1
|
Wu H, Lan Q, He YX, Xue JY, Liu H, Zou Y, Liu P, Luo G, Chen MT, Liu MN. Programmed cardiomyocyte death in myocardial infarction. Apoptosis 2025:10.1007/s10495-025-02075-3. [PMID: 39833636 DOI: 10.1007/s10495-025-02075-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2025] [Indexed: 01/22/2025]
Abstract
Cardiovascular disease (CVD) is a leading cause of human mortality worldwide, with patients often at high risk of heart failure (HF) in myocardial infarction (MI), a common form of CVD that results in cardiomyocyte death and myocardial necrosis due to inadequate myocardial perfusion. As terminally differentiated cells, cardiomyocytes possess a severely limited capacity for regeneration, and an excess of dead cardiomyocytes will further stress surviving cells, potentially exacerbating to more extensive heart disease. The article focuses on the relationship between programmed cell death (PCD) of cardiomyocytes, including different forms of apoptosis, necrosis, and autophagy, and MI, as well as the potential application of these mechanisms in the treatment of MI. By gaining a deeper understanding of the mechanisms of cardiomyocyte death, it aims to provide new insights into the prevention and treatment of MI.
Collapse
Affiliation(s)
- Hao Wu
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Qi Lan
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Yi-Xiang He
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Jin-Yi Xue
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Hao Liu
- Department of Pediatrics, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Yuan Zou
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Ping Liu
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Gang Luo
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China.
| | - Ming-Tai Chen
- Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, 518033, Shenzhen, People's Republic of China.
| | - Meng-Nan Liu
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China.
| |
Collapse
|
2
|
Zhou X, Wang H, Yan B, Nie X, Chen Q, Yang X, Lei M, Guo X, Ouyang C, Ren Z. Ferroptosis in Cardiovascular Diseases and Ferroptosis-Related Intervention Approaches. Cardiovasc Drugs Ther 2024:10.1007/s10557-024-07642-5. [PMID: 39641901 DOI: 10.1007/s10557-024-07642-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/08/2024] [Indexed: 12/07/2024]
Abstract
OBJECTIVE Cardiovascular diseases (CVDs) are major public health problems that threaten the lives and health of individuals. The article has reviewed recent progresses about ferroptosis and ferroptosis-related intervention approaches for the treatment of CVDs and provided more references and strategies for targeting ferroptosis to prevent and treat CVDs. METHODS A comprehensive review was conducted using the literature researches. RESULTS AND DISCUSSION Many ferroptosis-targeted compounds and ferroptosis-related genes may be prospective targets for treating CVDs and our review provides a solid foundation for further studies about the detailed pathological mechanisms of CVDs. CONCLUSION There are challenges and limitations about the translation of ferroptosis-targeted potential therapies from experimental research to clinical practice. It warrants further exploration to pursure safer and more effective ferroptosis-targeted thereapeutic approaches for CVDs.
Collapse
Affiliation(s)
- Xianpeng Zhou
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Hao Wang
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Biao Yan
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Xinwen Nie
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Qingjie Chen
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Xiaosong Yang
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Min Lei
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Xiying Guo
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Changhan Ouyang
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Zhanhong Ren
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China.
| |
Collapse
|
3
|
Miura T, Kouzu H, Tanno M, Tatekoshi Y, Kuno A. Role of AMP deaminase in diabetic cardiomyopathy. Mol Cell Biochem 2024; 479:3195-3211. [PMID: 38386218 DOI: 10.1007/s11010-024-04951-z] [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: 12/19/2023] [Accepted: 01/24/2024] [Indexed: 02/23/2024]
Abstract
Diabetes mellitus is one of the major causes of ischemic and nonischemic heart failure. While hypertension and coronary artery disease are frequent comorbidities in patients with diabetes, cardiac contractile dysfunction and remodeling occur in diabetic patients even without comorbidities, which is referred to as diabetic cardiomyopathy. Investigations in recent decades have demonstrated that the production of reactive oxygen species (ROS), impaired handling of intracellular Ca2+, and alterations in energy metabolism are involved in the development of diabetic cardiomyopathy. AMP deaminase (AMPD) directly regulates adenine nucleotide metabolism and energy transfer by adenylate kinase and indirectly modulates xanthine oxidoreductase-mediated pathways and AMP-activated protein kinase-mediated signaling. Upregulation of AMPD in diabetic hearts was first reported more than 30 years ago, and subsequent studies showed similar upregulation in the liver and skeletal muscle. Evidence for the roles of AMPD in diabetes-induced fatty liver, sarcopenia, and heart failure has been accumulating. A series of our recent studies showed that AMPD localizes in the mitochondria-associated endoplasmic reticulum membrane as well as the sarcoplasmic reticulum and cytosol and participates in the regulation of mitochondrial Ca2+ and suggested that upregulated AMPD contributes to contractile dysfunction in diabetic cardiomyopathy via increased generation of ROS, adenine nucleotide depletion, and impaired mitochondrial respiration. The detrimental effects of AMPD were manifested at times of increased cardiac workload by pressure loading. In this review, we briefly summarize the expression and functions of AMPD in the heart and discuss the roles of AMPD in diabetic cardiomyopathy, mainly focusing on contractile dysfunction caused by this disorder.
Collapse
Affiliation(s)
- Tetsuji Miura
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan.
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, 15-4-1, Maeda-7, Teine-Ku, Sapporo, 006-8585, Japan.
| | - Hidemichi Kouzu
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Masaya Tanno
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Nursing, Sapporo Medical University School of Health Sciences, Sapporo, Japan
| | - Yuki Tatekoshi
- Department of Pharmacology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Atsushi Kuno
- Department of Pharmacology, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
4
|
Li H, Wang M, Huang Y. Anthracycline-induced cardiotoxicity: An overview from cellular structural perspective. Biomed Pharmacother 2024; 179:117312. [PMID: 39167843 DOI: 10.1016/j.biopha.2024.117312] [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: 05/13/2024] [Revised: 07/28/2024] [Accepted: 08/13/2024] [Indexed: 08/23/2024] Open
Abstract
Anthracyclines are broad-spectrum anticancer drugs, but their clinical use is limited due to their severe cardiotoxicity. Anthracycline-induced cardiotoxicity (AIC) remains a significant cause of heart disease-related mortality in many cancer survivors. The underlying mechanisms of AIC have been explored over the past few decades. Reactive oxygen species and drug-induced inhibition of topoisomerase II beta are well-studied mechanisms, with mitochondria being a prominently investigated organelle. Emerging mechanisms such as ferroptosis, Ca2+ overload, autophagy and inflammation mediators have been implicated in recent years. In this review, our goal is to summarize and update the roles of various mechanisms in AIC, focusing on different cellular levels and further explore promising therapeutic approaches targeting these organelles or pathways.
Collapse
Affiliation(s)
- Hansheng Li
- Department of Cardiology and Cardiovascular Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei Province 430060, China.
| | - Meilun Wang
- Department of Cardiology and Cardiovascular Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei Province 430060, China.
| | - Yan Huang
- Department of Cardiology and Cardiovascular Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei Province 430060, China.
| |
Collapse
|
5
|
Zhang Z, Yang Z, Wang S, Wang X, Mao J. Overview of pyroptosis mechanism and in-depth analysis of cardiomyocyte pyroptosis mediated by NF-κB pathway in heart failure. Biomed Pharmacother 2024; 179:117367. [PMID: 39214011 DOI: 10.1016/j.biopha.2024.117367] [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: 07/17/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
The pyroptosis of cardiomyocytes has become an essential topic in heart failure research. The abnormal accumulation of these biological factors, including angiotensin II, advanced glycation end products, and various growth factors (such as connective tissue growth factor, vascular endothelial growth factor, transforming growth factor beta, among others), activates the nuclear factor-κB (NF-κB) signaling pathway in cardiovascular diseases, ultimately leading to pyroptosis of cardiomyocytes. Therefore, exploring the underlying molecular biological mechanisms is essential for developing novel drugs and therapeutic strategies. However, our current understanding of the precise regulatory mechanism of this complex signaling pathway in cardiomyocyte pyroptosis is still limited. Given this, this study reviews the milestone discoveries in the field of pyroptosis research since 1986, analyzes in detail the similarities, differences, and interactions between pyroptosis and other cell death modes (such as apoptosis, necroptosis, autophagy, and ferroptosis), and explores the deep connection between pyroptosis and heart failure. At the same time, it depicts in detail the complete pathway of the activation, transmission, and eventual cardiomyocyte pyroptosis of the NF-κB signaling pathway in the process of heart failure. In addition, the study also systematically summarizes various therapeutic approaches that can inhibit NF-κB to reduce cardiomyocyte pyroptosis, including drugs, natural compounds, small molecule inhibitors, gene editing, and other cutting-edge technologies, aiming to provide solid scientific support and new research perspectives for the prevention and treatment of heart failure.
Collapse
Affiliation(s)
- Zeyu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhihua Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shuai Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Xianliang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| | - Jingyuan Mao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
6
|
Wu YT, Zhang GY, Li L, Liu B, Wang RY, Song RQ, Hua Y, Bi YM, Han X, Zhang F, Wang D, Xie LP, Zhou YC. Salvia miltiorrhiza suppresses cardiomyocyte ferroptosis after myocardial infarction by activating Nrf2 signaling. JOURNAL OF ETHNOPHARMACOLOGY 2024; 330:118214. [PMID: 38641076 DOI: 10.1016/j.jep.2024.118214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/08/2024] [Accepted: 04/16/2024] [Indexed: 04/21/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ferroptosis, a recently identified non-apoptotic form of cell death reliant on iron, is distinguished by an escalation in lipid reactive oxygen species (ROS) that are iron-dependent. This phenomenon has a strong correlation with irregularities in iron metabolism and lipid peroxidation. Salvia miltiorrhiza Bunge (DS), a medicinal herb frequently utilized in China, is highly esteemed for its therapeutic effectiveness in enhancing blood circulation and ameliorating blood stasis, particularly during the treatment of cardiovascular diseases (CVDs). Numerous pharmacological studies have identified that DS manifests antioxidative stress effects as well as inhibits lipid peroxidation. However, ambiguity persists regarding the potential of DS to impede ferroptosis in cardiomyocytes and subsequently improve myocardial damage post-myocardial infarction (MI). AIM OF THE STUDY The present work focused on investigating whether DS could be used to prevent the ferroptosis of cardiomyocytes and improve post-MI myocardial damage. MATERIALS AND METHODS In vivo experiments: Through ligation of the left anterior descending coronary artery, we constructed both a wild-type (WT) and NF-E2 p45-related factor 2 knockout (Nrf2-/-) mouse model of MI. Effects of DS and ferrostatin-1 (Fer-1) on post-MI cardiomyocyte ferroptosis were examined through detecting ferroptosis and myocardial damage-related indicators as well as Nrf2 signaling-associated protein levels. In vitro experiments: Erastin was used for stimulating H9C2 cardiomyocytes to construct an in vitro ferroptosis cardiomyocyte model. Effects of DS and Fer-1 on cardiomyocyte ferroptosis were determined based on ferroptosis-related indicators and Nrf2 signaling-associated protein levels. Additionally, inhibitor and activator of Nrf2 were used for confirming the impact of Nrf2 signaling on DS's effect on cardiomyocyte ferroptosis. RESULTS In vivo: In comparison to the model group, DS suppressed ferroptosis in cardiomyocytes post-MI and ameliorated myocardial damage by inducing Nrf2 signaling-related proteins (Nrf2, xCT, GPX4), diminishing tissue ferrous iron and malondialdehyde (MDA) content. Additionally, it enhanced glutathione (GSH) levels and total superoxide dismutase (SOD) activity, effects that are aligned with those of Fer-1. Moreover, the effect of DS on alleviating cardiomyocyte ferroptosis after MI could be partly inhibited through Nrf2 knockdown. In vitro: Compared with the erastin group, DS inhibited cardiomyocyte ferroptosis by promoting the expression of Nrf2 signaling-related proteins, reducing ferrous iron, ROS, and MDA levels, but increasing GSH content and SOD activity, consistent with the effect of Fer-1. Additionally, Nrf2 inhibition increased erastin-mediated ferroptosis of cardiomyocytes through decreasing Nrf2 signaling-related protein expressions. Co-treatment with DS and Nrf2 activator failed to further enhance the anti-ferroptosis effect of DS. CONCLUSION MI is accompanied by cardiomyocyte ferroptosis, whose underlying mechanism is probably associated with Nrf2 signaling inhibition. DS possibly suppresses ferroptosis of cardiomyocytes and improves myocardial damage after MI through activating Nrf2 signaling.
Collapse
Affiliation(s)
- Yu-Ting Wu
- Binzhou Medical University Hospital, Binzhou, 256603, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou, 510515, China
| | - Guo-Yong Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou, 510515, China
| | - Lei Li
- Binzhou Medical University Hospital, Binzhou, 256603, China
| | - Bin Liu
- Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Ru-Yu Wang
- School of Clinical Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, China
| | | | - Yue Hua
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yi-Ming Bi
- The Affiliated Traditional Chinese Medicine Hospital of Guangzhou Medical University, Guangzhou, 510000, China
| | - Xin Han
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou, 510515, China
| | - Feng Zhang
- Binzhou Medical University Hospital, Binzhou, 256603, China
| | - Dong Wang
- Binzhou Medical University Hospital, Binzhou, 256603, China.
| | - Ling-Peng Xie
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510000, China.
| | - Ying-Chun Zhou
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
7
|
Sun Z, Liu K, Liang C, Wen L, Wu J, Liu X, Li X. Diosmetin as a promising natural therapeutic agent: In vivo, in vitro mechanisms, and clinical studies. Phytother Res 2024; 38:3660-3694. [PMID: 38748620 DOI: 10.1002/ptr.8214] [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: 11/08/2023] [Revised: 04/09/2024] [Accepted: 04/13/2024] [Indexed: 07/12/2024]
Abstract
Diosmetin, a natural occurring flavonoid, is primarily found in citrus fruits, beans, and other plants. Diosmetin demonstrates a variety of pharmacological activities, including anticancer, antioxidant, anti-inflammatory, antibacterial, metabolic regulation, cardiovascular function improvement, estrogenic effects, and others. The process of literature search was done using PubMed, Web of Science and ClinicalTrials databases with search terms containing Diosmetin, content, anticancer, anti-inflammatory, antioxidant, pharmacological activity, pharmacokinetics, in vivo, and in vitro. The aim of this review is to summarize the in vivo, in vitro and clinical studies of Diosmetin over the last decade, focusing on studies related to its anticancer, anti-inflammatory, and antioxidant activities. It is found that DIO has significant therapeutic effects on skin and cardiovascular system diseases, and its research in pharmacokinetics and toxicology is summarized. It provides the latest information for researchers and points out the limitations of current research and areas that should be strengthened in future research, so as to facilitate the relevant scientific research and clinical application of DIO.
Collapse
Affiliation(s)
- Zihao Sun
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kai Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chuipeng Liang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lin Wen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jijiao Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaolian Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaofang Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
8
|
Dai B, Liu H, Juan D, Wu K, Cao R. The role of miRNA-29b1 on the hypoxia-induced apoptosis in mammalian cardiomyocytes. Eur J Histochem 2024; 68:4021. [PMID: 38934067 PMCID: PMC11228570 DOI: 10.4081/ejh.2024.4021] [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: 03/12/2024] [Accepted: 05/06/2024] [Indexed: 06/28/2024] Open
Abstract
Cardiomyocyte apoptosis is a complex biological process involving the interaction of many factors and signaling pathways. In hypoxic environment, cardiomyocytes may trigger apoptosis due to insufficient energy supply, increased production of oxygen free radicals, and disturbance of intracellular calcium ion balance. The present research aimed to investigate the role of microRNA-29b1 (miR-29b1) in hypoxia-treated cardiomyocytes and its potential mechanism involved. We established an in vitro ischemia model using AC16 and H9C2 cardiomyocytes through hypoxia treatment (1% O2, 48 h). Cell apoptosis was evaluated by flow cytometry using Annexin V FITC-PI staining assay. Moreover, we used Western blot and immunofluorescence analysis to determine the expression of Bcl-2, Bax caspase-3 and Cx43 proteins. We found that miR-29b1 protected AC16 and H9C2 cells from hypoxia-induced injury as evidence that miR-29b1 attenuated the effects of hypoxia treatment on AC16 and H9C2 cell apoptosis after hypoxia treatment. In conclusion, our findings suggest that miR-29b1 may have potential cardiovascular protective effects during ischemia-related myocardial injury.
Collapse
Affiliation(s)
- Bo Dai
- Department of Cardiology, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan.
| | - Hailin Liu
- Department of Cardiology, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan.
| | - Dingmin Juan
- Department of Cardiology, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan.
| | - Kaize Wu
- Department of Cardiology, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan.
| | - Ruhao Cao
- Department of Cardiology, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan.
| |
Collapse
|
9
|
Chen Y, Li S, Yang Z, Wang T, Yin F, Zhao X, Zhang Y. Value of Bax and Bcl2 expression in peripheral blood mononuclear cells for clinical prognosis of patients with chronic heart failure. Medicine (Baltimore) 2024; 103:e36943. [PMID: 38241555 PMCID: PMC10798729 DOI: 10.1097/md.0000000000036943] [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: 03/20/2023] [Accepted: 07/28/2023] [Indexed: 01/21/2024] Open
Abstract
To investigate the expression of Bax and Bcl2 protein in peripheral blood mononuclear cells (PBMC) of patients with chronic heart failure (CHF), and to analyze their value for predicting major adverse cardiovascular event (MACE) in CHF patients. A total of 154 fasting venous blood samples from CHF patients were collected in our hospital from January 2017 to June 2019, and they were divided into 2 group according to whether MACE occurred during 3 years follow-up, MACE group and No-MACE group. Levels of Bax and Bcl2 protein expression in PBMC of CHF patients using enzyme-linked immunosorbent assay (ELISA), and then evaluated the predictive power of Bax and Bcl2 expression for MACE using logistic regression analysis and ROC curve. 62 (40.26%) of 154 CHF patients occurred MACE during follow-up, and there were significant differences in age, diabetes, LVEF, LDL-C and NYHA grade between MACE group and No-MACE group. Levels of Bax protein expression in PBMC of CHF patients in MACE group were significantly higher than those in No-MACE group, while levels of Bcl2 protein expression were significantly lower than those in No-MACE group, and Bax and Bcl2 protein levels increased and decreased with NYHA grades in MACE group and No-MACE group, respectively. Results of univariate and multivariate logistic regression analysis showed that Bax (OR, 1.026; 95% CI, 1.003-1.049; P = .027) and Bcl2 levels (OR, 0.952; 95% CI, 0.908-0.998; P = .041) were independent predictive factors for MACE in CHF patients. In addition, Bax and Bcl2 levels could be used to differentiate CHF patients at risk for MACE with an AUC of 0.744 (95% CI: 0.660-0.827) and an AUC of 0.743 (95% CI: 0.667-0.819), respectively. Levels of Bax and Bcl2 protein in PBMC could be used as independent predictive factors for MACE in CHF patients.
Collapse
Affiliation(s)
- Yangang Chen
- Department of Internal Medicine-Cardiovascular, Liangzhou Hospital of Wuwei City, Wuwei City, Gansu Province, China
| | - Shuiquan Li
- Department of Internal Medicine-Cardiovascular, Liangzhou Hospital of Wuwei City, Wuwei City, Gansu Province, China
| | - Zhenwen Yang
- Department of Internal Medicine-Cardiovascular, Liangzhou Hospital of Wuwei City, Wuwei City, Gansu Province, China
| | - Tianlu Wang
- Department of Internal Medicine-Cardiovascular, Liangzhou Hospital of Wuwei City, Wuwei City, Gansu Province, China
| | - Fahui Yin
- Department of Internal Medicine-Cardiovascular, Liangzhou Hospital of Wuwei City, Wuwei City, Gansu Province, China
| | - Xiangyu Zhao
- Department of Internal Medicine-Cardiovascular, Liangzhou Hospital of Wuwei City, Wuwei City, Gansu Province, China
| | - Yong Zhang
- Department of Internal Medicine-Cardiovascular, Liangzhou Hospital of Wuwei City, Wuwei City, Gansu Province, China
| |
Collapse
|
10
|
Tam E, Sweeney G. MitoNEET Provides Cardioprotection via Reducing Oxidative Damage and Conserving Mitochondrial Function. Int J Mol Sci 2023; 25:480. [PMID: 38203651 PMCID: PMC10779211 DOI: 10.3390/ijms25010480] [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: 12/01/2023] [Revised: 12/21/2023] [Accepted: 12/24/2023] [Indexed: 01/12/2024] Open
Abstract
Cardiometabolic diseases exert a significant health impact, leading to a considerable economic burden globally. The metabolic syndrome, characterized by a well-defined cluster of clinical parameters, is closely linked to an elevated risk of cardiovascular disease. Current treatment strategies often focus on addressing individual aspects of metabolic syndrome. We propose that exploring novel therapeutic approaches that simultaneously target multiple facets may prove more effective in alleviating the burden of cardiometabolic disease. There is a growing body of evidence suggesting that mitochondria can serve as a pivotal target for the development of therapeutics aimed at resolving both metabolic and vascular dysfunction. MitoNEET was identified as a binding target for the thiazolidinedione (TZD) class of antidiabetic drugs and is now recognized for its role in regulating various crucial cellular processes. Indeed, mitoNEET has demonstrated promising potential as a therapeutic target in various chronic diseases, encompassing cardiovascular and metabolic diseases. In this review, we present a thorough overview of the molecular mechanisms of mitoNEET, with an emphasis on their implications for cardiometabolic diseases in more recent years. Furthermore, we explore the potential impact of these findings on the development of novel therapeutic strategies and discuss potential directions for future research.
Collapse
Affiliation(s)
| | - Gary Sweeney
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada
| |
Collapse
|
11
|
Gawargi FI, Mishra PK. Ironing out the details: ferroptosis and its relevance to diabetic cardiomyopathy. Am J Physiol Regul Integr Comp Physiol 2023; 325:R665-R681. [PMID: 37746707 PMCID: PMC11178299 DOI: 10.1152/ajpregu.00117.2023] [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: 05/17/2023] [Revised: 09/05/2023] [Accepted: 09/14/2023] [Indexed: 09/26/2023]
Abstract
Ferroptosis is a newly identified myocardial cell death mechanism driven by iron-dependent lipid peroxidation. The presence of elevated intramyocardial lipid levels and excessive iron in patients with diabetes suggest a predominant role of ferroptosis in diabetic cardiomyopathy. As myocardial cell death is a precursor of heart failure, and intensive glycemic control cannot abate the increased risk of heart failure in patients with diabetes, targeting myocardial cell death via ferroptosis is a promising therapeutic avenue to prevent and/or treat diabetic cardiomyopathy. This review provides updated and comprehensive molecular mechanisms underpinning ferroptosis, clarifies several misconceptions about ferroptosis, emphasizes the importance of ferroptosis in diabetes-induced myocardial cell death, and offers valuable approaches to evaluate and target ferroptosis in the diabetic heart. Furthermore, basic concepts and ideas presented in this review, including glutathione peroxidase-4-independent and mitochondrial mechanisms of ferroptosis, are also important for investigating ferroptosis in other diabetic organs, as well as nondiabetic and metabolically compromised hearts.
Collapse
Affiliation(s)
- Flobater I Gawargi
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Paras K Mishra
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| |
Collapse
|
12
|
Li C, Zhou Y, Niu Y, He W, Wang X, Zhang X, Wu Y, Zhang W, Zhao L, Zheng H, Song W, Gao H. Deficiency of Pdk1 drives heart failure by impairing taurine homeostasis through Slc6a6. FASEB J 2023; 37:e23134. [PMID: 37561545 DOI: 10.1096/fj.202300272r] [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: 02/14/2023] [Revised: 06/19/2023] [Accepted: 07/27/2023] [Indexed: 08/11/2023]
Abstract
3-Phosphoinositide-dependent protein kinase-1 (Pdk1) as a serine/threonine protein kinase plays a critical role in multiple signaling pathways. Analysis of the gene expression omnibus database showed that Pdk1 was significantly downregulated in patients with heart diseases. Gene set enrichment analysis of the proteomics dataset identified apoptotic- and metabolism-related signaling pathways directly targeted by Pdk1. Previously, our research indicated that Pdk1 deletion-induced metabolic changes might be involved in the pathogenesis of heart failure; however, the underlying mechanism remains elusive. Here, we demonstrated that deficiency of Pdk1 resulted in apoptosis, oxidative damage, and disturbed metabolism, both in vivo and in vitro. Furthermore, profiling of metabonomics by 1 H-NMR demonstrated that taurine was the major differential metabolite in the heart of Pdk1-knockout mice. Taurine treatment significantly reduced the reactive oxygen species production and apoptosis, improved cardiac function, and prolonged the survival time in Pdk1 deficient mice. Proteomic screening identified solute carrier family 6 member 6 (Slc6a6) as the downstream that altered taurine levels in Pdk1-expression cells. Consistently, cellular apoptosis and oxidative damage were rescued by Slc6a6 in abnormal Pdk1 expression cells. These findings collectively suggest that Pdk1 deficiency induces heart failure via disturbances in taurine homeostasis, triggered by Slc6a6.
Collapse
Affiliation(s)
- Chen Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Efficacy Evaluation of Traditional Chinese Medicine and Encephalopathy Research of Zhejiang Province, Wenzhou, China
| | - Yi Zhou
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yan Niu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wenting He
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xinyi Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xi Zhang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yali Wu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wenli Zhang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Liangcai Zhao
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Hong Zheng
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Weihong Song
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, China
| | - Hongchang Gao
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Efficacy Evaluation of Traditional Chinese Medicine and Encephalopathy Research of Zhejiang Province, Wenzhou, China
| |
Collapse
|
13
|
Interdonato L, Impellizzeri D, D’Amico R, Cordaro M, Siracusa R, D’Agostino M, Genovese T, Gugliandolo E, Crupi R, Fusco R, Cuzzocrea S, Di Paola R. Modulation of TLR4/NFκB Pathways in Autoimmune Myocarditis. Antioxidants (Basel) 2023; 12:1507. [PMID: 37627502 PMCID: PMC10451772 DOI: 10.3390/antiox12081507] [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: 06/26/2023] [Revised: 07/14/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023] Open
Abstract
Myocarditis is an inflammatory and oxidative disorder characterized by immune cell recruitment in the damaged tissue and organ dysfunction. In this paper, we evaluated the molecular pathways involved in myocarditis using a natural compound, Coriolus versicolor, in an experimental model of autoimmune myocarditis (EAM). Animals were immunized with an emulsion of pig cardiac myosin and complete Freund's adjuvant supplemented with mycobacterium tuberculosis; thereafter, Coriolus versicolor (200 mg/Kg) was orally administered for 21 days. At the end of the experiment, blood pressure and heart rate measurements were recorded and the body and heart weights as well. From the molecular point of view, the Coriolus versicolor administration reduced the activation of the TLR4/NF-κB pathway and the levels of pro-inflammatory cytokines (INF-γ, TNF-α, IL-6, IL-17, and IL-2) and restored the levels of anti-inflammatory cytokines (IL-10). These anti-inflammatory effects were accompanied with a reduced lipid peroxidation and nitrite levels and restored the antioxidant enzyme activities (SOD and CAT) and GSH levels. Additionally, it reduced the histological injury and the immune cell recruitment (CD4+ and CD68+ cells). Moreover, we observed an antiapoptotic activity in both intrinsic (Fas/FasL/caspase-3) and extrinsic (Bax/Bcl-2) pathways. Overall, our data showed that Coriolus versicolor administration modulates the TLR4/NF-κB signaling in EAM.
Collapse
Affiliation(s)
- Livia Interdonato
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Ramona D’Amico
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Marika Cordaro
- Department of Biomedical, Dental and Morphological and Functional Imaging, University of Messina, Consolare Valeria, 98100 Messina, Italy
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Melissa D’Agostino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Tiziana Genovese
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Enrico Gugliandolo
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Rosalia Crupi
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Rosanna Di Paola
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| |
Collapse
|
14
|
Borger M, von Haefen C, Bührer C, Endesfelder S. Cardioprotective Effects of Dexmedetomidine in an Oxidative-Stress In Vitro Model of Neonatal Rat Cardiomyocytes. Antioxidants (Basel) 2023; 12:1206. [PMID: 37371938 DOI: 10.3390/antiox12061206] [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: 05/03/2023] [Revised: 05/25/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
Preterm birth is a risk factor for cardiometabolic disease. The preterm heart before terminal differentiation is in a phase that is crucial for the number and structure of cardiomyocytes in further development, with adverse effects of hypoxic and hyperoxic events. Pharmacological intervention could attenuate the negative effects of oxygen. Dexmedetomidine (DEX) is an α2-adrenoceptor agonist and has been mentioned in connection with cardio-protective benefits. In this study, H9c2 myocytes and primary fetal rat cardiomyocytes (NRCM) were cultured for 24 h under hypoxic condition (5% O2), corresponding to fetal physioxia (pO2 32-45 mmHg), ambient oxygen (21% O2, pO2 ~150 mmHg), or hyperoxic conditions (80% O2, pO2 ~300 mmHg). Subsequently, the effects of DEX preconditioning (0.1 µM, 1 µM, 10 µM) were analyzed. Modulated oxygen tension reduced both proliferating cardiomyocytes and transcripts (CycD2). High-oxygen tension induced hypertrophy in H9c2 cells. Cell-death-associated transcripts for caspase-dependent apoptosis (Casp3/8) increased, whereas caspase-independent transcripts (AIF) increased in H9c2 cells and decreased in NRCMs. Autophagy-related mediators (Atg5/12) were induced in H9c2 under both oxygen conditions, whereas they were downregulated in NRCMs. DEX preconditioning protected H9c2 and NRCMs from oxidative stress through inhibition of transcription of the oxidative stress marker GCLC, and inhibited the transcription of both the redox-sensitive transcription factors Nrf2 under hyperoxia and Hif1α under hypoxia. In addition, DEX normalized the gene expression of Hippo-pathway mediators (YAP1, Tead1, Lats2, Cul7) that exhibited abnormalities due to differential oxygen tensions compared with normoxia, suggesting that DEX modulates the activation of the Hippo pathway. This, in the context of the protective impact of redox-sensitive factors, may provide a possible rationale for the cardio-protective effects of DEX in oxygen-modulated requirements on survival-promoting transcripts of immortalized and fetal cardiomyocytes.
Collapse
Affiliation(s)
- Moritz Borger
- Department of Neonatology, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Clarissa von Haefen
- Department of Anesthesiology and Intensive Care Medicine, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Christoph Bührer
- Department of Neonatology, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Stefanie Endesfelder
- Department of Neonatology, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| |
Collapse
|
15
|
Hou Y, He Z, Han Y, Zhang T, Wang S, Wang X, Mao J. Mechanism of new optimized Sheng-Mai-San Formula to regulate cardiomyocyte apoptosis through NMDAR pathway. Heliyon 2023; 9:e16631. [PMID: 37416647 PMCID: PMC10320033 DOI: 10.1016/j.heliyon.2023.e16631] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 05/21/2023] [Accepted: 05/23/2023] [Indexed: 07/08/2023] Open
Abstract
Background and objectives Ischemic heart failure (HF) has become a disease that seriously endangers people's life and health. As a herbal formula widely used in clinical practice, new optimized Sheng-Mai-San (NO-SMS) has been shown to be significantly effective in improving cardiac function, increasing exercise tolerance, and slowing the progression of myocardial fibrosis in heart failure patients in multi-center clinical studies in various regions of China. In our previous pharmacodynamic and toxicological studies, we found that a medium-dose formulation (8.1 g of raw drug/kg) was the most effective in the treatment of heart failure, but its mechanism of action is still being investigated. The present study is exploring its relationship with cardiomyocyte apoptosis. Materials and methods We investigated and verified this through two parts of experiments, in vivo and in vitro. Firstly, we prepared male SD rats with heart failure models by ligating the left anterior descending branch of the coronary artery (EF ≤ 50%), which were treated with NO-SMS Formula (8.1 g of raw drug/kg/d), Ifenprodil (5.4 mg/kg/d) or Enalapril (0.9 mg/kg/d) prepared suspensions by gavage for 4 weeks. The cardiac and structural changes were evaluated by echocardiography, H&E, and MASSON staining. The apoptosis of cardiomyocytes in each group was detected by Western blot, qRT-PCR, and ELISA. In vitro cell experiments include H9c2 cardiomyocyte injury induced by H2O2 and NMDA respectively, and the groups were incubated with NO-SMS and Ifenprodil-containing serum for 24 h. Apoptosis was detected by Annexin V-FITC/PI double-staining method, and the rest of the assays were consistent with the in vivo experiments. Results Compared with the model group, the NO-SMS formula group and the Ifenprodil group could significantly improve cardiac function, delay myocardial fibrosis, reduce the expression of pro-apoptotic proteins, mRNA, and the concentration levels of Ca2+ and ROS in heart failure rats and H9c2 cardiomyocytes with H2O2 and NMDA-induced injury, which could significantly reduce the apoptosis rate of damaged cardiomyocytes and effectively inhibit the apoptosis of cardiomyocytes. Conclusion NO-SMS Formula improved cardiac function, inhibited ventricular remodeling and cardiomyocyte apoptosis in HF rats, and its mechanism may be related to the regulation of the NMDAR signaling pathway, inhibition of large intracellular Ca2+ inward flow, and ROS production in cardiomyocytes.
Collapse
Affiliation(s)
- Yazhu Hou
- Department of Cardiovascular Diseases, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Zixun He
- Department of Cardiovascular Diseases, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yixiao Han
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Tongyan Zhang
- Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300150, China
| | - Shuai Wang
- Department of Cardiovascular Diseases, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Xianliang Wang
- Department of Cardiovascular Diseases, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Jingyuan Mao
- Department of Cardiovascular Diseases, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| |
Collapse
|
16
|
Brandão SR, Reis-Mendes A, Araújo MD, Neuparth MJ, Rocha H, Carvalho F, Ferreira R, Costa VM. Cardiac Molecular Remodeling by Anticancer Drugs: Doxorubicin Affects More Metabolism While Mitoxantrone Impacts More Autophagy in Adult CD-1 Male Mice. Biomolecules 2023; 13:921. [PMID: 37371499 PMCID: PMC10296231 DOI: 10.3390/biom13060921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/14/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
Doxorubicin (DOX) and mitoxantrone (MTX) are classical chemotherapeutic agents used in cancer that induce similar clinical cardiotoxic effects, although it is not clear if they share similar underlying molecular mechanisms. We aimed to assess the effects of DOX and MTX on the cardiac remodeling, focusing mainly on metabolism and autophagy. Adult male CD-1 mice received pharmacologically relevant cumulative doses of DOX (18 mg/kg) and MTX (6 mg/kg). Both DOX and MTX disturbed cardiac metabolism, decreasing glycolysis, and increasing the dependency on fatty acids (FA) oxidation, namely, through decreased AMP-activated protein kinase (AMPK) and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) content and decreased free carnitine (C0) and increased acetylcarnitine (C2) concentration. Additionally, DOX heavily influenced glycolysis, oxidative metabolism, and amino acids turnover by exclusively decreasing phosphofructokinase (PFKM) and electron transfer flavoprotein-ubiquinone oxidoreductase (ETFDH) content, and the concentration of several amino acids. Conversely, both drugs downregulated autophagy given by the decreased content of autophagy protein 5 (ATG5) and microtubule-associated protein light chain 3 (LC3B), with MTX having also an impact on Beclin1. These results emphasize that DOX and MTX modulate cardiac remodeling differently, despite their clinical similarities, which is of paramount importance for future treatments.
Collapse
Affiliation(s)
- Sofia Reis Brandão
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Laboratory of Toxicology, UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Ana Reis-Mendes
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Laboratory of Toxicology, UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Margarida Duarte Araújo
- LAQV-REQUIMTE, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Department of Imuno-Physiology and Pharmacology, Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
| | - Maria João Neuparth
- Laboratory for Integrative and Translational Research in Population Health (ITR), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sports, University of Porto, 4200-450 Porto, Portugal
- TOXRUN-Toxicology Research Unit, University Institute of Health Sciences, CESPU, 4585-116 Gandra, Portugal
| | - Hugo Rocha
- Newborn Screening, Metabolism and Genetics Unit, Human Genetics Department, National Institute of Health Doutor Ricardo Jorge, 4000-053 Porto, Portugal
- Department of Pathological, Cytological and Thanatological Anatomy, School of Health, Polytechnic Institute of Porto, 4200-072 Porto, Portugal
| | - Félix Carvalho
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Laboratory of Toxicology, UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Rita Ferreira
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Vera Marisa Costa
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Laboratory of Toxicology, UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
17
|
Li J, Zhao X, Wu B, Ji Z, Liu H, Wang X, Zhang H, He Z. Non-invasive detection and differentiation of apoptotic and necroptotic cell death in vitro. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2023; 244:112730. [PMID: 37229972 DOI: 10.1016/j.jphotobiol.2023.112730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 03/03/2023] [Accepted: 05/18/2023] [Indexed: 05/27/2023]
Abstract
Cell death plays an important role in the development of multicellular organisms and the maintenance of adult homeostasis. However, traditional methods of cell death detection can damage cells and tissues. Here, we report the use of near-infrared (NIR) spectroscopy for non-invasively distinguishing between cell death types. We found a difference between normal, apoptotic, and necroptotic mouse dermal fibroblast cells in the wavelength range of 1100-1700 nm. In particular, the differences in scattering of NIR light between cells at different states are enough to be distinguished. This feature was exploited by measuring the attenuation coefficient (δμ), which specifies the ease at which light can pass through a substance. The results showed that δμ can be used to distinguish between different types of cell death. Therefore, this study proposes a new, non-invasive, and fast method to differentiate cell death types without the additional fluorescent labeling.
Collapse
Affiliation(s)
- Jinning Li
- Key Laboratory of Space Active Opto-Electronics Technology, Shanghai Institute of Technical Physics, Chinese Academy of Sciences, Shanghai 200083, PR China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoming Zhao
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200083, PR China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bing Wu
- Key Laboratory of Space Active Opto-Electronics Technology, Shanghai Institute of Technical Physics, Chinese Academy of Sciences, Shanghai 200083, PR China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhongpeng Ji
- Key Laboratory of Space Active Opto-Electronics Technology, Shanghai Institute of Technical Physics, Chinese Academy of Sciences, Shanghai 200083, PR China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Han Liu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200083, PR China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xuehan Wang
- Key Laboratory of Space Active Opto-Electronics Technology, Shanghai Institute of Technical Physics, Chinese Academy of Sciences, Shanghai 200083, PR China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Haibing Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200083, PR China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Zhiping He
- Key Laboratory of Space Active Opto-Electronics Technology, Shanghai Institute of Technical Physics, Chinese Academy of Sciences, Shanghai 200083, PR China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
18
|
Guo Y, Tang Y, Lu G, Gu J. p53 at the Crossroads between Doxorubicin-Induced Cardiotoxicity and Resistance: A Nutritional Balancing Act. Nutrients 2023; 15:nu15102259. [PMID: 37242146 DOI: 10.3390/nu15102259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/19/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
Doxorubicin (DOX) is a highly effective chemotherapeutic drug, but its long-term use can cause cardiotoxicity and drug resistance. Accumulating evidence demonstrates that p53 is directly involved in DOX toxicity and resistance. One of the primary causes for DOX resistance is the mutation or inactivation of p53. Moreover, because the non-specific activation of p53 caused by DOX can kill non-cancerous cells, p53 is a popular target for reducing toxicity. However, the reduction in DOX-induced cardiotoxicity (DIC) via p53 suppression is often at odds with the antitumor advantages of p53 reactivation. Therefore, in order to increase the effectiveness of DOX, there is an urgent need to explore p53-targeted anticancer strategies owing to the complex regulatory network and polymorphisms of the p53 gene. In this review, we summarize the role and potential mechanisms of p53 in DIC and resistance. Furthermore, we focus on the advances and challenges in applying dietary nutrients, natural products, and other pharmacological strategies to overcome DOX-induced chemoresistance and cardiotoxicity. Lastly, we present potential therapeutic strategies to address key issues in order to provide new ideas for increasing the clinical use of DOX and improving its anticancer benefits.
Collapse
Affiliation(s)
- Yuanfang Guo
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yufeng Tang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
| | - Guangping Lu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Junlian Gu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| |
Collapse
|
19
|
Leancă SA, Afrăsânie I, Crișu D, Matei IT, Duca ȘT, Costache AD, Onofrei V, Tudorancea I, Mitu O, Bădescu MC, Șerban LI, Costache II. Cardiac Reverse Remodeling in Ischemic Heart Disease with Novel Therapies for Heart Failure with Reduced Ejection Fraction. Life (Basel) 2023; 13:1000. [PMID: 37109529 PMCID: PMC10143569 DOI: 10.3390/life13041000] [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: 03/07/2023] [Revised: 04/03/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Despite the improvements in the treatment of coronary artery disease (CAD) and acute myocardial infarction (MI) over the past 20 years, ischemic heart disease (IHD) continues to be the most common cause of heart failure (HF). In clinical trials, over 70% of patients diagnosed with HF had IHD as the underlying cause. Furthermore, IHD predicts a worse outcome for patients with HF, leading to a substantial increase in late morbidity, mortality, and healthcare costs. In recent years, new pharmacological therapies have emerged for the treatment of HF, such as sodium-glucose cotransporter-2 inhibitors, angiotensin receptor-neprilysin inhibitors, selective cardiac myosin activators, and oral soluble guanylate cyclase stimulators, demonstrating clear or potential benefits in patients with HF with reduced ejection fraction. Interventional strategies such as cardiac resynchronization therapy, cardiac contractility modulation, or baroreflex activation therapy might provide additional therapeutic benefits by improving symptoms and promoting reverse remodeling. Furthermore, cardiac regenerative therapies such as stem cell transplantation could become a new therapeutic resource in the management of HF. By analyzing the existing data from the literature, this review aims to evaluate the impact of new HF therapies in patients with IHD in order to gain further insight into the best form of therapeutic management for this large proportion of HF patients.
Collapse
Affiliation(s)
- Sabina Andreea Leancă
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Irina Afrăsânie
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Daniela Crișu
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Iulian Theodor Matei
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Ștefania Teodora Duca
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Alexandru Dan Costache
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
- Department of Cardiovascular Rehabilitation, Clinical Rehabilitation Hospital, 700661 Iași, Romania
| | - Viviana Onofrei
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Ionuţ Tudorancea
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Ovidiu Mitu
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Minerva Codruța Bădescu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Lăcrămioara Ionela Șerban
- Department of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Irina Iuliana Costache
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| |
Collapse
|
20
|
Meng Z, Zhang Z, Zhao J, Liu C, Yao P, Zhang L, Xie D, Lau WB, Tsukuda J, Christopher TA, Lopez B, Zhu D, Liu D, Zhang JR, Gao E, Ischiropoulos H, Koch W, Ma X, Wang Y. Nitrative Modification of Caveolin-3: A Novel Mechanism of Cardiac Insulin Resistance and a Potential Therapeutic Target Against Ischemic Heart Failure in Prediabetic Animals. Circulation 2023; 147:1162-1179. [PMID: 36883479 PMCID: PMC10085855 DOI: 10.1161/circulationaha.122.063073] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 02/07/2023] [Indexed: 03/09/2023]
Abstract
BACKGROUND Myocardial insulin resistance is a hallmark of diabetic cardiac injury. However, the underlying molecular mechanisms remain unclear. Recent studies demonstrate that the diabetic heart is resistant to other cardioprotective interventions, including adiponectin and preconditioning. The "universal" resistance to multiple therapeutic interventions suggests impairment of the requisite molecule(s) involved in broad prosurvival signaling cascades. Cav (Caveolin) is a scaffolding protein coordinating transmembrane signaling transduction. However, the role of Cav3 in diabetic impairment of cardiac protective signaling and diabetic ischemic heart failure is unknown. METHODS Wild-type and gene-manipulated mice were fed a normal diet or high-fat diet for 2 to 12 weeks and subjected to myocardial ischemia and reperfusion. Insulin cardioprotection was determined. RESULTS Compared with the normal diet group, the cardioprotective effect of insulin was significantly blunted as early as 4 weeks of high-fat diet feeding (prediabetes), a time point where expression levels of insulin-signaling molecules remained unchanged. However, Cav3/insulin receptor-β complex formation was significantly reduced. Among multiple posttranslational modifications altering protein/protein interaction, Cav3 (not insulin receptor-β) tyrosine nitration is prominent in the prediabetic heart. Treatment of cardiomyocytes with 5-amino-3-(4-morpholinyl)-1,2,3-oxadiazolium chloride reduced the signalsome complex and blocked insulin transmembrane signaling. Mass spectrometry identified Tyr73 as the Cav3 nitration site. Phenylalanine substitution of Tyr73 (Cav3Y73F) abolished 5-amino-3-(4-morpholinyl)-1,2,3-oxadiazolium chloride-induced Cav3 nitration, restored Cav3/insulin receptor-β complex, and rescued insulin transmembrane signaling. It is most important that adeno-associated virus 9-mediated cardiomyocyte-specific Cav3Y73F reexpression blocked high-fat diet-induced Cav3 nitration, preserved Cav3 signalsome integrity, restored transmembrane signaling, and rescued insulin-protective action against ischemic heart failure. Last, diabetic nitrative modification of Cav3 at Tyr73 also reduced Cav3/AdipoR1 complex formation and blocked adiponectin cardioprotective signaling. CONCLUSIONS Nitration of Cav3 at Tyr73 and resultant signal complex dissociation results in cardiac insulin/adiponectin resistance in the prediabetic heart, contributing to ischemic heart failure progression. Early interventions preserving Cav3-centered signalsome integrity is an effective novel strategy against diabetic exacerbation of ischemic heart failure.
Collapse
Affiliation(s)
- Zhijun Meng
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Zhen Zhang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Jianli Zhao
- Department of Biomedical Engineering, the University of Alabama at Birmingham, AL 35005
| | - Caihong Liu
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Peng Yao
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Ling Zhang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Dina Xie
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Wayne Bond Lau
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Jumpei Tsukuda
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | | | - Bernard Lopez
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Di Zhu
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Demin Liu
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - John Ry Zhang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Erhe Gao
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140
| | - Harry Ischiropoulos
- Children’s Hospital of Philadelphia Research Institute, Philadelphia, PA 19104
| | - Walter Koch
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140
| | - Xinliang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Yajing Wang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
- Department of Biomedical Engineering, the University of Alabama at Birmingham, AL 35005
| |
Collapse
|
21
|
Liu Y, Wang Y, Bi Y, Zhao Z, Wang S, Lin S, Yang Z, Wang X, Mao J. Emerging role of mitophagy in heart failure: from molecular mechanism to targeted therapy. Cell Cycle 2023; 22:906-918. [PMID: 36658777 PMCID: PMC10054314 DOI: 10.1080/15384101.2023.2167949] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 01/09/2023] [Indexed: 01/21/2023] Open
Abstract
Heart failure is defined as a drop in heart's pump function, accounting for reduced blood output and venous stasis, and constitutes the end stage of various cardiovascular diseases. Although mild mitochondrial dysfunction may hinder cardiomyocyte metabolism and impair myocardial function, severe mitochondrial injury is accompanied by cardiomyocyte apoptosis, leading to irreversible damage of the heart. Selective autophagy of mitochondria, or mitophagy, serves to rapidly remove dysfunctional mitochondria and restore the health of the mitochondrial population within cells by allowing reutilization of degradative substrates such as amino acids, fatty acids, and nucleotides. Although mitophagy represents a protective program that prevents the accumulation of poorly structured or damaged mitochondria, excessive mitophagy leads to mitochondrial population decline, impaired oxidative phosphorylation, and decreased ATP production. In this review, we first discuss the molecular underpinnings of mitophagy and the roles of different mitophagy adaptors. Then, the multiple and complex influence of mitophagy on heart failure is summarized. Finally, novel pharmacological strategies targeting mitophagy to relieve heart failure are briefly summarized.
Collapse
Affiliation(s)
- Yu Liu
- Department of Cardiology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine/National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yizhou Wang
- Rehabilitation Department, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
| | - Yingfei Bi
- Department of Cardiology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine/National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Zhiqiang Zhao
- Department of Cardiology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine/National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Shuai Wang
- Department of Cardiology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine/National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Shanshan Lin
- Department of Cardiology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine/National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Zhihua Yang
- Department of Cardiology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine/National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Xianliang Wang
- Department of Cardiology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine/National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Jingyuan Mao
- Department of Cardiology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine/National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
22
|
Fratta Pasini AM, Stranieri C, Busti F, Di Leo EG, Girelli D, Cominacini L. New Insights into the Role of Ferroptosis in Cardiovascular Diseases. Cells 2023; 12:cells12060867. [PMID: 36980208 PMCID: PMC10047059 DOI: 10.3390/cells12060867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/14/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the principal cause of disease burden and death worldwide. Ferroptosis is a new form of regulated cell death mainly characterized by altered iron metabolism, increased polyunsaturated fatty acid peroxidation by reactive oxygen species, depletion of glutathione and inactivation of glutathione peroxidase 4. Recently, a series of studies have indicated that ferroptosis is involved in the death of cardiac and vascular cells and has a key impact on the mechanisms leading to CVDs such as ischemic heart disease, ischemia/reperfusion injury, cardiomyopathies, and heart failure. In this article, we reviewed the molecular mechanism of ferroptosis and the current understanding of the pathophysiological role of ferroptosis in ischemic heart disease and in some cardiomyopathies. Moreover, the comprehension of the machinery governing ferroptosis in vascular cells and cardiomyocytes may provide new insights into preventive and therapeutic strategies in CVDs.
Collapse
|
23
|
Xie J. Long Noncoding RNA XIST Regulates Myocardial Infarction via miR-486-5p/SIRT1 Axis. Appl Biochem Biotechnol 2023; 195:725-734. [PMID: 36129595 DOI: 10.1007/s12010-022-04165-3] [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] [Accepted: 07/15/2022] [Indexed: 01/25/2023]
Abstract
Myocardial infarction (MI) is severe heart disease leading to the death worldwide. Long noncoding RNAs (lncRNAs) play a vital role in progression of numerous heart diseases. In the present study, we examined the effects of lncRNA XIST and underlying mechanism on hypoxia-induced apoptosis. In vitro model of MI was established by inducing hypoxia in H9c2 cells. CCK-8 assay was used to measure the cell viability in hypoxia-induced H9c2 cells. The rate of cell apoptosis was assessed by using caspase-3 assay. Transfection was carried out to upregulate the expressions of lncRNA XIST, and miR-486-5p. RT-qPCR was used to measure the levels of lncRNA XIST and miR-486-5p. Also, the relation between XIST and miR-486-5p was confirmed by using Luciferase reporter assay. Our findings revealed that hypoxia significantly downregulated the expressions of XIST. Also, the cell viability markedly increased due to the overexpression of XIST in hypoxia-induced H9c2 cells, while overexpression of XIST significantly reduced the cell apoptosis in hypoxia-induced H9c2 cells. On the other hand, opposite effects were observed due to the downregulation of XIST in hypoxia-induced H9c2 cells. Moreover, XIST negatively regulated the expression of miR-4486-5p and upregulation of XIST inhibited hypoxic injury by downregulating miR-486-5p. Furthermore, SIRT1 expression was negatively regulated by miR-486-5p. We concluded that lncRNA XIST might provide protection against injury induced by hypoxia via miR-486-5p/SIRT1 axis.
Collapse
Affiliation(s)
- Jiayong Xie
- Department of of Nephrology, Xinghua People's Hospital, Xinghua, 225700, Jiangsu, China.
| |
Collapse
|
24
|
Dexmedetomidine Inhibits Parthanatos in Cardiomyocytes and in Aortic Banded Mice by the ROS-Mediated NLRP3 Inflammasome Activation. J Cardiovasc Transl Res 2022:10.1007/s12265-022-10340-y. [DOI: 10.1007/s12265-022-10340-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 11/04/2022] [Indexed: 11/24/2022]
|
25
|
Brandão SR, Carvalho F, Amado F, Ferreira R, Costa VM. Insights on the molecular targets of cardiotoxicity induced by anticancer drugs: A systematic review based on proteomic findings. Metabolism 2022; 134:155250. [PMID: 35809654 DOI: 10.1016/j.metabol.2022.155250] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 06/02/2022] [Accepted: 06/26/2022] [Indexed: 11/27/2022]
Abstract
Several anticancer agents have been associated with cardiac toxic effects. The currently proposed mechanisms to explain cardiotoxicity differ among anticancer agents, but in fact, the specific modulation is not completely elucidated. Thus, this systematic review aims to provide an integrative perspective of the molecular mechanisms underlying the toxicity of anticancer agents on heart muscle while using a high-throughput technology, mass spectrometry (MS)-based proteomics. A literature search using PubMed database led to the selection of 27 studies, of which 13 reported results exclusively on animal models, 13 on cardiomyocyte-derived cell lines and only one included both animal and a cardiomyocyte line. The reported anticancer agents were the proteasome inhibitor carfilzomib, the anthracyclines daunorubicin, doxorubicin, epirubicin and idarubicin, the antimicrotubule agent docetaxel, the alkylating agent melphalan, the anthracenedione mitoxantrone, the tyrosine kinase inhibitors (TKIs) erlotinib, lapatinib, sorafenib and sunitinib, and the monoclonal antibody trastuzumab. Regarding the MS-based proteomic approaches, electrophoretic separation using two-dimensional (2D) gels coupled with tandem MS (MS/MS) and liquid chromatography-MS/MS (LC-MS/MS) were the most common. Overall, the studies highlighted 1826 differentially expressed proteins across 116 biological processes. Most of them were grouped in larger processes and critically analyzed in the present review. The selection of studies using proteomics on heart muscle allowed to obtain information about the anticancer therapy-induced modulation of numerous proteins in this tissue and to establish connections that have been disregarded in other studies. This systematic review provides interesting points for a comprehensive understanding of the cellular cardiotoxicity mechanisms of different anticancer drugs.
Collapse
Affiliation(s)
- Sofia Reis Brandão
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira n° 28, 4050-313 Porto, Portugal; LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - Félix Carvalho
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira n° 28, 4050-313 Porto, Portugal
| | - Francisco Amado
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Rita Ferreira
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Vera Marisa Costa
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira n° 28, 4050-313 Porto, Portugal.
| |
Collapse
|
26
|
Liu M, Lv J, Pan Z, Wang D, Zhao L, Guo X. Mitochondrial dysfunction in heart failure and its therapeutic implications. Front Cardiovasc Med 2022; 9:945142. [PMID: 36093152 PMCID: PMC9448986 DOI: 10.3389/fcvm.2022.945142] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/04/2022] [Indexed: 11/18/2022] Open
Abstract
The ATP consumption in heart is very intensive to support muscle contraction and relaxation. Mitochondrion is the power plant of the cell. Mitochondrial dysfunction has long been believed as the primary mechanism responsible for the inability of energy generation and utilization in heart failure. In addition, emerging evidence has demonstrated that mitochondrial dysfunction also contributes to calcium dysregulation, oxidative stress, proteotoxic insults and cardiomyocyte death. These elements interact with each other to form a vicious circle in failing heart. The role of mitochondrial dysfunction in the pathogenesis of heart failure has attracted increasing attention. The complex signaling of mitochondrial quality control provides multiple targets for maintaining mitochondrial function. Design of therapeutic strategies targeting mitochondrial dysfunction holds promise for the prevention and treatment of heart failure.
Collapse
Affiliation(s)
- Miaosen Liu
- Clinical Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Jialan Lv
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhicheng Pan
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dongfei Wang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liding Zhao
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaogang Guo
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Xiaogang Guo,
| |
Collapse
|
27
|
Dutta A, Das M, Ghosh A, Rana S. Molecular and cellular pathophysiology of circulating cardiomyocyte-specific cell free DNA (cfDNA): Biomarkers of heart failure and potential therapeutic targets. Genes Dis 2022. [DOI: 10.1016/j.gendis.2022.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022] Open
|
28
|
Moloce MA, Costache II, Nicolae A, Onofrei Aursulesei V. Pharmacological Targets in Chronic Heart Failure with Reduced Ejection Fraction. Life (Basel) 2022; 12:1112. [PMID: 35892914 PMCID: PMC9394280 DOI: 10.3390/life12081112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 11/17/2022] Open
Abstract
Heart failure management has been repeatedly reviewed over time. This strategy has resulted in improved quality of life, especially in patients with heart failure with reduced ejection fraction (HFrEF). It is for this reason that new mechanisms involved in the development and progression of heart failure, along with specific therapies, have been identified. This review focuses on the most recent guidelines of therapeutic interventions, trials that explore novel therapies, and also new molecules that could improve prognosis of different HFrEF phenotypes.
Collapse
Affiliation(s)
- Maria-Angela Moloce
- Iasi “Saint Spiridon” County Hospital, 700111 Iasi, Romania; (M.-A.M.); (I.-I.C.); (V.O.A.)
| | - Irina-Iuliana Costache
- Iasi “Saint Spiridon” County Hospital, 700111 Iasi, Romania; (M.-A.M.); (I.-I.C.); (V.O.A.)
- Department of Internal Medicine (Cardiology), Iasi “Grigore T. Popa” University of Medicine and Pharmacy, 700111 Iasi, Romania
| | - Ana Nicolae
- Department of Internal Medicine (Cardiology), Iasi “Grigore T. Popa” University of Medicine and Pharmacy, 700111 Iasi, Romania
| | - Viviana Onofrei Aursulesei
- Iasi “Saint Spiridon” County Hospital, 700111 Iasi, Romania; (M.-A.M.); (I.-I.C.); (V.O.A.)
- Department of Internal Medicine (Cardiology), Iasi “Grigore T. Popa” University of Medicine and Pharmacy, 700111 Iasi, Romania
| |
Collapse
|
29
|
Zhou J, Xu J, Li P, Sun S, Kadier Y, Zhou S, Cheng A. Necroptosis and Viral Myocarditis: Tumor Necrosis Factor α as a Novel Biomarker for the Diagnosis of Viral Myocarditis. Front Cell Dev Biol 2022; 10:826904. [PMID: 35602592 PMCID: PMC9114881 DOI: 10.3389/fcell.2022.826904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Programmed cell death (PCD), including necroptosis, has emerged as a significant pathway in cardiovascular diseases. The infection of viral myocarditis (VMC) could cause cardiomyocytes degeneration, necrosis, and immune-inflammatory myocardial response. In this review, we summarized and evaluated the available evidence on the pathogenesis, molecule mechanism, diagnosis, and potential treatment strategies of viral myocarditis, with a special focus on the novel mechanism of necroptosis for cardiomyocytes death. Studies have shown that tumor necrosis factor-alpha (TNF-α) is an important cytokine involved in the activation of necroptosis; an elevated level of TNF-α is continually reported in patients suffering from VMC, implicating its involvement in the pathogenesis of VMC. It is of great interest to explore the clinical implication of TNF-α. We subsequently conducted a meta-analysis on the efficacy of serum TNF-α expression level and its diagnostic accuracy on acute viral myocarditis detection. Taken together, the review demonstrates a compelling role of necroptosis involved in the pathogenesis of VMC. Further, applying TNF-α as a serological marker for the diagnosis of VMC may be a useful strategy.
Collapse
Affiliation(s)
- Jin Zhou
- Tianjin Chest Hospital, Tianjin, China
| | - Jing Xu
- Tianjin Chest Hospital, Tianjin, China
| | - Peng Li
- Tianjin Chest Hospital, Tianjin, China
| | - Shan Sun
- Tianjin Chest Hospital, Tianjin, China
| | | | - Shiying Zhou
- Hotan District People’s Hospital, Tianjin, China
| | - Aijuan Cheng
- Tianjin Chest Hospital, Tianjin, China
- *Correspondence: Aijuan Cheng,
| |
Collapse
|
30
|
Signaling cascades in the failing heart and emerging therapeutic strategies. Signal Transduct Target Ther 2022; 7:134. [PMID: 35461308 PMCID: PMC9035186 DOI: 10.1038/s41392-022-00972-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/13/2022] [Accepted: 03/20/2022] [Indexed: 12/11/2022] Open
Abstract
Chronic heart failure is the end stage of cardiac diseases. With a high prevalence and a high mortality rate worldwide, chronic heart failure is one of the heaviest health-related burdens. In addition to the standard neurohormonal blockade therapy, several medications have been developed for chronic heart failure treatment, but the population-wide improvement in chronic heart failure prognosis over time has been modest, and novel therapies are still needed. Mechanistic discovery and technical innovation are powerful driving forces for therapeutic development. On the one hand, the past decades have witnessed great progress in understanding the mechanism of chronic heart failure. It is now known that chronic heart failure is not only a matter involving cardiomyocytes. Instead, chronic heart failure involves numerous signaling pathways in noncardiomyocytes, including fibroblasts, immune cells, vascular cells, and lymphatic endothelial cells, and crosstalk among these cells. The complex regulatory network includes protein-protein, protein-RNA, and RNA-RNA interactions. These achievements in mechanistic studies provide novel insights for future therapeutic targets. On the other hand, with the development of modern biological techniques, targeting a protein pharmacologically is no longer the sole option for treating chronic heart failure. Gene therapy can directly manipulate the expression level of genes; gene editing techniques provide hope for curing hereditary cardiomyopathy; cell therapy aims to replace dysfunctional cardiomyocytes; and xenotransplantation may solve the problem of donor heart shortages. In this paper, we reviewed these two aspects in the field of failing heart signaling cascades and emerging therapeutic strategies based on modern biological techniques.
Collapse
|
31
|
Fernandez Rico C, Konate K, Josse E, Nargeot J, Barrère-Lemaire S, Boisguérin P. Therapeutic Peptides to Treat Myocardial Ischemia-Reperfusion Injury. Front Cardiovasc Med 2022; 9:792885. [PMID: 35252383 PMCID: PMC8891520 DOI: 10.3389/fcvm.2022.792885] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 01/07/2022] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular diseases (CVD) including acute myocardial infarction (AMI) rank first in worldwide mortality and according to the World Health Organization (WHO), they will stay at this rank until 2030. Prompt revascularization of the occluded artery to reperfuse the myocardium is the only recommended treatment (by angioplasty or thrombolysis) to decrease infarct size (IS). However, despite beneficial effects on ischemic lesions, reperfusion leads to ischemia-reperfusion (IR) injury related mainly to apoptosis. Improvement of revascularization techniques and patient care has decreased myocardial infarction (MI) mortality however heart failure (HF) morbidity is increasing, contributing to the cost-intense worldwide HF epidemic. Currently, there is no treatment for reperfusion injury despite promising results in animal models. There is now an obvious need to develop new cardioprotective strategies to decrease morbidity/mortality of CVD, which is increasing due to the aging of the population and the rising prevalence rates of diabetes and obesity. In this review, we will summarize the different therapeutic peptides developed or used focused on the treatment of myocardial IR injury (MIRI). Therapeutic peptides will be presented depending on their interacting mechanisms (apoptosis, necroptosis, and inflammation) reported as playing an important role in reperfusion injury following myocardial ischemia. The search and development of therapeutic peptides have become very active, with increasing numbers of candidates entering clinical trials. Their optimization and their potential application in the treatment of patients with AMI will be discussed.
Collapse
Affiliation(s)
- Carlota Fernandez Rico
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
- Laboratory of Excellence Ion Channel Science and Therapeutics, Valbonne, France
| | - Karidia Konate
- PHYMEDEXP, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Emilie Josse
- PHYMEDEXP, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Joël Nargeot
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
- Laboratory of Excellence Ion Channel Science and Therapeutics, Valbonne, France
| | - Stéphanie Barrère-Lemaire
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
- Laboratory of Excellence Ion Channel Science and Therapeutics, Valbonne, France
| | - Prisca Boisguérin
- PHYMEDEXP, Université de Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
32
|
Tan Y, Li T, Hu M, Wang B, Zhou Q, Jiang Y, Zhang S, Duan X, Yang J, Liu X, Zhan Z. PHLPP1 deficiency ameliorates cardiomyocyte death and cardiac dysfunction through inhibiting Mcl-1 degradation. Cell Signal 2022; 92:110281. [DOI: 10.1016/j.cellsig.2022.110281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/25/2022] [Accepted: 02/07/2022] [Indexed: 12/31/2022]
|
33
|
Xu S, Xia X, Liu Y, Chen F, Gu R, Bian X, Xu X, Jia C, Lu S, Gu Y, Bai H, Zhang H. Remote cyclic compression ameliorates myocardial infarction injury in rats via AMPK-dependent pathway. Microvasc Res 2022; 141:104313. [PMID: 35041850 DOI: 10.1016/j.mvr.2022.104313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/10/2021] [Accepted: 01/03/2022] [Indexed: 12/25/2022]
Abstract
BACKGROUND Remote ischemic conditioning (RIC) displays a cardioprotective role in acute myocardial infarction (AMI). Since interruption of blood vessel is not an essential trigger of remote cardioprotection, tissue compression may play a prominent part in the effect. The purpose of this study was to confirm the protective effect of tissue compression on AMI and the underlying mechanisms. METHODS AND RESULTS Rat model of AMI was induced by ligation of the left anterior descending coronary artery. Remote cyclic compression (RCC) on forelimb was applied to AMI rats for 3 days after the operation. RCC postconditioning displayed cardioprotective effects against AMI injury by limiting infarct size, alleviating cardiac dysfunction, and suppressing cardiomyocyte apoptosis. In addition, RCC postconditioning induced myocardial autophagy as evidenced by increased LC3-II and Beclin-1 and reduced mTOR levels. Furthermore, RCC treatment upregulated AMPK phosphorylation in the context of AMI hearts. AMPK inhibitor Compound C administration markedly abrogated RCC-mediated cardioprotective effect, as evidenced by decreased infarct size and cardiac function. CONCLUSION Our results indicated that RCC postconditioning could attenuate AMI injury through inhibiting apoptosis and promoting autophagy via AMPK signaling pathway. The research provided a novel perspective for studying the cardioprotection of RIC and possible therapeutic strategy for managing AMI injury.
Collapse
Affiliation(s)
- Senlei Xu
- School of Acupuncture and Tuina, School of Regimen and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China; Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xuefeng Xia
- School of Acupuncture and Tuina, School of Regimen and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuchen Liu
- School of Acupuncture and Tuina, School of Regimen and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Fang Chen
- Dermatological Department, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Renjun Gu
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiangyu Bian
- School of Acupuncture and Tuina, School of Regimen and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xin Xu
- School of Acupuncture and Tuina, School of Regimen and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chengjie Jia
- Wuxi Municipal Rehabilitation Hospital, Wuxi, China
| | - Shengfeng Lu
- School of Acupuncture and Tuina, School of Regimen and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China; Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yihuang Gu
- School of Acupuncture and Tuina, School of Regimen and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hua Bai
- School of Acupuncture and Tuina, School of Regimen and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Hongru Zhang
- School of Acupuncture and Tuina, School of Regimen and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
34
|
Chang JC, Yang KT, Chao TH, Wang IC, Luo YP, Ting PC, Lin JH. Berberine protects cardiac cells against ferroptosis. Tzu Chi Med J 2022; 34:310-317. [PMID: 35912047 PMCID: PMC9333108 DOI: 10.4103/tcmj.tcmj_236_21] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/15/2021] [Accepted: 11/18/2021] [Indexed: 11/07/2022] Open
Abstract
Objectives: Cardiovascular diseases are one of the primary causes of death. Cardiomyocyte loss is a significant feature of cardiac injury. Ferroptosis is iron-dependent cell death, which occurs due to excess iron and reactive oxygen species (ROS) accumulation causing lipid peroxidation, and subsequent cell death. Ferroptosis has been confirmed to mediate ischemia/reperfusion-induced cardiomyopathy and chemotherapy-induced cardiotoxicity. Berberine (BBR) has been proven to protect the heart from cardiomyopathies, including cardiac hypertrophy, heart failure, myocardial infarction, and arrhythmias. It protects cardiomyocytes from apoptosis and autophagy. However, the relation between BBR and ferroptosis is still unknown. This study aimed to confirm if BBR reduces cardiac cell loss via inhibiting ferroptosis. Materials and Methods: We used erastin and Ras-selective lethal small molecule 3 (RSL3) to establish a ferroptosis model in an H9c2 cardiomyoblast cell line and rat neonatal cardiomyocytes to prove that BBR has a protective effect on cardiac cells via inhibiting ferroptosis. Results: In H9c2 cardiomyoblasts, the results showed that BBR reduced erastin and RSL3-induced cell viability loss. Moreover, BBR decreased ROS accumulation and lipid peroxidation in cells induced with ferroptosis. Furthermore, quantitative polymerase chain reaction results showed that Ptgs2 mRNA was reduced in BBR-treated cells. In rat neonatal cardiomyocytes, BBR reduced RSL3-induced loss of cell viability. Conclusion: These results indicated that BBR inhibited ferroptosis via reducing ROS generation and reducing lipid peroxidation in erastin and RSL3-treated cardiac cells.
Collapse
|
35
|
Wei J, Zhao Y, Liang H, Du W, Wang L. Preliminary evidence for the presence of multiple forms of cell death in diabetes cardiomyopathy. Acta Pharm Sin B 2022; 12:1-17. [PMID: 35127369 PMCID: PMC8799881 DOI: 10.1016/j.apsb.2021.08.026] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/25/2021] [Accepted: 07/30/2021] [Indexed: 12/11/2022] Open
Abstract
Diabetic mellitus (DM) is a common degenerative chronic metabolic disease often accompanied by severe cardiovascular complications (DCCs) as major causes of death in diabetic patients with diabetic cardiomyopathy (DCM) as the most common DCC. The metabolic disturbance in DCM generates the conditions/substrates and inducers/triggers and activates the signaling molecules and death executioners leading to cardiomyocyte death which accelerates the development of DCM and the degeneration of DCM to heart failure. Various forms of programmed active cell death including apoptosis, pyroptosis, autophagic cell death, autosis, necroptosis, ferroptosis and entosis have been identified and characterized in many types of cardiac disease. Evidence has also been obtained for the presence of multiple forms of cell death in DCM. Most importantly, published animal experiments have demonstrated that suppression of cardiomyocyte death of any forms yields tremendous protective effects on DCM. Herein, we provide the most updated data on the subject of cell death in DCM, critical analysis of published results focusing on the pathophysiological roles of cell death, and pertinent perspectives of future studies.
Collapse
Affiliation(s)
- Jinjing Wei
- Department of Endocrinology, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Yongting Zhao
- Department of Endocrinology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Haihai Liang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Weijie Du
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Lihong Wang
- Department of Endocrinology, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| |
Collapse
|
36
|
He WS, Wu Y, Ren MJ, Yu ZY, Zhao XS. Diosmetin inhibits apoptosis and activates AMPK-induced autophagy in myocardial damage under hypoxia environment. Kaohsiung J Med Sci 2021; 38:139-148. [PMID: 34713558 DOI: 10.1002/kjm2.12462] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 08/17/2021] [Accepted: 09/06/2021] [Indexed: 01/01/2023] Open
Abstract
Inhibition of hypoxia-induced cardiomyocyte apoptosis is considered as an important treatment method for ischemic heart diseases, but related drugs are still insufficient. The present study aims to explore the protective function and mechanism of the key Chinese medicine monomer diosmetin (DIOS) on the injury of cardiomyocytes induced by hypoxia. Here, AC16 and HCM-a cells were treated with 40 μM of DIOS under hypoxic environment and a hypoxic rat model was built to study the role of DIOS. The viability and autophagy of cardiomyocytes were increased, but the apoptosis of cells was suppressed by 40 μM DIOS, under hypoxic environment. Intriguingly, 10 mM 3-methyladenine, an inhibitor of autophagy, reversed the effect of DIOS on autophagy and apoptosis of the cardiomyocytes under hypoxia. Furthermore, DIOS induced AMP-activated protein kinase (AMPK) activation and Compound C (5 μM), an AMPK inhibitor, attenuated the inhibition of DIOS on the apoptosis of cardiomyocytes under hypoxia environment. In isoprenaline-induced hypoxic rats, it was verified that DIOS inhibited apoptosis, accelerated autophagy, and activated AMPKα pathway in vivo. Our findings indicated that DIOS alleviated hypoxia-induced myocardial apoptosis via inducing the activation of AMPK-induced autophagy. In summary, the study suggested that DIOS inhibited the apoptosis and induced the autophagy of hypoxia-induced cardiomyocytes through AMPK activation.
Collapse
Affiliation(s)
- Wen-Shuai He
- Department of Cardiology, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia Autonomous Region, China
| | - Yun Wu
- Department of Cardiology, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia Autonomous Region, China
| | - Mao-Jia Ren
- Department of Cardiology, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia Autonomous Region, China
| | - Zhong-Yu Yu
- Department of Cardiology, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia Autonomous Region, China
| | - Xing-Sheng Zhao
- Department of Cardiology, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia Autonomous Region, China
| |
Collapse
|
37
|
Zhang L, Yang C, Qiu B. LncRNA RP11-400K9.4 Aggravates Cardiomyocytes Apoptosis After Hypoxia/Reperfusion Injury by Targeting miR-423. Int Heart J 2021; 62:1124-1134. [PMID: 34497168 DOI: 10.1536/ihj.20-828] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Our study was aimed at exploring the roles of lncRNA RP11-400K9.4 (RP11-400K9.4) on hypoxia/reoxygenation (H/R) -induced cardiomyocytes apoptosis. H/R model was constructed in rat primary cardiomyocytes (PC) and H9c2 cells. In this study, the results showed that H/R significantly induced the apoptosis of PC and H9c2 cells. The expression of RP11-400K9.4 was upregulated in H/R-induced PC and H9c2 cells, but miR-423 expression was downregulated. Silencing RP11-400K9.4 could attenuate H/R-induced apoptosis in PC and H9c2 cells. We also found that miR-423 was a potential target of RP11-400K9.4. The effect of silencing RP11-400K9.4 on H/R-induced apoptosis of PC and H9c2 cells was significantly reversed by miR-423 inhibitor transfection. Furthermore, our data confirmed that silencing RP11-400K9.4 promoted the activation of phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) and mitogen-activated protein kinase/extracellular signal-regulated kinase (MEK) /extracellular signal-regulated kinase (ERK) pathways and these phenomena can be reversed by miR-423 inhibitor transfection. In conclusion, our study demonstrated that silencing RP11-400K9.4 could alleviate H/R-induced cardiomyocytes damages via suppressing apoptosis by targeting miR-423 with the activation of PI3K/AKT and MEK/ERK signaling pathways.
Collapse
Affiliation(s)
- Liqiong Zhang
- Department of Geriatrics, Qujing Number 1 People's Hospital
| | - Chao Yang
- Department of ECG Room, Weifang Yidu Central Hospital
| | - Binghua Qiu
- Department of General Practice, Weifang people's Hospital
| |
Collapse
|
38
|
Huang F, Yang R, Xiao Z, Xie Y, Lin X, Zhu P, Zhou P, Lu J, Zheng S. Targeting Ferroptosis to Treat Cardiovascular Diseases: A New Continent to Be Explored. Front Cell Dev Biol 2021; 9:737971. [PMID: 34527678 PMCID: PMC8435746 DOI: 10.3389/fcell.2021.737971] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 08/02/2021] [Indexed: 12/20/2022] Open
Abstract
Cardiovascular diseases, including cardiomyopathy, myocardial infarction, myocardial ischemia/reperfusion injury, heart failure, vascular injury, stroke, and arrhythmia, are correlated with cardiac and vascular cell death. Ferroptosis is a novel form of non-apoptotic regulated cell death which is characterized by an iron-driven accumulation of lethal lipid hydroperoxides. The initiation and execution of ferroptosis are under the control of several mechanisms, including iron metabolism, glutamine metabolism, and lipid peroxidation. Recently, emerging evidence has demonstrated that ferroptosis can play an essential role in the development of various cardiovascular diseases. Recent researches have shown the ferroptosis inhibitors, iron chelators, genetic manipulations, and antioxidants can alleviate myocardial injury by blocking ferroptosis pathway. In this review, we systematically described the mechanisms of ferroptosis and discussed the role of ferroptosis as a novel therapeutic strategy in the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Fangze Huang
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ronghua Yang
- Department of Burn Surgery, The First People's Hospital of Foshan, Foshan, China
| | - Zezhou Xiao
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yu Xie
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xuefeng Lin
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Peng Zhu
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Pengyu Zhou
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jun Lu
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shaoyi Zheng
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
39
|
Balogh V, MacAskill MG, Hadoke PWF, Gray GA, Tavares AAS. Positron Emission Tomography Techniques to Measure Active Inflammation, Fibrosis and Angiogenesis: Potential for Non-invasive Imaging of Hypertensive Heart Failure. Front Cardiovasc Med 2021; 8:719031. [PMID: 34485416 PMCID: PMC8416043 DOI: 10.3389/fcvm.2021.719031] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/22/2021] [Indexed: 12/11/2022] Open
Abstract
Heart failure, which is responsible for a high number of deaths worldwide, can develop due to chronic hypertension. Heart failure can involve and progress through several different pathways, including: fibrosis, inflammation, and angiogenesis. Early and specific detection of changes in the myocardium during the transition to heart failure can be made via the use of molecular imaging techniques, including positron emission tomography (PET). Traditional cardiovascular PET techniques, such as myocardial perfusion imaging and sympathetic innervation imaging, have been established at the clinical level but are often lacking in pathway and target specificity that is important for assessment of heart failure. Therefore, there is a need to identify new PET imaging markers of inflammation, fibrosis and angiogenesis that could aid diagnosis, staging and treatment of hypertensive heart failure. This review will provide an overview of key mechanisms underlying hypertensive heart failure and will present the latest developments in PET probes for detection of cardiovascular inflammation, fibrosis and angiogenesis. Currently, selective PET probes for detection of angiogenesis remain elusive but promising PET probes for specific targeting of inflammation and fibrosis are rapidly progressing into clinical use.
Collapse
Affiliation(s)
- Viktoria Balogh
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Imaging, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Mark G. MacAskill
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Imaging, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Patrick W. F. Hadoke
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Gillian A. Gray
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Adriana A. S. Tavares
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Imaging, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
40
|
Lin Y, Shen Y, Chen J, Hu C, Zhou Z, Yuan C. The Function of LncRNA FTX in Several Common Cancers. Curr Pharm Des 2021; 27:2381-2386. [PMID: 33121404 DOI: 10.2174/1381612826666201029164036] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 09/22/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND LncRNA is a kind of non-coding RNA and its research is more popular in recent years, which has more than 200 nucleotides. It plays a significant part in various biological functions, including chromosome modification, genome modification, transcriptional activation, transcriptional interference, and other processes. FTX, at the center of the X chromosome inactivation and it has been shown that lncRNA FTX regulates cancer cells' development, migration, and invasion in many studies. METHODS Relevant literature was collected through the PubMed system search and is summarized in this article. RESULTS LncRNA FTX abnormally increased in tumor cells, such as liver cancer, stomach cancer, leukemia, renal cell carcinoma, colorectal cancer, glioma, osteosarcoma, etc. However, the expression level decreased in temporal lobe epilepsy, liver cirrhosis, heart failure, etc. Conclusion: FTX may be an important regulatory factor and a potential therapeutic target in cancers.
Collapse
Affiliation(s)
- Yexiang Lin
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| | - Yujie Shen
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| | - Jinlan Chen
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| | - Chengyu Hu
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| | - Zixian Zhou
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| | - Chengfu Yuan
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| |
Collapse
|
41
|
Wu X, Iroegbu CD, Peng J, Guo J, Yang J, Fan C. Cell Death and Exosomes Regulation After Myocardial Infarction and Ischemia-Reperfusion. Front Cell Dev Biol 2021; 9:673677. [PMID: 34179002 PMCID: PMC8220218 DOI: 10.3389/fcell.2021.673677] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 05/18/2021] [Indexed: 12/21/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death in the global population, accounting for about one-third of all deaths each year. Notably, with CVDs, myocardial damages result from myocardial infarction (MI) or cardiac arrhythmias caused by interrupted blood flow. Significantly, in the process of MI or myocardial ischemic-reperfusion (I/R) injury, both regulated and non-regulated cell death methods are involved. The critical factor for patients’ prognosis is the infarct area’s size, which determines the myocardial cells’ survival. Cell therapy for MI has been a research hotspot in recent years; however, exosomes secreted by cells have attracted much attention following shortcomings concerning immunogens. Exosomes are extracellular vesicles containing several biologically active substances such as lipids, nucleic acids, and proteins. New evidence suggests that exosomes play a crucial role in regulating cell death after MI as exosomes of various stem cells can participate in the cell damage process after MI. Hence, in the review herein, we focused on introducing various cell-derived exosomes to reduce cell death after MI by regulating the cell death pathway to understand myocardial repair mechanisms better and provide a reference for clinical treatment.
Collapse
Affiliation(s)
- Xun Wu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chukwuemeka Daniel Iroegbu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jun Peng
- Hunan Provincial Key Laboratory of Cardiovascular Research, Changsha, China
| | - Jianjun Guo
- Hunan Fangsheng Pharmaceutical Co., Ltd., Changsha, China
| | - Jinfu Yang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chengming Fan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Provincial Key Laboratory of Cardiovascular Research, Changsha, China.,Hunan Fangsheng Pharmaceutical Co., Ltd., Changsha, China
| |
Collapse
|
42
|
Zhou NQ, Fang ZX, Huang N, Zuo Y, Qiu Y, Guo LJ, Song P, Xu J, Wan GR, Tian XQ, Yin YL, Li P. aFGF Targeted Mediated by Novel Nanoparticles-Microbubble Complex Combined With Ultrasound-Targeted Microbubble Destruction attenuates Doxorubicin-Induced Heart Failure via Anti-Apoptosis and Promoting Cardiac Angiogenesis. Front Pharmacol 2021; 12:607785. [PMID: 33986662 PMCID: PMC8111001 DOI: 10.3389/fphar.2021.607785] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 02/15/2021] [Indexed: 11/30/2022] Open
Abstract
The purpose of this study was to evaluate the protective effect of acidic fibroblast growth factor targeted mediated by novel nanoparticles–cationic lipid microbubbles complex (aFGF–NP + CPMBs) combined with ultrasound targeted microbubble destruction (UTMD)on doxorubicin–induced heart failure (HF)and its mechanism. Heart failure rats induced by intraperitoneal injection with doxorubicin (DOX) to achieve cummulative dose of 15mg/kg for continuous 6 weeks showed left ventricular dysfunction, seriously oxidative stress, cardiomyocyte apoptosis, and decrease of myocardial vascular density. In contrast, aFGF–NP + CPMBs combined with UTMD therapy (3ug/kg, caudal vein injection, twice a week, 6weeks)prominently ameliorated left ventricular dysfunction by increased ejection fraction (EF) and fractional shortening (FS), decreased brain natriuretic peptide (BNP); strengthened the ability of antioxidant stress confirmed by increasing the activity of SOD and reducing the production of MDA; exerted the effect of anti–cardiomyocyte apoptosis and promotion angiogenesis by inhibited Bax expression and increased Bcl–2 expression and platelet endothelial cell adhesion molecule (CD31) expression. Taken together, the research suggested that aFGF targeted mediated by novel nanoparticles–cationic lipid microbubbles complex combined with UTMD should be a promising targeted treatment for heart failure.
Collapse
Affiliation(s)
- Nan-Qian Zhou
- Department of Ultrasonography, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China.,Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,.Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Zhi-Xin Fang
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,.Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.,College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Ning Huang
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,.Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.,College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Yue Zuo
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Yue Qiu
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,.Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.,College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Li-Juan Guo
- Department of Oncology, First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Ping Song
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,.Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.,College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Jian Xu
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,.Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.,College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Guang-Rui Wan
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,.Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.,College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Xin-Qiao Tian
- Department of Ultrasonography, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Ya-Ling Yin
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Peng Li
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,.Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.,College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
43
|
Identification of feature autophagy-related genes in patients with acute myocardial infarction based on bioinformatics analyses. Biosci Rep 2021; 40:225582. [PMID: 32597946 PMCID: PMC7350888 DOI: 10.1042/bsr20200790] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 06/18/2020] [Accepted: 06/29/2020] [Indexed: 01/15/2023] Open
Abstract
Objective: To identify feature autophagy-related genes (ARGs) in patients with acute myocardial infarction (AMI) and further investigate their value in the diagnosis of AMI. Methods: Gene microarray expression data of AMI peripheral blood samples were downloaded from the GSE66360 dataset. The data were randomly classified into a discovery cohort (21 AMI patients and 22 healthy controls) and a validation cohort (28 AMI patients and 28 healthy controls). Differentially expressed ARGs between patients with AMI and healthy controls in the discovery cohort were identified using a statistical software package. Feature ARGs were screened based on support vector machine-recursive feature elimination (SVM-RFE), and an SVM classifier was constructed. Receiver operating characteristic (ROC) analysis was used to investigate the predictive value of the classifier, which was further verified in an independent external cohort. Results: A total of seven genes were identified based on SVM-RFE. The SVM classifier had an excellent discrimination ability in both the discovery cohort (area under the curve [AUC] = 0.968) and the validation cohort (AUC = 0.992), which was further confirmed in the GSE48060 dataset (AUC = 0.963). Furthermore, the SVM classifier showed outstanding discrimination between AMI patients with and without recurrent events in the independent external cohort (AUC = 0.992). The identified genes are mainly involved in the cellular response to autophagy, macroautophagy, apoptosis, and the FoxO signaling pathway. Conclusion: Our study identified feature ARGs and indicated their potential roles in AMI diagnosis to improve our understanding of the molecular mechanism underlying the occurrence of AMI.
Collapse
|
44
|
Piamsiri C, Maneechote C, Siri-Angkul N, Chattipakorn SC, Chattipakorn N. Targeting necroptosis as therapeutic potential in chronic myocardial infarction. J Biomed Sci 2021; 28:25. [PMID: 33836761 PMCID: PMC8034148 DOI: 10.1186/s12929-021-00722-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 03/29/2021] [Indexed: 12/20/2022] Open
Abstract
Cardiovascular diseases (CVDs) are considered the predominant cause of morbidity and mortality globally. Of these, myocardial infarction (MI) is the most common cause of CVD mortality. MI is a life-threatening condition which occurs when coronary perfusion is interrupted leading to cardiomyocyte death. Subsequent to MI, consequences include adverse cardiac remodeling and cardiac dysfunction mainly contribute to the development of heart failure (HF). It has been shown that loss of functional cardiomyocytes in MI-induced HF are associated with several cell death pathways, in particular necroptosis. Although the entire mechanism underlying necroptosis in MI progression is still not widely recognized, some recent studies have reported beneficial effects of necroptosis inhibitors on cell viability and cardiac function in chronic MI models. Therefore, extensive investigation into the necroptosis signaling pathway is indicated for further study. This article comprehensively reviews the context of the underlying mechanisms of necroptosis in chronic MI-induced HF in in vitro, in vivo and clinical studies. These findings could inform ways of developing novel therapeutic strategies to improve the clinical outcomes in MI patients from this point forward.
Collapse
Affiliation(s)
- Chanon Piamsiri
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Chayodom Maneechote
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Natthaphat Siri-Angkul
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand. .,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand. .,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
45
|
Zhang X, Chen Z, Zang J, Yao C, Shi J, Nie R, Wu G. LncRNA-mRNA co-expression analysis discovered the diagnostic and prognostic biomarkers and potential therapeutic agents for myocardial infarction. Aging (Albany NY) 2021; 13:8944-8959. [PMID: 33668039 PMCID: PMC8034908 DOI: 10.18632/aging.202713] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 02/09/2021] [Indexed: 12/14/2022]
Abstract
Currently, the role of lncRNA in myocardial infarction (MI) is poorly understood. 17 co-expression modules were determined, specifically, the greenyellow, saddlebrown, grey60, royalblue, lightgreen, white, and pink modules were specifically expressed in the acute phase of MI, and brown, darkred, and royalblue, while greenyellow modules were specifically expressed in MI compared with CAD. 12 time-dependent of lncRNA/mRNA clusters with consistent expression trends were also identified. MI-associated modules were mainly enriched to immune, cell cycle, and metabolic pathways. We further obtained a network of 1816 lncRNA-mRNAs with higher expression correlations among these lncRNAs by analyzing the topological properties of the network. Herein, lncRNA RP11-847H18.2 and KLHL28, SPRTN, and EPM2AIP1 were determined as gene markers specifically expressed in MI, and they demonstrated a high predictive performance for MI diagnosis and prognosis. Three drugs, namely, Calcium citrate, Calcium Phosphate, and Calcium phosphate dihydrate, were identified as potential precursors of MI. Finally, gene and lncRNA diagnostic models were developed based on these genes and lncRNAs, with their AUCs averaged above 0.89 in both training and validation datasets. The findings of this study improve the diagnosis and prognosis of MI and personalized treatment of MI.
Collapse
Affiliation(s)
- Xiaocong Zhang
- Department of Cardiology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, China.,Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Ziqi Chen
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Jiabin Zang
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Chun Yao
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Jian Shi
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Ruqiong Nie
- Department of Cardiology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Guifu Wu
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China.,Guangdong Innovative Engineering and Technology Research Center for Assisted Circulation, Shenzhen, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
46
|
Bai YT, Xiao FJ, Wang H, Ge RL, Wang LS. Hypoxia protects H9c2 cells against Ferroptosis through SENP1-mediated protein DeSUMOylation. Int J Med Sci 2021; 18:1618-1627. [PMID: 33746578 PMCID: PMC7976564 DOI: 10.7150/ijms.50804] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 01/04/2021] [Indexed: 01/19/2023] Open
Abstract
Hypoxia affects proliferation, differentiation, as well as death of cardiomyocyte, and plays an important role in the development of myocardial ischemia. However, the detailed mechanisms through which hypoxia regulates cardiomyocyte ferroptosis have not been explored. In this study, we revealed that hypoxia suppresses the proliferation, migration, and erastin-induced ferroptosis of H9c2 cells. First, we confirmed the upregulation of SENP1 in H9c2 cells cultured under hypoxic conditions. Through adenovirus-mediated SENP1 gene transfection, we demonstrated that SENP1 overexpression could enhance H9c2 cell proliferation and migration while also protecting H9c2 cells from erastin-induced ferroptosis. Furthermore, through immunoprecipitation and western blotting, we confirmed that SENP1 mediated deSUMOylation of HIF-1α and ACSL4 in H9c2 cells. In conclusion, this study describes the underlying mechanism through which hypoxia upregulates SENP1 expression, in turn protecting against ferroptosis via the regulation of HIF-1α and ACSL4 deSUMOylation. Our findings provide a theoretical foundation for the development of novel therapeutics for ischemic heart diseases.
Collapse
Affiliation(s)
- Yu-Ting Bai
- Qinghai Provincial People's Hospital, Xining, 810001, PR China.,Research Center for High Altitude Medicine, Qinghai University, Xining, 810001, PR China
| | - Feng-Jun Xiao
- Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
| | - Hua Wang
- Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
| | - Ri-Li Ge
- Research Center for High Altitude Medicine, Qinghai University, Xining, 810001, PR China
| | - Li-Sheng Wang
- Beijing Institute of Radiation Medicine, Beijing, 100850, PR China.,Department of Molecular Diagnosis and Regenerative Medicine, Medical Research Center, the Affiliate Hospital of Qingdao University, Qingdao 266000, PR. China
| |
Collapse
|
47
|
Lichý M, Szobi A, Hrdlička J, Neckář J, Kolář F, Adameová A. Programmed Cell Death in the Left and Right Ventricle of the Late Phase of Post-Infarction Heart Failure. Int J Mol Sci 2020; 21:E7782. [PMID: 33096720 PMCID: PMC7589581 DOI: 10.3390/ijms21207782] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 09/30/2020] [Accepted: 10/19/2020] [Indexed: 01/03/2023] Open
Abstract
While necroptosis has been shown to contribute to the pathogenesis of post-infarction heart failure (HF), the role of autophagy remains unclear. Likewise, linkage between these two cell death modalities has not been sufficiently investigated. HF was induced by 60-min left coronary occlusion in adult Wistar rats and heart function was assessed 6 weeks later followed by immunoblotting analysis of necroptotic and autophagic proteins in both the left (LV) and right ventricle (RV). HF had no effect on RIP1 and RIP3 expression. PhosphoSer229-RIP3, acting as a pro-necroptotic signal, was increased in LV while deceased in RV of failing hearts. Total MLKL was elevated in RV only. Decrease in pSer555-ULK1, increase in pSer473-Akt and no significant elevation in beclin-1 and LC3-II/I ratio indicated rather a lowered rate of autophagy in LV. No beclin-1 upregulation and decreased LC3 processing also suggested the inhibition of both autophagosome formation and maturation in RV of failing hearts. In contrast, p89 PARP1 fragment, a marker of executed apoptosis, was increased in RV only. This is the first study showing a different signaling in ventricles of the late phase of post-infarction HF, highlighting necroptosis itself rather than its linkage with autophagy in LV, and apoptosis in RV.
Collapse
Affiliation(s)
- Martin Lichý
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Kalinčiakova 8, 83232 Bratislava, Slovakia; (M.L.); (A.S.)
| | - Adrián Szobi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Kalinčiakova 8, 83232 Bratislava, Slovakia; (M.L.); (A.S.)
| | - Jaroslav Hrdlička
- Institute of Physiology of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic; (J.H.); (J.N.); (F.K.)
| | - Jan Neckář
- Institute of Physiology of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic; (J.H.); (J.N.); (F.K.)
| | - František Kolář
- Institute of Physiology of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic; (J.H.); (J.N.); (F.K.)
| | - Adriana Adameová
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Kalinčiakova 8, 83232 Bratislava, Slovakia; (M.L.); (A.S.)
| |
Collapse
|
48
|
Kuang Y, Zhang Y, Xiao Z, Xu L, Wang P, Ma Q. Protective effect of dimethyl fumarate on oxidative damage and signaling in cardiomyocytes. Mol Med Rep 2020; 22:2783-2790. [PMID: 32945364 PMCID: PMC7453509 DOI: 10.3892/mmr.2020.11342] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 06/26/2020] [Indexed: 12/14/2022] Open
Abstract
Myocardial ischemia/reperfusion (I/R) injury contributes to the pathogenesis of numerous diseases. Based on its antioxidant and anti‑inflammatory effects, dimethyl fumarate (DMF) has been reported to exert protective effects against I/R. However, to the best of our knowledge, its potential role as a myocardial protective agent in heart disease has received little attention. Previous studies have suggested that DMF may exert its protective effects by activating nuclear factor erythroid 2‑related factor 2 (Nrf2); however, the exact underlying mechanisms remain to be elucidated. The aim of the present study was to investigate the protective role of DMF in myocardial I/R injury, and to determine the role of Nrf2 in mediating the activity of DMF. H9c2 cells were incubated with DMF (20 µM) for 24 h before establishing the I/R model, and were then subjected to myocardial ischemia for 6 h, followed by reperfusion. Cell viability, lactate dehydrogenase levels, anti‑oxidant enzyme expression levels and anti‑apoptotic effects were evaluated, and AKT/Nrf2 pathway‑associated mechanisms were investigated. The results of the present study indicated that DMF may reduce myocardial I/R injury in a Nrf2‑dependent manner. DMF significantly improved cellular viability, suppressed the expression of apoptotic markers, decreased the production of reactive oxygen species and increased the expression of Nrf2‑regulated antioxidative genes. Notably, these beneficial DMF‑mediated effects were not observed in the control or I/R groups. In conclusion, the results of the present study suggested that DMF may exert protective effects against a myocardial I/R model, and further validated Nrf2 modulation as a primary mode of action. Thus suggesting that DMF may be a potential therapeutic agent for AKT/Nrf2 pathway activation in myocardial, and potentially systemic, diseases.
Collapse
Affiliation(s)
- Yuanyuan Kuang
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yinzhuang Zhang
- Department of Cardiovascular Medicine, The First Hospital of Changsha, Changsha, Hunan 410005, P.R. China
| | - Zhen Xiao
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Lijun Xu
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Ping Wang
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Qilin Ma
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
49
|
Sun F, Du J, Li H, Hao S, Zhao G, Lu F. FABP4 inhibitor BMS309403 protects against hypoxia-induced H9c2 cardiomyocyte apoptosis through attenuating endoplasmic reticulum stress. J Cell Mol Med 2020; 24:11188-11197. [PMID: 32896039 PMCID: PMC7576298 DOI: 10.1111/jcmm.15666] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/11/2020] [Accepted: 07/03/2020] [Indexed: 12/22/2022] Open
Abstract
Acute myocardial infarction is characterized by ischaemia-induced cardiomyocyte apoptosis, in which the endoplasmic reticulum (ER) stress plays an important role. The fatty acid-binding protein-4 (FABP4) has been implicated in regulating ER stress and apoptosis. Yet, whether FABP4 is involved in modulating cardiomyocyte apoptosis remains unclarified. By applying an in vitro model of hypoxia-induced apoptosis of H9c2 cardiomyocytes, we found that FABP4 expression was elevated upon hypoxia stimulation, which was further demonstrated to be transcriptionally activated by the hypoxia-inducible factor 1a (HIF-1α). In addition, the pharmacological inhibition of FABP4 with BMS309403 protected against hypoxia-induced apoptosis in cardiomyocytes, indicating that FABP4 induction is detrimental for cardiomyocyte survival under hypoxic condition. Moreover, BMS309403 attenuated ER stress in cardiomyocytes exposed to hypoxia, which, however, was reversed by tunicamycin, an ER stress activator. More importantly, the protective effect of BMS309403 on cardiomyocytes vanished in the presence of tunicamycin. Thus, these observations establish that FABP4 inhibitor BMS309403 reduces hypoxia-induced cardiomyocyte apoptosis through attenuating excessive ER stress, implying that FABP4 inhibition may be of clinical benefit for MI treatment.
Collapse
Affiliation(s)
- Fuqiang Sun
- Department of Cardiovascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jiangchuan Du
- Department of UltrasoundThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Hongbin Li
- Department of Critical Care MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Shuang Hao
- Department of Cardiovascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Guochang Zhao
- Department of Cardiovascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Fanfan Lu
- Department of Cardiovascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
50
|
Ji Y, Yao J, He Y. Extracellular ubiquitin protects cardiomyocytes during ischemia/hypoxia by inhibiting mitochondrial apoptosis pathway through CXCR4. Biomed Pharmacother 2020; 131:110787. [PMID: 33152945 DOI: 10.1016/j.biopha.2020.110787] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/13/2020] [Accepted: 09/18/2020] [Indexed: 10/23/2022] Open
Abstract
AIM Acute myocardial infarction (AMI) is one of the deadliest diseases worldwide. The search for countermeasures to reduce cardiomyocytes death in the infarcted area has always been the focus of research. Ubiquitin (UB) is a small polypeptide mainly involved in proteasome-mediated protein degradation in cells, whereas extracellular UB in body fluids can also function through its receptor CXC chemokine receptor type 4 (CXCR4). This study aimed to explore the functional roles of extracellular UB in cardiomyocytes during ischemia/hypoxia (I/H). METHODS H9C2 cells were subjected to I/H treatment and cell injury was evaluated by cell viability, morphology changes and apoptosis rate. UB expression and levels of ubiquitinated proteins after I/H injury were measured. The effects of extracellular UB on I/H-induced cardiomyocytes apoptosis and the possible underlying mechanisms were studied. RESULTS I/H injury induced the decrease of cell viability as well as enhanced impaired cell morphology and apoptosis rate in H9C2 cells. Levels of UB mRNA and ubiquitinated proteins were significantly up-regulated after I/H treatment, whereas the concentration of extracellular UB in the conditioned media did not show significant change and the intracellular mono-UB levels in cells were down-regulated. Extracellular UB treatment protected cardiomyocytes from I/H injury by inhibiting the overactivation of mitochondria-dependent apoptosis pathway and up-regulating autophagy level. Inhibition of CXCR4 receptor using AMD3100 abolished cardioprotective effects of extracellular UB. CONCLUSION The up-regulation of UB was suggested to be an adaptive response to resist I/H-induced cardiomyocytes apoptosis, and additional extracellular UB treatment might serve as a new potential therapeutic drug for AMI.
Collapse
Affiliation(s)
- Yiqun Ji
- MOE Engineering Center of Hematological Disease, MOH Key Lab of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Jialu Yao
- Department of Cardiology, Suzhou Municipal Hospital, Suzhou, Jiangsu Province, China
| | - Yang He
- MOE Engineering Center of Hematological Disease, MOH Key Lab of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China.
| |
Collapse
|