51
|
Dadson K, Kovacevic V, Rengasamy P, Kim GHE, Boo S, Li RK, George I, Schulze PC, Hinz B, Sweeney G. Cellular, structural and functional cardiac remodelling following pressure overload and unloading. Int J Cardiol 2016; 216:32-42. [PMID: 27140334 DOI: 10.1016/j.ijcard.2016.03.240] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 03/25/2016] [Accepted: 03/27/2016] [Indexed: 01/18/2023]
Abstract
BACKGROUND The cardiac remodelling process in advanced heart failure due to pressure overload has not been clearly defined but likely involves mechanisms of cardiac fibrosis and cardiomyocyte hypertrophy. The aim of this study was to examine pressure overload (PO)-induced cardiac remodelling processes and their reversibility after unloading in both humans with heart failure and a mouse model of PO induced by aortic constriction. METHODS & RESULTS Speckle tracking echocardiography showed PO-induced cardiac dysfunction in mice was reversible after removal of aortic constriction to unload. Masson's Trichrome staining suggested that PO-induced myocardial fibrosis was reversible, however detailed analysis of 3-dimensional collagen architecture by scanning electron microscopy demonstrated that matrix remodelling was not completely normalised as a disorganised network of thin collagen fibres was evident. Analysis of human left ventricular biopsy samples from HF patients revealed increased presence of large collagen fibres which were greatly reduced in paired samples from the same individuals after unloading by left ventricular assist device implantation. Again, an extensive network of small collagen fibres was still clearly seen to closely surround cardiomyocytes after unloading. Other features of PO-induced remodelling including increased myofibroblast content, cardiomyocyte disarray and hypertrophy were largely reversed upon unloading in both humans and mouse model. Previous work in humans demonstrated that receptors for adiponectin, an important mediator of cardiac fibrosis and hypertrophy, decreased in heart failure patients and returned to normal after unloading. Here we provide novel data showing a similar trend for adiponectin receptor adaptor protein APPL1, but not APPL2 isoform. CONCLUSIONS LV unloading diminishes PO-induced cardiac remodelling and improves function. These findings add new insights into the cardiac remodelling process, and provide novel targets for future pharmacologic therapies.
Collapse
Affiliation(s)
- Keith Dadson
- Department of Biology, York University, Toronto, Canada
| | | | | | | | - Stellar Boo
- Laboratory of Tissue Repair and Regeneration, Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, Canada
| | - Ren-Ke Li
- Division of Cardiovascular Surgery and Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Isaac George
- Department of Internal Medicine I, Division of Cardiology, Friedrich Schiller University Jena, Jena, Germany; Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, USA
| | - P Christian Schulze
- Department of Internal Medicine I, Division of Cardiology, Friedrich Schiller University Jena, Jena, Germany; Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, USA
| | - Boris Hinz
- Laboratory of Tissue Repair and Regeneration, Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, Canada
| | - Gary Sweeney
- Department of Biology, York University, Toronto, Canada.
| |
Collapse
|
52
|
Kanamori H, Takemura G, Goto K, Tsujimoto A, Mikami A, Ogino A, Watanabe T, Morishita K, Okada H, Kawasaki M, Seishima M, Minatoguchi S. Autophagic adaptations in diabetic cardiomyopathy differ between type 1 and type 2 diabetes. Autophagy 2016; 11:1146-60. [PMID: 26042865 DOI: 10.1080/15548627.2015.1051295] [Citation(s) in RCA: 190] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Little is known about the association between autophagy and diabetic cardiomyopathy. Also unknown are possible distinguishing features of cardiac autophagy in type 1 and type 2 diabetes. In hearts from streptozotocin-induced type 1 diabetic mice, diastolic function was impaired, though autophagic activity was significantly increased, as evidenced by increases in microtubule-associated protein 1 light chain 3/LC3 and LC3-II/-I ratios, SQSTM1/p62 (sequestosome 1) and CTSD (cathepsin D), and by the abundance of autophagic vacuoles and lysosomes detected electron-microscopically. AMP-activated protein kinase (AMPK) was activated and ATP content was reduced in type 1 diabetic hearts. Treatment with chloroquine, an autophagy inhibitor, worsened cardiac performance in type 1 diabetes. In addition, hearts from db/db type 2 diabetic model mice exhibited poorer diastolic function than control hearts from db/+ mice. However, levels of LC3-II, SQSTM1 and phosphorylated MTOR (mechanistic target of rapamycin) were increased, but CTSD was decreased and very few lysosomes were detected ultrastructurally, despite the abundance of autophagic vacuoles. AMPK activity was suppressed and ATP content was reduced in type 2 diabetic hearts. These findings suggest the autophagic process is suppressed at the final digestion step in type 2 diabetic hearts. Resveratrol, an autophagy enhancer, mitigated diastolic dysfunction, while chloroquine had the opposite effects in type 2 diabetic hearts. Autophagy in the heart is enhanced in type 1 diabetes, but is suppressed in type 2 diabetes. This difference provides important insight into the pathophysiology of diabetic cardiomyopathy, which is essential for the development of new treatment strategies.
Collapse
Key Words
- AMP-activated protein kinase
- AMPK, AMP-activated protein kinase
- CTSD, cathepsin D
- DM, diabetes mellitus
- GFP, green fluorescent protein
- HBA1c, glycated hemoglobin α 1
- LV, left ventricular
- MAP1LC3/LC3, microtubule-associated protein 1 light chain 3
- MTOR, mechanistic target of rapamycin
- Mn-SOD, superoxide dismutase 2, mitochondrial
- SIRT1, sirtuin 1
- SQSTM1/p62, sequestosome 1
- STZ, streptozotocin
- autophagy
- cardiomyopathy
- chloroquine
- diabetes mellitus
- insulin
- resveratrol
- type 1 diabetes
- type 2 diabetes
- ultrastructure
Collapse
Affiliation(s)
- Hiromitsu Kanamori
- a Department of Cardiology; Gifu University Graduate School of Medicine ; Gifu , Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
53
|
Li W, Sultana N, Siraj N, Ward LJ, Pawlik M, Levy E, Jovinge S, Bengtsson E, Yuan XM. Autophagy dysfunction and regulatory cystatin C in macrophage death of atherosclerosis. J Cell Mol Med 2016; 20:1664-72. [PMID: 27079462 PMCID: PMC4988293 DOI: 10.1111/jcmm.12859] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Accepted: 02/29/2016] [Indexed: 11/29/2022] Open
Abstract
Autophagy dysfunction in mouse atherosclerosis models has been associated with increased lipid accumulation, apoptosis and inflammation. Expression of cystatin C (CysC) is decreased in human atheroma, and CysC deficiency enhances atherosclerosis in mice. Here, we first investigated the association of autophagy and CysC expression levels with atheroma plaque severity in human atherosclerotic lesions. We found that autophagy proteins Atg5 and LC3β in advanced human carotid atherosclerotic lesions are decreased, while markers of dysfunctional autophagy p62/SQSTM1 and ubiquitin are increased together with elevated levels of lipid accumulation and apoptosis. The expressions of LC3β and Atg5 were positively associated with CysC expression. Second, we investigated whether CysC expression is involved in autophagy in atherosclerotic apoE-deficient mice, demonstrating that CysC deficiency (CysC(-/-) ) in these mice results in reduction of Atg5 and LC3β levels and induction of apoptosis. Third, macrophages isolated from CysC(-/-) mice displayed increased levels of p62/SQSTM1 and higher sensitivity to 7-oxysterol-mediated lysosomal membrane destabilization and apoptosis. Finally, CysC treatment minimized oxysterol-mediated cellular lipid accumulation. We conclude that autophagy dysfunction is a characteristic of advanced human atherosclerotic lesions and is associated with reduced levels of CysC. The deficiency of CysC causes autophagy dysfunction and apoptosis in macrophages and apoE-deficient mice. The results indicate that CysC plays an important regulatory role in combating cell death via the autophagic pathway in atherosclerosis.
Collapse
Affiliation(s)
- Wei Li
- Division of Obstetrics and Gynaecology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Nargis Sultana
- Occupational and Environmental Medicine Center, Heart and Medicine Center, County Council of Östergötland, Linköping, Sweden
| | - Nabeel Siraj
- Occupational and Environmental Medicine Center, Heart and Medicine Center, County Council of Östergötland, Linköping, Sweden.,Department of internal medicine, University of Alberta Edmonton, Alberta, Canada
| | - Liam J Ward
- Division of Obstetrics and Gynaecology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden.,Occupational and Environmental Medicine Center, Heart and Medicine Center, County Council of Östergötland, Linköping, Sweden
| | - Monika Pawlik
- Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Efrat Levy
- Nathan S. Kline Institute for Psychiatric Research, Departments of Psychiatry and Biochemistry and Molecular Pharmacology, New York University Langone School of Medicine, New York, NY, USA
| | - Stefan Jovinge
- Department of Clinical Sciences, Skåne University Hospital, Lund University, Lund, Sweden
| | - Eva Bengtsson
- Department of Clinical Sciences, Skåne University Hospital, Lund University, Lund, Sweden
| | - Xi-Ming Yuan
- Occupational and Environmental Medicine Center, Heart and Medicine Center, County Council of Östergötland, Linköping, Sweden
| |
Collapse
|
54
|
Wiersma M, Henning RH, Brundel BJJM. Derailed Proteostasis as a Determinant of Cardiac Aging. Can J Cardiol 2016; 32:1166.e11-20. [PMID: 27345610 DOI: 10.1016/j.cjca.2016.03.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 02/21/2016] [Accepted: 03/07/2016] [Indexed: 01/12/2023] Open
Abstract
Age comprises the single most important risk factor for cardiac disease development. The incidence and prevalence of cardiac diseases, which represents the main cause of death worldwide, will increase even more because of the aging population. A hallmark of aging is that it is accompanied by a gradual derailment of proteostasis (eg, the homeostasis of protein synthesis, folding, assembly, trafficking, function, and degradation). Loss of proteostasis is highly relevant to cardiomyocytes, because they are postmitotic cells and therefore not constantly replenished by proliferation. The derailment of proteostasis during aging is thus an important factor that preconditions for the development of age-related cardiac diseases, such as atrial fibrillation. In turn, frailty of proteostasis in aging cardiomyocytes is exemplified by its accelerated derailment in multiple cardiac diseases. Here, we review 2 major components of the proteostasis network, the stress-responsive and protein degradation pathways, in healthy and aged cardiomyocytes. Furthermore, we discuss the relation between derailment of proteostasis and age-related cardiac diseases, including atrial fibrillation. Finally, we introduce novel therapeutic targets that might possibly attenuate cardiac aging and thus limit cardiac disease progression.
Collapse
Affiliation(s)
- Marit Wiersma
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Robert H Henning
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Bianca J J M Brundel
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
55
|
Jiang DS, Yi X, Huo B, Liu XX, Li R, Zhu XH, Wei X. The potential role of lysosome-associated membrane protein 3 (LAMP3) on cardiac remodelling. Am J Transl Res 2016; 8:37-48. [PMID: 27069538 PMCID: PMC4759414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 01/02/2016] [Indexed: 06/05/2023]
Abstract
Lysosome-associated membrane protein 3 (LAMP3) was first identified as a cell surface marker of mature dendritic cells and specifically expressed in lung tissues. Recently studies demonstrated that LAMP3 plays a critical role in several cancers, and regulated by hypoxia. However, whether LAMP3 expressed in the heart and cardiomyocytes and changed its expression level in the hearts with cardiac remodelling was largely unknown. In this study, we first cultured H9C2 (a clonal muscle cell line from rat heart) and stimulated with 1 μM angiotensin II (Ang II), or 100 μM isoproterenol (ISO), or 100 μM phenylephrine (PE) for indicated times. We found that LAMP3 expression level was significantly increased after these stimulation. Next, the pressure overload-induced cardiac remodelling mouse model was performed in the wild type C57BL/6J mice. After 4 and 8 weeks of transverse aortic constriction (TAC), obvious cardiac remodelling was observed in the wild type mice compared with sham group. Importantly, LAMP3 expression level was gradually elevated from 2 weeks to 8 weeks after TAC surgery. Furthermore, in human dilated cardiomyopathy (DCM) hearts, severe cardiac remodelling was observed, as evidenced by remarkably increased cardiomyocytes cross sectional area and collagen deposition. Notably, the mRNA and protein level of LAMP3 were significantly increased in the DCM hearts compared with donor hearts. Immunohistochemistry assay showed that LAMP3 was expression in the cardiomyocytes and responsible for its increased expression in the hearts. Our data indicated that LAMP3 might have a potential role in the process of cardiac remodelling.
Collapse
Affiliation(s)
- Ding-Sheng Jiang
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
- Heart-Lung Transplantation Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
- Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| | - Xin Yi
- Department of Cardiology, Renmin Hospital of Wuhan UniversityWuhan 430060, China
- Cardiovascular Research Institute, Wuhan UniversityWuhan 430060, China
| | - Bo Huo
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
- Heart-Lung Transplantation Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
- Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| | - Xin-Xin Liu
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
- Heart-Lung Transplantation Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
- Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| | - Rui Li
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
- Heart-Lung Transplantation Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
- Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| | - Xue-Hai Zhu
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
- Heart-Lung Transplantation Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
- Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| | - Xiang Wei
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
- Heart-Lung Transplantation Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
- Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| |
Collapse
|
56
|
Zhang H, Wang X. Priming the proteasome by protein kinase G: a novel cardioprotective mechanism of sildenafil. Future Cardiol 2015; 11:177-89. [PMID: 25760877 DOI: 10.2217/fca.15.3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The proteasome mediates the degradation of most cellular proteins including misfolded proteins, pivotal to intracellular protein hemostasis. Proteasome functional insufficiency is implicated in a large subset of human failing hearts. Experimental studies have established proteasome functional insufficiency as a major pathogenic factor, rationalizing proteasome enhancement as a potentially new therapeutic strategy for congestive heart failure. Protein kinase G activation known to be cardioprotective was recently found to facilitate proteasomal degradation of misfolded proteins in cardiomyocytes; sildenafil was shown to activate myocardial protein kinase G, improve cardiac protein quality control and slow down the progression of cardiac proteinopathy in mice. This identifies the first clinically used drug that is capable of benign proteasome enhancement and unveils a potentially novel cardioprotective mechanism for sildenafil.
Collapse
Affiliation(s)
- Hanming Zhang
- Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion, SD 57069, USA
| | | |
Collapse
|
57
|
Abstract
Mitochondrial dynamics, fission and fusion, were first identified in yeast with investigation in heart cells beginning only in the last 5 to 7 years. In the ensuing time, it has become evident that these processes are not only required for healthy mitochondria, but also, that derangement of these processes contributes to disease. The fission and fusion proteins have a number of functions beyond the mitochondrial dynamics. Many of these functions are related to their membrane activities, such as apoptosis. However, other functions involve other areas of the mitochondria, such as OPA1's role in maintaining cristae structure and preventing cytochrome c leak, and its essential (at least a 10 kDa fragment of OPA1) role in mtDNA replication. In heart disease, changes in expression of these important proteins can have detrimental effects on mitochondrial and cellular function.
Collapse
Affiliation(s)
- A A Knowlton
- Molecular & Cellular Cardiology, Division of Cardiovascular Medicine and Pharmacology Department, University of California, Davis, and The Department of Veteran's Affairs, Northern California VA, Sacramento, California, USA
| | - T T Liu
- Molecular & Cellular Cardiology, Division of Cardiovascular Medicine and Pharmacology Department, University of California, Davis, and The Department of Veteran's Affairs, Northern California VA, Sacramento, California, USA
| |
Collapse
|
58
|
Nishida K, Otsu K. Autophagy during cardiac remodeling. J Mol Cell Cardiol 2015; 95:11-8. [PMID: 26678624 DOI: 10.1016/j.yjmcc.2015.12.003] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 11/23/2015] [Accepted: 12/04/2015] [Indexed: 11/18/2022]
Abstract
Despite progress in cardiovascular research and evidence-based therapies, heart failure is a leading cause of morbidity and mortality in industrialized countries. Cardiac remodeling is a chronic maladaptive process, characterized by progressive ventricular dilatation, cardiac hypertrophy, fibrosis, and deterioration of cardiac performance, and arises from interactions between adaptive modifications of cardiomyocytes and negative aspects of adaptation such as cardiomyocyte death and fibrosis. Autophagy has evolved as a conserved process for bulk degradation and recycling of cytoplasmic components, such as long-lived proteins and organelles. Accumulating evidence demonstrates that autophagy plays an essential role in cardiac remodeling to maintain cardiac function and cellular homeostasis in the heart. This review discusses some recent advances in understanding the role of autophagy during cardiac remodeling. This article is part of a Special Issue entitled: Autophagy in the Heart.
Collapse
Affiliation(s)
- Kazuhiko Nishida
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London, SE5 9NU, UK.
| | - Kinya Otsu
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London, SE5 9NU, UK
| |
Collapse
|
59
|
He H, Sun YP, Zheng L, Yue ZG. Steroidal saponins from Paris polyphylla induce apoptotic cell death and autophagy in A549 human lung cancer cells. Asian Pac J Cancer Prev 2015; 16:1169-73. [PMID: 25735350 DOI: 10.7314/apjcp.2015.16.3.1169] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Paris polyphylla (Chinese name: Chonglou) had been traditionally used for a long time and shown anti-cancer action. Based on the previous study that paris polyphylla steroidal saponins (PPSS) induced cytotoxic effect in human lung cancer A549 cells, this study was designed to further illustrate the mechanisms underlying. MATERIALS AND METHODS The mechanisms involved in PPSS-induced A549 cell death were investigated by phase contrast microscopy and fluorescence microscopy, flow cytometry and western blot analysis, respectively. RESULTS PPSS decreased the proportion of viable A549 cells, and exposure of A549 cells to PPSS led to both apoptosis and autophagy. Apoptosis was due to activations of caspase-8, caspase-3, as well as cleavage of PARP, and autophagy was confirmed by up-regulation of Beclin 1 and the conversion from LC3 I to LC3 II. CONCLUSIONS PPSS was able to induce lung cancer A549 cell apoptosis and autophagy in vitro, the results underlining the possibility that PPSS would be a potential candidate for intervention against lung cancer.
Collapse
Affiliation(s)
- Hao He
- School of Pharmaceutical Sciences, Xi'an Medical University, Xi'an, China E-mail :
| | | | | | | |
Collapse
|
60
|
Fabregat-Andres O, Paredes F, Monsalve M, Milara J, Ridocci-Soriano F, Gonzalez-Hervas S, Mena A, Facila L, Hornero F, Morell S, Martinez-Leon J, Cortijo J. mRNA PGC-1α levels in blood samples reliably correlates with its myocardial expression: study in patients undergoing cardiac surgery. Anatol J Cardiol 2015; 16:622-629. [PMID: 27004709 PMCID: PMC5368522 DOI: 10.5152/anatoljcardiol.2015.6466] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE Peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) is a transcriptional coactivator that has been proposed to play a protective role in mouse models of cardiac ischemia and heart failure, suggesting that PGC-1α could be relevant as a prognostic marker. Our previous studies showed that the estimation of peripheral mRNA PGC-1α expression was feasible and that its induction correlated with the extent of myocardial necrosis and left ventricular remodeling in patients with myocardial infarction. In this study, we sought to determine if the myocardial and peripheral expressions of PGC-1α are well correlated and to analyze the variability of PGC-1α expression depending on the prevalence of some metabolic disorders. METHODS This was a cohort of 35 consecutive stable heart failure patients with severe aortic stenosis who underwent an elective aortic valve replacement surgery. mRNA PGC-1α expression was simultaneously determined from myocardial biopsy specimens and blood samples obtained during surgery by quantitative PCR, and a correlation between samples was made using the Kappa index. Patients were divided into two groups according to the detection of baseline expression levels of PGC-1α in blood samples, and comparisons between both groups were made by chi-square test or unpaired Student's t-test as appropriate. RESULTS Based on myocardial biopsies, we found that mRNA PGC-1α expression in blood samples showed a statistically significant correlation with myocardial expression (Kappa index 0.66, p<0.001). The presence of higher systemic PGC-1α expression was associated with a greater expression of some target genes such as silent information regulator 2 homolog-1 (x-fold expression in blood samples: 4.43±5.22 vs. 1.09±0.14, p=0.044) and better antioxidant status in these patients (concentration of Trolox: 0.40±0.05 vs. 0.34±0.65, p=0.006). CONCLUSIONS Most patients with higher peripheral expression also had increased myocardial expression, so we conclude that the non-invasive estimation of mRNA PGC-1α expression from blood samples provides a good approach of the constitutive status of the mitochondrial protection system regulated by PGC-1α and that this could be used as prognostic indicator in cardiovascular disease.
Collapse
Affiliation(s)
| | - Federico Paredes
- Cardiovascular Institute, General University Hospital, Valencia-Spain
| | - María Monsalve
- Cardiovascular Institute, General University Hospital, Valencia-Spain
| | - Javier Milara
- Cardiovascular Institute, General University Hospital, Valencia-Spain
| | | | | | - Armando Mena
- Cardiovascular Institute, General University Hospital, Valencia-Spain
| | - Lorenzo Facila
- Cardiovascular Institute, General University Hospital, Valencia-Spain
| | - Fernando Hornero
- Cardiovascular Institute, General University Hospital, Valencia-Spain
| | - Salvador Morell
- Cardiovascular Institute, General University Hospital, Valencia-Spain
| | | | - Julio Cortijo
- Research Foundation, General University Hospital, Valencia-Spain
| |
Collapse
|
61
|
Therapeutic targeting of autophagy in cardiovascular disease. J Mol Cell Cardiol 2015; 95:86-93. [PMID: 26602750 DOI: 10.1016/j.yjmcc.2015.11.019] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 11/13/2015] [Accepted: 11/17/2015] [Indexed: 12/31/2022]
Abstract
Autophagy is an evolutionarily ancient process of intracellular catabolism necessary to preserve cellular homeostasis in response to a wide variety of stresses. In the case of post-mitotic cells, where cell replacement is not an option, finely tuned quality control of cytoplasmic constituents and organelles is especially critical. And due to the ubiquitous and critical role of autophagic flux in the maintenance of cell health, it comes as little surprise that perturbation of the autophagic process is observed in multiple disease processes. A large body of preclinical evidence suggests that autophagy is a double-edged sword in cardiovascular disease, acting in either beneficial or maladaptive ways, depending on the context. In light of this, the autophagic machinery in cardiomyocytes and other cardiovascular cell types has been proposed as a potential therapeutic target. Here, we summarize current knowledge regarding the dual functions of autophagy in cardiovascular disease. We go on to analyze recent evidence suggesting that titration of autophagic flux holds potential as a novel treatment strategy.
Collapse
|
62
|
Ren J, Taegtmeyer H. Too much or not enough of a good thing — The Janus faces of autophagy in cardiac fuel and protein homeostasis. J Mol Cell Cardiol 2015; 84:223-6. [DOI: 10.1016/j.yjmcc.2015.03.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 02/23/2015] [Accepted: 03/02/2015] [Indexed: 01/01/2023]
|
63
|
Nishida K, Yamaguchi O, Otsu K. Degradation systems in heart failure. J Mol Cell Cardiol 2015; 84:212-22. [PMID: 25981331 DOI: 10.1016/j.yjmcc.2015.05.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 05/05/2015] [Accepted: 05/07/2015] [Indexed: 11/29/2022]
Abstract
Heart failure is a complex clinical syndrome that results from any structural or functional impairment of ventricular filling or the ejection of blood, and is a leading cause of morbidity and mortality in industrialized countries. The mechanisms underlying the development of heart failure are multiple, complex and not well understood. Cardiac mass and its homeostasis are maintained by the balance between protein synthesis and degradation, and an imbalance is likely to result in cellular dysfunction and disease. The protein degradation systems are the principle mechanisms for maintaining cellular homeostasis via protein quality control. Three major protein degradation systems have been identified, namely the calpain system, autophagy, and the ubiquitin proteasome system. Proinflammatory mediators involve the development and progression of heart failure. DNA and RNA degradation systems play a critical role in regulating inflammation and maintaining cellular homeostasis mediated by damaged DNA clearance and posttranscriptional regulation, respectively. This review discusses some recent advances in understanding the role of these degradation systems in heart failure.
Collapse
Affiliation(s)
- Kazuhiko Nishida
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London SE5 9NU, UK
| | - Osamu Yamaguchi
- Department of Cardiovascular Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Kinya Otsu
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London SE5 9NU, UK.
| |
Collapse
|
64
|
Arabacilar P, Marber M. The case for inhibiting p38 mitogen-activated protein kinase in heart failure. Front Pharmacol 2015; 6:102. [PMID: 26029107 PMCID: PMC4428223 DOI: 10.3389/fphar.2015.00102] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 04/24/2015] [Indexed: 11/30/2022] Open
Abstract
This minireview discusses the evidence that the inhibition of p38 mitogen-activated protein kinases (p38 MAPKs) maybe of therapeutic value in heart failure. Most previous experimental studies, as well as past and ongoing clinical trials, have focussed on the role of p38 MAPKs in myocardial infarction and acute coronary syndromes. There is now growing evidence that these kinases are activated within the myocardium of the failing human heart and in the heart and blood vessels of animal models of heart failure. Furthermore, from a philosophical viewpoint the chronic activation of the adaptive stress pathways that lead to the activation of p38 MAPKs in heart failure is analogous to the chronic activation of the sympathetic, renin-aldosterone-angiotensin and neprilysin systems. These have provided some of the most effective therapies for heart failure. This minireview questions whether similar and synergistic advantages would follow the inhibition of p38 MAPKs.
Collapse
Affiliation(s)
- Pelin Arabacilar
- Cardiovascular Division, Department of Cardiology, King's College London British Heart Foundation Centre, The Rayne Institute, St Thomas' Hospital London, UK
| | - Michael Marber
- Cardiovascular Division, Department of Cardiology, King's College London British Heart Foundation Centre, The Rayne Institute, St Thomas' Hospital London, UK
| |
Collapse
|
65
|
Nandi SS, Duryee MJ, Shahshahan HR, Thiele GM, Anderson DR, Mishra PK. Induction of autophagy markers is associated with attenuation of miR-133a in diabetic heart failure patients undergoing mechanical unloading. Am J Transl Res 2015; 7:683-696. [PMID: 26064437 PMCID: PMC4455344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 04/11/2015] [Indexed: 06/04/2023]
Abstract
Autophagy is ubiquitous in all forms of heart failure and cardioprotective miR-133a is attenuated in human heart failure. Previous reports from heart failure patients undergoing left ventricular assist device (LVAD) implantation demonstrated that autophagy is upregulated in the LV of the failing human heart. Studies in the murine model show that diabetes downregulates miR-133a. However, the role of miR-133a in the regulation of autophagy in diabetic hearts is unclear. We tested the hypothesis that diabetes exacerbates cardiac autophagy by inhibiting miR-133a in heart failure patients undergoing LVAD implantation. The miRNA assay was performed on the LV of 15 diabetic (D) and 6 non-diabetic (ND) heart failure patients undergoing LVAD implantation. Four ND with highly upregulated and 5 D with highly downregulated miR-133a were analyzed for autophagy markers (Beclin1, LC3B, ATG3) and their upstream regulators (mTOR and AMPK), and hypertrophy marker (beta-myosin heavy chain) by RT-qPCR, Western blotting and immunofluorescence. Our results demonstrate that attenuation of miR-133a in diabetic hearts is associated with the induction of autophagy and hypertrophy, and suppression of mTOR without appreciable difference in AMPK activity. In conclusion, attenuation of miR-133a contributes to the exacerbation of diabetes mediated cardiac autophagy and hypertrophy in heart failure patients undergoing LVAD implantation.
Collapse
Affiliation(s)
- Shyam Sundar Nandi
- Department of Cellular, Integrative Physiology, University of Nebraska Medical Center Omaha, NE 68198, USA
| | - Michael J Duryee
- Department of Medicine, Division of Rheumatology, University of Nebraska Medical Center Omaha, NE 68198, USA ; Veterans Affair Nebraska-Western Iowa Health Care System, Research Services 151 4101 Woolworth Avenue, Omaha NE 68105, USA
| | - Hamid R Shahshahan
- Department of Cellular, Integrative Physiology, University of Nebraska Medical Center Omaha, NE 68198, USA
| | - Geoffrey M Thiele
- Department of Medicine, Division of Rheumatology, University of Nebraska Medical Center Omaha, NE 68198, USA ; Veterans Affair Nebraska-Western Iowa Health Care System, Research Services 151 4101 Woolworth Avenue, Omaha NE 68105, USA
| | - Daniel R Anderson
- Department of Medicine, Division of Cardiology, University of Nebraska Medical center Omaha, NE 68198, USA
| | - Paras K Mishra
- Department of Cellular, Integrative Physiology, University of Nebraska Medical Center Omaha, NE 68198, USA ; Department of Anesthesiology, University of Nebraska Medical Center Omaha, NE 68198, USA
| |
Collapse
|
66
|
Abstract
Autophagy is an important physiological process in the heart, and alterations in autophagic activity can exacerbate or mitigate injury during various pathological processes. Methods to assess autophagy have changed rapidly because the field of research has expanded. As with any new field, methods and standards for data analysis and interpretation evolve as investigators acquire experience and insight. The purpose of this review is to summarize current methods to measure autophagy, selective mitochondrial autophagy (mitophagy), and autophagic flux. We will examine several published studies where confusion arose in data interpretation, to illustrate the challenges. Finally, we will discuss methods to assess autophagy in vivo and in patients.
Collapse
Affiliation(s)
- Roberta A Gottlieb
- From the Cedars-Sinai Heart Institute and the Barbra Streisand Women's Heart Center Cedars-Sinai Medical Center, Los Angeles, CA.
| | - Allen M Andres
- From the Cedars-Sinai Heart Institute and the Barbra Streisand Women's Heart Center Cedars-Sinai Medical Center, Los Angeles, CA
| | - Jon Sin
- From the Cedars-Sinai Heart Institute and the Barbra Streisand Women's Heart Center Cedars-Sinai Medical Center, Los Angeles, CA
| | - David P J Taylor
- From the Cedars-Sinai Heart Institute and the Barbra Streisand Women's Heart Center Cedars-Sinai Medical Center, Los Angeles, CA
| |
Collapse
|
67
|
Abstract
Cardiovascular disease is the leading cause of death worldwide. As such, there is great interest in identifying novel mechanisms that govern the cardiovascular response to disease-related stress. First described in failing hearts, autophagy within the cardiovascular system has been widely characterized in cardiomyocytes, cardiac fibroblasts, endothelial cells, vascular smooth muscle cells, and macrophages. In all cases, a window of optimal autophagic activity appears to be critical to the maintenance of cardiovascular homeostasis and function; excessive or insufficient levels of autophagic flux can each contribute to heart disease pathogenesis. In this Review, we discuss the potential for targeting autophagy therapeutically and our vision for where this exciting biology may lead in the future.
Collapse
|
68
|
Linton PJ, Gurney M, Sengstock D, Mentzer RM, Gottlieb RA. This old heart: Cardiac aging and autophagy. J Mol Cell Cardiol 2014; 83:44-54. [PMID: 25543002 DOI: 10.1016/j.yjmcc.2014.12.017] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 12/10/2014] [Accepted: 12/16/2014] [Indexed: 01/02/2023]
Abstract
Autophagy, a cellular housekeeping process, is essential to maintain tissue homeostasis, particularly in long-lived cells such as cardiomyocytes. Autophagic activity declines with age and may explain many features of age-related cardiac dysfunction. In this review we summarize the current state of knowledge regarding age-related changes in autophagy in the heart. Recent findings from studies in human hearts are presented, including evidence that the autophagic response is intact in the aged human heart. Impaired autophagic clearance of protein aggregates or deteriorating mitochondria will have multiple consequences including increased arrhythmia risk, decreased contractile function, reduced tolerance to ischemic stress, and increased inflammation; thus autophagy represents a potentially important therapeutic target to mitigate the cardiac consequences of aging. This article is part of a Special Issue entitled CV Aging.
Collapse
Affiliation(s)
- Phyllis-Jean Linton
- Donald P. Shiley BioScience Center, San Diego State University, San Diego, CA, USA
| | - Michael Gurney
- Donald P. Shiley BioScience Center, San Diego State University, San Diego, CA, USA
| | - David Sengstock
- Division of Geriatric Medicine, Oakwood Hospital, Dearborn, MI, USA; Department of Internal Medicine, Wayne State University School of Medicine, Detroit, MI, USA
| | - Robert M Mentzer
- Cardiovascular Research Institute, Departments of Surgery and Physiology, Wayne State University School of Medicine, Detroit, MI, USA; Heart Institute and Barbra Streisand Women's Heart Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Roberta A Gottlieb
- Heart Institute and Barbra Streisand Women's Heart Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
69
|
Drews O, Taegtmeyer H. Targeting the ubiquitin-proteasome system in heart disease: the basis for new therapeutic strategies. Antioxid Redox Signal 2014; 21:2322-43. [PMID: 25133688 PMCID: PMC4241867 DOI: 10.1089/ars.2013.5823] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
SIGNIFICANCE Novel therapeutic strategies to treat heart failure are greatly needed. The ubiquitin-proteasome system (UPS) affects the structure and function of cardiac cells through targeted degradation of signaling and structural proteins. This review discusses both beneficial and detrimental consequences of modulating the UPS in the heart. RECENT ADVANCES Proteasome inhibitors were first used to test the role of the UPS in cardiac disease phenotypes, indicating therapeutic potential. In early cardiac remodeling and pathological hypertrophy with increased proteasome activities, proteasome inhibition prevented or restricted disease progression and contractile dysfunction. Conversely, enhancing proteasome activities by genetic manipulation, pharmacological intervention, or ischemic preconditioning also improved the outcome of cardiomyopathies and infarcted hearts with impaired cardiac and UPS function, which is, at least in part, caused by oxidative damage. CRITICAL ISSUES An understanding of the UPS status and the underlying mechanisms for its potential deregulation in cardiac disease is critical for targeted interventions. Several studies indicate that type and stage of cardiac disease influence the dynamics of UPS regulation in a nonlinear and multifactorial manner. Proteasome inhibitors targeting all proteasome complexes are associated with cardiotoxicity in humans. Furthermore, the type and dosage of proteasome inhibitor impact the pathogenesis in nonuniform ways. FUTURE DIRECTIONS Systematic analysis and targeting of individual UPS components with established and innovative tools will unravel and discriminate regulatory mechanisms that contribute to and protect against the progression of cardiac disease. Integrating this knowledge in drug design may reduce adverse effects on the heart as observed in patients treated with proteasome inhibitors against noncardiac diseases, especially cancer.
Collapse
Affiliation(s)
- Oliver Drews
- 1 Division of Cardiovascular Physiology, Institute of Physiology and Pathophysiology , Heidelberg University, Heidelberg, Germany
| | | |
Collapse
|
70
|
Mahr C, Gundry RL. Hold or fold--proteins in advanced heart failure and myocardial recovery. Proteomics Clin Appl 2014; 9:121-33. [PMID: 25331159 DOI: 10.1002/prca.201400100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 09/17/2014] [Accepted: 10/14/2014] [Indexed: 12/14/2022]
Abstract
Advanced heart failure (AHF) describes the subset of heart failure patients refractory to conventional medical therapy. For some AHF patients, the use of mechanical circulatory support (MCS) provides an intermediary "bridge" step for transplant-eligible patients or an alternative therapy for transplant-ineligible patients. Over the past 20 years, clinical observations have revealed that approximately 1% of patients with MCS undergo significant reverse remodeling to the point where the device can be explanted. Unfortunately, it is unclear why some patients experience durable, sustained myocardial remission, while others redevelop heart failure (i.e. which hearts "hold" and which hearts "fold"). In this review, we outline unmet clinical needs related to treating patients with MCS, provide an overview of protein dynamics in the reverse-remodeling process, and propose specific areas where we expect MS and proteomic analyses will have significant impact on our understanding of disease progression, molecular mechanisms of recovery, and provide new markers with prognostic value that can positively impact patient care. Complimentary perspectives are provided with the goal of making this important topic accessible and relevant to both a clinical and basic science audience, as the intersection of these disciplines is required to advance the field.
Collapse
Affiliation(s)
- Claudius Mahr
- Division of Cardiology, University of Washington, Seattle, WA, USA
| | | |
Collapse
|
71
|
Prescimone T, Masotti S, D’Amico A, Caruso R, Cabiati M, Caselli C, Viglione F, Verde A, Del Ry S, Giannessi D. Cardiac molecular markers of programmed cell death are activated in end-stage heart failure patients supported by left ventricular assist device. Cardiovasc Pathol 2014; 23:272-82. [DOI: 10.1016/j.carpath.2014.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Revised: 03/24/2014] [Accepted: 04/07/2014] [Indexed: 10/25/2022] Open
|
72
|
Nsair A, Liem DA, Cadeiras M, Cheng RK, Allareddy M, Kwon M, Shemin R, Deng MC. Molecular basis of recovering on mechanical circulatory support. Heart Fail Clin 2014; 10:S57-62. [PMID: 24262353 DOI: 10.1016/j.hfc.2013.08.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Our insights into different system levels of mechanisms by left ventricular assist device support are increasing and suggest a complex regulatory system of overlapping biological processes. To develop novel decision-making strategies and patient selection criteria, heart failure and reverse cardiac remodeling should be conceptualized and explored by a multifaceted research strategy of transcriptomics, metabolomics, proteomics, molecular biology, and bioinformatics. Knowledge of the molecular mechanisms of reverse cardiac remodeling is in its early stages, and comprehensive reconstruction of the underlying networks is necessary.
Collapse
Affiliation(s)
- Ali Nsair
- University of California, Ahmanson-UCLA Cardiomyopathy Center, 100 Medical Plaza, Suite 630, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | | | |
Collapse
|
73
|
Prescimone T, D'Amico A, Caselli C, Cabiati M, Viglione F, Caruso R, Verde A, Del Ry S, Trivella MG, Giannessi D. Caspase-1 transcripts in failing human heart after mechanical unloading. Cardiovasc Pathol 2014; 24:11-8. [PMID: 25200478 DOI: 10.1016/j.carpath.2014.08.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 08/06/2014] [Accepted: 08/06/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Caspase (Casp)-1 has been indicated as a molecular target capable of preventing the progression of cardiovascular diseases, including heart failure (HF), due to its central role in promoting inflammation and cardiomyocyte loss. The aim of this study was to assess whether Left Ventricular Assist Device (LVAD) implantation modifies the inflammatory and apoptotic profile in the heart through the modulation of Casp-1 expression level. METHODS Cardiac tissue was collected from end-stage HF patients before LVAD implant (pre-LVAD group, n=22) and at LVAD removal (post-LVAD, n=6), and from stable HF patients on medical therapy without prior circulatory support (HTx, n=7) at heart transplantation, as control. The cardiac expression of Casp-1, of its inhibitors caspase recruitment domain (CARD) only protein (COP) and CARD family, member 18 (ICEBERG), was evaluated by real-time PCR in the three groups of patients. RESULTS Casp-1 was increased in the pre-LVAD group compared to HTx (p=0.006), while on the contrary the ICEBERG level was significantly decreased in pre-LVAD with respect to HTx patients (p<0.001); no difference in COP expression level was found. CONCLUSIONS This study describes a specific pattern of the Casp-1 system associated with inflammation and apoptosis markers in patients who require LVAD insertion. The inflammation could be the key process regulating, in a negative loop, Casp-1 signaling and its down-stream effects, apoptosis included.
Collapse
Affiliation(s)
- Tommaso Prescimone
- CNR Institute of Clinical Physiology, Laboratory of Cardiovascular Biochemistry, Pisa, Italy
| | | | - Chiara Caselli
- CNR Institute of Clinical Physiology, Laboratory of Cardiovascular Biochemistry, Pisa, Italy
| | - Manuela Cabiati
- CNR Institute of Clinical Physiology, Laboratory of Cardiovascular Biochemistry, Pisa, Italy
| | - Federica Viglione
- CNR Institute of Clinical Physiology, Laboratory of Cardiovascular Biochemistry, Pisa, Italy
| | - Raffaele Caruso
- CNR Institute of Clinical Physiology, Cardiovascular Department, Niguarda Cà Granda Hospital, Milan, Italy
| | - Alessandro Verde
- CardioThoracic and Vascular Department, "A. De Gasperis" Niguarda Ca' Granda Hospital, Milan, Italy
| | - Silvia Del Ry
- CNR Institute of Clinical Physiology, Laboratory of Cardiovascular Biochemistry, Pisa, Italy
| | - Maria Giovanna Trivella
- CNR Institute of Clinical Physiology, Laboratory of Cardiovascular Biochemistry, Pisa, Italy
| | - Daniela Giannessi
- CNR Institute of Clinical Physiology, Laboratory of Cardiovascular Biochemistry, Pisa, Italy.
| |
Collapse
|
74
|
Wang Q, Yang L, Hua Y, Nair S, Xu X, Ren J. AMP-activated protein kinase deficiency rescues paraquat-induced cardiac contractile dysfunction through an autophagy-dependent mechanism. Toxicol Sci 2014; 142:6-20. [PMID: 25092649 DOI: 10.1093/toxsci/kfu158] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
AIM Paraquat, a quaternary nitrogen herbicide, is a highly toxic prooxidant resulting in multi-organ failure including the heart although the underlying mechanism still remains elusive. This study was designed to examine the role of the cellular fuel sensor AMP-activated protein kinase (AMPK) in paraquat-induced cardiac contractile and mitochondrial injury. RESULTS Wild-type and transgenic mice with overexpression of a mutant AMPK α2 subunit (kinase dead, KD), with reduced activity in both α1 and α2 subunits, were administered with paraquat (45 mg/kg) for 48 h. Paraquat elicited cardiac mechanical anomalies including compromised echocardiographic parameters (elevated left ventricular end-systolic diameter and reduced factional shortening), suppressed cardiomyocyte contractile function, intracellular Ca(2+) handling, reduced cell survival, and overt mitochondrial damage (loss in mitochondrial membrane potential). In addition, paraquat treatment promoted phosphorylation of AMPK and autophagy. Interestingly, deficiency in AMPK attenuated paraquat-induced cardiac contractile and intracellular Ca(2+) derangement. The beneficial effect of AMPK inhibition was associated with inhibition of the AMPK-TSC-mTOR-ULK1 signaling cascade. In vitro study revealed that inhibitors for AMPK and autophagy attenuated paraquat-induced cardiomyocyte contractile dysfunction. CONCLUSION Taken together, our findings revealed that AMPK may mediate paraquat-induced myocardial anomalies possibly by regulating the AMPK/mTOR-dependent autophagy.
Collapse
Affiliation(s)
- Qiurong Wang
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, Wyoming 82071
| | - Lifang Yang
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, Wyoming 82071 Department of Anesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Yinan Hua
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, Wyoming 82071
| | - Sreejayan Nair
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, Wyoming 82071
| | - Xihui Xu
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, Wyoming 82071
| | - Jun Ren
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, Wyoming 82071
| |
Collapse
|
75
|
|
76
|
Abstract
Cardiovascular disease, including heart failure, is a principal cause of death in individuals with obesity and diabetes. However, the mechanisms of obesity- and diabetes-induced heart disease are multifaceted and remain to be clearly defined. Of relevance to this review, there is currently great research and clinical interest in the endocrine effects of adipokines on the myocardium and their role in heart failure. We will discuss the potential significance of adipokines in the pathogenesis of heart failure via their ability to regulate remodeling events including metabolism, hypertrophy, fibrosis, and cell death. As an excellent example, we will first focus on adiponectin which is best known to confer numerous cardioprotective effects. However, we comprehensively discuss the existing literature that highlights it would be naive to assume that this was always the case. We also focus on lipocalin-2 which mediates pro-inflammatory and pro-apoptotic effects. It is important when studying actions of adipokines to integrate cellular and mechanistic analyses and translate these to physiologically relevant in vivo models and clinical studies. However, assimilating studies on numerous cardiac remodeling events which ultimately dictate cardiac dysfunction into a unifying conclusion is challenging. Nevertheless, there is undoubted potential for the use of adipokines as robust biomarkers and appropriate therapeutic targets in heart failure.
Collapse
Affiliation(s)
- Min Park
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
| | | |
Collapse
|
77
|
Abstract
Whether an element of routine housekeeping or in the setting of imminent disaster, it is a good idea to get one’s affairs in order. Autophagy, the process of recycling organelles and protein aggregates, is a basal homeostatic process and an evolutionarily conserved response to starvation and other forms of metabolic stress. Our understanding of the role of autophagy in the heart is changing rapidly as new information becomes available. This review examines the role of autophagy in the heart in the setting of cardioprotection, hypertrophy, and heart failure. Contradictory findings are reconciled in light of recent developments. The preponderance of evidence favors a beneficial role for autophagy in the heart under most conditions.
Collapse
Affiliation(s)
- Roberta A Gottlieb
- Donald P. Shiley BioScience Center, San Diego State University, San Diego, CA, USA,
| | | |
Collapse
|
78
|
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]
|
79
|
Mei Y, Thompson MD, Cohen RA, Tong X. Autophagy and oxidative stress in cardiovascular diseases. Biochim Biophys Acta Mol Basis Dis 2014; 1852:243-51. [PMID: 24834848 DOI: 10.1016/j.bbadis.2014.05.005] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 04/29/2014] [Accepted: 05/03/2014] [Indexed: 10/25/2022]
Abstract
Autophagy is a highly conserved degradation process by which intracellular components, including soluble macromolecules (e.g. nucleic acids, proteins, carbohydrates, and lipids) and dysfunctional organelles (e.g. mitochondria, ribosomes, peroxisomes, and endoplasmic reticulum) are degraded by the lysosome. Autophagy is orchestrated by the autophagy related protein (Atg) composed protein complexes to form autophagosomes, which fuse with lysosomes to generate autolysosomes where the contents are degraded to provide energy for cell survival in response to environmental and cellular stress. Autophagy is an important player in cardiovascular disease development such as atherosclerosis, cardiac ischemia/reperfusion, cardiomyopathy, heart failure and hypertension. Autophagy in particular contributes to cardiac ischemia, hypertension and diabetes by interaction with reactive oxygen species generated in endoplasmic reticulum and mitochondria. This review highlights the dual role of autophagy in cardiovascular disease development. Full recognition of autophagy as an adaptive or maladaptive response would provide potential new strategies for cardiovascular disease prevention and management. This article is part of a Special Issue entitled: Autophagy and protein quality control in cardiometabolic diseases.
Collapse
Affiliation(s)
- Yu Mei
- Vascular Biology Section, Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| | - Melissa D Thompson
- Vascular Biology Section, Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| | - Richard A Cohen
- Vascular Biology Section, Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| | - XiaoYong Tong
- Vascular Biology Section, Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA.
| |
Collapse
|
80
|
Gedik N, Thielmann M, Kottenberg E, Peters J, Jakob H, Heusch G, Kleinbongard P. No evidence for activated autophagy in left ventricular myocardium at early reperfusion with protection by remote ischemic preconditioning in patients undergoing coronary artery bypass grafting. PLoS One 2014; 9:e96567. [PMID: 24797938 PMCID: PMC4010496 DOI: 10.1371/journal.pone.0096567] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 04/08/2014] [Indexed: 11/30/2022] Open
Abstract
Objective Remote ischemic preconditioning (RIPC) by repeated brief limb ischemia/reperfusion reduces myocardial injury in patients undergoing coronary artery bypass grafting (CABG). Activation of signal transducer and activator of transcription 5 (STAT5) in left ventricular (LV) myocardium at early reperfusion is associated with such protection. Autophagy, i.e., removal of dysfunctional cellular components through lysosomes, has been proposed as one mechanism of cardioprotection. Therefore, we analyzed whether or not the protection by RIPC is associated with activated autophagy. Methods CABG patients were randomized to undergo RIPC (3×5 min blood pressure cuff inflation/5 min deflation) or placebo (cuff deflated) before skin incision (n = 10/10). Transmural myocardial biopsies were taken from the LV before cardioplegia (baseline) and at early (5–10 min) reperfusion. RIPC-induced protection was reflected by decreased serum troponin I concentration area under the curve (194±17 versus 709±129 ng/ml × 72 h, p = 0.002). Western blotting for beclin-1-phosphorylation and protein expression of autophagy-related gene 5–12 (ATG5-12) complex, light chain 3 (LC3), parkin, and p62 was performed. STAT3-, STAT5- and extracellular signal-regulated protein kinase 1/2 (ERK1/2)-phosphorylation was used as positive control to confirm signal activation by ischemia/reperfusion. Results Signals of all analyzed autophagy proteins did not differ between baseline and early reperfusion and not between RIPC and placebo. STAT5-phosphorylation was greater at early reperfusion only with RIPC (2.2-fold, p = 0.02). STAT3- and ERK1/2-phosphorylation were greater at early reperfusion with placebo and RIPC (≥2.7-fold versus baseline, p≤0.05). Conclusion Protection through RIPC in patients undergoing CABG surgery does not appear to be associated with enhanced autophagy in LV myocardium at early reperfusion.
Collapse
Affiliation(s)
- Nilgün Gedik
- Institut für Pathophysiologie, Universitätsklinikum Essen, Universität Duisburg-Essen, Essen, Germany
| | - Matthias Thielmann
- Klinik für Thorax- und Kardiovaskuläre Chirurgie, Universitätsklinikum Essen, Universität Duisburg-Essen, Essen, Germany
| | - Eva Kottenberg
- Klinik für Anästhesiologie und Intensivmedizin, Universitätsklinikum Essen, Universität Duisburg-Essen, Essen, Germany
| | - Jürgen Peters
- Klinik für Anästhesiologie und Intensivmedizin, Universitätsklinikum Essen, Universität Duisburg-Essen, Essen, Germany
| | - Heinz Jakob
- Klinik für Thorax- und Kardiovaskuläre Chirurgie, Universitätsklinikum Essen, Universität Duisburg-Essen, Essen, Germany
| | - Gerd Heusch
- Institut für Pathophysiologie, Universitätsklinikum Essen, Universität Duisburg-Essen, Essen, Germany
| | - Petra Kleinbongard
- Institut für Pathophysiologie, Universitätsklinikum Essen, Universität Duisburg-Essen, Essen, Germany
- * E-mail:
| |
Collapse
|
81
|
Abstract
Cardiovascular disease (CVD) remains the leading cause of death worldwide, despite the significant advances in medicine. Autophagy, a process of self-cannibalization employed by mammalian cells for the recycling of cellular contents, is altered not only in a number of CVDs, but in other diseases, as well. Many FDA-approved drugs are known to induce autophagy-mediated side effects in the cardiovascular system. In some cases, such drug-induced autophagy could be harnessed and used for treating CVD, greatly reducing the duration and cost of CVD treatments. However, because the induction of autophagy in cardiovascular targets can be both adaptive and maladaptive under specific settings, the challenge is to determine whether the changes stimulated by drug-induced autophagy are, in fact, beneficial. In this review, we surveyed a number of CVDs in which autophagy is known to occur, and we also address the role of FDA-approved drugs for which autophagy-mediated side effects occur within the cardiovascular system. The therapeutic potential of using small molecule modulators of autophagy in the management of CVD progression is discussed.
Collapse
|
82
|
Sabe AA, Elmadhun NY, Dalal RS, Robich MP, Sellke FW. Resveratrol regulates autophagy signaling in chronically ischemic myocardium. J Thorac Cardiovasc Surg 2014; 147:792-8; Discussion 798-9. [PMID: 24267781 PMCID: PMC3947125 DOI: 10.1016/j.jtcvs.2013.06.062] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 06/06/2013] [Accepted: 06/18/2013] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Autophagy is a cellular process by which damaged components are removed. Although autophagy can result in cell death, when optimally regulated, it might be cardioprotective. Resveratrol is a naturally occurring polyphenol also believed to be cardioprotective. Using a clinically relevant swine model of metabolic syndrome, we investigated the effects of resveratrol on autophagy in the chronically ischemic myocardium. METHODS Yorkshire swine were fed a regular diet (n = 7), a high cholesterol diet (n = 7), or a high cholesterol diet with supplemental resveratrol (n = 6). After 4 weeks, an ameroid constrictor was surgically placed on the left circumflex artery to induce chronic myocardial ischemia. The diets were continued another 7 weeks, and then the ischemic and nonischemic myocardium were harvested for protein analysis. RESULTS In the ischemic myocardium, a high cholesterol diet partly attenuated the autophagy, as determined by an increase in phosphorylated mammalian target of rapamycin (p-mTOR) and a decrease in p70 S6 kinase (P70S6K), lysosome-associated membrane protein (LAMP)-2, and autophagy-related gene 12-5 conjugate (ATG 12-5; P < .05). The addition of resveratrol blunted many of these changes, because the p-mTOR, P70S6K, and LAMP-2 levels were not significantly altered from those of the pigs fed a regular diet. Other autophagy markers were increased with a high cholesterol diet, including light chain 3A-II and beclin 1 (P < .05). In the nonischemic myocardium, beclin 1 was decreased in the high cholesterol-fed pigs (P < .05); otherwise no significant changes in protein expression were noted among the 3 groups. CONCLUSIONS In the chronically ischemic myocardium, resveratrol partly reversed the effects of a high cholesterol diet on autophagy. This might be a mechanism by which resveratrol exerts its cardioprotective effects.
Collapse
Affiliation(s)
- Ashraf A Sabe
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Brown University Warren Alpert School of Medicine, Providence, RI
| | - Nassrene Y Elmadhun
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Brown University Warren Alpert School of Medicine, Providence, RI
| | - Rahul S Dalal
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Brown University Warren Alpert School of Medicine, Providence, RI
| | - Michael P Robich
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Brown University Warren Alpert School of Medicine, Providence, RI
| | - Frank W Sellke
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Brown University Warren Alpert School of Medicine, Providence, RI.
| |
Collapse
|
83
|
Morales CR, Pedrozo Z, Lavandero S, Hill JA. Oxidative stress and autophagy in cardiovascular homeostasis. Antioxid Redox Signal 2014; 20:507-18. [PMID: 23641894 PMCID: PMC3894700 DOI: 10.1089/ars.2013.5359] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
SIGNIFICANCE Autophagy is an evolutionarily ancient process of intracellular protein and organelle recycling required to maintain cellular homeostasis in the face of a wide variety of stresses. Dysregulation of reactive oxygen species (ROS) and reactive nitrogen species (RNS) leads to oxidative damage. Both autophagy and ROS/RNS serve pathological or adaptive roles within cardiomyocytes, depending on the context. RECENT ADVANCES ROS/RNS and autophagy communicate with each other via both transcriptional and post-translational events. This cross talk, in turn, regulates the structural integrity of cardiomyocytes, promotes proteostasis, and reduces inflammation, events critical to disease pathogenesis. CRITICAL ISSUES Dysregulation of either autophagy or redox state has been implicated in many cardiovascular diseases. Cardiomyocytes are rich in mitochondria, which make them particularly sensitive to oxidative damage. Maintenance of mitochondrial homeostasis and elimination of defective mitochondria are each critical to the maintenance of redox homeostasis. FUTURE DIRECTIONS The complex interplay between autophagy and oxidative stress underlies a wide range of physiological and pathological events and its elucidation holds promise of potential clinical applicability.
Collapse
Affiliation(s)
- Cyndi R Morales
- 1 Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center , Dallas, Texas
| | | | | | | |
Collapse
|
84
|
Sun A, Cheng Y, Zhang Y, Zhang Q, Wang S, Tian S, Zou Y, Hu K, Ren J, Ge J. Aldehyde dehydrogenase 2 ameliorates doxorubicin-induced myocardial dysfunction through detoxification of 4-HNE and suppression of autophagy. J Mol Cell Cardiol 2014; 71:92-104. [PMID: 24434637 DOI: 10.1016/j.yjmcc.2014.01.002] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 12/06/2013] [Accepted: 01/03/2014] [Indexed: 01/24/2023]
Abstract
Mitochondrial aldehyde dehydrogenase (ALDH2) protects against cardiac injury via reducing production of 4-hydroxynonenal (4-HNE) and ROS. This study was designed to examine the impact of ALDH2 on doxorubicin (DOX)-induced cardiomyopathy and mechanisms involved with a focus on autophagy. 4-HNE and autophagic markers were detected by Western blotting in ventricular tissues from normal donors and patients with idiopathic dilated cardiomyopathy. Cardiac function, 4-HNE and levels of autophagic markers were detected in WT, ALDH2 knockout or ALDH2 transfected mice treated with or without DOX. Autophagy regulatory signaling including PI-3K, AMPK and Akt was examined in DOX-treated cardiomyocytes incubated with or without ALDH2 activator Alda-1. DOX-induced myocardial dysfunction, upregulation of 4-HNE and autophagic proteins were further aggravated in ALDH2 knockout mice while they were ameliorated in ALDH2 transfected mice. DOX downregulated Class I and upregulated Class III PI3-kinase, the effect of which was augmented by ALDH2 deletion. Accumulation of 4-HNE and autophagic protein markers in DOX-induced cardiomyocytes was significantly reduced by Alda-1. DOX depressed phosphorylated Akt but not AMPK, the effect was augmented by ALDH2 knockout. The autophagy inhibitor 3-MA attenuated, whereas autophagy inducer rapamycin mimicked DOX-induced cardiomyocyte contractile defects. In addition, rapamycin effectively mitigated Alda-1-offered protective action against DOX-induced cardiomyocyte dysfunction. Our data further revealed downregulated ALDH2 and upregulated autophagy levels in the hearts from patients with dilated cardiomyopathy. Taken together, our findings suggest that inhibition of 4-HNE and autophagy may be a plausible mechanism underscoring ALDH2-offered protection against DOX-induced cardiac defect. This article is part of a Special Issue entitled "Protein Quality Control, the Ubiquitin Proteasome System, and Autophagy".
Collapse
Affiliation(s)
- Aijun Sun
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China.
| | - Yong Cheng
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Heart Centre of Zhengzhou Ninth People's Hospital, Zhengzhou, Henan 450000, China
| | - Yingmei Zhang
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | - Qian Zhang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Cardiology, Branch of Shanghai First People's Hospital, Shanghai 200050, China
| | - Shijun Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Shan Tian
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Kai Hu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | - Junbo Ge
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
85
|
Abstract
Background Recent evidence has depicted a role of macrophage migration inhibitory factor (MIF) in cardiac homeostasis under pathological conditions. This study was designed to evaluate the role of MIF in doxorubicin‐induced cardiomyopathy and the underlying mechanism involved with a focus on autophagy. Methods and Results Wild‐type (WT) and MIF knockout (MIF−/−) mice were given saline or doxorubicin (20 mg/kg cumulative, i.p.). A cohort of WT and MIF−/− mice was given rapamycin (6 mg/kg, i.p.) with or without bafilomycin A1 (BafA1, 3 μmol/kg per day, i.p.) for 1 week prior to doxorubicin challenge. To consolidate a role for MIF in the maintenance of cardiac homeostasis following doxorubicin challenge, recombinant mouse MIF (rmMIF) was given to MIF−/− mice challenged with or without doxorubicin. Echocardiographic, cardiomyocyte function, and intracellular Ca2+ handling were evaluated. Autophagy and apoptosis were examined. Mitochondrial morphology and function were examined using transmission electron microscopy, JC‐1 staining, MitoSOX Red fluorescence, and mitochondrial respiration complex assay. DHE staining was used to evaluate reactive oxygen species (ROS) generation. MIF knockout exacerbated doxorubicin‐induced mortality and cardiomyopathy (compromised fractional shortening, cardiomyocyte and mitochondrial function, apoptosis, and ROS generation). These detrimental effects of doxorubicin were accompanied by defective autophagolysosome formation, the effect of which was exacerbated by MIF knockout. Rapamycin pretreatment rescued doxorubicin‐induced cardiomyopathy in WT and MIF−/− mice. Blocking autophagolysosome formation using BafA1 negated the cardioprotective effect of rapamycin and rmMIF. Conclusions Our data suggest that MIF serves as an indispensable cardioprotective factor against doxorubicin‐induced cardiomyopathy with an underlying mechanism through facilitating autophagolysosome formation.
Collapse
Affiliation(s)
- Xihui Xu
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, School of Pharmacy, Laramie, WY
| | | | | |
Collapse
|
86
|
Wang X, Robbins J. Proteasomal and lysosomal protein degradation and heart disease. J Mol Cell Cardiol 2013; 71:16-24. [PMID: 24239609 DOI: 10.1016/j.yjmcc.2013.11.006] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 11/04/2013] [Accepted: 11/06/2013] [Indexed: 01/08/2023]
Abstract
In the cell, the proteasome and lysosomes represent the most important proteolytic machineries, responsible for the protein degradation in the ubiquitin-proteasome system (UPS) and autophagy, respectively. Both the UPS and autophagy are essential to protein quality and quantity control. Alterations in cardiac proteasomal and lysosomal degradation are remarkably associated with most heart disease in humans and are implicated in the pathogenesis of congestive heart failure. Studies carried out in animal models and in cell culture have begun to establish both sufficiency and, in some cases, the necessity of proteasomal functional insufficiency or lysosomal insufficiency as a major pathogenic factor in the heart. This review article highlights some recent advances in the research into proteasome and lysosome protein degradation in relation to cardiac pathology and examines the emerging evidence for enhancing degradative capacities of the proteasome and/or lysosome as a new therapeutic strategy for heart disease. This article is part of a Special Issue entitled "Protein Quality Control, the Ubiquitin Proteasome System, and Autophagy".
Collapse
Affiliation(s)
- Xuejun Wang
- Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion, SD 57069, USA.
| | - Jeffrey Robbins
- Division of Molecular Cardiovascular Biology, The Heart Institute, Department of Pediatrics, The Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| |
Collapse
|
87
|
Nakayama H, Otsu K. Translation of hemodynamic stress to sterile inflammation in the heart. Trends Endocrinol Metab 2013; 24:546-53. [PMID: 23850260 DOI: 10.1016/j.tem.2013.06.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 06/13/2013] [Accepted: 06/15/2013] [Indexed: 12/19/2022]
Abstract
Recently, growing evidence suggests that cardiac inflammation contributes to progression of heart failure (HF). However, the precise mechanism has been elusive. Autophagy is well-known phenomenon which plays essential roles in the maintenance of cardiomyocyte homeostasis by clearing damaged proteins and organelles, and dysfunction of this system evokes HF. Although emerging roles of mitochondria in inflammasome development are highlighted in immune cells, an involvement in the heart has not been defined until recently. This review discusses recent advances in understanding the complex mechanisms underlying cardiac inflammation: these studies have revealed that a combination of mitochondrial autophagy and innate immune responses to mitochondrial DNA during increased hemodynamic stress contribute to cardiac inflammation.
Collapse
Affiliation(s)
- Hiroyuki Nakayama
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 565-0871, Japan
| | | |
Collapse
|
88
|
Chen WT, Hung KC, Wen MS, Hsu PY, Chen TH, Wang HD, Fang JT, Shie SS, Wang CY. Impaired leukocytes autophagy in chronic kidney disease patients. Cardiorenal Med 2013; 3:254-64. [PMID: 24474954 DOI: 10.1159/000356212] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 10/08/2013] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Proteins and cytoplasmic organelles undergo degradation and recycling via autophagy; its role in patients with chronic kidney disease (CKD) is still unclear. We hypothesize that impaired kidney function causes autophagy activation failure. METHODS We included 60 patients with stage 5 CKD and 30 age- and sex-matched healthy subjects as controls. Patients with conditions that could affect autophagy were excluded. Leukocytes were isolated and analyzed from peripheral blood samples collected after overnight fasting and 2 h after breakfast. RESULTS Overnight fasting induced conversion of microtubule-associated protein-1 light chain 3 I to II (γLC3) and increased mRNA levels of the autophagy-related gene 5 (Atg5) and Beclin-1 in healthy subjects, which were nearly absent in CKD patients (p = 0.0001). Moreover, no significant difference in autophagy activation was observed between CKD patients with or without hemodialysis. Correlation studies showed that γLC3 was negatively associated with the left atrium size. Changes in Atg5 transcript levels were negatively associated with the left ventricular end-diastolic diameter, and changes in Beclin-1 transcript levels were negatively associated with the mitral inflow E- and A-wave sizes. CONCLUSION These data suggest that CKD patients have impaired autophagy activation, which cannot be reversed with hemodialysis and is closely related to their cardiac abnormalities.
Collapse
Affiliation(s)
- Wei-Ting Chen
- Department of Cardiology, Chang Gung University College of Medicine, Taoyuan, ROC
| | - Kuo-Chun Hung
- Department of Cardiology, Chang Gung University College of Medicine, Taoyuan, ROC
| | - Ming-Shien Wen
- Department of Cardiology, Chang Gung University College of Medicine, Taoyuan, ROC
| | - Po-Yaur Hsu
- Kidney Research Center, Department of Nephrology, Chang Gung University College of Medicine, Taoyuan, ROC
| | - Tien-Hsing Chen
- Department of Cardiology, Chang Gung University College of Medicine, Taoyuan, ROC
| | - Horng-Dar Wang
- Institute of Biotechnology and Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan, ROC
| | - Ji-Tseng Fang
- Kidney Research Center, Department of Nephrology, Chang Gung University College of Medicine, Taoyuan, ROC
| | - Shian-Sen Shie
- Division of Infectious Diseases, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, ROC
| | - Chao-Yung Wang
- Department of Cardiology, Chang Gung University College of Medicine, Taoyuan, ROC
| |
Collapse
|
89
|
Beloukas AI, Magiorkinis E, Tsoumakas TL, Kosma AG, Diamantis A. Milestones in the History of Research on Cardiac Energy Metabolism. Can J Cardiol 2013; 29:1504-11. [DOI: 10.1016/j.cjca.2012.10.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Revised: 10/03/2012] [Accepted: 10/09/2012] [Indexed: 12/21/2022] Open
|
90
|
Brinks H, Giraud MN, Segiser A, Ferrié C, Longnus S, Ullrich ND, Koch WJ, Most P, Carrel TP, Tevaearai HT. Dynamic patterns of ventricular remodeling and apoptosis in hearts unloaded by heterotopic transplantation. J Heart Lung Transplant 2013; 33:203-10. [PMID: 24315785 DOI: 10.1016/j.healun.2013.10.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Revised: 09/22/2013] [Accepted: 10/01/2013] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Mechanical unloading of failing hearts can trigger functional recovery but results in progressive atrophy and possibly detrimental adaptation. In an unbiased approach, we examined the dynamic effects of unloading duration on molecular markers indicative of myocardial damage, hypothesizing that potential recovery may be improved by optimized unloading time. METHODS Heterotopically transplanted normal rat hearts were harvested at 3, 8, 15, 30, and 60 days. Forty-seven genes were analyzed using TaqMan-based microarray, Western blot, and immunohistochemistry. RESULTS In parallel with marked atrophy (22% to 64% volume loss at 3 respectively 60 days), expression of myosin heavy-chain isoforms (MHC-α/-β) was characteristically switched in a time-dependent manner. Genes involved in tissue remodeling (FGF-2, CTGF, TGFb, IGF-1) were increasingly upregulated with duration of unloading. A distinct pattern was observed for genes involved in generation of contractile force; an indiscriminate early downregulation was followed by a new steady-state below normal. For pro-apoptotic transcripts bax, bnip-3, and cCasp-6 and -9 mRNA levels demonstrated a slight increase up to 30 days unloading with pronunciation at 60 days. Findings regarding cell death were confirmed on the protein level. Proteasome activity indicated early increase of protein degradation but decreased below baseline in unloaded hearts at 60 days. CONCLUSIONS We identified incrementally increased apoptosis after myocardial unloading of the normal rat heart, which is exacerbated at late time points (60 days) and inversely related to loss of myocardial mass. Our findings suggest an irreversible detrimental effect of long-term unloading on myocardium that may be precluded by partial reloading and amenable to molecular therapeutic intervention.
Collapse
Affiliation(s)
- Henriette Brinks
- Department of Cardiac and Vascular Surgery, Inselspital University Hospital, Berne, Switzerland.
| | - Marie-Noelle Giraud
- Cardiology, Department of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Adrian Segiser
- Department of Cardiac and Vascular Surgery, Inselspital University Hospital, Berne, Switzerland
| | - Celine Ferrié
- Department of Cardiac and Vascular Surgery, Inselspital University Hospital, Berne, Switzerland
| | - Sarah Longnus
- Department of Cardiac and Vascular Surgery, Inselspital University Hospital, Berne, Switzerland
| | - Nina D Ullrich
- Institute of Physiology, University of Berne, Berne, Switzerland
| | - Walter J Koch
- Center for Translational Medicine, Department of Pharmacology, Temple University, Philadelphia, Pennsylvania
| | - Patrick Most
- Center for Molecular and Translational Cardiology, Department of Internal Medicine III, University of Heidelberg, Germany
| | - Thierry P Carrel
- Department of Cardiac and Vascular Surgery, Inselspital University Hospital, Berne, Switzerland
| | - Hendrik T Tevaearai
- Department of Cardiac and Vascular Surgery, Inselspital University Hospital, Berne, Switzerland
| |
Collapse
|
91
|
Proof of myocardial autophagy by combining antigen retrieval and the avidin–biotin peroxidase complex method. Int J Cardiol 2013; 168:4843-4. [DOI: 10.1016/j.ijcard.2013.07.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 07/02/2013] [Indexed: 11/22/2022]
|
92
|
Abstract
In contrast to their role in cell types with higher energy demands, mitochondria in endothelial cells primarily function in signaling cellular responses to environmental cues. This article provides an overview of key aspects of mitochondrial biology in endothelial cells, including subcellular location, biogenesis, dynamics, autophagy, reactive oxygen species production and signaling, calcium homeostasis, regulated cell death, and heme biosynthesis. In each section, we introduce key concepts and then review studies showing the importance of that mechanism to endothelial control of vasomotor tone, angiogenesis, and/or inflammatory activation. We particularly highlight the small number of clinical and translational studies that have investigated each mechanism in human subjects. Finally, we review interventions that target different aspects of mitochondrial function and their effects on endothelial function. The ultimate goal of such research is the identification of new approaches for therapy. The reviewed studies make it clear that mitochondria are important in endothelial physiology and pathophysiology. A great deal of work will be needed, however, before mitochondria-directed therapies are available for the prevention and treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Matthew A Kluge
- Evans Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| | | | | |
Collapse
|
93
|
Cao DJ, Jiang N, Blagg A, Johnstone JL, Gondalia R, Oh M, Luo X, Yang KC, Shelton JM, Rothermel BA, Gillette TG, Dorn GW, Hill JA. Mechanical unloading activates FoxO3 to trigger Bnip3-dependent cardiomyocyte atrophy. J Am Heart Assoc 2013; 2:e000016. [PMID: 23568341 PMCID: PMC3647287 DOI: 10.1161/jaha.113.000016] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Background Mechanical assist device therapy has emerged recently as an important and rapidly expanding therapy in advanced heart failure, triggering in some patients a beneficial reverse remodeling response. However, mechanisms underlying this benefit are unclear. Methods and Results In a model of mechanical unloading of the left ventricle, we observed progressive myocyte atrophy, autophagy, and robust activation of the transcription factor FoxO3, an established regulator of catabolic processes in other cell types. Evidence for FoxO3 activation was similarly detected in unloaded failing human myocardium. To determine the role of FoxO3 activation in cardiac muscle in vivo, we engineered transgenic mice harboring a cardiomyocyte‐specific constitutively active FoxO3 mutant (caFoxO3flox;αMHC‐Mer‐Cre‐Mer). Expression of caFoxO3 triggered dramatic and progressive loss of cardiac mass, robust increases in cardiomyocyte autophagy, declines in mitochondrial biomass and function, and early mortality. Whereas increases in cardiomyocyte apoptosis were not apparent, we detected robust increases in Bnip3 (Bcl2/adenovirus E1B 19‐kDa interacting protein 3), an established downstream target of FoxO3. To test the role of Bnip3, we crossed the caFoxO3flox;αMHC‐Mer‐Cre‐Mer mice with Bnip3‐null animals. Remarkably, the atrophy and autophagy phenotypes were significantly blunted, yet the early mortality triggered by FoxO3 activation persisted. Rather, declines in cardiac performance were attenuated by proteasome inhibitors. Consistent with involvement of FoxO3‐driven activation of the ubiquitin‐proteasome system, we detected time‐dependent activation of the atrogenes program and sarcomere protein breakdown. Conclusions In aggregate, these data point to FoxO3, a protein activated by mechanical unloading, as a master regulator that governs both the autophagy‐lysosomal and ubiquitin‐proteasomal pathways to orchestrate cardiac muscle atrophy.
Collapse
Affiliation(s)
- Dian J Cao
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Day SM, Divald A, Wang P, Davis F, Bartolone S, Jones R, Powell SR. Impaired assembly and post-translational regulation of 26S proteasome in human end-stage heart failure. Circ Heart Fail 2013; 6:544-9. [PMID: 23515276 DOI: 10.1161/circheartfailure.112.000119] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND This study examined the hypothesis that 26S proteasome dysfunction in human end-stage heart failure is associated with decreased docking of the 19S regulatory particle to the 20S proteasome. Previous studies have demonstrated that 26S proteasome activity is diminished in human end-stage heart failure associated with oxidation of the 19S regulatory particle Rpt5 subunit. Docking of the 19S regulatory particle to the 20S proteasome requires functional Rpt subunit ATPase activity and phosphorylation of the α-type subunits. METHODS AND RESULTS An enriched proteasome fraction was prepared from 7 human nonfailing and 10 failing heart explants. Native gel electrophoresis assessed docking of 19S to 20S proteasome revealing 3 proteasome populations (20S, 26S, and 30S proteasomes). In failing hearts, 30S proteasomes were significantly lower (P=0.048) by 37% suggesting diminished docking. Mass spectrometry-based phosphopeptide analysis demonstrated that the relative ratio of phosphorylated:non phosphorylated α7 subunit (serine250) of the 20S proteasome was significantly less (P=0.011) by almost 80% in failing hearts. Rpt ATPase activity was determined in the enriched fraction and after immunoprecipitation with an Rpt6 antibody. ATPase activity (ρmol PO4/μg protein per hour) of the total fraction was lowered from 291±97 to 194±27 and in the immunoprecipitated fraction from 42±12 to 3±2 (P=0.005) in failing hearts. CONCLUSIONS These studies suggest that diminished 26S activity in failing human hearts may be related to impaired docking of the 19S to the 20S as a result of decreased Rpt subunit ATPase activity and α7 subunit phosphorylation.
Collapse
Affiliation(s)
- Sharlene M Day
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | | | | | | | | | | |
Collapse
|
95
|
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
|
96
|
Affiliation(s)
- Monte S Willis
- McAllister Heart Institute, and Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, USA
| | | |
Collapse
|
97
|
Abstract
The ubiquitin proteasome system (UPS) has been the subject of intensive research over the past 20 years to define its role in normal physiology and in pathophysiology. Many of these studies have focused in on the cardiovascular system and have determined that the UPS becomes dysfunctional in several pathologies such as familial and idiopathic cardiomyopathies, atherosclerosis, and myocardial ischemia. This review presents a synopsis of the literature as it relates to the role of the UPS in myocardial ischemia. Studies have shown that the UPS is dysfunctional during myocardial ischemia, and recent studies have shed some light on possible mechanisms. Other studies have defined a role for the UPS in ischemic preconditioning which is best associated with myocardial ischemia and is thus presented here. Very recent studies have started to define roles for specific proteasome subunits and components of the ubiquitination machinery in various aspects of myocardial ischemia. Lastly, despite the evidence linking myocardial ischemia and proteasome dysfunction, there are continuing suggestions that proteasome inhibitors may be useful to mitigate ischemic injury. This review presents the rationale behind this and discusses both supportive and nonsupportive studies and presents possible future directions that may help in clarifying this controversy.
Collapse
Affiliation(s)
- Justine Calise
- Center for Heart and Lung Research, The Feinstein Institute for Medical Research, Manhasset, New York 11030, USA
| | | |
Collapse
|
98
|
Ramos FJ, Chen SC, Garelick MG, Dai DF, Liao CY, Schreiber KH, MacKay VL, An EH, Strong R, Ladiges WC, Rabinovitch PS, Kaeberlein M, Kennedy BK. Rapamycin reverses elevated mTORC1 signaling in lamin A/C-deficient mice, rescues cardiac and skeletal muscle function, and extends survival. Sci Transl Med 2012; 4:144ra103. [PMID: 22837538 DOI: 10.1126/scitranslmed.3003802] [Citation(s) in RCA: 264] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Mutations in LMNA, the gene that encodes A-type lamins, cause multiple diseases including dystrophies of the skeletal muscle and fat, dilated cardiomyopathy, and progeria-like syndromes (collectively termed laminopathies). Reduced A-type lamin function, however, is most commonly associated with skeletal muscle dystrophy and dilated cardiomyopathy rather than lipodystrophy or progeria. The mechanisms underlying these diseases are only beginning to be unraveled. We report that mice deficient in Lmna, which corresponds to the human gene LMNA, have enhanced mTORC1 (mammalian target of rapamycin complex 1) signaling specifically in tissues linked to pathology, namely, cardiac and skeletal muscle. Pharmacologic reversal of elevated mTORC1 signaling by rapamycin improves cardiac and skeletal muscle function and enhances survival in mice lacking A-type lamins. At the cellular level, rapamycin decreases the number of myocytes with abnormal desmin accumulation and decreases the amount of desmin in both muscle and cardiac tissue of Lmna(-/-) mice. In addition, inhibition of mTORC1 signaling with rapamycin improves defective autophagic-mediated degradation in Lmna(-/-) mice. Together, these findings point to aberrant mTORC1 signaling as a mechanistic component of laminopathies associated with reduced A-type lamin function and offer a potential therapeutic approach, namely, the use of rapamycin-related mTORC1 inhibitors.
Collapse
Affiliation(s)
- Fresnida J Ramos
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Mechanical stress meets autophagy: potential implications for physiology and pathology. Trends Mol Med 2012; 18:583-8. [PMID: 22981844 DOI: 10.1016/j.molmed.2012.08.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Revised: 08/07/2012] [Accepted: 08/13/2012] [Indexed: 02/08/2023]
Abstract
Changes in the mechanical environment are a universal challenge for cells, and mechanical cues regulate tissue structure and cell physiology throughout life. Autophagy is an important degradative pathway, fulfilling a wide range of roles in survival, homeostasis and adaptation. The two are connected, and in vitro, autophagy is rapidly induced in cells exposed to mechanical compression. In vivo, autophagy is also induced in several medically relevant circumstances that are also under mechanical stress such as bone and muscle homeostasis and tissue injury. The induction of autophagy has wide-ranging effects on cells. In this article, I propose that the autophagic response to mechanical stress is an important factor in a wide range of both physiological and pathological settings.
Collapse
|
100
|
Klionsky DJ. Look people, "Atg" is an abbreviation for "autophagy-related." That's it. Autophagy 2012; 8:1281-2. [PMID: 22889836 DOI: 10.4161/auto.21812] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|