1
|
Gong Y, Zhu W, Li Y, Lu T, Tan J, He C, Yang L, Zhu Y, Gong L. Dynamic regulation of proximal tubular autophagy from injury to repair after ischemic kidney damage. Cell Mol Biol Lett 2024; 29:151. [PMID: 39639205 PMCID: PMC11619129 DOI: 10.1186/s11658-024-00663-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 11/06/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND The role of proximal tubular autophagy in repairing kidney injury following ischemia remains unclear. METHODS In this study, we utilized mice with conditional deletion of the Atg5 gene in proximal tubules and monitored the long-term dynamic regulation of autophagy following ischemic acute kidney injury (AKI). RESULTS The results showed that Atg5-deficient proximal tubule epithelial cells exhibited damaged mitochondria, concentric membranes, and lysosomal accumulation 24 h after ischemia/reperfusion. However, 28 days after ischemia/reperfusion, concentric membrane bodies remained, but lysosomal accumulation was no longer observed. Notably, the absence of Atg5 in renal tubular epithelial cells impaired renal function and led to increased tubular cell proliferation and oxidative stress in the early stage of injury. However, during the repair period following AKI, Atg5 deficiency exhibited no significant difference in the expression of proliferating cell nuclear antigen (PCNA) and 4-hydoxynonenal (4HNE), suggesting that the improvement in renal fibrosis associated with Atg5 deficiency is unlikely to result from its effect on cell proliferation or reactive oxygen species levels. Additionally, Atg5 deficiency inhibits the secretion of profibrotic factor fibroblast growth factor 2 (FGF2) from the early stage of renal injury to the recovery stage of AKI, indicating that autophagy-specific regulation of FGF2 secretion is a dynamic process overlapping with other stages of injury. Furthermore, increased co-localization of ATG5 with 4HNE and FGF2 was observed in patient samples. CONCLUSION In summary, our results suggest that the dynamic regulation of autophagy on key molecules involved in kidney injury and repair varies with the stage of kidney injury.
Collapse
Affiliation(s)
- Yuhong Gong
- Experimental Animal Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Wei Zhu
- Department of Infectious Diseases, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yongqiang Li
- Department of General Practice, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, China
| | - Tao Lu
- Changzhou Geriatric Hospital Affiliated to Soochow University, Changzhou No. 7 People's Hospital, Changzhou, 213011, China
| | - Jiexing Tan
- Experimental Animal Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Changsheng He
- Experimental Animal Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Luodan Yang
- School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China.
| | - Yufeng Zhu
- Experimental Animal Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Li Gong
- Experimental Animal Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
2
|
Huang B, Nie G, Dai X, Cui T, Pu W, Zhang C. Environmentally relevant levels of Cd and Mo coexposure induces ferroptosis and excess ferritinophagy through AMPK/mTOR axis in duck myocardium. ENVIRONMENTAL TOXICOLOGY 2024; 39:4196-4206. [PMID: 38717027 DOI: 10.1002/tox.24302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/06/2024] [Accepted: 04/23/2024] [Indexed: 07/14/2024]
Abstract
Cadmium (Cd) and excess molybdenum (Mo) are multiorgan toxic, but the detrimental impacts of Cd and/or Mo on poultry have not been fully clarified. Thence, a 16-week sub-chronic toxic experiment was executed with ducks to assess the toxicity of Cd and/or Mo. Our data substantiated that Cd and Mo coexposure evidently reduced GSH-Px, GSH, T-SOD, and CAT activities and elevated H2O2 and MDA concentrations in myocardium. What is more, the study suggested that Cd and Mo united exposure synergistically elevated Fe2+ content in myocardium and activated AMPK/mTOR axis, then induced ferroptosis by obviously upregulating ACSL4, PTGS2, and TFRC expression levels and downregulating SLC7A11, GPX4, FPN1, FTL1, and FTH1 expression levels. Additionally, Cd and Mo coexposure further caused excessive ferritinophagy by observably increasing autophagosomes, the colocalization of endogenous FTH1 and LC3, ATG5, ATG7, LC3II/LC3I, NCOA4, and FTH1 expression levels. In brief, this study for the first time substantiated that Cd and Mo united exposure synergistically induced ferroptosis and excess ferritinophagy by AMPK/mTOR axis, finally augmenting myocardium injure in ducks, which will offer an additional view on united toxicity between two heavy metals on poultry.
Collapse
Affiliation(s)
- Bingyan Huang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Gaohui Nie
- Ministry of Public Education, Jiangxi Hongzhou Vocational College, Fengcheng, Jiangxi, China
| | - Xueyan Dai
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Ting Cui
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Wenjing Pu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Caiying Zhang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| |
Collapse
|
3
|
Jin C, Cao Y, Li Y. Bone Mesenchymal Stem Cells Origin Exosomes are Effective Against Sepsis-Induced Acute Kidney Injury in Rat Model. Int J Nanomedicine 2023; 18:7745-7758. [PMID: 38144514 PMCID: PMC10743757 DOI: 10.2147/ijn.s417627] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 12/13/2023] [Indexed: 12/26/2023] Open
Abstract
Introduction The incidence and mortality rates of sepsis-induced acute kidney injury (SAKI) remain high, posing a substantial healthcare burden. Studies have implicated a connection between the development of SAKI and inflammation response, apoptosis, and autophagy. Moreover, evidence suggests that manipulating autophagy could potentially influence the prognosis of this condition. Notably, exosomes derived from bone mesenchymal stem cells (BMSCs-Exo) have exhibited promise in mitigating cellular damage by modulating pathways associated with inflammation, apoptosis, and autophagy. Thus, this study aims to investigate the influence of BMSCs-Exo on SAKI and the potential mechanisms that drive this impact. Methods The SAKI model was induced in HK-2 cells using lipopolysaccharide (LPS), while rats underwent cecal ligation and puncture (CLP) to simulate the condition. Cell viability was assessed using the CCK-8 kit, and kidney damage was evaluated through HE staining, blood urea nitrogen (BUN), and serum creatinine (SCr) measurements. Inflammatory-related RNAs and proteins were quantified via qPCR and ELISA, respectively. Apoptosis was determined through apoptosis-related protein levels, flow cytometry, and TUNEL staining. Western blot analysis was utilized to measure associated protein expressions. Results In vivo, BMSCs-Exo ameliorated kidney injury in CLP-induced SAKI rats, reducing inflammatory cytokine production and apoptosis levels. Fluorescence microscope observed the absorption of BMSCs-Exo by renal cells following injection via tail vein. In the SAKI rat kidney tissue, there was an upregulation of LC3-II/LC3-I, p62, and phosphorylated AMP-activated protein kinase (p-AMPK) expressions, indicating blocked autophagic flux, while phosphorylated mammalian target of rapamycin (p-mTOR) expression was downregulated. However, BMSCs-Exo enhanced LC3-II/LC3-I and p-AMPK expression, concurrently reducing p62 and p-mTOR levels. In vitro, BMSCs-Exo enhanced cell viability in LPS-treated HK-2 cells, and exerted anti-inflammation and anti-apoptosis effects which were consistent with the results in vivo. Similarly, rapamycin (Rapa) exhibited a protective effect comparable to BMSCs-Exo, albeit partially abrogated by 3-methyladenine (3-MA). Conclusion BMSCs-Exo mitigate inflammation and apoptosis through autophagy in SAKI, offering a promising avenue for SAKI treatment.
Collapse
Affiliation(s)
- Cui Jin
- Department of Critical Care Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072, People’s Republic of China
| | - Yongmei Cao
- Department of Critical Care Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072, People’s Republic of China
| | - Yingchuan Li
- Department of Critical Care Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072, People’s Republic of China
| |
Collapse
|
4
|
Faria J, Calcat-I-Cervera S, Skovronova R, Broeksma BC, Berends AJ, Zaal EA, Bussolati B, O'Brien T, Mihăilă SM, Masereeuw R. Mesenchymal stromal cells secretome restores bioenergetic and redox homeostasis in human proximal tubule cells after ischemic injury. Stem Cell Res Ther 2023; 14:353. [PMID: 38072933 PMCID: PMC10712181 DOI: 10.1186/s13287-023-03563-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Ischemia/reperfusion injury is the leading cause of acute kidney injury (AKI). The current standard of care focuses on supporting kidney function, stating the need for more efficient and targeted therapies to enhance repair. Mesenchymal stromal cells (MSCs) and their secretome, either as conditioned medium (CM) or extracellular vesicles (EVs), have emerged as promising options for regenerative therapy; however, their full potential in treating AKI remains unknown. METHODS In this study, we employed an in vitro model of chemically induced ischemia using antimycin A combined with 2-deoxy-D-glucose to induce ischemic injury in proximal tubule epithelial cells. Afterwards we evaluated the effects of MSC secretome, CM or EVs obtained from adipose tissue, bone marrow, and umbilical cord, on ameliorating the detrimental effects of ischemia. To assess the damage and treatment outcomes, we analyzed cell morphology, mitochondrial health parameters (mitochondrial activity, ATP production, mass and membrane potential), and overall cell metabolism by metabolomics. RESULTS Our findings show that ischemic injury caused cytoskeletal changes confirmed by disruption of the F-actin network, energetic imbalance as revealed by a 50% decrease in the oxygen consumption rate, increased oxidative stress, mitochondrial dysfunction, and reduced cell metabolism. Upon treatment with MSC secretome, the morphological derangements were partly restored and ATP production increased by 40-50%, with umbilical cord-derived EVs being most effective. Furthermore, MSC treatment led to phenotype restoration as indicated by an increase in cell bioenergetics, including increased levels of glycolysis intermediates, as well as an accumulation of antioxidant metabolites. CONCLUSION Our in vitro model effectively replicated the in vivo-like morphological and molecular changes observed during ischemic injury. Additionally, treatment with MSC secretome ameliorated proximal tubule damage, highlighting its potential as a viable therapeutic option for targeting AKI.
Collapse
Affiliation(s)
- João Faria
- Division of Pharmacology, Department of Pharmaceutical Sciences, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| | - Sandra Calcat-I-Cervera
- College of Medicine, Nursing and Health Science, School of Medicine, Regenerative Medicine Institute (REMEDI), University of Galway, Galway, Ireland
| | - Renata Skovronova
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | | | - Alinda J Berends
- Division of Pharmacology, Department of Pharmaceutical Sciences, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| | - Esther A Zaal
- Division of Cell Biology, Metabolism and Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Timothy O'Brien
- College of Medicine, Nursing and Health Science, School of Medicine, Regenerative Medicine Institute (REMEDI), University of Galway, Galway, Ireland
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Silvia M Mihăilă
- Division of Pharmacology, Department of Pharmaceutical Sciences, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| | - Rosalinde Masereeuw
- Division of Pharmacology, Department of Pharmaceutical Sciences, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands.
| |
Collapse
|
5
|
Liu M, Chen Z, Zhang H, Cai Z, Liu T, Zhang M, Wu X, Ai F, Liu G, Zeng C, Shen J. Urolithin A alleviates early brain injury after subarachnoid hemorrhage by regulating the AMPK/mTOR pathway-mediated autophagy. Neurochirurgie 2023; 69:101480. [PMID: 37598622 DOI: 10.1016/j.neuchi.2023.101480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/22/2023]
Abstract
OBJECTIVE Unfavorable outcomes in patients with subarachnoid hemorrhage (SAH) are mainly attributed to early brain injury (EBI). Reduction of neuronal death can improve the prognosis in SAH patients. Autophagy and apoptosis are critical players in neuronal death. Urolithin A (UA) is a natural compound produced by gut bacteria from ingested ellagitannins and ellagic acid. Here, we detected the role of UA in EBI post-SAH. METHODS We established an animal model of SAH in rats by endovascular perforation, with administration of UA, 3-methyladenine (3-MA) and Compound C. SAH grading, neurological function, brain water content, western blotting analysis of levels of proteins related to apoptosis, autophagy and pathways, blood-brain barrier (BBB) integrity, TUNEL staining, and immunofluorescence staining of LC3 were evaluated at 24h after SAH. RESULTS SAH induction led to neurological dysfunctions, BBB disruption, and cerebral edema at 24h post-SAH in rats, which were relieved by UA. Additionally, cortical neuronal apoptosis in SAH rats was also attenuated by UA. Moreover, UA restored autophagy level in SAH rats. Mechanistically, UA activated the AMPK/mTOR pathway. Furthermore, inhibition of autophagy and AMPK limited UA-mediated protection against EBI post-SAH CONCLUSION: UA alleviates neurological deficits, BBB permeability, and cerebral edema by inhibiting cortical neuronal apoptosis through regulating the AMPK/mTOR pathway-dependent autophagy in rats following SAH.
Collapse
Affiliation(s)
- Meiqiu Liu
- Department of Neurosurgery, Ningde Municipal Hospital of Ningde Normal University, Ningde 352000, China
| | - Zhen Chen
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Huan Zhang
- Department of Obstetrics, Maternal and Child Health Hospital of Hubei Province Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Zhiji Cai
- Department of Neurosurgery, Ningde Municipal Hospital of Ningde Normal University, Ningde 352000, China
| | - Tiancheng Liu
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Mengli Zhang
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Xian Wu
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Fen Ai
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Ganzhe Liu
- Department of Neurology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Chao Zeng
- Department of Neurosurgery, Ningde Municipal Hospital of Ningde Normal University, Ningde 352000, China.
| | - Jiancheng Shen
- Department of Neurosurgery, Ningde Municipal Hospital of Ningde Normal University, Ningde 352000, China.
| |
Collapse
|
6
|
Xiang Y, Fu Y, Wu W, Tang C, Dong Z. Autophagy in acute kidney injury and maladaptive kidney repair. BURNS & TRAUMA 2023; 11:tkac059. [PMID: 36694860 PMCID: PMC9867874 DOI: 10.1093/burnst/tkac059] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/30/2022] [Accepted: 12/20/2022] [Indexed: 01/23/2023]
Abstract
Acute kidney injury (AKI) is a major renal disease characterized by a sudden decrease in kidney function. After AKI, the kidney has the ability to repair, but if the initial injury is severe the repair may be incomplete or maladaptive and result in chronic kidney problems. Autophagy is a highly conserved pathway to deliver intracellular contents to lysosomes for degradation. Autophagy plays an important role in maintaining renal function and is involved in the pathogenesis of renal diseases. Autophagy is activated in various forms of AKI and acts as a defense mechanism against kidney cell injury and death. After AKI, autophagy is maintained at a relatively high level in kidney tubule cells during maladaptive kidney repair but the role of autophagy in maladaptive kidney repair has been controversial. Nonetheless, recent studies have demonstrated that autophagy may contribute to maladaptive kidney repair after AKI by inducing tubular degeneration and promoting a profibrotic phenotype in renal tubule cells. In this review, we analyze the role and regulation of autophagy in kidney injury and repair and discuss the therapeutic strategies by targeting autophagy.
Collapse
Affiliation(s)
- Yu Xiang
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital at Central South University, Changsha 410000, Hunan Province, China
| | - Ying Fu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital at Central South University, Changsha 410000, Hunan Province, China
| | - Wenwen Wu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital at Central South University, Changsha 410000, Hunan Province, China
| | - Chengyuan Tang
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital at Central South University, Changsha 410000, Hunan Province, China
| | | |
Collapse
|
7
|
Chen T, Niu L, Wang L, Zhou Q, Zhao X, Lai S, He X, He H, He M. Ferulic acid protects renal tubular epithelial cells against anoxia/reoxygenation injury mediated by AMPKα1. Free Radic Res 2022; 56:173-184. [PMID: 35382666 DOI: 10.1080/10715762.2022.2062339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Anoxia/reoxygenation (A/R) injury causes dysfunction of rat renal tubular epithelial cells (NRK-52E), which is associated with excess reactive oxygen species (ROS) generation and eventually leads to apoptosis. Ferulic acid (FA), a phenolic acid, which is abundant in fruits and vegetables. FA possesses the properties of scavenging free radicals and cytoprotection against oxygen stress. In the study, the protective effects of FA against NRK-52E cells damage induced by A/R were explored and confirmed the role of AMP-activated protein kinaseα1 (AMPKα1). We found that after NRK-52E cells suffered A/R damage, FA pretreatment increased the cell viability and decreased LDH activity in culture medium in a concentration-dependent manner, the activities of endogenous antioxidant enzymes such as glutathione peroxidase, superoxide dismutase and catalase improved, intracellular ROS generation and malondialdehyde contents mitigated. In addition, pretreatment of 75 μM FA ameliorated mitochondrial dysfunction by A/R-injury and ultimately decreased apoptosis (25.3 ± 0.61 vs 12.1 ± 0.60), which was evidenced by preventing the release of cytochrome c from mitochondria to the cytoplasm. 75 μM FA pretreatment also significantly upregulated AMPKα1 expression (3.16 ± 0.18 folds) and phosphorylation (2.56 ± 0.13 folds). However, compound C, a specific AMPK inhibitor, significantly attenuated FA pretreatment's effects, as mentionedabove. These results firstly clarified that FA pretreatment attenuated NRK-52E cell damage induced by A/R via upregulating AMPKα1 expression and phosphorylation.
Collapse
Affiliation(s)
- Tianpeng Chen
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Li Niu
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Liang Wang
- Department of rehabilitation, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Qing Zhou
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Xiaoyu Zhao
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Songqing Lai
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Xinlan He
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Huan He
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang 330006, China
| | - Ming He
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| |
Collapse
|
8
|
Rodríguez C, Muñoz M, Contreras C, Prieto D. AMPK, metabolism, and vascular function. FEBS J 2021; 288:3746-3771. [PMID: 33825330 DOI: 10.1111/febs.15863] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/04/2021] [Accepted: 04/04/2021] [Indexed: 12/12/2022]
Abstract
Adenosine monophosphate-activated protein kinase (AMPK) is a cellular energy sensor activated during energy stress that plays a key role in maintaining energy homeostasis. This ubiquitous signaling pathway has been implicated in multiple functions including mitochondrial biogenesis, redox regulation, cell growth and proliferation, cell autophagy and inflammation. The protective role of AMPK in cardiovascular function and the involvement of dysfunctional AMPK in the pathogenesis of cardiovascular disease have been highlighted in recent years. In this review, we summarize and discuss the role of AMPK in the regulation of blood flow in response to metabolic demand and the basis of the AMPK physiological anticontractile, antioxidant, anti-inflammatory, and antiatherogenic actions in the vascular system. Investigations by others and us have demonstrated the key role of vascular AMPK in the regulation of endothelial function, redox homeostasis, and inflammation, in addition to its protective role in the hypoxia and ischemia/reperfusion injury. The pathophysiological implications of AMPK involvement in vascular function with regard to the vascular complications of metabolic disease and the therapeutic potential of AMPK activators are also discussed.
Collapse
Affiliation(s)
- Claudia Rodríguez
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - Mercedes Muñoz
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - Cristina Contreras
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - Dolores Prieto
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| |
Collapse
|
9
|
Oeing CU, Jun S, Mishra S, Dunkerly-Eyring BL, Chen A, Grajeda MI, Tahir UA, Gerszten RE, Paolocci N, Ranek MJ, Kass DA. MTORC1-Regulated Metabolism Controlled by TSC2 Limits Cardiac Reperfusion Injury. Circ Res 2021; 128:639-651. [PMID: 33401933 DOI: 10.1161/circresaha.120.317710] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
RATIONALE The mTORC1 (mechanistic target of rapamycin complex-1) controls metabolism and protein homeostasis and is activated following ischemia reperfusion (IR) injury and by ischemic preconditioning (IPC). However, studies vary as to whether this activation is beneficial or detrimental, and its influence on metabolism after IR is little reported. A limitation of prior investigations is their use of broad gain/loss of mTORC1 function, mostly applied before ischemic stress. This can be circumvented by regulating one serine (S1365) on TSC2 (tuberous sclerosis complex) to achieve bidirectional mTORC1 modulation but only with TCS2-regulated costimulation. OBJECTIVE We tested the hypothesis that reduced TSC2 S1365 phosphorylation protects the myocardium against IR and is required for IPC by amplifying mTORC1 activity to favor glycolytic metabolism. METHODS AND RESULTS Mice with either S1365A (TSC2SA; phospho-null) or S1365E (TSC2SE; phosphomimetic) knockin mutations were studied ex vivo and in vivo. In response to IR, hearts from TSC2SA mice had amplified mTORC1 activation and improved heart function compared with wild-type and TSC2SE hearts. The magnitude of protection matched IPC. IPC requited less S1365 phosphorylation, as TSC2SE hearts gained no benefit and failed to activate mTORC1 with IPC. IR metabolism was altered in TSC2SA, with increased mitochondrial oxygen consumption rate and glycolytic capacity (stressed/maximal extracellular acidification) after myocyte hypoxia-reperfusion. In whole heart, lactate increased and long-chain acylcarnitine levels declined during ischemia. The relative IR protection in TSC2SA was lost by lowering glucose in the perfusate by 36%. Adding fatty acid (palmitate) compensated for reduced glucose in wild type and TSC2SE but not TSC2SA which had the worst post-IR function under these conditions. CONCLUSIONS TSC2-S1365 phosphorylation status regulates myocardial substrate utilization, and its decline activates mTORC1 biasing metabolism away from fatty acid oxidation to glycolysis to confer protection against IR. This pathway is also engaged and reduced TSC2 S1365 phosphorylation required for effective IPC. Graphic Abstract: A graphic abstract is available for this article.
Collapse
Affiliation(s)
- Christian U Oeing
- Department of Internal Medicine and Cardiology, Charité University Medicine, Campus Virchow-Klinikum, Berlin, Germany, and German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany (C.U.O.).,Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD (C.U.O., S.J., S.M., B.L.D.-E., A.C., M.I.G., N.P., M.J.R., D.A.K.)
| | - Seungho Jun
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD (C.U.O., S.J., S.M., B.L.D.-E., A.C., M.I.G., N.P., M.J.R., D.A.K.)
| | - Sumita Mishra
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD (C.U.O., S.J., S.M., B.L.D.-E., A.C., M.I.G., N.P., M.J.R., D.A.K.)
| | - Brittany L Dunkerly-Eyring
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD (C.U.O., S.J., S.M., B.L.D.-E., A.C., M.I.G., N.P., M.J.R., D.A.K.).,Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, MD (B.L.D.-E., D.A.K.)
| | - Anna Chen
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD (C.U.O., S.J., S.M., B.L.D.-E., A.C., M.I.G., N.P., M.J.R., D.A.K.)
| | - Maria I Grajeda
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD (C.U.O., S.J., S.M., B.L.D.-E., A.C., M.I.G., N.P., M.J.R., D.A.K.)
| | - Usman A Tahir
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (U.A.T., R.E.G.)
| | - Robert E Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (U.A.T., R.E.G.)
| | - Nazareno Paolocci
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD (C.U.O., S.J., S.M., B.L.D.-E., A.C., M.I.G., N.P., M.J.R., D.A.K.).,Department of Biomedical Sciences, University of Padova, Italy (N.P.)
| | - Mark J Ranek
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD (C.U.O., S.J., S.M., B.L.D.-E., A.C., M.I.G., N.P., M.J.R., D.A.K.)
| | - David A Kass
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD (C.U.O., S.J., S.M., B.L.D.-E., A.C., M.I.G., N.P., M.J.R., D.A.K.).,Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, MD (B.L.D.-E., D.A.K.)
| |
Collapse
|
10
|
Packer M. Role of Impaired Nutrient and Oxygen Deprivation Signaling and Deficient Autophagic Flux in Diabetic CKD Development: Implications for Understanding the Effects of Sodium-Glucose Cotransporter 2-Inhibitors. J Am Soc Nephrol 2020; 31:907-919. [PMID: 32276962 DOI: 10.1681/asn.2020010010] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Growing evidence indicates that oxidative and endoplasmic reticular stress, which trigger changes in ion channels and inflammatory pathways that may undermine cellular homeostasis and survival, are critical determinants of injury in the diabetic kidney. Cells are normally able to mitigate these cellular stresses by maintaining high levels of autophagy, an intracellular lysosome-dependent degradative pathway that clears the cytoplasm of dysfunctional organelles. However, the capacity for autophagy in both podocytes and renal tubular cells is markedly impaired in type 2 diabetes, and this deficiency contributes importantly to the intensity of renal injury. The primary drivers of autophagy in states of nutrient and oxygen deprivation-sirtuin-1 (SIRT1), AMP-activated protein kinase (AMPK), and hypoxia-inducible factors (HIF-1α and HIF-2α)-can exert renoprotective effects by promoting autophagic flux and by exerting direct effects on sodium transport and inflammasome activation. Type 2 diabetes is characterized by marked suppression of SIRT1 and AMPK, leading to a diminution in autophagic flux in glomerular podocytes and renal tubules and markedly increasing their susceptibility to renal injury. Importantly, because insulin acts to depress autophagic flux, these derangements in nutrient deprivation signaling are not ameliorated by antihyperglycemic drugs that enhance insulin secretion or signaling. Metformin is an established AMPK agonist that can promote autophagy, but its effects on the course of CKD have been demonstrated only in the experimental setting. In contrast, the effects of sodium-glucose cotransporter-2 (SGLT2) inhibitors may be related primarily to enhanced SIRT1 and HIF-2α signaling; this can explain the effects of SGLT2 inhibitors to promote ketonemia and erythrocytosis and potentially underlies their actions to increase autophagy and mute inflammation in the diabetic kidney. These distinctions may contribute importantly to the consistent benefit of SGLT2 inhibitors to slow the deterioration in glomerular function and reduce the risk of ESKD in large-scale randomized clinical trials of patients with type 2 diabetes.
Collapse
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular Institute, Baylor University Medical Center, Dallas, Texas .,Imperial College, London, United Kingdom
| |
Collapse
|
11
|
Lee H, Zandkarimi F, Zhang Y, Meena JK, Kim J, Zhuang L, Tyagi S, Ma L, Westbrook TF, Steinberg GR, Nakada D, Stockwell BR, Gan B. Energy-stress-mediated AMPK activation inhibits ferroptosis. Nat Cell Biol 2020; 22:225-234. [PMID: 32029897 PMCID: PMC7008777 DOI: 10.1038/s41556-020-0461-8] [Citation(s) in RCA: 739] [Impact Index Per Article: 147.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 01/03/2020] [Indexed: 02/08/2023]
Abstract
Energy stress depletes ATP and induces cell death. Here, we identify an unexpected inhibitory role of energy stress on ferroptosis, a form of regulated cell death induced by iron-dependent lipid peroxidation. We found that ferroptotic cell death and lipid peroxidation can be inhibited by treatments that induce or mimic energy stress. Inactivation of AMP-activated protein kinase (AMPK), a sensor of cellular energy status, largely abolishes the protective effects of energy stress on ferroptosis in vitro and on ferroptosis-associated renal ischemia/reperfusion injury in vivo. Cancer cells with high basal AMPK activation are resistant to ferroptosis, and AMPK inactivation sensitizes these cells to ferroptosis. Functional and lipidomic analyses further link AMPK regulation of ferroptosis to AMPK-mediated phosphorylation of acetyl-CoA carboxylase (ACC) and polyunsaturated fatty acid biosynthesis. Together, our study demonstrates that energy stress inhibits ferroptosis partly through AMPK, and reveals an unexpected coupling between ferroptosis and AMPK-mediated energy stress signaling.
Collapse
Affiliation(s)
- Hyemin Lee
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Yilei Zhang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jitendra Kumar Meena
- Verna & Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Jongchan Kim
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,School of Natural Sciences, Department of Life Sciences, Sogang University, Seoul, Republic of Korea
| | - Li Zhuang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Siddhartha Tyagi
- Verna & Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Li Ma
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Thomas F Westbrook
- Verna & Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Therapeutic Innovation Center (THINC), Baylor College of Medicine, Houston, TX, USA
| | - Gregory R Steinberg
- Centre for Metabolism, Obesity and Diabetes Research, Department of Medicine and Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Daisuke Nakada
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Brent R Stockwell
- Department of Biological Sciences, Columbia University, New York, NY, USA. .,Department of Chemistry, Columbia University, New York, NY, USA.
| | - Boyi Gan
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA. .,Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
12
|
Shi M, Hu Z, Zhang X, You Q, Wang W, Yan R, Zhu Z. AMPK activation suppresses mTOR/S6K1 phosphorylation and induces leucine resistance in rats with sepsis. Cell Biol Int 2020; 44:1133-1141. [PMID: 31943518 DOI: 10.1002/cbin.11310] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 01/10/2020] [Indexed: 11/12/2022]
Abstract
Although it has been known that protein synthesis is suppressed in sepsis, which cannot be corrected by leucine supplement (also known as leucine resistance), the molecular signaling mechanism remains unclear. This study aimed to investigate the AMP-activated protein kinase/mammalian target of rapamycin (AMPK/mTOR) pathway in sepsis-induced leucine resistance and its upstream signals, and to seek a way to correct leucine resistance in sepsis. Sepsis was produced by cecal ligation and puncture (CLP) model in rat. Both septic rats and sham operation rat received total parenteral nutrition (TPN) with or without leucine for 24 h, and then protein synthesis and AMPK/mTOR and protein kinase B (PKB) were tested. In vitro C2C12 cells were treated with or without leucine, and we tested the AMPK/mTOR pathway and protein synthesis. We blocked AMPK by compound C and stimulated it by 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR) individually. The results showed that AMPK was highly phosphorylated and suppressed mTOR/S6K1 activation in CLP rats. In vitro when AMPK was activated by AICAR, protein synthesis was suppressed and leucine resistance was observed. High phosphorylation of AMPK was accompanied by PKB inactivation in CLP rats. When PKB was blocked, both AMPK activation and leucine resistance were observed. In CLP rats, nutrition support with intensive insulin therapy reversed leucine resistance by activating PKB and suppressing AMPK phosphorylation. These findings suggest that high phosphorylation of AMPK induced by PKB inactivation in sepsis suppresses mTOR, S6K1 phosphorylation, and protein synthesis and leads to leucine resistance. Intensive insulin treatment can reverse leucine resistance by suppressing AMPK activation through activation of PKB.
Collapse
Affiliation(s)
- Mengyao Shi
- Gastro-intestine Surgery Department, Shanghai Changzheng Hospital, Second Military Medical University, 415 FengYang Road, Shanghai, 200003, PR China
| | - Zunqi Hu
- Gastro-intestine Surgery Department, Shanghai Changzheng Hospital, Second Military Medical University, 415 FengYang Road, Shanghai, 200003, PR China
| | - Xin Zhang
- Gastro-intestine Surgery Department, Shanghai Changzheng Hospital, Second Military Medical University, 415 FengYang Road, Shanghai, 200003, PR China
| | - Qing You
- Gastro-intestine Surgery Department, Shanghai Changzheng Hospital, Second Military Medical University, 415 FengYang Road, Shanghai, 200003, PR China
| | - Weimin Wang
- Gastro-intestine Surgery Department, Shanghai Changzheng Hospital, Second Military Medical University, 415 FengYang Road, Shanghai, 200003, PR China
| | - Ronglin Yan
- Gastro-intestine Surgery Department, Shanghai Changzheng Hospital, Second Military Medical University, 415 FengYang Road, Shanghai, 200003, PR China
| | - Zhenxin Zhu
- Gastro-intestine Surgery Department, Shanghai Changzheng Hospital, Second Military Medical University, 415 FengYang Road, Shanghai, 200003, PR China
| |
Collapse
|
13
|
Zheng C, Zhou Y, Huang Y, Chen B, Wu M, Xie Y, Chen X, Sun M, Liu Y, Chen C, Pan J. Effect of ATM on inflammatory response and autophagy in renal tubular epithelial cells in LPS-induced septic AKI. Exp Ther Med 2019; 18:4707-4717. [PMID: 31777559 PMCID: PMC6862447 DOI: 10.3892/etm.2019.8115] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 07/29/2019] [Indexed: 12/19/2022] Open
Abstract
The aim of the present study was to explore the role of ataxia-telangiectasia mutated (ATM) in lipopolysaccharide (LPS)-induced in vitro model of septic acute kidney injury (AKI) and the association between ATM, tubular epithelial inflammatory response and autophagy. The renal tubular epithelial cell HK-2 cell line was cultured and passaged, with HK-2 cell injury induced by LPS. The effects of LPS on HK-2 cell morphology, viability, ATM expression and inflammation were observed. Lentiviral vectors encoding ATM shRNA were constructed to knock down ATM expression in HK-2 cells. The efficiency of ATM knockdown in HK-2 cells was detected by western blot analysis and reverse transcription-quantitative PCR (RT-qPCR). HK-2 cells transfected with the ATM shRNA lentivirus were used for subsequent experiments. Following ATM knockdown, corresponding controls were set up, and the effects of ATM on inflammation and autophagy were detected in HK-2 cells using RT-qPCR, western blotting and ELISA. After LPS stimulation, the HK-2 cells were rounded into a slender or fusiform shape with poorly defined outlines. LPS treatment reduced cell viability in a partly dose-dependent manner. LPS increased the expression of tumor necrosis factor-α, interleukin (IL)-1β and IL-6, with the levels reaching its highest value at 10 µg/ml. IL-6 and IL-1β expression increased with increasing LPS concentration. These findings suggest that LPS reduced HK-2 cell viability whilst increasing the expression of inflammatory factors. Following transfection with ATM shRNA, expression levels of key autophagy indicators microtubule associated protein 1 light chain 3α I/II ratio and beclin-1 in the two ATM shRNA groups were also significantly reduced compared with the NC shRNA group. In summary, downregulation of ATM expression in HK-2 cells reduced LPS-induced inflammation and autophagy in sepsis-induced AKI in vitro, suggesting that LPS may induce autophagy in HK-2 cells through the ATM pathway leading to the upregulation of inflammatory factors.
Collapse
Affiliation(s)
- Chenfei Zheng
- Department of Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Ying Zhou
- Department of Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yueyue Huang
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Bicheng Chen
- Zhejiang Provincial Top Key Discipline in Surgery, Wenzhou Key Laboratory of Surgery, Department of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Minmin Wu
- Zhejiang Provincial Top Key Discipline in Surgery, Wenzhou Key Laboratory of Surgery, Department of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yue Xie
- Zhejiang Provincial Top Key Discipline in Surgery, Wenzhou Key Laboratory of Surgery, Department of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Xinxin Chen
- Department of Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Mei Sun
- Department of Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yi Liu
- Department of Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Chaosheng Chen
- Department of Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Jingye Pan
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
14
|
Hou J, Rao M, Zheng W, Fan J, Law BYK. Advances on Cell Autophagy and Its Potential Regulatory Factors in Renal Ischemia-Reperfusion Injury. DNA Cell Biol 2019; 38:895-904. [PMID: 31347925 DOI: 10.1089/dna.2019.4767] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Ischemia-reperfusion injury is a major reason for acute kidney injury and various kidney diseases. Autophagy plays an important role during renal ischemia-reperfusion injury (RIRI), but it remains controversial whether autophagy contributes to cell survival or ischemia-reperfusion-induced cell death. In the review, we summarized the function of autophagy in the progression of acute ischemic kidney injury, as well as its related molecular mechanisms. While analyzing the opposite roles of autophagy in RIRI, it was concluded that the protective or detrimental function of autophagy was depending on the timing and amount of the activation of cell autophagy. We also summarized the regulatory agents, including active compounds, proteins, or microRNAs (miRNAs), which regulated the cell autophagy during renal acute ischemic kidney injury process. This explained why the opposite conclusion occurred when cell autophagy was studied in the RIRI models from different researchers. Therefore, the article provided a hypothesis to control cell autophagy at the appropriate timing and intensity so as to alleviate renal injury and sustain cell survival of the renal cell.
Collapse
Affiliation(s)
- Jing Hou
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, People's Republic of China.,State Key Laboratory of Quality Research in Chinese Medicines (Macau University of Science and Technology), Taipa, People's Republic of China.,Department of Nephrology, the Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| | - Mingyue Rao
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, People's Republic of China.,State Key Laboratory of Quality Research in Chinese Medicines (Macau University of Science and Technology), Taipa, People's Republic of China.,Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| | - Wenlu Zheng
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, People's Republic of China.,State Key Laboratory of Quality Research in Chinese Medicines (Macau University of Science and Technology), Taipa, People's Republic of China.,Department of Nuclear Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| | - Junming Fan
- Chengdu Medical College, Chengdu City, People's Republic of China.,Southwest Medical University, Luzhou, People's Republic of China
| | - Betty Yuen Kwan Law
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, People's Republic of China.,State Key Laboratory of Quality Research in Chinese Medicines (Macau University of Science and Technology), Taipa, People's Republic of China
| |
Collapse
|
15
|
Zhao W, Zhang L, Chen R, Lu H, Sui M, Zhu Y, Zeng L. SIRT3 Protects Against Acute Kidney Injury via AMPK/mTOR-Regulated Autophagy. Front Physiol 2018; 9:1526. [PMID: 30487750 PMCID: PMC6246697 DOI: 10.3389/fphys.2018.01526] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 10/11/2018] [Indexed: 01/04/2023] Open
Abstract
Acute kidney injury (AKI), which involves the loss of kidney function caused by damage to renal tubular cells, is an important public health concern. We previously showed that sirtuin (SIRT)3 protects the kidneys against mitochondrial damage by inhibiting the nucleotide-binding domain (NOD)-like receptor protein 3 (NLRP3) inflammasome, attenuating oxidative stress, and downregulating proinflammatory cytokines. In this article, we investigated the role of autophagy, mediated by a mammalian target of rapamycin (mTOR) and AMP-activated protein kinase (AMPK), in the protective effect of SIRT3, against sepsis-induced AKI, in a mouse model of cecal ligation and puncture (CLP). The AKI in CLP mice was associated with the upregulation of autophagy markers; this effect was abolished in SIRT3- mice in parallel with the downregulation of phospho (p)-AMPK and the upregulation of p-mTOR. Pretreatment with the autophagy inhibitor 3-methyladenine (3-MA) or AMPK inhibitor compound isotonic saline (C), exacerbated AKI. SIRT3 overexpression promoted autophagy, upregulated p-AMPK and downregulated p-mTOR in CLP mice, attenuating sepsis-induced AKI, tubular cell apoptosis, and inflammatory cytokine accumulation in the kidneys. The blockage of autophagy induction largely abolished the protective effect of SIRT3 in sepsis-induced AKI. These findings indicate that SIRT3 protects against CLP-induced AKI by inducing autophagy through regulation of the AMPK/mTOR pathway.
Collapse
Affiliation(s)
- Wenyu Zhao
- Department of Organ Transplantation, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Lei Zhang
- Department of Organ Transplantation, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Rui Chen
- Department of Organ Transplantation, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Hanlan Lu
- Department of Organ Transplantation, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Mingxing Sui
- Department of Organ Transplantation, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Youhua Zhu
- Department of Organ Transplantation, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Li Zeng
- Department of Organ Transplantation, Changhai Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
16
|
Zhang Y, Wang L, Meng L, Cao GK, Zhao YL, Wu Y. Expression changes of autophagy-related proteins in AKI patients treated with CRRT and their effects on prognosis of adult and elderly patients. IMMUNITY & AGEING 2018; 15:23. [PMID: 30305832 PMCID: PMC6169063 DOI: 10.1186/s12979-018-0128-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 08/29/2018] [Indexed: 01/22/2023]
Abstract
Background Sepsis is one of the common death factors in intensive care unit, which refers to the systemic inflammatory response syndrome caused by infection. It has many complications such as acute renal injury, shock, multiple organ dysfunction, and failure. The mortality of acute renal injury is the highest among the complications, which is a serious threat to the safety of patients and affects the quality of life. This study aimed to observe the changes in autophagy-related protein expressions in patients with acute kidney injury (AKI) after continuous renal replacement therapy (CRRT) and their impacts on prognosis. Methods 207 AKI patients visiting the Emergency Department of The First People's Hospital of Xuzhou from January 2014 to February 2018 were recruited and treated with CRRT. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) was applied to detect the expression of autophagy-related genes, including light chain 3 type II (LC3-II), autophagy-related 5 (Atg-5) and Beclin-1, in the monocytes of the patient's peripheral blood before and after treatment. The levels of inflammatory mediators interleukin (IL)-1β and IL-6 were determined via enzyme-linked immunosorbent assay before and after treatment. The patient's serum creatinine (Scr) level before and after treatment was measured using a full-automatic biochemistry analyser. Moreover, the treatment effect was graded after CRRT, and the relationship between the prognosis of patients and the autophagy-related proteins was observed. Results The Scr levels in the patients were significantly decreased after treatment with CRRT. Before treatment, the IL-1β and IL-6 blood levels were high in the patients, while the amounts were significantly reduced after CRTT. The expressions of LC3-II, Atg-5 and Beclin-1 in the monocytes of patients after treatment were significantly decreased compared with those before treatment. Compared with those in survived patients, the expression of autophagy-related proteins was significantly elevated in in patients died after one to three weeks after the treatment. IL-1β, IL-6, LC3-II and Beclin-1, but not Atg-5 values were significantly correlated with Scr. Conclusion The expression of LC3-II, Atg-5 and Beclin-1 in the monocytes of patients may change prominently after treatment with CRRT, so they are expected to be regarded as new prognostic indicators for AKI patients.
Collapse
Affiliation(s)
- Yang Zhang
- 1Department of Anesthesiology, Xuzhou Medical University, Xuzhou, 221000 Jiangsu China
| | - Ling Wang
- 2Department of Nephrology, The First People's Hospital of Xuzhou, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu 221000 People's Republic of China
| | - Lei Meng
- 1Department of Anesthesiology, Xuzhou Medical University, Xuzhou, 221000 Jiangsu China
| | - Guang-Ke Cao
- 2Department of Nephrology, The First People's Hospital of Xuzhou, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu 221000 People's Republic of China
| | - Yu-Liang Zhao
- 2Department of Nephrology, The First People's Hospital of Xuzhou, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu 221000 People's Republic of China
| | - Yu Wu
- 2Department of Nephrology, The First People's Hospital of Xuzhou, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu 221000 People's Republic of China
| |
Collapse
|
17
|
Kodiha M, Flamant E, Wang YM, Stochaj U. Defining the short-term effects of pharmacological 5'-AMP activated kinase modulators on mitochondrial polarization, morphology and heterogeneity. PeerJ 2018; 6:e5469. [PMID: 30186684 PMCID: PMC6119600 DOI: 10.7717/peerj.5469] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 07/27/2018] [Indexed: 01/09/2023] Open
Abstract
Background Under aerobic growth conditions, mitochondria are the major producers of cellular ATP and crucial for the proper performance of organs and tissues. This applies especially to cells with high energy demand, such as the renal proximal tubule epithelium. Mitochondrial dysfunction contributes to the pathology of human health conditions, including various kidney diseases. The improvement of mitochondrial function ameliorates some of these pathologies. This can potentially be achieved with pharmacological compounds. For example, long-term treatment with activators of 5'-AMP activated kinase (AMPK) enhances mitochondrial biogenesis. However, pharmacological damage control during acute cell injury requires that the short-term effects of these compounds and the impact on healthy cells are also understood. It was our objective to define the changes elicited by established modulators of AMPK activity in healthy renal proximal tubule cells. Methods Our work combines confocal microscopy with quantitative image analysis, 3D image reconstruction and Western blotting to provide novel insights into the biology of mitochondria. Specifically, we evaluated the effects of pharmacological AMPK modulators (compound C, AICAR, phenformin, resveratrol) on mitochondrial polarization, morphology and heterogeneity. Microscopic studies generated information at the single cell and subcellular levels. Our research focused on LLC-PK1 cells that are derived from the renal proximal tubule. Mitochondrial heterogeneity was also examined in MCF7 breast cancer cells. Results Pharmacological agents that affect AMPK activity in renal proximal tubule cells can alter mitochondrial organization and the electrochemical potential across the inner mitochondrial membrane. These changes were compound-specific. Short-term incubation with the AMPK inhibitor compound C caused mitochondrial hyperpolarization. This was accompanied by mitochondrial fragmentation. By contrast, AMPK activators AICAR, phenformin and resveratrol had little impact. We further show that the biological properties of mitochondria are determined by their subcellular location. Mitochondria at the cell periphery displayed higher MitoTracker/Tom70 values as compared to organelles located in the vicinity of the nucleus. This was not limited to renal proximal tubule cells, but also observed in MCF7 cells. Pharmacological AMPK modulators altered these location-dependent properties in a compound-specific fashion. While the region-dependent differences were enhanced with phenformin, they were ameliorated by resveratrol. Discussion We evaluated the rapid changes in mitochondrial characteristics that are induced by pharmacological AMPK modulators. Our research supports the concept that pharmacological agents that target AMPK can rearrange mitochondrial networks at the single cell level. Collectively, these insights are relevant to the development of proper strategies for the short-term adjustment of mitochondrial performance.
Collapse
Affiliation(s)
- Mohamed Kodiha
- Department of Physiology, McGill University, Montreal, Canada
| | - Etienne Flamant
- Department of Physiology, McGill University, Montreal, Canada
| | - Yi Meng Wang
- Department of Physiology, McGill University, Montreal, Canada
| | - Ursula Stochaj
- Department of Physiology, McGill University, Montreal, Canada
| |
Collapse
|
18
|
Jiang S, Li T, Ji T, Yi W, Yang Z, Wang S, Yang Y, Gu C. AMPK: Potential Therapeutic Target for Ischemic Stroke. Theranostics 2018; 8:4535-4551. [PMID: 30214637 PMCID: PMC6134933 DOI: 10.7150/thno.25674] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 07/16/2018] [Indexed: 02/07/2023] Open
Abstract
5'-AMP-activated protein kinase (AMPK), a member of the serine/threonine (Ser/Thr) kinase group, is universally distributed in various cells and organs. It is a significant endogenous defensive molecule that responds to harmful stimuli, such as cerebral ischemia, cerebral hemorrhage, and, neurodegenerative diseases (NDD). Cerebral ischemia, which results from insufficient blood flow or the blockage of blood vessels, is a major cause of ischemic stroke. Ischemic stroke has received increased attention due to its '3H' effects, namely high mortality, high morbidity, and high disability. Numerous studies have revealed that activation of AMPK plays a protective role in the brain, whereas its action in ischemic stroke remains elusive and poorly understood. Based on existing evidence, we introduce the basic structure, upstream regulators, and biological roles of AMPK. Second, we analyze the relationship between AMPK and the neurovascular unit (NVU). Third, the actions of AMPK in different phases of ischemia and current therapeutic methods are discussed. Finally, we evaluate existing controversy and provide a detailed analysis, followed by ethical issues, potential directions, and further prospects of AMPK. The information complied here may aid in clinical and basic research of AMPK, which may be a potent drug candidate for ischemic stroke treatment in the future.
Collapse
Affiliation(s)
- Shuai Jiang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
- Department of Aerospace Medicine, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Tian Li
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Ting Ji
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Wei Yi
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China
| | - Zhi Yang
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Simeng Wang
- Center for Human Nutrition, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Chunhu Gu
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China
| |
Collapse
|
19
|
Zhu J, Liu K, Huang K, Gu Y, Hu Y, Pan S, Ji Z. Metformin Improves Neurologic Outcome Via AMP-Activated Protein Kinase-Mediated Autophagy Activation in a Rat Model of Cardiac Arrest and Resuscitation. J Am Heart Assoc 2018; 7:e008389. [PMID: 29895585 PMCID: PMC6220525 DOI: 10.1161/jaha.117.008389] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 05/01/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Sudden cardiac arrest (CA) often results in severe injury to the brain, and neuroprotection after CA has proved to be difficult to achieve. Herein, we sought to investigate the effects of metformin pretreatment on brain injury secondary to CA and cardiopulmonary resuscitation. METHODS AND RESULTS Rats were subjected to 9-minute asphyxial CA after receiving daily metformin treatment for 2 weeks. Survival rate, neurologic deficit scores, neuronal loss, AMP-activated protein kinase (AMPK), and autophagy activation were assessed at indicated time points within the first 7 days after return of spontaneous circulation. Our results showed that metformin pretreatment elevated the 7-day survival rate from 55% to 85% and significantly reduced neurologic deficit scores. Moreover, metformin ameliorated CA-induced neuronal degeneration and glial activation in the hippocampal CA1 region, which was accompanied by augmented AMPK phosphorylation and autophagy activation in affected neuronal tissue. Inhibition of AMPK or autophagy with pharmacological inhibitors abolished metformin-afforded neuroprotection, and augmented autophagy induction by metformin treatment appeared downstream of AMPK activation. CONCLUSIONS Taken together, our data demonstrate, for the first time, that metformin confers neuroprotection against ischemic brain injury after CA/cardiopulmonary resuscitation by augmenting AMPK-dependent autophagy activation.
Collapse
Affiliation(s)
- Juan Zhu
- Department of Neurology, Nanfang Hospital Southern Medical University, Guangdong, China
| | - Kewei Liu
- Department of Neurology, Nanfang Hospital Southern Medical University, Guangdong, China
| | - Kaibin Huang
- Department of Neurology, Nanfang Hospital Southern Medical University, Guangdong, China
| | - Yong Gu
- Department of Neurology, Nanfang Hospital Southern Medical University, Guangdong, China
| | - Yafang Hu
- Department of Neurology, Nanfang Hospital Southern Medical University, Guangdong, China
| | - Suyue Pan
- Department of Neurology, Nanfang Hospital Southern Medical University, Guangdong, China
| | - Zhong Ji
- Department of Neurology, Nanfang Hospital Southern Medical University, Guangdong, China
| |
Collapse
|
20
|
Li X, Zhu G, Gou X, He W, Yin H, Yang X, Li J. Negative feedback loop of autophagy and endoplasmic reticulum stress in rapamycin protection against renal ischemia-reperfusion injury during initial reperfusion phase. FASEB J 2018; 32:fj201800299R. [PMID: 29771603 DOI: 10.1096/fj.201800299r] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Rapamycin, an immunosuppressant, is widely used in patients with kidney transplant. However, the therapeutic effects of rapamycin remain controversial. Additionally, previous studies have revealed deleterious effects of rapamycin predominantly when administered for ≥24 h. Few studies, however, have focused on the short-term effects of rapamycin administered only during the initial reperfusion phase. As such, we designed this study to explore the potential effects and mechanisms of rapamycin under a specific therapeutic regimen in which rapamycin is mixed in the perfusate during the initial reperfusion phase (within 24 h). Interestingly, we found that rapamycin maintained renal function and attenuated ischemia-reperfusion (I/R)-induced apoptosis in vivo and in vitro during the initial reperfusion phase, especially at 8 h after reperfusion. Simultaneously, rapamycin activated autophagy and inhibited endoplasmic reticulum (ER) stress and 3 pathways of unfolding protein response: ATF6, PERK, and IRE1α. Interestingly, we further found that the protective effects of rapamycin were suppressed when autophagy was inhibited by chloroquine and 3-methyladenine or when ER stress was induced by thapsigargin. Moreover, in terms of the regulatory effects of rapamycin, a negative-feedback loop between autophagy and ER stress occurred, with autophagy inhibiting ER stress and increased ER stress promoting autophagy during the initial reperfusion phase of renal I/R injury. Our study provides evidence that immediate reperfusion with rapamycin during the initial reperfusion phase repairs renal function and reduces apoptosis via activating autophagy, which could further inhibit ER stress. These results suggest a novel treatment modality for application during the initial reperfusion phase of renal I/R injury caused by kidney transplantation.-Li, X., Zhu, G., Gou, X., He, W., Yin, H., Yang, X., Li, J. Negative feedback loop of autophagy and endoplasmic reticulum stress in rapamycin protection against renal ischemia-reperfusion injury during initial reperfusion phase.
Collapse
Affiliation(s)
- Xinyuan Li
- Department of Urology, First Affiliated Hospital, Chongqing Medical University, Chongqing, China; and
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, Chongqing, China
| | - Gongmin Zhu
- Department of Urology, First Affiliated Hospital, Chongqing Medical University, Chongqing, China; and
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, Chongqing, China
| | - Xin Gou
- Department of Urology, First Affiliated Hospital, Chongqing Medical University, Chongqing, China; and
| | - Weiyang He
- Department of Urology, First Affiliated Hospital, Chongqing Medical University, Chongqing, China; and
| | - Hubin Yin
- Department of Urology, First Affiliated Hospital, Chongqing Medical University, Chongqing, China; and
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, Chongqing, China
| | - Xiaoyu Yang
- Department of Urology, First Affiliated Hospital, Chongqing Medical University, Chongqing, China; and
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, Chongqing, China
| | - Jie Li
- Department of Urology, First Affiliated Hospital, Chongqing Medical University, Chongqing, China; and
| |
Collapse
|
21
|
Li Q, Li L, Fei X, Zhang Y, Qi C, Hua S, Gong F, Fang M. Inhibition of autophagy with 3-methyladenine is protective in a lethal model of murine endotoxemia and polymicrobial sepsis. Innate Immun 2018; 24:231-239. [PMID: 29673286 PMCID: PMC6830927 DOI: 10.1177/1753425918771170] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Here, the regulatory role of autophagy is examined in both an LPS-induced lethal
endotoxic shock mouse model and cecal ligation and puncture (CLP) mouse model.
Autophagy-inhibitor 3-methyladenine (3-MA) and autophagy-enhancer rapamycin were
administrated to mice challenged with LPS or CLP. Animals challenged with LPS or
CLP combined with 3-MA displayed increased survival after endotoxemia, but LPS
combined with rapamycin worsened the endotoxic shock of the mice. Among the
different organs studied, the lungs and intestines exhibited significant
differences among LPS alone, LPS combined with 3-MA and LPS combined with
rapamycin. LPS combined with 3-MA attenuated the inflammatory damages of these
organs as compared with LPS alone. In contrast, LPS combined with rapamycin
increased damage in these organs. Consistently, serum inflammatory mediators
TNF-α and IL-6 were decreased by the treatment of LPS combined with 3-MA as
compared with LPS alone, while administration of LPS combined with rapamycin
increased the serum TNF-α and IL-6 levels. Similar results were found in mouse
bone marrow-derived macrophages exposed to LPS. Moreover, the regulatory effect
of autophagy to endotoxic shock is dependent on the TLR4 signaling pathway. Our
results demonstrate the central role of autophagy in the regulation of endotoxic
shock and its potential modulation for endotoxic shock treatment.
Collapse
Affiliation(s)
- Qirui Li
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Lingyun Li
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Xiaoyuan Fei
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Yuanyue Zhang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Chang Qi
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Shuyao Hua
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Feili Gong
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Min Fang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, China
| |
Collapse
|
22
|
Wang P, Shao BZ, Deng Z, Chen S, Yue Z, Miao CY. Autophagy in ischemic stroke. Prog Neurobiol 2018; 163-164:98-117. [DOI: 10.1016/j.pneurobio.2018.01.001] [Citation(s) in RCA: 183] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 12/04/2017] [Accepted: 01/10/2018] [Indexed: 02/07/2023]
|
23
|
Jia H, Yan Y, Liang Z, Tandra N, Zhang B, Wang J, Xu W, Qian H. Autophagy: A new treatment strategy for MSC-based therapy in acute kidney injury (Review). Mol Med Rep 2018; 17:3439-3447. [PMID: 29257336 DOI: 10.3892/mmr.2017.8311] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 11/09/2017] [Indexed: 11/09/2022] Open
Abstract
Acute kidney injury (AKI) is a common and serious medical condition associated with poor health outcomes. Autophagy is a conserved multistep pathway that serves a major role in many biological processes and diseases. Recent studies have demonstrated that autophagy is induced in proximal tubular cells during AKI. Autophagy serves a pro‑survival or pro‑death role under certain conditions. Furthermore, mesenchymal stem cells (MSCs) have therapeutic potential in the repair of renal injury. This review summarizes the recent progress on the role of autophagy in AKI and MSCs‑based therapy for AKI. Further research is expected to prevent and treat acute kidney injury.
Collapse
Affiliation(s)
- Haoyuan Jia
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Yongmin Yan
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Zhaofeng Liang
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Nitin Tandra
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Bin Zhang
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Juanjuan Wang
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Wenrong Xu
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Hui Qian
- Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| |
Collapse
|
24
|
Yu C, Li W, Liu J, Lu J, Feng J. Autophagy: novel applications of nonsteroidal anti-inflammatory drugs for primary cancer. Cancer Med 2018; 7:471-484. [PMID: 29282893 PMCID: PMC5806108 DOI: 10.1002/cam4.1287] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 11/15/2017] [Accepted: 11/19/2017] [Indexed: 12/17/2022] Open
Abstract
In eukaryotic cells, autophagy is a process associated with programmed cell death. During this process, cytoplasmic proteins and organelles are engulfed by double-membrane autophagosomes, which then fuse with lysosomes to form autolysosomes. These autolysosomes then degrade their contents to recycle the cellular components. Autophagy has been implicated in a wide variety of physiological and pathological processes that are closely related to tumorigenesis. In recent years, an increasing number of studies have indicated that nonsteroidal anti-inflammatory drugs, such as celecoxib, meloxicam, sulindac, aspirin, sildenafil, rofecoxib, and sodium salicylate, have diverse effects in cancer that are mediated by the autophagy pathway. These nonsteroidal anti-inflammatory drugs can modulate tumor autophagy through the PI3K/Akt/mTOR, MAPK/ERK1/2, P53/DRAM, AMPK/mTOR, Bip/GRP78, CHOP/ GADD153, and HGF/MET signaling pathways and inhibit lysosome function, leading to p53-dependent G1 cell-cycle arrest. In this review, we summarize the research progress in autophagy induced by nonsteroidal anti-inflammatory drugs and the molecular mechanisms of autophagy in cancer cells to provide a reference for the potential benefits of nonsteroidal anti-inflammatory drugs in cancer chemotherapy.
Collapse
Affiliation(s)
- Chen Yu
- Department of Integrated TCM & Western MedicineJiangsu Cancer HospitalJiangsu Institute of Cancer ResearchNanjing Medical University Affiliated Cancer HospitalNanjingJiang Su210000China
| | - Wei‐bing Li
- Department of Integrated TCM & Western MedicineJiangsu Cancer HospitalJiangsu Institute of Cancer ResearchNanjing Medical University Affiliated Cancer HospitalNanjingJiang Su210000China
| | - Jun‐bao Liu
- Department of Traditional Chinese MedicineHenan Provincial People's HospitalZhengzhouHenanChina
| | - Jian‐wei Lu
- Department of MedicineJiangsu Cancer HospitalJiangsu Institute of Cancer ResearchNanjing Medical University Affiliated Cancer HospitalNanjingJiang Su210000China
| | - Ji‐feng Feng
- Department of MedicineJiangsu Cancer HospitalJiangsu Institute of Cancer ResearchNanjing Medical University Affiliated Cancer HospitalNanjingJiang Su210000China
| |
Collapse
|
25
|
Dong W, Li Z, Chen Y, Zhang L, Ye Z, Liang H, Li R, Xu L, Zhang B, Liu S, Wang W, Li C, Shi W, Liang X. Necrostatin-1 attenuates sepsis-associated acute kidney injury by promoting autophagosome elimination in renal tubular epithelial cells. Mol Med Rep 2017; 17:3194-3199. [PMID: 29257238 DOI: 10.3892/mmr.2017.8214] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Accepted: 11/08/2017] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the protective effect of necrostatin‑1 (Nec‑1) in sepsis‑associated acute kidney injury (SA‑AKI). An SA‑AKI mouse model was established through an intraperitoneal injection of lipopolysaccharide (LPS), and Nec‑1 was administered to the mice prior to the establishment of SA‑AKI. Renal function and histological changes were evaluated, and the expression levels of microtubule‑associated protein light chain 3‑II (LC3‑II) and p62, as markers of autophagic flux, were detected. Autophagosomes and autolysosomes in renal tubular epithelial cells were also identified using electron microscopy. Pretreatment with Nec‑1 could attenuate the LPS‑induced increases in the concentrations of blood urea nitrogen (LPS+Nec‑1 vs. LPS group, 14.15±4.14 mmol/l vs. 32.54±5.46 mmol/l, respectively; P<0.001) and serum creatinine (11.50±1.67 µmol/l vs. 30.08±4.18 µmol/l, respectively; P<0.001). However, there were no significant differences in the rate of renal tubular epithelial cell necrosis between the groups. In the renal tissues of SA‑AKI mice, protein analysis showed that the LC3‑II and p62 proteins were increased, while a reverse transcription‑quantitative Reverse transcription‑polymerase chain reaction analysis detected no increase in LC3‑II or p62 mRNA. Additionally, a high number of autophagosomes, but not of autolysosomes, were observed by electron microscopy. When mice were pretreated with Nec‑1, the levels of LC3‑II and p62 decreased, and a large number of autolysosomes were observed by electron microscopy in the Nec‑1 pretreatment group. These results indicated that Nec‑1 improved autophagosome elimination, a process that is impaired by LPS, in renal tubular epithelial cells. This potentially enabled Nec‑1 to prevent SA‑AKI. Furthermore, the findings suggested that the protective effect of Nec‑1 may not have involved the inhibition of necroptosis, but may have occurred through the promotion of autophagosome elimination in renal tubular epithelial cells.
Collapse
Affiliation(s)
- Wei Dong
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Zhilian Li
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Yuanhan Chen
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Li Zhang
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Zhiming Ye
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Huaban Liang
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Ruizhao Li
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Lixia Xu
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Bin Zhang
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Shuangxin Liu
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Weidong Wang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Chunling Li
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Wei Shi
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Xinling Liang
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
26
|
Rao Z, Pan X, Zhang H, Sun J, Li J, Lu T, Gao M, Liu S, Yu D, Ding Z. Isoflurane Preconditioning Alleviated Murine Liver Ischemia and Reperfusion Injury by Restoring AMPK/mTOR-Mediated Autophagy. Anesth Analg 2017; 125:1355-1363. [PMID: 28857857 DOI: 10.1213/ane.0000000000002385] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Isoflurane has a pharmacological preconditioning effect against ischemia injury in the heart, kidney, and brain, but whether and how isoflurane preconditioning protects livers against ischemia and reperfusion (IR) injury is unclear. METHODS Mice were randomly divided into an isoflurane preconditioning (ISO) group and control group, receiving 1.5% isoflurane or carrier gas for 40 minutes, respectively (n = 8/group). A partial warm liver IR model was used, and liver injury was evaluated. Primary hepatocytes were pretreated with 1.5% isoflurane for 2 hours before the induction of cell death by hydrogen peroxide. Cell death and survival were evaluated with the lactate dehydrogenase and cell counting kit-8 assay. Autophagy and regulatory molecules in stressed livers and hepatocytes were analyzed by Western blot (n = 6/group). An autophagy inhibitor (3-methyladenine [3-MA]) and 5' adenosine monophosphate-activated protein kinase (AMPK) inhibitor (dorsomorphin) were administered in vivo (n = 8/group) and in vitro (n = 6/group). RESULTS Compared to that observed in the control group, mice in the ISO group showed reduced liver injury (alanine aminotransferase [ALT] levels, control versus ISO group, 8285 ± 769 vs 4896 ± 917 U/L, P < .001) and enhanced hepatocellular antiapoptosis in livers after IR. Furthermore, liver autophagy was restored by ISO as indicated by elevated LC3B II protein levels accompanied with increased p62 degradation. The in vitro study of primary hepatocytes also found that ISO effectively attenuated hepatocyte cell death induced by hydrogen peroxide. In addition, 3-MA pretreatment showed no significant influence in the control group, but abrogated the protective role of ISO both in stressed livers (ALT levels, phosphate-buffered saline + ISO versus 3-MA + ISO group, 5081 ± 294 vs 8663 ± 607 U/L, P < .001) and in hepatocytes. Finally, signaling pathway analysis demonstrated that AMPK was activated by ISO. Pretreatment with an AMPK inhibitor also abrogated liver protection by ISO (ALT levels, phosphate-buffered saline + ISO versus dorsomorphin [DOR] + ISO group, 5081 ± 294 vs 8710 ± 500 U/L, P < .001), with no significant effect in control mice. CONCLUSIONS Our results indicate that isoflurane preconditioning attenuates liver IR injury via AMPK/mTOR-mediated hepatocellular autophagy restoration. Our findings provide a novel potential therapeutic strategy for managing liver IR injury.
Collapse
Affiliation(s)
- Zhuqing Rao
- From the Departments of *Anesthesiology and †Liver Surgery, First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Zhang B, Lakshmanan J, Du Y, Smith JW, Harbrecht BG. Cell-specific regulation of iNOS by AMP-activated protein kinase in primary rat hepatocytes. J Surg Res 2017; 221:104-112. [PMID: 29229115 DOI: 10.1016/j.jss.2017.08.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 06/21/2017] [Accepted: 08/14/2017] [Indexed: 12/29/2022]
Abstract
BACKGROUND AMP-activated protein kinase (AMPK) regulates several metabolic pathways in hepatocytes that are critical to the hepatic response to sepsis and shock. Induction of nitric oxide synthesis is an important response to sepsis, inflammation and shock and many of the stimuli that upregulate inducible nitric oxide synthase (iNOS) also activate AMPK. AMPK inhibits nitric oxide (NO) production in skeletal and cardiac muscle cells, but the role of AMPK in regulating iNOS expression in hepatocytes has not been determined. MATERIALS AND METHODS Primary cultured rat hepatocytes were preincubated with an AMPK inhibitor, AMPK activators, or transfected with AMPK siRNA before being treated with the proinflammatory cytokines interleukin-1β (IL-1β) and interferon-γ (IFNγ). The hepatocyte cell lysate and culture supernatants were collected for Western blot analysis and Griess assay. RESULTS IL-1β and IFNγ markedly upregulated iNOS expression and AMPK phosphorylation. IL-1β + IFNγ-induced NO production and iNOS expression were significantly decreased in hepatocytes treated with the AMPK inhibitor compound C and AMPK knockdown by AMPK siRNA. Cytokine-induced iNOS expression was increased by AMPK activators 1-oxo-2-(2H-pyrrolium-1-yl)-1H-inden-3-olate, AMPK signaling activator III and AICA-riboside. Compound C upregulated Akt and c-Jun N-terminal kinase phosphorylation but decreased IκBα phosphorylation. AICA-riboside exerted opposite effects on these signaling pathways in hepatocytes. CONCLUSIONS In contrast to other cell types, AMPK increased IL-1β + IFNγ-induced NO production and iNOS expression through the Akt, c-Jun N-terminal kinase, and NF-κΒ signaling pathways in primary hepatocytes. These data suggest that AMPK-altering medications used clinically may have subsequent effects on iNOS expression and proinflammatory signaling pathways.
Collapse
Affiliation(s)
- Baochun Zhang
- Department of Surgery and Price Institute of Surgical Research, University of Louisville, Louisville, Kentucky.
| | - Jaganathan Lakshmanan
- Department of Surgery and Price Institute of Surgical Research, University of Louisville, Louisville, Kentucky
| | - Yibo Du
- Department of Surgery and Price Institute of Surgical Research, University of Louisville, Louisville, Kentucky
| | - Jason W Smith
- Department of Surgery and Price Institute of Surgical Research, University of Louisville, Louisville, Kentucky
| | - Brian G Harbrecht
- Department of Surgery and Price Institute of Surgical Research, University of Louisville, Louisville, Kentucky
| |
Collapse
|
28
|
Yu B, Ruan M, Liang T, Huang SW, Yu Y, Cheng HB, Shen XC. The Synergic Effect of Tetramethylpyrazine Phosphate and Borneol for Protecting Against Ischemia Injury in Cortex and Hippocampus Regions by Modulating Apoptosis and Autophagy. J Mol Neurosci 2017; 63:70-83. [PMID: 28779236 DOI: 10.1007/s12031-017-0958-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 07/26/2017] [Indexed: 01/07/2023]
Abstract
This study aimed to investigate the synergic effects of tetramethylpyrazine phosphate (TMPP) and borneol (BO) for protecting against ischemia in the cortex and hippocampus. A rat model of global cerebral ischemia-reperfusion (GCIR) was induced by four-vessel occlusion. The results showed that TMPP (13.3 mg/kg), BO (0.16 g/kg), and their combination improved the ultrastructure of neurons, reduced the apoptosis index, and reduced the intracellular calcium content in both the cortex and hippocampus. TMPP and the combined treatment increased cortex autophagy by modulating phosphorylated adenosine monophosphate-activated protein kinase (pAMPK) in the pAMPK-mammalian target of rapamycin (mTOR)-Unc-51-like kinase 1 (ULK1) signaling pathway, whereas BO only regulated ULK1. Moreover, BO increased neuron autophagy in the hippocampus by modulating mTOR, whereas TMPP targeted both mTOR and Beclin1. Similarly, the combination targeted both pAMPK and Beclin1. All three treatments decreased the expression of p53 and caspase-3 in the two areas. Additionally, TMPP and the combined therapy regulated Bax and Bcl-2. These results demonstrated the synergic effects between TMPP and BO for treating ischemia-reperfusion injury in the cortex and hippocampus regions. Their neuroprotective effects could be partly attributed to switching from apoptosis to protective autophagy. Additionally, the potential mechanism triggering this switching could be ascribed to the reduction of intracellular calcium content.
Collapse
Affiliation(s)
- Bin Yu
- Jiangsu Engineering Laboratory for Research and Industrialization of Empirical Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ming Ruan
- Jiangsu Provincial Key Construction Laboratory of Special Biomass Waste Resource Utilization, School of Food Science, Nanjing Xiaozhuang University, Nanjing, 211117, China
| | - Tao Liang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shi-Wen Huang
- Jiangsu Engineering Laboratory for Research and Industrialization of Empirical Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yun Yu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Hai-Bo Cheng
- Jiangsu Engineering Laboratory for Research and Industrialization of Empirical Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xiang-Chun Shen
- The Key Laboratory of Optimal Utilization of Natural Medicinal Resources, Guizhou Medical University, Huaxi College Station, Guian New District, Guiyang, 550025, China.
| |
Collapse
|
29
|
Lee SC, Kim KH, Kim OH, Lee SK, Hong HE, Won SS, Jeon SJ, Choi BJ, Jeong W, Kim SJ. Determination of optimized oxygen partial pressure to maximize the liver regenerative potential of the secretome obtained from adipose-derived stem cells. Stem Cell Res Ther 2017; 8:181. [PMID: 28774345 PMCID: PMC5543744 DOI: 10.1186/s13287-017-0635-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 07/11/2017] [Accepted: 07/20/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND A hypoxic-preconditioned secretome from stem cells reportedly promotes the functional and regenerative capacity of the liver more effectively than a control secretome. However, the optimum oxygen partial pressure (pO2) in the cell culture system that maximizes the therapeutic potential of the secretome has not yet been determined. METHODS We first determined the cellular alterations in adipose tissue-derived stem cells (ASCs) cultured under different pO2 (21%, 10%, 5%, and 1%). Subsequently, partially hepatectomized mice were injected with the secretome of ASCs cultured under different pO2, and then sera and liver specimens were obtained for analyses. RESULTS Of all AML12 cells cultured under different pO2, the AML12 cells cultured under 1% pO2 showed the highest mRNA expression of proliferation-associated markers (IL-6, HGF, and VEGF). In the cell proliferation assay, the AML12 cells cultured with the secretome of 1% pO2 showed the highest cell proliferation, followed by the cells cultured with the secretome of 21%, 10%, and 5% pO2, in that order. When injected into the partially hepatectomized mice, the 1% pO2 secretome most significantly increased the number of Ki67-positive cells, reduced serum levels of proinflammatory mediators (IL-6 and TNF-α), and reduced serum levels of liver transaminases. In addition, analysis of the liver specimens indicated that injection with the 1% pO2 secretome maximized the expression of the intermediate molecules of the PIP3/Akt and IL-6/STAT3 signaling pathways, all of which are known to promote liver regeneration. CONCLUSIONS The data of this study suggest that the secretome of ASCs cultured under 1% pO2 has the highest liver reparative and regenerative potential of all the secretomes tested here.
Collapse
Affiliation(s)
- Sang Chul Lee
- Department of Surgery, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Daeheung-dong 520-2, Joong-gu, Daejeon, Republic of Korea
| | - Kee-Hwan Kim
- Department of Surgery, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Uijeongbu, Republic of Korea
| | - Ok-Hee Kim
- Department of Surgery, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Daeheung-dong 520-2, Joong-gu, Daejeon, Republic of Korea
| | - Sang Kuon Lee
- Department of Surgery, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Daeheung-dong 520-2, Joong-gu, Daejeon, Republic of Korea
| | - Ha-Eun Hong
- Department of Surgery, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Daeheung-dong 520-2, Joong-gu, Daejeon, Republic of Korea
| | - Seong Su Won
- Department of Surgery, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Daeheung-dong 520-2, Joong-gu, Daejeon, Republic of Korea
| | - Sang-Jin Jeon
- Department of Surgery, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Daeheung-dong 520-2, Joong-gu, Daejeon, Republic of Korea
| | - Byung Jo Choi
- Department of Surgery, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Daeheung-dong 520-2, Joong-gu, Daejeon, Republic of Korea
| | - Wonjun Jeong
- Department of Surgery, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Daeheung-dong 520-2, Joong-gu, Daejeon, Republic of Korea
| | - Say-June Kim
- Department of Surgery, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Daeheung-dong 520-2, Joong-gu, Daejeon, Republic of Korea.
| |
Collapse
|
30
|
Zhou B, Leng Y, Lei SQ, Xia ZY. AMPK activation restores ischemic post-conditioning cardioprotection in STZ-induced type 1 diabetic rats: Role of autophagy. Mol Med Rep 2017; 16:3648-3656. [DOI: 10.3892/mmr.2017.7033] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 04/07/2017] [Indexed: 12/31/2022] Open
|
31
|
Pu T, Liao XH, Sun H, Guo H, Jiang X, Peng JB, Zhang L, Liu Q. Augmenter of liver regeneration regulates autophagy in renal ischemia–reperfusion injury via the AMPK/mTOR pathway. Apoptosis 2017; 22:955-969. [DOI: 10.1007/s10495-017-1370-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
32
|
Allouch S, Munusamy S. Metformin attenuates albumin-induced alterations in renal tubular cells in vitro. J Cell Physiol 2017; 232:3652-3663. [DOI: 10.1002/jcp.25838] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 02/01/2017] [Indexed: 12/14/2022]
|
33
|
Kasprowska D, Machnik G, Kost A, Gabryel B. Time-Dependent Changes in Apoptosis Upon Autophagy Inhibition in Astrocytes Exposed to Oxygen and Glucose Deprivation. Cell Mol Neurobiol 2017; 37:223-234. [PMID: 26983718 PMCID: PMC11482150 DOI: 10.1007/s10571-016-0363-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 03/08/2016] [Indexed: 12/19/2022]
Abstract
Recent studies have implicated the role of autophagy in brain ischemia pathophysiology. However, it remains unclear whether autophagy activation is protective or detrimental to astrocytes undergoing ischemic stress. This study evaluated the influence of ischemia-induced autophagy on cell death and the course of intrinsic and extrinsic apoptosis in primary cultures of rat cortical astrocytes exposed to combined oxygen-glucose deprivation (OGD). The role of autophagy was assessed by pharmacological inhibition with 3-methyladenine (3-MA). Cell viability was evaluated by measuring LDH release and through the use of the alamarBlue Assay. Apoptosis and necrosis were determined by fluorescence microscopy after Hoechst 33,342 and propidium iodide staining, respectively. The levels of apoptosis-related proteins were analyzed by immunoblotting. The downregulation of autophagy during OGD resulted in decreased cell viability and time-dependent changes in levels of apoptosis and necrosis. After short-term OGD (1, 4 h), cells treated with 3-MA showed higher level of cleaved caspase 3 compared with control cells. This result was consistent with an evaluation of apoptotic cell number by fluorescence microscopy. However, after prolonged exposure to OGD (8, 24 h), the number of apoptotic astrocytes (microscopically evaluated) did not differ or was even lower (as marked by caspase 3) in the presence of the autophagy inhibitor in comparison to the control. A higher level of necrosis was observed in 3-MA-treated cells compared to non-treated cells after 24 h OGD. The downregulation of autophagy caused time-dependent changes in both extrinsic (cleaved caspase 8, TNFα) and intrinsic (cleaved caspase 9) apoptotic pathways. Our results strongly indicate that the activation of autophagy in astrocytes undergoing ischemic stress is an adaptive mechanism, which allows for longer cell survival by delaying the initiation of apoptosis and necrosis.
Collapse
Affiliation(s)
- Daniela Kasprowska
- Department of Pharmacology, School of Medicine in Katowice, Medical University of Silesia, Medyków 18, 40-752, Katowice, Poland.
- Laboratory of Molecular Biology, Faculty of Physiotherapy, The Jerzy Kukuczka Academy of Physical Education, Mikołowska 72A, 40-065, Katowice, Poland.
| | - Grzegorz Machnik
- Department of Internal Medicine and Clinical Pharmacology, School of Medicine in Katowice, Medical University of Silesia, Medyków 18, 40-752, Katowice, Poland
| | - Alicja Kost
- Department of Histology, School of Medicine in Katowice, Medical University of Silesia, Medyków 18, 40-752, Katowice, Poland
| | - Bożena Gabryel
- Department of Pharmacology, School of Medicine in Katowice, Medical University of Silesia, Medyków 18, 40-752, Katowice, Poland
| |
Collapse
|
34
|
Liu Y, Lu Z, Cui M, Yang Q, Tang Y, Dong Q. Tissue kallikrein protects SH-SY5Y neuronal cells against oxygen and glucose deprivation-induced injury through bradykinin B2 receptor-dependent regulation of autophagy induction. J Neurochem 2016; 139:208-220. [PMID: 27248356 DOI: 10.1111/jnc.13690] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 05/21/2016] [Accepted: 05/24/2016] [Indexed: 12/22/2022]
Affiliation(s)
- Yanping Liu
- Department of Neurology; Huashan Hospital; State Key Laboratory of Medical Neurobiology; Fudan University; Shanghai China
| | - Zhengyu Lu
- Department of Neurology; Yueyang Hospital of Integrated Traditional Chinese and Western Medicine; Shanghai University of Traditional Chinese Medicine; Shanghai China
| | - Mei Cui
- Department of Neurology; Huashan Hospital; State Key Laboratory of Medical Neurobiology; Fudan University; Shanghai China
| | - Qi Yang
- Department of Neurology; Huashan Hospital; State Key Laboratory of Medical Neurobiology; Fudan University; Shanghai China
| | - Yuping Tang
- Department of Neurology; Huashan Hospital; State Key Laboratory of Medical Neurobiology; Fudan University; Shanghai China
| | - Qiang Dong
- Department of Neurology; Huashan Hospital; State Key Laboratory of Medical Neurobiology; Fudan University; Shanghai China
| |
Collapse
|
35
|
Lee SC, Kim KH, Kim OH, Lee SK, Kim SJ. Activation of Autophagy by Everolimus Confers Hepatoprotection Against Ischemia-Reperfusion Injury. Am J Transplant 2016; 16:2042-54. [PMID: 26814830 DOI: 10.1111/ajt.13729] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 12/16/2015] [Accepted: 01/08/2016] [Indexed: 01/25/2023]
Abstract
As the criteria for liver donation have been extended to include marginal donors, liver grafts are becoming particularly vulnerable to hepatic ischemia-reperfusion injury (IRI). However, no specific measures have been validated to ameliorate hepatic IRI. In this article, we explored whether everolimus has protective effects against hepatic IRI in relation with autophagy. The effects of everolimus were investigated in both in vitro and in vivo hepatic IRI models. Mouse hepatocyte AML12 cells and BALB/c mice were utilized for the establishment of each model. In the IRI-induced AML12 cells, everolimus treatment increased the expressions of autophagic markers (microtubule-associated protein 1 light chain 3 and p62) and decreased pro-apoptotic proteins (cleaved caspase 3 and cleaved poly-ADP ribose polymerase). The blockage of autophagy, using either bafilomycin A1 or si-autophagy-related protein 5, abrogated these anti-apoptosis effects of everolimus. Subsequently, everolimus administration to the hepatic IRI-induced mice provided hepatoprotective effects in terms of (1) decreasing the expressions of pro-apoptotic proteins, (2) inhibiting the release of pro-inflammatory cytokines (IL-6 and tumor necrosis factor-α), (3) reducing elevated liver enzymes (aspartate transaminase, alanine transaminase, and ammonia), and (4) restoring liver histopathology. These findings suggest that everolimus protects the liver against hepatic IRI by way of activating autophagy, and thus could be a potential therapeutic agent for hepatic IRI.
Collapse
Affiliation(s)
- S C Lee
- Department of Surgery, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Daejeon, Korea
| | - K H Kim
- Department of Surgery, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Uijeongbu, Korea
| | - O H Kim
- Department of Surgery, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Daejeon, Korea
| | - S K Lee
- Department of Surgery, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Daejeon, Korea
| | - S J Kim
- Department of Surgery, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Daejeon, Korea
| |
Collapse
|
36
|
Yang Y, Cheung HH, Law WN, Zhang C, Chan WY, Pei X, Wang Y. New Insights into the Role of Autophagy in Ovarian Cryopreservation by Vitrification. Biol Reprod 2016; 94:137. [PMID: 26911431 DOI: 10.1095/biolreprod.115.136374] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 02/03/2016] [Indexed: 12/13/2022] Open
Abstract
Ovarian cryopreservation by vitrification is a highly useful method for preserving female fertility during radiotherapy and chemotherapy. However, cryoinjury, osmotic stress during vitrification, and ischemia/reperfusion during transplantation lead to loss of ovarian follicles. Ovarian follicle loss may be partially reduced by several methods; however, studies regarding the mechanism of ovarian follicle loss have only investigated cell apoptosis, which consists of type I programmed cell death. Autophagy is type II programmed cell death, and cell homeostasis is maintained by autophagy during conditions of stress. The role of autophagy during cryopreservation by vitrification has rarely been reported. The potential role of autophagy during ovarian cryopreservation by vitrification is reviewed in this article.
Collapse
Affiliation(s)
- Yanzhou Yang
- Key Laboratory of Fertility Preservation and Maintenance, Ministry of Education, Key Laboratory of Reproduction and Genetics in Ningxia, Department of Histology and Embryology, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China The Chinese University of Hong Kong-Shandong University Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Hoi Hung Cheung
- The Chinese University of Hong Kong-Shandong University Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Wai Nok Law
- The Chinese University of Hong Kong-Shandong University Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Cheng Zhang
- College of Life Science, Capital Normal University, Beijing, People's Republic of China
| | - Wai Yee Chan
- The Chinese University of Hong Kong-Shandong University Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Xiuying Pei
- Key Laboratory of Fertility Preservation and Maintenance, Ministry of Education, Key Laboratory of Reproduction and Genetics in Ningxia, Department of Histology and Embryology, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Yanrong Wang
- Key Laboratory of Fertility Preservation and Maintenance, Ministry of Education, Key Laboratory of Reproduction and Genetics in Ningxia, Department of Histology and Embryology, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| |
Collapse
|
37
|
Lieberthal W, Tang M, Lusco M, Abate M, Levine JS. Preconditioning mice with activators of AMPK ameliorates ischemic acute kidney injury in vivo. Am J Physiol Renal Physiol 2016; 311:F731-F739. [PMID: 27252492 DOI: 10.1152/ajprenal.00541.2015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 04/01/2016] [Indexed: 12/25/2022] Open
Abstract
This study had two objectives: 1) to determine whether preconditioning cultured proximal tubular cells (PTCs) with pharmacological activators of AMP-activated protein kinase (AMPK) protects these cells from apoptosis induced by metabolic stress in vitro and 2) to assess the effects of preconditioning mice with these agents on the severity of ischemic acute renal kidney injury (AKI) in vivo. We demonstrate that preconditioning PTCs with 5-aminoimidazole-4-carboxamide-1-β-d-ribofuranoside (AICAR) or A-769662 reduces apoptosis of PTCs induced by subsequent stress. We also show that the reduction in cell death during metabolic stress associated with pretreatment by AMPK activators is associated with an increase in the cytosolic level of ATP, which is mediated by an increase in the rate of glycolysis. In addition, we provide evidence that the effect of AMPK activators on glycolysis is mediated, at least in part, by an increased uptake of glucose, and by the induction of hexokinase II (HK II) expression. Our data also show that the increased in HK II expression associated with preconditioning with AMPK activators is mediated by the activation (phosphorylation) of the cAMP-response element binding protein (CREB). We also provide entirely novel evidence that that A-79662 is substantially more effective than AICAR in mediating these alterations in PTCs in vitro. Finally, we demonstrate that preconditioning mice with AICAR or A-769662 substantially reduces the severity of renal dysfunction and tubular injury in a model of ischemic AKI in vivo and that the efficacy of AICAR and A-768662 in ameliorating ischemic AKI in vivo is comparable.
Collapse
Affiliation(s)
- Wilfred Lieberthal
- Section of Nephrology, Department of Medicine, Stony Brook University Medical Center, Stony Brook, New York; Section of Nephrology, Department of Medicine, Northport Veterans Affairs Hospital, Northport, New York;
| | - Meiyi Tang
- Section of Nephrology, Department of Medicine, Stony Brook University Medical Center, Stony Brook, New York
| | - Mark Lusco
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Mersema Abate
- Section of Nephrology, Department of Medicine, Stony Brook University Medical Center, Stony Brook, New York
| | - Jerrold S Levine
- Section of Nephrology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, Illinois; and Section of Nephrology, Department of Medicine, Jesse Brown Veterans Affairs Hospital, Chicago, Illinois
| |
Collapse
|
38
|
Jia Y, Zhao J, Liu M, Li B, Song Y, Li Y, Wen A, Shi L. Brazilin exerts protective effects against renal ischemia-reperfusion injury by inhibiting the NF-κB signaling pathway. Int J Mol Med 2016; 38:210-6. [PMID: 27247107 PMCID: PMC4899020 DOI: 10.3892/ijmm.2016.2616] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 05/19/2016] [Indexed: 01/28/2023] Open
Abstract
Renal ischemia-reperfusion (I/R) injury is associated with high morbidity and mortality as there is currently no available effective therapeutic strategy with which to treat this injury. Thus, the aim of this study was to investigate the potential protective effects of brazilin, a major active component of the Chinese medicine Caesalpinia sappan L., against renal I/R injury in vitro and in vivo. Rats were subjected to removal of the right kidney and I/R injury to the left kidney (ischemia for 45 min followed by reperfusion for 24 h). Treatment with brazilin (30 mg/kg, administered intravenously at 30 min prior to ischemia) led to the reversal of I/R-induced changes in serum creatinine (Scr) and blood urea nitrogen (BUN) levels, and also attenuated the histopathological damage induced by I/R. Furthermore, TUNEL assay revealed that brazilin reduced cell necrosis, and significantly decreased the expression of tumor necrosis factor (TNF)-α and interleukin (IL)-1β in renal tissue. Moreover, HK-2 cells were used in order to elucidate the mechanisms responsible for the protective effects of brazilin. The levels of phosphorylated IκBα and the nuclear translocation of nuclear factor-κB (NF-κB) were all evidently decreased by brazilin. These findings suggested that pre-treatment with brazilin protects against I/R-induced renal damage and suppresses the inflammatory response by inhibiting the activation of the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yanyan Jia
- Department of Pharmacy, General Hospital of Guangzhou Military Command of PLA, Guangzhou, Guangdong 510010, P.R. China
| | - Jinyi Zhao
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Meiyou Liu
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Bingling Li
- Department of Pharmacy, General Hospital of Guangzhou Military Command of PLA, Guangzhou, Guangdong 510010, P.R. China
| | - Ying Song
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Yuwen Li
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Aidong Wen
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Lei Shi
- Department of Pharmacy, General Hospital of Guangzhou Military Command of PLA, Guangzhou, Guangdong 510010, P.R. China
| |
Collapse
|
39
|
Ho J, Yu J, Wong SH, Zhang L, Liu X, Wong WT, Leung CCH, Choi G, Wang MHT, Gin T, Chan MTV, Wu WKK. Autophagy in sepsis: Degradation into exhaustion? Autophagy 2016; 12:1073-82. [PMID: 27172163 DOI: 10.1080/15548627.2016.1179410] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Autophagy is one of the innate immune defense mechanisms against microbial challenges. Previous in vitro and in vivo models of sepsis demonstrated that autophagy was activated initially in sepsis, followed by a subsequent phase of impairment. Autophagy modulation appears to be protective against multiple organ injuries in these murine sepsis models. This is achieved in part by preventing apoptosis, maintaining a balance between the productions of pro- and anti-inflammatory cytokines, and preserving mitochondrial functions. This article aims to discuss the role of autophagy in sepsis and the therapeutic potential of autophagy enhancers.
Collapse
Affiliation(s)
- Jeffery Ho
- a Department of Anesthesia and Intensive Care , The Chinese University of Hong Kong, Hong Kong Special Administrative Region , China
| | - Jun Yu
- b State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences and Department of Medicine and Therapeutics , The Chinese University of Hong Kong, Hong Kong Special Administrative Region , China
| | - Sunny H Wong
- b State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences and Department of Medicine and Therapeutics , The Chinese University of Hong Kong, Hong Kong Special Administrative Region , China
| | - Lin Zhang
- c School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region , China
| | - Xiaodong Liu
- a Department of Anesthesia and Intensive Care , The Chinese University of Hong Kong, Hong Kong Special Administrative Region , China
| | - Wai T Wong
- a Department of Anesthesia and Intensive Care , The Chinese University of Hong Kong, Hong Kong Special Administrative Region , China
| | - Czarina C H Leung
- a Department of Anesthesia and Intensive Care , The Chinese University of Hong Kong, Hong Kong Special Administrative Region , China
| | - Gordon Choi
- a Department of Anesthesia and Intensive Care , The Chinese University of Hong Kong, Hong Kong Special Administrative Region , China
| | - Maggie H T Wang
- d The Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong Special Administrative Region , China
| | - Tony Gin
- a Department of Anesthesia and Intensive Care , The Chinese University of Hong Kong, Hong Kong Special Administrative Region , China
| | - Matthew T V Chan
- a Department of Anesthesia and Intensive Care , The Chinese University of Hong Kong, Hong Kong Special Administrative Region , China
| | - William K K Wu
- a Department of Anesthesia and Intensive Care , The Chinese University of Hong Kong, Hong Kong Special Administrative Region , China.,b State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences and Department of Medicine and Therapeutics , The Chinese University of Hong Kong, Hong Kong Special Administrative Region , China
| |
Collapse
|
40
|
Heme oxygenase‑1 protects H2O2‑insulted glomerular mesangial cells from excessive autophagy. Mol Med Rep 2016; 13:5269-75. [PMID: 27122182 DOI: 10.3892/mmr.2016.5177] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 04/08/2016] [Indexed: 11/05/2022] Open
Abstract
Increasing evidence has demonstrated that the activation of heme oxygenase (HO)‑1 reduces autophagy stimulated by oxidative stress injury, in which the supraphysiological production of reactive oxygen species (ROS) is detected. However, the potential mechanism underlying this effect remains unclear. The present study aimed to investigate the function of HO‑1 activation in the regulation of autophagy in glomerular mesangial cells subjected to H2O2‑induced oxidative stress injury. The results demonstrated that the HO‑1 agonist, hemin, reduces the LC3 protein level, which was enhanced by H2O2 treatment. Furthermore, hemin‑activated HO‑1 may function as a regulator of oxidative stress‑induced autophagy in a dose‑dependent manner. Pharmacological activation of c‑Jun N‑terminal kinase (JNK) inhibited the effect of hemin, indicating that the JNK signaling pathway is associated with the mechanism of HO‑1 in impeding excessive autophagy. In addition to successfully alleviating H2O2‑induced oxidative stress and cellular apoptosis, hemin‑activated HO‑1 may provide cytoprotection against rapamycin, a specific autophagy agonist. The present result suggested the inhibitory action of HO‑1 in the avoidance of a potentially enhanced linkage between autophagy and apoptosis, particularly in the setting of excessive ROS. Therefore, enhancing the intracellular activity of HO‑1 may assist the crosstalk between oxidative stress, autophagy and apoptosis, and represent a novel therapeutic strategy for renal ischemic disease.
Collapse
|
41
|
Kaushal GP, Shah SV. Autophagy in acute kidney injury. Kidney Int 2016; 89:779-91. [PMID: 26924060 DOI: 10.1016/j.kint.2015.11.021] [Citation(s) in RCA: 304] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 11/12/2015] [Accepted: 11/19/2015] [Indexed: 02/09/2023]
Abstract
Autophagy is a conserved multistep pathway that degrades and recycles damaged organelles and macromolecules to maintain intracellular homeostasis. The autophagy pathway is upregulated under stress conditions including cell starvation, hypoxia, nutrient and growth-factor deprivation, endoplasmic reticulum stress, and oxidant injury, most of which are involved in the pathogenesis of acute kidney injury (AKI). Recent studies demonstrate that basal autophagy in the kidney is vital for the normal homeostasis of the proximal tubules. Deletion of key autophagy proteins impaired renal function and increased p62 levels and oxidative stress. In models of AKI, autophagy deletion in proximal tubules worsened tubular injury and renal function, highlighting that autophagy is renoprotective in models of AKI. In addition to nonselective sequestration of autophagic cargo, autophagy can facilitate selective degradation of damaged organelles, particularly mitochondrial degradation through the process of mitophagy. Damaged mitochondria accumulate in autophagy-deficient kidneys of mice subjected to ischemia-reperfusion injury, but the precise mechanisms of regulation of mitophagy in AKI are not yet elucidated. Recent progress in identifying the interplay of autophagy, apoptosis, and regulated necrosis has revived interest in examining shared pathways/molecules in this crosstalk during the pathogenesis of AKI. Autophagy and its associated pathways pose potentially unique targets for therapeutic interventions in AKI.
Collapse
Affiliation(s)
- Gur P Kaushal
- Renal Section, Medicine Service, Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA; Division of Nephrology, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.
| | - Sudhir V Shah
- Renal Section, Medicine Service, Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA; Division of Nephrology, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
42
|
Zhang L, Ding K, Wang H, Wu Y, Xu J. Traumatic Brain Injury-Induced Neuronal Apoptosis is Reduced Through Modulation of PI3K and Autophagy Pathways in Mouse by FTY720. Cell Mol Neurobiol 2016; 36:131-42. [PMID: 26099903 PMCID: PMC11482378 DOI: 10.1007/s10571-015-0227-1] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 06/12/2015] [Indexed: 01/13/2023]
Abstract
FTY720 is a synthetic compound produced by modification of metabolite from Isaria sinclairii. It is a novel type of immunosuppressive agent inhibiting lymphocyte egress from secondary lymphoid tissues, thereby causing peripheral lymphopenia. Growing evidences have suggested that apoptosis and autophagy were involved in the secondary brain injury after traumatic brain injury (TBI) although FTY720 exerted neuroprotective effects in a variety of neurological diseases except TBI. The present study was aimed to investigate the role of FTY720 in a mouse model of TBI. In experiment 1, ICR mice were divided into four groups: sham group, TBI group, TBI + vehicle group, and TBI + FTY720 group. And the injured cerebral cortex (including both contused and penumbra) was used for analysis. We found that FTY720 administration after TBI improved neurobehavioral function, alleviated brain edema, accompanied by modulation of apoptotic indicators such as Bcl-2, Bcl-xL, Bax, and cytochrome c. In experiment 2, ICR mice were also divided into four groups: sham group, TBI + vehicle group, TBI + FTY720 group, and TBI + FTY720 + inhibitors group. And the injured cerebral cortex (including both contused and penumbra) was used for analysis. We found that FTY720 increased the expression of phospho-protein kinase B (AKT) and some autophagy markers such as LC3 and Beclin 1. In addition, the apoptosis inhibition effect of FTY720 was partly abrogated by the phosphatidylinositide 3-kinases (PI3K)/AKT pathway inhibitor LY294002 and autophagy inhibitor 3-methyladenine. Collectively, our data provide the first evidence that FTY720 exerted neuroprotective effects after TBI, at least in part, through the activation of PI3K/AKT pathway and autophagy.
Collapse
Affiliation(s)
- Li Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Ke Ding
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Handong Wang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China.
| | - Yong Wu
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Jianguo Xu
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| |
Collapse
|
43
|
Yang S, Long L, Li D, Zhang J, Jin S, Wang F, Chen J. β-Guanidinopropionic acid extends the lifespan of Drosophila melanogaster via an AMP-activated protein kinase-dependent increase in autophagy. Aging Cell 2015; 14:1024-33. [PMID: 26120775 PMCID: PMC4693457 DOI: 10.1111/acel.12371] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2015] [Indexed: 01/01/2023] Open
Abstract
Previous studies have demonstrated that AMP‐activated protein kinase (AMPK) controls autophagy through the mammalian target of rapamycin (mTOR) and Unc‐51 like kinase 1 (ULK1/Atg1) signaling, which augments the quality of cellular housekeeping, and that β‐guanidinopropionic acid (β‐GPA), a creatine analog, leads to a chronic activation of AMPK. However, the relationship between β‐GPA and aging remains elusive. In this study, we hypothesized that feeding β‐GPA to adult Drosophila produces the lifespan extension via activation of AMPK‐dependent autophagy. It was found that dietary administration of β‐GPA at a concentration higher than 900 mm induced a significant extension of the lifespan of Drosophila melanogaster in repeated experiments. Furthermore, we found that Atg8 protein, the homolog of microtubule‐associated protein 1A/1B‐light chain 3 (LC3) and a biomarker of autophagy in Drosophila, was significantly upregulated by β‐GPA treatment, indicating that autophagic activity plays a role in the effect of β‐GPA. On the other hand, when the expression of Atg5 protein, an essential protein for autophagy, was reduced by RNA interference (RNAi), the effect of β‐GPA on lifespan extension was abolished. Moreover, we found that AMPK was also involved in this process. β‐GPA treatment significantly elevated the expression of phospho‐T172‐AMPK levels, while inhibition of AMPK by either AMPK‐RNAi or compound C significantly attenuated the expression of autophagy‐related proteins and lifespan extension in Drosophila. Taken together, our results suggest that β‐GPA can induce an extension of the lifespan of Drosophila via AMPK‐Atg1‐autophagy signaling pathway.
Collapse
Affiliation(s)
- Si Yang
- Department of Pharmacology School of Basic Medicine Tongji Medical College Huazhong University of Science and Technology Wuhan 430030 China
| | - Li‐Hong Long
- Department of Pharmacology School of Basic Medicine Tongji Medical College Huazhong University of Science and Technology Wuhan 430030 China
- The Key Laboratory of Neurological Diseases (HUST) Ministry of Education of China Wuhan 430030 China
- Hubei Key Laboratory of Drug Target Researches and Pharmacodynamic Evaluation (HUST) Wuhan 430030 China
- The Laboratory of Neuropsychiatric Diseases The Institute of Brain Research Huazhong University of Science and Technology Wuhan 430030 China
| | - Di Li
- Department of Pharmacology School of Basic Medicine Tongji Medical College Huazhong University of Science and Technology Wuhan 430030 China
| | - Jian‐Kang Zhang
- Department of Pharmacology School of Basic Medicine Tongji Medical College Huazhong University of Science and Technology Wuhan 430030 China
| | - Shan Jin
- College of Life Science Hubei University Wuhan 430062 China
| | - Fang Wang
- Department of Pharmacology School of Basic Medicine Tongji Medical College Huazhong University of Science and Technology Wuhan 430030 China
- The Key Laboratory of Neurological Diseases (HUST) Ministry of Education of China Wuhan 430030 China
- Hubei Key Laboratory of Drug Target Researches and Pharmacodynamic Evaluation (HUST) Wuhan 430030 China
- The Laboratory of Neuropsychiatric Diseases The Institute of Brain Research Huazhong University of Science and Technology Wuhan 430030 China
| | - Jian‐Guo Chen
- Department of Pharmacology School of Basic Medicine Tongji Medical College Huazhong University of Science and Technology Wuhan 430030 China
- The Key Laboratory of Neurological Diseases (HUST) Ministry of Education of China Wuhan 430030 China
- Hubei Key Laboratory of Drug Target Researches and Pharmacodynamic Evaluation (HUST) Wuhan 430030 China
- The Laboratory of Neuropsychiatric Diseases The Institute of Brain Research Huazhong University of Science and Technology Wuhan 430030 China
| |
Collapse
|
44
|
Hu S, Zhang Y, Zhang M, Guo Y, Yang P, Zhang S, Simsekyilmaz S, Xu JF, Li J, Xiang X, Yu Q, Wang CY. Aloperine Protects Mice against Ischemia-Reperfusion (IR)-Induced Renal Injury by Regulating PI3K/AKT/mTOR Signaling and AP-1 Activity. Mol Med 2015; 21:912-923. [PMID: 26552059 DOI: 10.2119/molmed.2015.00056] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 10/27/2015] [Indexed: 01/03/2023] Open
Abstract
Aloperine is a quinolizidine alkaloid extracted from the leaves of Sophora plants. It has been recognized with the potential to treat inflammatory and allergic diseases as well as tumors. In this report, we demonstrate that pretreatment with aloperine provided protection for mice against ischemia-reperfusion (IR)-induced acute renal injury as manifested by the attenuated inflammatory infiltration, reduced tubular apoptosis, and well-preserved renal function. Mechanistic studies revealed that aloperine selectively repressed IL-1β and IFN-γ expression by regulating PI3K/Akt/mTOR signaling and NF-κB transcriptional activity. However, aloperine did not show a perceptible impact on IL-6 and TGF-β expression and the related Jak2/Stat3 signaling. It was also noted that aloperine regulates AP-1 activity, through which it not only enhances SOD expression to increase reactive oxygen species (ROS) detoxification but also promotes the expression of antiapoptotic Bcl-2, thereby preventing tubular cells from IR-induced apoptosis. Collectively, our data suggest that administration of aloperine prior to IR insults, such as renal transplantation, could be a viable approach to prevent IR-induced injuries.
Collapse
Affiliation(s)
- Shuang Hu
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuxing Zhang
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meng Zhang
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanchao Guo
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Yang
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shu Zhang
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sakine Simsekyilmaz
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun-Fa Xu
- Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Medical College, Dongguan, China
| | - Jinxiu Li
- Department of Emergency Medicine, Institute of Emergency Medicine and Rare Diseases, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Xudong Xiang
- Department of Emergency Medicine, Institute of Emergency Medicine and Rare Diseases, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Qilin Yu
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cong-Yi Wang
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Medical College, Dongguan, China.,Department of Emergency Medicine, Institute of Emergency Medicine and Rare Diseases, the Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
45
|
Zhong J, Dong X, Xiu P, Wang F, Liu J, Wei H, Xu Z, Liu F, Li T, Li J. Blocking autophagy enhances meloxicam lethality to hepatocellular carcinoma by promotion of endoplasmic reticulum stress. Cell Prolif 2015; 48:691-704. [PMID: 26481188 DOI: 10.1111/cpr.12221] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 08/01/2015] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES Meloxicam, a selective cyclooxygenase-2 (COX-2) inhibitor, has been demonstrated to exert anti-tumour effects against various malignancies. However, up to now, mechanisms involved in meloxicam anti-hepatocellular carcinoma effects have remained unclear. MATERIALS AND METHODS Cell viability and apoptosis were assessed by CCK-8 and flow cytometry. Endoplasmic reticulum (ER) stress and autophagy-associated molecules were analysed by western blotting and immunofluorescence assay. GRP78 and Atg5 knock-down by siRNA or chemical inhibition was used to investigate cytotoxic effects of meloxicam treatment on HCC cells. RESULTS We found that meloxicam led to apoptosis and autophagy in HepG2 and Bel-7402 cells via a mechanism that involved ER stress. Up-regulation of GRP78 signalling pathway from meloxicam-induced ER stress was critical for activation of autophagy. Furthermore, autophagy activation attenuated ER stress-related cell death. Blocking autophagy by 3-methyladenine (3-MA) or Atg5 siRNA knock-down enhanced meloxicam lethality for HCC by activation of ER stress-related apoptosis. In addition, GRP78 seemed to lead to autophagic activation via the AMPK-mTOR signalling pathway. Blocking AMPK with a chemical inhibitor inhibited autophagy suggesting that meloxicam-regulated autophagy requires activation of AMPK. CONCLUSIONS Our results revealed that both ER stress and autophagy were involved in cell death evoked by meloxicam in HCC cells. This inhibition of autophagy to enhance meloxicam lethality, suggests a novel therapeutic strategy against HCC.
Collapse
Affiliation(s)
- Jingtao Zhong
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, China
| | - Xiaofeng Dong
- Department of Hepatobiliary Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, China
| | - Peng Xiu
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, China
| | - Fuhai Wang
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, China
| | - Ju Liu
- Laboratory of Medicrovascular Medicine and Medical Research Center, Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, China
| | - Honglong Wei
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, China
| | - Zongzhen Xu
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, China
| | - Feng Liu
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, China
| | - Tao Li
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, China
| | - Jie Li
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, China
| |
Collapse
|
46
|
Lee SH, Kim JS, Ravichandran K, Gil HW, Song HY, Hong SY. P-Glycoprotein Induction Ameliorates Colistin Induced Nephrotoxicity in Cultured Human Proximal Tubular Cells. PLoS One 2015; 10:e0136075. [PMID: 26287374 PMCID: PMC4545883 DOI: 10.1371/journal.pone.0136075] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 07/22/2015] [Indexed: 01/08/2023] Open
Abstract
The pathogenesis of colistin induced nephrotoxicity is poorly understood. Currently there are no effective therapeutic or prophylactic agents available. This study was aimed to determine the mechanism of colistin induced nephrotoxicity and to determine whether P-glycoprotein (P-gp) induction could prevent colistin induced nephrotoxicity. Colistin induced cell toxicity in cultured human proximal tubular cells in both dose and time dependent manner. Colistin provoked ROS in a dose dependent manner as measured by DCF-DA. To investigate apoptosis, caspase 3/7 activity was determined. Caspase 3/7 activity was increased dose dependently (25, 50, 100 μg/ml) at 6 h. Autophagosome formation was assessed by measuring LC3- II/LC3-I ratio. The ratio of LC3-II to LC3- I was increased at 2 h (25 μg/ml). Suppression of autophagosome formation increased colistin induced nephrotoxicity. The expression of P-gp and the cell toxicity was determined in colistin with or without dexamethasone (P-gp inducer) and verapamil (selective P-gp inhibitor). Colistin itself suppressed the expression of P-gp. P-gp expression and activity decreased colistin induced nephrotoxicity with dexamethasone treatment. In addition induced P-gp transporter was shown to improve the efflux effect on colistin treated HK2 cell line, which was demonstrated by calcein-AM fluorescence accumulation assay. The increased activity could be blocked by N-acetylcysteine. In conclusion, colistin induces nephrotoxicity by suppressing P-gp. Induction of P-gp could ameliorate colistin induced nephrotoxicity by decreasing apoptosis.
Collapse
Affiliation(s)
- Sun-hyo Lee
- Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Republic of Korea
| | - Jin-sun Kim
- Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Republic of Korea
| | - Kameswaran Ravichandran
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Hyo-Wook Gil
- Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Republic of Korea
- * E-mail:
| | - Ho-yeon Song
- Department of Microbiology, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea
| | - Sae-yong Hong
- Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Republic of Korea
| |
Collapse
|
47
|
The Role of Cardiolipin in Cardiovascular Health. BIOMED RESEARCH INTERNATIONAL 2015; 2015:891707. [PMID: 26301254 PMCID: PMC4537736 DOI: 10.1155/2015/891707] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 07/08/2015] [Indexed: 12/20/2022]
Abstract
Cardiolipin (CL), the signature phospholipid of mitochondrial membranes, is crucial for both mitochondrial function and cellular processes outside of the mitochondria. The importance of CL in cardiovascular health is underscored by the life-threatening genetic disorder Barth syndrome (BTHS), which manifests clinically as cardiomyopathy, skeletal myopathy, neutropenia, and growth retardation. BTHS is caused by mutations in the gene encoding tafazzin, the transacylase that carries out the second CL remodeling step. In addition to BTHS, CL is linked to other cardiovascular diseases (CVDs), including cardiomyopathy, atherosclerosis, myocardial ischemia-reperfusion injury, heart failure, and Tangier disease. The link between CL and CVD may possibly be explained by the physiological roles of CL in pathways that are cardioprotective, including mitochondrial bioenergetics, autophagy/mitophagy, and mitogen activated protein kinase (MAPK) pathways. In this review, we focus on the role of CL in the pathogenesis of CVD as well as the molecular mechanisms that may link CL functions to cardiovascular health.
Collapse
|
48
|
Hadj Ayed Tka K, Mahfoudh Boussaid A, Zaouali MA, Kammoun R, Bejaoui M, Ghoul Mazgar S, Rosello Catafau J, Ben Abdennebi H. Melatonin modulates endoplasmic reticulum stress and Akt/GSK3-beta signaling pathway in a rat model of renal warm ischemia reperfusion. Anal Cell Pathol (Amst) 2015; 2015:635172. [PMID: 26229743 PMCID: PMC4502281 DOI: 10.1155/2015/635172] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 05/28/2015] [Indexed: 12/17/2022] Open
Abstract
Melatonin (Mel) is widely used to attenuate ischemia/reperfusion (I/R) injury in several organs. Nevertheless, the underlying mechanisms remain unclear. This study was conducted to explore the effect of Mel on endoplasmic reticulum (ER) stress, Akt and MAPK cascades after renal warm I/R. Eighteen Wistar rats were randomized into three groups: Sham, I/R, and Mel + I/R. The ischemia period was 60 min followed by 120 min of reperfusion. Mel (10 mg/kg) was administrated 30 min prior to ischemia. The creatinine clearance, MDA, LDH levels, and histopathological changes were evaluated. In addition, Western blot was performed to study ER stress and its downstream apoptosis as well as phosphorylation of Akt, GSK-3β, VDAC, ERK, and P38. Mel decreased cytolysis and lipid peroxidation and improved renal function and morphology compared to I/R group. Parallely, it significantly reduced the ER stress parameters including GRP 78, p-PERK, XBP 1, ATF 6, CHOP, and JNK. Simultaneously, p-Akt level was significantly enhanced and its target molecules GSK-3β and VDAC were inhibited. Furthermore, the ERK and P38 phosphorylation were evidently augmented after Mel administration in comparison to I/R group. In conclusion, Mel improves the recovery of renal function by decreasing ER stress and stimulating Akt pathway after renal I/R injury.
Collapse
Affiliation(s)
- Kaouther Hadj Ayed Tka
- Unit of Molecular Biology and Anthropology Applied to Development and Health (UR12ES11), Faculty of Pharmacy, University of Monastir, rue Avicenne, 5000 Monastir, Tunisia
| | - Asma Mahfoudh Boussaid
- Unit of Molecular Biology and Anthropology Applied to Development and Health (UR12ES11), Faculty of Pharmacy, University of Monastir, rue Avicenne, 5000 Monastir, Tunisia
| | - Mohamed Amine Zaouali
- Unit of Molecular Biology and Anthropology Applied to Development and Health (UR12ES11), Faculty of Pharmacy, University of Monastir, rue Avicenne, 5000 Monastir, Tunisia
| | - Rym Kammoun
- Laboratory of Histology and Embryology, Faculty of Dental Medicine, University of Monastir, rue Avicenne, 5000 Monastir, Tunisia
| | - Mohamed Bejaoui
- Unit of Experimental Hepatic Ischemia-Reperfusion, Institute of Biomedical Investigations, Higher Council of Scientific Investigations, 08036 Barcelona, Spain
| | - Sonia Ghoul Mazgar
- Laboratory of Histology and Embryology, Faculty of Dental Medicine, University of Monastir, rue Avicenne, 5000 Monastir, Tunisia
| | - Joan Rosello Catafau
- Unit of Experimental Hepatic Ischemia-Reperfusion, Institute of Biomedical Investigations, Higher Council of Scientific Investigations, 08036 Barcelona, Spain
| | - Hassen Ben Abdennebi
- Unit of Molecular Biology and Anthropology Applied to Development and Health (UR12ES11), Faculty of Pharmacy, University of Monastir, rue Avicenne, 5000 Monastir, Tunisia
| |
Collapse
|
49
|
Wang Y, Reis C, Applegate R, Stier G, Martin R, Zhang JH. Ischemic conditioning-induced endogenous brain protection: Applications pre-, per- or post-stroke. Exp Neurol 2015; 272:26-40. [PMID: 25900056 DOI: 10.1016/j.expneurol.2015.04.009] [Citation(s) in RCA: 309] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 04/06/2015] [Accepted: 04/11/2015] [Indexed: 11/17/2022]
Abstract
In the area of brain injury and neurodegenerative diseases, a plethora of experimental and clinical evidence strongly indicates the promise of therapeutically exploiting the endogenous adaptive system at various levels like triggers, mediators and the end-effectors to stimulate and mobilize intrinsic protective capacities against brain injuries. It is believed that ischemic pre-conditioning and post-conditioning are actually the strongest known interventions to stimulate the innate neuroprotective mechanism to prevent or reverse neurodegenerative diseases including stroke and traumatic brain injury. Recently, studies showed the effectiveness of ischemic per-conditioning in some organs. Therefore the term ischemic conditioning, including all interventions applied pre-, per- and post-ischemia, which spans therapeutic windows in 3 time periods, has recently been broadly accepted by scientific communities. In addition, it is extensively acknowledged that ischemia-mediated protection not only affects the neurons but also all the components of the neurovascular network (consisting of neurons, glial cells, vascular endothelial cells, pericytes, smooth muscle cells, and venule/veins). The concept of cerebroprotection has been widely used in place of neuroprotection. Intensive studies on the cellular signaling pathways involved in ischemic conditioning have improved the mechanistic understanding of tolerance to cerebral ischemia. This has added impetus to exploration for potential pharmacologic mimetics, which could possibly induce and maximize inherent protective capacities. However, most of these studies were performed in rodents, and the efficacy of these mimetics remains to be evaluated in human patients. Several classical signaling pathways involving apoptosis, inflammation, or oxidation have been elaborated in the past decades. Newly characterized mechanisms are emerging with the advances in biotechnology and conceptual renewal. In this review we are going to focus on those recently reported methodological and mechanistic discoveries in the realm of ischemic conditioning. Due to the varied time differences of ischemic conditioning in different animal models and clinical trials, it is important to define optimal timing to achieve the best conditioning induced neuroprotection. This brings not only an opportunity in the treatment of stroke, but challenges as well, as data is just becoming available and the procedures are not yet optimized. The purpose of this review is to shed light on exploiting these ischemic conditioning modalities to protect the cerebrovascular system against diverse injuries and neurodegenerative disorders.
Collapse
Affiliation(s)
- Yuechun Wang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, USA; Department of Physiology, Jinan University School of Medicine, Guangzhou, China
| | - Cesar Reis
- Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA, USA
| | - Richard Applegate
- Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA, USA
| | - Gary Stier
- Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA, USA
| | - Robert Martin
- Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, USA; Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, USA; Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA, USA.
| |
Collapse
|
50
|
Zhang XY, Zhang TT, Song DD, Zhou JH, Han R, Qin ZH, Sheng R. Endoplasmic reticulum chaperone GRP78 is involved in autophagy activation induced by ischemic preconditioning in neural cells. Mol Brain 2015; 8:20. [PMID: 25885223 PMCID: PMC4381498 DOI: 10.1186/s13041-015-0112-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 03/15/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Our previous finding showed that brain ischemic preconditioning mediates neuroprotection through endoplasmic reticulum (ER) stress-induced autophagy. This study was aimed at exploring the role of ER chaperone GRP78 in IPC induced autophagy activation in neural cells. RESULTS Ischemic preconditioning (IPC) and oxygen glucose deprivation (OGD) models were established in rat pheochromocytoma (PC12) cells and primary cultured murine cortical neurons. IPC exerted neuroprotection against subsequent OGD injury in both PC12 cells and primary cortical neurons. IPC increased GRP78 expression and activated autophagy, as evidenced by upregulated LC3 and Beclin1, increased autophagic flux and formation of autophagosomes. BAPTA(dibromo-1,2-bis(aminophenoxy)ethane N,N,N9,N9 - tetra acetic acid, 0.125-2 μM) and small interfering RNA targeted GRP78 abrogated IPC induced neuroprotection and decreased the expression of GRP78, LC3II/LC3I and Beclin1. In contrast, lentiviral vector mediated GRP78 overexpression (LV-GRP78) strengthened resistance of PC12 cells to OGD injury and increased LC3 and Beclin1 expression. Moreover, knockdown of GRP78 in stable GRP78 overexpressing PC12 cells abolished the upregulation of LC3II/LC3I. GRP78 might activate autophagy through AMPK - mTOR pathway. CONCLUSION These results suggest that IPC- induced GRP78 upregulation is involved in autophagy activation, and hence exerts protection against ischemic injury in neural cells.
Collapse
Affiliation(s)
- Xiang-Yang Zhang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Soochow University School of Pharmaceutical Science, 199 Ren Ai Road, Suzhou, 215123, China.
| | - Tong-Tong Zhang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Soochow University School of Pharmaceutical Science, 199 Ren Ai Road, Suzhou, 215123, China.
| | - Dan-Dan Song
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Soochow University School of Pharmaceutical Science, 199 Ren Ai Road, Suzhou, 215123, China.
| | - Jun- Hao Zhou
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Soochow University School of Pharmaceutical Science, 199 Ren Ai Road, Suzhou, 215123, China.
| | - Rong Han
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Soochow University School of Pharmaceutical Science, 199 Ren Ai Road, Suzhou, 215123, China.
| | - Zheng-Hong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Soochow University School of Pharmaceutical Science, 199 Ren Ai Road, Suzhou, 215123, China.
| | - Rui Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Soochow University School of Pharmaceutical Science, 199 Ren Ai Road, Suzhou, 215123, China.
| |
Collapse
|