1
|
Gong X, Xu L, Cai P. Friend or foe of tripartite motif-containing protein 21 in cardiovascular disease: A review. Int J Biol Macromol 2025; 308:142682. [PMID: 40164260 DOI: 10.1016/j.ijbiomac.2025.142682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 04/02/2025]
Abstract
As an E3 ubiquitin ligase and an Fc receptor, tripartite motif-containing protein 21 (TRIM21) plays a crucial role in immune defense, signal transduction, and cellular regulation. TRIM21 is widely expressed in various tissues, but it is particularly abundant in cardiovascular tissues and is involved in the pathogenesis of various cardiovascular diseases (CVDs). However, although TRIM21 is involved in the regulation of several key molecular pathways in the immune system, its specific role in CVD remains unclear. In this review, we comprehensively summarize the regulatory role of TRIM21 in signaling pathways and discuss the function of TRIM21 in CVD, to provide a systematic understanding of this important protein in CVD and offer insights for further research into the pathogenesis of CVD and its potential applications.
Collapse
Affiliation(s)
- Xiangmei Gong
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Xu
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pengcheng Cai
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
2
|
Liu X, Shi X, Zhao H, Wang C. Exploring the molecular mechanisms of comorbidity of myocardial infarction and anxiety disorders by combining multiple data sets with in vivo experimental validation. Int Immunopharmacol 2025; 146:113852. [PMID: 39733641 DOI: 10.1016/j.intimp.2024.113852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/25/2024] [Accepted: 12/10/2024] [Indexed: 12/31/2024]
Abstract
BACKGROUND The incidence of comorbidity between myocardial infarction (MI) and anxiety disorders is increasing. However, the biological association between them has not been fully understood. OBJECTIVE This study aims to investigate the molecular mechanisms of comorbidity between MI and anxiety disorders and to predict their key genes and potential therapeutic drugs. METHODS We searched Gene Expression Omnibus databases and performed differential analyses using the limma package to identify the functional enrichment of differential genes. Next, we constructed regulatory networks to investigate the relationship between hub genes and autophagy, ferroptosis, and immunity. Furthermore, we predicted transcription factors by R package, constructed a miRNA network, performed the single-cell analysis of key gene expression, and predicted drug targeting of differential genes using the Connectivity Map database. RESULTS The datasets for MI and anxiety disorders were analyzed for up and down-regulated differential genes, resulting in 35 intersecting differential genes. The top 10 feature genes from each dataset were intersected using Random Forest, resulting in the identification of three intersecting genes: STK17B, AKIRIN2, and WDR77. Validation of the above key genes was carried out by in vitro experiments. We examined the gene expression of STK17B, WDR77 and AKIRIN2 in the hippocampus and myocardial infarction border zone respectively by qPCR and WB, and the results confirmed that the above are the key genes for myocardial infarction and anxiety. There is a significant correlation between the comorbidity mechanism of myocardial infarction and anxiety disorders with ferroptosis and immunity. The construction of the miRNA network revealed that miR-205 and let-7 had higher average connectivity among the three hub genes. The single-cell analysis revealed significant expression of key genes in Endothelial cells, Cardiomyocytes, Macrophages, and Fibroblasts datasets. Cd274 showed a higher correlation with key genes in myocardial infarction and anxiety disorders. CONCLUSION Validation by multiple datasets and in vitro experiments showed that STK17B, AKIRIN2, and WDR77 are the key genes in the comorbidity of myocardial infarction and anxiety disorders, and ferroptosis and immunity are the key links in the comorbidity mechanism of myocardial infarction and anxiety disorders.
Collapse
Affiliation(s)
- Xiang Liu
- Beijing University of Chinese Medicine, Beijing, China.
| | - Xiaojun Shi
- Beijing University of Chinese Medicine, Beijing, China
| | - Haibin Zhao
- Dongfang Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Chao Wang
- Dongfang Hospital of Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
3
|
Neeman-Egozi S, Livneh I, Dolgopyat I, Nussinovitch U, Milman H, Cohen N, Eisen B, Ciechanover A, Binah O. Stress-Induced Proteasome Sub-Cellular Translocation in Cardiomyocytes Causes Altered Intracellular Calcium Handling and Arrhythmias. Int J Mol Sci 2024; 25:4932. [PMID: 38732146 PMCID: PMC11084437 DOI: 10.3390/ijms25094932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
The ubiquitin-proteasome system (UPS) is an essential mechanism responsible for the selective degradation of substrate proteins via their conjugation with ubiquitin. Since cardiomyocytes have very limited self-renewal capacity, as they are prone to protein damage due to constant mechanical and metabolic stress, the UPS has a key role in cardiac physiology and pathophysiology. While altered proteasomal activity contributes to a variety of cardiac pathologies, such as heart failure and ischemia/reperfusion injury (IRI), the environmental cues affecting its activity are still unknown, and they are the focus of this work. Following a recent study by Ciechanover's group showing that amino acid (AA) starvation in cultured cancer cell lines modulates proteasome intracellular localization and activity, we tested two hypotheses in human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs, CMs): (i) AA starvation causes proteasome translocation in CMs, similarly to the observation in cultured cancer cell lines; (ii) manipulation of subcellular proteasomal compartmentalization is associated with electrophysiological abnormalities in the form of arrhythmias, mediated via altered intracellular Ca2+ handling. The major findings are: (i) starving CMs to AAs results in proteasome translocation from the nucleus to the cytoplasm, while supplementation with the aromatic amino acids tyrosine (Y), tryptophan (W) and phenylalanine (F) (YWF) inhibits the proteasome recruitment; (ii) AA-deficient treatments cause arrhythmias; (iii) the arrhythmias observed upon nuclear proteasome sequestration(-AA+YWF) are blocked by KB-R7943, an inhibitor of the reverse mode of the sodium-calcium exchanger NCX; (iv) the retrograde perfusion of isolated rat hearts with AA starvation media is associated with arrhythmias. Collectively, our novel findings describe a newly identified mechanism linking the UPS to arrhythmia generation in CMs and whole hearts.
Collapse
Affiliation(s)
- Shunit Neeman-Egozi
- Department of Physiology, Biophysics and Systems Biology, Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 3190601, Israel; (S.N.-E.); (B.E.)
| | - Ido Livneh
- The Rappaport-Technion Integrated Cancer Center (R-TICC) and The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa 319060, Israel; (I.L.); (N.C.)
| | - Irit Dolgopyat
- Department of Physiology, Biophysics and Systems Biology, Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 3190601, Israel; (S.N.-E.); (B.E.)
| | - Udi Nussinovitch
- Department of Cardiology, Edith Wolfson Medical Center, Holon 5822012, Israel
- The Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Helena Milman
- Department of Physiology, Biophysics and Systems Biology, Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 3190601, Israel; (S.N.-E.); (B.E.)
| | - Nadav Cohen
- The Rappaport-Technion Integrated Cancer Center (R-TICC) and The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa 319060, Israel; (I.L.); (N.C.)
| | - Binyamin Eisen
- Department of Physiology, Biophysics and Systems Biology, Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 3190601, Israel; (S.N.-E.); (B.E.)
| | - Aaron Ciechanover
- The Rappaport-Technion Integrated Cancer Center (R-TICC) and The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa 319060, Israel; (I.L.); (N.C.)
| | - Ofer Binah
- Department of Physiology, Biophysics and Systems Biology, Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 3190601, Israel; (S.N.-E.); (B.E.)
| |
Collapse
|
4
|
You JR, Wen ZJ, Tian JW, Lv XB, Li R, Li SP, Xin H, Li PF, Zhang YF, Zhang R. Crosstalk between ubiquitin ligases and ncRNAs drives cardiovascular disease progression. Front Immunol 2024; 15:1335519. [PMID: 38515760 PMCID: PMC10954775 DOI: 10.3389/fimmu.2024.1335519] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 02/26/2024] [Indexed: 03/23/2024] Open
Abstract
Cardiovascular diseases (CVDs) are multifactorial chronic diseases and have the highest rates of morbidity and mortality worldwide. The ubiquitin-proteasome system (UPS) plays a crucial role in posttranslational modification and quality control of proteins, maintaining intracellular homeostasis via degradation of misfolded, short-lived, or nonfunctional regulatory proteins. Noncoding RNAs (ncRNAs, such as microRNAs, long noncoding RNAs, circular RNAs and small interfering RNAs) serve as epigenetic factors and directly or indirectly participate in various physiological and pathological processes. NcRNAs that regulate ubiquitination or are regulated by the UPS are involved in the execution of target protein stability. The cross-linked relationship between the UPS, ncRNAs and CVDs has drawn researchers' attention. Herein, we provide an update on recent developments and perspectives on how the crosstalk of the UPS and ncRNAs affects the pathological mechanisms of CVDs, particularly myocardial ischemia/reperfusion injury, myocardial infarction, cardiomyopathy, heart failure, atherosclerosis, hypertension, and ischemic stroke. In addition, we further envision that RNA interference or ncRNA mimics or inhibitors targeting the UPS can potentially be used as therapeutic tools and strategies.
Collapse
Affiliation(s)
- Jia-Rui You
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, China
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Zeng-Jin Wen
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, China
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Jia-Wei Tian
- Department of Emergency Internal Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiao-Bing Lv
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, China
| | - Rong Li
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, China
| | - Shu-Ping Li
- Department of Cardiology, The Affiliated Qingdao Third People’s Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, China
| | - Hui Xin
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, China
| | - Pei-Feng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Yin-Feng Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Rui Zhang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
5
|
Huang Y, Wei C, Li P, Shao Y, Wang M, Wang F, Niu G, Sun K, Zhang Q, Gou Z, Yan X. FGF21 protects against doxorubicin-induced cardiotoxicity by inhibiting connexin 43 ubiquitination. Free Radic Biol Med 2023; 208:748-758. [PMID: 37774805 DOI: 10.1016/j.freeradbiomed.2023.09.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/26/2023] [Accepted: 09/26/2023] [Indexed: 10/01/2023]
Abstract
BACKGROUND Fibroblast growth factor 21 (FGF21) regulates glycolipid metabolism and insulin homeostasis and acts as a cardioprotective factor by protecting against myocardial ischemia/reperfusion injury, hypertension, and vascular dysfunction. FGF21 has been reported to prevent Doxorubicin (Dox)-induced cardiotoxicity, and the related signaling pathway is worthy of further study. Connexin43 (Cx43) protein was reduced by Dox treatment, especially low phosphorylated form of Cx43. Thus the aim of study is to explore the protection effect of FGF21 on Dox induced cardiotoxicity by improving the expression of Cx43 and the involved signaling pathway. METHODS AND RESULTS FGF21 inhibited apoptosis in Dox-treated mice and cardiomyocytes. FGF21 increased the levels of connexin43 phosphorylated at serine (S) 282 (p-Cx43 S282) and total Cx43 to inhibit Dox-induced apoptosis. By RNA sequencing, we found that deubiquitinase monocyte chemoattractant protein-induced protein 1 (MCPIP1) expression was increased by FGF21. We further found that FGF21 induced the phosphorylation of fibroblast growth factor receptor 1 (FGFR1), extracellular signal-regulated kinase 1 and 2 (Erk1/2), and Elk. Phosphorylated Elk translocated to the nucleus and increased the expression of MCPIP1. Then, MCPIP1 bound neural precursor cell expressed developmentally downregulated protein 4 (Nedd4), an E3 ubiquitination ligase, as shown by co-immunoprecipitation (Co-IP), and suppressed Cx43 ubiquitination and degradation, competitively inhibiting the binding of Cx43 with Nedd4. Thus Nedd4 could not bind and ubiquitinate Cx43, leading to the up-regulation of Cx43 and phosphorylation of Cx43 at S282. CONCLUSIONS FGF21 inhibited the effects of Dox on cardiomyocytes by elevating the phosphorylation of Cx43 at S282 and total Cx43 expression. This study suggests a previously unknown mechanism for the FGF21-mediated enhancement of cardiomyocyte survival and provides an effective approach to protect against the adverse cardiac effects of Dox.
Collapse
Affiliation(s)
- Ying Huang
- Center for Cardiovascular Disease, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, 215008, PR China
| | - Chenchen Wei
- Center for Cardiovascular Disease, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, 215008, PR China
| | - Ping Li
- Center for Cardiovascular Disease, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, 215008, PR China
| | - Yaqing Shao
- Center for Cardiovascular Disease, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, 215008, PR China
| | - Min Wang
- Center for Cardiovascular Disease, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, 215008, PR China
| | - Feng Wang
- Center for Cardiovascular Disease, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, 215008, PR China; Department of Pharmacology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, 215008, PR China
| | - Guanghao Niu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou, Jiangsu, 215000, PR China
| | - Kangyun Sun
- Center for Cardiovascular Disease, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, 215008, PR China
| | - Qian Zhang
- Department of Pharmacology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, 215008, PR China.
| | - Zhongshan Gou
- Center for Cardiovascular Disease, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, 215008, PR China.
| | - Xinxin Yan
- Center for Cardiovascular Disease, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, 215008, PR China; Department of Pharmacology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, 215008, PR China.
| |
Collapse
|
6
|
Xie Y, Gao R, Gao Y, Dong Z, Ge J. 11S Proteasome Activator REGγ Promotes Aortic Dissection by Inhibiting RBM3 (RNA Binding Motif Protein 3) Pathway. Hypertension 2023; 80:125-137. [PMID: 36330811 DOI: 10.1161/hypertensionaha.122.19618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Aortic dissection (AD) is a life-threatening cardiovascular disorder with high mortality and lacking underlying mechanisms or effective treatments. REGγ, the 11S proteasome activator known to promote the degradation of cellular proteins in a ubiquitin- and ATP-independent manner, emerges as a new regulator in the cardiovascular system. METHODS Using β-aminopropionitrile (BAPN)-subjected REGγ knockout AD mice and Ang II (angiotensin II)-treated REGγ deficiency vascular smooth muscle cells (VSMCs) to explore the effect of REGγ in AD progression. RESULTS REGγ was upregulated in mouse aorta of β-aminopropionitrile-induced AD model in vivo and Ang II-treated VSMCs in vitro. REGγ deficiency ameliorated AD progression in β-aminopropionitrile-induced mice by protecting against the switch in VSMCs from contractile to synthetic phenotype through suppressing RBM3 (RNA-binding motif protein 3) decay. Mechanically, REGγ interacted with and degraded the RNA-binding protein RBM3 directly, leading to decreased mRNA stability, lowered expression and transcriptional activity of transcription factor SRF (serum response factor), subsequently reduced transcription of VSMCs-specific contractile genes, α-SMA (alpha-smooth muscle actin) and SM22α (smooth muscle 22 alpha), caused the switch in VSMCs from contractile to synthetic phenotype and associated AD progression. Ablation of endogenous SRF or RBM3, or overexpressing exogenous RBM3 in VSMCs significantly blocked or reestablished the REGγ-dependent action on VSMCs phenotypic switch of Ang II stimulation in vitro. Furthermore, exogenously introducing RBM3 improved the switch in VSMCs from contractile to synthetic phenotype and associated AD features caused by REGγ in vivo. CONCLUSIONS Our results demonstrated that REGγ promoted the switch in VSMCs from contractile to synthetic phenotype and AD progression by inhibiting RBM3-SRF pathway, indicated that modulating REGγ-proteasome activity may be a potential therapeutic approach for AD-associated cardiovascular dysfunction.
Collapse
Affiliation(s)
- Yifan Xie
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China (Y.X., R.G., Y.G., Z.D., J.G.).,Shanghai Institute of Cardiovascular Diseases' Shanghai' China (Y.X., R.G., Y.G., Z.D., J.G.).,Institutes of Biomedical Science, Fudan University, Shanghai, China (Y.X., J.G.)
| | - Rifeng Gao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China (Y.X., R.G., Y.G., Z.D., J.G.).,Shanghai Institute of Cardiovascular Diseases' Shanghai' China (Y.X., R.G., Y.G., Z.D., J.G.)
| | - Yang Gao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China (Y.X., R.G., Y.G., Z.D., J.G.).,Shanghai Institute of Cardiovascular Diseases' Shanghai' China (Y.X., R.G., Y.G., Z.D., J.G.)
| | - Zheng Dong
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China (Y.X., R.G., Y.G., Z.D., J.G.).,Shanghai Institute of Cardiovascular Diseases' Shanghai' China (Y.X., R.G., Y.G., Z.D., J.G.)
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China (Y.X., R.G., Y.G., Z.D., J.G.).,Shanghai Institute of Cardiovascular Diseases' Shanghai' China (Y.X., R.G., Y.G., Z.D., J.G.).,Institutes of Biomedical Science, Fudan University, Shanghai, China (Y.X., J.G.)
| |
Collapse
|
7
|
Papanagnou E, Gumeni S, Sklirou AD, Rafeletou A, Terpos E, Keklikoglou K, Kastritis E, Stamatelopoulos K, Sykiotis GP, Dimopoulos MA, Trougakos IP. Autophagy activation can partially rescue proteasome dysfunction-mediated cardiac toxicity. Aging Cell 2022; 21:e13715. [PMID: 36259256 PMCID: PMC9649605 DOI: 10.1111/acel.13715] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 08/01/2022] [Accepted: 08/31/2022] [Indexed: 01/25/2023] Open
Abstract
The ubiquitin-proteasome pathway and its functional interplay with other proteostatic and/or mitostatic modules are crucial for cell viability, especially in post-mitotic cells like cardiomyocytes, which are constantly exposed to proteotoxic, metabolic, and mechanical stress. Consistently, treatment of multiple myeloma patients with therapeutic proteasome inhibitors may induce cardiac failure; yet the effects promoted by heart-targeted proteasome dysfunction are not completely understood. We report here that heart-targeted proteasome knockdown in the fly experimental model results in increased proteome instability and defective mitostasis, leading to disrupted cardiac activity, systemic toxicity, and reduced longevity. These phenotypes were partially rescued by either heart targeted- or by dietary restriction-mediated activation of autophagy. Supportively, activation of autophagy by Rapamycin or Metformin administration in flies treated with proteasome inhibitors reduced proteome instability, partially restored mitochondrial function, mitigated cardiotoxicity, and improved flies' longevity. These findings suggest that autophagic inducers represent a novel promising intervention against proteasome inhibitor-induced cardiovascular complications.
Collapse
Affiliation(s)
- Eleni‐Dimitra Papanagnou
- Department of Cell Biology and Biophysics, Faculty of BiologyNational and Kapodistrian University of AthensAthensGreece
| | - Sentiljana Gumeni
- Department of Cell Biology and Biophysics, Faculty of BiologyNational and Kapodistrian University of AthensAthensGreece
| | - Aimilia D. Sklirou
- Department of Cell Biology and Biophysics, Faculty of BiologyNational and Kapodistrian University of AthensAthensGreece
| | - Alexandra Rafeletou
- Department of Cell Biology and Biophysics, Faculty of BiologyNational and Kapodistrian University of AthensAthensGreece
| | - Evangelos Terpos
- Department of Clinical Therapeutics, School of MedicineNational and Kapodistrian University of AthensAthensGreece
| | - Kleoniki Keklikoglou
- Institute of Marine Biology, Biotechnology and Aquaculture, Hellenic Centre for Marine Research (HCMR)CreteGreece,Biology DepartmentUniversity of CreteHeraklionGreece
| | - Efstathios Kastritis
- Department of Clinical Therapeutics, School of MedicineNational and Kapodistrian University of AthensAthensGreece
| | - Kimon Stamatelopoulos
- Department of Clinical Therapeutics, School of MedicineNational and Kapodistrian University of AthensAthensGreece
| | - Gerasimos P. Sykiotis
- Service of Endocrinology, Diabetology and MetabolismLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Meletios A. Dimopoulos
- Department of Clinical Therapeutics, School of MedicineNational and Kapodistrian University of AthensAthensGreece
| | - Ioannis P. Trougakos
- Department of Cell Biology and Biophysics, Faculty of BiologyNational and Kapodistrian University of AthensAthensGreece
| |
Collapse
|
8
|
Chen T, Wang Y, Wang YH, Hang CH. The Mfn1-βIIPKC Interaction Regulates Mitochondrial Dysfunction via Sirt3 Following Experimental Subarachnoid Hemorrhage. Transl Stroke Res 2022; 13:845-857. [PMID: 35192161 DOI: 10.1007/s12975-022-00999-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 01/21/2022] [Accepted: 02/16/2022] [Indexed: 10/19/2022]
Abstract
Neuronal injury following subarachnoid hemorrhage (SAH) has been shown to be associated with mitochondrial dysfunction and oxidative stress. βIIPKC, a subtype of protein kinase C (PKC), accumulates on the mitochondrial outer membrane and phosphorylates mitofusin 1 (Mfn1) at serine 86. Here, we investigated the role of Mfn1-βIIPKC interaction in brain damage and neurological function in both in vivo and in vitro experimental SAH models. The expression of βIIPKC protein and the interaction of Mfn1-βIIPKC were found to be increased after OxyHb treatment in primary cultured cortical neurons and were also observed in the brain following SAH in rats. Treatment with the βIIPKC inhibitor βIIV5-3 or SAMβA, a peptide that selectively antagonizes Mfn1-βIIPKC association, significantly attenuated the OxyHb-induced neuronal injury and apoptosis. These protective effects were accompanied by inhibited mitochondrial dysfunction and preserved mitochondrial biogenesis. The results of western blot showed that βIIV5-3 or SAMβA markedly increased the expression of Sirt3 and enhanced the activities of its downstream mitochondrial antioxidant enzymes in OxyHb-treated neurons. Knockdown of Sirt3 via specific targeted small interfering RNA (siRNA) partially prevented the βIIV5-3- or SAMβA-induced protection and antioxidative effects. In addition, treatment with βIIV5-3 or SAMβA in vivo was found to obviously reduce brain edema, alleviate neuroinflammation, and preserve neurological function after experimental SAH in rats. In congruent with in vitro data, the protection induced by βIIV5-3 or SAMβA was reduced by Sirt3 knockdown in vivo. In summary, our present results showed that blocking Mfn1-βIIPKC interaction protects against brain damage and mitochondrial dysfunction via Sirt3 following experimental SAH.
Collapse
Affiliation(s)
- Tao Chen
- Department of Neurosurgery, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210000, Jiangsu, China
- Department of Neurosurgery, The 904Th Hospital of PLA, Medical School of Anhui Medical University, Wuxi, 214044, Jiangsu, China
| | - Yue Wang
- Department of Neurosurgery, The 904Th Hospital of PLA, Medical School of Anhui Medical University, Wuxi, 214044, Jiangsu, China
| | - Yu-Hai Wang
- Department of Neurosurgery, The 904Th Hospital of PLA, Medical School of Anhui Medical University, Wuxi, 214044, Jiangsu, China.
| | - Chun-Hua Hang
- Department of Neurosurgery, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210000, Jiangsu, China.
| |
Collapse
|
9
|
Lu Z, Deng M, Ma G, Chen L. TRIM38 protects H9c2 cells from hypoxia/reoxygenation injury via the TRAF6/TAK1/NF- κB signalling pathway. PeerJ 2022; 10:e13815. [PMID: 36061751 PMCID: PMC9435518 DOI: 10.7717/peerj.13815] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 07/08/2022] [Indexed: 01/18/2023] Open
Abstract
Tripartite motif (TRIM) 38 is a ubiquitin E3 protein ligase that is involved in various intracellular physiological processes. However, the role of TRIM38 in myocardial ischaemia/reperfusion (I/R) injury remains to be elucidated. We aimed to establish an in vitro cellular hypoxia/reperfusion (H/R) model to explore the role and potential mechanisms of TRIM38 in H9c2, a rat cardiomyoblast cell line. Recombinant adenoviruses for silencing or overexpressing TRIM38 were constructed and transfected into H9c2 cells. Western blotanalysisshowed that TRIM38 expression was significantly decreased after H/R injury. Functionally, TRIM38 expression relieved inflammatory responses and oxidative stress, and inhibited H/R-induced apoptosis in H9c2 cells. Mechanistically, TRIM38 overexpression inhibited H/R-induced transforming growth factor beta-activated kinase 1 (TAK1)/nuclear factor-kappa B (NF-κB) pathway activity in H9c2 cells. The opposite results were observed after TRIM38 knockdown. Furthermore, H/R-induced injury aggravated by TRIM38 deficiency in H9c2 cells was reversed upon treatment with 5Z-7-oxozeaenol, a TAK1 inhibitor. Therefore, TRIM38 reduction attenuated the anti-apoptotic capacity and anti-inflammatory potential of H/R-stimulated H9c2 cells by activating the TAK1/NF-κB signalling pathway. Specifically, TRIM38 alleviated H/R-induced H9c2 cell injury by promoting TNF receptor-associated factor 6 degradation, which led to the inactivation of the TAK1/NF-κB signalling pathway. Thus, our study provides new insights into the molecular mechanisms underlying H/R-induced myocardial injuries.
Collapse
Affiliation(s)
- Zhengri Lu
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, China
| | - Mengen Deng
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, China
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, China
| | - Lijuan Chen
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, China,Department of Cardiology, Nanjing Lishui People’s Hospital, Zhongda Hospital Lishui Branch, Nanjing, Jiangsu, China
| |
Collapse
|
10
|
The proteasome activator REGγ promotes diabetic endothelial impairment by inhibiting HMGA2-GLUT1 pathway. Transl Res 2022; 246:33-48. [PMID: 35367424 DOI: 10.1016/j.trsl.2022.03.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 11/22/2022]
Abstract
Diabetic vascular endothelial impairment is one of the main causes of death in patients with diabetes lacking adequately defined mechanisms or effective treatments. REGγ, the 11S proteasome activator known to promote the degradation of cellular proteins in a ubiquitin- and ATP-independent manner, emerges as a new regulator in the cardiovascular system. Here, we found that REGγ was upregulated in streptozocin (STZ)-induced diabetic mouse aortic endothelium in vivo and high glucose (HG)-treated vascular endothelial cells (ECs) in vitro. REGγ deficiency ameliorated endothelial impairment in STZ-induced diabetic mice by protecting against a decline in cellular glucose uptake and associated vascular ECs dysfunction by suppressing high mobility group AT-hook 2 (HMGA2) decay. Mechanically, REGγ interacted with and degraded the transcription factor HMGA2 directly, leading to decreased HMGA2 transcriptional activity, subsequently lowered expression of glucose transporter type 1 (GLUT1), and reduced cellular glucose uptake, vascular endothelial dysfunction, and impaired diabetic endothelium. Ablation of endogenous GLUT1 or HMGA2 or overexpressing exogenous HMGA2 in vascular ECs significantly blocked or reestablished the REGγ-dependent action on cellular glucose uptake and vascular endothelial functions of HG stimulation in vitro. Furthermore, exogenously introducing HMGA2 improved diabetic mice endothelial impairment features caused by REGγ in vivo, thereby substantiating a REGγ-HMGA2-GLUT1 pathway in diabetic endothelial impairment. Our findings indicate that modulating REGγ-proteasome activity may be a potential therapeutic approach for diabetic disorders with endothelial impairment.
Collapse
|
11
|
The Role of HECT-Type E3 Ligase in the Development of Cardiac Disease. Int J Mol Sci 2021; 22:ijms22116065. [PMID: 34199773 PMCID: PMC8199989 DOI: 10.3390/ijms22116065] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 05/26/2021] [Accepted: 06/01/2021] [Indexed: 12/12/2022] Open
Abstract
Despite advances in medicine, cardiac disease remains an increasing health problem associated with a high mortality rate. Maladaptive cardiac remodeling, such as cardiac hypertrophy and fibrosis, is a risk factor for heart failure; therefore, it is critical to identify new therapeutic targets. Failing heart is reported to be associated with hyper-ubiquitylation and impairment of the ubiquitin–proteasome system, indicating an importance of ubiquitylation in the development of cardiac disease. Ubiquitylation is a post-translational modification that plays a pivotal role in protein function and degradation. In 1995, homologous to E6AP C-terminus (HECT) type E3 ligases were discovered. E3 ligases are key enzymes in ubiquitylation and are classified into three families: really interesting new genes (RING), HECT, and RING-between-RINGs (RBRs). Moreover, 28 HECT-type E3 ligases have been identified in human beings. It is well conserved in evolution and is characterized by the direct attachment of ubiquitin to substrates. HECT-type E3 ligase is reported to be involved in a wide range of human diseases and health. The role of HECT-type E3 ligases in the development of cardiac diseases has been uncovered in the last decade. There are only a few review articles summarizing recent advancements regarding HECT-type E3 ligase in the field of cardiac disease. This study focused on cardiac remodeling and described the role of HECT-type E3 ligases in the development of cardiac disease. Moreover, this study revealed that the current knowledge could be exploited for the development of new clinical therapies.
Collapse
|
12
|
Goto J, Otaki Y, Watanabe T, Kobayashi Y, Aono T, Watanabe K, Wanezaki M, Kutsuzawa D, Kato S, Tamura H, Nishiyama S, Arimoto T, Takahashi H, Shishido T, Watanabe M. HECT (Homologous to the E6-AP Carboxyl Terminus)-Type Ubiquitin E3 Ligase ITCH Attenuates Cardiac Hypertrophy by Suppressing the Wnt/β-Catenin Signaling Pathway. Hypertension 2020; 76:1868-1878. [PMID: 33131309 DOI: 10.1161/hypertensionaha.120.15487] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The HECT (homologous to the E6-AP carboxyl terminus)-type ubiquitin E3 ligase ITCH is an enzyme that plays an important role in ubiquitin-proteasomal protein degradation. Disheveled proteins (Dvl1 [disheveled protein 1], Dvl2, and Dvl3) are the main components of the Wnt/β-catenin signaling pathway, which is involved in cardiac hypertrophy. The aim of this study was to examine the role of ITCH during development of cardiac hypertrophy. Thoracic transverse aortic constriction (TAC) was performed in transgenic mice with cardiac-specific overexpression of ITCH (ITCH-Tg) and wild-type mice. Cardiac hypertrophy after TAC was attenuated in ITCH-Tg mice, and the survival rate was higher for ITCH-Tg mice than for wild-type mice. Protein interaction between ITCH and Dvls was confirmed with immunoprecipitation in vivo and in vitro. Expression of key molecules of the Wnt/β-catenin signaling pathway (Dvl1, Dvl2, GSK3β [glycogen synthase kinase 3β], and β-catenin) was inhibited in ITCH-Tg mice compared with wild-type mice. Notably, the ubiquitination level of Dvl proteins increased in ITCH-Tg mice. Protein and mRNA expression levels of ITCH increased in response to Wnt3a stimulation in neonatal rat cardiomyocytes. Knockdown of ITCH using small-interfering RNA increased cardiomyocyte size and augmented protein expression levels of Dvl proteins, phospho-GSK3β, and β-catenin after Wnt3a stimulation in cardiomyocytes. Conversely, overexpression of ITCH attenuated cardiomyocyte hypertrophy and decreased protein expression levels of Dvl proteins, phospho-GSK3β and β-catenin. In conclusion, ITCH targets Dvl proteins for ubiquitin-proteasome degradation in cardiomyocytes and attenuates cardiac hypertrophy by suppressing the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Jun Goto
- From the Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Japan
| | - Yoichiro Otaki
- From the Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Japan
| | - Tetsu Watanabe
- From the Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Japan
| | - Yuta Kobayashi
- From the Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Japan
| | - Tomonori Aono
- From the Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Japan
| | - Ken Watanabe
- From the Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Japan
| | - Masahiro Wanezaki
- From the Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Japan
| | - Daisuke Kutsuzawa
- From the Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Japan
| | - Shigehiko Kato
- From the Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Japan
| | - Harutoshi Tamura
- From the Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Japan
| | - Satoshi Nishiyama
- From the Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Japan
| | - Takanori Arimoto
- From the Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Japan
| | - Hiroki Takahashi
- From the Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Japan
| | - Tetsuro Shishido
- From the Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Japan
| | - Masafumi Watanabe
- From the Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Japan
| |
Collapse
|
13
|
Zhang W, Chen Q, Xu L, Cai J, Zhang J. The potential role of PSMA6 in modulating fat deposition in pigs by promoting preadipocyte proliferation and differentiation. Gene 2020; 769:145228. [PMID: 33096182 DOI: 10.1016/j.gene.2020.145228] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/21/2020] [Accepted: 10/07/2020] [Indexed: 01/07/2023]
Abstract
To investigate whether the proteasome subunit alpha 6 (PSMA6) gene has an effect on fat deposition, the gene expression profile was first detected in Berkshire pigs and Jinhua pigs (JHP). The results demonstrated that significantly higher levels of mRNA expression were identified in adipose tissues and the liver. Interestingly, when compared to the longissimus dorsi muscle (LDM) in each breed, it was discovered that the expression levels of the PSMA6 gene in these tissues of JHP were considerably higher than those in Berkshire pigs. Meantime, some significant correlations of PSMA6 mRNA expression in lipid metabolism-related tissues such as the liver and LDM with the marbling score, as well as the content of intramuscular fat (IMF), in pigs were found by correlation coefficient analysis. To further explore the effects of PSMA6 expression on fat deposition, we performed PSMA6 overexpression in 3T3-L1 cells via Lentivirus infection. Our results indicated that PSMA6 could promote cell proliferation and accelerate cell division. It was also found that the transcription factors CCAAT/enhancer-binding protein alpha (CEBPA) and peroxisome proliferator-activated receptor gamma (PPARG), as well as the key genes related to adipogenesis, were upregulated, while the genes related to fat oxidation were significantly downregulated, which indicated that the PSMA6 gene could stimulate the differentiation of preadipocytes.
Collapse
Affiliation(s)
- Wei Zhang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Qiangqiang Chen
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Liaoyi Xu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Jianfeng Cai
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Jinzhi Zhang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, People's Republic of China.
| |
Collapse
|
14
|
Zhang Y, Qian H, Wu B, You S, Wu S, Lu S, Wang P, Cao L, Zhang N, Sun Y. E3 Ubiquitin ligase NEDD4 family‑regulatory network in cardiovascular disease. Int J Biol Sci 2020; 16:2727-2740. [PMID: 33110392 PMCID: PMC7586430 DOI: 10.7150/ijbs.48437] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 08/06/2020] [Indexed: 12/17/2022] Open
Abstract
Protein ubiquitination represents a critical modification occurring after translation. E3 ligase catalyzes the covalent binding of ubiquitin to the protein substrate, which could be degraded. Ubiquitination as an important protein post-translational modification is closely related to cardiovascular disease. The NEDD4 family, belonging to HECT class of E3 ubiquitin ligases can recognize different substrate proteins, including PTEN, ENaC, Nav1.5, SMAD2, PARP1, Septin4, ALK1, SERCA2a, TGFβR3 and so on, via the WW domain to catalyze ubiquitination, thus participating in multiple cardiovascular-related disease such as hypertension, arrhythmia, myocardial infarction, heart failure, cardiotoxicity, cardiac hypertrophy, myocardial fibrosis, cardiac remodeling, atherosclerosis, pulmonary hypertension and heart valve disease. However, there is currently no review comprehensively clarifying the important role of NEDD4 family proteins in the cardiovascular system. Therefore, the present review summarized recent studies about NEDD4 family members in cardiovascular disease, providing novel insights into the prevention and treatment of cardiovascular disease. In addition, assessing transgenic animals and performing gene silencing would further identify the ubiquitination targets of NEDD4. NEDD4 quantification in clinical samples would also constitute an important method for determining NEDD4 significance in cardiovascular disease.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Hao Qian
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Boquan Wu
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Shilong You
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Shaojun Wu
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Saien Lu
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Pingyuan Wang
- Staff scientist, Center for Molecular Medicine National Heart Lung and Blood Institute, National Institutes of Health, the United States
| | - Liu Cao
- Key Laboratory of Medical Cell Biology, Ministry of Education; Institute of Translational Medicine, China Medical University; Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, Shenyang, Liaoning, China
| | - Naijin Zhang
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Yingxian Sun
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| |
Collapse
|
15
|
Li B, Li S, He Q, Du S. Generation of MuRF-GFP transgenic zebrafish models for investigating murf gene expression and protein localization in Smyd1b and Hsp90α1 knockdown embryos. Comp Biochem Physiol B Biochem Mol Biol 2019; 240:110368. [PMID: 31669374 DOI: 10.1016/j.cbpb.2019.110368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/24/2019] [Accepted: 09/26/2019] [Indexed: 12/18/2022]
Abstract
Muscle-specific RING-finger proteins (MuRFs) are E3 ubiquitin ligases that play important roles in protein quality control in skeletal and cardiac muscles. Here we characterized murf gene expression and protein localization in zebrafish embryos. We found that the zebrafish genome contains six murf genes, including murf1a, murf1b, murf2a, murf2b, murf3 and a murf2-like gene that are specifically expressed in skeletal and cardiac muscles of zebrafish embryos. To analyze the subcellular localization, we generated transgenic zebrafish models expressing MurF1a-GFP or MuRF2a-GFP fusion proteins. MuRF1a-GFP and MuRF2a-GFP showed distinct patterns of subcellular localization. MuRF1a-GFP displayed a striated pattern of localization in myofibers, whereas MuRF2a-GFP mainly exhibited a random pattern of punctate distribution. The MuRF1a-GFP signal appeared as small dots aligned along the M-lines of the sarcomeres in skeletal myofibers. To determine whether knockdown of smyd1b or hsp90α1 that increased myosin protein degradation could alter murf gene expression or MuRF protein localization, we knocked down smyd1b or hsp90α1 in wild type, Tg(ef1a:MurF1a-GFP) and Tg(ef1a:MuRF2a-GFP) transgenic zebrafish embryos. Knockdown of smyd1b or hsp90α1 had no effect on murf gene expression. However, the sarcomeric distribution of MuRF1a-GFP was abolished in the knockdown embryos. This was accompanied by an increased random punctate distribution of MuRF1a-GFP in muscle cells of zebrafish embryos. Collectively, these studies demonstrate that MuRFs are specifically expressed in developing muscles of zebrafish embryos. The M-line localization MuRF1a is altered by sarcomere disruption in smyd1b or hsp90α1 knockdown embryos.
Collapse
Affiliation(s)
- Baojun Li
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, Shanxi, China; Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 701 East Pratt Street, Baltimore, MD 21202, USA
| | - Siping Li
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 701 East Pratt Street, Baltimore, MD 21202, USA
| | - Qiuxia He
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 701 East Pratt Street, Baltimore, MD 21202, USA
| | - Shaojun Du
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 701 East Pratt Street, Baltimore, MD 21202, USA.
| |
Collapse
|
16
|
Gavazzoni M, Lombardi CM, Vizzardi E, Gorga E, Sciatti E, Rossi L, Belotti A, Rossi G, Metra M, Raddino R. Irreversible proteasome inhibition with carfilzomib as first line therapy in patients with newly diagnosed multiple myeloma: Early in vivo cardiovascular effects. Eur J Pharmacol 2018; 838:85-90. [DOI: 10.1016/j.ejphar.2018.09.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 09/05/2018] [Accepted: 09/06/2018] [Indexed: 01/22/2023]
|
17
|
Dorsch LM, Schuldt M, Knežević D, Wiersma M, Kuster DWD, van der Velden J, Brundel BJJM. Untying the knot: protein quality control in inherited cardiomyopathies. Pflugers Arch 2018; 471:795-806. [PMID: 30109411 PMCID: PMC6475634 DOI: 10.1007/s00424-018-2194-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 08/06/2018] [Indexed: 12/27/2022]
Abstract
Mutations in genes encoding sarcomeric proteins are the most important causes of inherited cardiomyopathies, which are a major cause of mortality and morbidity worldwide. Although genetic screening procedures for early disease detection have been improved significantly, treatment to prevent or delay mutation-induced cardiac disease onset is lacking. Recent findings indicate that loss of protein quality control (PQC) is a central factor in the disease pathology leading to derailment of cellular protein homeostasis. Loss of PQC includes impairment of heat shock proteins, the ubiquitin-proteasome system, and autophagy. This may result in accumulation of misfolded and aggregation-prone mutant proteins, loss of sarcomeric and cytoskeletal proteins, and, ultimately, loss of cardiac function. PQC derailment can be a direct effect of the mutation-induced activation, a compensatory mechanism due to mutation-induced cellular dysfunction or a consequence of the simultaneous occurrence of the mutation and a secondary hit. In this review, we discuss recent mechanistic findings on the role of proteostasis derailment in inherited cardiomyopathies, with special focus on sarcomeric gene mutations and possible therapeutic applications.
Collapse
Affiliation(s)
- Larissa M Dorsch
- Amsterdam UMC, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, O2 building 11W53, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands.
| | - Maike Schuldt
- Amsterdam UMC, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, O2 building 11W53, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands.
| | - Dora Knežević
- Amsterdam UMC, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, O2 building 11W53, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - Marit Wiersma
- Amsterdam UMC, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, O2 building 11W53, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - Diederik W D Kuster
- Amsterdam UMC, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, O2 building 11W53, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - Jolanda van der Velden
- Amsterdam UMC, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, O2 building 11W53, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - Bianca J J M Brundel
- Amsterdam UMC, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, O2 building 11W53, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| |
Collapse
|
18
|
Exercise prevents impaired autophagy and proteostasis in a model of neurogenic myopathy. Sci Rep 2018; 8:11818. [PMID: 30087400 PMCID: PMC6081439 DOI: 10.1038/s41598-018-30365-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 07/26/2018] [Indexed: 01/07/2023] Open
Abstract
Increased proteolytic activity has been widely associated with skeletal muscle atrophy. However, elevated proteolysis is also critical for the maintenance of cellular homeostasis by disposing cytotoxic proteins and non-functioning organelles. We recently demonstrated that exercise activates autophagy and re-establishes proteostasis in cardiac diseases. Here, we characterized the impact of exercise on skeletal muscle autophagy and proteostasis in a model of neurogenic myopathy induced by sciatic nerve constriction in rats. Neurogenic myopathy, characterized by progressive atrophy and impaired contractility, was paralleled by accumulation of autophagy-related markers and loss of acute responsiveness to both colchicine and chloroquine. These changes were correlated with elevated levels of damaged proteins, chaperones and pro-apoptotic markers compared to control animals. Sustained autophagy inhibition using chloroquine in rats (50 mg.kg-1.day-1) or muscle-specific deletion of Atg7 in mice was sufficient to impair muscle contractility in control but not in neurogenic myopathy, suggesting that dysfunctional autophagy is critical in skeletal muscle pathophysiology. Finally, 4 weeks of aerobic exercise training (moderate treadmill running, 5x/week, 1 h/day) prior to neurogenic myopathy improved skeletal muscle autophagic flux and proteostasis. These changes were followed by spared muscle mass and better contractility properties. Taken together, our findings suggest the potential value of exercise in maintaining skeletal muscle proteostasis and slowing down the progression of neurogenic myopathy.
Collapse
|
19
|
Gavazzoni M, Vizzardi E, Gorga E, Bonadei I, Rossi L, Belotti A, Rossi G, Ribolla R, Metra M, Raddino R. Mechanism of cardiovascular toxicity by proteasome inhibitors: New paradigm derived from clinical and pre-clinical evidence. Eur J Pharmacol 2018; 828:80-88. [DOI: 10.1016/j.ejphar.2018.03.022] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 03/11/2018] [Accepted: 03/14/2018] [Indexed: 01/08/2023]
|
20
|
Bozi LH, Campos JC. Targeting the ubiquitin proteasome system in diabetic cardiomyopathy. J Mol Cell Cardiol 2017; 109:61-63. [DOI: 10.1016/j.yjmcc.2017.06.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 06/19/2017] [Accepted: 06/26/2017] [Indexed: 12/14/2022]
|
21
|
Shintani-Domoto Y, Hayasaka T, Maeda D, Masaki N, Ito TK, Sakuma K, Tanaka M, Kabashima K, Takei S, Setou M, Fukayama M. Different desmin peptides are distinctly deposited in cytoplasmic aggregations and cytoplasm of desmin-related cardiomyopathy patients. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2017; 1865:828-836. [PMID: 28341603 DOI: 10.1016/j.bbapap.2017.03.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 03/15/2017] [Accepted: 03/16/2017] [Indexed: 11/16/2022]
Abstract
Desmin-related cardiomyopathy is a heterogeneous group of myofibrillar myopathies characterized by aggregates of desmin and related proteins in myocytes. It has been debated how the expression and protein structure are altered in the aggregates and other parts of myocytes in patients. To address this question, we investigated the proteome quantification as well as localization in formalin-fixed and paraffin-embedded specimens of the heart of patients by imaging mass spectrometry and liquid chromatography-mass spectrometry analyses. Fifteen tryptic peptide signals were enriched in the desmin-related cardiomyopathy myocardium, twelve of which were identified as desmin peptides with 14.3- to 27.3-fold increase compared to normal hearts. High-intensity signals at m/z 1032.5 and 1002.5, which were desmin peptides 59-70 at the head portion and 213-222 at the 1B domain, were with infrequent colocalization distributed not only in desmin-positive intracytoplasmic aggregates but also in histologically normal cytoplasm, indicating that desmin protein is fragmented and different types of naturally-occurring truncated proteins ectopically assemble throughout the heart of patients. Thus, in addition to conventional histological identification of protein aggregates, specific desmin peptides show a marked difference in quantity and localization in a tissue section of desmin-related cardiomyopathy and differentiate from other cardiomyopathies. This article is part of a Special Issue entitled: MALDI Imaging, edited by Dr. Corinna Henkel and Prof. Peter Hoffmann.
Collapse
Affiliation(s)
- Yukako Shintani-Domoto
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takahiro Hayasaka
- Laboratory for Advanced Lipid Analysis, Health Innovation & Technology Center, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan; Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Daichi Maeda
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cellular and Organ Pathology, Akita University, Akita, Japan
| | - Noritaka Masaki
- Department of Medical Spectroscopy, Institute for Medical Photonics Research, Preeminent Medical Photonics Education &Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan; Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takashi K Ito
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan; International Mass Imaging Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kei Sakuma
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Michio Tanaka
- Department of Pathology, Tokyo Metropolitan Hiroo Hospital, Tokyo, Japan
| | - Katsuya Kabashima
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan; International Mass Imaging Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Shiro Takei
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan; International Mass Imaging Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Mitsutoshi Setou
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan; International Mass Imaging Center, Hamamatsu University School of Medicine, Hamamatsu, Japan; Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan; Riken Center for Molecular Imaging Science, Kobe, Japan; Department of Anatomy, The University of Hong Kong, Hong Kong, China; Division of Neural Systematics, National Institute for Physiological Sciences, Okazaki, Japan.
| | - Masashi Fukayama
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
22
|
Gilda JE, Gomes AV. Proteasome dysfunction in cardiomyopathies. J Physiol 2017; 595:4051-4071. [PMID: 28181243 DOI: 10.1113/jp273607] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 01/13/2017] [Indexed: 12/16/2022] Open
Abstract
The ubiquitin-proteasome system (UPS) plays a critical role in removing unwanted intracellular proteins and is involved in protein quality control, signalling and cell death. Because the heart is subject to continuous metabolic and mechanical stress, the proteasome plays a particularly important role in the heart, and proteasome dysfunction has been suggested as a causative factor in cardiac dysfunction. Proteasome impairment has been detected in cardiomyopathies, heart failure, myocardial ischaemia, and hypertrophy. Proteasome inhibition is also sufficient to cause cardiac dysfunction in healthy pigs, and patients using a proteasome inhibitor for cancer therapy have a higher incidence of heart failure. In this Topical Review we discuss the experimental data which suggest UPS dysfunction is a common feature of cardiomyopathies, with an emphasis on hypertrophic cardiomyopathy caused by sarcomeric mutations. We also propose potential mechanisms by which cardiomyopathy-causing mutations may lead to proteasome impairment, such as altered calcium handling and increased oxidative stress due to mitochondrial dysfunction.
Collapse
Affiliation(s)
- Jennifer E Gilda
- Department of Neurobiology, Physiology, and Behaviour, University of California, Davis, CA, 95616, USA
| | - Aldrin V Gomes
- Department of Neurobiology, Physiology, and Behaviour, University of California, Davis, CA, 95616, USA.,Department of Physiology and Membrane Biology, University of California, Davis, CA, 95616, USA
| |
Collapse
|
23
|
Evans TD, Sergin I, Zhang X, Razani B. Target acquired: Selective autophagy in cardiometabolic disease. Sci Signal 2017; 10:eaag2298. [PMID: 28246200 PMCID: PMC5451512 DOI: 10.1126/scisignal.aag2298] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The accumulation of damaged or excess proteins and organelles is a defining feature of metabolic disease in nearly every tissue. Thus, a central challenge in maintaining metabolic homeostasis is the identification, sequestration, and degradation of these cellular components, including protein aggregates, mitochondria, peroxisomes, inflammasomes, and lipid droplets. A primary route through which this challenge is met is selective autophagy, the targeting of specific cellular cargo for autophagic compartmentalization and lysosomal degradation. In addition to its roles in degradation, selective autophagy is emerging as an integral component of inflammatory and metabolic signaling cascades. In this Review, we focus on emerging evidence and key questions about the role of selective autophagy in the cell biology and pathophysiology of metabolic diseases such as obesity, diabetes, atherosclerosis, and steatohepatitis. Essential players in these processes are the selective autophagy receptors, defined broadly as adapter proteins that both recognize cargo and target it to the autophagosome. Additional domains within these receptors may allow integration of information about autophagic flux with critical regulators of cellular metabolism and inflammation. Details regarding the precise receptors involved, such as p62 and NBR1, and their predominant interacting partners are just beginning to be defined. Overall, we anticipate that the continued study of selective autophagy will prove to be informative in understanding the pathogenesis of metabolic diseases and to provide previously unrecognized therapeutic targets.
Collapse
Affiliation(s)
- Trent D Evans
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ismail Sergin
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Xiangyu Zhang
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Babak Razani
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
24
|
Aghajani M, Faghihi M, Imani A, Vaez Mahdavi MR, Shakoori A, Rastegar T, Parsa H, Mehrabi S, Moradi F, Kazemi Moghaddam E. Post-infarct sleep disruption and its relation to cardiac remodeling in a rat model of myocardial infarction. Chronobiol Int 2017; 34:587-600. [PMID: 28156163 DOI: 10.1080/07420528.2017.1281823] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Sleep disruption after myocardial infarction (MI) by affecting ubiquitin-proteasome system (UPS) is thought to contribute to myocardial remodeling and progressive worsening of cardiac function. The aim of current study was to test the hypothesis about the increased risk of developing heart failure due to experience of sleep restriction (SR) after MI. Male Wistar rats (n = 40) were randomly assigned to four experimental groups: (1) Sham, (2) MI, (3) MI and SR (MI + SR) (4) Sham and SR (Sham + SR). MI was induced by permanent ligation of left anterior descending coronary artery. Twenty-four hours after surgery, animals were subjected to chronic SR paradigm. Blood sampling was performed at days 1, 8 and 21 after MI for determination of serum levels of creatine kinase-MB (CK-MB), corticosterone, malondialdehyde (MDA) and nitric oxide (NO). Finally, at 21 days after MI, echocardiographic parameters and expression of MuRF1, MaFBx, A20, eNOS, iNOS and NF-kB in the heart were evaluated. We used H&E staining to detect myocardial hypertrophy. We found out that post infarct SR increased corticosterone levels. Our results highlighted deteriorating effects of post-MI SR on NO production, oxidative stress, and echocardiographic indexes (p < 0.05). Moreover, its detrimental effects on myocardial damage were confirmed by overexpression of MuRF1, MaFBx, iNOS and NF-kB (p < 0.001) in left ventricle and downregulation of A20 and eNOS (p < 0.05). Furthermore, histological examination revealed that experience of SR after MI increased myocardial diameter as compared to Sham subjects (p < 0.05). Our data suggest that SR after MI leads to an enlargement of the heart within 21 days, marked by an increase in oxidative stress and NO production as well as an imbalance in UPS that ultimately results in cardiac dysfunction and heart failure.
Collapse
Affiliation(s)
- Marjan Aghajani
- a Physiology Department , Faculty of Medicine, Tehran University of Medical Sciences , Tehran , Iran
| | - Mahdieh Faghihi
- a Physiology Department , Faculty of Medicine, Tehran University of Medical Sciences , Tehran , Iran
| | - Alireza Imani
- a Physiology Department , Faculty of Medicine, Tehran University of Medical Sciences , Tehran , Iran.,b Occupational Sleep Research Center, Tehran University of Medical Sciences , Tehran , Iran
| | - Mohammad Reza Vaez Mahdavi
- c Traditional Medicine Clinical Trial Research Center, Shahed University , Tehran , Iran.,d Department of Physiology , Medical Faculty, Shahed University , Tehran , Iran
| | - Abbas Shakoori
- e Genetic Department , Faculty of Medicine, Tehran University of Medical Sciences , Tehran , Iran
| | - Tayebeh Rastegar
- f Anatomy Department , Faculty of Medicine, Tehran University of Medical Sciences , Tehran , Iran
| | - Hoda Parsa
- a Physiology Department , Faculty of Medicine, Tehran University of Medical Sciences , Tehran , Iran
| | - Saman Mehrabi
- e Genetic Department , Faculty of Medicine, Tehran University of Medical Sciences , Tehran , Iran
| | - Fatemeh Moradi
- a Physiology Department , Faculty of Medicine, Tehran University of Medical Sciences , Tehran , Iran
| | - Ehsan Kazemi Moghaddam
- g Shiraz Burn and Wound Healing Research Center, Amir-al-momenin Burn Hospital, Shiraz University of Medical Sciences , Iran.,h Department of Microbiology , Medical Faculty, Shahed University , Tehran , Iran
| |
Collapse
|
25
|
Dadson K, Hauck L, Hao Z, Grothe D, Rao V, Mak TW, Billia F. The E3 ligase Mule protects the heart against oxidative stress and mitochondrial dysfunction through Myc-dependent inactivation of Pgc-1α and Pink1. Sci Rep 2017; 7:41490. [PMID: 28148912 PMCID: PMC5288653 DOI: 10.1038/srep41490] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 12/21/2016] [Indexed: 01/03/2023] Open
Abstract
Cardiac homeostasis requires proper control of protein turnover. Protein degradation is principally controlled by the Ubiquitin-Proteasome System. Mule is an E3 ubiquitin ligase that regulates cellular growth, DNA repair and apoptosis to maintain normal tissue architecture. However, Mule’s function in the heart has yet to be described. In a screen, we found reduced Mule expression in left ventricular samples from end-stage heart failure patients. Consequently, we generated conditional cardiac-specific Mule knockout (Mule fl/fl(y);mcm) mice. Mule ablation in adult Mule fl/fl(y);mcm mice prevented myocardial c-Myc polyubiquitination, leading to c-Myc accumulation and subsequent reduced expression of Pgc-1α, Pink1, and mitochondrial complex proteins. Furthermore, these mice developed spontaneous cardiac hypertrophy, left ventricular dysfunction, and early mortality. Co-deletion of Mule and c-Myc rescued this phenotype. Our data supports an indispensable role for Mule in cardiac homeostasis through the regulation of mitochondrial function via maintenance of Pgc-1α and Pink1 expression and persistent negative regulation of c-Myc.
Collapse
Affiliation(s)
- Keith Dadson
- Toronto General Research Institute, Toronto, 100 College St., M5G 1L7, Ontario Canada
| | - Ludger Hauck
- Toronto General Research Institute, Toronto, 100 College St., M5G 1L7, Ontario Canada
| | - Zhenyue Hao
- Campbell Family Cancer Research Institute, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Daniela Grothe
- Toronto General Research Institute, Toronto, 100 College St., M5G 1L7, Ontario Canada
| | - Vivek Rao
- Division of Cardiovascular Surgery, UHN, Toronto, ON, M5G 2C4, Canada
| | - Tak W Mak
- Campbell Family Cancer Research Institute, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Filio Billia
- Toronto General Research Institute, Toronto, 100 College St., M5G 1L7, Ontario Canada.,Division of Cardiology, University Health Network (UHN), Toronto, Ontario, Canada.,Heart and Stroke Richard Lewar Centre of Excellence, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, 1 King's College Circle, Toronto, M5G 1A8, Ontario Canada
| |
Collapse
|
26
|
Shimizu Y, Nicholson CK, Lambert JP, Barr LA, Kuek N, Herszenhaut D, Tan L, Murohara T, Hansen JM, Husain A, Naqvi N, Calvert JW. Sodium Sulfide Attenuates Ischemic-Induced Heart Failure by Enhancing Proteasomal Function in an Nrf2-Dependent Manner. Circ Heart Fail 2016; 9:e002368. [PMID: 27056879 DOI: 10.1161/circheartfailure.115.002368] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 02/29/2016] [Indexed: 12/17/2022]
Abstract
BACKGROUND Therapeutic strategies aimed at increasing hydrogen sulfide (H2S) levels exert cytoprotective effects in various models of cardiovascular injury. However, the underlying mechanism(s) responsible for this protection remain to be fully elucidated. Nuclear factor E2-related factor 2 (Nrf2) is a cellular target of H2S and facilitator of H2S-mediated cardioprotection after acute myocardial infarction. Here, we tested the hypothesis that Nrf2 mediates the cardioprotective effects of H2S therapy in the setting of heart failure. METHODS AND RESULTS Mice (12 weeks of age) deficient in Nrf2 (Nrf2 KO; C57BL/6J background) and wild-type littermates were subjected to ischemic-induced heart failure. Wild-type mice treated with H2S in the form of sodium sulfide (Na2S) displayed enhanced Nrf2 signaling, improved left ventricular function, and less cardiac hypertrophy after the induction of heart failure. In contrast, Na2S therapy failed to provide protection against heart failure in Nrf2 KO mice. Studies aimed at evaluating the underlying cardioprotective mechanisms found that Na2S increased the expression of proteasome subunits, resulting in an increased proteasome activity and a reduction in the accumulation of damaged proteins. In contrast, Na2S therapy failed to enhance the proteasome and failed to attenuate the accumulation of damaged proteins in Nrf2 KO mice. Additionally, Na2S failed to improve cardiac function when the proteasome was inhibited. CONCLUSIONS These findings indicate that Na2S therapy enhances proteasomal activity and function during the development of heart failure in an Nrf2-dependent manner and that this enhancement leads to attenuation in cardiac dysfunction.
Collapse
Affiliation(s)
- Yuuki Shimizu
- From the Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center (Y.S., C.K.N., J.P.L., L.A.B., N.K., D.H., J.W.C.), Department of Medicine, Division of Cardiology (L.T., A.H., N.N.), and Department of Pediatrics (J.M.H.), Emory University School of Medicine, Atlanta, GA; and Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (T.M.)
| | - Chad K Nicholson
- From the Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center (Y.S., C.K.N., J.P.L., L.A.B., N.K., D.H., J.W.C.), Department of Medicine, Division of Cardiology (L.T., A.H., N.N.), and Department of Pediatrics (J.M.H.), Emory University School of Medicine, Atlanta, GA; and Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (T.M.)
| | - Jonathan P Lambert
- From the Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center (Y.S., C.K.N., J.P.L., L.A.B., N.K., D.H., J.W.C.), Department of Medicine, Division of Cardiology (L.T., A.H., N.N.), and Department of Pediatrics (J.M.H.), Emory University School of Medicine, Atlanta, GA; and Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (T.M.)
| | - Larry A Barr
- From the Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center (Y.S., C.K.N., J.P.L., L.A.B., N.K., D.H., J.W.C.), Department of Medicine, Division of Cardiology (L.T., A.H., N.N.), and Department of Pediatrics (J.M.H.), Emory University School of Medicine, Atlanta, GA; and Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (T.M.)
| | - Nicholas Kuek
- From the Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center (Y.S., C.K.N., J.P.L., L.A.B., N.K., D.H., J.W.C.), Department of Medicine, Division of Cardiology (L.T., A.H., N.N.), and Department of Pediatrics (J.M.H.), Emory University School of Medicine, Atlanta, GA; and Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (T.M.)
| | - David Herszenhaut
- From the Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center (Y.S., C.K.N., J.P.L., L.A.B., N.K., D.H., J.W.C.), Department of Medicine, Division of Cardiology (L.T., A.H., N.N.), and Department of Pediatrics (J.M.H.), Emory University School of Medicine, Atlanta, GA; and Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (T.M.)
| | - Lin Tan
- From the Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center (Y.S., C.K.N., J.P.L., L.A.B., N.K., D.H., J.W.C.), Department of Medicine, Division of Cardiology (L.T., A.H., N.N.), and Department of Pediatrics (J.M.H.), Emory University School of Medicine, Atlanta, GA; and Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (T.M.)
| | - Toyoaki Murohara
- From the Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center (Y.S., C.K.N., J.P.L., L.A.B., N.K., D.H., J.W.C.), Department of Medicine, Division of Cardiology (L.T., A.H., N.N.), and Department of Pediatrics (J.M.H.), Emory University School of Medicine, Atlanta, GA; and Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (T.M.)
| | - Jason M Hansen
- From the Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center (Y.S., C.K.N., J.P.L., L.A.B., N.K., D.H., J.W.C.), Department of Medicine, Division of Cardiology (L.T., A.H., N.N.), and Department of Pediatrics (J.M.H.), Emory University School of Medicine, Atlanta, GA; and Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (T.M.)
| | - Ahsan Husain
- From the Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center (Y.S., C.K.N., J.P.L., L.A.B., N.K., D.H., J.W.C.), Department of Medicine, Division of Cardiology (L.T., A.H., N.N.), and Department of Pediatrics (J.M.H.), Emory University School of Medicine, Atlanta, GA; and Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (T.M.)
| | - Nawazish Naqvi
- From the Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center (Y.S., C.K.N., J.P.L., L.A.B., N.K., D.H., J.W.C.), Department of Medicine, Division of Cardiology (L.T., A.H., N.N.), and Department of Pediatrics (J.M.H.), Emory University School of Medicine, Atlanta, GA; and Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (T.M.)
| | - John W Calvert
- From the Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center (Y.S., C.K.N., J.P.L., L.A.B., N.K., D.H., J.W.C.), Department of Medicine, Division of Cardiology (L.T., A.H., N.N.), and Department of Pediatrics (J.M.H.), Emory University School of Medicine, Atlanta, GA; and Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan (T.M.).
| |
Collapse
|
27
|
Bairwa SC, Parajuli N, Dyck JRB. The role of AMPK in cardiomyocyte health and survival. Biochim Biophys Acta Mol Basis Dis 2016; 1862:2199-2210. [PMID: 27412473 DOI: 10.1016/j.bbadis.2016.07.001] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 07/05/2016] [Accepted: 07/05/2016] [Indexed: 01/09/2023]
Abstract
Cellular energy homeostasis is a fundamental process that governs the overall health of the cell and is paramount to cell survival. Central to this is the control of ATP generation and utilization, which is regulated by a complex myriad of enzymatic reactions controlling cellular metabolism. In the cardiomyocyte, ATP generated from substrate catabolism is used for numerous cellular processes including maintaining ionic homeostasis, cell repair, protein synthesis and turnover, organelle turnover, and contractile function. In many instances, cardiovascular disease is associated with impaired cardiac energetics and thus the signalling that regulates pathways involved in cardiomyocyte metabolism may be potential targets for pharmacotherapy designed to help treat cardiovascular disease. An important regulator of cardiomyocyte energy homeostasis is adenosine monophosphate-activated protein kinase (AMPK). AMPK is a serine-threonine kinase that functions primarily as a metabolic sensor to coordinate anabolic and catabolic activities in the cell via the phosphorylation of multiple proteins involved in metabolic pathways. In addition to the direct role that AMPK plays in the regulation of cardiomyocyte metabolism, AMPK can also either directly or indirectly influence other cellular processes such as regulating mitochondrial function, post-translation acetylation, autophagy, mitophagy, endoplasmic reticulum stress, and apoptosis. Thus, AMPK is implicated in the control of a wide variety of cellular processes that can influence cardiomyocyte health and survival. In this review, we will discuss the important role that AMPK plays in regulating cardiac metabolism, as well as the additional cellular processes that may contribute to cardiomyocyte function and survival in the healthy and the diseased heart. 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)
- Suresh C Bairwa
- Department of Medicine, Faculty of Medicine and Dentistry, Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Nirmal Parajuli
- Department of Medicine, Faculty of Medicine and Dentistry, Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Jason R B Dyck
- Department of Medicine, Faculty of Medicine and Dentistry, Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada; Department of Pediatrics, Faculty of Medicine and Dentistry, Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
28
|
Gilda JE, Lai X, Witzmann FA, Gomes AV. Delineation of Molecular Pathways Involved in Cardiomyopathies Caused by Troponin T Mutations. Mol Cell Proteomics 2016; 15:1962-81. [PMID: 27022107 DOI: 10.1074/mcp.m115.057380] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Indexed: 11/06/2022] Open
Abstract
Familial hypertrophic cardiomyopathy (FHC) is associated with mild to severe cardiac problems and is the leading cause of sudden death in young people and athletes. Although the genetic basis for FHC is well-established, the molecular mechanisms that ultimately lead to cardiac dysfunction are not well understood. To obtain important insights into the molecular mechanism(s) involved in FHC, hearts from two FHC troponin T models (Ile79Asn [I79N] and Arg278Cys [R278C]) were investigated using label-free proteomics and metabolomics. Mutations in troponin T are the third most common cause of FHC, and the I79N mutation is associated with a high risk of sudden cardiac death. Most FHC-causing mutations, including I79N, increase the Ca(2+) sensitivity of the myofilament; however, the R278C mutation does not alter Ca(2+) sensitivity and is associated with a better prognosis than most FHC mutations. Out of more than 1200 identified proteins, 53 and 76 proteins were differentially expressed in I79N and R278C hearts, respectively, when compared with wild-type hearts. Interestingly, more than 400 proteins were differentially expressed when the I79N and R278C hearts were directly compared. The three major pathways affected in I79N hearts relative to R278C and wild-type hearts were the ubiquitin-proteasome system, antioxidant systems, and energy production pathways. Further investigation of the proteasome system using Western blotting and activity assays showed that proteasome dysfunction occurs in I79N hearts. Metabolomic results corroborate the proteomic data and suggest the glycolytic, citric acid, and electron transport chain pathways are important pathways that are altered in I79N hearts relative to R278C or wild-type hearts. Our findings suggest that impaired energy production and protein degradation dysfunction are important mechanisms in FHCs associated with poor prognosis and that cardiac hypertrophy is not likely needed for a switch from fatty acid to glucose metabolism.
Collapse
Affiliation(s)
| | - Xianyin Lai
- ¶Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Frank A Witzmann
- ¶Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Aldrin V Gomes
- From the ‡Department of Neurobiology, Physiology, and Behavior, §Department of Physiology and Membrane Biology, University of California, Davis, California 95616;
| |
Collapse
|
29
|
Otaki Y, Takahashi H, Watanabe T, Funayama A, Netsu S, Honda Y, Narumi T, Kadowaki S, Hasegawa H, Honda S, Arimoto T, Shishido T, Miyamoto T, Kamata H, Nakajima O, Kubota I. HECT-Type Ubiquitin E3 Ligase ITCH Interacts With Thioredoxin-Interacting Protein and Ameliorates Reactive Oxygen Species-Induced Cardiotoxicity. J Am Heart Assoc 2016; 5:JAHA.115.002485. [PMID: 26796253 PMCID: PMC4859366 DOI: 10.1161/jaha.115.002485] [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] [Indexed: 12/17/2022]
Abstract
Background The homologous to the E6‐AP carboxyl terminus (HECT)–type ubiquitin E3 ligase ITCH is an enzyme that plays a pivotal role in posttranslational modification by ubiquitin proteasomal protein degradation. Thioredoxin‐interacting protein (TXNIP) is a negative regulator of the thioredoxin system and an endogenous reactive oxygen species scavenger. In the present study, we focused on the functional role of ubiquitin E3 ligase ITCH and its interaction with TXNIP to elucidate the mechanism of cardiotoxicity induced by reactive oxygen species, such as doxorubicin and hydrogen peroxide. Methods and Results Protein interaction between TXNIP and ITCH in cardiomyocyte was confirmed by immunoprecipitation assays. Overexpression of ITCH increased proteasomal TXNIP degradation and augmented thioredoxin activity, leading to inhibition of reactive oxygen species generation, p38 MAPK, p53, and subsequent intrinsic pathway cardiomyocyte apoptosis in reactive oxygen species–induced cardiotoxicity. Conversely, knockdown of ITCH using small interfering RNA inhibited TXNIP degradation and resulted in a subsequent increase in cardiomyocyte apoptosis. Next, we generated a transgenic mouse with cardiac‐specific overexpression of ITCH, called the ITCH‐Tg mouse. The expression level of TXNIP in the myocardium in ITCH‐Tg mice was significantly lower than WT littermates. In ITCH‐Tg mice, cardiac dysfunction and remodeling were restored compared with WT littermates after doxorubicin injection and myocardial infarction surgery. Kaplan–Meier analysis revealed that ITCH‐Tg mice had a higher survival rate than WT littermates after doxorubicin injection and myocardial infarction surgery. Conclusion We demonstrated, for the first time, that ITCH targets TXNIP for ubiquitin‐proteasome degradation in cardiomyocytes and ameliorates reactive oxygen species–induced cardiotoxicity through the thioredoxin system.
Collapse
Affiliation(s)
- Yoichiro Otaki
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan (Y.O., H.T., T.W., A.F., S.N., Y.H., T.N., S.K., H.H., S.H., T.A., T.S., T.M., I.K.)
| | - Hiroki Takahashi
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan (Y.O., H.T., T.W., A.F., S.N., Y.H., T.N., S.K., H.H., S.H., T.A., T.S., T.M., I.K.)
| | - Tetsu Watanabe
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan (Y.O., H.T., T.W., A.F., S.N., Y.H., T.N., S.K., H.H., S.H., T.A., T.S., T.M., I.K.)
| | - Akira Funayama
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan (Y.O., H.T., T.W., A.F., S.N., Y.H., T.N., S.K., H.H., S.H., T.A., T.S., T.M., I.K.)
| | - Shunsuke Netsu
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan (Y.O., H.T., T.W., A.F., S.N., Y.H., T.N., S.K., H.H., S.H., T.A., T.S., T.M., I.K.)
| | - Yuki Honda
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan (Y.O., H.T., T.W., A.F., S.N., Y.H., T.N., S.K., H.H., S.H., T.A., T.S., T.M., I.K.)
| | - Taro Narumi
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan (Y.O., H.T., T.W., A.F., S.N., Y.H., T.N., S.K., H.H., S.H., T.A., T.S., T.M., I.K.)
| | - Shinpei Kadowaki
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan (Y.O., H.T., T.W., A.F., S.N., Y.H., T.N., S.K., H.H., S.H., T.A., T.S., T.M., I.K.)
| | - Hiromasa Hasegawa
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan (Y.O., H.T., T.W., A.F., S.N., Y.H., T.N., S.K., H.H., S.H., T.A., T.S., T.M., I.K.)
| | - Shintaro Honda
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan (Y.O., H.T., T.W., A.F., S.N., Y.H., T.N., S.K., H.H., S.H., T.A., T.S., T.M., I.K.)
| | - Takanori Arimoto
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan (Y.O., H.T., T.W., A.F., S.N., Y.H., T.N., S.K., H.H., S.H., T.A., T.S., T.M., I.K.)
| | - Tetsuro Shishido
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan (Y.O., H.T., T.W., A.F., S.N., Y.H., T.N., S.K., H.H., S.H., T.A., T.S., T.M., I.K.)
| | - Takuya Miyamoto
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan (Y.O., H.T., T.W., A.F., S.N., Y.H., T.N., S.K., H.H., S.H., T.A., T.S., T.M., I.K.)
| | - Hideaki Kamata
- Laboratory of Biomedical Chemistry, Department of Molecular Medical Science, Graduate School of Medicine, University of Hiroshima, Japan (H.K.)
| | - Osamu Nakajima
- Research Laboratory for Molecular Genetics, Yamagata University School of Medicine, Yamagata, Japan (O.N.)
| | - Isao Kubota
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan (Y.O., H.T., T.W., A.F., S.N., Y.H., T.N., S.K., H.H., S.H., T.A., T.S., T.M., I.K.)
| |
Collapse
|
30
|
Sjögren B, Swaney S, Neubig RR. FBXO44-Mediated Degradation of RGS2 Protein Uniquely Depends on a Cullin 4B/DDB1 Complex. PLoS One 2015; 10:e0123581. [PMID: 25970626 PMCID: PMC4430315 DOI: 10.1371/journal.pone.0123581] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 03/05/2015] [Indexed: 12/30/2022] Open
Abstract
The ubiquitin-proteasome system for protein degradation plays a major role in regulating cell function and many signaling proteins are tightly controlled by this mechanism. Among these, Regulator of G Protein Signaling 2 (RGS2) is a target for rapid proteasomal degradation, however, the specific enzymes involved are not known. Using a genomic siRNA screening approach, we identified a novel E3 ligase complex containing cullin 4B (CUL4B), DNA damage binding protein 1 (DDB1) and F-box protein 44 (FBXO44) that mediates RGS2 protein degradation. While the more typical F-box partners CUL1 and Skp1 can bind FBXO44, that E3 ligase complex does not bind RGS2 and is not involved in RGS2 degradation. These observations define an unexpected DDB1/CUL4B-containing FBXO44 E3 ligase complex. Pharmacological targeting of this mechanism provides a novel therapeutic approach to hypertension, anxiety, and other diseases associated with RGS2 dysregulation.
Collapse
Affiliation(s)
- Benita Sjögren
- Department of Pharmacology & Toxicology, Michigan State University, 1355 Bogue Street, East Lansing, MI 48824, United States of America
| | - Steven Swaney
- Center for Chemical Genomics, University of Michigan, 210 Washtenaw Avenue, Ann Arbor, MI 48109, United States of America
| | - Richard R Neubig
- Department of Pharmacology & Toxicology, Michigan State University, 1355 Bogue Street, East Lansing, MI 48824, United States of America
| |
Collapse
|
31
|
Liem DA, Nsair A, Setty SP, Cadeiras M, Wang D, Maclellan R, Lotz C, Lin AJ, Tabaraki J, Li H, Ge J, Odeberg J, Ponten F, Larson E, Mulder J, Lundberg E, Weiss JN, Uhlen M, Ping P, Deng MC. Molecular- and organelle-based predictive paradigm underlying recovery by left ventricular assist device support. Circ Heart Fail 2014; 7:359-66. [PMID: 24643888 DOI: 10.1161/circheartfailure.113.000250] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
32
|
Zhang X, Guo L, Niu T, Shao L, Li H, Wu W, Wang W, Lv L, Qin Q, Wang F, Tang D, Wang XL, Cui T. Ubiquitin carboxyl terminal hydrolyase L1-suppressed autophagic degradation of p21WAF1/Cip1 as a novel feedback mechanism in the control of cardiac fibroblast proliferation. PLoS One 2014; 9:e94658. [PMID: 24732420 PMCID: PMC3986084 DOI: 10.1371/journal.pone.0094658] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 03/19/2014] [Indexed: 11/24/2022] Open
Abstract
Aims Deubiquitinating enzymes (DUBs) appear to be critical regulators of a multitude of processes such as proliferation, apoptosis, differentiation, and inflammation; however, the potential roles of DUBs in the heart remain to be determined. This study was aimed to explore the role of a DUB, ubiquitin carboxyl terminal hydrolyase L1 (UCH-L1) in maladaptive cardiac remodeling and dysfunction. Methods and Results Maladaptive cardiac remodeling and dysfunction were induced in mice by transverse aortic constriction (TAC). UCH-L1 expression was transiently increased and then declined near to the basal level while impairment of cardiac function proceeded. The upregulation of UCH-L1 was observed in cardiac myocytes and fibroblasts. In primary culture of cardiac fibroblasts, UCH-L1 was upregulated by platelet-derived growth factor (PDGF)-BB and PDGF-DD. Adenoviral overexpession of UCH-L1 inhibited the PDGF-induced cardiac fibroblast proliferation without affecting the activation of mitogen activated protein kinases (MAPKs), Akt, and signal transducers and activators of transcription 3 (STAT3). Further signaling dissection revealed that PDGF-BB posttranscriptional upregulated p21WAF1/Cip1 protein expression, which was inhibited by rapamycin, an activator of autophagy via suppressing mammalian target of rapamycin (mTOR), rather than MG132, a proteasome inhibitor. Overexpression of UCH-L1 enhanced PDGF-BB-induced mTOR phosphorylation and upregulation of p21WAF1/Cip1 protein expression while suppressed autophagic flux in cardiac fibroblasts. Conclusion UCH-L1 facilitates PDGF-BB-induced suppression of autophagic degradation of p21WAF1/Cip1 proteins in cardiac fibroblasts, which may serve as a novel negative feedback mechanism in the control of cardiac fibroblast proliferation contributing to cardiac fibrosis and dysfunction.
Collapse
Affiliation(s)
- Xiaoming Zhang
- Shandong University Qilu Hospital Research Center for Cell Therapy, Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Department of Pathophysiology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Linlin Guo
- Shandong University Qilu Hospital Research Center for Cell Therapy, Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Ting Niu
- Shandong University Qilu Hospital Research Center for Cell Therapy, Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Lei Shao
- Shandong University Qilu Hospital Research Center for Cell Therapy, Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Huanjie Li
- Shandong University Qilu Hospital Research Center for Cell Therapy, Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Weiwei Wu
- Shandong University Qilu Hospital Research Center for Cell Therapy, Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Wenjuan Wang
- Shandong University Qilu Hospital Research Center for Cell Therapy, Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Linmao Lv
- Shandong University Qilu Hospital Research Center for Cell Therapy, Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Qingyun Qin
- Shandong University Qilu Hospital Research Center for Cell Therapy, Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Fang Wang
- Shandong University Qilu Hospital Research Center for Cell Therapy, Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Dongqi Tang
- Shandong University Qilu Hospital Research Center for Cell Therapy, Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, United States of America
| | - Xing Li Wang
- Shandong University Qilu Hospital Research Center for Cell Therapy, Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan, Shandong, China
- * E-mail: (XW); (TC)
| | - Taixing Cui
- Shandong University Qilu Hospital Research Center for Cell Therapy, Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, United States of America
- * E-mail: (XW); (TC)
| |
Collapse
|
33
|
Hwee DT, Baehr LM, Philp A, Baar K, Bodine SC. Maintenance of muscle mass and load-induced growth in Muscle RING Finger 1 null mice with age. Aging Cell 2014; 13:92-101. [PMID: 23941502 PMCID: PMC3947038 DOI: 10.1111/acel.12150] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2013] [Indexed: 12/18/2022] Open
Abstract
Age-related loss of muscle mass occurs to varying degrees in all individuals and has a detrimental effect on morbidity and mortality. Muscle RING Finger 1 (MuRF1), a muscle-specific E3 ubiquitin ligase, is believed to mediate muscle atrophy through the ubiquitin proteasome system (UPS). Deletion of MuRF1 (KO) in mice attenuates the loss of muscle mass following denervation, disuse, and glucocorticoid treatment; however, its role in age-related muscle loss is unknown. In this study, skeletal muscle from male wild-type (WT) and MuRF1 KO mice was studied up to the age of 24 months. Muscle mass and fiber cross-sectional area decreased significantly with age in WT, but not in KO mice. In aged WT muscle, significant decreases in proteasome activities, especially 20S and 26S β5 (20–40% decrease), were measured and were associated with significant increases in the maladaptive endoplasmic reticulum (ER) stress marker, CHOP. Conversely, in aged MuRF1 KO mice, 20S or 26S β5 proteasome activity was maintained or decreased to a lesser extent than in WT mice, and no increase in CHOP expression was measured. Examination of the growth response of older (18 months) mice to functional overload revealed that old WT mice had significantly less growth relative to young mice (1.37- vs. 1.83-fold), whereas old MuRF1 KO mice had a normal growth response (1.74- vs. 1.90-fold). These data collectively suggest that with age, MuRF1 plays an important role in the control of skeletal muscle mass and growth capacity through the regulation of cellular stress.
Collapse
Affiliation(s)
- Darren T. Hwee
- Departments of Neurobiology, Physiology, and Behavior; University of California Davis; Davis CA 95616 USA
- Molecular, Cellular and Integrative Physiology Graduate Group; University of California Davis; Davis CA 95616 USA
| | - Leslie M. Baehr
- Physiology and Membrane Biology; University of California Davis; Davis CA 95616 USA
| | - Andrew Philp
- Departments of Neurobiology, Physiology, and Behavior; University of California Davis; Davis CA 95616 USA
| | - Keith Baar
- Departments of Neurobiology, Physiology, and Behavior; University of California Davis; Davis CA 95616 USA
- Physiology and Membrane Biology; University of California Davis; Davis CA 95616 USA
| | - Sue C. Bodine
- Departments of Neurobiology, Physiology, and Behavior; University of California Davis; Davis CA 95616 USA
- Physiology and Membrane Biology; University of California Davis; Davis CA 95616 USA
| |
Collapse
|
34
|
Proteomic remodeling of proteasome in right heart failure. J Mol Cell Cardiol 2014; 66:41-52. [DOI: 10.1016/j.yjmcc.2013.10.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Revised: 09/13/2013] [Accepted: 10/22/2013] [Indexed: 12/30/2022]
|
35
|
Katz G, Shainberg A, Hochhauser E, Kurtzwald-Josefson E, Issac A, El-Ani D, Aravot D, Afek A, Seidman JG, Seidman CE, Eldar M, Arad M. The role of mutant protein level in autosomal recessive catecholamine dependent polymorphic ventricular tachycardia (CPVT2). Biochem Pharmacol 2013; 86:1576-83. [PMID: 24070655 DOI: 10.1016/j.bcp.2013.09.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Revised: 09/12/2013] [Accepted: 09/12/2013] [Indexed: 01/05/2023]
Abstract
Humans and genetically engineered mice with recessively inherited CPVT develop arrhythmia which may arise due to malfunction or degradation of calsequestrin (CASQ2). We investigated the relation between protein level and arrhythmia severity in CASQ2(D307H/D307H) (D307H), compared to CASQ2(Δ/Δ) (KO) and wild type (WT) mice. CASQ2 expression and Ca²⁺ transients were recorded in cardiomyocytes from neonatal or adult mice. Arrhythmia was studied in vivo using heart rhythm telemetry at rest, exercise and after epinephrine injection. CASQ2 protein was absent in KO heart. Neonatal D307H and WT hearts expressed significantly less CASQ2 protein than the level found in the adult WT. Adult D307H expressed only 20% of CASQ2 protein found in WT. Spontaneous Ca²⁺ release was more prevalent in neonatal KO cardiomyocytes (89%) compared to 33-36% of either WT or D307H, respectively, p<0.001. Adult cardiomyocytes from both mutant mice had more Ca²⁺ abnormalities compared to control (KO: 82%, D307H 63%, WT 12%, p<0.01). Calcium oscillations were most common in KO cardiomyocytes. We then treated mice with bortezomib to inhibit CASQ2(D307H) degradation. Bortezomib increased CASQ2 expression in D307H hearts by ∼50% (p<0.05). Bortezomib-treated D307H mice had lower CPVT prevalence and less premature ventricular beats during peak exercise. No benefit against arrhythmia was observed in bortezomib treated KO mice. These results indicate that the mutant CASQ2(D307H) protein retains some of its physiological function. Its expression decreases with age and is inversely related to arrhythmia severity. Preventing the degradation of mutant protein should be explored as a possible therapeutic strategy in appropriate CPVT2 patients.
Collapse
Affiliation(s)
- Guy Katz
- Leviev Heart Center, Sheba Medical Center, Tel Hashomer and Sackler School of, Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Honton B, Despas F, Dumonteil N, Rouvellat C, Roussel M, Carrie D, Galinier M, Montastruc JL, Pathak A. Bortezomib and heart failure: case-report and review of the French Pharmacovigilance database. Fundam Clin Pharmacol 2013; 28:349-52. [PMID: 23781941 DOI: 10.1111/fcp.12039] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Revised: 04/17/2013] [Accepted: 05/21/2013] [Indexed: 11/30/2022]
Abstract
Bortezomib is a proteasome inhibitor commonly indicated for the treatment of multiple myeloma and non Hodgkin lymphoma. Cardiac adverse drug reactions of this drug are not clearly established. We report case where direct involvement of bortezomib in the occurrence of heart failure is strongly suspected and 22 other cases spontaneously reported to the French Pharmacovigilance System. This report should increase cardiologist awareness about the risk of heart failure related to this drug. Moreover, these cases underline the need for a systematic cardiac screening in patients exposed to bortezomib.
Collapse
Affiliation(s)
- Benjamin Honton
- Centre Hospitalier et Universitaire de Toulouse, Service de Cardiologie, F-31432, Toulouse, France; Institut National de la Santé et de la Recherche Médicale INSERM, Institut des Maladies Métaboliques et cardiovasculaires I2MC, Université de Toulouse, UPS, F-31432, Toulouse, France
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Costa MW, Guo G, Wolstein O, Vale M, Castro ML, Wang L, Otway R, Riek P, Cochrane N, Furtado M, Semsarian C, Weintraub RG, Yeoh T, Hayward C, Keogh A, Macdonald P, Feneley M, Graham RM, Seidman JG, Seidman CE, Rosenthal N, Fatkin D, Harvey RP. Functional characterization of a novel mutation in NKX2-5 associated with congenital heart disease and adult-onset cardiomyopathy. ACTA ACUST UNITED AC 2013; 6:238-47. [PMID: 23661673 DOI: 10.1161/circgenetics.113.000057] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND The transcription factor NKX2-5 is crucial for heart development, and mutations in this gene have been implicated in diverse congenital heart diseases and conduction defects in mouse models and humans. Whether NKX2-5 mutations have a role in adult-onset heart disease is unknown. METHODS AND RESULTS Mutation screening was performed in 220 probands with adult-onset dilated cardiomyopathy. Six NKX2-5 coding sequence variants were identified, including 3 nonsynonymous variants. A novel heterozygous mutation, I184M, located within the NKX2-5 homeodomain, was identified in 1 family. A subset of family members had congenital heart disease, but there was an unexpectedly high prevalence of dilated cardiomyopathy. Functional analysis of I184M in vitro demonstrated a striking increase in protein expression when transfected into COS-7 cells or HL-1 cardiomyocytes because of reduced degradation by the Ubiquitin-proteasome system. In functional assays, DNA-binding activity of I184M was reduced, resulting in impaired activation of target genes despite increased expression levels of mutant protein. CONCLUSIONS Certain NKX2-5 homeodomain mutations show abnormal protein degradation via the Ubiquitin-proteasome system and partially impaired transcriptional activity. We propose that this class of mutation can impair heart development and mature heart function and contribute to NKX2-5-related cardiomyopathies with graded severity.
Collapse
Affiliation(s)
- Mauro W Costa
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Sishi BJN, Loos B, van Rooyen J, Engelbrecht AM. Doxorubicin induces protein ubiquitination and inhibits proteasome activity during cardiotoxicity. Toxicology 2013; 309:23-9. [PMID: 23639627 DOI: 10.1016/j.tox.2013.04.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Revised: 04/02/2013] [Accepted: 04/22/2013] [Indexed: 02/04/2023]
Abstract
Anthracycline-induced cardiotoxicity is a clinically complex syndrome that leads to substantial morbidity and mortality for cancer survivors. Despite several years of research, the underlying molecular mechanisms remain largely undefined and thus effective therapies to manage this condition are currently non-existent. This study therefore aimed to determine the contribution of the ubiquitin-proteasome pathway (UPP) and endoplasmic reticulum (ER)-stress within this context. Cardiotoxicity was induced with the use of doxorubicin (DXR) in H9C2 rat cardiomyoblasts (3 μM) for 24 h, whereas the tumour-bearing GFP-LC3 mouse model was treated with a cumulative dose of 20 mg/kg. Markers for proteasome-specific protein degradation were significantly upregulated in both models following DXR treatment, however proteasome activity was lost. Moreover, ER-stress as assessed by increased ER load was considerably augmented (in vitro) with modest binding of DXR with ER. These results suggest that DXR induces intrinsic activation of the UPP and ER stress which ultimately contributes to dysfunction of the myocardium during this phenomenon.
Collapse
Affiliation(s)
- Balindiwe J N Sishi
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, 7600, South Africa.
| | | | | | | |
Collapse
|
39
|
Herrmann J, Wohlert C, Saguner AM, Flores A, Nesbitt LL, Chade A, Lerman LO, Lerman A. Primary proteasome inhibition results in cardiac dysfunction. Eur J Heart Fail 2013; 15:614-23. [PMID: 23616520 DOI: 10.1093/eurjhf/hft034] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
AIMS The proteasome prevents the intracellular accumulation of proteins and its impairment can lead to structural and functional alterations, as noted for the coronary vasculature in a previous study. Utilizing the same model, this study was designed to test the hypothesis that chronic proteasome inhibition (PSI) also leads to structural and functional changes of the heart. METHODS AND RESULTS Female domestic pigs were randomized to a normal diet without (N) or with twice-weekly subcutaneous injections of the proteasome inhibitor MLN-273 (0.08 mg/kg, N + PSI, n = 5 each group). In vivo data on cardiac structure and function as well as myocardial perfusion and microvascular permeability response to adenosine and dobutamine were obtained by electron beam computed tomography after 11 weeks. Subsequent ex vivo myocardial analyses included immunoblotting, immunostaining, TUNEL (terminal deoxynucleotidyl transferase dUTP nick end labelling), Masson trichrome, and Congo red staining. Compared with N, an increase in LV mass was observed in N + PSI (106.5 ± 16.4 g vs. 183.1 ± 24.2 g, P < 0.05). The early to late diastolic filling ratio was increased in N + PSI vs. N (3.5 ± 0.6 vs. 1.8 ± 0.1, P < 0.05). The EF tended to be lower (46 ± 12% and 53 ± 9%, respectively) and cardiac output was significantly lower in N + PSI than in N (2.9 ± 1.1 vs. 4.7 ± 1.1 L/min, P < 0.05). Tissue analyses demonstrated an accumulation of proteasome substrates, apoptosis, and fibrosis in the PSI group. Compared with N, the myocardial perfusion response was reduced and microvascular permeability was increased in N + PSI. CONCLUSION The current study demonstrates that chronic proeasome inhibition affects the cardiovascular system, leading to functional and structural alteration of the heart consistent with a hypertrophic-restrictive cardiomyopathy phenotype.
Collapse
Affiliation(s)
- Joerg Herrmann
- Division of Cardiovascular Diseases, Mayo Clinic Rochester, 200 First Street, SW, Rochester, MN 55905, USA.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Maintenance of protein quality control is a critical function of the ubiquitin proteasome system (UPS). Evidence is rapidly mounting to link proteasome dysfunction with a multitude of cardiac diseases, including ischemia, reperfusion, atherosclerosis, hypertrophy, heart failure, and cardiomyopathies. Recent studies have demonstrated a remarkable level of complexity in the regulation of the UPS in the heart and suggest that our understanding of how UPS dysfunction might contribute to the pathophysiology of such a wide range of cardiac afflictions is still very limited. Whereas experimental systems, including animal models, are invaluable for exploring mechanisms and establishing pathogenicity of UPS dysfunction in cardiac disease, studies using human heart tissue provide a vital adjunct for establishing clinical relevance of experimental findings and promoting new hypotheses. Accordingly, this review will focus on UPS dysfunction in human dilated and hypertrophic cardiomyopathies and highlight areas rich for further study in this expanding field.
Collapse
Affiliation(s)
- Sharlene M Day
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA.
| |
Collapse
|
41
|
Campos JC, Queliconi BB, Dourado PMM, Cunha TF, Zambelli VO, Bechara LRG, Kowaltowski AJ, Brum PC, Mochly-Rosen D, Ferreira JCB. Exercise training restores cardiac protein quality control in heart failure. PLoS One 2012; 7:e52764. [PMID: 23300764 PMCID: PMC3531365 DOI: 10.1371/journal.pone.0052764] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Accepted: 11/22/2012] [Indexed: 12/16/2022] Open
Abstract
Exercise training is a well-known coadjuvant in heart failure treatment; however, the molecular mechanisms underlying its beneficial effects remain elusive. Despite the primary cause, heart failure is often preceded by two distinct phenomena: mitochondria dysfunction and cytosolic protein quality control disruption. The objective of the study was to determine the contribution of exercise training in regulating cardiac mitochondria metabolism and cytosolic protein quality control in a post-myocardial infarction-induced heart failure (MI-HF) animal model. Our data demonstrated that isolated cardiac mitochondria from MI-HF rats displayed decreased oxygen consumption, reduced maximum calcium uptake and elevated H₂O₂ release. These changes were accompanied by exacerbated cardiac oxidative stress and proteasomal insufficiency. Declined proteasomal activity contributes to cardiac protein quality control disruption in our MI-HF model. Using cultured neonatal cardiomyocytes, we showed that either antimycin A or H₂O₂ resulted in inactivation of proteasomal peptidase activity, accumulation of oxidized proteins and cell death, recapitulating our in vivo model. Of interest, eight weeks of exercise training improved cardiac function, peak oxygen uptake and exercise tolerance in MI-HF rats. Moreover, exercise training restored mitochondrial oxygen consumption, increased Ca²⁺-induced permeability transition and reduced H₂O₂ release in MI-HF rats. These changes were followed by reduced oxidative stress and better cardiac protein quality control. Taken together, our findings uncover the potential contribution of mitochondrial dysfunction and cytosolic protein quality control disruption to heart failure and highlight the positive effects of exercise training in re-establishing cardiac mitochondrial physiology and protein quality control, reinforcing the importance of this intervention as a non-pharmacological tool for heart failure therapy.
Collapse
Affiliation(s)
- Juliane C. Campos
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Bruno B. Queliconi
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Sao Paulo, Brazil
| | | | - Telma F. Cunha
- School of Physical Education and Sport, University of Sao Paulo, Sao Paulo, Brazil
| | | | - Luiz R. G. Bechara
- School of Physical Education and Sport, University of Sao Paulo, Sao Paulo, Brazil
| | - Alicia J. Kowaltowski
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Sao Paulo, Brazil
| | - Patricia C. Brum
- School of Physical Education and Sport, University of Sao Paulo, Sao Paulo, Brazil
| | - Daria Mochly-Rosen
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Julio C. B. Ferreira
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
42
|
Gielen S, Sandri M, Kozarez I, Kratzsch J, Teupser D, Thiery J, Erbs S, Mangner N, Lenk K, Hambrecht R, Schuler G, Adams V. Exercise training attenuates MuRF-1 expression in the skeletal muscle of patients with chronic heart failure independent of age: the randomized Leipzig Exercise Intervention in Chronic Heart Failure and Aging catabolism study. Circulation 2012; 125:2716-27. [PMID: 22565934 DOI: 10.1161/circulationaha.111.047381] [Citation(s) in RCA: 148] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Muscle wasting occurs in both chronic heart failure (CHF) and normal aging and contributes to exercise intolerance and increased morbidity/mortality. However, the molecular mechanisms of muscle atrophy in CHF and their interaction with aging are still largely unknown. We therefore measured the activation of the ubiquitin-proteasome system and the lysosomal pathway of intracellular proteolysis in muscle biopsies of CHF patients and healthy controls in two age strata and assessed the age-dependent effects of a 4-week endurance training program on the catabolic-anabolic balance. METHODS AND RESULTS Sixty CHF patients (30 patients aged ≤55 years, mean age 46±5 years; 30 patients aged ≥65 years, mean age 72±5 years) and 60 healthy controls (30 subjects aged ≤55 years, mean age 50±5 years; 30 subjects aged ≥65 years, mean age 72±4 years) were randomized to 4 weeks of supervised endurance training or to a control group. Before and after the intervention, vastus lateralis muscle biopsies were obtained. The expressions of cathepsin-L and the muscle-specific E3 ligases MuRF-1 and MAFbx were measured by real-time polymerase chain reaction and confirmed by Western blot. At baseline, MuRF-1 expression was significantly higher in CHF patients versus healthy controls (mRNA: 624±59 versus 401±25 relative units; P=0.007). After 4 weeks of exercise training, MuRF-1 mRNA expression was reduced by -32.8% (P=0.02) in CHF patients aged ≤55 years and by -37.0% (P<0.05) in CHF patients aged ≥65 years. CONCLUSIONS MuRF-1, a component of the ubiquitin-proteasome system involved in muscle proteolysis, is increased in the skeletal muscle of patients with heart failure. Exercise training results in reduced MuRF-1 levels, suggesting that it blocks ubiquitin-proteasome system activation and does so in both younger and older CHF patients. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov. Unique identifier: NCT00176319.
Collapse
Affiliation(s)
- Stephan Gielen
- Department of Internal Medicine III, University Hospital, Martin Luther University of Halle/Wittenberg, Ernst-Grube Strasse 40, 06120 Halle, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Ferreira JCB, Boer BN, Grinberg M, Brum PC, Mochly-Rosen D. Protein quality control disruption by PKCβII in heart failure; rescue by the selective PKCβII inhibitor, βIIV5-3. PLoS One 2012; 7:e33175. [PMID: 22479367 PMCID: PMC3316563 DOI: 10.1371/journal.pone.0033175] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Accepted: 02/05/2012] [Indexed: 11/18/2022] Open
Abstract
Myocardial remodeling and heart failure (HF) are common sequelae of many forms of cardiovascular disease and a leading cause of mortality worldwide. Accumulation of damaged cardiac proteins in heart failure has been described. However, how protein quality control (PQC) is regulated and its contribution to HF development are not known. Here, we describe a novel role for activated protein kinase C isoform βII (PKCβII) in disrupting PQC. We show that active PKCβII directly phosphorylated the proteasome and inhibited proteasomal activity in vitro and in cultured neonatal cardiomyocytes. Importantly, inhibition of PKCβII, using a selective PKCβII peptide inhibitor (βIIV5-3), improved proteasomal activity and conferred protection in cultured neonatal cardiomyocytes. We also show that sustained inhibition of PKCβII increased proteasomal activity, decreased accumulation of damaged and misfolded proteins and increased animal survival in two rat models of HF. Interestingly, βIIV5-3-mediated protection was blunted by sustained proteasomal inhibition in HF. Finally, increased cardiac PKCβII activity and accumulation of misfolded proteins associated with decreased proteasomal function were found also in remodeled and failing human hearts, indicating a potential clinical relevance of our findings. Together, our data highlights PKCβII as a novel inhibitor of proteasomal function. PQC disruption by increased PKCβII activity in vivo appears to contribute to the pathophysiology of heart failure, suggesting that PKCβII inhibition may benefit patients with heart failure. (218 words).
Collapse
Affiliation(s)
- Julio C B Ferreira
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, California, United States of America
| | | | | | | | | |
Collapse
|
44
|
Shi Y, Moon M, Dawood S, McManus B, Liu PP. Mechanisms and management of doxorubicin cardiotoxicity. Herz 2012; 36:296-305. [PMID: 21656050 DOI: 10.1007/s00059-011-3470-3] [Citation(s) in RCA: 150] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Doxorubicin is an effective anti-tumor agent with a cumulative dose-dependent cardiotoxicity. In addition to its principal toxic mechanisms involving iron and redox reactions, recent studies have described new mechanisms of doxorubicin-induced cell death, including abnormal protein processing, hyper-activated innate immune responses, inhibition of neuregulin-1 (NRG1)/ErbB(HER) signalling, impaired progenitor cell renewal/cardiac repair, and decreased vasculogenesis. Although multiple mechanisms involved in doxorubicin cardiotoxicity have been studied, there is presently no clinically proven treatment established for doxorubicin cardiomyopathy. Iron chelator dexrazoxane, angiotensin converting enzyme (ACE) inhibitors, and β-blockade have been proposed as potential preventive strategies for doxorubicin cardiotoxicity. Novel approaches such as anti-miR-146 or recombinant NRG1 to increase cardiomyocyte resistance to toxicity may be of interest in the future.
Collapse
Affiliation(s)
- Y Shi
- Division of Cardiology, Heart and Stroke/Richard Lewar Centre of Excellence, University Health Network, University of Toronto, Toronto General Hospital, Ontario, Canada
| | | | | | | | | |
Collapse
|
45
|
Sishi BJN, Bester DJ, Wergeland A, Loos B, Jonassen AK, van Rooyen J, Engelbrecht AM. Daunorubicin therapy is associated with upregulation of E3 ubiquitin ligases in the heart. Exp Biol Med (Maywood) 2012; 237:219-26. [PMID: 22328594 DOI: 10.1258/ebm.2011.011106] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Daunorubicin (DNR) and doxorubicin (DOX) are two of the most effective anthracycline drugs known for the treatment of systemic neoplasms and solid tumors. However, their clinical use is hampered due to profound cardiotoxicity. The mechanism by which DNR injures the heart remains to be fully elucidated. Recent reports have indicated that DOX activates ubiquitin proteasome-mediated degradation of specific transcription factors; however, no reports exist on the effect of DNR on the E3 ubiquitin ligases, MURF-1 (muscle ring finger 1) and MAFbx (muscle atrophy F-box). The aim of this study was to investigate the effect of DNR treatment on the protein and organelle degradation systems in the heart and to elucidate some of the signalling mechanisms involved. Adult rats were divided into two groups where one group received six intraperitoneal injections of 2 mg/kg DNR on alternate days and the other group received saline injections as control. Hearts were excised and perfused on a working heart system the day after the last injection and freeze-clamped for biochemical analysis. DNR treatment significantly attenuated cardiac function and increased apoptosis in the heart. DNR-induced cardiac cytotoxicity was associated with upregulation of the E3 ligases, MURF-1 and MAFbx and also caused significant increases in two markers of autophagy, beclin-1 and LC3. These changes observed in the heart were also associated with attenuation of the phosphoinositide 3-kinase/Akt signalling pathway.
Collapse
Affiliation(s)
- Balindiwe J N Sishi
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | | | | | | | | | | | | |
Collapse
|
46
|
Zhang Y, Yuan M, Bradley KM, Dong F, Anversa P, Ren J. Insulin-like growth factor 1 alleviates high-fat diet-induced myocardial contractile dysfunction: role of insulin signaling and mitochondrial function. Hypertension 2012; 59:680-93. [PMID: 22275536 DOI: 10.1161/hypertensionaha.111.181867] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Obesity is often associated with reduced plasma insulin-like growth factor 1 (IGF-1) levels, oxidative stress, mitochondrial damage, and cardiac dysfunction. This study was designed to evaluate the impact of IGF-1 on high-fat diet-induced oxidative, myocardial, geometric, and mitochondrial responses. FVB and cardiomyocyte-specific IGF-1 overexpression transgenic mice were fed a low- (10%) or high-fat (45%) diet to induce obesity. High-fat diet feeding led to glucose intolerance, elevated plasma levels of leptin, interleukin 6, insulin, and triglyceride, as well as reduced circulating IGF-1 levels. Echocardiography revealed reduced fractional shortening, increased end-systolic and end-diastolic diameter, increased wall thickness, and cardiac hypertrophy in high-fat-fed FVB mice. High-fat diet promoted reactive oxygen species generation, apoptosis, protein and mitochondrial damage, reduced ATP content, cardiomyocyte cross-sectional area, contractile and intracellular Ca(2+) dysregulation (including depressed peak shortening and maximal velocity of shortening/relengthening), prolonged duration of relengthening, and dampened intracellular Ca(2+) rise and clearance. Western blot analysis revealed disrupted phosphorylation of insulin receptor and postreceptor signaling molecules insulin receptor substrate 1 (tyrosine/serine phosphorylation), Akt, glycogen synthase kinase 3β, forkhead transcriptional factors, and mammalian target of rapamycin, as well as downregulated expression of mitochondrial proteins peroxisome proliferator-activated receptor-γ coactivator 1α and uncoupling protein 2. Intriguingly, IGF-1 mitigated high-fat-diet feeding-induced alterations in reactive oxygen species, protein and mitochondrial damage, ATP content, apoptosis, myocardial contraction, intracellular Ca(2+) handling, and insulin signaling but not whole body glucose intolerance and cardiac hypertrophy. Exogenous IGF-1 treatment also alleviated high-fat diet-induced cardiac dysfunction. Our data revealed that IGF-1 alleviates high-fat diet-induced cardiac dysfunction despite persistent cardiac remodeling, possibly because of preserved cell survival, mitochondrial function, and insulin signaling.
Collapse
Affiliation(s)
- Yingmei Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | | | | | | | | | | |
Collapse
|
47
|
Abstract
Protein ubiquitination, the covalent attachment of ubiquitin to target proteins, has emerged as one of the most prevalent posttranslational modifications (PTMs), regulating nearly every cellular pathway. The diversity of signaling associated with this particular PTM stems from the myriad ways in which a target protein can be modified by ubiquitin, e.g., monoubiquitin, multi-monoubiquitin, and polyubiquitin linkages. In this Review, we focus on developments in both enzymatic and chemical methods that engender ubiquitin with new chemical and physical properties. Moreover, we highlight how these methods have enabled studies directed toward (i) characterizing enzymes responsible for reversing the ubiquitin modification, (ii) understanding the influence of ubiquitin on protein function and crosstalk with other PTMs, and (iii) uncovering the impact of polyubiquitin chain linkage and length on downstream signaling events.
Collapse
Affiliation(s)
- Eric R. Strieter
- Department of Chemistry, University of Wisconsin−Madison, Madison, Wisconsin 53706,
United States
| | - David A. Korasick
- Department of Chemistry, University of Wisconsin−Madison, Madison, Wisconsin 53706,
United States
| |
Collapse
|
48
|
Opitz E, Koch A, Klingel K, Schmidt F, Prokop S, Rahnefeld A, Sauter M, Heppner FL, Völker U, Kandolf R, Kuckelkorn U, Stangl K, Krüger E, Kloetzel PM, Voigt A. Impairment of immunoproteasome function by β5i/LMP7 subunit deficiency results in severe enterovirus myocarditis. PLoS Pathog 2011; 7:e1002233. [PMID: 21909276 PMCID: PMC3164653 DOI: 10.1371/journal.ppat.1002233] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Accepted: 07/07/2011] [Indexed: 12/18/2022] Open
Abstract
Proteasomes recognize and degrade poly-ubiquitinylated proteins. In infectious disease, cells activated by interferons (IFNs) express three unique catalytic subunits β1i/LMP2, β2i/MECL-1 and β5i/LMP7 forming an alternative proteasome isoform, the immunoproteasome (IP). The in vivo function of IPs in pathogen-induced inflammation is still a matter of controversy. IPs were mainly associated with MHC class I antigen processing. However, recent findings pointed to a more general function of IPs in response to cytokine stress. Here, we report on the role of IPs in acute coxsackievirus B3 (CVB3) myocarditis reflecting one of the most common viral disease entities among young people. Despite identical viral load in both control and IP-deficient mice, IP-deficiency was associated with severe acute heart muscle injury reflected by large foci of inflammatory lesions and severe myocardial tissue damage. Exacerbation of acute heart muscle injury in this host was ascribed to disequilibrium in protein homeostasis in viral heart disease as indicated by the detection of increased proteotoxic stress in cytokine-challenged cardiomyocytes and inflammatory cells from IP-deficient mice. In fact, due to IP-dependent removal of poly-ubiquitinylated protein aggregates in the injured myocardium IPs protected CVB3-challenged mice from oxidant-protein damage. Impaired NFκB activation in IP-deficient cardiomyocytes and inflammatory cells and proteotoxic stress in combination with severe inflammation in CVB3-challenged hearts from IP-deficient mice potentiated apoptotic cell death in this host, thus exacerbating acute tissue damage. Adoptive T cell transfer studies in IP-deficient mice are in agreement with data pointing towards an effective CD8 T cell immune. This study therefore demonstrates that IP formation primarily protects the target organ of CVB3 infection from excessive inflammatory tissue damage in a virus-induced proinflammatory cytokine milieu.
Collapse
Affiliation(s)
- Elisa Opitz
- Medizinische Klinik für Kardiologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Annett Koch
- Institut für Biochemie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Karin Klingel
- Abteilung Molekulare Pathologie, Institut für Pathologie und Neuropathologie, Eberhard-Karls-Universität, Tuebingen, Germany
| | - Frank Schmidt
- Interfakultäres Institut für Genetik und Funktionelle Genomforschung, Ernst-Moritz-Arndt-Universität, Greifswald, Germany
| | - Stefan Prokop
- Institut für Neuropathologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Anna Rahnefeld
- Medizinische Klinik für Kardiologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Martina Sauter
- Abteilung Molekulare Pathologie, Institut für Pathologie und Neuropathologie, Eberhard-Karls-Universität, Tuebingen, Germany
| | - Frank L. Heppner
- Institut für Neuropathologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Uwe Völker
- Interfakultäres Institut für Genetik und Funktionelle Genomforschung, Ernst-Moritz-Arndt-Universität, Greifswald, Germany
| | - Reinhard Kandolf
- Abteilung Molekulare Pathologie, Institut für Pathologie und Neuropathologie, Eberhard-Karls-Universität, Tuebingen, Germany
| | - Ulrike Kuckelkorn
- Institut für Biochemie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Karl Stangl
- Medizinische Klinik für Kardiologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Elke Krüger
- Institut für Biochemie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Peter M. Kloetzel
- Institut für Biochemie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Antje Voigt
- Medizinische Klinik für Kardiologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
49
|
Abstract
Chronic heart failure continues to impose a substantial health-care burden, despite recent treatment advances. The key pathophysiological process that ultimately leads to chronic heart failure is cardiac remodelling in response to chronic disease stresses. Here, we review recent advances in our understanding of molecular and cellular mechanisms that play a part in the complex remodelling process, with a focus on key molecules and pathways that might be suitable targets for therapeutic manipulation. Such pathways include those that regulate cardiac myocyte hypertrophy, calcium homoeostasis, energetics, and cell survival, and processes that take place outside the cardiac myocyte--eg, in the myocardial vasculature and extracellular matrix. We also discuss major gaps in our current understanding, take a critical look at conventional approaches to target discovery that have been used to date, and consider new investigational avenues that might accelerate clinically relevant discovery.
Collapse
Affiliation(s)
- Ajay M Shah
- King's College London British Heart Foundation Centre of Excellence, London, UK.
| | | |
Collapse
|
50
|
Usui S, Maejima Y, Pain J, Hong C, Cho J, Park JY, Zablocki D, Tian B, Glass DJ, Sadoshima J. Endogenous muscle atrophy F-box mediates pressure overload-induced cardiac hypertrophy through regulation of nuclear factor-kappaB. Circ Res 2011; 109:161-71. [PMID: 21617130 PMCID: PMC3257317 DOI: 10.1161/circresaha.110.238717] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Accepted: 05/17/2010] [Indexed: 01/19/2023]
Abstract
RATIONALE Overexpression of muscle atrophy F-box (MAFbx/atrogin-1), an E3 ubiquitin ligase, induces proteasomal degradation in cardiomyocytes. The role of endogenous MAFbx in regulating cardiac hypertrophy and failure remains unclear. OBJECTIVE We investigated the role of MAFbx in regulating cardiac hypertrophy and function in response to pressure overload. Transverse aortic constriction (TAC) was applied to MAFbx knockout (KO) and wild-type (WT) mice. METHODS AND RESULTS Expression of MAFbx in WT mice was significantly increased by TAC. TAC-induced increases in cardiac hypertrophy were significantly smaller in MAFbx KO than in WT mice. There was significantly less lung congestion and interstitial fibrosis in MAFbx KO than in WT mice. MAFbx KO also inhibited β-adrenergic cardiac hypertrophy. DNA microarray analysis revealed that activation of genes associated with the transcription factor binding site for the nuclear factor-κB family were inhibited in MAFbx KO mice compared with WT mice after TAC. Although the levels of IκB-α were significantly decreased after TAC in WT mice, they were increased in MAFbx KO mice. MAFbx regulates ubiquitination and proteasomal degradation of IκB-α in cardiomyocytes. In primary cultured rat cardiomyocytes, phenylephrine-induced activation of nuclear factor-κB and hypertrophy were significantly suppressed by MAFbx knockdown but were partially rescued by overexpression of nuclear factor-κB p65. CONCLUSIONS MAFbx plays an essential role in mediating cardiac hypertrophy in response to pressure overload. Downregulation of MAFbx inhibits cardiac hypertrophy in part through stabilization of IκB-α and inactivation of nuclear factor-κB. Taken together, inhibition of MAFbx attenuates pathological hypertrophy, thereby protecting the heart from progression into heart failure.
Collapse
Affiliation(s)
- Soichiro Usui
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, UMDNJ, New Jersey Medical School, NJ, USA
- Department of Disease Control and Homeostasis, Kanazawa University Graduate School of Medical Science, Ishikawa, Japan
| | - Yasuhiro Maejima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, UMDNJ, New Jersey Medical School, NJ, USA
| | - Jayashree Pain
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, UMDNJ, New Jersey Medical School, NJ, USA
| | - Chull Hong
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, UMDNJ, New Jersey Medical School, NJ, USA
| | - Jaeyeaon Cho
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, UMDNJ, New Jersey Medical School, NJ, USA
| | - Ji Yeon Park
- Department of Biochemistry and Molecular Biology, UMDNJ, New Jersey Medical School, NJ, USA
| | - Daniela Zablocki
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, UMDNJ, New Jersey Medical School, NJ, USA
| | - Bin Tian
- Department of Biochemistry and Molecular Biology, UMDNJ, New Jersey Medical School, NJ, USA
| | - David J. Glass
- Novartis Institutes for Biomedical Research, 400 Technology Square, Cambridge, MA, USA
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, UMDNJ, New Jersey Medical School, NJ, USA
| |
Collapse
|