1
|
Yang ZJ, Zhang WF, Jin QQ, Wu ZR, Du YY, Shi H, Qu ZS, Han XJ, Jiang LP. Lactate Contributes to Remote Ischemic Preconditioning-Mediated Protection Against Myocardial Ischemia Reperfusion Injury by Facilitating Autophagy via the AMP-Activated Protein Kinase-Mammalian Target of Rapamycin-Transcription Factor EB-Connexin 43 Axis. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1857-1878. [PMID: 39069170 DOI: 10.1016/j.ajpath.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/06/2024] [Accepted: 07/09/2024] [Indexed: 07/30/2024]
Abstract
Remote ischemic preconditioning (RIPC) exerts a protective role on myocardial ischemia/reperfusion (I/R) injury by the release of various humoral factors. Lactate is a common metabolite in ischemic tissues. Nevertheless, little is known about the role lactate plays in myocardial I/R injury and its underlying mechanism. This investigation revealed that RIPC elevated the level of lactate in blood and myocardium. Furthermore, AZD3965, a selective monocarboxylate transporter 1 inhibitor, and 2-deoxy-d-glucose, a glycolysis inhibitor, mitigated the effects of RIPC-induced elevated lactate in the myocardium and prevented RIPC against myocardial I/R injury. In an in vitro hypoxia/reoxygenation model, lactate markedly mitigated hypoxia/reoxygenation-induced cell damage in H9c2 cells. Further studies suggested that lactate contributed to RIPC, rescuing I/R-induced autophagy deficiency by promoting transcription factor EB (TFEB) translocation to the nucleus through activating the AMP-activated protein kinase (AMPK)-mammalian target of rapamycin (mTOR) pathway without influencing the phosphatidylinositol 3-kinase-Akt pathway, thus reducing cardiomyocyte damage. Interestingly, lactate up-regulated the mRNA and protein expression of connexin 43 (CX43) by facilitating the binding of TFEB to CX43 promoter in the myocardium. Functionally, silencing of TFEB attenuated the protective effect of lactate on cell damage, which was reversed by overexpression of CX43. Further mechanistic studies suggested that lactate facilitated CX43-regulated autophagy via the AMPK-mTOR-TFEB signaling pathway. Collectively, this research demonstrates that RIPC protects against myocardial I/R injury through lactate-mediated myocardial autophagy via the AMPK-mTOR-TFEB-CX43 axis.
Collapse
Affiliation(s)
- Zhang-Jian Yang
- Jiangxi Provincial Key Laboratory of Drug Targets and Drug Screening, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, China; Department of Pharmacy, 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Wei-Fang Zhang
- Department of Pharmacy, 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Qing-Qing Jin
- Jiangxi Provincial Key Laboratory of Drug Targets and Drug Screening, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Zhi-Rong Wu
- Jiangxi Provincial Key Laboratory of Drug Targets and Drug Screening, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yun-Yan Du
- Jiangxi Provincial Key Laboratory of Drug Targets and Drug Screening, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Hao Shi
- Jiangxi Provincial Key Laboratory of Drug Targets and Drug Screening, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Zhen-Sheng Qu
- Jiangxi Provincial Key Laboratory of Drug Targets and Drug Screening, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xiao-Jian Han
- Institute of Geriatrics, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China.
| | - Li-Ping Jiang
- Jiangxi Provincial Key Laboratory of Drug Targets and Drug Screening, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, China.
| |
Collapse
|
2
|
Koutsifeli P, Daniels LJ, Neale J, Fong S, Varma U, Annandale M, Li X, Nursalim Y, Bell JR, Weeks KL, Stotland A, Taylor DJ, Gottlieb RA, Delbridge LMD, Mellor KM. Methods for detection of cardiac glycogen-autophagy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.11.607511. [PMID: 39211188 PMCID: PMC11361060 DOI: 10.1101/2024.08.11.607511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Glycogen-autophagy ('glycophagy') is a selective autophagy process involved in delivering glycogen to the lysosome for bulk degradation. Glycophagy protein intermediaries include STBD1 as a glycogen tagging receptor, delivering the glycogen cargo into the forming phagosome by partnering with the Atg8 homolog, GABARAPL1. Glycophagy is emerging as a key process of energy metabolism and development of reliable tools for assessment of glycophagy activity is an important priority. Here we show that antibodies raised against the N-terminus of the GABARAPL1 protein (but not the full-length protein) detected a specific endogenous GABARAPL1 immunoblot band at 18kDa. A stable GFP-GABARAPL1 cardiac cell line was used to quantify GABARAPL1 lysosomal flux via measurement of GFP puncta in response to lysosomal inhibition with bafilomycin. Endogenous glycophagy flux was quantified in primary rat ventricular myocytes by the extent of glycogen accumulation with bafilomycin combined with chloroquine treatment (no effect observed with bafilomycin or chloroquine alone). In wild-type isolated mouse hearts, bafilomycin alone and bafilomycin combined with chloroquine (but not chloroquine alone) elicited a significant increase in glycogen content signifying basal glycophagy flux. Collectively, these methodologies provide a comprehensive toolbox for tracking cardiac glycophagy activity to advance research into the role of glycophagy in health and disease.
Collapse
|
3
|
Popov SV, Maslov LN, Mukhomedzyanov AV, Kurbatov BK, Gorbunov AS, Kilin M, Azev VN, Khlestkina MS, Sufianova GZ. Apelin Is a Prototype of Novel Drugs for the Treatment of Acute Myocardial Infarction and Adverse Myocardial Remodeling. Pharmaceutics 2023; 15:pharmaceutics15031029. [PMID: 36986889 PMCID: PMC10056827 DOI: 10.3390/pharmaceutics15031029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/03/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
In-hospital mortality in patients with ST-segment elevation myocardial infarction (STEMI) is 5-6%. Consequently, it is necessary to develop fundamentally novel drugs capable of reducing mortality in patients with acute myocardial infarction. Apelins could be the prototype for such drugs. Chronic administration of apelins mitigates adverse myocardial remodeling in animals with myocardial infarction or pressure overload. The cardioprotective effect of apelins is accompanied by blockage of the MPT pore, GSK-3β, and the activation of PI3-kinase, Akt, ERK1/2, NO-synthase, superoxide dismutase, glutathione peroxidase, matrix metalloproteinase, the epidermal growth factor receptor, Src kinase, the mitoKATP channel, guanylyl cyclase, phospholipase C, protein kinase C, the Na+/H+ exchanger, and the Na+/Ca2+ exchanger. The cardioprotective effect of apelins is associated with the inhibition of apoptosis and ferroptosis. Apelins stimulate the autophagy of cardiomyocytes. Synthetic apelin analogues are prospective compounds for the development of novel cardioprotective drugs.
Collapse
Affiliation(s)
- Sergey V Popov
- Tomsk National Research Medical Center, Cardiology Research Institute, The Russian Academy of Sciences, Kyevskaya 111A, Tomsk 634012, Russia
| | - Leonid N Maslov
- Tomsk National Research Medical Center, Cardiology Research Institute, The Russian Academy of Sciences, Kyevskaya 111A, Tomsk 634012, Russia
| | - Alexandr V Mukhomedzyanov
- Tomsk National Research Medical Center, Cardiology Research Institute, The Russian Academy of Sciences, Kyevskaya 111A, Tomsk 634012, Russia
| | - Boris K Kurbatov
- Tomsk National Research Medical Center, Cardiology Research Institute, The Russian Academy of Sciences, Kyevskaya 111A, Tomsk 634012, Russia
| | - Alexandr S Gorbunov
- Tomsk National Research Medical Center, Cardiology Research Institute, The Russian Academy of Sciences, Kyevskaya 111A, Tomsk 634012, Russia
| | - Michail Kilin
- Tomsk National Research Medical Center, Cardiology Research Institute, The Russian Academy of Sciences, Kyevskaya 111A, Tomsk 634012, Russia
| | - Viacheslav N Azev
- Branch of Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, The Russian Academy of Sciences, Pushchino 142290, Russia
| | - Maria S Khlestkina
- Department of Pharmacology, Tyumen State Medical University, Tyumen 625023, Russia
| | - Galina Z Sufianova
- Department of Pharmacology, Tyumen State Medical University, Tyumen 625023, Russia
| |
Collapse
|
4
|
Martins WK, Silva MDND, Pandey K, Maejima I, Ramalho E, Olivon VC, Diniz SN, Grasso D. Autophagy-targeted therapy to modulate age-related diseases: Success, pitfalls, and new directions. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 2:100033. [PMID: 34909664 PMCID: PMC8663935 DOI: 10.1016/j.crphar.2021.100033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 04/15/2021] [Accepted: 05/02/2021] [Indexed: 02/08/2023] Open
Abstract
Autophagy is a critical metabolic process that supports homeostasis at a basal level and is dynamically regulated in response to various physiological and pathological processes. Autophagy has some etiologic implications that support certain pathological processes due to alterations in the lysosomal-degradative pathway. Some of the conditions related to autophagy play key roles in highly relevant human diseases, e.g., cardiovascular diseases (15.5%), malignant and other neoplasms (9.4%), and neurodegenerative conditions (3.7%). Despite advances in the discovery of new strategies to treat these age-related diseases, autophagy has emerged as a therapeutic option after preclinical and clinical studies. Here, we discuss the pitfalls and success in regulating autophagy initiation and its lysosome-dependent pathway to restore its homeostatic role and mediate therapeutic effects for cancer, neurodegenerative, and cardiac diseases. The main challenge for the development of autophagy regulators for clinical application is the lack of specificity of the repurposed drugs, due to the low pharmacological uniqueness of their target, including those that target the PI3K/AKT/mTOR and AMPK pathway. Then, future efforts must be conducted to deal with this scenery, including the disclosure of key components in the autophagy machinery that may intervene in its therapeutic regulation. Among all efforts, those focusing on the development of novel allosteric inhibitors against autophagy inducers, as well as those targeting autolysosomal function, and their integration into therapeutic regimens should remain a priority for the field.
Collapse
Affiliation(s)
- Waleska Kerllen Martins
- Laboratory of Cell and Membrane (LCM), Anhanguera University of São Paulo (UNIAN), Rua Raimundo Pereira de Magalhães, 3,305. Pirituba, São Paulo, 05145-200, Brazil
| | - Maryana do Nascimento da Silva
- Laboratory of Cell and Membrane (LCM), Anhanguera University of São Paulo (UNIAN), Rua Raimundo Pereira de Magalhães, 3,305. Pirituba, São Paulo, 05145-200, Brazil
| | - Kiran Pandey
- Center for Neural Science, New York University, Meyer Building, Room 823, 4 Washington Place, New York, NY, 10003, USA
| | - Ikuko Maejima
- Laboratory of Molecular Traffic, Institute for Molecular and Cellular Regulation, Gunma University, 3-39-15 Showa Machi, Maebashi, Gunma, 3718512, Japan
| | - Ercília Ramalho
- Laboratory of Cell and Membrane (LCM), Anhanguera University of São Paulo (UNIAN), Rua Raimundo Pereira de Magalhães, 3,305. Pirituba, São Paulo, 05145-200, Brazil
| | - Vania Claudia Olivon
- Laboratory of Pharmacology and Physiology, UNIDERP, Av. Ceará, 333. Vila Miguel Couto, Campo Grande, MS, 79003-010, Brazil
| | - Susana Nogueira Diniz
- Laboratory of Molecular Biology and Functional Genomics, Anhanguera University of São Paulo (UNIAN), Rua Raimundo Pereira de Magalhães, 3,305. Pirituba, São Paulo, 05145-200, Brazil
| | - Daniel Grasso
- Instituto de Estudios de la Inmunidad Humoral (IDEHU), Universidad de Buenos Aires, CONICET, Junín 954 p4, Buenos Aires, C1113AAD, Argentina
| |
Collapse
|
5
|
Kulek AR, Anzell A, Wider JM, Sanderson TH, Przyklenk K. Mitochondrial Quality Control: Role in Cardiac Models of Lethal Ischemia-Reperfusion Injury. Cells 2020; 9:cells9010214. [PMID: 31952189 PMCID: PMC7016592 DOI: 10.3390/cells9010214] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/10/2020] [Accepted: 01/12/2020] [Indexed: 02/07/2023] Open
Abstract
The current standard of care for acute myocardial infarction or 'heart attack' is timely restoration of blood flow to the ischemic region of the heart. While reperfusion is essential for the salvage of ischemic myocardium, re-introduction of blood flow paradoxically kills (rather than rescues) a population of previously ischemic cardiomyocytes-a phenomenon referred to as 'lethal myocardial ischemia-reperfusion (IR) injury'. There is long-standing and exhaustive evidence that mitochondria are at the nexus of lethal IR injury. However, during the past decade, the paradigm of mitochondria as mediators of IR-induced cardiomyocyte death has been expanded to include the highly orchestrated process of mitochondrial quality control. Our aims in this review are to: (1) briefly summarize the current understanding of the pathogenesis of IR injury, and (2) incorporating landmark data from a broad spectrum of models (including immortalized cells, primary cardiomyocytes and intact hearts), provide a critical discussion of the emerging concept that mitochondrial dynamics and mitophagy (the components of mitochondrial quality control) may contribute to the pathogenesis of cardiomyocyte death in the setting of ischemia-reperfusion.
Collapse
Affiliation(s)
- Andrew R. Kulek
- Cardiovascular Research Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA; (A.R.K.); (A.A.); (T.H.S.)
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Anthony Anzell
- Cardiovascular Research Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA; (A.R.K.); (A.A.); (T.H.S.)
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Departments of Emergency Medicine and Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA;
| | - Joseph M. Wider
- Departments of Emergency Medicine and Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA;
| | - Thomas H. Sanderson
- Cardiovascular Research Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA; (A.R.K.); (A.A.); (T.H.S.)
- Departments of Emergency Medicine and Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA;
| | - Karin Przyklenk
- Cardiovascular Research Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA; (A.R.K.); (A.A.); (T.H.S.)
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Correspondence: ; Tel.: +1-313-577-9047
| |
Collapse
|
6
|
Wang MJ, Yang CH, Jin Y, Wan CB, Qian WH, Xing F, Li X, Liu YY. Baicalin Inhibits Coxsackievirus B3 Replication by Reducing Cellular Lipid Synthesis. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2020; 48:143-160. [PMID: 31903780 DOI: 10.1142/s0192415x20500081] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Baicalin is a flavonoid extracted from Scutellariae Radix and shows a variety of biological activities as reducing lipids, diminishing inflammation, and inhibiting bacterial infection. However, there is no report of baicalin against CVB3 infection. In this study, we found that baicalin can reduce viral titer in a dose-dependent manner in vitro at a dose with no direct virucidal effect. Moreover, we revealed that baicalin can also improve survival rate, reduce heart weight/body weight ratio, prevent virus replication, and relieve myocardial inflammation in the acute viral myocarditis mouse model induced by CVB3. Then, in order to explore the mechanism of baicalin inhibiting CVB3 replication, we respectively examined the expression of autophagosome marker LC3-II by Western blot, tested the concentration of free fatty acid (FFA) and cholesterol (CHO) by commercial kits, detected the mRNA levels of fatty acid synthase (Fasn) and acetyl coenzyme a carboxylase (ACC) by RT-PCR, and observed the lipid content of cells by fluorescence staining. The results showed that CVB3 infection increased autophagosome formation and lipid content in HeLa cells, but these changes were significantly blocked by baicalin. Finally, in order to confirm that baicalin inhibits viral replication and reduces autophagosome formation by reducing cellular lipids, we added exogenous palmitate to cell culture supernatants to promote intracellular lipid synthesis and found that palmitate did not alter LC3-II and CVB3/VP1 expression in HeLa cells with or without CVB3 infection. Interestingly, palmitate can reverse the inhibitory effect of baicalin on autophagosome formation and viral replication. In conclusion, our results indicated that lipids play an important role in CVB3 replication, and the effect of baicalin against CVB3 was associated with its ability to reduce cellular lipid synthesis to limit autophagosome formation.
Collapse
Affiliation(s)
- Meng-Jie Wang
- Department of Clinical Laboratory, Lian'shui County People's Hospital, 6 East of Hongri Avenue, Huai'an, Jiangsu 223400, P. R. China
| | - Chun-Hua Yang
- Department of Clinical Laboratory, Huai'an Hospital of Huaian District, 14 Yuemiao East Street, Huai'an, Jiangsu 223200, P. R. China
| | - Yue Jin
- Department of Clinical Laboratory, The Affiliated Huai'an Hospital of Xuzhou Medical University, 62 Huaihai South Road, Huaian, Jiangsu 223002, P. R. China
| | - Chang-Biao Wan
- Department of Clinical Laboratory, The Affiliated Huai'an Hospital of Xuzhou Medical University, 62 Huaihai South Road, Huaian, Jiangsu 223002, P. R. China
| | - Wei-He Qian
- Department of Clinical Laboratory, The Affiliated Huai'an Hospital of Xuzhou Medical University, 62 Huaihai South Road, Huaian, Jiangsu 223002, P. R. China
| | - Fei Xing
- Department of Clinical Laboratory, The Affiliated Huai'an Hospital of Xuzhou Medical University, 62 Huaihai South Road, Huaian, Jiangsu 223002, P. R. China
| | - Xiang Li
- Department of Clinical Laboratory, The Affiliated Huai'an Hospital of Xuzhou Medical University, 62 Huaihai South Road, Huaian, Jiangsu 223002, P. R. China
| | - Yuan-Yuan Liu
- Department of Endocrinology, The First Affiliated Hospital of Soochow University, 188 Shizhi Street, Suzhou, Jiangsu 215006, P. R. China.,Department of Endocrinology, Huai'an First Affiliated Hospital of Nanjing Medical University, 6 Beijing West Road, Huaian, Jiangsu 223300, P. R. China
| |
Collapse
|
7
|
Kore RA, Henson JC, Hamzah RN, Griffin RJ, Tackett AJ, Ding Z, Mehta JL. Molecular events in MSC exosome mediated cytoprotection in cardiomyocytes. Sci Rep 2019; 9:19276. [PMID: 31848380 PMCID: PMC6917778 DOI: 10.1038/s41598-019-55694-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 11/25/2019] [Indexed: 01/15/2023] Open
Abstract
A host of hormonal-metabolic alterations take place following exposure of cardiomyocytes to hypoxia and other noxious stimuli. Here, we demonstrate that exposure of cultured rat cardiomyocytes to lipopolysaccharide (LPS) resulted in upregulation (~1.5 fold) of oxidized low-density lipoprotein receptor-1 (LOX-1). There was also a marked increase in apoptosis 12 hrs after LPS treatment with caspase-3 levels being significantly elevated (~1.3 fold) and a significant increase in LDH release at 24 hrs. Interestingly, there was a ~1.4-fold upregulation of LC-3 expression post-LPS treatment indicating development of autophagy, which probably is a compensatory response to combat cellular injury induced by LPS. Treatment with LPS also reduced the size and morphology of cardiomyocyte spheroids. In an attempt to limit LPS-induced injury, cardiomyocytes were treated with exosomes derived from mesenchymal stromal cells (MSCs). We noted a significant suppression of LOX-1 expression that in turn suppressed apoptosis as well as autophagic response and restored spheroid morphology. Mass spectrophotometric analysis of MSC exosomes revealed a cargo rich in proteins which are involved in pathways negatively modulating cell death and apoptosis while promoting cell survival. This is first report to our knowledge on the initial molecular events in MSC exosome mediated cytoprotection of stressed cardiomyocytes.
Collapse
Affiliation(s)
- Rajshekhar A Kore
- Department of Medicine, Cardiology Division, University of Arkansas for Medical Sciences, Little Rock, AR and the Central Arkansas Veterans Healthcare system, Little Rock, AR, 72205, USA
| | - Jeffrey C Henson
- Department of Medicine, Cardiology Division, University of Arkansas for Medical Sciences, Little Rock, AR and the Central Arkansas Veterans Healthcare system, Little Rock, AR, 72205, USA
| | - Rabab N Hamzah
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.,Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, AR, 72204, USA
| | - Robert J Griffin
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Alan J Tackett
- Department of Biochemistry and Molecular biology, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Zufeng Ding
- Department of Medicine, Cardiology Division, University of Arkansas for Medical Sciences, Little Rock, AR and the Central Arkansas Veterans Healthcare system, Little Rock, AR, 72205, USA
| | - Jawahar L Mehta
- Department of Medicine, Cardiology Division, University of Arkansas for Medical Sciences, Little Rock, AR and the Central Arkansas Veterans Healthcare system, Little Rock, AR, 72205, USA.
| |
Collapse
|
8
|
Hu S, Cao S, Tong Z, Liu J. FGF21 protects myocardial ischemia-reperfusion injury through reduction of miR-145-mediated autophagy. Am J Transl Res 2018; 10:3677-3688. [PMID: 30662618 PMCID: PMC6291727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 10/21/2018] [Indexed: 06/09/2023]
Abstract
AIMS Fibroblast growth factor 21 (FGF21) plays a critical role in protecting against myocardial ischemia/reperfusion (I/R) injury. However, the molecular mechanism is not completely understood. Here, we aimed to examine whether miRNA-145 (miR-145) is involved in FGF21 protection against myocardial I/R injury through angiopoietin-2 (Angpt2) and autophagy. METHODS We established a rat myocardial I/R model and H9c2 hypoxia/reoxygenation (H/R) model. After administration of FGF21 in the rat I/R model, the infarct size, morphological changes and apoptosis in myocardium were determined by 2,3,5-triphenyltetrazolium chloride (TTC), hematoxylin and eosin (HE), and Masson's trichrome staining, and TUNEL assay, respectively. The expression levels of miR-145 and Angpt2 were evaluated by quantitative real-time PCR (qRT-PCR), Western blotting and immunohistochemical (IHC) staining. The activity of lactate dehydrogenase (LDH), TNF-α and IL-6 were assayed. Using a dual-luciferase reporter system, the targeted role of miR-145 on Angpt2 was studied. After transfection with miR-145 inhibitor, H9c2 cells were subjected to stimulated H/R with or without FGF21 treatment. The expression of Angpt2 was assessed while cell apoptosis and cell migration assays were performed. RESULTS FGF21 significantly decreased infarction after I/R, ameliorated I/R-induced cell apoptosis, and inhibited I/R-induced LDH, TNF-α and IL-6 in serum. FGF21 inhibited I/R-induced decrease in miR-145 level, increase in Angpt2 expression and decrease in autophagy; FGF21 also upregulated LC3-B and Beclin1 levels. miR-145 directly targeted Angpt2. The roles of FGF21 in expression of miR-145 and Angpt2 and activation of autophagy after H/R were reversed by miR-145 inhibitor. In addition, the FGF21-inhibited cell apoptosis and FGF21-promoted migration after H/R were restored by miR-145 inhibitor. CONCLUSION FGF21 protects myocardial cells against I/R injury by promoting an increase in miR-145 levels and autophagy while inhibiting Angpt2 expression, suggesting a novel therapeutic strategy for protecting against myocardial I/R injury.
Collapse
Affiliation(s)
- Shuoqiang Hu
- Department of Cardiology, Beijing Daxing Teaching Hospital, Capital Medical UniversityBeijing 102600, P. R. China
| | - Shujun Cao
- Department of Cardiology, Beijing Daxing Teaching Hospital, Capital Medical UniversityBeijing 102600, P. R. China
| | - Zichuan Tong
- Department of Cardiology, Beijing Daxing Teaching Hospital, Capital Medical UniversityBeijing 102600, P. R. China
| | - Jinghua Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel DiseasesBeijing 100029, P. R. China
| |
Collapse
|
9
|
Ferdinandy P, Baczkó I, Bencsik P, Giricz Z, Görbe A, Pacher P, Varga ZV, Varró A, Schulz R. Definition of hidden drug cardiotoxicity: paradigm change in cardiac safety testing and its clinical implications. Eur Heart J 2018; 40:1771-1777. [PMID: 29982507 PMCID: PMC6554653 DOI: 10.1093/eurheartj/ehy365] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 03/12/2018] [Accepted: 06/11/2018] [Indexed: 12/18/2022] Open
Abstract
Unexpected cardiac adverse effects are the leading causes of discontinuation of clinical trials and withdrawal of drugs from the market. Since the original observations in the mid-90s, it has been well established that cardiovascular risk factors and comorbidities (such as ageing, hyperlipidaemia, and diabetes) and their medications (e.g. nitrate tolerance, adenosine triphosphate-dependent potassium inhibitor antidiabetic drugs, statins, etc.) may interfere with cardiac ischaemic tolerance and endogenous cardioprotective signalling pathways. Indeed drugs may exert unwanted effects on the diseased and treated heart that is hidden in the healthy myocardium. Hidden cardiotoxic effects may be due to (i) drug-induced enhancement of deleterious signalling due to ischaemia/reperfusion injury and/or the presence of risk factors and/or (ii) inhibition of cardioprotective survival signalling pathways, both of which may lead to ischaemia-related cell death and/or pro-arrhythmic effects. This led to a novel concept of ‘hidden cardiotoxicity’, defined as cardiotoxity of a drug that manifests only in the diseased heart with e.g. ischaemia/reperfusion injury and/or in the presence of its major comorbidities. Little is known on the mechanism of hidden cardiotoxocity, moreover, hidden cardiotoxicity cannot be revealed by the routinely used non-clinical cardiac safety testing methods on healthy animals or tissues. Therefore, here, we emphasize the need for development of novel cardiac safety testing platform involving combined experimental models of cardiac diseases (especially myocardial ischaemia/reperfusion and ischaemic conditioning) in the presence and absence of major cardiovascular comorbidities and/or cotreatments.
Collapse
Affiliation(s)
- Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4, Budapest, Hungary
- Pharmahungary Group, Hajnoczy u. 6, Szeged, Hungary
| | - István Baczkó
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Dóm tér 12, Szeged, Hungary
| | | | - Zoltán Giricz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4, Budapest, Hungary
- Pharmahungary Group, Hajnoczy u. 6, Szeged, Hungary
| | - Anikó Görbe
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4, Budapest, Hungary
- Pharmahungary Group, Hajnoczy u. 6, Szeged, Hungary
| | - Pál Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Fishers Lane, Bethesda, MD, USA
| | - Zoltán V Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4, Budapest, Hungary
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Fishers Lane, Bethesda, MD, USA
| | - András Varró
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Dóm tér 12, Szeged, Hungary
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University of Giessen, Aulweg 129, Giessen, Germany
| |
Collapse
|