201
|
Davran F, Yilmaz VT, Erdem BK, Gultekin M, Suleymanlar G, Akbas H. Association of interleukin 18-607A/C and -137C/G polymorphisms with oxidative stress in renal transplant recipients. Ren Fail 2016; 38:717-22. [PMID: 26983036 DOI: 10.3109/0886022x.2016.1158034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Objectives IL-18 mediates various inflammatory and oxidative responses including renal injury, fibrosis, and graft rejection. It has been reported that the promoter -607 and -137 polymorphisms of IL-18 influence the level of IL-18. This prospective observational study investigated the association between oxidative stress with IL-18-607 and -137 polymorphisms in renal transplant recipients. Patients and methods This study included 75 renal transplant recipients (28 female, 47 male) from living-related donors. Blood samples were collected immediately before and after transplantation at day 7 and month 1. Serum IL-18, creatinine, cystatin C, CRP, and oxidative stress markers (TOS, TAC) were measured. The Oxidative Stress Index (OSI) was calculated. Polymorphisms of the promoter region of the IL-18 gene, IL18-607A/C, and -137C/G were determined by analysis of a "real-time PCR/Melting curve". Results Serum creatinine, cystatin C, CRP, IL-18, TOS, and OSI levels significantly decreased after transplantation. Post-transplant levels of serum TAC and estimated GFR demonstrated consistent significant increases. Serum IL-18 levels were significantly higher in patients with IL-18-137 GG and IL-18-607 CC genotypes before transplantation. Conclusion Our results indicate that the IL-18-137 GG and -607 CC genotypes contribute to higher IL-18 levels; however, the influence of these polymorphisms on oxidative stress has not been observed.
Collapse
Affiliation(s)
- Fatih Davran
- a Department of Biochemistry, Faculty of Medicine , Akdeniz University , Antalya , Turkey
| | - Vural Taner Yilmaz
- b Division of Nephrology, Department of Internal Medicine, Faculty of Medicine , Akdeniz University , Antalya , Turkey
| | - Bilge Karatoy Erdem
- a Department of Biochemistry, Faculty of Medicine , Akdeniz University , Antalya , Turkey
| | - Meral Gultekin
- c Department of Microbiology, Faculty of Medicine , Akdeniz University , Antalya , Turkey
| | - Gultekin Suleymanlar
- b Division of Nephrology, Department of Internal Medicine, Faculty of Medicine , Akdeniz University , Antalya , Turkey
| | - Halide Akbas
- a Department of Biochemistry, Faculty of Medicine , Akdeniz University , Antalya , Turkey
| |
Collapse
|
202
|
Prada D, Harris A, Guidoboni G, Siesky B, Huang AM, Arciero J. Autoregulation and neurovascular coupling in the optic nerve head. Surv Ophthalmol 2016; 61:164-86. [DOI: 10.1016/j.survophthal.2015.10.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 10/02/2015] [Accepted: 10/02/2015] [Indexed: 12/23/2022]
|
203
|
Hull TD, Boddu R, Guo L, Tisher CC, Traylor AM, Patel B, Joseph R, Prabhu SD, Suliman HB, Piantadosi CA, Agarwal A, George JF. Heme oxygenase-1 regulates mitochondrial quality control in the heart. JCI Insight 2016; 1:e85817. [PMID: 27110594 DOI: 10.1172/jci.insight.85817] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The cardioprotective inducible enzyme heme oxygenase-1 (HO-1) degrades prooxidant heme into equimolar quantities of carbon monoxide, biliverdin, and iron. We hypothesized that HO-1 mediates cardiac protection, at least in part, by regulating mitochondrial quality control. We treated WT and HO-1 transgenic mice with the known mitochondrial toxin, doxorubicin (DOX). Relative to WT mice, mice globally overexpressing human HO-1 were protected from DOX-induced dilated cardiomyopathy, cardiac cytoarchitectural derangement, and infiltration of CD11b+ mononuclear phagocytes. Cardiac-specific overexpression of HO-1 ameliorated DOX-mediated dilation of the sarcoplasmic reticulum as well as mitochondrial disorganization in the form of mitochondrial fragmentation and increased numbers of damaged mitochondria in autophagic vacuoles. HO-1 overexpression promotes mitochondrial biogenesis by upregulating protein expression of NRF1, PGC1α, and TFAM, which was inhibited in WT animals treated with DOX. Concomitantly, HO-1 overexpression inhibited the upregulation of the mitochondrial fission mediator Fis1 and resulted in increased expression of the fusion mediators, Mfn1 and Mfn2. It also prevented dynamic changes in the levels of key mediators of the mitophagy pathway, PINK1 and parkin. Therefore, these findings suggest that HO-1 has a novel role in protecting the heart from oxidative injury by regulating mitochondrial quality control.
Collapse
Affiliation(s)
- Travis D Hull
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA; Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Ravindra Boddu
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA; Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Lingling Guo
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA; Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Cornelia C Tisher
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA; Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Amie M Traylor
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA; Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Bindiya Patel
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA; Comprehensive Cardiovascular Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Reny Joseph
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA; Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Sumanth D Prabhu
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA; Comprehensive Cardiovascular Center, University of Alabama at Birmingham, Birmingham, Alabama, USA; Department of Veterans Affairs, Birmingham, Alabama, USA
| | - Hagir B Suliman
- Department of Anesthesiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Claude A Piantadosi
- Department of Pulmonary, Allergy and Critical Care, Duke University School of Medicine, Durham, North Carolina, USA
| | - Anupam Agarwal
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA; Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama, USA; Department of Veterans Affairs, Birmingham, Alabama, USA
| | - James F George
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA; Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama, USA; Comprehensive Cardiovascular Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
204
|
Shirakabe A, Zhai P, Ikeda Y, Saito T, Maejima Y, Hsu CP, Nomura M, Egashira K, Levine B, Sadoshima J. Drp1-Dependent Mitochondrial Autophagy Plays a Protective Role Against Pressure Overload-Induced Mitochondrial Dysfunction and Heart Failure. Circulation 2016; 133:1249-63. [PMID: 26915633 DOI: 10.1161/circulationaha.115.020502] [Citation(s) in RCA: 325] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Accepted: 01/29/2016] [Indexed: 01/14/2023]
Abstract
BACKGROUND Mitochondrial autophagy is an important mediator of mitochondrial quality control in cardiomyocytes. The occurrence of mitochondrial autophagy and its significance during cardiac hypertrophy are not well understood. METHODS AND RESULTS Mice were subjected to transverse aortic constriction (TAC) and observed at multiple time points up to 30 days. Cardiac hypertrophy developed after 5 days, the ejection fraction was reduced after 14 days, and heart failure was observed 30 days after TAC. General autophagy was upregulated between 1 and 12 hours after TAC but was downregulated below physiological levels 5 days after TAC. Mitochondrial autophagy, evaluated by electron microscopy, mitochondrial content, and Keima with mitochondrial localization signal, was transiently activated at ≈3 to 7 days post-TAC, coinciding with mitochondrial translocation of Drp1. However, it was downregulated thereafter, followed by mitochondrial dysfunction. Haploinsufficiency of Drp1 abolished mitochondrial autophagy and exacerbated the development of both mitochondrial dysfunction and heart failure after TAC. Injection of Tat-Beclin 1, a potent inducer of autophagy, but not control peptide, on day 7 after TAC, partially rescued mitochondrial autophagy and attenuated mitochondrial dysfunction and heart failure induced by overload. Haploinsufficiency of either drp1 or beclin 1 prevented the rescue by Tat-Beclin 1, suggesting that its effect is mediated in part through autophagy, including mitochondrial autophagy. CONCLUSIONS Mitochondrial autophagy is transiently activated and then downregulated in the mouse heart in response to pressure overload. Downregulation of mitochondrial autophagy plays an important role in mediating the development of mitochondrial dysfunction and heart failure, whereas restoration of mitochondrial autophagy attenuates dysfunction in the heart during pressure overload.
Collapse
Affiliation(s)
- Akihiro Shirakabe
- From Department of Cell Biology and Molecular Medicine, Rutgers-New Jersey Medical School, Newark (A.S., P.Z., Y.I., T.S., Y.M., J.S.); Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Japan (Y.I.); Department of Medicine and Bioregulatory Science, Kyushu University, Fukuoka, Japan (M.N.); Division of Cardiovascular Surgery, Department of Surgery, Veterans General Hospital, National Yang-Ming University School of Medicine, Taiwan (C.-P.H.); Department of Cardiovascular Medicine, Kyushu University Hospital, Fukuoka, Japan (K.E.); Department of Cardiovascular Research, Development, and Translational Medicine, Graduate School of Medical Science, Kyushu University Hospital, Fukuoka, Japan (K.E.); and Center for Autophagy Research, Department of Internal Medicine, and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas (B.L.)
| | - Peiyong Zhai
- From Department of Cell Biology and Molecular Medicine, Rutgers-New Jersey Medical School, Newark (A.S., P.Z., Y.I., T.S., Y.M., J.S.); Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Japan (Y.I.); Department of Medicine and Bioregulatory Science, Kyushu University, Fukuoka, Japan (M.N.); Division of Cardiovascular Surgery, Department of Surgery, Veterans General Hospital, National Yang-Ming University School of Medicine, Taiwan (C.-P.H.); Department of Cardiovascular Medicine, Kyushu University Hospital, Fukuoka, Japan (K.E.); Department of Cardiovascular Research, Development, and Translational Medicine, Graduate School of Medical Science, Kyushu University Hospital, Fukuoka, Japan (K.E.); and Center for Autophagy Research, Department of Internal Medicine, and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas (B.L.)
| | - Yoshiyuki Ikeda
- From Department of Cell Biology and Molecular Medicine, Rutgers-New Jersey Medical School, Newark (A.S., P.Z., Y.I., T.S., Y.M., J.S.); Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Japan (Y.I.); Department of Medicine and Bioregulatory Science, Kyushu University, Fukuoka, Japan (M.N.); Division of Cardiovascular Surgery, Department of Surgery, Veterans General Hospital, National Yang-Ming University School of Medicine, Taiwan (C.-P.H.); Department of Cardiovascular Medicine, Kyushu University Hospital, Fukuoka, Japan (K.E.); Department of Cardiovascular Research, Development, and Translational Medicine, Graduate School of Medical Science, Kyushu University Hospital, Fukuoka, Japan (K.E.); and Center for Autophagy Research, Department of Internal Medicine, and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas (B.L.)
| | - Toshiro Saito
- From Department of Cell Biology and Molecular Medicine, Rutgers-New Jersey Medical School, Newark (A.S., P.Z., Y.I., T.S., Y.M., J.S.); Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Japan (Y.I.); Department of Medicine and Bioregulatory Science, Kyushu University, Fukuoka, Japan (M.N.); Division of Cardiovascular Surgery, Department of Surgery, Veterans General Hospital, National Yang-Ming University School of Medicine, Taiwan (C.-P.H.); Department of Cardiovascular Medicine, Kyushu University Hospital, Fukuoka, Japan (K.E.); Department of Cardiovascular Research, Development, and Translational Medicine, Graduate School of Medical Science, Kyushu University Hospital, Fukuoka, Japan (K.E.); and Center for Autophagy Research, Department of Internal Medicine, and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas (B.L.)
| | - Yasuhiro Maejima
- From Department of Cell Biology and Molecular Medicine, Rutgers-New Jersey Medical School, Newark (A.S., P.Z., Y.I., T.S., Y.M., J.S.); Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Japan (Y.I.); Department of Medicine and Bioregulatory Science, Kyushu University, Fukuoka, Japan (M.N.); Division of Cardiovascular Surgery, Department of Surgery, Veterans General Hospital, National Yang-Ming University School of Medicine, Taiwan (C.-P.H.); Department of Cardiovascular Medicine, Kyushu University Hospital, Fukuoka, Japan (K.E.); Department of Cardiovascular Research, Development, and Translational Medicine, Graduate School of Medical Science, Kyushu University Hospital, Fukuoka, Japan (K.E.); and Center for Autophagy Research, Department of Internal Medicine, and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas (B.L.)
| | - Chiao-Po Hsu
- From Department of Cell Biology and Molecular Medicine, Rutgers-New Jersey Medical School, Newark (A.S., P.Z., Y.I., T.S., Y.M., J.S.); Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Japan (Y.I.); Department of Medicine and Bioregulatory Science, Kyushu University, Fukuoka, Japan (M.N.); Division of Cardiovascular Surgery, Department of Surgery, Veterans General Hospital, National Yang-Ming University School of Medicine, Taiwan (C.-P.H.); Department of Cardiovascular Medicine, Kyushu University Hospital, Fukuoka, Japan (K.E.); Department of Cardiovascular Research, Development, and Translational Medicine, Graduate School of Medical Science, Kyushu University Hospital, Fukuoka, Japan (K.E.); and Center for Autophagy Research, Department of Internal Medicine, and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas (B.L.)
| | - Masatoshi Nomura
- From Department of Cell Biology and Molecular Medicine, Rutgers-New Jersey Medical School, Newark (A.S., P.Z., Y.I., T.S., Y.M., J.S.); Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Japan (Y.I.); Department of Medicine and Bioregulatory Science, Kyushu University, Fukuoka, Japan (M.N.); Division of Cardiovascular Surgery, Department of Surgery, Veterans General Hospital, National Yang-Ming University School of Medicine, Taiwan (C.-P.H.); Department of Cardiovascular Medicine, Kyushu University Hospital, Fukuoka, Japan (K.E.); Department of Cardiovascular Research, Development, and Translational Medicine, Graduate School of Medical Science, Kyushu University Hospital, Fukuoka, Japan (K.E.); and Center for Autophagy Research, Department of Internal Medicine, and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas (B.L.)
| | - Kensuke Egashira
- From Department of Cell Biology and Molecular Medicine, Rutgers-New Jersey Medical School, Newark (A.S., P.Z., Y.I., T.S., Y.M., J.S.); Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Japan (Y.I.); Department of Medicine and Bioregulatory Science, Kyushu University, Fukuoka, Japan (M.N.); Division of Cardiovascular Surgery, Department of Surgery, Veterans General Hospital, National Yang-Ming University School of Medicine, Taiwan (C.-P.H.); Department of Cardiovascular Medicine, Kyushu University Hospital, Fukuoka, Japan (K.E.); Department of Cardiovascular Research, Development, and Translational Medicine, Graduate School of Medical Science, Kyushu University Hospital, Fukuoka, Japan (K.E.); and Center for Autophagy Research, Department of Internal Medicine, and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas (B.L.)
| | - Beth Levine
- From Department of Cell Biology and Molecular Medicine, Rutgers-New Jersey Medical School, Newark (A.S., P.Z., Y.I., T.S., Y.M., J.S.); Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Japan (Y.I.); Department of Medicine and Bioregulatory Science, Kyushu University, Fukuoka, Japan (M.N.); Division of Cardiovascular Surgery, Department of Surgery, Veterans General Hospital, National Yang-Ming University School of Medicine, Taiwan (C.-P.H.); Department of Cardiovascular Medicine, Kyushu University Hospital, Fukuoka, Japan (K.E.); Department of Cardiovascular Research, Development, and Translational Medicine, Graduate School of Medical Science, Kyushu University Hospital, Fukuoka, Japan (K.E.); and Center for Autophagy Research, Department of Internal Medicine, and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas (B.L.)
| | - Junichi Sadoshima
- From Department of Cell Biology and Molecular Medicine, Rutgers-New Jersey Medical School, Newark (A.S., P.Z., Y.I., T.S., Y.M., J.S.); Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Japan (Y.I.); Department of Medicine and Bioregulatory Science, Kyushu University, Fukuoka, Japan (M.N.); Division of Cardiovascular Surgery, Department of Surgery, Veterans General Hospital, National Yang-Ming University School of Medicine, Taiwan (C.-P.H.); Department of Cardiovascular Medicine, Kyushu University Hospital, Fukuoka, Japan (K.E.); Department of Cardiovascular Research, Development, and Translational Medicine, Graduate School of Medical Science, Kyushu University Hospital, Fukuoka, Japan (K.E.); and Center for Autophagy Research, Department of Internal Medicine, and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas (B.L.).
| |
Collapse
|
205
|
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: 8.7] [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
|
206
|
The dual role of autophagy under hypoxia-involvement of interaction between autophagy and apoptosis. Apoptosis 2016; 20:769-77. [PMID: 25721361 DOI: 10.1007/s10495-015-1110-8] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Hypoxia is one of severe cellular stress and it is well known to be associated with a worse outcome since a lack of oxygen accelerates the induction of apoptosis. Autophagy, an important and evolutionarily conserved mechanism for maintaining cellular homeostasis, is closely related to the apoptosis caused by hypoxia. Generally autophagy blocks the induction of apoptosis and inhibits the activation of apoptosis-associated caspase which could reduce cellular injury. However, in special cases, autophagy or autophagy-relevant proteins may help to induce apoptosis, which could aggravate cell damage under hypoxia condition. In addition, the activation of apoptosis-related proteins-caspase can also degrade autophagy-related proteins, such as Atg3, Atg4, Beclin1 protein, inhibiting autophagy. Although the relationship between autophagy and apoptosis has been known for rather complex for more than a decade, the underlying regulatory mechanisms have not been clearly understood. This short review discusses and summarizes the dual role of autophagy and the interaction and molecular regulatory mechanisms between autophagy and apoptosis under hypoxia.
Collapse
|
207
|
Chang C, Su H, Zhang D, Wang Y, Shen Q, Liu B, Huang R, Zhou T, Peng C, Wong C, Shen HM, Lippincott-Schwartz J, Liu W. AMPK-Dependent Phosphorylation of GAPDH Triggers Sirt1 Activation and Is Necessary for Autophagy upon Glucose Starvation. Mol Cell 2015; 60:930-40. [PMID: 26626483 DOI: 10.1016/j.molcel.2015.10.037] [Citation(s) in RCA: 220] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 09/10/2015] [Accepted: 10/22/2015] [Indexed: 12/23/2022]
Abstract
Eukaryotes initiate autophagy to cope with the lack of external nutrients, which requires the activation of the nicotinamide adenine dinucleotide (NAD(+))-dependent deacetylase Sirtuin 1 (Sirt1). However, the mechanisms underlying the starvation-induced Sirt1 activation for autophagy initiation remain unclear. Here, we demonstrate that glyceraldehyde 3-phosphate dehydrogenase (GAPDH), a conventional glycolytic enzyme, is a critical mediator of AMP-activated protein kinase (AMPK)-driven Sirt1 activation. Under glucose starvation, but not amino acid starvation, cytoplasmic GAPDH is phosphorylated on Ser122 by activated AMPK. This causes GAPDH to redistribute into the nucleus. Inside the nucleus, GAPDH interacts directly with Sirt1, displacing Sirt1's repressor and causing Sirt1 to become activated. Preventing this shift of GAPDH abolishes Sirt1 activation and autophagy, while enhancing it, through overexpression of nuclear-localized GAPDH, increases Sirt1 activation and autophagy. GAPDH is thus a pivotal and central regulator of autophagy under glucose deficiency, undergoing AMPK-dependent phosphorylation and nuclear translocation to activate Sirt1 deacetylase activity.
Collapse
|
208
|
Kaus A, Sareen D. ALS Patient Stem Cells for Unveiling Disease Signatures of Motoneuron Susceptibility: Perspectives on the Deadly Mitochondria, ER Stress and Calcium Triad. Front Cell Neurosci 2015; 9:448. [PMID: 26635528 PMCID: PMC4652136 DOI: 10.3389/fncel.2015.00448] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 11/02/2015] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a largely sporadic progressive neurodegenerative disease affecting upper and lower motoneurons (MNs) whose specific etiology is incompletely understood. Mutations in superoxide dismutase-1 (SOD1), TAR DNA-binding protein 43 (TARDBP/TDP-43) and C9orf72, have been identified in subsets of familial and sporadic patients. Key associated molecular and neuropathological features include ubiquitinated TDP-43 inclusions, stress granules, aggregated dipeptide proteins from mutant C9orf72 transcripts, altered mitochondrial ultrastructure, dysregulated calcium homeostasis, oxidative and endoplasmic reticulum (ER) stress, and an unfolded protein response (UPR). Such impairments have been documented in ALS animal models; however, whether these mechanisms are initiating factors or later consequential events leading to MN vulnerability in ALS patients is debatable. Human induced pluripotent stem cells (iPSCs) are a valuable tool that could resolve this “chicken or egg” causality dilemma. Relevant systems for probing pathophysiologically affected cells from large numbers of ALS patients and discovering phenotypic disease signatures of early MN susceptibility are described. Performing unbiased ‘OMICS and high-throughput screening in relevant neural cells from a cohort of ALS patient iPSCs, and rescuing mitochondrial and ER stress impairments, can identify targeted therapeutics for increasing MN longevity in ALS.
Collapse
Affiliation(s)
- Anjoscha Kaus
- Board of Governors-Regenerative Medicine Institute, Cedars-Sinai Medical Center Los Angeles, CA, USA ; Department of Biomedical Sciences, Cedars-Sinai Medical Center Los Angeles, CA, USA
| | - Dhruv Sareen
- Board of Governors-Regenerative Medicine Institute, Cedars-Sinai Medical Center Los Angeles, CA, USA ; Department of Biomedical Sciences, Cedars-Sinai Medical Center Los Angeles, CA, USA ; iPSC Core, The David and Janet Polak Stem Cell Laboratory, Cedars-Sinai Medical Center Los Angeles, CA, USA
| |
Collapse
|
209
|
Wang XY, Fan XS, Cai L, Liu S, Cong XF, Chen X. Lysophosphatidic acid rescues bone mesenchymal stem cells from hydrogen peroxide-induced apoptosis. Apoptosis 2015; 20:273-84. [PMID: 25633408 DOI: 10.1007/s10495-014-1074-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The increase of reactive oxygen species in infracted heart significantly reduces the survival of donor mesenchymal stem cells, thereby attenuating the therapeutic efficacy for myocardial infarction. In our previous study, we demonstrated that lysophosphatidic acid (LPA) protects bone marrow-derived mesenchymal stem cells (BMSCs) against hypoxia and serum deprivation-induced apoptosis. However, whether LPA protects BMSCs from H2O2-induced apoptosis was not examined. In this study, we report that H2O2 induces rat BMSC apoptosis whereas LPA pre-treatment effectively protects BMSCs from H2O2-induced apoptosis. LPA protection of BMSC from the induced apoptosis is mediated mostly through LPA3 receptor. Furthermore, we found that membrane G protein Gi2 and Gi3 are involved in LPA-elicited anti-apoptotic effects through activation of ERK1/2- and PI3 K-pathways. Additionally, H2O2 increases levels of type II of light chain 3B (LC3B II), an autophagy marker, and H2O2-induced autophagy thus protected BMSCs from apoptosis. LPA further increases the expression of LC3B II in the presence of H2O2. In contrast, autophagy flux inhibitor bafilomycin A1 has no effect on LPA's protection of BMSC from H2O2-induced apoptosis. Taken together, our data suggest that LPA rescues H2O2-induced apoptosis mainly by interacting with Gi-coupled LPA3, resulting activation of the ERK1/2- and PI3 K/AKT-pathways and inhibition caspase-3 cleavage, and LPA protection of BMSCs against the apoptosis is independent of it induced autophagy.
Collapse
Affiliation(s)
- Xian-Yun Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Centre for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | | | | | | | | | | |
Collapse
|
210
|
Maejima Y, Isobe M, Sadoshima J. Regulation of autophagy by Beclin 1 in the heart. J Mol Cell Cardiol 2015; 95:19-25. [PMID: 26546165 DOI: 10.1016/j.yjmcc.2015.10.032] [Citation(s) in RCA: 176] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 10/19/2015] [Accepted: 10/29/2015] [Indexed: 12/12/2022]
Abstract
Dysregulation of autophagy in cardiomyocytes is implicated in various heart disease conditions. Beclin 1, a mammalian ortholog of yeast Atg6 and a core component of the autophagy machinery, plays a central role in the regulation of autophagy through activation of Vps34. Beclin 1's ability to activate Vps34 is tightly regulated via transcriptional regulation, miRNA, post-translational modification, and interaction with Beclin 1 binding proteins. Of these mechanisms, binding of Beclin 1 with Bcl-2 family proteins (Bcl-2/XL) that negatively regulate autophagy activity has been shown to be both positively and negatively regulated by various kinases, including DAPK, ROCK1, Mst1 and JNK1, in response to external stimuli. Beclin 1's interaction with Bcl-2/XL also secondarily affects apoptosis through regulation of pro-apoptotic BH3 domain containing proteins. Thus, modulation of Beclin 1 significantly influences both autophagy and apoptosis, thereby deeply affecting the survival and death of cardiomyocytes in the heart. In this review, we discuss the signaling mechanism of autophagy modulation through Beclin 1 and therapeutic potential of Beclin 1 in heart diseases.
Collapse
Affiliation(s)
- Yasuhiro Maejima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, Newark, NJ, USA; Department of Cardiovascular Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mitsuaki Isobe
- Department of Cardiovascular Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, Newark, NJ, USA.
| |
Collapse
|
211
|
Jian J, Xuan F, Qin F, Huang R. Bauhinia championii flavone inhibits apoptosis and autophagy via the PI3K/Akt pathway in myocardial ischemia/reperfusion injury in rats. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:5933-45. [PMID: 26604691 PMCID: PMC4642812 DOI: 10.2147/dddt.s92549] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This study aimed to determine the effects of Bauhinia championii flavone (BCF) on myocardial ischemia/reperfusion injury (MI/RI) in rats and to explore potential mechanisms. The MI/RI model in rats was established by ligating the left anterior descending coronary artery for 30 minutes, then reperfusing for 3 hours. BCF at 20 mg/kg was given 20 minutes prior to ischemia via sublingual intravenous injection, with 24 μg/kg phosphoinositide 3-kinase inhibitor (PI3K; wortmannin) as a control. The creatine kinase-MB and nitric oxide content were assessed by colorimetry. The levels of mitochondrial permeability transition pores and tumor necrosis factor alpha were determined by an enzyme-linked immunosorbent assay. Cardiomyocyte apoptosis was detected by the terminal deoxynucleotidyl transferase dUTP nick end labeling assay. Additionally, the expression of PI3K, endothelial nitric oxide synthase, caspase-3, and Beclin1 was analyzed by fluorescence quantitative polymerase chain reaction and Western blotting, respectively. Akt and microtubule-associated protein 1 light chain 3-II protein levels were also evaluated. Pretreatment with BCF significantly decreased the levels of creatine kinase-MB, tumor necrosis factor alpha, and mitochondrial permeability transition pores, but increased the nitric oxide content. Furthermore, BCF inhibited apoptosis, downregulated caspase-3, Beclin1, and microtubule-associated protein 1 light chain 3-II, upregulated PI3K, and increased the protein levels of phosphorylated Akt and endothelial nitric oxide synthase. However, all of the previously mentioned effects of BCF were blocked when BCF was coadministered with wortmannin. In conclusion, these observations indicated that BCF has cardioprotective effects against MI/RI by reducing cell apoptosis and excessive autophagy, which might be related to the activation of the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Jie Jian
- Department of Pharmacology, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Feifei Xuan
- Department of Pharmacology, Guangxi Medical University, Nanning, People's Republic of China
| | - Feizhang Qin
- Department of Pharmacology, Guangxi Medical University, Nanning, People's Republic of China
| | - Renbin Huang
- Department of Pharmacology, Guangxi Medical University, Nanning, People's Republic of China
| |
Collapse
|
212
|
Cardiac-specific overexpression of metallothionein attenuates myocardial remodeling and contractile dysfunction in l-NAME-induced experimental hypertension: Role of autophagy regulation. Toxicol Lett 2015; 237:121-32. [DOI: 10.1016/j.toxlet.2015.06.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 06/05/2015] [Accepted: 06/05/2015] [Indexed: 12/20/2022]
|
213
|
Jiang H, Xiao J, Kang B, Zhu X, Xin N, Wang Z. PI3K/SGK1/GSK3β signaling pathway is involved in inhibition of autophagy in neonatal rat cardiomyocytes exposed to hypoxia/reoxygenation by hydrogen sulfide. Exp Cell Res 2015; 345:134-40. [PMID: 26163895 DOI: 10.1016/j.yexcr.2015.07.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 06/19/2015] [Accepted: 07/06/2015] [Indexed: 11/16/2022]
Abstract
Excessive autophagy aggravates myocardial ischemia/reperfusion (IR) injury. Hydrogen sulfide (H2S) has been shown to possess a strong cardioprotective effect due to its anti-necrosis, anti-apoptosis, anti-oxidant and anti-inflammatory properties. Our previous study showed that H2S could also protect the myocardium against IR injury through its anti-autophagy effect in vivo, but the underlying mechanism remains unclear. The aim of the present study was to determine whether PI3K/SGK1/GSK3β signaling pathway was involved in the anti-autophagy effect of H2S against myocardial hypoxia/reoxygenation (HR) injury in vitro. Autophagy was significantly increased in cardiomyocytes subjected to HR, but it was down-regulated by H2S (NaHS donor). Blocking PI3K by LY294002 (a PI3K inhibitor) or knocking down SGK1 by SGK1 siRNA augmented autophagy and attenuated the anti-autophagy effect of H2S. However, blocking GSK3β by tws119 (a GSK3β inhibitor) produced an opposite effect. In addition, while treatment of neonatal rat cardiomyocytes with HR reduced cell viability and augmented cell injury, H2S significantly reversed it. Blocking PI3K or knocking down SGK1 aggravated HR injury and weakened the protective effect of H2S, while blocking GSK3β produced an opposite effect. In conclusion, H2S can inhibit autophagy in neonatal rat cardiomyocytes exposed to H/R and exert a cardioprotective effect at least partly by regulating PI3K/SGK1/GSK3β signaling pathway.
Collapse
Affiliation(s)
- Huan Jiang
- Department of Cardiothoracic Surgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Jian Xiao
- Department of Cardiothoracic Surgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Bo Kang
- Department of Cardiothoracic Surgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Xiaoyan Zhu
- Department of Physiology, Second Military Medical University, Shanghai, China
| | - Ni Xin
- Department of Physiology, Second Military Medical University, Shanghai, China.
| | - Zhinong Wang
- Department of Cardiothoracic Surgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China.
| |
Collapse
|
214
|
Ebselen reduces autophagic activation and cell death in the ipsilateral thalamus following focal cerebral infarction. Neurosci Lett 2015; 600:206-12. [DOI: 10.1016/j.neulet.2015.06.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 06/09/2015] [Accepted: 06/11/2015] [Indexed: 12/19/2022]
|
215
|
Ren J, Taegtmeyer H. Too much or not enough of a good thing — The Janus faces of autophagy in cardiac fuel and protein homeostasis. J Mol Cell Cardiol 2015; 84:223-6. [DOI: 10.1016/j.yjmcc.2015.03.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 02/23/2015] [Accepted: 03/02/2015] [Indexed: 01/01/2023]
|
216
|
Li L, Tan J, Miao Y, Lei P, Zhang Q. ROS and Autophagy: Interactions and Molecular Regulatory Mechanisms. Cell Mol Neurobiol 2015; 35:615-21. [PMID: 25722131 DOI: 10.1007/s10571-015-0166-x] [Citation(s) in RCA: 593] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 02/17/2015] [Indexed: 12/15/2022]
Abstract
Reactive oxygen species (ROS) and antioxidant ingredients are a series of crucial signaling molecules in oxidative stress response. Under some pathological conditions such as traumatic brain injury, ischemia/reperfusion, and hypoxia in tumor, the relative excessive accumulation of ROS could break cellular homeostasis, resulting in oxidative stress and mitochondrial dysfunction. Meanwhile, autophagy is also induced. In this process, oxidative stress could promote the formation of autophagy. Autophagy, in turn, may contribute to reduce oxidative damages by engulfing and degradating oxidized substance. This short review summarizes these interactions between ROS and autophagy in related pathological conditions referred to as above with a focus on discussing internal regulatory mechanisms. The tight interactions between ROS and autophagy reflected in two aspects: the induction of autophagy by oxidative stress and the reduction of ROS by autophagy. The internal regulatory mechanisms of autophagy by ROS can be summarized as transcriptional and post-transcriptional regulation, which includes various molecular signal pathways such as ROS-FOXO3-LC3/BNIP3-autophagy, ROS-NRF2-P62-autophagy, ROS-HIF1-BNIP3/NIX-autophagy, and ROS-TIGAR-autophagy. Autophagy also may regulate ROS levels through several pathways such as chaperone-mediated autophagy pathway, mitophagy pathway, and P62 delivery pathway, which might provide a further theoretical basis for the pathogenesis of the related diseases and still need further research.
Collapse
Affiliation(s)
- Lulu Li
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Anshan Road NO.154, Tianjin, 300052, China
| | | | | | | | | |
Collapse
|
217
|
Levine B, Liu R, Dong X, Zhong Q. Beclin orthologs: integrative hubs of cell signaling, membrane trafficking, and physiology. Trends Cell Biol 2015; 25:533-44. [PMID: 26071895 PMCID: PMC4554927 DOI: 10.1016/j.tcb.2015.05.004] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 05/04/2015] [Accepted: 05/07/2015] [Indexed: 12/25/2022]
Abstract
Beclin orthologs are crucial regulators of autophagy and related membrane-trafficking pathways. Multiple signaling pathways converge on Beclin 1 to regulate cellular stress responses, membrane trafficking, and physiology.
The Beclin family, including yeast Atg6 (autophagy related gene 6), its orthologs in higher eukaryotic species, and the more recently characterized mammalian-specific Beclin 2, are essential molecules in autophagy and other membrane-trafficking events. Extensive studies of Beclin orthologs have provided considerable insights into the regulation of autophagy, the diverse roles of autophagy in physiology and disease, and potential new strategies to modulate autophagy in a variety of clinical diseases. In this review we discuss the functions of Beclin orthologs, the regulation of such functions by diverse cellular signaling pathways, and the effects of such regulation on downstream cellular processes including tumor suppression and metabolism. These findings suggest that Beclin orthologs serve as crucial molecules that integrate diverse environmental signals with membrane trafficking events to ensure optimal responses of the cell to stressful stimuli.
Collapse
Affiliation(s)
- Beth Levine
- Center for Autophagy Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Rong Liu
- Center for Autophagy Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xiaonan Dong
- Center for Autophagy Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Qing Zhong
- Center for Autophagy Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
218
|
Lin WJ, Kuang HY. Oxidative stress induces autophagy in response to multiple noxious stimuli in retinal ganglion cells. Autophagy 2015; 10:1692-701. [PMID: 25207555 PMCID: PMC4198355 DOI: 10.4161/auto.36076] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Retinal ganglion cells (RGCs) are the only afferent neurons that can transmit visual information to the brain. The death of RGCs occurs in the early stages of glaucoma, diabetic retinopathy, and many other retinal diseases. Autophagy is a highly conserved lysosomal pathway, which is crucial for maintaining cellular homeostasis and cell survival under stressful conditions. Research has established that autophagy exists in RGCs after increasing intraocular pressure (IOP), retinal ischemia, optic nerve transection (ONT), axotomy, or optic nerve crush. However, the mechanism responsible for defining how autophagy is induced in RGCs has not been elucidated. Accumulating data has pointed to an essential role of reactive oxygen species (ROS) in the activation of autophagy. RGCs have long axons with comparatively high densities of mitochondria. This makes them more sensitive to energy deficiency and vulnerable to oxidative stress. In this review, we explore the role of oxidative stress in the activation of autophagy in RGCs, and discuss the possible mechanisms that are involved in this process. We aim to provide a more theoretical basis of oxidative stress-induced autophagy, and provide innovative targets for therapeutic intervention in retinopathy.
Collapse
|
219
|
Chen D, Zhang Z, Yao H, Liang Y, Xing H, Xu S. Effects of atrazine and chlorpyrifos on oxidative stress-induced autophagy in the immune organs of common carp (Cyprinus carpio L.). FISH & SHELLFISH IMMUNOLOGY 2015; 44:12-20. [PMID: 25652291 DOI: 10.1016/j.fsi.2015.01.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 01/11/2015] [Accepted: 01/22/2015] [Indexed: 06/04/2023]
Abstract
Atrazine (ATR) and chlorpyrifos (CPF) are the most common agrochemical in the freshwater ecosystems of the world. This study assessed the effects of ATR (4.28, 42.8 and 428 μg/L), CPF (1.16, 11.6 and 116 μg/L) and combined ATR/CPF (1.13, 11.3 and 113 μg/L) on common carp head kidneys and spleens following 40 d exposure and 40 d recovery treatments. Nitric oxide (NO) content, activities of anti hydroxyl radical (AHR), anti superoxide anion (ASA), peroxidase (POD) and inducible nitric oxide synthase (iNOS), and the mRNA levels of the autophagy genes (LC3-II, dynein, TOR) were determined. The results indicate that the antioxidant enzyme (AHR, ASA, POD and iNOS) activities and NO content in the head kidney and spleen of the common carp increased significantly after a 40 d exposure to ATR and CPF alone or in combination. The mRNA levels of LC3-II and dynein in common carp increased significantly after exposure to ATR and CPF alone, or in combination. Moreover, the mRNA levels of LC3-II and dynein decreased significantly after a 40-d recovery. However, the mRNA levels of TOR gene for all decreased significantly at the end of the exposure and the recovery. To our knowledge, this is the first study to report the oxidative stress-induced autophagic effects in the common carp by exposure to ATR, CPF and the ATR/CPF combination. The information presented in the present study may be helpful to understanding the mechanisms of autophagy induced by ATR, CPF and the ATR/CPF combination in fish.
Collapse
Affiliation(s)
- Dechun Chen
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Harbin 150030, PR China; Department of Biological Engineering, Jilin Engineering Vocational College, 1299 Changfa Road, Siping 136001, China
| | - Ziwei Zhang
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Harbin 150030, PR China
| | - Haidong Yao
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Harbin 150030, PR China
| | - Yang Liang
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Harbin 150030, PR China
| | - Houjuan Xing
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Harbin 150030, PR China; Animal Health Supervision Institute of Heilongjiang Province, 243 Haping Road, Xiangfang District, Harbin 150069, PR China.
| | - Shiwen Xu
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Harbin 150030, PR China.
| |
Collapse
|
220
|
Abstract
Autophagy is an important physiological process in the heart, and alterations in autophagic activity can exacerbate or mitigate injury during various pathological processes. Methods to assess autophagy have changed rapidly because the field of research has expanded. As with any new field, methods and standards for data analysis and interpretation evolve as investigators acquire experience and insight. The purpose of this review is to summarize current methods to measure autophagy, selective mitochondrial autophagy (mitophagy), and autophagic flux. We will examine several published studies where confusion arose in data interpretation, to illustrate the challenges. Finally, we will discuss methods to assess autophagy in vivo and in patients.
Collapse
Affiliation(s)
- Roberta A Gottlieb
- From the Cedars-Sinai Heart Institute and the Barbra Streisand Women's Heart Center Cedars-Sinai Medical Center, Los Angeles, CA.
| | - Allen M Andres
- From the Cedars-Sinai Heart Institute and the Barbra Streisand Women's Heart Center Cedars-Sinai Medical Center, Los Angeles, CA
| | - Jon Sin
- From the Cedars-Sinai Heart Institute and the Barbra Streisand Women's Heart Center Cedars-Sinai Medical Center, Los Angeles, CA
| | - David P J Taylor
- From the Cedars-Sinai Heart Institute and the Barbra Streisand Women's Heart Center Cedars-Sinai Medical Center, Los Angeles, CA
| |
Collapse
|
221
|
Huang L, Dai K, Chen M, Zhou W, Wang X, Chen J, Zhou W. The AMPK Agonist PT1 and mTOR Inhibitor 3HOI-BA-01 Protect Cardiomyocytes After Ischemia Through Induction of Autophagy. J Cardiovasc Pharmacol Ther 2015; 21:70-81. [PMID: 25868658 DOI: 10.1177/1074248415581177] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Myocardial ischemia has become one of the main causes of sudden cardiac death worldwide. Autophagy has been demonstrated to protect cardiomyocytes from ischemia/reperfusion (I/R)-induced damage. A novel small molecule compound 2-Chloro-5-[[5-[[5-(4,5-Dimethyl-2-nitrophenyl)-2-furanyl]methylene]-4,5-dihydro-4-oxo-2-thiazolyl]amino]benzoic acid (PT1) has been previously shown to specifically activate 5'-adenosine monophosphate-activated protein kinase (AMPK). Because AMPK activation effectively induces autophagy, we tested the protective efficacy of PT1 on cardiomyocytes after oxygen glucose deprivation/reoxygenation (OGD/R) in vitro. Mouse neonatal cardiomyocytes were treated with PT1 after OGD/R. 3-[4-(1,3-benzodioxol-5-yl)-2-oxo-3-buten-1-yl]-3-hydroxy-1,3-dihydro-2H-indol-2-one (3HOI-BA-01), a novel small compound showing potent inhibitory effect on mammalian target of rapamycin (mTOR) activation, was also tested for its cardioprotective effect, based on the established relationship between mTOR signaling and autophagy. Cell survival and autophagy-related signal pathways were examined after treatment with these agents. Our data indicate that both PT1 and 3HOI-BA-01 enhance cell survival after OGD/R. As expected, both PT1 and 3HOI-BA-01 induced autophagy in cardiomyocytes through activating AMPK pathway and inhibiting mTOR signaling, respectively. Induction of autophagy by PT1 and 3HOI-BA-01 was responsible for their cardioprotective effect, since inhibition of autophagy abolished the protective efficacy. Furthermore, simultaneous administration of PT1 and 3HOI-BA-01 profoundly upregulated autophagy after OGD/R and significantly promoted survival of cardiomyocytes. In vivo administration of PT1 and 3HOI-BA-01 in a murine myocardial (I/R injury model remarkably reduced infarct size and induced autophagy. Taken together, our research suggests that PT1 and 3HOI-BA-01 could be promising therapeutic agents for myocardial ischemia.
Collapse
Affiliation(s)
- Ling Huang
- Department of Cardiology, The Central Hospital of Wuhan, Wuhan, China
| | - Kai Dai
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Manhua Chen
- Department of Cardiology, The Central Hospital of Wuhan, Wuhan, China
| | - Wenping Zhou
- Department of Cardiology, The Central Hospital of Wuhan, Wuhan, China
| | - Xiaoling Wang
- Department of Cardiology, The Central Hospital of Wuhan, Wuhan, China
| | - Jing Chen
- Department of Emergency, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Zhou
- Department of Cardiology, The Central Hospital of Wuhan, Wuhan, China
| |
Collapse
|
222
|
Orogo AM, Gustafsson ÅB. Therapeutic targeting of autophagy: potential and concerns in treating cardiovascular disease. Circ Res 2015; 116:489-503. [PMID: 25634972 DOI: 10.1161/circresaha.116.303791] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Autophagy is an evolutionarily conserved process by which long-lived proteins and organelles are sequestered by autophagosomes and subsequently degraded by lysosomes for recycling. Autophagy is important for maintaining cardiac homeostasis and is a survival mechanism that is upregulated during stress or starvation. Accumulating evidence suggests that dysregulated or reduced autophagy is associated with heart failure and aging. Thus, modulating autophagy represents an attractive future therapeutic target for treating cardiovascular disease. Activation of autophagy is generally considered to be cardioprotective, whereas excessive autophagy can lead to cell death and cardiac atrophy. It is important to understand how autophagy is regulated to identify ideal therapeutic targets for treating disease. Here, we discuss the key proteins in the core autophagy machinery and describe upstream regulators that respond to extracellular and intracellular signals to tightly coordinate autophagic activity. We review various genetic and pharmacological studies that demonstrate the important role of autophagy in the heart and consider the advantages and limitations of approaches that modulate autophagy.
Collapse
Affiliation(s)
- Amabel M Orogo
- From the Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla
| | - Åsa B Gustafsson
- From the Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla.
| |
Collapse
|
223
|
Guo L, Xu JM, Mo XY. Ischemic postconditioning regulates cardiomyocyte autophagic activity following ischemia/reperfusion injury. Mol Med Rep 2015; 12:1169-76. [PMID: 25816157 DOI: 10.3892/mmr.2015.3533] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 02/13/2015] [Indexed: 11/05/2022] Open
Abstract
Ischemic postconditioning (IPostC) is a promising protective mechanism for combating reperfusion injury. However, the role of autophagy in the protective effects of IPostC and the associated signaling pathways have remained to be elucidated. Male Sprague Dawley rats were subjected to 30 min ischemia and 1, 2, 3, 6, 12 and 24 h of reperfusion, with or without IPostC treatment. Autophagic flux was evaluated by detecting mRNA and protein expression levels of microtubule associated protein 1 light chain 3 and p62. Phosphorylated (p)-P70S6 kinase (P70S6K), p-adenosine monophosphate-activated kinase (AMPK) and Beclin 1 protein levels were measured by western blot analysis. Myocardial infarct size was measured using staining with Evans blue dye and myocardial apoptosis was evaluated by terminal deoxynucleotidyl transferase mediated dUTP nick end labeling staining. Autophagic activity was observed to be inhibited within the first hour of reperfusion, increased during 2-6 h and reduced from 12-24 h following IPostC compared with reperfusion without IPostC. Inhibition of autophagy by chloroquine significantly reversed the effects of IPostC on myocardial infarct size and the levels of apoptosis. IPostC significantly increased Beclin 1 levels, inhibited AMPK activation and increased P70S6K activation within the first hour compared with reperfusion without IPostC. IPostC attenuated the upregulation of Beclin 1 levels, increased AMPK activation and reduced P70S6K activation between 2 and 12 h, and subsequently exerted the opposite effects on these molecules between 12 and 24 h. IPostC was demonstrated to regulate autophagic activity in a time‑dependent manner. The Beclin 1 and AMPK-mammalian target of rapamycin signaling pathways are suggested to be involved in the regulation of IPostC in autophagy.
Collapse
Affiliation(s)
- Liang Guo
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Jun-Mei Xu
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Xiao-Ye Mo
- Department of Emergency, Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
224
|
Degradation of misfolded proteins in neurodegenerative diseases: therapeutic targets and strategies. Exp Mol Med 2015; 47:e147. [PMID: 25766616 PMCID: PMC4351408 DOI: 10.1038/emm.2014.117] [Citation(s) in RCA: 589] [Impact Index Per Article: 58.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 11/19/2014] [Indexed: 12/13/2022] Open
Abstract
Mammalian cells remove misfolded proteins using various proteolytic systems, including the ubiquitin (Ub)-proteasome system (UPS), chaperone mediated autophagy (CMA) and macroautophagy. The majority of misfolded proteins are degraded by the UPS, in which Ub-conjugated substrates are deubiquitinated, unfolded and cleaved into small peptides when passing through the narrow chamber of the proteasome. The substrates that expose a specific degradation signal, the KFERQ sequence motif, can be delivered to and degraded in lysosomes via the CMA. Aggregation-prone substrates resistant to both the UPS and the CMA can be degraded by macroautophagy, in which cargoes are segregated into autophagosomes before degradation by lysosomal hydrolases. Although most misfolded and aggregated proteins in the human proteome can be degraded by cellular protein quality control, some native and mutant proteins prone to aggregation into β-sheet-enriched oligomers are resistant to all known proteolytic pathways and can thus grow into inclusion bodies or extracellular plaques. The accumulation of protease-resistant misfolded and aggregated proteins is a common mechanism underlying protein misfolding disorders, including neurodegenerative diseases such as Huntington's disease (HD), Alzheimer's disease (AD), Parkinson's disease (PD), prion diseases and Amyotrophic Lateral Sclerosis (ALS). In this review, we provide an overview of the proteolytic pathways in neurons, with an emphasis on the UPS, CMA and macroautophagy, and discuss the role of protein quality control in the degradation of pathogenic proteins in neurodegenerative diseases. Additionally, we examine existing putative therapeutic strategies to efficiently remove cytotoxic proteins from degenerating neurons.
Collapse
|
225
|
Yao T, Ying X, Zhao Y, Yuan A, He Q, Tong H, Ding S, Liu J, Peng X, Gao E, Pu J, He B. Vitamin D receptor activation protects against myocardial reperfusion injury through inhibition of apoptosis and modulation of autophagy. Antioxid Redox Signal 2015; 22:633-50. [PMID: 25365634 PMCID: PMC4346660 DOI: 10.1089/ars.2014.5887] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
AIMS To determine the roles of vitamin D receptor (VDR) in ischemia/reperfusion-induced myocardial injury and to investigate the underlying mechanisms involved. RESULTS The endogenous VDR expression was detected in the mouse heart, and myocardial ischemia/reperfusion (MI/R) upregulated VDR expression. Activation of VDR by natural and synthetic agonists reduced myocardial infarct size and improved cardiac function. Mechanistically, VDR activation inhibited endoplasmic reticulum (ER) stress (determined by the reduction of CCAAT/enhancer-binding protein homologous protein expression and caspase-12 activation), attenuated mitochondrial impairment (determined by the decrease of mitochondrial cytochrome c release and caspase-9 activation), and reduced cardiomyocyte apoptosis. Furthermore, VDR activation significantly inhibited MI/R-induced autophagy dysfunction (determined by the inhibition of Beclin 1 over-activation, the reduction of autophagosomes, the LC3-II/LC3-I ratio, p62 protein abundance, and the restoration of autophagy flux). Moreover, VDR activation inhibited MI/R-induced oxidative stress through a metallothionein-dependent mechanism. The cardioprotective effects of VDR agonists mentioned earlier were impaired in the setting of cardiac-specific VDR silencing. In contrast, adenovirus-mediated cardiac VDR overexpression decreased myocardial infarct size and improved cardiac function through attenuating oxidative stress, and inhibiting apoptosis and autophagy dysfunction. INNOVATION AND CONCLUSION Our data demonstrate that VDR is a novel endogenous self-defensive and cardioprotective receptor against MI/R injury, via mechanisms (at least in part) reducing oxidative stress, and inhibiting apoptosis and autophagy dysfunction-mediated cell death.
Collapse
Affiliation(s)
- Tianbao Yao
- 1 Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
226
|
Assessment of the effect of laser irradiations at different wavelengths (660, 810, 980, and 1064 nm) on autophagy in a rat model of mucositis. Lasers Med Sci 2015; 30:1289-95. [PMID: 25732242 DOI: 10.1007/s10103-015-1727-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 02/18/2015] [Indexed: 02/07/2023]
Abstract
It is known that high-dose radiation has an effect on tissue healing, but tissue healing does not occur when low dose radiation is applied. To clarify this issue, we compare the treatment success of low dose radiation with programmed cell death mechanisms on wounded tissue. In this study, we aimed to investigate the interactions of low and high-dose radiation using an autophagic mechanism. We included 35 adult Wistar-Albino rats in this study. All animals were injected with 100 mg/kg of 5-fluorouracil (5-FU) on the first day and 65 mg/kg of 5-FU on the third day. The tips of 18-gauge needles were used to develop a superficial scratching on the left cheek pouch mucosa by dragging in a linear movement on third and fifth days. After mucositis formation was clinically detected, animals were divided into five groups (n = 7). Different wavelengths of laser irradiations (1064 nm, Fidelis Plus, Fotona, Slovenia; 980 nm, FOX laser, A.R.C., Germany; 810 nm, Fotona XD, Fotona, Slovenia; 660 nm, HELBO, Medizintechnik GmbH, Wels, Austria) were performed on four groups once daily for 4 days. The laser irradiation was not performed on the control group. To get the tissue from the left cheek at the end of fourth day from all animals, oval excisional biopsy was performed. Molecular analysis assessments of pathological and normal tissue taken were performed. For this purpose, the expression analysis of autophagy genes was performed. The results were evaluated by normalization and statistics analysis. We found that Ulk1, Beclin1, and Atg5 expression levels were increased in the rats when the Nd:YAG laser was applied. This increase showed that a 1064-nm laser is needed to activate the autophagic mechanism. However, in the diode applications, we found that Beclin1, Atg10, Atg5, and Atg7 expressions numerically decreased. Atg5 is responsible for the elongation of autophagosome. Becn1 is a control gene in the control mechanism of autophagy. The reduction of the expression of these genes leads us to think that it may depend on the effect of drug (5-FU) used to form model. Expressions of therapeutic genes increase to ensure hemostasis, but in our study, expressions were found to decrease. More detailed studies are needed.
Collapse
|
227
|
Rapamycin protection of livers from ischemia and reperfusion injury is dependent on both autophagy induction and mammalian target of rapamycin complex 2-Akt activation. Transplantation 2015; 99:48-55. [PMID: 25340604 DOI: 10.1097/tp.0000000000000476] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
BACKGROUND Although rapamycin (RPM) have been studied extensively in ischemia models, its functional mechanisms remains to be defined. METHODS We determined how RPM impacted the pathogenesis of ischemia-reperfusion injury (IRI) in a murine liver partial warm ischemia model, with emphasis on its regulation of hepatocyte death. RESULTS Rapamycin protected livers from IRI in the presence of fully developed liver inflammatory immune response. Rapamycin enhanced liver autophagy induction at the reperfusion stage. Dual mammalian (mechanistic) target of rapamycin (mTOR)1/2 inhibitor Torin 1, despite its ability to induced autophagy, failed to protect livers from IRI. The treatment with RPM, but not Torin 1, resulted in the enhanced activation of the mTORC2-Akt signaling pathway activation in livers after reperfusion. Inactivation of Akt by Triciribine abolished the liver protective effect of RPM. The differential cytoprotective effect of RPM and Torin 1 was confirmed in vitro in hepatocyte cultures. Rapamycin, but not Trin 1, protected hepatocytes from stress and tumor necrosis factor-α induced cell death; and inhibition of autophagy by chloroquine or Akt by Triciribine abolished RPM-mediated cytoprotection. CONCLUSION Rapamycin protected livers from IRI by both autophagy and mTORC2-Akt activation mechanisms.
Collapse
|
228
|
Iwasaki M, Sato M, Minagawa K, Manz MC, Yoshihara A, Miyazaki H. Longitudinal relationship between metabolic syndrome and periodontal disease among Japanese adults aged ≥70 years: the Niigata Study. J Periodontol 2015; 86:491-8. [PMID: 25579498 DOI: 10.1902/jop.2015.140398] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND There has been little evaluation in longitudinal epidemiologic studies of the effect of metabolic syndrome (MetS) on periodontal status. The specific aim of this longitudinal study is to investigate whether MetS in the Japanese population could be a risk factor for periodontal disease. METHODS A total of 125 older adults from Japan for whom data were available for the years 2003 to 2006 were selected for the current study. Full-mouth periodontal status, measured as clinical attachment level (CAL), was recorded at baseline and in follow-up examinations. Development of periodontal disease was considered to be ≥2 teeth demonstrating a longitudinal loss of proximal attachment of ≥3 mm at the follow-up dental examination. A multivariable Poisson regression model with robust error variance was used to evaluate the association of MetS defined by the modified National Cholesterol Education Program Adult Treatment Panel III criteria with development of periodontal disease. Adjustments for sex, income, education, smoking status, number of teeth at baseline, mean CAL at baseline, pattern of visits to a dentist, and brushing frequency were considered. RESULTS The prevalence of MetS was 21.6% (27/125). Study participants with MetS were approximately 2.6 times more likely to develop periodontal disease (adjusted relative risk 2.58, 95% confidence interval 1.17 to 5.67) after simultaneous adjustment for other covariates. CONCLUSIONS These findings support the hypothesis that MetS may be a risk factor for periodontal disease in older Japanese individuals. Additional studies with larger, more diverse populations and more complete information are needed to substantiate the findings.
Collapse
Affiliation(s)
- Masanori Iwasaki
- Division of Community Oral Health Development, Kyushu Dental University, Kitakyushu, Japan
| | | | | | | | | | | |
Collapse
|
229
|
Xu W, Jiang H, Hu X, Fu W. Effects of high-mobility group box 1 on the expression of Beclin-1 and LC3 proteins following hypoxia and reoxygenation injury in rat cardiomyocytes. Int J Clin Exp Med 2014; 7:5353-5357. [PMID: 25664043 PMCID: PMC4307490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 12/08/2014] [Indexed: 06/04/2023]
Abstract
The mechanisms underlying autophagy during myocardial ischemia and reperfusion remain unclear. The present study investigated the relationship between high-mobility group box 1 protein (HMGB1) and autophagy in hypoxia/reoxygenation (H/R)-induced neonatal rat cardiomyocytes. Neonatal rat cardiomyocytes were treated with recombinant HMGB1 (200 ng/L) or ammonium glycyrrhizinate (100 μM) at appropriate concentrations. Cell viabilities and lactate dehydrogenase (LDH) and creatine kinase (CK) activity levels were measured. HMGB1, LC3 and Beclin-1 expression were assessed by Western blot. The results demonstrated that HMGB1-induced myocardial cells have increased levels of Beclin-1 protein and even higher levels of LC3 protein, while HMGB1-inhibited myocardial cells have decreased levels of Beclin-1 and LC3 proteins. In addition, HMGB1 induction significantly increased LDH and CK levels in the cell culture medium; the inhibition of HMGB1 significantly reduced LDH and CK expression in cardiomyocyte culture medium. In conclusion, the results of the present study suggest that HMGB1 is able to regulate Beclin-1 and LC3 levels following hypoxia and reoxygenation injury in rat cardiomyocytes.
Collapse
Affiliation(s)
- Weipan Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University Wuhan, China
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University Wuhan, China
| | - Xiaorong Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University Wuhan, China
| | - Wenwen Fu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University Wuhan, China
| |
Collapse
|
230
|
Jimenez RE, Kubli DA, Gustafsson ÅB. Autophagy and mitophagy in the myocardium: therapeutic potential and concerns. Br J Pharmacol 2014; 171:1907-16. [PMID: 24148024 DOI: 10.1111/bph.12477] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 08/30/2013] [Accepted: 09/23/2013] [Indexed: 01/10/2023] Open
Abstract
The autophagic-lysosomal degradation pathway is critical for cardiac homeostasis, and defects in this pathway are associated with development of cardiomyopathy. Autophagy is responsible for the normal turnover of organelles and long-lived proteins. Autophagy is also rapidly up-regulated in response to stress, where it rapidly clears dysfunctional organelles and cytotoxic protein aggregates in the cell. Autophagy is also important in clearing dysfunctional mitochondria before they can cause harm to the cell. This quality control mechanism is particularly important in cardiac myocytes, which contain a very high volume of mitochondria. The degradation of proteins and organelles also generates free fatty acids and amino acids, which help maintain energy levels in myocytes during stress conditions. Increases in autophagy have been observed in various cardiovascular diseases, but a major question that remains to be answered is whether enhanced autophagy is an adaptive or maladaptive response to stress. This review discusses the regulation and role of autophagy in the myocardium under baseline conditions and in various aetiologies of heart disease. It also discusses whether this pathway represents a new therapeutic target to treat or prevent cardiovascular disease and the concerns associated with modulating autophagy.
Collapse
Affiliation(s)
- Rebecca E Jimenez
- Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | | | | |
Collapse
|
231
|
Zaouali MA, Bejaoui M, Calvo M, Folch-Puy E, Pantazi E, Pasut G, Rimola A, Ben Abdennebi H, Adam R, Roselló-Catafau J. Polyethylene glycol rinse solution: An effective way to prevent ischemia-reperfusion injury. World J Gastroenterol 2014; 20:16203-16214. [PMID: 25473175 PMCID: PMC4239509 DOI: 10.3748/wjg.v20.i43.16203] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 05/02/2014] [Accepted: 06/13/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To test whether a new rinse solution containing polyethylene glycol 35 (PEG-35) could prevent ischemia-reperfusion injury (IRI) in liver grafts.
METHODS: Sprague-Dawley rat livers were stored in University of Wisconsin preservation solution and then washed with different rinse solutions (Ringer’s lactate solution and a new rinse solution enriched with PEG-35 at either 1 or 5 g/L) before ex vivo perfusion with Krebs-Heinseleit buffer solution. We assessed the following: liver injury (transaminase levels), mitochondrial damage (glutamate dehydrogenase activity), liver function (bile output and vascular resistance), oxidative stress (malondialdehyde), nitric oxide, liver autophagy (Beclin-1 and LCB3) and cytoskeleton integrity (filament and globular actin fraction); as well as levels of metalloproteinases (MMP2 and MMP9), adenosine monophosphate-activated protein kinase (AMPK), heat shock protein 70 (HSP70) and heme oxygenase 1 (HO-1).
RESULTS: When we used the PEG-35 rinse solution, reduced hepatic injury and improved liver function were noted after reperfusion. The PEG-35 rinse solution prevented oxidative stress, mitochondrial damage, and liver autophagy. Further, it increased the expression of cytoprotective heat shock proteins such as HO-1 and HSP70, activated AMPK, and contributed to the restoration of cytoskeleton integrity after IRI.
CONCLUSION: Using the rinse solution containing PEG-35 was effective for decreasing liver graft vulnerability to IRI.
Collapse
|
232
|
Maejima Y, Chen Y, Isobe M, Gustafsson ÅB, Kitsis RN, Sadoshima J. Recent progress in research on molecular mechanisms of autophagy in the heart. Am J Physiol Heart Circ Physiol 2014; 308:H259-68. [PMID: 25398984 DOI: 10.1152/ajpheart.00711.2014] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Dysregulation of autophagy, an evolutionarily conserved process for degradation of long-lived proteins and organelles, has been implicated in the pathogenesis of human disease. Recent research has uncovered pathways that control autophagy in the heart and molecular mechanisms by which alterations in this process affect cardiac structure and function. Although initially thought to be a nonselective degradation process, autophagy, as it has become increasingly clear, can exhibit specificity in the degradation of molecules and organelles, such as mitochondria. Furthermore, it has been shown that autophagy is involved in a wide variety of previously unrecognized cellular functions, such as cell death and metabolism. A growing body of evidence suggests that deviation from appropriate levels of autophagy causes cellular dysfunction and death, which in turn leads to heart disease. Here, we review recent advances in understanding the role of autophagy in heart disease, highlight unsolved issues, and discuss the therapeutic potential of modulating autophagy in heart disease.
Collapse
Affiliation(s)
- Yasuhiro Maejima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, Newark, New Jersey; Department of Cardiovascular Medicine, Tokyo Medical and Dental University, Tokyo, Japan; and
| | - Yun Chen
- Departments of Medicine and Cell Biology, Wilf Family Cardiovascular Research Institute, Diabetes Research Center, Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, New York
| | - Mitsuaki Isobe
- Department of Cardiovascular Medicine, Tokyo Medical and Dental University, Tokyo, Japan; and
| | - Åsa B Gustafsson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California
| | - Richard N Kitsis
- Departments of Medicine and Cell Biology, Wilf Family Cardiovascular Research Institute, Diabetes Research Center, Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, New York
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, Newark, New Jersey;
| |
Collapse
|
233
|
Chen F, Sun ZW, Ye LF, Fu GS, Mou Y, Hu SJ. Lycopene protects against apoptosis in hypoxia/reoxygenation‑induced H9C2 myocardioblast cells through increased autophagy. Mol Med Rep 2014; 11:1358-65. [PMID: 25351505 DOI: 10.3892/mmr.2014.2771] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 09/24/2014] [Indexed: 11/06/2022] Open
Abstract
Lycopene (Ly), the most common type of antioxidant in the majority of diet types, provides tolerance to ischemia/reperfusion injury. However, the underlying mechanism of the protective effects observed following Ly administration remains poorly investigated. The aim of the current study was to investigate whether Ly prevents damage to hypoxia/reoxygenation (HR)‑induced H9C2 myocardioblasts in an autophagy‑dependent manner. The levels of autophagic markers were detected using western blotting, the level of apoptosis was detected using western blotting and flow cytometry. The activation of autophagy was impaired via knockdown of the expression of 'microtubule‑associated protein 1‑light chain 3β (MAP1LC3B)' and 'Beclin 1'. After 16 h hypoxia, followed by 2 h reoxygenation, the expression levels of the microtubule‑associated protein 1A/1B‑light chain 3 (LC3) and Βeclin 1 autophagic biomarkers, and cell viability were reduced, whereas the percentage of apoptotic cells, and the expression levels of the Bax/B‑cell lymphoma 2 (Bcl‑2) and active caspase‑3 apoptotic biomarkers were increased. Pre‑incubation of the cells with different Ly concentrations reversed the HR‑induced inhibition of autophagy and cell viability, and the HR‑induced elevation in apoptotic levels. The induction of autophagy was accompanied by reduced apoptosis, and decreased expression levels of Bax/Bcl‑2 and active caspase‑3. In addition, the impairment of autophagy by silencing the expression of MAP1LC3B and Beclin 1 accelerated HR‑induced H9C2 cell apoptosis and the Ly‑mediated protective effects disappeared. Furthermore, Bax/Bcl‑2 and active caspase‑3 expression levels were increased. Moreover, Ly‑induced autophagy was associated with increased adenosine monophosphate kinase (AMPK) phosphorylation. Suppressed AMPK phosphorylation using compound C terminates Ly‑mediated cytoprotective effects. Ly treatment improves cell viability and reduces apoptosis as a result of the activation of the adaptive autophagic response on HR‑induced H9C2 myocardioblasts. AMPK phosphorylation may be involved in the progression.
Collapse
Affiliation(s)
- Fei Chen
- Institution of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Ze-Wei Sun
- Institution of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Li-Fang Ye
- Institution of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Guo-Sheng Fu
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310012, P.R. China
| | - Yun Mou
- Department of Ultrasound, The Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Shen-Jiang Hu
- Institution of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| |
Collapse
|
234
|
DRAM1 protects neuroblastoma cells from oxygen-glucose deprivation/reperfusion-induced injury via autophagy. Int J Mol Sci 2014; 15:19253-64. [PMID: 25342320 PMCID: PMC4227272 DOI: 10.3390/ijms151019253] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 09/18/2014] [Accepted: 10/09/2014] [Indexed: 01/01/2023] Open
Abstract
DNA damage-regulated autophagy modulator protein 1 (DRAM1), a multi-pass membrane lysosomal protein, is reportedly a tumor protein p53 (TP53) target gene involved in autophagy. During cerebral ischemia/reperfusion (I/R) injury, DRAM1 protein expression is increased, and autophagy is activated. However, the functional significance of DRAM1 and the relationship between DRAM1 and autophagy in brain I/R remains uncertain. The aim of this study is to investigate whether DRAM1 mediates autophagy activation in cerebral I/R injury and to explore its possible effects and mechanisms. We adopt the oxygen-glucose deprivation and reperfusion (OGD/R) Neuro-2a cell model to mimic cerebral I/R conditions in vitro, and RNA interference is used to knock down DRAM1 expression in this model. Cell viability assay is performed using the LIVE/DEAD viability/cytotoxicity kit. Cell phenotypic changes are analyzed through Western blot assays. Autophagy flux is monitored through the tandem red fluorescent protein-Green fluorescent protein-microtubule associated protein 1 light chain 3 (RFP-GFP-LC3) construct. The expression levels of DRAM1 and microtubule associated protein 1 light chain 3II/I (LC3II/I) are strongly up-regulated in Neuro-2a cells after OGD/R treatment and peaked at the 12 h reperfusion time point. The autophagy-specific inhibitor 3-Methyladenine (3-MA) inhibits the expression of DRAM1 and LC3II/I and exacerbates OGD/R-induced cell injury. Furthermore, DRAM1 knockdown aggravates OGD/R-induced cell injury and significantly blocks autophagy through decreasing autophagosome-lysosome fusion. In conclusion, our data demonstrate that DRAM1 knockdown in Neuro-2a cells inhibits autophagy by blocking autophagosome-lysosome fusion and exacerbated OGD/R-induced cell injury. Thus, DRAM1 might constitute a new therapeutic target for I/R diseases.
Collapse
|
235
|
Ikeda Y, Shirakabe A, Maejima Y, Zhai P, Sciarretta S, Toli J, Nomura M, Mihara K, Egashira K, Ohishi M, Abdellatif M, Sadoshima J. Endogenous Drp1 mediates mitochondrial autophagy and protects the heart against energy stress. Circ Res 2014; 116:264-78. [PMID: 25332205 DOI: 10.1161/circresaha.116.303356] [Citation(s) in RCA: 451] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
RATIONALE Both fusion and fission contribute to mitochondrial quality control. How unopposed fusion affects survival of cardiomyocytes and left ventricular function in the heart is poorly understood. OBJECTIVE We investigated the role of dynamin-related protein 1 (Drp1), a GTPase that mediates mitochondrial fission, in mediating mitochondrial autophagy, ventricular function, and stress resistance in the heart. METHODS AND RESULTS Drp1 downregulation induced mitochondrial elongation, accumulation of damaged mitochondria, and increased apoptosis in cardiomyocytes at baseline. Drp1 downregulation also suppressed autophagosome formation and autophagic flux at baseline and in response to glucose deprivation in cardiomyocytes. The lack of lysosomal translocation of mitochondrially targeted Keima indicates that Drp1 downregulation suppressed mitochondrial autophagy. Mitochondrial elongation and accumulation of damaged mitochondria were also observed in tamoxifen-inducible cardiac-specific Drp1 knockout mice. After Drp1 downregulation, cardiac-specific Drp1 knockout mice developed left ventricular dysfunction, preceded by mitochondrial dysfunction, and died within 13 weeks. Autophagic flux is significantly suppressed in cardiac-specific Drp1 knockout mice. Although left ventricular function in cardiac-specific Drp1 heterozygous knockout mice was normal at 12 weeks of age, left ventricular function decreased more severely after 48 hours of fasting, and the infarct size/area at risk after ischemia/reperfusion was significantly greater in cardiac-specific Drp1 heterozygous knockout than in control mice. CONCLUSIONS Disruption of Drp1 induces mitochondrial elongation, inhibits mitochondrial autophagy, and causes mitochondrial dysfunction, thereby promoting cardiac dysfunction and increased susceptibility to ischemia/reperfusion.
Collapse
Affiliation(s)
- Yoshiyuki Ikeda
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (Y.I., A.S., Y.M., P.Z., S.S., J.T., M.A., J.S.); IRCCS Neuromed, Pozzilli, Italy (S.S.); Department of Medicine and Bioregulatory Science (M.N.), Department of Molecular Biology (K.M.), Department of Cardiovascular Medicine, Department of Cardiovascular Research, Development, and Translational Medicine (K.E.), Graduate School of Medical Science, Kyushu University Hospital, Fukuoka, Japan; and Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Kagoshima, Japan (M.O.)
| | - Akihiro Shirakabe
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (Y.I., A.S., Y.M., P.Z., S.S., J.T., M.A., J.S.); IRCCS Neuromed, Pozzilli, Italy (S.S.); Department of Medicine and Bioregulatory Science (M.N.), Department of Molecular Biology (K.M.), Department of Cardiovascular Medicine, Department of Cardiovascular Research, Development, and Translational Medicine (K.E.), Graduate School of Medical Science, Kyushu University Hospital, Fukuoka, Japan; and Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Kagoshima, Japan (M.O.)
| | - Yasuhiro Maejima
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (Y.I., A.S., Y.M., P.Z., S.S., J.T., M.A., J.S.); IRCCS Neuromed, Pozzilli, Italy (S.S.); Department of Medicine and Bioregulatory Science (M.N.), Department of Molecular Biology (K.M.), Department of Cardiovascular Medicine, Department of Cardiovascular Research, Development, and Translational Medicine (K.E.), Graduate School of Medical Science, Kyushu University Hospital, Fukuoka, Japan; and Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Kagoshima, Japan (M.O.)
| | - Peiyong Zhai
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (Y.I., A.S., Y.M., P.Z., S.S., J.T., M.A., J.S.); IRCCS Neuromed, Pozzilli, Italy (S.S.); Department of Medicine and Bioregulatory Science (M.N.), Department of Molecular Biology (K.M.), Department of Cardiovascular Medicine, Department of Cardiovascular Research, Development, and Translational Medicine (K.E.), Graduate School of Medical Science, Kyushu University Hospital, Fukuoka, Japan; and Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Kagoshima, Japan (M.O.)
| | - Sebastiano Sciarretta
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (Y.I., A.S., Y.M., P.Z., S.S., J.T., M.A., J.S.); IRCCS Neuromed, Pozzilli, Italy (S.S.); Department of Medicine and Bioregulatory Science (M.N.), Department of Molecular Biology (K.M.), Department of Cardiovascular Medicine, Department of Cardiovascular Research, Development, and Translational Medicine (K.E.), Graduate School of Medical Science, Kyushu University Hospital, Fukuoka, Japan; and Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Kagoshima, Japan (M.O.)
| | - Jessica Toli
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (Y.I., A.S., Y.M., P.Z., S.S., J.T., M.A., J.S.); IRCCS Neuromed, Pozzilli, Italy (S.S.); Department of Medicine and Bioregulatory Science (M.N.), Department of Molecular Biology (K.M.), Department of Cardiovascular Medicine, Department of Cardiovascular Research, Development, and Translational Medicine (K.E.), Graduate School of Medical Science, Kyushu University Hospital, Fukuoka, Japan; and Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Kagoshima, Japan (M.O.)
| | - Masatoshi Nomura
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (Y.I., A.S., Y.M., P.Z., S.S., J.T., M.A., J.S.); IRCCS Neuromed, Pozzilli, Italy (S.S.); Department of Medicine and Bioregulatory Science (M.N.), Department of Molecular Biology (K.M.), Department of Cardiovascular Medicine, Department of Cardiovascular Research, Development, and Translational Medicine (K.E.), Graduate School of Medical Science, Kyushu University Hospital, Fukuoka, Japan; and Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Kagoshima, Japan (M.O.)
| | - Katsuyoshi Mihara
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (Y.I., A.S., Y.M., P.Z., S.S., J.T., M.A., J.S.); IRCCS Neuromed, Pozzilli, Italy (S.S.); Department of Medicine and Bioregulatory Science (M.N.), Department of Molecular Biology (K.M.), Department of Cardiovascular Medicine, Department of Cardiovascular Research, Development, and Translational Medicine (K.E.), Graduate School of Medical Science, Kyushu University Hospital, Fukuoka, Japan; and Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Kagoshima, Japan (M.O.)
| | - Kensuke Egashira
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (Y.I., A.S., Y.M., P.Z., S.S., J.T., M.A., J.S.); IRCCS Neuromed, Pozzilli, Italy (S.S.); Department of Medicine and Bioregulatory Science (M.N.), Department of Molecular Biology (K.M.), Department of Cardiovascular Medicine, Department of Cardiovascular Research, Development, and Translational Medicine (K.E.), Graduate School of Medical Science, Kyushu University Hospital, Fukuoka, Japan; and Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Kagoshima, Japan (M.O.)
| | - Mitsuru Ohishi
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (Y.I., A.S., Y.M., P.Z., S.S., J.T., M.A., J.S.); IRCCS Neuromed, Pozzilli, Italy (S.S.); Department of Medicine and Bioregulatory Science (M.N.), Department of Molecular Biology (K.M.), Department of Cardiovascular Medicine, Department of Cardiovascular Research, Development, and Translational Medicine (K.E.), Graduate School of Medical Science, Kyushu University Hospital, Fukuoka, Japan; and Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Kagoshima, Japan (M.O.)
| | - Maha Abdellatif
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (Y.I., A.S., Y.M., P.Z., S.S., J.T., M.A., J.S.); IRCCS Neuromed, Pozzilli, Italy (S.S.); Department of Medicine and Bioregulatory Science (M.N.), Department of Molecular Biology (K.M.), Department of Cardiovascular Medicine, Department of Cardiovascular Research, Development, and Translational Medicine (K.E.), Graduate School of Medical Science, Kyushu University Hospital, Fukuoka, Japan; and Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Kagoshima, Japan (M.O.)
| | - Junichi Sadoshima
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (Y.I., A.S., Y.M., P.Z., S.S., J.T., M.A., J.S.); IRCCS Neuromed, Pozzilli, Italy (S.S.); Department of Medicine and Bioregulatory Science (M.N.), Department of Molecular Biology (K.M.), Department of Cardiovascular Medicine, Department of Cardiovascular Research, Development, and Translational Medicine (K.E.), Graduate School of Medical Science, Kyushu University Hospital, Fukuoka, Japan; and Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Kagoshima, Japan (M.O.).
| |
Collapse
|
236
|
Oxidative stress in kidney transplantation: malondialdehyde is an early predictive marker of graft dysfunction. Transplantation 2014; 97:1058-65. [PMID: 24406454 DOI: 10.1097/01.tp.0000438626.91095.50] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Oxidative stress is one of the most important components of the ischemia-reperfusion process after kidney transplantation (KTx) and increases with graft dysfunction. METHODS This prospective study was conducted on 40 consecutive KTx recipients to evaluate time-dependent changes in oxidative stress-related parameters within the first week after KTx and to assess their performance in predicting delayed graft function (DGF=dialysis requirement during initial posttransplant week) and graft function at 1 year. Blood samples were collected before (day 0) and after KTx (days 1, 2, 4, and 7). Total antioxidant capacity, plasma levels of malondialdehyde (MDA), and activities of glutathione peroxidase, glutathione reductase and superoxide dismutase were measured. Multivariable linear mixed and linear regression models, receiver-operating characteristic (ROC), and areas under ROC curves (AUC-ROC) were used. RESULTS At all time points after KTx, mean MDA levels were significantly higher in patients developing DGF (n=18). Shortly after KTx (8-12 hr), MDA values were higher in DGF recipients (on average, +0.16 μmol/L) and increased further on following day, contrasting with prompt functioning recipients. Day 1 MDA levels accurately predicted DGF (AUC-ROC=0.90), with a performance higher than SCr (AUC-ROC=0.73) and similar to cystatin C (AUC-ROC=0.91). Multivariable analysis revealed that MDA levels on day 7 represented an independent predictor of 1-year graft function. Antioxidant enzyme activities were not significantly changed during the study period and were not predictors of 1-year graft function. CONCLUSIONS Increased MDA levels on day 1 after KTx might be an early prognostic indicator of DGF, and levels on day 7 might represent a useful predictor of 1-year graft function.
Collapse
|
237
|
Zeng M, Wei X, Wu Z, Li W, Li B, Fei Y, He Y, Chen J, Wang P, Liu X. Reactive oxygen species contribute to simulated ischemia/reperfusion-induced autophagic cell death in human umbilical vein endothelial cells. Med Sci Monit 2014; 20:1017-23. [PMID: 24943908 PMCID: PMC4074109 DOI: 10.12659/msm.890897] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Autophagy is important for cells to degrade protein aggregates and organelles. Our preliminary study suggests that ischemia/reperfusion in rabbit hearts promoted autophagic myocardial injury, resulting in no-reflow phenomenon. In this study, we sought to further understand the mechanism and outcome of the upregulation of autophagy in ischemia/reperfusion. Material/Methods We employed a simulated ischemia/reperfusion (sI/R) model in human umbilical vein endothelial cells (HUVECs) in vitro, in the presence or absence of antioxidants. Results Our study confirms that sI/R induces autophagy in HUVECs as measured by increased expression of Beclin 1 and microtubule-associated protein 1 light chain 3 (LC3), electron microscopic analysis, and special biofluorescent staining with monodansylcadaverine. This sI/R-induced autophagy was also accompanied by increased levels of p65 protein expression and cell death. In addition, we detected the accumulation of reactive oxygen species (ROS) after sI/R. Moreover, with the application of ROS scavengers that block the release of ROS, we were able to demonstrate that inhibition of autophagy increases cell survival. Conclusions The study suggests that ROS accumulation is involved in the sI/R-induced autophagic cell death in HUVECs.
Collapse
Affiliation(s)
- Min Zeng
- Department of Cardiology, People's Hospital of Hainan Province, Haikou, China (mainland)
| | - Xin Wei
- Department of Cardiology, People's Hospital of Hainan Province, Haikou, China (mainland)
| | - Zhiyong Wu
- Department of Cardiology, People's Hospital of Hainan Province, Haikou, China (mainland)
| | - Wei Li
- Department of Cardiology, People's Hospital of Hainan Province, Haikou, China (mainland)
| | - Bing Li
- Department of Cardiology, People's Hospital of Hainan Province, Haikou, China (mainland)
| | - Yi Fei
- Department of Cardiology, People's Hospital of Hainan Province, Haikou, China (mainland)
| | - Yangli He
- Department of Cardiology, People's Hospital of Hainan Province, Haikou, China (mainland)
| | - Jixiong Chen
- Department of Cardiology, People's Hospital of Hainan Province, Haikou, China (mainland)
| | - Ping Wang
- Department of Cardiology, People's Hospital of Hainan Province, Haikou, China (mainland)
| | - Xiaojun Liu
- Department of Cardiology, People's Hospital of Hainan Province, Haikou, China (mainland)
| |
Collapse
|
238
|
Nicotinamide mononucleotide, an intermediate of NAD+ synthesis, protects the heart from ischemia and reperfusion. PLoS One 2014; 9:e98972. [PMID: 24905194 PMCID: PMC4048236 DOI: 10.1371/journal.pone.0098972] [Citation(s) in RCA: 221] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2013] [Accepted: 05/08/2014] [Indexed: 12/17/2022] Open
Abstract
Nicotinamide phosphoribosyltransferase (Nampt), the rate-limiting enzyme for nicotinamide adenine dinucleotide (NAD+) synthesis, and Sirt1, an NAD+-dependent histone deacetylase, protect the heart against ischemia/reperfusion (I/R). It remains unknown whether Nampt mediates the protective effect of ischemic preconditioning (IPC), whether nicotinamide mononucleotide (NMN, 500 mg/kg), a product of Nampt in the NAD+ salvage pathway, mimics the effect of IPC, or whether caloric restriction (CR) upregulates Nampt and protects the heart through a Sirt1-dependent mechanism. IPC upregulated Nampt protein, and the protective effect of IPC against ischemia (30 minutes) and reperfusion (24 hours) was attenuated at both early and late phases in Nampt +/− mice, suggesting that Nampt plays an essential role in mediating the protective effect of IPC. In order to mimic the effect of Nampt, NMN was administered by intraperitoneal injection. NMN significantly increased the level of NAD+ in the heart at baseline and prevented a decrease in NAD+ during ischemia. NMN protected the heart from I/R injury when it was applied once 30 minutes before ischemia or 4 times just before and during reperfusion, suggesting that exogenous NMN protects the heart from I/R injury in both ischemic and reperfusion phases. The protective effect of NMN was accompanied by decreases in acetylation of FoxO1, but it was not obvious in Sirt1 KO mice, suggesting that the effect of NMN is mediated through activation of Sirt1. Compared to control diet (90% calories), CR (60% calories for 6 weeks) in mice led to a significant reduction in I/R injury, accompanied by upregulation of Nampt. The protective effect of CR against I/R injury was not significant in cardiac-specific Sirt1 KO mice, suggesting that the protective effect of CR is in part mediated through the Nampt-Sirt1 pathway. In conclusion, exogenous application of NMN and CR protects the heart by both mimicking IPC and activating Sirt1.
Collapse
|
239
|
Zhang YL, Yao YT, Fang NX, Zhou CH, Gong JS, Li LH. Restoration of autophagic flux in myocardial tissues is required for cardioprotection of sevoflurane postconditioning in rats. Acta Pharmacol Sin 2014; 35:758-69. [PMID: 24793309 DOI: 10.1038/aps.2014.20] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 03/04/2014] [Indexed: 01/03/2023] Open
Abstract
AIM Sevoflurane postconditioning (SpostC) has been shown to protect the heart from ischemia-reperfusion (I/R) injury. In this study, we examined whether SpostC affected autophagic flux in myocardial tissues that contributed to its cardioprotective effects in rats following acute I/R injury. METHODS SD rats underwent 30 min of left anterior descending coronary artery ligation followed by 120 min of reperfusion. The rats were subjected to inhalation of 2.4% (v/v) sevoflurane during the first 5 min of reperfusion, and chloroquine (10 mg/kg, ip) was injected 1 h before I/R. Myocardial infarct size was estimated using TTC staining. Autophagosomes in myocardial tissues were detected under TEM. Expression of LC3B-II, beclin-1, p62/SQSTM1, cathepsin B, caspase-3 and cleaved PARP was assessed using Western blot analysis. Plasma cardiac troponin I was measured using ELISA. Cardiomyocyte apoptosis was evaluated with TUNEL staining. RESULTS I/R procedure produced severe myocardium infarct and apoptosis accompanied by markedly increased number of autophagosomes, as well as increased levels of LC3B-II, beclin-1 and p62 in myocardial tissues. SpostC significantly reduced infarct size, attenuated myocardial apoptosis, restored intact autophagic flux and improved the lysosomal function in myocardial tissues. Administration of chloroquine that blocked autophagic flux abrogated the cardioprotective effects of SpostC. CONCLUSION SpostC exerts its cardioprotective effects in rats following I/R injury via restoring autophagic flux in myocardial tissues.
Collapse
|
240
|
Ma S, Wang Y, Chen Y, Cao F. The role of the autophagy in myocardial ischemia/reperfusion injury. Biochim Biophys Acta Mol Basis Dis 2014; 1852:271-6. [PMID: 24859226 DOI: 10.1016/j.bbadis.2014.05.010] [Citation(s) in RCA: 232] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 04/29/2014] [Accepted: 05/12/2014] [Indexed: 02/08/2023]
Abstract
Autophagy is an intracellular process responsible for damaged or unnecessary protein and organelle degradation. In the heart, autophagy occurs at basal level and dysregulated autophagy is associated with a variety of cardiovascular diseases. Autophagy is enhanced in ischemia as well as in the reperfusion phase during cardiac ischemia reperfusion (I/R) injury. More importantly, recent studies revealed that autophagy exerted both beneficial and detrimental effects in pathology of cardiac ischemia reperfusion. This paper is to review the functional significance of autophagy in cardiac ischemia reperfusion injury and discuss underlying signaling pathways. This article is part of a Special Issue entitled: Autophagy and protein quality control in cardiometabolic diseases.
Collapse
Affiliation(s)
- Sai Ma
- Department of Cardiology, Chinese PLA General Hospital, 28# Fuxing Street, Beijing 100852, China; Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127# Changle West Road, Xi'an, Shaanxi 710032, China
| | - Yabin Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127# Changle West Road, Xi'an, Shaanxi 710032, China
| | - Yundai Chen
- Department of Cardiology, Chinese PLA General Hospital, 28# Fuxing Street, Beijing 100852, China
| | - Feng Cao
- Department of Cardiology, Chinese PLA General Hospital, 28# Fuxing Street, Beijing 100852, China; Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127# Changle West Road, Xi'an, Shaanxi 710032, China.
| |
Collapse
|
241
|
Mei Y, Thompson MD, Cohen RA, Tong X. Autophagy and oxidative stress in cardiovascular diseases. Biochim Biophys Acta Mol Basis Dis 2014; 1852:243-51. [PMID: 24834848 DOI: 10.1016/j.bbadis.2014.05.005] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 04/29/2014] [Accepted: 05/03/2014] [Indexed: 10/25/2022]
Abstract
Autophagy is a highly conserved degradation process by which intracellular components, including soluble macromolecules (e.g. nucleic acids, proteins, carbohydrates, and lipids) and dysfunctional organelles (e.g. mitochondria, ribosomes, peroxisomes, and endoplasmic reticulum) are degraded by the lysosome. Autophagy is orchestrated by the autophagy related protein (Atg) composed protein complexes to form autophagosomes, which fuse with lysosomes to generate autolysosomes where the contents are degraded to provide energy for cell survival in response to environmental and cellular stress. Autophagy is an important player in cardiovascular disease development such as atherosclerosis, cardiac ischemia/reperfusion, cardiomyopathy, heart failure and hypertension. Autophagy in particular contributes to cardiac ischemia, hypertension and diabetes by interaction with reactive oxygen species generated in endoplasmic reticulum and mitochondria. This review highlights the dual role of autophagy in cardiovascular disease development. Full recognition of autophagy as an adaptive or maladaptive response would provide potential new strategies for cardiovascular disease prevention and management. This article is part of a Special Issue entitled: Autophagy and protein quality control in cardiometabolic diseases.
Collapse
Affiliation(s)
- Yu Mei
- Vascular Biology Section, Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| | - Melissa D Thompson
- Vascular Biology Section, Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| | - Richard A Cohen
- Vascular Biology Section, Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| | - XiaoYong Tong
- Vascular Biology Section, Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA.
| |
Collapse
|
242
|
Morales CR, Pedrozo Z, Lavandero S, Hill JA. Oxidative stress and autophagy in cardiovascular homeostasis. Antioxid Redox Signal 2014; 20:507-18. [PMID: 23641894 PMCID: PMC3894700 DOI: 10.1089/ars.2013.5359] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
SIGNIFICANCE Autophagy is an evolutionarily ancient process of intracellular protein and organelle recycling required to maintain cellular homeostasis in the face of a wide variety of stresses. Dysregulation of reactive oxygen species (ROS) and reactive nitrogen species (RNS) leads to oxidative damage. Both autophagy and ROS/RNS serve pathological or adaptive roles within cardiomyocytes, depending on the context. RECENT ADVANCES ROS/RNS and autophagy communicate with each other via both transcriptional and post-translational events. This cross talk, in turn, regulates the structural integrity of cardiomyocytes, promotes proteostasis, and reduces inflammation, events critical to disease pathogenesis. CRITICAL ISSUES Dysregulation of either autophagy or redox state has been implicated in many cardiovascular diseases. Cardiomyocytes are rich in mitochondria, which make them particularly sensitive to oxidative damage. Maintenance of mitochondrial homeostasis and elimination of defective mitochondria are each critical to the maintenance of redox homeostasis. FUTURE DIRECTIONS The complex interplay between autophagy and oxidative stress underlies a wide range of physiological and pathological events and its elucidation holds promise of potential clinical applicability.
Collapse
Affiliation(s)
- Cyndi R Morales
- 1 Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center , Dallas, Texas
| | | | | | | |
Collapse
|
243
|
Yang Y, Duan W, Li Y, Jin Z, Yan J, Yu S, Yi D. Novel role of silent information regulator 1 in myocardial ischemia. Circulation 2014; 128:2232-40. [PMID: 24218438 DOI: 10.1161/circulationaha.113.002480] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Yang Yang
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China (Y.Y., W.D., Z.J., S.Y., D.Y.); Team 10, School of Stomatology, The Fourth Military Medical University, Xi'an, China (Y.L.); and Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, China (J.Y.)
| | | | | | | | | | | | | |
Collapse
|
244
|
Zhang Y, Ren J. Targeting autophagy for the therapeutic application of histone deacetylase inhibitors in ischemia/reperfusion heart injury. Circulation 2014; 129:1088-91. [PMID: 24396040 DOI: 10.1161/circulationaha.113.008115] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Yingmei Zhang
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie (Y.Z., J.R.); and Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China (Y.Z.)
| | | |
Collapse
|
245
|
Hardas SS, Sultana R, Warrier G, Dan M, Wu P, Grulke EA, Tseng MT, Unrine JM, Graham UM, Yokel RA, Butterfield DA. Rat hippocampal responses up to 90 days after a single nanoceria dose extends a hierarchical oxidative stress model for nanoparticle toxicity. Nanotoxicology 2013; 8 Suppl 1:155-66. [DOI: 10.3109/17435390.2013.868059] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
| | | | | | - Mo Dan
- Department of Pharmaceutical Sciences and
| | - Peng Wu
- Chemical and Materials Engineering Department, University of Kentucky, Lexington, KY, USA,
| | - Eric A. Grulke
- Chemical and Materials Engineering Department, University of Kentucky, Lexington, KY, USA,
| | - Michael T. Tseng
- Department of Anatomical Sciences & Neurobiology, University of Louisville, Louisville, KY, USA,
| | - Jason M. Unrine
- Department of Plant and Soil Sciences, University of Kentucky, Lexington, KY, USA,
| | - Uschi M. Graham
- Center for Applied Energy Research, University of Kentucky, Lexington, KY, USA,
| | - Robert A. Yokel
- Department of Pharmaceutical Sciences and
- Graduate Center for Toxicology, University of Kentucky Academic Medical Center, Lexington, KY, USA, and
| | - D. Allan Butterfield
- Department of Chemistry,
- Center of Membrane Sciences, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
246
|
Shiomi M, Miyamae M, Takemura G, Kaneda K, Inamura Y, Onishi A, Koshinuma S, Momota Y, Minami T, Figueredo VM. Sevoflurane induces cardioprotection through reactive oxygen species-mediated upregulation of autophagy in isolated guinea pig hearts. J Anesth 2013; 28:593-600. [PMID: 24337890 DOI: 10.1007/s00540-013-1755-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 11/11/2013] [Indexed: 12/23/2022]
Abstract
PURPOSE Sevoflurane increases reactive oxygen species (ROS), which mediate cardioprotection against myocardial ischemia-reperfusion injury. Emerging evidence suggests that autophagy is involved in cardioprotection. We examined whether reactive oxygen species mediate sevoflurane preconditioning through autophagy. METHODS Isolated guinea pigs hearts were subjected to 30 min ischemia followed by 120 min reperfusion (control). Anesthetic preconditioning was elicited with 2 % sevoflurane for 10 min before ischemia (SEVO). The ROS-scavenger, N-(2-mercaptopropionyl) glycine (MPG, 1 mmol/l), was administered starting 30 min before ischemia to sevoflurane-treated (SEVO + MPG) or non-sevoflurane-treated (MPG) hearts. Infarct size was determined by triphenyltetrazolium chloride stain. Tissue samples were obtained after reperfusion to determine autophagy-related protein (microtubule-associated protein light chain I and II: LC3-I, -II) and 5' AMP-activated protein kinase (AMPK) expression using Western blot analysis. Electron microscopy was used to detect autophagosomes. RESULTS Infarct size was significantly reduced and there were more abundant autophagosomes in SEVO compared with control. Western blot analysis revealed that the ratio of LC3-II/I and phosphorylation of AMPK were significantly increased in SEVO. These effects were abolished by MPG. CONCLUSIONS Sevoflurane induces cardioprotection through ROS-mediated upregulation of autophagy.
Collapse
Affiliation(s)
- Mayumi Shiomi
- Department of Anesthesiology, Osaka Medical College, Takatsuki, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
247
|
Rodrigo R, Libuy M, Feliú F, Hasson D. Oxidative stress-related biomarkers in essential hypertension and ischemia-reperfusion myocardial damage. DISEASE MARKERS 2013; 35:773-90. [PMID: 24347798 PMCID: PMC3856219 DOI: 10.1155/2013/974358] [Citation(s) in RCA: 142] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 10/07/2013] [Indexed: 12/22/2022]
Abstract
Cardiovascular diseases are a leading cause of mortality and morbidity worldwide, with hypertension being a major risk factor. Numerous studies support the contribution of reactive oxygen and nitrogen species in the pathogenesis of hypertension, as well as other pathologies associated with ischemia/reperfusion. However, the validation of oxidative stress-related biomarkers in these settings is still lacking and novel association of these biomarkers and other biomarkers such as endothelial progenitor cells, endothelial microparticles, and ischemia modified albumin, is just emerging. Oxidative stress has been suggested as a pathogenic factor and therapeutic target in early stages of essential hypertension. Systolic and diastolic blood pressure correlated positively with plasma F2-isoprostane levels and negatively with total antioxidant capacity of plasma in hypertensive and normotensive patients. Cardiac surgery with extracorporeal circulation causes an ischemia/reperfusion event associated with increased lipid peroxidation and protein carbonylation, two biomarkers associated with oxidative damage of cardiac tissue. An enhancement of the antioxidant defense system should contribute to ameliorating functional and structural abnormalities derived from this metabolic impairment. However, data have to be validated with the analysis of the appropriate oxidative stress and/or nitrosative stress biomarkers.
Collapse
Affiliation(s)
- Ramón Rodrigo
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Independencia 1027, Casilla 70058, Santiago 7, Chile
| | - Matías Libuy
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Independencia 1027, Casilla 70058, Santiago 7, Chile
| | - Felipe Feliú
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Independencia 1027, Casilla 70058, Santiago 7, Chile
| | - Daniel Hasson
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Independencia 1027, Casilla 70058, Santiago 7, Chile
| |
Collapse
|
248
|
Wang X, Robbins J. Proteasomal and lysosomal protein degradation and heart disease. J Mol Cell Cardiol 2013; 71:16-24. [PMID: 24239609 DOI: 10.1016/j.yjmcc.2013.11.006] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 11/04/2013] [Accepted: 11/06/2013] [Indexed: 01/08/2023]
Abstract
In the cell, the proteasome and lysosomes represent the most important proteolytic machineries, responsible for the protein degradation in the ubiquitin-proteasome system (UPS) and autophagy, respectively. Both the UPS and autophagy are essential to protein quality and quantity control. Alterations in cardiac proteasomal and lysosomal degradation are remarkably associated with most heart disease in humans and are implicated in the pathogenesis of congestive heart failure. Studies carried out in animal models and in cell culture have begun to establish both sufficiency and, in some cases, the necessity of proteasomal functional insufficiency or lysosomal insufficiency as a major pathogenic factor in the heart. This review article highlights some recent advances in the research into proteasome and lysosome protein degradation in relation to cardiac pathology and examines the emerging evidence for enhancing degradative capacities of the proteasome and/or lysosome as a new therapeutic strategy for heart disease. This article is part of a Special Issue entitled "Protein Quality Control, the Ubiquitin Proteasome System, and Autophagy".
Collapse
Affiliation(s)
- Xuejun Wang
- Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion, SD 57069, USA.
| | - Jeffrey Robbins
- Division of Molecular Cardiovascular Biology, The Heart Institute, Department of Pediatrics, The Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| |
Collapse
|
249
|
Bhuiyan MS, Pattison JS, Osinska H, James J, Gulick J, McLendon PM, Hill JA, Sadoshima J, Robbins J. Enhanced autophagy ameliorates cardiac proteinopathy. J Clin Invest 2013; 123:5284-97. [PMID: 24177425 DOI: 10.1172/jci70877] [Citation(s) in RCA: 241] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 08/22/2013] [Indexed: 01/06/2023] Open
Abstract
Basal autophagy is a crucial mechanism in cellular homeostasis, underlying both normal cellular recycling and the clearance of damaged or misfolded proteins, organelles and aggregates. We showed here that enhanced levels of autophagy induced by either autophagic gene overexpression or voluntary exercise ameliorated desmin-related cardiomyopathy (DRC). To increase levels of basal autophagy, we generated an inducible Tg mouse expressing autophagy-related 7 (Atg7), a critical and rate-limiting autophagy protein. Hearts from these mice had enhanced autophagy, but normal morphology and function. We crossed these mice with CryABR120G mice, a model of DRC in which autophagy is significantly attenuated in the heart, to test the functional significance of autophagy activation in a proteotoxic model of heart failure. Sustained Atg7-induced autophagy in the CryABR120G hearts decreased interstitial fibrosis, ameliorated ventricular dysfunction, decreased cardiac hypertrophy, reduced intracellular aggregates and prolonged survival. To determine whether different methods of autophagy upregulation have additive or even synergistic benefits, we subjected the autophagy-deficient CryABR120G mice and the Atg7-crossed CryABR120G mice to voluntary exercise, which also upregulates autophagy. The entire exercised Atg7-crossed CryABR120G cohort survived to 7 months. These findings suggest that activating autophagy may be a viable therapeutic strategy for improving cardiac performance under proteotoxic conditions.
Collapse
|
250
|
Shafique E, Choy WC, Liu Y, Feng J, Cordeiro B, Lyra A, Arafah M, Yassin-Kassab A, Zanetti AVD, Clements RT, Bianchi C, Benjamin LE, Sellke FW, Abid MR. Oxidative stress improves coronary endothelial function through activation of the pro-survival kinase AMPK. Aging (Albany NY) 2013; 5:515-30. [PMID: 24018842 PMCID: PMC3765580 DOI: 10.18632/aging.100569] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Age-associated decline in cardiovascular function is believed to occur from the deleterious effects of reactive oxygen species (ROS). However, failure of recent clinical trials using antioxidants in patients with cardiovascular disease, and the recent findings showing paradoxical role for NADPH oxidase-derived ROS in endothelial function challenge this long-held notion against ROS. Here, we examine the effects of endothelium-specific conditional increase in ROS on coronary endothelial function. We have generated a novel binary (Tet-ON/OFF) conditional transgenic mouse (Tet-Nox2:VE-Cad-tTA) that induces endothelial cell (EC)-specific overexpression of Nox2/gp91 (NADPH oxidase) and 1.8±0.42-fold increase in EC-ROS upon tetracycline withdrawal (Tet-OFF). We examined ROS effects on EC signaling and function. First, we demonstrate that endothelium-dependent coronary vasodilation was significantly improved in Tet-OFF Nox2 compared to Tet-ON (control) littermates. Using EC isolated from mouse heart, we show that endogenous ROS increased eNOS activation and nitric oxide (NO) synthesis through activation of the survival kinase AMPK. Coronary vasodilation in Tet-OFF Nox2 animals was CaMKKβ-AMPK-dependent. Finally, we demonstrate that AMPK activation induced autophagy and thus, protected ECs from oxidant-induced cell death. Together, these findings suggest that increased ROS levels, often associated with cardiovascular conditions in advanced age, play a protective role in endothelial homeostasis by inducing AMPK-eNOS axis.
Collapse
Affiliation(s)
- Ehtesham Shafique
- Cardiovascular Research Center, Division of Cardiothoracic Surgery, Department of Surgery, Rhode Island Hospital, Providence, RI 02903, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|