1
|
Fu J, Su C, Ge Y, Ao Z, Xia L, Chen Y, Yang Y, Chen S, Xu R, Yang X, Huang K, Fu Q. PDE4D inhibition ameliorates cardiac hypertrophy and heart failure by activating mitophagy. Redox Biol 2025; 81:103563. [PMID: 40015131 PMCID: PMC11909752 DOI: 10.1016/j.redox.2025.103563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/13/2025] [Accepted: 02/21/2025] [Indexed: 03/01/2025] Open
Abstract
Cyclic adenosine monophosphate (cAMP) plays a major role in normal and pathologic signaling in the heart. Phosphodiesterase 4 (PDE4) is a major PDE degrading cAMP in the heart. There are inconsistencies concerning the roles of the PDE4 isoforms 4B and 4D in regulation of cardiac function. Cardiac PDE4B overexpression is beneficial in remodeling and heart failure (HF), however, the effect of PDE4D and PDE4 inhibitor in HF remains unclear. We generated global and conditional cardiac-specific heterozygous PDE4D knockout mice and adeno-associated virus serotype 9-PDE4D overexpression to determine the role of PDE4D in cardiac hypertrophy and HF. PDE4D upregulation was observed in failing hearts from human and isoproterenol injection and TAC mice. In vitro, isoproterenol stimulation increased PDE4D expression via PKA but had no effect on PDE4B expression in cardiomyocytes. PDE4D overexpression per se induced oxidative stress, mitochondrial damage and cardiomyocyte hypertrophy by decreasing PINK1/Parkin-mediated mitophagy through inhibiting cAMP-PKA-CREB-Sirtuin1 (SIRT1) signaling pathway, while PDE4B overexpression did not affect CREB-SIRT1 pathway and mitophagy but exhibited a protective effect on isoproterenol-induced oxidative stress and hypertrophy in cardiomyocytes. PDE4D silencing or inhibition with PDE4 inhibitor roflumilast ameliorated isoproterenol-induced mitochondrial injury and cardiomyocyte hypertrophy. In vivo, ISO injection or TAC inhibited cardiac mitophagy and caused cardiac hypertrophy and HF, which were ameliorated by roflumilast or cardiac-specific PDE4D haploinsufficiency. Conversely, cardiac PDE4D overexpression suppressed cardiac mitophagy and abolished the protective effects of global PDE4D haploinsufficiency on TAC-induced cardiac hypertrophy and HF. In conclusion, these studies elucidate a novel mechanism by which sustained adrenergic stimulation contributes to cardiac hypertrophy and HF by increasing PDE4D via cAMP-PKA signaling, which in turn reduces cAMP-PKA activity, resulting in cardiomyocyte hypertrophy and mitochondrial injury via inhibition of CREB-SIRT1 signaling-mediated mitophagy. PDE4D inhibition may represent a novel therapeutic strategy for HF.
Collapse
Affiliation(s)
- Jing Fu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Pharmacy, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Congping Su
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yin Ge
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhou Ao
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Xia
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingxiang Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yizheng Yang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shiwei Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Xu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyan Yang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Kai Huang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qin Fu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China.
| |
Collapse
|
2
|
Wu H, Diao H, Zhang F, Jiang W, Pan T, Bian Y. Organelle interplay in cardiovascular diseases: Mechanisms, pathogenesis, and therapeutic perspectives. Biomed Pharmacother 2025; 185:117978. [PMID: 40073746 DOI: 10.1016/j.biopha.2025.117978] [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: 10/31/2024] [Revised: 02/16/2025] [Accepted: 03/07/2025] [Indexed: 03/14/2025] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of rising morbidity and mortality among humans worldwide; however, our approach to the pathogenesis, exploration, and management of CVDs still remains limited. As the heart consists of cardiomyocytes, cardiac fibroblasts, endothelial cells, smooth muscle cells, and several types of cells, different types of dysfunction in the interplay between organelles play an important damaging role, resulting in cardiac pathologies. The interplay between cellular organelles is intricate and vital for maintaining cellular homeostasis, as highlighted by multiple diseases linked to the dysfunction of both organelles. Many studies have revealed the potential mechanisms by which organelles communicate with each other and regulate the pathological processes of CVDs together. However, gaps remain in fully understanding the complexity of these interactions and translating these insights into therapeutic approaches. In this review, we summarized how the interplay between cellular organelles in the cardiomyocytes alters in various heart diseases. We find underexplored areas, such as the crucial signaling pathways governing organelle communication, and discuss their implications for disease future progression. Furthermore, we evaluate emerging potential medicines aimed at restoring organelle interactions. Finally, we propose future directions for researching to advance the development of novel medicines and therapies, addressing current gaps and providing a theoretical basis for improved clinical outcomes in CVDs.
Collapse
Affiliation(s)
- Han Wu
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Hongtao Diao
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Feng Zhang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Weitao Jiang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Tengfei Pan
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yu Bian
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China.
| |
Collapse
|
3
|
Liu Z, Wang Y, Li L, Liu L, Li Y, Li Z, Xie Y, Yu F. SNAI2, a potential crossing point between cancer and cardiovascular disease. FASEB J 2025; 39:e70459. [PMID: 40059450 DOI: 10.1096/fj.202500198r] [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: 01/20/2025] [Revised: 02/18/2025] [Accepted: 03/03/2025] [Indexed: 05/13/2025]
Abstract
Cancer and cardiovascular disease remain the leading causes of morbidity and mortality worldwide, and the two separate disease entities share several similarities and possible interactions. Patients with cancer may have underlying cardiovascular disease, which is often exacerbated by the stress of tumor growth or treatment. At the same time, cardiotoxicity induced by anti-cancer therapies or the malignant process itself can lead to new cardiovascular diseases. Efforts have been made to find a rational explanation for this phenomenon. As a classical tumor-promoting factor, we notice that SNAI2 simultaneously plays an important pathogenic role in cardiovascular diseases. Moreover, there are several striking parallels in the mechanisms of cancer and CVD, such as shared risk factors (e.g., smoking and diabetes), cellular phenotypic switching, and metabolic remodeling, all of which are mediated by SNAI2. This review aims to summarize SNAI2's role in the core mechanisms linking cancer and CVD, as well as explore therapeutic approaches targeting SNAI2 and also seeks to provide insights into the common mechanisms underlying both cancer and CVD.
Collapse
Affiliation(s)
- Zihao Liu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yingzi Wang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Lei Li
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Linlu Liu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yuhao Li
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Zhixin Li
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yucheng Xie
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Fengxu Yu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Cardiovascular Remodeling and Dysfunction Key Laboratory of Luzhou, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| |
Collapse
|
4
|
Liu H, Wang H, Lin X, Xu M, Lan W, Wang J. Harnessing natural saponins: Advancements in mitochondrial dysfunction and therapeutic applications. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156383. [PMID: 39848019 DOI: 10.1016/j.phymed.2025.156383] [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: 11/06/2024] [Revised: 12/26/2024] [Accepted: 01/09/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND Mitochondrial dysfunction plays a crucial role in the development of a variety of diseases, notably neurodegenerative disorders, cardiovascular diseases, metabolic syndrome, and cancer. Natural saponins, which are intricate glycosides characterized by steroidal or triterpenoid structures, have attracted interest due to their diverse pharmacological benefits, including anti-inflammatory, antiviral, and anti-aging effects. PURPOSE This review synthesizes recent advancements in understanding mitochondrial dysfunction and explores how saponins can modulate mitochondrial function. It focuses on their potential applications in neuroprotection, cardiovascular health, and oncology. STUDY DESIGN The review incorporates a comprehensive literature analysis, highlighting the interplay between saponins and mitochondrial signaling pathways. Specific attention is given to the effects of saponins like ginsenoside Rg2 and 20(S)-protopanaxatriol on mitophagy and their neuroprotective, anti-aging, and synergistic therapeutic effects when combined. METHODS We conducted a comprehensive review of current research and clinical trials using PubMed, Google Scholar, and SciFinder databases. The search focused on saponins' role in mitochondrial function and their therapeutic effects, including "saponins", "mitochondria" and "mitochondrial function". The analysis primarily focused on articles published between 2011 and 2024. RESULTS The findings indicate that certain saponins can enhance mitophagy and modulate mitochondrial signaling pathways, showing promise in neuroprotection and anti-aging. Additionally, combinations of saponins have demonstrated synergistic effects in myocardial protection and cancer therapy, potentially improving therapeutic outcomes. CONCLUSION Although saponins exhibit significant potential in modulating mitochondrial functions and developing innovative therapeutic strategies, their clinical applications are constrained by low bioavailability. Rigorous clinical trials are essential to translate these findings into effective clinical therapies, ultimately improving patient outcomes through a deeper understanding of saponins' impact on mitochondrial function.
Collapse
Affiliation(s)
- Hongmei Liu
- Department of Pharmacy, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China; Department of pharmacy, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, Sichuan 610000, China
| | - Huan Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xinyu Lin
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Min Xu
- Department of Pharmacy, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China; Department of pharmacy, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, Sichuan 610000, China
| | - Wenying Lan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jinlian Wang
- Traditional Chinese Medicine Hospital of Meishan, Meishan 620010, China.
| |
Collapse
|
5
|
Li J, Li H, Niu N, Zhu Y, Hou S, Zhao W. NRF-1 promotes FUNDC1-mediated mitophagy as a protective mechanism against hypoxia-induced injury in cardiomyocytes. Exp Cell Res 2025; 446:114472. [PMID: 39978717 DOI: 10.1016/j.yexcr.2025.114472] [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: 09/30/2024] [Revised: 01/17/2025] [Accepted: 02/16/2025] [Indexed: 02/22/2025]
Abstract
Hypoxia-induced apoptosis and mitochondrial dysfunction in cardiomyocytes are involved in the mechanisms of heart failure. Our previous studies have confirmed that NRF-1 alleviates hypoxia-induced injury by promoting mitochondrial function and inhibiting apoptosis in cardiomyocytes. However, the mechanism by which NRF-1 attenuates hypoxia-induced injury in cardiomyocytes is still unclear. Mitophagy, a selective autophagy, has recently shown a remarkable correlation with hypoxia-induced cardiomyocyte injury. In this study, we evaluated whether NRF-1 protects cardiomyocytes from hypoxia-induced injury by regulating mitophagy. The findings indicate that hypoxia prevents H9c2 cells from growing, encourages mitochondrial dysfunction, and triggers mitophagy. In addition, promoting mitophagy by rapamycin reduces hypoxia-induced injury in H9c2 cells. Overexpression of NRF-1 in hypoxia-induced H9c2 cells promotes mitophagy and alleviates cell injury, and this effect can be inhibited by 3-MA. Further study found that NRF-1 promotes the expression of FUNDC1 by binding to its promoter region. Knockdown of FUNDC1 in NRF-1 over-expression H9c2 cells inhibited mitophagy and aggravated hypoxia-induced injury. In conclusion, our study suggests that NRF-1 protects against hypoxia-induced injury by regulating FUNDC1-mediated mitophagy in cardiomyocytes.
Collapse
Affiliation(s)
- Junliang Li
- School of Basic Medicine, Ningxia Medical University, Yinchuan, China.
| | - Hui Li
- Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases, Yinchuan, China.
| | - Nan Niu
- Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases, Yinchuan, China.
| | - Yazhou Zhu
- School of Basic Medicine, Ningxia Medical University, Yinchuan, China.
| | - Siyu Hou
- School of Basic Medicine, Ningxia Medical University, Yinchuan, China.
| | - Wei Zhao
- School of Basic Medicine, Ningxia Medical University, Yinchuan, China; Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases, Yinchuan, China.
| |
Collapse
|
6
|
Liu Z, Gan Y, Shen Z, Cai S, Wang X, Li Y, Li X, Fu H, Chen J, Li N. Role of copper homeostasis and cuproptosis in heart failure pathogenesis: implications for therapeutic strategies. Front Pharmacol 2025; 15:1527901. [PMID: 39850564 PMCID: PMC11754225 DOI: 10.3389/fphar.2024.1527901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 12/23/2024] [Indexed: 01/25/2025] Open
Abstract
Copper is an essential micronutrient involved in various physiological processes in various cell types. Consequently, dysregulation of copper homeostasis-either excessive or deficient-can lead to pathological changes, such as heart failure (HF). Recently, a new type of copper-dependent cell death known as cuproptosis has drawn increasing attention to the impact of copper dyshomeostasis on HF. Notably, copper dyshomeostasis was associated with the occurrence of HF. Hence, this review aimed to investigate the biological processes involved in copper uptake, transport, excretion, and storage at both the cellular and systemic levels in terms of cuproptosis and HF, along with the underlying mechanisms of action. Additionally, the role of cuproptosis and its related mitochondrial dysfunction in HF pathogenesis was analyzed. Finally, we reviewed the therapeutic potential of current drugs that target copper metabolism for treating HF. Overall, the conclusions of this review revealed the therapeutic potential of copper-based therapies that target cuproptosis for the development of strategies for the treatment of HF.
Collapse
Affiliation(s)
- Zhichao Liu
- School of Rehabilitation Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Yongkang Gan
- Department of Vascular Surgery, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
| | - Zhen Shen
- Department of Clinical Laboratory, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Siqi Cai
- College of Art, Nanjing University of Information Science and Technology, Nanjing, Jiangsu, China
| | - Xizhen Wang
- School of Rehabilitation Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Yong Li
- Experimental Center for Medical Research, Shandong Second Medical University, Weifang, Shandong, China
| | - Xiaofeng Li
- Department of Cardiovascular, Second Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Huanjie Fu
- Department of Cardiovascular, Second Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jinhong Chen
- School of Rehabilitation Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Ningcen Li
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
7
|
Hu D, Cui Y, Hou X, Wang X, Shen Z, Pang H, Ge Y, Ning H. Drp1-Dependent Mitochondrial Fission Contributes to Lactic Acid-Induced Chicken Cardiomyocyte Damage. J Biochem Mol Toxicol 2025; 39:e70128. [PMID: 39756064 DOI: 10.1002/jbt.70128] [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: 11/18/2024] [Revised: 12/18/2024] [Accepted: 12/25/2024] [Indexed: 01/07/2025]
Abstract
Enhanced glycolysis and elevated lactic acid (LA) production are observed during sudden death syndrome (SDS) in broilers. However, the mechanism underlying LA-induced cardiomyocyte damage and heart failure in fast-growing broilers remains unclear. In this study, chicken embryo cardiomyocytes (CECs) were cultured and treated with LA to investigate LA-induced CEC injury and its mechanism, aiming to develop strategies to prevent LA-induced SDS in broilers. Results showed that LA inhibited CEC proliferation and contraction whereas inducing apoptosis. Furthermore, LA disrupted mitochondrial ultrastructure, reduced mitochondrial membrane potential, activated mitophagy, and disturbed mitochondrial dynamics. Treatment with Mdivi-1, a selective Drp1 inhibitor, improved CEC viability, restored mitochondrial network integrity, reduced reactive oxygen species production, and inhibited LA-induced apoptosis. These findings suggest that LA-induced cardiomyocyte injury during SDS in broilers is associated with mitochondrial damage and increased mitochondrial fission. The inhibition of mitochondrial hyperfission by Mdivi-1 effectively preserves CEC morphology, structure, and function, playing a critical role in preventing LA-induced damage. This study provides a foundation for strategies to prevent and control SDS in broilers.
Collapse
Affiliation(s)
- Dongfang Hu
- College of Animal Science and Technology, Henan Institute of Science and Technology, Xinxiang, China
| | - Yunli Cui
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| | - Xueke Hou
- College of Animal Science and Technology, Henan Institute of Science and Technology, Xinxiang, China
| | - Xueying Wang
- College of Animal Science and Technology, Henan Institute of Science and Technology, Xinxiang, China
| | - Zihui Shen
- College of Animal Science and Technology, Henan Institute of Science and Technology, Xinxiang, China
| | - Huiqing Pang
- College of Animal Science and Technology, Henan Institute of Science and Technology, Xinxiang, China
| | - Yaming Ge
- College of Animal Science and Technology, Henan Institute of Science and Technology, Xinxiang, China
| | - Hongmei Ning
- College of Animal Science and Technology, Henan Institute of Science and Technology, Xinxiang, China
| |
Collapse
|
8
|
Sun Q, Karwi QG, Wong N, Lopaschuk GD. Advances in myocardial energy metabolism: metabolic remodelling in heart failure and beyond. Cardiovasc Res 2024; 120:1996-2016. [PMID: 39453987 PMCID: PMC11646102 DOI: 10.1093/cvr/cvae231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/28/2024] [Accepted: 07/03/2024] [Indexed: 10/27/2024] Open
Abstract
The very high energy demand of the heart is primarily met by adenosine triphosphate (ATP) production from mitochondrial oxidative phosphorylation, with glycolysis providing a smaller amount of ATP production. This ATP production is markedly altered in heart failure, primarily due to a decrease in mitochondrial oxidative metabolism. Although an increase in glycolytic ATP production partly compensates for the decrease in mitochondrial ATP production, the failing heart faces an energy deficit that contributes to the severity of contractile dysfunction. The relative contribution of the different fuels for mitochondrial ATP production dramatically changes in the failing heart, which depends to a large extent on the type of heart failure. A common metabolic defect in all forms of heart failure [including heart failure with reduced ejection fraction (HFrEF), heart failure with preserved EF (HFpEF), and diabetic cardiomyopathies] is a decrease in mitochondrial oxidation of pyruvate originating from glucose (i.e. glucose oxidation). This decrease in glucose oxidation occurs regardless of whether glycolysis is increased, resulting in an uncoupling of glycolysis from glucose oxidation that can decrease cardiac efficiency. The mitochondrial oxidation of fatty acids by the heart increases or decreases, depending on the type of heart failure. For instance, in HFpEF and diabetic cardiomyopathies myocardial fatty acid oxidation increases, while in HFrEF myocardial fatty acid oxidation either decreases or remains unchanged. The oxidation of ketones (which provides the failing heart with an important energy source) also differs depending on the type of heart failure, being increased in HFrEF, and decreased in HFpEF and diabetic cardiomyopathies. The alterations in mitochondrial oxidative metabolism and glycolysis in the failing heart are due to transcriptional changes in key enzymes involved in the metabolic pathways, as well as alterations in redox state, metabolic signalling and post-translational epigenetic changes in energy metabolic enzymes. Of importance, targeting the mitochondrial energy metabolic pathways has emerged as a novel therapeutic approach to improving cardiac function and cardiac efficiency in the failing heart.
Collapse
Affiliation(s)
- Qiuyu Sun
- Cardiovascular Research Center, University of Alberta, Edmonton, AB T6G 2S2, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Qutuba G Karwi
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, Saint John’s, NL A1B 3V6, Canada
| | - Nathan Wong
- Cardiovascular Research Center, University of Alberta, Edmonton, AB T6G 2S2, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Gary D Lopaschuk
- Cardiovascular Research Center, University of Alberta, Edmonton, AB T6G 2S2, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2S2, Canada
| |
Collapse
|
9
|
Bruszt K, Horvath O, Ordog K, Toth S, Juhasz K, Vamos E, Fekete K, Gallyas F, Toth K, Halmosi R, Deres L. Cardiac effects of OPA1 protein promotion in a transgenic animal model. PLoS One 2024; 19:e0310394. [PMID: 39570915 PMCID: PMC11581344 DOI: 10.1371/journal.pone.0310394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 08/30/2024] [Indexed: 11/24/2024] Open
Abstract
Mitochondria form a dynamic network in cells, regulated by the balance between mitochondrial fusion and fission. The inhibition of mitochondrial fission can have positive effects in acute ischemic/reperfusion injury models by preventing the fall in mitochondrial membrane potential associated with fission processes. However, inhibition of fission in chronic models is disadvantageous because it obstructs the elimination of damaged mitochondrial fragments. OPA1, in view of previous results, is a possible therapeutic target as a fusion promoter and structure stabilizer protein. We used transgenic mice in which the OMA1 cleavage sites of OPA1 were deleted. This resulted in a higher representation of L-OPA1 compared to S-OPA1. After genotyping and model validation, all animals were examined by echocardiograph on two occasions, at weeks 11 and 36. Histological samples were taken from hearts to examine mitochondrial morphology and structure remodeling. The signaling pathways related to mitochondrial dynamic processes were evaluated. Cardiomyocytes were isolated from neonatal mice to determine the efficiency of mitochondrial respiration using the SeaHorse assay method. OPA1 protein promotion has a negative effect on systolic function during aging. We confirmed that volume overload and ventricular remodeling did not manifest. The reason behind the loss of pump function might be, at least partly, due to the energy deficit caused by mitochondrial respiratory failure and damage in mitochondrial quality control pathways.
Collapse
Affiliation(s)
- Kitti Bruszt
- 1st Department of Medicine, University of Pecs Medical School, Pecs, Hungary
- Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Orsolya Horvath
- 1st Department of Medicine, University of Pecs Medical School, Pecs, Hungary
- Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Katalin Ordog
- 1st Department of Medicine, University of Pecs Medical School, Pecs, Hungary
- Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Szilard Toth
- 1st Department of Medicine, University of Pecs Medical School, Pecs, Hungary
- Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Kata Juhasz
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, Pecs, Hungary
| | - Eszter Vamos
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, Pecs, Hungary
| | - Katalin Fekete
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, Pecs, Hungary
| | - Ferenc Gallyas
- Szentagothai Research Centre, University of Pecs, Pecs, Hungary
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, Pecs, Hungary
| | - Kalman Toth
- 1st Department of Medicine, University of Pecs Medical School, Pecs, Hungary
- Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Robert Halmosi
- 1st Department of Medicine, University of Pecs Medical School, Pecs, Hungary
- Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Laszlo Deres
- 1st Department of Medicine, University of Pecs Medical School, Pecs, Hungary
- Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| |
Collapse
|
10
|
Zhou G, Wang X, Guo M, Qu C, Gao L, Yu J, Li Y, Luo S, Shi Q, Guo Y. Mitophagy deficiency activates stimulator of interferon genes activation and aggravates pathogenetic cardiac remodeling. Genes Dis 2024; 11:101074. [PMID: 39281830 PMCID: PMC11399633 DOI: 10.1016/j.gendis.2023.08.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/11/2023] [Accepted: 08/04/2023] [Indexed: 09/12/2024] Open
Abstract
Stimulator of interferon genes (STING) has recently been found to play a crucial role in cardiac sterile inflammation and dysfunction. The role of stimulator of interferon genes (STING) in cardiac sterile inflammation and dysfunction has been recently discovered. This study aims to examine the involvement of STING in pathological cardiac remodeling and the mechanisms that govern the activation of the STING pathway. To investigate this, transverse aortic constriction (TAC) was performed on STING knockout mice to induce pressure overload-induced cardiac remodeling. Subsequently, cardiac function, remodeling, and inflammation levels were evaluated. The STING pathway was found to be activated in the pressure overload-stressed heart and angiotensin II (Ang II)-stimulated cardiac fibroblasts. Loss of STING expression led to a significant reduction in inflammatory responses, mitochondrial fragmentation, and oxidative stress in the heart, resulting in attenuated cardiac remodeling and dysfunction. Furthermore, the exacerbation of pressure overload-induced STING-mediated inflammation and pathological cardiac remodeling was observed when mitophagy was suppressed through the silencing of Parkin, an E3 ubiquitin ligase. Taken together, these findings indicate that STING represents a newly identified and significant molecule implicated in the process of pathological cardiac remodeling and that mitophagy is an upstream mechanism that regulates STING activation. Targeting STING may therefore provide a novel therapeutic strategy for pathological cardiac remodeling and heart failure.
Collapse
Affiliation(s)
- Guoxiang Zhou
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xiaowen Wang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Mingyu Guo
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Can Qu
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Lei Gao
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jiang Yu
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yuanjing Li
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Suxin Luo
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Qiong Shi
- The Department of Laboratory Medicine, M.O.E. Key Laboratory of Laboratory Medical Diagnostics, Chongqing Medical University, Chongqing 400016, China
| | - Yongzheng Guo
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
11
|
Rahmani S, Roohbakhsh A, Pourbarkhordar V, Hayes AW, Karimi G. Melatonin regulates mitochondrial dynamics and mitophagy: Cardiovascular protection. J Cell Mol Med 2024; 28:e70074. [PMID: 39333694 PMCID: PMC11436317 DOI: 10.1111/jcmm.70074] [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: 02/26/2024] [Revised: 07/27/2024] [Accepted: 08/28/2024] [Indexed: 09/29/2024] Open
Abstract
Despite extensive progress in the knowledge and understanding of cardiovascular diseases and significant advances in pharmacological treatments and procedural interventions, cardiovascular diseases (CVD) remain the leading cause of death globally. Mitochondrial dynamics refers to the repetitive cycle of fission and fusion of the mitochondrial network. Fission and fusion balance regulate mitochondrial shape and influence physiology, quality and homeostasis. Mitophagy is a process that eliminates aberrant mitochondria. Melatonin (Mel) is a pineal-synthesized hormone with a range of pharmacological properties. Numerous nonclinical trials have demonstrated that Mel provides cardioprotection against ischemia/reperfusion, cardiomyopathies, atherosclerosis and cardiotoxicity. Recently, interest has grown in how mitochondrial dynamics contribute to melatonin cardioprotective effects. This review assesses the literature on the protective effects of Mel against CVD via the regulation of mitochondrial dynamics and mitophagy in both in-vivo and in-vitro studies. The signalling pathways underlying its cardioprotective effects were reviewed. Mel modulated mitochondrial dynamics and mitophagy proteins by upregulation of mitofusin, inhibition of DRP1 and regulation of mitophagy-related proteins. The evidence supports a significant role of Mel in mitochondrial dynamics and mitophagy quality control in CVD.
Collapse
Affiliation(s)
- Sohrab Rahmani
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Roohbakhsh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vahid Pourbarkhordar
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - A Wallace Hayes
- Center for Environmental Occupational Risk Analysis and Management, College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
12
|
Zhou X, Wang J, Yu L, Qiao G, Qin D, Yuen-Kwan Law B, Ren F, Wu J, Wu A. Mitophagy and cGAS-STING crosstalk in neuroinflammation. Acta Pharm Sin B 2024; 14:3327-3361. [PMID: 39220869 PMCID: PMC11365416 DOI: 10.1016/j.apsb.2024.05.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/10/2024] [Accepted: 04/12/2024] [Indexed: 09/04/2024] Open
Abstract
Mitophagy, essential for mitochondrial health, selectively degrades damaged mitochondria. It is intricately linked to the cGAS-STING pathway, which is crucial for innate immunity. This pathway responds to mitochondrial DNA and is associated with cellular stress response. Our review explores the molecular details and regulatory mechanisms of mitophagy and the cGAS-STING pathway. We critically evaluate the literature demonstrating how dysfunctional mitophagy leads to neuroinflammatory conditions, primarily through the accumulation of damaged mitochondria, which activates the cGAS-STING pathway. This activation prompts the production of pro-inflammatory cytokines, exacerbating neuroinflammation. This review emphasizes the interaction between mitophagy and the cGAS-STING pathways. Effective mitophagy may suppress the cGAS-STING pathway, offering protection against neuroinflammation. Conversely, impaired mitophagy may activate the cGAS-STING pathway, leading to chronic neuroinflammation. Additionally, we explored how this interaction influences neurodegenerative disorders, suggesting a common mechanism underlying these diseases. In conclusion, there is a need for additional targeted research to unravel the complexities of mitophagy-cGAS-STING interactions and their role in neurodegeneration. This review highlights potential therapies targeting these pathways, potentially leading to new treatments for neuroinflammatory and neurodegenerative conditions. This synthesis enhances our understanding of the cellular and molecular foundations of neuroinflammation and opens new therapeutic avenues for neurodegenerative disease research.
Collapse
Affiliation(s)
- Xiaogang Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Jing Wang
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Lu Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Gan Qiao
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Dalian Qin
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Betty Yuen-Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR 999078, China
| | - Fang Ren
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chongqing Traditional Chinese Medicine Hospital, Chongqing 400021, China
| | - Jianming Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Anguo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
13
|
Nag S, Mitra O, Maturi B, Kaur SP, Saini A, Nama M, Roy S, Samanta S, Chacko L, Dutta R, Sayana SB, Subramaniyan V, Bhatti JS, Kandimalla R. Autophagy and mitophagy as potential therapeutic targets in diabetic heart condition: Harnessing the power of nanotheranostics. Asian J Pharm Sci 2024; 19:100927. [PMID: 38948399 PMCID: PMC11214300 DOI: 10.1016/j.ajps.2024.100927] [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: 07/02/2023] [Revised: 03/29/2024] [Accepted: 04/13/2024] [Indexed: 07/02/2024] Open
Abstract
Autophagy and mitophagy pose unresolved challenges in understanding the pathology of diabetic heart condition (DHC), which encompasses a complex range of cardiovascular issues linked to diabetes and associated cardiomyopathies. Despite significant progress in reducing mortality rates from cardiovascular diseases (CVDs), heart failure remains a major cause of increased morbidity among diabetic patients. These cellular processes are essential for maintaining cellular balance and removing damaged or dysfunctional components, and their involvement in the development of diabetic heart disease makes them attractive targets for diagnosis and treatment. While a variety of conventional diagnostic and therapeutic strategies are available, DHC continues to present a significant challenge. Point-of-care diagnostics, supported by nanobiosensing techniques, offer a promising alternative for these complex scenarios. Although conventional medications have been widely used in DHC patients, they raise several concerns regarding various physiological aspects. Modern medicine places great emphasis on the application of nanotechnology to target autophagy and mitophagy in DHC, offering a promising approach to deliver drugs beyond the limitations of traditional therapies. This article aims to explore the potential connections between autophagy, mitophagy and DHC, while also discussing the promise of nanotechnology-based theranostic interventions that specifically target these molecular pathways.
Collapse
Affiliation(s)
- Sagnik Nag
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor, Malaysia
| | - Oishi Mitra
- Department of Bio-Sciences, School of Bio-Sciences & Technology (SBST), Vellore Institute of Technology (VIT), Tiruvalam Road, Vellore 632014, Tamil Nadu, India
| | - Bhanu Maturi
- Department of Internal Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Simran Preet Kaur
- Department of Microbiology, University of Delhi (South Campus), Benito Juarez Road, New Delhi 110021, India
| | - Ankita Saini
- Department of Microbiology, University of Delhi (South Campus), Benito Juarez Road, New Delhi 110021, India
| | - Muskan Nama
- Department of Bio-Sciences, School of Bio-Sciences & Technology (SBST), Vellore Institute of Technology (VIT), Tiruvalam Road, Vellore 632014, Tamil Nadu, India
| | - Soumik Roy
- Department of Biotechnology, Indian Institute of Technology, Hyderabad (IIT-H), Sangareddy, Telangana 502284, India
| | - Souvik Samanta
- Department of Bio-Sciences, School of Bio-Sciences & Technology (SBST), Vellore Institute of Technology (VIT), Tiruvalam Road, Vellore 632014, Tamil Nadu, India
| | - Leena Chacko
- BioAnalytical Lab, Meso Scale Discovery, 1601 Research Blvd, Rockville, MD, USA
| | - Rohan Dutta
- Department of Bio-Sciences, School of Bio-Sciences & Technology (SBST), Vellore Institute of Technology (VIT), Tiruvalam Road, Vellore 632014, Tamil Nadu, India
| | - Suresh Babu Sayana
- Department of Pharmacology, Government Medical College, Suryapet, Telangana, India
| | - Vetriselvan Subramaniyan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor, Malaysia
| | - Jasvinder Singh Bhatti
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, India
| | - Ramesh Kandimalla
- Department of Biochemistry, Kakatiya Medical College, Warangal 506007, India
| |
Collapse
|
14
|
Wang Y, Ren T, Li C, Wu Q, Liu J, Guan X, Chang X, Liu Z, Liu R. Mechanisms involved in the regulation of mitochondrial quality control by PGAM5 in heart failure. Cell Stress Chaperones 2024; 29:510-518. [PMID: 38821173 PMCID: PMC11214171 DOI: 10.1016/j.cstres.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/02/2024] Open
Abstract
Heart failure (HF) refers to a group of clinical syndromes in which various heart diseases lead to the inability of cardiac output to meet the metabolic needs of the body's tissues. Cardiac metabolism requires enormous amounts of energy; thus, impaired myocardial energy metabolism is considered a key factor in the occurrence and development of HF. Mitochondria serve as the primary energy source for cardiomyocytes, and their regular functionality underpins healthy cardiac function. The mitochondrial quality control system is a crucial mechanism for regulating the functionality of cardiomyocytes, and any abnormality in this system can potentially impact the morphology and structure of mitochondria, as well as the energy metabolism of cardiomyocytes. Phosphoglycerate mutase 5 (PGAM5), a multifunctional protein, plays a key role in the regulation of mitochondrial quality control through multiple pathways. Therefore, abnormal PGAM5 function is closely related to mitochondrial damage. This article reviews the mechanism of PGAM5's involvement in the regulation of the mitochondrial quality control system in the occurrence and development of HF, thereby providing a theoretical basis for future in-depth research.
Collapse
Affiliation(s)
- Yanli Wang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tiantian Ren
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Cuizhi Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qiaomin Wu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jinfeng Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xuanke Guan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xing Chang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Zhiming Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Ruxiu Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
15
|
Martin TG, Sherer LA, Kirk JA. BAG3 localizes to mitochondria in cardiac fibroblasts and regulates mitophagy. Am J Physiol Heart Circ Physiol 2024; 326:H1124-H1130. [PMID: 38488519 PMCID: PMC11381031 DOI: 10.1152/ajpheart.00736.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 03/01/2024] [Accepted: 03/08/2024] [Indexed: 04/14/2024]
Abstract
The co-chaperone Bcl2-associated athanogene 3 (BAG3) is a central node in protein quality control in the heart. In humans and animal models, decreased BAG3 expression is associated with cardiac dysfunction and dilated cardiomyopathy. Although previous studies focused on BAG3 in cardiomyocytes, cardiac fibroblasts are also critical drivers of pathologic remodeling. Yet, the role of BAG3 in cardiac fibroblasts is almost completely unexplored. Here, we show that BAG3 is expressed in primary rat neonatal cardiac fibroblasts and preferentially localizes to mitochondria. Knockdown of BAG3 reduces mitophagy and enhances fibroblast activation, which is associated with fibrotic remodeling. Heat shock protein 70 (Hsp70) is a critical binding partner for BAG3 and inhibiting this interaction in fibroblasts using the drug JG-98 decreased autophagy, decreased mitofusin-2 expression, and disrupted mitochondrial morphology. Together, these data indicate that BAG3 is expressed in cardiac fibroblasts, where it facilitates mitophagy and promotes fibroblast quiescence. This suggests that depressed BAG3 levels in heart failure may exacerbate fibrotic pathology, thus contributing to myocardial dysfunction through sarcomere-independent pathways.NEW & NOTEWORTHY We report BAG3's localization to mitochondria and its role in mitophagy for the first time in primary ventricular cardiac fibroblasts. We have also collected the first evidence showing that loss of BAG3 increases cardiac fibroblast activation into myofibroblasts, which are major drivers of cardiac fibrosis and pathological remodeling during heart disease.
Collapse
Affiliation(s)
- Thomas G Martin
- Department of Cell and Molecular Physiology, Loyola University Stritch School of Medicine, Maywood, Illinois, United States
| | - Laura A Sherer
- Department of Cell and Molecular Physiology, Loyola University Stritch School of Medicine, Maywood, Illinois, United States
| | - Jonathan A Kirk
- Department of Cell and Molecular Physiology, Loyola University Stritch School of Medicine, Maywood, Illinois, United States
| |
Collapse
|
16
|
Mone P, Agyapong ED, Morciano G, Jankauskas SS, De Luca A, Varzideh F, Pinton P, Santulli G. Dysfunctional mitochondria elicit bioenergetic decline in the aged heart. THE JOURNAL OF CARDIOVASCULAR AGING 2024; 4:13. [PMID: 39015481 PMCID: PMC11250775 DOI: 10.20517/jca.2023.50] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Aging represents a complex biological progression affecting the entire body, marked by a gradual decline in tissue function, rendering organs more susceptible to stress and diseases. The human heart holds significant importance in this context, as its aging process poses life-threatening risks. It entails macroscopic morphological shifts and biochemical changes that collectively contribute to diminished cardiac function. Among the numerous pivotal factors in aging, mitochondria play a critical role, intersecting with various molecular pathways and housing several aging-related agents. In this comprehensive review, we provide an updated overview of the functional role of mitochondria in cardiac aging.
Collapse
Affiliation(s)
- Pasquale Mone
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
- Department of Medicine and Health Sciences, University of Molise, Campobasso 86100, Italy
| | - Esther Densu Agyapong
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
- Department of Medical Sciences, University of Ferrara, Ferrara 44121, Italy
| | - Giampaolo Morciano
- Department of Medical Sciences, University of Ferrara, Ferrara 44121, Italy
- Maria Cecilia Hospital, GVM Care & Research, Cotignola 48033, Italy
| | - Stanislovas S. Jankauskas
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Antonio De Luca
- Department of Mental and Physical Health and Preventive Medicine, Vanvitelli University, Naples 80100, Italy
| | - Fahimeh Varzideh
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Paolo Pinton
- Department of Medical Sciences, University of Ferrara, Ferrara 44121, Italy
- Maria Cecilia Hospital, GVM Care & Research, Cotignola 48033, Italy
| | - Gaetano Santulli
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
- Department of Medicine and Health Sciences, University of Molise, Campobasso 86100, Italy
- Department of Advanced Biomedical Sciences, “Federico II” University, International Translational Research and Medical Education (ITME) Consortium, Academic Research Unit, Naples 80131, Italy
- Department of Molecular Pharmacology, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Neuroimmunology and Inflammation (INI), Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, New York, NY 10461, USA
| |
Collapse
|
17
|
He DW, Liu DZ, Luo XZ, Chen CB, Lu CH, Na N, Huang F. HMGB1-RAGE axis contributes to myocardial ischemia/reperfusion injury via regulation of cardiomyocyte autophagy and apoptosis in diabetic mice. Biol Chem 2024; 405:167-176. [PMID: 37768929 DOI: 10.1515/hsz-2023-0134] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 08/22/2023] [Indexed: 09/30/2023]
Abstract
Patients with acute myocardial infarction complicated with diabetes are more likely to develop myocardial ischemia/reperfusion (I/R) injury (MI/RI) during reperfusion therapy. Both HMGB1 and RAGE play important roles in MI/RI. However, the specific mechanisms of HMGB1 associated with RAGE are not fully clarified in diabetic MI/RI. This study aimed to investigate whether the HMGB1-RAGE axis induces diabetic MI/RI via regulating autophagy and apoptosis. A db/db mouse model of MI/RI was established, where anti-HMGB1 antibody and RAGE inhibitor (FPS-ZM1) were respectively injected after 10 min of reperfusion. The results showed that treatment with anti-HMGB1 significantly reduced the infarct size, serum LDH, and CK-MB level. Similar situations also occurred in mice administrated with FPS-ZM1, though the HMGB1 level was unchanged. Then, we found that treatment with anti-HMGB1 or FPS-ZM1 performed the same effects in suppressing the autophagy and apoptosis, as reflected by the results of lower LAMP2 and LC3B levels, increased Bcl-2 level, reduced BAX and caspase-3 levels. Moreover, the Pink1/Parkin levels were also inhibited at the same time. Collectively, this study indicates that the HMGB1-RAGE axis aggravated diabetic MI/RI via apoptosis and Pink1/Parkin mediated autophagy pathways, and inhibition of HMGB1 or RAGE contributes to alleviating those adverse situations.
Collapse
Affiliation(s)
- De-Wei He
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, No.6 Shuangyong Road, Nanning 530021, Guangxi, China
| | - De-Zhao Liu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, No.6 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Xiao-Zhi Luo
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, No.6 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Chuan-Bin Chen
- Department of Cardiology, The Second Affiliated Hospital of Hainan Medical University, 368 Yihai Avenue, Haikou 570216, Hainan, China
| | - Chuang-Hong Lu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, No.6 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Na Na
- Department of Chemistry, Scripps Research Institute, No.10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Feng Huang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, No.6 Shuangyong Road, Nanning 530021, Guangxi, China
| |
Collapse
|
18
|
Chen C, Wang J, Zhu X, Hu J, Liu C, Liu L. Energy metabolism and redox balance: How phytochemicals influence heart failure treatment. Biomed Pharmacother 2024; 171:116136. [PMID: 38215694 DOI: 10.1016/j.biopha.2024.116136] [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: 10/11/2023] [Revised: 12/31/2023] [Accepted: 01/04/2024] [Indexed: 01/14/2024] Open
Abstract
Heart Failure (HF) epitomizes a formidable global health quandary characterized by marked morbidity and mortality. It has been established that severe derangements in energy metabolism are central to the pathogenesis of HF, culminating in an inadequate cardiac energy milieu, which, in turn, precipitates cardiac pump dysfunction and systemic energy metabolic failure, thereby steering the trajectory and potential recuperation of HF. The conventional therapeutic paradigms for HF predominantly target amelioration of heart rate, and cardiac preload and afterload, proffering symptomatic palliation or decelerating the disease progression. However, the realm of therapeutics targeting the cardiac energy metabolism remains largely uncharted. This review delineates the quintessential characteristics of cardiac energy metabolism in healthy hearts, and the metabolic aberrations observed during HF, alongside the associated metabolic pathways and targets. Furthermore, we delve into the potential of phytochemicals in rectifying the redox disequilibrium and the perturbations in energy metabolism observed in HF. Through an exhaustive analysis of recent advancements, we underscore the promise of phytochemicals in modulating these pathways, thereby unfurling a novel vista on HF therapeutics. Given their potential in orchestrating cardiac energy metabolism, phytochemicals are emerging as a burgeoning frontier for HF treatment. The review accentuates the imperative for deeper exploration into how these phytochemicals specifically intervene in cardiac energy metabolism, and the subsequent translation of these findings into clinical applications, thereby broadening the horizon for HF treatment modalities.
Collapse
Affiliation(s)
- Cong Chen
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Jie Wang
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China.
| | - Xueying Zhu
- Department of Anatomy, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jun Hu
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Chao Liu
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Lanchun Liu
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| |
Collapse
|
19
|
Jamialahmadi T, Hasanpour M, Vakilian F, Penson PE, Iranshahy M, Sahebkar A. Evaluation of Urolithin A Efficacy in Heart Failure Patients with Reduced Ejection Fraction: A Randomized, Double-blind, Crossover, Placebo-controlled Clinical Trial. Rev Recent Clin Trials 2024; 19:221-228. [PMID: 38415449 DOI: 10.2174/0115748871279354240209101604] [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: 09/28/2023] [Revised: 01/16/2024] [Accepted: 01/23/2024] [Indexed: 02/29/2024]
Abstract
BACKGROUND Mitochondrial dysfunction and impaired mitophagy are integral to myocyte loss and the progression of heart failure. Urolithin A (UA), a microbiota-produced metabolite of ellagitannins and ellagic acid, is a known stimulator of mitophagy and mitochondrial biogenesis that has shown cardioprotective effects in experimental models. METHODS A randomized, double-blind, placebo-controlled 2×2 crossover trial was conducted on 10 patients with HF with reduced ejection fraction (HFrEF). The trial design involved two 4- week intervention periods of UA (500 mg BID) and placebo, separated by a 2-week washout phase. The patients underwent two-dimensional echocardiogram examination as well as blood sampling at the beginning and end of each period. RESULTS All patients completed the study. The results failed to reveal any significant effect of UA supplementation on echocardiographic measures (LVEF, LVEDD, LVESV, and TAPSE). Plasma concentrations of pro-BNP, glucose, and CRP (p >0.05) were also not altered. Serum HDL-C levels were increased with UA compared with placebo (+6.46 ± 2.33 mg/dL, p =0.026), whereas other lipid indices (LDL-C, triglycerides, total cholesterol, and VLDL-C) remained unchanged (p >0.05). CONCLUSION The results of the present study do not support any positive effect of UA supplementation in improving echocardiographic and biochemical indices of HFrEF. Further studies with higher doses of UA and longer supplementation duration are encouraged to be conducted.
Collapse
Affiliation(s)
- Tannaz Jamialahmadi
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maede Hasanpour
- Department of Pharmacognosy and Medicinal Plants Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farveh Vakilian
- Atherosclerosis Prevention Research Centre, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Peter E Penson
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Milad Iranshahy
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
20
|
Carvalho RA. The glycolytic pathway to heart failure. GLYCOLYSIS 2024:235-266. [DOI: 10.1016/b978-0-323-91704-9.00010-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
21
|
Qin S, Ren YC, Liu JY, Chen WB, Fu B, Zheng J, Fu XY. ANXA1sp attenuates sepsis-induced myocardial injury by promoting mitochondrial biosynthesis and inhibiting oxidative stress and autophagy via SIRT3 upregulation. Kaohsiung J Med Sci 2024; 40:35-45. [PMID: 37877496 DOI: 10.1002/kjm2.12767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 10/26/2023] Open
Abstract
Sepsis-induced myocardial injury is one of the most difficult complications of sepsis in intensive care units. Annexin A1 (ANXA1) short peptide (ANXA1sp) protects organs during the perioperative period. However, the protective effect of ANXA1sp against sepsis-induced myocardial injury remains unclear. We aimed to explore the protective effects and mechanisms of ANXA1sp against sepsis-induced myocardial injury both in vitro and in vivo. Cellular and animal models of myocardial injury in sepsis were established with lipopolysaccharide. The cardiac function of mice was assessed by high-frequency echocardiography. Elisa assay detected changes in inflammatory mediators and markers of myocardial injury. Western blotting detected autophagy and mitochondrial biosynthesis-related proteins. Autophagic flux changes were observed by confocal microscopy, and autophagosomes were evaluated by TEM. ATP, SOD, ROS, and MDA levels were also detected.ANXA1sp pretreatment enhanced the 7-day survival rate, improved cardiac function, and reduced TNF-α, IL-6, IL-1β, CK-MB, cTnI, and LDH levels. ANXA1sp significantly increased the expression of sirtuin-3 (SIRT3), mitochondrial biosynthesis-related proteins peroxisome proliferator-activated receptor γ co-activator 1α (PGC-1α), and mitochondrial transcription factor A (TFAM). ANXA1sp increased mitochondrial membrane potential (△Ψm), ATP, and SOD, and decreased ROS, autophagy flux, the production of autophagosomes per unit area, and MDA levels. The protective effect of ANXA1sp decreased significantly after SIRT3 silencing in vitro and in vivo, indicating that the key factor in ANXA1sp's protective role is the upregulation of SIRT3. In summary, ANXA1sp attenuated sepsis-induced myocardial injury by upregulating SIRT3 to promote mitochondrial biosynthesis and inhibit oxidative stress and autophagy.
Collapse
Affiliation(s)
- Song Qin
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
| | - Ying-Cong Ren
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
| | - Jun-Ya Liu
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
| | - Wen-Bo Chen
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
| | - Bao Fu
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
| | - Jie Zheng
- Department of anesthesiology, Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
| | - Xiao-Yun Fu
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
| |
Collapse
|
22
|
Jin G, Wang K, Zhao Y, Yuan S, He Z, Zhang J. Targeting histone deacetylases for heart diseases. Bioorg Chem 2023; 138:106601. [PMID: 37224740 DOI: 10.1016/j.bioorg.2023.106601] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/17/2023] [Accepted: 05/05/2023] [Indexed: 05/26/2023]
Abstract
Histone deacetylases (HDACs) are responsible for the deacetylation of lysine residues in histone or non-histone substrates, leading to the regulation of many biological functions, such as gene transcription, translation and remodeling chromatin. Targeting HDACs for drug development is a promising way for human diseases, including cancers and heart diseases. In particular, numerous HDAC inhibitors have revealed potential clinical value for the treatment of cardiac diseases in recent years. In this review, we systematically summarize the therapeutic roles of HDAC inhibitors with different chemotypes on heart diseases. Additionally, we discuss the opportunities and challenges in developing HDAC inhibitors for the treatment of cardiac diseases.
Collapse
Affiliation(s)
- Gang Jin
- Pharmacy College, Henan University of Chinese Medicine, 450046 Zhengzhou, China
| | - Kaiyue Wang
- Pharmacy College, Henan University of Chinese Medicine, 450046 Zhengzhou, China
| | - Yaohui Zhao
- Pharmacy College, Henan University of Chinese Medicine, 450046 Zhengzhou, China
| | - Shuo Yuan
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Zhangxu He
- Pharmacy College, Henan University of Chinese Medicine, 450046 Zhengzhou, China.
| | - Jingyu Zhang
- Pharmacy College, Henan University of Chinese Medicine, 450046 Zhengzhou, China.
| |
Collapse
|
23
|
Yang Z, Sun L, Wang H. Identification of mitophagy-related genes with potential clinical utility in myocardial infarction at transcriptional level. Front Cardiovasc Med 2023; 10:1166324. [PMID: 37304955 PMCID: PMC10250750 DOI: 10.3389/fcvm.2023.1166324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 05/09/2023] [Indexed: 06/13/2023] Open
Abstract
Background Myocardial infarction (MI) ranks among the most prevalent cardiovascular diseases. Insufficient blood flow to the coronary arteries always leads to ischemic necrosis of the cardiac muscle. However, the mechanism of myocardial injury after MI remains unclear. This article aims to explore the potential common genes between mitophagy and MI and to construct a suitable prediction model. Methods Two Gene Expression Omnibus (GEO) datasets (GSE62646 and GSE59867) were used to screen the differential expression genes in peripheral blood. SVM, RF, and LASSO algorithm were employed to find MI and mitophagy-related genes. Moreover, DT, KNN, RF, SVM and LR were conducted to build the binary models, and screened the best model to further external validation (GSE61144) and internal validation (10-fold cross validation and Bootstrap), respectively. The performance of various machine learning models was compared. In addition, immune cell infiltration correlation analysis was conducted with MCP-Counter and CIBERSORT. Results We finally identified ATG5, TOMM20, MFN2 transcriptionally differed between MI and stable coronary artery diseases. Both internal and external validation supported that these three genes could accurately predict MI withAUC = 0.914 and 0.930 by logistic regression, respectively. Additionally, functional analysis suggested that monocytes and neutrophils might be involved in mitochondrial autophagy after myocardial infarction. Conclusion The data showed that the transcritional levels of ATG5, TOMM20 and MFN2 in patients with MI were significantly different from the control group, which might be helpful to further accurately diagnose diseases and have potential application value in clinical practice.
Collapse
Affiliation(s)
- Zhikai Yang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Liang Sun
- The NHC Key Laboratory of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| | - Hua Wang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
24
|
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
|
25
|
Soh JEC, Shimizu A, Molla MR, Zankov DP, Nguyen LKC, Khan MR, Tesega WW, Chen S, Tojo M, Ito Y, Sato A, Hitosugi M, Miyagawa S, Ogita H. RhoA rescues cardiac senescence by regulating Parkin-mediated mitophagy. J Biol Chem 2023; 299:102993. [PMID: 36758801 PMCID: PMC10020657 DOI: 10.1016/j.jbc.2023.102993] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/11/2023] Open
Abstract
Heart failure is one of the leading causes of death worldwide. RhoA, a small GTPase, governs actin dynamics in various tissue and cell types, including cardiomyocytes; however, its involvement in cardiac function has not been fully elucidated. Here, we generated cardiomyocyte-specific RhoA conditional knockout (cKO) mice, which demonstrated a significantly shorter lifespan with left ventricular dilation and severely impaired ejection fraction. We found that the cardiac tissues of the cKO mice exhibited structural disorganization with fibrosis and also exhibited enhanced senescence compared with control mice. In addition, we show that cardiomyocyte mitochondria were structurally abnormal in the aged cKO hearts. Clearance of damaged mitochondria by mitophagy was remarkably inhibited in both cKO cardiomyocytes and RhoA-knockdown HL-1 cultured cardiomyocytes. In RhoA-depleted cardiomyocytes, we reveal that the expression of Parkin, an E3 ubiquitin ligase that plays a crucial role in mitophagy, was reduced, and expression of N-Myc, a negative regulator of Parkin, was increased. We further reveal that the RhoA-Rho kinase axis induced N-Myc phosphorylation, which led to N-Myc degradation and Parkin upregulation. Re-expression of Parkin in RhoA-depleted cardiomyocytes restored mitophagy, reduced mitochondrial damage, attenuated cardiomyocyte senescence, and rescued cardiac function both in vitro and in vivo. Finally, we found that patients with idiopathic dilated cardiomyopathy without causal mutations for dilated cardiomyopathy showed reduced cardiac expression of RhoA and Parkin. These results suggest that RhoA promotes Parkin-mediated mitophagy as an indispensable mechanism contributing to cardioprotection in the aging heart.
Collapse
Affiliation(s)
- Joanne Ern Chi Soh
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Akio Shimizu
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Md Rasel Molla
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Dimitar P Zankov
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Le Kim Chi Nguyen
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Mahbubur Rahman Khan
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Wondwossen Wale Tesega
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Si Chen
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan; Department of Emergency, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Misa Tojo
- Division of Legal Medicine, Department of Social Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Yoshito Ito
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Akira Sato
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Masahito Hitosugi
- Division of Legal Medicine, Department of Social Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hisakazu Ogita
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan.
| |
Collapse
|
26
|
Chen H, Zhou H, Yang J, Wan H, He Y. Guhong injection mitigates myocardial ischemia/reperfusion injury by activating GST P to inhibit ASK1-JNK/p38 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 109:154603. [PMID: 36610111 DOI: 10.1016/j.phymed.2022.154603] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/30/2022] [Accepted: 12/12/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Guhong injection (GHI), a novel compound preparation that is composed of a chemical drug, namely aceglutamide, and the aqueous extract of safflower (Carthamus tinctorius L.), exhibits extreme antioxidative, antiapoptotic, anti-inflammatory, and neuroprotective effects. Since oxidative stress, apoptosis, and inflammatory response are all the dominant mechanisms of myocardial ischemia/reperfusion (MI/R) injury, we probe into the protective mechanism of GHI on MI/R injury for the first time. METHODS In this research, we first employed molecular docking to determine whether three active ingredients in GHI, acetylglutamine (NAG), hydroxysafflor yellow A (HSYA), and syringin, possessed the potential activity to modulate the protein, glutathione S-transferase P (GST P). We further identified the protective effect of GHI on myocardial tissue with TTC staining, HE staining, TUNEL staining, and ELISA, and on H9c2 with flow cytometry and ELISA. We next explored whether the cardioprotective effect of GHI on left anterior descending ligation-reperfusion in rats and hypoxia/reoxygenation (H/R) in H9c2 cells was related to activate GST P to inhibit ASK1-JNK/p38 pathway via approaches of qRT-PCR and Western blot. RESULTS Results of molecular docking indicated that all three compounds spontaneously docked to GST P, among them the binding affinities of both HSYA and syringin to GST P were higher than NAG. In vivo, GHI reduced myocardial infarction size and mitigated myocardial pathological injury. In vitro, GHI enhanced cell viability and extenuated depolarization of mitochondrial membrane potential. In addition, the results of in vivo and in vitro studies demonstrated that the cardioprotection of GHI was associated with improving the mRNA and protein expression levels of GST P to modulate oxidative stress, and inhibiting the levels of mRNA expression and protein phosphorylation of ASK1, JNK, and p38. However, the suppressed effect of GHI on ASK1-JNK/p38 pathway was reversed by ethacrynic acid (EA, a GST inhibitor), indicating that the regulation of GHI on ASK1-JNK/p38 was related to the activity of GST P. Besides, the in vitro results of qRT-PCR and western-blot also certified that the inhibited JNK and p38 further reduced Bax expression and elevated Bcl-2 expression to reduce the expression of caspase-3 to exert anti-apoptosis effects. CONCLUSION Taken together, the cardioprotection of GHI mainly incarnated in activating GST P to relieve oxidation properties, thereby inhibiting ASK1-JNK/p38 pathway to suppress apoptosis.
Collapse
Affiliation(s)
- Haiyang Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Huifen Zhou
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Jiehong Yang
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Haitong Wan
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Yu He
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| |
Collapse
|
27
|
Lu Y, Li Z, Zhang S, Zhang T, Liu Y, Zhang L. Cellular mitophagy: Mechanism, roles in diseases and small molecule pharmacological regulation. Theranostics 2023; 13:736-766. [PMID: 36632220 PMCID: PMC9830443 DOI: 10.7150/thno.79876] [Citation(s) in RCA: 265] [Impact Index Per Article: 132.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/23/2022] [Indexed: 01/06/2023] Open
Abstract
Cellular mitophagy means that cells selectively wrap and degrade damaged mitochondria through an autophagy mechanism, thus maintaining mitochondria and intracellular homeostasis. In recent years, mitophagy has received increasing attention as a research hotspot related to the pathogenesis of clinical diseases, such as neurodegenerative diseases, cardiovascular diseases, cancer, metabolic diseases, and so on. It has been found that the regulation of mitophagy may become a new direction for the treatment of some diseases. In addition, numerous small molecule modulators of mitophagy have also been reported, which provides new opportunities to comprehend the procedure and potential of therapeutic development. Taken together, in this review, we summarize current understanding of the mechanism of mitophagy, discuss the roles of mitophagy and its relationship with diseases, introduce the existing small-molecule pharmacological modulators of mitophagy and further highlight the significance of their development.
Collapse
Affiliation(s)
- Yingying Lu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Zhijia Li
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Shuangqian Zhang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Tongtong Zhang
- The Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China,Medical Research Center, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China
| | - Yanjun Liu
- The Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China,Medical Research Center, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China,✉ Corresponding authors: Yanjun Liu, E-mail: ; Lan Zhang, E-mail:
| | - Lan Zhang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China,✉ Corresponding authors: Yanjun Liu, E-mail: ; Lan Zhang, E-mail:
| |
Collapse
|
28
|
Pedriali G, Ramaccini D, Bouhamida E, Wieckowski MR, Giorgi C, Tremoli E, Pinton P. Perspectives on mitochondrial relevance in cardiac ischemia/reperfusion injury. Front Cell Dev Biol 2022; 10:1082095. [PMID: 36561366 PMCID: PMC9763599 DOI: 10.3389/fcell.2022.1082095] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease is the most common cause of death worldwide and in particular, ischemic heart disease holds the most considerable position. Even if it has been deeply studied, myocardial ischemia-reperfusion injury (IRI) is still a side-effect of the clinical treatment for several heart diseases: ischemia process itself leads to temporary damage to heart tissue and obviously the recovery of blood flow is promptly required even if it worsens the ischemic injury. There is no doubt that mitochondria play a key role in pathogenesis of IRI: dysfunctions of these important organelles alter cell homeostasis and survival. It has been demonstrated that during IRI the system of mitochondrial quality control undergoes alterations with the disruption of the complex balance between the processes of mitochondrial fusion, fission, biogenesis and mitophagy. The fundamental role of mitochondria is carried out thanks to the finely regulated connection to other organelles such as plasma membrane, endoplasmic reticulum and nucleus, therefore impairments of these inter-organelle communications exacerbate IRI. This review pointed to enhance the importance of the mitochondrial network in the pathogenesis of IRI with the aim to focus on potential mitochondria-targeting therapies as new approach to control heart tissue damage after ischemia and reperfusion process.
Collapse
Affiliation(s)
- Gaia Pedriali
- Maria Cecilia Hospital, GVM Care and Research, Cotignola, Italy
| | | | - Esmaa Bouhamida
- Maria Cecilia Hospital, GVM Care and Research, Cotignola, Italy
| | - Mariusz R. Wieckowski
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Carlotta Giorgi
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Science, Section of Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Elena Tremoli
- Maria Cecilia Hospital, GVM Care and Research, Cotignola, Italy,*Correspondence: Paolo Pinton, ; Elena Tremoli,
| | - Paolo Pinton
- Maria Cecilia Hospital, GVM Care and Research, Cotignola, Italy,Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Science, Section of Experimental Medicine, University of Ferrara, Ferrara, Italy,*Correspondence: Paolo Pinton, ; Elena Tremoli,
| |
Collapse
|
29
|
Ronconi-Krüger N, Pinheiro J, Simioni C, Nazari EM. Methylmercury Toxicity During Heart Development: A Combined Analysis of Morphological and Functional Parameters. Cardiovasc Toxicol 2022; 22:962-970. [DOI: 10.1007/s12012-022-09772-4] [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: 07/22/2022] [Accepted: 10/27/2022] [Indexed: 11/11/2022]
|
30
|
Viloria MAD, Li Q, Lu W, Nhu NT, Liu Y, Cui ZY, Cheng YJ, Lee SD. Effect of exercise training on cardiac mitochondrial respiration, biogenesis, dynamics, and mitophagy in ischemic heart disease. Front Cardiovasc Med 2022; 9:949744. [PMID: 36304547 PMCID: PMC9592995 DOI: 10.3389/fcvm.2022.949744] [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: 05/21/2022] [Accepted: 08/12/2022] [Indexed: 12/07/2022] Open
Abstract
Objective Cardiac mitochondrial dysfunction was found in ischemic heart disease (IHD). Hence, this study determined the effects of exercise training (ET) on cardiac mitochondrial respiration and cardiac mitochondrial quality control in IHD. Methods A narrative synthesis was conducted after searching animal studies written in English in three databases (PubMed, Web of Science, and EMBASE) until December 2020. Studies that used aerobic exercise as an intervention for at least 3 weeks and had at least normal, negative (sedentary IHD), and positive (exercise-trained IHD) groups were included. The CAMARADES checklist was used to check the quality of the included studies. Results The 10 included studies (CAMARADES score: 6–7/10) used swimming or treadmill exercise for 3–8 weeks. Seven studies showed that ET ameliorated cardiac mitochondrial respiratory function as manifested by decreased reactive oxygen species (ROS) production and increased complexes I-V activity, superoxide dismutase 2 (SOD2), respiratory control ratio (RCR), NADH dehydrogenase subunits 1 and 6 (ND1/6), Cytochrome B (CytB), and adenosine triphosphate (ATP) production. Ten studies showed that ET improved cardiac mitochondrial quality control in IHD as manifested by enhanced and/or controlled mitochondrial biogenesis, dynamics, and mitophagy. Four other studies showed that ET resulted in better cardiac mitochondrial physiological characteristics. Conclusion Exercise training could improve cardiac mitochondrial functions, including respiration, biogenesis, dynamics, and mitophagy in IHD. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=226817, identifier: CRD42021226817.
Collapse
Affiliation(s)
- Mary Audrey D. Viloria
- Department of Physical Therapy, Graduate Institute of Rehabilitation Science, China Medical University, Taichung, Taiwan,Department of Physical Therapy, College of Health Sciences, Mariano Marcos State University, Batac, Philippines
| | - Qing Li
- Department of Rehabilitation, Shanghai Xuhui Central Hospital, Shanghai, China
| | - Wang Lu
- Department of Traditional Treatment, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Nguyen Thanh Nhu
- Faculty of Medicine, Can Tho University of Medicine and Pharmacy, Can Tho, Vietnam
| | - Yijie Liu
- School of Rehabilitation Medicine, Shanghai University of Traditional Medicine, Shanghai, China,Institute of Rehabilitation Medicine, Shanghai University of Traditional Medicine, Shanghai, China
| | - Zhen-Yang Cui
- School of Rehabilitation Medicine, Weifang Medical University, Weifang, China
| | - Yu-Jung Cheng
- Department of Physical Therapy, Graduate Institute of Rehabilitation Science, China Medical University, Taichung, Taiwan,Yu-Jung Cheng
| | - Shin-Da Lee
- Department of Physical Therapy, Graduate Institute of Rehabilitation Science, China Medical University, Taichung, Taiwan,School of Rehabilitation Medicine, Weifang Medical University, Weifang, China,Department of Physical Therapy, Asia University, Taichung, Taiwan,*Correspondence: Shin-Da Lee
| |
Collapse
|
31
|
Beckley MA, Shrestha S, Singh KK, Portman MA. The role of mitochondria in the pathogenesis of Kawasaki disease. Front Immunol 2022; 13:1017401. [PMID: 36300112 PMCID: PMC9592088 DOI: 10.3389/fimmu.2022.1017401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/20/2022] [Indexed: 11/28/2022] Open
Abstract
Kawasaki disease is a systemic vasculitis, especially of the coronary arteries, affecting children. Despite extensive research, much is still unknown about the principal driver behind the amplified inflammatory response. We propose mitochondria may play a critical role. Mitochondria serve as a central hub, influencing energy generation, cell proliferation, and bioenergetics. Regulation of these biological processes, however, comes at a price. Release of mitochondrial DNA into the cytoplasm acts as damage-associated molecular patterns, initiating the development of inflammation. As a source of reactive oxygen species, they facilitate activation of the NLRP3 inflammasome. Kawasaki disease involves many of these inflammatory pathways. Progressive mitochondrial dysfunction alters the activity of immune cells and may play a role in the pathogenesis of Kawasaki disease. Because they contain their own genome, mitochondria are susceptible to mutation which can propagate their dysfunction and immunostimulatory potential. Population-specific variants in mitochondrial DNA have also been linked to racial disparities in disease risk and treatment response. Our objective is to critically examine the current literature of mitochondria's role in coordinating proinflammatory signaling pathways, focusing on potential mitochondrial dysfunction in Kawasaki disease. No association between impaired mitochondrial function and Kawasaki disease exists, but we suggest a relationship between the two. We hypothesize a framework of mitochondrial determinants that may contribute to ethnic/racial disparities in the progression of Kawasaki disease.
Collapse
Affiliation(s)
- Mikayla A. Beckley
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Sadeep Shrestha
- Department of Epidemiology, School of Public Health University of Alabama at Birmingham, Birmingham, AL, United States
| | - Keshav K. Singh
- Department of Genetics, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Michael A. Portman
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, United States
- Department of Pediatrics, Division of Cardiology, University of Washington, Seattle, WA, United States
| |
Collapse
|
32
|
Protective Effect of Natural Medicinal Plants on Cardiomyocyte Injury in Heart Failure: Targeting the Dysregulation of Mitochondrial Homeostasis and Mitophagy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3617086. [PMID: 36132224 PMCID: PMC9484955 DOI: 10.1155/2022/3617086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/16/2022] [Accepted: 08/18/2022] [Indexed: 11/22/2022]
Abstract
Heart failure occurs because of various cardiovascular pathologies, such as coronary artery disease or cardiorenal syndrome, eventually reaching end-stage disease. Various factors contribute to cardiac structural or functional changes that result in systolic or diastolic dysfunction. Several studies have confirmed that the key factor in heart failure progression is myocardial cell death, and mitophagy is the major mechanism regulating myocardial cell death in heart failure. The clinical mechanisms of heart failure are well understood in practice. However, the essential role of mitophagic regulation in heart failure has only recently received widespread attention. Receptor-mediated mitophagy is involved in various mitochondrial processes like oxidative stress injury, energy metabolism disorders, and calcium homeostasis, which are also the main causes of heart failure. Understanding of the diverse regulatory mechanisms in mitophagy and the complexity of its pathophysiology in heart failure remains incomplete. Related studies have found that various natural medicinal plants and active ingredients, such as flavonoids and saponins, can regulate mitophagy to a certain extent, improve myocardial function, and protect myocardial cells. This review comprehensively covers the relevant mechanisms of different types of mitophagy in regulating heart failure pathology and controlling mitochondrial adaptability to stress injury. Further, it explores the relationship between mitophagy and cardiac ejection dysfunction. Natural medicinal plant-targeted regulation strategies and scientific evidence on mitophagy were provided to elucidate current and potential strategies to apply mitophagy-targeted therapy for heart failure.
Collapse
|
33
|
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
|
34
|
Mendoza A, Karch J. Keeping the beat against time: Mitochondrial fitness in the aging heart. FRONTIERS IN AGING 2022; 3:951417. [PMID: 35958271 PMCID: PMC9360554 DOI: 10.3389/fragi.2022.951417] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 06/30/2022] [Indexed: 11/21/2022]
Abstract
The process of aging strongly correlates with maladaptive architectural, mechanical, and biochemical alterations that contribute to the decline in cardiac function. Consequently, aging is a major risk factor for the development of heart disease, the leading cause of death in the developed world. In this review, we will summarize the classic and recently uncovered pathological changes within the aged heart with an emphasis on the mitochondria. Specifically, we describe the metabolic changes that occur in the aging heart as well as the loss of mitochondrial fitness and function and how these factors contribute to the decline in cardiomyocyte number. In addition, we highlight recent pharmacological, genetic, or behavioral therapeutic intervention advancements that may alleviate age-related cardiac decline.
Collapse
Affiliation(s)
- Arielys Mendoza
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, United States
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, United States
| | - Jason Karch
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, United States
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
35
|
Lin J, Duan J, Wang Q, Xu S, Zhou S, Yao K. Mitochondrial Dynamics and Mitophagy in Cardiometabolic Disease. Front Cardiovasc Med 2022; 9:917135. [PMID: 35783853 PMCID: PMC9247260 DOI: 10.3389/fcvm.2022.917135] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 05/20/2022] [Indexed: 12/17/2022] Open
Abstract
Mitochondria play a key role in cellular metabolism. Mitochondrial dynamics (fusion and fission) and mitophagy, are critical to mitochondrial function. Fusion allows organelles to share metabolites, proteins, and mitochondrial DNA, promoting complementarity between damaged mitochondria. Fission increases the number of mitochondria to ensure that they are passed on to their offspring during mitosis. Mitophagy is a process of selective removal of excess or damaged mitochondria that helps improve energy metabolism. Cardiometabolic disease is characterized by mitochondrial dysfunction, high production of reactive oxygen species, increased inflammatory response, and low levels of ATP. Cardiometabolic disease is closely related to mitochondrial dynamics and mitophagy. This paper reviewed the mechanisms of mitochondrial dynamics and mitophagy (focus on MFN1, MFN2, OPA1, DRP1, and PINK1 proteins) and their roles in diabetic cardiomyopathy, myocardial infarction, cardiac hypertrophy, heart failure, atherosclerosis, and obesity.
Collapse
Affiliation(s)
- Jianguo Lin
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jinlong Duan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qingqing Wang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Siyu Xu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Simin Zhou
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Kuiwu Yao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Eye Hospital China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Kuiwu Yao
| |
Collapse
|
36
|
Abstract
As a muscular pump that contracts incessantly throughout life, the heart must constantly generate cellular energy to support contractile function and fuel ionic pumps to maintain electrical homeostasis. Thus, mitochondrial metabolism of multiple metabolic substrates such as fatty acids, glucose, ketones, and lactate is essential to ensuring an uninterrupted supply of ATP. Multiple metabolic pathways converge to maintain myocardial energy homeostasis. The regulation of these cardiac metabolic pathways has been intensely studied for many decades. Rapid adaptation of these pathways is essential for mediating the myocardial adaptation to stress, and dysregulation of these pathways contributes to myocardial pathophysiology as occurs in heart failure and in metabolic disorders such as diabetes. The regulation of these pathways reflects the complex interactions of cell-specific regulatory pathways, neurohumoral signals, and changes in substrate availability in the circulation. Significant advances have been made in the ability to study metabolic regulation in the heart, and animal models have played a central role in contributing to this knowledge. This review will summarize metabolic pathways in the heart and describe their contribution to maintaining myocardial contractile function in health and disease. The review will summarize lessons learned from animal models with altered systemic metabolism and those in which specific metabolic regulatory pathways have been genetically altered within the heart. The relationship between intrinsic and extrinsic regulators of cardiac metabolism and the pathophysiology of heart failure and how these have been informed by animal models will be discussed.
Collapse
Affiliation(s)
- Heiko Bugger
- University Heart Center Graz, Department of Cardiology, Medical University of Graz, Graz, Austria, Austria (H.B., N.J.B.)
| | - Nikole J Byrne
- University Heart Center Graz, Department of Cardiology, Medical University of Graz, Graz, Austria, Austria (H.B., N.J.B.)
| | - E Dale Abel
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (E.D.A.)
| |
Collapse
|
37
|
Yang J, Guo Q, Feng X, Liu Y, Zhou Y. Mitochondrial Dysfunction in Cardiovascular Diseases: Potential Targets for Treatment. Front Cell Dev Biol 2022; 10:841523. [PMID: 35646910 PMCID: PMC9140220 DOI: 10.3389/fcell.2022.841523] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 04/13/2022] [Indexed: 12/20/2022] Open
Abstract
Cardiovascular diseases (CVDs) are serious public health issues and are responsible for nearly one-third of global deaths. Mitochondrial dysfunction is accountable for the development of most CVDs. Mitochondria produce adenosine triphosphate through oxidative phosphorylation and inevitably generate reactive oxygen species (ROS). Excessive ROS causes mitochondrial dysfunction and cell death. Mitochondria can protect against these damages via the regulation of mitochondrial homeostasis. In recent years, mitochondria-targeted therapy for CVDs has attracted increasing attention. Various studies have confirmed that clinical drugs (β-blockers, angiotensin-converting enzyme inhibitors/angiotensin receptor-II blockers) against CVDs have mitochondrial protective functions. An increasing number of cardiac mitochondrial targets have shown their cardioprotective effects in experimental and clinical studies. Here, we briefly introduce the mechanisms of mitochondrial dysfunction and summarize the progression of mitochondrial targets against CVDs, which may provide ideas for experimental studies and clinical trials.
Collapse
|
38
|
Li A, Gao M, Liu B, Qin Y, Chen L, Liu H, Wu H, Gong G. Mitochondrial autophagy: molecular mechanisms and implications for cardiovascular disease. Cell Death Dis 2022; 13:444. [PMID: 35534453 PMCID: PMC9085840 DOI: 10.1038/s41419-022-04906-6] [Citation(s) in RCA: 195] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/27/2022] [Accepted: 05/03/2022] [Indexed: 12/13/2022]
Abstract
Mitochondria are highly dynamic organelles that participate in ATP generation and involve calcium homeostasis, oxidative stress response, and apoptosis. Dysfunctional or damaged mitochondria could cause serious consequences even lead to cell death. Therefore, maintaining the homeostasis of mitochondria is critical for cellular functions. Mitophagy is a process of selectively degrading damaged mitochondria under mitochondrial toxicity conditions, which plays an essential role in mitochondrial quality control. The abnormal mitophagy that aggravates mitochondrial dysfunction is closely related to the pathogenesis of many diseases. As the myocardium is a highly oxidative metabolic tissue, mitochondria play a central role in maintaining optimal performance of the heart. Dysfunctional mitochondria accumulation is involved in the pathophysiology of cardiovascular diseases, such as myocardial infarction, cardiomyopathy and heart failure. This review discusses the most recent progress on mitophagy and its role in cardiovascular disease.
Collapse
Affiliation(s)
- Anqi Li
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Meng Gao
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Bilin Liu
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yuan Qin
- Department of Pharmacy, Shanghai East Hospital, Tongji University, Shanghai, 200120, China
| | - Lei Chen
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Hanyu Liu
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Huayan Wu
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Guohua Gong
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
39
|
Honokiol Provides Cardioprotection from Myocardial Ischemia/Reperfusion Injury (MI/RI) by Inhibiting Mitochondrial Apoptosis via the PI3K/AKT Signaling Pathway. Cardiovasc Ther 2022; 2022:1001692. [PMID: 35414825 PMCID: PMC8977331 DOI: 10.1155/2022/1001692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 03/10/2022] [Indexed: 11/17/2022] Open
Abstract
Background Myocardial injury refers to a major complication that occurs in myocardial ischemia/reperfusion injury (MI/RI). Honokiol is a well-recognized active compound extracted from the traditional Chinese herb known as Magnolia officinalis and is utilized in treating different vascular diseases. This research is aimed at examining whether Honokiol might alleviate myocardial injury in an MI/RI model. Methods Seventy-eight male C57BL/6 mice were categorized randomly into three cohorts including the Sham operation (Sham) cohort, the MI/RI cohort (Con), and the Honokiol cohort (n = 26 for each cohort). The mice in the Honokiol cohort were treated with Honokiol before MI/RI surgery (0.2 mg/kg/day for 14 days, intraperitoneal), while the mice in the Con cohort were given an intraperitoneal injection with an equivalent volume of vehicle (DMSO) daily in 14 days prior to exposure to MI/RI. After the surgery, creatine kinase- (CK-) MB and cardiac troponin T (cTnT) levels, as well as the infarct area, were measured to assess the degree of myocardial damage. Apoptotic levels were detected using terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) staining. Electron microscopy was utilized to identify mitochondrial damage. Lastly, the expression levels of glyceraldehyde-3-phosphate dehydrogenase (GAPDH), cleaved caspase-9, cytochrome C (Cyt-C), B cell lymphoma/leukemia-2 (Bcl-2), B cell lymphoma/leukemia-2 associated X (Bax), AKT, p-AKT, PI3K, and p-PI3K were analyzed utilizing western blotting. Results Honokiol can reduce the MI/RI-induced cTnT and CK-MB levels, apoptosis index, and mitochondrial swelling in cardiomyocytes via activating the PI3K/AKT signaling pathway. Conclusion Honokiol provides cardiac protection from MI/RI by suppressing mitochondrial apoptosis through the PI3K/AKT signaling pathway.
Collapse
|
40
|
Wachoski-Dark E, Zhao T, Khan A, Shutt TE, Greenway SC. Mitochondrial Protein Homeostasis and Cardiomyopathy. Int J Mol Sci 2022; 23:3353. [PMID: 35328774 PMCID: PMC8953902 DOI: 10.3390/ijms23063353] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/15/2022] [Accepted: 03/17/2022] [Indexed: 12/06/2022] Open
Abstract
Human mitochondrial disorders impact tissues with high energetic demands and can be associated with cardiac muscle disease (cardiomyopathy) and early mortality. However, the mechanistic link between mitochondrial disease and the development of cardiomyopathy is frequently unclear. In addition, there is often marked phenotypic heterogeneity between patients, even between those with the same genetic variant, which is also not well understood. Several of the mitochondrial cardiomyopathies are related to defects in the maintenance of mitochondrial protein homeostasis, or proteostasis. This essential process involves the importing, sorting, folding and degradation of preproteins into fully functional mature structures inside mitochondria. Disrupted mitochondrial proteostasis interferes with mitochondrial energetics and ATP production, which can directly impact cardiac function. An inability to maintain proteostasis can result in mitochondrial dysfunction and subsequent mitophagy or even apoptosis. We review the known mitochondrial diseases that have been associated with cardiomyopathy and which arise from mutations in genes that are important for mitochondrial proteostasis. Genes discussed include DnaJ heat shock protein family member C19 (DNAJC19), mitochondrial import inner membrane translocase subunit TIM16 (MAGMAS), translocase of the inner mitochondrial membrane 50 (TIMM50), mitochondrial intermediate peptidase (MIPEP), X-prolyl-aminopeptidase 3 (XPNPEP3), HtraA serine peptidase 2 (HTRA2), caseinolytic mitochondrial peptidase chaperone subunit B (CLPB) and heat shock 60-kD protein 1 (HSPD1). The identification and description of disorders with a shared mechanism of disease may provide further insights into the disease process and assist with the identification of potential therapeutics.
Collapse
Affiliation(s)
- Emily Wachoski-Dark
- Department of Cardiac Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada;
- Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Tian Zhao
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada;
| | - Aneal Khan
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada;
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
- M.A.G.I.C. Inc., Calgary, AB T2E 7Z4, Canada
| | - Timothy E. Shutt
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada;
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Steven C. Greenway
- Department of Cardiac Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada;
- Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada;
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada;
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
41
|
Zhao M, Lian A, Zhong L, Guo R. The regulatory mechanism between lysosomes and mitochondria in the aetiology of cardiovascular diseases. Acta Physiol (Oxf) 2022; 234:e13757. [PMID: 34978753 DOI: 10.1111/apha.13757] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/16/2021] [Accepted: 01/01/2022] [Indexed: 11/28/2022]
Abstract
Coordinated action among various organelles maintains cellular functions. For instance, mitochondria and lysosomes are the main organelles contributing to cellular metabolism and provide energy for cardiomyocyte contraction. They also provide essential signalling platforms in the cell that regulate many key processes such as autophagy, apoptosis, oxidative stress, inflammation and cell death. Often, abnormalities in mitochondrial or lysosomal structures and functions bring about cardiovascular diseases (CVDs). Although the communication between mitochondria and lysosomes throughout the cardiovascular system is intensely studied, the regulatory mechanisms have not been completely understood. Thus, we summarize the most recent studies related to mitochondria and lysosomes' role in CVDs and their potential connections and communications under cardiac pathophysiological conditions. Further, we discuss limitations and future perspectives regarding diagnosis, therapeutic strategies and drug discovery in CVDs.
Collapse
Affiliation(s)
- Mengxue Zhao
- College of Life Sciences Institute of Life Science and Green Development Hebei University Baoding China
| | - Andrew Lian
- College of Osteopathic Medicine of the Pacific Western University of Health Sciences Pomona California USA
| | - Li Zhong
- College of Life Sciences Institute of Life Science and Green Development Hebei University Baoding China
- College of Osteopathic Medicine of the Pacific Western University of Health Sciences Pomona California USA
| | - Rui Guo
- College of Life Sciences Institute of Life Science and Green Development Hebei University Baoding China
- The Key Laboratory of Zoological Systematics and Application College of Life Sciences Hebei University Baoding China
| |
Collapse
|
42
|
Isoform-selective HDAC Inhibitor Mocetinostat (MGCD0103) Alleviates Myocardial Ischemia/Reperfusion Injury via Mitochondrial Protection through the HDACs/CREB/PGC-1α Signaling Pathway. J Cardiovasc Pharmacol 2021; 79:217-228. [PMID: 34983914 DOI: 10.1097/fjc.0000000000001174] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 09/28/2021] [Indexed: 11/25/2022]
Abstract
ABSTRACT Over the past decade, histone deacetylases (HDACs) has been proven to manipulate development and exacerbation of cardiovascular diseases, including myocardial ischemia/reperfusion injury (MIRI), cardiac hypertrophy, ventricular remodeling, myocardial fibrosis. Inhibition of histone deacetylases, especially class-I HDACs, is potent to protection of ischemic myocardium after ischemia/reperfusion. Herein, we examine whether mocetinostat (MGCD0103, MOCE), a class-I selective HDAC inhibitor in phase-II clinical trial, conducts cardioprotection under ischemia/reperfusion (I/R) in vivo and vitro, if so, reveal its potential pharmacological mechanism to provide an experimental and theoretical basis for mocetinostat usage in a clinical setting. HCMs were exposed to hypoxia and reoxygenation (H/R), with or without mocetinostat treatment. H/R reduced mitochondrial membrane potential (MMP) and induced HCMs apoptosis. Mocetinostat pre-treatment reversed these H/R-induced mitochondrial damage and cellular apoptosis and upregulated CREB, p-CREB and PGC-1α in HCMs during H/R. Transfection with siRNA against PGC-1α or CREB abolished the protective effects of mocetinostat on cardiomyocytes undergoing H/R. In vivo, mocetinostat was demonstrated to protect myocardial injury posed by myocardial ischemia/reperfusion (I/R) via activation of CREB and upregulation of PGC-1α. Mocetinostat (MGCD0103) can protect myocardium from ischemia/reperfusion injury through mitochondrial protection mediated by CREB/PGC-1α pathway. Therefore, activation of the CREB/PGC-1α signaling pathway via inhibition of Class-I HDACs may be a promising new therapeutic strategy for alleviating myocardial reperfusion injury.
Collapse
|
43
|
Jiang X, Cai S, Jin Y, Wu F, He J, Wu X, Tan Y, Wang Y. Irisin Attenuates Oxidative Stress, Mitochondrial Dysfunction, and Apoptosis in the H9C2 Cellular Model of Septic Cardiomyopathy through Augmenting Fundc1-Dependent Mitophagy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:2989974. [PMID: 34457111 PMCID: PMC8390168 DOI: 10.1155/2021/2989974] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 07/25/2021] [Accepted: 08/06/2021] [Indexed: 12/22/2022]
Abstract
In the present study, we used lipopolysaccharide- (LPS-) stimulated H9C2 cardiomyocytes to investigate whether irisin treatment attenuates septic cardiomyopathy via Fundc1-related mitophagy. Fundc1 levels and mitophagy were significantly reduced in LPS-stimulated H9C2 cardiomyocytes but were significantly increased by irisin treatment. Irisin significantly increased ATP production and the activities of mitochondrial complexes I and III in the LPS-stimulated cardiomyocytes. Irisin also improved glucose metabolism and significantly reduced LPS-induced levels of reactive oxygen species by increasing the activities of antioxidant enzymes, glutathione peroxidase (GPX), and superoxide dismutase (SOD), as well as levels of reduced glutathione (GSH). TUNEL assays showed that irisin significantly reduced LPS-stimulated cardiomyocyte apoptosis by suppressing the activation of caspase-3 and caspase-9. However, the beneficial effects of irisin on oxidative stress, mitochondrial metabolism, and viability of LPS-stimulated H9C2 cardiomyocytes were abolished by silencing Fundc1. These results demonstrate that irisin abrogates mitochondrial dysfunction, oxidative stress, and apoptosis through Fundc1-related mitophagy in LPS-stimulated H9C2 cardiomyocytes. This suggests irisin is a potentially useful treatment for septic cardiomyopathy, though further investigations are necessary to confirm our findings.
Collapse
Affiliation(s)
- Xiaoqing Jiang
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Shumin Cai
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yinghui Jin
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Feng Wu
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jing He
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xixuan Wu
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ying Tan
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yu Wang
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
44
|
Marek-Iannucci S, Ozdemir AB, Moreira D, Gomez AC, Lane M, Porritt RA, Lee Y, Shimada K, Abe M, Stotland A, Zemmour D, Parker S, Sanchez-Lopez E, Van Eyk J, Gottlieb RA, Fishbein M, Karin M, Crother TR, Noval Rivas M, Arditi M. Autophagy-mitophagy induction attenuates cardiovascular inflammation in a murine model of Kawasaki disease vasculitis. JCI Insight 2021; 6:e151981. [PMID: 34403365 PMCID: PMC8492304 DOI: 10.1172/jci.insight.151981] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/11/2021] [Indexed: 01/18/2023] Open
Abstract
Kawasaki disease (KD) is the leading cause of acquired heart disease among children. Murine and human data suggest that the NLRP3-IL-1β pathway is the main driver of KD pathophysiology. NLRP3 can be activated during defective autophagy/mitophagy. We used the Lactobacillus casei cell wall extract (LCWE) murine model of KD vasculitis, to examine the role of autophagy/mitophagy on cardiovascular lesion development. LCWE-injected mice had impaired autophagy/mitophagy and increased levels of ROS in cardiovascular lesions, together with increased systemic 8-OHdG release. Enhanced autophagic flux significantly reduced cardiovascular lesions in LCWE-injected mice, whereas autophagy blockade increased inflammation. Vascular smooth muscle cell specific deletion of Atg16l1 and global Parkin-/- significantly increased disease formation, supporting the importance of autophagy/mitophagy in this model. Ogg1-/- mice had significantly increased lesions with increased NLRP3 activity, whereas treatment with MitoQ, reduced vascular tissue inflammation, ROS production and systemic 8-OHdG release. Treatment with MN58b or Metformin (increasing AMPK and reducing ROS), resulted in decreased disease formation. Our results demonstrate that impaired autophagy/mitophagy and ROS-dependent damage exacerbate the development of murine KD vasculitis. This pathway can be efficiently targeted to reduce disease severity. These findings enhance our understanding of KD pathogenesis and identify novel therapeutic avenues for KD treatment.
Collapse
Affiliation(s)
- Stefanie Marek-Iannucci
- Graduate School of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - A Beyza Ozdemir
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Debbie Moreira
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Angela C Gomez
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Malcolm Lane
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Rebecca A Porritt
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Youngho Lee
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Kenichi Shimada
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Masanori Abe
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Aleksandr Stotland
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - David Zemmour
- Department of Pathology, Harvard Medical School and Brigham and Women's Hospital, Boston, United States of America
| | - Sarah Parker
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | | | - Jennifer Van Eyk
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Roberta A Gottlieb
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Michael Fishbein
- Department of Pathology, UCLA, Los Angeles, United States of America
| | - Michael Karin
- Department of Pathology, UCSD, San Diego, United States of America
| | - Timothy R Crother
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Magali Noval Rivas
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, United States of America
| | - Moshe Arditi
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, United States of America
| |
Collapse
|
45
|
Mitochondrial Modulations, Autophagy Pathways Shifts in Viral Infections: Consequences of COVID-19. Int J Mol Sci 2021; 22:ijms22158180. [PMID: 34360945 PMCID: PMC8347486 DOI: 10.3390/ijms22158180] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/24/2021] [Accepted: 07/24/2021] [Indexed: 01/05/2023] Open
Abstract
Mitochondria are vital intracellular organelles that play an important role in regulating various intracellular events such as metabolism, bioenergetics, cell death (apoptosis), and innate immune signaling. Mitochondrial fission, fusion, and membrane potential play a central role in maintaining mitochondrial dynamics and the overall shape of mitochondria. Viruses change the dynamics of the mitochondria by altering the mitochondrial processes/functions, such as autophagy, mitophagy, and enzymes involved in metabolism. In addition, viruses decrease the supply of energy to the mitochondria in the form of ATP, causing viruses to create cellular stress by generating ROS in mitochondria to instigate viral proliferation, a process which causes both intra- and extra-mitochondrial damage. SARS-COV2 propagates through altering or changing various pathways, such as autophagy, UPR stress, MPTP and NLRP3 inflammasome. Thus, these pathways act as potential targets for viruses to facilitate their proliferation. Autophagy plays an essential role in SARS-COV2-mediated COVID-19 and modulates autophagy by using various drugs that act on potential targets of the virus to inhibit and treat viral infection. Modulated autophagy inhibits coronavirus replication; thus, it becomes a promising target for anti-coronaviral therapy. This review gives immense knowledge about the infections, mitochondrial modulations, and therapeutic targets of viruses.
Collapse
|
46
|
Abstract
Alterations in cardiac energy metabolism contribute to the severity of heart failure. However, the energy metabolic changes that occur in heart failure are complex and are dependent not only on the severity and type of heart failure present but also on the co-existence of common comorbidities such as obesity and type 2 diabetes. The failing heart faces an energy deficit, primarily because of a decrease in mitochondrial oxidative capacity. This is partly compensated for by an increase in ATP production from glycolysis. The relative contribution of the different fuels for mitochondrial ATP production also changes, including a decrease in glucose and amino acid oxidation, and an increase in ketone oxidation. The oxidation of fatty acids by the heart increases or decreases, depending on the type of heart failure. For instance, in heart failure associated with diabetes and obesity, myocardial fatty acid oxidation increases, while in heart failure associated with hypertension or ischemia, myocardial fatty acid oxidation decreases. Combined, these energy metabolic changes result in the failing heart becoming less efficient (ie, a decrease in cardiac work/O2 consumed). The alterations in both glycolysis and mitochondrial oxidative metabolism in the failing heart are due to both transcriptional changes in key enzymes involved in these metabolic pathways, as well as alterations in NAD redox state (NAD+ and nicotinamide adenine dinucleotide levels) and metabolite signaling that contribute to posttranslational epigenetic changes in the control of expression of genes encoding energy metabolic enzymes. Alterations in the fate of glucose, beyond flux through glycolysis or glucose oxidation, also contribute to the pathology of heart failure. Of importance, pharmacological targeting of the energy metabolic pathways has emerged as a novel therapeutic approach to improving cardiac efficiency, decreasing the energy deficit and improving cardiac function in the failing heart.
Collapse
Affiliation(s)
- Gary D Lopaschuk
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada (G.D.L., Q.G.K.)
| | - Qutuba G Karwi
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada (G.D.L., Q.G.K.)
| | - Rong Tian
- Mitochondria and Metabolism Center, University of Washington, Seattle (R.T.)
| | - Adam R Wende
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.)
| | - E Dale Abel
- Division of Endocrinology and Metabolism, University of Iowa Carver College of Medicine, Iowa City (E.D.A.).,Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City (E.D.A.)
| |
Collapse
|
47
|
Morciano G, Vitto VAM, Bouhamida E, Giorgi C, Pinton P. Mitochondrial Bioenergetics and Dynamism in the Failing Heart. Life (Basel) 2021; 11:436. [PMID: 34066065 PMCID: PMC8151847 DOI: 10.3390/life11050436] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/07/2021] [Accepted: 05/07/2021] [Indexed: 12/13/2022] Open
Abstract
The heart is responsible for pumping blood, nutrients, and oxygen from its cavities to the whole body through rhythmic and vigorous contractions. Heart function relies on a delicate balance between continuous energy consumption and generation that changes from birth to adulthood and depends on a very efficient oxidative metabolism and the ability to adapt to different conditions. In recent years, mitochondrial dysfunctions were recognized as the hallmark of the onset and development of manifold heart diseases (HDs), including heart failure (HF). HF is a severe condition for which there is currently no cure. In this condition, the failing heart is characterized by a disequilibrium in mitochondrial bioenergetics, which compromises the basal functions and includes the loss of oxygen and substrate availability, an altered metabolism, and inefficient energy production and utilization. This review concisely summarizes the bioenergetics and some other mitochondrial features in the heart with a focus on the features that become impaired in the failing heart.
Collapse
Affiliation(s)
- Giampaolo Morciano
- Maria Cecilia Hospital, GVM Care&Research, 48033 Cotignola, Italy
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (V.A.M.V.); (E.B.); (C.G.)
| | - Veronica Angela Maria Vitto
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (V.A.M.V.); (E.B.); (C.G.)
| | - Esmaa Bouhamida
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (V.A.M.V.); (E.B.); (C.G.)
| | - Carlotta Giorgi
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (V.A.M.V.); (E.B.); (C.G.)
| | - Paolo Pinton
- Maria Cecilia Hospital, GVM Care&Research, 48033 Cotignola, Italy
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (V.A.M.V.); (E.B.); (C.G.)
| |
Collapse
|
48
|
Ji W, Wan T, Zhang F, Zhu X, Guo S, Mei X. Aldehyde Dehydrogenase 2 Protects Against Lipopolysaccharide-Induced Myocardial Injury by Suppressing Mitophagy. Front Pharmacol 2021; 12:641058. [PMID: 34025411 PMCID: PMC8139555 DOI: 10.3389/fphar.2021.641058] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 04/19/2021] [Indexed: 01/18/2023] Open
Abstract
Sepsis is defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. Sepsis-induced circulatory and cardiac dysfunction is associated with high mortality rates. Mitophagy, a specific form of autophagy, is excessively activated in lipopolysaccharide-induced myocardial injury. The present study investigated whether aldehyde dehydrogenase 2 (ALDH2) regulates mitophagy in sepsis-induced myocardial dysfunction. After lipopolysaccharide administration, cardiac dysfunction, inflammatory cell infiltration, biochemical indicators of myocardial cell injury, and cardiomyocyte apoptosis were ameliorated in mice by ALDH2 activation or overexpression. In contrast, cardiac dysfunction and cardiomyocyte apoptosis were exacerbated in mice followed ALDH2 inhibition. Moreover, ALDH2 activation or overexpression regulated mitophagy by suppressing the expression of phosphatase and tensin homolog-induced putative kinase 1 (PINK1)/Parkin, by preventing the accumulation of 4-hydroxy-trans-nonenal. Conversely, ALDH2 inhibition promoted the expression of LC3B by increasing 4-hydroxy-trans-2-nonenal accumulation. Consequently, ALDH2 may protect the heart from lipopolysaccharide-induced injury by suppressing PINK1/Parkin-dependent mitophagy.
Collapse
Affiliation(s)
- Wenqing Ji
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, China
| | - Tiantian Wan
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, China
| | - Fang Zhang
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, China
| | - Xiaomei Zhu
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, China
| | - Shubin Guo
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, China
| | - Xue Mei
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, China
| |
Collapse
|
49
|
Doblado L, Lueck C, Rey C, Samhan-Arias AK, Prieto I, Stacchiotti A, Monsalve M. Mitophagy in Human Diseases. Int J Mol Sci 2021; 22:3903. [PMID: 33918863 PMCID: PMC8069949 DOI: 10.3390/ijms22083903] [Citation(s) in RCA: 143] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/23/2021] [Accepted: 03/26/2021] [Indexed: 02/06/2023] Open
Abstract
Mitophagy is a selective autophagic process, essential for cellular homeostasis, that eliminates dysfunctional mitochondria. Activated by inner membrane depolarization, it plays an important role during development and is fundamental in highly differentiated post-mitotic cells that are highly dependent on aerobic metabolism, such as neurons, muscle cells, and hepatocytes. Both defective and excessive mitophagy have been proposed to contribute to age-related neurodegenerative diseases, such as Parkinson's and Alzheimer's diseases, metabolic diseases, vascular complications of diabetes, myocardial injury, muscle dystrophy, and liver disease, among others. Pharmacological or dietary interventions that restore mitophagy homeostasis and facilitate the elimination of irreversibly damaged mitochondria, thus, could serve as potential therapies in several chronic diseases. However, despite extraordinary advances in this field, mainly derived from in vitro and preclinical animal models, human applications based on the regulation of mitochondrial quality in patients have not yet been approved. In this review, we summarize the key selective mitochondrial autophagy pathways and their role in prevalent chronic human diseases and highlight the potential use of specific interventions.
Collapse
Affiliation(s)
- Laura Doblado
- Instituto de Investigaciones Biomédicas “Alberto Sols” (CSIC-UAM), Arturo Duperier 4, 28029 Madrid, Spain; (L.D.); (C.L.); (C.R.)
| | - Claudia Lueck
- Instituto de Investigaciones Biomédicas “Alberto Sols” (CSIC-UAM), Arturo Duperier 4, 28029 Madrid, Spain; (L.D.); (C.L.); (C.R.)
| | - Claudia Rey
- Instituto de Investigaciones Biomédicas “Alberto Sols” (CSIC-UAM), Arturo Duperier 4, 28029 Madrid, Spain; (L.D.); (C.L.); (C.R.)
| | - Alejandro K. Samhan-Arias
- Department of Biochemistry, Universidad Autónoma de Madrid e Instituto de Investigaciones Biomédicas “Alberto Sols” (CSIC-UAM), Arturo Duperier 4, 28029 Madrid, Spain;
| | - Ignacio Prieto
- Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Isaac Peral 42, 28015 Madrid, Spain;
| | - Alessandra Stacchiotti
- Department of Biomedical Sciences for Health, Universita’ Degli Studi di Milano, Via Mangiagalli 31, 20133 Milan, Italy
- U.O. Laboratorio di Morfologia Umana Applicata, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy
| | - Maria Monsalve
- Instituto de Investigaciones Biomédicas “Alberto Sols” (CSIC-UAM), Arturo Duperier 4, 28029 Madrid, Spain; (L.D.); (C.L.); (C.R.)
| |
Collapse
|
50
|
Xu X, Xie X, Zhang H, Wang P, Li G, Chen J, Chen G, Cao X, Xiong L, Peng F, Peng C. Water-soluble alkaloids extracted from Aconiti Radix lateralis praeparata protect against chronic heart failure in rats via a calcium signaling pathway. Biomed Pharmacother 2021; 135:111184. [PMID: 33418305 DOI: 10.1016/j.biopha.2020.111184] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/15/2020] [Accepted: 12/26/2020] [Indexed: 11/16/2022] Open
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Many studies have shown the beneficial effects of aconite water-soluble alkaloid extract (AWA) in experimental models of heart disease, which have been ascribed to the presence of aconine, hypaconine, talatisamine, fuziline, neoline, and songorine. This study evaluated the effects of a chemically characterized AWA by chemical content, evaluated its effects in suprarenal abdominal aortic coarctation surgery (AAC)-induced chronic heart failure (CHF) in rats, and revealed the underlying mechanisms of action by proteomics. METHODS Rats were distributed into different groups: sham, model, and AWA-treated groups (10, 20, and 40 mg/kg/day). Sham rats received surgery without AAC, whereas model rats an AWA-treated groups underwent AAC surgery. after 8 weeks, the treatment group was fed AWA for 4 weeks, and body weight was assessed weekly. At the end of the treatment, heart function was tested by echocardiography. AAC-induced chronic heart failure, including myocardial fibrosis, cardiomyocyte hypertrophy, and apoptosis, was evaluated in heart tissue and plasma by RT-qPCR, ELISA, hematoxylin and eosin (H&E) staining, Masson's trichrome staining, TUNEL staining, and immunofluorescence staining of α-SMA, Col Ⅰ, and Col Ⅲ. Then, a proteomics approach was used to explore the underlying mechanisms of action of AWA in chronic heart failure. RESULTS AWA administration reduced body weight gain, myocardial fibrosis, cardiomyocyte hypertrophy, and apoptosis, and rats showed improvement in cardiac function compared to model group. The extract significantly ameliorated the AAC-induced altered expression of heart failure markers such as ANP, NT-proBNP, and β-MHC, as well as fibrosis, hypertrophy markers MMP-2 and MMP-9, and other heart failure-related factors including plasma levels of TNF-α and IL-6. Furthermore, the extract reduced the protein expression of α-SMA, Col Ⅰ, and Col Ⅲ in the left ventricular (LV), thus inhibiting the LV remodeling associated with CHF. In addition, proteomics characterization of differentially expressed proteins showed that AWA administration inhibited left ventricular remodeling in CHF rats via a calcium signaling pathway, and reversed the expression of RyR2 and SERCA2a. CONCLUSIONS AWA extract exerts beneficial effects in an AAC-induced CHF model in rats, which was associated with an improvement in LV function, hypertrophy, fibrosis, and apoptotic status. These effects may be related to the regulation of calcium signaling by the altered expression of RyR2 and SERCA2a.
Collapse
MESH Headings
- Aconitum/chemistry
- Animals
- Apoptosis/drug effects
- Calcium Signaling/drug effects
- Cardiovascular Agents/isolation & purification
- Cardiovascular Agents/pharmacology
- Chronic Disease
- Disease Models, Animal
- Fibrosis
- Heart Failure/drug therapy
- Heart Failure/metabolism
- Heart Failure/pathology
- Heart Failure/physiopathology
- Hypertrophy, Left Ventricular/drug therapy
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/pathology
- Hypertrophy, Left Ventricular/physiopathology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Plant Extracts/isolation & purification
- Plant Extracts/pharmacology
- Rats, Sprague-Dawley
- Ryanodine Receptor Calcium Release Channel/metabolism
- Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
- Solubility
- Solvents/chemistry
- Ventricular Dysfunction, Left/drug therapy
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/pathology
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Function, Left/drug effects
- Ventricular Remodeling/drug effects
- Water/chemistry
- Rats
Collapse
Affiliation(s)
- Xin Xu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu 611137, China; Key Laboratory of the Ministry of Education for Standardization of Chinese Medicine Co-founded by Sichuan Province and MOST, Chengdu 611137, China
| | - Xiaofang Xie
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu 611137, China; Key Laboratory of the Ministry of Education for Standardization of Chinese Medicine Co-founded by Sichuan Province and MOST, Chengdu 611137, China
| | - Huiqiong Zhang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu 611137, China; Key Laboratory of the Ministry of Education for Standardization of Chinese Medicine Co-founded by Sichuan Province and MOST, Chengdu 611137, China
| | - Pei Wang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Gangmin Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu 611137, China; Key Laboratory of the Ministry of Education for Standardization of Chinese Medicine Co-founded by Sichuan Province and MOST, Chengdu 611137, China
| | - Junren Chen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu 611137, China; Key Laboratory of the Ministry of Education for Standardization of Chinese Medicine Co-founded by Sichuan Province and MOST, Chengdu 611137, China
| | - Guanru Chen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu 611137, China; Key Laboratory of the Ministry of Education for Standardization of Chinese Medicine Co-founded by Sichuan Province and MOST, Chengdu 611137, China
| | - Xiaoyu Cao
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Liang Xiong
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu 611137, China; Key Laboratory of the Ministry of Education for Standardization of Chinese Medicine Co-founded by Sichuan Province and MOST, Chengdu 611137, China
| | - Fu Peng
- West China School of Pharmacy, Sichuan University, Chengdu 611137, China.
| | - Cheng Peng
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu 611137, China; Key Laboratory of the Ministry of Education for Standardization of Chinese Medicine Co-founded by Sichuan Province and MOST, Chengdu 611137, China.
| |
Collapse
|