1
|
Zhou H, Ji Y, Li J, Sun L. The study on the role of O-GlcNAcylation of SIRT3 in regulating mitochondrial oxidative stress during simulate myocardial ischemia-reperfusion. Sci Rep 2024; 14:21201. [PMID: 39261577 PMCID: PMC11390985 DOI: 10.1038/s41598-024-72324-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 09/05/2024] [Indexed: 09/13/2024] Open
Abstract
Myocardial ischemia-reperfusion injury (MIRI) is a significant complication following reperfusion therapy after myocardial infarction. Mitochondrial oxidative stress is a critical factor in MIRI, and Sirtuin 3 (SIRT3), as a major mitochondrial deacetylase, plays a key protective role, with its activity potentially regulated by O-GlcNAcylation. This study used the H9C2 cell line to establish a simulated ischemia/reperfusion (SI/R) model, we utilized co-immunoprecipitated to validate the relationship between O-GlcNAc transferase (OGT) and SIRT3, demonstrated SIRT3 O-GlcNAcylation sites through LC-MS/MS, and performed site mutations using CRISPR/Cas9 technology. The results were validated using immunoblotting. SIRT3 and superoxide dismutase 2 (SOD2) activities were detected using a fluorometric assay, while mitochondrial reactive oxygen species (MROS) levels and cellular apoptosis were assessed using immunofluorescence. We have identified an interaction between SIRT3 and OGT, where SIRT3 undergoes dynamic O-GlcNAcylation at the S190 site, facilitating SIRT3 deacetylase activity. During SI/R, elevated levels of O-GlcNAcylation activate SOD2 by promoting SIRT3 enzyme activity, thereby inhibiting excessive MROS production. This significantly mitigates the occurrence of malignant autophagy in myocardial cells during reperfusion, promoting their survival. Conversely, blocking SIRT3 O-GlcNAcylation at the S190 site exacerbates SI/R injury. We demonstrate that O-GlcNAcylation is a crucial post-translational modification (PTM) of SIRT3 during SI/R, shedding light on a promising mechanism for future therapeutic approaches.
Collapse
Affiliation(s)
- Han Zhou
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yingjie Ji
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jingjie Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Lin Sun
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
2
|
Yu W, Chen J, Jin S, Fan X, Cai X. Identification and Validation of Glycosylation-Related Genes in Obesity and MASH: Insights from Human Liver Samples and a High-Fat Diet Mouse Model. Pharmgenomics Pers Med 2024; 17:363-381. [PMID: 38983907 PMCID: PMC11232960 DOI: 10.2147/pgpm.s463608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/12/2024] [Indexed: 07/11/2024] Open
Abstract
Background Obesity is reaching epidemic proportions in the developed world. The biosynthesis and degradation of human glycoproteins take place at the highest level in the liver. However, the association between glycosylation and the factors affecting obesity and metabolism-associated steatohepatitis (MASH) is still unclear. Materials and Methods Gene expression data of liver samples from obese patients were retrieved from GSE83452 and GSE89632 databases. Difference analysis and machine learning were used to identify hub genes involved in glycosylation and associated with the response of weight loss treatment. A total of 7 glycosylation-related hub genes were identified and then subjected to correlation analysis, immune cells infiltration analysis and ROC (Receiver Operating Characteristic) analysis. We also evaluated the potential function of 7 hub genes in obesity patients. MASH mice were used to validate the glycosylation-related hub genes. Results A total of 25 overlapped glycosylation-related genes were identified by DEGs analysis. ACER2, STX17, ARF5, GPC4, ENTPD5, NANP, and DPY19L2 were identified as hub genes. Among these hub genes, ACER2, STX17, ARF5, and ENTPD5 were also differential expressed in MASH patients. ENTPD5 showed increased transcription in obese MASH mice. Conclusion The current study identified seven glycosylation-related genes, ACER2, STX17, ARF5, GPC4, ENTPD5, NANP, and DPY19L2, that might play key roles in the development of obesity. ENTPD5 might play a key role in the development of MASH. These findings provide fresh perspectives for expanding the investigation of obesity and MASH.
Collapse
Affiliation(s)
- Weihua Yu
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Jionghuang Chen
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Shengxi Jin
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Xiaoxiao Fan
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Xiujun Cai
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| |
Collapse
|
3
|
Chen S, Guan S, Yan Z, Ouyang F, Li S, Liu L, Zhong J. Role of RIPK3‑CaMKII‑mPTP signaling pathway‑mediated necroptosis in cardiovascular diseases (Review). Int J Mol Med 2023; 52:98. [PMID: 37654208 PMCID: PMC10495754 DOI: 10.3892/ijmm.2023.5301] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 07/27/2023] [Indexed: 09/02/2023] Open
Abstract
Necroptosis, which is distinct from apoptosis and necrosis, serves a crucial role in ontogeny and the maintenance of homeostasis. In the last decade, it has been demonstrated that the pathogenesis of cardiovascular diseases is also linked to necroptosis. Receptor interaction protein kinase (RIPK) 1, RIPK3 and mixed lineage kinase domain‑like protein serve vital roles in necroptosis. In addition to the aforementioned necroptosis‑related components, calcium/calmodulin‑dependent protein kinase II (CaMKII) has been identified as a novel substrate for RIPK3 that promotes the opening of the mitochondrial permeability transition pore (mPTP), and thus, mediates necroptosis of myocardial cells through the RIPK3‑CaMKII‑mPTP signaling pathway. The present review provides an overview of the current knowledge of the RIPK3‑CaMKII‑mPTP‑mediated necroptosis signaling pathway in cardiovascular diseases, focusing on the role of the RIPK3‑CaMKII‑mPTP signaling pathway in acute myocardial infarction, ischemia‑reperfusion injury, heart failure, abdominal aortic aneurysm, atherosclerosis, diabetic cardiomyopathy, hypertrophic cardiomyopathy, atrial fibrillation, and the cardiotoxicity associated with antitumor drugs and other chemicals. Finally, the present review discusses the research status of drugs targeting the RIPK3‑CaMKII‑mPTP signaling pathway.
Collapse
Affiliation(s)
- Sheng Chen
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong 528308, P.R. China
| | - Senhong Guan
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong 528308, P.R. China
| | - Zhaohan Yan
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong 528308, P.R. China
| | - Fengshan Ouyang
- Department of Rehabilitation Medicine, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong 528308, P.R. China
| | - Shuhuan Li
- Department of Pediatrics, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong 528308, P.R. China
| | - Lanyuan Liu
- Department of Ultrasound Medicine, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong 528308, P.R. China
| | - Jiankai Zhong
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong 528308, P.R. China
| |
Collapse
|
4
|
Kanlioz M, Ekici U. A New and Effective Technique in the Endoscopic Treatment of Obesity and Regulation of Diabetes: The Pyloric Revision. Cureus 2023; 15:e43357. [PMID: 37701000 PMCID: PMC10493824 DOI: 10.7759/cureus.43357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2023] [Indexed: 09/14/2023] Open
Abstract
AIM This study aimed to investigate the role of the functional structure of the pylorus in obesity and diabetes and to determine the efficacy of a new method, pyloric revision (PR), in the treatment. METHODS The pyloric structures of the patients who applied for endoscopic obesity treatment were examined, and the patients were classified as normotonic (NP), hypotonic (HP), and atonic (AP) according to their pyloric structures. PR was applied to those with pyloric structural disorders. Patients with NP were also given the preferred endoscopic treatment (balloon, botulinum toxin, Kanlioz technique). In addition, the pre-procedure fasting blood glucose (FBG) and glycated hemoglobin levels (HbA1c) of the patients were compared with the sixth-month post-procedure status. In order to compare the pyloric structure and other parameters in normal weights with the obese group, a second group of 100 normal-weight (BMI<25) individuals was formed and compared with the study group. RESULTS In patients with HP (93 patients) and AP (61 patients), a statistically significant decrease was found between HbA1c and FBG levels before treatment and six months after treatment (p˂0.02, p<0.001, respectively). There was a statistically significant difference in favor of the endoscopic obesity treatment group (EOTG) in terms of pyloric disorder, HbA1c, and FPG levels between the EOTG and the normal weight group (NWG) (p<0.0001). CONCLUSION We recommend using PR as an easy-to-perform, effective, minimally invasive, reproducible, and cost-effective technique that does not require hospitalization.
Collapse
Affiliation(s)
- Murat Kanlioz
- General Surgery, Flora Transplantation Institute, Istanbul, TUR
| | - Ugur Ekici
- General Surgery, Flora Transplantation Institute, İstanbul, TUR
| |
Collapse
|
5
|
Emodin Regulates lncRNA XIST/miR-217 Axis to Protect Myocardial Ischemia-Reperfusion Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:3612814. [PMID: 36760350 PMCID: PMC9904883 DOI: 10.1155/2023/3612814] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/22/2022] [Accepted: 11/24/2022] [Indexed: 02/04/2023]
Abstract
Purpose This study is aimed at investigating the effect of emodin on myocardial ischemia-reperfusion injury (MIRI) and mechanism. Methods Eighty healthy adult male SD rats (weighing 230-250 g) were utilized to establish I/R model, which were randomly divided into five groups (16 rats in each group): sham operation group, myocardial ischemia-reperfusion injury group (I/R group), emodin group, emodin +NC group, and emodin +XIST group. The contents of CK, CK-MB, LDH, and HBDH in serum were determined by ELISA kit. LDH was detected by ELISA assay, SOD was detected by the xanthine oxidase method, and MDA was detected by the thiobarbituric acid method. The relative expression of XIST and miR-217 was evaluated by RT-qPCR. Western blot was applied to detect the protein expression. Flow cytometry was applied to detect cardiomyocyte apoptosis. Results Myocardial infarction area was obviously increased in I/R model rats, while emodin decreased the myocardial infarction in I/R model rats. In addition, cardiac enzymes (CK, CK-MB, LDH, and HBDH) and apoptosis were obviously increased in MIRI model rats, while emodin obviously decreased cardiac enzymes and apoptosis. The ROS and MDA levels were raised while the activities of SOD were declined in the I/R model group. The ROS and MDA levels were decreased while the activities of SOD were raised in the emodin group. The XIST expression was markedly raised in the I/R model group while decreased in the emodin group, and the overexpression of XIST reversed the protective effect of emodin on myocardial infarction, oxidative stress, and cardiomyocyte apoptosis. In addition, XIST directly regulated the expression of miR-217, and si-XIST inhibited H/R-induced oxidative damage of cardiomyocytes via inhibiting miR-217. Conclusion Emodin protected MIRI both in vitro and in vivo via inhibiting lncRNA XIST to upregulate miR-217.
Collapse
|
6
|
Yu YW, Liu S, Zhou YY, Huang KY, Wu BS, Lin ZH, Zhu CX, Xue YJ, Ji KT. Shexiang Baoxin Pill attenuates myocardial ischemia/reperfusion injury by activating autophagy via modulating the ceRNA-Map3k8 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 104:154336. [PMID: 35849969 DOI: 10.1016/j.phymed.2022.154336] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/04/2022] [Accepted: 07/10/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND The pathogenesis of myocardial ischemia/reperfusion is complex, involving multiple regulatory genes and environmental factors, and requiring the simultaneous regulation of multiple targets. Meanwhile, Traditional Chinese Medicine (TCM) has certain advantages in the comprehensive treatment of multi-site, multi-target conditions and overall regulation of this condition. This study explores the effect of the well-known TCM, the Shexiang Baoxin Pill (SBP) on myocardial ischemia/reperfusion injury in mice. MATERIALS AND METHODS In vivo, 20 mg/kg/day SBP was administered by gavage for 28 days. In vitro, cardiomyocytes were pretreated with 25 μg/ml SBP for 24 h. Evans blue/TTC double-staining was employed to determine the infarct size. Markers of myocardial injury were detected in the serum and cell supernatants. The changes of pyroptosis and autophagy proteins were detected by western blot. Immunofluorescence, immunohistochemistry and PCR were performed to further illustrate the results. RESULTS SBP significantly reduced the myocardial infarct size, decreased the myocardial injury markers, inhibited cardiomyocyte pyroptosis and oxidative stress, and promoted autophagy in vivo. In vitro, SBP alleviated cardiomyocyte pyroptosis, inhibited oxidative stress, reduced IL-1β and IL-18 secretion, and unblocked autophagy flux. Myocardial injury is mitigated by SBP via the rapid degradation of autophagosomes, and SBP promotes the accumulation of autophagosomes by downregulating mmu_circ_0005874, Map3k8 and upregulating mmu-miR-543-3p. CONCLUSION We found for the first time that SBP can inhibit pyroptosis and oxidative stress, and protect from myocardial I/R injury. In addition, it inhibits pyroptosis and improves H/R injury by promoting autophagosome generation and accelerating autophagic flux. SBP interferes with autophagy through the interaction between mmu_circ_0005874/mmu-miR-543-3p/Map3k8.
Collapse
Affiliation(s)
- Yong-Wei Yu
- Department of Cardiology, The Second Affliated and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 312500, China; Intensive Care Unit, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Shuai Liu
- Department of Cardiology, The Second Affliated and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 312500, China
| | - Ying-Ying Zhou
- Department of Endocrinology, The Second Affliated and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 312500, China
| | - Kai-Yu Huang
- Department of Cardiology, The Second Affliated and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 312500, China
| | - Bo-Sen Wu
- Department of Cardiology, The Second Affliated and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 312500, China
| | - Zhi-Hui Lin
- Department of Cardiology, The Second Affliated and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 312500, China
| | - Chen-Xi Zhu
- Department of Cardiology, The Second Affliated and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 312500, China
| | - Yang-Jing Xue
- Department of Cardiology, The Second Affliated and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 312500, China.
| | - Kang-Ting Ji
- Department of Cardiology, The Second Affliated and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 312500, China.
| |
Collapse
|
7
|
Alkouri A, Cybularz M, Mierke J, Nowack T, Biedermann J, Ulbrich S, Fischer J, Heidrich FM, Jellinghaus S, Speiser U, Linke A, Pfluecke C. The predictive role of early CRP values for one-year mortality in the first two days after acute myocardial infarction. Biomarkers 2022; 27:293-298. [PMID: 35138209 DOI: 10.1080/1354750x.2022.2040591] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND An excessive inflammatory reaction after acute myocardial infarction (AMI) is known to be harmful. New anti-inflammatory therapies are required. PURPOSE This study assessed the predictive role of early CRP in patients with STEMI. METHODS 1003 patients with STEMI were analyzed. 180 patients with proven infection were excluded. CRP after 12 h, 24 h, and 48 h after pain onset were evaluated. RESULTS Of 823 patients, 103 (12.5%) died within one year after AMI. The deceased patients showed higher CRP, even after already 12 h (6 vs. 13 mg/l, p < 0.001), 24 h (13 vs. 25 mg/l, p < 0.001) and after 48 h (40 vs. 92 mg/l, p < 0.001). A CRP of ≥8 mg/l, 12 h after AMI, was found in 45% and was independently associated with long-term mortality (OR: 2.7, p = 0.03), after 24 h: CRP ≥18 mg/l in 44% (OR: 2.5, p = 0.03), after 48 h: CRP ≥53 mg/l in 44% (OR 1.9, p = 0.03). Early CRP values correlated strongly with the later maximum value of CRP (p < 0.001). CONCLUSIONS Already early CRP values are accurate for risk-prediction following AMI. By identifying patients who are beginning to develop an excessive inflammatory response, it may be possible to identify those who benefit from anti-inflammatory therapies.
Collapse
Affiliation(s)
- Ahmad Alkouri
- Department for Internal Medicine and Cardiology, Herzzentrum Dresden, University Clinic, Technische Universität Dresden.,Department for Internal Medicine and Cardiology, Städtisches Klinikum Görlitz, Germany
| | - Maria Cybularz
- Department for Internal Medicine and Cardiology, Herzzentrum Dresden, University Clinic, Technische Universität Dresden
| | - Johannes Mierke
- Department for Internal Medicine and Cardiology, Herzzentrum Dresden, University Clinic, Technische Universität Dresden
| | - Thomas Nowack
- Department for Internal Medicine and Cardiology, Herzzentrum Dresden, University Clinic, Technische Universität Dresden
| | - Jonathan Biedermann
- Department for Internal Medicine and Cardiology, Herzzentrum Dresden, University Clinic, Technische Universität Dresden
| | - Stefan Ulbrich
- Department for Internal Medicine and Cardiology, Herzzentrum Dresden, University Clinic, Technische Universität Dresden
| | - Julia Fischer
- Department for Internal Medicine and Cardiology, Herzzentrum Dresden, University Clinic, Technische Universität Dresden
| | - Felix M Heidrich
- Department for Internal Medicine and Cardiology, Herzzentrum Dresden, University Clinic, Technische Universität Dresden
| | - Stefanie Jellinghaus
- Department for Internal Medicine and Cardiology, Herzzentrum Dresden, University Clinic, Technische Universität Dresden
| | - Uwe Speiser
- Department for Internal Medicine and Cardiology, Herzzentrum Dresden, University Clinic, Technische Universität Dresden
| | - Axel Linke
- Department for Internal Medicine and Cardiology, Herzzentrum Dresden, University Clinic, Technische Universität Dresden
| | - Christian Pfluecke
- Department for Internal Medicine and Cardiology, Herzzentrum Dresden, University Clinic, Technische Universität Dresden.,Department for Internal Medicine and Cardiology, Städtisches Klinikum Görlitz, Germany
| |
Collapse
|
8
|
Ma LL, Kong FJ, Dong Z, Xin KY, Wang XX, Sun AJ, Zou YZ, Ge JB. Hypertrophic Preconditioning Attenuates Myocardial Ischaemia-Reperfusion Injury by Modulating SIRT3-SOD2-mROS-Dependent Autophagy. Cell Prolif 2021; 54:e13051. [PMID: 33973685 PMCID: PMC8249780 DOI: 10.1111/cpr.13051] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/01/2021] [Accepted: 04/13/2021] [Indexed: 12/19/2022] Open
Abstract
Background Ischaemic preconditioning elicited by brief periods of coronary occlusion and reperfusion protects the heart from a subsequent prolonged ischaemic insult. Here, we test the hypothesis that short‐term non‐ischaemic stimulation of hypertrophy renders the heart resistant to subsequent ischaemic injury. Methods and Results Transient transverse aortic constriction (TAC) was performed for 3 days in mice and then withdrawn for 4 days by aortic debanding, followed by subsequent exposure to myocardial ischaemia‐reperfusion (I/R) injury. Following I/R injury, myocardial infarct size and apoptosis were significantly decreased, and cardiac dysfunction was markedly improved in the TAC preconditioning group compared with the control group. Mechanistically, TAC preconditioning markedly suppressed I/R‐induced autophagy and preserved autophagic flux by deacetylating SOD2 via a SIRT3‐dependent mechanism. Moreover, treatment with an adenovirus encoding SIRT3 partially mimicked the effects of hypertrophic preconditioning, whereas genetic ablation of SIRT3 in mice blocked the cardioprotective effects of hypertrophic preconditioning. Furthermore, in vivo lentiviral‐mediated knockdown of Beclin 1 in the myocardium ameliorated the I/R‐induced impairment of autophagic flux and was associated with a reduction in cell death, whereas treatment with a lentivirus encoding Beclin 1 abolished the cardioprotective effect of TAC preconditioning. Conclusions The present study identifies TAC preconditioning as a novel strategy for induction of an endogenous self‐defensive and cardioprotective mechanism against cardiac injury. Specifically, TAC preconditioning reduced myocardial autophagic cell death in a SIRT3/SOD2 pathway‐dependent manner.
Collapse
Affiliation(s)
- Lei-Lei Ma
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Viral Heart Diseases, Shanghai, China.,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Fei-Juan Kong
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Dong
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Viral Heart Diseases, Shanghai, China.,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Kai-Yue Xin
- Department of Cardiology, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xing-Xu Wang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Viral Heart Diseases, Shanghai, China.,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Ai-Jun Sun
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Viral Heart Diseases, Shanghai, China.,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Yun-Zeng Zou
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Viral Heart Diseases, Shanghai, China.,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Jun-Bo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Viral Heart Diseases, Shanghai, China.,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| |
Collapse
|
9
|
Kompa AR, Greening DW, Kong AM, McMillan PJ, Fang H, Saxena R, Wong RCB, Lees JG, Sivakumaran P, Newcomb AE, Tannous BA, Kos C, Mariana L, Loudovaris T, Hausenloy DJ, Lim SY. Sustained subcutaneous delivery of secretome of human cardiac stem cells promotes cardiac repair following myocardial infarction. Cardiovasc Res 2021; 117:918-929. [PMID: 32251516 PMCID: PMC7898942 DOI: 10.1093/cvr/cvaa088] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/13/2020] [Accepted: 03/31/2020] [Indexed: 12/12/2022] Open
Abstract
AIMS To establish pre-clinical proof of concept that sustained subcutaneous delivery of the secretome of human cardiac stem cells (CSCs) can be achieved in vivo to produce significant cardioreparative outcomes in the setting of myocardial infarction. METHODS AND RESULTS Rats were subjected to permanent ligation of left anterior descending coronary artery and randomized to receive subcutaneous implantation of TheraCyte devices containing either culture media as control or 1 × 106 human W8B2+ CSCs, immediately following myocardial ischaemia. At 4 weeks following myocardial infarction, rats treated with W8B2+ CSCs encapsulated within the TheraCyte device showed preserved left ventricular ejection fraction. The preservation of cardiac function was accompanied by reduced fibrotic scar tissue, interstitial fibrosis, cardiomyocyte hypertrophy, as well as increased myocardial vascular density. Histological analysis of the TheraCyte devices harvested at 4 weeks post-implantation demonstrated survival of human W8B2+ CSCs within the devices, and the outer membrane was highly vascularized by host blood vessels. Using CSCs expressing plasma membrane reporters, extracellular vesicles of W8B2+ CSCs were found to be transferred to the heart and other organs at 4 weeks post-implantation. Furthermore, mass spectrometry-based proteomic profiling of extracellular vesicles of W8B2+ CSCs identified proteins implicated in inflammation, immunoregulation, cell survival, angiogenesis, as well as tissue remodelling and fibrosis that could mediate the cardioreparative effects of secretome of human W8B2+ CSCs. CONCLUSIONS Subcutaneous implantation of TheraCyte devices encapsulating human W8B2+ CSCs attenuated adverse cardiac remodelling and preserved cardiac function following myocardial infarction. The TheraCyte device can be employed to deliver stem cells in a minimally invasive manner for effective secretome-based cardiac therapy.
Collapse
Affiliation(s)
- Andrew R Kompa
- Departments of Medicine and Surgery, University of Melbourne,
Melbourne, VIC, Australia
- Department of Epidemiology and Preventive Medicine, Centre of Cardiovascular
Research and Education in Therapeutics, Monash University, Melbourne, VIC,
Australia
| | - David W Greening
- Molecular Proteomics, Baker Heart and Diabetes Institute,
Melbourne, VIC, Australia
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular
Science, La Trobe University, Melbourne, VIC, Australia
| | - Anne M Kong
- O’Brien Institute Department, St Vincent’s Institute of Medical
Research, 9 Princes Street, Fitzroy, VIC 3065, Australia
| | - Paul J McMillan
- Department of Biochemistry and Molecular Biology, Biological Optical Microscopy
Platform, University of Melbourne, Melbourne, VIC, Australia
| | - Haoyun Fang
- Molecular Proteomics, Baker Heart and Diabetes Institute,
Melbourne, VIC, Australia
| | - Ritika Saxena
- O’Brien Institute Department, St Vincent’s Institute of Medical
Research, 9 Princes Street, Fitzroy, VIC 3065, Australia
- School of Life and Environmental Sciences, Faculty of Science, Deakin
University, Burwood, VIC, Australia
| | - Raymond C B Wong
- Departments of Medicine and Surgery, University of Melbourne,
Melbourne, VIC, Australia
- Cellular Reprogramming Unit, Centre for Eye Research Australia, Royal Victorian
Eye and Ear Hospital, East Melbourne, VIC, Australia
- Shenzhen Eye Hospital, Shenzhen University School of Medicine,
Shenzhen, China
| | - Jarmon G Lees
- Departments of Medicine and Surgery, University of Melbourne,
Melbourne, VIC, Australia
- O’Brien Institute Department, St Vincent’s Institute of Medical
Research, 9 Princes Street, Fitzroy, VIC 3065, Australia
| | - Priyadharshini Sivakumaran
- O’Brien Institute Department, St Vincent’s Institute of Medical
Research, 9 Princes Street, Fitzroy, VIC 3065, Australia
| | - Andrew E Newcomb
- Department of Cardiothoracic Surgery, St Vincent’s Hospital
Melbourne, Melbourne, VIC, Australia
| | - Bakhos A Tannous
- Department of Neurology and Pathology, Massachusetts General
Hospital, Charlestown, MA, USA
- Program in Neuroscience, Harvard Medical School, Boston, MA,
USA
| | - Cameron Kos
- O'Brien Institute Department & Immunology & Diabetes Unit, St Vincent’s
Institute of Medical Research, VIC, Australia
| | - Lina Mariana
- O'Brien Institute Department & Immunology & Diabetes Unit, St Vincent’s
Institute of Medical Research, VIC, Australia
| | - Thomas Loudovaris
- O'Brien Institute Department & Immunology & Diabetes Unit, St Vincent’s
Institute of Medical Research, VIC, Australia
| | - Derek J Hausenloy
- Cardiovascular and Metabolic Disorders Program, Duke-National University of
Singapore Medical School, Singapore, Singapore
- National Heart Research Institute Singapore, National Heart
Centre, Singapore, Singapore
- The Hatter Cardiovascular Institute, University College London,
London, UK
- Cardiovascular Research Center, College of Medical and Health Sciences, Asia
University, Taichung, Taiwan
- Yong Loo Lin School of Medicine, National University Singapore,
Singapore, Singapore
| | - Shiang Y Lim
- Departments of Medicine and Surgery, University of Melbourne,
Melbourne, VIC, Australia
- O’Brien Institute Department, St Vincent’s Institute of Medical
Research, 9 Princes Street, Fitzroy, VIC 3065, Australia
| |
Collapse
|
10
|
Mayoral LPC, Andrade GM, Mayoral EPC, Huerta TH, Canseco SP, Rodal Canales FJ, Cabrera-Fuentes HA, Cruz MM, Pérez Santiago AD, Alpuche JJ, Zenteno E, Ruíz HM, Cruz RM, Jeronimo JH, Perez-Campos E. Obesity subtypes, related biomarkers & heterogeneity. Indian J Med Res 2021; 151:11-21. [PMID: 32134010 PMCID: PMC7055173 DOI: 10.4103/ijmr.ijmr_1768_17] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Obesity is a serious medical condition worldwide, which needs new approaches and recognized international consensus in treating diseases leading to morbidity. The aim of this review was to examine heterogeneous links among the various phenotypes of obesity in adults. Proteins and associated genes in each group were analysed to differentiate between biomarkers. A variety of terms for classification and characterization within this pathology are currently in use; however, there is no clear consensus in terminology. The most significant groups reviewed include metabolically healthy obese, metabolically abnormal obese, metabolically abnormal, normal weight and sarcopenic obese. These phenotypes do not define particular genotypes or epigenetic gene regulation, or proteins related to inflammation. There are many other genes linked to obesity, though the value of screening all of those for diagnosis has low predictive results, as there are no significant biomarkers. It is important to establish a consensus in the terminology used and the characteristics attributed to obesity subtypes. The identification of specific molecular biomarkers is also required for better diagnosis in subtypes of obesity.
Collapse
Affiliation(s)
- Laura Perez-Campos Mayoral
- Research Centre-Faculty of Medicine, National Autonomous University of Mexico-Benito Juárez Autonomous University of Oaxaca, Oaxaca, Mexico
| | - Gabriel Mayoral Andrade
- Research Centre-Faculty of Medicine, National Autonomous University of Mexico-Benito Juárez Autonomous University of Oaxaca, Oaxaca, Mexico
| | - Eduardo Perez-Campos Mayoral
- Research Centre-Faculty of Medicine, National Autonomous University of Mexico-Benito Juárez Autonomous University of Oaxaca, Oaxaca, Mexico
| | | | - Socorro Pina Canseco
- Research Centre-Faculty of Medicine, National Autonomous University of Mexico-Benito Juárez Autonomous University of Oaxaca, Oaxaca, Mexico
| | - Francisco J Rodal Canales
- Research Centre-Faculty of Medicine, National Autonomous University of Mexico-Benito Juárez Autonomous University of Oaxaca, Oaxaca, Mexico
| | - Héctor Alejandro Cabrera-Fuentes
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, Singapore; Institute of Biochemistry, Medical School, Justus-Liebig University, Giessen, Germany
| | | | | | - Juan José Alpuche
- Research Centre-Faculty of Medicine, National Autonomous University of Mexico-Benito Juárez Autonomous University of Oaxaca, Oaxaca, Mexico
| | - Edgar Zenteno
- Research Centre-Faculty of Medicine, National Autonomous University of Mexico-Benito Juárez Autonomous University of Oaxaca, Oaxaca, Mexico
| | - Hector Martínez Ruíz
- Research Centre-Faculty of Medicine, National Autonomous University of Mexico-Benito Juárez Autonomous University of Oaxaca, Oaxaca, Mexico
| | - Ruth Martínez Cruz
- Research Centre-Faculty of Medicine, National Autonomous University of Mexico-Benito Juárez Autonomous University of Oaxaca, Oaxaca, Mexico
| | - Julia Hernandez Jeronimo
- Research Centre-Faculty of Medicine, National Autonomous University of Mexico-Benito Juárez Autonomous University of Oaxaca, Oaxaca, Mexico
| | - Eduardo Perez-Campos
- National Technological Institute of Mexico, ITOaxaca; Clinical Pathology Laboratory 'Dr. Eduardo Pérez Ortega' Oaxaca, Mexico
| |
Collapse
|
11
|
Szabó MR, Pipicz M, Csont T, Csonka C. Modulatory Effect of Myokines on Reactive Oxygen Species in Ischemia/Reperfusion. Int J Mol Sci 2020; 21:ijms21249382. [PMID: 33317180 PMCID: PMC7763329 DOI: 10.3390/ijms21249382] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/06/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022] Open
Abstract
There is a growing body of evidence showing the importance of physical activity against acute ischemic events in various organs. Ischemia/reperfusion injury (I/R) is characterized by tissue damage as a result of restriction and subsequent restoration of blood supply to an organ. Oxidative stress due to increased reactive oxygen species formation and/or insufficient antioxidant defense is considered to play an important role in I/R. Physical activity not only decreases the general risk factors for ischemia but also confers direct anti-ischemic protection via myokine production. Myokines are skeletal muscle-derived cytokines, representing multifunctional communication channels between the contracting skeletal muscle and other organs through an endocrine manner. In this review, we discuss the most prominent members of the myokines (i.e., brain-derived neurotrophic factor (BDNF), cathepsin B, decorin, fibroblast growth factors-2 and -21, follistatin, follistatin-like, insulin-like growth factor-1; interleukin-6, interleukin-7, interleukin-15, irisin, leukemia inhibitory factor, meteorin-like, myonectin, musclin, myostatin, and osteoglycin) with a particular interest in their potential influence on reactive oxygen and nitrogen species formation or antioxidant capacity. A better understanding of the mechanism of action of myokines and particularly their participation in the regulation of oxidative stress may widen their possible therapeutic use and, thereby, may support the fight against I/R.
Collapse
Affiliation(s)
- Márton Richárd Szabó
- Metabolic Diseases and Cell Signaling (MEDICS) Research Group, Department of Biochemistry, University of Szeged, Dóm tér 9, 6720 Szeged, Hungary; (M.R.S.); (M.P.); (T.C.)
- Interdisciplinary Centre of Excellence, University of Szeged, Dugonics tér 13, 6720 Szeged, Hungary
| | - Márton Pipicz
- Metabolic Diseases and Cell Signaling (MEDICS) Research Group, Department of Biochemistry, University of Szeged, Dóm tér 9, 6720 Szeged, Hungary; (M.R.S.); (M.P.); (T.C.)
- Interdisciplinary Centre of Excellence, University of Szeged, Dugonics tér 13, 6720 Szeged, Hungary
| | - Tamás Csont
- Metabolic Diseases and Cell Signaling (MEDICS) Research Group, Department of Biochemistry, University of Szeged, Dóm tér 9, 6720 Szeged, Hungary; (M.R.S.); (M.P.); (T.C.)
- Interdisciplinary Centre of Excellence, University of Szeged, Dugonics tér 13, 6720 Szeged, Hungary
| | - Csaba Csonka
- Metabolic Diseases and Cell Signaling (MEDICS) Research Group, Department of Biochemistry, University of Szeged, Dóm tér 9, 6720 Szeged, Hungary; (M.R.S.); (M.P.); (T.C.)
- Interdisciplinary Centre of Excellence, University of Szeged, Dugonics tér 13, 6720 Szeged, Hungary
- Department of Sports Medicine, University of Szeged, Tisza Lajos krt 107, 6725 Szeged, Hungary
- Correspondence: ; Tel.: +36-30-5432-693
| |
Collapse
|
12
|
A Potential Involvement of Anandamide in the Modulation of HO/NOS Systems: Women, Menopause, and "Medical Cannabinoids". Int J Mol Sci 2020; 21:ijms21228801. [PMID: 33233803 PMCID: PMC7699915 DOI: 10.3390/ijms21228801] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/16/2020] [Accepted: 11/19/2020] [Indexed: 12/27/2022] Open
Abstract
Endocannabinoids and their receptors are present in the cardiovascular system; however, their actions under different pathological conditions remain controversial. The aim of our study was to examine the effects of anandamide (AEA) on heme oxygenase (HO) and nitric oxide synthase (NOS) systems in an estrogen-depleted rat model. Sham-operated (SO) and surgically induced estrogen-deficient (OVX) female Wistar rats were used. During a two-week period, a group of OVX rats received 0.1 mg/kg estrogen (E2) per os, while AEA-induced alterations were analyzed after two weeks of AEA treatment at the dose of 1.0 mg/kg. At the end of the experiment, cardiac activity and expression of HO and NOS enzymes, content of cannabinoid 1 receptor, as well as concentrations of transient potential vanilloid 1 (TRPV1) and calcitonin gene-related peptide (CGRP) were measured. Our results show that estrogen withdrawal caused a significant decrease in both NOS and HO systems, and a similar tendency was observed regarding the TRPV1/CGRP pathway. Two weeks of either AEA or E2 treatment restored the adverse changes; however, the combined administration of these two molecules did not result in a further improvement. In light of the potential relationship between AEA and HO/NOS systems, AEA-induced upregulation of HO/NOS enzymes may be a therapeutic strategy in estrogen-deficient conditions.
Collapse
|
13
|
Castaño D, Rattanasopa C, Monteiro-Cardoso VF, Corlianò M, Liu Y, Zhong S, Rusu M, Liehn EA, Singaraja RR. Lipid efflux mechanisms, relation to disease and potential therapeutic aspects. Adv Drug Deliv Rev 2020; 159:54-93. [PMID: 32423566 DOI: 10.1016/j.addr.2020.04.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 02/06/2023]
Abstract
Lipids are hydrophobic and amphiphilic molecules involved in diverse functions such as membrane structure, energy metabolism, immunity, and signaling. However, altered intra-cellular lipid levels or composition can lead to metabolic and inflammatory dysfunction, as well as lipotoxicity. Thus, intra-cellular lipid homeostasis is tightly regulated by multiple mechanisms. Since most peripheral cells do not catabolize cholesterol, efflux (extra-cellular transport) of cholesterol is vital for lipid homeostasis. Defective efflux contributes to atherosclerotic plaque development, impaired β-cell insulin secretion, and neuropathology. Of these, defective lipid efflux in macrophages in the arterial walls leading to foam cell and atherosclerotic plaque formation has been the most well studied, likely because a leading global cause of death is cardiovascular disease. Circulating high density lipoprotein particles play critical roles as acceptors of effluxed cellular lipids, suggesting their importance in disease etiology. We review here mechanisms and pathways that modulate lipid efflux, the role of lipid efflux in disease etiology, and therapeutic options aimed at modulating this critical process.
Collapse
|
14
|
Tuscany Sangiovese grape juice imparts cardioprotection by regulating gene expression of cardioprotective C-type natriuretic peptide. Eur J Nutr 2019; 59:2953-2968. [PMID: 31707544 DOI: 10.1007/s00394-019-02134-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 10/29/2019] [Indexed: 01/02/2023]
Abstract
PURPOSE A regular intake of red grape juice has cardioprotective properties, but its role on the modulation of natriuretic peptides (NPs), in particular of C-type NP (CNP), has not yet been proven. The aims were to evaluate: (1) in vivo the effects of long-term intake of Tuscany Sangiovese grape juice (SGJ) on the NPs system in a mouse model of myocardial infarction (MI); (2) in vitro the response to SGJ small RNAs of murine MCEC-1 under physiological and ischemic condition; (3) the activation of CNP/NPR-B/NPR-C in healthy human subjects after 7 days' SGJ regular intake. METHODS (1) C57BL/6J male and female mice (n = 33) were randomly subdivided into: SHAM (n = 7), MI (n = 15) and MI fed for 4 weeks with a normal chow supplemented with Tuscany SGJ (25% vol/vol, 200 µl/per day) (MI + SGJ, n = 11). Echocardiography and histological analyses were performed. Myocardial NPs transcriptional profile was investigated by Real-Time PCR. (2) MCEC-1 were treated for 24 h with a pool of SGJ small RNAs and cell viability under 24 h exposure to H2O2 was evaluated by MTT assay. (3) Human blood samples were collected from seven subjects before and after the 7 days' intake of Tuscany SGJ. NPs and miRNA transcriptional profile were investigated by Real-Time PCR in MCEC-1 and human blood. RESULTS Our experimental data, obtained in a multimodal pipeline, suggest that the long-term intake of SGJ promotes an adaptive response of the myocardium to the ischemic microenvironment through the modulation of the cardiac CNP/NPR-B/NPR-C system. CONCLUSIONS Our results open new avenue in the development of functional foods aimed at enhancing cardioprotection of infarcted hearts through action on the myocardial epigenome.
Collapse
|
15
|
Two pharmacological epoxyeicosatrienoic acid-enhancing therapies are effectively antihypertensive and reduce the severity of ischemic arrhythmias in rats with angiotensin II-dependent hypertension. J Hypertens 2019; 36:1326-1341. [PMID: 29570510 DOI: 10.1097/hjh.0000000000001708] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVE We examined the effects of treatment with soluble epoxide hydrolase inhibitor (sEHi) and epoxyeicosatrienoic acids (EETs) analogue (EET-A), given alone or combined, on blood pressure (BP) and ischemia/reperfusion myocardial injury in rats with angiotensin II (ANG II)-dependent hypertension. METHODS Ren-2 transgenic rats (TGR) were used as a model of ANG II-dependent hypertension and Hannover Sprague-Dawley rats served as controls. Rats were treated for 14 days with sEHi or EET-A and BP was measured by radiotelemetry. Albuminuria, cardiac hypertrophy and concentrations of ANG II and EETs were determined. Separate groups were subjected to acute myocardial ischemia/reperfusion injury and the infarct size and ventricular arrhythmias were determined. RESULTS Treatment of TGR with sEHi and EET-A, given alone or combined, decreased BP to a similar degree, reduced albuminuria and cardiac hypertrophy to similar extent; only treatment regimens including sEHi increased myocardial and renal tissue concentrations of EETs. sEHi and EET-A, given alone or combined, suppressed kidney ANG II levels in TGR. Remarkably, infarct size did not significantly differ between TGR and Hannover Sprague-Dawley rats, but the incidence of ischemia-induced ventricular fibrillations was higher in TGR. Application of sEHi and EET-A given alone and combined sEHi and EET-A treatment were all equally effective in reducing life-threatening ventricular fibrillation in TGR. CONCLUSION The findings indicate that chronic treatment with either sEHi or EET-A exerts distinct antihypertensive and antiarrhythmic actions in our ANG II-dependent model of hypertension whereas combined administration of sEHi and EET-A does not provide additive antihypertensive or cardioprotective effects.
Collapse
|
16
|
Ong SB, Kwek XY, Katwadi K, Hernandez-Resendiz S, Crespo-Avilan GE, Ismail NI, Lin YH, Yap EP, Lim SY, Ja KPMM, Ramachandra CJA, Tee N, Toh JJ, Shim W, Wong P, Cabrera-Fuentes HA, Hausenloy DJ. Targeting Mitochondrial Fission Using Mdivi-1 in A Clinically Relevant Large Animal Model of Acute Myocardial Infarction: A Pilot Study. Int J Mol Sci 2019; 20:E3972. [PMID: 31443187 PMCID: PMC6720595 DOI: 10.3390/ijms20163972] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/09/2019] [Accepted: 08/14/2019] [Indexed: 12/19/2022] Open
Abstract
Background: New treatments are needed to reduce myocardial infarct size (MI) and prevent heart failure (HF) following acute myocardial infarction (AMI), which are the leading causes of death and disability worldwide. Studies in rodent AMI models showed that genetic and pharmacological inhibition of mitochondrial fission, induced by acute ischemia and reperfusion, reduced MI size. Whether targeting mitochondrial fission at the onset of reperfusion is also cardioprotective in a clinically-relevant large animal AMI model remains to be determined. Methods: Adult pigs (30-40 kg) were subjected to closed-chest 90-min left anterior descending artery ischemia followed by 72 h of reperfusion and were randomized to receive an intracoronary bolus of either mdivi-1 (1.2 mg/kg, a small molecule inhibitor of the mitochondrial fission protein, Drp1) or vehicle control, 10-min prior to reperfusion. The left ventricular (LV) size and function were both assessed by transthoracic echocardiography prior to AMI and after 72 h of reperfusion. MI size and the area-at-risk (AAR) were determined using dual staining with Tetrazolium and Evans blue. Heart samples were collected for histological determination of fibrosis and for electron microscopic analysis of mitochondrial morphology. Results: A total of 14 pigs underwent the treatment protocols (eight control and six mdivi-1). Administration of mdivi-1 immediately prior to the onset of reperfusion did not reduce MI size (MI size as % of AAR: Control 49.2 ± 8.6 vs. mdivi-1 50.5 ± 11.4; p = 0.815) or preserve LV systolic function (LV ejection fraction %: Control 67.5 ± 0.4 vs. mdivi-1 59.6 ± 0.6; p = 0.420), when compared to vehicle control. Similarly, there were no differences in mitochondrial morphology or myocardial fibrosis between mdivi-1 and vehicle control groups. Conclusion: Our pilot study has shown that treatment with mdivi-1 (1.2 mg/kg) at the onset of reperfusion did not reduce MI size or preserve LV function in the clinically-relevant closed-chest pig AMI model. A larger study, testing different doses of mdivi-1 or using a more specific Drp1 inhibitor are required to confirm these findings.
Collapse
Affiliation(s)
- Sang-Bing Ong
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore.
- Department of Cardiovascular, Renal and Metabolic Medicine, School of Medicine, Sapporo Medical University, Hokkaido 060-8543, Japan.
| | - Xiu-Yi Kwek
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore
| | - Khairunnisa Katwadi
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore
| | - Sauri Hernandez-Resendiz
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore
| | - Gustavo E Crespo-Avilan
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore
- Institute of Biochemistry, Medical School, Justus-Liebig University, 35392 Giessen, Germany
| | - Nur Izzah Ismail
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
| | - Ying-Hsi Lin
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore
| | - En Ping Yap
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore
| | - Song-Yi Lim
- Innoheart Pte Ltd., Singapore 119844, Singapore
| | - K P Myu Mai Ja
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore
| | - Chrishan J A Ramachandra
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore
| | - Nicole Tee
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore
| | | | - Winston Shim
- Innoheart Pte Ltd., Singapore 119844, Singapore
- Health and Social Sciences Cluster, Singapore Institute of Technology, Singapore 138683, Singapore
| | - Philip Wong
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore
- Innoheart Pte Ltd., Singapore 119844, Singapore
| | - Hector A Cabrera-Fuentes
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore.
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore.
- Institute of Biochemistry, Medical School, Justus-Liebig University, 35392 Giessen, Germany.
- Tecnologico de Monterrey, Centro de Biotecnologia-FEMSA, Monterrey, NL 64849, Mexico.
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russian.
| | - Derek J Hausenloy
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore
- Tecnologico de Monterrey, Centro de Biotecnologia-FEMSA, Monterrey, NL 64849, Mexico
- Yong Loo Lin School of Medicine, National University Singapore, Singapore 119228, Singapore
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, London WC1E 6HX, UK
- The National Institute of Health Research University College London Hospitals Biomedical Research Centre, London W1T 7DN, UK
| |
Collapse
|
17
|
He JT, Li H, Yang L, Cheng KL. Involvement of Endothelin-1, H 2S and Nrf2 in Beneficial Effects of Remote Ischemic Preconditioning in Global Cerebral Ischemia-Induced Vascular Dementia in Mice. Cell Mol Neurobiol 2019; 39:671-686. [PMID: 31025223 DOI: 10.1007/s10571-019-00670-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 03/12/2019] [Indexed: 12/11/2022]
Abstract
The present study explored the role of endothelin-1, H2S, and Nrf2 in remote preconditioning (RIPC)-induced beneficial effects in ischemia-reperfusion (I/R)-induced vascular dementia. Mice were subjected to 20 min of global ischemia by occluding both carotid arteries to develop vascular dementia, which was assessed using Morris water maze test on 7th day. RIPC was given by subjecting hind limb to four cycles of ischemia (5 min) and reperfusion (5 min) and it significantly restored I/R-induced locomotor impairment, neurological severity score, cerebral infarction, apoptosis markers along with deficits in learning and memory. Biochemically, there was increase in the plasma levels of endothelin-1 along with increase in the brain levels of H2S and its biosynthetic enzymes viz., cystathionine-β-synthase (CBS) and cystathionine-γ-lyase (CLS). There was also an increase in the expression of Nrf2 and glutathione reductase in the brain in response to RIPC. Pretreatment with bosentan (dual blocker of ETA and ETB receptors), amino-oxyacetic acid (CBS synthase inhibitor), and DL-propargylglycine (CLS inhibitor) significantly attenuated RIPC-mediated beneficial effects and biochemical alterations. The effects of bosentan on behavioral and biochemical parameters were more significant than individual treatments with CBS or CLS inhibitors. Moreover, CBS and CLS inhibitors did not alter the endothelin-1 levels possibly suggesting that endothelin-1 may act as upstream mediator of H2S. It is concluded that RIPC may stimulate the release endothelin-1, which may activate CBS and CLS to increase the levels of H2S and latter may increase the expression of Nrf2 to decrease oxidative stress and prevent vascular dementia.
Collapse
Affiliation(s)
- Jin-Ting He
- Department of Neurology, China-Japan Union Hospital, Jilin University, Changchun, 130033, Jilin, China
| | - Haiqi Li
- Department of Neurology, China-Japan Union Hospital, Jilin University, Changchun, 130033, Jilin, China
| | - Le Yang
- Department of Endocrinology, The People's Hospital of Jilin Province, Changchun, 130031, China.
| | - Kai-Liang Cheng
- Department of Radiology, China-Japan Union Hospital, Jilin University, 126 Xiantai Street, Changchun, 130033, Jilin, China.
| |
Collapse
|
18
|
Yap J, Cabrera-Fuentes HA, Irei J, Hausenloy DJ, Boisvert WA. Role of Macrophages in Cardioprotection. Int J Mol Sci 2019; 20:E2474. [PMID: 31109146 PMCID: PMC6566352 DOI: 10.3390/ijms20102474] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 05/14/2019] [Accepted: 05/17/2019] [Indexed: 12/21/2022] Open
Abstract
Cardiovascular diseases are the leading cause of mortality worldwide. It is widely known that non-resolving inflammation results in atherosclerotic conditions, which are responsible for a host of downstream pathologies including thrombosis, myocardial infarction (MI), and neurovascular events. Macrophages, as part of the innate immune response, are among the most important cell types in every stage of atherosclerosis. In this review we discuss the principles governing macrophage function in the healthy and infarcted heart. More specifically, how cardiac macrophages participate in myocardial infarction as well as cardiac repair and remodeling. The intricate balance between phenotypically heterogeneous populations of macrophages in the heart have profound and highly orchestrated effects during different phases of myocardial infarction. In the early "inflammatory" stage of MI, resident cardiac macrophages are replaced by classically activated macrophages derived from the bone marrow and spleen. And while the macrophage population shifts towards an alternatively activated phenotype, the inflammatory response subsides giving way to the "reparative/proliferative" phase. Lastly, we describe the therapeutic potential of cardiac macrophages in the context of cell-mediated cardio-protection. Promising results demonstrate innovative concepts; one employing a subset of yolk sac-derived, cardiac macrophages that have complete restorative capacity in the injured myocardium of neonatal mice, and in another example, post-conditioning of cardiac macrophages with cardiosphere-derived cells significantly improved patient's post-MI diagnoses.
Collapse
Affiliation(s)
- Jonathan Yap
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA.
| | - Hector A Cabrera-Fuentes
- Tecnologico de Monterrey, Centro de Biotecnologia-FEMSA, Monterrey, NL 264610, Mexico.
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore.
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore.
- Institute of Biochemistry, Medical School, Justus-Liebig University, 35392 Giessen, Germany.
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia.
| | - Jason Irei
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA.
| | - Derek J Hausenloy
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore.
- Institute of Biochemistry, Medical School, Justus-Liebig University, 35392 Giessen, Germany.
- Yong Loo Lin School of Medicine, National University Singapore, Singapore 117597, Singapore.
- The Hatter Cardiovascular Institute, University College London, London WC1E 6HX, UK.
- The National Institute of Health Research University College London Hospitals Biomedical Research Centre, Research & Development, London W1T 7DN, UK.
| | - William A Boisvert
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA.
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia.
| |
Collapse
|
19
|
Paez DT, Garces M, Calabró V, Bin EP, D'Annunzio V, Del Mauro J, Marchini T, Höcht C, Evelson P, Gelpi RJ, Donato M. Adenosine A 1 receptors and mitochondria: targets of remote ischemic preconditioning. Am J Physiol Heart Circ Physiol 2019; 316:H743-H750. [PMID: 30681368 DOI: 10.1152/ajpheart.00071.2018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Adenosine is involved in classic preconditioning in most species and acts especially through adenosine A1 and A3 receptors. The aim of the present study was to evaluate whether remote ischemic preconditioning (rIPC) activates adenosine A1 receptors and improves mitochondrial function, thereby reducing myocardial infarct size. Isolated rat hearts were subjected to 30 min of global ischemia and 60 min of reperfusion [ischemia-reperfusion (I/R)]. In a second group, before isolation of the heart, a rIPC protocol (3 cycles of hindlimb I/R) was performed. Infarct size was measured with tetrazolium staining, and Akt/endothelial nitric oxide (NO) synthase (eNOS) expression/phosphorylation and mitochondrial function were evaluated after ischemia at 10 and 60 min of reperfusion. As expected, rIPC significantly decreased infarct size. This beneficial effect was abolished only when 8-cyclopentyl-1,3-dipropylxanthine (adenosine A1 receptor blocker) and NG-nitro-l-arginine methyl ester (NO synthesis inhibitor) were administered during the reperfusion phase. At the early reperfusion phase, rIPC induced significant Akt and eNOS phosphorylation, which was abolished by the perfusion with an adenosine A1 receptor blocker. I/R led to impaired mitochondrial function, which was attenuated by rIPC and mediated by adenosine A1 receptors. In conclusion, we demonstrated that rIPC limits myocardial infarct by activation of adenosine A1 receptors at early reperfusion in the isolated rat heart. Interestingly, rIPC appears to reduce myocardial infarct size by the Akt/eNOS pathway and improves mitochondrial function during myocardial reperfusion. NEW & NOTEWORTHY Adenosine is involved in classic preconditioning and acts especially through adenosine A1 and A3 receptors. However, its role in the mechanism of remote ischemic preconditioning is controversial. In this study, we demonstrated that remote ischemic preconditioning activates adenosine A1 receptors during early reperfusion, inducing Akt/endothelial nitric oxide synthase phosphorylation and improving mitochondrial function, thereby reducing myocardial infarct size.
Collapse
Affiliation(s)
- Diamela T Paez
- Faculty of Medicine, Department of Pathology, Institute of Cardiovascular Pathophysiology, University of Buenos Aires , Buenos Aires , Argentina.,National Council of Scientific and Technological Research (CONICET), Institute of Biochemistry and Molecular Medicine (IBIMOL), Faculty of Medicine, University of Buenos Aires , Buenos Aires , Argentina
| | - Mariana Garces
- CONICET, IBIMOL, Faculty of Pharmacy and Biochemistry, University of Buenos Aires , Buenos Aires , Argentina.,Faculty of Pharmacy and Biochemistry, Department of Analytical Chemistry and Physic Chemistry, General and Inorganic Chemistry, University of Buenos Aires , Buenos Aires , Argentina
| | - Valeria Calabró
- CONICET, IBIMOL, Faculty of Pharmacy and Biochemistry, University of Buenos Aires , Buenos Aires , Argentina.,Faculty of Pharmacy and Biochemistry, Department of Analytical Chemistry and Physic Chemistry, General and Inorganic Chemistry, University of Buenos Aires , Buenos Aires , Argentina
| | - Eliana P Bin
- Faculty of Medicine, Department of Pathology, Institute of Cardiovascular Pathophysiology, University of Buenos Aires , Buenos Aires , Argentina.,National Council of Scientific and Technological Research (CONICET), Institute of Biochemistry and Molecular Medicine (IBIMOL), Faculty of Medicine, University of Buenos Aires , Buenos Aires , Argentina
| | - Verónica D'Annunzio
- Faculty of Medicine, Department of Pathology, Institute of Cardiovascular Pathophysiology, University of Buenos Aires , Buenos Aires , Argentina.,National Council of Scientific and Technological Research (CONICET), Institute of Biochemistry and Molecular Medicine (IBIMOL), Faculty of Medicine, University of Buenos Aires , Buenos Aires , Argentina
| | - Julieta Del Mauro
- Faculty of Pharmacy and Biochemistry, Department of Pharmacology, University of Buenos Aires , Buenos Aires , Argentina
| | - Timoteo Marchini
- CONICET, IBIMOL, Faculty of Pharmacy and Biochemistry, University of Buenos Aires , Buenos Aires , Argentina.,Faculty of Pharmacy and Biochemistry, Department of Analytical Chemistry and Physic Chemistry, General and Inorganic Chemistry, University of Buenos Aires , Buenos Aires , Argentina
| | - Christian Höcht
- Faculty of Pharmacy and Biochemistry, Department of Pharmacology, University of Buenos Aires , Buenos Aires , Argentina
| | - Pablo Evelson
- CONICET, IBIMOL, Faculty of Pharmacy and Biochemistry, University of Buenos Aires , Buenos Aires , Argentina.,Faculty of Pharmacy and Biochemistry, Department of Analytical Chemistry and Physic Chemistry, General and Inorganic Chemistry, University of Buenos Aires , Buenos Aires , Argentina
| | - Ricardo J Gelpi
- Faculty of Medicine, Department of Pathology, Institute of Cardiovascular Pathophysiology, University of Buenos Aires , Buenos Aires , Argentina.,National Council of Scientific and Technological Research (CONICET), Institute of Biochemistry and Molecular Medicine (IBIMOL), Faculty of Medicine, University of Buenos Aires , Buenos Aires , Argentina
| | - Martín Donato
- Faculty of Medicine, Department of Pathology, Institute of Cardiovascular Pathophysiology, University of Buenos Aires , Buenos Aires , Argentina.,National Council of Scientific and Technological Research (CONICET), Institute of Biochemistry and Molecular Medicine (IBIMOL), Faculty of Medicine, University of Buenos Aires , Buenos Aires , Argentina
| |
Collapse
|
20
|
Han MX, Xu XW, Lu SQ, Zhang GX. Effect of olprinone on ischemia-reperfusion induced myocardial injury in rats. Biomed Pharmacother 2019; 111:1005-1012. [PMID: 30841413 DOI: 10.1016/j.biopha.2019.01.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 01/02/2019] [Accepted: 01/05/2019] [Indexed: 01/22/2023] Open
Abstract
AIMS This study investigated the effect of olprinone on ischemia-reperfusion (I/R) induced cardiac injury, and the underlying mechanism. MAIN METHODS Male Sprague-Dawley rats were subjected to a 30-min coronary arterial occlusion followed by 24 h reperfusion. After the start of reperfusion, rats were respectively treated with olprinone in three different dosages (0.2, 0.6, 2 mg/kg, intraperitoneal injection, i.p./12 h). Twenty-four hours later, a mean arterial pressure (MAP) heart function analysis system was used to monitor hemodynamic parameters; TTC staining method was used to detect the myocardial infarct size; 24-hour mortality of rats was recorded; western blot was used to detect the protein expressions of Caspase-3, Bax, Bcl-2, Beclin-1 and LC3-II/LC3-I. RESULTS Cardiac function in I/R group was lower than that in sham group (dp/dt max: 1348.29 ± 266.01 vs. 3333.73 ± 1258.03, -dp/dt max: 1163.23 ± 588.18 vs. 3198.93 ± 1416.00, P < 0.05), which was significantly improved by treatment with high dosage of olprinone (dp/dt max: 1348.29±266.01 vs. 2022.43±493.39, -dp/dt max: 1163.23±588.18 vs. 1784.50±418.92, P < 0.05). The percentage of myocardial infarct size in medium and high dosages of olprinone group was lower than that in I/R group (42.67 ± 2.94, 22.33 ± 3.63 vs. 63.67 ± 5.86, P < 0.05). There was no significant difference in mortality among each group within 24 h. Compared with sham group, the expression of Caspase-3 was significantly up-regulated in I/R group (3.44±0.47-fold of sham, P < 0.05), which was inhibited by medium dosage of olprinone treatment (2.00±0.52-fold of sham, P < 0.05 vs. I/R group); also, expression of Bax was increased compared with sham group (4.06±0.25-fold of sham, P < 0.05), which was markedly inhibited by all dosages of olprinone treatment (low: 2.16±0.61-fold, medium: 2.74±0.66-fold, high 1.65±0.55-fold, P < 0.05 vs. I/R group). Expression of Bcl-2 was increased after I/R (1.17±0.06-fold, P < 0.05), which was further elevated in all dosages of olprinone treatment (low: 1.62 ± 0.13-fold, medium: 1.46 ± 0.13-fold, high: 1.82 ± 0.39-fold, P < 0.05 vs. I/R group). In addition, compared with sham group, the expression of Beclin-1 was up-regulated to 1.44±0.05-fold of sham in I/R group (P < 0.05), which was further increased in low and medium dosages of olprinone group (low: 2.46±0.44-fold, medium: 2.80±0.75-fold, P < 0.05 vs. I/R group). Moreover, expression of LC3-II was elevated in low dosage of olprinone treated group (low: 4.50±0.47-fold, P < 0.05 vs. I/R group). CONCLUSIONS Olprinone improves the cardiac function in response to myocardial I/R injury by regulation of anti-apoptotic, pro-apoptotic. In addition, autophagic signal pathways may also play a role in olprinone's therapeutic effect.
Collapse
Affiliation(s)
- Meng-Xiao Han
- Department of Physiology, Medical College of Soochow University, 199 Ren-Ai Road, Dushu Lake Campus, Suzhou Industrial Park, Suzhou, 215123, PR China
| | - Xiao-Wen Xu
- Department of Emergency, The First Affiliated Hospital, Soochow University, 188 Shi-Zi Road, Suzhou, 215006, PR China; Department of Emergency, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, PR China
| | - Shi-Qi Lu
- Department of Emergency, The First Affiliated Hospital, Soochow University, 188 Shi-Zi Road, Suzhou, 215006, PR China.
| | - Guo-Xing Zhang
- Department of Physiology, Medical College of Soochow University, 199 Ren-Ai Road, Dushu Lake Campus, Suzhou Industrial Park, Suzhou, 215123, PR China.
| |
Collapse
|
21
|
Davidson SM, Arjun S, Basalay MV, Bell RM, Bromage DI, Bøtker HE, Carr RD, Cunningham J, Ghosh AK, Heusch G, Ibanez B, Kleinbongard P, Lecour S, Maddock H, Ovize M, Walker M, Wiart M, Yellon DM. The 10th Biennial Hatter Cardiovascular Institute workshop: cellular protection-evaluating new directions in the setting of myocardial infarction, ischaemic stroke, and cardio-oncology. Basic Res Cardiol 2018; 113:43. [PMID: 30310998 PMCID: PMC6182684 DOI: 10.1007/s00395-018-0704-z] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 10/04/2018] [Indexed: 12/13/2022]
Abstract
Due to its poor capacity for regeneration, the heart is particularly sensitive to the loss of contractile cardiomyocytes. The onslaught of damage caused by ischaemia and reperfusion, occurring during an acute myocardial infarction and the subsequent reperfusion therapy, can wipe out upwards of a billion cardiomyocytes. A similar program of cell death can cause the irreversible loss of neurons in ischaemic stroke. Similar pathways of lethal cell injury can contribute to other pathologies such as left ventricular dysfunction and heart failure caused by cancer therapy. Consequently, strategies designed to protect the heart from lethal cell injury have the potential to be applicable across all three pathologies. The investigators meeting at the 10th Hatter Cardiovascular Institute workshop examined the parallels between ST-segment elevation myocardial infarction (STEMI), ischaemic stroke, and other pathologies that cause the loss of cardiomyocytes including cancer therapeutic cardiotoxicity. They examined the prospects for protection by remote ischaemic conditioning (RIC) in each scenario, and evaluated impasses and novel opportunities for cellular protection, with the future landscape for RIC in the clinical setting to be determined by the outcome of the large ERIC-PPCI/CONDI2 study. It was agreed that the way forward must include measures to improve experimental methodologies, such that they better reflect the clinical scenario and to judiciously select combinations of therapies targeting specific pathways of cellular death and injury.
Collapse
Affiliation(s)
- Sean M Davidson
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Sapna Arjun
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Maryna V Basalay
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Robert M Bell
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Daniel I Bromage
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
| | - Richard D Carr
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
- MSD A/S, Copenhagen, Denmark
| | - John Cunningham
- Centre for Nephrology, UCL Medical School, Royal Free Campus, Rowland Hill Street, London, NW3 2PF, UK
| | - Arjun K Ghosh
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Gerd Heusch
- West German Heart and Vascular Center, Institute for Pathophysiology, University of Essen Medical School, Essen, Germany
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades CardioVasculares, Madrid, Spain
- IIS-Fundación Jiménez Díaz University Hospital, Madrid, Spain
| | - Petra Kleinbongard
- West German Heart and Vascular Center, Institute for Pathophysiology, University of Essen Medical School, Essen, Germany
| | - Sandrine Lecour
- Cardioprotection Group, Hatter Institute for Cardiovascular Research in Africa, University of Cape Town, Cape Town, South Africa
| | - Helen Maddock
- Centre for Sport, Exercise and Life Sciences, Faculty of Health and Life Sciences, Coventry University, Priory Street, Coventry, CV1 5FB, UK
| | - Michel Ovize
- INSERM U1060, CarMeN Laboratory, Université de Lyon and Service d'explorations Fonctionnelles Cardiovasculaires Groupement Hospitalier Est, 59 Boulevard Pinel, 69500, Bron, France
| | - Malcolm Walker
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Marlene Wiart
- INSERM U1060, CarMeN Laboratory, Université de Lyon and Service d'explorations Fonctionnelles Cardiovasculaires Groupement Hospitalier Est, 59 Boulevard Pinel, 69500, Bron, France
- CNRS, Lyon, France
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, 67 Chenies Mews, London, WC1E 6HX, UK.
| |
Collapse
|
22
|
Alpuche J, Quírino L, Sánchez-Vega JT, Yap J, Pérez-Campos E, Cabrera-Fuentes HA. The Role of Platelets in Ischemic Conditioning. CONDITIONING MEDICINE 2018; 1:313-318. [PMID: 30556056 PMCID: PMC6291202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Ischemic heart disease (IHD) is one of the leading causes of death and disability worldwide. Platelets, as the main regulators of hemostasis, are major players in acute myocardial ischemia/reperfusion injury (IRI). Additionally, platelets are modified by endogenous cardioprotective strategies such as ischemic preconditioning, postconditioning, and remote ischemic conditioning. In this article, we provide an overview of the functionional role of platelets in acute myocardial IRI, and highlight their potential as targets for cardioprotection to improve health outcomes in patients with IHD.
Collapse
Affiliation(s)
- Juan Alpuche
- CONACyT-Facultad de Medicina, Centro de Investigación Facultad de Medicina, UNAM-UABJO. Universidad Autónoma Benito Juárez de Oaxaca. México
- Centro de Investigación Facultad de Medicina, UNAM-UABJO. Universidad Autónoma Benito Juárez de Oaxaca
| | - Luz Quírino
- Centro de Investigación Facultad de Medicina, UNAM-UABJO. Universidad Autónoma Benito Juárez de Oaxaca
- Facultad de Odontología, Universidad Autónoma Benito Juárez de Oaxaca, México
| | - José T Sánchez-Vega
- Parasitology Laboratory, Department of Microbiology and Parasitology, Faculty of Medicine, Universidad Nacional Autónoma de México, México City, México
| | - Jonathan Yap
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Hawaii, USA
| | - Eduardo Pérez-Campos
- Centro de Investigación Facultad de Medicina, UNAM-UABJO. Universidad Autónoma Benito Juárez de Oaxaca
- Tecnológico Nacional de México/IT Oaxaca. Oaxaca. México
| | - Hector A. Cabrera-Fuentes
- Kazan Federal University, Department of Microbiology, Kazan, Russian Federation
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
- Escuela de Ingeniería y Ciencias, Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, Monterrey, NL, México
- Institute of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
23
|
Liehn EA, Ponomariov V, Diaconu R, Streata I, Ioana M, Crespo-Avilan GE, Hernández-Reséndiz S, Cabrera-Fuentes HA. Apolipoprotein E in Cardiovascular Diseases: Novel Aspects of an Old-fashioned Enigma. Arch Med Res 2018; 49:522-529. [PMID: 30213474 DOI: 10.1016/j.arcmed.2018.08.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 08/24/2018] [Indexed: 12/30/2022]
Abstract
The presence of different APOE isoforms represents a well-known risk factor for cardiovascular diseases. Besides the pleiotropic effects of APOE polymorphism on heart and neurological diseases, this review summarizes the less-known functions of APOE and the possible implications for cardiovascular disorders. Beyond the role as lipid transporting protein, its involvement in lipid membrane homeostasis and signaling, as well as its nuclear transcriptional effects suggests a more complex role of APOE, receiving great interest from researchers and physicians from all medical fields. Due to the presence of different APOE isoforms in human population, understanding APOE's role in pathological processes represents not only a challenge, but a demand for further development of therapeutic strategies for cardiovascular diseases.
Collapse
Affiliation(s)
- Elisa A Liehn
- Institute for Molecular Cardiovascular Research, Rheinisch Westfälische Technische Hochschule Aachen University, Aachen, Germany; Human Genomics Laboratory, University of Medicine and Pharmacy Craiova, Craiova, Romania; Department of Cardiology, Pulmonology, Angiology and Intensive Care, University Hospital, Rheinisch Westfälische Technische Hochschule, Aachen, Germany
| | - Victor Ponomariov
- Institute for Molecular Cardiovascular Research, Rheinisch Westfälische Technische Hochschule Aachen University, Aachen, Germany; Human Genomics Laboratory, University of Medicine and Pharmacy Craiova, Craiova, Romania
| | - Rodica Diaconu
- Human Genomics Laboratory, University of Medicine and Pharmacy Craiova, Craiova, Romania
| | - Ioana Streata
- Human Genomics Laboratory, University of Medicine and Pharmacy Craiova, Craiova, Romania
| | - Mihai Ioana
- Human Genomics Laboratory, University of Medicine and Pharmacy Craiova, Craiova, Romania
| | - Gustavo E Crespo-Avilan
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, Singapore; National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | - Sauri Hernández-Reséndiz
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, Singapore; National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | - Hector A Cabrera-Fuentes
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, Singapore; National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore; Kazan Federal University, Department of Microbiology, Kazan, Russian Federation; Escuela de Ingenieria y Ciencias, Centro de Biotecnologia-FEMSA, Tecnologico de Monterrey, Nuevo Leon, México; Institute of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany.
| |
Collapse
|
24
|
A modified approach for programmed electrical stimulation in mice: Inducibility of ventricular arrhythmias. PLoS One 2018; 13:e0201910. [PMID: 30133474 PMCID: PMC6104969 DOI: 10.1371/journal.pone.0201910] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 07/24/2018] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Electrophysiological studies in mice, the prevailing model organism in the field of basic cardiovascular research, are impeded by the low yield of programmed electrical stimulation (PES). OBJECTIVE To investigate a modified approach for ventricular arrhythmia (VA) induction and a novel scoring system in mice. METHOD A systematic review of literature on current methods for PES in mice searching the PubMed database revealed that VA inducibility was low and ranged widely (4.6 ± 10.7%). Based on this literature review, a modified PES protocol with 3 to 10 extrastimuli was developed and tested in comparison to the conventional PES protocol using up to 3 extrastimuli in anesthetized wildtype mice (C57BL/6J, n = 12). Induced VA, classified according to the Lambeth Convention, were assessed by established arrhythmia scores as well as a novel arrhythmia score based on VA duration. RESULTS PES with the modified approach raised both the occurrence and the duration of VA compared to conventional PES (0% vs 50%; novel VA score p = 0.0002). Particularly, coupling of >6 extrastimuli raised the induction of VA. Predominantly, premature ventricular complexes (n = 6) and ventricular tachycardia <1s (n = 4) were observed. Repeated PES after adrenergic stimulation using isoprenaline resulted in enhanced induction of ventricular tachycardia <1s in both protocols. CONCLUSION Our findings suggest that the presented approach of modified PES enables effective induction and quantification of VA in wildtype mice and may well be suited to document and evaluate detailed VA characteristics in mice.
Collapse
|
25
|
Hernández-Reséndiz S, Muñoz-Vega M, Contreras WE, Crespo-Avilan GE, Rodriguez-Montesinos J, Arias-Carrión O, Pérez-Méndez O, Boisvert WA, Preissner KT, Cabrera-Fuentes HA. Responses of Endothelial Cells Towards Ischemic Conditioning Following Acute Myocardial Infarction. CONDITIONING MEDICINE 2018; 1:247-258. [PMID: 30338315 PMCID: PMC6191189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
One of the primary therapeutic goals of modern cardiology is to design strategies aimed at minimizing myocardial infarct size and optimizing cardiac function following acute myocardial infarction (AMI). Patients with AMI who underwent reperfusion therapy display dysfunction of the coronary endothelium. Consequently, ischemic endothelial cells become more permeable and weaken their natural anti-thrombotic and anti-inflammatory potential. Ischemia-reperfusion injury (IRI) is associated with activation of the humoral and cellular components of the hemostatic and innate immune system, and also with excessive production of reactive oxygen species (ROS), the inhibition of nitric oxide synthase, and with inflammatory processes. Given its essential role in the regulation of vascular homeostasis, involving platelets and leukocytes among others, dysfunctional endothelium can lead to increased risk of coronary vasospasm and thrombosis. Endothelial dysfunction can be prevented by ischemic conditioning with a protective intervention based on limited intermittent periods of ischemia and reperfusion. The molecular mechanisms and signal transduction pathways underlying conditioning phenomena in the coronary endothelium have been described as involving less ROS production, reduced adhesion of neutrophils to endothelial cells and diminished inflammatory reactions. This review summarizes our current understanding of the cellular and molecular mechanisms regulating IRI-affected and -damaged coronary endothelium, and how ischemic conditioning may preserve its function.
Collapse
Affiliation(s)
- Sauri Hernández-Reséndiz
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
- Escuela de Ingeniería y Ciencias, Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, Monterrey, NL, México
| | - Mónica Muñoz-Vega
- Unidad de Trastornos del Movimiento y Sueño, Hospital General Dr. Manuel Gea González. Ciudad de México, México
| | - Whendy E Contreras
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | - Gustavo E Crespo-Avilan
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | | | - Oscar Arias-Carrión
- Molecular Biology Department, Instituto Nacional de Cardiología "Ignacio Chávez", C.D de México, México
| | - Oscar Pérez-Méndez
- Molecular Biology Department, Instituto Nacional de Cardiología "Ignacio Chávez", C.D de México, México
| | - William A Boisvert
- Kazan Federal University, Department of Microbiology, Kazan, Russian Federation
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Hawaii, USA
| | - Klaus T Preissner
- Kazan Federal University, Department of Microbiology, Kazan, Russian Federation
- Institute of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany
| | - Hector A Cabrera-Fuentes
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
- Escuela de Ingeniería y Ciencias, Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, Monterrey, NL, México
- Kazan Federal University, Department of Microbiology, Kazan, Russian Federation
- Institute of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
26
|
Bernhagen J. "Remote" myokine protects from pulmonary ischemia/reperfusion injury by a surprising "proximal" control mechanism. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:275. [PMID: 30094261 DOI: 10.21037/atm.2018.05.45] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Jürgen Bernhagen
- Department of Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Munich, Germany.,Munich Heart Alliance, Munich, Germany.,Munich Cluster for Systems Neurology, Munich, Germany
| |
Collapse
|
27
|
Abstract
Despite considerable advances in medicine, cardiovascular disease is still rising, with ischemic heart disease being the leading cause of death and disability worldwide. Thus extensive efforts are continuing to establish effective therapeutic modalities that would improve both quality of life and survival in this patient population. Novel therapies are being investigated not only to protect the myocardium against ischemia-reperfusion injury but also to regenerate the heart. Stem cell therapy, such as potential use of human mesenchymal stem cells and induced pluripotent stem cells and their exosomes, will make it possible not only to address molecular mechanisms of cardiac conditioning, but also to develop new therapies for ischemic heart disease. Despite the studies and progress made over the last 15 years on the use of stem cell therapy for cardiovascular disease, the efforts are still in their infancy. Even though the expectations have been high, the findings indicate that most of the clinical trials generally have been small and the results inconclusive. Because of many negative findings, there is certain pessimism that cardiac cell therapy is likely to yield any meaningful results over the next decade or so. Similar to other new technologies, early failures are not unusual and they may be followed by impressive success. Nevertheless, there has been considerable attention to safety by the clinical investigators because the adverse events of stem cell therapy have been impressively rare. In summary, although regenerative biology might not help the cardiovascular patient in the near term, it is destined to do so over the next several decades.
Collapse
Affiliation(s)
- Maia Terashvili
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI
| | - Zeljko J Bosnjak
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI.
| |
Collapse
|
28
|
Ong SB, Katwadi K, Kwek XY, Ismail NI, Chinda K, Ong SG, Hausenloy DJ. Non-coding RNAs as therapeutic targets for preventing myocardial ischemia-reperfusion injury. Expert Opin Ther Targets 2018; 22:247-261. [DOI: 10.1080/14728222.2018.1439015] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Sang-Bing Ong
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Khairunnisa Katwadi
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Xiu-Yi Kwek
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Nur Izzah Ismail
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School, Singapore, Singapore
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur, Malaysia
| | - Kroekkiat Chinda
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
- Biomedical Research Unit in Cardiovascular Sciences (BRUCS), Naresuan University, Phitsanulok, Thailand
| | - Sang-Ging Ong
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Derek J Hausenloy
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School, Singapore, Singapore
- National Heart Research Institute of Singapore, National Heart CentreSingapore, Singapore
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, London, UK
- Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore
| |
Collapse
|
29
|
Inflammation following acute myocardial infarction: Multiple players, dynamic roles, and novel therapeutic opportunities. Pharmacol Ther 2018; 186:73-87. [PMID: 29330085 PMCID: PMC5981007 DOI: 10.1016/j.pharmthera.2018.01.001] [Citation(s) in RCA: 547] [Impact Index Per Article: 78.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Acute myocardial infarction (AMI) and the heart failure that often follows, are major causes of death and disability worldwide. As such, new therapies are required to limit myocardial infarct (MI) size, prevent adverse left ventricular (LV) remodeling, and reduce the onset of heart failure following AMI. The inflammatory response to AMI, plays a critical role in determining MI size, and a persistent pro-inflammatory reaction can contribute to adverse post-MI LV remodeling, making inflammation an important therapeutic target for improving outcomes following AMI. In this article, we provide an overview of the multiple players (and their dynamic roles) involved in the complex inflammatory response to AMI and subsequent LV remodeling, and highlight future opportunities for targeting inflammation as a therapeutic strategy for limiting MI size, preventing adverse LV remodeling, and reducing heart failure in AMI patients.
Collapse
|
30
|
Hong H, Tao T, Chen S, Liang C, Qiu Y, Zhou Y, Zhang R. MicroRNA-143 promotes cardiac ischemia-mediated mitochondrial impairment by the inhibition of protein kinase Cepsilon. Basic Res Cardiol 2017; 112:60. [PMID: 28887629 DOI: 10.1007/s00395-017-0649-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Accepted: 09/05/2017] [Indexed: 12/30/2022]
Abstract
The cardioprotection of protein kinase Cepsilon (PKCε) against myocardial infarction (MI) mediated by its anti-apoptotic property and underlying mechanism of targeted regulation by microRNA (miRNA) are not established. MI-induced injury, PKCε expression, and targeted regulation of miRNA-143 (miR-143) to PKCε have been evaluated using animal MI and cellular hypoxic models conjugated with series of state-of-art molecular techniques. The results demonstrated that PKCε significantly downregulated along with increased infarcted area and apoptotic and necrotic damage in MI model, and the targeted relationship and potential binding profile were established between miR-143 and PKCε. Both in vivo and in vitro ischemic tests showed that miR-143 induced apoptosis and necrosis, which was reversed by antagomiR-143 or AMO-143. The upregulation of miR-143 by transfection of miR-143 in vitro also induced cell loss, and this effect of miR-143 was completely reversed by co-transfection of miR-143 with AMO-143. The identically deleterious action of miR-143 on mitochondrial membrane potential and ATP synthesis was also observed in both animal MI and cellular hypoxic models, as well as miR-143 overexpressed models and converted by either antagomiR or AMO. Importantly, overexpression of miR-143 downregulated PKCε in all tested models and this downregulation was reversed in the presence of antagomiR or AMO. The direct targeted regulation of miR-143 on PKCε was confirmed by luciferase reporter and miRNA-masking tests. In conclusion, MI-mediated upregulation of miR-143 inhibits PKCε expression and consequently interference with the cardioprotection of PKCε to mitochondrial, and leads to mitochondrial membrane potential dissipation and myocardial death eventually.
Collapse
Affiliation(s)
- Hong Hong
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Ting Tao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Si Chen
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Chaoqi Liang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yue Qiu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yuhong Zhou
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Rong Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China.
| |
Collapse
|
31
|
Activation of Endocannabinoid Receptor 2 as a Mechanism of Propofol Pretreatment-Induced Cardioprotection against Ischemia-Reperfusion Injury in Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:2186383. [PMID: 28814985 PMCID: PMC5549482 DOI: 10.1155/2017/2186383] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 04/20/2017] [Accepted: 04/30/2017] [Indexed: 01/16/2023]
Abstract
Propofol pretreatment before reperfusion, or propofol conditioning, has been shown to be cardioprotective, while its mechanism is unclear. The current study investigated the roles of endocannabinoid signaling in propofol cardioprotection in an in vivo model of myocardial ischemia/reperfusion (I/R) injury and in in vitro primary cardiomyocyte hypoxia/reoxygenation (H/R) injury. The results showed that propofol conditioning increased both serum and cell culture media concentrations of endocannabinoids including anandamide (AEA) and 2-arachidonoylglycerol (2-AG) detected by LC-MS/MS. The reductions of myocardial infarct size in vivo and cardiomyocyte apoptosis and death in vitro were accompanied with attenuations of oxidative injuries manifested as decreased reactive oxygen species (ROS), malonaldehyde (MDA), and MPO (myeloperoxidase) and increased superoxide dismutase (SOD) production. These effects were mimicked by either URB597, a selective endocannabinoids degradation inhibitor, or VDM11, a selective endocannabinoids reuptake inhibitor. In vivo study further validated that the cardioprotective and antioxidative effects of propofol were reversed by selective CB2 receptor antagonist AM630 but not CB1 receptor antagonist AM251. We concluded that enhancing endogenous endocannabinoid release and subsequent activation of CB2 receptor signaling represent a major mechanism whereby propofol conditioning confers antioxidative and cardioprotective effects against myocardial I/R injury.
Collapse
|
32
|
Cardioprotection by the transfer of coronary effluent from ischaemic preconditioned rat hearts: identification of cardioprotective humoral factors. Basic Res Cardiol 2017; 112:52. [PMID: 28695353 DOI: 10.1007/s00395-017-0641-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 07/06/2017] [Indexed: 01/24/2023]
Abstract
Ischaemic preconditioning (IPC) provides myocardial resistance to ischaemia/reperfusion (I/R) injuries. The protection afforded by IPC is not limited to the target tissue but extends to remote tissues, suggesting a mechanism mediated by humoral factors. The aim of the present study was to identify the humoral factors that are responsible for the cardioprotection induced by the coronary effluent transferred from IPC to naïve hearts. Isolated rat hearts were submitted to IPC (three cycles of 5 min I/R) before 30-min global ischaemia and 60-min reperfusion. The coronary effluent (Efl_IPC) collected during IPC was fractionated by ultrafiltration in different molecular weight ranges (<3, 3-5, 5-10, 10-30, 30-50, and >50 kDa) and evaluated for cardioprotective effects by perfusion before I/R in naïve hearts. Only the <3, 5-10 and <10 kDa fractions of hydrophobic eluate reduced I/R injuries. The cardioprotective effect of the 5-10 fraction was blocked by KATP channel blockers and a PKC inhibitor. An Efl_IPC proteomic analysis revealed 14 cytoprotection-related proteins in 4-12 kDa peptides. HSP10 perfusion protected the heart against I/R injuries. These data provide insights into the mechanisms of cardioprotection in humoral factors released by IPC. Cardioprotection is afforded by hydrophobic peptides in the 4-12 kDa size range, which activate pathways that are dependent on PKC and KATP. Fourteen 4-12 kDa peptides were identified, suggesting a potential therapeutic role for these molecules in ischaemic diseases. One of these, HSP10, identified by mass spectrometry, reduced I/R injuries and may be a potential candidate as a therapeutic target.
Collapse
|
33
|
Chun N, Haddadin AS, Liu J, Hou Y, Wong KA, Lee D, Rushbrook JI, Gulaya K, Hines R, Hollis T, Nistal Nuno B, Mangi AA, Hashim S, Pekna M, Catalfamo A, Chin HY, Patel F, Rayala S, Shevde K, Heeger PS, Zhang M. Activation of complement factor B contributes to murine and human myocardial ischemia/reperfusion injury. PLoS One 2017; 12:e0179450. [PMID: 28662037 PMCID: PMC5491012 DOI: 10.1371/journal.pone.0179450] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 05/29/2017] [Indexed: 12/28/2022] Open
Abstract
The pathophysiology of myocardial injury that results from cardiac ischemia and reperfusion (I/R) is incompletely understood. Experimental evidence from murine models indicates that innate immune mechanisms including complement activation via the classical and lectin pathways are crucial. Whether factor B (fB), a component of the alternative complement pathway required for amplification of complement cascade activation, participates in the pathophysiology of myocardial I/R injury has not been addressed. We induced regional myocardial I/R injury by transient coronary ligation in WT C57BL/6 mice, a manipulation that resulted in marked myocardial necrosis associated with activation of fB protein and myocardial deposition of C3 activation products. In contrast, in fB-/- mice, the same procedure resulted in significantly reduced myocardial necrosis (% ventricular tissue necrotic; fB-/- mice, 20 ± 4%; WT mice, 45 ± 3%; P < 0.05) and diminished deposition of C3 activation products in the myocardial tissue (fB-/- mice, 0 ± 0%; WT mice, 31 ± 6%; P<0.05). Reconstitution of fB-/- mice with WT serum followed by cardiac I/R restored the myocardial necrosis and activated C3 deposition in the myocardium. In translational human studies we measured levels of activated fB (Bb) in intracoronary blood samples obtained during cardio-pulmonary bypass surgery before and after aortic cross clamping (AXCL), during which global heart ischemia was induced. Intracoronary Bb increased immediately after AXCL, and the levels were directly correlated with peripheral blood levels of cardiac troponin I, an established biomarker of myocardial necrosis (Spearman coefficient = 0.465, P < 0.01). Taken together, our results support the conclusion that circulating fB is a crucial pathophysiological amplifier of I/R-induced, complement-dependent myocardial necrosis and identify fB as a potential therapeutic target for prevention of human myocardial I/R injury.
Collapse
Affiliation(s)
- Nicholas Chun
- Nephrology Division, Department of Medicine and Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Ala S. Haddadin
- Department of Anesthesiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Junying Liu
- Department of Anesthesiology, College of Medicine, SUNY Downstate Medical Center, Brooklyn, New York, United States of America
| | - Yunfang Hou
- Department of Anesthesiology, College of Medicine, SUNY Downstate Medical Center, Brooklyn, New York, United States of America
| | - Karen A. Wong
- Department of Anesthesiology, College of Medicine, SUNY Downstate Medical Center, Brooklyn, New York, United States of America
| | - Daniel Lee
- Department of Surgery, College of Medicine, SUNY Downstate Medical Center, Brooklyn, New York, United States of America
| | - Julie I. Rushbrook
- Department of Anesthesiology, College of Medicine, SUNY Downstate Medical Center, Brooklyn, New York, United States of America
| | - Karan Gulaya
- Department of Anesthesiology, College of Medicine, SUNY Downstate Medical Center, Brooklyn, New York, United States of America
| | - Roberta Hines
- Department of Anesthesiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Tamika Hollis
- Department of Anesthesiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Beatriz Nistal Nuno
- Department of Anesthesiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Abeel A. Mangi
- Department of Surgery, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Sabet Hashim
- Department of Surgery, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Marcela Pekna
- Department of Medical Chemistry and Cell Biology, Göteborg University, Göteborg, Sweden
| | - Amy Catalfamo
- Department of Anesthesiology, College of Medicine, SUNY Downstate Medical Center, Brooklyn, New York, United States of America
| | - Hsiao-ying Chin
- Department of Anesthesiology, College of Medicine, SUNY Downstate Medical Center, Brooklyn, New York, United States of America
| | - Foramben Patel
- Department of Biomedical Sciences, Long Island University, Brookville, New York, United States of America
| | - Sravani Rayala
- Department of Biomedical Sciences, Long Island University, Brookville, New York, United States of America
| | - Ketan Shevde
- Department of Anesthesiology, College of Medicine, SUNY Downstate Medical Center, Brooklyn, New York, United States of America
| | - Peter S. Heeger
- Nephrology Division, Department of Medicine and Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Ming Zhang
- Department of Anesthesiology, College of Medicine, SUNY Downstate Medical Center, Brooklyn, New York, United States of America
- Department of Cell Biology, College of Medicine, SUNY Downstate Medical Center, Brooklyn, New York, United States of America
| |
Collapse
|
34
|
Stieger P, Daniel JM, Thölen C, Dutzmann J, Knöpp K, Gündüz D, Aslam M, Kampschulte M, Langheinrich A, Fischer S, Cabrera-Fuentes H, Wang Y, Wollert KC, Bauersachs J, Braun-Dullaeus R, Preissner KT, Sedding DG. Targeting of Extracellular RNA Reduces Edema Formation and Infarct Size and Improves Survival After Myocardial Infarction in Mice. J Am Heart Assoc 2017. [PMID: 28637776 PMCID: PMC5669142 DOI: 10.1161/jaha.116.004541] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Following myocardial infarction (MI), peri-infarct myocardial edema formation further impairs cardiac function. Extracellular RNA (eRNA) released from injured cells strongly increases vascular permeability. This study aimed to assess the role of eRNA in MI-induced cardiac edema formation, infarct size, cardiac function, and survival after acute MI and to evaluate the therapeutic potential of ribonuclease 1 (RNase-1) treatment as an eRNA-degrading intervention. METHODS AND RESULTS C57BL/6J mice were subjected to MI by permanent ligation of the left anterior descending coronary artery. Plasma eRNA levels were significantly increased compared with those in controls starting from 30 minutes after ligation. Systemic application of RNase-1, but not DNase, significantly reduced myocardial edema formation 24 hours after ligation compared with controls. Consequently, eRNA degradation by RNase-1 significantly improved the perfusion of collateral arteries in the border zone of the infarcted myocardium 24 hours after ligation of the left anterior descending coronary artery, as detected by micro-computed tomography imaging. Although there was no significant difference in the area at risk, the area of vital myocardium was markedly larger in mice treated with RNase-1 compared with controls, as detected by Evans blue and 2,3,5-triphenyltetrazolium chloride staining. The increase in viable myocardium was associated with significantly preserved left ventricular function, as assessed by echocardiography. Moreover, RNase-1 significantly improved 8-week survival following MI. CONCLUSIONS eRNA is an unrecognized permeability factor in vivo, associated with myocardial edema formation after acute MI. RNase-1 counteracts eRNA-induced edema formation and preserves perfusion of the infarction border zone, reducing infarct size and protecting cardiac function after MI.
Collapse
Affiliation(s)
- Philipp Stieger
- Department of Cardiology and Angiology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Jan-Marcus Daniel
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Christiane Thölen
- Department of Cardiology and Angiology, University Hospital Giessen and Marburg, Giessen, Germany
| | - Jochen Dutzmann
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Kai Knöpp
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Dursun Gündüz
- Department of Cardiology and Angiology, University Hospital Giessen and Marburg, Giessen, Germany
| | - Muhammad Aslam
- Department of Cardiology and Angiology, University Hospital Giessen and Marburg, Giessen, Germany
| | - Marian Kampschulte
- Department of Radiology, University Hospital Giessen and Marburg, Giessen, Germany
| | | | - Silvia Fischer
- Institute of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany
| | - Hector Cabrera-Fuentes
- Institute of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany.,National Heart Research Institute, Singapore.,National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore.,Department of Microbiology, Kazan Federal University, Kazan, Russian Federation
| | - Yong Wang
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany.,Division of Molecular and Translational Cardiology, Hannover Medical School, Hannover, Germany
| | - Kai C Wollert
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany.,Division of Molecular and Translational Cardiology, Hannover Medical School, Hannover, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Rüdiger Braun-Dullaeus
- Department of Cardiology and Angiology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Klaus T Preissner
- Institute of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany
| | - Daniel G Sedding
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany .,Institute of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
35
|
Wang L, Yu J, Fordjour PA, Xing X, Gao H, Li Y, Li L, Zhu Y, Gao X, Fan G. Danshen injection prevents heart failure by attenuating post-infarct remodeling. JOURNAL OF ETHNOPHARMACOLOGY 2017; 205:22-32. [PMID: 28465251 DOI: 10.1016/j.jep.2017.04.027] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Revised: 04/16/2017] [Accepted: 04/26/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Danshen Injection (DSI) is a traditional Chinese medicine extracted from Danshen, prepared from the dried root and rhizome of Salvia miltiorrhiza Bunge. Danshen is an ancient antipyretic traditional Chinese medicine which is mostly used to improve blood circulation and dispel blood stasis. Danshen decoction or liquor-fried Danshen (with grain-based liquor) which is cool in nature is traditionally used to 'cool the blood' and reduce the swelling of sores and abscesses. AIM OF STUDY The present study aimed to examine the effect and mechanism of DSI in LAD induced heart injury. MATERIALS AND METHODS One day after LAD surgery, adult male Sprague-Dawley rats were randomized to 3 groups: MI group; DSI group (1.5ml/kg/d, intramuscular); and Valsartan group (10mg/kg/d, intragastric). Echocardiography and hemodynamic measurements (Pressure-Volume loop) were performed to evaluate cardiac function. Pathological methods (Masson, and Sirus red staining) were used to check myocardial fibrosis. Western blotting assay was used to detect the protein expression of MMP-2. RT-PCR was used to detect the gene expression of MMP-9, MPO, iNOS, Bcl-2 and Bax. RESULTS DSI administration to LAD rats resulted in improved cardiac functions, hemodynamic parameters and normalized ventricular mass. Furthermore, DSI-treated group demonstrated potential regulation of myocardial collagen I and III deposition associated with MMP-2 expression. Also, DSI administration decreased gene expression of iNOS, MPO and MMP-9, and increased Bcl-2/Bax ratio. CONCLUSION Myocardial fibrosis, cardiac hypertrophy, hemodynamic deterioration as well as systolic and diastolic dysfunctions which characterize a failing hearts were significantly prevented by DSI. Our study may provide future directions to focus on the anti-hypertrophic mechanisms of DSI and pathological roles played by MMP-2 in myocardial hypertrophy. Meanwhile, DSI also performed the effect of anti-inflammation by the way of decreasing iNOS and MPO. The way Danshen Injection increasing Bcl-2/Bax presented the possibility that it may has the effect of inhibiting cell death.
Collapse
Affiliation(s)
- Lingyan Wang
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of traditional Chinese Medicine, Tianjin, PR China.
| | - Jiahui Yu
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of traditional Chinese Medicine, Tianjin, PR China.
| | - Patrick Asare Fordjour
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of traditional Chinese Medicine, Tianjin, PR China.
| | - Xiaoxue Xing
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of traditional Chinese Medicine, Tianjin, PR China.
| | - Hui Gao
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of traditional Chinese Medicine, Tianjin, PR China.
| | - Yanyan Li
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of traditional Chinese Medicine, Tianjin, PR China.
| | - Lingyan Li
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of traditional Chinese Medicine, Tianjin, PR China.
| | - Yan Zhu
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of traditional Chinese Medicine, Tianjin, PR China.
| | - Xiumei Gao
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of traditional Chinese Medicine, Tianjin, PR China; State Key Laboratory of Modern Chinese Medicine, Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Medicine, Nankai District, Tianjin, PR China.
| | - Guanwei Fan
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of traditional Chinese Medicine, Tianjin, PR China; Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, NanKai District, Tianjin, PR China.
| |
Collapse
|
36
|
Jamaiyar A, Wan W, Ohanyan V, Enrick M, Janota D, Cumpston D, Song H, Stevanov K, Kolz CL, Hakobyan T, Dong F, Newby BMZ, Chilian WM, Yin L. Alignment of inducible vascular progenitor cells on a micro-bundle scaffold improves cardiac repair following myocardial infarction. Basic Res Cardiol 2017; 112:41. [PMID: 28540527 DOI: 10.1007/s00395-017-0631-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 05/18/2017] [Indexed: 12/26/2022]
Abstract
Ischemic heart disease is still the leading cause of death even with the advancement of pharmaceutical therapies and surgical procedures. Early vascularization in the ischemic heart is critical for a better outcome. Although stem cell therapy has great potential for cardiovascular regeneration, the ideal cell type and delivery method of cells have not been resolved. We tested a new approach of stem cell therapy by delivery of induced vascular progenitor cells (iVPCs) grown on polymer micro-bundle scaffolds in a rat model of myocardial infarction. iVPCs partially reprogrammed from vascular endothelial cells (ECs) had potent angiogenic potential and were able to simultaneously differentiate into vascular smooth muscle cells (SMCs) and ECs in 2D culture. Under hypoxic conditions, iVPCs also secreted angiogenic cytokines such as vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) as measured by enzyme-linked immunosorbent assay (ELISA). A longitudinal micro-scaffold made from poly(lactic-co-glycolic acid) was sufficient for the growth and delivery of iVPCs. Co-cultured ECs and SMCs aligned well on the micro-bundle scaffold similarly as in the vessels. 3D cell/polymer micro-bundles formed by iVPCs and micro-scaffolds were transplanted into the ischemic myocardium in a rat model of myocardial infarction (MI) with ligation of the left anterior descending artery. Our in vivo data showed that iVPCs on the micro-bundle scaffold had higher survival, and better retention and engraftment in the myocardium than free iVPCs. iVPCs on the micro-bundles promoted better cardiomyocyte survival than free iVPCs. Moreover, iVPCs and iVPC/polymer micro-bundles treatment improved cardiac function (ejection fraction and fractional shortening, endocardial systolic volume) measured by echocardiography, increased vessel density, and decreased infarction size [endocardial and epicardial infarct (scar) length] better than untreated controls at 8 weeks after MI. We conclude that iVPCs grown on a polymer micro-bundle scaffold are new promising approach for cell-based therapy designed for cardiovascular regeneration in ischemic heart disease.
Collapse
Affiliation(s)
- Anurag Jamaiyar
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH, 44272, USA.,School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Weiguo Wan
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH, 44272, USA
| | - Vahagn Ohanyan
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH, 44272, USA
| | - Molly Enrick
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH, 44272, USA
| | - Danielle Janota
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH, 44272, USA
| | - Devan Cumpston
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH, 44272, USA
| | - Hokyung Song
- Department of Chemical and Biomolecular Engineering, The University of Akron, Akron, OH, 44325, USA
| | - Kelly Stevanov
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH, 44272, USA
| | - Christopher L Kolz
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH, 44272, USA
| | - Tatev Hakobyan
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH, 44272, USA
| | - Feng Dong
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH, 44272, USA
| | - Bi-Min Zhang Newby
- Department of Chemical and Biomolecular Engineering, The University of Akron, Akron, OH, 44325, USA
| | - William M Chilian
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH, 44272, USA
| | - Liya Yin
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH, 44272, USA.
| |
Collapse
|
37
|
Quantifying the area-at-risk of myocardial infarction in-vivo using arterial spin labeling cardiac magnetic resonance. Sci Rep 2017; 7:2271. [PMID: 28536472 PMCID: PMC5442118 DOI: 10.1038/s41598-017-02544-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 04/12/2017] [Indexed: 01/05/2023] Open
Abstract
T2-weighted cardiovascular magnetic resonance (T2-CMR) of myocardial edema can quantify the area-at-risk (AAR) following acute myocardial infarction (AMI), and has been used to assess myocardial salvage by new cardioprotective therapies. However, some of these therapies may reduce edema, leading to an underestimation of the AAR by T2-CMR. Here, we investigated arterial spin labeling (ASL) perfusion CMR as a novel approach to quantify the AAR following AMI. Adult B6sv129-mice were subjected to in vivo left coronary artery ligation for 30 minutes followed by 72 hours reperfusion. T2-mapping was used to quantify the edema-based AAR (% of left ventricle) following ischemic preconditioning (IPC) or cyclosporin-A (CsA) treatment. In control animals, the AAR by T2-mapping corresponded to that delineated by histology. As expected, both IPC and CsA reduced MI size. However, IPC, but not CsA, also reduced myocardial edema leading to an underestimation of the AAR by T2-mapping. In contrast, regions of reduced myocardial perfusion delineated by cardiac ASL were able to delineate the AAR when compared to both T2-mapping and histology in control animals, and were not affected by either IPC or CsA. Therefore, ASL perfusion CMR may be an alternative method for quantifying the AAR following AMI, which unlike T2-mapping, is not affected by IPC.
Collapse
|
38
|
Bei Y, Xu T, Lv D, Yu P, Xu J, Che L, Das A, Tigges J, Toxavidis V, Ghiran I, Shah R, Li Y, Zhang Y, Das S, Xiao J. Exercise-induced circulating extracellular vesicles protect against cardiac ischemia-reperfusion injury. Basic Res Cardiol 2017; 112:38. [PMID: 28534118 DOI: 10.1007/s00395-017-0628-z] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 05/15/2017] [Indexed: 12/12/2022]
Abstract
Extracellular vesicles (EVs) serve an important function as mediators of intercellular communication. Exercise is protective for the heart, although the signaling mechanisms that mediate this cardioprotection have not been fully elucidated. Here using nano-flow cytometry, we found a rapid increase in plasma EVs in human subjects undergoing exercise stress testing. We subsequently identified that serum EVs were increased by ~1.85-fold in mice after 3-week swimming. Intramyocardial injection of equivalent quantities of EVs from exercised mice and non-exercised controls provided similar protective effects against acute ischemia/reperfusion (I/R) injury in mice. However, injection of exercise-induced EVs in a quantity equivalent to the increase seen with exercise (1.85 swim group) significantly enhanced the protective effect. Similarly, treatment with exercise-induced increased EVs provided additional anti-apoptotic effect in H2O2-treated H9C2 cardiomyocytes mediated by the activation of ERK1/2 and HSP27 signaling. Finally, by treating H9C2 cells with insulin-like growth factor-1 to mimic exercise stimulus in vitro, we found an increased release of EVs from cardiomyocytes associated with ALIX and RAB35 activation. Collectively, our results show that exercise-induced increase in circulating EVs enhances the protective effects of endogenous EVs against cardiac I/R injury. Exercise-derived EVs might serve as a potent therapy for myocardial injury in the future.
Collapse
Affiliation(s)
- Yihua Bei
- Cardiac Regeneration and Ageing Lab, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai, 200444, China
| | - Tianzhao Xu
- Cardiac Regeneration and Ageing Lab, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai, 200444, China
| | - Dongchao Lv
- Cardiac Regeneration and Ageing Lab, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai, 200444, China
| | - Pujiao Yu
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Jiahong Xu
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Lin Che
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Avash Das
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - John Tigges
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Vassilios Toxavidis
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Ionita Ghiran
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Ravi Shah
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Yongqin Li
- Cardiac Regeneration and Ageing Lab, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai, 200444, China
| | - Yuhui Zhang
- Heart Failure Care Unit, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Saumya Das
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai, 200444, China.
| |
Collapse
|
39
|
Antitumor Macrophage Response to Bacillus pumilus Ribonuclease (Binase). Mediators Inflamm 2017; 2017:4029641. [PMID: 28804220 PMCID: PMC5540387 DOI: 10.1155/2017/4029641] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 03/22/2017] [Indexed: 11/18/2022] Open
Abstract
Extracellular bacterial ribonucleases such as binase from Bacillus pumilus possess cytotoxic activity against tumor cells with a potential for clinical application. Moreover, they may induce activation of tumor-derived macrophages either into the M1-phenotype with well-documented functions in the regulation of the antitumor immune response or into M2-macrophages that may stimulate tumor growth, metastasis, and angiogenesis. In this study, binase or endogenous RNase1 (but not RNA or short oligonucleotides) stimulated the expression of activated NF-κB p65 subunit in macrophages. Since no changes in MyD88 and TRIF adaptor protein expression were observed, toll-like receptors may not be involved in RNase-related NF-κB pathway activation. In addition, short exposure (0.5 hr) to binase induced the release of cytokines such as IL-6, МСР-1, or TNF-α (but not IL-4 and IL-10), indicative for the polarization into antitumor M1-macrophages. Thus, we revealed increased expression of activated NF-κB p65 subunit in macrophages upon stimulation by binase and RNase1, but not RNA or short oligonucleotides.
Collapse
|
40
|
Kalkhoran SB, Munro P, Qiao F, Ong SB, Hall AR, Cabrera-Fuentes H, Chakraborty B, Boisvert WA, Yellon DM, Hausenloy DJ. Unique morphological characteristics of mitochondrial subtypes in the heart: the effect of ischemia and ischemic preconditioning. Discoveries (Craiova) 2017; 5. [PMID: 28736742 DOI: 10.15190/d.2017.1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
RATIONALE Three subsets of mitochondria have been described in adult cardiomyocytes - intermyofibrillar (IMF), subsarcolemmal (SSM), and perinuclear (PN). They have been shown to differ in physiology, but whether they also vary in morphological characteristics is unknown. Ischemic preconditioning (IPC) is known to prevent mitochondrial dysfunction induced by acute myocardial ischemia/reperfusion injury (IRI), but whether IPC can also modulate mitochondrial morphology is not known. AIMS Morphological characteristics of three different subsets of adult cardiac mitochondria along with the effect of ischemia and IPC on mitochondrial morphology will be investigated. METHODS Mouse hearts were subjected to the following treatments (N=6 for each group): stabilization only, IPC (3x5 min cycles of global ischemia and reperfusion), ischemia only (20 min global ischemia); and IPC and ischemia. Hearts were then processed for electron microscopy and mitochondrial morphology was assessed subsequently. RESULTS In adult cardiomyocytes, IMF mitochondria were found to be more elongated and less spherical than PN and SSM mitochondria. PN mitochondria were smaller in size when compared to the other two subsets. SSM mitochondria had similar area to IMF mitochondria but their sphericity measures were similar to PN mitochondria. Ischemia was shown to increase the sphericity parameters of all 3 subsets of mitochondria; reduce the length of IMF mitochondria, and increase the size of PN mitochondria. IPC had no effect on mitochondrial morphology either at baseline or after ischemia. CONCLUSION The three subsets of mitochondria in the adult heart are morphologically different. IPC does not appear to modulate mitochondrial morphology in adult cardiomyocytes.
Collapse
Affiliation(s)
- Siavash Beikoghli Kalkhoran
- Hatter Cardiovascular Institute, University College London, UK.,National Institute of Health Research University College London Hospitals Biomedical Research Ctr., UK
| | - Peter Munro
- Institute of Ophthalmology, University College London, UK
| | - Fan Qiao
- Centre for Quantitative Medicine, Duke-NUS Graduate Medical School, Singapore
| | - Sang-Bing Ong
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore.,National Heart Research Institute Singapore, National Heart Centre Singapore
| | - Andrew R Hall
- Hatter Cardiovascular Institute, University College London, UK.,National Institute of Health Research University College London Hospitals Biomedical Research Ctr., UK
| | - Hector Cabrera-Fuentes
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore.,National Heart Research Institute Singapore, National Heart Centre Singapore.,Kazan Federal University, Department of Microbiology, Kazan, Russian Federation.,Escuela de Ingenieria y Ciencias, Centro de Biotecnologia-FEMSA, Tecnologico de Monterrey, Mexico
| | - Bibhas Chakraborty
- Centre for Quantitative Medicine, Duke-NUS Graduate Medical School, Singapore
| | - William A Boisvert
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii
| | - Derek M Yellon
- Hatter Cardiovascular Institute, University College London, UK.,National Institute of Health Research University College London Hospitals Biomedical Research Ctr., UK
| | - Derek J Hausenloy
- Hatter Cardiovascular Institute, University College London, UK.,National Institute of Health Research University College London Hospitals Biomedical Research Ctr., UK.,Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore.,National Heart Research Institute Singapore, National Heart Centre Singapore.,Yong Loo Lin School of Medicine, National University Singapore, Singapore.,Barts Heart Centre, St Bartholomew's Hospital, London, UK
| |
Collapse
|
41
|
Amifostine Pretreatment Attenuates Myocardial Ischemia/Reperfusion Injury by Inhibiting Apoptosis and Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:4130824. [PMID: 28392886 PMCID: PMC5368387 DOI: 10.1155/2017/4130824] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 01/23/2017] [Accepted: 02/14/2017] [Indexed: 11/17/2022]
Abstract
The present study was aimed at investigating the effect of amifostine on myocardial ischemia/reperfusion (I/R) injury of mice and H9c2 cells cultured with TBHP (tert-butyl hydroperoxide). The results showed that pretreatment with amifostine significantly attenuated cell apoptosis and death, accompanied by decreased reactive oxygen species (ROS) production and lower mitochondrial potential (ΔΨm). In vivo, amifostine pretreatment alleviated I/R injury and decreased myocardial apoptosis and infarct area, which was paralleled by increased superoxide dismutase (SOD) and reduced malondialdehyde (MDA) in myocardial tissues, increased Bcl2 expression, decreased Bax expression, lower cleaved caspase-3 level, fewer TUNEL positive cells, and fewer DHE-positive cells in heart. Our results indicate that amifostine pretreatment has a protective effect against myocardial I/R injury via scavenging ROS.
Collapse
|
42
|
Civitarese RA, Kapus A, McCulloch CA, Connelly KA. Role of integrins in mediating cardiac fibroblast–cardiomyocyte cross talk: a dynamic relationship in cardiac biology and pathophysiology. Basic Res Cardiol 2016; 112:6. [DOI: 10.1007/s00395-016-0598-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 12/14/2016] [Indexed: 12/16/2022]
|