201
|
Gupta MK, Kaminski R, Mullen B, Gordon J, Burdo TH, Cheung JY, Feldman AM, Madesh M, Khalili K. HIV-1 Nef-induced cardiotoxicity through dysregulation of autophagy. Sci Rep 2017; 7:8572. [PMID: 28819214 PMCID: PMC5561171 DOI: 10.1038/s41598-017-08736-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 07/13/2017] [Indexed: 12/26/2022] Open
Abstract
Cardiovascular disease is a leading cause of co-morbidity in HIV-1 positive patients, even those in whom plasma virus levels are well-controlled. The pathogenic mechanism of HIV-1-associated cardiomyopathy is unknown, but has been presumed to be mediated indirectly, owing to the absence of productive HIV-1 replication in cardiomyocytes. We sought to investigate the effect of the HIV-1 auxiliary protein, Nef, which is suspected of extracellular release by infected CD4+ T cells on protein quality control and autophagy in cardiomyocytes. After detection of Nef in the serum of HIV-1 positive patients and the accumulation of this protein in human and primate heart tissue from HIV-1/SIV-infected cells we employed cell and molecular biology approaches to investigate the effect of Nef on cardiomyocyte-homeostasis by concentrating on protein quality control (PQC) pathway and autophagy. We found that HIV-1 Nef-mediated inhibition of autophagy flux leads to cytotoxicity and death of cardiomyocytes. Nef compromises autophagy at the maturation stage of autophagosomes by interacting with Beclin 1/Rab7 and dysregulating TFEB localization and cellular lysosome content. These effects were reversed by rapamycin treatment. Our results indicate that HIV-1 Nef-mediated inhibition of cellular PQC is one possible mechanism involved in the development of HIV-associated cardiomyopathy.
Collapse
Affiliation(s)
- Manish K Gupta
- Department of Neuroscience, Center for Neurovirology and Comprehensive NeuroAIDS Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Rafal Kaminski
- Department of Neuroscience, Center for Neurovirology and Comprehensive NeuroAIDS Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Brian Mullen
- Department of Neuroscience, Center for Neurovirology and Comprehensive NeuroAIDS Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Jennifer Gordon
- Department of Neuroscience, Center for Neurovirology and Comprehensive NeuroAIDS Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Tricia H Burdo
- Department of Neuroscience, Center for Neurovirology and Comprehensive NeuroAIDS Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Joseph Y Cheung
- Department of Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA.,Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Arthur M Feldman
- Department of Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA.,Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Muniswamy Madesh
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA.,Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Kamel Khalili
- Department of Neuroscience, Center for Neurovirology and Comprehensive NeuroAIDS Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA.
| |
Collapse
|
202
|
Mohan N, Jiang J, Wu WJ. Implications of Autophagy and Oxidative Stress in Trastuzumab-Mediated Cardiac Toxicities. AUSTIN PHARMACOLOGY & PHARMACEUTICS 2017; 2:1005. [PMID: 30288503 PMCID: PMC6168002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Trastuzumab, a humanized monoclonal antibody directed against human epidermal growth factor receptor 2 (HER2), remains the standard of care as part of adjuvant therapy for patients diagnosed with HER2-positive breast cancers. Despite high therapeutic efficacy, trastuzumab-based regimens can cause serious cardiotoxic side effects. Effective mitigation of cardiotoxic risk relies on thorough understanding of molecular mechanisms of cardiotoxicity induced by trastuzumab. Among the probable mechanisms responsible for trastuzumab-mediated cardiotoxicity, generation of free radicals causing oxidative stress has garnered notable attention in recent years. More recently, role of autophagy in trastuzumab-induced cardiomyopathy was explored. Trastuzumab-mediated HER2 signaling dysregulation activated Erk/mTOR signaling cascade resulting in autophagy inhibition. Consequently, autophagy impairment leads to massive accumulation of damaged mitochondria and free radicals causing oxidative stress and toxicity in cardiomyocytes. This review will discuss recent advances in understanding the mechanism of oxidative stress and highlight the role of autophagy in trastuzumab-mediated cardiac dysfunctions.
Collapse
Affiliation(s)
- N Mohan
- Division of Biotechnology Review and Research I, Center for Drug Evaluation and Research, USA
| | - J Jiang
- Division of Biotechnology Review and Research I, Center for Drug Evaluation and Research, USA
| | - W J Wu
- Division of Biotechnology Review and Research I, Center for Drug Evaluation and Research, USA
| |
Collapse
|
203
|
Perrotta I. The origin of the autophagosomal membrane in human atherosclerotic plaque: a preliminary ultrastructural study. Ultrastruct Pathol 2017; 41:327-334. [PMID: 28796583 DOI: 10.1080/01913123.2017.1349853] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Autophagy is an evolutionarily conserved process that occurs ubiquitously and functions as a primary route for the degradation of damaged organelles and proteins in response to starvation, oxidative stress, and other harmful conditions. The initial event upon autophagy induction is the formation of a membranous cistern called the phagophore or isolation membrane, a cup-shaped structure that elongates, engulfs cytoplasmic "cargo", and fuses at its rims to give rise to the autophagosome within which cytoplasmic material is enclosed. Although thoroughly studied in diverse cell culture systems, few attempts have been made to analyze the membrane dynamics during phagophore biogenesis in tissues. With respect to the cardiovascular system, no structural information is currently available regarding the sources that may contribute to the nucleation and growth of the phagophore membrane. The results presented here demonstrate that in the cells of human atherosclerotic plaque the phagophores are in contact with the endoplasmic reticulum (ER) membranes. Initially, the phagophore appears as a membrane sac that enwraps injured organelles and dysfunctional proteins and then matures into a double-membrane, closed structure often containing portions of the ER. These structural data indicate that the membrane source that elongates the phagophore might probably come from the ER. The topographical relationship between the ER tubules and the phagophore might also favor an efficient mechanism to transfer lipids from their site of synthesis to the nascent membrane, thus promoting its elongation and, ultimately, the formation of the autophagosome.
Collapse
Affiliation(s)
- Ida Perrotta
- a Department of Biology, Ecology and Earth Sciences, Centre for Microscopy and Microanalysis, Transmission Electron Microscopy Laboratory , University of Calabria , Arcavacata di Rende , Cosenza , Italy
| |
Collapse
|
204
|
Apigenin Alleviates Endotoxin-Induced Myocardial Toxicity by Modulating Inflammation, Oxidative Stress, and Autophagy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:2302896. [PMID: 28828145 PMCID: PMC5554558 DOI: 10.1155/2017/2302896] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 06/08/2017] [Accepted: 06/15/2017] [Indexed: 12/19/2022]
Abstract
Apigenin, a component in daily diets, demonstrates antioxidant and anti-inflammatory properties. Here, we intended to explore the mechanism of apigenin-mediated endotoxin-induced myocardial injury and its role in the interplay among inflammation, oxidative stress, and autophagy. In our lipopolysaccharide- (LPS-) induced myocardial injury model, apigenin ameliorated cardiac injury (lactate dehydrogenase (LDH) and creatine kinase (CK)), cell death (TUNEL staining, DNA fragmentation, and PARP activity), and tissue damage (cardiac troponin I (cTnI) and cardiac myosin light chain-1 (cMLC1)) and improved cardiac function (ejection fraction (EF) and end diastolic left ventricular inner dimension (LVID)). Apigenin also alleviated endotoxin-induced myocardial injury by modulating oxidative stress (nitrotyrosine and protein carbonyl) and inflammatory cytokines (TNF-α, IL-1β, MIP-1α, and MIP-2) along with their master regulator NFκB. Apigenin modulated redox homeostasis, and its anti-inflammatory role might be associated with its ability to control autophagy. Autophagy (determined by LAMP1, ATG5, and p62), its transcriptional regulator transcription factor EB (TFEB), and downstream target genes including vacuolar protein sorting-associated protein 11 (Vps11) and microtubule-associated proteins 1A/1B light chain 3B (Map1lc3) were modulated by apigenin. Thus, our study demonstrated that apigenin may lead to potential development of new target in sepsis treatment or other myocardial oxidative and/or inflammation-induced injuries.
Collapse
|
205
|
Chen L, Wang FY, Zeng ZY, Cui L, Shen J, Song XW, Li P, Zhao XX, Qin YW. MicroRNA-199a acts as a potential suppressor of cardiomyocyte autophagy through targeting Hspa5. Oncotarget 2017; 8:63825-63834. [PMID: 28969032 PMCID: PMC5609964 DOI: 10.18632/oncotarget.19133] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 06/02/2017] [Indexed: 12/18/2022] Open
Abstract
Autophagy is an adaptive response to cardiomyocytes survival under stress conditions. MicroRNAs (miRNAs, miR) have been described to act as potent modulators of autophagy. To investigate whether and how miR-199a modulated autophagy in vitro, primary cardiomyocytes were treated under starvation to induce autophagy. Results showed that down-regulation of miR-199a was sufficient to activate cardiomyocytes autophagy. MiR-199a suppressed cardiomyocytes autophagy through direct inhibiting heat shock protein family A member 5 (Hspa5). Forced overexpression of Hspa5 recovered the inhibitory effect of miR-199a in autophagy activation. Our results suggested miR-199a as an effective suppressor of starvation-induced cardiomyocytes autophagy and that Hspa5 was a direct target during this process. These results extend the understanding of the role and pathway of miR-199a in cardiomyocytes autophagy, and may introduce a potential therapeutic strategy for the protection of cardiomyocytes in myocardial infarction or ischemic heart disease.
Collapse
Affiliation(s)
- Liang Chen
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China.,Department of Laboratory Medicine, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Fei-Yu Wang
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Zhen-Yu Zeng
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Ling Cui
- Department of Cardiology, People's Hospital of Inner Mongolia, Hohhot 010017, China
| | - Jian Shen
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China.,Department of Cardiology, 411 Hospital of PLA Navy, Shanghai 200081, China
| | - Xiao-Wei Song
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Pan Li
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Xian-Xian Zhao
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Yong-Wen Qin
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
206
|
Wu P, Yuan X, Li F, Zhang J, Zhu W, Wei M, Li J, Wang X. Myocardial Upregulation of Cathepsin D by Ischemic Heart Disease Promotes Autophagic Flux and Protects Against Cardiac Remodeling and Heart Failure. Circ Heart Fail 2017; 10:CIRCHEARTFAILURE.117.004044. [PMID: 28694354 DOI: 10.1161/circheartfailure.117.004044] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 06/07/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND Lysosomal dysfunction is implicated in human heart failure for which ischemic heart disease is the leading cause. Altered myocardial expression of CTSD (cathepsin D), a major lysosomal protease, was observed in human heart failure, but its pathophysiological significance has not been determined. METHODS AND RESULTS Western blot analyses revealed an increase in the precursor but not the mature form of CTSD in myocardial samples from explanted human failing hearts with ischemic heart disease, which is recapitulated in chronic myocardial infarction produced via coronary artery ligation in Ctsd+/+ but not Ctsd+/- mice. Mice deficient of Ctsd displayed impaired myocardial autophagosome removal, reduced autophagic flux, and restrictive cardiomyopathy. After induction of myocardial infarction, weekly serial echocardiography detected earlier occurrence of left ventricle chamber dilatation, greater decreases in ejection fraction and fractional shortening, and lesser wall thickening throughout the first 4 weeks; pressure-volume relationship analyses at 4 weeks revealed greater decreases in systolic and diastolic functions, stroke work, stroke volume, and cardiac output; greater increases in the ventricular weight to body weight and the lung weight to body weight ratios and larger scar size were also detected in Ctsd+/- mice compared with Ctsd+/+ mice. Significant increases of myocardial autophagic flux detected at 1 and 4 weeks after induction of myocardial infarction in the Ctsd+/+ mice were diminished in the Ctsd+/- mice. CONCLUSIONS Myocardial CTSD upregulation induced by myocardial infarction protects against cardiac remodeling and malfunction, which is at least in part through promoting myocardial autophagic flux.
Collapse
Affiliation(s)
- Penglong Wu
- From the Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China (P.W., M.W., J.L., X.W.); Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion (P.W., X.W.); Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis (X.Y., F.L.); Department of Pathology, University of Alabama at Birmingham (J.Z.); Clinical Research Center and Division of Cardiology, Zhejiang University School of Medicine 2nd Affiliated Hospital, Hangzhou, China (W.Z.); Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou, China (X.W.); and Guangzhou Institute of Oncology, Tumor Hospital, Guangzhou Medical University, China (X.W.)
| | - Xun Yuan
- From the Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China (P.W., M.W., J.L., X.W.); Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion (P.W., X.W.); Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis (X.Y., F.L.); Department of Pathology, University of Alabama at Birmingham (J.Z.); Clinical Research Center and Division of Cardiology, Zhejiang University School of Medicine 2nd Affiliated Hospital, Hangzhou, China (W.Z.); Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou, China (X.W.); and Guangzhou Institute of Oncology, Tumor Hospital, Guangzhou Medical University, China (X.W.)
| | - Faqian Li
- From the Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China (P.W., M.W., J.L., X.W.); Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion (P.W., X.W.); Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis (X.Y., F.L.); Department of Pathology, University of Alabama at Birmingham (J.Z.); Clinical Research Center and Division of Cardiology, Zhejiang University School of Medicine 2nd Affiliated Hospital, Hangzhou, China (W.Z.); Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou, China (X.W.); and Guangzhou Institute of Oncology, Tumor Hospital, Guangzhou Medical University, China (X.W.)
| | - Jianhua Zhang
- From the Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China (P.W., M.W., J.L., X.W.); Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion (P.W., X.W.); Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis (X.Y., F.L.); Department of Pathology, University of Alabama at Birmingham (J.Z.); Clinical Research Center and Division of Cardiology, Zhejiang University School of Medicine 2nd Affiliated Hospital, Hangzhou, China (W.Z.); Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou, China (X.W.); and Guangzhou Institute of Oncology, Tumor Hospital, Guangzhou Medical University, China (X.W.)
| | - Wei Zhu
- From the Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China (P.W., M.W., J.L., X.W.); Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion (P.W., X.W.); Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis (X.Y., F.L.); Department of Pathology, University of Alabama at Birmingham (J.Z.); Clinical Research Center and Division of Cardiology, Zhejiang University School of Medicine 2nd Affiliated Hospital, Hangzhou, China (W.Z.); Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou, China (X.W.); and Guangzhou Institute of Oncology, Tumor Hospital, Guangzhou Medical University, China (X.W.)
| | - Meng Wei
- From the Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China (P.W., M.W., J.L., X.W.); Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion (P.W., X.W.); Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis (X.Y., F.L.); Department of Pathology, University of Alabama at Birmingham (J.Z.); Clinical Research Center and Division of Cardiology, Zhejiang University School of Medicine 2nd Affiliated Hospital, Hangzhou, China (W.Z.); Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou, China (X.W.); and Guangzhou Institute of Oncology, Tumor Hospital, Guangzhou Medical University, China (X.W.)
| | - Jingbo Li
- From the Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China (P.W., M.W., J.L., X.W.); Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion (P.W., X.W.); Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis (X.Y., F.L.); Department of Pathology, University of Alabama at Birmingham (J.Z.); Clinical Research Center and Division of Cardiology, Zhejiang University School of Medicine 2nd Affiliated Hospital, Hangzhou, China (W.Z.); Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou, China (X.W.); and Guangzhou Institute of Oncology, Tumor Hospital, Guangzhou Medical University, China (X.W.).
| | - Xuejun Wang
- From the Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China (P.W., M.W., J.L., X.W.); Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion (P.W., X.W.); Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis (X.Y., F.L.); Department of Pathology, University of Alabama at Birmingham (J.Z.); Clinical Research Center and Division of Cardiology, Zhejiang University School of Medicine 2nd Affiliated Hospital, Hangzhou, China (W.Z.); Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou, China (X.W.); and Guangzhou Institute of Oncology, Tumor Hospital, Guangzhou Medical University, China (X.W.).
| |
Collapse
|
207
|
Bharath LP, Cho JM, Park SK, Ruan T, Li Y, Mueller R, Bean T, Reese V, Richardson RS, Cai J, Sargsyan A, Pires K, Anandh Babu PV, Boudina S, Graham TE, Symons JD. Endothelial Cell Autophagy Maintains Shear Stress-Induced Nitric Oxide Generation via Glycolysis-Dependent Purinergic Signaling to Endothelial Nitric Oxide Synthase. Arterioscler Thromb Vasc Biol 2017; 37:1646-1656. [PMID: 28684613 DOI: 10.1161/atvbaha.117.309510] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 06/19/2017] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Impaired endothelial cell (EC) autophagy compromises shear stress-induced nitric oxide (NO) generation. We determined the responsible mechanism. APPROACH AND RESULTS On autophagy compromise in bovine aortic ECs exposed to shear stress, a decrease in glucose uptake and EC glycolysis attenuated ATP production. We hypothesized that decreased glycolysis-dependent purinergic signaling via P2Y1 (P2Y purinoceptor 1) receptors, secondary to impaired autophagy in ECs, prevents shear-induced phosphorylation of eNOS (endothelial nitric oxide synthase) at its positive regulatory site S1117 (p-eNOSS1177) and NO generation. Maneuvers that restore glucose transport and glycolysis (eg, overexpression of GLUT1 [glucose transporter 1]) or purinergic signaling (eg, addition of exogenous ADP) rescue shear-induced p-eNOSS1177 and NO production in ECs with impaired autophagy. Conversely, inhibiting glucose transport via GLUT1 small interfering RNA, blocking purinergic signaling via ectonucleotidase-mediated ATP/ADP degradation (eg, apyrase), or inhibiting P2Y1 receptors using pharmacological (eg, MRS2179 [2'-deoxy-N6-methyladenosine 3',5'-bisphosphate tetrasodium salt]) or genetic (eg, P2Y1-receptor small interfering RNA) procedures inhibit shear-induced p-eNOSS1177 and NO generation in ECs with intact autophagy. Supporting a central role for PKCδT505 (protein kinase C delta T505) in relaying the autophagy-dependent purinergic-mediated signal to eNOS, we find that (1) shear stress-induced activating phosphorylation of PKCδT505 is negated by inhibiting autophagy, (2) shear-induced p-eNOSS1177 and NO generation are restored in autophagy-impaired ECs via pharmacological (eg, bryostatin) or genetic (eg, constitutively active PKCδ) activation of PKCδT505, and (3) pharmacological (eg, rottlerin) and genetic (eg, PKCδ small interfering RNA) PKCδ inhibition prevents shear-induced p-eNOSS1177 and NO generation in ECs with intact autophagy. Key nodes of dysregulation in this pathway on autophagy compromise were revealed in human arterial ECs. CONCLUSIONS Targeted reactivation of purinergic signaling and PKCδ has strategic potential to restore compromised NO generation in pathologies associated with suppressed EC autophagy.
Collapse
Affiliation(s)
- Leena P Bharath
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Jae Min Cho
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Seul-Ki Park
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Ting Ruan
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Youyou Li
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Robert Mueller
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Tyler Bean
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Van Reese
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Russel S Richardson
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Jinjin Cai
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Ashot Sargsyan
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Karla Pires
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Pon Velayutham Anandh Babu
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Sihem Boudina
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Timothy E Graham
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - J David Symons
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.).
| |
Collapse
|
208
|
Protoporphyrin IX-mediated sonodynamic therapy promotes autophagy in vascular smooth muscle cells. Oncol Lett 2017; 14:2097-2102. [PMID: 28789437 PMCID: PMC5530015 DOI: 10.3892/ol.2017.6394] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 04/13/2017] [Indexed: 12/17/2022] Open
Abstract
Sonodynamic therapy (SDT) is effective in treating intimal hyperplasia and promoting plaque stability in animal models. The present study aimed to evaluate the effects of SDT with the sonosensitizer protoporphyrin IX (PpIX) on vascular smooth muscle cell (VSMC) viability and autophagy. Cultured VSMCs cells were divided into the following groups: i) Control, ii) ultrasound, iii) PpIX and iv) SDT. Flow cytometry and laser confocal detection were used to measure Annexin V stained VSMCs following different treatments. Alterations in mitochondrial membrane potential (MMP) were evaluated via JC-1 staining. Autophagosome formation was observed using electron and fluorescence microscopy. Western blotting was used to analyze the expression levels of the autophagy markers light chain 3 (LC3-I) and LC3-II. The results demonstrated that SDT did not trigger apoptosis nor induce a significant decline in MMP of VSMCs. However, SDT significantly increased autophagasome formation and increased the LC3-II/LC3-I ratio. The findings demonstrated that PpIX-SDT increased autophagy without inducing mitochondrial-dependent apoptosis in VSMCs.
Collapse
|
209
|
Hsp90 inhibitor geldanamycin attenuates the cytotoxicity of sunitinib in cardiomyocytes via inhibition of the autophagy pathway. Toxicol Appl Pharmacol 2017. [PMID: 28624441 DOI: 10.1016/j.taap.2017.06.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Sunitinib malate (sunitinib) is an orally available, multitargeted tyrosine kinase inhibitor with antitumor and antiangiogenic activities. Although sunitinib is effective for the treatment of patients with gastrointestinal stromal tumor, advanced renal cell carcinoma, or pancreatic neuroendocrine tumor, adverse cardiac events associated with sunitinib administration have been reported. Here, we examined the effect of geldanamycin, an inhibitor of heat shock protein (Hsp) 90, on sunitinib-induced cytotoxicity in cardiomyocytes. First, we found that treatment with geldanamycin or other Hsp90 inhibitors (tanespimycin, ganetespib, or BIIB021) significantly attenuated sunitinib-induced cytotoxicity in rat H9c2 cardiomyocytes, suggesting a drug-class effect of Hsp90 inhibitors. We then examined the mechanisms underlying sunitinib-induced cytotoxicity and found that sunitinib induced autophagy in H9c2 cells and that pretreatment with geldanamycin inhibited the induction of autophagy by promoting degradation of the autophagy-related proteins Atg7, Beclin-1, and ULK1. Pharmacological assessment with autophagy inhibitors confirmed that geldanamycin attenuated the cytotoxicity of sunitinib by interfering with autophagy. In addition, we found that the molecular chaperone Hsp70, which is induced by geldanamycin, was not involved in the attenuation of sunitinib-induced cytotoxicity. Finally, to provide more clinically relevant data, we confirmed that geldanamycin attenuated sunitinib-induced cytotoxicity in human induced pluripotent stem cell-derived cardiomyocytes. Together, these data suggest that geldanamycin attenuates sunitinib-induced cytotoxicity in cardiomyocytes by inhibiting the autophagy pathway. Thus, the further investigation of combination or sequential treatment with an Hsp90 inhibitor and sunitinib is warranted as a potential strategy of attenuating the cardiotoxicity associated with sunitinib administration in the clinical setting.
Collapse
|
210
|
Wang Y, Wang G. Polymeric nanomicelles: a potential hazard for the cardiovascular system? Nanomedicine (Lond) 2017; 12:1355-1358. [PMID: 28524805 DOI: 10.2217/nnm-2017-0099] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Affiliation(s)
- Yazhou Wang
- Key Laboratory of Biorheological Science & Technology, Ministry of Education, State & Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Guixue Wang
- Key Laboratory of Biorheological Science & Technology, Ministry of Education, State & Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing 400044, China
| |
Collapse
|
211
|
Zhang C, Syed TW, Liu R, Yu J. Role of Endoplasmic Reticulum Stress, Autophagy, and Inflammation in Cardiovascular Disease. Front Cardiovasc Med 2017; 4:29. [PMID: 28553639 PMCID: PMC5427082 DOI: 10.3389/fcvm.2017.00029] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 04/25/2017] [Indexed: 01/07/2023] Open
Abstract
Cardiovascular diseases are a class of heart or blood vessels diseases, which are now considered to be the leading cause of death globally. A number of recent studies have reported key roles for inflammation in the progression of diseased vessels and systematic heart failure. In particular, endoplasmic reticulum (ER) stress, which is mechanistically implicated in inflammation and autophagy, has now been associated with pathophysiological states in the cardiovascular system. Autophagy has also been identified as an important process in the progression of multiple cardiovascular diseases such as in atherosclerosis plaque progression and ischemia and/or reperfusion. In light of the above, it has been proposed that a link between inflammation, autophagy, and ER stress may exist that contribute to diseases of the heart and its supporting vessels. This review highlights current knowledge on the cross talk between these three biological processes, and the potential of targeting this pathway as a therapeutic approach for cardiovascular disorders and its related diseases.
Collapse
Affiliation(s)
- Cheng Zhang
- Center for Metabolic Disease Research, Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Taha Wasim Syed
- Center for Metabolic Disease Research, Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Renjing Liu
- Agnes Ginges Laboratory for Diseases of the Aorta, Centenary Institute, University of Sydney, Camperdown, NSW, Australia,Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Jun Yu
- Center for Metabolic Disease Research, Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA,*Correspondence: Jun Yu,
| |
Collapse
|
212
|
Wei H, Qu H, Wang H, Ji B, Ding Y, Liu D, Duan Y, Liang H, Peng C, Xiao X, Deng H. 1,25-Dihydroxyvitamin-D3 prevents the development of diabetic cardiomyopathy in type 1 diabetic rats by enhancing autophagy via inhibiting the β-catenin/TCF4/GSK-3β/mTOR pathway. J Steroid Biochem Mol Biol 2017; 168:71-90. [PMID: 28216152 DOI: 10.1016/j.jsbmb.2017.02.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 02/09/2017] [Accepted: 02/09/2017] [Indexed: 12/31/2022]
Abstract
Diabetic cardiomyopathy (DCM) can increase the risk of heart failure and death in diabetic patients. However, no effective approaches are available to prevent its progression and development. Studies have shown that vitamin D is greatly implicated in cardiac hypertrophy and fibrosis, and there is a high prevalence of vitamin D deficiency in diabetic patients. In this study, we investigated whether 1,25-Dihydroxyvitamin-D3 (1,25D3) can improve DCM through a vitamin D receptor (VDR)-dependent mechanism associated with autophagy and the β-catenin/T-cell factor/lymphoid enhancer factor (TCF4)/glycogen synthase kinase-3β (GSK-3β)/mammalian target of rapamycin (mTOR) pathway. In this study, streptozotocin (STZ)-induced type 1 diabetic rats were established and were treated with 1,25D3 and/or chloroquine and/or VDR gene silencing for 8 weeks before being sacrificed. Compared with untreated diabetic rats, 1,25D3 partly attenuated the myocardial hypertrophy and interstitial fibrosis, improved cardiac function and restored the impaired cardiac autophagy in diabetic rats, all of which were reversed by silencing the VDR gene in diabetic rats. In high-glucose cultured H9C2 cells, 1,25D3 increased autophagy in a dose-dependent manner. Besides, the β-catenin/TCF4/GSK-3β and mTOR signaling were activated both in diabetic rats and in high-glucose cultured H9C2 cells. Treatment with 1,25D3 inhibited the β-catenin/TCF4/GSK-3β and mTOR signaling in H9C2 cells, whereas co-treatment with lithium chloride (LiCl) reversed this situation and abolished the beneficial effect of 1,25D3 on autophagy. These data suggest that 1,25D3 may improve DCM in type 1 diabetic rats by modulating autophagy through the β-catenin/TCF4/GSK-3β and mTOR pathway. Vitamin D may exist as a new therapeutic target for the treatment of DCM.
Collapse
Affiliation(s)
- Huili Wei
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuan Jiagang, Yuzhong District, Chongqing 400016, China.
| | - Hua Qu
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuan Jiagang, Yuzhong District, Chongqing 400016, China.
| | - Hang Wang
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuan Jiagang, Yuzhong District, Chongqing 400016, China.
| | - Baolan Ji
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuan Jiagang, Yuzhong District, Chongqing 400016, China
| | - Yao Ding
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuan Jiagang, Yuzhong District, Chongqing 400016, China
| | - Dan Liu
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuan Jiagang, Yuzhong District, Chongqing 400016, China
| | - Yang Duan
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuan Jiagang, Yuzhong District, Chongqing 400016, China
| | - Huimin Liang
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuan Jiagang, Yuzhong District, Chongqing 400016, China
| | - Chuan Peng
- Laboratory of Lipid and Glucose Metabolism, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xiaoqiu Xiao
- Laboratory of Lipid and Glucose Metabolism, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Huacong Deng
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuan Jiagang, Yuzhong District, Chongqing 400016, China.
| |
Collapse
|
213
|
The role of autophagy in asparaginase-induced immune suppression of macrophages. Cell Death Dis 2017; 8:e2721. [PMID: 28358370 PMCID: PMC5386542 DOI: 10.1038/cddis.2017.144] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 02/25/2017] [Accepted: 02/28/2017] [Indexed: 02/07/2023]
Abstract
Erwinia asparaginase, a bacteria-derived enzyme drug, has been used in the treatment of various cancers, especially acute lymphoblastic leukemia (ALL). One of the most significant side effects associated with asparaginase administration is immune suppression, which limits its application in clinic. Macrophages are phagocytic immune cells and have a central role in inflammation and host defense. We reported here that asparaginase disturbed the function of macrophages including phagocytosis, proliferation, ROS and nitric oxide secretion, interleukin 6 (IL-6) and tumor necrosis factor α (TNF-α) secretion, and major histocompatibility complex II (MHC-II) molecule expression, thus induced immune suppression in interferon-γ and lipopolysaccharide-stimulated macrophages. We also observed that asparaginase inhibited autophagy in macrophages via activating Akt/mTOR and suppressing Erk1/2 signaling pathway as evidenced by less formation of autophagosomes, downregulation of autophagy-related protein LC3-II, and decreased number of autophagy-like vacuoles. Further study discovered that treatment with autophagy inhibitor 3-MA in place of asparaginase on activated macrophages could also downregulate phagocytosis, cytokine secretion, and MHC-II expression. Moreover, incubation with autophagy inducer trehalose restored the capacity of phagocytosis, IL-6 and TNF-α secretion, and MHC-II expression in macrophages. These results prove the important role of autophagy in the function of macrophages, and activation of autophagy can overcome asparaginase-induced immune suppression in macrophages.
Collapse
|
214
|
Yau JW, Singh KK, Hou Y, Lei X, Ramadan A, Quan A, Teoh H, Kuebler WM, Al-Omran M, Yanagawa B, Ni H, Verma S. Endothelial-specific deletion of autophagy-related 7 (ATG7) attenuates arterial thrombosis in mice. J Thorac Cardiovasc Surg 2017; 154:978-988.e1. [PMID: 28400112 DOI: 10.1016/j.jtcvs.2017.02.058] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 02/01/2017] [Accepted: 02/17/2017] [Indexed: 01/29/2023]
Abstract
BACKGROUND Thrombosis persists as a leading cause of morbidity and mortality. Given that endothelial cells (ECs) play a central role in regulating thrombosis, understanding the molecular endothelial cues that regulate susceptibility or resistance to thrombosis have important translational implications. Accordingly, we evaluated the role of endothelial autophagy in the development of thrombosis. METHODS We generated mice in which the essential autophagy-related 7 (ATG7) gene was conditionally deleted from ECs (EC-ATG7-/- mice). Three in vivo models of thrombosis were used, and mechanistic studies were conducted with cultured human umbilical vein endothelial cells (HUVECs). RESULTS We silenced ATG7 in HUVECs and observed >60% decreases in tumor necrosis factor (TNF)-α-induced tissue factor (TF) transcript levels, protein expression, and activity. TF mRNA levels in the carotid arteries of EC-ATG7-/- mice subjected to the prothrombotic stimulus FeCl3 were lower than those in the similarly treated wild-type (WT) littermate group. Compared with WT mice, EC-ATG7-/- mice exhibited prolonged time to carotid (2-fold greater) and mesenteric (1.3-fold greater) artery occlusion following FeCl3 injury. The thrombi generated in laser-injured cremasteric arterioles were smaller in EC-ATG7-/- mice compared with WT mice, and took 2.3-fold longer to appear. CONCLUSIONS Taken together, these results provide definitive evidence that loss of endothelial ATG7 attenuates thrombosis and reduces the expression of TF. Our findings demonstrate that endothelial ATG7, and thus autophagy, is a critical and previously unrecognized target for modulating the susceptibility to thrombosis.
Collapse
Affiliation(s)
- Jonathan W Yau
- Division of Cardiac Surgery, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Krishna K Singh
- Division of Cardiac Surgery, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Division of Vascular Surgery, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Department of Surgery, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada.
| | - Yan Hou
- Division of Cardiac Surgery, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Xi Lei
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Azza Ramadan
- Division of Cardiac Surgery, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Adrian Quan
- Division of Cardiac Surgery, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Hwee Teoh
- Division of Cardiac Surgery, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Division of Endocrinology and Metabolism, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Wolfgang M Kuebler
- Department of Surgery, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, Canada; Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Mohammed Al-Omran
- Division of Vascular Surgery, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Department of Surgery, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, King Saud University and the King Saud University-Li Ka Shing Collaborative Research Program, Riyadh, Kingdom of Saudi Arabia
| | - Bobby Yanagawa
- Division of Cardiac Surgery, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Department of Surgery, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Heyu Ni
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Subodh Verma
- Division of Cardiac Surgery, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Department of Surgery, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
215
|
Pestana CR, Urbaczek AC, Alberici JV, Rodrigues GJ, Carrilho E. Metabolic profiling of human endothelial cells during autophagy assessed in a biomimetic microfluidic device model. Life Sci 2017; 172:42-47. [PMID: 28011226 DOI: 10.1016/j.lfs.2016.12.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 12/11/2016] [Accepted: 12/19/2016] [Indexed: 01/22/2023]
Abstract
AIMS Autophagy is critical to endothelial function. We explored the effects of autophagy induced by serum deprivation on Human Umbilical Vascular Endothelial Cells (HUVEC) metabolome profile and its inhibition by the antimalarial drug chloroquine (CLQ) using a microfluidic biomimetic model. MAIN METHODS The metabolites secreted by HUVEC into the circulating microfluidics were determined by liquid chromatography mass spectrometry (LC-MS) and further analyzed using Metaboanalyst 3.0 multivariate and pathway analysis tools. KEY FINDINGS Principal component analysis showed the discrimination of metabolites between treated and control groups. The results also identified alterations in metabolites relevant to endothelial function such as arginine, glutamate and energy metabolism pathways. Interestingly, CLQ mostly reversed the changes induced by serum deprivation. SIGNIFICANCE The knowledge of endothelial metabolic profile during autophagy may contribute to the identification of clinical biomarkers and potential therapeutic approaches based on the regulation of autophagy.
Collapse
Affiliation(s)
- Cezar Rangel Pestana
- Instituto Latino-Americano de Ciências da Vida e da Natureza, Universidade Federal da Integração Latino-Americana, Foz do Iguaçu, PR, Brazil.
| | - Ana Carolina Urbaczek
- Instituto de Química de São Carlos, Universidade de São Paulo, São Carlos, SP, Brazil
| | | | | | - Emanuel Carrilho
- Instituto de Química de São Carlos, Universidade de São Paulo, São Carlos, SP, Brazil
| |
Collapse
|
216
|
Gαq Signaling in the Regulation of Autophagy and Heart Failure. J Cardiovasc Pharmacol 2017; 69:212-214. [PMID: 28166086 DOI: 10.1097/fjc.0000000000000471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
217
|
Parra V, Rothermel BA. Calcineurin signaling in the heart: The importance of time and place. J Mol Cell Cardiol 2017; 103:121-136. [PMID: 28007541 PMCID: PMC5778886 DOI: 10.1016/j.yjmcc.2016.12.006] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 12/12/2016] [Accepted: 12/16/2016] [Indexed: 12/20/2022]
Abstract
The calcium-activated protein phosphatase, calcineurin, lies at the intersection of protein phosphorylation and calcium signaling cascades, where it provides an essential nodal point for coordination between these two fundamental modes of intracellular communication. In excitatory cells, such as neurons and cardiomyocytes, that experience rapid and frequent changes in cytoplasmic calcium, calcineurin protein levels are exceptionally high, suggesting that these cells require high levels of calcineurin activity. Yet, it is widely recognized that excessive activation of calcineurin in the heart contributes to pathological hypertrophic remodeling and the progression to failure. How does a calcium activated enzyme function in the calcium-rich environment of the continuously contracting heart without pathological consequences? This review will discuss the wide range of calcineurin substrates relevant to cardiovascular health and the mechanisms calcineurin uses to find and act on appropriate substrates in the appropriate location while potentially avoiding others. Fundamental differences in calcineurin signaling in neonatal verses adult cardiomyocytes will be addressed as well as the importance of maintaining heterogeneity in calcineurin activity across the myocardium. Finally, we will discuss how circadian oscillations in calcineurin activity may facilitate integration with other essential but conflicting processes, allowing a healthy heart to reap the benefits of calcineurin signaling while avoiding the detrimental consequences of sustained calcineurin activity that can culminate in heart failure.
Collapse
Affiliation(s)
- Valentina Parra
- Advanced Centre for Chronic Disease (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas, Universidad de Chile, Santiago,Chile; Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Quimicas y Farmaceuticas, Universidad de Chie, Santiago, Chile
| | - Beverly A Rothermel
- Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Centre, Dallas, TX, USA; Department of Molecular Biology, University of Texas Southwestern Medical Centre, Dallas, TX, USA.
| |
Collapse
|
218
|
Lu H, Fan Y, Qiao C, Liang W, Hu W, Zhu T, Zhang J, Chen YE. TFEB inhibits endothelial cell inflammation and reduces atherosclerosis. Sci Signal 2017; 10:10/464/eaah4214. [PMID: 28143903 DOI: 10.1126/scisignal.aah4214] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Transcription factor EB (TFEB) is a master regulator of autophagy and lysosome biogenesis. We investigated the function of TFEB in vascular biology and pathophysiology and demonstrated that TFEB in endothelial cells inhibited inflammation and reduced atherosclerosis development. Laminar shear stress, which protects against atherosclerosis, increased TFEB abundance in cultured primary human endothelial cells. Furthermore, TFEB overexpression in these cells was anti-inflammatory, whereas TFEB knockdown aggravated inflammation. The anti-inflammatory effect of TFEB was, at least, partially due to reduced oxidative stress because TFEB overexpression in endothelial cells decreased the concentrations of reactive oxygen species and increased the expression of the antioxidant genes HO1 (which encodes heme oxygenase 1) and SOD2 (which encodes superoxide dismutase 2). In addition, transgenic mice with endothelial cell-specific expression of TFEB exhibited reduced leukocyte recruitment to endothelial cells and decreased atherosclerosis development. Our study suggests that TFEB is a protective transcription factor against endothelial cell inflammation and a potential target for treating atherosclerosis and associated cardiovascular diseases.
Collapse
Affiliation(s)
- Haocheng Lu
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Yanbo Fan
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI 48109, USA.
| | - Congzhen Qiao
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Wenying Liang
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Wenting Hu
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Tianqing Zhu
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Jifeng Zhang
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Y Eugene Chen
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI 48109, USA.
| |
Collapse
|
219
|
Suárez-Rivero JM, Villanueva-Paz M, de la Cruz-Ojeda P, de la Mata M, Cotán D, Oropesa-Ávila M, de Lavera I, Álvarez-Córdoba M, Luzón-Hidalgo R, Sánchez-Alcázar JA. Mitochondrial Dynamics in Mitochondrial Diseases. Diseases 2016; 5:diseases5010001. [PMID: 28933354 PMCID: PMC5456341 DOI: 10.3390/diseases5010001] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 12/13/2016] [Accepted: 12/21/2016] [Indexed: 01/09/2023] Open
Abstract
Mitochondria are very versatile organelles in continuous fusion and fission processes in response to various cellular signals. Mitochondrial dynamics, including mitochondrial fission/fusion, movements and turnover, are essential for the mitochondrial network quality control. Alterations in mitochondrial dynamics can cause neuropathies such as Charcot-Marie-Tooth disease in which mitochondrial fusion and transport are impaired, or dominant optic atrophy which is caused by a reduced mitochondrial fusion. On the other hand, mitochondrial dysfunction in primary mitochondrial diseases promotes reactive oxygen species production that impairs its own function and dynamics, causing a continuous vicious cycle that aggravates the pathological phenotype. Mitochondrial dynamics provides a new way to understand the pathophysiology of mitochondrial disorders and other diseases related to mitochondria dysfunction such as diabetes, heart failure, or Hungtinton’s disease. The knowledge about mitochondrial dynamics also offers new therapeutics targets in mitochondrial diseases.
Collapse
Affiliation(s)
- Juan M Suárez-Rivero
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Sevilla 41013, Spain.
| | - Marina Villanueva-Paz
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Sevilla 41013, Spain.
| | - Patricia de la Cruz-Ojeda
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Sevilla 41013, Spain.
| | - Mario de la Mata
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Sevilla 41013, Spain.
| | - David Cotán
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Sevilla 41013, Spain.
| | - Manuel Oropesa-Ávila
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Sevilla 41013, Spain.
| | - Isabel de Lavera
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Sevilla 41013, Spain.
| | - Mónica Álvarez-Córdoba
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Sevilla 41013, Spain.
| | - Raquel Luzón-Hidalgo
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Sevilla 41013, Spain.
| | - José A Sánchez-Alcázar
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red Enfermedades Raras, Instituto de Salud Carlos III, Sevilla 41013, Spain.
| |
Collapse
|
220
|
Xue Z, Yuan W, Li J, Zhou H, Xu L, Weng J, Li X, Zhang X, Wang Z, Yan J. Cyclophilin A mediates the ox-LDL-induced activation and apoptosis of macrophages via autophagy. Int J Cardiol 2016; 230:142-148. [PMID: 28038796 DOI: 10.1016/j.ijcard.2016.12.042] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 11/03/2016] [Accepted: 12/16/2016] [Indexed: 11/25/2022]
Abstract
BACKGROUND Oxidized low-density lipoprotein (ox-LDL) is the most common inflammatory factor that mediates the activation and apoptosis of macrophages. Cyclophilin A (CyPA) is expressed following oxidative stress, hypoxia, and infection. However, the role of CyPA in the activation and apoptosis of macrophages is unclear. The aims of the study were to determine whether CyPA mediates the ox-LDL-induced activation and apoptosis in RAW264.7 cells and to analyze potential mechanisms. METHODS AND RESULTS Through Western blot and ELISA test, the expression of CyPA induced by ox-LDL is time-dependent in RAW264.7 cells. Gene silencing of CyPA reduced the generation of lipid droplets in the cytoplasm and downregulated the expression of the surface markers of macrophage activation, namely, CD80, CD86, and major histocompatibility complex class 2 antigen. Cell apoptosis is significantly decreased and the level of anti-apoptosis protein bcl-2 is increased in CyPA silent cells compared with the control group. Finally, autophagy-related protein LC3-II/LC3-I ratio level significantly decreased in CyPA silent cells with less autophagosome formation while the blocked autophagy flux was recovered. The differences in the activation and apoptosis between CyPA silent cells and the control cells were inhibited by pre-treatment with class III PI 3-kinase inhibitor 3-MA. CONCLUSIONS These results indicate that CyPA mediates the ox-LDL-induced activation and apoptosis in RAW264.7 cells by regulating autophagy.
Collapse
Affiliation(s)
- Zhiqiang Xue
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, People's Republic of China
| | - Wei Yuan
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, People's Republic of China.
| | - Jing Li
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, People's Republic of China
| | - Hong Zhou
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, People's Republic of China
| | - Lihua Xu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, People's Republic of China
| | - Jiayi Weng
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, People's Republic of China
| | - Xiaoyang Li
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, People's Republic of China
| | - Xinru Zhang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, People's Republic of China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, People's Republic of China
| | - Jinchuan Yan
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, People's Republic of China.
| |
Collapse
|
221
|
Lippai M, Szatmári Z. Autophagy-from molecular mechanisms to clinical relevance. Cell Biol Toxicol 2016; 33:145-168. [PMID: 27957648 DOI: 10.1007/s10565-016-9374-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 12/02/2016] [Indexed: 12/14/2022]
Abstract
Autophagy is a lysosomal degradation pathway of eukaryotic cells that is highly conserved from yeast to mammals. During this process, cooperating protein complexes are recruited in a hierarchic order to the phagophore assembly site (PAS) to mediate the elongation and closure of double-membrane vesicles called autophagosomes, which sequester cytosolic components and deliver their content to the endolysosomal system for degradation. As a major cytoprotective mechanism, autophagy plays a key role in the stress response against nutrient starvation, hypoxia, and infections. Although numerous studies reported that impaired function of core autophagy proteins also contributes to the development and progression of various human diseases such as neurodegenerative disorders, cardiovascular and muscle diseases, infections, and different types of cancer, the function of this process in human diseases remains unclear. Evidence often suggests a controversial role for autophagy in the pathomechanisms of these severe disorders. Here, we provide an overview of the molecular mechanisms of autophagy and summarize the recent advances on its function in human health and disease.
Collapse
Affiliation(s)
- Mónika Lippai
- Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, Pázmány Péter stny. 1/C, Budapest, 1117, Hungary
| | - Zsuzsanna Szatmári
- Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, Pázmány Péter stny. 1/C, Budapest, 1117, Hungary.
| |
Collapse
|
222
|
Yang J, Yu J, Li D, Yu S, Ke J, Wang L, Wang Y, Qiu Y, Gao X, Zhang J, Huang L. Store-operated calcium entry-activated autophagy protects EPC proliferation via the CAMKK2-MTOR pathway in ox-LDL exposure. Autophagy 2016; 13:82-98. [PMID: 27791458 DOI: 10.1080/15548627.2016.1245261] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Improving biological functions of endothelial progenitor cells (EPCs) is beneficial to maintaining endothelium homeostasis and promoting vascular re-endothelialization. Because macroautophagy/autophagy has been documented as a double-edged sword in cell functions, its effects on EPCs remain to be elucidated. This study was designed to explore the role and molecular mechanisms of store-operated calcium entry (SOCE)-activated autophagy in proliferation of EPCs under hypercholesterolemia. We employed oxidized low-density lipoprotein (ox-LDL) to mimic hypercholesterolemia in bone marrow-derived EPCs from rat. Ox-LDL dose-dependently activated autophagy flux, while inhibiting EPC proliferation. Importantly, inhibition of autophagy either by silencing Atg7 or by 3-methyladenine treatment, further aggravated proliferative inhibition by ox-LDL, suggesting the protective effects of autophagy against ox-LDL. Interestingly, ox-LDL increased STIM1 expression and intracellular Ca2+ concentration. Either Ca2+ chelators or deficiency in STIM1 attenuated ox-LDL-induced autophagy activation, confirming the involvement of SOCE in the process. Furthermore, CAMKK2 (calcium/calmodulin-dependent protein kinase kinase 2, β) activation and MTOR (mechanistic target of rapamycin [serine/threonine kinase]) deactivation were associated with autophagy modulation. Together, our results reveal a novel signaling pathway of SOCE-CAMKK2 in the regulation of autophagy and offer new insights into the important roles of autophagy in maintaining proliferation and promoting the survival capability of EPCs. This may be beneficial to improving EPC transplantation efficacy and enhancing vascular re-endothelialization in patients with hypercholesterolemia.
Collapse
Affiliation(s)
- Jie Yang
- a Institute of Cardiovascular Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing , China
| | - Jie Yu
- a Institute of Cardiovascular Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing , China
| | - Dongdong Li
- a Institute of Cardiovascular Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing , China
| | - Sanjiu Yu
- a Institute of Cardiovascular Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing , China
| | - Jingbin Ke
- a Institute of Cardiovascular Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing , China
| | - Lianyou Wang
- a Institute of Cardiovascular Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing , China
| | - Yanwei Wang
- a Institute of Cardiovascular Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing , China
| | - Youzhu Qiu
- a Institute of Cardiovascular Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing , China
| | - Xubin Gao
- a Institute of Cardiovascular Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing , China
| | - Jihang Zhang
- a Institute of Cardiovascular Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing , China
| | - Lan Huang
- a Institute of Cardiovascular Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing , China
| |
Collapse
|
223
|
Alloza I, Goikuria H, Freijo MDM, Vandenbroeck K. A role for autophagy in carotid atherosclerosis. Eur Stroke J 2016; 1:255-263. [PMID: 31008286 DOI: 10.1177/2396987316674085] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 09/20/2016] [Indexed: 12/15/2022] Open
Abstract
Purpose Autophagy has emerged in recent years as a critical cellular survival mechanism for cell homeostasis and may play a protective role in atherosclerosis. We aimed to review here the role autophagy plays in different cell types present in carotid atherosclerotic plaques and that may be associated with the development of unstable carotid atheroma plaque. Methods We performed a thorough literature exploration in this area of research covering the three main cell types present in carotid atheroma plaques. Findings Reviewed reports indicate that the role of autophagy in stable or unstable carotid atherosclerotic plaques depends on the different cell types and phenotypes, the stage and morphology of the plaque and the specific autophagy factor/s involved. Discussion Although defective autophagy could be one of the causes for carotid atheroma plaques to become unstable, it is important to take into account that autophagic players can act differentially in different cell types and different stages of the developed plaque. Conclusion This review provides an overview of the role of autophagy in the main cell types in carotid atherosclerosis (i.e. macrophages, endothelial cells and smooth muscle cells).
Collapse
Affiliation(s)
- Iraide Alloza
- Neurogenomiks Group, Neuroscience Department, Faculty of Medicine and Odontology, University of Basque Country, UPV/EHU, Leioa, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.,ACHUCARRO Basque Centre for Neuroscience, Zamudio, Spain
| | - Haize Goikuria
- Neurogenomiks Group, Neuroscience Department, Faculty of Medicine and Odontology, University of Basque Country, UPV/EHU, Leioa, Spain.,ACHUCARRO Basque Centre for Neuroscience, Zamudio, Spain
| | | | - Koen Vandenbroeck
- Neurogenomiks Group, Neuroscience Department, Faculty of Medicine and Odontology, University of Basque Country, UPV/EHU, Leioa, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.,ACHUCARRO Basque Centre for Neuroscience, Zamudio, Spain
| |
Collapse
|
224
|
Abstract
Heart failure with reduced ejection fraction (HFrEF) develops when cardiac output falls as a result of cardiac injury. The most well-recognized of the compensatory homeostatic responses to a fall in cardiac output are activation of the sympathetic nervous system and the renin-angiotensin-aldosterone system (RAAS). In the short term, these 'neurohormonal' systems induce a number of changes in the heart, kidneys, and vasculature that are designed to maintain cardiovascular homeostasis. However, with chronic activation, these responses result in haemodynamic stress and exert deleterious effects on the heart and the circulation. Neurohormonal activation is now known to be one of the most important mechanisms underlying the progression of heart failure, and therapeutic antagonism of neurohormonal systems has become the cornerstone of contemporary pharmacotherapy for heart failure. In this Review, we discuss the effects of neurohormonal activation in HFrEF and highlight the mechanisms by which these systems contribute to disease progression.
Collapse
|
225
|
Regina C, Panatta E, Candi E, Melino G, Amelio I, Balistreri CR, Annicchiarico-Petruzzelli M, Di Daniele N, Ruvolo G. Vascular ageing and endothelial cell senescence: Molecular mechanisms of physiology and diseases. Mech Ageing Dev 2016; 159:14-21. [DOI: 10.1016/j.mad.2016.05.003] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 04/12/2016] [Accepted: 05/03/2016] [Indexed: 01/21/2023]
|
226
|
Abstract
INTRODUCTION/BACKGROUND Heart failure is a major cause of cardiovascular morbidity and mortality. This review covers current heart failure treatment guidelines, emerging therapies that are undergoing clinical trial, and potential new therapeutic targets arising from basic science advances. SOURCES OF DATA A non-systematic search of MEDLINE was carried out. International guidelines and relevant reviews were searched for additional articles. AREAS OF AGREEMENT Angiotensin-converting enzyme inhibitors and beta-blockers are first line treatments for chronic heart failure with reduced left ventricular function. AREAS OF CONTROVERSY Treatment strategies to improve mortality in heart failure with preserved left ventricular function are unclear. GROWING POINTS Many novel therapies are being tested for clinical efficacy in heart failure, including those that target natriuretic peptides and myosin activators. A large number of completely novel targets are also emerging from laboratory-based research. Better understanding of pathophysiological mechanisms driving heart failure in different settings (e.g. hypertension, post-myocardial infarction, metabolic dysfunction) may allow for targeted therapies. AREAS TIMELY FOR DEVELOPING RESEARCH Therapeutic targets directed towards modifying the extracellular environment, angiogenesis, cell viability, contractile function and microRNA-based therapies.
Collapse
Affiliation(s)
- Adam Nabeebaccus
- Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, London, UK
| | - Sean Zheng
- Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, London, UK
| | - Ajay M Shah
- Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, London, UK
| |
Collapse
|
227
|
Boya P, Esteban-Martínez L, Serrano-Puebla A, Gómez-Sintes R, Villarejo-Zori B. Autophagy in the eye: Development, degeneration, and aging. Prog Retin Eye Res 2016; 55:206-245. [PMID: 27566190 DOI: 10.1016/j.preteyeres.2016.08.001] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 08/15/2016] [Accepted: 08/18/2016] [Indexed: 02/06/2023]
Abstract
Autophagy is a catabolic pathway that promotes the degradation and recycling of cellular components. Proteins, lipids, and even whole organelles are engulfed in autophagosomes and delivered to the lysosome for elimination. In response to stress, autophagy mediates the degradation of cell components, which are recycled to generate the nutrients and building blocks required to sustain cellular homeostasis. Moreover, it plays an important role in cellular quality control, particularly in neurons, in which the total burden of altered proteins and damaged organelles cannot be reduced by redistribution to daughter cells through cell division. Research has only begun to examine the role of autophagy in the visual system. The retina, a light-sensitive tissue, detects and transmits electrical impulses through the optic nerve to the visual cortex in the brain. Both the retina and the eye are exposed to a variety of environmental insults and stressors, including genetic mutations and age-associated alterations that impair their function. Here, we review the main studies that have sought to explain autophagy's importance in visual function. We describe the role of autophagy in retinal development and cell differentiation, and discuss the implications of autophagy dysregulation both in physiological aging and in important diseases such as age-associated macular degeneration and glaucoma. We also address the putative role of autophagy in promoting photoreceptor survival and discuss how selective autophagy could provide alternative means of protecting retinal cells. The findings reviewed here underscore the important role of autophagy in maintaining proper retinal function and highlight novel therapeutic approaches for blindness and other diseases of the eye.
Collapse
Affiliation(s)
- Patricia Boya
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain.
| | - Lorena Esteban-Martínez
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Ana Serrano-Puebla
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Raquel Gómez-Sintes
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Beatriz Villarejo-Zori
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
| |
Collapse
|
228
|
Lezoualc'h F, Fazal L, Laudette M, Conte C. Cyclic AMP Sensor EPAC Proteins and Their Role in Cardiovascular Function and Disease. Circ Res 2016; 118:881-97. [PMID: 26941424 DOI: 10.1161/circresaha.115.306529] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
cAMP is a universal second messenger that plays central roles in cardiovascular regulation influencing gene expression, cell morphology, and function. A crucial step toward a better understanding of cAMP signaling came 18 years ago with the discovery of the exchange protein directly activated by cAMP (EPAC). The 2 EPAC isoforms, EPAC1 and EPAC2, are guanine-nucleotide exchange factors for the Ras-like GTPases, Rap1 and Rap2, which they activate independently of the classical effector of cAMP, protein kinase A. With the development of EPAC pharmacological modulators, many reports in the literature have demonstrated the critical role of EPAC in the regulation of various cAMP-dependent cardiovascular functions, such as calcium handling and vascular tone. EPAC proteins are coupled to a multitude of effectors into distinct subcellular compartments because of their multidomain architecture. These novel cAMP sensors are not only at the crossroads of different physiological processes but also may represent attractive therapeutic targets for the treatment of several cardiovascular disorders, including cardiac arrhythmia and heart failure.
Collapse
Affiliation(s)
- Frank Lezoualc'h
- From the Department of Cardiac and Renal Remodeling of the Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM), UMR-1048, Toulouse, France (F.L., L.F., M.L., C.C.); and Université Toulouse III-Paul Sabatier, Toulouse, France (F.L., L.F., M.L., C.C.).
| | - Loubina Fazal
- From the Department of Cardiac and Renal Remodeling of the Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM), UMR-1048, Toulouse, France (F.L., L.F., M.L., C.C.); and Université Toulouse III-Paul Sabatier, Toulouse, France (F.L., L.F., M.L., C.C.)
| | - Marion Laudette
- From the Department of Cardiac and Renal Remodeling of the Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM), UMR-1048, Toulouse, France (F.L., L.F., M.L., C.C.); and Université Toulouse III-Paul Sabatier, Toulouse, France (F.L., L.F., M.L., C.C.)
| | - Caroline Conte
- From the Department of Cardiac and Renal Remodeling of the Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM), UMR-1048, Toulouse, France (F.L., L.F., M.L., C.C.); and Université Toulouse III-Paul Sabatier, Toulouse, France (F.L., L.F., M.L., C.C.)
| |
Collapse
|
229
|
Xie S, Deng Y, Pan YY, Ren J, Jin M, Wang Y, Wang ZH, Zhu D, Guo XL, Yuan X, Shang J, Liu HG. Chronic intermittent hypoxia induces cardiac hypertrophy by impairing autophagy through the adenosine 5'-monophosphate-activated protein kinase pathway. Arch Biochem Biophys 2016; 606:41-52. [PMID: 27412517 DOI: 10.1016/j.abb.2016.07.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 07/05/2016] [Accepted: 07/08/2016] [Indexed: 12/19/2022]
Abstract
Autophagy is tightly regulated to maintain cardiac homeostasis. Impaired autophagy is closely associated with pathological cardiac hypertrophy. However, the relationship between autophagy and cardiac hypertrophy induced by chronic intermittent hypoxia (CIH) is not known. In the present study, we measured autophagy-related genes and autophagosomes during 10 weeks of CIH in rats, and 6 days in H9C2 cardiomyocytes, and showed that autophagy was impaired. This conclusion was confirmed by the autophagy flux assay. We detected significant hypertrophic changes in myocardium with impaired autophagy. Rapamycin, an autophagy enhancer, attenuated the cardiac hypertrophy induced by CIH. Moreover, silencing autophagy-related gene 5 (ATG5) exerted the opposite effect. The role of adenosine monophosphate-activated protein kinase (AMPK) in regulating autophagy under CIH was confirmed using AICAR to upregulate this enzyme and restore autophagy flux. Restoring autophagy by AICAR or rapamycin significantly reversed the hypertrophic changes in cardiomyocytes. To investigate the mechanism of autophagy impairment, we compared phospho (p)-AMPK, p-Akt, cathepsin D, and NFAT3 levels, along with calcineurin activity, between sham and CIH groups. CIH activated calcineurin, and inhibited AMPK and AMPK-mediated autophagy in an Akt- and NFAT3-independent manner. Collectively, these data demonstrated that impaired autophagy induced by CIH through the AMPK pathway contributed to cardiac hypertrophy.
Collapse
Affiliation(s)
- Sheng Xie
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, China
| | - Yan Deng
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, China
| | - Yue-Ying Pan
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, China
| | - Jie Ren
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, China
| | - Meng Jin
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, China
| | - Yu Wang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, China
| | - Zhi-Hua Wang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, China
| | - Die Zhu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, China
| | - Xue-Ling Guo
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, China
| | - Xiao Yuan
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, China
| | - Jin Shang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, China
| | - Hui-Guo Liu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, China.
| |
Collapse
|
230
|
Riquelme JA, Chavez MN, Mondaca-Ruff D, Bustamante M, Vicencio JM, Quest AFG, Lavandero S. Therapeutic targeting of autophagy in myocardial infarction and heart failure. Expert Rev Cardiovasc Ther 2016; 14:1007-19. [PMID: 27308848 DOI: 10.1080/14779072.2016.1202760] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Myocardial infarction (MI) is the leading cause of death. When MI is not lethal, heart failure (HF) is a major consequence with high prevalence and poor prognosis. The targeting of autophagy represents a potentially therapeutic approach for the treatment of both pathologies. AREAS COVERED PubMed searches were performed to discuss the current state of the art regarding the role of autophagy in MI and HF. We review available and potential approaches to modulate autophagy from a pharmacological and genetic perspective. We also discuss the targeting of autophagy in myocardial regeneration. Expert commentary: The targeting of autophagy has potential for the treatment of MI and HF. Autophagy is a process that takes place in virtually all cells of the body and thus, in order to evaluate this therapeutic approach in clinical trials, strategies that specifically target this process in the myocardium is required to avoid unwanted effects in other organs.
Collapse
Affiliation(s)
- Jaime A Riquelme
- a Advanced Center for Chronic Disease (ACCDiS) & Center for Molecular Studies of the Cell (CEMC), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina , Universidad de Chile , Santiago , Chile
| | - Myra N Chavez
- a Advanced Center for Chronic Disease (ACCDiS) & Center for Molecular Studies of the Cell (CEMC), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina , Universidad de Chile , Santiago , Chile.,b FONDAP Center for Genome Regulation, Facultad de Ciencias , Universidad de Chile , Santiago , Chile
| | - David Mondaca-Ruff
- a Advanced Center for Chronic Disease (ACCDiS) & Center for Molecular Studies of the Cell (CEMC), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina , Universidad de Chile , Santiago , Chile
| | - Mario Bustamante
- a Advanced Center for Chronic Disease (ACCDiS) & Center for Molecular Studies of the Cell (CEMC), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina , Universidad de Chile , Santiago , Chile.,c Advanced Center for Chronic Disease (ACCDiS), Division Enfermedades Cardiovasculares, Facultad de Medicina , Pontificia Universidad Catolica de Chile , Santiago , Chile
| | - Jose Miguel Vicencio
- a Advanced Center for Chronic Disease (ACCDiS) & Center for Molecular Studies of the Cell (CEMC), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina , Universidad de Chile , Santiago , Chile.,d Cancer Institute , University College London , London , UK
| | - Andrew F G Quest
- a Advanced Center for Chronic Disease (ACCDiS) & Center for Molecular Studies of the Cell (CEMC), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina , Universidad de Chile , Santiago , Chile
| | - Sergio Lavandero
- a Advanced Center for Chronic Disease (ACCDiS) & Center for Molecular Studies of the Cell (CEMC), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina , Universidad de Chile , Santiago , Chile.,e Department of Internal Medicine, Cardiology Division , University of Texas Southwestern Medical Center , Dallas , TX , USA
| |
Collapse
|
231
|
Schiattarella GG, Cattaneo F, Pironti G, Magliulo F, Carotenuto G, Pirozzi M, Polishchuk R, Borzacchiello D, Paolillo R, Oliveti M, Boccella N, Avvedimento M, Sepe M, Lombardi A, Busiello RA, Trimarco B, Esposito G, Feliciello A, Perrino C. Akap1 Deficiency Promotes Mitochondrial Aberrations and Exacerbates Cardiac Injury Following Permanent Coronary Ligation via Enhanced Mitophagy and Apoptosis. PLoS One 2016; 11:e0154076. [PMID: 27136357 PMCID: PMC4852950 DOI: 10.1371/journal.pone.0154076] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 04/08/2016] [Indexed: 11/19/2022] Open
Abstract
A-kinase anchoring proteins (AKAPs) transmit signals cues from seven-transmembrane receptors to specific sub-cellular locations. Mitochondrial AKAPs encoded by the Akap1 gene have been shown to modulate mitochondrial function and reactive oxygen species (ROS) production in the heart. Under conditions of hypoxia, mitochondrial AKAP121 undergoes proteolytic degradation mediated, at least in part, by the E3 ubiquitin ligase Seven In-Absentia Homolog 2 (Siah2). In the present study we hypothesized that Akap1 might be crucial to preserve mitochondrial function and structure, and cardiac responses to myocardial ischemia. To test this, eight-week-old Akap1 knockout mice (Akap1-/-), Siah2 knockout mice (Siah2-/-) or their wild-type (wt) littermates underwent myocardial infarction (MI) by permanent left coronary artery ligation. Age and gender matched mice of either genotype underwent a left thoracotomy without coronary ligation and were used as controls (sham). Twenty-four hours after coronary ligation, Akap1-/- mice displayed larger infarct size compared to Siah2-/- or wt mice. One week after MI, cardiac function and survival were also significantly reduced in Akap1-/- mice, while cardiac fibrosis was significantly increased. Akap1 deletion was associated with remarkable mitochondrial structural abnormalities at electron microscopy, increased ROS production and reduced mitochondrial function after MI. These alterations were associated with enhanced cardiac mitophagy and apoptosis. Autophagy inhibition by 3-methyladenine significantly reduced apoptosis and ameliorated cardiac dysfunction following MI in Akap1-/- mice. These results demonstrate that Akap1 deficiency promotes cardiac mitochondrial aberrations and mitophagy, enhancing infarct size, pathological cardiac remodeling and mortality under ischemic conditions. Thus, mitochondrial AKAPs might represent important players in the development of post-ischemic cardiac remodeling and novel therapeutic targets.
Collapse
Affiliation(s)
- Gabriele Giacomo Schiattarella
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Fabio Cattaneo
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Gianluigi Pironti
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Fabio Magliulo
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Giuseppe Carotenuto
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Marinella Pirozzi
- Institute of Protein Biochemistry, Italian National Research Council (CNR-IBP), Naples, Italy
| | - Roman Polishchuk
- Telethon Institute of Genetic and Medicine (TIGEM), Naples, Italy
| | | | - Roberta Paolillo
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Marco Oliveti
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Nicola Boccella
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Marisa Avvedimento
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Maria Sepe
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Naples, Italy
| | | | | | - Bruno Trimarco
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Giovanni Esposito
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
- * E-mail: (CP); (GE)
| | - Antonio Feliciello
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Naples, Italy
| | - Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
- * E-mail: (CP); (GE)
| |
Collapse
|
232
|
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: 1.5] [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
|
233
|
Shaikh S, Troncoso R, Criollo A, Bravo-Sagua R, García L, Morselli E, Cifuentes M, Quest AFG, Hill JA, Lavandero S. Regulation of cardiomyocyte autophagy by calcium. Am J Physiol Endocrinol Metab 2016; 310:E587-E596. [PMID: 26884385 PMCID: PMC4835942 DOI: 10.1152/ajpendo.00374.2015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 02/10/2016] [Indexed: 11/22/2022]
Abstract
Calcium signaling plays a crucial role in a multitude of events within the cardiomyocyte, including cell cycle control, growth, apoptosis, and autophagy. With respect to calcium-dependent regulation of autophagy, ion channels and exchangers, receptors, and intracellular mediators play fundamental roles. In this review, we discuss calcium-dependent regulation of cardiomyocyte autophagy, a lysosomal mechanism that is often cytoprotective, serving to defend against disease-related stress and nutrient insufficiency. We also highlight the importance of the subcellular distribution of calcium and related proteins, interorganelle communication, and other key signaling events that govern cardiomyocyte autophagy.
Collapse
Affiliation(s)
- Soni Shaikh
- Advanced Center for Chronic Disease and Center for Molecular Studies of the Cell, Facultad de Ciencias Quimicas y Farmaceuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Rodrigo Troncoso
- Advanced Center for Chronic Disease and Center for Molecular Studies of the Cell, Facultad de Ciencias Quimicas y Farmaceuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile, Santiago, Chile
| | - Alfredo Criollo
- Advanced Center for Chronic Disease and Center for Molecular Studies of the Cell, Facultad de Ciencias Quimicas y Farmaceuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Roberto Bravo-Sagua
- Advanced Center for Chronic Disease and Center for Molecular Studies of the Cell, Facultad de Ciencias Quimicas y Farmaceuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Lorena García
- Advanced Center for Chronic Disease and Center for Molecular Studies of the Cell, Facultad de Ciencias Quimicas y Farmaceuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Bioquimica y Biologia Molecular, Facultad de Ciencias Quimicas y Farmaceuticas, Universidad de Chile, Santiago, Chile
| | - Eugenia Morselli
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mariana Cifuentes
- Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile, Santiago, Chile
| | - Andrew F G Quest
- Advanced Center for Chronic Disease and Center for Molecular Studies of the Cell, Facultad de Ciencias Quimicas y Farmaceuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile; and
| | - Joseph A Hill
- Departments of Internal Medicine (Cardiology Division) and
- Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Sergio Lavandero
- Advanced Center for Chronic Disease and Center for Molecular Studies of the Cell, Facultad de Ciencias Quimicas y Farmaceuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile;
- Departamento de Bioquimica y Biologia Molecular, Facultad de Ciencias Quimicas y Farmaceuticas, Universidad de Chile, Santiago, Chile
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile; and
- Departments of Internal Medicine (Cardiology Division) and
| |
Collapse
|
234
|
Affiliation(s)
- Shigeki Miyamoto
- From Department of Pharmacology, University of California San Diego, La Jolla, CA
| | - Joan Heller Brown
- From Department of Pharmacology, University of California San Diego, La Jolla, CA.
| |
Collapse
|
235
|
Bultman SJ, Holley DW, G de Ridder G, Pizzo SV, Sidorova TN, Murray KT, Jensen BC, Wang Z, Bevilacqua A, Chen X, Quintana MT, Tannu M, Rosson GB, Pandya K, Willis MS. BRG1 and BRM SWI/SNF ATPases redundantly maintain cardiomyocyte homeostasis by regulating cardiomyocyte mitophagy and mitochondrial dynamics in vivo. Cardiovasc Pathol 2016; 25:258-269. [PMID: 27039070 DOI: 10.1016/j.carpath.2016.02.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 02/24/2016] [Accepted: 02/27/2016] [Indexed: 12/13/2022] Open
Abstract
There has been an increasing recognition that mitochondrial perturbations play a central role in human heart failure. Mitochondrial networks, whose function is to maintain the regulation of mitochondrial biogenesis, autophagy ('mitophagy') and mitochondrial fusion/fission, are new potential therapeutic targets. Yet our understanding of the molecular underpinning of these processes is just emerging. We recently identified a role of the SWI/SNF ATP-dependent chromatin remodeling complexes in the metabolic homeostasis of the adult cardiomyocyte using cardiomyocyte-specific and inducible deletion of the SWI/SNF ATPases BRG1 and BRM in adult mice (Brg1/Brm double mutant mice). To build upon these observations in early altered metabolism, the present study looks at the subsequent alterations in mitochondrial quality control mechanisms in the impaired adult cardiomyocyte. We identified that Brg1/Brm double-mutant mice exhibited increased mitochondrial biogenesis, increases in 'mitophagy', and alterations in mitochondrial fission and fusion that led to small, fragmented mitochondria. Mechanistically, increases in the autophagy and mitophagy-regulated proteins Beclin1 and Bnip3 were identified, paralleling changes seen in human heart failure. Evidence for perturbed cardiac mitochondrial dynamics included decreased mitochondria size, reduced numbers of mitochondria, and an altered expression of genes regulating fusion (Mfn1, Opa1) and fission (Drp1). We also identified cardiac protein amyloid accumulation (aggregated fibrils) during disease progression along with an increase in pre-amyloid oligomers and an upregulated unfolded protein response including increased GRP78, CHOP, and IRE-1 signaling. Together, these findings described a role for BRG1 and BRM in mitochondrial quality control, by regulating mitochondrial number, mitophagy, and mitochondrial dynamics not previously recognized in the adult cardiomyocyte. As critical to the pathogenesis of heart failure, epigenetic mechanisms like SWI/SNF chromatin remodeling seem more intimately linked to cardiac function and mitochondrial quality control mechanisms than previously realized.
Collapse
Affiliation(s)
- Scott J Bultman
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Darcy Wood Holley
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | | - Tatiana N Sidorova
- Departments of Medicine and Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Katherine T Murray
- Departments of Medicine and Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Brian C Jensen
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Zhongjing Wang
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Ariana Bevilacqua
- Department of Pathology & Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Xin Chen
- Department of Neurosurgery, Shandong Provincial Hospital affiliated to Shandong University, 250021, Jinan, PR China
| | - Megan T Quintana
- Department of Surgery, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Manasi Tannu
- School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Gary B Rosson
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | - Monte S Willis
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA; Department of Pathology & Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
236
|
Yao P, Zhao H, Mo W, He P. Laminar Shear Stress Promotes Vascular Endothelial Cell Autophagy Through Upregulation with Rab4. DNA Cell Biol 2016; 35:118-23. [PMID: 26716952 DOI: 10.1089/dna.2015.3041] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Affiliation(s)
- Pingbo Yao
- ICU of the Affiliated Nanhua Hospital of University of South China, Hengyang, China
| | - Hong Zhao
- Nursing College, University of South China, Hengyang, China
| | - Wenjuan Mo
- Nursing College, University of South China, Hengyang, China
| | - Pingping He
- Nursing College, University of South China, Hengyang, China
| |
Collapse
|
237
|
García-Rúa V, Feijóo-Bandín S, Rodríguez-Penas D, Mosquera-Leal A, Abu-Assi E, Beiras A, María Seoane L, Lear P, Parrington J, Portolés M, Roselló-Lletí E, Rivera M, Gualillo O, Parra V, Hill JA, Rothermel B, González-Juanatey JR, Lago F. Endolysosomal two-pore channels regulate autophagy in cardiomyocytes. J Physiol 2016; 594:3061-77. [PMID: 26757341 DOI: 10.1113/jp271332] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 12/28/2015] [Indexed: 01/12/2023] Open
Abstract
KEY POINTS Two-pore channels (TPCs) were identified as a novel family of endolysosome-targeted calcium release channels gated by nicotinic acid adenine dinucleotide phosphate, as also as intracellular Na(+) channels able to control endolysosomal fusion, a key process in autophagic flux. Autophagy, an evolutionarily ancient response to cellular stress, has been implicated in the pathogenesis of a wide range of cardiovascular pathologies, including heart failure. We report direct evidence indicating that TPCs are involved in regulating autophagy in cardiomyocytes, and that TPC knockout mice show alterations in the cardiac lysosomal system. TPC downregulation implies a decrease in the viability of cardiomyocytes under starvation conditions. In cardiac tissues from both humans and rats, TPC transcripts and protein levels were higher in females than in males, and correlated negatively with markers of autophagy. We conclude that the endolysosomal channels TPC1 and TPC2 are essential for appropriate basal and induced autophagic flux in cardiomyocytes, and also that they are differentially expressed in male and female hearts. ABSTRACT Autophagy participates in physiological and pathological remodelling of the heart. The endolysosomal two-pore channels (TPCs), TPC1 and TPC2, have been implicated in the regulation of autophagy. The present study aimed to investigate the role of TPC1 and TPC2 in basal and induced cardiac autophagic activity. In cultured cardiomyocytes, starvation induced a significant increase in TPC1 and TPC2 transcripts and protein levels that paralleled the increase in autophagy identified by increased LC3-II and decreased p62 levels. Small interfering RNA depletion of TPC2 alone or together with TPC1 increased both LC3II and p62 levels under basal conditions and in response to serum starvation, suggesting that, under conditions of severe energy depletion (serum plus glucose starvation), changes in the autophagic flux (as assessed by use of bafilomycin A1) occurred either when TPC1 or TPC2 were downregulated. The knockdown of TPCs diminished cardiomyocyte viability under starvation and simulated ischaemia. Electron micrographs of hearts from TPC1/2 double knockout mice showed that cardiomyocytes contained large numbers of immature lysosomes with diameters significantly smaller than those of wild-type mice. In cardiac tissues from humans and rats, TPC1 and TPC2 transcripts and protein levels were higher in females than in males. Furthermore, transcript levels of TPCs correlated negatively with p62 levels in heart tissues. TPC1 and TPC2 are essential for appropriate basal and induced autophagic flux in cardiomyocytes (i.e. there is a negative effect on cell viability under stress conditions in their absence) and they are differentially expressed in male and female human and murine hearts, where they correlate with markers of autophagy.
Collapse
Affiliation(s)
- Vanessa García-Rúa
- Department of Cellular and Molecular Cardiology, Institute of Biomedical Research (IDIS-SERGAS), Santiago de Compostela, Spain
| | - Sandra Feijóo-Bandín
- Department of Cellular and Molecular Cardiology, Institute of Biomedical Research (IDIS-SERGAS), Santiago de Compostela, Spain
| | - Diego Rodríguez-Penas
- Department of Cellular and Molecular Cardiology, Institute of Biomedical Research (IDIS-SERGAS), Santiago de Compostela, Spain
| | - Ana Mosquera-Leal
- Department of Cellular and Molecular Cardiology, Institute of Biomedical Research (IDIS-SERGAS), Santiago de Compostela, Spain
| | - Emad Abu-Assi
- Department of Cellular and Molecular Cardiology, Institute of Biomedical Research (IDIS-SERGAS), Santiago de Compostela, Spain
| | - Andrés Beiras
- Department of Pathology, Institute of Biomedical Research (IDIS-SERGAS), Santiago de Compostela, Spain
| | - Luisa María Seoane
- Department of Endocrine Pathophysiology, Institute of Biomedical Research (IDIS-SERGAS), Santiago de Compostela, Spain
| | - Pamela Lear
- Department of Pharmacology, Oxford University, UK
| | | | | | | | | | - Oreste Gualillo
- Department of Neuroendocrine Interactions in Rheumatic and Inflammatory Diseases, Institute of Biomedical Research (IDIS-SERGAS), Santiago de Compostela, Spain
| | - Valentina Parra
- Department of Internal Medicine (Cardiology) and Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joseph A Hill
- Department of Internal Medicine (Cardiology) and Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Beverly Rothermel
- Department of Internal Medicine (Cardiology) and Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - José Ramón González-Juanatey
- Department of Cellular and Molecular Cardiology, Institute of Biomedical Research (IDIS-SERGAS), Santiago de Compostela, Spain
| | - Francisca Lago
- Department of Cellular and Molecular Cardiology, Institute of Biomedical Research (IDIS-SERGAS), Santiago de Compostela, Spain
| |
Collapse
|
238
|
Wang L, Jiang Y, Song X, Guo C, Zhu F, Wang X, Wang Q, Shi Y, Wang J, Gao F, Zhao W, Chen YH, Zhang L. Pdcd4 deficiency enhances macrophage lipoautophagy and attenuates foam cell formation and atherosclerosis in mice. Cell Death Dis 2016; 7:e2055. [PMID: 26775706 PMCID: PMC4816189 DOI: 10.1038/cddis.2015.416] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 12/14/2015] [Accepted: 12/16/2015] [Indexed: 12/11/2022]
Abstract
Macrophage foam cells, a major component of the atherosclerotic lesion, have vital roles in the development of atherosclerosis. Lipoautophagy, a type of autophagy characterized by selective delivery of lipid droplet for lysosomal degradation, may impact atherosclerosis by regulating macrophage foam cell formation. Previously, we reported that programmed cell death 4 (PDCD4), a tumor suppressor, negatively regulated autophagy in tumor cells. However, its roles in macrophage lipoautophagy, foam cell formation and atherosclerosis remain to be established. Here we found that Pdcd4 deficiency clearly improved oxidized low-density lipoproteins-impaired autophagy efflux, promoted autophagy-mediated lipid breakdown in murine macrophages and thus prevented macrophage conversion into foam cells. Importantly, Pdcd4 deficiency in mice significantly upregulated macrophage autophagy in local plaques along with attenuated lipid accumulation and atherosclerotic lesions in high-fat-fed Apolipoprotein E knockout mice. Bone marrow transplantation experiment demonstrated that PDCD4-mediated autophagy in hematopoietic cells contributed to the development of atherosclerosis. These results indicate that endogenous PDCD4 promotes for macrophage foam cell formation and atherosclerosis development via inhibiting autophagy and provides new insights into atherogenesis, suggesting that promoting macrophage autophagy through downregulating PDCD4 expression may be beneficial for treating atherosclerosis.
Collapse
Affiliation(s)
- L Wang
- Department of Immunology, Shandong University School of Medicine, Jinan, China
| | - Y Jiang
- Department of Immunology, Shandong University School of Medicine, Jinan, China.,Department of Hematology, the Second Hospital of Shandong University, Jinan, China
| | - X Song
- Department of Immunology, Shandong University School of Medicine, Jinan, China.,Basic Research Center, Shandong Cancer Hospital, Jinan, China
| | - C Guo
- Department of Immunology, Shandong University School of Medicine, Jinan, China
| | - F Zhu
- Department of Immunology, Shandong University School of Medicine, Jinan, China
| | - X Wang
- Department of Immunology, Shandong University School of Medicine, Jinan, China
| | - Q Wang
- Department of Immunology, Shandong University School of Medicine, Jinan, China
| | - Y Shi
- Department of Immunology, Shandong University School of Medicine, Jinan, China
| | - J Wang
- Department of Immunology, Shandong University School of Medicine, Jinan, China
| | - F Gao
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, China
| | - W Zhao
- Department of Immunology, Shandong University School of Medicine, Jinan, China
| | - Y H Chen
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - L Zhang
- Department of Immunology, Shandong University School of Medicine, Jinan, China
| |
Collapse
|
239
|
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
|
240
|
Vásquez-Trincado C, García-Carvajal I, Pennanen C, Parra V, Hill JA, Rothermel BA, Lavandero S. Mitochondrial dynamics, mitophagy and cardiovascular disease. J Physiol 2016; 594:509-25. [PMID: 26537557 DOI: 10.1113/jp271301] [Citation(s) in RCA: 421] [Impact Index Per Article: 52.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 08/30/2015] [Indexed: 12/14/2022] Open
Abstract
Cardiac hypertrophy is often initiated as an adaptive response to haemodynamic stress or myocardial injury, and allows the heart to meet an increased demand for oxygen. Although initially beneficial, hypertrophy can ultimately contribute to the progression of cardiac disease, leading to an increase in interstitial fibrosis and a decrease in ventricular function. Metabolic changes have emerged as key mechanisms involved in the development and progression of pathological remodelling. As the myocardium is a highly oxidative tissue, mitochondria play a central role in maintaining optimal performance of the heart. 'Mitochondrial dynamics', the processes of mitochondrial fusion, fission, biogenesis and mitophagy that determine mitochondrial morphology, quality and abundance have recently been implicated in cardiovascular disease. Studies link mitochondrial dynamics to the balance between energy demand and nutrient supply, suggesting that changes in mitochondrial morphology may act as a mechanism for bioenergetic adaptation during cardiac pathological remodelling. Another critical function of mitochondrial dynamics is the removal of damaged and dysfunctional mitochondria through mitophagy, which is dependent on the fission/fusion cycle. In this article, we discuss the latest findings regarding the impact of mitochondrial dynamics and mitophagy on the development and progression of cardiovascular pathologies, including diabetic cardiomyopathy, atherosclerosis, damage from ischaemia-reperfusion, cardiac hypertrophy and decompensated heart failure. We will address the ability of mitochondrial fusion and fission to impact all cell types within the myocardium, including cardiac myocytes, cardiac fibroblasts and vascular smooth muscle cells. Finally, we will discuss how these findings can be applied to improve the treatment and prevention of cardiovascular diseases.
Collapse
Affiliation(s)
- César Vásquez-Trincado
- Advanced Centre for Chronic Disease (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile.,Centre for Molecular Studies of the Cell, Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile
| | - Ivonne García-Carvajal
- Advanced Centre for Chronic Disease (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile.,Centre for Molecular Studies of the Cell, Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile
| | - Christian Pennanen
- Advanced Centre for Chronic Disease (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile.,Centre for Molecular Studies of the Cell, Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile
| | - Valentina Parra
- Advanced Centre for Chronic Disease (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile.,Centre for Molecular Studies of the Cell, Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile.,Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Centre, Dallas, TX, USA
| | - Joseph A Hill
- Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Centre, Dallas, TX, USA.,Department of Molecular Biology, University of Texas Southwestern Medical Centre, Dallas, TX, USA
| | - Beverly A Rothermel
- Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Centre, Dallas, TX, USA.,Department of Molecular Biology, University of Texas Southwestern Medical Centre, Dallas, TX, USA
| | - Sergio Lavandero
- Advanced Centre for Chronic Disease (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile.,Centre for Molecular Studies of the Cell, Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile.,Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Centre, Dallas, TX, USA
| |
Collapse
|
241
|
Cho GW, Altamirano F, Hill JA. Chronic heart failure: Ca(2+), catabolism, and catastrophic cell death. Biochim Biophys Acta Mol Basis Dis 2016; 1862:763-777. [PMID: 26775029 DOI: 10.1016/j.bbadis.2016.01.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 12/28/2015] [Accepted: 01/06/2016] [Indexed: 12/12/2022]
Abstract
Robust successes have been achieved in recent years in conquering the acutely lethal manifestations of heart disease. Many patients who previously would have died now survive to enjoy happy and productive lives. Nevertheless, the devastating impact of heart disease continues unabated, as the spectrum of disease has evolved with new manifestations. In light of this ever-evolving challenge, insights that culminate in novel therapeutic targets are urgently needed. Here, we review fundamental mechanisms of heart failure, both with reduced (HFrEF) and preserved (HFpEF) ejection fraction. We discuss pathways that regulate cardiomyocyte remodeling and turnover, focusing on Ca(2+) signaling, autophagy, and apoptosis. In particular, we highlight recent insights pointing to novel connections among these events. We also explore mechanisms whereby potential therapeutic approaches targeting these processes may improve morbidity and mortality in the devastating syndrome of heart failure.
Collapse
Affiliation(s)
- Geoffrey W Cho
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Francisco Altamirano
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Joseph A Hill
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
242
|
Tan VP, Miyamoto S. Nutrient-sensing mTORC1: Integration of metabolic and autophagic signals. J Mol Cell Cardiol 2016; 95:31-41. [PMID: 26773603 DOI: 10.1016/j.yjmcc.2016.01.005] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 12/11/2015] [Accepted: 01/04/2016] [Indexed: 12/26/2022]
Abstract
The ability of adult cardiomyocytes to regenerate is limited, and irreversible loss by cell death plays a crucial role in heart diseases. Autophagy is an evolutionarily conserved cellular catabolic process through which long-lived proteins and damaged organelles are targeted for lysosomal degradation. Autophagy is important in cardiac homeostasis and can serve as a protective mechanism by providing an energy source, especially in the face of sustained starvation. Cellular metabolism is closely associated with cell survival, and recent evidence suggests that metabolic and autophagic signaling pathways exhibit a high degree of crosstalk and are functionally interdependent. In this review, we discuss recent progress in our understanding of regulation of autophagy and its crosstalk with metabolic signaling, with a focus on the nutrient-sensing mTOR complex 1 (mTORC1) pathway.
Collapse
Affiliation(s)
- Valerie P Tan
- Department of Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0636, USA
| | - Shigeki Miyamoto
- Department of Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0636, USA.
| |
Collapse
|
243
|
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: 121] [Impact Index Per Article: 13.4] [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
|
244
|
Grootaert MOJ, da Costa Martins PA, Bitsch N, Pintelon I, De Meyer GRY, Martinet W, Schrijvers DM. Defective autophagy in vascular smooth muscle cells accelerates senescence and promotes neointima formation and atherogenesis. Autophagy 2015; 11:2014-2032. [PMID: 26391655 PMCID: PMC4824610 DOI: 10.1080/15548627.2015.1096485] [Citation(s) in RCA: 210] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 09/03/2015] [Accepted: 09/15/2015] [Indexed: 12/13/2022] Open
Abstract
Autophagy is triggered in vascular smooth muscle cells (VSMCs) of diseased arterial vessels. However, the role of VSMC autophagy in cardiovascular disease is poorly understood. Therefore, we investigated the effect of defective autophagy on VSMC survival and phenotype and its significance in the development of postinjury neointima formation and atherosclerosis. Tissue-specific deletion of the essential autophagy gene Atg7 in murine VSMCs (atg7-/- VSMCs) caused accumulation of SQSTM1/p62 and accelerated the development of stress-induced premature senescence as shown by cellular and nuclear hypertrophy, CDKN2A-RB-mediated G1 proliferative arrest and senescence-associated GLB1 activity. Transfection of SQSTM1-encoding plasmid DNA in Atg7+/+ VSMCs induced similar features, suggesting that accumulation of SQSTM1 promotes VSMC senescence. Interestingly, atg7-/- VSMCs were resistant to oxidative stress-induced cell death as compared to controls. This effect was attributed to nuclear translocation of the transcription factor NFE2L2 resulting in upregulation of several antioxidative enzymes. In vivo, defective VSMC autophagy led to upregulation of MMP9, TGFB and CXCL12 and promoted postinjury neointima formation and diet-induced atherogenesis. Lesions of VSMC-specific atg7 knockout mice were characterized by increased total collagen deposition, nuclear hypertrophy, CDKN2A upregulation, RB hypophosphorylation, and GLB1 activity, all features typical of cellular senescence. To conclude, autophagy is crucial for VSMC function, phenotype, and survival. Defective autophagy in VSMCs accelerates senescence and promotes ligation-induced neointima formation and diet-induced atherogenesis, implying that autophagy inhibition as therapeutic strategy in the treatment of neointimal stenosis and atherosclerosis would be unfavorable. Conversely, stimulation of autophagy could be a valuable new strategy in the treatment of arterial disease.
Collapse
Affiliation(s)
- Mandy OJ Grootaert
- Laboratory of Physiopharmacology; University of Antwerp; Antwerp, Belgium
| | - Paula A da Costa Martins
- Department of Cardiology; Cardiovascular Research Institute Maastricht; Maastricht, The Netherlands
| | - Nicole Bitsch
- Department of Cardiology; Cardiovascular Research Institute Maastricht; Maastricht, The Netherlands
| | - Isabel Pintelon
- Laboratory of Cell Biology and Histology; University of Antwerp; Antwerp, Belgium
| | - Guido RY De Meyer
- Laboratory of Physiopharmacology; University of Antwerp; Antwerp, Belgium
| | - Wim Martinet
- Laboratory of Physiopharmacology; University of Antwerp; Antwerp, Belgium
| | | |
Collapse
|
245
|
Novel players in cardioprotection: Insulin like growth factor-1, angiotensin-(1–7) and angiotensin-(1–9). Pharmacol Res 2015; 101:41-55. [DOI: 10.1016/j.phrs.2015.06.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 06/27/2015] [Accepted: 06/28/2015] [Indexed: 12/14/2022]
|
246
|
Low Shear Stress Inhibited Endothelial Cell Autophagy Through TET2 Downregulation. Ann Biomed Eng 2015; 44:2218-27. [PMID: 26493943 DOI: 10.1007/s10439-015-1491-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 10/15/2015] [Indexed: 12/16/2022]
Abstract
Low shear stress plays a crucial role in the initiation and progression of atherosclerotic lesions. However, the detailed mechanisms of these processes remain unclear. In this study, the effect of low shear stress on endothelial cell autophagy and its potential mechanism were investigated. Results showed autophagy dysfunction and ten-eleven translocation 2 (TET2) protein downregulation during atherosclerotic lesion progression. Autophagic markers BECLIN 1 and LC3II/LC3I under low shear stress (5 dyne/cm(2)) obviously decreased compared with those under physiological shear stress (15 dyne/cm(2)), whereas autophagic substrate p62 increased. TET2 expression was also downregulated under low shear stress. Endothelial cell autophagy was improved with TET2 overexpression but was impaired by TET2 siRNA treatment. Moreover, TET2 overexpression upregulated the expression of endothelial cell nitric oxide synthase (eNOS) and downregulated the expression of endothelin-1 (ET-1). TET2 siRNA further attenuated eNOS expression and stimulated ET-1 expression. Overall, the results showed that low shear stress downregulated endothelial cell autophagy by impaired TET2 expression, which might contribute to the atherogenic process.
Collapse
|
247
|
Spaich S, Katus HA, Backs J. Ongoing controversies surrounding cardiac remodeling: is it black and white-or rather fifty shades of gray? Front Physiol 2015; 6:202. [PMID: 26257654 PMCID: PMC4510775 DOI: 10.3389/fphys.2015.00202] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 07/03/2015] [Indexed: 01/02/2023] Open
Abstract
Cardiac remodeling describes the heart's multimodal response to a myriad of external or intrinsic stimuli and stressors most of which are probably only incompletely elucidated to date. Over many years the signaling molecules involved in these remodeling processes have been dichotomized according to a classic antagonistic view of black and white, i.e., attributed either a solely maladaptive or entirely beneficial character. By dissecting controversies, recent developments and shifts in perspective surrounding the three major cardiac signaling molecules calcineurin (Cn), protein kinase A (PKA) and calcium/calmodulin-dependent kinase II (CaMKII), this review challenges this dualistic view and advocates the nature and dignity of each of these key mediators of cardiac remodeling as a multilayered, highly context-sensitive and sophisticated continuum that can be markedly swayed and influenced by a multitude of environmental factors and crosstalk mechanisms. Furthermore this review delineates the importance and essential contributions of degradation and proteolysis to cardiac plasticity and homeostasis and finally aims to integrate the various aspects of protein synthesis and turnover into a comprehensive picture.
Collapse
Affiliation(s)
- Sebastian Spaich
- Research Unit Cardiac Epigenetics, Department of Cardiology, Angiology and Pneumology, University of HeidelbergHeidelberg, Germany
- German Centre for Cardiovascular Research, Partner Site Heidelberg/MannheimHeidelberg, Germany
- Department of Cardiology, Angiology and Pneumology, University of HeidelbergHeidelberg, Germany
| | - Hugo A. Katus
- Research Unit Cardiac Epigenetics, Department of Cardiology, Angiology and Pneumology, University of HeidelbergHeidelberg, Germany
- German Centre for Cardiovascular Research, Partner Site Heidelberg/MannheimHeidelberg, Germany
- Department of Cardiology, Angiology and Pneumology, University of HeidelbergHeidelberg, Germany
| | - Johannes Backs
- Research Unit Cardiac Epigenetics, Department of Cardiology, Angiology and Pneumology, University of HeidelbergHeidelberg, Germany
- German Centre for Cardiovascular Research, Partner Site Heidelberg/MannheimHeidelberg, Germany
| |
Collapse
|
248
|
Affiliation(s)
- Gabriele G Schiattarella
- From Departments of Internal Medicine (Cardiology) (G.G.S., J.A.H.) and Molecular Biology (J.A.H.), University of Texas Southwestern Medical Center, Dallas, TX
| | - Joseph A Hill
- From Departments of Internal Medicine (Cardiology) (G.G.S., J.A.H.) and Molecular Biology (J.A.H.), University of Texas Southwestern Medical Center, Dallas, TX.
| |
Collapse
|
249
|
Abstract
Dynamic packaging of DNA into strings of nucleosomes is a major mechanism whereby eukaryotic cells regulate gene expression. Intricate control of nucleosomal structure and assembly governs access of RNA polymerase II to DNA and consequent RNA synthesis. As part of this, post-translational modifications of histone proteins are central to the regulation of chromatin structure, playing vital roles in regulating the activation and repression of gene transcription. In the heart, dynamic homeostasis of histone modification-driven by the actions of modifiers and recruitment of downstream effectors-is a fundamental regulator of the transcriptional reprogramming that occurs in the setting of disease-related stress. Here, we examine the growing evidence for histone modification as a key mechanism governing pathological growth and remodeling of the myocardium.
Collapse
Affiliation(s)
- Thomas G Gillette
- From the Departments of Internal Medicine (Cardiology) (T.G.G., J.A.H.) and Molecular Biology (J.A.H.), University of Texas Southwestern Medical Center, Dallas.
| | - Joseph A Hill
- From the Departments of Internal Medicine (Cardiology) (T.G.G., J.A.H.) and Molecular Biology (J.A.H.), University of Texas Southwestern Medical Center, Dallas
| |
Collapse
|
250
|
Kroemer G. Autophagy: a druggable process that is deregulated in aging and human disease. J Clin Invest 2015; 125:1-4. [PMID: 25654544 DOI: 10.1172/jci78652] [Citation(s) in RCA: 227] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Autophagy ("self-eating") constitutes one of the most spectacular yet subtly regulated phenomena in cell biology. Similarly to cell division, differentiation, and death, autophagy is perturbed in multiple diseases, in that excessive or deficient autophagy may contribute to pathogenesis. Numerous attempts have been launched to identify specific inducers or inhibitors of autophagy and to use them for the therapeutic correction of its deregulation. At present, several major disease categories (including but not limited to age-related, cardiovascular, infectious, neoplastic, neurodegenerative, and metabolic pathologies) are being investigated for pathogenic aberrations in autophagy and their pharmacologic rectification. Driven by promising preclinical results, several clinical trials are exploring autophagy as a therapeutic target.
Collapse
|