1
|
Rai AK, Sanghvi S, Muthukumaran NS, Chandrasekera D, Kadam A, Kishore J, Kyriazis ID, Tomar D, Ponnalagu D, Shettigar V, Khan M, Singh H, Goukassian D, Katare R, Garikipati VNS. Role of mitochondrial ribosomal protein L7/L12 (MRPL12) in diabetic ischemic heart disease. Free Radic Biol Med 2024; 222:531-538. [PMID: 38977138 DOI: 10.1016/j.freeradbiomed.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/19/2024] [Accepted: 07/04/2024] [Indexed: 07/10/2024]
Abstract
BACKGROUND Myocardial infarction (MI) is a significant cause of death in diabetic patients. Growing evidence suggests that mitochondrial dysfunction contributes to heart failure in diabetes. However, the molecular mechanisms of mitochondrial dysfunction mediating heart failure in diabetes are still poorly understood. METHODS We examined MRPL12 levels in right atrial appendage tissues from diabetic patients undergoing coronary artery bypass graft (CABG) surgery. Using AC-16 cells overexpressing MRPL12 under normal and hyperglycemic conditions we performed mitochondrial functional assays OXPHOS, bioenergetics, mitochondrial membrane potential, ATP production and cell death. RESULTS We observed elevated MRPL12 levels in heart tissue samples from diabetic patients with ischemic heart disease compared to non-diabetic patients. Overexpression of MRPL12 under hyperglycemic conditions did not affect oxidative phosphorylation (OXPHOS) levels, cellular ATP levels, or cardiomyocyte cell death. However, notable impairment in mitochondrial membrane potential (MMP) was observed under hyperglycemic conditions, along with alterations in both basal respiration oxygen consumption rate (OCR) and maximal respiratory capacity OCR. CONCLUSIONS Overall, our results suggest that MRPL12 may have a compensatory role in the diabetic myocardium with ischemic heart disease, suggesting that MRPL12 may implicate in the pathophysiology of MI in diabetes.
Collapse
MESH Headings
- Aged
- Animals
- Female
- Humans
- Male
- Middle Aged
- Adenosine Triphosphate/metabolism
- Atrial Appendage/metabolism
- Atrial Appendage/pathology
- Coronary Artery Bypass
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/complications
- Membrane Potential, Mitochondrial
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Mitochondria, Heart/genetics
- Mitochondrial Proteins/metabolism
- Mitochondrial Proteins/genetics
- Myocardial Ischemia/metabolism
- Myocardial Ischemia/pathology
- Myocardial Ischemia/genetics
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Oxidative Phosphorylation
- Ribosomal Proteins/metabolism
- Ribosomal Proteins/genetics
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Cell Cycle Proteins/genetics
- Cell Cycle Proteins/metabolism
Collapse
Affiliation(s)
- Amit Kumar Rai
- Aging + Cardiovascular Discovery Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, USA
| | - Shridhar Sanghvi
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Molecular Cellular and Developmental Biology, The Ohio State University, Columbus, OH, USA
| | | | - Dhananjie Chandrasekera
- Department of Physiology, HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Ashlesha Kadam
- Section of Cardiovascular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Jahnavi Kishore
- Aging + Cardiovascular Discovery Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, USA
| | - Ioannis D Kyriazis
- Department of Biology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Dhanendra Tomar
- Section of Cardiovascular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Devasena Ponnalagu
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Pharmacology, University of Washington, Seattle, WA, USA
| | - Vikram Shettigar
- Aging + Cardiovascular Discovery Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, USA
| | - Mahmood Khan
- Dorothy M. Davis Heart Lung and Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Harpreet Singh
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Dorothy M. Davis Heart Lung and Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - David Goukassian
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Rajesh Katare
- Department of Physiology, HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Venkata Naga Srikanth Garikipati
- Aging + Cardiovascular Discovery Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, USA; Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
2
|
Ott C. Mapping the interplay of immunoproteasome and autophagy in different heart failure phenotypes. Free Radic Biol Med 2024; 218:149-165. [PMID: 38570171 DOI: 10.1016/j.freeradbiomed.2024.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/25/2024] [Accepted: 03/30/2024] [Indexed: 04/05/2024]
Abstract
Proper protein degradation is required for cellular protein homeostasis and organ function. Particularly, in post-mitotic cells, such as cardiomyocytes, unbalanced proteolysis due to inflammatory stimuli and oxidative stress contributes to organ dysfunction. To ensure appropriate protein turnover, eukaryotic cells exert two main degradation systems, the ubiquitin-proteasome-system and the autophagy-lysosome-pathway. It has been shown that proteasome activity affects the development of cardiac dysfunction differently, depending on the type of heart failure. Studies analyzing the inducible subtype of the proteasome, the immunoproteasome (i20S), demonstrated that the i20S plays a double role in diseased hearts. While i20S subunits are increased in cardiac hypertrophy, atrial fibrillation and partly in myocarditis, the opposite applies to diabetic cardiomyopathy and ischemia/reperfusion injury. In addition, the i20S appears to play a role in autophagy modulation depending on heart failure phenotype. This review summarizes the current literature on the i20S in different heart failure phenotypes, emphasizing the two faces of i20S in injured hearts. A selection of established i20S inhibitors is introduced and signaling pathways linking the i20S to autophagy are highlighted. Mapping the interplay of the i20S and autophagy in different types of heart failure offers potential approaches for developing treatment strategies against heart failure.
Collapse
Affiliation(s)
- Christiane Ott
- German Institute of Human Nutrition Potsdam-Rehbruecke, Department of Molecular Toxicology, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.
| |
Collapse
|
3
|
Shen J, Yang H, Qiao X, Chen Y, Zheng L, Lin J, Lang J, Yu Q, Wang Z. The E3 ubiquitin ligase TRIM17 promotes gastric cancer survival and progression via controlling BAX stability and antagonizing apoptosis. Cell Death Differ 2023; 30:2322-2335. [PMID: 37697039 PMCID: PMC10589321 DOI: 10.1038/s41418-023-01221-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 08/20/2023] [Accepted: 08/31/2023] [Indexed: 09/13/2023] Open
Abstract
Tripartite motif 17 (TRIM17) belongs to a subfamily of the RING-type E3 ubiquitin ligases, and regulates several cellular processes and pathological conditions including cancer. However, its potential function in gastric cancer (GC) remains obscure. Here, we have found TRIM17 mRNA and protein levels are both upregulated in human GC compared with normal specimens, and TRIM17 upregulation indicates poor survival for GC patients. Functionally, TRIM17 was found to act as an oncogene by promoting the proliferation and survival of GC cell lines AGS and HGC-27. Mechanistically, TRIM17 acts to interact with BAX and promote its ubiquitination and proteasomal degradation, leading to a deficiency in BAX-dependent apoptosis in GC cells in the absence and presence of apoptosis stimuli. Moreover, TRIM17 and BAX expression levels are inversely correlated in human GC specimens. Our data thus suggest TRIM17 contributes to gastric cancer survival through regulating BAX protein stability and antagonizing apoptosis, which provides a promising therapeutic target for GC treatment and a biomarker for prognosis.
Collapse
Affiliation(s)
- Jiajia Shen
- Department of Biochemistry, Institute of Medicinal Biotecnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Hang Yang
- Department of Biochemistry, Institute of Medicinal Biotecnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Xinran Qiao
- Department of Biochemistry, Institute of Medicinal Biotecnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yang Chen
- Department of Biochemistry, Institute of Medicinal Biotecnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Liyun Zheng
- Department of Biochemistry, Institute of Medicinal Biotecnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jingyu Lin
- Department of Biochemistry, Institute of Medicinal Biotecnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jingyu Lang
- CAS_Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qiang Yu
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Agency for Science, Technology, and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Zhen Wang
- Department of Biochemistry, Institute of Medicinal Biotecnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
4
|
Li D, Son Y, Jang M, Wang S, Zhu W. Nanoparticle Based Cardiac Specific Drug Delivery. BIOLOGY 2023; 12:biology12010082. [PMID: 36671774 PMCID: PMC9856055 DOI: 10.3390/biology12010082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/06/2023]
Abstract
Heart failure secondary to myocardial injuries is a leading cause of death worldwide. Recently, a growing number of novel therapies have emerged for injured myocardium repairment. However, delivering therapeutic agents specifically to the injured heart remains a significant challenge. Nanoparticles are the most commonly used vehicles for targeted drug delivery. Various nanoparticles have been synthesized to deliver drugs and other therapeutic molecules to the injured heart via passive or active targeting approaches, and their targeting specificity and therapeutic efficacies have been investigated. Here, we summarized nanoparticle-based, cardiac-specific drug delivery systems, their potency for treating heart diseases, and the mechanisms underlying these cardiac-targeting strategies. We also discussed the clinical studies that have employed nanoparticle-based cardiac-specific drug delivery.
Collapse
Affiliation(s)
- Dong Li
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
- Department of Cardiology, Dongfang Hospital, The Second Affiliated Hospital of Beijing University of Chinese Medicine, Beijing 100078, China
| | - Yura Son
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
| | - Michelle Jang
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
- Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ 85281, USA
| | - Shu Wang
- College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA
- Correspondence: (S.W.); (W.Z.)
| | - Wuqiang Zhu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
- Correspondence: (S.W.); (W.Z.)
| |
Collapse
|
5
|
Liu Z, Ji M, Jin F, Jiang F, Liu X. Expression and clinical significance of miR-17-5p in tumor tissues of patients with colorectal cancer. J Gastrointest Oncol 2022; 13:3067-3079. [PMID: 36636078 PMCID: PMC9830340 DOI: 10.21037/jgo-22-1185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 12/07/2022] [Indexed: 12/29/2022] Open
Abstract
Background At present, there is a lack of novel biomarkers for the early diagnosis and targeted therapy of colorectal cancer (CRC). The current study investigated the expression of miR-17-5p in colorectal tumors and adjacent tissues, and examined the effects of miR-17-5p on the cellular growth of the colon cancer cell line HCT-116. Methods A total of 30 paired tissue specimens were obtained from patients with CRC who were admitted to the Department of General Surgery V of the First Affiliated Hospital, Gannan Medical College between December 2019 and January 2021. The clinical information for the corresponding patients was collated. Real-time fluorescence quantitative polymerase chain reaction (RT-qPCR) was performed to detect the expression of miR-17-5p in tumor tissues and the paracancerous tissues. The relationship between miR-17-5p and clinicopathology was analyzed. The CRC cell line HCT-116 was transfected with miR-17-5p mimics and inhibitors using liposomes. The effects of miR-17-5p on cell proliferation, invasion and migration, and apoptosis were determined using colon formation assays, transwell and scratch assays, and flow cytometry, respectively. The effects of miR-17-5p on CD44 (homing cell adhesion molecule), MMP2 (matrix metalloproteinase 2), BCL2 (B-cell lymphoma-2), BAX (BCL2-associated X), and E-cadherin protein and gene expression were investigated using Western blot and RT-qPCR, respectively. Results MiR-17-5p expression was higher in tumor tissues compared to the normal adjacent tissues. While miR-17-5p expression levels were unrelated to age nor gender, they were related to the degree of differentiation, stage, lymph node metastasis, and tumor size in patients with CRC. Upregulation of miR-17-5p promoted CRC cell proliferation, metastasis, and invasion, while inhibiting apoptosis. However, downregulation of miR-17-5p had the opposite effect. Furthermore, miR-17-5p upregulation increased E-cadherin, CD44, MMP2, and BCL2 expression while decreasing BAX expression, whereas miR-17-5p downregulation had the opposite effect. Conclusions MiR-17-5p is highly expressed in tumor tissues and correlates with increased proliferation, invasion, and migration, as well as reduced apoptosis in HCT-116 cells. Indeed, miR-17-5p may be a potential indicator for the early diagnosis of CRC and may guide clinical targeted therapy.
Collapse
Affiliation(s)
- Zheng Liu
- Department of General Surgery, The First Affiliated Hospital of Gannan Medical College, Ganzhou, China;,Ganzhou City Key Laboratory of Colorectal and Anal Diseases Research, Ganzhou, China
| | - Mengmeng Ji
- Department of General Surgery, The First Affiliated Hospital of Gannan Medical College, Ganzhou, China;,Ganzhou City Key Laboratory of Colorectal and Anal Diseases Research, Ganzhou, China
| | - Fangtong Jin
- Department of General Surgery, The First Affiliated Hospital of Gannan Medical College, Ganzhou, China;,Ganzhou City Key Laboratory of Colorectal and Anal Diseases Research, Ganzhou, China
| | - Feng Jiang
- Department of General Surgery, The First Affiliated Hospital of Gannan Medical College, Ganzhou, China
| | - Xiaoping Liu
- Department of General Surgery, The First Affiliated Hospital of Gannan Medical College, Ganzhou, China;,Ganzhou City Key Laboratory of Colorectal and Anal Diseases Research, Ganzhou, China
| |
Collapse
|
6
|
Zhang X, Wang A, Wang X, Zhao Q, Xing H. Evaluation of L-Selenomethionine on Ameliorating Cardiac Injury Induced by Environmental Ammonia. Biol Trace Elem Res 2022; 200:4712-4725. [PMID: 35094233 DOI: 10.1007/s12011-021-03071-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 12/15/2021] [Indexed: 12/27/2022]
Abstract
L-Selenomethionine is one of the important organic selenium sources. The supplementation of L-selenomethionine in diets is significant to improve the health of pigs. Ammonia is a major pollutant in the atmosphere and piggery, posing a threat to human and animal health. Although ammonia exposure can damage the heart, the mechanism of cardiac toxicity by ammonia is still unknown. In this study, we investigated the mechanism of cardiac injury induced by ammonia exposure in pigs and the protective effect of L-selenomethionine on its cardiotoxicity. The results showed that the blood ammonia content of pig increased significantly in ammonia group, the expressions of energy metabolism-related genes (LDHA, PDK4, HK2, and CPTIB) and the oxidative stress indexes were significantly changed (P < 0.05), the AMPK/PPAR-γ/NF-κB signaling pathways were activated, the chromatin edge aggregation and nuclear pyknosis were observed in ultrastructure, the apoptotic cells were significantly increased (P < 0.05), and the mRNA and protein expressions of apoptosis-related genes (Bcl-2, Bax, Cyt-c, caspase-3, and caspase-9) were significantly affected (P < 0.05). The above changes were significantly alleviated in ammonia + L-selenomethionine group, but there were still significant differences compared with the C group (P < 0.05). Our results indicated that ammonia exposure could cause energy metabolism disorder and oxidative stress and induce apoptosis of cardiomyocytes through AMPK/PPAR-γ/NF-κB pathways, which could lead to cardiac injury and affect cardiac function. L-Selenomethionine could effectively alleviate the cardiac damage caused by ammonia and antagonize the cardiotoxicity of ammonia.
Collapse
Affiliation(s)
- Xinxin Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Anqi Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Xinqiao Wang
- College of Chemical Engineering and Environment, China University of Petroleum, Beijing, 102249, People's Republic of China
| | - Qian Zhao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Houjuan Xing
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, People's Republic of China.
| |
Collapse
|
7
|
Ponnalagu D, Hamilton S, Sanghvi S, Antelo D, Schwieterman N, Hansra I, Xu X, Gao E, Edwards JC, Bansal SS, Wold LE, Terentyev D, Janssen PML, Hund TJ, Khan M, Kohut AR, Koch WJ, Singh H. CLIC4 localizes to mitochondrial-associated membranes and mediates cardioprotection. SCIENCE ADVANCES 2022; 8:eabo1244. [PMID: 36269835 PMCID: PMC9586484 DOI: 10.1126/sciadv.abo1244] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 08/25/2022] [Indexed: 06/12/2023]
Abstract
Mitochondrial-associated membranes (MAMs) are known to modulate organellar and cellular functions and can subsequently affect pathophysiology including myocardial ischemia-reperfusion (IR) injury. Thus, identifying molecular targets in MAMs that regulate the outcome of IR injury will hold a key to efficient therapeutics. Here, we found chloride intracellular channel protein (CLIC4) presence in MAMs of cardiomyocytes and demonstrate its role in modulating ER and mitochondrial calcium homeostasis under physiological and pathological conditions. In a murine model, loss of CLIC4 increased myocardial infarction and substantially reduced cardiac function after IR injury. CLIC4 null cardiomyocytes showed increased apoptosis and mitochondrial dysfunction upon hypoxia-reoxygenation injury in comparison to wild-type cardiomyocytes. Overall, our results indicate that MAM-CLIC4 is a key mediator of cellular response to IR injury and therefore may have a potential implication on other pathophysiological processes.
Collapse
Affiliation(s)
- Devasena Ponnalagu
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Shanna Hamilton
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Shridhar Sanghvi
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Diego Antelo
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Neill Schwieterman
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
- College of Nursing, The Ohio State University, Columbus, OH, USA
| | - Inderjot Hansra
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
| | - Xianyao Xu
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
- Departments of Biomedical Engineering and Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Erhe Gao
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - John C. Edwards
- Nephrology Division, Department of Internal Medicine, St. Louis University, St. Louis, MO, USA
| | - Shyam S. Bansal
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Loren E. Wold
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
- College of Nursing, The Ohio State University, Columbus, OH, USA
| | - Dmitry Terentyev
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Paul M. L. Janssen
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Thomas J. Hund
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
- Departments of Biomedical Engineering and Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Mahmood Khan
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
- Department of Emergency Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Andrew R. Kohut
- Penn Heart and Vascular Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Walter J. Koch
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Harpreet Singh
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
8
|
Cheng X, Hu J, Liu X, Tibenda JJ, Wang X, Zhao Q. Therapeutic targets by traditional Chinese medicine for ischemia-reperfusion injury induced apoptosis on cardiovascular and cerebrovascular diseases. Front Pharmacol 2022; 13:934256. [PMID: 36060007 PMCID: PMC9437626 DOI: 10.3389/fphar.2022.934256] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 06/30/2022] [Indexed: 12/02/2022] Open
Abstract
Traditional Chinese medicine (TCM) has a significant role in treating and preventing human diseases. Ischemic heart and cerebrovascular injuries are two types of diseases with different clinical manifestations with high prevalence and incidence. In recent years, it has been reported that many TCM has beneficial effects on ischemic diseases through the inhibition of apoptosis, which is the key target to treat myocardial and cerebral ischemia. This review provides a comprehensive summary of the mechanisms of various TCMs in treating ischemic cardiovascular and cerebrovascular diseases through anti-apoptotic targets and pathways. However, clinical investigations into elucidating the pharmacodynamic ingredients of TCM are still lacking, which should be further demystified in the future. Overall, the inhibition of apoptosis by TCM may be an effective strategy for treating ischemic cardio-cerebrovascular diseases.
Collapse
Affiliation(s)
- Xiuli Cheng
- Department of Pharmacy, People’s Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| | - Jin Hu
- Department of Preparation Center, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Xiaofeng Liu
- Department of Pharmacy, People’s Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| | | | - Xiaobo Wang
- Research Institute of Integrated TCM and Western Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Xiaobo Wang, ; Qipeng Zhao,
| | - Qipeng Zhao
- School of Pharmacy, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education (Ningxia Medical University), Yinchuan, China
- *Correspondence: Xiaobo Wang, ; Qipeng Zhao,
| |
Collapse
|
9
|
Integrated Gut-Heart Axis and Network Pharmacology to Reveal the Mechanisms of the Huoxue Wentong Formula Against Myocardial Ischemia. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:9538512. [PMID: 35600966 PMCID: PMC9117028 DOI: 10.1155/2022/9538512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 04/18/2022] [Indexed: 11/20/2022]
Abstract
Background Myocardial ischemia (MI) is a major public health problem with high mortality and morbidity worldwide. Huoxue Wentong formula (HX), a traditional Chinese medicine (TCM) formula, exhibits unambiguous effects on treating MI and preventing cardiovascular diseases. However, the molecular mechanism of the therapeutic effects of HX on MI remains largely unknown. Objective This study combined microbiology, metabolomics, and network pharmacology to explore the relationship between the gut microbiota and its metabolites in MI rats and the efficacy of HX. Methods First, the MI rat model was established by ligation of left anterior descending. Echocardiography, Masson's staining, and hematoxylin and eosin staining were used to evaluate the effect of HX on MI. Then, fecal metabolomics and 16S rRNA sequencing were used to obtain the microbial and metabolic characteristics of HX on MI. After that, network pharmacology was used to predict the target and action pathway of HX in treating MI. Finally, the relationship between fecal metabolites and target was explored through bioinformatics. Results HX can improve the cardiac function and ameliorated myocardial fibrosis in MI rats. Moreover, HX can affect the gut microbiota community and metabolites of MI rats, especially Bacteroides, Deferribacteres, Ruminococcus_sp._zagget7, Acidobacteria, daidzein, L-lactic acid, and malate. Network pharmacology found that HX can function through tumor necrosis factor (TNF), tumor protein p53 (TP53), interleukin 6 (IL6), vascular endothelial growth factor A (VEGFA), fos proto-oncogene (FOS), bcl2-associated X (BAX), myeloperoxidase (MPO), PI3K-Akt signaling pathways, and HIF-1 signaling pathway. The mechanism study showed that the anti-MI effect of HX was related to valine, leucine, and isoleucine biosynthesis, fatty acid biosynthesis, and arachidonic acid metabolism. Conclusion This study demonstrates that HX treated MI rats in a multitarget and multipathway manner. Its mechanism is related to the change of gut microbiota and the regulation of valine, leucine and isoleucine biosynthesis, fatty acid biosynthesis, and arachidonic acid metabolism.
Collapse
|
10
|
Chen YY, Liu QP, An P, Jia M, Luan X, Tang JY, Zhang H. Ginsenoside Rd: A promising natural neuroprotective agent. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 95:153883. [PMID: 34952508 DOI: 10.1016/j.phymed.2021.153883] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/05/2021] [Accepted: 12/05/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Neurological diseases seriously affect human health, which are arousing wider attention, and it is a great challenge to discover neuroprotective drugs with minimal side-effects and better efficacies. Natural agents derived from herbs or plants have become unparalleled resources for the discovery of novel drug candidates. Panax ginseng C. A. Meyer, a well-known herbal medicine in China, occupies a very important position in traditional Chinese medicines (TCMs) with a long history of clinical application. Ginsenoside Rd is the active compound in P. ginseng known to have broad-spectrum pharmacological effects to reduce neurological damage that can lead to neurological diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease, depression, cognitive impairment, and cerebral ischemia. PURPOSE To review and discuss the effects and mechanisms of ginsenoside Rd in the treatment of neurological diseases. STUDY DESIGN & METHODS The related information was compiled by the major scientific databases, such as Chinese National Knowledge Infrastructure (CNKI), Elsevier, ScienceDirect, PubMed, SpringerLink, Web of Science, and GeenMedical. Using 'Ginsenoside Rd', 'Ginsenosides', 'Anti-inflammation', 'Antioxidant', 'Apoptosis' and 'Neuroprotection' as keywords, the correlated literature was extracted and conducted from the databases mentioned above. RESULTS Through summarizing the existing research progress, we found that the general effects of ginsenoside Rd are anti-inflammatory, antioxidant, anti-apoptosis, inhibition of Ca2+ influx and protection of mitochondria, and through these pathways, the compound can inhibit excitatory toxicity, regulate nerve growth factor, and promote nerve regeneration. CONCLUSION Ginsenoside Rd is a promising natural neuroprotective agent. This review would contribute to the future development of ginsenoside Rd as a novel clinical candidate drug for treating neurological diseases.
Collapse
Affiliation(s)
- Yu-Ying Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Qiu-Ping Liu
- Shanghai Frontiers Science Center of TCM Chemical Biology; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Pei An
- Shanghai Frontiers Science Center of TCM Chemical Biology; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Min Jia
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xin Luan
- Shanghai Frontiers Science Center of TCM Chemical Biology; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Jian-Yuan Tang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China.
| | - Hong Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
11
|
Astragaloside IV protects diabetic cardiomyopathy against inflammation and apoptosis via regulating TLR4/MyD88/NF-κB signaling pathway. J Funct Foods 2022. [DOI: 10.1016/j.jff.2021.104905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
12
|
Li J, Jiang H, Wu P, Li S, Han B, Yang Q, Wang X, Han B, Deng N, Qu B, Zhang Z. Toxicological effects of deltamethrin on quail cerebrum: Weakened antioxidant defense and enhanced apoptosis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 286:117319. [PMID: 33990053 DOI: 10.1016/j.envpol.2021.117319] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/16/2021] [Accepted: 05/03/2021] [Indexed: 06/12/2023]
Abstract
Deltamethrin is the most common type II synthetic pyrethroid insecticide, and has posed widespread residues to environment. However, whether deltamethrin has potential toxic effects on quail cerebrum remains greatly obscure. Accordingly, we investigated the impact of chronic exposure to deltamethrin on oxidative stress and apoptosis in quail cerebrum. Quails upon 12-week exposure of deltamethrin (0, 15, 30, or 45 mg/kg body weight intragastric administration) were used as a cerebrum injury model. The results showed that deltamethrin treatment led to cerebral injury dose-dependently through the weakened antioxidant defense by downregulating nuclear factor erythroid-2-related factor 2 (Nrf2) and its downstream proteins levels and mRNA expression. Furthermore, deltamethrin treatment induced apoptosis in cerebrum by decreasing B-cell lymphoma gene 2 (Bcl-2) level, as well as increasing Jun N-terminal kinase3, caspase-3, and Bcl-2-associated X protein levels. Simultaneously, toll-like receptor 4 (TLR4) downstream inflammation-related genes or proteins were significantly up-regulated by deltamethrin dose-dependently. Altogether, our study demonstrated that chronic exposure to deltamethrin induces inflammation and apoptosis in quail cerebrums by promoting oxidative stress linked to inhibition of the Nrf2/TLR4 signaling pathway. These results provide a novel knowledge on the chronic toxic effect of deltamethrin, and establish a theoretical foundation for the evaluation of pesticide-induced health risk.
Collapse
Affiliation(s)
- Jiayi Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Huijie Jiang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Pengfei Wu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Siyu Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Bing Han
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Qingyue Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Xiaoqiao Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Biqi Han
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Ning Deng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Bing Qu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Zhigang Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Harbin, 150030, China.
| |
Collapse
|
13
|
Yang F, Jiang X, Cao H, Shuai W, Zhang L, Wang G, Quan D, Jiang X. Daphnetin Preconditioning Decreases Cardiac Injury and Susceptibility to Ventricular Arrhythmia following Ischaemia-Reperfusion through the TLR4/MyD88/NF-Κb Signalling Pathway. Pharmacology 2021; 106:369-383. [PMID: 33902056 DOI: 10.1159/000513631] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 11/24/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND/AIMS Daphnetin (7,8-dihydroxycoumarin, DAP) exhibits various bioactivities, such as anti-inflammatory and antioxidant activities. However, the role of DAP in myocardial ischaemia/reperfusion (I/R) injury and I/R-related arrhythmia is still uncertain. This study aimed to investigate the mechanisms underlying the effects of DAP on myocardial I/R injury and electrophysiological properties in vivo and in vitro. METHODS Myocardial infarct size was measured by triphenyltetrazolium chloride staining. Cardiac function was assessed by echocardiographic and haemodynamic analyses. The levels of creatine kinase-MB, lactate dehydrogenase, malondialdehyde, superoxide dismutase, interleukin-6 (IL-6), and tumour necrosis factor-alpha (TNF-α) were detected using commercial kits. Apoptosis was measured by terminal deoxynucleotidyl-transferase-mediated dUTP nick-end labelling staining and flow cytometry. The viability of H9c2 cells was determined by the Cell Counting Kit-8 assay. In vitro, the levels of IL-6 and TNF-α were measured by quantitative PCR. The expression levels of proteins associated with apoptosis, inflammation, and the Toll-like receptor 4/myeloid differentiation factor 88/nuclear factor kappa B (TLR4/MyD88/NF-κB) signalling pathway were detected by Western blot analysis. The RR, PR, QRS, and QTc intervals were assessed by surface ECG. The 90% action potential duration (APD90), threshold of APD alternans, and ventricular tachycardia inducibility were measured by the Langendorff perfusion technique. RESULTS DAP preconditioning decreased myocardial I/R injury and hypoxia/reoxygenation (H/R) injury in cells. DAP preconditioning improved cardiac function after myocardial I/R injury. DAP preconditioning also suppressed apoptosis, attenuated oxidative stress, and inhibited inflammatory responses in vivo and in vitro. Furthermore, DAP preconditioning decreased the susceptibility to ventricular arrhythmia after myocardial I/R. Finally, DAP preconditioning inhibited the expression of TLR4, MyD88, and phosphorylated NF-κB (p-NF-κB)/P65 in mice subjected to I/R and cells subjected to H/R. CONCLUSIONS DAP preconditioning protected against myocardial I/R injury and decreased susceptibility to ventricular arrhythmia by inhibiting the TLR4/MyD88/NF-κB signalling pathway.
Collapse
Affiliation(s)
- Fan Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Xiaobo Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Hongyi Cao
- Department of Endocrinology, Chengdu Fifth People's Hospital, Chengdu, China
| | - Wei Shuai
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Lijun Zhang
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Guangji Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Dajun Quan
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Xuejun Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
14
|
Meng S, Hu Y, Zhu J, Feng T, Quan X. miR-30c-5p acts as a therapeutic target for ameliorating myocardial ischemia-reperfusion injury. Am J Transl Res 2021; 13:2198-2212. [PMID: 34017383 PMCID: PMC8129376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 10/16/2020] [Indexed: 06/12/2023]
Abstract
Coronary heart disease (CHD) is one of the most vital reasons for death and disability all over the world. miRNA, as a plasma index, is quite valuable for disease screening and prognosis prediction in CHD. Mining the molecular mechanism behind miRNA is also helpful for us to find molecular therapeutic strategies. In this research, we found that the expression of plasma miR-30c-5p in CHD patients was obviously lower than that in the control group (CG), which had a high differential value for CHD. We also discovered that miR-30c-5p was obviously correlated with clinical characteristics of CHD patients such as age, NYHA grade, smoking history, hypertension, hyperlipidemia, etc. In prognosis analysis, the miR-30c-5p expression in patients with poor prognosis was dramatically lower than that in those with good one, and the AUC for predicting poor prognosis of CHD was not lower than 0.850. In addition, we also induced myocardial ischemia/reperfusion (I/R) injury model of H9C2 cells through hypoxia/reoxygenation, and found that H9C2 cells also had abnormally down-regulated miR-30c-5p and up-regulated BCL2-like 11 (BCL2L11). Up-regulating miR-30c-5p or down-regulating BCL2L11 were helpful to improve proliferation and apoptosis of I/R injury model. Mechanically, BCL2L11 was also negatively regulated by miR-30c-5p, and up-regulating the former could cancel the in vitro protective effect of up-regulating the latter on H9C2 cell I/R injury model. In vivo research, up-regulating miR-30c-5p or down-regulating BCL2L11 can improve myocardial injury, histopathological changes and apoptosis in rat I/R model.
Collapse
Affiliation(s)
- Shuping Meng
- Cardiovascular Surgery Intensive Care Unit, Heart Center of Henan Province People’s Hospital (Central China Fuwai Hospital)Zhengzhou 450003, Henan, China
| | - Yanlei Hu
- Cardiovascular Surgery Intensive Care Unit, Heart Center of Henan Province People’s Hospital (Central China Fuwai Hospital)Zhengzhou 450003, Henan, China
| | - Jialu Zhu
- Cardiovascular Surgery Intensive Care Unit, Heart Center of Henan Province People’s Hospital (Central China Fuwai Hospital)Zhengzhou 450003, Henan, China
| | - Tao Feng
- Cardiovascular Surgery Intensive Care Unit, Heart Center of Henan Province People’s Hospital (Central China Fuwai Hospital)Zhengzhou 450003, Henan, China
| | - Xiaoqiang Quan
- Department of Cardiovascular Surgery, Heart Center of Henan Province People’s Hospital (Central China Fuwai Hospital)Zhengzhou 450003, Henan, China
| |
Collapse
|
15
|
Xin T, Lv W, Liu D, Jing Y, Hu F. Opa1 Reduces Hypoxia-Induced Cardiomyocyte Death by Improving Mitochondrial Quality Control. Front Cell Dev Biol 2020; 8:853. [PMID: 32984338 PMCID: PMC7483501 DOI: 10.3389/fcell.2020.00853] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 08/10/2020] [Indexed: 12/14/2022] Open
Abstract
Mitochondrial dysfunction contributes to cardiovascular disorders, especially post-infarction cardiac injury, through incompletely characterized mechanisms. Among the latter, increasing evidence points to alterations in mitochondrial quality control, a range of adaptive responses regulating mitochondrial morphology and function. Optic atrophy 1 (Opa1) is a mitochondrial inner membrane GTPase known to promote mitochondrial fusion. In this study, hypoxia-mediated cardiomyocyte damage was induced to mimic post-infarction cardiac injury in vitro. Loss- and gain-of-function assays were then performed to evaluate the impact of Opa1 expression on mitochondrial quality control and cardiomyocyte survival and function. Hypoxic stress reduced cardiomyocyte viability, impaired contractile/relaxation functions, and augmented the synthesis of pro-inflammatory mediators. These effects were exacerbated by Opa1 knockdown, and significantly attenuated by Opa1 overexpression. Mitochondrial quality control was disturbed by hypoxia, as reflected by multiple mitochondrial deficits; i.e., increased fission, defective fusion, impaired mitophagy, decreased biogenesis, increased oxidative stress, and blunted respiration. By contrast, overexpression of Opa1 normalized mitochondrial quality control and sustained cardiomyocyte function. We also found that ERK, AMPK, and YAP signaling can regulate Opa1 expression. These results identify Opa1 as a novel regulator of mitochondrial quality control and highlight a key role for Opa1 in protecting cardiomyocytes against post-infarction cardiac injury.
Collapse
Affiliation(s)
- Ting Xin
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, China
| | - Wei Lv
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, China
| | - Dongmei Liu
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, China
| | - Yongle Jing
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, China
| | - Fang Hu
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, China
| |
Collapse
|
16
|
Meng X, Mei L, Zhao C, Chen W, Zhang N. miR-885 mediated cardioprotection against hypoxia/reoxygenation-induced apoptosis in human cardiomyocytes via inhibition of PTEN and BCL2L11 and modulation of AKT/mTOR signaling. J Cell Physiol 2020; 235:8048-8057. [PMID: 31960416 DOI: 10.1002/jcp.29460] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 01/07/2020] [Indexed: 12/11/2022]
Abstract
Ischemia/reperfusion (I/R) injury could cause the enhanced cell apoptosis of cardiomyocytes, which is one of key contributors for the development of ischemic heart disease. Recent studies emphasized the role of microRNAs (miRNAs) in regulating cardiomyocyte apoptosis. The study planned to elucidate the molecular actions of miR-885 on mediating human cardiomyocytes (HCMs) apoptosis induced by hypoxia/reoxygenation (H/R) and to explore the potential molecular mechanisms. The present data revealed that H/R stimulation inhibited HCM viability and potentiated HCM apoptosis, and more importantly, the expression of miR-885 in HCMs was markedly repressed after H/R stimulation. Further experimental examinations demonstrated that overexpression of miR-885 attenuated H/R-induced increased in HCM apoptotic rates, while miR-885 knockdown impaired HCM viability and increased HCM apoptotic rates. Moreover, the mechanistic studies showed that miR-885 inversely regulated the expression of phosphatase and tensin homolog (PTEN) and BCL2 like 11 (BCL2L11) in HCMs, and enforced expression of PTEN and BCL2L11 partially antagonized the protective actions of miR-885 overexpression on H/R-induced HCM injury. Moreover, H/R suppressed AKT/mTOR signaling, which was attenuated by miR-885 overexpression in HCMs. In conclusion, the present study for the first time showed the downregulation of miR-885 induced by H/R in HCMs, and provided the evidence that miR-885 attenuated H/R-induced cell apoptosis via inhibiting PTEN and BLC2L11 and modulation of AKT/mTOR signaling in HCMs.
Collapse
Affiliation(s)
- Xin Meng
- Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lijun Mei
- Department of Blood Transfusion, Ankang Central Hospital, Ankang, China
| | - Chedong Zhao
- Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wei Chen
- Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ning Zhang
- Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
17
|
Yuan Y, Wang Y, Liu X, Luo B, Zhang L, Zheng F, Li X, Guo L, Wang L, Jiang M, Pan Y, Yan Y, Yang J, Chen S, Wang J, Tang J. KPC1 alleviates hypoxia/reoxygenation-induced apoptosis in rat cardiomyocyte cells though BAX degradation. J Cell Physiol 2019; 234:22921-22934. [PMID: 31148189 PMCID: PMC6771896 DOI: 10.1002/jcp.28854] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 04/24/2019] [Accepted: 04/30/2019] [Indexed: 12/19/2022]
Abstract
Bax triggers cell apoptosis by permeabilizing the outer mitochondrial membrane, leading to membrane potential loss and cytochrome c release. However, it is unclear if proteasomal degradation of Bax is involved in the apoptotic process, especially in heart ischemia-reperfusion (I/R)-induced injury. In the present study, KPC1 expression was heightened in left ventricular cardiomyocytes of patients with coronary heart disease (CHD), in I/R-myocardium in vivo and in hypoxia and reoxygenation (H/R)-induced cardiomyocytes in vitro. Overexpression of KPC1 reduced infarction size and cell apoptosis in I/R rat hearts. Similarly, the forced expression of KPC1 restored mitochondrial membrane potential (MMP) and cytochrome c release driven by H/R in H9c2 cells, whereas reducing cell apoptosis, and knockdown of KPC1 by short-hairpin RNA (shRNA) deteriorated cell apoptosis induced by H/R. Mechanistically, forced expression of KPC1 promoted Bax protein degradation, which was abolished by proteasome inhibitor MG132, suggesting that KPC1 promoted proteasomal degradation of Bax. Furthermore, KPC1 prevented basal and apoptotic stress-induced Bax translocation to mitochondria. Bax can be a novel target for the antiapoptotic effects of KPC1 on I/R-induced cardiomyocyte apoptosis and render mechanistic penetration into at least a subset of the mitochondrial effects of KPC1.
Collapse
Affiliation(s)
- Ye Yuan
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of MedicineShiyanHubeiChina
- Institute of Biomedicine and Key Lab of Human Embryonic Stem Cell of Hubei ProvinceHubei University of MedicineHubeiChina
| | - Yong‐yi Wang
- Department of Cardiovascular Surgery, Ren Ji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Xin Liu
- Laboratory Animal CenterHubeiChina
| | - Bin Luo
- Department of Physiology, School of Basic Medicine ScienceHubei University of MedicineHubeiChina
| | - Lei Zhang
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of MedicineShiyanHubeiChina
- Institute of Biomedicine and Key Lab of Human Embryonic Stem Cell of Hubei ProvinceHubei University of MedicineHubeiChina
| | - Fei Zheng
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of MedicineShiyanHubeiChina
| | - Xing‐Yuan Li
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of MedicineShiyanHubeiChina
| | - Ling‐Yun Guo
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of MedicineShiyanHubeiChina
| | - Lu Wang
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of MedicineShiyanHubeiChina
| | - Miao Jiang
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of MedicineShiyanHubeiChina
| | - Ya‐mu Pan
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of MedicineShiyanHubeiChina
| | - Yu‐wen Yan
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of MedicineShiyanHubeiChina
| | - Jian‐ye Yang
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of MedicineShiyanHubeiChina
| | - Shi‐You Chen
- Department of Physiology & PharmacologyThe University of GeorgiaAthensUSA
| | - Jia‐Ning Wang
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of MedicineShiyanHubeiChina
- Institute of Biomedicine and Key Lab of Human Embryonic Stem Cell of Hubei ProvinceHubei University of MedicineHubeiChina
| | - Jun‐Ming Tang
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of MedicineShiyanHubeiChina
- Department of Physiology, School of Basic Medicine ScienceHubei University of MedicineHubeiChina
- Institute of Biomedicine and Key Lab of Human Embryonic Stem Cell of Hubei ProvinceHubei University of MedicineHubeiChina
| |
Collapse
|