1
|
Pan J, Wang J, Lei Z, Wang H, Zeng N, Zou J, Zhang X, Sun J, Guo D, Luan F, Shi Y. Therapeutic Potential of Chinese Herbal Medicine and Underlying Mechanism for the Treatment of Myocardial Infarction. Phytother Res 2025; 39:189-232. [PMID: 39523856 DOI: 10.1002/ptr.8368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/23/2024] [Accepted: 08/23/2024] [Indexed: 11/16/2024]
Abstract
Myocardial infarction (MI) is a prevalent disease with high mortality rates worldwide. The course of MI is intricate and variable, necessitating personalized treatment strategies based on different mechanisms. However, variety of postoperative complications and rejections, such as heart failure, arrhythmias, cardiac rupture, and left ventricular thrombus, contribute to a poor prognosis. Despite the inclusion of antiplatelet agents and statins in the conventional treatment regimen, their clinical applicability is constrained by potential adverse effects and limited efficacy. Additionally, the mechanisms leading to MI are complex and diverse. Therefore, the development of novel compounds for MI treatment. The use of traditional Chinese medicine (TCM) in the prevention and treatment of cardiovascular diseases, including MI, is grounded in its profound historical background, comprehensive theoretical system, and accumulated knowledge. An increasing number of contemporary evidence-based medical studies have demonstrated that TCM plays a significant role in alleviating symptoms and improving the quality of life for MI patients. Chinese herbal formulations and active ingredients can intervene in the pathological process of MI through key factors such as inflammation, oxidative stress, apoptosis, ferroptosis, pyroptosis, myocardial fibrosis, angiogenesis, and autophagy. This article critically reviews existing herbal formulations from an evidence-based medicine perspective, evaluating their research status and potential clinical applications. Additionally, it explores recent advancements in the use of herbal medicines and their components for the prevention and treatment of MI, offering detailed insights into their mechanisms of action.
Collapse
Affiliation(s)
- Jiaojiao Pan
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| | - Jinhui Wang
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| | - Ziwen Lei
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| | - He Wang
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| | - Nan Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Junbo Zou
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| | - Xiaofei Zhang
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| | - Jing Sun
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| | - Dongyan Guo
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| | - Fei Luan
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| | - Yajun Shi
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| |
Collapse
|
2
|
Alhegaili AS, Bafail DA, Bawahab AA, Alsubaie N, Abd-Elhakim YM, Mohamed AAR, Khamis T, Khalifa NE, Elhamouly M, Dahran N, El Shetry ES. The interplay of oxidative stress, apoptotic signaling, and impaired mitochondrial function in the pyrethroid-induced cardiac injury: Alleviative role of curcumin-loaded chitosan nanoparticle. Food Chem Toxicol 2024; 194:115095. [PMID: 39515510 DOI: 10.1016/j.fct.2024.115095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
This study assessed the consequence of exposure to a pyrethroid insecticide, fenpropathrin (FPN), on the heart and the probable underlying mechanisms in rats. Moreover, the probable protective effect of curcumin-loaded chitosan nanoparticles (CMN-CNP) was evaluated. Forty male Sprague Dawley rats were distributed into four groups orally given corn oil, CMN-CNP (50 mg/kg b.wt), FPN (15 mg/kg b.wt), or CMN-CNP + FPN for 60 days. The results revealed that FPN exposure increased serum cardiac damage indicators. In addition, a substantial increase in the reactive oxygen species and malondialdehyde content but reduced enzymatic and non-enzymatic antioxidants and altered architecture was recorded in the cardiac tissue of FPN-exposed rats. Additionally, a significant down-regulation of expression of the mitochondrial complexes I-V, mitochondrial dynamics, and antioxidants-related genes but up-regulation of apoptosis-related genes was detected in the FPN-exposed group. Immunofluorescence analyses revealed higher amounts of the harmful protein 4-hydroxynonenal in the heart tissue of FPN-exposed rats. Nevertheless, the earlier disturbances were significantly rescued in the FPN + CMN-CNP treated group. Conclusively, our findings reported the cardiotoxic activity of FPN and the involvement of several mitochondrial imbalances as a probable underlying mechanism. Also, the study findings proved the efficacy of CMN-CNP in combating FPN cardiotoxic effects.
Collapse
Affiliation(s)
- Alaa S Alhegaili
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Duaa Abdullah Bafail
- Department of Clinical Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ahmed Abdulwahab Bawahab
- Department of Basic Medical Sciences, College of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Nawal Alsubaie
- Department of Pharmacy Practice, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia.
| | - Yasmina M Abd-Elhakim
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Amany Abdel-Rahman Mohamed
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Tarek Khamis
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt; Laboratory of Biotechnology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Norhan E Khalifa
- Department of Physiology, Faculty of Veterinary Medicine, Matrouh University, Matrouh 51511, Egypt
| | - Moustafa Elhamouly
- Department of Histology and Cytology Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Egypt
| | - Naief Dahran
- Department of Basic Medical Sciences, College of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Eman S El Shetry
- Department of Anatomy, College of Medicine, University of Hail, Hail, Saudi Arabia; Department of Human Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
3
|
Guo Q, Wang J, Ni C, Pan J, Zou J, Shi Y, Sun J, Zhang X, Wang D, Luan F. Research progress on the natural products in the intervention of myocardial infarction. Front Pharmacol 2024; 15:1445349. [PMID: 39239656 PMCID: PMC11374734 DOI: 10.3389/fphar.2024.1445349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/05/2024] [Indexed: 09/07/2024] Open
Abstract
Coronary heart disease is a prevalent cardiovascular ailment globally, with myocardial infarction (MI) being one of its most severe manifestations. The morbidity and mortality of MI are escalating, showing an increasing trend among younger, highly educated individuals, thereby posing a serious threat to public health. Currently, thrombolysis, percutaneous coronary intervention, and coronary artery bypass grafting are the primary clinical treatments for MI. Although these methods significantly reduce patient mortality, complications often result in poor prognoses. Due to limitations in chemical synthetic drug research, the focus has shifted towards developing herbs based on natural substances. Natural medicines represent a novel approach for safer and more effective MI management and treatment. They can control multiple pathogenic variables by targeting various pathways and systems. This paper investigates the molecular mechanisms of MI and evaluates the application of natural products and medicinal plants in MI treatment over the past 5 years, demonstrating their specific good therapeutic potential and superior tolerance. These natural therapies have been shown to mitigate myocardial cell damage caused by MI through mechanisms such as oxidative stress, inflammation, apoptosis, angiogenesis, myocardial fibrosis, autophagy, endoplasmic reticulum stress, mitophagy, and pyroptosis. This review offers the latest insights into the application of natural products and medicinal plants in MI treatment, elucidating their mechanisms of action and serving as an important reference for MI prevention.
Collapse
Affiliation(s)
- Qiuting Guo
- College of Pharmacy, Xianyang Polytechnic Institute, Xianyang, China
| | - Jinhui Wang
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi, China
| | - Caixia Ni
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, Sichuan, China
| | - Jiaojiao Pan
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi, China
| | - Junbo Zou
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi, China
| | - Yajun Shi
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi, China
| | - Jing Sun
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi, China
| | - Xiaofei Zhang
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi, China
| | - Deng Wang
- Department of Pharmacy, Xi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, Shaanxi, China
| | - Fei Luan
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi, China
| |
Collapse
|
4
|
Shorthill SK, Jones TLM, Woulfe KC, Cherrington BD, Bruns DR. The influence of estrogen on myocardial post-translational modifications and cardiac function in women. Can J Physiol Pharmacol 2024; 102:452-464. [PMID: 38266237 DOI: 10.1139/cjpp-2023-0412] [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] [Indexed: 01/26/2024]
Abstract
The lifetime risk of heart failure (HF) is comparable in men and women; nevertheless, disparities exist in our understanding of how HF differs between sexes. Several differences in cardiac physiology exist between men and women including the propensity to develop specific HF phenotypes. Men are more likely to be diagnosed with HF failure with reduced ejection fraction, while women have a greater propensity to develop HF with preserved ejection fraction. The mechanisms responsible for these differences remain unclear. Post-translational modifications (PTMs) of myofilament proteins likely contribute to these sex-specific propensities. The role of PTMs in heart disease is an expanding field with immense potential therapeutic targets. However, numerous PTMs remain underexplored, particularly in the context of the female heart. Estrogen, a key gonadal hormone, cardioprotective in pre-menopausal women and its loss with menopause likely contributes to disease in aging women. However, how estrogen regulates PTMs to contribute to HF development is not fully clear. This review outlines key sex differences in HF along with characterizing the contributions of novel myocardial PTMs in cardiac physiology and their regulation by estrogen. Collectively, we highlight the necessity for further investigation into women's heart health and the distinctive mechanisms distinguishing women from men.
Collapse
Affiliation(s)
| | - Timothy L M Jones
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kathleen C Woulfe
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Brian D Cherrington
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY, USA
| | - Danielle R Bruns
- Division of Kinesiology and Health, University of Wyoming, Laramie, WY, USA
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY, USA
| |
Collapse
|
5
|
Lv X, Liu B, Su X, Tian X, Wang H. Unlocking cardioprotection: iPSC exosomes deliver Nec-1 to target PARP1/AIFM1 axis, alleviating HF oxidative stress and mitochondrial dysfunction. J Transl Med 2024; 22:681. [PMID: 39061056 PMCID: PMC11282728 DOI: 10.1186/s12967-024-05204-9] [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: 01/01/2024] [Accepted: 04/15/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Heart failure (HF) is characterized by oxidative stress and mitochondrial dysfunction. This study investigates the therapeutic potential of Necrostatin-1 (Nec-1) delivered through exosomes derived from induced pluripotent stem cells (iPSCs) to address these pathologies in HF. METHODS An HF rat model was established, and comprehensive assessments were performed using echocardiography, hemodynamics, and ventricular mass index measurements. iPSCs were used to isolate exosomes, loaded with Nec-1, and characterized for efficient delivery into cardiomyocytes. The interaction between Nec-1-loaded exosomes (Nec-1-Exos), poly (ADP-ribose) polymerase 1 (PARP1), and apoptosis-inducing factor mitochondria-associated 1 (AIFM1) was explored. Gain-of-function experiments assessed changes in cardiomyocyte parameters, and histological analyses were conducted on myocardial tissues. RESULTS Cardiomyocytes successfully internalized Nec-1-loaded exosomes, leading to downregulation of PARP1, inhibition of AIFM1 nuclear translocation, increased ATP and superoxide dismutase levels, reduced reactive oxygen species and malonaldehyde levels, and restored mitochondrial membrane potential. Histological examinations confirmed the modulation of the PARP1/AIFM1 axis by Nec-1, mitigating HF. CONCLUSIONS iPSC-derived exosomes carrying Nec-1 attenuate oxidative stress and mitochondrial dysfunction in HF by targeting the PARP1/AIFM1 axis. This study proposes a promising therapeutic strategy for HF management and highlights the potential of exosome-mediated drug delivery.
Collapse
Affiliation(s)
- Xiaobing Lv
- Department of Cardiology, Jinan Central Hospital, Shandong University, No.105 Jiefang Road, Lixia District, Jinan, Shandong Province, 250013, P. R. China
- Department of Cardiology, the Affiliated Hospital of Qingdao University, Qingdao, 266000, P.R. China
| | - Boqin Liu
- Department of Cardiology, Qingdao Municipal Hospital (West Yard), Qingdao, 266000, P.R. China
| | - Xiaoting Su
- Department of Obstetric, the Affiliated Hospital of Qingdao University, Qingdao, 266000, P.R. China
| | - Xintao Tian
- Department of Emergency Internal Medicine, the Affiliated Hospital of Qingdao University, Qingdao, 266000, P.R. China
| | - Huating Wang
- Department of Cardiology, Jinan Central Hospital, Shandong University, No.105 Jiefang Road, Lixia District, Jinan, Shandong Province, 250013, P. R. China.
| |
Collapse
|
6
|
Chen S, Guan S, Yan Z, Ouyang F, Li S, Liu L, Zhong J. Role of RIPK3‑CaMKII‑mPTP signaling pathway‑mediated necroptosis in cardiovascular diseases (Review). Int J Mol Med 2023; 52:98. [PMID: 37654208 PMCID: PMC10495754 DOI: 10.3892/ijmm.2023.5301] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 07/27/2023] [Indexed: 09/02/2023] Open
Abstract
Necroptosis, which is distinct from apoptosis and necrosis, serves a crucial role in ontogeny and the maintenance of homeostasis. In the last decade, it has been demonstrated that the pathogenesis of cardiovascular diseases is also linked to necroptosis. Receptor interaction protein kinase (RIPK) 1, RIPK3 and mixed lineage kinase domain‑like protein serve vital roles in necroptosis. In addition to the aforementioned necroptosis‑related components, calcium/calmodulin‑dependent protein kinase II (CaMKII) has been identified as a novel substrate for RIPK3 that promotes the opening of the mitochondrial permeability transition pore (mPTP), and thus, mediates necroptosis of myocardial cells through the RIPK3‑CaMKII‑mPTP signaling pathway. The present review provides an overview of the current knowledge of the RIPK3‑CaMKII‑mPTP‑mediated necroptosis signaling pathway in cardiovascular diseases, focusing on the role of the RIPK3‑CaMKII‑mPTP signaling pathway in acute myocardial infarction, ischemia‑reperfusion injury, heart failure, abdominal aortic aneurysm, atherosclerosis, diabetic cardiomyopathy, hypertrophic cardiomyopathy, atrial fibrillation, and the cardiotoxicity associated with antitumor drugs and other chemicals. Finally, the present review discusses the research status of drugs targeting the RIPK3‑CaMKII‑mPTP signaling pathway.
Collapse
Affiliation(s)
- Sheng Chen
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong 528308, P.R. China
| | - Senhong Guan
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong 528308, P.R. China
| | - Zhaohan Yan
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong 528308, P.R. China
| | - Fengshan Ouyang
- Department of Rehabilitation Medicine, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong 528308, P.R. China
| | - Shuhuan Li
- Department of Pediatrics, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong 528308, P.R. China
| | - Lanyuan Liu
- Department of Ultrasound Medicine, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong 528308, P.R. China
| | - Jiankai Zhong
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong 528308, P.R. China
| |
Collapse
|
7
|
Liang Y, Huang Y, Shao R, Xiao F, Lin F, Dai H, Pan L. Propofol produces neurotoxicity by inducing mitochondrial apoptosis. Exp Ther Med 2022; 24:630. [PMID: 36160898 PMCID: PMC9468839 DOI: 10.3892/etm.2022.11567] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 05/10/2022] [Indexed: 11/13/2022] Open
Abstract
Propofol is a fast and short-acting intravenous anesthetic widely used in clinical anesthesia and intensive care unit sedation. However, its use can cause abnormal effects on the central nervous system. Thus, the purpose of this study was to investigate the mechanism of propofol on primary hippocampal neuron injury. In addition, we aimed to determine whether a correlation exists between propofol and mitochondrial apoptosis-induced neurotoxicity. Hippocampal neurons cultured for 4 days were exposed to different drugs. The treatment groups were divided according to drug exposure into propofol, a rotenone inhibitor, and a coenzyme Q10 agonist groups. The final concentrations of propofol were 1, 10 and 100 µM. The content of ATP and reactive oxygen species (ROS) in the neurons of each group were detected using commercial kits in the culture supernatant after 3 h of drug exposure. Western blotting was used to analyze the expression of apoptosis-related proteins. The JC-1 kit was used to detect the mitochondrial membrane potential. The results revealed that, compared with the non-propofol treatment groups, the expression of apoptosis-related proteins, ATP content, and mitochondrial membrane potential were significantly decreased while the ROS content was markedly increased in the propofol treatment group. In conclusion, propofol treatment promoted damage to hippocampal neuronal mitochondria in a dose-dependent manner. This damage may lead to neuronal apoptosis and neurotoxicity by inducing the inhibition of mitochondrial respiratory chain complex I.
Collapse
Affiliation(s)
- Yubing Liang
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Yu Huang
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Rongge Shao
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Fei Xiao
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Fei Lin
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Huijun Dai
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Linghui Pan
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| |
Collapse
|
8
|
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
|
9
|
Dabravolski SA, Sadykhov NK, Kartuesov AG, Borisov EE, Sukhorukov VN, Orekhov AN. The Role of Mitochondrial Abnormalities in Diabetic Cardiomyopathy. Int J Mol Sci 2022; 23:ijms23147863. [PMID: 35887211 PMCID: PMC9321738 DOI: 10.3390/ijms23147863] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/14/2022] [Accepted: 07/15/2022] [Indexed: 02/06/2023] Open
Abstract
Diabetic cardiomyopathy (DCM) is defined as the presence in diabetic patients of abnormal cardiac structure and performance (such as left ventricular hypertrophy, fibrosis, and arrhythmia) in the absence of other cardiac risk factors (such as hypertension or coronary artery disease). Although the pathogenesis of DCM remains unclear currently, mitochondrial structural and functional dysfunctions are recognised as a central player in the DCM development. In this review, we focus on the role of mitochondrial dynamics, biogenesis and mitophagy, Ca2+ metabolism and bioenergetics in the DCM development and progression. Based on the crucial role of mitochondria in DCM, application of mitochondria-targeting therapies could be effective strategies to slow down the progression of the disease.
Collapse
Affiliation(s)
- Siarhei A. Dabravolski
- Department of Clinical Diagnostics, Vitebsk State Academy of Veterinary Medicine [UO VGAVM], 7/11 Dovatora Str., 210026 Vitebsk, Belarus
- Correspondence:
| | - Nikolay K. Sadykhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia; (N.K.S.); (A.G.K.)
| | - Andrey G. Kartuesov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia; (N.K.S.); (A.G.K.)
| | - Evgeny E. Borisov
- Petrovsky National Research Centre of Surgery, 2, Abrikosovsky Lane, 119991 Moscow, Russia; (E.E.B.); (V.N.S.)
| | - Vasily N. Sukhorukov
- Petrovsky National Research Centre of Surgery, 2, Abrikosovsky Lane, 119991 Moscow, Russia; (E.E.B.); (V.N.S.)
- Institute for Atherosclerosis Research, Osennyaya 4-1-207, 121609 Moscow, Russia;
| | - Alexander N. Orekhov
- Institute for Atherosclerosis Research, Osennyaya 4-1-207, 121609 Moscow, Russia;
| |
Collapse
|
10
|
Luo Y, Apaijai N, Liao S, Maneechote C, Chunchai T, Arunsak B, Benjanuwattra J, Yanpiset P, Chattipakorn SC, Chattipakorn N. Therapeutic potentials of cell death inhibitors in rats with cardiac ischaemia/reperfusion injury. J Cell Mol Med 2022; 26:2462-2476. [PMID: 35315192 PMCID: PMC8995446 DOI: 10.1111/jcmm.17275] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/16/2022] [Accepted: 02/28/2022] [Indexed: 12/13/2022] Open
Abstract
Growing evidence demonstrated that cell death pathways including ferroptosis, apoptosis and necroptosis contribute to cardiac ischaemia/reperfusion (I/R) injury. We hypothesized that ferroptosis, apoptosis and necroptosis contribute differently to myocardial damage during acute cardiac I/R injury. Rats underwent cardiac I/R or sham operation. I/R-operated rats were divided into 4 groups: vehicle, apoptosis (Z-vad), ferroptosis (Fer-1) and necroptosis (Nec-1) inhibition. Rats in each cell death inhibitor group were subdivided into 3 different dose regimens: low, medium and high. Infarct size, left ventricular (LV) function, arrhythmias and molecular mechanism were investigated. Cardiac I/R caused myocardial infarction, LV dysfunction, arrhythmias, mitochondrial dysfunction, mitochondrial dynamic imbalance, inflammation, apoptosis and ferroptosis. Infarct size, LV dysfunction, mitochondrial dysfunction, apoptosis and ferroptosis were all reduced to a similar extent in rats treated with Z-vad (low and medium doses) or Fer-1 (medium and high doses). Fer-1 treatment also reduced mitochondrial dynamic imbalance and inflammation. No evidence of necroptosis was found in association with acute I/R injury, therefore Nec-1 treatment could not be assessed. Apoptosis and ferroptosis, not necroptosis, contributed to myocardial damage in acute I/R injury. Inhibitors of these 2 pathways provided effective cardioprotection in rats with I/R injury though modulation of mitochondrial function and attenuated apoptosis and ferroptosis.
Collapse
Affiliation(s)
- Ying Luo
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nattayaporn Apaijai
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Suchan Liao
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Chayodom Maneechote
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Titikorn Chunchai
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Busarin Arunsak
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Juthipong Benjanuwattra
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Panat Yanpiset
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.,Department of Oral Biology and Diagnostic Science, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
11
|
Zhuo Y, Yuan R, Chen X, He J, Chen Y, Zhang C, Sun K, Yang S, Liu Z, Gao H. Tanshinone I exerts cardiovascular protective effects in vivo and in vitro through inhibiting necroptosis via Akt/Nrf2 signaling pathway. Chin Med 2021; 16:48. [PMID: 34183021 PMCID: PMC8240219 DOI: 10.1186/s13020-021-00458-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 06/16/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Tanshinone I (TI) is a primary component of Salvia miltiorrhiza Bunge (Danshen), which confers a favorable role in a variety of pharmacological activities including cardiovascular protection. However, the exact mechanism of the cardiovascular protection activity of TI remains to be illustrated. In this study, the cardiovascular protective effect and its mechanism of TI were investigated. METHODS In this study, tert-butyl hydroperoxide (t-BHP)-stimulated H9c2 cells model was employed to investigate the protective effect in vitro. The cell viability was determined by 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl tetrazolium bromide (MTT) assay and lactate dehydrogenase (LDH) kit. The reactive-oxygen-species (ROS) level and mitochondrial membrane potential (MMP) were investigated by the flow cytometry and JC-1 assay, respectively. While in vivo experiment, the cardiovascular protective effect of TI was determined by using myocardial ischemia-reperfusion (MI/R) model including hematoxylin-eosin (H&E) staining assay and determination of superoxide dismutase (SOD) and malondialdehyde (MDA). Tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) release were detected by Enzyme-linked immunosorbent assay (ELISA). Receptor interacting protein kinase 1 (RIP1), receptor interacting protein kinase 3 (RIP3), receptor interacting protein kinase 3 (MLKL), protein kinase B (Akt), Nuclear factor erythroid 2 related factor 2 (Nrf2), Heme oxygenase-1 (HO-1) and NAD(P)H: quinone oxidoreductase-1 (NQO-1) were determined by western blotting. RESULTS Our data demonstrated that TI pretreatment attenuated t-BHP and MI/R injury-induced necroptosis by inhibiting the expression of p-RIP1, p-RIP3, and p-MLKL. TI activated the Akt/Nrf2 pathway to promote the expression of antioxidant-related proteins such as phosphorylation of Akt, nuclear factor erythroid 2 related factor 2 (Nrf2), quinone oxidoreductase-1 (NQO-1) and heme oxygenase-1 (HO-1) expression in t-BHP-stimulated H9c2 cells. TI relieved oxidative stress by mitigating ROS generation and reversing MMP loss. In vivo experiment, TI made electrocardiograph (ECG) recovery better and lessened the degree of myocardial tissue damage. The counts of white blood cell (WBC), neutrophil (Neu), lymphocyte (Lym), and the release of TNF-α and IL-6 were reversed by TI treatment. SOD level was increased, while MDA level was decreased by TI treatment. CONCLUSION Collectively, our findings indicated that TI exerted cardiovascular protective activities in vitro and in vivo through suppressing RIP1/RIP3/MLKL and activating Akt/Nrf2 signaling pathways, which could be developed into a cardiovascular protective agent.
Collapse
Affiliation(s)
- Youqiong Zhuo
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530200, China
| | - Renyikun Yuan
- State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, China
| | - Xinxin Chen
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530200, China
| | - Jia He
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530200, China
| | - Yangling Chen
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530200, China
| | - Chenwei Zhang
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530200, China
| | - Kaili Sun
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530200, China
| | - Shilin Yang
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530200, China
| | - Zhenjie Liu
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530200, China
| | - Hongwei Gao
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China.
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530200, China.
| |
Collapse
|
12
|
Yoshimura C, Nagasaka A, Kurose H, Nakaya M. Efferocytosis during myocardial infarction. J Biochem 2021; 168:1-6. [PMID: 32302392 DOI: 10.1093/jb/mvaa051] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 04/07/2020] [Indexed: 02/07/2023] Open
Abstract
Myocardial infarction is one of the major causes of death worldwide. Many heart cells die during myocardial infarction through various processes such as necrosis, apoptosis, necroptosis, autophagy-related cell death, pyroptosis and ferroptosis. These dead cells in infarcted hearts expose the so-called 'eat-me' signals, such as phosphatidylserine, on their surfaces, enhancing their removal by professional and non-professional phagocytes. Clearance of dead cells by phagocytes in the diseased hearts plays a crucial role in the pathology of myocardial infarction by inhibiting the inflammatory responses caused by the leakage of contents from dead cells. This review focuses on the rapidly growing understanding of the molecular mechanisms of dead cell phagocytosis, termed efferocytosis, during myocardial infarction, which contributes to the pathophysiology of myocardial infarction.
Collapse
Affiliation(s)
- Chikashi Yoshimura
- Department of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Akiomi Nagasaka
- Department of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Hitoshi Kurose
- Department of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Michio Nakaya
- Department of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.,AMED-PRIME, Japan Agency for Medical Research and Development, Tokyo 100-0004, Japan
| |
Collapse
|
13
|
da Silva JS, Montagnoli TL, Rocha BS, Tacco MLCA, Marinho SCP, Zapata-Sudo G. Estrogen Receptors: Therapeutic Perspectives for the Treatment of Cardiac Dysfunction after Myocardial Infarction. Int J Mol Sci 2021; 22:E525. [PMID: 33430254 PMCID: PMC7825655 DOI: 10.3390/ijms22020525] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/24/2020] [Accepted: 12/28/2020] [Indexed: 02/07/2023] Open
Abstract
Estrogen receptors (ER) mediate functions beyond their endocrine roles, as modulation of cardiovascular, renal, and immune systems through anti-inflammatory and anti-apoptotic effects, preventing necrosis of cardiomyocytes and endothelial cells, and attenuating cardiac hypertrophy. Estradiol (E2) prevents cardiac dysfunction, increases nitric oxide synthesis, and reduces the proliferation of vascular cells, yielding protective effects, regardless of gender. Such actions are mediated by ER (ER-alpha (ERα), ER-beta (ERβ), or G protein-coupled ER (GPER)) through genomic or non-genomic pathways, which regulate cardiovascular function and prevent tissue remodeling. Despite the extensive knowledge on the cardioprotective effects of estrogen, clinical studies conducted on myocardial infarction (MI) and cardiovascular diseases still include favorable and unfavorable profiles. The purpose of this review is to provide up-to-date information regarding molecular, preclinical, and clinical aspects of cardiovascular E2 effects and ER modulation as a potential therapeutic target for the treatment of MI-induced cardiac dysfunction.
Collapse
Affiliation(s)
- Jaqueline S. da Silva
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (J.S.d.S.); (T.L.M.); (B.S.R.); (M.L.C.A.T.); (S.C.P.M.)
| | - Tadeu L. Montagnoli
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (J.S.d.S.); (T.L.M.); (B.S.R.); (M.L.C.A.T.); (S.C.P.M.)
| | - Bruna S. Rocha
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (J.S.d.S.); (T.L.M.); (B.S.R.); (M.L.C.A.T.); (S.C.P.M.)
| | - Matheus L. C. A. Tacco
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (J.S.d.S.); (T.L.M.); (B.S.R.); (M.L.C.A.T.); (S.C.P.M.)
| | - Sophia C. P. Marinho
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (J.S.d.S.); (T.L.M.); (B.S.R.); (M.L.C.A.T.); (S.C.P.M.)
| | - Gisele Zapata-Sudo
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (J.S.d.S.); (T.L.M.); (B.S.R.); (M.L.C.A.T.); (S.C.P.M.)
- Instituto de Cardiologia Edson Saad, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| |
Collapse
|
14
|
Stamenkovska M, Hadzi-Petrushev N, Nikodinovski A, Gagov H, Atanasova-Panchevska N, Mitrokhin V, Kamkin A, Mladenov M. Application of curcumine and its derivatives in the treatment of cardiovascular diseases: a review. INTERNATIONAL JOURNAL OF FOOD PROPERTIES 2021. [DOI: 10.1080/10942912.2021.1977655] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Mimoza Stamenkovska
- Institute of Biology, Faculty of Natural Science and Mathematics, Ss Cyril and Methodius University, Skopje, North Macedonia
- Faculty of Dental Medicine, European University Skopje, Skopje, North Macedonia
| | - Nikola Hadzi-Petrushev
- Institute of Biology, Faculty of Natural Science and Mathematics, Ss Cyril and Methodius University, Skopje, North Macedonia
| | - Aleksandar Nikodinovski
- Institut for Preclinical and Clinical Pharmacology and Toxicology, Medical Faculty, Ss Cyril and Methodius University, Skopje, North Macedonia
| | - Hristo Gagov
- Faculty of Biology, St. Kliment Ohridski University, Sofia, Bulgaria
| | - Natalija Atanasova-Panchevska
- Institute of Biology, Faculty of Natural Science and Mathematics, Ss Cyril and Methodius University, Skopje, North Macedonia
| | - Vadim Mitrokhin
- Department of Fundamental and Applied Physiology, Russian National Research Medical University, Moscow, Russia
| | - Andre Kamkin
- Department of Fundamental and Applied Physiology, Russian National Research Medical University, Moscow, Russia
| | - Mitko Mladenov
- Institute of Biology, Faculty of Natural Science and Mathematics, Ss Cyril and Methodius University, Skopje, North Macedonia
- Department of Fundamental and Applied Physiology, Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
15
|
Abstract
Acute myocardial infarction (AMI) is associated with the induction of a sterile inflammatory response that leads to further injury. The NACHT, leucine-rich repeat, and pyrin domain-containing protein 3 (NLRP3) inflammasome is a macromolecular structure responsible for the inflammatory response to injury or infection. NLRP3 can sense intracellular danger signals, such as ischemia and extracellular or intracellular alarmins during tissue injury. The NLRP3 inflammasome is primed and triggered by locally released damage-associated molecular patterns and amplifies the inflammatory response and cell death through caspase-1 activation. Here, we examine the scientific evidence supporting a role for NLRP3 in AMI and the available strategies to inhibit the effects of the inflammasome. Our focus is on the beneficial effects seen in experimental models of AMI in preclinical animal models and the initial results of clinical trials.
Collapse
|
16
|
Perrino C, Ferdinandy P, Bøtker HE, Brundel BJJM, Collins P, Davidson SM, den Ruijter HM, Engel FB, Gerdts E, Girao H, Gyöngyösi M, Hausenloy DJ, Lecour S, Madonna R, Marber M, Murphy E, Pesce M, Regitz-Zagrosek V, Sluijter JPG, Steffens S, Gollmann-Tepeköylü C, Van Laake LW, Van Linthout S, Schulz R, Ytrehus K. Improving translational research in sex-specific effects of comorbidities and risk factors in ischaemic heart disease and cardioprotection: position paper and recommendations of the ESC Working Group on Cellular Biology of the Heart. Cardiovasc Res 2020; 117:367-385. [PMID: 32484892 DOI: 10.1093/cvr/cvaa155] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 03/29/2020] [Accepted: 05/27/2020] [Indexed: 12/17/2022] Open
Abstract
Ischaemic heart disease (IHD) is a complex disorder and a leading cause of death and morbidity in both men and women. Sex, however, affects several aspects of IHD, including pathophysiology, incidence, clinical presentation, diagnosis as well as treatment and outcome. Several diseases or risk factors frequently associated with IHD can modify cellular signalling cascades, thus affecting ischaemia/reperfusion injury as well as responses to cardioprotective interventions. Importantly, the prevalence and impact of risk factors and several comorbidities differ between males and females, and their effects on IHD development and prognosis might differ according to sex. The cellular and molecular mechanisms underlying these differences are still poorly understood, and their identification might have important translational implications in the prediction or prevention of risk of IHD in men and women. Despite this, most experimental studies on IHD are still undertaken in animal models in the absence of risk factors and comorbidities, and assessment of potential sex-specific differences are largely missing. This ESC WG Position Paper will discuss: (i) the importance of sex as a biological variable in cardiovascular research, (ii) major biological mechanisms underlying sex-related differences relevant to IHD risk factors and comorbidities, (iii) prospects and pitfalls of preclinical models to investigate these associations, and finally (iv) will provide recommendations to guide future research. Although gender differences also affect IHD risk in the clinical setting, they will not be discussed in detail here.
Collapse
Affiliation(s)
- Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Via Pansini 5, 80131 Naples, Italy
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary.,Pharmahungary Group, Hajnoczy str. 6., H-6722 Szeged, Hungary
| | - Hans E Bøtker
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Blvd. 161, 8200 Aarhus, Denmark
| | - Bianca J J M Brundel
- Department of Physiology, Amsterdam UMC, Vrije Universiteit, Amsterdam Cardiovascular Sciences, De Boelelaan 1117, Amsterdam, 1108 HV, the Netherlands
| | - Peter Collins
- Imperial College, Faculty of Medicine, National Heart & Lung Institute, South Kensington Campus, London SW7 2AZ, UK.,Royal Brompton Hospital, Sydney St, Chelsea, London SW3 6NP, UK
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, WC1E 6HX London, UK
| | - Hester M den Ruijter
- Experimental Cardiology Laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Muscle Research Center Erlangen (MURCE), Schwabachanlage 12, 91054 Erlangen, Germany
| | - Eva Gerdts
- Department for Clinical Science, University of Bergen, PO Box 7804, 5020 Bergen, Norway
| | - Henrique Girao
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Azinhaga Santa Comba, Celas, 3000-548 Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, and Clinical Academic Centre of Coimbra (CACC), 3000-548 Coimbra, Portugal
| | - Mariann Gyöngyösi
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, A-1090 Vienna, Austria
| | - Derek J Hausenloy
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, 169857, Singapore.,National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, 169609, Singapore.,Yong Loo Lin School of Medicine, National University Singapore, 1E Kent Ridge Road, 119228, Singapore.,The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London WC1E 6HX, UK.,Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, 500, Lioufeng Rd., Wufeng, Taichung 41354, Taiwan
| | - Sandrine Lecour
- Hatter Institute for Cardiovascular Research in Africa, Faculty of Health Sciences, Chris Barnard Building, University of Cape Town, Private Bag X3 7935 Observatory, Cape Town, South Africa
| | - Rosalinda Madonna
- Institute of Cardiology, University of Pisa, Lungarno Antonio Pacinotti 43, 56126 Pisa, Italy.,Department of Internal Medicine, University of Texas Medical School in Houston, 6410 Fannin St #1014, Houston, TX 77030, USA
| | - Michael Marber
- King's College London BHF Centre, The Rayne Institute, St Thomas' Hospital, Westminster Bridge Road, London SE1 7EH, UK
| | - Elizabeth Murphy
- Laboratory of Cardiac Physiology, Cardiovascular Branch, NHLBI, NIH, 10 Center Drive, Bethesda, MD 20892, USA
| | - Maurizio Pesce
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS Via Parea, 4, I-20138 Milan, Italy
| | - Vera Regitz-Zagrosek
- Berlin Institute of Gender in Medicine, Center for Cardiovascular Research, DZHK, partner site Berlin, Geschäftsstelle Potsdamer Str. 58, 10785 Berlin, Germany.,University of Zürich, Rämistrasse 71, 8006 Zürich, Germany
| | - Joost P G Sluijter
- Experimental Cardiology Laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 8, 3584 CS Utrecht, the Netherlands.,Circulatory Health Laboratory, Regenerative Medicine Center, University Medical Center Utrecht, Utrecht University, Heidelberglaan 8, 3584 CS Utrecht, the Netherlands
| | - Sabine Steffens
- Institute for Cardiovascular Prevention and German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Pettenkoferstr. 9, Ludwig-Maximilians-University, 80336 Munich, Germany
| | - Can Gollmann-Tepeköylü
- Department of Cardiac Surgery, Medical University of Innsbruck, Anichstr.35, A - 6020 Innsbruck, Austria
| | - Linda W Van Laake
- Cardiology and UMC Utrecht Regenerative Medicine Center, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Sophie Van Linthout
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité, University Medicine Berlin, 10178 Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité, University Medicine Berlin, 10178 Berlin, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University Giessen, Ludwigstraße 23, 35390 Giessen, Germany
| | - Kirsti Ytrehus
- Department of Medical Biology, UiT The Arctic University of Norway, Hansine Hansens veg 18, 9037 Tromsø, Norway
| |
Collapse
|
17
|
Quantitative proteomic analyses reveal that GPX4 downregulation during myocardial infarction contributes to ferroptosis in cardiomyocytes. Cell Death Dis 2019; 10:835. [PMID: 31685805 PMCID: PMC6828761 DOI: 10.1038/s41419-019-2061-8] [Citation(s) in RCA: 244] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 08/05/2019] [Accepted: 09/23/2019] [Indexed: 12/17/2022]
Abstract
Ischaemic heart disease (IHD) is the leading cause of death worldwide. Although myocardial cell death plays a significant role in myocardial infarction (MI), its underlying mechanism remains to be elucidated. To understand the progression of MI and identify potential therapeutic targets, we performed tandem mass tag (TMT)-based quantitative proteomic analysis using an MI mouse model. Gene ontology (GO) analysis and gene set enrichment analysis (GSEA) revealed that the glutathione metabolic pathway and reactive oxygen species (ROS) pathway were significantly downregulated during MI. In particular, glutathione peroxidase 4 (GPX4), which protects cells from ferroptosis (an iron-dependent programme of regulated necrosis), was downregulated in the early and middle stages of MI. RNA-seq and qRT-PCR analyses suggested that GPX4 downregulation occurred at the transcriptional level. Depletion or inhibition of GPX4 using specific siRNA or the chemical inhibitor RSL3, respectively, resulted in the accumulation of lipid peroxide, leading to cell death by ferroptosis in H9c2 cardiomyoblasts. Although neonatal rat ventricular myocytes (NRVMs) were less sensitive to GPX4 inhibition than H9c2 cells, NRVMs rapidly underwent ferroptosis in response to GPX4 inhibition under cysteine deprivation. Our study suggests that downregulation of GPX4 during MI contributes to ferroptotic cell death in cardiomyocytes upon metabolic stress such as cysteine deprivation.
Collapse
|
18
|
Boarescu PM, Boarescu I, Bocșan IC, Pop RM, Gheban D, Bulboacă AE, Nicula C, Râjnoveanu RM, Bolboacă SD. Curcumin Nanoparticles Protect against Isoproterenol Induced Myocardial Infarction by Alleviating Myocardial Tissue Oxidative Stress, Electrocardiogram, and Biological Changes. Molecules 2019; 24:molecules24152802. [PMID: 31374848 PMCID: PMC6696485 DOI: 10.3390/molecules24152802] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 07/25/2019] [Accepted: 07/31/2019] [Indexed: 12/13/2022] Open
Abstract
Curcumin from Curcuma longa is a nutraceutical compound reported to possess strong antioxidant activity that makes it a candidate for use in counteracting oxidative stress-induced damage. The effect of pre-treatment with curcumin nanoparticles (nC) compared to conventional curcumin (Cs) on blood pressure, electrocardiogram, and biological changes on isoproterenol (ISO)-induced myocardial infarction (MI) in rats had been investigated. The Cs doses of 150 and 200 mg/kg bw and all nC doses (100, 150 and 200 mg/kg bw) significantly reduced heart rate before ISO administration and prevented QRS complex enlargement after MI induction (p < 0.026). All doses of Cs and nC prevented prolongation of the QT and QT corrected (QTc) intervals, with better results for higher doses (p < 0.048). The nC solution had more significant results than Cs in all metabolic parameters assessed (lactate dehydrogenase, glycaemia, aspartate transaminase, and alanine transaminase, p < 0.009). nC was more efficient than Cs in limiting myocardial oxidative stress and enhancing antioxidative capacity (p < 0.004). Compared to Cs, nC better prevented myocardial damage extension, reduced interstitial oedema, and inflammation. Curcumin nanoparticles as compared to conventional curcumin exert better antioxidative effects. Moreover, nC better prevent cardiomyocytes damage, and electrocardiogram alterations, in the case of ISO-induced MI in rats.
Collapse
Affiliation(s)
- Paul-Mihai Boarescu
- Department of Pathophysiology, Iuliu Haţieganu University of Medicine and Pharmacy Cluj-Napoca, Victor Babeş Street, no. 2-4, 400012 Cluj-Napoca, Romania
- Department of Medical Informatics and Biostatistics, Iuliu Haţieganu University of Medicine and Pharmacy Cluj-Napoca, Louis Pasteur Street, no. 6, 400349 Cluj-Napoca, Romania
| | - Ioana Boarescu
- Department of Neurology, County Clinical Emergency Hospital of Cluj-Napoca, Victor Babes Street, no 43, 400012 Cluj-Napoca, Romania
| | - Ioana Corina Bocșan
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Haţieganu University of Medicine and Pharmacy Cluj-Napoca, Gheorghe Marinescu Street, no 23, 400337 Cluj-Napoca, Romania
| | - Raluca Maria Pop
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Haţieganu University of Medicine and Pharmacy Cluj-Napoca, Gheorghe Marinescu Street, no 23, 400337 Cluj-Napoca, Romania
| | - Dan Gheban
- Department of Pathological Anatomy, Iuliu Haţieganu University of Medicine and Pharmacy Cluj-Napoca, Clinicilor Street, no 3-5, 400006 Cluj-Napoca, Romania
| | - Adriana Elena Bulboacă
- Department of Pathophysiology, Iuliu Haţieganu University of Medicine and Pharmacy Cluj-Napoca, Victor Babeş Street, no. 2-4, 400012 Cluj-Napoca, Romania.
| | - Cristina Nicula
- Department of Ophthalmology, Iuliu Haţieganu University of Medicine and Pharmacy Cluj-Napoca, Clinicilor Street, no 3-5, 400006 Cluj-Napoca, Romania.
| | - Ruxandra-Mioara Râjnoveanu
- Department of Pneumology, Iuliu Haţieganu University of Medicine and Pharmacy Cluj-Napoca, B.P. Hasdeu Street, no. 6, 400371 Cluj-Napoca, Romania
| | - Sorana D Bolboacă
- Department of Medical Informatics and Biostatistics, Iuliu Haţieganu University of Medicine and Pharmacy Cluj-Napoca, Louis Pasteur Street, no. 6, 400349 Cluj-Napoca, Romania
| |
Collapse
|
19
|
Rahnavard M, Hassanpour M, Ahmadi M, Heidarzadeh M, Amini H, Javanmard MZ, Nouri M, Rahbarghazi R, Safaie N. Curcumin ameliorated myocardial infarction by inhibition of cardiotoxicity in the rat model. J Cell Biochem 2019; 120:11965-11972. [PMID: 30775806 DOI: 10.1002/jcb.28480] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 12/17/2018] [Accepted: 01/02/2019] [Indexed: 01/24/2023]
Abstract
Cardiovascular diseases are the main cause of death globally. Many attempts have been done to ameliorate the pathological changes after the occurrence of myocardial infarction. Curcumin is touted as a polyphenol phytocompound with appropriate cardioprotective properties. In this study, the therapeutic effect of curcumin was investigated on acute myocardial infarction in the model of rats. Rats were classified into four groups; control, isoproterenol hydrochloride (ISO) (100 mg/kbw), curcumin (50 mg/kbw), and curcumin plus ISO treatment groups. After 9-day administration of curcumin, levels of lactate dehydrogenase (LDH), creatine kinase (CK), and cardiac troponin I (cTnI) were determined. Superoxide dismutase (SOD) and malondialdehyde (MDA) contents were measured to investigate the oxidative status in infarct rats received curcumin. By using H & E staining, tissue inflammation was performed. Masson's trichrome staining was conducted to show cardiac remodeling and collagen deposition. The number of apoptotic cells was determined by using the terminal deoxynucleotidyl transferase dUTP nick end labeling assay. Data showed the serum decrease of LDH, CK, and cTnI in infarct rats after curcumin intake compared to the rats given (ISO) ( P < 0.05). Curcumin was found to reduce oxidative status by reducing SOD and MDA contents ( P < 0.05). Gross and microscopic examinations revealed that the decrease of infarct area, inflammation response and collagen deposition in rats given ISO plus curcumin ( P < 0.05). We noted the superior effect of curcumin to reduce the number of apoptotic cardiomyocytes after 9 days. Data point the cardioprotective effect of curcumin to diminish the complication of infarction by the reduction of cell necrosis and apoptosis in a rat model of experimental infarction.
Collapse
Affiliation(s)
- Mehdi Rahnavard
- Department of Anatomy and Histology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mehdi Hassanpour
- Stem Cell And Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Ahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Heidarzadeh
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hassan Amini
- Stem Cell And Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of General and Vascular Surgery, Imam Reza Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoumeh Zirak Javanmard
- Department of Anatomy and Histology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mohammad Nouri
- Stem Cell And Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nasser Safaie
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
20
|
Effects of Curcumin Nanoparticles in Isoproterenol-Induced Myocardial Infarction. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7847142. [PMID: 31205590 PMCID: PMC6530192 DOI: 10.1155/2019/7847142] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 03/15/2019] [Accepted: 03/21/2019] [Indexed: 12/30/2022]
Abstract
Curcumin has anti-inflammatory, antioxidative, anticarcinogenic, and cardiovascular protective effects. Our study is aimed at evaluating the effects of pretreatment with curcumin nanoparticles (CCNP) compared to conventional curcumin (CC) on isoproterenol (ISO) induced myocardial infarction (MI) in rats. Fifty-six Wistar-Bratislava white rats were randomly divided into eight groups of seven rats each. Curcumin and curcumin nanoparticles were given by gavage in three different doses (100 mg/kg body weight (bw), 150 mg/kg bw, and 200 mg/kg bw) for 15 days. The MI was induced on day 13 using 100 mg/kg bw ISO administered twice, with the second dose 24 h after the initial dose. The blood samples were taken 24 h after the last dose of ISO. The antioxidant, anti-inflammatory, and cardioprotective effects were evaluated in all groups. All doses of CC and CCNP offered a cardioprotective effect by preventing creatine kinase-MB leakage from cardiomyocytes, with the best result for CCNP. All the oxidative stress parameters were significantly improved after CCNP compared to CC pretreatment. CCNP was more efficient than CC in limiting the increase in inflammatory cytokine levels (such as TNF-α, IL-6, IL-1α, IL-1β, MCP-1, and RANTES) after MI. MMP-2 and MMP-9 levels decreased more after pretreatment with CCNP than with CC. CCNP better prevented myocardial necrosis and reduced interstitial edema and neutrophil infiltration than CC, on histopathological examination. Therefore, improving the bioactivity of curcumin by nanotechnology may help limit cardiac injury after myocardial infarction.
Collapse
|