1
|
Avolio E, Bassani B, Campanile M, Mohammed KA, Muti P, Bruno A, Spinetti G, Madeddu P. Shared molecular, cellular, and environmental hallmarks in cardiovascular disease and cancer: Any place for drug repurposing? Pharmacol Rev 2025; 77:100033. [PMID: 40148035 DOI: 10.1016/j.pharmr.2024.100033] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 12/17/2024] [Indexed: 03/29/2025] Open
Abstract
Cancer and cardiovascular disease (CVD) are the 2 biggest killers worldwide. Specific treatments have been developed for the 2 diseases. However, mutual therapeutic targets should be considered because of the overlap of cellular and molecular mechanisms. Cancer research has grown at a fast pace, leading to an increasing number of new mechanistic treatments. Some of these drugs could prove useful for treating CVD, which realizes the concept of cancer drug repurposing. This review provides a comprehensive outline of the shared hallmarks of cancer and CVD, primarily ischemic heart disease and heart failure. We focus on chronic inflammation, altered immune response, stromal and vascular cell activation, and underlying signaling pathways causing pathological tissue remodeling. There is an obvious scope for targeting those shared mechanisms, thereby achieving reciprocal preventive and therapeutic benefits. Major attention is devoted to illustrating the logic, advantages, challenges, and viable examples of drug repurposing and discussing the potential influence of sex, gender, age, and ethnicity in realizing this approach. Artificial intelligence will help to refine the personalized application of drug repurposing for patients with CVD. SIGNIFICANCE STATEMENT: Cancer and cardiovascular disease (CVD), the 2 biggest killers worldwide, share several underlying cellular and molecular mechanisms. So far, specific therapies have been developed to tackle the 2 diseases. However, the development of new cardiovascular drugs has been slow compared with cancer drugs. Understanding the intersection between pathological mechanisms of the 2 diseases provides the basis for repurposing cancer therapeutics for CVD treatment. This approach could allow the rapid development of new drugs for patients with CVDs.
Collapse
Affiliation(s)
- Elisa Avolio
- Bristol Heart Institute, Laboratory of Experimental Cardiovascular Medicine, Translational Health Sciences, Bristol Medical School, University of Bristol, United Kingdom.
| | - Barbara Bassani
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, IRCCS MultiMedica, Milan, Italy
| | - Marzia Campanile
- Laboratory of Cardiovascular Pathophysiology - Regenerative Medicine, IRCCS MultiMedica, Milan, Italy; Department of Biosciences, University of Milan, Milan, Italy
| | - Khaled Ak Mohammed
- Bristol Heart Institute, Laboratory of Experimental Cardiovascular Medicine, Translational Health Sciences, Bristol Medical School, University of Bristol, United Kingdom; Department of Cardiothoracic Surgery, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Paola Muti
- IRCCS MultiMedica, Milan, Italy; Department of Biomedical, Surgical and Dental Health Sciences, University of Milan, Italy
| | - Antonino Bruno
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, IRCCS MultiMedica, Milan, Italy; Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy.
| | - Gaia Spinetti
- Laboratory of Cardiovascular Pathophysiology - Regenerative Medicine, IRCCS MultiMedica, Milan, Italy.
| | - Paolo Madeddu
- Bristol Heart Institute, Laboratory of Experimental Cardiovascular Medicine, Translational Health Sciences, Bristol Medical School, University of Bristol, United Kingdom.
| |
Collapse
|
2
|
Wu Y, Zou Y, Song C, Cao K, Cai K, Chen S, Zhang Z, Geng D, Zhang N, Feng H, Tang M, Li Z, Sun G, Zhang Y, Sun Y, Zhang Y. The role of serine/threonine protein kinases in cardiovascular disease and potential therapeutic methods. Biomed Pharmacother 2024; 177:117093. [PMID: 38971012 DOI: 10.1016/j.biopha.2024.117093] [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: 04/17/2024] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 07/08/2024] Open
Abstract
Protein phosphorylation is an important link in a variety of signaling pathways, and most of the important life processes in cells involve protein phosphorylation. Based on the amino acid residues of phosphorylated proteins, protein kinases can be categorized into the following families: serine/threonine protein kinases, tyrosine-specific protein kinases, histidine-specific protein kinases, tryptophan kinases, and aspartate/glutamyl protein kinases. Of all the protein kinases, most are serine/threonine kinases, where serine/threonine protein kinases are protein kinases that catalyze the phosphorylation of serine or threonine residues on target proteins using ATP as a phosphate donor. The current socially accepted classification of serine/threonine kinases is to divide them into seven major groups: protein kinase A, G, C (AGC), CMGC, Calmodulin-dependent protein kinase (CAMK), Casein kinase (CK1), STE, Tyrosine kinase (TKL) and others. After decades of research, a preliminary understanding of the specific classification and respective functions of serine/threonine kinases has entered a new period of exploration. In this paper, we review the literature of the previous years and introduce the specific signaling pathways and related therapeutic modalities played by each of the small protein kinases in the serine/threonine protein kinase family, respectively, in some common cardiovascular system diseases such as heart failure, myocardial infarction, ischemia-reperfusion injury, and diabetic cardiomyopathy. To a certain extent, the current research results, including molecular mechanisms and therapeutic methods, are fully summarized and a systematic report is made for the prevention and treatment of cardiovascular diseases in the future.
Collapse
Affiliation(s)
- Yanjiao Wu
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province 110001, People's Republic of China
| | - Yuanming Zou
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province 110001, People's Republic of China
| | - Chunyu Song
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province 110001, People's Republic of China
| | - Kexin Cao
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province 110001, People's Republic of China
| | - Kexin Cai
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province 110001, People's Republic of China
| | - Shuxian Chen
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province 110001, People's Republic of China
| | - Zhaobo Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province 110001, People's Republic of China
| | - Danxi Geng
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province 110001, People's Republic of China
| | - Naijin Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province 110001, People's Republic of China; Institute of health sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning Province 110001, People's Republic of China; Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, Shenyang 110004, China.
| | - Hao Feng
- Department of Ophthalmology, The First Affiliated Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province 110001, People's Republic of China.
| | - Man Tang
- Department of clinical pharmacology, College of Pharmacy, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning Province 110001, People's Republic of China.
| | - Zhao Li
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province 110001, People's Republic of China.
| | - Guozhe Sun
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province 110001, People's Republic of China.
| | - Yixiao Zhang
- Department of Urology Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning Province 110004, People's Republic of China.
| | - Yingxian Sun
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province 110001, People's Republic of China; Institute of health sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning Province 110001, People's Republic of China; Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning Province 110001, People's Republic of China.
| | - Ying Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province 110001, People's Republic of China; Institute of health sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning Province 110001, People's Republic of China.
| |
Collapse
|
3
|
Yang C, Rubin L, Yu X, Lazarovici P, Zheng W. Preclinical evidence using synthetic compounds and natural products indicates that AMPK represents a potential pharmacological target for the therapy of pulmonary diseases. Med Res Rev 2024; 44:1326-1369. [PMID: 38229486 DOI: 10.1002/med.22014] [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: 06/05/2023] [Revised: 12/07/2023] [Accepted: 12/30/2023] [Indexed: 01/18/2024]
Abstract
Adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) is a highly conserved eukaryotic enzyme discovered as a key regulator of cellular energy homeostasis, with anti-inflammation, antioxidative stress, anticancer, and antifibrosis beneficial effects. AMPK is dysregulated in human pulmonary diseases such as acute lung injury, nonsmall cell lung cancer, pulmonary fibrosis, chronic obstructive pulmonary disease, and asthma. This review provides an overview of the beneficial role of natural, synthetic, and Chinese traditional medicines AMPK modulators in pulmonary diseases, and highlights the role of the AMPK signaling pathway in the lung, emphasizing the importance of finding lead compounds and drugs that can target and modulate AMPK to treat the lung diseases.
Collapse
Affiliation(s)
- Chao Yang
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Limor Rubin
- Allergy and Clinical Immunology Unit, Department of Medicine, Jerusalem, Israel
| | - Xiyong Yu
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Philip Lazarovici
- School of Pharmacy Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wenhua Zheng
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| |
Collapse
|
4
|
Boengler K, Eickelmann C, Kleinbongard P. Mitochondrial Kinase Signaling for Cardioprotection. Int J Mol Sci 2024; 25:4491. [PMID: 38674076 PMCID: PMC11049936 DOI: 10.3390/ijms25084491] [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: 03/01/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Myocardial ischemia/reperfusion injury is reduced by cardioprotective adaptations such as local or remote ischemic conditioning. The cardioprotective stimuli activate signaling cascades, which converge on mitochondria and maintain the function of the organelles, which is critical for cell survival. The signaling cascades include not only extracellular molecules that activate sarcolemmal receptor-dependent or -independent protein kinases that signal at the plasma membrane or in the cytosol, but also involve kinases, which are located to or within mitochondria, phosphorylate mitochondrial target proteins, and thereby modify, e.g., respiration, the generation of reactive oxygen species, calcium handling, mitochondrial dynamics, mitophagy, or apoptosis. In the present review, we give a personal and opinionated overview of selected protein kinases, localized to/within myocardial mitochondria, and summarize the available data on their role in myocardial ischemia/reperfusion injury and protection from it. We highlight the regulation of mitochondrial function by these mitochondrial protein kinases.
Collapse
Affiliation(s)
- Kerstin Boengler
- Institute of Physiology, Justus-Liebig University, 35392 Giessen, Germany
| | - Chantal Eickelmann
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, 45147 Essen, Germany; (C.E.); (P.K.)
| | - Petra Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, 45147 Essen, Germany; (C.E.); (P.K.)
| |
Collapse
|
5
|
Maiese K. Innovative therapeutic strategies for cardiovascular disease. EXCLI JOURNAL 2023; 22:690-715. [PMID: 37593239 PMCID: PMC10427777 DOI: 10.17179/excli2023-6306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 07/24/2023] [Indexed: 08/19/2023]
Abstract
As a significant non-communicable disease, cardiovascular disease is the leading cause of death for both men and women, comprises almost twenty percent of deaths in most racial and ethnic groups, can affect greater than twenty-five million individuals worldwide over the age of twenty, and impacts global economies with far-reaching financial challenges. Multiple factors can affect the onset of cardiovascular disease that include high serum cholesterol levels, elevated blood pressure, tobacco consumption and secondhand smoke exposure, poor nutrition, physical inactivity, obesity, and concurrent diabetes mellitus. Yet, addressing any of these factors cannot completely eliminate the onset or progression of cardiovascular disorders. Novel strategies are necessary to target underlying cardiovascular disease mechanisms. The silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), a histone deacetylase, can limit cardiovascular injury, assist with stem cell development, oversee metabolic homeostasis through nicotinamide adenine dinucleotide (NAD+) pathways, foster trophic factor protection, and control cell senescence through the modulation of telomere function. Intimately tied to SIRT1 pathways are mammalian forkhead transcription factors (FoxOs) which can modulate cardiac disease to reduce oxidative stress, repair microcirculation disturbances, and reduce atherogenesis through pathways of autophagy, apoptosis, and ferroptosis. AMP activated protein kinase (AMPK) also is critical among these pathways for the oversight of cardiac cellular metabolism, insulin sensitivity, mitochondrial function, inflammation, and the susceptibility to viral infections such as severe acute respiratory syndrome coronavirus that can impact cardiovascular disease. Yet, the relationship among these pathways is both intricate and complex and requires detailed insight to successfully translate these pathways into clinical care for cardiovascular disorders.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, New York, New York 10022
| |
Collapse
|
6
|
Duan S, Zhang M, Zeng H, Song J, Zhang M, Gao S, Yang H, Ding M, Li P. Integrated proteomics and phosphoproteomics profiling reveals the cardioprotective mechanism of bioactive compounds derived from Salvia miltiorrhiza Burge. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 117:154897. [PMID: 37307738 DOI: 10.1016/j.phymed.2023.154897] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/29/2023] [Accepted: 05/21/2023] [Indexed: 06/14/2023]
Abstract
BACKGROUND Natural products are an important source for discovering novel drugs due to their various pharmacological activities. Salvia miltiorrhiza Burge (Danshen) has been shown to have promising therapeutic potential in the management of heart diseases, making it a candidate for cardiovascular drug discovery. Currently, there is limited quantitative analysis of the phosphorylation levels of Danshen-derived natural products on a proteome-wide, which may bias the study of their mechanisms of action. PURPOSE This study aimed to evaluate the global signaling perturbation induced by Danshen-derived bioactive compounds and their potential relationship with myocardial ischemia/reperfusion (IR) injury therapy. STUDY DESIGN We employed quantitative proteome and phosphoproteome analysis to identify dysregulated signaling in IR injury hearts from mice. We compared changes induced by Danshen-derived compounds based on IR-associated phospho-events, using an integrative approach that maps relative abundance of proteins and phosphorylation sites. METHODS Isobaric chemical tandem mass tags (TMT) labeled multiplexing strategy was used to generate unbiased quantitative proteomics and phosphoproteomics data. Highly accurate and precise TMT quantitation was performed using the Orbitrap Fusion Tribrid Mass Spectrometer with synchronous precursor selection MS3 detection mode. Mass spectrometric raw files were analyzed with MaxQuant (2.0.1.0) and statistical and bioinformatics analysis was conducted with Perseus (1.6.15). RESULTS We quantified 3661 proteins and over 11,000 phosphosites in impaired heart tissue of the IR mice model, expanding our knowledge of signaling pathways and other biological processes disrupted in IR injury. Next, 1548 and 5545 differently expressed proteins and phosphosites were identified by quantifying the proteome and phosphoproteome of H9c2 cells treated by five Danshen bioactive compounds respectively. Results revealed the vast differences in abilities of five Danshen-derived bioactive compounds to regulate phosphorylation modifications in cardiomyocytes, with dihydrotanshinone I (DHT) showing potential for protecting against IR injury by modulating the AMPK/mTOR signaling pathway. CONCLUSIONS This study provides a new strategy for analyzing drug/natural product-regulated phosphorylation modification levels on a proteome-wide scale, leading to a better understanding of cell signaling pathways and downstream phenotypic responses.
Collapse
Affiliation(s)
- Shengnan Duan
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Meiting Zhang
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Hao Zeng
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tongjia Lane, Nanjing 210009, China
| | - Jinyi Song
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Min Zhang
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Song Gao
- Department of Pharmaceutical and Environmental Health Sciences, Texas Southern University, 3100 Cleburne Street, Houston, TX 77004, USA
| | - Hua Yang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tongjia Lane, Nanjing 210009, China
| | - Ming Ding
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China.
| | - Ping Li
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tongjia Lane, Nanjing 210009, China.
| |
Collapse
|
7
|
Oxytocin ameliorates high glucose- and ischemia/reperfusion-induced myocardial injury by suppressing pyroptosis via AMPK signaling pathway. Biomed Pharmacother 2022; 153:113498. [DOI: 10.1016/j.biopha.2022.113498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/23/2022] [Accepted: 07/30/2022] [Indexed: 11/20/2022] Open
|
8
|
Sun M, Chen Z, Song Y, Zhang B, Yang J, Tan H. PLXND1-mediated calcium dyshomeostasis impairs endocardial endothelial autophagy in atrial fibrillation. Front Physiol 2022; 13:960480. [PMID: 36017337 PMCID: PMC9395636 DOI: 10.3389/fphys.2022.960480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 07/14/2022] [Indexed: 11/26/2022] Open
Abstract
Left atrial appendage (LAA) thrombus detachment resulting in intracranial embolism is a major complication of atrial fibrillation (AF). Endocardial endothelial cell (EEC) injury leads to thrombosis, whereas autophagy protects against EEC dysfunction. However, the role and underlying mechanisms of autophagy in EECs during AF have not been elucidated. In this study, we isolated EECs from AF model mice and observed reduced autophagic flux and intracellular calcium concentrations in EECs from AF mice. In addition, we detected an increased expression of the mechanosensitive protein PLXND1 in the cytomembranes of EECs. PLXND1 served as a scaffold protein to bind with ORAI1 and further decreased ORAI1-mediated calcium influx. The decrease in the calcium influx-mediated phosphorylation of CAMK2 is associated with the inhibition of autophagy, which results in EEC dysfunction in AF. Our study demonstrated that the change in PLXND1 expression contributes to intracellular calcium dyshomeostasis, which inhibits autophagy flux and results in EEC dysfunction in AF. This study provides a potential intervention target for EEC dysfunction to prevent and treat intracardiac thrombosis in AF and its complications.
Collapse
Affiliation(s)
- Mengjia Sun
- Institute of Cardiovascular Diseases of PLA, the Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- Department of Cardiology, the Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhen Chen
- Institute of Cardiovascular Diseases of PLA, the Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- Department of Cardiology, the Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yuanbin Song
- Institute of Cardiovascular Diseases of PLA, the Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- Department of Cardiology, the Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Bo Zhang
- Institute of Cardiovascular Diseases of PLA, the Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- Department of Cardiology, the Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jie Yang
- Institute of Cardiovascular Diseases of PLA, the Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- Department of Cardiology, the Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- *Correspondence: Jie Yang, ; Hu Tan,
| | - Hu Tan
- Institute of Cardiovascular Diseases of PLA, the Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- Department of Cardiology, the Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- *Correspondence: Jie Yang, ; Hu Tan,
| |
Collapse
|
9
|
Melis N, Carcy R, Rubera I, Cougnon M, Duranton C, Tauc M, Pisani DF. Akt Inhibition as Preconditioning Treatment to Protect Kidney Cells against Anoxia. Int J Mol Sci 2021; 23:ijms23010152. [PMID: 35008578 PMCID: PMC8745656 DOI: 10.3390/ijms23010152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/15/2021] [Accepted: 12/21/2021] [Indexed: 11/16/2022] Open
Abstract
Lesions issued from the ischemia/reperfusion (I/R) stress are a major challenge in human pathophysiology. Of human organs, the kidney is highly sensitive to I/R because of its high oxygen demand and poor regenerative capacity. Previous studies have shown that targeting the hypusination pathway of eIF5A through GC7 greatly improves ischemic tolerance and can be applied successfully to kidney transplants. The protection process correlates with a metabolic shift from oxidative phosphorylation to glycolysis. Because the protein kinase B Akt is involved in ischemic protective mechanisms and glucose metabolism, we looked for a link between the effects of GC7 and Akt in proximal kidney cells exposed to anoxia or the mitotoxic myxothiazol. We found that GC7 treatment resulted in impaired Akt phosphorylation at the Ser473 and Thr308 sites, so the effects of direct Akt inhibition as a preconditioning protocol on ischemic tolerance were investigated. We evidenced that Akt inhibitors provide huge protection for kidney cells against ischemia and myxothiazol. The pro-survival effect of Akt inhibitors, which is reversible, implied a decrease in mitochondrial ROS production but was not related to metabolic changes or an antioxidant defense increase. Therefore, the inhibition of Akt can be considered as a preconditioning treatment against ischemia.
Collapse
Affiliation(s)
- Nicolas Melis
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA;
| | - Romain Carcy
- Université Côte d’Azur, CNRS, LP2M, 06103 Nice, France; (R.C.); (I.R.); (M.C.); (C.D.); (M.T.)
- CHU Nice, Hôpital Pasteur 2, Service de Réanimation Polyvalente et Service de Réanimation des Urgences Vitales, 06103 Nice, France
- Laboratories of Excellence Ion Channel Science and Therapeutics, 06103 Nice, France
| | - Isabelle Rubera
- Université Côte d’Azur, CNRS, LP2M, 06103 Nice, France; (R.C.); (I.R.); (M.C.); (C.D.); (M.T.)
- Laboratories of Excellence Ion Channel Science and Therapeutics, 06103 Nice, France
| | - Marc Cougnon
- Université Côte d’Azur, CNRS, LP2M, 06103 Nice, France; (R.C.); (I.R.); (M.C.); (C.D.); (M.T.)
- Laboratories of Excellence Ion Channel Science and Therapeutics, 06103 Nice, France
| | - Christophe Duranton
- Université Côte d’Azur, CNRS, LP2M, 06103 Nice, France; (R.C.); (I.R.); (M.C.); (C.D.); (M.T.)
- Laboratories of Excellence Ion Channel Science and Therapeutics, 06103 Nice, France
| | - Michel Tauc
- Université Côte d’Azur, CNRS, LP2M, 06103 Nice, France; (R.C.); (I.R.); (M.C.); (C.D.); (M.T.)
- Laboratories of Excellence Ion Channel Science and Therapeutics, 06103 Nice, France
| | - Didier F. Pisani
- Université Côte d’Azur, CNRS, LP2M, 06103 Nice, France; (R.C.); (I.R.); (M.C.); (C.D.); (M.T.)
- Laboratories of Excellence Ion Channel Science and Therapeutics, 06103 Nice, France
- Correspondence:
| |
Collapse
|
10
|
Ma J, Chen C, Yu Q, Han L. AMP-activated protein kinase contributes to myofibrillar protein hydrolysis in bovine skeletal muscle through postmortem mitochondrial dysfunction-induced apoptosis. J Food Biochem 2021; 46:e14028. [PMID: 34894156 DOI: 10.1111/jfbc.14028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/17/2021] [Accepted: 11/09/2021] [Indexed: 11/30/2022]
Abstract
This study aimed to verify the role of AMP-activated protein kinase (AMPK) in mitochondrial dysfunction-induced apoptosis and postmortem bovine muscle tenderization. AMPK phosphorylation levels, mitochondrial dysfunction, mitochondrial apoptotic factors, and myofibrillar protein hydrolysis were assessed in the control group and Compound C (AMPK inhibitor) group over a 168 hr aging period. Compared with the Compound C group, the control group had an extremely significantly increased AMPK activity at 6-120 hr (p < .01) and a 62.3% and 42.1% higher mitochondrial Bax/Bcl-2 ratio at 6 and 12 hr, respectively (p < .05). Moreover, the control group had a significantly or extremely significantly higher mitochondrial dysfunction and cytoplasmic cytochrome c content at 6-72 and 12-72 hr, respectively (p < .05, p < .01); a 23.2%, 26.5%, and 26.1% increased caspase-3 expression levels at 12, 24, and 72 hr, respectively (p < .05); a significantly higher proportion of apoptotic nuclei at 24-168 hr (p < .05); and a 30.8%, 35.8%, 43.9%, and 39.5% increased production of 45-, 38-, 36-, 30-, and 28-kDa proteins at 168 hr, respectively (p < .05). Taken together, these results suggested that activated AMPK promoted mitochondrial apoptosis and bovine muscle tenderization during postmortem aging by increasing the Bax/Bcl-2 ratio on the mitochondrial membrane. PRACTICAL APPLICATIONS: Based on consumer preference, chilled fresh meat is gradually becoming the future trend of the meat industry. Poorly tenderized beef often affects consumers' desire to make secondary purchases and leads to large losses to the meat industry. Therefore, AMP-activated protein kinase, which regulates postmortem mitochondrial apoptosis and bovine muscle tenderization, is a valid research target.
Collapse
Affiliation(s)
- Jibing Ma
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou, China
| | - Cheng Chen
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou, China
| | - Qunli Yu
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou, China
| | - Ling Han
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou, China
| |
Collapse
|
11
|
Yin J, Zhan D, Ma H, Liu H, Yu L, Zhang Y, Nan F. Optimization and Development of a Scalable Palladium-Catalyzed C–H Activation Process for the Geometry-Selective Preparation of Kilograms of YLF466D, a Potent AMP-Activated Protein Kinase Activator. Org Process Res Dev 2021. [DOI: 10.1021/acs.oprd.1c00150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Jianpeng Yin
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai, Shandong 264000, China
| | - Desheng Zhan
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai, Shandong 264000, China
| | - Hui Ma
- The National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Huanan Liu
- The National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Lifang Yu
- The National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yangming Zhang
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai, Shandong 264000, China
- The National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Fajun Nan
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai, Shandong 264000, China
- The National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| |
Collapse
|
12
|
Marino A, Hausenloy DJ, Andreadou I, Horman S, Bertrand L, Beauloye C. AMP-activated protein kinase: A remarkable contributor to preserve a healthy heart against ROS injury. Free Radic Biol Med 2021; 166:238-254. [PMID: 33675956 DOI: 10.1016/j.freeradbiomed.2021.02.047] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/13/2021] [Accepted: 02/26/2021] [Indexed: 12/19/2022]
Abstract
Heart failure is one of the leading causes of death and disability worldwide. Left ventricle remodeling, fibrosis, and ischemia/reperfusion injury all contribute to the deterioration of cardiac function and predispose to the onset of heart failure. Adenosine monophosphate-activated protein kinase (AMPK) is the universally recognized energy sensor which responds to low ATP levels and restores cellular metabolism. AMPK activation controls numerous cellular processes and, in the heart, it plays a pivotal role in preventing onset and progression of disease. Excessive reactive oxygen species (ROS) generation, known as oxidative stress, can activate AMPK, conferring an additional role of AMPK as a redox-sensor. In this review, we discuss recent insights into the crosstalk between ROS and AMPK. We describe the molecular mechanisms by which ROS activate AMPK and how AMPK signaling can further prevent heart failure progression. Ultimately, we review the potential therapeutic approaches to target AMPK for the treatment of cardiovascular disease and prevention of heart failure.
Collapse
Affiliation(s)
- Alice Marino
- Pôle de Recherche Cardiovasculaire, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Derek J Hausenloy
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore; National Heart Research Institute Singapore, National Heart Centre, Singapore; Yong Loo Lin School of Medicine, National University Singapore, Singapore; The Hatter Cardiovascular Institute, University College London, London, UK; Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Sandrine Horman
- Pôle de Recherche Cardiovasculaire, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Luc Bertrand
- Pôle de Recherche Cardiovasculaire, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Christophe Beauloye
- Pôle de Recherche Cardiovasculaire, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium; Division of Cardiology, Cliniques universitaires Saint Luc, Brussels, Belgium.
| |
Collapse
|
13
|
Maissan P, Mooij EJ, Barberis M. Sirtuins-Mediated System-Level Regulation of Mammalian Tissues at the Interface between Metabolism and Cell Cycle: A Systematic Review. BIOLOGY 2021; 10:194. [PMID: 33806509 PMCID: PMC7999230 DOI: 10.3390/biology10030194] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/20/2021] [Accepted: 02/25/2021] [Indexed: 02/06/2023]
Abstract
Sirtuins are a family of highly conserved NAD+-dependent proteins and this dependency links Sirtuins directly to metabolism. Sirtuins' activity has been shown to extend the lifespan of several organisms and mainly through the post-translational modification of their many target proteins, with deacetylation being the most common modification. The seven mammalian Sirtuins, SIRT1 through SIRT7, have been implicated in regulating physiological responses to metabolism and stress by acting as nutrient sensors, linking environmental and nutrient signals to mammalian metabolic homeostasis. Furthermore, mammalian Sirtuins have been implicated in playing major roles in mammalian pathophysiological conditions such as inflammation, obesity and cancer. Mammalian Sirtuins are expressed heterogeneously among different organs and tissues, and the same holds true for their substrates. Thus, the function of mammalian Sirtuins together with their substrates is expected to vary among tissues. Any therapy depending on Sirtuins could therefore have different local as well as systemic effects. Here, an introduction to processes relevant for the actions of Sirtuins, such as metabolism and cell cycle, will be followed by reasoning on the system-level function of Sirtuins and their substrates in different mammalian tissues. Their involvement in the healthy metabolism and metabolic disorders will be reviewed and critically discussed.
Collapse
Affiliation(s)
- Parcival Maissan
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands;
| | - Eva J. Mooij
- Systems Biology, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, Surrey, UK;
- Centre for Mathematical and Computational Biology, CMCB, University of Surrey, Guildford GU2 7XH, Surrey, UK
| | - Matteo Barberis
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands;
- Systems Biology, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, Surrey, UK;
- Centre for Mathematical and Computational Biology, CMCB, University of Surrey, Guildford GU2 7XH, Surrey, UK
| |
Collapse
|
14
|
Griffiths K, Lee JJ, Frenneaux MP, Feelisch M, Madhani M. Nitrite and myocardial ischaemia reperfusion injury. Where are we now? Pharmacol Ther 2021; 223:107819. [PMID: 33600852 DOI: 10.1016/j.pharmthera.2021.107819] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 01/25/2021] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease remains the leading cause of death worldwide despite major advances in technology and treatment, with coronary heart disease (CHD) being a key contributor. Following an acute myocardial infarction (AMI), it is imperative that blood flow is rapidly restored to the ischaemic myocardium. However, this restoration is associated with an increased risk of additional complications and further cardiomyocyte death, termed myocardial ischaemia reperfusion injury (IRI). Endogenously produced nitric oxide (NO) plays an important role in protecting the myocardium from IRI. It is well established that NO mediates many of its downstream functions through the 'canonical' NO-sGC-cGMP pathway, which is vital for cardiovascular homeostasis; however, this pathway can become impaired in the face of inadequate delivery of necessary substrates, in particular L-arginine, oxygen and reducing equivalents. Recently, it has been shown that during conditions of ischaemia an alternative pathway for NO generation exists, which has become known as the 'nitrate-nitrite-NO pathway'. This pathway has been reported to improve endothelial dysfunction, protect against myocardial IRI and attenuate infarct size in various experimental models. Furthermore, emerging evidence suggests that nitrite itself provides multi-faceted protection, in an NO-independent fashion, against a myriad of pathophysiologies attributed to IRI. In this review, we explore the existing pre-clinical and clinical evidence for the role of nitrate and nitrite in cardioprotection and discuss the lessons learnt from the clinical trials for nitrite as a perconditioning agent. We also discuss the potential future for nitrite as a pre-conditioning intervention in man.
Collapse
Affiliation(s)
- Kayleigh Griffiths
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Jordan J Lee
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Michael P Frenneaux
- Norwich Medical School, University of East Anglia, Bob Champion Research and Education Building, Norwich Research Park, Norwich NR4 7UQ, UK
| | - Martin Feelisch
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Melanie Madhani
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| |
Collapse
|
15
|
Drzewoski J, Hanefeld M. The Current and Potential Therapeutic Use of Metformin-The Good Old Drug. Pharmaceuticals (Basel) 2021; 14:122. [PMID: 33562458 PMCID: PMC7915435 DOI: 10.3390/ph14020122] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/02/2021] [Accepted: 02/02/2021] [Indexed: 02/07/2023] Open
Abstract
Metformin, one of the oldest oral antidiabetic agents and still recommended by almost all current guidelines as the first-line treatment for type 2 diabetes mellitus (T2DM), has become the medication with steadily increasing potential therapeutic indications. A broad spectrum of experimental and clinical studies showed that metformin has a pleiotropic activity and favorable effect in different pathological conditions, including prediabetes, type 1 diabetes mellitus (T1DM) and gestational diabetes mellitus (GDM). Moreover, there are numerous studies, meta-analyses and population studies indicating that metformin is safe and well tolerated and may be associated with cardioprotective and nephroprotective effect. Recently, it has also been reported in some studies, but not all, that metformin, besides improvement of glucose homeostasis, may possibly reduce the risk of cancer development, inhibit the incidence of neurodegenerative disease and prolong the lifespan. This paper presents some arguments supporting the initiation of metformin in patients with newly diagnosed T2DM, especially those without cardiovascular risk factors or without established cardiovascular disease or advanced kidney insufficiency at the time of new guidelines favoring new drugs with pleotropic effects complimentary to glucose control. Moreover, it focuses on the potential beneficial effects of metformin in patients with T2DM and coexisting chronic diseases.
Collapse
Affiliation(s)
- Józef Drzewoski
- Central Teaching Hospital of Medical University of Lodz, 92-213 Lodz, Poland
| | - Markolf Hanefeld
- Medical Clinic III, Department of Medicine Technical University Dresden, 01307 Dresden, Germany;
| |
Collapse
|
16
|
Ding R, Wu W, Sun Z, Li Z. AMP-activated protein kinase: An attractive therapeutic target for ischemia-reperfusion injury. Eur J Pharmacol 2020; 888:173484. [DOI: 10.1016/j.ejphar.2020.173484] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/26/2020] [Accepted: 08/11/2020] [Indexed: 02/07/2023]
|
17
|
Liu XD, Li YG, Wang GY, Bi YG, Zhao Y, Yan ML, Liu X, Wei M, Wan LL, Zhang QY. Metformin protects high glucose‑cultured cardiomyocytes from oxidative stress by promoting NDUFA13 expression and mitochondrial biogenesis via the AMPK signaling pathway. Mol Med Rep 2020; 22:5262-5270. [PMID: 33174032 PMCID: PMC7646981 DOI: 10.3892/mmr.2020.11599] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 06/01/2020] [Indexed: 01/16/2023] Open
Abstract
Tissue damage in diabetes is at least partly due to elevated reactive oxygen species production by the mitochondrial respiratory chain during hyperglycemia. Sustained hyperglycemia results in mitochondrial dysfunction and the abnormal expression of mitochondrial genes, such as NADH: Ubiquinone oxidoreductase subunit A13 (NDUFA13). Metformin, an AMP-activated protein kinase (AMPK) activator, protects cardiomyocytes from oxidative stress by improving mitochondrial function; however, the exact underlying mechanisms are not completely understood. The aim of the present study was to investigated the molecular changes and related regulatory mechanisms in the response of H9C2 cardiomyocytes to metformin under high glucose conditions. H9C2 cells were subjected to CCK-8 assay to assess cell viability. Reactive oxygen species generation was measured with DCFH-DA assay. Western blotting was used to analyze the expression levels of NDUFA13, AMPK, p-AMPK and GAPDH. Reverse transcription-quantitative PCR was used to evaluate the expression levels of mitochondrial genes and transcription factors. It was observed that metformin protected H9C2 cardiomyocytes by suppressing high glucose (HG)-induced elevated oxidative stress. In addition, metformin stimulated mitochondrial biogenesis, as indicated by increased expression levels of mitochondrial genes (NDUFA1, NDUFA2, NDUFA13 and manganese superoxide dismutase) and mitochondrial biogenesis-related transcription factors [peroxisome proliferator-activated receptor-gamma coactivator-1α, nuclear respiratory factor (NRF)-1, and NRF-2] in the metformin + HG group compared with the HG group. Moreover, metformin promoted mitochondrial NDUFA13 protein expression via the AMPK signaling pathway, which was abolished by pretreatment with the AMPK inhibitor, Compound C. The results suggested that metformin protected cardiomyocytes against HG-induced oxidative stress via a mechanism involving AMPK, NDUFA13 and mitochondrial biogenesis.
Collapse
Affiliation(s)
- Xiang-Dong Liu
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, P.R. China
| | - Yong-Guang Li
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Guang-Yu Wang
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Ya-Guang Bi
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Yu Zhao
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Mei-Ling Yan
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Xuebo Liu
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, P.R. China
| | - Meng Wei
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Li-Li Wan
- Division of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Qing-Yong Zhang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, P.R. China
| |
Collapse
|
18
|
Xin C, Zhang Z, Gao G, Ding L, Yang C, Wang C, Liu Y, Guo Y, Yang X, Zhang L, Zhang L, Liu Y, Jin Z, Tao L. Irisin Attenuates Myocardial Ischemia/Reperfusion Injury and Improves Mitochondrial Function Through AMPK Pathway in Diabetic Mice. Front Pharmacol 2020; 11:565160. [PMID: 33013403 PMCID: PMC7516196 DOI: 10.3389/fphar.2020.565160] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 08/14/2020] [Indexed: 01/10/2023] Open
Abstract
Aims Several recent reports have shown irisin protects the heart against ischemia/reperfusion injury. However, the effect of irisin on I/R injury in diabetic mice has not been described. The present study was designed to investigate the role of irisin in myocardial ischemia-reperfusion (MI/R) injury in diabetic mice. Methods A mouse model of diabetes was established by feeding wild type or gene-manipulated adult male mice with a high-fat diet. All the mice received intraperitoneal injection of irisin or PBS. Thirty minutes after injection, mice were subjected to 30 min of myocardial ischemia followed by 3h (for cell apoptosis and protein determination), 24 h (for infarct size and cardiac function). Results Knock-out of gene FNDC5 augmented MI/R injury in diabetic mice, while irisin treatment attenuated MI/R injury, improved cardiac function, cellular ATP biogenetics, mitochondria potential, and impaired mitochondrion-related cell death. More severely impaired AMPK pathway was observed in diabetic FNDC5-/- mice received MI/R. Knock-out of gene AMPK blocks the beneficial effects of irisin on MI/R injury, cardiac function, cellular ATP biogenetics, mitochondria potential, and mitochondrion-related cell death. Conclusions Our present study demonstrated that irisin improves the mitochondria function and attenuates MI/R injury in diabetic mice through AMPK pathway.
Collapse
Affiliation(s)
- Chao Xin
- Department of Cardiology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Zheng Zhang
- Department of Cardiology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Guojie Gao
- Department of Cardiology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Liping Ding
- Department of Cardiology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Chao Yang
- Department of Cardiology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Chengzhu Wang
- Department of Cardiology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Yanjun Liu
- Department of Cardiology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Yufei Guo
- Department of Cardiology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Xueqing Yang
- Department of Cardiology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Lijuan Zhang
- Department of Cardiology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Lina Zhang
- Department of Cardiology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Yi Liu
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Zhitao Jin
- Department of Cardiology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Ling Tao
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| |
Collapse
|
19
|
Zhu J, Wang YF, Chai XM, Qian K, Zhang LW, Peng P, Chen PM, Cao JF, Qin ZH, Sheng R, Xie H. Exogenous NADPH ameliorates myocardial ischemia-reperfusion injury in rats through activating AMPK/mTOR pathway. Acta Pharmacol Sin 2020; 41:535-545. [PMID: 31776448 PMCID: PMC7470878 DOI: 10.1038/s41401-019-0301-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 08/16/2019] [Indexed: 12/18/2022]
Abstract
Our previous study shows that nicotinamide adenine dinucleotide phosphate (NADPH) plays an important role in protecting against cerebral ischemia injury. In this study we investigated whether NADPH exerted cardioprotection against myocardial ischemia/reperfusion (I/R) injury. To induce myocardial I/R injury, rats were subjected to ligation of the left anterior descending branch of coronary artery for 30 min followed by reperfusion for 2 h. At the onset of reperfusion, NADPH (4, 8, 16 mg· kg−1· d−1, iv) was administered to the rats. We found that NADPH concentrations in plasma and heart were significantly increased at 4 h after intravenous administration. Exogenous NADPH (8−16 mg/kg) significantly decreased myocardial infarct size and reduced serum levels of lactate dehydrogenase (LDH) and cardiac troponin I (cTn-I). Exogenous NADPH significantly decreased the apoptotic rate of cardiomyocytes, and reduced the cleavage of PARP and caspase-3. In addition, exogenous NADPH reduced mitochondrial vacuolation and increased mitochondrial membrane protein COXIV and TOM20, decreased BNIP3L and increased Bcl-2 to protect mitochondrial function. We conducted in vitro experiments in neonatal rat cardiomyocytes (NRCM) subjected to oxygen–glucose deprivation/restoration (OGD/R). Pretreatment with NADPH (60, 500 nM) significantly rescued the cell viability and inhibited OGD/R-induced apoptosis. Pretreatment with NADPH significantly increased the phosphorylation of AMPK and downregulated the phosphorylation of mTOR in OGD/R-treated NRCM. Compound C, an AMPK inhibitor, abolished NADPH-induced AMPK phosphorylation and cardioprotection in OGD/R-treated NRCM. In conclusion, exogenous NADPH exerts cardioprotection against myocardial I/R injury through the activation of AMPK/mTOR pathway and inhibiting mitochondrial damage and cardiomyocyte apoptosis. NADPH may be a potential candidate for the prevention and treatment of myocardial ischemic diseases.
Collapse
|
20
|
Mitochondrial ROS-Modulated mtDNA: A Potential Target for Cardiac Aging. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:9423593. [PMID: 32308810 PMCID: PMC7139858 DOI: 10.1155/2020/9423593] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 03/05/2020] [Accepted: 03/16/2020] [Indexed: 02/05/2023]
Abstract
Mitochondrial DNA (mtDNA) damage is associated with the development of cardiovascular diseases. Cardiac aging plays a central role in cardiovascular diseases. There is accumulating evidence linking cardiac aging to mtDNA damage, including mtDNA mutation and decreased mtDNA copy number. Current wisdom indicates that mtDNA is susceptible to damage by mitochondrial reactive oxygen species (mtROS). This review presents the cellular and molecular mechanisms of cardiac aging, including autophagy, chronic inflammation, mtROS, and mtDNA damage, and the effects of mitochondrial biogenesis and oxidative stress on mtDNA. The importance of nucleoid-associated proteins (Pol γ), nuclear respiratory factors (NRF1 and NRF2), the cGAS-STING pathway, and the mitochondrial biogenesis pathway concerning the development of mtDNA damage during cardiac aging is discussed. Thus, the repair of damaged mtDNA provides a potential clinical target for preventing cardiac aging.
Collapse
|
21
|
Mechanical Postconditioning Promotes Glucose Metabolism and AMPK Activity in Parallel with Improved Post-Ischemic Recovery in an Isolated Rat Heart Model of Donation after Circulatory Death. Int J Mol Sci 2020; 21:ijms21030964. [PMID: 32024002 PMCID: PMC7039237 DOI: 10.3390/ijms21030964] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 01/27/2020] [Accepted: 01/30/2020] [Indexed: 12/25/2022] Open
Abstract
Donation after circulatory death (DCD) could improve donor heart availability; however, warm ischemia-reperfusion injury raises concerns about graft quality. Mechanical postconditioning (MPC) may limit injury, but mechanisms remain incompletely characterized. Therefore, we investigated the roles of glucose metabolism and key signaling molecules in MPC using an isolated rat heart model of DCD. Hearts underwent 20 min perfusion, 30 min global ischemia, and 60 minu reperfusion with or without MPC (two cycles: 30 s reperfusion—30 s ischemia). Despite identical perfusion conditions, MPC either significantly decreased (low recovery = LoR; 32 ± 5%; p < 0.05), or increased (high recovery = HiR; 59 ± 7%; p < 0.05) the recovery of left ventricular work compared with no MPC (47 ± 9%). Glucose uptake and glycolysis were increased in HiR vs. LoR hearts (p < 0.05), but glucose oxidation was unchanged. Furthermore, in HiR vs. LoR hearts, phosphorylation of raptor, a downstream target of AMPK, increased (p < 0.05), cytochrome c release (p < 0.05) decreased, and TNFα content tended to decrease. Increased glucose uptake and glycolysis, lower mitochondrial damage, and a trend towards decreased pro-inflammatory cytokines occurred specifically in HiR vs. LoR MPC hearts, which may result from greater AMPK activation. Thus, we identify endogenous cellular mechanisms that occur specifically with cardioprotective MPC, which could be elicited in the development of effective reperfusion strategies for DCD cardiac grafts.
Collapse
|
22
|
Sun ZG, Tian G, Zheng XC, Liu WY, Luo XT, Xiao J, Song H, Xu X. AMPKα2 Deficiency Does Not Affect the Exercise-Induced Improvements in Glucose Tolerance and Metabolic Disorders in Mice Fed a High-Fat Diet. J Nutr Sci Vitaminol (Tokyo) 2020; 65:491-497. [PMID: 31902862 DOI: 10.3177/jnsv.65.491] [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: 11/27/2022]
Abstract
Exercise can improve obesity and metabolic disorders in mice fed a high-fat diet (HFD), but the role of AMPKα2 in the process remains unclear. The aim of this study was to investigate the role of AMPKα2 in the exercise-induced improvements in glucose tolerance and metabolic turnover in obesity mice. Male wild-type mice (n=12) and AMPKα2 knockout (AMPKα2 KO) mice (n=12) were fed a HFD for 16 wk and were then randomly divided into four groups: WT HFD group (WT HF), AMPKα2 KO HFD group (AMPKα2 KO HF), WT HFD exercise group (WT HE), and AMPK HFD exercise group (AMPKα2 KO HE). The HF groups continue to be fed a HFD from 16 wk to 24 wk, and the HE groups were fed a HFD and performed exercise training. After 8 wk of exercise, all mice were placed in an energy metabolism chamber to test their metabolic turnover, include locomotor activity, food intake, oxygen consumption (VO2), carbon dioxide production (VCO2), energy expenditure (EE) and respiratory exchange ratio (RER), over a period of 3 d. Exercise improved glucose tolerance, VO2, VCO2 and EE in mice fed a HFD (p<0.05). The VO2, VCO2 and EE in AMPKα2 KO HE group were lower than these in WT HE group (p<0.05). Our findings revealed exercise improved glucose tolerance and metabolic disorders in C57 and AMPKα2 KO mice fed a HFD. AMPKα2 is not essential for exercise-induced improvements in glucose tolerance and metabolic disorders.
Collapse
Affiliation(s)
- Zhong-Guang Sun
- Department of Exercise Rehabilitation, Shanghai University of Sport
| | - Ge Tian
- Beijing Xian Nong Tan Sports Technical College
| | - Xiao-Ci Zheng
- Department of Exercise Rehabilitation, Shanghai University of Sport
| | - Wen-Ying Liu
- Department of Exercise Rehabilitation, Shanghai University of Sport
| | - Xue-Ting Luo
- Department of Exercise Rehabilitation, Shanghai University of Sport
| | - Jing Xiao
- Department of Exercise Rehabilitation, Shanghai University of Sport
| | - Hui Song
- Department of Exercise Rehabilitation, Shanghai University of Sport
| | - Xin Xu
- Department of Exercise Rehabilitation, Shanghai University of Sport
| |
Collapse
|
23
|
Jiang S, Shi F, Lin H, Ying Y, Luo L, Huang D, Luo Z. Inonotus obliquus polysaccharides induces apoptosis of lung cancer cells and alters energy metabolism via the LKB1/AMPK axis. Int J Biol Macromol 2019; 151:1277-1286. [PMID: 31751687 DOI: 10.1016/j.ijbiomac.2019.10.174] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 10/17/2019] [Accepted: 10/19/2019] [Indexed: 12/21/2022]
Abstract
The present study explores the mechanisms underlying the anti-cancer action of Inonotus obliquus polysaccharides (IOP). Thus, we characterized the IOP components extracted from Chaga sclerotium and, found that the extracts contained 70% polysaccharides with an average molecular weight of 4.5 × 104 Da consisting of 75% glucose. We then showed that IOP extract activated AMPK in lung cancer cells expressing LKB1, suppressed cell viability, colony-formation, and triggered cell apoptosis. In conjunction, IOP downregulated Bcl-2, upregulated Bax, and enhanced cleavage of Caspase-3 and PARP. All of these effects were prevented by treatment with Compound C, a chemical inhibitor of AMPK. IOP diminished mitochondrial membrane potential (MMP), concurrent with decreases in oxidative phosphorylation and glycolysis, which was dependent on LKB1/AMPK. Finally, IOP at a dosage of 50 mg/kg significantly inhibited allograft tumor growth of the LLC1 cells in association with increased apoptosis. Collectively, our results demonstrate that IOP acts on cancer cells through a mechanism by which AMPK triggers the apoptotic pathway via the opening of mitochondrial permeability transition pore, and reducing MMP, leading to an inhibition of ATP production. Therefore, our study provides a solid foundation for the use of IOP as a promising alternative or supplementary medicine for cancer therapy.
Collapse
Affiliation(s)
- Shuping Jiang
- Jiangxi Provincial Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Nanchang University Jiangxi Medical College, China
| | - Fuli Shi
- Jiangxi Provincial Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Nanchang University Jiangxi Medical College, China
| | - Hui Lin
- Jiangxi Provincial Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Nanchang University Jiangxi Medical College, China
| | - Ying Ying
- Jiangxi Provincial Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Nanchang University Jiangxi Medical College, China
| | - Lingyu Luo
- Department of Gastroenterology, Research Institute of Digestive Diseases, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Deqiang Huang
- Department of Gastroenterology, Research Institute of Digestive Diseases, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Zhijun Luo
- Jiangxi Provincial Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Nanchang University Jiangxi Medical College, China.
| |
Collapse
|
24
|
Naryzhnaya NV, Maslov LN, Oeltgen PR. Pharmacology of mitochondrial permeability transition pore inhibitors. Drug Dev Res 2019; 80:1013-1030. [PMID: 31823411 DOI: 10.1002/ddr.21593] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 08/08/2019] [Accepted: 08/12/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Natalia V. Naryzhnaya
- Laboratory of Experimental CardiologyCardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Science Tomsk Russia
| | - Leonid N. Maslov
- Laboratory of Experimental CardiologyCardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Science Tomsk Russia
| | - Peter R. Oeltgen
- Department of PathologyUniversity of Kentucky College of Medicine Lexington Kentucky
| |
Collapse
|
25
|
Li Y, Peng J, Li P, Du H, Li Y, Liu X, Zhang L, Wang LL, Zuo Z. Identification of potential AMPK activator by pharmacophore modeling, molecular docking and QSAR study. Comput Biol Chem 2019; 79:165-176. [DOI: 10.1016/j.compbiolchem.2019.02.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/20/2019] [Accepted: 02/22/2019] [Indexed: 12/31/2022]
|
26
|
Wang Z, Cao Y, Yin Q, Han Y, Wang Y, Sun G, Zhu H, Xu M, Gu C. Activation of AMPK alleviates cardiopulmonary bypass-induced cardiac injury via ameliorating acute cardiac glucose metabolic disorder. Cardiovasc Ther 2019; 36:e12482. [PMID: 30632675 DOI: 10.1111/1755-5922.12482] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 12/10/2018] [Accepted: 01/03/2019] [Indexed: 01/15/2023] Open
Abstract
Recent years, studies have demonstrated that hyperglycemia is one of the main manifestations after cardiac surgeries, which contributes to myocardial injuries and increases the chance of subsequent complications and mortality in such patients. However, strategies targeting at glucose metabolic disorder after cardiac surgeries to attenuate myocardial injuries are inadequately studied. In this study, a rat model of cardiopulmonary bypass (CPB) was applied to investigate the role of Adenosine 5'-monophosphate-activated protein kinase (AMPK) in modulating myocardial glucose metabolic-induced cardiac injuries after cardiac surgery. The results revealed that CPB elicited significant cardiac dysfunction, and pronouncedly elevated the markers of myocardial injuries including serum creatine kinase MB and cardiac troponin I. Additionally, blunted myocardial glucose uptake after CPB was associated with decreased membrane glucose transporter 4 (GLUT4) content. However, pretreatment of AMPK agonist 5-aminoimidazole-4-carboxamide1-β-D-ribofuranoside (AICAR) at the beginning of CPB activated AMPK, enhanced phosphorylation of Akt substrate 160 (AS160), and increased myocardial membrane content of GLUT4. Meanwhile, improved myocardial glucose uptake and more importantly alleviated cardiac injury were also observed after CPB pretreated with AICAR. Moreover, the application of a mutant form of AS160 (AS160-4P) abolished the beneficial effect of AICAR, as evidenced by impaired cardiac glucose uptake, reduced myocardial membrane GLUT-4 translocation, increased cardiac injury markers, and deterioration of cardiac function after CPB. In conclusion, it was suggested in this study that preactivation of AMPK by AICAR improved myocardial glucose uptake by promoting AS160 dependent myocardial membrane GLUT-4 translocation, which ultimately provided a potent cardioprotective effect.
Collapse
Affiliation(s)
- Zhifa Wang
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,Department of Pediatrics, First Affiliated Hospital of Xi'an Medical University, Xi'an Medical University, Xi'an, China
| | - Yukun Cao
- Department of Heart Center, Airforce General Hospital, Beijing, China
| | - Qiang Yin
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yuehu Han
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yunya Wang
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Guocheng Sun
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Hailong Zhu
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ming Xu
- Department of Physiology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chunhu Gu
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
27
|
Li Q, Zhang Z, Li H, Pan X, Chen S, Cui Z, Ma J, Zhou Z, Xing B. Lycium barbarum polysaccharides protects H9c2 cells from hypoxia-induced injury by down-regulation of miR-122. Biomed Pharmacother 2019; 110:20-28. [DOI: 10.1016/j.biopha.2018.11.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 10/22/2018] [Accepted: 11/02/2018] [Indexed: 02/07/2023] Open
|
28
|
Uncoupling proteins as a therapeutic target to protect the diabetic heart. Pharmacol Res 2018; 137:11-24. [PMID: 30223086 DOI: 10.1016/j.phrs.2018.09.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 09/07/2018] [Accepted: 09/13/2018] [Indexed: 12/16/2022]
Abstract
Myocardial remodeling and dysfunction caused by accelerated oxidative damage is a widely reported phenomenon within a diabetic state. Altered myocardial substrate preference appears to be the major cause of enhanced oxidative stress-mediated cell injury within a diabetic heart. During this process, exacerbated free fatty acid flux causes an abnormal increase in mitochondrial membrane potential leading to the overproduction of free radical species and subsequent cell damage. Uncoupling proteins (UCPs) are expressed within the myocardium and can protect against free radical damage by modulating mitochondrial respiration, leading to reduced production of reactive oxygen species. Moreover, transgenic animals lacking UCPs have been shown to be more susceptible to oxidative damage and display reduced cardiac function when compared to wild type animals. This suggests that tight regulation of UCPs is necessary for normal cardiac function and in the prevention of diabetes-induced oxidative damage. This review aims to enhance our understanding of the pathophysiological mechanisms relating to the role of UCPs in a diabetic heart, and further discuss known pharmacological compounds and hormones that can protect a diabetic heart through the modulation of UCPs.
Collapse
|
29
|
Metformin prevents the development of severe chronic kidney disease and its associated mineral and bone disorder. Kidney Int 2018; 94:102-113. [DOI: 10.1016/j.kint.2018.01.027] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 01/17/2018] [Accepted: 01/25/2018] [Indexed: 12/15/2022]
|
30
|
Chen Y, Guo Z, Peng X, Xie W, Chen L, Tan Z. Nimodipine represses AMPK phosphorylation and excessive autophagy after chronic cerebral hypoperfusion in rats. Brain Res Bull 2018; 140:88-96. [PMID: 29625150 DOI: 10.1016/j.brainresbull.2018.03.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 01/29/2018] [Accepted: 03/30/2018] [Indexed: 12/29/2022]
Abstract
Chronic cerebral hypofusion (CCH) after bilateral carotid artery occlusion (2VO) causes cognitive damage and neuronal degeneration in the cortex and hippocampal CA1 area, and influences the oxygen and glucose supply in the brain which often results in metabolic alterations and oxidative stress. AMP-activated protein kinase (AMPK) phosphorylation, a sensor of cellular energy status, directs metabolic adaptation to support cellular growth and survival after CCH. Autophagy is also likely to be involved in metabolic adaptation and plays an important role in neuronal deterioration and cognitive decline after CCH. Nimodipine, an L-type calcium channel antagonist, has been reported to exert neuroprotective effects. However, the potential role of nimodipine in autophagy and the energy sensing AMPK signal is not well understood. In addition, little is known about the relationship between autophagy and AMPK signal. Here, we designed a way to evaluate these issues. Adult male Wistar rats were subjected to 2VO and randomly divided into three groups: the Vehicle (2VO), Nimodipine (2VO + nimodipine 10 mg/kg) groups. A third group served as sham controls. Each group was investigated at 2 and 4 weeks post gavage and tested using the Morris water maze. The activities of LC3B and AMPK signal were examined using immunohistochemistry and western blotting. Nimodipine significantly alleviated spatial learning and memory impairments and the number of lesion neurons. At 2 weeks of durg administration, these drug effects, suppressing AMPK activation and excessive autophagy, were more pronounced at the cortex than at hippocampal CA1 area. The effects of nimodipine were significant in the hippocampal CA1 area after 4 weeks of administration. Furthermore, nimodipine inhibited expression of eIF2α/ATF4 signaling related to energy deficit stress in 2VO rats. These results suggest that excessive autophagy has promoted neuronal and tissue injury after 2VO in rats. Nimodipine protected the brain from CCH by inhibiting the autophagy activity. The p-AMPK and eIF2α/ATF4 pathway is likely part of an integrated pro-autophagy signaling network after CCH.
Collapse
Affiliation(s)
- Yan Chen
- Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, Hubei 430061, China; Hubei University of Chinese Medicine, Wuhan, Hubei 430061, China; Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, Hubei 430015, China
| | - Zhenli Guo
- Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, Hubei 430015, China
| | - Xingming Peng
- Hubei University of Chinese Medicine, Wuhan, Hubei 430061, China
| | - Wenting Xie
- Hubei University of Chinese Medicine, Wuhan, Hubei 430061, China
| | - Lizhu Chen
- Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, Hubei 430015, China
| | - Zihu Tan
- Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, Hubei 430061, China; Hubei Province Academy of Traditional Chinese Medicine, Wuhan, Hubei 430074, China.
| |
Collapse
|
31
|
Samidurai A, Salloum FN, Durrant D, Chernova OB, Kukreja RC, Das A. Chronic treatment with novel nanoformulated micelles of rapamycin, Rapatar, protects diabetic heart against ischaemia/reperfusion injury. Br J Pharmacol 2017; 174:4771-4784. [PMID: 28967097 PMCID: PMC5727242 DOI: 10.1111/bph.14059] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 08/17/2017] [Accepted: 09/13/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE Enhanced mammalian target of rapamycin (mTOR) signalling contributes to the pathogenesis of diabetes and plays a critical role in myocardial ischaemia/reperfusion (I/R) injury. Rapatar is a novel nanoformulated micellar of rapamycin, a putative inhibitor of mTOR that has been rationally designed to increase water solubility of rapamycin to facilitate p.o. administration and enhance bioavailability. We examined the effect of Rapatar on the metabolic status and protection against myocardial I/R injury in type 2 diabetic mice. EXPERIMENTAL APPROACH Adult male db/db mice were treated daily for 10 weeks with Rapatar (0.75 mg·kg-1 ·day-1 , p.o.) or vehicle. Isolated hearts were connected to a Langendorff perfusion system and subjected to global ischaemia (30 min) and reperfusion (1 h). KEY RESULTS Rapatar reduced fasting plasma glucose and triglyceride levels, prevented the gain in body weight and also improved glucose tolerance and insulin sensitivity in db/db mice compared with control. Cardiac function was improved following Rapatar treatment in db/db mice. Myocardial infarct size was reduced in Rapatar-treated mice with improved post-ischaemic rate-force product. Western blot analyses demonstrated a significant inhibition of phosphorylation of ribosomal protein S6 (downstream target of mTORC1), but not Akt (Ser473 , target of mTORC2) following chronic treatment with Rapatar. Rapatar also induced phosphorylation of AMPK, STAT3, ERK1/2 and glycogen synthase kinase 3β, without interfering with phosphorylation of p38. CONCLUSION AND IMPLICATIONS Our studies indicate that chronic treatment with Rapatar improves metabolic status and cardiac function with a reduction of infarct size following myocardial I/R injury in diabetic mice.
Collapse
Affiliation(s)
- Arun Samidurai
- Pauley Heart Center, Division of CardiologyVirginia Commonwealth UniversityRichmondVAUSA
| | - Fadi N Salloum
- Pauley Heart Center, Division of CardiologyVirginia Commonwealth UniversityRichmondVAUSA
| | - David Durrant
- Pauley Heart Center, Division of CardiologyVirginia Commonwealth UniversityRichmondVAUSA
| | | | - Rakesh C Kukreja
- Pauley Heart Center, Division of CardiologyVirginia Commonwealth UniversityRichmondVAUSA
| | - Anindita Das
- Pauley Heart Center, Division of CardiologyVirginia Commonwealth UniversityRichmondVAUSA
| |
Collapse
|
32
|
Biochemical targets of drugs mitigating oxidative stress via redox-independent mechanisms. Biochem Soc Trans 2017; 45:1225-1252. [PMID: 29101309 DOI: 10.1042/bst20160473] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 09/24/2017] [Accepted: 09/26/2017] [Indexed: 12/13/2022]
Abstract
Acute or chronic oxidative stress plays an important role in many pathologies. Two opposite approaches are typically used to prevent the damage induced by reactive oxygen and nitrogen species (RONS), namely treatment either with antioxidants or with weak oxidants that up-regulate endogenous antioxidant mechanisms. This review discusses options for the third pharmacological approach, namely amelioration of oxidative stress by 'redox-inert' compounds, which do not inactivate RONS but either inhibit the basic mechanisms leading to their formation (i.e. inflammation) or help cells to cope with their toxic action. The present study describes biochemical targets of many drugs mitigating acute oxidative stress in animal models of ischemia-reperfusion injury or N-acetyl-p-aminophenol overdose. In addition to the pro-inflammatory molecules, the targets of mitigating drugs include protein kinases and transcription factors involved in regulation of energy metabolism and cell life/death balance, proteins regulating mitochondrial permeability transition, proteins involved in the endoplasmic reticulum stress and unfolded protein response, nuclear receptors such as peroxisome proliferator-activated receptors, and isoprenoid synthesis. The data may help in identification of oxidative stress mitigators that will be effective in human disease on top of the current standard of care.
Collapse
|
33
|
Karam BS, Chavez-Moreno A, Koh W, Akar JG, Akar FG. Oxidative stress and inflammation as central mediators of atrial fibrillation in obesity and diabetes. Cardiovasc Diabetol 2017; 16:120. [PMID: 28962617 PMCID: PMC5622555 DOI: 10.1186/s12933-017-0604-9] [Citation(s) in RCA: 329] [Impact Index Per Article: 41.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 09/22/2017] [Indexed: 02/07/2023] Open
Abstract
Atrial fibrillation (AF) is the most common sustained cardiac arrhythmia in humans. Several risk factors promote AF, among which diabetes mellitus has emerged as one of the most important. The growing recognition that obesity, diabetes and AF are closely intertwined disorders has spurred major interest in uncovering their mechanistic links. In this article we provide an update on the growing evidence linking oxidative stress and inflammation to adverse atrial structural and electrical remodeling that leads to the onset and maintenance of AF in the diabetic heart. We then discuss several therapeutic strategies to improve atrial excitability by targeting pathways that control oxidative stress and inflammation.
Collapse
Affiliation(s)
- Basil S Karam
- Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Wonjoon Koh
- Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joseph G Akar
- Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Fadi G Akar
- Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
34
|
Youssef N, Campbell S, Barr A, Gandhi M, Hunter B, Dolinsky V, Dyck JRB, Clanachan AS, Light PE. Hearts lacking plasma membrane KATP channels display changes in basal aerobic metabolic substrate preference and AMPK activity. Am J Physiol Heart Circ Physiol 2017; 313:H469-H478. [DOI: 10.1152/ajpheart.00612.2016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 06/13/2017] [Accepted: 06/13/2017] [Indexed: 12/12/2022]
Abstract
Cardiac ATP-sensitive K+ (KATP) channels couple changes in cellular metabolism to membrane excitability and are activated during metabolic stress, although under basal aerobic conditions, KATP channels are thought to be predominately closed. Despite intense research into the roles of KATP channels during metabolic stress, their contribution to aerobic basal cardiac metabolism has not been previously investigated. Hearts from Kir6.2+/+ and Kir6.2−/− mice were perfused in working mode, and rates of glycolysis, fatty acid oxidation, and glucose oxidation were measured. Changes in activation/expression of proteins regulating metabolism were probed by Western blot analysis. Despite cardiac mechanical function and metabolic efficiency being similar in both groups, hearts from Kir6.2−/− mice displayed an approximately twofold increase in fatty acid oxidation and a 0.45-fold reduction in glycolytic rates but similar glucose oxidation rates compared with hearts from Kir6.2+/+ mice. Kir6.2−/− hearts also possessed elevated levels of activated AMP-activated protein kinase (AMPK), higher glycogen content, and reduced mitochondrial density. Moreover, activation of AMPK by isoproterenol or diazoxide was significantly blunted in Kir6.2−/− hearts. These data indicate that KATP channel ablation alters aerobic basal cardiac metabolism. The observed increase in fatty acid oxidation and decreased glycolysis before any metabolic insult may contribute to the poor recovery observed in Kir6.2−/− hearts in response to exercise or ischemia-reperfusion injury. Therefore, KATP channels may play an important role in the regulation of cardiac metabolism through AMPK signaling. NEW & NOTEWORTHY In this study, we show that genetic ablation of plasma membrane ATP-sensitive K+ channels results in pronounced changes in cardiac metabolic substrate preference and AMP-activated protein kinase activity. These results suggest that ATP-sensitive K+ channels may play a novel role in regulating metabolism in addition to their well-documented effects on ionic homeostasis during periods of stress.
Collapse
Affiliation(s)
- Nermeen Youssef
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Scott Campbell
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Amy Barr
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Manoj Gandhi
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Beth Hunter
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Vernon Dolinsky
- Children’s Hospital Research Institute of Manitoba, Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jason R. B. Dyck
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; and
| | - Alexander S. Clanachan
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Peter E. Light
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
35
|
Cao Y, Bojjireddy N, Kim M, Li T, Zhai P, Nagarajan N, Sadoshima J, Palmiter RD, Tian R. Activation of γ2-AMPK Suppresses Ribosome Biogenesis and Protects Against Myocardial Ischemia/Reperfusion Injury. Circ Res 2017; 121:1182-1191. [PMID: 28835357 DOI: 10.1161/circresaha.117.311159] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 08/17/2017] [Accepted: 08/23/2017] [Indexed: 11/16/2022]
Abstract
RATIONALE AMPK (AMP-activated protein kinase) is a heterotrimeric protein that plays an important role in energy homeostasis and cardioprotection. Two isoforms of each subunit are expressed in the heart, but the isoform-specific function of AMPK remains unclear. OBJECTIVE We sought to determine the role of γ2-AMPK in cardiac stress response using bioengineered cell lines and mouse models containing either isoform of the γ-subunit in the heart. METHODS AND RESULTS We found that γ2 but not γ1 or γ3 subunit translocated into nucleus on AMPK activation. Nuclear accumulation of AMPK complexes containing γ2-subunit phosphorylated and inactivated RNA Pol I (polymerase I)-associated transcription factor TIF-IA at Ser-635, precluding the assembly of transcription initiation complexes for rDNA. The subsequent downregulation of pre-rRNA level led to attenuated endoplasmic reticulum (ER) stress and cell death. Deleting γ2-AMPK led to increases in pre-rRNA level, ER stress markers, and cell death during glucose deprivation, which could be rescued by inhibition of rRNA processing or ER stress. To study the function of γ2-AMPK in the heart, we generated a mouse model with cardiac-specific deletion of γ2-AMPK (cardiac knockout [cKO]). Although the total AMPK activity was unaltered in cKO hearts because of upregulation of γ1-AMPK, the lack of γ2-AMPK sensitizes the heart to myocardial ischemia/reperfusion injury. The cKO failed to suppress pre-rRNA level during ischemia/reperfusion and showed a greater infarct size. Conversely, cardiac-specific overexpression of γ2-AMPK decreased ribosome biosynthesis and ER stress during ischemia/reperfusion insult, and the infarct size was reduced. CONCLUSIONS The γ2-AMPK translocates into the nucleus to suppress pre-rRNA transcription and ribosome biosynthesis during stress, thus ameliorating ER stress and cell death. Increased γ2-AMPK activity is required to protect against ischemia/reperfusion injury. Our study reveals an isoform-specific function of γ2-AMPK in modulating ribosome biosynthesis, cell survival, and cardioprotection.
Collapse
Affiliation(s)
- Yang Cao
- From the Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center (Y.C., N.B., M.K., T.L., R.T.) and Department of Biochemistry (R.D.P.), University of Washington, Seattle; Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark (P.Z., N.N., J.S.); and Howard Hughes Medical Institute, Seattle, WA (R.D.P.)
| | - Naveen Bojjireddy
- From the Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center (Y.C., N.B., M.K., T.L., R.T.) and Department of Biochemistry (R.D.P.), University of Washington, Seattle; Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark (P.Z., N.N., J.S.); and Howard Hughes Medical Institute, Seattle, WA (R.D.P.)
| | - Maengjo Kim
- From the Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center (Y.C., N.B., M.K., T.L., R.T.) and Department of Biochemistry (R.D.P.), University of Washington, Seattle; Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark (P.Z., N.N., J.S.); and Howard Hughes Medical Institute, Seattle, WA (R.D.P.)
| | - Tao Li
- From the Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center (Y.C., N.B., M.K., T.L., R.T.) and Department of Biochemistry (R.D.P.), University of Washington, Seattle; Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark (P.Z., N.N., J.S.); and Howard Hughes Medical Institute, Seattle, WA (R.D.P.)
| | - Peiyong Zhai
- From the Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center (Y.C., N.B., M.K., T.L., R.T.) and Department of Biochemistry (R.D.P.), University of Washington, Seattle; Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark (P.Z., N.N., J.S.); and Howard Hughes Medical Institute, Seattle, WA (R.D.P.)
| | - Narayani Nagarajan
- From the Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center (Y.C., N.B., M.K., T.L., R.T.) and Department of Biochemistry (R.D.P.), University of Washington, Seattle; Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark (P.Z., N.N., J.S.); and Howard Hughes Medical Institute, Seattle, WA (R.D.P.)
| | - Junichi Sadoshima
- From the Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center (Y.C., N.B., M.K., T.L., R.T.) and Department of Biochemistry (R.D.P.), University of Washington, Seattle; Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark (P.Z., N.N., J.S.); and Howard Hughes Medical Institute, Seattle, WA (R.D.P.)
| | - Richard D Palmiter
- From the Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center (Y.C., N.B., M.K., T.L., R.T.) and Department of Biochemistry (R.D.P.), University of Washington, Seattle; Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark (P.Z., N.N., J.S.); and Howard Hughes Medical Institute, Seattle, WA (R.D.P.)
| | - Rong Tian
- From the Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center (Y.C., N.B., M.K., T.L., R.T.) and Department of Biochemistry (R.D.P.), University of Washington, Seattle; Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark (P.Z., N.N., J.S.); and Howard Hughes Medical Institute, Seattle, WA (R.D.P.).
| |
Collapse
|
36
|
Dludla PV, Joubert E, Muller CJF, Louw J, Johnson R. Hyperglycemia-induced oxidative stress and heart disease-cardioprotective effects of rooibos flavonoids and phenylpyruvic acid-2- O-β-D-glucoside. Nutr Metab (Lond) 2017; 14:45. [PMID: 28702068 PMCID: PMC5504778 DOI: 10.1186/s12986-017-0200-8] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 06/23/2017] [Indexed: 12/15/2022] Open
Abstract
Diabetic patients are at an increased risk of developing heart failure when compared to their non-diabetic counter parts. Accumulative evidence suggests chronic hyperglycemia to be central in the development of myocardial infarction in these patients. At present, there are limited therapies aimed at specifically protecting the diabetic heart at risk from hyperglycemia-induced injury. Oxidative stress, through over production of free radical species, has been hypothesized to alter mitochondrial function and abnormally augment the activity of the NADPH oxidase enzyme system resulting in accelerated myocardial injury within a diabetic state. This has led to a dramatic increase in the exploration of plant-derived materials known to possess antioxidative properties. Several edible plants contain various natural constituents, including polyphenols that may counteract oxidative-induced tissue damage through their modulatory effects of intracellular signaling pathways. Rooibos, an indigenous South African plant, well-known for its use as herbal tea, is increasingly studied for its metabolic benefits. Prospective studies linking diet rich in polyphenols from rooibos to reduced diabetes associated cardiovascular complications have not been extensively assessed. Aspalathin, a flavonoid, and phenylpyruvic acid-2-O-β-D-glucoside, a phenolic precursor, are some of the major compounds found in rooibos that can ameliorate hyperglycemia-induced cardiomyocyte damage in vitro. While the latter has demonstrated potential to protect against cell apoptosis, the proposed mechanism of action of aspalathin is linked to its capacity to enhance the expression of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) expression, an intracellular antioxidant response element. Thus, here we review literature on the potential cardioprotective properties of flavonoids and a phenylpropenoic acid found in rooibos against diabetes-induced oxidative injury.
Collapse
Affiliation(s)
- Phiwayinkosi V Dludla
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, P.O. Box 19070, Tygerberg, 7505 South Africa.,Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Elizabeth Joubert
- Plant Bioactives Group, Post-Harvest and Wine Technology Division, Agricultural Research Council (ARC) Infruitec- Nietvoorbij, Stellenbosch, South Africa.,Department of Food Science, Stellenbosch University, Stellenbosch, South Africa
| | - Christo J F Muller
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, P.O. Box 19070, Tygerberg, 7505 South Africa.,Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg, South Africa.,Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, South Africa
| | - Johan Louw
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, P.O. Box 19070, Tygerberg, 7505 South Africa.,Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, South Africa
| | - Rabia Johnson
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, P.O. Box 19070, Tygerberg, 7505 South Africa.,Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg, South Africa
| |
Collapse
|
37
|
Li J, Zhong L, Wang F, Zhu H. Dissecting the role of AMP-activated protein kinase in human diseases. Acta Pharm Sin B 2017; 7:249-259. [PMID: 28540163 PMCID: PMC5430814 DOI: 10.1016/j.apsb.2016.12.003] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/12/2016] [Accepted: 11/17/2016] [Indexed: 12/18/2022] Open
Abstract
AMP-activated protein kinase (AMPK), known as a sensor and a master of cellular energy balance, integrates various regulatory signals including anabolic and catabolic metabolic processes. Accompanying the application of genetic methods and a plethora of AMPK agonists, rapid progress has identified AMPK as an attractive therapeutic target for several human diseases, such as cancer, type 2 diabetes, atherosclerosis, myocardial ischemia/reperfusion injury and neurodegenerative disease. The role of AMPK in metabolic and energetic modulation both at the intracellular and whole body levels has been reviewed elsewhere. In the present review, we summarize and update the paradoxical role of AMPK implicated in the diseases mentioned above and put forward the challenge encountered. Thus it will be expected to provide important clues for exploring rational methods of intervention in human diseases.
Collapse
Affiliation(s)
- Jin Li
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| | - Liping Zhong
- Life Science College of Tarim University, Xinjiang 843300, China
| | - Fengzhong Wang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
- Corresponding author. Tel./fax: +86 10 62810295.
| | - Haibo Zhu
- State Key Laboratory for Bioactive Substances and Functions of Natural Medicines, Beijing 100050, China
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Beijing 100050, China
- Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- Corresponding author at: Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China. Tel./fax: +86 10 63188106.
| |
Collapse
|
38
|
Li T, Jiang S, Yang Z, Ma Z, Yi W, Wang D, Yang Y. Targeting the energy guardian AMPK: another avenue for treating cardiomyopathy? Cell Mol Life Sci 2017; 74:1413-1429. [PMID: 27815596 PMCID: PMC11107559 DOI: 10.1007/s00018-016-2407-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 10/25/2016] [Accepted: 11/01/2016] [Indexed: 12/11/2022]
Abstract
5'-AMP-activated protein kinase (AMPK) is a pivotal regulator of endogenous defensive molecules in various pathological processes. The AMPK signaling regulates a variety of intracellular intermedial molecules involved in biological reactions, including glycogen metabolism, protein synthesis, and cardiac fibrosis, in response to hypertrophic stimuli. Studies have revealed that the activation of AMPK performs a protective role in cardiovascular diseases, whereas its function in cardiac hypertrophy and cardiomyopathy remains elusive and poorly understood. In view of the current evidence of AMPK, we introduce the biological information of AMPK and cardiac hypertrophy as well as some upstream activators of AMPK. Next, we discuss two important types of cardiomyopathy involving AMPK, RKAG2 cardiomyopathy, and hypertrophic cardiomyopathy. Eventually, therapeutic research, genetic screening, conflicts, obstacles, challenges, and potential directions are also highlighted in this review, aimed at providing a comprehensive understanding of AMPK for readers.
Collapse
Affiliation(s)
- Tian Li
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an, 710032, China
| | - Shuai Jiang
- Department of Aerospace Medicine, The Fourth Military Medical University, 169 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
| | - Zhiqiang Ma
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an, 710038, China
| | - Wei Yi
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710038, China
| | - Dongjin Wang
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China.
| | - Yang Yang
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China.
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an, 710032, China.
| |
Collapse
|
39
|
N-Acetylcysteine Attenuates Diabetic Myocardial Ischemia Reperfusion Injury through Inhibiting Excessive Autophagy. Mediators Inflamm 2017; 2017:9257291. [PMID: 28265179 PMCID: PMC5317145 DOI: 10.1155/2017/9257291] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 12/14/2016] [Accepted: 12/28/2016] [Indexed: 01/03/2023] Open
Abstract
Background. Excessive autophagy is a major mechanism of myocardial ischemia reperfusion injury (I/RI) in diabetes with enhanced oxidative stress. Antioxidant N-acetylcysteine (NAC) reduces myocardial I/RI. It is unknown if inhibition of autophagy may represent a mechanism whereby NAC confers cardioprotection in diabetes. Methods and Results. Diabetes was induced in Sprague-Dawley rats with streptozotocin and they were treated without or with NAC (1.5 g/kg/day) for four weeks before being subjected to 30-minute coronary occlusion and 2-hour reperfusion. The results showed that cardiac levels of 15-F2t-Isoprostane were increased and that autophagy was evidenced as increases in ratio of LC3 II/I and protein P62 and AMPK and mTOR expressions were significantly increased in diabetic compared to nondiabetic rats, concomitant with increased postischemic myocardial infarct size and CK-MB release but decreased Akt and eNOS activation. Diabetes was also associated with increased postischemic apoptotic cell death manifested as increases in TUNEL positive cells, cleaved-caspase-3, and ratio of Bax/Bcl-2 protein expression. NAC significantly attenuated I/RI-induced increases in oxidative stress and cardiac apoptosis, prevented postischemic autophagy formation in diabetes, and reduced postischemic myocardial infarction (all p < 0.05). Conclusions. NAC confers cardioprotection against diabetic heart I/RI primarily through inhibiting excessive autophagy which might be a major mechanism why diabetic hearts are less tolerant to I/RI.
Collapse
|
40
|
Xue RQ, Sun L, Yu XJ, Li DL, Zang WJ. Vagal nerve stimulation improves mitochondrial dynamics via an M 3 receptor/CaMKKβ/AMPK pathway in isoproterenol-induced myocardial ischaemia. J Cell Mol Med 2017; 21:58-71. [PMID: 27491814 PMCID: PMC5192749 DOI: 10.1111/jcmm.12938] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 06/27/2016] [Indexed: 12/25/2022] Open
Abstract
Mitochondrial dynamics-fission and fusion-are associated with ischaemic heart disease (IHD). This study explored the protective effect of vagal nerve stimulation (VNS) against isoproterenol (ISO)-induced myocardial ischaemia in a rat model and tested whether VNS plays a role in preventing disorders of mitochondrial dynamics and function. Isoproterenol not only caused cardiac injury but also increased the expression of mitochondrial fission proteins [dynamin-related peptide1 (Drp1) and mitochondrial fission protein1 (Fis-1)) and decreased the expression of fusion proteins (optic atrophy-1 (OPA1) and mitofusins1/2 (Mfn1/2)], thereby disrupting mitochondrial dynamics and leading to increase in mitochondrial fragments. Interestingly, VNS restored mitochondrial dynamics through regulation of Drp1, Fis-1, OPA1 and Mfn1/2; enhanced ATP content and mitochondrial membrane potential; reduced mitochondrial permeability transition pore (MPTP) opening; and improved mitochondrial ultrastructure and size. Furthermore, VNS reduced the size of the myocardial infarction and ameliorated cardiomyocyte apoptosis and cardiac dysfunction induced by ISO. Moreover, VNS activated AMP-activated protein kinase (AMPK), which was accompanied by phosphorylation of Ca2+ /calmodulin-dependent protein kinase kinase β (CaMKKβ) during myocardial ischaemia. Treatment with subtype-3 of muscarinic acetylcholine receptor (M3 R) antagonist 4-diphenylacetoxy-N-methylpiperidine methiodide or AMPK inhibitor Compound C abolished the protective effects of VNS on mitochondrial dynamics and function, suggesting that M3 R/CaMKKβ/AMPK signalling are involved in mediating beneficial effects of VNS. This study demonstrates that VNS modulates mitochondrial dynamics and improves mitochondrial function, possibly through the M3 R/CaMKKβ/AMPK pathway, to attenuate ISO-induced cardiac damage in rats. Targeting mitochondrial dynamics may provide a novel therapeutic strategy in IHD.
Collapse
Affiliation(s)
- Run-Qing Xue
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Lei Sun
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Xiao-Jiang Yu
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Dong-Ling Li
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Wei-Jin Zang
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| |
Collapse
|
41
|
Lu YM, Jiao B, Lee J, Zhang L, Yu ZB. Simulated microgravity increases myocardial susceptibility to ischemia-reperfusion injury via a deficiency of AMP-activated protein kinase. Can J Physiol Pharmacol 2016; 95:59-71. [PMID: 27831744 DOI: 10.1139/cjpp-2015-0456] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Gravitation is an important factor in maintaining cardiac contractility. Our study investigated whether simulated microgravity increases myocardial susceptibility to ischemia-reperfusion (IR) injury. Using the Langendorff-perfused heart model with 300 beats/min pacing, 4-week tail suspension (SUS) and control (CON) male Sprague-Dawley rats (n = 10 rats/group) were subjected to 60 min of left anterior descending coronary artery (LAD) occlusion followed by 120 min of reperfusion. Left ventricular end-systolic pressure (LVESP), left ventricular end-diastolic pressure (LVEDP), creatine kinase (CK) and lactate dehydrogenase (LDH) activity, and infarct size were assessed. Data demonstrated that there were significantly increased LVEDP, CK, LDH, and infarct size in SUS compared with CON (P < 0.05), accompanied by decreased LVESP (P < 0.05). Furthermore, TUNEL-positive cardiomyocytes were higher in SUS than that in CON (P < 0.01), and AMP-activated protein kinase (AMPK) phosphorylation and Bcl-2/Bax in SUS were less compared with CON (P < 0.05). Similarly, isolated hearts pre-treated with A-769662 exhibited better recovery of cardiac function, increased AMPK phosphorylation, and reduced necrosis and apoptosis. Furthermore, AMPKα protein showed a significant suppression in 4-week hindlimb unweighting rats. These results suggest that AMPK deficiency increases myocardial susceptibility to IR injury in rats subjected to simulated microgravity.
Collapse
Affiliation(s)
- Yuan-Ming Lu
- a Department of Aerospace Physiology, Fourth Military Medical University, Key Laboratory of Aerospace Medicine, Ministry of China, Xi'an, 710032, China
| | - Bo Jiao
- a Department of Aerospace Physiology, Fourth Military Medical University, Key Laboratory of Aerospace Medicine, Ministry of China, Xi'an, 710032, China
| | - Jun Lee
- b Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Lin Zhang
- a Department of Aerospace Physiology, Fourth Military Medical University, Key Laboratory of Aerospace Medicine, Ministry of China, Xi'an, 710032, China
| | - Zhi-Bin Yu
- a Department of Aerospace Physiology, Fourth Military Medical University, Key Laboratory of Aerospace Medicine, Ministry of China, Xi'an, 710032, China
| |
Collapse
|
42
|
Jiang S, Tian S, Wu X, Tao Y, Jiang D. Effect of AMP-activated protein kinase activation on cardiac fibroblast proliferation induced by coxsackievirus B3. Exp Ther Med 2016; 11:2547-2552. [PMID: 27284347 DOI: 10.3892/etm.2016.3174] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 03/01/2016] [Indexed: 12/31/2022] Open
Abstract
Excessive fibroblast proliferation and collagen production are the major pathogenic mechanisms in the progression of viral myocarditis, which is most frequently associated with infection by coxsackievirus B3 (CVB3). AMP-activated protein kinase (AMPK) has been confirmed to be involved in the progression of myocardial remodeling. However, it remains unclear whether AMPK has an effect on CVB3-induced cardiac fibroblast proliferation. In the present study, the effects of AMPK on cardiac fibroblast proliferation and collagen secretion induced by CVB3 were investigated. Proliferation of neonatal cardiac fibroblasts was determined by cell counting and detection of newly synthesized DNA in cells, and the proportion of cells in the S phase was measured. Hydroxyproline ELISA was used to detect collagen secretion. Phosphorylation of AMPKα-Thr172 was evaluated by western blotting. It was found that neonatal cardiac fibroblasts were clearly proliferating markedly and secreting collagen at 24 h after CVB3 infection, and peaked at 48 h. These effects were inhibited following pretreatment of the fibroblasts with 5-aminoimidazole-4-carboxamide-ribonucleoside (AICAR), a specific AMPK activator, for 2 h prior to CVB3 infection. However, if the cells were preincubated with compound C for 30 min, the inhibitive effects of AICAR were reversed. Western blotting results indicated that AMPK α-Thr172 phosphorylation was increased by AICAR and attenuated by Compound C. The results of the present study suggest that CVB3 infection increases cardiac fibroblast proliferation and collagen secretion, and that these phenomena can be inhibited by activated AMPK. These findings contribute to our understanding of AMPK function and the future design of therapeutic approaches for the treatment of cardiac fibrosis caused by chronic viral infection, such as CVB3-induced myocarditis.
Collapse
Affiliation(s)
- Shengyang Jiang
- Department of Cardiology, The Third Affiliated Hospital of Nantong University, Wuxi Institute of Integrated Traditional Chinese and Western Medicine, Wuxi, Jiangsu 214041, P.R. China
| | - Shunli Tian
- Department of Geratology, Tianjin Geriatric Institute, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Xueming Wu
- Department of Cardiology, The Third Affiliated Hospital of Nantong University, Wuxi Institute of Integrated Traditional Chinese and Western Medicine, Wuxi, Jiangsu 214041, P.R. China
| | - Yijia Tao
- Department of Cardiology, The Third Affiliated Hospital of Nantong University, Wuxi Institute of Integrated Traditional Chinese and Western Medicine, Wuxi, Jiangsu 214041, P.R. China
| | - Donglin Jiang
- Clinical Central Laboratory, The Third Affiliated Hospital of Nantong University, Wuxi Institute of Integrated Traditional Chinese and Western Medicine, Wuxi, Jiangsu 214041, P.R. China
| |
Collapse
|
43
|
Abstract
Aims There are reports that ataxia telangiectasia mutated (ATM) can activate the AMP-activated protein kinase (AMPK) and also Akt, two kinases that play integral parts in cardioprotection and metabolic function. We hypothesized that chloroquine and resveratrol, both known ATM activators, would also activate AMPK and Akt. Main methods Phosphorylation of AMPK and Akt was assessed after C2C12 myotubes were exposed to chloroquine or resveratrol. Additional experiments were done in cells expressing shRNA against ATM or in the presence of the ATM inhibitor KU55933. The effects of chloroquine on intracellular calcium were assessed with the fluorescent probe Calcium Green-1 AM. Key findings 0.5 mM chloroquine increased AMPK phosphorylation by nearly four-fold (P < 0.05), and 0.25 mM chloroquine roughly doubled Akt phosphorylation (P < 0.05). Chloroquine also increased autophosphorylation of ATM by ∼50% (P < 0.05). Resveratrol (0.15 mM) increased AMPK phosphorylation about three-fold (P < 0.05) but in contrast to chloroquine sharply decreased Akt phosphorylation. Chloroquine increased AMPK and Akt phosphorylation in myotubes expressing shRNA against ATM that reduced ATM protein levels by about 90%. Likewise, chloroquine-stimulated phosphorylation of AMPK and Akt and resveratrol-stimulated phosphorylation of AMPK were not altered by inhibition of ATM. Chloroquine decreased intracellular calcium by >50% concomitant with a decrease in glucose transport. Significance These ATM-independent effects of chloroquine on AMPK and Akt and the additional effect to decrease intracellular calcium are likely to partially underlie the positive metabolic effects of chloroquine that have been reported in the literature.
Collapse
|
44
|
Abstract
Improvements in healthcare and nutrition have generated remarkable increases in life expectancy worldwide. This is one of the greatest achievements of the modern world yet it also presents a grave challenge: as more people survive into later life, more also experience the diseases of old age, including type 2 diabetes (T2D), cardiovascular disease (CVD) and cancer. Developing new ways to improve health in the elderly is therefore a top priority for biomedical research. Although our understanding of the molecular basis of these morbidities has advanced rapidly, effective novel treatments are still lacking. Alternative drug development strategies are now being explored, such as the repurposing of existing drugs used to treat other diseases. This can save a considerable amount of time and money since the pharmacokinetics, pharmacodynamics and safety profiles of these drugs are already established, effectively enabling preclinical studies to be bypassed. Metformin is one such drug currently being investigated for novel applications. The present review provides a thorough and detailed account of our current understanding of the molecular pharmacology and signalling mechanisms underlying biguanide-protein interactions. It also focuses on the key role of the microbiota in regulating age-associated morbidities and a potential role for metformin to modulate its function. Research in this area holds the key to solving many of the mysteries of our current understanding of drug action and concerted effects to provide sustained and long-life health.
Collapse
|
45
|
AMPK in cardiac fibrosis and repair: Actions beyond metabolic regulation. J Mol Cell Cardiol 2016; 91:188-200. [PMID: 26772531 DOI: 10.1016/j.yjmcc.2016.01.001] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 12/28/2015] [Accepted: 01/04/2016] [Indexed: 02/06/2023]
Abstract
Fibrosis is a general term encompassing a plethora of pathologies that span all systems and is marked by increased deposition of collagen. Injury of variable etiology gives rise to complex cascades involving several cell-types and molecular signals, leading to the excessive accumulation of extracellular matrix that promotes fibrosis and eventually leads to organ failure. Cardiac fibrosis is a dynamic process associated notably with ischemia, hypertrophy, volume- and pressure-overload, aging and diabetes mellitus. It has profoundly deleterious consequences on the normal architecture and functioning of the myocardium and is associated with considerable mortality and morbidity. The AMP-activated protein kinase (AMPK) is a ubiquitously expressed cellular energy sensor and an essential component of the adaptive response to cardiomyocyte stress that occurs during ischemia. Nevertheless, its actions extend well beyond its energy-regulating role and it appears to possess an essential role in regulating fibrosis of the myocardium. In this review paper, we will summarize the main elements and crucial players of cardiac fibrosis. In addition, we will provide an overview of the diverse roles of AMPK in the heart and discuss in detail its implication in cardiac fibrosis. Lastly, we will highlight the recently published literature concerning AMPK-targeting current therapy and novel strategies aiming to attenuate fibrosis.
Collapse
|
46
|
Daskalopoulos EP, Dufeys C, Beauloye C, Bertrand L, Horman S. AMPK in Cardiovascular Diseases. EXPERIENTIA SUPPLEMENTUM (2012) 2016; 107:179-201. [PMID: 27812981 DOI: 10.1007/978-3-319-43589-3_8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This chapter summarizes the implication of AMP-activated protein kinase (AMPK) in the regulation of various physiological and pathological cellular events of great importance for the maintenance of cardiac function. These include the control of both metabolic and non-metabolic elements targeting the different cellular components of the cardiac tissue, i.e., cardiomyocytes, fibroblasts, and vascular cells. The description of the multifaceted action of the two AMPK catalytic isoforms, α1 and α2, emphasizes the general protective action of this protein kinase against the development of critical pathologies like myocardial ischemia, cardiac hypertrophy, diabetic cardiomyopathy, and heart failure.
Collapse
Affiliation(s)
- Evangelos P Daskalopoulos
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium.,Cardiovascular Research (Care) Institute, Athens, Ioannina, Greece
| | - Cécile Dufeys
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium
| | - Christophe Beauloye
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium.,Division of Cardiology, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Luc Bertrand
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium.
| | - Sandrine Horman
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium
| |
Collapse
|
47
|
AMPK is critical for mitochondrial function during reperfusion after myocardial ischemia. J Mol Cell Cardiol 2015; 91:104-13. [PMID: 26746142 DOI: 10.1016/j.yjmcc.2015.12.032] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Revised: 12/18/2015] [Accepted: 12/29/2015] [Indexed: 02/06/2023]
Abstract
AMP-activated kinase (AMPK) is a stress responsive kinase that regulates cellular metabolism and protects against cardiomyocyte injury during ischemia-reperfusion (IR). Mitochondria play an important role in cell survival, but the specific actions of activated AMPK in maintaining mitochondrial integrity and function during reperfusion are unknown. Thus, we assessed the consequences of AMPK inactivation on heart mitochondrial function during reperfusion. Mouse hearts expressing wild type (WT) or kinase-dead (KD) AMPK were studied. Mitochondria isolated from KD hearts during reperfusion had intact membrane integrity, but demonstrated reduced oxidative capacity, increased hydrogen peroxide production and decreased resistance to mitochondrial permeability transition pore opening compared to WT. KD hearts showed increased activation of the mitogen activated protein kinase kinase 4 (MKK4) and downstream c-Jun terminal kinase (JNK) and greater necrosis during reperfusion after coronary occlusion. Transgenic expression of mitochondrial catalase (MCAT) prevented the excessive cardiac JNK activation and attenuated the increased myocardial necrosis observed during reperfusion in KD mice. Inhibition of JNK increased the resistance of KD hearts to mPTP opening, contractile dysfunction and necrosis during IR. Thus, intrinsic activation of AMPK is critical to prevent excess mitochondrial reactive oxygen production and consequent JNK signaling during reperfusion, thereby protecting against mPTP opening, irreversible mitochondrial damage and myocardial injury.
Collapse
|
48
|
Wu X, Li Y, Wang Q, Li W, Feng Y. Effects of berberine and pomegranate seed oil on plasma phospholipid metabolites associated with risks of type 2 diabetes mellitus by U-HPLC/Q-TOF-MS. J Chromatogr B Analyt Technol Biomed Life Sci 2015; 1007:110-20. [DOI: 10.1016/j.jchromb.2015.11.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 11/03/2015] [Accepted: 11/04/2015] [Indexed: 11/26/2022]
|
49
|
Maiese K. Erythropoietin and diabetes mellitus. World J Diabetes 2015; 6:1259-1273. [PMID: 26516410 PMCID: PMC4620106 DOI: 10.4239/wjd.v6.i14.1259] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 08/25/2015] [Accepted: 09/28/2015] [Indexed: 02/05/2023] Open
Abstract
Erythropoietin (EPO) is a 30.4 kDa growth factor and cytokine that governs cell proliferation, immune modulation, metabolic homeostasis, vascular function, and cytoprotection. EPO is under investigation for the treatment of variety of diseases, but appears especially suited for the treatment of disorders of metabolism that include diabetes mellitus (DM). DM and the complications of this disease impact a significant portion of the global population leading to disability and death with currently limited therapeutic options. In addition to its utility for the treatment of anemia, EPO can improve cardiac function, reduce fatigue, and improve cognition in patients with DM as well as regulate cellular energy metabolism, obesity, tissue repair and regeneration, apoptosis, and autophagy in experimental models of DM. Yet, EPO can have adverse effects that involve the vasculature system and unchecked cellular proliferation. Critical to the cytoprotective capacity and the potential for a positive clinical outcome with EPO are the control of signal transduction pathways that include protein kinase B, the mechanistic target of rapamycin, Wnt signaling, mammalian forkhead transcription factors of the O class, silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae), and AMP activated protein kinase. Therapeutic strategies that can specifically target and control EPO and its signaling pathways hold great promise for the development of new and effective clinical treatments for DM and the complications of this disorder.
Collapse
|
50
|
Basnet S, Kozikowski A, Makaryus AN, Pekmezaris R, Zeltser R, Akerman M, Lesser M, Wolf-Klein G. Metformin and Myocardial Injury in Patients With Diabetes and ST-Segment Elevation Myocardial Infarction: A Propensity Score Matched Analysis. J Am Heart Assoc 2015; 4:e002314. [PMID: 26494519 PMCID: PMC4845135 DOI: 10.1161/jaha.115.002314] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Background Although animal studies have documented metformin's cardioprotective effects, the impact in humans remains elusive. The study objective was to explore the association between metformin and myocardial infarct size in patients with diabetes presenting with ST‐segment elevation myocardial infarction. Methods and Results Data extraction used the National Cardiovascular Data CathPCI Registry in all patients with diabetes aged >18 years presenting with ST‐segment elevation myocardial infarction at 2 academic medical centers from January 2010 to December 2013. The exposure of interest was ongoing metformin use before the event. Propensity score matching was used for the metformin and nonmetformin groups on key prognostic variables. All matched pairs had acceptable D scores of <10%, confirming an efficient matching procedure. The primary outcome was myocardial infarct size, reflected by peak serum creatine kinase–myocardial band, troponin T, and hospital discharge left ventricular ejection fraction. Of all 1726 ST‐segment elevation myocardial infarction cases reviewed, 493 patients had diabetes (28.5%), with 208 metformin users (42.1%) and 285 nonusers. Matched pairs analysis yielded 137 cases per group. The difference between metformin and nonmetformin groups was −18.1 ng/mL (95% CI −55.0 to 18.8; P=0.56) for total peak serum creatine kinase–myocardial band and −1.1 ng/mL (95% CI −2.8 to 0.5; P=0.41) for troponin T. Median discharge left ventricular ejection fraction in both groups was 45, and the difference between metformin and nonmetformin users was 0.7% (95% CI −2.2 to 3.6; P=0.99). Conclusions No statistically significant association of cardioprotection was found between metformin and myocardial infarct size in patients with diabetes and acute ST‐segment elevation myocardial infarction.
Collapse
Affiliation(s)
- Suresh Basnet
- Department of Critical Care Medicine, Valley Health System, Winchester, VA (S.B.)
| | - Andrzej Kozikowski
- Department of Medicine, North Shore-LIJ Health System, Great Neck, NY (A.K., R.P.)
| | - Amgad N Makaryus
- Department of Cardiology, North Shore-LIJ Health System, Manhasset, NY (A.N.M., R.Z.) Nassau University Medical Center, East Meadow, NY (A.N.M., R.Z.)
| | - Renee Pekmezaris
- Department of Medicine, North Shore-LIJ Health System, Great Neck, NY (A.K., R.P.)
| | - Roman Zeltser
- Department of Cardiology, North Shore-LIJ Health System, Manhasset, NY (A.N.M., R.Z.) Nassau University Medical Center, East Meadow, NY (A.N.M., R.Z.)
| | - Meredith Akerman
- Biostatistics Unit, The Feinstein Institute for Medical Research, North Shore-LIJ Health System, Manhasset, NY (M.A., M.L.)
| | - Martin Lesser
- Biostatistics Unit, The Feinstein Institute for Medical Research, North Shore-LIJ Health System, Manhasset, NY (M.A., M.L.)
| | - Gisele Wolf-Klein
- Department of Geriatrics, North Shore-LIJ Health System, New Hyde Park, NY (G.W.K.)
| |
Collapse
|