1
|
Lei F, Zhang J, Deng Y, Wang X, Tang J, Tian J, Wan Y, Wang L, Zhou X, Zhang Y, Li C. Biomimetic nanoplatform treats myocardial ischemia/reperfusion injury by synergistically promoting angiogenesis and inhibiting inflammation. Colloids Surf B Biointerfaces 2024; 243:114159. [PMID: 39137530 DOI: 10.1016/j.colsurfb.2024.114159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 08/15/2024]
Abstract
After myocardial ischemia/reperfusion injury (MI/RI), endothelial cell injury causes impaired angiogenesis and obstruction of microcirculation, resulting in an inflammatory outburst that exacerbates the damage. Therefore, synergistic blood vessel repair and inflammation inhibition are effective therapeutic strategies. In this study, we developed a platelet membrane (PM)-encapsulated baicalin nanocrystalline (BA NC) nanoplatform with a high drug load, BA NC@PM, which co-target to endothelial cells and macrophages through the transmembrane proteins of the PM to promote angiogenesis and achieve anti-inflammatory effects. In vitro cell scratch assays and transwell assay manifested that BA NC@PM could promote endothelial cell migration, as well as increase mRNA expression of CD31 and VEGF in the heart after treatment of MI/RI mice, suggesting its favorable vascular repair function. In addition, the preparation significantly reduced the expression of pro-inflammatory factors and increased the expression of anti-inflammatory factors in plasma, promoting the polarization of macrophages. Our study highlights a strategy for enhancing the treatment of MI/RI by promoting angiogenesis and regulating macrophage polarization via the biomimetic BA NC@PM nanoplatform.
Collapse
Affiliation(s)
- Fenting Lei
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jie Zhang
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yiping Deng
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xueqin Wang
- Department of Thyroid Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jun Tang
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Ji Tian
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Ying Wan
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Li Wang
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiangyu Zhou
- Department of Thyroid Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Basic Medicine Research Innovation Center for Cardiometabolic Disease, Ministry of Education, Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Yingying Zhang
- Department of Anaesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.
| | - Chunhong Li
- Basic Medicine Research Innovation Center for Cardiometabolic Disease, Ministry of Education, Southwest Medical University, Luzhou, Sichuan 646000, China; Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
2
|
Nording H, Müller OJ. CITED4 gene therapy: Can it be cited for preventing maladaptive post-ischemic cardiac remodeling? Mol Ther 2024; 32:3217-3219. [PMID: 39317189 PMCID: PMC11489530 DOI: 10.1016/j.ymthe.2024.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 09/04/2024] [Accepted: 09/04/2024] [Indexed: 09/26/2024] Open
Affiliation(s)
- Henry Nording
- Department of Internal Medicine V, University of Kiel, and German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany; DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Lübeck/Kiel, 23562 Lübeck, Germany
| | - Oliver J Müller
- Department of Internal Medicine V, University of Kiel, and German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany; DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Lübeck/Kiel, 23562 Lübeck, Germany.
| |
Collapse
|
3
|
Wisniewski AM, Chancellor WZ, Young A, Money D, Beller JP, Charlton J, Lunardi N, Yang Z, Laubach VE, Mehaffey JH, Kron IL, Roeser ME. Adenosine 2A Receptor Agonism Improves Survival in Extracorporeal Cardiopulmonary Resuscitation. J Surg Res 2024; 301:404-412. [PMID: 39029264 DOI: 10.1016/j.jss.2024.06.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/20/2024] [Accepted: 06/22/2024] [Indexed: 07/21/2024]
Abstract
INTRODUCTION Despite resuscitation advances including extracorporeal cardiopulmonary resuscitation (ECPR), freedom from neurologic and myocardial insult after cardiac arrest remains unlikely. We hypothesized that adenosine 2A receptor (A2AR) agonism, which attenuates reperfusion injury, would improve outcomes in a porcine model of ECPR. METHODS Adult swine underwent 20 min of circulatory arrest followed by defibrillation and 6 h of ECPR. Animals were randomized to receive saline vehicle or A2AR agonist (ATL1223 or Regadenoson) infusion during extracorporeal membrane oxygenation. Animals were weaned off extracorporeal membrane oxygenation and monitored for 24 h. Clinical and biochemical end points were compared. RESULTS The administration of A2AR agonists increased survival (P = 0.01) after cardiac arrest compared to vehicle. Markers of neurologic damage including S100 calcium binding protein B and glial fibrillary acidic protein were significantly lower with A2AR agonist treatment. CONCLUSIONS In a model of cardiac arrest treated with ECPR, A2AR agonism increased survival at 24 h and reduced neurologic damage suggesting A2AR activation may be a promising therapeutic target after cardiac arrest.
Collapse
Affiliation(s)
- Alex M Wisniewski
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia
| | - William Z Chancellor
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia
| | - Andrew Young
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia
| | - Dustin Money
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia
| | - Jared P Beller
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia
| | - Jennifer Charlton
- Department of Pediatrics, University of Virginia Health System, Charlottesville, Virginia
| | - Nadia Lunardi
- Department of Anesthesiology, University of Virginia Health System, Charlottesville, Virginia
| | - Zequan Yang
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia
| | - Victor E Laubach
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia
| | - J Hunter Mehaffey
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia
| | - Irving L Kron
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia
| | - Mark E Roeser
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia.
| |
Collapse
|
4
|
Wang Y, Li F, Wei S, Li W, Wu J, Li S, Hu X, Tang T, Liu X. Puerarin-Loaded Liposomes Co-Modified by Ischemic Myocardium-Targeting Peptide and Triphenylphosphonium Cations Ameliorate Myocardial Ischemia-Reperfusion Injury. Int J Nanomedicine 2024; 19:7997-8014. [PMID: 39130683 PMCID: PMC11317047 DOI: 10.2147/ijn.s468394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/29/2024] [Indexed: 08/13/2024] Open
Abstract
Purpose Mitochondrial damage may lead to uncontrolled oxidative stress and massive apoptosis, and thus plays a pivotal role in the pathological processes of myocardial ischemia-reperfusion (I/R) injury. However, it is difficult for the drugs such as puerarin (PUE) to reach the mitochondrial lesion due to lack of targeting ability, which seriously affects the expected efficacy of drug therapy for myocardial I/R injury. Methods We prepared triphenylphosphonium (TPP) cations and ischemic myocardium-targeting peptide (IMTP) co-modified puerarin-loaded liposomes (PUE@T/I-L), which effectively deliver the drug to mitochondria and improve the effectiveness of PUE in reducing myocardial I/R injury. Results In vitro test results showed that PUE@T/I-L had sustained release and excellent hemocompatibility. Fluorescence test results showed that TPP cations and IMTP double-modified liposomes (T/I-L) enhanced the intracellular uptake, escaped lysosomal capture and promoted drug targeting into the mitochondria. Notably, PUE@T/I-L inhibited the opening of the mitochondrial permeability transition pore, reduced intracellular reactive oxygen species (ROS) levels and increased superoxide dismutase (SOD) levels, thereby decreasing the percentage of Hoechst-positive cells and improving the survival of hypoxia-reoxygenated (H/R)-injured H9c2 cells. In a mouse myocardial I/R injury model, PUE@T/I-L showed a significant myocardial protective effect against myocardial I/R injury by protecting mitochondrial integrity, reducing myocardial apoptosis and decreasing infarct size. Conclusion This drug delivery system exhibited excellent mitochondrial targeting and reduction of myocardial apoptosis, which endowed it with good potential extension value in the precise treatment of myocardial I/R injury.
Collapse
Affiliation(s)
- Yan Wang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Institution of Clinical Pharmacy, Central South University, Changsha, People’s Republic of China
| | - Fengmei Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Institution of Clinical Pharmacy, Central South University, Changsha, People’s Republic of China
| | - Shanshan Wei
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Institution of Clinical Pharmacy, Central South University, Changsha, People’s Republic of China
| | - Wenqun Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Institution of Clinical Pharmacy, Central South University, Changsha, People’s Republic of China
| | - Junyong Wu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Institution of Clinical Pharmacy, Central South University, Changsha, People’s Republic of China
| | - Shengnan Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Institution of Clinical Pharmacy, Central South University, Changsha, People’s Republic of China
| | - Xiongbin Hu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Institution of Clinical Pharmacy, Central South University, Changsha, People’s Republic of China
| | - Tiantian Tang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Institution of Clinical Pharmacy, Central South University, Changsha, People’s Republic of China
| | - Xinyi Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Institution of Clinical Pharmacy, Central South University, Changsha, People’s Republic of China
| |
Collapse
|
5
|
Hu S, Wen J, Fan XD, Li P. Study on therapeutic mechanism of total salvianolic acids against myocardial ischemia-reperfusion injury based on network pharmacology, molecular docking, and experimental study. JOURNAL OF ETHNOPHARMACOLOGY 2024; 326:117902. [PMID: 38360382 DOI: 10.1016/j.jep.2024.117902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/04/2024] [Accepted: 02/08/2024] [Indexed: 02/17/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Radix Salviae miltiorrhizae, also known as Danshen in Chinese, effectively activates the blood and resolves stasis. Total salvianolic acids (SA) is the main active ingredient of Danshen, and related preparations, such as salvianolate injection are commonly used clinically to treat myocardial ischemia-reperfusion injury (MIRI). However, the potential targets and key active ingredients of SA have not been sufficiently investigated. AIM OF THE STUDY This study aimed to investigate the mechanism of action of SA in treating MIRI. MATERIALS AND METHODS Network pharmacology and molecular docking techniques were used to predict SA targets against MIRI. The key acting pathway of SA were validated by performing experiments in a rat MIRI model. RESULTS Twenty potential ingredients and 54 targets of SA in treating MIRI were identified. Ingredient-target-pathway network analysis revealed that salvianolic acid B and rosmarinic acid had the highest degree value. Pathway enrichment analysis showed that SA may regulate MIRI through the IL-17 signaling pathway, and this result was confirmed in the rat MIRI experiment. CONCLUSION The results of this study indicate that SA may protect MIRI by regulating the IL-17 pathway.
Collapse
Affiliation(s)
- Shuang Hu
- Institute of Basic Medical Sciences, XiYuan Hospital of China Academy of Chinese Medical Sciences, No.1 XiYuan CaoChang, Haidian District, Beijing, 100091, China; Key Laboratory of Pharmacology of Chinese Materia Medica of Beijing, No.1 XiYuan CaoChang, Haidian District, Beijing, 100091, China; Graduate School of China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Jing Wen
- Institute of Basic Medical Sciences, XiYuan Hospital of China Academy of Chinese Medical Sciences, No.1 XiYuan CaoChang, Haidian District, Beijing, 100091, China; Key Laboratory of Pharmacology of Chinese Materia Medica of Beijing, No.1 XiYuan CaoChang, Haidian District, Beijing, 100091, China; Graduate School of China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Xiao-di Fan
- Institute of Basic Medical Sciences, XiYuan Hospital of China Academy of Chinese Medical Sciences, No.1 XiYuan CaoChang, Haidian District, Beijing, 100091, China; Key Laboratory of Pharmacology of Chinese Materia Medica of Beijing, No.1 XiYuan CaoChang, Haidian District, Beijing, 100091, China.
| | - Peng Li
- Institute of Basic Medical Sciences, XiYuan Hospital of China Academy of Chinese Medical Sciences, No.1 XiYuan CaoChang, Haidian District, Beijing, 100091, China; Key Laboratory of Pharmacology of Chinese Materia Medica of Beijing, No.1 XiYuan CaoChang, Haidian District, Beijing, 100091, China.
| |
Collapse
|
6
|
Qiu L, Yan Y, Zhong G, Hou Z, Ye Y, Lin J, Luo D. Hydromorphone hydrochloride preconditioning combined with postconditioning attenuates myocardial ischemia/reperfusion injury in rats by improving mitochondrial function and activating the PI3K/Akt signaling pathway. Chem Biol Drug Des 2024; 103:e14474. [PMID: 38342769 DOI: 10.1111/cbdd.14474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/18/2024] [Accepted: 01/23/2024] [Indexed: 02/13/2024]
Abstract
Thrombolytic therapy or percutaneous coronary intervention for myocardial infarction often cause myocardial ischemia/reperfusion injury (MIRI) and poor prognosis of patients. This study aimed to explore the protective effect and potential mechanism of hydromorphone hydrochloride (HH) on MIRI. Fifty Sprague-Dawley male rats were randomly divided into Sham group, I/R group, HH-pre group, HH-post group, and HH-pre + post group. Except Sham group, MIRI models were established by ligating and relaxing the left anterior descending coronary artery, followed by tail vein injection of HH (0.3 μmol/L) 10 min before ligation (HH-pre group), 10 min after reperfusion (HH-post group), and twice at the above two time points (HH-pre + post group). After intervention, the cardiac function of rats was evaluated by echocardiography, and the levels of myocardial injury markers, oxidative stress indicators, and mitochondrial function indicators were detected. Next, the myocardial infarction area was evaluated by 2,3,5-triphenyltetrazolium chloride staining, mitochondrial biogenesis, and phosphoinositide 3 kinase (PI3K)/protein kinase B (Akt) signaling pathway by western blot. Compared with the I/R group, HH intervention improved cardiac function, decreased myocardial infarction area, reduced serum myocardial injury markers, alleviated oxidative stress, improved mitochondrial function, up-regulated mitochondrial biogenesis, and activated PI3K/Akt signaling pathway. Moreover, the HH-pre + post group was superior to the HH-pre and HH-post groups in the above aspects. Collectively, HH had protective effect on MIRI rats, and HH preconditioning combined with postconditioning showed optimal efficacy. Such efficacy may be achieved by promoting mitochondrial biogenesis to improve mitochondrial function and reduce oxidative stress, and activating the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Liuji Qiu
- Department of Anesthesiology, Huizhou Central People's Hospital, Huizhou, Guangdong, China
| | - Yan Yan
- Department of Anesthesiology, Huizhou Central People's Hospital, Huizhou, Guangdong, China
| | - Guocheng Zhong
- Department of Anesthesiology, Huizhou Central People's Hospital, Huizhou, Guangdong, China
| | - Zhiqi Hou
- Department of Anesthesiology, Huizhou Central People's Hospital, Huizhou, Guangdong, China
| | - Yongcai Ye
- Department of Anesthesiology, Huizhou Central People's Hospital, Huizhou, Guangdong, China
| | - Jiaying Lin
- Department of Anesthesiology, Huizhou Central People's Hospital, Huizhou, Guangdong, China
| | - Dexing Luo
- Department of Anesthesiology, Huizhou Central People's Hospital, Huizhou, Guangdong, China
| |
Collapse
|
7
|
Li YG, Li JH, Wang HQ, Liao J, Du XY. Cinnamaldehyde protects cardiomyocytes from oxygen-glucose deprivation/reoxygenation-induced lipid peroxidation and DNA damage via activating the Nrf2 pathway. Chem Biol Drug Des 2024; 103:e14489. [PMID: 38404216 DOI: 10.1111/cbdd.14489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/01/2024] [Accepted: 02/07/2024] [Indexed: 02/27/2024]
Abstract
Rapid restoration of perfusion in ischemic myocardium is the most direct and effective treatment for coronary heart disease but may cause myocardial ischemia/reperfusion injury (MIRI). Cinnamaldehyde (CA, C9H8O), a key component in the well-known Chinese medicine cinnamomum cassia, has cardioprotective effects against MIRI. This study aimed to observe the therapeutic effect of CA on MIRI and to elucidate its potential mechanism. H9C2 rat cardiomyocytes were pretreated with CA solution at 0, 10, and 100 μM, respectively and subjected to oxygen-glucose deprivation/reoxygenation (OGD/R). Then the cell viability, the NF-κB and caspase3 gene levels, the reduced glutathione (GSH)/oxidized glutathione (GSSG) ratio, superoxide dismutase (SOD) level, reactive oxygen species (ROS) generation, 4-hydroxynonenal (4-HNE), and malondialdehyde (MDA) were detected. The severity of DNA damage was assessed by tail moment (TM) values using alkaline comet assay. Besides, the DNA damage-related proteins and the key proteins of the Nrf2 pathway were detected by western blot. CA treatment increased the cell viability, GHS/GSSG ratio, SOD level, PARP1, Nrf2, PPAR-γ, and HO-1 protein levels of H9C2 cardiomyocytes, while reducing NF-κB, caspase3, ROS level, 4-HNE and MDA content, γ-H2AX protein level, and TM values. Inhibition of the Nrf2 pathway reversed the effect of CA on cell viability and apoptosis of OGD/R induced H9C2 cardiomyocytes. Besides, 100 μM CA was more effective than 10 μM CA. In the OGD/R-induced H9C2 cardiomyocyte model, CA can protect cardiomyocytes from MIRI by attenuating lipid peroxidation and repairing DNA damage. The mechanism may be related to the activation of the Nrf2 pathway.
Collapse
Affiliation(s)
- Yan-Guang Li
- Department of Cardiology, Central Hospital of Jiaozuo Coal Industry (Group) Co., LTD, Jiaozuo, Henan, China
| | - Jiang-Hong Li
- Department of Cardiology, Central Hospital of Jiaozuo Coal Industry (Group) Co., LTD, Jiaozuo, Henan, China
| | - Hai-Qin Wang
- Department of Cardiology, Central Hospital of Jiaozuo Coal Industry (Group) Co., LTD, Jiaozuo, Henan, China
| | | | - Xiao-Ya Du
- Department of Cardiology, Central Hospital of Jiaozuo Coal Industry (Group) Co., LTD, Jiaozuo, Henan, China
| |
Collapse
|
8
|
Qin D, Jia XF, Hanna A, Lee J, Pekson R, Elrod JW, Calvert JW, Frangogiannis NG, Kitsis RN. BAK contributes critically to necrosis and infarct generation during reperfused myocardial infarction. J Mol Cell Cardiol 2023; 184:1-12. [PMID: 37709008 PMCID: PMC10841630 DOI: 10.1016/j.yjmcc.2023.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 08/29/2023] [Accepted: 09/11/2023] [Indexed: 09/16/2023]
Abstract
At least seven cell death programs are activated during myocardial infarction (MI), but which are most important in causing heart damage is not understood. Two of these programs are mitochondrial-dependent necrosis and apoptosis. The canonical function of the pro-cell death BCL-2 family proteins BAX and BAK is to mediate permeabilization of the outer mitochondrial membrane during apoptosis allowing apoptogen release. BAX has also been shown to sensitize cells to mitochondrial-dependent necrosis, although the underlying mechanisms remain ill-defined. Genetic deletion of Bax or both Bax and Bak in mice reduces infarct size following reperfused myocardial infarction (MI/R), but the contribution of BAK itself to cardiomyocyte apoptosis and necrosis and infarction has not been investigated. In this study, we use Bak-deficient mice and isolated adult cardiomyocytes to delineate the role of BAK in the pathogenesis of infarct generation and post-infarct remodeling during MI/R and non-reperfused MI. Generalized homozygous deletion of Bak reduced infarct size ∼50% in MI/R in vivo, which was attributable primarily to decreases in necrosis. Protection from necrosis was also observed in BAK-deficient isolated cardiomyocytes suggesting that the cardioprotection from BAK loss in vivo is at least partially cardiomyocyte-autonomous. Interestingly, heterozygous Bak deletion, in which the heart still retains ∼28% of wild type BAK levels, reduced infarct size to a similar extent as complete BAK absence. In contrast to MI/R, homozygous Bak deletion did not attenuate acute infarct size or long-term scar size, post-infarct remodeling, cardiac dysfunction, or mortality in non-reperfused MI. We conclude that BAK contributes significantly to cardiomyocyte necrosis and infarct generation during MI/R, while its absence does not appear to impact the pathogenesis of non-reperfused MI. These observations suggest BAK may be a therapeutic target for MI/R and that even partial pharmacological antagonism may provide benefit.
Collapse
Affiliation(s)
- Dongze Qin
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, United States of America; Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Xiaotong F Jia
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, United States of America; Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Anis Hanna
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, United States of America; Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Jaehoon Lee
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, United States of America; Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Ryan Pekson
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, United States of America; Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - John W Elrod
- Department of Cardiovascular Sciences and Cardiovascular Research Center, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States of America
| | - John W Calvert
- Department of Surgery Emory University School of Medicine, Atlanta, GA, United States of America
| | - Nikolaos G Frangogiannis
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, United States of America; Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, United States of America; Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Richard N Kitsis
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, United States of America; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, United States of America; Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, United States of America.
| |
Collapse
|
9
|
Diab FMA, Ayobe MH, Abdel-Salam MF, Otman MFS, Abdel-Hady EA. Increased nitric oxide availability worsens the cardiac performance during early re-perfusion period in adult rats. J Basic Clin Physiol Pharmacol 2023; 34:629-637. [PMID: 34144641 DOI: 10.1515/jbcpp-2020-0358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 05/18/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Re-perfusion is the standard therapy for acute myocardial infarction, despite the associated pathologies that may contribute to irreversible myocardial injury. The present study aims to clarify the alterations in cardiac activities in response to experimental cardiac ischemic arrest followed by re-perfusion in isolated hearts perfused with nitric oxide (NO) donor, l-arginine, or NO inhibitor, Nω-Nitro-l-arginine methyl ester hydrochloride (l-NAME), to shed light on the possible role of NO in the re-perfusion process. METHODS Hearts isolated from adult Wistar rats were studied on Langendorff preparation under basal conditions and during 30 min re-perfusion following 30 min of total global ischemia. Rats were randomly divided into three groups; control and l-arginine or l-NAME infused heart groups. Cardiac tissue content of malondialdhyde, catalase and nitrite was also measured. RESULTS Compared to the control group, both l-arginine and l-NAME infused hearts showed increased basal chronotropy and myocardial flow rate. Following ischemia and during the whole period of re-perfusion, the three groups demonstrated significant deterioration in the inotropic activity and compromised myocardial flow rate. l-arginine infused hearts revealed depressed inotropy and chronotropy, weak systolic and diastolic functions with compromised myocardial flow at early 5 min of re-perfusion, yet with significantly higher myocardial flow rate by the end of re-perfusion. CONCLUSIONS Reducing NO availability by l-NAME revealed mild impact on the ischemia re-perfusion induced contractile dysfunction, whereas excess NO worsens cardiac performance at the early re-perfusion period.
Collapse
Affiliation(s)
- Faten M A Diab
- Physiology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mahmoud H Ayobe
- Physiology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | - Mohammed F S Otman
- Physiology Department, Faculty of Medicine, University of Elmergib, Al Khums, Libya
| | - Enas A Abdel-Hady
- Physiology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
10
|
Zeng JJ, Shi HQ, Ren FF, Zhao XS, Chen QY, Wang DJ, Wu LP, Chu MP, Lai TF, Li L. Notoginsenoside R1 protects against myocardial ischemia/reperfusion injury in mice via suppressing TAK1-JNK/p38 signaling. Acta Pharmacol Sin 2023; 44:1366-1379. [PMID: 36721009 PMCID: PMC10310839 DOI: 10.1038/s41401-023-01057-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 01/14/2023] [Indexed: 02/01/2023] Open
Abstract
Previous studies show that notoginsenoside R1 (NG-R1), a novel saponin isolated from Panax notoginseng, protects kidney, intestine, lung, brain and heart from ischemia-reperfusion injury. In this study we investigated the cardioprotective mechanisms of NG-R1 in myocardial ischemia/reperfusion (MI/R) injury in vivo and in vitro. MI/R injury was induced in mice by occluding the left anterior descending coronary artery for 30 min followed by 4 h reperfusion. The mice were treated with NG-R1 (25 mg/kg, i.p.) every 2 h for 3 times starting 30 min prior to ischemic surgery. We showed that NG-R1 administration significantly decreased the myocardial infarction area, alleviated myocardial cell damage and improved cardiac function in MI/R mice. In murine neonatal cardiomyocytes (CMs) subjected to hypoxia/reoxygenation (H/R) in vitro, pretreatment with NG-R1 (25 μM) significantly inhibited apoptosis. We revealed that NG-R1 suppressed the phosphorylation of transforming growth factor β-activated protein kinase 1 (TAK1), JNK and p38 in vivo and in vitro. Pretreatment with JNK agonist anisomycin or p38 agonist P79350 partially abolished the protective effects of NG-R1 in vivo and in vitro. Knockdown of TAK1 greatly ameliorated H/R-induced apoptosis of CMs, and NG-R1 pretreatment did not provide further protection in TAK1-silenced CMs under H/R injury. Overexpression of TAK1 abolished the anti-apoptotic effect of NG-R1 and diminished the inhibition of NG-R1 on JNK/p38 signaling in MI/R mice as well as in H/R-treated CMs. Collectively, NG-R1 alleviates MI/R injury by suppressing the activity of TAK1, subsequently inhibiting JNK/p38 signaling and attenuating cardiomyocyte apoptosis.
Collapse
Affiliation(s)
- Jing-Jing Zeng
- Department of Cardiology, Key Laboratory of Panvascular Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Department of Cardiology, Ningbo No. 2 Hospital, Ningbo, 315000, China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, 315000, China
| | - Han-Qing Shi
- Department of Cardiology, Key Laboratory of Panvascular Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Fang-Fang Ren
- Department of Cardiology, Key Laboratory of Panvascular Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Xiao-Shan Zhao
- Department of Cardiology, Key Laboratory of Panvascular Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Qiao-Ying Chen
- Department of Cardiology, Key Laboratory of Panvascular Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Dong-Juan Wang
- Department of Cardiology, Ningbo No. 2 Hospital, Ningbo, 315000, China
| | - Lian-Pin Wu
- Department of Cardiology, Key Laboratory of Panvascular Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Mao-Ping Chu
- Department of Cardiology, Key Laboratory of Panvascular Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Teng-Fang Lai
- Department of Cardiology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, China.
| | - Lei Li
- Department of Cardiology, Key Laboratory of Panvascular Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
11
|
Farkašová F, Kindernay L, Ferko M, Rajtík T, Szobi A, Ravingerová T. Age-Dependent Effects of Remote Preconditioning in Hypertensive Rat Hearts are Associated With Activation of RISK Signaling. Physiol Res 2023; 72:S11-S22. [PMID: 37294114 DOI: 10.33549/physiolres.935019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/23/2023] Open
Abstract
Remote ischemic preconditioning (RIPC) represents one of the forms of innate cardioprotection. While being effective in animal models, its application in humans has not been always beneficial, which might be attributed to the presence of various comorbidities, such as hypertension, or being related to the confounding factors, such as patients' sex and age. RIPC has been shown to mediate its cardioprotective effects through the activation of Reperfusion Injury Salvage Kinase (RISK) pathway in healthy animals, however, scarce evidence supports this effect of RIPC in the hearts of spontaneously hypertensive (SHR) rats, in particular, in relationship with aging. The study aimed to investigate the effectiveness of RIPC in male SHR rats of different age and to evaluate the role of RISK pathway in the effect of RIPC on cardiac ischemic tolerance. RIPC was performed using three cycles of inflation/deflation of the pressure cuff placed on the hind limb of anesthetized rats aged three, five and eight months. Subsequently, hearts were excised, Langendorff-perfused and exposed to 30-min global ischemia and 2-h reperfusion. Infarct-sparing and antiarrhythmic effects of RIPC were observed only in three and five months-old animals but not in eight months-old rats. Beneficial effects of RIPC were associated with increased activity of RISK and decreased apoptotic signaling only in three and five months-old animals. In conclusion, RIPC showed cardioprotective effects in SHR rats that were partially age-dependent and might be attributed to the differences in the activation of RISK pathway and various aspects of ischemia/reperfusion injury in aging animals.
Collapse
Affiliation(s)
- Farkašová Farkašová
- Institute for Heart Research, Centre of Experimental Medicine Slovak Academy of Sciences, Bratislava, Slovak Republic.
| | | | | | | | | | | |
Collapse
|
12
|
Lu CH, Chen DX, Dong K, Wu YJ, Na N, Wen H, Hu YS, Liang YY, Wu SY, Lin BY, Huang F, Zeng ZY. Inhibition of miR-143-3p alleviates myocardial ischemia reperfusion injury via limiting mitochondria-mediated apoptosis. Biol Chem 2023; 404:619-631. [PMID: 36780323 DOI: 10.1515/hsz-2022-0334] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 01/13/2023] [Indexed: 02/14/2023]
Abstract
MicroRNA (miR)-143-3p is a potential regulatory molecule in myocardial ischemia/reperfusion injury (MI/RI), wherein its expression and pathological effects remains controversial. Thus, a mouse MI/RI and cell hypoxia/reoxygenation (H/R) models were built for clarifying the miR-143-3p's role in MI/RI. Following myocardial ischemia for 30 min, mice underwent reperfusion for 3, 6, 12 and 24 h. It was found miR-143-3p increased in the ischemic heart tissue over time after reperfusion. Cardiomyocytes transfected with miR-143-3p were more susceptible to apoptosis. Mechanistically, miR-143-3p targeted B cell lymphoma 2 (bcl-2). And miR-143-3p inhibition reduced cardiomyocytes apoptosis upon H/R, whereas it was reversed by a specific bcl-2 inhibitor ABT-737. Of note, miR-143-3p inhibition upregulated bcl-2 with better mitochondrial membrane potential (Δψm), reduced cytoplasmic cytochrome c (cyto-c) and caspase proteins, and minimized infarction area in mice upon I/R. Collectively, inhibition of miR-143-3p might alleviate MI/RI via targeting bcl-2 to limit mitochondria-mediated apoptosis. To our knowledge, this study further clarifies the miR-143-3p's pathological role in the early stages of MI/RI, and inhibiting miR-143-3p could be an effective treatment for ischemic myocardial disease.
Collapse
Affiliation(s)
- Chuang-Hong Lu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, No.6 Shuangyong Road, Nanning 530021, Guangxi, China
| | - De-Xin Chen
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, No.6 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Kun Dong
- Department of Organ Transplantation, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Yun-Jiao Wu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, No.6 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Na Na
- Department of Chemistry, Scripps Research Institute, No.10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Hong Wen
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, No.6 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Yao-Shi Hu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, No.6 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Yuan-Ying Liang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, No.6 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Si-Yi Wu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, No.6 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Bei-You Lin
- Department of Cardiology, Zhuhai City People's Hospital, No.79 Kangning Road, Zhuhai 519050, Guangdong, China
| | - Feng Huang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, No.6 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Zhi-Yu Zeng
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, No.6 Shuangyong Road, Nanning 530021, Guangxi, China
| |
Collapse
|
13
|
Vargas I, Grabau RP, Chen J, Weinheimer C, Kovacs A, Dominguez-Viqueira W, Mitchell A, Wickline SA, Pan H. Simultaneous Inhibition of Thrombosis and Inflammation Is Beneficial in Treating Acute Myocardial Infarction. Int J Mol Sci 2023; 24:7333. [PMID: 37108494 PMCID: PMC10138953 DOI: 10.3390/ijms24087333] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/28/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Myocardial ischemia reperfusion injury (IRI) in acute coronary syndromes is a condition in which ischemic/hypoxic injury to cells subtended by the occluded vessel continues despite successful resolution of the thrombotic obstruction. For decades, most efforts to attenuate IRI have focused on interdicting singular molecular targets or pathways, but none have successfully transitioned to clinical use. In this work, we investigate a nanoparticle-based therapeutic strategy for profound but local thrombin inhibition that may simultaneously mitigate both thrombosis and inflammatory signaling pathways to limit myocardial IRI. Perfluorocarbon nanoparticles (PFC NP) were covalently coupled with an irreversible thrombin inhibitor, PPACK (Phe[D]-Pro-Arg-Chloromethylketone), and delivered intravenously to animals in a single dose prior to ischemia reperfusion injury. Fluorescent microscopy of tissue sections and 19F magnetic resonance images of whole hearts ex vivo demonstrated abundant delivery of PFC NP to the area at risk. Echocardiography at 24 h after reperfusion demonstrated preserved ventricular structure and improved function. Treatment reduced thrombin deposition, suppressed endothelial activation, inhibited inflammasome signaling pathways, and limited microvascular injury and vascular pruning in infarct border zones. Accordingly, thrombin inhibition with an extraordinarily potent but locally acting agent suggested a critical role for thrombin and a promising therapeutic strategy in cardiac IRI.
Collapse
Affiliation(s)
- Ian Vargas
- University of South Florida Heart Institute, University of South Florida, Tampa, FL 33602, USA
| | - Ryan P. Grabau
- University of South Florida Heart Institute, University of South Florida, Tampa, FL 33602, USA
| | - Junjie Chen
- Consortium for Translational Research in Advanced Imaging and Nanomedicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carla Weinheimer
- Cardiovascular Division, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Attila Kovacs
- Cardiovascular Division, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | - Adam Mitchell
- Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Samuel A. Wickline
- University of South Florida Heart Institute, University of South Florida, Tampa, FL 33602, USA
| | - Hua Pan
- Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63105, USA
| |
Collapse
|
14
|
Zhuang Y, Yu ML, Lu SF. Purinergic signaling in myocardial ischemia-reperfusion injury. Purinergic Signal 2023; 19:229-243. [PMID: 35254594 PMCID: PMC9984618 DOI: 10.1007/s11302-022-09856-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 02/18/2022] [Indexed: 10/18/2022] Open
Abstract
Purines and their derivatives, extensively distributed in the body, act as a class of extracellular signaling molecules via a rich array of receptors, also known as purinoceptors (P1, P2X, and P2Y). They mediate multiple intracellular signal transduction pathways and participate in various physiological and pathological cell behaviors. Since the function in myocardial ischemia-reperfusion injury (MIRI), this review summarized the involvement of purinergic signal transduction in diversified pathological processes, including energy metabolism disorder, oxidative stress injury, calcium overload, inflammatory immune response, platelet aggregation, coronary vascular dysfunction, and cell necrosis and apoptosis. Moreover, increasing evidence suggests that purinergic signaling also mediates the prevention and treatment of MIRI, such as ischemic conditioning, pharmacological intervention, and some other therapies. In conclusion, this review exhibited that purinergic signaling mediates the complex processes of MIRI which shows its promising application and prospecting in the future.
Collapse
Affiliation(s)
- Yi Zhuang
- College of Acupuncture and Tuina, Nanjing University of Chinese Medicine, 138 Xian-lin Avenue, Qixia District, Nanjing, 210023, Jiangsu Province, China
| | - Mei-Ling Yu
- College of Acupuncture and Tuina, Nanjing University of Chinese Medicine, 138 Xian-lin Avenue, Qixia District, Nanjing, 210023, Jiangsu Province, China
| | - Sheng-Feng Lu
- College of Acupuncture and Tuina, Nanjing University of Chinese Medicine, 138 Xian-lin Avenue, Qixia District, Nanjing, 210023, Jiangsu Province, China. .,Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
15
|
Chen W, Zhang Y, Wang Z, Tan M, Lin J, Qian X, Li H, Jiang T. Dapagliflozin alleviates myocardial ischemia/reperfusion injury by reducing ferroptosis via MAPK signaling inhibition. Front Pharmacol 2023; 14:1078205. [PMID: 36891270 PMCID: PMC9986553 DOI: 10.3389/fphar.2023.1078205] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 02/08/2023] [Indexed: 02/22/2023] Open
Abstract
Reperfusion is essential for ischemic myocardium but paradoxically leads to myocardial damage that worsens cardiac functions. Ferroptosis often occurs in cardiomyocytes during ischemia/reperfusion (I/R). The SGLT2 inhibitor dapagliflozin (DAPA) exerts cardioprotective effects independent of hypoglycemia. Here, we investigated the effect and potential mechanism of DAPA against myocardial ischemia/reperfusion injury (MIRI)-related ferroptosis using the MIRI rat model and hypoxia/reoxygenation (H/R)-induced H9C2 cardiomyocytes. Our results show that DAPA significantly ameliorated myocardial injury, reperfusion arrhythmia, and cardiac function, as evidenced by alleviated ST-segment elevation, ameliorated cardiac injury biomarkers including cTnT and BNP and pathological features, prevented H/R-triggered cell viability loss in vitro. In vitro and in vivo experiments showed that DAPA inhibited ferroptosis by upregulating the SLC7A11/GPX4 axis and FTH and inhibiting ACSL4. DAPA notably mitigated oxidative stress, lipid peroxidation, ferrous iron overload, and reduced ferroptosis. Subsequently, network pharmacology and bioinformatics analysis suggested that the MAPK signaling pathway was a potential target of DAPA and a common mechanism of MIRI and ferroptosis. DAPA treatment significantly reduced MAPK phosphorylation in vitro and in vivo, suggesting that DAPA might protect against MIRI by reducing ferroptosis through the MAPK signaling pathway.
Collapse
Affiliation(s)
- Weixiang Chen
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yue Zhang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zuoxiang Wang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Mingyue Tan
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jia Lin
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaodong Qian
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hongxia Li
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Tingbo Jiang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
16
|
Wang Y, Chen Y, Zhang T. Integrated whole-genome gene expression analysis reveals an atlas of dynamic immune landscapes after myocardial infarction. Front Cardiovasc Med 2023; 10:1087721. [PMID: 36937942 PMCID: PMC10020602 DOI: 10.3389/fcvm.2023.1087721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 02/10/2023] [Indexed: 03/06/2023] Open
Abstract
Introduction Myocardial infarction (MI) is a deadly medical condition leading to irreversible damage to the inflicted cardiac tissue. Elevated inflammatory response marks the severity of MI and is associated with the development of heart failure (HF), a long-term adverse outcome of MI. However, the efficacy of anti-inflammatory therapies for MI remains controversial. Deciphering the dynamic transcriptional signatures in peripheral blood mononuclear cells (PBMCs) is a viable and translatable route to better understand post-MI inflammation, which may help guide post-MI anti-inflammatory treatments. Methods In this work, integrated whole-genome gene expression analysis was performed to explore dynamic immune landscapes associated with MI. Results GSEA and GSVA showed that pathways involved in the inflammatory response and metabolic reprogramming were significantly enriched in PBMCs from MI patients. Based on leukocyte profiles generated by xCell algorithm, the relative abundance of monocytes and neutrophils was significantly increased in PBMCs from MI patients and had positive correlations with typical inflammation-associated transcripts. Mfuzz clustering revealed temporal gene expression profiles of PBMCs during the 6-month post-MI follow-up. Analysis of DEGs and gene sets indicated that PBMCs from HF group were characterized by elevated and lasting expression of genes implicated in inflammation and coagulation. Consensus clustering generated 4 metabolic subtypes of PBMCs with molecular heterogeneity in HF patients. Discussion In summary, integrated whole-genome gene expression analysis here outlines a transcriptomic framework that may improve the understanding of dynamic signatures present in PBMCs, as well as the heterogeneity of PBMCs in MI patients with or without long-term clinical outcome of HF. Moreover, the work here uncovers the diversity and heterogeneity of PBMCs from HF patients, providing novel bioinformatic evidence supporting the mechanistic implications of metabolic reprogramming and mitochondrial dysfunction in the post-MI inflammation and HF. Therefore, our work here supports the notion that individualized anti-inflammatory therapies are needed to improve the clinical management of post-MI patients.
Collapse
Affiliation(s)
- Yujue Wang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Chen
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Clinical Research Institute of Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- Laboratory of Clinical and Molecular Pharmacology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Yu Chen, ; Teng Zhang,
| | - Teng Zhang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Clinical Research Institute of Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Yu Chen, ; Teng Zhang,
| |
Collapse
|
17
|
Weng X, Tan H, Huang Z, Chen J, Zhang N, Wang Q, Li Q, Gao J, Sun D, Yakufu W, Wang Z, Li W, Zhu G, Pang Z, Song Y, Qian J, Ge J. Targeted delivery and ROS-responsive release of Resolvin D1 by platelet chimeric liposome ameliorates myocardial ischemia-reperfusion injury. J Nanobiotechnology 2022; 20:454. [PMID: 36266658 PMCID: PMC9585729 DOI: 10.1186/s12951-022-01652-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 09/27/2022] [Indexed: 11/10/2022] Open
Abstract
Resolvin D1 (RvD1) has been shown to provide effective protection against ischemia–reperfusion injury in multiple vital organs such as the heart, brain, kidney. However, the clinical translational potential of systemic administration of RvD1 in the treatment of ischemia–reperfusion injury is greatly limited due to biological instability and lack of targeting ability. Combining the natural inflammatory response and reactive oxygen species (ROS) overproduction after reperfusion injury, we developed a platelet-bionic, ROS-responsive RvD1 delivery platform. The resulting formulation enables targeted delivery of RvD1 to the injury site by hijacking circulating chemotactic monocytes, while achieving locally controlled release. In a mouse model of myocardial ischemia repefusuin (MI/R) injury, intravenous injection of our formula resulted in the enrichment of RvD1 in the injured area, which in turn promotes clearance of dead cells, production of specialized proresolving mediators (SPMs), and angiogenesis during injury repair, effectively improving cardiac function. This delivery system integrates drug bio-protection, targeted delivery and controlled release, which endow it with great clinical translational value.
Collapse
Affiliation(s)
- Xueyi Weng
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases , Fudan University, 180 Feng Lin Road, Shanghai, 200032, China.,National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine, 180 Feng Lin Road, Shanghai, 200032, China
| | - Haipeng Tan
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases , Fudan University, 180 Feng Lin Road, Shanghai, 200032, China.,National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine, 180 Feng Lin Road, Shanghai, 200032, China
| | - Zheyong Huang
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases , Fudan University, 180 Feng Lin Road, Shanghai, 200032, China.,National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine, 180 Feng Lin Road, Shanghai, 200032, China
| | - Jing Chen
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases , Fudan University, 180 Feng Lin Road, Shanghai, 200032, China.,National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine, 180 Feng Lin Road, Shanghai, 200032, China
| | - Ning Zhang
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases , Fudan University, 180 Feng Lin Road, Shanghai, 200032, China.,National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine, 180 Feng Lin Road, Shanghai, 200032, China
| | - Qiaozi Wang
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases , Fudan University, 180 Feng Lin Road, Shanghai, 200032, China.,National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine, 180 Feng Lin Road, Shanghai, 200032, China
| | - Qiyu Li
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases , Fudan University, 180 Feng Lin Road, Shanghai, 200032, China.,National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine, 180 Feng Lin Road, Shanghai, 200032, China
| | - Jinfeng Gao
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases , Fudan University, 180 Feng Lin Road, Shanghai, 200032, China.,National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine, 180 Feng Lin Road, Shanghai, 200032, China
| | - Dili Sun
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases , Fudan University, 180 Feng Lin Road, Shanghai, 200032, China.,National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine, 180 Feng Lin Road, Shanghai, 200032, China
| | - Wusiman Yakufu
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases , Fudan University, 180 Feng Lin Road, Shanghai, 200032, China.,National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine, 180 Feng Lin Road, Shanghai, 200032, China
| | - Zhengmin Wang
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases , Fudan University, 180 Feng Lin Road, Shanghai, 200032, China.,National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine, 180 Feng Lin Road, Shanghai, 200032, China
| | - Weiyan Li
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases , Fudan University, 180 Feng Lin Road, Shanghai, 200032, China.,National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine, 180 Feng Lin Road, Shanghai, 200032, China
| | - Guangrui Zhu
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases , Fudan University, 180 Feng Lin Road, Shanghai, 200032, China.,National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine, 180 Feng Lin Road, Shanghai, 200032, China
| | - Zhiqing Pang
- School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, 826 Zhangheng Road, Shanghai, 201203, China.
| | - Yanan Song
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases , Fudan University, 180 Feng Lin Road, Shanghai, 200032, China. .,National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine, 180 Feng Lin Road, Shanghai, 200032, China.
| | - Juying Qian
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases , Fudan University, 180 Feng Lin Road, Shanghai, 200032, China. .,National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine, 180 Feng Lin Road, Shanghai, 200032, China.
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases , Fudan University, 180 Feng Lin Road, Shanghai, 200032, China.,National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine, 180 Feng Lin Road, Shanghai, 200032, China.,Institute of Biomedical Science, Fudan University, 180 Feng Lin Road, Shanghai, 200032, China
| |
Collapse
|
18
|
Rao P, Li C, Wang L, Jiang Y, Yang L, Li H, Yang P, Tao J, Lu D, Sun L. ZNF143 regulates autophagic flux to alleviate myocardial ischemia/reperfusion injury through Raptor. Cell Signal 2022; 99:110444. [PMID: 35988805 DOI: 10.1016/j.cellsig.2022.110444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 08/06/2022] [Accepted: 08/16/2022] [Indexed: 11/03/2022]
Abstract
The exact role of autophagy in myocardial ischemia/reperfusion (I/R) injury is still controversial. Excessive or insufficient autophagy may lead to cell death. Therefore, how to regulate autophagic balance during myocardial ischemia/reperfusion is critical to the treatment of myocardial I/R injury. Raptor is an mTOR regulatory related protein and closely related to the induction of autophagy. ZNF143 is widely expressed in various cells and acts as a transcription factor, which is involved in the regulation of autophagy, cell growth and development. In this study, we aimed to explore the mechanism by which ZNF143 regulated autophagy in myocardial I/R injury and the relationship between ZNF143 and Raptor. In our results, we found that ZNF143 expression was down-regulated in myocardial I/R. Inhibition of ZNF143 expression further enhanced autophagy and restored the deficiency of autophagic flux caused by myocardial I/R, subsequently alleviating myocardial I/R injury. On the other hand, overexpression of ZNF143 up-regulated Raptor expression and reduced autophagic activity, consequently exacerbating myocardial I/R injury. Taken together, our study revealed that ZNF143 might be a key target of the regulation of autophagy and a novel therapeutic target of myocardial I/R injury.
Collapse
Affiliation(s)
- Peng Rao
- Department of Cardiology, The Second Affiliated Hospital, Kunming Medical University, Kunming 650101, China
| | - Changyan Li
- Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming 650500, China
| | - Limeiting Wang
- Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming 650500, China
| | - Yongliang Jiang
- Department of Cardiology, The Second Affiliated Hospital, Kunming Medical University, Kunming 650101, China
| | - Lin Yang
- Department of Cardiology, The Second Affiliated Hospital, Kunming Medical University, Kunming 650101, China
| | - Hao Li
- Department of Cardiology, The Second Affiliated Hospital, Kunming Medical University, Kunming 650101, China
| | - Ping Yang
- Department of Cardiology, The Second Affiliated Hospital, Kunming Medical University, Kunming 650101, China
| | - Jun Tao
- Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming 650500, China
| | - Di Lu
- Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming 650500, China.
| | - Lin Sun
- Department of Cardiology, The Second Affiliated Hospital, Kunming Medical University, Kunming 650101, China.
| |
Collapse
|
19
|
Circ_0050908 up-regulates TRAF3 by sponging miR-324-5p to aggravate myocardial ischemia-reperfusion injury. Int Immunopharmacol 2022; 108:108740. [DOI: 10.1016/j.intimp.2022.108740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/25/2021] [Accepted: 03/28/2022] [Indexed: 11/21/2022]
|
20
|
Feng J, Wu Y. Interleukin-35 ameliorates cardiovascular disease by suppressing inflammatory responses and regulating immune homeostasis. Int Immunopharmacol 2022; 110:108938. [PMID: 35759811 DOI: 10.1016/j.intimp.2022.108938] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/05/2022] [Accepted: 06/07/2022] [Indexed: 12/14/2022]
Abstract
The immune response is of great significance in the initiation and progression of a diversity of cardiovascular diseases involving pro-and anti-inflammatory cytokines. Interleukin-35 (IL-35), a cytokine of the interleukin-12 family, is a novel anti-inflammation and immunosuppressive cytokine, maintaining inflammatory suppression and regulating immune homeostasis. The role of IL-35 in cardiovascular diseases (CVDs) has aroused enthusiastic attention, a diversity of experimental or clinical evidence has indicated that IL-35 potentially has a pivot role in protecting against cardiovascular diseases, especially atherosclerosis and myocarditis. In this review, we initiate an overview of the relationship between Interleukin-35 and cardiovascular diseases, including atherosclerosis, acute coronary syndrome, pulmonary hypertension, abdominal aortic aneurysm, heart failure, myocardial ischemia-reperfusion, aortic dissection and myocarditis. Although the specific molecular mechanisms entailing the protective effects of IL-35 remain an unsolved issue, targeted therapies with IL-35 might provide a promising and effective solution to prevent and cure cardiovascular diseases.
Collapse
Affiliation(s)
- Jie Feng
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Yanqing Wu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
21
|
Yu D, Gong X, Zhang Y, Li Q, Zhang M. Tropisetron Preconditioning Decreases Myocardial Biomarkers in Patients Undergoing Heart Valve Replacement Surgery. Front Med (Lausanne) 2022; 9:690272. [PMID: 35425785 PMCID: PMC9002259 DOI: 10.3389/fmed.2022.690272] [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: 04/02/2021] [Accepted: 03/01/2022] [Indexed: 12/01/2022] Open
Abstract
Background Cardioplegic arrest during the heart valve replacement surgery frequently leads to myocardial damage. Tropisetron (TRP) has been demonstrated to reduce myocardial ischemia-reperfusion injury and inflammation in animals. We examined the efficacy of TRP in lowering myocardial biomarkers in patients undergoing heart valve replacement surgery. Methods A total of seventy-five patients, scheduled for elective heart valve replacement surgery, were randomly chosen to receive either 10 ml of normal saline or 10 mg/10 ml of TRP immediately after anesthesia induction. Blood samples for the measurement of cardiac troponin I (cTnI), creatine kinase (CK-MB), lactate dehydrogenase (LDH), tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-10 (IL-10) were taken before anesthesia, as well as 4, 12, and 24 h after aortic cross-clamp release to evaluate myocardial injury using two-way ANOVA for repeated measurements. The study was registered at www.chictr.org.cn (number, ChiCTR-1800018681). Results Treatment with TRP decreased the increment of cTnI (Fgroup = 4.911, p = 0.030; Ftime = 55.356, p = 0.001; Fgroup × time = 5.340, p = 0.002) at 12 and 24 h; of CK-MB (Fgroup = 6.552, p = 0.013; Ftime = 49.276, p = 0.001; Fgroup × time = 7.627, p = 0.003) at 4, 12, and 24 h; of TNF-α (Fgroup = 4.153, p = 0.046; Ftime = 28.244, p = 0.002; Fgroup × time = 4.692, p = 0.006) at 4 and 12 h; and of LDH (Fgroup = 4.275, p = 0.043; Ftime = 63.225, p = 0.001; Fgroup × time = 2.501, p = 0.083) at 24 h after the release of the aortic cross-clamp. It increased IL-10 (Fgroup = 5.958, p = 0.018; Ftime = 31.226, p = 0.002; Fgroup × time = 1.464, p = 0.236) at 12 h after the release of the aortic cross-clamp. Multiple linear regression analysis showed that cardiopulmonary bypass (CPB) time was a risk factor, and that TRP treatment was a protective factor for postoperative cTNI change (β = 4.449, 95% CI [0.97–7.92], p = 0.013 for CPB time; and β = −381, 95% CI [−613.4 to −148.5], p = 0.002 for TRP treatment). Conclusions Tropisetron had cardioprotective and anti-inflammatory effects in patients undergoing heart valve replacement surgery with cardioplegic arrest. The addition of TRP and reduction of CPB time should be considered for myocardial protection in heart valve replacement surgery. Clinical Trial Registration [www.chictr.org.cn/index.aspx], identifier [ChiCTR1800018681].
Collapse
Affiliation(s)
- Di Yu
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, China.,Hubei No. 3 People's Hospital of Jianghan University, Wuhan, China
| | - Xingrui Gong
- Department of Anesthesiology, Xiangyang Central Hospital, Hubei University of Arts and Science, Xiangyang, China
| | - Yufei Zhang
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Qing Li
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Mazhang Zhang
- Department of Anesthesiology, Shanghai Children' Medical Central, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
22
|
Recombinant Klotho Protein Ameliorates Myocardial Ischemia/Reperfusion Injury by Attenuating Sterile Inflammation. Biomedicines 2022; 10:biomedicines10040894. [PMID: 35453645 PMCID: PMC9032004 DOI: 10.3390/biomedicines10040894] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/31/2022] [Accepted: 04/08/2022] [Indexed: 02/01/2023] Open
Abstract
Currently, no effective therapy and potential target have been elucidated for preventing myocardial ischemia and reperfusion injury (I/R). We hypothesized that the administration of recombinant klotho (rKL) protein could attenuate the sterile inflammation in peri-infarct regions by inhibiting the extracellular release of high mobility group box-1 (HMGB1). This hypothesis was examined using a rat coronary artery ligation model. Rats were divided into sham, sham+ rKL, I/R, and I/R+ rKL groups (n = 5/group). Administration of rKL protein reduced infarct volume and attenuated extracellular release of HMGB1 from peri-infarct tissue after myocardial I/R injury. The administration of rKL protein inhibited the expression of pro-inflammatory cytokines in the peri-infarct regions and significantly attenuated apoptosis and production of intracellular reactive oxygen species by myocardial I/R injury. Klotho treatment significantly reduced the increase in the levels of circulating HMGB1 in blood at 4 h after myocardial ischemia. rKL regulated the levels of inflammation-related proteins. This is the first study to suggest that exogenous administration of rKL exerts myocardial protection effects after I/R injury and provides new mechanistic insights into rKL that can provide the theoretical basis for clinical application of new adjunctive modality for critical care of acute myocardial infarction.
Collapse
|
23
|
Risk Factors for No-Reflow in Patients with ST-Elevation Myocardial Infarction Who Underwent Percutaneous Coronary Intervention: A Case-Control Study. Cardiol Res Pract 2022; 2022:3482518. [PMID: 35308062 PMCID: PMC8930256 DOI: 10.1155/2022/3482518] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/06/2022] [Accepted: 02/25/2022] [Indexed: 11/17/2022] Open
Abstract
Methods This case-control study retrospectively reviewed the medical data of patients treated with primary percutaneous coronary intervention within 12 h after STEMI onset between January 2010 and January 2013 at the Department of Cardiology of the Beijing Anzhen Hospital. Results A total of 902 patients were included in the analysis. The basic characteristics between the reflow and no-reflow groups were similar, except for time-to-hospital admission, heart rate, plasma glucose, high-sensitivity C-reactive protein (hsCRP)/prealbumin (PAB), neutrophil count, intraaortic balloon pump, and aspiration thrombectomy. The multivariable analysis showed that hsCRP/PAB (OR = 1.003, 95% CI: 1.000–1.006, P=0.022), neutrophil count (OR = 1.085, 95% CI: 1.028–1.146, P=0.003), plasma glucose levels (OR = 1.086, 95% CI: 1.036–1.138, P=0.001), diabetes mellitus (OR = 0.596, 95% CI: 0.371–0.958, P=0.033), Killip classification >1 (OR = 2.002, 95% CI: 1.273–3.148, P=0.003), intraoperative intraaortic balloon pump (IABP) use (OR = 3.257, 95% CI: 1.954–5.428, P=0.001), and aspiration thrombectomy (OR = 3.412, 95% CI: 2.259–5.152, P=0.001) were independently associated with no-reflow. Conclusion hsCRP/PAB, neutrophil count, plasma glucose levels, diabetes mellitus, Killip classification, intraoperative IABP use, and aspiration thrombectomy were independent risk factors for no-reflow in patients with STEMI.
Collapse
|
24
|
Hakovirta H, Jalkanen J, Saimanen E, Kukkonen T, Romsi P, Suominen V, Vikatmaa L, Valtonen M, Karvonen MK, Venermo M. Induction of CD73 prevents death after emergency open aortic surgery for a ruptured abdominal aortic aneurysm: a randomized, double-blind, placebo-controlled study. Sci Rep 2022; 12:1839. [PMID: 35115574 PMCID: PMC8813993 DOI: 10.1038/s41598-022-05771-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 01/18/2022] [Indexed: 11/09/2022] Open
Abstract
Mortality remains high after emergency open surgery for a ruptured abdominal aortic aneurysm (RAAA). The aim of the present study was to assess, if intravenous (IV) Interferon (IFN) beta-1a improve survival after surgery by up-regulating Cluster of differentiation (CD73). This is a multi-center phase II double-blind, 2:1 randomized, parallel group comparison of the efficacy and safety of IV IFN beta-1a vs. placebo for the prevention of death after open surgery for an infra-renal RAAA. All study patients presented a confirmed infra-renal RAAA, survived the primary emergency surgery and were treated with IFN beta-1a (10 μg) or matching placebo for 6 days after surgery. Major exclusion criteria included fatal hemorrhagic shock, chronic renal replacement therapy, diagnosed liver cirrhosis, severe congestive heart failure, advanced malignant disease, primary attempt of endovascular aortic repair (EVAR), and per-operative suprarenal clamping over 30 min. Main outcome measure was all-cause mortality at day 30 (D30) from initial emergency aortic reconstruction. The study was pre-maturely stopped due to a reported drug-drug interaction and was left under-powered. Out of 40 randomized patients 38 were included in the outcome analyses (27 IFN beta-1a and 11 placebo). There was no statistically significant difference between treatment groups at baseline except more open-abdomen and intestinal ischemia was present in the IFN beta-1a arm. D30 all-cause mortality was 22.2% (6/27) in the IFN beta-1a arm and 18.2% (2/11) in the placebo arm (OR 1.30; 95% CI 0.21-8.19). The most common adverse event relating to the IFN beta-1a was pyrexia (20.7% in the IFN beta-1a arm vs. 9.1% in the placebo arm). Patients with high level of serum CD73 associated with survival (P = 0.001) whereas the use of glucocorticoids and the presence of IFN beta-1a neutralizing antibodies associated with a poor CD73 response and survival. The initial aim of the trial, if postoperative INF beta-1a treatment results on better RAAA survival, could not be demonstrated. Nonetheless the anticipated target mechanism up-regulation of CD73 was associated with 100% survival. According to present results the INF beta-1a induced up-regulation of serum CD73 was blocked with both use of glucocorticoids and serum IFN beta-1a neutralizing antibodies. The study was pre-maturely stopped due to interim analysis after a study concerning the use if IV IFN beta-1a in ARDS suggested that the concomitant use of glucocorticoids and IFN beta-1a block the CD73 induction. Trial registration: ClinicalTrials.gov NCT03119701. Registered 19/04/2017 (retrospectively registered).
Collapse
Affiliation(s)
- Harri Hakovirta
- Turku University, Kiinanmyllynkatu 4-8, 20520, Turku, Finland. .,Satasairaala, Pori, Finland. .,Department of Vascular Surgery, Turku University Hospital, Turku, Finland.
| | | | - Eija Saimanen
- Department of Surgery, South Karelia Central Hospital, Lappeenranta, Finland
| | - Tiia Kukkonen
- Department of Vascular Surgery, Hospital Nova of Central Finland, Jyvaskyla, Finland
| | - Pekka Romsi
- Department of Vascular Surgery, Oulu University Hospital, Oulu, Finland
| | - Velipekka Suominen
- Department of Vascular Surgery, Tampere University Hospital, Tampere, Finland
| | - Leena Vikatmaa
- Department of Anesthesiology, Intensive Care, and Pain Medicine, Helsinki University Hospital, Helsinki, Finland
| | - Mika Valtonen
- Department of Perioperative Services, Intensive Care and Pain Management, Turku University Hospital, Turku, Finland
| | | | - Maarit Venermo
- Department of Vascular Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | | |
Collapse
|
25
|
Yu YW, Que JQ, Liu S, Huang KY, Qian L, Weng YB, Rong FN, Wang L, Zhou YY, Xue YJ, Ji KT. Sodium-Glucose Co-transporter-2 Inhibitor of Dapagliflozin Attenuates Myocardial Ischemia/Reperfusion Injury by Limiting NLRP3 Inflammasome Activation and Modulating Autophagy. Front Cardiovasc Med 2022; 8:768214. [PMID: 35083298 PMCID: PMC8785320 DOI: 10.3389/fcvm.2021.768214] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 12/14/2021] [Indexed: 12/20/2022] Open
Abstract
Background: The sodium-glucose co-transporter-2 (SGLT-2) inhibitor dapagliflozin improves cardiovascular outcomes in patients with type 2 diabetes in a manner that is partially independent of its hypoglycemic effect. These observations suggest that it may exert a cardioprotective effect by another mechanism. This study explored the effects of dapagliflozin on myocardial ischemia/reperfusion injury in a mouse model. Materials and Methods: For the in vivo I/R studies, mice received 40 mg/kg/d dapagliflozin, starting 7 days before I/R. Evans Blue/TTC double-staining was used to determine the infarct size. Serum levels of cTnI, CK-MB, and LDH were measured. Inflammation, autophagy protein expression, and caspase-1 activity changes were measured at the protein level. Primary cardiomyocytes were used to investigate the direct effect of dapagliflozin on cardiomyocytes and to verify whether they have the same effect as observed in in vivo experiments. Result: A high dose of dapagliflozin significantly reduced infarct size and decreased the serum levels of cTnI, CK-MB, and LDH. Dapagliflozin also reduced serum levels of IL-1β, reduced expression of myocardial inflammation-related proteins, and inhibited cardiac caspase-1 activity. The treatment restored autophagy flux and promoted the degradation of autophagosomes. Relief of inflammation relied on autophagosome phagocytosis of NLRP3 and autophagosome clearance after lysosome improvement. 10 μM dapagliflozin reduced intracellular Ca2+ and Na+ in primary cardiomyocytes, and increasing NHE1 and NCX expression mitigated dapagliflozin effects on autophagy. Conclusion: Dapagliflozin protects against myocardial ischemia/reperfusion injury independently of its hypoglycemic effect. High-dose dapagliflozin pretreatment might limit NLRP3 inflammasome activation and mediate its selective autophagy. Dapagliflozin directly acts on cardiomyocytes through NHE1/NCX.
Collapse
Affiliation(s)
- Yong-Wei Yu
- Department of Cardiology, The Second Affiliated and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China.,Intensive Care Unit, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Jia-Qun Que
- Department of Cardiology, The Second Affiliated and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Shuai Liu
- Department of Cardiology, The Second Affiliated and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Kai-Yu Huang
- Department of Cardiology, The Second Affiliated and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Lu Qian
- Department of Cardiology, The Second Affiliated and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Ying-Bei Weng
- Department of Cardiology, The Second Affiliated and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Fang-Ning Rong
- Department of Cardiology, The Second Affiliated and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Lei Wang
- Department of Cardiology, The Second Affiliated and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Ying-Ying Zhou
- Department of Endocrinology, The Second Affiliated and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yang-Jing Xue
- Department of Cardiology, The Second Affiliated and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Kang-Ting Ji
- Department of Cardiology, The Second Affiliated and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
26
|
Liu T, Howarth AG, Chen Y, Nair AR, Yang HJ, Ren D, Tang R, Sykes J, Kovacs MS, Dey D, Slomka P, Wood JC, Finney R, Zeng M, Prato FS, Francis J, Berman DS, Shah PK, Kumar A, Dharmakumar R. Intramyocardial Hemorrhage and the "Wave Front" of Reperfusion Injury Compromising Myocardial Salvage. J Am Coll Cardiol 2022; 79:35-48. [PMID: 34991787 DOI: 10.1016/j.jacc.2021.10.034] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 10/04/2021] [Accepted: 10/12/2021] [Indexed: 01/29/2023]
Abstract
BACKGROUND Reperfusion therapy for acute myocardial infarction (MI) is lifesaving. However, the benefit of reperfusion therapy can be paradoxically diminished by reperfusion injury, which can increase MI size. OBJECTIVES Hemorrhage is known to occur in reperfused MIs, but whether hemorrhage plays a role in reperfusion-mediated MI expansion is not known. METHODS We studied cardiac troponin kinetics (cTn) of ST-segment elevation MI patients (n = 70) classified by cardiovascular magnetic resonance to be hemorrhagic (70%) or nonhemorrhagic following primary percutaneous coronary intervention. To isolate the effects of hemorrhage from ischemic burden, we performed controlled canine studies (n = 25), and serially followed both cTn and MI size with time-lapse imaging. RESULTS CTn was not different before reperfusion; however, an increase in cTn following primary percutaneous coronary intervention peaked earlier (12 hours vs 24 hours; P < 0.05) and was significantly higher in patients with hemorrhage (P < 0.01). In hemorrhagic animals, reperfusion led to rapid expansion of myocardial necrosis culminating in epicardial involvement, which was not present in nonhemorrhagic cases (P < 0.001). MI size and salvage were not different at 1 hour postreperfusion in animals with and without hemorrhage (P = 0.65). However, within 72 hours of reperfusion, a 4-fold greater loss in salvageable myocardium was evident in hemorrhagic MIs (P < 0.001). This paralleled observations in patients with larger MIs occurring in hemorrhagic cases (P < 0.01). CONCLUSIONS Myocardial hemorrhage is a determinant of MI size. It drives MI expansion after reperfusion and compromises myocardial salvage. This introduces a clinical role of hemorrhage in acute care management, risk assessment, and future therapeutics.
Collapse
Affiliation(s)
- Ting Liu
- Cedars-Sinai Medical Center, Los Angeles, California, USA; Department of Radiology, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Andrew G Howarth
- Cedars-Sinai Medical Center, Los Angeles, California, USA; University of Calgary, Calgary, Alberta, Canada
| | - Yinyin Chen
- Cedars-Sinai Medical Center, Los Angeles, California, USA; Zhongshan Hospital, Fudan University and Shanghai Institute of Medical Imaging, Shanghai, China
| | - Anand R Nair
- Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Hsin-Jung Yang
- Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Daoyuan Ren
- Zhongshan Hospital, Fudan University and Shanghai Institute of Medical Imaging, Shanghai, China
| | - Richard Tang
- Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Jane Sykes
- Lawson Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Michael S Kovacs
- Lawson Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Damini Dey
- Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Piotr Slomka
- Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - John C Wood
- University of Southern California, Los Angeles, California, USA
| | | | - Mengsu Zeng
- Zhongshan Hospital, Fudan University and Shanghai Institute of Medical Imaging, Shanghai, China
| | - Frank S Prato
- Lawson Research Institute, University of Western Ontario, London, Ontario, Canada
| | | | | | | | - Andreas Kumar
- Northern Ontario School of Medicine, Sudbury, Ontario, Canada
| | - Rohan Dharmakumar
- Cedars-Sinai Medical Center, Los Angeles, California, USA; Krannert Cardiovascular Research Center, Indiana University School of Medicine/IU Health Cardiovascular Institute, Indianapolis, Indiana, USA.
| |
Collapse
|
27
|
Tubeimoside I Ameliorates Myocardial Ischemia-Reperfusion Injury through SIRT3-Dependent Regulation of Oxidative Stress and Apoptosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5577019. [PMID: 34795840 PMCID: PMC8595016 DOI: 10.1155/2021/5577019] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 09/13/2021] [Accepted: 10/23/2021] [Indexed: 12/31/2022]
Abstract
Myocardial ischemia-reperfusion injury (MIRI) is a phenomenon that reperfusion leads to irreversible damage to the myocardium and increases mortality in acute myocardial infarction (AMI) patients. There is no effective drug to treat MIRI. Tubeimoside I (TBM) is a triterpenoid saponin purified from Chinese traditional medicine tubeimu. In this study, 4 mg/kg TBM was given to mice intraperitoneally at 15 min after ischemia. And TBM treatment improved postischemic cardiac function, decreased infarct size, diminished lactate dehydrogenase release, ameliorated oxidative stress, and reduced apoptotic index. Notably, ischemia-reperfusion induced a significant decrease in cardiac SIRT3 expression and activity, while TBM treatment upregulated SIRT3's expression and activity. However, the cardioprotective effects of TBM were largely abolished by a SIRT3 inhibitor 3-(1H-1,2,3-triazol-4-yl) pyridine (3-TYP). This suggests that SIRT3 plays an essential role in TBM's cardioprotective effects. In vitro, TBM also protected H9c2 cells against simulated ischemia/reperfusion (SIR) injury by attenuating oxidative stress and apoptosis, and siSIRT3 diminished its protective effects. Taken together, our results demonstrate for the first time that TBM protects against MIRI through SIRT3-dependent regulation of oxidative stress and apoptosis. TBM might be a potential drug candidate for MIRI treatment.
Collapse
|
28
|
Chen X, Xie Q, Zhu Y, Xu J, Lin G, Liu S, Su Z, Lai X, Li Q, Xie J, Yang X. Cardio-protective effect of tetrahydrocurcumin, the primary hydrogenated metabolite of curcumin in vivo and in vitro: Induction of apoptosis and autophagy via PI3K/AKT/mTOR pathways. Eur J Pharmacol 2021; 911:174495. [PMID: 34555398 DOI: 10.1016/j.ejphar.2021.174495] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/14/2021] [Accepted: 09/06/2021] [Indexed: 12/14/2022]
Abstract
Tetrahydrocurcumin (THC) is an essential metabolite of curcumin, a major active component of the Curcuma species, which have been used traditionally for the treatment of cardiovascular diseases. The PI3K/AKT/mTOR signaling pathways serve a vital role during myocardial ischemia-reperfusion (MI/R) injury. The aim of the present study was to investigate the cardioprotective potential and mechanism of THC. In the in vivo study, an animal model of MI/R was induced by coronary occlusion. Results indicated that THC (50 mg/kg/day) protected the rat hearts from MI/R-induced heart failure by increasing ejection fraction (EF) and fractional shortening (FS) and decreasing left ventricular end systolic diameter (LVESD) and left ventricular end systolic volume (LVESV). THC also reduced myocardial infarct size and apoptosis. Furthermore, H9c2 cells were incubated with THC (20 μM) to explore its potential effect following exposure to hypoxia and reoxygenation (H/R). THC post-treatment significantly augmented cell viability and prevented lactate dehydrogenase (LDH) release after H/R exposure. THC effectively improved antioxidant activity by increasing SOD and CAT activities and decreasing MDA level. THC also enhanced mitochondrial membrane potential, inhibited apoptotic cell death, diminished the Bax/Bcl-2 ratio and cleaved caspase-3 level relative to the H/R model. In addition, THC effectively decreased Beclin1 expression and LC3 II/LC3 I ratio, but increased p62 expression, compared with the H/R model group, and decreased the formation of H/R-induced autophagosomes and autolysosomes. Furthermore, THC promoted the phosphorylation of PI3K/AKT/mTOR and induced the expression of hypoxia-inducible factor 1α (HIF-1α) after H/R. However, these effects on H9c2 cells were notably abolished by the PI3K inhibitor LY294002 and mTOR inhibitor rapamycin. In conclusion, THC effectively inhibited H/R-induced autophagy and apoptosis via, at least partially, activating the PI3K/AKT/mTOR pathways. THC might have the potential to be further developed into a potential candidate for the treatment of MI/R injury.
Collapse
Affiliation(s)
- Xiaoying Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, PR China
| | - Qingfeng Xie
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, PR China; The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China
| | - Ying Zhu
- The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, PR China
| | - Jiamin Xu
- The Second School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou, PR China
| | - Guoshu Lin
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, PR China
| | - Shujun Liu
- The Second School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou, PR China
| | - Ziren Su
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, PR China; Dongguan Institute of Guangzhou University of Chinese Medicine, Dongguan, PR China
| | - Xiaoping Lai
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, PR China; Dongguan Institute of Guangzhou University of Chinese Medicine, Dongguan, PR China.
| | - Qian Li
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, PR China
| | - Jianhui Xie
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, PR China
| | - Xiaobo Yang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, PR China.
| |
Collapse
|
29
|
Zhang M, Lei YS, Meng XW, Liu HY, Li LG, Zhang J, Zhang JX, Tao WH, Peng K, Lin J, Ji FH. Iguratimod Alleviates Myocardial Ischemia/Reperfusion Injury Through Inhibiting Inflammatory Response Induced by Cardiac Fibroblast Pyroptosis via COX2/NLRP3 Signaling Pathway. Front Cell Dev Biol 2021; 9:746317. [PMID: 34760889 PMCID: PMC8573346 DOI: 10.3389/fcell.2021.746317] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/27/2021] [Indexed: 12/31/2022] Open
Abstract
Background: NLRP3 inflammasome contributes a lot to sterile inflammatory response and pyroptosis in ischemia/reperfusion (I/R) injury. Cardiac fibroblasts (CFs) are regarded as semi-professional inflammatory cells and they exert an immunomodulatory role in heart. Iguratimod provides a protective role in several human diseases through exerting a powerful anti-inflammatory effect. However, it is still unclear whether iguratimod could alleviate myocardial I/R injury and whether inflammation triggered by NLRP3-related pyroptosis of CFs is involved in this process. Methods: Transcriptomics analysis for GSE160516 dataset was conducted to explore the biological function of differentially expressed genes during myocardial I/R. In vivo, mice underwent ligation of left anterior descending coronary artery for 30 min followed by 24 h reperfusion. In vitro, primary CFs were subjected to hypoxia for 1 h followed by reoxygenation for 3 h (H/R). Iguratimod was used prior to I/R or H/R. Myocardial infarct area, serum level of cardiac troponin I (cTnI), pathology of myocardial tissue, cell viability, lactate dehydrogenase (LDH) release, and the expression levels of mRNA and protein for pyroptosis-related molecules were measured. Immunofluorescence was applied to determine the cellular localization of NLRP3 protein in cardiac tissue. Results: During myocardial I/R, inflammatory response was found to be the most significantly enriched biological process, and nucleotide-binding oligomerization domain (NOD)-like receptor signaling was a crucial pathway in mediating cardiac inflammation. In our experiments, pretreatment with iguratimod significantly ameliorated I/R-induced myocardial injury and H/R-induced pyroptosis of CFs, as evidenced by reduced myocardial infarct area, serum cTnI level, and LDH release in supernatants, as well as improved pathology of cardiac tissue and cell viability. Immunofluorescence analysis showed that NLRP3 was mainly localized in CFs. Moreover, iguratimod inhibited the expression of pro-inflammatory cytokines and pyroptosis-related molecules, including NLRP3, cleaved caspase-1, and GSDMD-N. Conclusion: Our results suggested that inflammatory response mediated by NOD-like receptor signaling is of vital importance in myocardial I/R injury. Iguratimod protected cardiomyocytes through reducing the cascade of inflammation in heart by inhibiting cardiac fibroblast pyroptosis via the COX2/NLRP3 signaling pathway.
Collapse
Affiliation(s)
- Mian Zhang
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Anesthesiology, Soochow University, Suzhou, China
| | - Yi-Shan Lei
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Anesthesiology, Soochow University, Suzhou, China
| | - Xiao-Wen Meng
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Anesthesiology, Soochow University, Suzhou, China
| | - Hua-Yue Liu
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Anesthesiology, Soochow University, Suzhou, China
| | - Lin-Gui Li
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Anesthesiology, Soochow University, Suzhou, China
| | - Jun Zhang
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Anesthesiology, Soochow University, Suzhou, China
| | - Jia-Xin Zhang
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Anesthesiology, Soochow University, Suzhou, China
| | - Wen-Hui Tao
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Anesthesiology, Soochow University, Suzhou, China
| | - Ke Peng
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Anesthesiology, Soochow University, Suzhou, China
| | - Jun Lin
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Fu-Hai Ji
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Anesthesiology, Soochow University, Suzhou, China
| |
Collapse
|
30
|
Wang H, Zheng B, Che K, Han X, Li L, Wang H, Liu Y, Shi J, Sun S. Protective effects of safranal on hypoxia/reoxygenation-induced injury in H9c2 cardiac myoblasts via the PI3K/AKT/GSK3β signaling pathway. Exp Ther Med 2021; 22:1400. [PMID: 34675994 PMCID: PMC8524664 DOI: 10.3892/etm.2021.10836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 09/14/2021] [Indexed: 12/22/2022] Open
Abstract
Safranal (SFR), an active ingredient extracted from saffron, exhibits a protective effect on the cardiovascular system. However, the mechanism of SFR against hypoxia/reoxygenation (H/R)-induced cardiomyocyte injury has previously not been investigated in vitro. The aim of the present study was therefore to observe the protective effects of SFR on H/R-induced cardiomyocyte injury and to explore its mechanisms. A H/R injury model of H9c2 cardiac myoblasts was established by administering 800 µmol/l CoCl2 to H9c2 cells for 24 h and reoxygenating the cells for 4 h to induce hypoxia. H9c2 cardiac myoblasts were pretreated with SFR for 12 h to evaluate the associated protective effects. A Cell Counting Kit-8 assay was used for cell viability detection, and the expression levels of lactate dehydrogenase (LDH), creatine kinase-MB (CK-MB), glutathione peroxidase (GSH-px), catalase (CAT), superoxide dismutase (SOD), malondialdehyde (MDA) and caspase-3, and the intracellular Ca2+ concentration were measured using the corresponding commercial kits. Levels of reactive oxygen species (ROS) in the cells were detected using 2,7-dichlorodihydrofluorescein diacetate. Flow cytometry was used to determine the degree of apoptosis and the level of mitochondrial membrane potential (MMP). Moreover, the expression levels of phosphorylated (p-)PI3K, AKT, p-AKT, glycogen synthase kinase 3β (GSK3β), p-GSK3β, Bcl-2, Bax, caspase-3 and cleaved caspase-3 were measured using western blot analysis. Results of the present study demonstrated that the H9c2 cardiac myoblasts treated with SFR exhibited significantly improved levels of viability and significantly reduced levels of ROS, compared with the H/R group. Furthermore, compared with the H/R group, SFR treatment significantly increased the MMP levels and antioxidant enzyme levels, including CAT, SOD and GSH-px; whereas the levels of CK-MB, LDH, MDA and intracellular Ca2+ concentration were significantly decreased. Moreover, the results of the present study demonstrated that SFR significantly reduced caspase-3, cleaved caspase-3 and Bax protein expression levels, but upregulated the Bcl-2 protein expression levels. SFR also increased the protein expressions of PI3K/AKT/GSK3β. In summary, the results suggested that SFR may exert a protective effect against H/R-induced cardiomyocyte injury, which occurs in connection with the inhibition of oxidative stress and apoptosis via regulation of the PI3K/AKT/GSK3β signaling pathway.
Collapse
Affiliation(s)
- Hefei Wang
- Department of Traditional Chinese Medicine and Medical History Literature, School of Basic Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Bin Zheng
- Department of Traditional Chinese Medicine, School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Kaimeng Che
- Department of Traditional Chinese Medicine and Medical History Literature, School of Basic Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Xue Han
- Department of Traditional Chinese Medicine and Medical History Literature, School of Basic Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Li Li
- Department of Pharmacognosy, School of Pharmacy, Hebei Medical University, Shijiazhuang, Hebei 050200, P.R. China
| | - Hongfang Wang
- Department of Traditional Chinese Medicine, School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Yanshuang Liu
- Department of Diagnostics, Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Jing Shi
- Department of Scientific Research Management, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Shijiang Sun
- Department of Hospital Management and Medical History Literature, Hebei Province Hospital of Chinese Medicine, The First Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| |
Collapse
|
31
|
Qin H, Li S, Liu Z. Protective Effect of Shexiang Baoxin Pill on Myocardial Ischemia/Reperfusion Injury in Patients With STEMI. Front Pharmacol 2021; 12:721011. [PMID: 34603032 PMCID: PMC8479593 DOI: 10.3389/fphar.2021.721011] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 08/25/2021] [Indexed: 01/11/2023] Open
Abstract
Background: There is no definite effect in the treatment of myocardial ischemia/reperfusion (I/R) injury in patients with acute ST-segment elevation myocardial infarction (STEMI). We evaluated the protective effect of Shexiang Baoxin Pill (SBP) on I/R injury in STEMI patients. Methods: STEMI patients were randomly divided into a primary percutaneous coronary intervention (PPCI) group (n = 52) and a PPCI + SBP group (n = 51). The area at risk of infarction (AAR) and final infarct size (FIS) were examined by single-photon emission computed tomography (SPECT). I/R injury was assessed using myocardial salvage (MS) and salvage index (SI) calculated from AAR and FIS. Results: The ST-segment resolution (STR) in the PPCI + SBP group was significantly higher than that in the PPCI group (p = 0.036), and the peak value of high-sensitivity troponin T (hsTNT) was lower than that in the PPCI group (p = 0.048). FIS in the PPCI + SBP group was smaller than that in the PPCI group (p = 0.047). MS (p = 0.023) and SI (p = 0.006) in the PPCI + SBP group were larger than those in the PPCI group. The left ventricular ejection fraction (LVEF) in the PPCI + SBP group was higher than that in the PPCI group (p = 0.049), and N-terminal pro-B type natriuretic peptide (NT-proBNP) level in the PPCI + SBP group was lower than that in the PPCI group (p = 0.048). Conclusions: SBP can alleviate I/R injury (MS and SI), decrease myocardial infarction area (peak value of hsTNT and FIS), and improve myocardial reperfusion (MBG and STR) and cardiac function (LVEF and NT-proBNP).
Collapse
Affiliation(s)
- Haixia Qin
- Ordos Central Hospital, Ordos Clinical Medical College, Inner Mongolia Medical University, Ordos, China
| | - Siyuan Li
- Ordos Central Hospital, Ordos Clinical Medical College, Inner Mongolia Medical University, Ordos, China
| | - Zhenbing Liu
- Ordos Central Hospital, Ordos Clinical Medical College, Inner Mongolia Medical University, Ordos, China
| |
Collapse
|
32
|
Zhang XJ, Liu X, Hu M, Zhao GJ, Sun D, Cheng X, Xiang H, Huang YP, Tian RF, Shen LJ, Ma JP, Wang HP, Tian S, Gan S, Xu H, Liao R, Zou T, Ji YX, Zhang P, Cai J, Wang ZV, Meng G, Xu Q, Wang Y, Ma XL, Liu PP, Huang Z, Zhu L, She ZG, Zhang X, Bai L, Yang H, Lu Z, Li H. Pharmacological inhibition of arachidonate 12-lipoxygenase ameliorates myocardial ischemia-reperfusion injury in multiple species. Cell Metab 2021; 33:2059-2075.e10. [PMID: 34536344 DOI: 10.1016/j.cmet.2021.08.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 04/01/2020] [Accepted: 08/25/2021] [Indexed: 12/18/2022]
Abstract
Myocardial ischemia-reperfusion (MIR) injury is a major cause of adverse outcomes of revascularization after myocardial infarction. To identify the fundamental regulator of reperfusion injury, we performed metabolomics profiling in plasma of individuals before and after revascularization and identified a marked accumulation of arachidonate 12-lipoxygenase (ALOX12)-dependent 12-HETE following revascularization. The potent induction of 12-HETE proceeded by reperfusion was conserved in post-MIR in mice, pigs, and monkeys. While genetic inhibition of Alox12 protected mouse hearts from reperfusion injury and remodeling, Alox12 overexpression exacerbated MIR injury. Remarkably, pharmacological inhibition of ALOX12 significantly reduced cardiac injury in mice, pigs, and monkeys. Unexpectedly, ALOX12 promotes cardiomyocyte injury beyond its enzymatic activity and production of 12-HETE but also by its suppression of AMPK activity via a direct interaction with its upstream kinase TAK1. Taken together, our study demonstrates that ALOX12 is a novel AMPK upstream regulator in the post-MIR heart and that it represents a conserved therapeutic target for the treatment of myocardial reperfusion injury.
Collapse
Affiliation(s)
- Xiao-Jing Zhang
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Xiaolan Liu
- Institute of Model Animal of Wuhan University, Wuhan 430071, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Manli Hu
- Institute of Model Animal of Wuhan University, Wuhan 430071, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Guo-Jun Zhao
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Dating Sun
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Xu Cheng
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Hui Xiang
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Yong-Ping Huang
- Institute of Model Animal of Wuhan University, Wuhan 430071, China; College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Rui-Feng Tian
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Li-Jun Shen
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Jun-Peng Ma
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Hai-Ping Wang
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Song Tian
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Shanyu Gan
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Haibo Xu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Rufang Liao
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Toujun Zou
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Yan-Xiao Ji
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Peng Zhang
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Jingjing Cai
- Department of Cardiology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Zhao V Wang
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Guannan Meng
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China
| | - Qingbo Xu
- Centre for Clinic Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Yibin Wang
- Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Xin-Liang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19004, USA
| | - Peter P Liu
- Division of Cardiology, University of Ottawa Heart Institute, Ottawa, ON K1Y 4W7, Canada
| | - Zan Huang
- College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Lihua Zhu
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Zhi-Gang She
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Xin Zhang
- Gannan Institute of Translational Medicine, Ganzhou 341000, China
| | - Lan Bai
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China.
| | - Hailong Yang
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China.
| | - Zhibing Lu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan 430060, China.
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| |
Collapse
|
33
|
Qi H, Zhang J, Shang Y, Yuan S, Meng C. Argon inhibits reactive oxygen species oxidative stress via the miR-21-mediated PDCD4/PTEN pathway to prevent myocardial ischemia/reperfusion injury. Bioengineered 2021; 12:5529-5539. [PMID: 34506261 PMCID: PMC8806883 DOI: 10.1080/21655979.2021.1965696] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The objective of this study was to explore the effect of argon preconditioning on myocardial ischemia reperfusion (MI/R) injury and its mechanism. Cardiomyocytes H2C9 were pre-treated with 50% argon, and a cell model of oxygen-glucose deprivation (OGD) was established. CCK-8 and cytotoxicity detection kits were used to detect cell viability and lactate dehydrogenase (LDH) release. The miR-21 expression was detected using quantitative real-time polymerase chain reaction. Western blot analysis was performed to detect the expression of programmed cell death protein 4 (PDCD4) and homologous phosphatase and tensin homolog (PTEN) proteins. The levels of inflammatory factors (IL-1β, IL-6, and IL-8) and oxidative stress factors (reactive oxygen species ROS], malondialdehyde [MDA], and superoxide dismutase [SOD]) were measured using an enzyme-linked immunosorbent assay. The effect of argon on cell apoptosis was detected using flow cytometry. Argon increased the proliferation of cardiomyocytes induced by OGD, decreased the release of LDH in cell culture medium, increased miR-21 expression in cells, decreased the expression of miR-21 target proteins PDCD4 and PTEN, decreased the levels of inflammatory factors (interleukin-1β [IL-1β], interleukin-6 [IL-6], and interleukin-8 [IL-8]) and oxidative stress factors (ROS and MDA), increased the SOD content, and decreased the cell apoptosis rate. Our results suggest that argon preconditioning inhibited the PDCD4/PTEN pathway via miR-21, thereby inhibiting ROS oxidative stress and preventing MI/R injury.
Collapse
Affiliation(s)
- Hong Qi
- Department of ICU, Union Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Jiancheng Zhang
- Department of ICU, Union Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - You Shang
- Department of ICU, Union Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Shiying Yuan
- Department of ICU, Union Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Chunqing Meng
- Department of Orthopedic Surgery, Union Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
34
|
Liu Y, Shu JC, Wang MF, Xu ZJ, Yang L, Meng XW, Duan WB, Zhang N, Shao F, Liu RH, Chen LY. Melanoxylonin A-G, neoflavonoids from the heartwood of Dalbergia melanoxylon and their cardioprotective effects. PHYTOCHEMISTRY 2021; 189:112845. [PMID: 34171505 DOI: 10.1016/j.phytochem.2021.112845] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/10/2021] [Accepted: 06/12/2021] [Indexed: 06/13/2023]
Abstract
Seven undescribed neoflavonoids, named melanoxylonins A-G, were isolated from the heartwood of Dalbergia melanoxylon, and all the non-toxic isolates were evaluated for their cardioprotective effect against ischemia/reoxygenation (I/R) injury in H9c2 cells. Of these, melanoxylonin A-D containing the 8-OH group showed better potent cardioprotective effects than the other four congeners. Molecular docking studies confirmed the capacity of melanoxylonin D to interact with the myeloperoxidase (MPO) protein. These results indicated that the potential cardioprotective effects of melanoxylonin D in H9c2 cells with I/R injury may be imparted through suppression of MPO. These results may provide a new medicinal usage of D. melanoxylon.
Collapse
Affiliation(s)
- Yang Liu
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, China
| | - Ji-Cheng Shu
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, China
| | - Meng-Fei Wang
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, China
| | - Zhang-Jun Xu
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, China
| | - Li Yang
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, China
| | - Xiao-Wei Meng
- School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, China
| | - Wen-Bing Duan
- National Pharmaceutical Engineering Center for Solid Preparation of Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, China
| | - Ni Zhang
- School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, China
| | - Feng Shao
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, China
| | - Rong-Hua Liu
- School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, China.
| | - Lan-Ying Chen
- National Pharmaceutical Engineering Center for Solid Preparation of Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, China.
| |
Collapse
|
35
|
Quercetin Protects H9c2 Cardiomyocytes against Oxygen-Glucose Deprivation/Reoxygenation-Induced Oxidative Stress and Mitochondrial Apoptosis by Regulating the ERK1/2/DRP1 Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:7522175. [PMID: 34457029 PMCID: PMC8390138 DOI: 10.1155/2021/7522175] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/11/2021] [Indexed: 01/08/2023]
Abstract
Reperfusion of blood flow during ischemic myocardium resuscitation induces ischemia/reperfusion (I/R) injury. Oxidative stress has been identified as a major cause in this process. Quercetin (QCT) is a member of the flavonoid family that exerts antioxidant effects. The aim of this study was to investigate the preventive effects of QCT on I/R injury and its underlying mechanism. To this end, H9c2 cardiomyocytes were treated with different concentrations of QCT (10, 20, and 40 μM) and subsequently subjected to oxygen-glucose deprivation/reperfusion (OGD/R) administration. The results indicated that OGD/R-induced oxidative stress, apoptosis, and mitochondrial dysfunction in H9c2 cardiomyocytes were aggravated following 40 μM QCT treatment and alleviated following the administration of 10 and 20 μM QCT prior to OGD/R treatment. In addition, OGD/R treatment inactivated ERK1/2 signaling activation. The effect was mitigated using 10 and 20 μM QCT prior to OGD/R treatment. In conclusion, these results suggested that low concentrations of QCT might alleviate I/R injury by suppressing oxidative stress and improving mitochondrial function through the regulation of ERK1/2-DRP1 signaling, providing a potential candidate for I/R injury prevention.
Collapse
|
36
|
Tan H, Song Y, Chen J, Zhang N, Wang Q, Li Q, Gao J, Yang H, Dong Z, Weng X, Wang Z, Sun D, Yakufu W, Pang Z, Huang Z, Ge J. Platelet-Like Fusogenic Liposome-Mediated Targeting Delivery of miR-21 Improves Myocardial Remodeling by Reprogramming Macrophages Post Myocardial Ischemia-Reperfusion Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100787. [PMID: 34137511 PMCID: PMC8336489 DOI: 10.1002/advs.202100787] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/10/2021] [Indexed: 05/25/2023]
Abstract
Inflammatory modulations focusing on macrophage phenotype are promising candidates to promote better cardiac healing post myocardial ischemia-reperfusion (MI/R) injury. However, the peak of monocyte/macrophage recruitment is later than the time when enhanced permeability and retention effect disappears, which greatly increases the difficulty of reprogramming macrophages through systemic administration. Meanwhile, the inability of nanomaterials to release their contents to specific intracellular locations through reasonable cellular internalization pathways is another obstacle to achieving macrophage reprogramming. Here, inspired by the increase in circulating platelet-monocyte aggregates in patients' post-MI/R and the high efficiency of fusogenic liposomes to deliver contents to the cytoplasm of target cells, a platelet-like fusogenic liposome (PLPs) is constructed. Under the coating of PLPs, mesoporous silica nanospheres with a payload of miR-21, an anti-inflammatory agent, can be specifically delivered to inflammatory monocytes in the blood circulation of MI/R induced mice. Then it directly enters the cytoplasm of monocytes through membrane fusion, thereby realizing the reparative reprogramming of the inflamed macrophages derived from it. In vivo administration of the resulting formula can effectively preserve the cardiac function of mice undergone MI/R. Minimal invasiveness and biological safety make this nano-platform a promising approach of immunotherapy.
Collapse
Affiliation(s)
- Haipeng Tan
- Department of CardiologyZhongshan HospitalFudan UniversityShanghai Institute of Cardiovascular Diseases180 Fenglin Road, Xuhui DistrictShanghai20032P. R. China
| | - Ya'nan Song
- Department of CardiologyZhongshan HospitalFudan UniversityShanghai Institute of Cardiovascular Diseases180 Fenglin Road, Xuhui DistrictShanghai20032P. R. China
| | - Jing Chen
- Department of CardiologyZhongshan HospitalFudan UniversityShanghai Institute of Cardiovascular Diseases180 Fenglin Road, Xuhui DistrictShanghai20032P. R. China
| | - Ning Zhang
- Department of CardiologyZhongshan HospitalFudan UniversityShanghai Institute of Cardiovascular Diseases180 Fenglin Road, Xuhui DistrictShanghai20032P. R. China
| | - Qiaozi Wang
- Department of CardiologyZhongshan HospitalFudan UniversityShanghai Institute of Cardiovascular Diseases180 Fenglin Road, Xuhui DistrictShanghai20032P. R. China
| | - Qiyu Li
- Department of CardiologyZhongshan HospitalFudan UniversityShanghai Institute of Cardiovascular Diseases180 Fenglin Road, Xuhui DistrictShanghai20032P. R. China
| | - Jinfeng Gao
- Department of CardiologyZhongshan HospitalFudan UniversityShanghai Institute of Cardiovascular Diseases180 Fenglin Road, Xuhui DistrictShanghai20032P. R. China
| | - Hongbo Yang
- Department of CardiologyZhongshan HospitalFudan UniversityShanghai Institute of Cardiovascular Diseases180 Fenglin Road, Xuhui DistrictShanghai20032P. R. China
| | - Zheng Dong
- Department of CardiologyZhongshan HospitalFudan UniversityShanghai Institute of Cardiovascular Diseases180 Fenglin Road, Xuhui DistrictShanghai20032P. R. China
| | - Xueyi Weng
- Department of CardiologyZhongshan HospitalFudan UniversityShanghai Institute of Cardiovascular Diseases180 Fenglin Road, Xuhui DistrictShanghai20032P. R. China
| | - Zhengmin Wang
- Department of CardiologyZhongshan HospitalFudan UniversityShanghai Institute of Cardiovascular Diseases180 Fenglin Road, Xuhui DistrictShanghai20032P. R. China
| | - Dili Sun
- Department of CardiologyZhongshan HospitalFudan UniversityShanghai Institute of Cardiovascular Diseases180 Fenglin Road, Xuhui DistrictShanghai20032P. R. China
| | - Wusiman Yakufu
- Department of CardiologyZhongshan HospitalFudan UniversityShanghai Institute of Cardiovascular Diseases180 Fenglin Road, Xuhui DistrictShanghai20032P. R. China
| | - Zhiqing Pang
- School of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of Education826 Zhangheng Road, Pudong New AreaShanghai201210P. R. China
| | - Zheyong Huang
- Department of CardiologyZhongshan HospitalFudan UniversityShanghai Institute of Cardiovascular Diseases180 Fenglin Road, Xuhui DistrictShanghai20032P. R. China
| | - Junbo Ge
- Department of CardiologyZhongshan HospitalFudan UniversityShanghai Institute of Cardiovascular Diseases180 Fenglin Road, Xuhui DistrictShanghai20032P. R. China
| |
Collapse
|
37
|
Zhao W, Zhang X, Rong J. SUMOylation as a Therapeutic Target for Myocardial Infarction. Front Cardiovasc Med 2021; 8:701583. [PMID: 34395563 PMCID: PMC8355363 DOI: 10.3389/fcvm.2021.701583] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 07/01/2021] [Indexed: 12/23/2022] Open
Abstract
Myocardial infarction is a prevalent and life-threatening cardiovascular disease. The main goal of existing interventional therapies is to restore coronary reperfusion while few are designed to ameliorate the pathology of heart diseases via targeting the post-translational modifications of those critical proteins. Small ubiquitin-like modifier (SUMO) proteins are recently discovered to form a new type of protein post-translational modifications (PTM), known as SUMOylation. SUMOylation and deSUMOylation are dynamically balanced in the maintenance of various biological processes including cell division, DNA repair, epigenetic transcriptional regulation, and cellular metabolism. Importantly, SUMOylation plays a critical role in the regulation of cardiac functions and the pathology of cardiovascular diseases, especially in heart failure and myocardial infarction. This review summarizes the current understanding on the effects of SUMOylation and SUMOylated proteins in the pathophysiology of myocardial infarction and identifies the potential treatments against myocardial injury via targeting SUMO. Ultimately, this review recommends SUMOylation as a key therapeutic target for treating cardiovascular diseases.
Collapse
Affiliation(s)
- Wei Zhao
- Li Ka Shing Faculty of Medicine, School of Chinese Medicine, University of Hong Kong, Hong Kong, China.,Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiuying Zhang
- Li Ka Shing Faculty of Medicine, School of Chinese Medicine, University of Hong Kong, Hong Kong, China
| | - Jianhui Rong
- Li Ka Shing Faculty of Medicine, School of Chinese Medicine, University of Hong Kong, Hong Kong, China.,Shenzhen Institute of Research and Innovation, The University of Hong Kong, Shenzhen, China
| |
Collapse
|
38
|
Xie M, Cho GW, Kong Y, Li DL, Altamirano F, Luo X, Morales CR, Jiang N, Schiattarella GG, May HI, Medina J, Shelton J, Ferdous A, Gillette TG, Hill JA. Activation of Autophagic Flux Blunts Cardiac Ischemia/Reperfusion Injury. Circ Res 2021; 129:435-450. [PMID: 34111934 PMCID: PMC8317428 DOI: 10.1161/circresaha.120.318601] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 06/09/2021] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Min Xie
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Geoffrey W. Cho
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Yongli Kong
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Dan L. Li
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Francisco Altamirano
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Xiang Luo
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Cyndi R. Morales
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Nan Jiang
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Gabriele G. Schiattarella
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Herman I. May
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Jessica Medina
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - John Shelton
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Anwarul Ferdous
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Thomas G. Gillette
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Joseph A. Hill
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| |
Collapse
|
39
|
Helix B Surface Peptide Protects Cardiomyocytes From Hypoxia/Reoxygenation-induced Autophagy Through the PI3K/Akt Pathway. J Cardiovasc Pharmacol 2021; 76:181-188. [PMID: 32404595 DOI: 10.1097/fjc.0000000000000849] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Helix B surface peptide (HBSP) is a newly discovered tissue-protective erythropoietin derivative that provides benefits after myocardial ischemia/reperfusion. This study explores the cardioprotective effects of HBSP in myocardial cells in response to hypoxia/reoxygenation injury and its potential mechanism. METHODS In this study, rat ventricular (H9c2) cell cultures were established and pretreated with HBSP. H9c2 cardiomyocytes were randomly assigned to the control, H/R, H/R + LY294002 (a PI3K inhibitor), HBSP + H/R, and HBSP + H/R + LY294002 groups. The pretreated cardiomyocytes underwent H/R, and the cardiomyocytes were monitored for viability through a CCK-8 assay, whereas flow cytometry was used to test cell apoptosis. Orgotein Superoxide Dismutase (SOD) and lactate dehydrogenase (LDH) expression were monitored by SOD and LDH kits, respectively. The expression of LC3 autophagosomes was determined by immunocytochemistry. The expression of LC3II/LC3I, p-Mammalian Target of Rapamycin (mTOR) mTOR, mTOR, Beclin 1, p-PI3K, PI3K p-Akt, and Akt was determined by Western blotting. RESULTS HBSP increased cell viability and reduced SOD and LDH production, and it also reduced H/R-induced cell apoptosis. Moreover, the expression of the autophagy-related proteins (LC3II/LC3I) was inhibited by HBSP, whereas the expression of p-PI3K, p-Akt, and p-mTOR was enhanced. However, the PI3K inhibitor (LY294002) notably abolished these effects in H9c2 cells. CONCLUSIONS HBSP inhibits excessive autophagy and apoptosis induced by H/R by activating the PI3K/Akt pathway. HBSP may potentially be a therapeutic intervention for myocardial ischemia/reperfusion injury.
Collapse
|
40
|
Long L, Yu Z, Chen S, Wu J, Liu Y, Peng J, Qu H, Fu C. Pretreatment of Huoxue Jiedu Formula Ameliorates Myocardial Ischaemia/Reperfusion Injury by Decreasing Autophagy via Activation of the PI3K/AKT/mTOR Pathway. Front Pharmacol 2021; 12:608790. [PMID: 33716739 PMCID: PMC7952439 DOI: 10.3389/fphar.2021.608790] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 01/26/2021] [Indexed: 12/31/2022] Open
Abstract
Background: Myocardial ischaemia/reperfusion (I/R) results in myocardial injury via excessive autophagy. Huoxue Jiedu Formula (HJF) has been widely applied in China for the treatment of ischaemic heart disease. However, the mechanisms of HJF are still poorly understood. Thus, the present experiment was designed to observe the effects of HJF on myocardial I/R injury and explore the possible mechanism. Methods: Myocardial injury in rats subjected to myocardial I/R was reflected by nitrotetrazolium blue chloride staining, thioflavin S staining, serum creatine kinase-MB (CK-MB) and cardiac troponin T (cTnT). Autophagy was determined by electron microscopy, laser confocal microscopy, Q-PCR and western blot. The possible pathway was predicted by network pharmacology and validated in vivo and in vitro. Results: Pretreatment of HJF decreased the no-reflow area, infarcted area, serum CK-MB levels and serum cTnT levels in I/R rat model. In addition, pretreatment of HJF decreased autophagy in heart tissues (decrease in Beclin-1 and LC3-II, and increase in Bcl-2, p62 and ratio of LC3-I/LC3-II). In the vivo study, pretreatment of HJF significantly decreased hypoxia/reoxygenation (H/R)-induced autophagy in H9C2 cells. Network pharmacology was applied to predict the possible mechanism by which HJF affects cardiac autophagy, and the PI3K/AKT/mTOR signalling pathway was the most significantly enriched pathway. And experimental studies demonstrated that pretreatment of HJF increased the phosphorylation of AKT and mTOR, and the effects of HJF on autophagy would be offset by PI3K inhibitor LY294002. Conclusion: Pretreatment of HJF ameliorates myocardial I/R injury by decreasing autophagy through activating PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Linzi Long
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zikai Yu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - ShengJun Chen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Jiangyin Tianjiang Pharmaceutical Co., Ltd., Jiangsu, China
| | - Jiarui Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yingying Liu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jun Peng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Hua Qu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Changgeng Fu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
41
|
Beom JH, Kim JH, Seo J, Lee JH, Chung YE, Chung HS, Chung SP, Kim CH, You JS. Targeted temperature management at 33°C or 36℃ induces equivalent myocardial protection by inhibiting HMGB1 release in myocardial ischemia/reperfusion injury. PLoS One 2021; 16:e0246066. [PMID: 33503060 PMCID: PMC7840046 DOI: 10.1371/journal.pone.0246066] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 01/13/2021] [Indexed: 01/03/2023] Open
Abstract
Acute myocardial infarction (AMI) is lethal and causes myocardial necrosis via time-dependent ischemia due to prolonged occlusion of the infarct-related artery. No effective therapy or potential therapeutic targets can prevent myocardial ischemia/reperfusion (I/R) injury. Targeted temperature management (TTM) may reduce peri-infarct regions by inhibiting the extracellular release of high mobility group box-1 (HMGB1) as a primary mediator of the innate immune response. We used a rat left anterior descending (LAD) coronary artery ligation model to determine if TTM at 33°C and 36°C had similar myocardial protective effects. Rats were divided into sham, LAD I/R+37°C normothermia, LAD I/R+33°C TTM, and LAD I/R+36°C TTM groups (n = 5 per group). To verify the cardioprotective effect of TTM by specifically inhibiting HMGB1, rats were assigned to sham, LAD I/R, and LAD I/R after pre-treatment with glycyrrhizin (known as a pharmacological inhibitor of HMGB1) groups (n = 5 per group). Different target temperatures of 33°C and 36°C caused equivalent reductions in infarct volume after myocardial I/R, inhibited the extracellular release of HMGB1 from infarct tissue, and suppressed the expression of inflammatory cytokines from peri-infarct regions. TTM at 33°C and 36°C significantly attenuated the elevation of cardiac troponin, a sensitive and specific marker of heart muscle damage, after injury. Similarly, glycyrrhizin alleviated myocardial damage by suppressing the extracellular release of HMGB1. TTM at 33°C and 36°C had equivalent myocardial protective effects by similar inhibiting HMGB1 release against myocardial I/R injury. This is the first study to suggest that a target core temperature of 36°C is applicable for cardioprotection.
Collapse
Affiliation(s)
- Jin Ho Beom
- Department of Emergency Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ju Hee Kim
- Department of Emergency Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jeho Seo
- Department of Emergency Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jung Ho Lee
- Department of Pharmacology, BK21 PLUS Project for Medical Science, Brain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Pharmacology, Eulji University School of Medicine, Daejeon, Republic of Korea
| | - Yong Eun Chung
- Department of Radiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyun Soo Chung
- Department of Emergency Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sung Phil Chung
- Department of Emergency Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Chul Hoon Kim
- Department of Pharmacology, BK21 PLUS Project for Medical Science, Brain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Je Sung You
- Department of Emergency Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
42
|
Zeng H, Wang L, Zhang J, Pan T, Yu Y, Lu J, Zhou P, Yang H, Li P. Activated PKB/GSK-3 β synergizes with PKC- δ signaling in attenuating myocardial ischemia/reperfusion injury via potentiation of NRF2 activity: Therapeutic efficacy of dihydrotanshinone-I. Acta Pharm Sin B 2021; 11:71-88. [PMID: 33532181 PMCID: PMC7838031 DOI: 10.1016/j.apsb.2020.09.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/30/2020] [Accepted: 08/31/2020] [Indexed: 12/16/2022] Open
Abstract
Disrupted redox status primarily contributes to myocardial ischemia/reperfusion injury (MIRI). NRF2, the endogenous antioxidant regulator, might provide therapeutic benefits. Dihydrotanshinone-I (DT) is an active component in Salvia miltiorrhiza with NRF2 induction potency. This study seeks to validate functional links between NRF2 and cardioprotection of DT and to investigate the molecular mechanism particularly emphasizing on NRF2 cytoplasmic/nuclear translocation. DT potently induced NRF2 nuclear accumulation, ameliorating post-reperfusion injuries via redox alterations. Abrogated cardioprotection in NRF2-deficient mice and cardiomyocytes strongly supports NRF2-dependent cardioprotection of DT. Mechanistically, DT phosphorylated NRF2 at Ser40, rendering its nuclear-import by dissociating from KEAP1 and inhibiting degradation. Importantly, we identified PKC-δ-(Thr505) phosphorylation as primary upstream event triggering NRF2-(Ser40) phosphorylation. Knockdown of PKC-δ dramatically retained NRF2 in cytoplasm, convincing its pivotal role in mediating NRF2 nuclear-import. NRF2 activity was further enhanced by activated PKB/GSK-3β signaling via nuclear-export signal blockage independent of PKC-δ activation. By demonstrating independent modulation of PKC-δ and PKB/GSK-3β/Fyn signaling, we highlight the ability of DT to exploit both nuclear import and export regulation of NRF2 in treating reperfusion injury harboring redox homeostasis alterations. Coactivation of PKC and PKB phenocopied cardioprotection of DT in vitro and in vivo, further supporting the potential applicability of this rationale.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Hua Yang
- Corresponding authors. Tel./fax: +86 25 83271379.
| | - Ping Li
- Corresponding authors. Tel./fax: +86 25 83271379.
| |
Collapse
|
43
|
Pharmacological Modulation of Cardiac Remodeling after Myocardial Infarction. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8815349. [PMID: 33488934 PMCID: PMC7790555 DOI: 10.1155/2020/8815349] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/13/2020] [Accepted: 12/21/2020] [Indexed: 12/14/2022]
Abstract
Cardiac remodeling describes a series of structural and functional changes in the heart after myocardial infarction (MI). Adverse post-MI cardiac remodeling directly jeopardizes the recovery of cardiac functions and the survival rate in MI patients. Several classes of drugs are proven to be useful to reduce the mortality of MI patients. However, it is an ongoing challenge to prevent the adverse effects of cardiac remodeling. The present review aims to identify the pharmacological therapies from the existing clinical drugs for the treatment of adverse post-MI cardiac remodeling. Post-MI cardiac remodeling is a complex process involving ischemia/reperfusion, inflammation, cell death, and deposition of extracellular matrix (ECM). Thus, the present review included two parts: (1) to examine the basic pathophysiology in the cardiovascular system and the molecular basis of cardiac remodeling and (2) to identify the pathological aspects of cardiac remodeling and the potential of the existing pharmacotherapies. Ultimately, the present review highlights drug repositioning as a strategy to discover effective therapies from the existing drugs against post-MI cardiac remodeling.
Collapse
|
44
|
Matveev DV, Kuznetsov MR, Matveev AD, Evteev AV, Fedorov EE. [Reperfusion syndrome: state of the art]. ANGIOLOGII︠A︡ I SOSUDISTAI︠A︡ KHIRURGII︠A︡ = ANGIOLOGY AND VASCULAR SURGERY 2020; 26:176-183. [PMID: 33332321 DOI: 10.33529/angio2020421] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Reperfusion syndrome is a complex series of clinical manifestations resulting from restoration of blood flow to previously ischaemic tissues. It is accompanied by damage to cells, tissues and organs at various levels, followed by the development of multiple organ failure. This review deals with the main pathophysiological mechanisms of the development of reperfusion syndrome in lesions of cardiac, cerebral and lower-limb vessels. Oxidative stress is considered to be the most important marker of ischaemia-reperfusion injury irrespective of the type of tissues affected. Presented herein are the data on contemporary possibilities of influencing various stages and components of the development of reperfusion injury by means of drug therapy, demonstrating that due to the importance of oxidative stress as a key link of reperfusion injury, antioxidant therapy should be the main component of prevention and treatment of reperfusion injury.
Collapse
Affiliation(s)
- D V Matveev
- Department of Surgery, Russian Medical Academy of Continuous Professional Education, RF Ministry of Public Health, Moscow, Russia
| | - M R Kuznetsov
- Institute of Cluster Oncology named after L.L. Levshin, I.M. Sechenov First Moscow Medical University, Moscow, Russia
| | - A D Matveev
- Department of Surgery, Russian Medical Academy of Continuous Professional Education, RF Ministry of Public Health, Moscow, Russia
| | - A V Evteev
- Scientific Company "Flamena", Reutov, Moscow Region, Russia
| | - E E Fedorov
- Surgical Department #1, Municipal Clinical Hospital #29 named after N.E. Bauman, Moscow, Russia
| |
Collapse
|
45
|
An S, Wang X, Shi H, Zhang X, Meng H, Li W, Chen D, Ge J. Apelin protects against ischemia-reperfusion injury in diabetic myocardium via inhibiting apoptosis and oxidative stress through PI3K and p38-MAPK signaling pathways. Aging (Albany NY) 2020; 12:25120-25137. [PMID: 33342766 PMCID: PMC7803490 DOI: 10.18632/aging.104106] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 08/19/2020] [Indexed: 12/12/2022]
Abstract
Among all diabetes mellitus-associated cardiovascular diseases, morbidity of diabetic myocardium with ischemia reperfusion injury (D-IRI) is increasing year by year. We aimed to discover a therapeutic biomarker and investigate its mechanism in D-IRI. High-fat diet and streptozotocin-induced diabetes rats were operated with IRI or sham. Recombined lentiviral vector encoding Apelin was injected into D-IRI rat via tail vein. Cardiac function, infarct size, cellular death and oxidative stress were major outcome measures. Cardiomyocyte ischemia reperfusion injury was more serious in D-IRI rats than in non-diabetes ischemia reperfusion injury (ND-IRI) rats. The secretion of NTproBNP was increased in D-IRI compared with ND-IRI. Bcl-2 expression was decreased, and Bax and cleaved caspase-3 expression was increased in D-IRI rats compared with ND-IRI rats, which were reversed after treatment with Apelin. Apelin-upregulation improved cardiomyocyte ischemia reperfusion injury and decreased NT-proBNP levels in D-IRI rats. Apelin overexpression enhanced PI3K and eNOS levels while reduced those of p38-MAPK and iNOS in D-IRI rats. Apelin overexpression protected against D-IRI through inhibiting apoptosis and oxidative stress via PI3K and p38MAPK signaling pathways in D-IRI rats. These findings provide critical new insight into understanding of Apelin's cardio-protective effects, which may become a novel therapeutic target for the diabetic IRI patients.
Collapse
Affiliation(s)
- Songtao An
- Department of Cardiology, Henan Province People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan, China
| | - Xi Wang
- Department of Cardiology, Henan Province People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan, China
| | - Huairui Shi
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xueqiang Zhang
- Department of Cardiology, Hongxing Hospital, Hami 839000, Xinjiang, China
| | - Hua Meng
- Department of Cardiology, Henan Province People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan, China
| | - Wenbo Li
- Department of Cardiology, Henan Province People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan, China
| | - Dongchang Chen
- Department of Cardiology, Henan Province People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
46
|
Zeng Z, Ma H, Chen J, Huang N, Zhang Y, Su Y, Zhang H. Knockdown of miR-1275 protects against cardiomyocytes injury through promoting neuromedin U type 1 receptor. Cell Cycle 2020; 19:3639-3649. [PMID: 33323026 DOI: 10.1080/15384101.2020.1860310] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The present study aimed to assess the role of miR-1275 in cardiac ischemia reperfusion injury. H9 human embryonic stem cell (hESC)-derived cardiomyocytes stimulated by oxygen-glucose deprivation/reoxygenation (OGD/R) were used to simulate myocardial injury in vitro. miR-1275 expression levels in cells were measured by RT-qPCR. The release of lactate dehydrogenase (LDH) and creatine kinase (CK) was examined through LDH and CK ELISA kits. Cell apoptosis was detected through flow cytometry. A Fura-2 Calcium Flux Assay Kit and a Fluo-4 assay kit were used to determine the Ca2+ concentration. Expression levels of proteins were tested by Western blotting. The binding effect of miR-1275 and neuromedin U type 1 receptor (NMUR1) was detected by dual-luciferase activity assay. The results showed that miR-1275 was upregulated in OGD/R-stimulated cardiomyocytes. Inhibition of miR-1275 suppressed the increased activity of LDH and CK, cell apoptosis, reactive oxygen species (ROS) production, intracellular Ca2+ concentration and sarcoplasmic reticulum (SR) Ca2+ leak induced by OGD/R treatment in cardiomyocytes. miR-1275 directly targets 3'UTR of NMUR1 and negatively regulates NMUR1 expression. Silence of NMUR1 abolished the protecting effect of the miR-1275 antagomir on myocardial OGD/R injury. Our study indicated that the miR-1275 antagomir protects cardiomyocytes from OGD/R injury through the promotion of NMUR1.
Collapse
Affiliation(s)
- Zhu Zeng
- Department of Emergency, The Affiliated Children Hospital of Xi'an Jiaotong University , Xi'an, Shaanxi, China
| | - Haixin Ma
- Medical Department, The Affiliated Children Hospital of Xi'an Jiaotong University , Xi'an, Shaanxi, China
| | - Jing Chen
- Department of Emergency, The Affiliated Children Hospital of Xi'an Jiaotong University , Xi'an, Shaanxi, China
| | - Nina Huang
- Department of Emergency, The Affiliated Children Hospital of Xi'an Jiaotong University , Xi'an, Shaanxi, China
| | - Yudan Zhang
- Department of Emergency, The Affiliated Children Hospital of Xi'an Jiaotong University , Xi'an, Shaanxi, China
| | - Yufei Su
- Department of Emergency, The Affiliated Children Hospital of Xi'an Jiaotong University , Xi'an, Shaanxi, China
| | - Huifang Zhang
- Department of Emergency, The Affiliated Children Hospital of Xi'an Jiaotong University , Xi'an, Shaanxi, China
| |
Collapse
|
47
|
Gong L, Wang X, Pan J, Zhang M, Liu D, Liu M, Li L, An F. The co-treatment of rosuvastatin with dapagliflozin synergistically inhibited apoptosis via activating the PI3K/AKt/mTOR signaling pathway in myocardial ischemia/reperfusion injury rats. Open Med (Wars) 2020; 15:47-57. [PMID: 33385063 PMCID: PMC7754177 DOI: 10.1515/med-2021-0005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 08/10/2020] [Accepted: 11/09/2020] [Indexed: 01/05/2023] Open
Abstract
Objective The purpose of the present study was to evaluate the role of co-treatment of rosuvastatin (RSV) and dapagliflozin (DGZ) preconditioning in myocardium ischemia/reperfusion (I/R) injury and to further investigate the underlying mechanism. Methods Sprague-Dawley (SD) rats (n = 25) were divided into five groups randomly: (1) Sham, (2) I/R, (3) I/R + RSV (10 mg/kg), (4) IR + DGZ (1 mg/kg), and (5) I/R + RSV (10 mg/kg) + DGZ (1 mg/kg). The I/R model was induced with 30 min of left anterior descending occlusion followed by 120 min of reperfusion. Results In vivo pretreatment with RSV and DGZ, respectively, showed a significant reduction of infarction size, a significant increase in the levels of left ventricular systolic pressure, and maximal rate increase in left ventricular pressure (+dp/dtmax), decrease in the levels of left ventricular end-diastolic pressure (LVEDP), maximal rate of decrease of left ventricular pressure (−dp/dtmax) and activity of cardiac enzymes of creatine kinase (CK), creatine kinase MB isoenzymes (CK-MB), and hyper-tensive cardiac troponin I compared with the I/R group. H9C2 cells were exposed to hypoxia/reoxygenation to simulate an I/R model. In vitro administration of 25 µM RSV and 50 µM DGZ significantly enhanced cell viability, upregulated the expression levels of p-PI3K, p-Akt, p-mTOR, and Bcl-2, whereas it downregulated cleaved-caspase3, Bax. TUNEL assay indicated that pretreatment with RSV and DGZ decreased the apoptosis of H9C2 cells. Conclusion The combination of RSV and DGZ significantly enhances the cardioprotective effects compared with RSV or DGZ alone. RSV and DGZ have the potential cardioprotective effects against I/R injury by activating the PI3K/AKt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Lei Gong
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, No. 107 WenHuaXi Road, Jinan, Shandong 250012, China.,The Second Affiliated Hospital of Xuzhou Medical University, No.32 MeiJian Road, Quanshan District, Xuzhou, Jiangsu 221000, China
| | - Xuyang Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, No. 107 WenHuaXi Road, Jinan, Shandong 250012, China
| | - Jinyu Pan
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, No. 107 WenHuaXi Road, Jinan, Shandong 250012, China
| | - Mingjun Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, No. 107 WenHuaXi Road, Jinan, Shandong 250012, China
| | - Dian Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, No. 107 WenHuaXi Road, Jinan, Shandong 250012, China
| | - Ming Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, No. 107 WenHuaXi Road, Jinan, Shandong 250012, China
| | - Li Li
- The Second Affiliated Hospital of Xuzhou Medical University, No.32 MeiJian Road, Quanshan District, Xuzhou, Jiangsu 221000, China
| | - Fengshuang An
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, No. 107 WenHuaXi Road, Jinan, Shandong 250012, China
| |
Collapse
|
48
|
Goodwin ML, Selzman CH. Take a load off: Unloading and reperfusion. J Thorac Cardiovasc Surg 2020; 161:2046-2050. [PMID: 32859423 DOI: 10.1016/j.jtcvs.2020.07.078] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/30/2020] [Accepted: 07/08/2020] [Indexed: 11/16/2022]
Affiliation(s)
- Matthew L Goodwin
- Division of Cardiothoracic Surgery, University of Utah, Salt Lake City, Utah
| | - Craig H Selzman
- Division of Cardiothoracic Surgery, University of Utah, Salt Lake City, Utah.
| |
Collapse
|
49
|
Selective protection of human cardiomyocytes from anthracycline cardiotoxicity by small molecule inhibitors of MAP4K4. Sci Rep 2020; 10:12060. [PMID: 32694738 PMCID: PMC7374628 DOI: 10.1038/s41598-020-68907-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 06/25/2020] [Indexed: 12/11/2022] Open
Abstract
Given the poor track record to date of animal models for creating cardioprotective drugs, human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) have been proposed as a therapeutically relevant human platform to guide target validation and cardiac drug development. Mitogen-Activated Protein Kinase Kinase Kinase Kinase-4 (MAP4K4) is an “upstream” member of the MAPK superfamily that is implicated in human cardiac muscle cell death from oxidative stress, based on gene silencing and pharmacological inhibition in hPSC-CMs. A further role for MAP4K4 was proposed in heart muscle cell death triggered by cardiotoxic anti-cancer drugs, given its reported activation in failing human hearts with doxorubicin (DOX) cardiomyopathy, and its activation acutely by DOX in cultured cardiomyocytes. Here, we report successful protection from DOX in two independent hPSC-CM lines, using two potent, highly selective MAP4K4 inhibitors. The MAP4K4 inhibitors enhanced viability and reduced apoptosis at otherwise lethal concentrations of DOX, and preserved cardiomyocyte function, as measured by spontaneous calcium transients, at sub-maximal ones. Notably, in contrast, no intereference was seen in tumor cell killing, caspase activation, or mitochondrial membrane dissipation by DOX, in human cancer cell lines. Thus, MAP4K4 is a plausible, tractable, selective therapeutic target in DOX-induced human heart muscle cell death.
Collapse
|
50
|
Zeng G, Lian C, Yang P, Zheng M, Ren H, Wang H. E3-ubiquitin ligase TRIM6 aggravates myocardial ischemia/reperfusion injury via promoting STAT1-dependent cardiomyocyte apoptosis. Aging (Albany NY) 2020; 11:3536-3550. [PMID: 31171760 PMCID: PMC6594808 DOI: 10.18632/aging.101995] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 05/22/2019] [Indexed: 12/26/2022]
Abstract
Cardiomyocyte apoptosis is a major cause of myocardial ischemia/reperfusion (MI/R) injury, in which the activation of the signal transducer and activator of transcription 1 (STAT1) plays an important role. The E3-ubiquitin ligase TRIM6 has been implicated in regulating STAT1 activity, however, whether it is associated with MI/R injury and the underlying mechanism are not determined. In this study, by investigating a mouse MI/R injury model, we show that TRIM6 expression is induced in mouse heart following MI/R injury. Additionally, TRIM6 depletion reduces and its overexpression increases myocardial infarct size, serum creatine phosphokinase (CPK) level and cardiomyocyte apoptosis in mice subjected to MI/R injury, indicating that TRIM6 functions to aggravate MI/R injury. Mechanistically, TRIM6 promotes IKKε-dependent STAT1 activation, and the inhibition of IKKε or STAT1 with the specific inhibitor, CAY10576 or fludarabine, abolishes TRIM6 effects on cardiomyocyte apoptosis and MI/R injury. Similarly, TRIM6 mutant lacking the ability to ubiquitinate IKKε and induce IKKε/STAT1 activation also fails to promote cardiomyocyte apoptosis and MI/R injury. Thus, these results suggest that TRIM6 aggravates MI/R injury through promoting IKKε/STAT1 activation-dependent cardiomyocyte apoptosis, and that TRIM6 might represent a novel therapeutic target for alleviating MI/R injury.
Collapse
Affiliation(s)
- Guangwei Zeng
- Department of Cardiology, The Second Affiliated Hospital of Air Force Medical University, Shaanxi, China
| | - Chen Lian
- Department of Cardiology, The Second Affiliated Hospital of Air Force Medical University, Shaanxi, China
| | - Pei Yang
- Department of Cardiology, The Second Affiliated Hospital of Air Force Medical University, Shaanxi, China.,Jiajiang Oil Storage Warehouse, Xining Joint Service Centre, Xining, China
| | - Mingming Zheng
- Department of Health Economic Managment, The Second Affiliated Hospital of Air Force Medical University, Shaanxi, China
| | - He Ren
- Department of Cardiology, The Second Affiliated Hospital of Air Force Medical University, Shaanxi, China
| | - Haiyan Wang
- Department of Cardiology, The Second Affiliated Hospital of Air Force Medical University, Shaanxi, China
| |
Collapse
|