pediatrics, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Amber E. ten Hoedt
- Department of Pediatrics, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | | | - David Manchester
- Clinical Genetics and Metabolism, Department of Pediatrics, University of Colorado School of Medicine, Children’s Hospital Colorado, Aurora, CO, USA
| | - Rie Miyata
- Department of Pediatrics, Tokyo Kita Shakai Hoken Hospital, Tokyo, Japan
| | - Masaharu Hayashi
- Department of Brain Development and Neural Regeneration, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Elizabeth Said
- Section of Medical Genetics, Mater dei Hospital, Msida, Malta
- Department of Anatomy & Cell Biology, University of Malta, Msida, Malta
| | - Doriette Soler
- Department of Paediatrics, Mater dei Hospital, Msida, Malta
| | - Peter M. Kroisel
- Institute of Human Genetics, Medical University of Graz, Austria
| | | | - Francis M. Filloux
- University of Utah School of Medicine, Division of Pediatric Neurology, Salt Lake City, UT, USA
| | | | | | | | - Stefan Buk
- Department of Clinical Neuropathology, Academic Neuroscience Centre, King’s College Hospital, London, UK
| | - Istvan Bodi
- Department of Clinical Neuropathology, Academic Neuroscience Centre, King’s College Hospital, London, UK
| | - Hans-Hilmar Goebel
- Department of Neuropathology, Johannes Gutenberg University Medical Centre, Mainz, Germany
| | - Caroline A. Sewry
- Dubowitz Neuromuscular Centre, Institute of Child Health, University College, London, UK
| | - Stephen Abbs
- DNA Laboratory, Guy’s and St. Thomas’ Serco Pathology, Guy’s Hospital, London, UK
| | | | | | - Mathias Gautel
- Randall Division of Cell and Molecular Biophysics, King’s College, London, UK
- Cardiovascular Division, King’s College London BHF Centre of Research Excellence, London, UK
| | - Heinz Jungbluth
- Laboratory of Molecular Medicine, Bambino Gesu Children’s Hospital, Istituto di Ricovero e Cure a Carattere Scientifico, Rome, Italy
- Clinical Neuroscience Division, IOP, King’s College, London, UK
| |
Collapse
|
47
|
Urban ETR, Bury SD, Barbay HS, Guggenmos DJ, Dong Y, Nudo RJ. Gene expression changes of interconnected spared cortical neurons 7 days after ischemic infarct of the primary motor cortex in the rat. Mol Cell Biochem 2012; 369:267-86. [PMID: 22821175 PMCID: PMC3694431 DOI: 10.1007/s11010-012-1390-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 07/07/2012] [Indexed: 12/11/2022]
Abstract
After cortical injury resulting from stroke, some recovery can occur and may involve spared areas of the cerebral cortex reorganizing to assume functions previously controlled by the damaged cortical areas. No studies have specifically assessed gene expression changes in remote neurons with axonal processes that terminate in the infarcted tissue, i.e., the subset of neurons most likely to be involved in regenerative processes. By physiologically identifying the primary motor area controlling forelimb function in adult rats (caudal forelimb area = CFA), and injecting a retrograde tract-tracer, we labeled neurons within the non-primary motor cortex (rostral forelimb area = RFA) that project to CFA. Then, 7 days after a CFA infarct (n = 6), we used laser capture microdissection techniques to harvest labeled neurons in RFA. Healthy, uninjured rats served as controls (n = 6). Biological interactions and functions of gene profiling were investigated by Affymetrix Microarray, and Ingenuity Pathway Analysis. A total of 143 up- and 128 down-regulated genes showed significant changes (fold change ≥1.3 and p < 0.05). The canonical pathway, "Axonal Guidance Signaling," was overrepresented (p value = 0.002). Significantly overrepresented functions included: branching of neurites, organization of cytoskeleton, dendritic growth and branching, organization of cytoplasm, guidance of neurites, development of cellular protrusions, density of dendritic spines, and shape change (p = 0.000151-0.0487). As previous studies have shown that spared motor areas are important in recovery following injury to the primary motor area, the results suggest that these gene expression changes in remote, interconnected neurons may underlie reorganization and recovery mechanisms.
Collapse
Affiliation(s)
- Edward T. R. Urban
- Department of Molecular & Integrative Physiology, Kansas University Medical Center, 3901 Rainbow Boulevard, Mail Stop 3043, Kansas City, KS 66160, USA. Landon Center on Aging, Kansas University Medical Center, 3901 Rainbow Boulevard, Mail Stop 1005, Kansas City, KS 66160, USA
| | - Scott D. Bury
- Landon Center on Aging, Kansas University Medical Center, 3901 Rainbow Boulevard, Mail Stop 1005, Kansas City, KS 66160, USA
| | - H. Scott Barbay
- Landon Center on Aging, Kansas University Medical Center, 3901 Rainbow Boulevard, Mail Stop 1005, Kansas City, KS 66160, USA
| | - David J. Guggenmos
- Department of Molecular & Integrative Physiology, Kansas University Medical Center, 3901 Rainbow Boulevard, Mail Stop 3043, Kansas City, KS 66160, USA. Landon Center on Aging, Kansas University Medical Center, 3901 Rainbow Boulevard, Mail Stop 1005, Kansas City, KS 66160, USA
| | - Yafeng Dong
- Department of Obstetrics and Gynecology, Kansas University Medical Center, 3901 Rainbow Boulevard, Mail Stop 2028, Kansas City, KS 66160, USA
| | - Randolph J. Nudo
- Department of Molecular & Integrative Physiology, Kansas University Medical Center, 3901 Rainbow Boulevard, Mail Stop 3043, Kansas City, KS 66160, USA. Landon Center on Aging, Kansas University Medical Center, 3901 Rainbow Boulevard, Mail Stop 1005, Kansas City, KS 66160, USA. Intellectual & Developmental Disabilities Research Center, Kansas University Medical Center, 3901 Rainbow Boulevard, Mail Stop 3051, Kansas City, KS 66160, USA
| |
Collapse
|
48
|
Kho AL, Perera S, Alexandrovich A, Gautel M. The sarcomeric cytoskeleton as a target for pharmacological intervention. Curr Opin Pharmacol 2012; 12:347-54. [DOI: 10.1016/j.coph.2012.03.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Revised: 03/14/2012] [Accepted: 03/16/2012] [Indexed: 01/10/2023]
|
49
|
Lange S, Perera S, Teh P, Chen J. Obscurin and KCTD6 regulate cullin-dependent small ankyrin-1 (sAnk1.5) protein turnover. Mol Biol Cell 2012; 23:2490-504. [PMID: 22573887 PMCID: PMC3386213 DOI: 10.1091/mbc.e12-01-0052] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Small ankyrin-1 isoform 5 (sAnk1.5) turnover is regulated by posttranslational modification (ubiquitylation, neddylation, and acetylation), the presence of obscurin, and KCTD6 (a novel tissue-specific interaction partner). KCTD6 links sAnk1.5 to cullin-3. The absence of obscurin results in translocation of sAnk1.5/KCTD6 to the Z-disk and loss of sAnk1.5 on the protein level. Protein turnover through cullin-3 is tightly regulated by posttranslational modifications, the COP9 signalosome, and BTB/POZ-domain proteins that link cullin-3 to specific substrates for ubiquitylation. In this paper, we report how potassium channel tetramerization domain containing 6 (KCTD6) represents a novel substrate adaptor for cullin-3, effectively regulating protein levels of the muscle small ankyrin-1 isoform 5 (sAnk1.5). Binding of sAnk1.5 to KCTD6, and its subsequent turnover is regulated through posttranslational modification by nedd8, ubiquitin, and acetylation of C-terminal lysine residues. The presence of the sAnk1.5 binding partner obscurin, and mutation of lysine residues increased sAnk1.5 protein levels, as did knockdown of KCTD6 in cardiomyocytes. Obscurin knockout muscle displayed reduced sAnk1.5 levels and mislocalization of the sAnk1.5/KCTD6 complex. Scaffolding functions of obscurin may therefore prevent activation of the cullin-mediated protein degradation machinery and ubiquitylation of sAnk1.5 through sequestration of sAnk1.5/KCTD6 at the sarcomeric M-band, away from the Z-disk–associated cullin-3. The interaction of KCTD6 with ankyrin-1 may have implications beyond muscle for hereditary spherocytosis, as KCTD6 is also present in erythrocytes, and erythrocyte ankyrin isoforms contain its mapped minimal binding site.
Collapse
Affiliation(s)
- Stephan Lange
- School of Medicine, University of California, San Diego, La Jolla, CA 92093-0613, USA.
| | | | | | | |
Collapse
|
50
|
Developmental regulation of MURF E3 ubiquitin ligases in skeletal muscle. J Muscle Res Cell Motil 2012; 33:107-22. [PMID: 22426552 PMCID: PMC3353113 DOI: 10.1007/s10974-012-9288-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Accepted: 03/01/2012] [Indexed: 12/23/2022]
Abstract
The striated muscle-specific tripartite motif (TRIM) proteins TRIM63/MURF1, TRIM55/MURF2 and TRIM54/MURF3 can function as E3 ubiquitin ligases in ubiquitin-mediated muscle protein turnover. Despite the well-characterised role of MURF1 in skeletal muscle atrophy, the dynamics of MURF isogene expression in the development and early postnatal adaptation of skeletal muscle is unknown. Here, we show that MURF2 is the isogene most highly expressed in embryonic skeletal muscle at E15.5, with the 50 kDa A isoform predominantly expressed. MURF1 and MURF3 are upregulated only postnatally. Knockdown of MURF2 p50A by isoform-specific siRNA results in delayed myogenic differentiation and myotube formation in vitro, with perturbation of the stable, glutamylated microtubule population. This underscores that MURF2 plays an important role in the earliest stages of skeletal muscle differentiation and myofibrillogenesis. During further development, there is a shift towards the 60 kDa A isoform, which dominates postnatally. Analysis of the fibre-type expression shows that MURF2 A isoforms are predominantly slow-fibre associated, whilst MURF1 is largely excluded from these fibres, and MURF3 is ubiquitously distributed in both type I and II fibres.
Collapse
|