1
|
Carulli E, McGarvey M, Chabok M, Panoulas V, Rosser G, Akhtar M, Smith R, Chandra N, Al-Hussaini A, Kabir T, Barker L, Bruno F, Konstantinou K, de Silva R, Hill J, Xu Y, Lane R, Bucciarelli-Ducci C, Luescher T, Dalby M. Transcoronary cooling and dilution for cardioprotection during revascularisation for ST-segment elevation myocardial infarction: design and rationale of the STEMI-Cool study. Am Heart J 2024:S0002-8703(24)00338-7. [PMID: 39742936 DOI: 10.1016/j.ahj.2024.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/11/2024] [Accepted: 12/20/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND ST-segment elevation myocardial infarction (STEMI) is treated with immediate primary percutaneous coronary intervention (pPCI) to restore coronary blood flow in the acutely ischaemic territory, but is associated with reperfusion injury limiting the benefit of the therapy. No treatment has proven effective in reducing reperfusion injury. Transcoronary hypothermia has been tested in clinical studies and is well tolerated, but is generally established after crossing the occlusion with a guidewire therefore after initial reperfusion, which might have contributed to the neutral outcomes. Transcatheter strategies may also offer additional benefit through haemodilution and the resultant controlled reperfusion, but this has not been fully investigated for pPCI. DESIGN STEMI-Cool is a pragmatic, registry-based randomised clinical pilot trial to test the recruitment rate, feasibility, and safety of a simple transcoronary cooling and dilution protocol. Sixty STEMI patients undergoing pPCI will be randomised 1:1 to standard of care or continuous infusion of room temperature saline through the guiding catheter to achieve intracoronary temperature reductions of 6-8°C, commencing before crossing the coronary occlusion with a guidewire. Mechanistic outcome measures will include microvascular resistance, biomarkers of inflammation before infusion and at 24h, and magnetic resonance imaging of myocardial salvage and infarct size. CONCLUSIONS STEMI-Cool will investigate the recruitment rate, feasibility and safety of an innovative and simple cooling and diluting strategy for cardioprotection before and during reperfusion with pPCI, aiming to address limitations faced in other studies. Mechanistic outcome measures will allow insight into inflammatory, microvascular and structural changes induced by transcoronary cooling and dilution.
Collapse
Affiliation(s)
- Ermes Carulli
- Department of cardiology, Guy's and St Thomas' NHS Foundation Trust, Harefield Hospital, London, UK; Doctoral school in Translational Medicine, University of Milan, Milan, Italy.
| | - Michael McGarvey
- Department of cardiology, Guy's and St Thomas' NHS Foundation Trust, Harefield Hospital, London, UK
| | - Mohssen Chabok
- Department of cardiology, Guy's and St Thomas' NHS Foundation Trust, Harefield Hospital, London, UK
| | - Vasileios Panoulas
- Department of cardiology, Guy's and St Thomas' NHS Foundation Trust, Harefield Hospital, London, UK
| | - Gareth Rosser
- Department of cardiology, Guy's and St Thomas' NHS Foundation Trust, Harefield Hospital, London, UK
| | - Mohammed Akhtar
- Department of cardiology, Guy's and St Thomas' NHS Foundation Trust, Harefield Hospital, London, UK
| | - Robert Smith
- Department of cardiology, Guy's and St Thomas' NHS Foundation Trust, Harefield Hospital, London, UK
| | - Navin Chandra
- Department of cardiology, Guy's and St Thomas' NHS Foundation Trust, Harefield Hospital, London, UK
| | - Abtehale Al-Hussaini
- Department of cardiology, Guy's and St Thomas' NHS Foundation Trust, Harefield Hospital, London, UK
| | - Tito Kabir
- Department of cardiology, Guy's and St Thomas' NHS Foundation Trust, Harefield Hospital, London, UK
| | - Laura Barker
- Department of cardiology, Guy's and St Thomas' NHS Foundation Trust, Harefield Hospital, London, UK
| | - Francesco Bruno
- Department of cardiology, Guy's and St Thomas' NHS Foundation Trust, Harefield Hospital, London, UK
| | | | - Ranil de Silva
- Department of cardiology, Guy's and St Thomas' NHS Foundation Trust, Harefield Hospital, London, UK
| | - Jonathan Hill
- Department of cardiology, Guy's and St Thomas' NHS Foundation Trust, Harefield Hospital, London, UK
| | - Yun Xu
- Department of Chemical Engineering, Imperial College London, London, UK
| | - Rebecca Lane
- Department of cardiology, Guy's and St Thomas' NHS Foundation Trust, Harefield Hospital, London, UK
| | - Chiara Bucciarelli-Ducci
- Department of cardiology, Guy's and St Thomas' NHS Foundation Trust, Harefield Hospital, London, UK
| | - Thomas Luescher
- Department of cardiology, Guy's and St Thomas' NHS Foundation Trust, Harefield Hospital, London, UK; King's College London, London, UK
| | - Miles Dalby
- Department of cardiology, Guy's and St Thomas' NHS Foundation Trust, Harefield Hospital, London, UK; King's College London, London, UK
| |
Collapse
|
2
|
Zhang Q, Ruan H, Wang X, Luo A, Ran X. Ulinastatin attenuated cardiac ischaemia/reperfusion injury by suppressing activation of the tissue kallikrein-kinin system. Br J Pharmacol 2024; 181:4988-5008. [PMID: 39294926 DOI: 10.1111/bph.16477] [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: 08/14/2023] [Revised: 02/29/2024] [Accepted: 05/13/2024] [Indexed: 09/21/2024] Open
Abstract
BACKGROUND AND PURPOSE Ulinastatin has beneficial effects in patients undergoing coronary artery bypass grafting (CABG) surgery due to its anti-inflammatory properties, but the underlying mechanism remains unclear. EXPERIMENTAL APPROACH We used samples from patients undergoing CABG, a model of cardiac ischaemia-reperfusion injury (IRI) in mice and murine cardiac endothelial cell cultures to investigate links between ulinastatin, the kallikrein-kinin system (KKS), endothelial dysfunction and cardiac inflammation in the response to ischaemia/reperfusion injury (IRI). These links were assessed using clinical investigations, in vitro and in vivo experiments and RNA sequencing analysis. KEY RESULTS Ulinastatin inhibited the activity of tissue kallikrein, a key enzyme of the KKS, at 24 h after CABG surgery, which was verified in our murine cardiac ischaemia-reperfusion model. Under normal conditions, ulinastatin only inhibited kallikrein activity but did not affect bradykinin (B1/B2) receptors. Ulinastatin protected against IRI, in vivo and in vitro, by suppressing activation of the kallikrein-kinin system and down-regulating B1/B2 receptor-related signalling pathways including ERK/ iNOS, which resulted in enhanced endothelial barrier function, mitigation of inflammation and oedema, decreased infarct size, improved cardiac function and decreased mortality. Inhibition of kallikrein and knockdown of B1, but not B2 receptors prevented ERK translocation into the nucleus, reducing reperfusion-induced injury in murine cardiac endothelial cells. CONCLUSIONS AND IMPLICATIONS Treatment with ulinastatin exerts a protective influence on cardiac reperfusion by suppressing activation of the kallikrein-kinin system. Our findings highlight the potential of targeting kallikrein /bradykinin receptors to alleviate endothelial dysfunction, thus improving cardiac IRI.
Collapse
Affiliation(s)
- Qin Zhang
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hang Ruan
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaochuan Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ailin Luo
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao Ran
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
3
|
Qiu ZY, Shi KN, Li HH, Zhang B. CBR-470-1 protects against cardiomyocyte death in ischaemia/reperfusion injury by activating the Nrf2-GPX4 cascade. Toxicol Appl Pharmacol 2024; 492:117113. [PMID: 39343043 DOI: 10.1016/j.taap.2024.117113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/14/2024] [Accepted: 09/25/2024] [Indexed: 10/01/2024]
Abstract
Cardiac ischaemia/reperfusion (I/R) impairs mitochondrial function, resulting in excessive oxidative stress and cardiomyocyte ferroptosis and death. Nuclear factor E2-related factor 2 (Nrf2) is a key regulator of redox homeostasis and has cardioprotective effects against various stresses. Here, we tested whether CBR-470-1, a noncovalent Nrf2 activator, can protect against cardiomyocyte death caused by I/R stress. Compared with vehicle treatment, the administration of CBR-470-1 (2 mg/kg) to mice significantly increased Nrf2 protein levels and ameliorated the infarct size, the I/R-induced decrease in cardiac contractile performance, and the I/R-induced increases in cell apoptosis, ROS levels, and inflammation. Consistently, the beneficial effects of CBR-470-1 on cardiomyocytes were verified in a hypoxia/reoxygenation (H/R) model in vitro, but this cardioprotection was dramatically attenuated by the GPX4 inhibitor RSL3. Mechanistically, CBR-470-1 upregulated Nrf2 expression, which increased the expression levels of antioxidant enzymes (NQO1, SOD1, Prdx1, and Gclc) and antiferroptotic proteins (SLC7A11 and GPX4) and downregulated the protein expression of p53 and Nlrp3, leading to the inhibition of ROS production and inflammation and subsequent cardiomyocyte death (apoptosis, ferroptosis and pyroptosis). In summary, CBR-470-1 prevented I/R-mediated cardiac injury possibly through inhibiting cardiomyocyte apoptosis, ferroptosis and pyroptosis via Nrf2-mediated inhibition of p53 and Nlrp3 and activation of the SLC7A11/GPX4 pathway. Our data also highlight that CBR-470-1 may serve as a valuable agent for treating ischaemic heart disease.
Collapse
Affiliation(s)
- Ze-Yang Qiu
- Department of Cardiology, First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning 116011, China
| | - Kai-Na Shi
- Department of Emergency Medicine, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Hui-Hua Li
- Department of Emergency Medicine, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China.
| | - Bo Zhang
- Department of Cardiology, First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning 116011, China.
| |
Collapse
|
4
|
Li J, Song X, Liao X, Shi Y, Chen H, Xiao Q, Liu F, Zhan J, Cai Y. Adaptive enzyme-responsive self-assembling multivalent apelin ligands for targeted myocardial infarction therapy. J Control Release 2024; 372:571-586. [PMID: 38897292 DOI: 10.1016/j.jconrel.2024.06.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/06/2024] [Accepted: 06/14/2024] [Indexed: 06/21/2024]
Abstract
Microvascular dysfunction following myocardial infarction exacerbates coronary flow obstruction and impairs the preservation of ventricular function. The apelinergic system, known for its pleiotropic effects on improving vascular function and repairing ischemic myocardium, has emerged as a promising therapeutic target for myocardial infarction. Despite its potential, the natural apelin peptide has an extremely short circulating half-life. Current apelin analogs have limited receptor binding efficacy and poor targeting, which restricts their clinical applications. In this study, we utilized an enzyme-responsive peptide self-assembly technique to develop an enzyme-responsive small molecule peptide that adapts to the expression levels of matrix metalloproteinases in myocardial infarction lesions. This peptide is engineered to respond to the high concentration of matrix metalloproteinases in the lesion area, allowing for precise and abundant presentation of the apelin motif. The changes in hydrophobicity allow the apelin motif to self-assemble into a supramolecular multivalent peptide ligand-SAMP. This self-assembly behavior not only prolongs the residence time of apelin in the myocardial infarction lesion but also enhances the receptor-ligand interaction through increased receptor binding affinity due to multivalency. Studies have demonstrated that SAMP significantly promotes angiogenesis after ischemia, reduces cardiomyocyte apoptosis, and improves cardiac function. This novel therapeutic strategy offers a new approach to restoring coronary microvascular function and improving damaged myocardium after myocardial infarction.
Collapse
Affiliation(s)
- Jiejing Li
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xudong Song
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xu Liao
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yihan Shi
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Huiming Chen
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Qiuqun Xiao
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Fengjiao Liu
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jie Zhan
- Department of Laboratory Medicine, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Yanbin Cai
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China; Department of Cardiovascular Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
5
|
Li F, Wang Y, Li W, Wu J, Li S, Hu X, Tang T, Liu X. Enhanced protection against hypoxia/reoxygenation-induced apoptosis in H9c2 cells by puerarin-loaded liposomes modified with matrix metalloproteinases-targeting peptide and triphenylphosphonium. J Liposome Res 2023; 33:378-391. [PMID: 37017315 DOI: 10.1080/08982104.2023.2193845] [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: 10/19/2022] [Accepted: 03/06/2023] [Indexed: 04/06/2023]
Abstract
Based on the inhibition of mitochondrial permeability transition pore (mPTP) opening, puerarin (PUE) has a good potential to reduce myocardial ischemia/reperfusion injury (MI/RI). However, the lack of targeting of free PUE makes it difficult to reach the mitochondria. In this paper, we constructed matrix metalloproteinase-targeting peptide (MMP-TP) and triphenylphosphonium (TPP) cation co-modified liposomes loaded with PUE (PUE@T/M-L) for mitochondria-targeted drug delivery. PUE@T/M-L had a favorable particle size of 144.9 ± 0.8 nm, an encapsulation efficiency of 78.9 ± 0.6%, and a sustained-release behavior. The results of cytofluorimetric experiments showed that MMP-TP and TPP double-modified liposomes (T/M-L) enhanced intracellular uptake, escaped lysosomal capture, and promoted drug targeting into mitochondria. In addition, PUE@T/M-L enhanced the viability of hypoxia-reoxygenation (H/R) injured H9c2 cells by inhibiting mPTP opening and reactive oxygen species (ROS) production, reducing Bax expression and increasing Bcl-2 expression. It was inferred that PUE@T/M-L delivered PUE into the mitochondria of H/R injured H9c2 cells, resulting in a significant increase in cellular potency. Based on the ability of MMP-TP to bind the elevated expression of matrix metalloproteinases (MMPs), T/M-L had excellent tropism for Lipopolysaccharide (LPS) -stimulated macrophages and can significantly reduce TNF-α and ROS levels, thus allowing both drug accumulation in ischemic cardiomyocytes and reducing inflammatory stimulation during MI/RI. Fluorescence imaging results of the targeting effect using a DiR probe also indicated that DiR@T/M-L could accumulate and retain in the ischemic myocardium. Taken together, these results demonstrated the promising application of PUE@T/M-L for mitochondria-targeted drug delivery to achieve maximum therapeutic efficacy of PUE.
Collapse
Affiliation(s)
- Fengmei Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institution of Clinical Pharmacy, Central South University, Changsha, China
| | - Yan Wang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institution of Clinical Pharmacy, Central South University, Changsha, China
| | - Wenqun Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institution of Clinical Pharmacy, Central South University, Changsha, China
| | - Junyong Wu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institution of Clinical Pharmacy, Central South University, Changsha, China
| | - Shengnan Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institution of Clinical Pharmacy, Central South University, Changsha, China
| | - Xiongbin Hu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institution of Clinical Pharmacy, Central South University, Changsha, China
| | - Tiantian Tang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institution of Clinical Pharmacy, Central South University, Changsha, China
| | - Xinyi Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institution of Clinical Pharmacy, Central South University, Changsha, China
| |
Collapse
|
6
|
Wang T, Wu S, Ibrahim IAA, Fan L. Cardioprotective Role of Swertiamarin, a Plant Glycoside Against Experimentally Induced Myocardial Infarction via Antioxidant and Anti-inflammatory Functions. Appl Biochem Biotechnol 2023; 195:5394-5408. [PMID: 35960488 DOI: 10.1007/s12010-022-04094-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/15/2022] [Indexed: 12/07/2022]
Abstract
The study examined the protective effects of swertiamarin on rats with experimentally induced myocardial infarction. Three to six week-old male albino Wistar rats were used in this study and experimental myocardial infarction (MI) was induced using isoproterenol. Our results showed that swertiamarin restored the alteration in heart weight, body weight, and heart weight/tibia length ratio of MI-induced rats to basal levels significantly (p < 0.05). Swertiamarin significantly (p < 0.05) restored the levels of cardiac pathophysiological marker creatine kinase (CKMB), lactate dehydrogenase (LDH), aspartate aminotransferase (AST), alanine transaminase (ALT), and cardiac troponin I (cTn-1) to near normalcy in MI-induced rats. Levels of oxidative stress markers malondialdehyde (MDA), protein carbonyls (PC), and levels of Vitamin C and Vitamin E were significantly (p < 0.05) reverted to near basal levels in MI-induced rats by swertiamarin. Levels of the antioxidant glutathione (GSH) and antioxidant enzymes which include superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), glutathione-s-transferase (GST), glutathione reductase (GR), and plasma total antioxidant capacity (TAC) were (p < 0.05) brought to near normalcy in MI-induced rats by swertiamarin. Levels of sodium (Na), potassium (k), and calcium (Ca) ATPases were significantly (p < 0.05) restored to near normalcy in MI-induced rats by swertiamarin. Status of pro-inflammatory cytokines including tumor necrosis factor (TNF-α), interleukin-6 (IL-6), and histological aberrations were also significantly (p < 0.05) restored to near normalcy in MI-induced rats by swertiamarin. Together, our results concluded that swertiamarin exerts significant cardioprotective functions in experimental MI in rats.
Collapse
Affiliation(s)
- Tao Wang
- Department of Cardiology, Shandong Provincial Third Hospital, Shandong University, Jinan, 250031, Shandong, China
| | - Shubin Wu
- Department of Cardiology, Shandong Provincial Third Hospital, Shandong University, Jinan, 250031, Shandong, China
| | - Ibrahim Abdel Aziz Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Leilei Fan
- Department of Cardiology, Shandong Provincial Third Hospital, Shandong University, Jinan, 250031, Shandong, China.
| |
Collapse
|
7
|
Reventun P, Sánchez-Esteban S, Cook-Calvete A, Delgado-Marín M, Roza C, Jorquera-Ortega S, Hernandez I, Tesoro L, Botana L, Zamorano JL, Zaragoza C, Saura M. Endothelial ILK induces cardioprotection by preventing coronary microvascular dysfunction and endothelial-to-mesenchymal transition. Basic Res Cardiol 2023; 118:28. [PMID: 37452166 PMCID: PMC10348984 DOI: 10.1007/s00395-023-00997-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 06/13/2023] [Accepted: 06/30/2023] [Indexed: 07/18/2023]
Abstract
Endothelial dysfunction is an early event in coronary microvascular disease. Integrin-linked kinase (ILK) prevents endothelial nitric oxide synthase (eNOS) uncoupling and, thus, endothelial dysfunction. However, the specific role of endothelial ILK in cardiac function remains to be fully elucidated. We hypothesised that endothelial ILK plays a crucial role in maintaining coronary microvascular function and contractile performance in the heart. We generated an endothelial cell-specific ILK conditional knock-out mouse (ecILK cKO) and investigated cardiovascular function. Coronary endothelial ILK deletion significantly impaired cardiac function: ejection fraction, fractional shortening and cardiac output decreased, whilst left ventricle diastolic internal diameter decreased and E/A and E/E' ratios increased, indicating not only systolic but also diastolic dysfunction. The functional data correlated with extensive extracellular matrix remodelling and perivascular fibrosis, indicative of adverse cardiac remodelling. Mice with endothelial ILK deletion suffered early ischaemic-like events with ST elevation and transient increases in cardiac troponins, which correlated with fibrotic remodelling. In addition, ecILK cKO mice exhibited many features of coronary microvascular disease: reduced cardiac perfusion, impaired coronary flow reserve and arterial remodelling with patent epicardial coronary arteries. Moreover, endothelial ILK deletion induced a moderate increase in blood pressure, but the antihypertensive drug Losartan did not affect microvascular remodelling whilst only partially ameliorated fibrotic remodelling. The plasma miRNA profile reveals endothelial-to-mesenchymal transition (endMT) as an upregulated pathway in endothelial ILK conditional KO mice. Our results show that endothelial cells in the microvasculature in endothelial ILK conditional KO mice underwent endMT. Moreover, endothelial cells isolated from these mice and ILK-silenced human microvascular endothelial cells underwent endMT, indicating that decreased endothelial ILK contributes directly to this endothelial phenotype shift. Our results identify ILK as a crucial regulator of microvascular endothelial homeostasis. Endothelial ILK prevents microvascular dysfunction and cardiac remodelling, contributing to the maintenance of the endothelial cell phenotype.
Collapse
Affiliation(s)
- P Reventun
- Facultad Medicina, Depto. Biología Sistemas (UD Fisiología), Universidad de Alcalá, IRYCIS, Mod 2 Planta 0, Ctra Madrid, Barcelona Km 33,500, Alcalá de Henares, Madrid, Spain
- School of Medicine, Department of Medicine, Cardiology Division, Johns Hopkins University, Baltimore, MD, United States
| | - S Sánchez-Esteban
- Facultad Medicina, Depto. Biología Sistemas (UD Fisiología), Universidad de Alcalá, IRYCIS, Mod 2 Planta 0, Ctra Madrid, Barcelona Km 33,500, Alcalá de Henares, Madrid, Spain
| | - A Cook-Calvete
- Facultad Medicina, Depto. Biología Sistemas (UD Fisiología), Universidad de Alcalá, IRYCIS, Mod 2 Planta 0, Ctra Madrid, Barcelona Km 33,500, Alcalá de Henares, Madrid, Spain
| | - M Delgado-Marín
- Facultad Medicina, Depto. Biología Sistemas (UD Fisiología), Universidad de Alcalá, IRYCIS, Mod 2 Planta 0, Ctra Madrid, Barcelona Km 33,500, Alcalá de Henares, Madrid, Spain
| | - C Roza
- Facultad Medicina, Depto. Biología Sistemas (UD Fisiología), Universidad de Alcalá, IRYCIS, Mod 2 Planta 0, Ctra Madrid, Barcelona Km 33,500, Alcalá de Henares, Madrid, Spain
| | - S Jorquera-Ortega
- Facultad Medicina, Depto. Biología Sistemas (UD Fisiología), Universidad de Alcalá, IRYCIS, Mod 2 Planta 0, Ctra Madrid, Barcelona Km 33,500, Alcalá de Henares, Madrid, Spain
| | - I Hernandez
- Unidad Mixta de Investigación Cardiovascular, Universidad Francisco de Vitoria, IRYCIS, Pozuelo de Alarcón, Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - L Tesoro
- Unidad Mixta de Investigación Cardiovascular, Universidad Francisco de Vitoria, IRYCIS, Pozuelo de Alarcón, Madrid, Spain
| | - L Botana
- Unidad Mixta de Investigación Cardiovascular, Universidad Francisco de Vitoria, IRYCIS, Pozuelo de Alarcón, Madrid, Spain
| | - J L Zamorano
- Servicio Cardiología, Hospital Universitario Ramón y Cajal, Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - C Zaragoza
- Unidad Mixta de Investigación Cardiovascular, Universidad Francisco de Vitoria, IRYCIS, Pozuelo de Alarcón, Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - M Saura
- Facultad Medicina, Depto. Biología Sistemas (UD Fisiología), Universidad de Alcalá, IRYCIS, Mod 2 Planta 0, Ctra Madrid, Barcelona Km 33,500, Alcalá de Henares, Madrid, Spain.
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.
| |
Collapse
|
8
|
Wang Y, Guo L, Zhang Z, Fu S, Huang P, Wang A, Liu M, Ma X. A bibliometric analysis of myocardial ischemia/reperfusion injury from 2000 to 2023. Front Cardiovasc Med 2023; 10:1180792. [PMID: 37383699 PMCID: PMC10293770 DOI: 10.3389/fcvm.2023.1180792] [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: 03/06/2023] [Accepted: 05/22/2023] [Indexed: 06/30/2023] Open
Abstract
Background Myocardial ischemia/reperfusion injury (MIRI) refers to the more severe damage that occurs in the previously ischemic myocardium after a short-term interruption of myocardial blood supply followed by restoration of blood flow within a certain period of time. MIRI has become a major challenge affecting the therapeutic efficacy of cardiovascular surgery. Methods A scientific literature search on MIRI-related papers published from 2000 to 2023 in the Web of Science Core Collection database was conducted. VOSviewer was used for bibliometric analysis to understand the scientific development and research hotspots in this field. Results A total of 5,595 papers from 81 countries/regions, 3,840 research institutions, and 26,202 authors were included. China published the most papers, but the United States had the most significant influence. Harvard University was the leading research institution, and influential authors included Lefer David J., Hausenloy Derek J., Yellon Derek M., and others. All keywords can be divided into four different directions: risk factors, poor prognosis, mechanisms and cardioprotection. Conclusion Research on MIRI is flourishing. It is necessary to conduct an in-depth investigation of the interaction between different mechanisms and multi-target therapy will be the focus and hotspot of MIRI research in the future.
Collapse
Affiliation(s)
- Yifei Wang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Lijun Guo
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Zhibo Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Shuangqing Fu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Pingping Huang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Anzhu Wang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Mi Liu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Xiaochang Ma
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| |
Collapse
|
9
|
Kloka JA, Friedrichson B, Wülfroth P, Henning R, Zacharowski K. Microvascular Leakage as Therapeutic Target for Ischemia and Reperfusion Injury. Cells 2023; 12:1345. [PMID: 37408180 DOI: 10.3390/cells12101345] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/03/2023] [Accepted: 05/07/2023] [Indexed: 07/07/2023] Open
Abstract
Reperfusion injury is a very common complication of various indicated therapies such as the re-opening of vessels in the myocardium or brain as well as reflow in hemodynamic shutdown (cardiac arrest, severe trauma, aortic cross-clamping). The treatment and prevention of reperfusion injury has therefore been a topic of immense interest in terms of mechanistic understanding, the exploration of interventions in animal models and in the clinical setting in major prospective studies. While a wealth of encouraging results has been obtained in the lab, the translation into clinical success has met with mixed outcomes at best. Considering the still very high medical need, progress continues to be urgently needed. Multi-target approaches rationally linking interference with pathophysiological pathways as well as a renewed focus on aspects of microvascular dysfunction, especially on the role of microvascular leakage, are likely to provide new insights.
Collapse
Affiliation(s)
- Jan Andreas Kloka
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Goethe University, 60590 Frankfurt, Germany
| | - Benjamin Friedrichson
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Goethe University, 60590 Frankfurt, Germany
| | | | | | - Kai Zacharowski
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Goethe University, 60590 Frankfurt, Germany
| |
Collapse
|
10
|
Brlecic PE, Bonham CA, Rosengart TK, Mathison M. Direct cardiac reprogramming: A new technology for cardiac repair. J Mol Cell Cardiol 2023; 178:51-58. [PMID: 36965701 PMCID: PMC10124164 DOI: 10.1016/j.yjmcc.2023.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 03/03/2023] [Accepted: 03/21/2023] [Indexed: 03/27/2023]
Abstract
Cardiovascular disease is one of the leading causes of morbidity and mortality worldwide, with myocardial infarctions being amongst the deadliest manifestations. Reduced blood flow to the heart can result in the death of cardiac tissue, leaving affected patients susceptible to further complications and recurrent disease. Further, contemporary management typically involves a pharmacopeia to manage the metabolic conditions contributing to atherosclerotic and hypertensive heart disease, rather than regeneration of the damaged myocardium. With modern healthcare extending lifespan, a larger demographic will be at risk for heart disease, driving the need for novel therapeutics that surpass those currently available in efficacy. Transdifferentiation and cellular reprogramming have been looked to as potential methods for the treatment of diseases throughout the body. Specifically targeting the fibrotic cells in cardiac scar tissue as a source to be reprogrammed into induced cardiomyocytes remains an appealing option. This review aims to highlight the history of and advances in cardiac reprogramming and describe its translational potential as a treatment for cardiovascular disease.
Collapse
Affiliation(s)
- Paige E Brlecic
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Clark A Bonham
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Todd K Rosengart
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Megumi Mathison
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
11
|
Yang Y, Shao M, Yao J, Yang S, Cheng W, Ma L, Li W, Cao J, Zhang Y, Hu Y, Li C, Wang Y, Wang W. Neocryptotanshinone protects against myocardial ischemia-reperfusion injury by promoting autolysosome degradation of protein aggregates via the ERK1/2-Nrf2-LAMP2 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 110:154625. [PMID: 36586206 DOI: 10.1016/j.phymed.2022.154625] [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: 07/21/2022] [Revised: 12/04/2022] [Accepted: 12/23/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Aggrephagy is a critical compensatory mechanism for the elimination of misfolded proteins resulting from stress and depends on the autolysosome degradation of protein aggregates. However, there have been few mechanism research related to aggrephagy in myocardial ischemia/reperfusion (I/R) injury. Neocryptotanshinone (NCTS) is a fat-soluble active compound extracted from Salvia miltiorrhiza, and may be cardioprotective against I/R. However, the efficacy and specific mechanism of NCTS on I/R have not been studied. PURPOSE The current study aimed to investigate the molecular mechanism of NCTS involved in the therapeutic effect on I/R, with a special emphasis on the up-regulation of the ERK1/2-Nrf2-LAMP2 pathway to increase autolysosomal degradation during aggrephagy. METHODS A rat model of myocardial I/R injury was constructed by left anterior descending (LAD) ligation-reperfusion. To verify cardiac protection, autolysosome clearance of protein aggregates, and their intracellular biological mechanism, an oxygen-glucose deprivation/recovery (OGD/R)-induced H9c2 cardiomyocyte model was created. RESULTS NCTS was found to have a significant cardioprotective effect in I/R rats as evidenced by remarkably improved pathological anatomy, decreased myocardial damage indicators, and substantially enhanced cardiac performance. Mechanistically, NCTS might boost the levels of LAMP2 mRNA and protein, total and Ser40 phosphorylated Nrf2, and Thr202/Tyr204p-ERK1/2 protein. Simultaneously, the cytoplasmic Nrf2 level was reduced after NCTS administration, which was contrary to the total Nrf2 content. However, these beneficial changes were reversed by the co-administration with ERK1/2 inhibitor, PD98059. NCTS therapy up-regulated Rab7 protein content, Cathepsin B activity, and lysosomal acidity, while down-regulating autophagosome numbers, Ubiquitin (Ub), and autophagosome marker protein accumulations through the above signaling pathway. This might indicate that NCTS enhanced lysosomal fusion and hydrolytic capacity. It was also found that NCTS intervention limited oxidative stress and cellular apoptosis both in vivo and in vitro. CONCLUSIONS We reported for the first time that NCTS promoted the autolysosome removal of protein aggregation both in vivo and in vitro, to exert the therapeutic advantages of myocardial I/R injury. This was reliant on the up-regulation of the ERK1/2-Nrf2-LAMP2 signaling pathway.
Collapse
Affiliation(s)
- Ye Yang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China; Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing 100700, China
| | - Mingyan Shao
- Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing 100700, China; School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Junkai Yao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China; Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing 100700, China
| | - Shuangjie Yang
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Wenkun Cheng
- Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing 100700, China; School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Lin Ma
- Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing 100700, China; School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Weili Li
- Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing 100700, China; School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jing Cao
- Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing 100700, China; School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yawen Zhang
- Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing 100700, China; School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yueyao Hu
- Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing 100700, China; School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Chun Li
- Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing 100700, China; Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Yong Wang
- Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing 100700, China; School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Wei Wang
- Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing 100700, China; Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| |
Collapse
|
12
|
Cobas Paz R, Caneiro Queija B, Íñiguez Romo A. No-reflow phenomenon in STEMI: beyond a good angiographic result. REVISTA ESPANOLA DE CARDIOLOGIA (ENGLISH ED.) 2022; 75:706-708. [PMID: 35623972 DOI: 10.1016/j.rec.2022.02.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 06/15/2023]
Affiliation(s)
- Rafael Cobas Paz
- Servicio de Cardiología, Hospital Álvaro Cunqueiro, Vigo, Pontevedra, Spain.
| | | | | |
Collapse
|
13
|
Kaluza GL, Creech JL, Furer A, Afari ME, Milewski K, Yi GH, Cheng Y, Conditt GB, McGregor JC, Blum D, Rousselle SD, Granada JF, Burkhoff D. Chronic myocardial and coronary arterial effects of intracoronary supersaturated oxygen therapy in swine with normal and ischemic-reperfused myocardium. Sci Rep 2022; 12:5785. [PMID: 35388096 PMCID: PMC8987078 DOI: 10.1038/s41598-022-09776-8] [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: 06/29/2021] [Accepted: 03/16/2022] [Indexed: 11/08/2022] Open
Abstract
The study assessed chronic myocardial, coronary and systemic effects of intracoronary supersaturated oxygen (SSO2) therapy. Left anterior descending coronary arteries of 40 swine were stented and randomized to 90-min selective intracoronary infusion of SSO2 (pO2 760-1000 mmHg) or normoxemic saline. In 20 out of 40 animals, SSO2 delivery followed a 60-min balloon occlusion to induce myocardial infarction (MI). In both normal and MI models, intracoronary treatment with hyperoxemic SSO2 therapy showed no evidence of coronary thrombosis. There were no biologically relevant differences between treatments at either time point in regard to coronary intervention site healing and neointimal growth. No signs of any myocardial or systemic toxicity were observed after 7 or 30 days. A trend was observed toward reduced incidence of microscopic MI scars and reduced infarct size in histopathology, as well as toward better recovery of echocardiographically evaluated global and regional contractility at 30 days. No treatment related infarcts or thromboemboli were observed in the downstream organs.
Collapse
Affiliation(s)
- Grzegorz L Kaluza
- Skirball Center for Innovation, Cardiovascular Research Foundation, Orangeburg, NY, USA.
| | | | - Ariel Furer
- Sheba Tel HaShomer City of Health, Ramat Gan, Israel
| | | | | | - Geng-Hua Yi
- Skirball Center for Innovation, Cardiovascular Research Foundation, Orangeburg, NY, USA
| | - Yanping Cheng
- Skirball Center for Innovation, Cardiovascular Research Foundation, Orangeburg, NY, USA
| | - Gerard B Conditt
- Skirball Center for Innovation, Cardiovascular Research Foundation, Orangeburg, NY, USA
| | - Jenn C McGregor
- Skirball Center for Innovation, Cardiovascular Research Foundation, Orangeburg, NY, USA
| | | | | | - Juan F Granada
- Skirball Center for Innovation, Cardiovascular Research Foundation, Orangeburg, NY, USA
| | - Daniel Burkhoff
- Skirball Center for Innovation, Cardiovascular Research Foundation, Orangeburg, NY, USA
| |
Collapse
|
14
|
Cobas Paz R, Caneiro Queija B, Íñiguez Romo A. Fenómeno de no-reflow en el IAMCEST: más allá de un buen resultado angiográfico. Rev Esp Cardiol 2022. [DOI: 10.1016/j.recesp.2022.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
15
|
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Jaganathan Subramani
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, Texas 79430
| | - Venkatesh Kundumani-Sridharan
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, Texas 79430
| | - Kumuda C Das
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, Texas 79430
| |
Collapse
|
16
|
Zhang XJ, Liu X, Hu M, Zhao GJ, Sun D, Cheng X, Xiang H, Huang YP, Tian RF, Shen LJ, Ma JP, Wang HP, Tian S, Gan S, Xu H, Liao R, Zou T, Ji YX, Zhang P, Cai J, Wang ZV, Meng G, Xu Q, Wang Y, Ma XL, Liu PP, Huang Z, Zhu L, She ZG, Zhang X, Bai L, Yang H, Lu Z, Li H. Pharmacological inhibition of arachidonate 12-lipoxygenase ameliorates myocardial ischemia-reperfusion injury in multiple species. Cell Metab 2021; 33:2059-2075.e10. [PMID: 34536344 DOI: 10.1016/j.cmet.2021.08.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 04/01/2020] [Accepted: 08/25/2021] [Indexed: 12/18/2022]
Abstract
Myocardial ischemia-reperfusion (MIR) injury is a major cause of adverse outcomes of revascularization after myocardial infarction. To identify the fundamental regulator of reperfusion injury, we performed metabolomics profiling in plasma of individuals before and after revascularization and identified a marked accumulation of arachidonate 12-lipoxygenase (ALOX12)-dependent 12-HETE following revascularization. The potent induction of 12-HETE proceeded by reperfusion was conserved in post-MIR in mice, pigs, and monkeys. While genetic inhibition of Alox12 protected mouse hearts from reperfusion injury and remodeling, Alox12 overexpression exacerbated MIR injury. Remarkably, pharmacological inhibition of ALOX12 significantly reduced cardiac injury in mice, pigs, and monkeys. Unexpectedly, ALOX12 promotes cardiomyocyte injury beyond its enzymatic activity and production of 12-HETE but also by its suppression of AMPK activity via a direct interaction with its upstream kinase TAK1. Taken together, our study demonstrates that ALOX12 is a novel AMPK upstream regulator in the post-MIR heart and that it represents a conserved therapeutic target for the treatment of myocardial reperfusion injury.
Collapse
Affiliation(s)
- Xiao-Jing Zhang
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Xiaolan Liu
- Institute of Model Animal of Wuhan University, Wuhan 430071, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Manli Hu
- Institute of Model Animal of Wuhan University, Wuhan 430071, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Guo-Jun Zhao
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Dating Sun
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Xu Cheng
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Hui Xiang
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Yong-Ping Huang
- Institute of Model Animal of Wuhan University, Wuhan 430071, China; College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Rui-Feng Tian
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Li-Jun Shen
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Jun-Peng Ma
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Hai-Ping Wang
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Song Tian
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Shanyu Gan
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Haibo Xu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Rufang Liao
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Toujun Zou
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Yan-Xiao Ji
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Peng Zhang
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Jingjing Cai
- Department of Cardiology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Zhao V Wang
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Guannan Meng
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China
| | - Qingbo Xu
- Centre for Clinic Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Yibin Wang
- Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Xin-Liang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19004, USA
| | - Peter P Liu
- Division of Cardiology, University of Ottawa Heart Institute, Ottawa, ON K1Y 4W7, Canada
| | - Zan Huang
- College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Lihua Zhu
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Zhi-Gang She
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Xin Zhang
- Gannan Institute of Translational Medicine, Ganzhou 341000, China
| | - Lan Bai
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China.
| | - Hailong Yang
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China.
| | - Zhibing Lu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan 430060, China.
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| |
Collapse
|
17
|
Roostalu U, Thisted L, Skytte JL, Salinas CG, Pedersen PJ, Hecksher-Sørensen J, Rolin B, Hansen HH, MacKrell JG, Christie RM, Vrang N, Jelsing J, Zois NE. Effect of captopril on post-infarction remodelling visualized by light sheet microscopy and echocardiography. Sci Rep 2021; 11:5241. [PMID: 33664407 PMCID: PMC7933438 DOI: 10.1038/s41598-021-84812-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 02/22/2021] [Indexed: 02/08/2023] Open
Abstract
Angiotensin converting enzyme inhibitors, among them captopril, improve survival following myocardial infarction (MI). The mechanisms of captopril action remain inadequately understood due to its diverse effects on multiple signalling pathways at different time periods following MI. Here we aimed to establish the role of captopril in late-stage post-MI remodelling. Left anterior descending artery (LAD) ligation or sham surgery was carried out in male C57BL/6J mice. Seven days post-surgery LAD ligated mice were allocated to daily vehicle or captopril treatment continued over four weeks. To provide comprehensive characterization of the changes in mouse heart following MI a 3D light sheet imaging method was established together with automated image analysis workflow. The combination of echocardiography and light sheet imaging enabled to assess cardiac function and the underlying morphological changes. We show that delayed captopril treatment does not affect infarct size but prevents left ventricle dilation and hypertrophy, resulting in improved ejection fraction. Quantification of lectin perfused blood vessels showed improved vascular density in the infarct border zone in captopril treated mice in comparison to vehicle dosed control mice. These results validate the applicability of combined echocardiographic and light sheet assessment of drug mode of action in preclinical cardiovascular research.
Collapse
Affiliation(s)
- Urmas Roostalu
- Gubra, Hørsholm Kongevej 11, B, 2970, Hørsholm, Denmark.
| | | | | | | | | | | | - Bidda Rolin
- Gubra, Hørsholm Kongevej 11, B, 2970, Hørsholm, Denmark
- Novo Nordisk, 2760, Maaloev, Denmark
| | | | - James G MacKrell
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Robert M Christie
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Niels Vrang
- Gubra, Hørsholm Kongevej 11, B, 2970, Hørsholm, Denmark
| | - Jacob Jelsing
- Gubra, Hørsholm Kongevej 11, B, 2970, Hørsholm, Denmark
| | | |
Collapse
|
18
|
The long noncoding RNA lncCIRBIL disrupts the nuclear translocation of Bclaf1 alleviating cardiac ischemia-reperfusion injury. Nat Commun 2021; 12:522. [PMID: 33483496 PMCID: PMC7822959 DOI: 10.1038/s41467-020-20844-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 12/22/2020] [Indexed: 11/08/2022] Open
Abstract
Cardiac ischemia-reperfusion (I/R) injury is a pathological process resulting in cardiomyocyte death. The present study aims to evaluate the role of the long noncoding RNA Cardiac Injury-Related Bclaf1-Inhibiting LncRNA (lncCIRBIL) on cardiac I/R injury and delineate its mechanism of action. The level of lncCIRBIL is reduced in I/R hearts. Cardiomyocyte-specific transgenic overexpression of lncCIRBIL reduces infarct area following I/R injury. Knockout of lncCIRBIL in mice exacerbates cardiac I/R injury. Qualitatively, the same results are observed in vitro. LncCIRBIL directly binds to BCL2-associated transcription factor 1 (Bclaf1), to inhibit its nuclear translocation. Cardiomyocyte-specific transgenic overexpression of Bclaf1 worsens, while partial knockout of Bclaf1 mitigates cardiac I/R injury. Meanwhile, partial knockout of Bclaf1 abrogates the detrimental effects of lncCIRBIL knockout on cardiac I/R injury. Collectively, the protective effect of lncCIRBIL on I/R injury is accomplished by inhibiting the nuclear translocation of Bclaf1. LncCIRBIL and Bclaf1 are potential therapeutic targets for ischemic cardiac disease.
Collapse
|
19
|
Rao Z, Shen D, Chen J, Jin L, Wu X, Chen M, Li L, Chu M, Lin J. Basic Fibroblast Growth Factor Attenuates Injury in Myocardial Infarction by Enhancing Hypoxia-Inducible Factor-1 Alpha Accumulation. Front Pharmacol 2020; 11:1193. [PMID: 32848793 PMCID: PMC7427464 DOI: 10.3389/fphar.2020.01193] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 07/22/2020] [Indexed: 11/13/2022] Open
Abstract
Background The combination of antiapoptotic and angiogenic actions may represent a pharmacotherapeutic strategy for the treatment of myocardial infarction. Fibroblast growth factor (FGF) is expressed in various cell types including endothelial and muscle cells and promotes their survival, migration, and proliferation. Methods and Results Myocardial microvascular endothelial cells were divided into four treatment groups, the sham, hypoxia, basic FGF (bFGF), and bFGF plus 2-methoxyestradiol groups, and subjected to in vitro apoptotic analysis and Matrigel assays. An in vivo model of myocardial infarction was established by ligaturing the left coronary artery of mice in the four treatment groups. Cardiac performance, myocardial injury, endothelial cell angiogenesis, and myocardial apoptosis were assessed. bFGF administration after myocardial infarction improved cardiac function and cell viability, attenuated myocardial injury and apoptosis, and enhanced angiogenesis. Western blotting of HIF-1α, p-AKT, VEGF, p53, BAX, and Bcl-2 showed that bFGF increased HIF-1α, p-AKT, VEGF, and Bcl-2 and decreased BAX protein levels. Conclusion The results of the present study indicated that bFGF attenuates myocardial injury by inhibiting apoptosis and promoting angiogenesis via a novel HIF-1α-mediated mechanism and a potential utility of bFGF in protecting against myocardial infarction.
Collapse
Affiliation(s)
- Zhiheng Rao
- Department of Cardiology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Clinical Medicine, The Second School of Wenzhou Medical University, Wenzhou, China
| | - Danping Shen
- Department of Clinical Medicine, The Second School of Wenzhou Medical University, Wenzhou, China.,Department of Pediatric Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiahui Chen
- Department of Cardiology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Clinical Medicine, The Second School of Wenzhou Medical University, Wenzhou, China.,Department of Cardiology, Taishun People's Hospital, Wenzhou, China
| | - Lushen Jin
- Department of Pediatric Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xueping Wu
- Department of Clinical Medicine, The Second School of Wenzhou Medical University, Wenzhou, China
| | - Ming Chen
- Department of Clinical Medicine, The First School of Wenzhou Medical University, Wenzhou, China
| | - Lei Li
- Department of Cardiology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Clinical Medicine, The Second School of Wenzhou Medical University, Wenzhou, China
| | - Maoping Chu
- Department of Clinical Medicine, The Second School of Wenzhou Medical University, Wenzhou, China.,Department of Pediatric Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiafeng Lin
- Department of Cardiology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Clinical Medicine, The Second School of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
20
|
Wang J, Lu L, Chen S, Xie J, Lu S, Zhou Y, Jiang H. Up-regulation of PERK/Nrf2/HO-1 axis protects myocardial tissues of mice from damage triggered by ischemia-reperfusion through ameliorating endoplasmic reticulum stress. Cardiovasc Diagn Ther 2020; 10:500-511. [PMID: 32695629 DOI: 10.21037/cdt-20-126] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Ischemia-reperfusion (I/R) injury, which leads to additionally cardiac tissue damage, is a severe adverse effect of reperfusion therapeutics used for the treatment of acute myocardial infarction. Agents capable of alleviating I/R-induced myocardial injury are urgently needed. In this study, we investigated whether up-regulation of PERK/Nrf2/HO-1 axis provided protective roles for murine myocardium suffering I/R intervention. Methods The in vivo I/R model was formed by ligation of the left anterior descending (LAD) coronary artery of C57BL/6J mice. All animals were assigned into the following groups at random: sham, I/R, rAAV9-PERK + I/R, rAAV9-Nrf2 + I/R, rAAV9-HO-1 + I/R, siRNA-HO-1 + rAAV9-PERK + I/R. The ligation of LAD was released after 30 min of ischemia, which was followed by reperfusion of LAD for 4 h. Then the cardiac tissues and blood serum were collected. TUNEL staining, ELISA assay, TTC staining, Western blotting and real-time PCR were used to determine I/R injury-related indicators. Results Our results showed that I/R administration triggered cardiomyocytes apoptosis and LDH and CK-MB release, yet overexpression of PERK decreased cellular apoptosis index in the cardiac tissue and reduced levels of LDH and CK-MB in the serum. We further found that the protective actions of PERK against I/R-evoked cardiac damage might be attributed to up-regulation of Nrf2/HO-1 signaling transduction, given that overexpression of Nrf2 and HO-1 ameliorated cardiac cell apoptosis and reduced the size of infarction and ischemia in the myocardial tissue, yet gene silencing of HO-1 invalidated the beneficial roles of PERK overexpression in improving I/R-induced cardiac injury. Then, we investigated whether PERK-activated Nrf2/HO-1 cascade affected endoplasmic reticulum stress (ERS), considering the crucial roles of ERS-associated apoptosis in the development of I/R damage. Our findings indicated that up-regulation of PERK-mediated Nrf2/HO-1 pathway induced the expression reduction of GRP78, CRT, CHOP and caspase-12 both at the transcriptional and translational level. Conclusions We, for the first time, discovered that up-regulation of PERK/Nrf2/HO-1 axis improved I/R-induced myocardial injury via reducing ERS-related signal molecules and downstream pro-apoptotic factors.
Collapse
Affiliation(s)
- Jichun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Li Lu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Sisi Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jing Xie
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Shuai Lu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yanli Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
21
|
Chen S, David SW, Khan ZA, Metzger DC, Wasserman HS, Lotfi AS, Hanson ID, Dixon SR, LaLonde TA, Généreux P, Ozan MO, Maehara A, Stone GW. One-year outcomes of supersaturated oxygen therapy in acute anterior myocardial infarction: The IC-HOT study. Catheter Cardiovasc Interv 2020; 97:1120-1126. [PMID: 32649037 PMCID: PMC8246818 DOI: 10.1002/ccd.29090] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 05/26/2020] [Accepted: 05/31/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Supersaturated oxygen (SSO2 ) has recently been approved by the U.S. Food and Drug Administration for administration after primary percutaneous coronary intervention (pPCI) in patients with anterior ST-segment elevation myocardial infarction (STEMI) based on its demonstration of infarct size reduction in the IC-HOT study. OBJECTIVES To describe the 1-year clinical outcomes of intracoronary SSO2 treatment after pPCI in patients with anterior STEMI. METHODS IC-HOT was a prospective, open-label, single-arm study in which 100 patients without cardiogenic shock undergoing successful pPCI of an occluded left anterior descending coronary artery were treated with a 60-min SSO2 infusion. One-year clinical outcomes were compared with a propensity-matched control group of similar patients with anterior STEMI enrolled in the INFUSE-AMI trial. RESULTS Baseline and postprocedural characteristics were similar in the two groups except for pre-PCI thrombolysis in myocardial infarction 3 flow, which was less prevalent in patients treated with SSO2 (9.6% vs. 22.9%, p = .02). Treatment with SSO2 was associated with a lower 1-year rate of the composite endpoint of all-cause death or new-onset heart failure (HF) or hospitalization for HF (0.0% vs. 12.3%, p = .001). All-cause mortality, driven by cardiovascular mortality, and new-onset HF or HF hospitalization were each individually lower in SSO2 -treated patients. There were no significant differences between groups in the 1-year rates of reinfarction or clinically driven target vessel revascularization. CONCLUSIONS Infusion of SSO2 following pPCI in patients with anterior STEMI was associated with improved 1-year clinical outcomes including lower rates of death and new-onset HF or HF hospitalizations.
Collapse
Affiliation(s)
- Shmuel Chen
- Clinical Trials Center, Cardiovascular Research Foundation, New York.,Division of Cardiology, NewYork-Presbyterian Hospital/Columbia University Irving Medical Center, New York
| | | | | | | | | | - Amir S Lotfi
- Baystate Medical Center, Springfield, Massachusetts
| | | | | | - Thomas A LaLonde
- Ascension St. John Hospital, Detroit, Michigan.,Wayne State University School of Medicine, Detroit, Michigan
| | - Philippe Généreux
- Clinical Trials Center, Cardiovascular Research Foundation, New York.,Gagnon Cardiovascular Institute, Morristown Medical Center, Morristown, New Jersey.,Hôpital du Sacré-Coeur de Montréal, Montréal, Québec, Canada
| | - M Ozgu Ozan
- Clinical Trials Center, Cardiovascular Research Foundation, New York
| | - Akiko Maehara
- Clinical Trials Center, Cardiovascular Research Foundation, New York.,Division of Cardiology, NewYork-Presbyterian Hospital/Columbia University Irving Medical Center, New York
| | - Gregg W Stone
- Clinical Trials Center, Cardiovascular Research Foundation, New York.,The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York
| |
Collapse
|
22
|
Zeng H, He X, Chen JX. Endothelial Sirtuin 3 Dictates Glucose Transport to Cardiomyocyte and Sensitizes Pressure Overload-Induced Heart Failure. J Am Heart Assoc 2020; 9:e015895. [PMID: 32468895 PMCID: PMC7428981 DOI: 10.1161/jaha.120.015895] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Background Alterations of energetic metabolism are suggested to be an important contributor to pressure overload (PO)‐induced heart failure. Our previous study reveals that knockout of endothelial Sirtuin 3 (SIRT3) alters glycolysis and impairs diastolic function. We hypothesize that endothelial SIRT3 regulates glucose utilization of cardiomyocytes and sensitizes PO‐induced heart failure. Methods and Results SIRT3 endothelial cell knockout mice and their control SIRT3 LoxP mice were subjected to PO by transverse aortic constriction for 7 weeks. The ratio of heart weight to tibia length was increased in both strains of mice, in which SIRT3 endothelial cell knockout mice+transverse aortic constriction exhibited more severe cardiac hypertrophy. Coronary blood flow and systolic function were significantly decreased in SIRT3 endothelial cell knockout mice+transverse aortic constriction compared with SIRT3 LoxP mice+transverse aortic constriction, as evidenced by lower systolic/diastolic ratio, ejection fraction, and fractional shortening. PO‐induced upregulation of apelin and glucose transporter 4 were significantly reduced in the hearts of SIRT3 endothelial cell knockout mice. In vitro, levels of hypoxia‐inducible factor‐1α and glucose transporter 1 and glucose uptake were significantly reduced in SIRT3 knockout endothelial cells. Furthermore, hypoxia‐induced apelin expression was abolished together with reduced apelin‐mediated glucose uptake in SIRT3 knockout endothelial cells. Exposure of cardiomyocyte with apelin increased expression of glucose transporter 1 and glucose transporter 4. This was accompanied by a significant increase in glycolysis. Supplement of apelin in SIRT3 knockout hypoxic endothelial cell media increased glycolysis in the cardiomyocytes. Conclusions Knockout of SIRT3 disrupts glucose transport from endothelial cells to cardiomyocytes, reduces cardiomyocyte glucose utilization via apelin in a paracrine manner, and sensitizes PO‐induced heart failure. Endothelial SIRT3 may regulate cardiomyocyte glucose availability and govern the function of the heart.
Collapse
Affiliation(s)
- Heng Zeng
- Department of Pharmacology and Toxicology School of Medicine University of Mississippi Medical Center Jackson MS
| | - Xiaochen He
- Department of Pharmacology and Toxicology School of Medicine University of Mississippi Medical Center Jackson MS
| | - Jian-Xiong Chen
- Department of Pharmacology and Toxicology School of Medicine University of Mississippi Medical Center Jackson MS
| |
Collapse
|
23
|
Xu Y, Tang C, Tan S, Duan J, Tian H, Yang Y. Cardioprotective effect of isorhamnetin against myocardial ischemia reperfusion (I/R) injury in isolated rat heart through attenuation of apoptosis. J Cell Mol Med 2020; 24:6253-6262. [PMID: 32307912 PMCID: PMC7294129 DOI: 10.1111/jcmm.15267] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/19/2020] [Accepted: 03/26/2020] [Indexed: 12/16/2022] Open
Abstract
In this study, we investigated the effects of isorhamnetin on myocardial ischaemia reperfusion (I/R) injury in Langendorff‐perfused rat hearts. Isorhamnetin treatment (5, 10 and 20 μg/mL) significantly alleviated cardiac morphological injury, reduced myocardial infarct size, decreased the levels of marker enzymes (LDH and CK) and improved the haemodynamic parameters, reflected by the elevated levels of the left ventricular developed pressure (LVDP), coronary flow (CF) and the maximum up/down velocity of left ventricular pressure (+dp/dtmax). Moreover, isorhamnetin reperfusion inhibited apoptosis of cardiomyocytes in the rats subjected to cardiac I/R in a dose‐dependent manner concomitant with decreased protein expression of Bax and cleaved‐caspase‐3, as well as increased protein expression of Bcl‐2. In addition, I/R‐induced oxidative stress was manifestly mitigated by isorhamnetin treatment, as showed by the decreased malondialdehyde (MDA) level and increased antioxidant enzymes activities of superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GSH‐Px). These results indicated that isorhamnetin exerts a protective effect against I/R‐induced myocardial injury through the attenuation of apoptosis and oxidative stress.
Collapse
Affiliation(s)
- Yan Xu
- Department of Geriatrics, the Second Xiangya Hospital, Central South University, Changsha, PR China
| | - Chun Tang
- Department of Nephrology, Center of Nephrology and Urology, the Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, PR China
| | - Shengyu Tan
- Department of Geriatrics, the Second Xiangya Hospital, Central South University, Changsha, PR China
| | - Juan Duan
- Department of Geriatrics, the Second Xiangya Hospital, Central South University, Changsha, PR China
| | - Hongmei Tian
- Department of Geriatrics, the Second Xiangya Hospital, Central South University, Changsha, PR China
| | - Yu Yang
- Department of Geriatrics, the Second Xiangya Hospital, Central South University, Changsha, PR China
| |
Collapse
|
24
|
Hofmann R, Witt N, Lagerqvist B, Jernberg T, Lindahl B, Erlinge D, Herlitz J, Alfredsson J, Linder R, Omerovic E, Angerås O, Venetsanos D, Kellerth T, Sparv D, Lauermann J, Barmano N, Verouhis D, Östlund O, Svensson L, James SK. Oxygen therapy in ST-elevation myocardial infarction. Eur Heart J 2019; 39:2730-2739. [PMID: 29912429 DOI: 10.1093/eurheartj/ehy326] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 05/21/2018] [Indexed: 01/05/2023] Open
Abstract
Aims To determine whether supplemental oxygen in patients with ST-elevation myocardial infarction (STEMI) impacts on procedure-related and clinical outcomes. Methods and results The DETermination of the role of Oxygen in suspected Acute Myocardial Infarction (DETO2X-AMI) trial randomized patients with suspected myocardial infarction (MI) to receive oxygen at 6 L/min for 6-12 h or ambient air. In this pre-specified analysis, we included only STEMI patients who underwent percutaneous coronary intervention (PCI). In total, 2807 patients were included, 1361 assigned to receive oxygen, and 1446 assigned to ambient air. The pre-specified primary composite endpoint of all-cause death, rehospitalization with MI, cardiogenic shock, or stent thrombosis at 1 year occurred in 6.3% (86 of 1361) of patients allocated to oxygen compared to 7.5% (108 of 1446) allocated to ambient air [hazard ratio (HR) 0.85, 95% confidence interval (95% CI) 0.64-1.13; P = 0.27]. There was no difference in the rate of death from any cause (HR 0.86, 95% CI 0.61-1.22; P = 0.41), rate of rehospitalization for MI (HR 0.92, 95% CI 0.57-1.48; P = 0.73), rehospitalization for cardiogenic shock (HR 1.05, 95% CI 0.21-5.22; P = 0.95), or stent thrombosis (HR 1.27, 95% CI 0.46-3.51; P = 0.64). The primary composite endpoint was consistent across all subgroups, as well as at different time points, such as during hospital stay, at 30 days and the total duration of follow-up up to 1356 days. Conclusions Routine use of supplemental oxygen in normoxemic patients with STEMI undergoing primary PCI did not significantly affect 1-year all-cause death, rehospitalization with MI, cardiogenic shock, or stent thrombosis.
Collapse
Affiliation(s)
- Robin Hofmann
- Division of Cardiology, Department of Clinical Science and Education, Karolinska Institutet, Södersjukhuset, Sjukhusbacken 10, Stockholm, Sweden
| | - Nils Witt
- Division of Cardiology, Department of Clinical Science and Education, Karolinska Institutet, Södersjukhuset, Sjukhusbacken 10, Stockholm, Sweden
| | - Bo Lagerqvist
- Cardiology, Department of Medical Sciences, Uppsala University, Akademiska sjukhuset, Entrance 40, floor 5, Uppsala, Sweden
| | - Tomas Jernberg
- Cardiology, Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, Mörbygårdsvägen 5, Stockholm, Sweden
| | - Bertil Lindahl
- Cardiology, Department of Medical Sciences, Uppsala University, Akademiska sjukhuset, Entrance 40, floor 5, Uppsala, Sweden.,Uppsala Clinical Research Center, Uppsala University, Dag Hammarskjölds väg 38, Uppsala, Sweden
| | - David Erlinge
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden
| | - Johan Herlitz
- Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden.,Department of Health Sciences, University of Borås, Borås, Sweden
| | - Joakim Alfredsson
- Department of Cardiology, Linköping University Hospital, Linköping, Sweden
| | - Rikard Linder
- Cardiology, Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, Mörbygårdsvägen 5, Stockholm, Sweden
| | - Elmir Omerovic
- Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Oskar Angerås
- Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Dimitrios Venetsanos
- Department of Medical and Health Sciences, Linköping University, Sandbäcksgatan 7, Linköping, Sweden.,Department of Cardiology, Linköping University Hospital, Linköping, Sweden
| | - Thomas Kellerth
- Department of Cardiology, Örebro University Hospital, Örebro, Sweden
| | - David Sparv
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden
| | - Jörg Lauermann
- Division of Cardiology, Department of Internal Medicine, Ryhov Hospital, Sjukhusgatan, Jönköping, Sweden
| | - Neshro Barmano
- Division of Cardiology, Department of Internal Medicine, Ryhov Hospital, Sjukhusgatan, Jönköping, Sweden
| | - Dinos Verouhis
- Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.,Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| | - Ollie Östlund
- Uppsala Clinical Research Center, Uppsala University, Dag Hammarskjölds väg 38, Uppsala, Sweden
| | - Leif Svensson
- Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.,Centre for Resuscitation Science, Karolinska Institutet, Södersjukhuset, Jägargatan 20, Stockholm, Sweden
| | - Stefan K James
- Cardiology, Department of Medical Sciences, Uppsala University, Akademiska sjukhuset, Entrance 40, floor 5, Uppsala, Sweden.,Uppsala Clinical Research Center, Uppsala University, Dag Hammarskjölds väg 38, Uppsala, Sweden
| | | |
Collapse
|
25
|
A predictive computational model to estimate myocardial temperature during intracoronary hypothermia in acute myocardial infarction. Med Eng Phys 2019; 68:65-75. [DOI: 10.1016/j.medengphy.2019.03.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 03/15/2019] [Accepted: 03/31/2019] [Indexed: 11/20/2022]
|
26
|
Yang C, Li B, Liu Y, Xing Y. Ginsenoside Rb1 protects cardiomyocytes from oxygen-glucose deprivation injuries by targeting microRNA-21. Exp Ther Med 2019; 17:3709-3716. [PMID: 30988756 PMCID: PMC6447784 DOI: 10.3892/etm.2019.7330] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 08/09/2018] [Indexed: 12/11/2022] Open
Abstract
Ginsenoside Rb1 (GS-Rb1) is one of the most important active pharmacological extracts of the traditional Chinese medicine, ginseng, and there is extensive evidence of its cardioprotective properties. However, the microRNA (miR) targets of GS-Rb1 and the underlying mechanisms of GS-Rb1 and miR-21 in the progression of cardiomyocyte apoptosis have not been clearly elucidated. The aim of the current study was to investigate the impact of miR-21 and its target gene, programmed cell death protein 4 (PDCD4), on the protective effect of GS-Rb1 in cardiomyocytes injured by oxygen-glucose deprivation (OGD). The miR-21 expression levels were downregulated, and the percentage of the apoptotic cells and reactive oxygen species (ROS) was increased in OGD-cultured neonatal rat cardiomyocytes; however, the effects were reversed by GS-Rb1 treatment. It was demonstrated that GS-Rb1 could reduce intracellular ROS content, and the expression of cytochrome C and the pro-apoptosis protein, apoptosis regulator B-cell lymphoma associated X (Bax) protein while increasing the expression of the anti-apoptosis protein, apoptosis regulator Bcl-2. The target gene, PDCD4, was significantly upregulated in the OGD group; however, the expression of PDCD4 was inhibited by GS-Rb1 treatment. Furthermore, miR-21 inhibitor transfection reduced GS-Rb1-induced miR-21 upregulation compared with the OGD+GS-Rb1 group, indicating that the miR-21 was involved in the anti-apoptotic effect of GS-Rb1 in cardiomyocytes. The results of the current study highlighted that GS-Rb1 could target miR-21 and its target gene, PDCD4, to protect OGD-injured cardiomyocytes. The results of the current study may provide a novel insight for the treatment of myocardial infarction with Traditional Chinese Medicines, involving miRs as targets.
Collapse
Affiliation(s)
- Chuang Yang
- Department of Cardiology, Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Bo Li
- Department of Cardiology, Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Yongsheng Liu
- Department of Cardiology, Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Yue Xing
- Department of Cardiology, Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| |
Collapse
|
27
|
Houang EM, Bartos J, Hackel BJ, Lodge TP, Yannopoulos D, Bates FS, Metzger JM. Cardiac Muscle Membrane Stabilization in Myocardial Reperfusion Injury. JACC Basic Transl Sci 2019; 4:275-287. [PMID: 31061929 PMCID: PMC6488758 DOI: 10.1016/j.jacbts.2019.01.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 01/11/2019] [Accepted: 01/26/2019] [Indexed: 12/11/2022]
Abstract
The phospholipid bilayer membrane that surrounds each cell in the body represents the first and last line of defense for preserving overall cell viability. In several forms of cardiac and skeletal muscle disease, deficits in the integrity of the muscle membrane play a central role in disease pathogenesis. In Duchenne muscular dystrophy, an inherited and uniformly fatal disease of progressive muscle deterioration, muscle membrane instability is the primary cause of disease, including significant heart disease, for which there is no cure or highly effective treatment. Further, in multiple clinical forms of myocardial ischemia-reperfusion injury, the cardiac sarcolemma is damaged and this plays a key role in disease etiology. In this review, cardiac muscle membrane stability is addressed, with a focus on synthetic block copolymers as a unique chemical-based approach to stabilize damaged muscle membranes. Recent advances using clinically relevant small and large animal models of heart disease are discussed. In addition, mechanistic insights into the copolymer-muscle membrane interface, featuring atomistic, molecular, and physiological structure-function approaches are highlighted. Collectively, muscle membrane instability contributes significantly to morbidity and mortality in prominent acquired and inherited heart diseases. In this context, chemical-based muscle membrane stabilizers provide a novel therapeutic approach for a myriad of heart diseases wherein the integrity of the cardiac muscle membrane is at risk.
Collapse
Affiliation(s)
- Evelyne M. Houang
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Jason Bartos
- Department of Medicine-Cardiovascular Division, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Benjamin J. Hackel
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota
| | - Timothy P. Lodge
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota
| | - Demetris Yannopoulos
- Department of Medicine-Cardiovascular Division, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Frank S. Bates
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota
| | - Joseph M. Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota
| |
Collapse
|
28
|
Patel MD, Platts-Mills TF, Grover JM, Thomas SM, Rossi JS. Feasibility of prehospital delivery of remote ischemic conditioning by emergency medical services in chest pain patients: protocol for a pilot study. Pilot Feasibility Stud 2019; 5:42. [PMID: 30911405 PMCID: PMC6415490 DOI: 10.1186/s40814-019-0431-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 03/06/2019] [Indexed: 12/22/2022] Open
Abstract
Background Remote ischemic conditioning (RIC) is a non-invasive procedure with hypothesized therapeutic benefits for patients experiencing an acute ST-elevation myocardial infarction (STEMI). Further study of emergency medical services (EMS) delivery of RIC in the prehospital setting is needed to inform the design and methods for future clinical trials of RIC in STEMI patients. The main objective of this pilot study is to assess the feasibility of prehospital delivery of RIC by EMS providers in the United States. Methods We will conduct a single-arm study of the standard RIC procedure (i.e., up to 4 cycles of alternating 5-min inflation and 5-min deflation of an upper arm cuff) administered by EMS paramedics in 50 patients experiencing acute onset chest pain. The investigational autoRIC® device (CellAegis Devices, Inc., Toronto, Ontario) will be initiated by paramedics during ground ambulance transport. Automated RIC cycles will continue through emergency department arrival and stay. The primary endpoint will be the completion of all 4 cycles of RIC without interruption. We will also examine study procedures and collect qualitative data from study participants and paramedics. Discussion To our knowledge, this will be the first study in the United States to assess the feasibility of completing the 40-min RIC procedure when initiated during ground ambulance transport. Findings from this pilot study will be used to optimize the design and methods for a future efficacy trial of RIC in acute STEMI patients. Trial registration NCT03400579 (ClinicalTrials.gov). Registered on 17 January 2018.
Collapse
Affiliation(s)
- Mehul D Patel
- 1Department of Emergency Medicine, University of North Carolina at Chapel Hill, 170 Manning Drive, CB #7594, Chapel Hill, NC 27599-7594 USA
| | - Timothy F Platts-Mills
- 1Department of Emergency Medicine, University of North Carolina at Chapel Hill, 170 Manning Drive, CB #7594, Chapel Hill, NC 27599-7594 USA
| | - Joseph M Grover
- 1Department of Emergency Medicine, University of North Carolina at Chapel Hill, 170 Manning Drive, CB #7594, Chapel Hill, NC 27599-7594 USA.,Orange County Emergency Services, Hillsborough, USA
| | - Sonia M Thomas
- 3Division of Biostatistics and Epidemiology, RTI International, Raleigh, USA
| | - Joseph S Rossi
- 4Division of Cardiology, University of North Carolina at Chapel Hill, Chapel Hill, USA
| |
Collapse
|
29
|
Cheng Y, Liu DZ, Zhang CX, Cui H, Liu M, Zhang BL, Mei QB, Lu ZF, Zhou SY. Mitochondria-targeted antioxidant delivery for precise treatment of myocardial ischemia–reperfusion injury through a multistage continuous targeted strategy. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 16:236-249. [DOI: 10.1016/j.nano.2018.12.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 11/29/2018] [Accepted: 12/28/2018] [Indexed: 02/06/2023]
|
30
|
Wei SJ, Luan FY, He DY, Xu F, Chen YG. Intracoronary administration of nicorandil-induced cardiac arrest during primary percutaneous coronary intervention: A case report. Medicine (Baltimore) 2019; 98:e14473. [PMID: 30762768 PMCID: PMC6408065 DOI: 10.1097/md.0000000000014473] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
RATIONALE Primary percutaneous coronary intervention (PPCI) is the most effective therapy for patients with an acute ST-segment elevation myocardial infarction (STEMI). However, up to half of STEMI patients suffer from coronary microvascular dysfunction, presenting as the slow flow or no-reflow phenomenon. PATIENTS CONCERNS A 78-year-old man was admitted to the chest pain center with sudden chest pain and tightness for about an hour. DIAGNOSES Electrocardiography demonstrated ST-segment elevation in leads II, III, aVF, and third-degree atrioventricular block. Coronary angiography showed acute total occlusion in the distal right coronary artery (RCA). INTERVENTIONS PPCI was performed on the patient. After thrombus aspiration, a stent was placed in the distal RCA. As coronary angiography showed TIMI grade 2 flow in RCA, 6 mg nicorandil was intracoronary administrated in twice. Immediately, cardiac arrest occurred and cardiopulmonary resuscitation (CPR) was performed. OUTCOMES The patient survived and had a good outcome during follow-up for >6 months. LESSONS Up to now, there has been no case report of cardiac arrest caused by nicorandil. Although intracoronary nicorandil is one of the most commonly used methods to improve coronary flow, much more attention should be paid to side effects of nicorandil.
Collapse
Affiliation(s)
- Shu-Jian Wei
- Department of Emergency, Qilu Hospital
- Chest Pain Center, Qilu Hospital
- Institute of Emergency and Critical Care Medicine
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, China
| | | | - Da-yu He
- Department of Emergency, Qilu Hospital
- Chest Pain Center, Qilu Hospital
- Institute of Emergency and Critical Care Medicine
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, China
| | - Feng Xu
- Department of Emergency, Qilu Hospital
- Chest Pain Center, Qilu Hospital
- Institute of Emergency and Critical Care Medicine
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, China
| | - Yu-Guo Chen
- Department of Emergency, Qilu Hospital
- Chest Pain Center, Qilu Hospital
- Institute of Emergency and Critical Care Medicine
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, China
| |
Collapse
|
31
|
David SW, Khan ZA, Patel NC, Metzger DC, Wood FO, Wasserman HS, Lotfi AS, Hanson ID, Dixon SR, LaLonde TA, Généreux P, Ozan MO, Maehara A, Stone GW. Evaluation of intracoronary hyperoxemic oxygen therapy in acute anterior myocardial infarction: The IC‐HOT study. Catheter Cardiovasc Interv 2018; 93:882-890. [DOI: 10.1002/ccd.27905] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/20/2018] [Accepted: 08/29/2018] [Indexed: 01/30/2023]
Affiliation(s)
| | - Zubair A. Khan
- Providence‐Providence Park Hospital Southfield Michigan
- North Alabama Medical Center Florence Alabama
| | | | | | | | | | | | | | | | | | - Philippe Généreux
- Gagnon Cardiovascular Institute, Morristown Medical Center Morristown New Jersey
- Hôpital du Sacré‐Coeur de Montréal Montréal Québec Canada
- Cardiovascular Research Foundation New York New York
| | | | - Akiko Maehara
- Cardiovascular Research Foundation New York New York
- Columbia University Medical Center New York New York
| | - Gregg W. Stone
- Cardiovascular Research Foundation New York New York
- Columbia University Medical Center New York New York
| |
Collapse
|
32
|
Zhang YS, Tang LJ, Tu H, Wang SJ, Liu B, Zhang XJ, Li NS, Luo XJ, Peng J. Fasudil ameliorates the ischemia/reperfusion oxidative injury in rat hearts through suppression of myosin regulatory light chain/NADPH oxidase 2 pathway. Eur J Pharmacol 2018; 822:1-12. [PMID: 29337194 DOI: 10.1016/j.ejphar.2018.01.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Revised: 12/17/2017] [Accepted: 01/10/2018] [Indexed: 11/18/2022]
Abstract
Fasudil is a potent Rho-kinase (ROCK) inhibitor and can relax smooth muscle or cardiac muscle contraction through decreasing the phosphorylation level of myosin regulatory light chain (p-MLC20 or p-MLC2v), while p-MLC2v can function as a transcription factor to promote the NADPH oxidase 2 (NOX2) expression in rat hearts subjected to ischemia/reperfusion (I/R). This study aims to explore whether fasudil can protect the rat hearts against I/R oxidative injury through suppressing NOX2 expression via reduction of p-MLC2v level. The SD rat hearts were subjected to 1h-ischemia plus 3h-reperfusion, which showed myocardial injuries (myocardial fiber loss and disarray, increase of creatine kinase release and myocardial infarction/apoptosis), increase in ROCK activity and nuclear p-MLC2v level concomitant with up-regulation of NOX2 and H2O2 production; these phenomena were attenuated by fasudil in a dose-dependent manner. Next, we verified the cardioprotective effect of fasudil and the underlying mechanisms in hypoxia-reoxygenation (H/R) -treated H9c2 cells. Consistent with the results in vivo, the H/R-treated H9c2 cells showed cellular injury (increase in apoptotic ratio), elevation in ROCK activity and nuclear p-MLC2v level, accompanied by up-regulation of NOX2 and H2O2 production; these effects were blocked in the presence of fasudil in a dose-dependent way. Based on these observations, we conclude that beneficial effect of fasudil against myocardial I/R or H/R oxidative injury is related to the suppression of NOX2 expression through decrease of the p-MLC2v level. Our findings also highlight that intervention of MLC2v phosphorylation by drugs may provide a novel strategy to protect heart from I/R oxidative injury.
Collapse
Affiliation(s)
- Yi-Shuai Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Li-Jing Tang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Hua Tu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Shi-Jing Wang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Bin Liu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Xiao-Jie Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Nian-Sheng Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Xiu-Ju Luo
- Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Jun Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China.
| |
Collapse
|
33
|
Otterspoor LC, van Nunen LX, van ‘t Veer M, Johnson NP, Pijls NH. Intracoronary Hypothermia Before Reperfusion to Reduce Reperfusion Injury in Acute Myocardial Infarction: A Novel Hypothesis and Technique. Ther Hypothermia Temp Manag 2017; 7:199-205. [DOI: 10.1089/ther.2017.0006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Affiliation(s)
- Luuk C. Otterspoor
- Department of Cardiology, Catharina Hospital, Eindhoven, the Netherlands
| | | | - Marcel van ‘t Veer
- Department of Cardiology, Catharina Hospital, Eindhoven, the Netherlands
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Nils P. Johnson
- Division of Cardiology, Weatherhead PET Center, McGovern Medical School, UTHealth and Memorial Hermann Hospital, Houston, Texas
| | - Nico H.J. Pijls
- Department of Cardiology, Catharina Hospital, Eindhoven, the Netherlands
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| |
Collapse
|
34
|
Otterspoor L, Van 't Veer M, Van Nunen L, Brueren G, Tonino P, Wijnbergen I, Helmes H, Zimmermann F, Van Hagen E, Johnson N, Pijls N. Safety and feasibility of selective intracoronary hypothermia in acute myocardial infarction. EUROINTERVENTION 2017; 13:e1475-e1482. [DOI: 10.4244/eij-d-17-00240] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
35
|
Cyrne Carvalho H. Microvascular reperfusion in myocardial infarction: The new concept of the open artery in the 21st century. Rev Port Cardiol 2017; 36:743-745. [PMID: 29037834 DOI: 10.1016/j.repc.2017.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Affiliation(s)
- Henrique Cyrne Carvalho
- Centro Hospitalar e Universitário do Porto, Porto, Portugal; Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal.
| |
Collapse
|
36
|
Microvascular reperfusion in myocardial infarction: The new concept of the open artery in the 21st century. REVISTA PORTUGUESA DE CARDIOLOGIA (ENGLISH EDITION) 2017. [DOI: 10.1016/j.repce.2017.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
37
|
Granger DN, Kvietys PR. Reperfusion therapy-What's with the obstructed, leaky and broken capillaries? ACTA ACUST UNITED AC 2017; 24:213-228. [PMID: 29102280 DOI: 10.1016/j.pathophys.2017.09.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Microvascular dysfunction is well established as an early and rate-determining factor in the injury response of tissues to ischemia and reperfusion (I/R). Severe endothelial cell dysfunction, which can develop without obvious morphological cell injury, is a major underlying cause of the microvascular abnormalities that accompany I/R. While I/R-induced microvascular dysfunction is manifested in different ways, two responses that have received much attention in both the experimental and clinical setting are impaired capillary perfusion (no-reflow) and endothelial barrier failure with a transition to hemorrhage. These responses are emerging as potentially important determinants of the severity of the tissue injury response, and there is growing clinical evidence that they are predictive of clinical outcome following reperfusion therapy. This review provides a summary of animal studies that have focused on the mechanisms that may underlie the genesis of no-reflow and hemorrhage following reperfusion of ischemic tissues, and addresses the clinical evidence that implicates these vascular events in the responses of the ischemic brain (stroke) and heart (myocardial infarction) to reperfusion therapy. Inasmuch as reactive oxygen species (ROS) and matrix metalloproteinases (MMP) are frequently invoked as triggers of the microvascular dysfunction elicited by I/R, the potential roles and sources of these mediators are also discussed. The available evidence in the literature justifies the increased interest in the development of no-reflow and hemorrhage in heart and brain following reperfusion therapy, and suggests that these vascular events may be predictive of poor clinical outcome and warrant the development of targeted treatment strategies.
Collapse
Affiliation(s)
- D Neil Granger
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, United States.
| | - Peter R Kvietys
- Department of Physiological Sciences, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| |
Collapse
|
38
|
Zhu Y, Matsumura Y, Wagner WR. Ventricular wall biomaterial injection therapy after myocardial infarction: Advances in material design, mechanistic insight and early clinical experiences. Biomaterials 2017; 129:37-53. [PMID: 28324864 PMCID: PMC5827941 DOI: 10.1016/j.biomaterials.2017.02.032] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 02/07/2017] [Accepted: 02/26/2017] [Indexed: 12/11/2022]
Abstract
Intramyocardial biomaterial injection therapy for myocardial infarction has made significant progress since concept initiation more than 10 years ago. The interim successes and progress in the first 5 years have been extensively reviewed. During the last 5 years, two phase II clinical trials have reported their long term follow up results and many additional biomaterial candidates have reached preclinical and clinical testing. Also in recent years deeper investigations into the mechanisms behind the beneficial effects associated with biomaterial injection therapy have been pursued, and a variety of process and material parameters have been evaluated for their impact on therapeutic outcomes. This review explores the advances made in this biomaterial-centered approach to ischemic cardiomyopathy and discusses potential future research directions as this therapy seeks to positively impact patients suffering from one of the world's most common sources of mortality.
Collapse
Affiliation(s)
- Yang Zhu
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Yasumoto Matsumura
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - William R Wagner
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15219, USA; Department of Surgery, University of Pittsburgh, Pittsburgh, PA, 15219, USA; Department of Chemical Engineering, University of Pittsburgh, Pittsburgh, PA, 15219, USA.
| |
Collapse
|
39
|
Zhong CB, Chen X, Zhou XY, Wang XB. The Role of Peroxisome Proliferator-Activated Receptor γ in Mediating Cardioprotection Against Ischemia/Reperfusion Injury. J Cardiovasc Pharmacol Ther 2017; 23:46-56. [PMID: 28466688 DOI: 10.1177/1074248417707049] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Myocardial infarction (MI) is a serious cardiovascular disease resulting in high rates of morbidity and mortality. Although advances have been made in restoring myocardial perfusion in ischemic areas, decreases in cardiomyocyte death and infarct size are still limited, attributing to myocardial ischemia/reperfusion (I/R) injury. It is necessary to develop therapies to restrict myocardial I/R injury and protect cardiomyocytes against further damage after MI. Many studies have suggested that peroxisome proliferator-activated receptor γ (PPARγ), a ligand-inducible nuclear receptor that predominantly regulates glucose and lipid metabolism, is a promising therapeutic target for ameliorating myocardial I/R injury. Thus, this review focuses on the role of PPARγ in cardioprotection during myocardial I/R. The cardioprotective effects of PPARγ, including attenuating oxidative stress, inhibiting inflammatory responses, improving glucose and lipid metabolism, and antagonizing apoptosis, are described. Additionally, the underlying mechanisms of cardioprotective effects of PPARγ, such as regulating the expression of target genes, influencing other transcription factors, and modulating kinase signaling pathways, are further discussed.
Collapse
Affiliation(s)
- Chong-Bin Zhong
- 1 The Second Clinical Institute of Southern Medical University, Guangzhou, China
| | - Xi Chen
- 1 The Second Clinical Institute of Southern Medical University, Guangzhou, China
| | - Xu-Yue Zhou
- 1 The Second Clinical Institute of Southern Medical University, Guangzhou, China
| | - Xian-Bao Wang
- 2 Department of Cardiology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
40
|
Babadjouni RM, Walcott BP, Liu Q, Tenser MS, Amar AP, Mack WJ. Neuroprotective delivery platforms as an adjunct to mechanical thrombectomy. Neurosurg Focus 2017; 42:E4. [PMID: 28366053 DOI: 10.3171/2017.1.focus16514] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Despite the success of numerous neuroprotective strategies in animal and preclinical stroke models, none have effectively translated to clinical medicine. A multitude of influences are likely responsible. Two such factors are inefficient recanalization strategies for large vessel occlusions and suboptimal delivery methods/platforms for neuroprotective agents. The recent endovascular stroke trials have established a new paradigm for large vessel stroke treatment. The associated advent of advanced mechanical revascularization devices and new stroke technologies help address each of these existing gaps. A strategy combining effective endovascular revascularization with administration of neuroprotective therapies is now practical and could have additive, if not synergistic, effects. This review outlines past and current neuroprotective strategies assessed in acute stroke trials. The discussion focuses on delivery platforms and their potential applicability to endovascular stoke treatment.
Collapse
Affiliation(s)
| | - Brian P Walcott
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, California
| | | | - Matthew S Tenser
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Arun P Amar
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - William J Mack
- Zilkha Neurogenetic Institute and.,Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
41
|
Intramyocardial Injection of Recombinant Adeno-Associated Viral Vector Coexpressing PR39/Adrenomedullin Enhances Angiogenesis and Reduces Apoptosis in a Rat Myocardial Infarction Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:1271670. [PMID: 28348718 PMCID: PMC5352904 DOI: 10.1155/2017/1271670] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 01/29/2017] [Accepted: 02/09/2017] [Indexed: 02/04/2023]
Abstract
Cotransfer of angiogenic and antiapoptotic genes could be the basis of new gene therapy strategies for myocardial infarction. In this study, rAAV-PR39-ADM, coexpressing antimicrobial peptide (PR39) and adrenomedullin (ADM), was designed with the mediation of recombinant adeno-associated virus. In vitro, CRL-1730 cells were divided into four groups, namely, the sham group, the AAV-null group, the NS (normal saline) group, and the PR39-ADM group. Immunocytochemistry analysis, CCK-8 assays, Matrigel assays, and apoptotic analysis were performed; in vivo, myocardial infarction model was established through ligation of the left coronary artery on rats, and treatment groups corresponded to those used in vitro. Myocardial injury, cardiac performance, and the extent of myocardial apoptosis were assessed. Results suggested that rAAV-PR39-ADM administration after myocardial infarction improved cell viability and cardiac function, attenuated apoptosis and myocardial injury, and promoted angiogenesis. Subsequently, levels of 6×His, HIF-1α, VEGF, p-Akt, Akt, ADM, Bcl-2, and Bax were measured by western blot. rAAV-PR39-ADM increased p-Akt, HIF-1α, and VEGF levels and induced higher Bcl-2 expression and lower Bax expression. In conclusion, our results demonstrate that rAAV-PR39-ADM mitigates myocardial injury by promoting angiogenesis and reducing apoptosis. This study suggests a potential novel gene therapy-based method that could be used clinically for myocardial infarction.
Collapse
|
42
|
Wang Y, Zhong L, Liu X, Zhu YZ. ZYZ-772 Prevents Cardiomyocyte Injury by Suppressing Nox4-Derived ROS Production and Apoptosis. Molecules 2017; 22:molecules22020331. [PMID: 28230797 PMCID: PMC6155929 DOI: 10.3390/molecules22020331] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 02/10/2017] [Accepted: 02/13/2017] [Indexed: 01/17/2023] Open
Abstract
Nox-dependent signaling plays critical roles in the development of heart failure, cardiac hypertrophy, and myocardial infarction. NADPH oxidase 4 (Nox4) as a major source of oxidative stress in the heart offers a new therapeutic target in cardiovascular disease. In the present work, a novel flavonoid was isolated from Zanthoxylum bungeanum. Its structure was elucidated as Quercetin-3-O-(6′′-O-α-l-rhamnopyransoyl)-β-d-glucopyranoside-7-O-β-d-glucopyranoside (ZYZ-772) for the first time. ZYZ-772 exhibited significant cardio-protective property against CoCl2 induced H9c2 cardiomyocyte cells injury. In CoCl2 stimulated cardiomyocyte injury, ZYZ-772 inhibited expression of Nox4, and alleviated ROS overproduction. Importantly, ROS triggered MAPKs phosphorylation and P53 signaling mediated apoptosis were restored by ZYZ-772. Our findings present the first piece of evidence for the therapeutic properties of ZYZ-772 in preventing cardiomyocyte injury, which could be attributed to the suppression of Nox4/MAPKs/P53 axis. This will offer a novel therapeutic strategy for the treatment of cardiac ischemia disease.
Collapse
Affiliation(s)
- Ying Wang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China.
| | - Liangjie Zhong
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China.
| | - Xinhua Liu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China.
| | - Yi Zhun Zhu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China.
- School of Pharmacy, Macao University of Science and Technology, Macao.
| |
Collapse
|
43
|
Ren M, Liu Y, Zhao H, Dong S, Jiang Z, Li K, Tian J. Adenosine triphosphate postconditioning is associated with better preserved global and regional cardiac function during myocardial ischemia and reperfusion: a speckle tracking imaging-based echocardiologic study. Cardiovasc Ther 2017; 34:343-51. [PMID: 27328167 DOI: 10.1111/1755-5922.12205] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Effects of ischemic postconditioning (IPostC) and adenosine triphosphate (ATP)-mediated pharmacologic postconditioning (ATP-PPostC) on cardiac function were evaluated by speckle tracking imaging (STI)-based echocardiography. AIMS A myocardial I/R model was induced in rabbits by reversible ligation of the left ventricular branch of coronary artery. Rabbits were randomized into three groups: ischemia and reperfusion (IR) (no further intervention), IPostC, and ATP-PPostC groups. Cardiac function was evaluated by conventional and STI-based echocardiography. Myocardial necrosis, apoptosis, and myocardial mRNAs of apoptosis-related proteins (Bcl-2 and Bax) were evaluated. RESULTS Speckle tracking imaging (STI)-based echocardiography revealed that IPostC and ATP-PPostC were associated with better preserved global and regional cardiac function, as indicated by significantly increased GLSrsys, GLSrd, GLSsys, SrLsys, SrLd, and SLsys in both groups (all P<.5). Subsequent pathologic studies indicate that the percentage of necrotic myocardium and permillage of apoptotic cells were significantly lower in the IPostC and ATP-PPostC groups than in the IR group (all P<.05). Moreover, both IPostC and ATP-PPostC were associated with increased Bcl-2 mRNA levels and reduced Bax mRNA levels. CONCLUSIONS IPostC and ATP-PPostC may exert cardioprotective functions by better preservation of cardiac function during the I/R process and at least partly via attenuation of myocardial apoptosis.
Collapse
Affiliation(s)
- Min Ren
- Department of Ultrasound, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yujie Liu
- Department of Ultrasound, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | | | - Shixia Dong
- Department of Special Diagnosis, PLA 313 Hospital, Huludao, China
| | - Zhonghui Jiang
- Department of Ultrasound, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Keting Li
- Department of Ultrasound, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiawei Tian
- Department of Ultrasound, Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
44
|
Zhou Y, Yang P, Li A, Ye X, Ren S, Li X. Prostaglandin E 2 reduces swine myocardial ischemia reperfusion injury via increased endothelial nitric oxide synthase and vascular endothelial growth factor expression levels. Biomed Rep 2016; 6:188-194. [PMID: 28357071 PMCID: PMC5351385 DOI: 10.3892/br.2016.834] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 12/12/2016] [Indexed: 02/05/2023] Open
Abstract
Prostaglandin E2 (PGE2) has been demonstrated to attenuate cardiac ischemia-reperfusion (I/R) injury. However, the underlying mechanism of PGE2 in cardiac I/R injury remains unknown. Upregulated expression levels of vascular endothelial growth factor (VEGF) and endothelial nitric oxide synthase (eNOS) were reported in acute myocardial infarction (AMI), and were demonstrated to diminish I/R injury. In the current study the involvement of VEGF and eNOS in the myocardial protective effect of PGE2 were investigated in a catheter-based porcine model of AMI. Twenty-two Chinese miniature pigs were randomized into sham-surgery (n=6), control (n=8) and PGE2 (n=8) groups. PGE2 (1 µg/kg) was injected from 10 min prior to left anterior descending occlusion up to 1 h after reperfusion in the PGE2 group. Subsequently, the hemodynamic parameters were evaluated. Thioflavin-S and Evans Blue double staining were performed to evaluate the extent of the myocardial reperfusion area (RA) and no-reflow area (NRA). Immunohistochemical and western blot analysis were used to evaluate protein expression levels of VEGF and eNOS. Left ventricular (LV) systolic pressure significantly improved and LV end-diastolic pressure significantly decreased in the PGE2 group when compared with the control group 2 h after occlusion and 3 h after reperfusion (P<0.05, respectively). The RA and NRA were smaller in the PGE2 group than in the control group (P<0.05, respectively). Furthermore, PGE2 treatment increased the myocardial content of VEGF and eNOS when compared with the control group (P<0.05, respectively). Thus, the results of the present study demonstrate the cardio-protective mechanisms of PGE2, which may protect the heart from I/R injury via enhancement of VEGF and eNOS expression levels.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Cardiology, China-Japan Friendship Hospital, Beijing 100029, P.R. China
| | - Peng Yang
- Department of Cardiology, China-Japan Friendship Hospital, Beijing 100029, P.R. China
| | - Aili Li
- Department of Cardiology, China-Japan Friendship Hospital, Beijing 100029, P.R. China
| | - Xiaojun Ye
- Department of Cardiology, China-Japan Friendship Hospital, Beijing 100029, P.R. China
| | - Shiyan Ren
- Department of Cardiology, China-Japan Friendship Hospital, Beijing 100029, P.R. China
| | - Xianlun Li
- Department of Cardiology, China-Japan Friendship Hospital, Beijing 100029, P.R. China
| |
Collapse
|
45
|
Spath NB, Mills NL, Cruden NL. Novel cardioprotective and regenerative therapies in acute myocardial infarction: a review of recent and ongoing clinical trials. Future Cardiol 2016; 12:655-672. [PMID: 27791385 PMCID: PMC5985502 DOI: 10.2217/fca-2016-0044] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 08/24/2016] [Indexed: 12/15/2022] Open
Abstract
Following the original large-scale randomized trials of aspirin and β-blockade, there have been a number of major advances in pharmacological and mechanical treatments for acute myocardial infarction. Despite this progress, myocardial infarction remains a major global cause of mortality and morbidity, driving a quest for novel treatments in this area. As the understanding of mitochondrial dynamics and the pathophysiology of reperfusion injury has evolved, the last three decades have seen advances in ischemic conditioning, pharmacological and metabolic cardioprotection, as well as biological and stem-cell therapies. The aim of this review is to provide a synopsis of adjunctive cardioprotective and regenerative therapies currently undergoing or entering early clinical trials in the treatment of patients with acute myocardial infarction.
Collapse
Affiliation(s)
- Nicholas B Spath
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Nicholas L Mills
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | | |
Collapse
|
46
|
He X, Zeng H, Chen JX. Ablation of SIRT3 causes coronary microvascular dysfunction and impairs cardiac recovery post myocardial ischemia. Int J Cardiol 2016; 215:349-57. [PMID: 27128560 PMCID: PMC4890543 DOI: 10.1016/j.ijcard.2016.04.092] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 04/11/2016] [Indexed: 11/21/2022]
Abstract
RATIONALE Sirtuin (SIRT3), a major nicotinamide adenine dinucleotide (NAD(+))-dependent deacetylase in mitochondria, declines with aging and its ablation is associated with accelerated development of cardiovascular diseases. However, the role of SIRT3 in coronary microvascular function and post-MI recovery has not been completely understood. OBJECTIVE The goal was to investigate whether ablation of SIRT3 causes coronary microvascular dysfunction, exacerbates post-myocardial ischemia (MI) cardiac dysfunction and impairs cardiac recovery. METHODS AND RESULTS Using endothelial cells (ECs) isolated from SIRT3 knockout (KO) mice, we revealed that the angiogenic capabilities were significantly reduced in SIRT3 deficient ECs. SIRT3 KO mice presented a pre-existing coronary microvascular dysfunction and microvascular rarefaction, as evidenced by a reduction in hyperemic peak diastolic blood flow velocity and coronary flow reserve (CFR), accompanied by loss of capillary-pericytes in the heart. Furthermore, SIRT3 KO mice subjected to myocardial ischemia by the ligation of left anterior descending coronary artery (LAD) exhibited more severe cardiac dysfunction together with decreased pericyte/EC coverage than that of wild type (WT) mice. In contrast, overexpression of SIRT3 preserved cardiac function in post-MI mice. Immunoblot analysis further showed that the expression of angiopoietin-1 (Ang-1), vascular endothelial growth factor (VEGF) and 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) were significantly decreased in the SIRT3-deficient ischemic hearts than those of WT ischemic hearts. This was accompanied by higher levels of cleaved caspase-3 and apoptosis. CONCLUSION Our results reveal a potential mechanism by which SIRT3 deletion exacerbates post-MI cardiac dysfunction and impairment of cardiac recovery involving microvascular rarefaction and pre-existing coronary microvascular dysfunction.
Collapse
Affiliation(s)
- Xiaochen He
- Dept. of Pharmacology and Toxicology, University of Mississippi Medical Center, United States
| | - Heng Zeng
- Dept. of Pharmacology and Toxicology, University of Mississippi Medical Center, United States
| | - Jian-Xiong Chen
- Dept. of Pharmacology and Toxicology, University of Mississippi Medical Center, United States.
| |
Collapse
|
47
|
Novel therapeutic strategies targeting fibroblasts and fibrosis in heart disease. Nat Rev Drug Discov 2016; 15:620-638. [PMID: 27339799 DOI: 10.1038/nrd.2016.89] [Citation(s) in RCA: 221] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Our understanding of the functions of cardiac fibroblasts has moved beyond their roles in heart structure and extracellular matrix generation and now includes their contributions to paracrine, mechanical and electrical signalling during ontogenesis and normal cardiac activity. Fibroblasts also have central roles in pathogenic remodelling during myocardial ischaemia, hypertension and heart failure. As key contributors to scar formation, they are crucial for tissue repair after interventions including surgery and ablation. Novel experimental approaches targeting cardiac fibroblasts are promising potential therapies for heart disease. Indeed, several existing drugs act, at least partially, through effects on cardiac connective tissue. This Review outlines the origins and roles of fibroblasts in cardiac development, homeostasis and disease; illustrates the involvement of fibroblasts in current and emerging clinical interventions; and identifies future targets for research and development.
Collapse
|
48
|
Intravenous Beta-Blockers for Cardioprotection in STEMI. J Am Coll Cardiol 2016; 67:2716-2718. [DOI: 10.1016/j.jacc.2016.03.532] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 03/29/2016] [Indexed: 11/21/2022]
|
49
|
Borea PA, Gessi S, Merighi S, Varani K. Adenosine as a Multi-Signalling Guardian Angel in Human Diseases: When, Where and How Does it Exert its Protective Effects? Trends Pharmacol Sci 2016; 37:419-434. [PMID: 26944097 DOI: 10.1016/j.tips.2016.02.006] [Citation(s) in RCA: 210] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 02/08/2016] [Accepted: 02/09/2016] [Indexed: 12/20/2022]
Abstract
The importance of adenosine for human health cannot be overstated. Indeed, this ubiquitous nucleoside is an integral component of ATP, and regulates the function of every tissue and organ in the body. Acting via receptor-dependent and -independent mechanisms [the former mediated via four G-protein-coupled receptors (GPCRs), A1, A2A, A2B, and A3,], it has a significant role in protecting against cell damage in areas of increased tissue metabolism, and combating organ dysfunction in numerous pathological states. Accordingly, raised levels of adenosine have been demonstrated in epilepsy, ischaemia, pain, inflammation, and cancer, in which its behaviour can be likened to that of a guardian angel, even though there are instances in which overproduction of adenosine is pathological. In this review, we condense the current body of knowledge on the issue, highlighting when, where, and how adenosine exerts its protective effects in both the brain and the periphery.
Collapse
Affiliation(s)
- Pier Andrea Borea
- Department of Medical Sciences, Pharmacology Section, University of Ferrara, Via Fossato di Mortara, 17-19, 44121 Ferrara, Italy.
| | - Stefania Gessi
- Department of Medical Sciences, Pharmacology Section, University of Ferrara, Via Fossato di Mortara, 17-19, 44121 Ferrara, Italy.
| | - Stefania Merighi
- Department of Medical Sciences, Pharmacology Section, University of Ferrara, Via Fossato di Mortara, 17-19, 44121 Ferrara, Italy.
| | - Katia Varani
- Department of Medical Sciences, Pharmacology Section, University of Ferrara, Via Fossato di Mortara, 17-19, 44121 Ferrara, Italy
| |
Collapse
|
50
|
LeBlanc AJ, Hoying JB. Adaptation of the Coronary Microcirculation in Aging. Microcirculation 2016; 23:157-67. [DOI: 10.1111/micc.12264] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 12/08/2015] [Indexed: 02/06/2023]
Affiliation(s)
- Amanda J. LeBlanc
- Department of Physiology; Cardiovascular Innovation Institute; University of Louisville; Louisville Kentucky USA
| | - James B. Hoying
- Department of Physiology; Cardiovascular Innovation Institute; University of Louisville; Louisville Kentucky USA
| |
Collapse
|