51
|
Peng H, Xu J, Yang XP, Kassem KM, Rhaleb IA, Peterson E, Rhaleb NE. N-acetyl-seryl-aspartyl-lysyl-proline treatment protects heart against excessive myocardial injury and heart failure in mice. Can J Physiol Pharmacol 2019; 97:753-765. [PMID: 30998852 PMCID: PMC6824427 DOI: 10.1139/cjpp-2019-0047] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Myocardial infarction (MI) in mice results in cardiac rupture at 4-7 days after MI, whereas cardiac fibrosis and dysfunction occur later. N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP) has anti-inflammatory, anti-fibrotic, and pro-angiogenic properties. We hypothesized that Ac-SDKP reduces cardiac rupture and adverse cardiac remodeling, and improves function by promoting angiogenesis and inhibiting detrimental reactive fibrosis and inflammation after MI. C57BL/6J mice were subjected to MI and treated with Ac-SDKP (1.6 mg/kg per day) for 1 or 5 weeks. We analyzed (1) intercellular adhesion molecule-1 (ICAM-1) expression; (2) inflammatory cell infiltration and angiogenesis; (3) gelatinolytic activity; (4) incidence of cardiac rupture; (5) p53, the endoplasmic reticulum stress marker CCAAT/enhancer binding protein homology protein (CHOP), and cardiomyocyte apoptosis; (6) sarcoplasmic reticulum Ca2+ ATPase (SERCA2) expression; (7) interstitial collagen fraction and capillary density; and (8) cardiac remodeling and function. Acutely, Ac-SDKP reduced cardiac rupture, decreased ICAM-1 expression and the number of infiltrating macrophages, decreased gelatinolytic activity, p53 expression, and myocyte apoptosis, but increased capillary density in the infarction border. Chronically, Ac-SDKP improved cardiac structures and function, reduced CHOP expression and interstitial collagen fraction, and preserved myocardium SERCA2 expression. Thus, Ac-SDKP decreased cardiac rupture, ameliorated adverse cardiac remodeling, and improved cardiac function after MI, likely through preserved SERCA2 expression and inhibition of endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Hongmei Peng
- a Hypertension and Vascular Research Division, Department of Internal Medicine, Detroit, MI 48202, USA
| | - Jiang Xu
- a Hypertension and Vascular Research Division, Department of Internal Medicine, Detroit, MI 48202, USA
| | - Xiao-Ping Yang
- a Hypertension and Vascular Research Division, Department of Internal Medicine, Detroit, MI 48202, USA
| | - Kamal M Kassem
- b Department of Internal Medicine, University of Cincinnati Medical Center, Cincinnati, OH 45219, USA
| | - Imane A Rhaleb
- a Hypertension and Vascular Research Division, Department of Internal Medicine, Detroit, MI 48202, USA
| | - Ed Peterson
- c Department of Public Health Sciences, Henry Ford Hospital, Detroit, MI 48202, USA
| | - Nour-Eddine Rhaleb
- a Hypertension and Vascular Research Division, Department of Internal Medicine, Detroit, MI 48202, USA
- d Department of Physiology, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
52
|
Kassem KM, Vaid S, Peng H, Sarkar S, Rhaleb NE. Tβ4-Ac-SDKP pathway: Any relevance for the cardiovascular system? Can J Physiol Pharmacol 2019; 97:589-599. [PMID: 30854877 PMCID: PMC6824425 DOI: 10.1139/cjpp-2018-0570] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The last 20 years witnessed the emergence of the thymosin β4 (Tβ4)-N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP) pathway as a new source of future therapeutic tools to treat cardiovascular and renal diseases. In this review article, we attempted to shed light on the numerous experimental findings pertaining to the many promising cardiovascular therapeutic avenues for Tβ4 and (or) its N-terminal derivative, Ac-SDKP. Specifically, Ac-SDKP is endogenously produced from the 43-amino acid Tβ4 by 2 successive enzymes, meprin α and prolyl oligopeptidase. We also discussed the possible mechanisms involved in the Tβ4-Ac-SDKP-associated cardiovascular biological effects. In infarcted myocardium, Tβ4 and Ac-SDKP facilitate cardiac repair after infarction by promoting endothelial cell migration and myocyte survival. Additionally, Tβ4 and Ac-SDKP have antifibrotic and anti-inflammatory properties in the arteries, heart, lungs, and kidneys, and stimulate both in vitro and in vivo angiogenesis. The effects of Tβ4 can be mediated directly through a putative receptor (Ku80) or via its enzymatically released N-terminal derivative Ac-SDKP. Despite the localization and characterization of Ac-SDKP binding sites in myocardium, more studies are needed to fully identify and clone Ac-SDKP receptors. It remains promising that Ac-SDKP or its degradation-resistant analogs could serve as new therapeutic tools to treat cardiac, vascular, and renal injury and dysfunction to be used alone or in combination with the already established pharmacotherapy for cardiovascular diseases.
Collapse
Affiliation(s)
- Kamal M Kassem
- a Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202, USA
- b Internal Medicine Department, University of Cincinnati Medical Center, Cincinnati, OH 45219, USA
| | - Sonal Vaid
- a Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202, USA
- c Internal Medicine Department, St. Vincent Indianapolis Hospital, Indianapolis, IN 46260, USA
| | - Hongmei Peng
- a Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202, USA
| | - Sarah Sarkar
- a Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202, USA
| | - Nour-Eddine Rhaleb
- a Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202, USA
- d Department of Physiology, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
53
|
Toor IS, Rückerl D, Mair I, Thomson A, Rossi AG, Newby DE, Allen JE, Gray GA. Enhanced monocyte recruitment and delayed alternative macrophage polarization accompanies impaired repair following myocardial infarction in C57BL/6 compared to BALB/c mice. Clin Exp Immunol 2019; 198:83-93. [PMID: 31119724 PMCID: PMC6718279 DOI: 10.1111/cei.13330] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2019] [Indexed: 12/24/2022] Open
Abstract
Activation of the innate immune response following myocardial infarction (MI) is essential for infarct repair. Preclinical models of MI commonly use C57BL/6 mice, which have a type 1‐dominant immune response, whereas other mouse strains such as BALB/c mice have a type 2‐dominant immune response. We compared C57BL/6 and BALB/c mice to investigate whether predisposition towards a proinflammatory phenotype influences the dynamics of the innate immune response to MI and associated infarct healing and the risk of cardiac rupture. MI was induced by permanent coronary artery ligation in 12–15‐week‐old male wild‐type BALB/c and C57BL/6 mice. Prior to MI, C57BL/6 mice had a lower proportion of CD206+ anti‐inflammatory macrophages in the heart and an expanded blood pool of proinflammatory Ly6Chigh monocytes in comparison to BALB/c mice. The systemic inflammatory response in C57BL/6 mice following MI was more pronounced, with greater peripheral blood Ly6Chigh monocytosis, splenic Ly6Chigh monocyte mobilization and myeloid cell infiltration of pericardial adipose tissue. This led to an increased and prolonged macrophage accumulation, as well as delayed transition towards anti‐inflammatory macrophage polarization in the infarct zone and surrounding tissues of C57BL/6 mice. These findings accompanied a higher rate of mortality due to cardiac rupture in C57BL/6 mice compared with BALB/c mice. We conclude that lower post‐MI survival of C57BL/6 mice over BALB/c mice is mediated in part by a more pronounced and prolonged inflammatory response. Outcomes in BALB/c mice highlight the therapeutic potential of modulating resolution of the innate immune response following MI for the benefit of successful infarct healing.
Collapse
Affiliation(s)
- I S Toor
- BHF/University Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - D Rückerl
- Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Edinburgh, UK
| | - I Mair
- MRC Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - A Thomson
- BHF/University Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - A G Rossi
- MRC Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - D E Newby
- BHF/University Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - J E Allen
- Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Edinburgh, UK
| | - G A Gray
- BHF/University Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
54
|
Qin CX, Rosli S, Deo M, Cao N, Walsh J, Tate M, Alexander AE, Donner D, Horlock D, Li R, Kiriazis H, Lee MKS, Bourke JE, Yang Y, Murphy AJ, Du XJ, Gao XM, Ritchie RH. Cardioprotective Actions of the Annexin-A1 N-Terminal Peptide, Ac 2-26, Against Myocardial Infarction. Front Pharmacol 2019; 10:269. [PMID: 31001111 PMCID: PMC6457169 DOI: 10.3389/fphar.2019.00269] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 03/04/2019] [Indexed: 12/22/2022] Open
Abstract
The anti-inflammatory, pro-resolving annexin-A1 protein acts as an endogenous brake against exaggerated cardiac necrosis, inflammation, and fibrosis following myocardial infarction (MI) in vivo. Little is known, however, regarding the cardioprotective actions of the N-terminal-derived peptide of annexin A1, Ac2-26, particularly beyond its anti-necrotic actions in the first few hours after an ischemic insult. In this study, we tested the hypothesis that exogenous Ac2-26 limits cardiac injury in vitro and in vivo. Firstly, we demonstrated that Ac2-26 limits cardiomyocyte death both in vitro and in mice subjected to ischemia-reperfusion (I-R) injury in vivo (Ac2-26, 1 mg/kg, i.v. just prior to post-ischemic reperfusion). Further, Ac2-26 (1 mg/kg i.v.) reduced cardiac inflammation (after 48 h reperfusion), as well as both cardiac fibrosis and apoptosis (after 7-days reperfusion). Lastly, we investigated whether Ac2-26 preserved cardiac function after MI. Ac2-26 (1 mg/kg/day s.c., osmotic pump) delayed early cardiac dysfunction 1 week post MI, but elicited no further improvement 4 weeks after MI. Taken together, our data demonstrate the first evidence that Ac2-26 not only preserves cardiomyocyte survival in vitro, but also offers cardioprotection beyond the first few hours after an ischemic insult in vivo. Annexin-A1 mimetics thus represent a potential new therapy to improve cardiac outcomes after MI.
Collapse
Affiliation(s)
- Cheng Xue Qin
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, VIC, Australia.,Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Sarah Rosli
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Minh Deo
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Nga Cao
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Jesse Walsh
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Mitchel Tate
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Amy E Alexander
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Daniel Donner
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Duncan Horlock
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Renming Li
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Helen Kiriazis
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Man K S Lee
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Jane E Bourke
- Department of Pharmacology, Monash University, Clayton, VIC, Australia
| | - Yuan Yang
- Centre for Inflammatory Diseases, Monash University, Clayton, VIC, Australia
| | - Andrew J Murphy
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Xiao-Jun Du
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Xiao Ming Gao
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Rebecca H Ritchie
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, VIC, Australia.,Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Pharmacology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
55
|
Macrophage migration inhibitory factor plays an essential role in ischemic preconditioning-mediated cardioprotection. Clin Sci (Lond) 2019; 133:665-680. [PMID: 30804219 DOI: 10.1042/cs20181013] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 01/26/2019] [Accepted: 02/22/2019] [Indexed: 12/23/2022]
Abstract
Ischemic preconditioning (IPC) is an endogenous protection strategy against myocardial ischemia-reperfusion (I/R) injury. Macrophage migration inhibitory factor (MIF) released from the myocardium subjected to brief periods of ischemia confers cardioprotection. We hypothesized that MIF plays an essential role in IPC-induced cardioprotection. I/R was induced either ex vivo or in vivo in male wild-type (WT) and MIF knockout (MIFKO) mice with or without proceeding IPC (three cycles of 5-min ischemia and 5-min reperfusion). Indices of myocardial injury, regional inflammation and cardiac function were determined to evaluate the extent of I/R injury. Activations of the reperfusion injury salvage kinase (RISK) pathway, AMP-activated protein kinase (AMPK) and their downstream components were investigated to explore the underlying mechanisms. IPC conferred prominent protection in WT hearts evidenced by reduced infarct size (by 33-35%), myocyte apoptosis and enzymatic markers of tissue injury, ROS production, inflammatory cell infiltration and MCP1/CCR2 expression (all P<0.05). IPC also ameliorated cardiac dysfunction both ex vivo and in vivo These protective effects were abolished in MIFKO hearts. Notably, IPC mediated further activations of RISK pathway, AMPK and the membrane translocation of GLUT4 in WT hearts. Deletion of MIF blunted these changes in response to IPC, which is the likely basis for the absence of protective effects of IPC against I/R injury. In conclusion, MIF plays a critical role in IPC-mediated cardioprotection under ischemic stress by activating RISK signaling pathway and AMPK. These results provide an insight for developing a novel therapeutic strategy that target MIF to protect ischemic hearts.
Collapse
|
56
|
Hao W, Lu S, Guo R, Fan J, Zhen L, Nie S. Risk factors for cardiac rupture complicating myocardial infarction: a PRISMA meta-analysis and systematic review. J Investig Med 2018; 67:720-728. [PMID: 30487185 DOI: 10.1136/jim-2018-000841] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2018] [Indexed: 11/04/2022]
Abstract
Cardiac rupture (CR) is a complication of acute myocardial infarction (AMI) that is associated with a high mortality rate. This study aimed to identify the risk factors for CR in patients with AMI. Medline, Cochrane, EMBASE, and Google Scholar databases were searched for relevant literature published through September 16, 2018. Eligible studies included patients with AMI and compared factors between patients with and without CR. Sixteen studies were identified and included in the meta-analysis. Results revealed that female gender (pooled OR=2.72, 95% CI 2.04 to 3.63, p<0.001), older age (pooled difference in means=6.91, 95% CI 4.20 to 9.62, p<0.001), infarction at left anterior descending coronary artery (LAD) (pooled OR=1.85, 95% CI 1.03 to 3.32, p=0.039), and anterior wall infarction (pooled OR=1.87, 95% CI 1.30 to 2.68, p=0.001) were associated with increased risk of CR, whereas history of MI, smoking, and multivessel disease were associated with reduced risk of CR. Patients treated with primary percutaneous coronary intervention (PCI) had reduced risk of CR, while patients who had received any thrombolysis had increased risk of CR. In conclusion, results of systematic review and meta-analysis of existing literature suggest that risk factors for CR in patients with AMI include female gender, older age, new-onset MI, non-smoking status, LAD infarction, anterior wall infarction, and single-vessel disease. Furthermore, treatment with primary PCI may help reduce the risk for CR, while thrombolysis might increase the risk for CR.
Collapse
Affiliation(s)
- Wen Hao
- Emergency and Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, and Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Shangxin Lu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Ruifeng Guo
- Emergency and Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, and Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Jingyao Fan
- Emergency and Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, and Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Lei Zhen
- Emergency and Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, and Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Shaoping Nie
- Emergency and Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, and Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| |
Collapse
|
57
|
Hua J, Liu Z, Liu Z, An D, Lai W, Zhan Q, Zeng Q, Ren H, Xu D. Metformin Increases Cardiac Rupture After Myocardial Infarction via the AMPK-MTOR/PGC-1α Signaling Pathway in Rats with Acute Myocardial Infarction. Med Sci Monit 2018; 24:6989-7000. [PMID: 30275441 PMCID: PMC6180847 DOI: 10.12659/msm.910930] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background Cardiac rupture often occurs after acute myocardial infarction due to complex and unclear pathogenesis. This study investigated whether metformin increases the incidence of cardiac rupture after myocardial infarction through the AMPK-MTOR/PGC-1α signaling pathway. Material/Methods An acute myocardial infarction (MI) mouse model was established. A series of experiments involving RT-qPCR, Western blot, TUNEL staining, and Masson staining were performed in this study. Results Myocardial infarction occurred, resulting in the cardiac rupture, and the expression level of PGC-1α increased in the cardiac myocardium. Meanwhile, the proportion of myocardial NT-PGC-1α/PGC-1α decreased. The expression level of myocardial PGC-1α in MI mice with cardiac rupture after MI was significantly higher than that in the mice without cardiac rupture, and the ratio of myocardial NT-PGC-1α/PGC-1α was low. In addition, increasing the dose of metformin significantly increased the incidence of cardiac rupture after myocardial infarction in MI mice. High-dose metformin caused cardiac rupture in MI mice. Moreover, high-dose metformin (Met 2.0 nM) reduces the proportion of NT-PGC-1α/PGC-1α in primary cardiomyocytes of SD mice (SD-NRVCs [Neonatal rat ventricular cardiomyocytes]), and its effect was inhibited by Compound C (AMPK inhibitor). Further, after 3 days of treatment with high-dose metformin in MI mice, myocardial fibrin synthesis decreased and fibrosis was significantly inhibited. Meanwhile, cardiomyocyte apoptosis increased significantly. With the increase in metformin concentration, the expression level of myocardial LC3b gradually increased in MI mice, suggesting that metformin enhances the autophagy of cardiomyocytes. Conclusions These results suggest that metformin increases cardiac rupture after myocardial infarction through the AMPK-MTOR/PGC-1α signaling pathway.
Collapse
Affiliation(s)
- Jinghai Hua
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland).,Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, Guangdong, China (mainland)
| | - Zhanghua Liu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland).,Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, Guangdong, China (mainland)
| | - Zuheng Liu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland).,Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, Guangdong, China (mainland)
| | - Dongqi An
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland).,Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, Guangdong, China (mainland)
| | - Wenyan Lai
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland).,Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, Guangdong, China (mainland)
| | - Qiong Zhan
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland).,Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, Guangdong, China (mainland)
| | - Qingchun Zeng
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland).,Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, Guangdong, China (mainland)
| | - Hao Ren
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, Guangdong, China (mainland).,Department of Rheumatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland)
| | - Dingli Xu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (mainland).,Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, Guangdong, China (mainland)
| |
Collapse
|
58
|
Gong W, Ma Y, Li A, Shi H, Nie S. Trimetazidine suppresses oxidative stress, inhibits MMP-2 and MMP-9 expression, and prevents cardiac rupture in mice with myocardial infarction. Cardiovasc Ther 2018; 36:e12460. [PMID: 30019466 DOI: 10.1111/1755-5922.12460] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 06/28/2018] [Accepted: 07/14/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND/AIMS Cardiac rupture (CR) is a catastrophic complication of acute myocardial infarction (MI). At present, there are no effective pharmacological strategies for preventing post-MI rupture. Here we investigated the effect of trimetazidine (TMZ) on post-MI CR. METHODS MI models were induced by left coronary artery ligation in male C57BL/6 mice. Animals allocated to the rupture incidence were closely monitored for 7 days; autopsy was performed once animals were found dead to determine the reason of death. Heart function was detected by echocardiography. Oxidative stress markers and matrix metalloproteinases (MMPs) were analyzed by Western Blotting. RESULTS TMZ markedly reduced the post-MI CR incidence of mice. We found that the expression of metalloproteinase (MMP) -2 and MMP-9 in the TMZ-treated group was significantly lower than the saline-treated group. Further, TMZ markedly attenuated MI-induced oxidative stress. To investigate the mechanism of the effect of TMZ on CR, we pretreated H9c2 cells with H2 O2 and found that TMZ treatment markedly decreased H2 O2 -induced MMP-2 and MMP-9 expression. TMZ prevents CR through inhibition of oxidative stress, which is attributable to the down-regulation of MMP-2, MMP-9 expression. CONCLUSIONS Our findings indicate that TMZ suppresses oxidative stress, inhibits MMP-2 and MMP-9 expression, and prevents CR in mice with MI.
Collapse
Affiliation(s)
- Wei Gong
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Youcai Ma
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Aobo Li
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Han Shi
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Shaoping Nie
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| |
Collapse
|
59
|
Kasneci A, Lee JS, Yun TJ, Shang J, Lampen S, Gomolin T, Cheong CC, Chalifour LE. From the Cover: Lifelong Exposure of C57bl/6n Male Mice to Bisphenol A or Bisphenol S Reduces Recovery From a Myocardial Infarction. Toxicol Sci 2018; 159:189-202. [PMID: 28903498 DOI: 10.1093/toxsci/kfx133] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Bisphenol A (BPA) leaches from plastics to contaminate foodstuffs. Analogs, such as bisphenol S (BPS), are now used increasingly in manufacturing. Greater BPA exposure has been correlated with exacerbation of cardiovascular disease, including myocardial infarction (MI). To test the hypothesis that bisphenol exposure impairs cardiac healing, we exposed C57bl/6n mice to water containing 25ng/ml BPA or BPS from conception and surgically induced an MI in adult male progeny. Increased early death and cardiac dilation, and reduced cardiac function were found post-MI in BPA- and BPS-exposed mice. Flow cytometry revealed increased monocyte and macrophage infiltration that correlated with increased chemokine C-C motif ligand-2 expression in the infarct. In vitro BPA and BPS addition increased matrix metalloproteinase-9 (MMP) protein and secreted activity in RAW264.7 macrophage cells suggesting that invivo increases in MMP2 and MMP9 in exposed infarcts were myeloid-derived. Bone marrow-derived monocytes isolated from exposed mice had greater expression of pro-inflammatory polarization markers when chemokine stimulated indicating an enhanced susceptibility to develop a pro-inflammatory monocyte population. Chronic BPA exposure of estrogen receptor beta (ERβ) deficient mice did not worsen early death, cardiac structure/function, or expression of myeloid markers after an MI. In contrast, BPS exposure of ERβ-deficient mice resulted in greater death and expression of myeloid markers. We conclude that lifelong exposure to BPA or BPS augmented the monocyte/macrophage inflammatory response and adverse remodeling from an MI thereby reducing the ability to survive and successfully recover, and that the adverse effect of BPA, but not BPS, is downstream of ERβ signaling.
Collapse
Affiliation(s)
- Amanda Kasneci
- Lady Davis Institute for Medical Research, Montréal, Québec H3T 1E2, Canada
| | - Jun Seong Lee
- Institut de Recherches Cliniques de Montréal, Montréal, Québec H2W 1R7, Canada
| | - Tae Jin Yun
- Institut de Recherches Cliniques de Montréal, Montréal, Québec H2W 1R7, Canada
| | - Jijun Shang
- Lady Davis Institute for Medical Research, Montréal, Québec H3T 1E2, Canada
| | - Shaun Lampen
- Lady Davis Institute for Medical Research, Montréal, Québec H3T 1E2, Canada
| | - Tamar Gomolin
- Lady Davis Institute for Medical Research, Montréal, Québec H3T 1E2, Canada
| | - Cheolho C Cheong
- Institut de Recherches Cliniques de Montréal, Montréal, Québec H2W 1R7, Canada.,Division of Experimental Medicine, Department of Medicine, McGill University, Montréal, Québec H3A 1A2, Canada
| | - Lorraine E Chalifour
- Lady Davis Institute for Medical Research, Montréal, Québec H3T 1E2, Canada.,Division of Experimental Medicine, Department of Medicine, McGill University, Montréal, Québec H3A 1A2, Canada
| |
Collapse
|
60
|
Manning JR, Chelvarajan L, Levitan BM, Withers CN, Nagareddy PR, Haggerty CM, Fornwalt BK, Gao E, Tripathi H, Abdel-Latif A, Andres DA, Satin J. Rad GTPase deletion attenuates post-ischemic cardiac dysfunction and remodeling. ACTA ACUST UNITED AC 2018; 3:83-96. [PMID: 29732439 PMCID: PMC5931223 DOI: 10.1016/j.jacbts.2017.09.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Rad-GTPase is an LTCC component that functions to govern calcium current in the myocardium. Deletion of Rad increases myocardial contractility secondary to increased trigger calcium entry. AMI induces heart failure, including reduced calcium homeostasis, but deletion of Rad prevents AMI myocardial calcium alterations. Rad deletion prevents post-MI scar spread by attenuating the inflammatory response. Future studies will explore whether Rad deletion is an effective therapeutic direction for providing combined safe, stable inotropic support to the failing heart in concert with protection against inflammatory signaling.
The protein Rad interacts with the L-type calcium channel complex to modulate trigger Ca2+ and hence to govern contractility. Reducing Rad levels increases cardiac output. Ablation of Rad also attenuated the inflammatory response following acute myocardial infarction. Future studies to target deletion of Rad in the heart could be conducted to establish a novel treatment paradigm whereby pathologically stressed hearts would be given safe, stable positive inotropic support without arrhythmias and without pathological structural remodeling. Future investigations will also focus on establishing inhibitors of Rad and testing the efficacy of Rad deletion in cardioprotection relative to the time of onset of acute myocardial infarction.
Collapse
Affiliation(s)
- Janet R Manning
- Department of Physiology, University of Kentucky, Lexington KY.,Department of Biochemistry, University of Kentucky, Lexington KY
| | - Lakshman Chelvarajan
- Saha Cardiovascular Research Center, Department of Medicine, University of Kentucky, Lexington, KY
| | - Bryana M Levitan
- Department of Physiology, University of Kentucky, Lexington KY.,Gill Heart and Vascular Institute, University of Kentucky, Lexington KY
| | | | | | - Christopher M Haggerty
- Saha Cardiovascular Research Center, Department of Medicine, University of Kentucky, Lexington, KY.,Department of Imaging Science and Innovation, Geisinger, Danville PA
| | - Brandon K Fornwalt
- Saha Cardiovascular Research Center, Department of Medicine, University of Kentucky, Lexington, KY.,Department of Imaging Science and Innovation, Geisinger, Danville PA
| | - Erhe Gao
- Department of Physiology, University of Kentucky, Lexington KY.,Center for Translational Medicine, Temple University School of Medicine, Philadelphia PA
| | - Himi Tripathi
- Saha Cardiovascular Research Center, Department of Medicine, University of Kentucky, Lexington, KY
| | - Ahmed Abdel-Latif
- Saha Cardiovascular Research Center, Department of Medicine, University of Kentucky, Lexington, KY.,Gill Heart and Vascular Institute, University of Kentucky, Lexington KY
| | - Douglas A Andres
- Department of Biochemistry, University of Kentucky, Lexington KY
| | - Jonathan Satin
- Department of Physiology, University of Kentucky, Lexington KY
| |
Collapse
|
61
|
Du X. Post-infarct cardiac injury, protection and repair: roles of non-cardiomyocyte multicellular and acellular components. SCIENCE CHINA-LIFE SCIENCES 2018; 61:266-276. [DOI: 10.1007/s11427-017-9223-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 10/23/2017] [Indexed: 02/06/2023]
|
62
|
Glovaci D, Naqvi A, Yu K, Patel P, Krishnam M. Utility of cardiac MRI in determining percutaneous versus surgical post-infarction ventricular septal defect repair. Future Cardiol 2018; 14:125-130. [PMID: 29355029 DOI: 10.2217/fca-2017-0062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM Postmyocardial infarction ventricular septal defect (VSD) is a rare complication that can lead to rapid hemodynamic patient decompensation. The type of VSD repair relies on several factors including: size, location, timing and surgical expertise. CASE A 63-year-old man with a ST-elevation myocardial infarction underwent percutaneous coronary intervention of the right coronary artery. A holosystolic murmur was notable postcatheterization, and transthoracic echocardiogram confirmed a VSD. To characterize the VSD, a cardiac MRI demonstrated a large, serpiginous VSD and longitudinal septal tear. Given the anatomic complexity and stable hemodynamics, a surgical trans-left ventricular patch repair was performed. CONCLUSION We emphasize the importance of cardiac magnetic resonance as a decision-making tool, utilizing imaging to ascertain the anatomy combined with hemodynamics to determine optimal individualized therapy.
Collapse
Affiliation(s)
- Diana Glovaci
- Department of Internal Medicine, University California Irvine Medical Center, Orange, CA 92868, USA
| | - Ali Naqvi
- Department of Internal Medicine, University California Irvine Medical Center, Orange, CA 92868, USA
| | - Katherine Yu
- Department of Cardiology, University California Irvine Medical Center, Orange, CA 92868, USA
| | - Pranav Patel
- Department of Cardiology, University California Irvine Medical Center, Orange, CA 92868, USA
| | - Mayil Krishnam
- Department of Radiology, University California Irvine Medical Center, Orange, CA 92868, USA
| |
Collapse
|
63
|
Lindsey ML, Bolli R, Canty JM, Du XJ, Frangogiannis NG, Frantz S, Gourdie RG, Holmes JW, Jones SP, Kloner RA, Lefer DJ, Liao R, Murphy E, Ping P, Przyklenk K, Recchia FA, Schwartz Longacre L, Ripplinger CM, Van Eyk JE, Heusch G. Guidelines for experimental models of myocardial ischemia and infarction. Am J Physiol Heart Circ Physiol 2018; 314:H812-H838. [PMID: 29351451 PMCID: PMC5966768 DOI: 10.1152/ajpheart.00335.2017] [Citation(s) in RCA: 369] [Impact Index Per Article: 52.7] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Myocardial infarction is a prevalent major cardiovascular event that arises from myocardial ischemia with or without reperfusion, and basic and translational research is needed to better understand its underlying mechanisms and consequences for cardiac structure and function. Ischemia underlies a broad range of clinical scenarios ranging from angina to hibernation to permanent occlusion, and while reperfusion is mandatory for salvage from ischemic injury, reperfusion also inflicts injury on its own. In this consensus statement, we present recommendations for animal models of myocardial ischemia and infarction. With increasing awareness of the need for rigor and reproducibility in designing and performing scientific research to ensure validation of results, the goal of this review is to provide best practice information regarding myocardial ischemia-reperfusion and infarction models. Listen to this article’s corresponding podcast at ajpheart.podbean.com/e/guidelines-for-experimental-models-of-myocardial-ischemia-and-infarction/.
Collapse
Affiliation(s)
- Merry L Lindsey
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi.,Research Service, G. V. (Sonny) Montgomery Veterans Affairs Medical Center , Jackson, Mississippi
| | - Roberto Bolli
- Division of Cardiovascular Medicine and Institute of Molecular Cardiology, University of Louisville , Louisville, Kentucky
| | - John M Canty
- Division of Cardiovascular Medicine, Departments of Biomedical Engineering and Physiology and Biophysics, The Veterans Affairs Western New York Health Care System and Clinical and Translational Science Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo , Buffalo, New York
| | - Xiao-Jun Du
- Baker Heart and Diabetes Institute , Melbourne, Victoria , Australia
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York
| | - Stefan Frantz
- Department of Internal Medicine I, University Hospital , Würzburg , Germany
| | - Robert G Gourdie
- Center for Heart and Regenerative Medicine Research, Virginia Tech Carilion Research Institute , Roanoke, Virginia
| | - Jeffrey W Holmes
- Department of Biomedical Engineering, University of Virginia Health System , Charlottesville, Virginia
| | - Steven P Jones
- Department of Medicine, Institute of Molecular Cardiology, Diabetes and Obesity Center, University of Louisville , Louisville, Kentucky
| | - Robert A Kloner
- HMRI Cardiovascular Research Institute, Huntington Medical Research Institutes , Pasadena, California.,Division of Cardiovascular Medicine, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - David J Lefer
- Cardiovascular Center of Excellence, Louisiana State University Health Science Center , New Orleans, Louisiana
| | - Ronglih Liao
- Harvard Medical School , Boston, Massachusetts.,Division of Genetics and Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital , Boston, Massachusetts
| | - Elizabeth Murphy
- Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health , Bethesda, Maryland
| | - Peipei Ping
- National Institutes of Health BD2KBig Data to Knowledge (BD2K) Center of Excellence and Department of Physiology, Medicine and Bioinformatics, University of California , Los Angeles, California
| | - Karin Przyklenk
- Cardiovascular Research Institute and Departments of Physiology and Emergency Medicine, Wayne State University School of Medicine , Detroit, Michigan
| | - Fabio A Recchia
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Fondazione G. Monasterio, Pisa , Italy.,Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University , Philadelphia, Pennsylvania
| | - Lisa Schwartz Longacre
- Heart Failure and Arrhythmias Branch, Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health , Bethesda, Maryland
| | - Crystal M Ripplinger
- Department of Pharmacology, School of Medicine, University of California , Davis, California
| | - Jennifer E Van Eyk
- The Smidt Heart Institute, Department of Medicine, Cedars Sinai Medical Center , Los Angeles, California
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School , Essen , Germany
| |
Collapse
|
64
|
Endogenous Annexin-A1 Regulates Haematopoietic Stem Cell Mobilisation and Inflammatory Response Post Myocardial Infarction in Mice In Vivo. Sci Rep 2017; 7:16615. [PMID: 29192208 PMCID: PMC5709412 DOI: 10.1038/s41598-017-16317-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 11/09/2017] [Indexed: 12/25/2022] Open
Abstract
Endogenous anti-inflammatory annexin-A1 (ANX-A1) plays an important role in preserving left ventricular (LV) viability and function after ischaemic insults in vitro, but its long-term cardioprotective actions in vivo are largely unknown. We tested the hypothesis that ANX-A1-deficiency exaggerates inflammation, haematopoietic stem progenitor cell (HSPC) activity and LV remodelling in response to myocardial ischaemia in vivo. Adult ANX-A1−/− mice subjected to coronary artery occlusion exhibited increased infarct size and LV macrophage content after 24–48 h reperfusion compared with wildtype (WT) counterparts. In addition, ANX-A1−/− mice exhibited greater expansion of HSPCs and altered pattern of HSPC mobilisation 8 days post-myocardial infarction, with increased circulating neutrophils and platelets, consistent with increased cardiac inflammation as a result of increased myeloid invading injured myocardium in response to MI. Furthermore, ANX-A1−/− mice exhibited significantly increased expression of LV pro-inflammatory and pro-fibrotic genes and collagen deposition after MI compared to WT counterparts. ANX-A1-deficiency increased cardiac necrosis, inflammation, hypertrophy and fibrosis following MI, accompanied by exaggerated HSPC activity and impaired macrophage phenotype. These findings suggest that endogenous ANX-A1 regulates mobilisation and differentiation of HSPCs. Limiting excessive monocyte/neutrophil production may limit LV damage in vivo. Our findings support further development of novel ANX-A1-based therapies to improve cardiac outcomes after MI.
Collapse
|
65
|
Olsen MB, Hildrestrand GA, Scheffler K, Vinge LE, Alfsnes K, Palibrk V, Wang J, Neurauter CG, Luna L, Johansen J, Øgaard JDS, Ohm IK, Slupphaug G, Kuśnierczyk A, Fiane AE, Brorson SH, Zhang L, Gullestad L, Louch WE, Iversen PO, Østlie I, Klungland A, Christensen G, Sjaastad I, Sætrom P, Yndestad A, Aukrust P, Bjørås M, Finsen AV. NEIL3-Dependent Regulation of Cardiac Fibroblast Proliferation Prevents Myocardial Rupture. Cell Rep 2017; 18:82-92. [PMID: 28052262 DOI: 10.1016/j.celrep.2016.12.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 10/06/2016] [Accepted: 12/01/2016] [Indexed: 12/15/2022] Open
Abstract
Myocardial infarction (MI) triggers a reparative response involving fibroblast proliferation and differentiation driving extracellular matrix modulation necessary to form a stabilizing scar. Recently, it was shown that a genetic variant of the base excision repair enzyme NEIL3 was associated with increased risk of MI in humans. Here, we report elevated myocardial NEIL3 expression in heart failure patients and marked myocardial upregulation of Neil3 after MI in mice, especially in a fibroblast-enriched cell fraction. Neil3-/- mice show increased mortality after MI caused by myocardial rupture. Genome-wide analysis of 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC) reveals changes in the cardiac epigenome, including in genes related to the post-MI transcriptional response. Differentially methylated genes are enriched in pathways related to proliferation and myofibroblast differentiation. Accordingly, Neil3-/- ruptured hearts show increased proliferation of fibroblasts and myofibroblasts. We propose that NEIL3-dependent modulation of DNA methylation regulates cardiac fibroblast proliferation and thereby affects extracellular matrix modulation after MI.
Collapse
Affiliation(s)
- Maria B Olsen
- Research Institute of Internal Medicine, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway; Center for Heart Failure Research, University of Oslo, 0317 Oslo, Norway; K.G. Jebsen Inflammation Research Centre, University of Oslo, 0317 Oslo, Norway
| | - Gunn A Hildrestrand
- Department of Microbiology, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway
| | - Katja Scheffler
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Leif Erik Vinge
- Research Institute of Internal Medicine, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway; Department of Cardiology, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway; Center for Heart Failure Research, University of Oslo, 0317 Oslo, Norway
| | - Katrine Alfsnes
- Research Institute of Internal Medicine, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway; Center for Heart Failure Research, University of Oslo, 0317 Oslo, Norway; K.G. Jebsen Inflammation Research Centre, University of Oslo, 0317 Oslo, Norway
| | - Vuk Palibrk
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Junbai Wang
- Department of Pathology, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway
| | - Christine G Neurauter
- Department of Microbiology, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway
| | - Luisa Luna
- Department of Microbiology, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway
| | - Jostein Johansen
- Bioinformatics Core Facility-BioCore , Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Jonas D S Øgaard
- Research Institute of Internal Medicine, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway
| | - Ingrid K Ohm
- Research Institute of Internal Medicine, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway; Center for Heart Failure Research, University of Oslo, 0317 Oslo, Norway
| | - Geir Slupphaug
- Proteomics and Metabolomics Core Facility-PROMEC, Norwegian University of Science and Technology, 7491 Trondheim, Norway; Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Anna Kuśnierczyk
- Proteomics and Metabolomics Core Facility-PROMEC, Norwegian University of Science and Technology, 7491 Trondheim, Norway; Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Arnt E Fiane
- Department of Cardiothoracic Surgery, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway
| | - Sverre-Henning Brorson
- Department of Pathology, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway
| | - Lili Zhang
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway
| | - Lars Gullestad
- Research Institute of Internal Medicine, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway; Department of Cardiology, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway; Center for Heart Failure Research, University of Oslo, 0317 Oslo, Norway
| | - William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway; Center for Heart Failure Research, University of Oslo, 0317 Oslo, Norway
| | - Per Ole Iversen
- Department of Haematology, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway; Department of Nutrition, University of Oslo, 0317 Oslo, Norway
| | - Ingunn Østlie
- Department of Pathology, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway
| | - Arne Klungland
- Department of Microbiology, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway
| | - Geir Christensen
- Department of Cardiology, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway; Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway; Center for Heart Failure Research, University of Oslo, 0317 Oslo, Norway
| | - Ivar Sjaastad
- Department of Cardiology, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway; Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway; Center for Heart Failure Research, University of Oslo, 0317 Oslo, Norway
| | - Pål Sætrom
- Bioinformatics Core Facility-BioCore , Norwegian University of Science and Technology, 7491 Trondheim, Norway; Department of Computer and Information Science, Norwegian University of Science and Technology, 7491 Trondheim, Norway; Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Arne Yndestad
- Research Institute of Internal Medicine, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway; Center for Heart Failure Research, University of Oslo, 0317 Oslo, Norway; K.G. Jebsen Inflammation Research Centre, University of Oslo, 0317 Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway; Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway; K.G. Jebsen Inflammation Research Centre, University of Oslo, 0317 Oslo, Norway
| | - Magnar Bjørås
- Department of Microbiology, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway; Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, 7491 Trondheim, Norway.
| | - Alexandra V Finsen
- Research Institute of Internal Medicine, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway; Department of Cardiology, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway; Center for Heart Failure Research, University of Oslo, 0317 Oslo, Norway
| |
Collapse
|
66
|
Abstract
The heart is extremely sensitive to ischaemic injury. During an acute myocardial infarction (AMI) event, the injury is initially caused by reduced blood supply to the tissues, which is then further exacerbated by an intense and highly specific inflammatory response that occurs during reperfusion. Numerous studies have highlighted the central role of the NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome in this process. The inflammasome, an integral part of the innate immune system, is a macromolecular protein complex that finely regulates the activation of caspase 1 and the production and secretion of powerful pro-inflammatory cytokines such as IL-1β and IL-18. In this Review, we summarize evidence supporting the therapeutic value of NLRP3 inflammasome-targeted strategies in experimental models, and the data supporting the role of the NLRP3 inflammasome in AMI and its consequences on adverse cardiac remodelling, cytokine-mediated systolic dysfunction, and heart failure.
Collapse
Affiliation(s)
- Stefano Toldo
- Pauley Heart Center, Sanger Hall, 1201 East Marshall Street, Richmond, Virginia 23298, USA.,VCU Johnson Center for Critical Care and Pulmonary Research, Molecular Medicine Research Building, 1220 East Broad Street, Richmond, Virginia 23298, USA.,Division of Cardiothoracic Surgery, Sanger Hall, 1201 East Marshall Street, Richmond, Virginia 23298, USA
| | - Antonio Abbate
- Pauley Heart Center, Sanger Hall, 1201 East Marshall Street, Richmond, Virginia 23298, USA.,VCU Johnson Center for Critical Care and Pulmonary Research, Molecular Medicine Research Building, 1220 East Broad Street, Richmond, Virginia 23298, USA
| |
Collapse
|
67
|
Wang Z, Huang S, Sheng Y, Peng X, Liu H, Jin N, Cai J, Shu Y, Li T, Li P, Fan C, Hu X, Zhang W, Long R, You Y, Huang C, Song Y, Xiang C, Wang J, Yang Y, Liu K. Topiramate modulates post-infarction inflammation primarily by targeting monocytes or macrophages. Cardiovasc Res 2017; 113:475-487. [PMID: 28339742 DOI: 10.1093/cvr/cvx027] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 03/09/2017] [Indexed: 12/24/2022] Open
Abstract
Aims Monocytes/macrophages response plays a key role in post-infarction inflammation that contributes greatly to post-infarction ventricular remodelling and cardiac rupture. Therapeutic targeting of the GABAA receptor, which is enriched in monocytes/macrophages but not expressed in the myocardium, may be possible after myocardial infarction (MI). Methods and results After MI was induced by ligation of the coronary artery, C57BL/6 mice were intraperitoneally administered with one specific agonist or antagonist of the GABAA receptor (topiramate or bicuculline), in the setting of presence or depletion of monocytes/macrophages. Our data showed that within the first 2 weeks after MI, when monocytes/macrophages dominated, in contrast with bicuculline, topiramate treatment significantly reduced Ly-6Chigh monocyte numbers by regulating splenic monocytopoiesis and promoted foetal derived macrophages preservation and conversion of M1 to M2 or Ly-6Chigh to Ly-6Clow macrophage phenotype in the infarcted heart, though GABAAergic drugs failed to affect M1/M2 or Ly-6Chigh/Ly-6Clow macrophage polarization directly. Accordingly, pro-inflammatory activities mediated by M1 or Ly-6Chigh macrophages were decreased and reparative processes mediated by M2 or Ly-6Clow macrophages were augmented. As a result, post-infarction ventricular remodelling was attenuated, as reflected by reduced infarct size and increased collagen density within infarcts. Echocardiographic indices, mortality and rupture rates were reduced. After depletion of monocytes/macrophages by clodronate liposomes, GABAAergic drugs exhibited no effect on cardiac dysfunction and surrogate clinical outcomes. Conclusion Control of the GABAA receptor activity in monocytes/macrophages can potently modulate post-infarction inflammation. Topiramate emerges as a promising drug, which may be feasible to translate for MI therapy in the future.
Collapse
Affiliation(s)
- Zhaohui Wang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, China
| | - Shiyuan Huang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, China
| | - Yuling Sheng
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, China
| | - Xu Peng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, China
| | - Hui Liu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, China
| | - Nan Jin
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, China
| | - Jun Cai
- Department of Emergency Surgery, the Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, 26 Shengli Ave, Wuhan 430014, China
| | - Yanwen Shu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, China
| | - Ting Li
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, China
| | - Ping Li
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, China
| | - Cheng Fan
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, China
| | - Xiaofan Hu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, China
| | - Wenyong Zhang
- Department of Geriatrics, LIYUAN Hospital, Tongji Medical College, Huazhong University of Science and Technology, 39 Yanhu Ave, Wuhan 430077, China
| | - Rui Long
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, China
| | - Ya You
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, China
| | - Caihong Huang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, China
| | - Yi Song
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, China
| | - Chunhua Xiang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, China
| | - Jue Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, China
| | - Yong Yang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, China
| | - Kun Liu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, China
| |
Collapse
|
68
|
Richardson WJ, Holmes JW. Emergence of Collagen Orientation Heterogeneity in Healing Infarcts and an Agent-Based Model. Biophys J 2017; 110:2266-77. [PMID: 27224491 DOI: 10.1016/j.bpj.2016.04.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 12/30/2015] [Accepted: 04/07/2016] [Indexed: 01/01/2023] Open
Abstract
Spatial heterogeneity of matrix structure can be an important determinant of tissue function. Although bulk properties of collagen structure in healing myocardial infarcts have been characterized previously, regional heterogeneity in infarct structure has received minimal attention. Herein, we quantified regional variations of collagen and nuclear orientations over the initial weeks of healing after infarction in rats, and employed a computational model of infarct remodeling to test potential explanations for the heterogeneity we observed in vivo. Fiber and cell orientation maps were generated from infarct samples acquired previously at 1, 2, 3, and 6 weeks postinfarction in a rat ligation model. We analyzed heterogeneity by calculating the dot product of each fiber or cell orientation vector with every other fiber or cell orientation vector, and plotting that dot product versus distance between the fibers or cells. This analysis revealed prominent regional heterogeneity, with alignment of both fibers and cell nuclei in local pockets far exceeding the global average. Using an agent-based model of fibroblast-mediated collagen remodeling, we found that similar levels of heterogeneity can spontaneously emerge from initially isotropic matrix via locally reinforcing cell-matrix interactions. Specifically, cells that sensed fiber orientation at a distance or remodeled fibers at a distance by traction-mediated reorientation or aligned deposition gave rise to regionally heterogeneous structures. However, only the simulations in which cells deposited collagen fibers aligned with their own orientation reproduced experimentally measured patterns of heterogeneity across all time points. These predictions warrant experimental follow-up to test the role of such mechanisms in vivo and identify opportunities to control heterogeneity for therapeutic benefit.
Collapse
Affiliation(s)
- William J Richardson
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia; Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia
| | - Jeffrey W Holmes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia; Department of Medicine, University of Virginia, Charlottesville, Virginia; Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia.
| |
Collapse
|
69
|
Arunachalam SP, Arani A, Baffour F, Rysavy JA, Rossman PJ, Glaser KJ, Lake DS, Trzasko JD, Manduca A, McGee KP, Ehman RL, Araoz PA. Regional assessment of in vivo myocardial stiffness using 3D magnetic resonance elastography in a porcine model of myocardial infarction. Magn Reson Med 2017; 79:361-369. [PMID: 28382658 DOI: 10.1002/mrm.26695] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 03/03/2017] [Accepted: 03/09/2017] [Indexed: 12/11/2022]
Abstract
PURPOSE The stiffness of a myocardial infarct affects the left ventricular pump function and remodeling. Magnetic resonance elastography (MRE) is a noninvasive imaging technique for measuring soft-tissue stiffness in vivo. The purpose of this study was to investigate the feasibility of assessing in vivo regional myocardial stiffness with high-frequency 3D cardiac MRE in a porcine model of myocardial infarction, and compare the results with ex vivo uniaxial tensile testing. METHODS Myocardial infarct was induced in a porcine model by embolizing the left circumflex artery. Fourteen days postinfarction, MRE imaging was performed in diastole using an echocardiogram-gated spin-echo echo-planar-imaging sequence with 140-Hz vibrations and 3D MRE processing. The MRE stiffness and tensile modulus from uniaxial testing were compared between the remote and infarcted myocardium. RESULTS Myocardial infarcts showed increased in vivo MRE stiffness compared with remote myocardium (4.6 ± 0.7 kPa versus 3.0 ± 0.6 kPa, P = 0.02) within the same pig. Ex vivo uniaxial mechanical testing confirmed the in vivo MRE results, showing that myocardial infarcts were stiffer than remote myocardium (650 ± 80 kPa versus 110 ± 20 kPa, P = 0.01). CONCLUSIONS These results demonstrate the feasibility of assessing in vivo regional myocardial stiffness with high-frequency 3D cardiac MRE. Magn Reson Med 79:361-369, 2018. © 2017 International Society for Magnetic Resonance in Medicine.
Collapse
Affiliation(s)
| | - Arvin Arani
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Francis Baffour
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Joseph A Rysavy
- Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Kevin J Glaser
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - David S Lake
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Armando Manduca
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Kiaran P McGee
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Richard L Ehman
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Philip A Araoz
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
70
|
Inhibition of the Renin-Angiotensin System Post Myocardial Infarction Prevents Inflammation-Associated Acute Cardiac Rupture. Cardiovasc Drugs Ther 2017; 31:145-156. [DOI: 10.1007/s10557-017-6717-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
71
|
Gong W, Feng S, Wang X, Fan J, Li A, Nie SP. Beta-blockers reduced the risk of cardiac rupture in patients with acute myocardial infarction: A meta-analysis of randomized control trials. Int J Cardiol 2017; 232:171-175. [PMID: 28109576 DOI: 10.1016/j.ijcard.2017.01.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 01/02/2017] [Accepted: 01/04/2017] [Indexed: 11/29/2022]
Abstract
BACKGROUND Cardiac rupture (CR) is a catastrophic complication that occurs after acute myocardial infarction (MI) and, at present, there are no effective pharmacological strategies for preventing this condition. The objective of this meta-analysis was to assess the effect of beta-blockers on CR in patients with acute MI. METHODS An extensive search of the PUBMED, EMBASE, ISI Web of Science, MEDLINE and Cochrane was performed to retrieve the studies of beta-blockers treatment in patients with acute MI. Data were combined using a random effects model. A meta-analysis was performed using Review Manager 5.3. RESULTS Four randomized controlled trials (RCTs) involving 68, 842 patients, 603 of whom occurred CR, were met criteria. Meta analysis showed that beta-blockers caused a statistically and clinically significant decrease in the incidence of CR of 32% (RR: 0.68, 95% CI: 0.47 to 0.99, P=0.04). CONCLUSIONS The findings of this meta-analysis confirmed that the early use of beta-blockers is associated with decreased incidence of CR, suggesting some beneficial effects of beta-blockers on infarct healing after acute MI.
Collapse
Affiliation(s)
- Wei Gong
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China; Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China
| | - Siting Feng
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China; Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China
| | - Xiao Wang
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China; Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China
| | - Jingyao Fan
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China; Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China
| | - Aobo Li
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China; Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China
| | - Shao-Ping Nie
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China; Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China.
| |
Collapse
|
72
|
Qian G, Jin RJ, Fu ZH, Yang YQ, Su HL, Dong W, Guo J, Jing J, Guo YL, Chen YD. Development and validation of clinical risk score to predict the cardiac rupture in patients with STEMI. Am J Emerg Med 2016; 35:589-593. [PMID: 28132793 DOI: 10.1016/j.ajem.2016.12.033] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 12/12/2016] [Accepted: 12/12/2016] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Cardiac rupture (CR) is a fatal complication of ST-elevation myocardial infarction (STEMI) with poor prognosis. The aim of this study was to develop and validate practical risk score to predict the CR after STEMI. METHODS A total of 11,234 STEMI patients from 7 centers in China were enrolled in our study, we firstly developed a simplified fast-track CR risk model from 7455 STEMI patients, and then prospectively validated the CR risk model using receiver-operating characteristic (ROC) curves by the other 3779 consecutive STEMI patients. This trial is registered with ClinicalTrials.gov, number NCT02484326. RESULTS The incidence of CR was 2.12% (238/11,234), and the thirty-day mortality in CR patients was 86%. We developed a risk model which had 7 independent baseline clinical predictors (female sex, advanced age, anterior myocardial infarction, delayed admission, heart rate, elevated white blood cell count and anemia). The CR risk score system differentiated STEMI patients with incidence of CR ranging from 0.2% to 13%. The risk score system demonstrated good predictive value with area under the ROC of 0.78 (95% CI 0.73-0.84) in validation cohort. Primary percutaneous coronary intervention decreased the incidence of CR in high risk group (3.9% vs. 6.2%, p<0.05) and very high risk group (8.0% vs. 15.2%, p<0.05). CONCLUSIONS A simple risk score system based on 7 baseline clinical variables could identify patients with high risk of CR, for whom appropriate treatment strategies can be implemented.
Collapse
Affiliation(s)
- Geng Qian
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Rong-Jie Jin
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Zhen-Hong Fu
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Yong-Qiang Yang
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Hong-Liang Su
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Wei Dong
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Jun Guo
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Jing Jing
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Yuan-Lin Guo
- Department of Cardiology, Beijing Fuwai Hospital, Beijing, China
| | - Yun-Dai Chen
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
73
|
Omiya S, Omori Y, Taneike M, Protti A, Yamaguchi O, Akira S, Shah AM, Nishida K, Otsu K. Toll-like receptor 9 prevents cardiac rupture after myocardial infarction in mice independently of inflammation. Am J Physiol Heart Circ Physiol 2016; 311:H1485-H1497. [PMID: 27769998 DOI: 10.1152/ajpheart.00481.2016] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 10/07/2016] [Accepted: 10/12/2016] [Indexed: 12/20/2022]
Abstract
We have reported that the Toll-like receptor 9 (TLR9) signaling pathway plays an important role in the development of pressure overload-induced inflammatory responses and heart failure. However, its role in cardiac remodeling after myocardial infarction has not been elucidated. TLR9-deficient and control C57Bl/6 wild-type mice were subjected to left coronary artery ligation. The survival rate 14 days postoperation was significantly lower in TLR9-deficient mice than that in wild-type mice with evidence of cardiac rupture in all dead mice. Cardiac magnetic resonance imaging showed no difference in infarct size and left ventricular wall thickness and function between TLR9-deficient and wild-type mice. There were no differences in the number of infiltrating inflammatory cells and the levels of inflammatory cytokine mRNA in infarct hearts between TLR9-deficient and wild-type mice. The number of α-smooth muscle actin (αSMA)-positive myofibroblasts and αSMA/Ki67-double-positive proliferative myofibroblasts was increased in the infarct and border areas in infarct hearts compared with those in sham-operated hearts in wild-type mice, but not in TLR9-deficient mice. The class B CpG oligonucleotide increased the phosphorylation level of NF-κB and the number of αSMA-positive and αSMA/Ki67-double-positive cells and these increases were attenuated by BAY1-7082, an NF-κB inhibitor, in cardiac fibroblasts isolated from wild-type hearts. The CpG oligonucleotide showed no effect on NF-κB activation or the number of αSMA-positive and αSMA/Ki67-double-positive cells in cardiac fibroblasts from TLR9-deficient hearts. Although the TLR9 signaling pathway is not involved in the acute inflammatory response in infarct hearts, it ameliorates cardiac rupture possibly by promoting proliferation and differentiation of cardiac fibroblasts.
Collapse
Affiliation(s)
- Shigemiki Omiya
- Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, London, United Kingdom
| | - Yosuke Omori
- Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, London, United Kingdom
| | - Manabu Taneike
- Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, London, United Kingdom
| | - Andrea Protti
- Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, London, United Kingdom
| | - Osamu Yamaguchi
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan; and
| | - Shizuo Akira
- Laboratory of Host Defense, WPI Immunology Frontier Research Centre, Osaka University, Suita, Japan
| | - Ajay M Shah
- Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, London, United Kingdom
| | - Kazuhiko Nishida
- Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, London, United Kingdom
| | - Kinya Otsu
- Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, London, United Kingdom;
| |
Collapse
|
74
|
Cao H, Zhang Q, He Y, Feng X, Liu Z. Teflon-buttressed sutures plus pericardium patch repair left ventricular rupture caused by radiofrequency catheter ablation: A case report. Medicine (Baltimore) 2016; 95:e4933. [PMID: 27661047 PMCID: PMC5044917 DOI: 10.1097/md.0000000000004933] [Citation(s) in RCA: 3] [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
BACKGROUND Cardiac rupture often occurs after myocardial infarction or chest trauma with a high mortality rate. However, left ventricular rupture caused by radiofrequency catheter ablation (RFCA) is extremely rare. METHODS We describe a case of a 61-year-old male who survived from left ventricular rupture caused by a RFCA procedure for frequent ventricular premature contractions. Surgical exploration with cardiopulmonary bypass (CPB) was performed when the signs of cardiac tamponade developed 7 hours after the ablation surgery. RESULTS Teflon-buttressed sutures of the tear in the left ventricular posterolateral wall and pericardium patch applied to the contusion region on the wall repaired the rupture safely and effectively. CONCLUSION Timely surgical intervention under CPB facilitated the survival of the patient. Teflon-buttressed sutures plus pericardium patch achieved the successful repair of the rupture.
Collapse
Affiliation(s)
| | | | | | - Xiaodong Feng
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Correspondence: Xiaodong Feng, Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China (e-mail: ); Zhongmin Liu, Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China (e-mail: )
| | - Zhongmin Liu
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Correspondence: Xiaodong Feng, Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China (e-mail: ); Zhongmin Liu, Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China (e-mail: )
| |
Collapse
|
75
|
Clarke SA, Richardson WJ, Holmes JW. Modifying the mechanics of healing infarcts: Is better the enemy of good? J Mol Cell Cardiol 2015; 93:115-24. [PMID: 26631496 DOI: 10.1016/j.yjmcc.2015.11.028] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 11/09/2015] [Accepted: 11/26/2015] [Indexed: 02/06/2023]
Abstract
Myocardial infarction (MI) is a major source of morbidity and mortality worldwide, with over 7 million people suffering infarctions each year. Heart muscle damaged during MI is replaced by a collagenous scar over a period of several weeks, and the mechanical properties of that scar tissue are a key determinant of serious post-MI complications such as infarct rupture, depression of heart function, and progression to heart failure. Thus, there is increasing interest in developing therapies that modify the structure and mechanics of healing infarct scar. Yet most prior attempts at therapeutic scar modification have failed, some catastrophically. This article reviews available information about the mechanics of healing infarct scar and the functional impact of scar mechanical properties, and attempts to infer principles that can better guide future attempts to modify scar. One important conclusion is that collagen structure, mechanics, and remodeling of healing infarct scar vary so widely among experimental models that any novel therapy should be tested across a range of species, infarct locations, and reperfusion protocols. Another lesson from past work is that the biology and mechanics of healing infarcts are sufficiently complex that the effects of interventions are often counterintuitive; for example, increasing infarct stiffness has little effect on heart function, and inhibition of matrix metalloproteases (MMPs) has little effect on scar collagen content. Computational models can help explain such counterintuitive results, and are becoming an increasingly important tool for integrating known information to better identify promising therapies and design experiments to test them. Moving forward, potentially exciting new opportunities for therapeutic modification of infarct mechanics include modulating anisotropy and promoting scar compaction.
Collapse
Affiliation(s)
- Samantha A Clarke
- Department of Biomedical Engineering, University of Virginia, United States
| | - William J Richardson
- Department of Biomedical Engineering, University of Virginia, United States; Robert M. Berne Cardiovascular Research Center, University of Virginia, United States
| | - Jeffrey W Holmes
- Department of Biomedical Engineering, University of Virginia, United States; Department of Medicine, University of Virginia, United States; Robert M. Berne Cardiovascular Research Center, University of Virginia, United States.
| |
Collapse
|
76
|
Holmes JW, Laksman Z, Gepstein L. Making better scar: Emerging approaches for modifying mechanical and electrical properties following infarction and ablation. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2015; 120:134-48. [PMID: 26615948 DOI: 10.1016/j.pbiomolbio.2015.11.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 11/13/2015] [Accepted: 11/20/2015] [Indexed: 12/31/2022]
Abstract
Following myocardial infarction (MI), damaged myocytes are replaced by collagenous scar tissue, which serves an important mechanical function - maintaining integrity of the heart wall against enormous mechanical forces - but also disrupts electrical function as structural and electrical remodeling in the infarct and borderzone predispose to re-entry and ventricular tachycardia. Novel emerging regenerative approaches aim to replace this scar tissue with viable myocytes. Yet an alternative strategy of therapeutically modifying selected scar properties may also prove important, and in some cases may offer similar benefits with lower risk or regulatory complexity. Here, we review potential goals for such modifications as well as recent proof-of-concept studies employing specific modifications, including gene therapy to locally increase conduction velocity or prolong the refractory period in and around the infarct scar, and modification of scar anisotropy to improve regional mechanics and pump function. Another advantage of scar modification techniques is that they have applications well beyond MI. In particular, ablation treats electrical abnormalities of the heart by intentionally generating scar to block aberrant conduction pathways. Yet in diseases such as atrial fibrillation (AF) where ablation can be extensive, treating the electrical disorder can significantly impair mechanical function. Creating smaller, denser scars that more effectively block conduction, and choosing the location of those lesions by balancing their electrical and mechanical impacts, could significantly improve outcomes for AF patients. We review some recent advances in this area, including the use of computational models to predict the mechanical effects of specific lesion sets and gene therapy for functional ablation. Overall, emerging techniques for modifying scar properties represents a potentially important set of tools for improving patient outcomes across a range of heart diseases, whether used in place of or as an adjunct to regenerative approaches.
Collapse
Affiliation(s)
- Jeffrey W Holmes
- Departments of Biomedical Engineering and Medicine, Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States.
| | - Zachary Laksman
- Cardiac Electrophysiology, University of British Columbia, Vancouver, BC, Canada
| | - Lior Gepstein
- Departments of Cardiology (Ramban Health Care Campus) and Physiology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
77
|
Wan F, Letavernier E, Le Saux CJ, Houssaini A, Abid S, Czibik G, Sawaki D, Marcos E, Dubois-Rande JL, Baud L, Adnot S, Derumeaux G, Gellen B. Calpastatin overexpression impairs postinfarct scar healing in mice by compromising reparative immune cell recruitment and activation. Am J Physiol Heart Circ Physiol 2015; 309:H1883-93. [PMID: 26453333 DOI: 10.1152/ajpheart.00594.2015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 10/03/2015] [Indexed: 12/15/2022]
Abstract
The activation of the calpain system is involved in the repair process following myocardial infarction (MI). However, the impact of the inhibition of calpain by calpastatin, its natural inhibitor, on scar healing and left ventricular (LV) remodeling is elusive. Male mice ubiquitously overexpressing calpastatin (TG) and wild-type (WT) controls were subjected to an anterior coronary artery ligation. Mortality at 6 wk was higher in TG mice (24% in WT vs. 44% in TG, P < 0.05) driven by a significantly higher incidence of cardiac rupture during the first week post-MI, despite comparable infarct size and LV dysfunction and dilatation. Calpain activation post-MI was blunted in TG myocardium. In TG mice, inflammatory cell infiltration and activation were reduced in the infarct zone (IZ), particularly affecting M2 macrophages and CD4(+) T cells, which are crucial for scar healing. To elucidate the role of calpastatin overexpression in macrophages, we stimulated peritoneal macrophages obtained from TG and WT mice in vitro with IL-4, yielding an abrogated M2 polarization in TG but not in WT cells. Lymphopenic Rag1(-/-) mice receiving TG splenocytes before MI demonstrated decreased T-cell recruitment and M2 macrophage activation in the IZ day 5 after MI compared with those receiving WT splenocytes. Calpastatin overexpression prevented the activation of the calpain system after MI. It also impaired scar healing, promoted LV rupture, and increased mortality. Defective scar formation was associated with blunted CD4(+) T-cell and M2-macrophage recruitment.
Collapse
Affiliation(s)
- Feng Wan
- Institut National de la Santé et de la Recherche Médicale U955, Université Paris-Est Creteil, Créteil, France
| | - Emmanuel Letavernier
- Department of Physiology, Assistance Publique-Hôpitaux de Paris (AP-HP), Tenon Hospital, Paris, France; Inflammation-Immunopathology-Biotherapy Department (DHU i2B), F-75020, Sorbonne Universités, Université Pierre et Marie Curie, Université Paris 06, Unités Mixtes de Recherche Scientifique 1155, Paris, France; and
| | - Claude Jourdan Le Saux
- Department of Medicine/Cardiology Division, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Amal Houssaini
- Institut National de la Santé et de la Recherche Médicale U955, Université Paris-Est Creteil, Créteil, France
| | - Shariq Abid
- Institut National de la Santé et de la Recherche Médicale U955, Université Paris-Est Creteil, Créteil, France
| | - Gabor Czibik
- Institut National de la Santé et de la Recherche Médicale U955, Université Paris-Est Creteil, Créteil, France
| | - Daigo Sawaki
- Institut National de la Santé et de la Recherche Médicale U955, Université Paris-Est Creteil, Créteil, France
| | - Elisabeth Marcos
- Institut National de la Santé et de la Recherche Médicale U955, Université Paris-Est Creteil, Créteil, France
| | - Jean-Luc Dubois-Rande
- Institut National de la Santé et de la Recherche Médicale U955, Université Paris-Est Creteil, Créteil, France; Département Hospitalo-Universitairé Ageing Thorax-Vessels Blood (DHU A-TVB), Department of Physiology, AP-HP, Henri Mondor Hospital, Créteil, France; DHU A-TVB, Department of Cardiology, AP-HP, Henri Mondor Hospital, Créteil, France
| | - Laurent Baud
- Department of Physiology, Assistance Publique-Hôpitaux de Paris (AP-HP), Tenon Hospital, Paris, France; Inflammation-Immunopathology-Biotherapy Department (DHU i2B), F-75020, Sorbonne Universités, Université Pierre et Marie Curie, Université Paris 06, Unités Mixtes de Recherche Scientifique 1155, Paris, France; and
| | - Serge Adnot
- Institut National de la Santé et de la Recherche Médicale U955, Université Paris-Est Creteil, Créteil, France; Département Hospitalo-Universitairé Ageing Thorax-Vessels Blood (DHU A-TVB), Department of Physiology, AP-HP, Henri Mondor Hospital, Créteil, France
| | - Geneviève Derumeaux
- Institut National de la Santé et de la Recherche Médicale U955, Université Paris-Est Creteil, Créteil, France; Département Hospitalo-Universitairé Ageing Thorax-Vessels Blood (DHU A-TVB), Department of Physiology, AP-HP, Henri Mondor Hospital, Créteil, France
| | - Barnabas Gellen
- Institut National de la Santé et de la Recherche Médicale U955, Université Paris-Est Creteil, Créteil, France; DHU A-TVB, Department of Cardiology, AP-HP, Henri Mondor Hospital, Créteil, France; Department of Cardiology, Poitiers University Hospital, F-86000, Poitiers, France
| |
Collapse
|
78
|
Richardson WJ, Clarke SA, Quinn TA, Holmes JW. Physiological Implications of Myocardial Scar Structure. Compr Physiol 2015; 5:1877-909. [PMID: 26426470 DOI: 10.1002/cphy.c140067] [Citation(s) in RCA: 192] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Once myocardium dies during a heart attack, it is replaced by scar tissue over the course of several weeks. The size, location, composition, structure, and mechanical properties of the healing scar are all critical determinants of the fate of patients who survive the initial infarction. While the central importance of scar structure in determining pump function and remodeling has long been recognized, it has proven remarkably difficult to design therapies that improve heart function or limit remodeling by modifying scar structure. Many exciting new therapies are under development, but predicting their long-term effects requires a detailed understanding of how infarct scar forms, how its properties impact left ventricular function and remodeling, and how changes in scar structure and properties feed back to affect not only heart mechanics but also electrical conduction, reflex hemodynamic compensations, and the ongoing process of scar formation itself. In this article, we outline the scar formation process following a myocardial infarction, discuss interpretation of standard measures of heart function in the setting of a healing infarct, then present implications of infarct scar geometry and structure for both mechanical and electrical function of the heart and summarize experiences to date with therapeutic interventions that aim to modify scar geometry and structure. One important conclusion that emerges from the studies reviewed here is that computational modeling is an essential tool for integrating the wealth of information required to understand this complex system and predict the impact of novel therapies on scar healing, heart function, and remodeling following myocardial infarction.
Collapse
Affiliation(s)
- William J Richardson
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA.,Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
| | - Samantha A Clarke
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - T Alexander Quinn
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jeffrey W Holmes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA.,Department of Medicine, University of Virginia, Charlottesville, Virginia, USA.,Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
79
|
Qian G, Wu C, Chen YD, Tu CC, Wang JW, Qian YA. Predictive factors of cardiac rupture in patients with ST-elevation myocardial infarction. J Zhejiang Univ Sci B 2015; 15:1048-54. [PMID: 25471834 DOI: 10.1631/jzus.b1400095] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Cardiac rupture (CR) is a potentially fatal mechanical complication of ST-elevation myocardial infarction (STEMI). We aimed to determine the incidence and risk factors of CR in Chinese STEMI patients. A total of 9798 consecutive STEMI patients from four centers in China were retrospectively analyzed, among which 178 patients had CR. STEMI patients without CR were chosen as a control group. Clinical characteristics were compared between STEMI patients with CR and those without CR. The incidence of CR in STEMI patients was 1.82%, and the 30-d mortality was up to 61.2%. CR patients were significantly older, more female, and associated with a longer time from onset of pain to hospital admission than their non-CR counterparts (P<0.001). More patients with anterior myocardial infarction (82.1%) were found in the CR group, and CR patients had significantly higher heart rates than the control group ((91±19) bpm vs. (71±16) bpm; P<0.001). In multiple-adjusted models, the independent risk factors of CR were advanced age, female gender, anaemia, increased heart rate, anterior myocardial infarction, increased white blood cell (WBC) count, delayed admission, and renal dysfunction. The level of hemoglobin remained a significant determinant factor of CR (OR (95% CI): 0.82 (0.75-0.89); P<0.001) after adjusting for various potential confounding factors. Counts of WBC also remained a significant determinant of the CR (OR (95% CI): 1.08 (1.04-1.12); P<0.001). A number of variables were independently related to CR. This study indicated, for the first time, that both hemoglobin and WBC levels were independently correlated with occurrence of CR.
Collapse
Affiliation(s)
- Geng Qian
- Department of Cardiology, Chinese PLA General Hospital, Beijing 100853, China; Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China; Department of Cardiology, Lujiang People's Hospital, Anhui 231500, China
| | | | | | | | | | | |
Collapse
|
80
|
Trivedi KR, Aldebert P, Riberi A, Mancini J, Levy G, Macia JC, Quilicci J, Habib G, Fraisse A. Sequential management of post-myocardial infarction ventricular septal defects. Arch Cardiovasc Dis 2015; 108:321-30. [DOI: 10.1016/j.acvd.2015.01.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 12/23/2014] [Accepted: 01/12/2015] [Indexed: 01/20/2023]
|
81
|
Patel BB, Kasneci A, Bolt AM, Di Lalla V, Di Iorio MR, Raad M, Mann KK, Chalifour LE. Chronic Exposure to Bisphenol A Reduces Successful Cardiac Remodeling After an Experimental Myocardial Infarction in Male C57bl/6n Mice. Toxicol Sci 2015; 146:101-15. [PMID: 25862758 DOI: 10.1093/toxsci/kfv073] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Estrogenic compounds such as bisphenol A (BPA) leach from plastics into food and beverage containers. Increased BPA exposure has been correlated with increased cardiovascular disease. To test the hypothesis that increased BPA exposure reduces cardiovascular remodeling, we chronically exposed C57bl/6n male mice to BPA and performed a myocardial infarction (MI). We measured cardiac function, as well as myeloid and cardiac fibroblast accumulation and activity. We found increased early death as well as increased cardiac dilation and reduced cardiac function in surviving BPA-exposed mice. Matrix metalloproteinase-2 (MMP2) protein and activity were increased 1.5-fold in BPA-exposed heart. BPA-exposed mice had similar neutrophil infiltration; however, monocyte and macrophage (MΦ) infiltration into the ischemic area was 5-fold greater than VEH mice potentially due to a 2-fold increase in monocyte chemoattractant protein-1. Monocyte and MΦ exposure to BPA in vitro in primary bone marrow cultures or in isolated peritoneal MΦ increased polarization to an activated MΦ, increased MMP2 and MMP9 expression 2-fold and activity 3-fold, and increased uptake of microspheres 3-fold. Cardiac fibroblasts (CF) differentiate to α-smooth muscle actin (αSMA) expressing myofibroblasts, migrate to the ischemic area and secrete collagen to strengthen the scar. Collagen and αSMA expression were reduced 50% in BPA-exposed hearts. Chronic in vivo or continuous in vitro BPA exposure ablated transforming growth factor beta-mediated differentiation of CF, reduced αSMA expression 50% and reduced migration 40% yet increased secreted MMP2 activity 2-fold. We conclude that chronic BPA exposure reduces the ability to successfully remodel after an MI by increasing MΦ-based inflammation and reducing myofibroblast repair function.
Collapse
Affiliation(s)
- Bhavini B Patel
- *Lady Davis Institute for Medical Research, Montréal, Québec H3T 1E2, Canada, Department of Oncology, Jewish General Hospital, 3755 Chemin Cote Ste Catherine, Montréal, Québec H3T 1E2, Canada, Division of Experimental Medicine, Department of Medicine, McGill University, 850 Sherbrooke Street, Montréal, Québec H3A 1A2, Canada, Division of Cardiology and Division of Endocrinology, Jewish General Hospital, 3755 Chemin Cote Ste Catherine, Montréal, Québec H3T 1E2, Canada
| | - Amanda Kasneci
- *Lady Davis Institute for Medical Research, Montréal, Québec H3T 1E2, Canada, Department of Oncology, Jewish General Hospital, 3755 Chemin Cote Ste Catherine, Montréal, Québec H3T 1E2, Canada, Division of Experimental Medicine, Department of Medicine, McGill University, 850 Sherbrooke Street, Montréal, Québec H3A 1A2, Canada, Division of Cardiology and Division of Endocrinology, Jewish General Hospital, 3755 Chemin Cote Ste Catherine, Montréal, Québec H3T 1E2, Canada
| | - Alicia M Bolt
- *Lady Davis Institute for Medical Research, Montréal, Québec H3T 1E2, Canada, Department of Oncology, Jewish General Hospital, 3755 Chemin Cote Ste Catherine, Montréal, Québec H3T 1E2, Canada, Division of Experimental Medicine, Department of Medicine, McGill University, 850 Sherbrooke Street, Montréal, Québec H3A 1A2, Canada, Division of Cardiology and Division of Endocrinology, Jewish General Hospital, 3755 Chemin Cote Ste Catherine, Montréal, Québec H3T 1E2, Canada *Lady Davis Institute for Medical Research, Montréal, Québec H3T 1E2, Canada, Department of Oncology, Jewish General Hospital, 3755 Chemin Cote Ste Catherine, Montréal, Québec H3T 1E2, Canada, Division of Experimental Medicine, Department of Medicine, McGill University, 850 Sherbrooke Street, Montréal, Québec H3A 1A2, Canada, Division of Cardiology and Division of Endocrinology, Jewish General Hospital, 3755 Chemin Cote Ste Catherine, Montréal, Québec H3T 1E2, Canada
| | - Vanessa Di Lalla
- *Lady Davis Institute for Medical Research, Montréal, Québec H3T 1E2, Canada, Department of Oncology, Jewish General Hospital, 3755 Chemin Cote Ste Catherine, Montréal, Québec H3T 1E2, Canada, Division of Experimental Medicine, Department of Medicine, McGill University, 850 Sherbrooke Street, Montréal, Québec H3A 1A2, Canada, Division of Cardiology and Division of Endocrinology, Jewish General Hospital, 3755 Chemin Cote Ste Catherine, Montréal, Québec H3T 1E2, Canada
| | - Massimo R Di Iorio
- *Lady Davis Institute for Medical Research, Montréal, Québec H3T 1E2, Canada, Department of Oncology, Jewish General Hospital, 3755 Chemin Cote Ste Catherine, Montréal, Québec H3T 1E2, Canada, Division of Experimental Medicine, Department of Medicine, McGill University, 850 Sherbrooke Street, Montréal, Québec H3A 1A2, Canada, Division of Cardiology and Division of Endocrinology, Jewish General Hospital, 3755 Chemin Cote Ste Catherine, Montréal, Québec H3T 1E2, Canada
| | - Mohamad Raad
- *Lady Davis Institute for Medical Research, Montréal, Québec H3T 1E2, Canada, Department of Oncology, Jewish General Hospital, 3755 Chemin Cote Ste Catherine, Montréal, Québec H3T 1E2, Canada, Division of Experimental Medicine, Department of Medicine, McGill University, 850 Sherbrooke Street, Montréal, Québec H3A 1A2, Canada, Division of Cardiology and Division of Endocrinology, Jewish General Hospital, 3755 Chemin Cote Ste Catherine, Montréal, Québec H3T 1E2, Canada
| | - Koren K Mann
- *Lady Davis Institute for Medical Research, Montréal, Québec H3T 1E2, Canada, Department of Oncology, Jewish General Hospital, 3755 Chemin Cote Ste Catherine, Montréal, Québec H3T 1E2, Canada, Division of Experimental Medicine, Department of Medicine, McGill University, 850 Sherbrooke Street, Montréal, Québec H3A 1A2, Canada, Division of Cardiology and Division of Endocrinology, Jewish General Hospital, 3755 Chemin Cote Ste Catherine, Montréal, Québec H3T 1E2, Canada *Lady Davis Institute for Medical Research, Montréal, Québec H3T 1E2, Canada, Department of Oncology, Jewish General Hospital, 3755 Chemin Cote Ste Catherine, Montréal, Québec H3T 1E2, Canada, Division of Experimental Medicine, Department of Medicine, McGill University, 850 Sherbrooke Street, Montréal, Québec H3A 1A2, Canada, Division of Cardiology and Division of Endocrinology, Jewish General Hospital, 3755 Chemin Cote Ste Catherine, Montréal, Québec H3T 1E2, Canada *Lady Davis Institute for Medical Research, Montréal, Québec H3T 1E2, Canada, Department of Oncology, Jewish General Hospital, 3755 Chemin Cote Ste Catherine, Montréal, Québec H3T 1E2, Canada, Division of Experimental Medicine, Department of Medicine, McGill University, 850 Sherbrooke Street, Montréal, Québec H3A 1A2, Canada, Division of Cardiology and Division of Endocrinology, Jewish General Hospital, 3755 Chemin Cote Ste Catherine, Montréal, Québec H3T 1E2, Canada
| | - Lorraine E Chalifour
- *Lady Davis Institute for Medical Research, Montréal, Québec H3T 1E2, Canada, Department of Oncology, Jewish General Hospital, 3755 Chemin Cote Ste Catherine, Montréal, Québec H3T 1E2, Canada, Division of Experimental Medicine, Department of Medicine, McGill University, 850 Sherbrooke Street, Montréal, Québec H3A 1A2, Canada, Division of Cardiology and Division of Endocrinology, Jewish General Hospital, 3755 Chemin Cote Ste Catherine, Montréal, Québec H3T 1E2, Canada *Lady Davis Institute for Medical Research, Montréal, Québec H3T 1E2, Canada, Department of Oncology, Jewish General Hospital, 3755 Chemin Cote Ste Catherine, Montréal, Québec H3T 1E2, Canada, Division of Experimental Medicine, Department of Medicine, McGill University, 850 Sherbrooke Street, Montréal, Québec H3A 1A2, Canada, Division of Cardiology and Division of Endocrinology, Jewish General Hospital, 3755 Chemin Cote Ste Catherine, Montréal, Québec H3T 1E2, Canada *Lady Davis Institute for Medical Research, Montréal, Québec H3T 1E2, Canada, Department of Oncology, Jewish General Hospital, 3755 Chemin Cote Ste Catherine, Montréal, Québec H3T 1E2, Canada, Division of Experimental Medicine, Department of Medicine, McGill University, 850 Sherbrooke Street, Montréal, Québec H3A 1A2, Canada, Division of Cardiology and Division of Endocrinology, Jewish General Hospital, 3755 Chemin Cote Ste Catherine, Montréal, Québec H3T 1E2, Canada *Lady Davis Institute for Medical Research, Montréal, Québec H3T 1E2, Canada, Department of Oncology, Jewish General Hospital, 3755 Chemin Cote Ste Catherine, Montréal, Québec H3T 1E2, Canada, Division of Experimental Medicine, Department of Medicine, McGill University, 850 Sherbrooke Street, Montréal, Québec H3A 1A2, Canada, Division of Cardiology and Division of Endocrinology, Jewish General Hospital, 3755 Chemin Cote Ste Catherine, Montréal, Québec H3T 1E2, Canada
| |
Collapse
|
82
|
Qin C, Yang YH, May L, Gao X, Stewart AG, Tu Y, Woodman OL, Ritchie RH. Cardioprotective potential of annexin-A1 mimetics in myocardial infarction. Pharmacol Ther 2014; 148:47-65. [PMID: 25460034 DOI: 10.1016/j.pharmthera.2014.11.012] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 11/14/2014] [Indexed: 12/15/2022]
Abstract
Myocardial infarction (MI) and its resultant heart failure remains a major cause of death in the world. The current treatments for patients with MI are revascularization with thrombolytic agents or interventional procedures. These treatments have focused on restoring blood flow to the ischemic tissue to prevent tissue necrosis and preserve organ function. The restoration of blood flow after a period of ischemia, however, may elicit further myocardial damage, called reperfusion injury. Pharmacological interventions, such as antioxidant and Ca(2+) channel blockers, have shown premises in experimental settings; however, clinical studies have shown limited success. Thus, there is a need for the development of novel therapies to treat reperfusion injury. The therapeutic potential of glucocorticoid-regulated anti-inflammatory mediator annexin-A1 (ANX-A1) has recently been recognized in a range of systemic inflammatory disorders. ANX-A1 binds to and activates the family of formyl peptide receptors (G protein-coupled receptor family) to inhibit neutrophil activation, migration and infiltration. Until recently, studies on the cardioprotective actions of ANX-A1 and its peptide mimetics (Ac2-26, CGEN-855A) have largely focused on its anti-inflammatory effects as a mechanism of preserving myocardial viability following I-R injury. Our laboratory provided the first evidence of the direct protective action of ANX-A1 on myocardium, independent of inflammatory cells in vitro. We now review the potential for ANX-A1 based therapeutics to be seen as a "triple shield" therapy against myocardial I-R injury, limiting neutrophil infiltration and preserving both cardiomyocyte viability and contractile function. This novel therapy may thus represent a valuable clinical approach to improve outcome after MI.
Collapse
Affiliation(s)
- Chengxue Qin
- Baker IDI Heart & Diabetes Institute, Melbourne, Victoria, Australia; Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, Victoria, Australia
| | - Yuan H Yang
- Centre for Inflammatory Diseases Monash University and Monash Medical Centre, Clayton, Victoria, Australia
| | - Lauren May
- Department of Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, and Department of Pharmacology, Monash University, Parkville, Victoria, Australia
| | - Xiaoming Gao
- Baker IDI Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Alastair G Stewart
- Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, Victoria, Australia
| | - Yan Tu
- Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, Victoria, Australia
| | - Owen L Woodman
- School of Medical Sciences, RMIT University, Bundoora 3083, Victoria, Australia
| | - Rebecca H Ritchie
- Baker IDI Heart & Diabetes Institute, Melbourne, Victoria, Australia; Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, Victoria, Australia; Department of Medicine, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
83
|
Luo T, Kim JK, Chen B, Abdel-Latif A, Kitakaze M, Yan L. Attenuation of ER stress prevents post-infarction-induced cardiac rupture and remodeling by modulating both cardiac apoptosis and fibrosis. Chem Biol Interact 2014; 225:90-8. [PMID: 25450231 DOI: 10.1016/j.cbi.2014.10.032] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 10/15/2014] [Accepted: 10/30/2014] [Indexed: 12/20/2022]
Abstract
Endoplasmic reticulum (ER) stress is implicated in the pathophysiology of various cardiovascular diseases, but the role of ER stress in cardiac rupture and/or remodeling after myocardial infarction (MI) is still unclear. Here we investigated whether ER stress plays a major role for these processes in mice. We ligated the left coronary artery (LCA) without reperfusion in mice and administered either NaCl or 4-phenylbutyric acid (4-PBA, 20 mg/kg/d) intraperitoneally for 4 weeks. Cardiac rupture rates during the first week of MI were 37.5% and 18.2% in the control and 4-PBA groups, respectively. The extent of ventricular aneurysm and fibrosis was less, and the cardiac function better, in the 4-PBA group compared with the control group. The protein levels of ER stress markers in the heart tissues of the control group remained elevated during the entire 4-week period after MI, while pro-apoptotic proteins mainly increased in the early phase, and the pro-fibrotic proteins markedly increased in the late phase post MI; 4-PBA decreased all of these protein levels. In the primary cultured neonatal rat cardiomyocytes or fibroblasts, hypoxia (3% O2) increased the number of apoptotic cardiomyocytes and promoted the proliferation and migration of fibroblasts, all of which were attenuated by 4-PBA (0.5 mM). These findings indicate that MI induces ER stress and provokes cardiac apoptosis and fibrosis, culminating in cardiac rupture and remodeling, and that the attenuation of ER stress could be an effective therapeutic target to prevent post-MI complications.
Collapse
Affiliation(s)
- Tao Luo
- Division of Cardiology, Department of Medicine, University of California Irvine Medical Center, Orange, CA 92868, USA; Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou 510632, China.
| | - Jin Kyung Kim
- Division of Cardiology, Department of Medicine, University of California Irvine Medical Center, Orange, CA 92868, USA
| | - Baihe Chen
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ahmed Abdel-Latif
- Saha Cardiovascular Research Center, University of Kentucky, Lexington 40536-0509, USA
| | - Masafumi Kitakaze
- Department of Clinical Research and Development, National Cerebral and Cardiovascular Center, 5-7-1 Fujishirodai, Suita 5675-8565, Japan
| | - Liang Yan
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou 510632, China
| |
Collapse
|
84
|
Whittaker P, Przyklenk K. From ischemic conditioning to 'hyperconditioning': clinical phenomenon and basic science opportunity. Dose Response 2014; 12:650-63. [PMID: 25552962 DOI: 10.2203/dose-response.14-035.whittaker] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Thousands of articles have been published on the topic of ischemic conditioning. Nevertheless, relatively little attention has been given to assessment of conditioning's dose-response characteristics. Specifically, the consequences of multiple conditioning episodes, what we will term "hyperconditioning", have seldom been examined. We propose that hyperconditioning warrants investigation because it; (1) may be of clinical importance, (2) could provide insight into conditioning mechanisms, and (3) might result in development of novel models of human disease. The prevalence of angina pectoris and intermittent claudication is sufficiently high and the potential for daily ischemia-reperfusion episodes sufficiently large that hyperconditioning is a clinically relevant phenomenon. In basic science, attenuation of conditioning-mediated infarct size reduction found in some studies after hyperconditioning offers a possible means to facilitate further discernment of cardioprotective signaling pathways. Moreover, hyperconditioning's impact extends beyond cytoprotection to tissue structural elements. Several studies demonstrate that hyperconditioning produces collagen injury (primarily fiber breakage). Such structural impairment could have adverse clinical consequences; however, in laboratory studies, selective collagen damage could provide the basis for models of cardiac rupture and dilated cardiomyopathy. Accordingly, we propose that hyperconditioning represents the dark, but potentially illuminating, side of ischemic conditioning - a paradigm that merits attention and prospective evaluation.
Collapse
Affiliation(s)
- Peter Whittaker
- Cardiovascular Research Institute and Department of Emergency Medicine, Wayne State University School of Medicine, Detroit 48201
| | - Karin Przyklenk
- Cardiovascular Research Institute and Department of Physiology, and Department of Emergency Medicine, Wayne State University School of Medicine, Detroit 48201
| |
Collapse
|
85
|
Chen B, Lu D, Fu Y, Zhang J, Huang X, Cao S, Xu D, Bin J, Kitakaze M, Huang Q, Liao Y. Olmesartan prevents cardiac rupture in mice with myocardial infarction by modulating growth differentiation factor 15 and p53. Br J Pharmacol 2014; 171:3741-53. [PMID: 24749959 PMCID: PMC4128070 DOI: 10.1111/bph.12736] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Revised: 04/02/2014] [Accepted: 04/11/2014] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND AND PURPOSE Cardiac rupture is a catastrophic complication that occurs after acute myocardial infarction (MI) and, at present, there are no effective pharmacological strategies for preventing this condition. Here we investigated the effect of the angiotensin II receptor blocker olmesartan (Olm) on post-infarct cardiac rupture and its underlying mechanisms of action. EXPERIMENTAL APPROACH C57Bl/6 mice with MI were treated with Olm, aldosterone (Aldo) or vehicle. Cultured neonatal cardiomyocytes and fibroblasts were exposed to normoxia or anoxia and treated with angiotensin II (Ang II), RNH6270 (active ingredient of Olm) or Aldo. KEY RESULTS The mortality rate and incidence of cardiac rupture in MI mice during the first week in the Olm-treated group were significantly lower than in the vehicle-treated group. Olm or RNH6270 reduced myeloperoxidase staining in the infarcted myocardium, decreased apoptosis in cultured cardiomyocytes and fibroblasts, as assessed by Hoechst staining and TUNEL assay, attenuated the accumulation of p53 and phosphorylated p53 and cleaved caspase 3 induced by MI or Ang II, as assessed by Western blotting, and up-regulated growth differentiation factor-15 (GDF-15). In cultured cardiomyocytes and fibroblasts, treatment with Ang II, Aldo or anoxia significantly down-regulated the expression of GDF-15. CONCLUSIONS AND IMPLICATIONS Olm prevents cardiac rupture through inhibition of apoptosis and inflammation, which is attributable to the down-regulation of p53 activity and up-regulation of GDF-15. Our findings suggest that early administration of an AT1 receptor anatagonist to patients with acute MI is a potential preventive approach for cardiac rupture.
Collapse
Affiliation(s)
- Baihe Chen
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang HospitalGuangzhou, China
| | - Di Lu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang HospitalGuangzhou, China
| | - Yujuan Fu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang HospitalGuangzhou, China
| | - Jingwen Zhang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang HospitalGuangzhou, China
| | - Xiaobo Huang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang HospitalGuangzhou, China
| | - Shiping Cao
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang HospitalGuangzhou, China
| | - Dingli Xu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang HospitalGuangzhou, China
| | - Jianping Bin
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang HospitalGuangzhou, China
| | - Masafumi Kitakaze
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang HospitalGuangzhou, China
- Cardiovascular Division of the Department of Medicine, National Cerebral and Cardiovascular CenterSuita, Osaka, Japan
| | - Qiaobing Huang
- Department of Pathophysiology, School of Basic Medicine, Southern Medical UniversityGuangzhou, China
| | - Yulin Liao
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang HospitalGuangzhou, China
| |
Collapse
|
86
|
Peng H, Xu J, Yang XP, Dai X, Peterson EL, Carretero OA, Rhaleb NE. Thymosin-β4 prevents cardiac rupture and improves cardiac function in mice with myocardial infarction. Am J Physiol Heart Circ Physiol 2014; 307:H741-51. [PMID: 25015963 DOI: 10.1152/ajpheart.00129.2014] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Thymosin-β4 (Tβ4) promotes cell survival, angiogenesis, and tissue regeneration and reduces inflammation. Cardiac rupture after myocardial infarction (MI) is mainly the consequence of excessive regional inflammation, whereas cardiac dysfunction after MI results from a massive cardiomyocyte loss and cardiac fibrosis. It is possible that Tβ4 reduces the incidence of cardiac rupture post-MI via anti-inflammatory actions and that it decreases adverse cardiac remodeling and improves cardiac function by promoting cardiac cell survival and cardiac repair. C57BL/6 mice were subjected to MI and treated with either vehicle or Tβ4 (1.6 mg·kg(-1)·day(-1) ip via osmotic minipump) for 7 days or 5 wk. Mice were assessed for 1) cardiac remodeling and function by echocardiography; 2) inflammatory cell infiltration, capillary density, myocyte apoptosis, and interstitial collagen fraction histopathologically; 3) gelatinolytic activity by in situ zymography; and 4) expression of ICAM-1 and p53 by immunoblot analysis. Tβ4 reduced cardiac rupture that was associated with a decrease in the numbers of infiltrating inflammatory cells and apoptotic myocytes, a decrease in gelatinolytic activity and ICAM-1 and p53 expression, and an increase in the numbers of CD31-positive cells. Five-week treatment with Tβ4 ameliorated left ventricular dilation, improved cardiac function, markedly reduced interstitial collagen fraction, and increased capillary density. In a murine model of acute MI, Tβ4 not only decreased mortality rate as a result of cardiac rupture but also significantly improved cardiac function after MI. Thus, the use of Tβ4 could be explored as an alternative therapy in preventing cardiac rupture and restoring cardiac function in patients with MI.
Collapse
Affiliation(s)
- Hongmei Peng
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Jiang Xu
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Xiao-Ping Yang
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Xiangguo Dai
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Edward L Peterson
- Department of Public Health Sciences, Henry Ford Hospital, Detroit, Michigan; and
| | - Oscar A Carretero
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Nour-Eddine Rhaleb
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan; Department of Physiology, Wayne State University, Detroit, Michigan
| |
Collapse
|
87
|
Role of MIF in myocardial ischaemia and infarction: insight from recent clinical and experimental findings. Clin Sci (Lond) 2014; 127:149-61. [PMID: 24697297 DOI: 10.1042/cs20130828] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
First discovered in 1966 as an inflammatory cytokine, MIF (macrophage migration inhibitory factor) has been extensively studied for its pivotal role in a variety of inflammatory diseases, including rheumatoid arthritis and atherosclerosis. Although initial studies over a decade ago reported increases in circulating MIF levels following acute MI (myocardial infarction), the dynamic changes in MIF and its pathophysiological significance following MI have been unknown until recently. In the present review, we summarize recent experimental and clinical studies examining the diverse functions of MIF across the spectrum of acute MI from brief ischaemia to post-infarct healing. Following an acute ischaemic insult, MIF is rapidly released from jeopardized cardiomyocytes, followed by a persistent MIF production and release from activated immune cells, resulting in a sustained increase in circulating levels of MIF. Recent studies have documented two distinct actions of MIF following acute MI. In the supra-acute phase of ischaemia, MIF mediates cardioprotection via several distinct mechanisms, including metabolic activation, apoptosis suppression and antioxidative stress. In prolonged myocardial ischaemia, however, MIF promotes inflammatory responses with largely detrimental effects on cardiac function and remodelling. The pro-inflammatory properties of MIF are complex and involve MIF derived from cardiac and immune cells contributing sequentially to the innate immune response evoked by MI. Emerging evidence on the role of MIF in myocardial ischaemia and infarction highlights a significant potential for the clinical use of MIF agonists or antagonists and as a unique cardiac biomarker.
Collapse
|
88
|
Unsöld B, Kaul A, Sbroggiò M, Schubert C, Regitz-Zagrosek V, Brancaccio M, Damilano F, Hirsch E, Van Bilsen M, Munts C, Sipido K, Bito V, Detre E, Wagner NM, Schäfer K, Seidler T, Vogt J, Neef S, Bleckmann A, Maier LS, Balligand JL, Bouzin C, Ventura-Clapier R, Garnier A, Eschenhagen T, El-Armouche A, Knöll R, Tarone G, Hasenfuß G. Melusin protects from cardiac rupture and improves functional remodelling after myocardial infarction. Cardiovasc Res 2013; 101:97-107. [DOI: 10.1093/cvr/cvt235] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Abstract
Aims
Melusin is a muscle-specific chaperone protein whose expression is required for a compensatory hypertrophy response to pressure overload. Here, we evaluated the consequences of melusin overexpression in the setting of myocardial infarction (MI) using a comprehensive multicentre approach.
Methods and results
Mice overexpressing melusin in the heart (TG) and wild-type controls (WT) were subjected to permanent LAD ligation and both the acute response (Day 3) and subsequent remodelling (2 weeks) were examined. Mortality in wild-type mice was significant between Days 3 and 7, primarily due to cardiac rupture, but melusin's overexpression strongly reduced mortality (43.2% in wild-type vs. 27.3% in melusin-TG, P = 0.005). At Day 3 after MI, a time point preceding the mortality peak, TG hearts had increased heat shock protein 70 expression, increased ERK1/2 signalling, reduced cardiomyocyte hyper-contractility and inflammatory cell infiltrates, and increased matricellular protein expression in the infarcted area.
At 2 weeks after MI, melusin overexpression conferred a favourable adaptive remodelling characterized by reduced left ventricle dilatation and better preserved contractility in the presence of a comparable degree of hypertrophy. Adaptive remodelling in melusin TG mice was characterized by reduced apoptosis and fibrosis as well as increased cardiomyocyte contractility.
Conclusions
Consistent with its function as a chaperone protein, melusin overexpression exerts a dual protective action following MI reducing an array of maladaptive processes. In the early phase after MI, reduced inflammation and myocyte remodelling protect against cardiac rupture. Chronically, reduced myocyte loss and matrix remodelling, with preserved myocyte contractility, confer adaptive LV remodelling.
Collapse
Affiliation(s)
- Bernhard Unsöld
- Department of Cardiology, University of Göttingen, Heart Research Center Göttingen, Göttingen, Germany
| | - Axel Kaul
- Department of Cardiology, University of Göttingen, Heart Research Center Göttingen, Göttingen, Germany
| | - Mauro Sbroggiò
- Dipartimento di Biotecnologie Molecolari e Scienze per la Salute, Università di Torino, Turin, Italy
| | - Carola Schubert
- Institute of Gender in Medicine, Charité-Universitätsmedizin, Berlin, Germany
| | | | - Mara Brancaccio
- Dipartimento di Biotecnologie Molecolari e Scienze per la Salute, Università di Torino, Turin, Italy
| | - Federico Damilano
- Dipartimento di Biotecnologie Molecolari e Scienze per la Salute, Università di Torino, Turin, Italy
| | - Emilio Hirsch
- Dipartimento di Biotecnologie Molecolari e Scienze per la Salute, Università di Torino, Turin, Italy
| | - Marc Van Bilsen
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Chantal Munts
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Karin Sipido
- Division of Experimental Cardiology, Department of Cardiovascular Medicine, University of Leuven, Leuven, Belgium
| | - Virginie Bito
- Division of Experimental Cardiology, Department of Cardiovascular Medicine, University of Leuven, Leuven, Belgium
| | - Elke Detre
- Division of Experimental Cardiology, Department of Cardiovascular Medicine, University of Leuven, Leuven, Belgium
| | - Nana Maria Wagner
- Department of Cardiology, University of Göttingen, Heart Research Center Göttingen, Göttingen, Germany
| | - Katrin Schäfer
- Department of Cardiology, University of Göttingen, Heart Research Center Göttingen, Göttingen, Germany
| | - Tim Seidler
- Department of Cardiology, University of Göttingen, Heart Research Center Göttingen, Göttingen, Germany
| | - Johannes Vogt
- Department of Cardiology, University of Göttingen, Heart Research Center Göttingen, Göttingen, Germany
| | - Stefan Neef
- Department of Cardiology, University of Göttingen, Heart Research Center Göttingen, Göttingen, Germany
| | - Annalen Bleckmann
- Department of Hematology and Oncology, University of Göttingen, Göttingen, Germany
| | - Lars S. Maier
- Department of Cardiology, University of Göttingen, Heart Research Center Göttingen, Göttingen, Germany
| | - Jean Luc Balligand
- Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Caroline Bouzin
- Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | | | | | - Thomas Eschenhagen
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Ali El-Armouche
- Department of Pharmacology, University of Göttingen, Heart Research Center Göttingen, Göttingen, Germany
| | - Ralph Knöll
- Myocardial Genetics, Imperial College London, London, UK
| | - Guido Tarone
- Dipartimento di Biotecnologie Molecolari e Scienze per la Salute, Università di Torino, Turin, Italy
| | - Gerd Hasenfuß
- Department of Cardiology, University of Göttingen, Heart Research Center Göttingen, Göttingen, Germany
| |
Collapse
|
89
|
Arslan F, Smeets MB, Buttari B, Profumo E, Riganò R, Akeroyd L, Kara E, Timmers L, Sluijter JP, van Middelaar B, den Ouden K, Pasterkamp G, Lim SK, de Kleijn DPV. Lack of haptoglobin results in unbalanced VEGFα/angiopoietin-1 expression, intramural hemorrhage and impaired wound healing after myocardial infarction. J Mol Cell Cardiol 2012; 56:116-28. [PMID: 23274064 DOI: 10.1016/j.yjmcc.2012.12.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Revised: 12/05/2012] [Accepted: 12/17/2012] [Indexed: 12/20/2022]
Abstract
Decreased haptoglobin (Hp) functionality due to allelic variations is associated with worsened outcome in patients after myocardial infarction (MI). However, mechanisms through which haptoglobin deficiency impairs cardiac repair remain to be elucidated. In the present study, we identified novel molecular alterations mediated by Hp involved in early and late cardiac repair responses after left coronary artery ligation in Hp(-/-) and wild-type (WT) mice. We observed a higher mortality rate in Hp(-/-) mice despite similar infarct size between groups. Deaths were commonly caused by cardiac rupture in Hp(-/-) animals. Histological analysis of 3 and 7days old non-ruptured infarcted hearts revealed more frequent and more severe intramural hemorrhage and increased leukocyte infiltration in Hp(-/-) mice. Analyses of non-ruptured hearts revealed increased oxidative stress, reduced PAI-1 activity and enhanced VEGFα transcription in Hp(-/-) mice. In line with these observations, we found increased microvascular permeability in Hp(-/-) hearts 3days after infarction. In vitro, haptoglobin prevented hemoglobin-induced oxidative stress and restored VEGF/Ang-1 balance in endothelial cell cultures. During long-term follow-up of the surviving animals, we observed altered matrix turnover, impaired scar formation and worsened cardiac function and geometry in Hp(-/-)mice. In conclusion, haptoglobin deficiency severely deteriorates tissue repair and cardiac performance after experimental MI. Haptoglobin plays a crucial role in both short- and long-term cardiac repair responses by reducing oxidative stress, maintaining microvascular integrity, myocardial architecture and proper scar formation.
Collapse
Affiliation(s)
- Fatih Arslan
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|