1
|
Jiang L, Xiong W, Yang Y, Qian J. Insight into Cardioprotective Effects and Mechanisms of Dexmedetomidine. Cardiovasc Drugs Ther 2024; 38:1139-1159. [PMID: 38869744 DOI: 10.1007/s10557-024-07579-9] [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] [Accepted: 05/17/2024] [Indexed: 06/14/2024]
Abstract
PURPOSE Cardiovascular disease remains the leading cause of death worldwide. Dexmedetomidine is a highly selective α2 adrenergic receptor agonist with sedative, analgesic, anxiolytic, and sympatholytic properties, and several studies have shown its possible protective effects in cardiac injury. The aim of this review is to further elucidate the underlying cardioprotective mechanisms of dexmedetomidine, thus suggesting its potential in the clinical management of cardiac injury. RESULTS AND CONCLUSION Our review summarizes the findings related to the involvement of dexmedetomidine in cardiac injury and discusses the results in the light of different mechanisms. We found that numerous mechanisms may contribute to the cardioprotective effects of dexmedetomidine, including the regulation of programmed cell death, autophagy and fibrosis, alleviation of inflammatory response, endothelial dysfunction and microcirculatory derangements, improvement of mitochondrial dysregulation, hemodynamics, and arrhythmias. Dexmedetomidine may play a promising and beneficial role in the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Leyu Jiang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Wei Xiong
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuqiao Yang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jinqiao Qian
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, China.
| |
Collapse
|
2
|
Zhang Q, Ruan H, Wang X, Luo A, Ran X. Ulinastatin attenuated cardiac ischaemia/reperfusion injury by suppressing activation of the tissue kallikrein-kinin system. Br J Pharmacol 2024; 181:4988-5008. [PMID: 39294926 DOI: 10.1111/bph.16477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 02/29/2024] [Accepted: 05/13/2024] [Indexed: 09/21/2024] Open
Abstract
BACKGROUND AND PURPOSE Ulinastatin has beneficial effects in patients undergoing coronary artery bypass grafting (CABG) surgery due to its anti-inflammatory properties, but the underlying mechanism remains unclear. EXPERIMENTAL APPROACH We used samples from patients undergoing CABG, a model of cardiac ischaemia-reperfusion injury (IRI) in mice and murine cardiac endothelial cell cultures to investigate links between ulinastatin, the kallikrein-kinin system (KKS), endothelial dysfunction and cardiac inflammation in the response to ischaemia/reperfusion injury (IRI). These links were assessed using clinical investigations, in vitro and in vivo experiments and RNA sequencing analysis. KEY RESULTS Ulinastatin inhibited the activity of tissue kallikrein, a key enzyme of the KKS, at 24 h after CABG surgery, which was verified in our murine cardiac ischaemia-reperfusion model. Under normal conditions, ulinastatin only inhibited kallikrein activity but did not affect bradykinin (B1/B2) receptors. Ulinastatin protected against IRI, in vivo and in vitro, by suppressing activation of the kallikrein-kinin system and down-regulating B1/B2 receptor-related signalling pathways including ERK/ iNOS, which resulted in enhanced endothelial barrier function, mitigation of inflammation and oedema, decreased infarct size, improved cardiac function and decreased mortality. Inhibition of kallikrein and knockdown of B1, but not B2 receptors prevented ERK translocation into the nucleus, reducing reperfusion-induced injury in murine cardiac endothelial cells. CONCLUSIONS AND IMPLICATIONS Treatment with ulinastatin exerts a protective influence on cardiac reperfusion by suppressing activation of the kallikrein-kinin system. Our findings highlight the potential of targeting kallikrein /bradykinin receptors to alleviate endothelial dysfunction, thus improving cardiac IRI.
Collapse
Affiliation(s)
- Qin Zhang
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hang Ruan
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaochuan Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ailin Luo
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao Ran
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
3
|
Guo F, Han X, You Y, Xu SJ, Zhang YH, Chen YY, Xin GJ, Liu ZX, Ren JG, Cao C, Li LM, Fu JH. Hydroxysafflor Yellow A Inhibits Pyroptosis and Protecting HUVECs from OGD/R via NLRP3/Caspase-1/GSDMD Pathway. Chin J Integr Med 2024; 30:1027-1034. [PMID: 38319525 DOI: 10.1007/s11655-023-3716-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/03/2023] [Indexed: 02/07/2024]
Abstract
OBJECTIVE To observe the protective effect and mechanism of hydroxyl safflower yellow A (HSYA) from myocardial ischemia-reperfusion injury on human umbilical vein endothelial cells (HUVECs). METHODS HUVECs were treated with oxygen-glucose deprivation reperfusion (OGD/R) to simulate the ischemia reperfusion model, and cell counting kit-8 was used to detect the protective effect of different concentrations (1.25-160 µ mol/L) of HSYA on HUVECs after OGD/R. HSYA 80 µ mol/L was used for follow-up experiments. The contents of inflammatory cytokines interleukin (IL)-18, IL-1 β, monocyte chemotactic protein 1 (MCP-1), tumor necrosis factor α (TNF-α) and IL-6 before and after administration were measured by enzyme-linked immunosorbent assay. The protein expressions of toll-like receptor, NOD-like receptor containing pyrin domain 3 (NLRP3), gasdermin D (GSDMD) and GSDMD-N-terminal domain (GSDMD-N) before and after administration were detected by Western blot. NLRP3 inflammasome inhibitor cytokine release inhibitory drug 3 sodium salt (CRID3 sodium salt, also known as MCC950) and agonist were added, and the changes of NLRP3, cysteine-aspartic acid protease 1 (Caspase-1), GSDMD and GSDMD-N protein expressions were detected by Western blot. RESULTS HSYA inhibited OGD/R-induced inflammation and significantly decreased the contents of inflammatory cytokines IL-18, IL-1 β, MCP-1, TNF-α and IL-6 (P<0.01 or P<0.05). At the same time, by inhibiting NLRP3/Caspase-1/GSDMD pathway, HSYA can reduce the occurrence of pyroptosis after OGD/R and reduce the expression of NLRP3, Caspase-1, GSDMD and GSDMD-N proteins (P<0.01). CONCLUSIONS The protective effect of HSYA on HUVECs after OGD/R is related to down-regulating the expression of NLRP3 inflammasome and inhibiting pyroptosis.
Collapse
Affiliation(s)
- Fan Guo
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- Beijing Key Laboratory of Chinese Materia Pharmacology, Beijing, 100091, China
| | - Xiao Han
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- Beijing Key Laboratory of Chinese Materia Pharmacology, Beijing, 100091, China
| | - Yue You
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- Beijing Key Laboratory of Chinese Materia Pharmacology, Beijing, 100091, China
| | - Shu-Juan Xu
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- Beijing Key Laboratory of Chinese Materia Pharmacology, Beijing, 100091, China
| | - Ye-Hao Zhang
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- Beijing Key Laboratory of Chinese Materia Pharmacology, Beijing, 100091, China
| | - Yuan-Yuan Chen
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- Beijing Key Laboratory of Chinese Materia Pharmacology, Beijing, 100091, China
| | - Gao-Jie Xin
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- Beijing Key Laboratory of Chinese Materia Pharmacology, Beijing, 100091, China
| | - Zi-Xin Liu
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- Beijing Key Laboratory of Chinese Materia Pharmacology, Beijing, 100091, China
| | - Jun-Guo Ren
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- Beijing Key Laboratory of Chinese Materia Pharmacology, Beijing, 100091, China
| | - Ce Cao
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- Beijing Key Laboratory of Chinese Materia Pharmacology, Beijing, 100091, China
| | - Ling-Mei Li
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China.
- Beijing Key Laboratory of Chinese Materia Pharmacology, Beijing, 100091, China.
- Department of Central Laboratory, Kunshan Hospital of Chinese Medicine, Kunshan, Jiangsu Province, 215300, China.
| | - Jian-Hua Fu
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- Beijing Key Laboratory of Chinese Materia Pharmacology, Beijing, 100091, China
| |
Collapse
|
4
|
Mylonas N, Nikolaou PE, Karakasis P, Stachteas P, Fragakis N, Andreadou I. Endothelial Protection by Sodium-Glucose Cotransporter 2 Inhibitors: A Literature Review of In Vitro and In Vivo Studies. Int J Mol Sci 2024; 25:7274. [PMID: 39000380 PMCID: PMC11242615 DOI: 10.3390/ijms25137274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/26/2024] [Accepted: 06/30/2024] [Indexed: 07/16/2024] Open
Abstract
Endothelial dysfunction often precedes the development of cardiovascular diseases, including heart failure. The cardioprotective benefits of sodium-glucose cotransporter 2 inhibitors (SGLT2is) could be explained by their favorable impact on the endothelium. In this review, we summarize the current knowledge on the direct in vitro effects of SGLT2is on endothelial cells, as well as the systematic observations in preclinical models. Four putative mechanisms are explored: oxidative stress, nitric oxide (NO)-mediated pathways, inflammation, and endothelial cell survival and proliferation. Both in vitro and in vivo studies suggest that SGLT2is share a class effect on attenuating reactive oxygen species (ROS) and on enhancing the NO bioavailability by increasing endothelial nitric oxide synthase activity and by reducing NO scavenging by ROS. Moreover, SGLT2is significantly suppress inflammation by preventing endothelial expression of adhesion receptors and pro-inflammatory chemokines in vivo, indicating another class effect for endothelial protection. However, in vitro studies have not consistently shown regulation of adhesion molecule expression by SGLT2is. While SGLT2is improve endothelial cell survival under cell death-inducing stimuli, their impact on angiogenesis remains uncertain. Further experimental studies are required to accurately determine the interplay among these mechanisms in various cardiovascular complications, including heart failure and acute myocardial infarction.
Collapse
Affiliation(s)
- Nikolaos Mylonas
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupolis, Zografou, 15771 Athens, Greece; (N.M.); (P.E.N.)
| | - Panagiota Efstathia Nikolaou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupolis, Zografou, 15771 Athens, Greece; (N.M.); (P.E.N.)
| | - Paschalis Karakasis
- Second Department of Cardiology, Aristotle University of Thessaloniki, Hippokration General Hospital of Thessaloniki, 54642 Thessaloniki, Greece; (P.K.); (P.S.); (N.F.)
| | - Panagiotis Stachteas
- Second Department of Cardiology, Aristotle University of Thessaloniki, Hippokration General Hospital of Thessaloniki, 54642 Thessaloniki, Greece; (P.K.); (P.S.); (N.F.)
| | - Nikolaos Fragakis
- Second Department of Cardiology, Aristotle University of Thessaloniki, Hippokration General Hospital of Thessaloniki, 54642 Thessaloniki, Greece; (P.K.); (P.S.); (N.F.)
- Outpatient Department of Cardiometabolic Medicine, Second Department of Cardiology, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupolis, Zografou, 15771 Athens, Greece; (N.M.); (P.E.N.)
| |
Collapse
|
5
|
Zaruba MM, Staggl S, Ghadge SK, Maurer T, Gavranovic-Novakovic J, Jeyakumar V, Schönherr P, Wimmer A, Pölzl G, Bauer A, Messner M. Roxadustat Attenuates Adverse Remodeling Following Myocardial Infarction in Mice. Cells 2024; 13:1074. [PMID: 38994928 PMCID: PMC11240812 DOI: 10.3390/cells13131074] [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: 03/26/2024] [Revised: 06/07/2024] [Accepted: 06/12/2024] [Indexed: 07/13/2024] Open
Abstract
Activation of the CXCL12/CXCR4/ACKR3 axis is known to aid myocardial repair through ischemia-triggered hypoxia-inducible factor-1α (HIF-1α). To enhance the upregulation of HIF-1α, we administered roxadustat, a novel prolyl hydroxylase inhibitor (PHI) clinically approved by the European Medicines Agency 2021 for the treatment of renal anemia, with the purpose of improving LV function and attenuating ischemic cardiomyopathy. METHODS We evaluated roxadustat's impact on HIF-1 stimulation, cardiac remodeling, and function after MI. Therefore, we analyzed nuclear HIF-1 expression, the mRNA and protein expression of key HIF-1 target genes (RT-PCR, Western blot), inflammatory cell infiltration (immunohistochemistry), and apoptosis (TUNEL staining) 7 days after MI. Additionally, we performed echocardiography in male and female C57BL/6 mice 28 days post-MI. RESULTS We found a substantial increase in nuclear HIF-1, associated with an upregulation of HIF-1α target genes like CXCL12/CXCR4/ACKR3 at the mRNA and protein levels. Roxadustat increased the proportion of myocardial reparative M2 CD206+ cells, suggesting beneficial alterations in immune cell migration and a trend towards reduced apoptosis. Echocardiography showed that roxadustat treatment significantly preserved ejection fraction and attenuated subsequent ventricular dilatation, thereby reducing adverse remodeling. CONCLUSIONS Our findings suggest that roxadustat is a promising clinically approved treatment option to preserve myocardial function by attenuating adverse remodeling.
Collapse
Affiliation(s)
- Marc-Michael Zaruba
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria; (M.-M.Z.); (S.S.); (S.K.G.); (T.M.); (J.G.-N.); (P.S.); (G.P.); (A.B.)
| | - Simon Staggl
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria; (M.-M.Z.); (S.S.); (S.K.G.); (T.M.); (J.G.-N.); (P.S.); (G.P.); (A.B.)
| | - Santhosh Kumar Ghadge
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria; (M.-M.Z.); (S.S.); (S.K.G.); (T.M.); (J.G.-N.); (P.S.); (G.P.); (A.B.)
- Valneva Austria GmbH, Campus Vienna Biocenter 3, 1030 Vienna, Austria
| | - Thomas Maurer
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria; (M.-M.Z.); (S.S.); (S.K.G.); (T.M.); (J.G.-N.); (P.S.); (G.P.); (A.B.)
| | - Jasmina Gavranovic-Novakovic
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria; (M.-M.Z.); (S.S.); (S.K.G.); (T.M.); (J.G.-N.); (P.S.); (G.P.); (A.B.)
| | - Vivek Jeyakumar
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria; (M.-M.Z.); (S.S.); (S.K.G.); (T.M.); (J.G.-N.); (P.S.); (G.P.); (A.B.)
| | - Patric Schönherr
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria; (M.-M.Z.); (S.S.); (S.K.G.); (T.M.); (J.G.-N.); (P.S.); (G.P.); (A.B.)
| | - Andreas Wimmer
- Department of Surgery, Kardinal Schwarzenberg Klinikum GmbH, 5620 Salzburg, Austria;
| | - Gerhard Pölzl
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria; (M.-M.Z.); (S.S.); (S.K.G.); (T.M.); (J.G.-N.); (P.S.); (G.P.); (A.B.)
| | - Axel Bauer
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria; (M.-M.Z.); (S.S.); (S.K.G.); (T.M.); (J.G.-N.); (P.S.); (G.P.); (A.B.)
| | - Moritz Messner
- Department of Internal Medicine III, Cardiology and Angiology, Medical University Innsbruck, 6020 Innsbruck, Austria; (M.-M.Z.); (S.S.); (S.K.G.); (T.M.); (J.G.-N.); (P.S.); (G.P.); (A.B.)
| |
Collapse
|
6
|
Kumphune S, Seenak P, Paiyabhrom N, Songjang W, Pankhong P, Jumroon N, Thaisakun S, Phaonakrop N, Roytrakul S, Malakul W, Jiraviriyakul A, Nernpermpisooth N. Cardiac endothelial ischemia/reperfusion injury-derived protein damage-associated molecular patterns disrupt the integrity of the endothelial barrier. Heliyon 2024; 10:e24600. [PMID: 38312663 PMCID: PMC10835233 DOI: 10.1016/j.heliyon.2024.e24600] [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: 09/05/2023] [Revised: 01/05/2024] [Accepted: 01/10/2024] [Indexed: 02/06/2024] Open
Abstract
Human cardiac microvascular endothelial cells (HCMECs) are sensitive to ischemia and vulnerable to damage during reperfusion. The release of damage-associated molecular patterns (DAMPs) during reperfusion induces additional tissue damage. The current study aimed to identify early protein DAMPs in human cardiac microvascular endothelial cells subjected to ischemia-reperfusion injury (IRI) using a proteomic approach and their effect on endothelial cell injury. HCMECs were subjected to 60 min of simulated ischemia and 6 h of reperfusion, which can cause lethal damage. DAMPs in the culture media were subjected to liquid chromatography-tandem mass spectrometry proteomic analysis. The cells were treated with endothelial IRI-derived DAMP medium for 24 h. Endothelial injury was assessed by measuring lactate dehydrogenase activity, morphological features, and the expression of endothelial cadherin, nitric oxide synthase (eNOS), and caveolin-1. The top two upregulated proteins, DNAJ homolog subfamily B member 11 and pyrroline-5-carboxylate reductase 2, are promising and sensitive predictors of cardiac microvascular endothelial damage. HCMECs expose to endothelial IRI-derived DAMP, the lactate dehydrogenase activity was significantly increased compared with the control group (10.15 ± 1.03 vs 17.67 ± 1.19, respectively). Following treatment with endothelial IRI-derived DAMPs, actin-filament dysregulation, and downregulation of vascular endothelial cadherin, caveolin-1, and eNOS expressions were observed, along with cell death. In conclusion, the early protein DAMPs released during cardiac microvascular endothelial IRI could serve as novel candidate biomarkers for acute myocardial IRI. Distinct features of impaired plasma membrane integrity can help identify therapeutic targets to mitigate the detrimental consequences mediated of endothelial IRI-derived DAMPs.
Collapse
Affiliation(s)
- Sarawut Kumphune
- Biomedical Engineering and Innovation Research Centre, Chiang Mai University, Muang, Chiang Mai, 50200, Thailand
- Biomedical Engineering Institute, Chiang Mai University, Muang, Chiang Mai, 50200, Thailand
- Integrative Biomedical Research Unit (IBRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand
| | - Porrnthanate Seenak
- Integrative Biomedical Research Unit (IBRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand
- Department of Cardio-Thoracic Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand
| | - Nitchawat Paiyabhrom
- Department of Medical Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand
| | - Worawat Songjang
- Integrative Biomedical Research Unit (IBRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand
- Department of Medical Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand
| | - Panyupa Pankhong
- Integrative Biomedical Research Unit (IBRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand
- Department of Medical Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand
| | - Noppadon Jumroon
- Integrative Biomedical Research Unit (IBRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand
- Department of Medical Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand
| | - Siriwan Thaisakun
- National Centre for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani, 12120, Thailand
| | - Narumon Phaonakrop
- National Centre for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani, 12120, Thailand
| | - Sittiruk Roytrakul
- National Centre for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani, 12120, Thailand
| | - Wachirawadee Malakul
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok, 65000, Thailand
| | - Arunya Jiraviriyakul
- Integrative Biomedical Research Unit (IBRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand
- Department of Medical Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand
| | - Nitirut Nernpermpisooth
- Integrative Biomedical Research Unit (IBRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand
- Department of Cardio-Thoracic Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand
| |
Collapse
|
7
|
Jiang Y, Cai Y, Han R, Xu Y, Xia Z, Xia W. Salvianolic acids and its potential for cardio-protection against myocardial ischemic reperfusion injury in diabetes. Front Endocrinol (Lausanne) 2024; 14:1322474. [PMID: 38283744 PMCID: PMC10811029 DOI: 10.3389/fendo.2023.1322474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 12/21/2023] [Indexed: 01/30/2024] Open
Abstract
The incidence of diabetes and related mortality rate increase yearly in modern cities. Additionally, elevated glucose levels can result in an increase of reactive oxygen species (ROS), ferroptosis, and the disruption of protective pathways in the heart. These factors collectively heighten the vulnerability of diabetic individuals to myocardial ischemia. Reperfusion therapies have been effectively used in clinical practice. There are limitations to the current clinical methods used to treat myocardial ischemia-reperfusion injury. As a result, reducing post-treatment ischemia/reperfusion injury remains a challenge. Therefore, efforts are underway to provide more efficient therapy. Salvia miltiorrhiza Bunge (Danshen) has been used for centuries in ancient China to treat cardiovascular diseases (CVD) with rare side effects. Salvianolic acid is a water-soluble phenolic compound with potent antioxidant properties and has the greatest hydrophilic property in Danshen. It has recently been discovered that salvianolic acids A (SAA) and B (SAB) are capable of inhibiting apoptosis by targeting the JNK/Akt pathway and the NF-κB pathway, respectively. This review delves into the most recent discoveries regarding the therapeutic and cardioprotective benefits of salvianolic acid for individuals with diabetes. Salvianolic acid shows great potential in myocardial protection in diabetes mellitus. A thorough understanding of the protective mechanism of salvianolic acid could expand its potential uses in developing medicines for treating diabetes mellitus related myocardial ischemia-reperfusion.
Collapse
Affiliation(s)
- Yuxin Jiang
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Yin Cai
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Ronghui Han
- Faculty of Chinese Medicine State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao SAR, China
| | - Youhua Xu
- Faculty of Chinese Medicine State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao SAR, China
| | - Zhengyuan Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Doctoral Training Platform for Research and Translation, BoShiWan, GuanChong Village, Shuanghe Town, ZhongXiang City, Hubei, China
| | - Weiyi Xia
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
- Doctoral Training Platform for Research and Translation, BoShiWan, GuanChong Village, Shuanghe Town, ZhongXiang City, Hubei, China
| |
Collapse
|
8
|
Luan Y, Luan Y, Jiao Y, Liu H, Huang Z, Feng Q, Pei J, Yang Y, Ren K. Broadening Horizons: Exploring mtDAMPs as a Mechanism and Potential Intervention Target in Cardiovascular Diseases. Aging Dis 2023; 15:2395-2416. [PMID: 38270118 PMCID: PMC11567272 DOI: 10.14336/ad.2023.1130] [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: 09/06/2023] [Accepted: 11/30/2023] [Indexed: 01/26/2024] Open
Abstract
Cardiovascular diseases (CVDs) have been recognized as the leading cause of premature mortality and morbidity worldwide despite significant advances in therapeutics. Inflammation is a key factor in CVD progression. Once stress stimulates cells, they release cellular compartments known as damage-associated molecular patterns (DAMPs). Mitochondria can release mitochondrial DAMPs (mtDAMPs) to initiate an immune response when stimulated with cellular stress. Investigating the molecular mechanisms underlying the DAMPs that regulate CVD progression is crucial for improving CVDs. Herein, we discuss the composition and mechanism of DAMPs, the significance of mtDAMPs in cellular inflammation, the presence of mtDAMPs in different types of cells, and the main signaling pathways associated with mtDAMPs. Based on this, we determined the role of DAMPs in CVDs and the effects of mtDAMP intervention on CVD progression. By offering a fresh perspective and comprehensive insights into the molecular mechanisms of DAMPs, this review seeks to provide important theoretical foundations for developing drugs targeting CVDs.
Collapse
Affiliation(s)
- Yi Luan
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Ying Luan
- State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing, China.
| | - Yuxue Jiao
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Hui Liu
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China.
| | - Zhen Huang
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China.
| | - Qi Feng
- Department ofIntegrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Jinyan Pei
- Quality Management Department, The Third People’s Hospital of Henan Provine, Zhengzhou, China.
| | - Yang Yang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Kaidi Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
9
|
Bumroongthai K, Kavanagh DPJ, Genever P, Kalia N. Improving vasculoprotective effects of MSCs in coronary microvessels - benefits of 3D culture, sub-populations and heparin. Front Immunol 2023; 14:1257497. [PMID: 37954606 PMCID: PMC10635425 DOI: 10.3389/fimmu.2023.1257497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 10/02/2023] [Indexed: 11/14/2023] Open
Abstract
Introduction Opening occluded coronary arteries in patients with myocardial infarction (MI) damages the delicate coronary microvessels through a process called myocardial ischaemia-reperfusion injury. Although mesenchymal stromal cells (MSCs) have the potential to limit this injury, clinical success remains limited. This may be due to (i) poor MSC homing to the heart (ii) infused MSCs, even if derived from the same site, being a heterogeneous population with varying therapeutic efficacy and (iii) conventional 2D culture of MSCs decreasing their homing and beneficial properties. This study investigated whether 3D culture of two distinctly different bone marrow (BM)-derived MSC sub-populations could improve their homing and coronary vasculoprotective efficacy. Methods Intravital imaging of the anaesthetised mouse beating heart was used to investigate the trafficking and microvascular protective effects of two clonally-derived BM-derived MSC lines, namely CD317neg MSCs-Y201 and CD317pos MSCs-Y202, cultured using conventional monolayer and 3D hanging drop methods. Results 3D culture consistently improved the adhesive behaviour of MSCs-Y201 to various substrates in vitro. However, it was their differential ability to reduce neutrophil events within the coronary capillaries and improve ventricular perfusion in vivo that was most remarkable. Moreover, dual therapy combined with heparin further improved the vasculoprotection afforded by 3D cultured MSCs-Y201 by also modifying platelet as well as neutrophil recruitment, which subsequently led to the greatest salvage of viable myocardium. Therapeutic benefit could mechanistically be explained by reductions in coronary endothelial oxidative stress and intercellular adhesion molecule-1 (ICAM-1)/vascular cell adhesion molecule-1 (VCAM-1) expression. However, since this was noted by both 2D and 3D cultured MSCs-Y201, therapeutic benefit is likely explained by the fact that 3D cultured MSCs-Y201 were the most potent sub-population at reducing serum levels of several pro-inflammatory cytokines. Conclusion This novel study highlights the importance of not only 3D culture, but also of a specific CD317neg MSC sub-population, as being critical to realising their full coronary vasculoprotective potential in the injured heart. Since the smallest coronary blood vessels are increasingly recognised as a primary target of reperfusion injury, therapeutic interventions must be able to protect these delicate structures from inflammatory cells and maintain perfusion in the heart. We propose that relatively feasible technical modifications in a specific BM-derived MSC sub-population could achieve this.
Collapse
Affiliation(s)
- Kobkaew Bumroongthai
- Microcirculation Research Group, Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Dean P. J. Kavanagh
- Microcirculation Research Group, Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Paul Genever
- Department of Biology, University of York, York, United Kingdom
| | - Neena Kalia
- Microcirculation Research Group, Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
10
|
Haj-Yehia E, Korste S, Jochem R, Lusha A, Roth A, Dietzel N, Niroomand J, Stock P, Westendorf AM, Buer J, Hendgen-Cotta UB, Rassaf T, Totzeck M. CD47 blockade enhances phagocytosis of cardiac cell debris by neutrophils. IJC HEART & VASCULATURE 2023; 48:101269. [PMID: 37731517 PMCID: PMC10507185 DOI: 10.1016/j.ijcha.2023.101269] [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: 08/04/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 09/22/2023]
Abstract
CD47 is a cell surface protein controlling phagocytotic activity of innate immune cells. CD47 blockade was investigated as an immune checkpoint therapy in cancer treatment, enhancing phagocytosis of tumor cells by macrophages. Anti-CD47 treatment also reduced injury size during reperfused acute myocardial infarction (repAMI) by enhancing phagocytotic acitivity of macrophages. Little is known about the impact of CD47 blockade on neutrophils, representing the main portion of early infiltrating immune cells after repAMI. Therefore, we performed 45 min of cardiac ischemia followed by 24 h of reperfusion, observing a decreased cardiac injury size measured by triphenyl tetrazolium chloride (TTC) Evan's blue staining. We were able to detect this effect with an innovative three-dimensional method based on light sheet fluorescence microscopy (LSFM). This further allowed us a simultaneous analysis of neutrophil infiltration, showing an unaltered amount of injury-associated neutrophils with reduced cardiac injury volume from repAMI. This observation suggests modulated phagocytosis of cell debris by neutrophils. Therefore, we performed flow cytometry analysis, revealing an increased phagocytotic activity of neutrophils in vitro. These findings highlight that CD47 blockade also enhances phagocytosis of cardiac cell debris by neutrophils, which might be an additional protective effect of anti-CD47 treatment after repAMI.
Collapse
Affiliation(s)
- Elias Haj-Yehia
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, 45147 Essen, Germany
| | - Sebastian Korste
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, 45147 Essen, Germany
| | - Robert Jochem
- Department of Nephrology, University Hospital Essen, 45147 Essen, Germany
| | - Aldona Lusha
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, 45147 Essen, Germany
| | - Anna Roth
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, 45147 Essen, Germany
| | - Nina Dietzel
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, 45147 Essen, Germany
| | - Josefine Niroomand
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, 45147 Essen, Germany
| | - Pia Stock
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, 45147 Essen, Germany
| | - Astrid M. Westendorf
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Jan Buer
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ulrike B. Hendgen-Cotta
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, 45147 Essen, Germany
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, 45147 Essen, Germany
| | - Matthias Totzeck
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, 45147 Essen, Germany
| |
Collapse
|
11
|
Yassaghi Y, Jeddi S, Yousefzadeh N, Kashfi K, Ghasemi A. Long-term inorganic nitrate administration protects against myocardial ischemia-reperfusion injury in female rats. BMC Cardiovasc Disord 2023; 23:411. [PMID: 37605135 PMCID: PMC10441752 DOI: 10.1186/s12872-023-03425-2] [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: 02/13/2023] [Accepted: 08/01/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND The favorable effects of nitrate against myocardial ischemia-reperfusion injury (MIRI) have primarily focused on male rats and in short term. Here we determine the impact of long-term nitrate intervention on baseline cardiac function and the resistance to MIRI in female rats. METHODS Female Wistar rats were randomly divided into untreated and nitrate-treated (100 mg/L sodium nitrate in drinking water for 9 months) groups (n = 14/group). At intervention end, levels of serum progesterone, nitric oxide metabolites (NOx), heart NOx concentration, and mRNA expressions of NO synthase isoforms (NOS), i.e., endothelial (eNOS), neuronal (nNOS), and inducible (iNOS), were measured. Isolated hearts were exposed to ischemia, and cardiac function indices (CFI) recorded. When the ischemia-reperfusion (IR) period ended, infarct size, NO metabolites, eNOS, nNOS, and iNOS expression were measured. RESULTS Nitrate-treated rats had higher serum progesterone (29.8%, P = 0.013), NOx (31.6%, P = 0.035), and higher heart NOx (60.2%, P = 0.067), nitrite (131%, P = 0.018), and eNOS expression (200%, P = 0.005). Nitrate had no significant effects on baseline CFI but it increased recovery of left ventricular developed pressure (LVDP, 19%, P = 0.020), peak rate of positive (+ dp/dt, 16%, P = 0.006) and negative (-dp/dt, 14%, P = 0.014) changes in left ventricular pressure and decreased left ventricular end-diastolic pressure (LVEDP, 17%, P < 0.001) and infarct size (34%, P < 0.001). After the IR, the two groups had significantly different heart nitrite, nitrate, NOx, and eNOS and iNOS mRNA expressions. CONCLUSIONS Long-term nitrate intervention increased the resistance to MIRI in female rats; this was associated with increased heart eNOS expression and circulating progesterone before ischemia and blunting ischemia-induced increased iNOS and decreased eNOS after MIRI.
Collapse
Affiliation(s)
- Younes Yassaghi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, No. 24, Parvaneh Street, Yaman Street, P.O. Box: 19395-4763, Velenjak, Tehran, Iran
| | - Sajad Jeddi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, No. 24, Parvaneh Street, Yaman Street, P.O. Box: 19395-4763, Velenjak, Tehran, Iran
| | - Nasibeh Yousefzadeh
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, No. 24, Parvaneh Street, Yaman Street, P.O. Box: 19395-4763, Velenjak, Tehran, Iran
| | - Khosrow Kashfi
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, USA
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, No. 24, Parvaneh Street, Yaman Street, P.O. Box: 19395-4763, Velenjak, Tehran, Iran.
| |
Collapse
|
12
|
Handley E, Callanan A. Effects of electrospun fibers containing ascorbic acid on oxidative stress reduction for cardiac tissue engineering. J Appl Polym Sci 2023; 140:e54242. [PMID: 38439767 PMCID: PMC10909520 DOI: 10.1002/app.54242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/17/2023] [Accepted: 05/15/2023] [Indexed: 03/06/2024]
Abstract
Tissue engineering provides promise for regeneration of cardiac tissue following myocardial infarction. However, the harsh microenvironment of the infarct hampers the efficacy of regenerative therapies. Ischemia-reperfusion injury dramatically increases the levels of reactive oxygen species (ROS) within the infarcted area, causing a cascade of further cellular injury. Implantable tissue engineered grafts can target this oxidative stress by delivering pharmaceutical compounds directly into the diseased tissue. Herein, we successfully fabricated electrospun polycaprolactone (PCL) fibers containing varying concentrations of ascorbic acid, a potent antioxidant well known for its ROS-scavenging capabilities. The antioxidant scaffolds displayed significantly improved scavenging of DPPH radicals, superoxide anions and hydroxyl radicals, in a dose dependent manner. Mechanical properties testing indicated that incorporation of ascorbic acid enhanced the strength and Young's modulus of the material, correlating with a moderate but non-significant increase in the crystallinity. Moreover, the scaffolds supported adhesion and maintained survival of human umbilical vein endothelial cells in vitro, indicating good cytocompatibility. These results provide motivation for the use of ascorbic acid-containing fibrous scaffolds to regulate the highly oxidative microenvironment following myocardial infarction.
Collapse
Affiliation(s)
- Ella‐Louise Handley
- Institute for Bioengineering, School of EngineeringUniversity of EdinburghEdinburghUK
| | - Anthony Callanan
- Institute for Bioengineering, School of EngineeringUniversity of EdinburghEdinburghUK
| |
Collapse
|
13
|
Manohar-Sindhu S, Merfeld-Clauss S, Goddard Y, March KL, Traktuev DO. Diminished vasculogenesis under inflammatory conditions is mediated by Activin A. Angiogenesis 2023; 26:423-436. [PMID: 36977946 DOI: 10.1007/s10456-023-09873-w] [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: 12/09/2022] [Accepted: 03/06/2023] [Indexed: 03/30/2023]
Abstract
Severe inflammatory stress often leads to vessel rarefaction and fibrosis, resulting in limited tissue recovery. However, signaling pathways mediating these processes are not completely understood. Patients with ischemic and inflammatory conditions have increased systemic Activin A level, which frequently correlates with the severity of pathology. Yet, Activin A's contribution to disease progression, specifically to vascular homeostasis and remodeling, is not well defined. This study investigated vasculogenesis in an inflammatory environment with an emphasis on Activin A's role. Exposure of endothelial cells (EC) and perivascular cells (adipose stromal cells, ASC) to inflammatory stimuli (represented by blood mononuclear cells from healthy donors activated with lipopolysaccharide, aPBMC) dramatically decreased EC tubulogenesis or caused vessel rarefaction compared to control co-cultures, concurrent with increased Activin A secretion. Both EC and ASC upregulated Inhibin Ba mRNA and Activin A secretion in response to aPBMC or their secretome. We identified TNFα (in EC) and IL-1β (in EC and ASC) as the exclusive inflammatory factors, present in aPBMC secretome, responsible for induction of Activin A. Similar to ASC, brain and placental pericytes upregulated Activin A in response to aPBMC and IL-1β, but not TNFα. Both these cytokines individually diminished EC tubulogenesis. Blocking Activin A with neutralizing IgG mitigated detrimental effects of aPBMC or TNFα/IL-1β on tubulogenesis in vitro and vessel formation in vivo. This study delineates the signaling pathway through which inflammatory cells have a detrimental effect on vessel formation and homeostasis, and highlights the central role of Activin A in this process. Transitory interference with Activin A during early phases of inflammatory or ischemic insult, with neutralizing antibodies or scavengers, may benefit vasculature preservation and overall tissue recovery.
Collapse
Affiliation(s)
- Sahana Manohar-Sindhu
- UF Center for Regenerative Medicine, Division of Cardiovascular Medicine, Department of Medicine, UF College of Medicine, University of Florida, 1600 SW Archer Road, PO Box 100277, Gainesville, FL, 32610, USA
| | - Stephanie Merfeld-Clauss
- UF Center for Regenerative Medicine, Division of Cardiovascular Medicine, Department of Medicine, UF College of Medicine, University of Florida, 1600 SW Archer Road, PO Box 100277, Gainesville, FL, 32610, USA
| | - Yana Goddard
- UF Center for Regenerative Medicine, Division of Cardiovascular Medicine, Department of Medicine, UF College of Medicine, University of Florida, 1600 SW Archer Road, PO Box 100277, Gainesville, FL, 32610, USA
| | - Keith L March
- UF Center for Regenerative Medicine, Division of Cardiovascular Medicine, Department of Medicine, UF College of Medicine, University of Florida, 1600 SW Archer Road, PO Box 100277, Gainesville, FL, 32610, USA
| | - Dmitry O Traktuev
- UF Center for Regenerative Medicine, Division of Cardiovascular Medicine, Department of Medicine, UF College of Medicine, University of Florida, 1600 SW Archer Road, PO Box 100277, Gainesville, FL, 32610, USA.
| |
Collapse
|
14
|
Li X, Ou W, Xie M, Yang J, Li Q, Li T. Nanomedicine-Based Therapeutics for Myocardial Ischemic/Reperfusion Injury. Adv Healthc Mater 2023; 12:e2300161. [PMID: 36971662 PMCID: PMC11468948 DOI: 10.1002/adhm.202300161] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 03/05/2023] [Indexed: 03/29/2023]
Abstract
Myocardial ischemic/reperfusion (IR) injury is a global cardiovascular disease with high mortality and morbidity. Therapeutic interventions for myocardial ischemia involve restoring the occluded coronary artery. However, reactive oxygen species (ROS) inevitably impair the cardiomyocytes during the ischemic and reperfusion phases. Antioxidant therapy holds great promise against myocardial IR injury. The current therapeutic methodologies for ROS scavenging depend predominantly on administering antioxidants. Nevertheless, the intrinsic drawbacks of antioxidants limit their further clinical transformation. The use of nanoplatforms with versatile characteristics greatly benefits drug delivery in myocardial ischemic therapy. Nanoplatform-mediated drug delivery significantly improves drug bioavailability, increases therapeutic index, and reduces systemic toxicity. Nanoplatforms can be specifically and reasonably designed to enhance molecule accumulation at the myocardial site. The present review initially summarizes the mechanism of ROS generation during the process of myocardial ischemia. The understanding of this phenomenon will facilitate the advancement of innovative therapeutic strategies against myocardial IR injury. The latest developments in nanomedicine for treating myocardial ischemic injury are then discussed. Finally, the current challenges and perspectives in antioxidant therapy for myocardial IR injury are addressed.
Collapse
Affiliation(s)
- Xi Li
- Department of AnesthesiologyLaboratory of Mitochondria and MetabolismNational Clinical Research Center for GeriatricsWest China Hospital of Sichuan UniversityChengdu610041P. R. China
| | - Wei Ou
- Department of AnesthesiologyLaboratory of Mitochondria and MetabolismNational Clinical Research Center for GeriatricsWest China Hospital of Sichuan UniversityChengdu610041P. R. China
- Department of AnesthesiologyNanchong Central HospitalNanchong637000P. R. China
| | - Maodi Xie
- Department of AnesthesiologyLaboratory of Mitochondria and MetabolismNational Clinical Research Center for GeriatricsWest China Hospital of Sichuan UniversityChengdu610041P. R. China
| | - Jing Yang
- Department of AnesthesiologyLaboratory of Mitochondria and MetabolismNational Clinical Research Center for GeriatricsWest China Hospital of Sichuan UniversityChengdu610041P. R. China
| | - Qian Li
- Department of AnesthesiologyLaboratory of Mitochondria and MetabolismNational Clinical Research Center for GeriatricsWest China Hospital of Sichuan UniversityChengdu610041P. R. China
| | - Tao Li
- Department of AnesthesiologyLaboratory of Mitochondria and MetabolismNational Clinical Research Center for GeriatricsWest China Hospital of Sichuan UniversityChengdu610041P. R. China
| |
Collapse
|
15
|
Yousefzadeh N, Jeddi S. Long-term Ovariectomy Reduces Tolerance of Rats to Myocardial Ischemia-reperfusion Injury. Int J Endocrinol Metab 2023; 21:e135101. [PMID: 38028249 PMCID: PMC10676666 DOI: 10.5812/ijem-135101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 05/07/2023] [Accepted: 05/30/2023] [Indexed: 12/01/2023] Open
Abstract
Background The harmful impact of ovariectomy on myocardial ischemia-reperfusion (M/IR) injury has been established in the short term. Objectives In this study, we aimed to investigate the long-term effects of ovariectomy on M/IR injury. Methods Two methods involving dorsolateral skin incisions were used to induce the ovariectomized (OVX) rat model. The rats were divided into 2 groups: Control and OVX (n = 6). At the end of the study, the hearts were isolated and subjected to global ischemia using the Langendorff apparatus. Cardiac function indices (CFIs) were recorded, including left ventricular end-diastolic pressure (LVEDP), peak rates of positive (+dp/dt) and negative (-dp/dt) changes in LV pressure, and LV-developed pressure (LVDP). At the end of the reperfusion period, the hearts were used to measure the size of the infarct, levels of nitric oxide metabolites (NOx), and mRNA expression of NO synthase (NOS) enzymes, including endothelial (eNOS), neuronal (nNOS), and inducible (iNOS). Results Compared to controls, OVX rats had larger infarct size by 51%, higher LVEDP by 29%, and lower recovery of +dp/dt, -dp/dt, and LVDP by 29%, 22%, and 35%, respectively. Furthermore, in heart tissue, rats that underwent OVX had significantly higher concentrations of nitrate, nitrite, and NOx by 79%, 82%, and 83%, respectively. Additionally, these rats had lower mRNA levels of eNOS by 38% and higher mRNA levels of iNOS by 71%. Conclusions The long-term deficiency of estrogen increased the expression of iNOS and decreased the expression of eNOS in the heart tissue of OVX rats. Imbalanced NOS expressions were associated with exacerbated responses to M/IR injury in OVX rats.
Collapse
Affiliation(s)
- Nasibeh Yousefzadeh
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajad Jeddi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Yao L, He F, Zhao Q, Li D, Fu S, Zhang M, Zhang X, Zhou B, Wang L. Spatial Multiplexed Protein Profiling of Cardiac Ischemia-Reperfusion Injury. Circ Res 2023; 133:86-103. [PMID: 37249015 DOI: 10.1161/circresaha.123.322620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/05/2023] [Indexed: 05/31/2023]
Abstract
BACKGROUND Reperfusion therapy is critical to myocardial salvage in the event of a myocardial infarction but is complicated by ischemia-reperfusion injury (IRI). Limited understanding of the spatial organization of cardiac cells, which governs cellular interaction and function, has hindered the search for targeted interventions minimizing the deleterious effects of IRI. METHODS We used imaging mass cytometry to characterize the spatial distribution and dynamics of cell phenotypes and communities in the mouse left ventricle following IRI. Heart sections were collected from 12 cardiac segments (basal, mid-cavity, apical, and apex of the anterior, lateral, and inferior wall) and 8 time points (before ischemia [I-0H], and postreperfusion [R-0H, R-2H, R-6H, R-12H, R-1D, R-3D, R-7D]), and stained with 29 metal-isotope-tagged antibodies. Cell community analysis was performed on reconstructed images, and the most disease-relevant cell type and target protein were selected for intervention of IRI. RESULTS We obtained a total of 251 multiplexed images, and identified 197 063 single cells, which were grouped into 23 distinct cell communities based on the structure of cellular neighborhoods. The cellular architecture was heterogeneous throughout the ventricular wall and exhibited swift changes following IRI. Analysis of proteins with posttranslational modifications in single cells unveiled 13 posttranslational modification intensity clusters and highlighted increased H3K9me3 (tri-methylated lysine 9 of histone H3) as a key regulatory response in endothelial cells during the middle stage of IRI. Erasing H3K9 methylation, by silencing its methyltransferase Suv39h1 or overexpressing its demethylase Kdm4d in isolated endothelial cells, attenuated cardiac dysfunction and pathological remodeling following IRI. in vitro, H3K9me3 binding significantly increased at endothelial cell function-related genes upon hypoxia, suppressing tube formation, which was rescued by inhibiting H3K9me3. CONCLUSIONS We mapped the spatiotemporal heterogeneity of cellular phenotypes in the adult heart upon IRI, and uncovered H3K9me3 in endothelial cells as a potential therapeutic target for alleviating pathological remodeling of the heart following myocardial IRI.
Collapse
Affiliation(s)
- Luyan Yao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (L.Y., F.H., Q.Z., D.L., S.F., M.Z., X.Z., B.Z., L.W.)
| | - Funan He
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (L.Y., F.H., Q.Z., D.L., S.F., M.Z., X.Z., B.Z., L.W.)
| | - Quanyi Zhao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (L.Y., F.H., Q.Z., D.L., S.F., M.Z., X.Z., B.Z., L.W.)
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Bejing (Q.Z., B.Z., L.W.)
| | - Dandan Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (L.Y., F.H., Q.Z., D.L., S.F., M.Z., X.Z., B.Z., L.W.)
| | - Shufang Fu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (L.Y., F.H., Q.Z., D.L., S.F., M.Z., X.Z., B.Z., L.W.)
| | - Mingzhi Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (L.Y., F.H., Q.Z., D.L., S.F., M.Z., X.Z., B.Z., L.W.)
| | - Xingzhong Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (L.Y., F.H., Q.Z., D.L., S.F., M.Z., X.Z., B.Z., L.W.)
| | - Bingying Zhou
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (L.Y., F.H., Q.Z., D.L., S.F., M.Z., X.Z., B.Z., L.W.)
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Bejing (Q.Z., B.Z., L.W.)
| | - Li Wang
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Bejing (Q.Z., B.Z., L.W.)
- Key Laboratory of Application of Pluripotent Stem Cells in Heart Regeneration, Chinese Academy of Medical Sciences, Beijing (L.W.)
| |
Collapse
|
17
|
Fang G, Shen Y, Liao D. ENPP2 alleviates hypoxia/reoxygenation injury and ferroptosis by regulating oxidative stress and mitochondrial function in human cardiac microvascular endothelial cells. Cell Stress Chaperones 2023; 28:253-263. [PMID: 37052764 PMCID: PMC10167086 DOI: 10.1007/s12192-023-01324-1] [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: 10/13/2022] [Revised: 01/19/2023] [Accepted: 01/22/2023] [Indexed: 04/14/2023] Open
Abstract
This study aimed to elucidate the molecular mechanisms of hypoxia/reoxygenation (H/R) injury in human cardiac microvascular endothelial cells (HCMECs) by regulating ferroptosis. H/R model was established with HCMECs and before the reperfusion, ferroptosis inhibitor ferrostatin-1 or ferroptosis inducer erastin was all administered. Wound-healing assay was performed to detect the migration ability of cells in each group, and the angiogenesis ability was determined by tube formation assay. The level of reactive oxygen species (ROS) was detected by flow cytometry. Transmission electron microscopy (TEM) was used to observe the state of mitochondria. The expressions of related proteins in HCMECs were assessed by Western blot. From the results, H/R injury could inhibit the migration and angiogenesis, induce the ROS production, and cause the mitochondrial damage of HCMECs. Ferroptosis activator erastin could aggravate H/R injury in HCMECs, while the ferroptosis inhibitor ferrostatin-1 could reverse the effects of H/R on HCMECs. Western blot results showed that H/R or/and erastin treatment could significantly induce ACSL4, HGF, VEGF, p-ERK, and uPA protein expression and inhibit GPX4 expression. The addition of ferrostatin-1 resulted in the opposite trend of the proteins expression above to erastin treatment. What is more, overexpression of ENPP2 markedly suppressed the damaging effect of H/R on HCMECs and reversed the effects of H/R or erastin treatment on the expression of related proteins. These results demonstrated a great therapeutic efficacy of ENPP2 overexpression in preventing the development of H/R injury through inhibiting oxidative stress and ferroptosis.
Collapse
Affiliation(s)
- Guanhua Fang
- Department of Cardiovascular Surgery, Union Hospital, Fujian Medical University, Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, 350001 Fujian China
| | - Yanming Shen
- Fujian Medical University, Fuzhou, 350001 Fujian China
| | - Dongshan Liao
- Department of Cardiovascular Surgery, Union Hospital, Fujian Medical University, Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, 350001 Fujian China
| |
Collapse
|
18
|
Simonson B, Chaffin M, Hill MC, Atwa O, Guedira Y, Bhasin H, Hall AW, Hayat S, Baumgart S, Bedi KC, Margulies KB, Klattenhoff CA, Ellinor PT. Single-nucleus RNA sequencing in ischemic cardiomyopathy reveals common transcriptional profile underlying end-stage heart failure. Cell Rep 2023; 42:112086. [PMID: 36790929 PMCID: PMC10423750 DOI: 10.1016/j.celrep.2023.112086] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 11/14/2022] [Accepted: 01/23/2023] [Indexed: 02/16/2023] Open
Abstract
Ischemic cardiomyopathy (ICM) is the leading cause of heart failure worldwide, yet the cellular and molecular signature of this disease is largely unclear. Using single-nucleus RNA sequencing (snRNA-seq) and integrated computational analyses, we profile the transcriptomes of over 99,000 human cardiac nuclei from the non-infarct region of the left ventricle of 7 ICM transplant recipients and 8 non-failing (NF) controls. We find the cellular composition of the ischemic heart is significantly altered, with decreased cardiomyocytes and increased proportions of lymphatic, angiogenic, and arterial endothelial cells in patients with ICM. We show that there is increased LAMININ signaling from endothelial cells to other cell types in ICM compared with NF. Finally, we find that the transcriptional changes that occur in ICM are similar to those in hypertrophic and dilated cardiomyopathies and that the mining of these combined datasets can identify druggable genes that could be used to target end-stage heart failure.
Collapse
Affiliation(s)
- Bridget Simonson
- Precision Cardiology Laboratory, The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Mark Chaffin
- Precision Cardiology Laboratory, The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Matthew C Hill
- Precision Cardiology Laboratory, The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Ondine Atwa
- Precision Cardiology Laboratory, The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Yasmine Guedira
- Precision Cardiology Laboratory, The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Harshit Bhasin
- Precision Cardiology Laboratory, The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Amelia W Hall
- Precision Cardiology Laboratory, The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Gene Regulation Observatory, The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sikander Hayat
- Precision Cardiology Laboratory, Bayer US, LLC, Cambridge, MA 02142, USA
| | - Simon Baumgart
- Precision Cardiology Laboratory, Bayer US, LLC, Cambridge, MA 02142, USA
| | - Kenneth C Bedi
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kenneth B Margulies
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Patrick T Ellinor
- Precision Cardiology Laboratory, The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
19
|
Min S, Cho SW. Engineered human cardiac tissues for modeling heart diseases. BMB Rep 2023; 56:32-42. [PMID: 36443005 PMCID: PMC9887099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Indexed: 01/28/2023] Open
Abstract
Heart disease is one of the major life-threatening diseases with high mortality and incidence worldwide. Several model systems, such as primary cells and animals, have been used to understand heart diseases and establish appropriate treatments. However, they have limitations in accuracy and reproducibility in recapitulating disease pathophysiology and evaluating drug responses. In recent years, three-dimensional (3D) cardiac tissue models produced using tissue engineering technology and human cells have outperformed conventional models. In particular, the integration of cell reprogramming techniques with bioengineering platforms (e.g., microfluidics, scaffolds, bioprinting, and biophysical stimuli) has facilitated the development of heart-ona- chip, cardiac spheroid/organoid, and engineered heart tissue (EHT) to recapitulate the structural and functional features of the native human heart. These cardiac models have improved heart disease modeling and toxicological evaluation. In this review, we summarize the cell types for the fabrication of cardiac tissue models, introduce diverse 3D human cardiac tissue models, and discuss the strategies to enhance their complexity and maturity. Finally, recent studies in the modeling of various heart diseases are reviewed. [BMB Reports 2023; 56(1): 32-42].
Collapse
Affiliation(s)
- Sungjin Min
- Department of Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 03722, Korea,Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Korea,Corresponding author. Tel: +82-2-2123-5662; Fax: +82-2-362-7265; E-mail:
| |
Collapse
|
20
|
Min S, Cho SW. Engineered human cardiac tissues for modeling heart diseases. BMB Rep 2023; 56:32-42. [PMID: 36443005 PMCID: PMC9887099 DOI: 10.5483/bmbrep.2022-0185] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/28/2022] [Accepted: 11/28/2022] [Indexed: 07/30/2023] Open
Abstract
Heart disease is one of the major life-threatening diseases with high mortality and incidence worldwide. Several model systems, such as primary cells and animals, have been used to understand heart diseases and establish appropriate treatments. However, they have limitations in accuracy and reproducibility in recapitulating disease pathophysiology and evaluating drug responses. In recent years, three-dimensional (3D) cardiac tissue models produced using tissue engineering technology and human cells have outperformed conventional models. In particular, the integration of cell reprogramming techniques with bioengineering platforms (e.g., microfluidics, scaffolds, bioprinting, and biophysical stimuli) has facilitated the development of heart-ona- chip, cardiac spheroid/organoid, and engineered heart tissue (EHT) to recapitulate the structural and functional features of the native human heart. These cardiac models have improved heart disease modeling and toxicological evaluation. In this review, we summarize the cell types for the fabrication of cardiac tissue models, introduce diverse 3D human cardiac tissue models, and discuss the strategies to enhance their complexity and maturity. Finally, recent studies in the modeling of various heart diseases are reviewed. [BMB Reports 2023; 56(1): 32-42].
Collapse
Affiliation(s)
- Sungjin Min
- Department of Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 03722, Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Korea
| |
Collapse
|
21
|
Madder ( Rubia cordifolia L.) Alleviates Myocardial Ischemia-Reperfusion Injury by Protecting Endothelial Cells from Apoptosis and Inflammation. Mediators Inflamm 2023; 2023:5015039. [PMID: 36875688 PMCID: PMC9981279 DOI: 10.1155/2023/5015039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/23/2022] [Accepted: 09/26/2022] [Indexed: 02/25/2023] Open
Abstract
Objective Ischemia-reperfusion injury often occurs in organ transplantation, coronary heart disease, ischemic heart disease, and other diseases, which greatly reduces clinical efficacy. This study examined the effectiveness of madder as a medicine to treat ischemia-reperfusion injury. Methods The efficacy of madder was evaluated by measuring myocardial infarction size, coronary outflow volume, myocardial contraction rate, activation of inflammatory factors, autophagy factors, apoptosis factors, and related pathway genes in mice. Results The results indicated that treatment with madder can effectively reduce the area of myocardial infarction and restore arterial blood flow velocity and myocardial contractility in mice. Additionally, madder treatment inhibited the expression of inflammatory factors, autophagy factors, and apoptosis factors in mice and reduced the degree of myocardial cell injury. Studies have also shown that madder treatment can alleviate myocardial ischemia-reperfusion injury in mice and inhibit the occurrence of inflammatory response by inhibiting the activity of the NF-κB pathway. Conclusion The results showed that madder was effective against ischemia-reperfusion injury, thus showing potential as a clinical drug for treating ischemia-reperfusion injury.
Collapse
|
22
|
Yeh KC, Lee CJ, Song JS, Wu CH, Yeh TK, Wu SH, Hsieh TC, Chen YT, Tseng HY, Huang CL, Chen CT, Jan JJ, Chou MC, Shia KS, Chiang KH. Protective Effect of CXCR4 Antagonist DBPR807 against Ischemia-Reperfusion Injury in a Rat and Porcine Model of Myocardial Infarction: Potential Adjunctive Therapy for Percutaneous Coronary Intervention. Int J Mol Sci 2022; 23:ijms231911730. [PMID: 36233031 PMCID: PMC9570210 DOI: 10.3390/ijms231911730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/23/2022] [Accepted: 09/28/2022] [Indexed: 11/25/2022] Open
Abstract
CXCR4 antagonists have been claimed to reduce mortality after myocardial infarction in myocardial infarction (MI) animals, presumably due to suppressing inflammatory responses caused by myocardial ischemia-reperfusion injury, thus, subsequently facilitating tissue repair and cardiac function recovery. This study aims to determine whether a newly designed CXCR4 antagonist DBPR807 could exert better vascular-protective effects than other clinical counterparts (e.g., AMD3100) to alleviate cardiac damage further exacerbated by reperfusion. Consequently, we find that instead of traditional continuous treatment or multiple-dose treatment at different intervals of time, a single-dose treatment of DBPR807 before reperfusion in MI animals could attenuate inflammation via protecting oxidative stress damage and preserve vascular/capillary density and integrity via mobilizing endothelial progenitor cells, leading to a desirable fibrosis reduction and recovery of cardiac function, as evaluated with the LVEF (left ventricular ejection fraction) in infarcted hearts in rats and mini-pigs, respectively. Thus, it is highly suggested that CXCR4 antagonists should be given at a single high dose prior to reperfusion to provide the maximal cardiac functional improvement. Based on its favorable efficacy and safety profiles indicated in tested animals, DBPR807 has a great potential to serve as an adjunctive medicine for percutaneous coronary intervention (PCI) therapies in acute MI patients.
Collapse
Affiliation(s)
- Kai-Chia Yeh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Chia-Jui Lee
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Jen-Shin Song
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Chien-Huang Wu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Teng-Kuang Yeh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Szu-Huei Wu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Tsung-Chin Hsieh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Yen-Ting Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Huan-Yi Tseng
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Chen-Lung Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Chiung-Tong Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Jiing-Jyh Jan
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Ming-Chen Chou
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Kak-Shan Shia
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan 35053, Taiwan
- Correspondence: (K.-S.S.); (K.-H.C.)
| | - Kuang-Hsing Chiang
- Taipei Heart Institute, Taipei Medical University, Taipei 11031, Taiwan
- Department of Cardiology, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei 106319, Taiwan
- Correspondence: (K.-S.S.); (K.-H.C.)
| |
Collapse
|
23
|
Gallinat A, Badimon L. DJ-1 interacts with the ectopic ATP-synthase in endothelial cells during acute ischemia and reperfusion. Sci Rep 2022; 12:12753. [PMID: 35882968 PMCID: PMC9325725 DOI: 10.1038/s41598-022-16998-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 07/19/2022] [Indexed: 01/28/2023] Open
Abstract
Endothelial cells (ECs) play a central role in ischemia. ATP-Synthase is now recognized to be ectopically expressed in the cell surface of many cell types, with putative roles described in angiogenesis, proliferation, and intracellular pH regulation. DJ-1 is a multifunctional protein, involved in cell protection against ischemia, ischemia–reperfusion (I/R), and oxidative stress, that regulates mitochondrial ATP-synthase. Here we focused on the characterization of the endothelial dynamics of DJ-1, and its implication in the regulation of the ectopic ATP-synthase (ecATP-S) activity, during acute ischemia and I/R in ECs. We found that DJ-1 is secreted from ECs, by a mechanism enhanced in ischemia and I/R. A cleaved form of DJ-1 (DJ-1∆C) was found only in the secretome of ischemic cells. The ecATP-S activity increased following acute ischemia in ECs, coinciding with DJ-1 and DJ-1∆C secretion. The inhibition of DJ-1 expression inhibited the ecATP-S response to ischemia by ∼ 50%, and its exogenous administration maximized the effect, together with an enhanced Akt phosphorylation and angiotube-formation potential at reperfusion. Immunoprecipitation studies showed direct interaction between DJ-1 and the ecATP-S. Altogether suggesting that DJ-1 is actively cleaved and released from ischemic ECs and plays an important role in the regulation of the ecATP-S activity during acute ischemia and reperfusion.
Collapse
Affiliation(s)
- Alex Gallinat
- Cardiovascular Program-ICCC, IR-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, c/Sant Antoni María Claret, 167, 08025, Barcelona, Spain.,Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Lina Badimon
- Cardiovascular Program-ICCC, IR-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, c/Sant Antoni María Claret, 167, 08025, Barcelona, Spain. .,CIBERCV-Instituto de Salud Carlos III, Madrid, Spain. .,UAB-Chair Cardiovascular Research, Barcelona, Spain.
| |
Collapse
|
24
|
Ai Y, He M, Wan C, Luo H, Xin H, Wang Y, Liang Q. Nanoplatform‐Based Reactive Oxygen Species Scavengers for Therapy of Ischemia‐Reperfusion Injury. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202200066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Yongjian Ai
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology Tsinghua University‐Peking University Joint Centre for Life Sciences Beijing Key Lab of Microanalytical Methods & Instrumentation Department of Chemistry Center for Synthetic and Systems Biology Tsinghua University Beijing 100084 P. R. China
| | - Meng‐Qi He
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology Tsinghua University‐Peking University Joint Centre for Life Sciences Beijing Key Lab of Microanalytical Methods & Instrumentation Department of Chemistry Center for Synthetic and Systems Biology Tsinghua University Beijing 100084 P. R. China
| | - Chengxian Wan
- Jiangxi Provincial People's Hospital The First Affiliated Hospital of Nanchang Medical College The Affiliated People's Hospital of Nanchang University Nanchang Jiangxi 330006 P. R. China
| | - Hua Luo
- State Key Laboratory of Quality Research in Chinese Medicine Institute of Chinese Medical Sciences University of Macau Macau SAR 999078 China
| | - Hongbo Xin
- The National Engineering Research Center for Bioengineering Drugs and the Technologies Institute of Translational Medicine Nanchang University Nanchang Jiangxi 330088 P. R. China
| | - Yitao Wang
- State Key Laboratory of Quality Research in Chinese Medicine Institute of Chinese Medical Sciences University of Macau Macau SAR 999078 China
| | - Qionglin Liang
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology Tsinghua University‐Peking University Joint Centre for Life Sciences Beijing Key Lab of Microanalytical Methods & Instrumentation Department of Chemistry Center for Synthetic and Systems Biology Tsinghua University Beijing 100084 P. R. China
| |
Collapse
|
25
|
Current knowledge of pyroptosis in heart diseases. J Mol Cell Cardiol 2022; 171:81-89. [PMID: 35868567 DOI: 10.1016/j.yjmcc.2022.07.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 06/22/2022] [Accepted: 07/13/2022] [Indexed: 10/17/2022]
Abstract
Pyroptosis is a form of pro-inflammatory, necrotic cell death mediated by proteins of the gasdermin family. Various heart diseases, including myocardial ischemia/reperfusion injury, myocardial infarction, and heart failure, involve cardiomyocyte and non-myocyte pyroptosis. Cardiomyocyte pyroptosis also causes the release of pro-inflammatory cytokines. Recent studies have confirmed that pyroptosis is predominantly triggered by both the canonical and non-canonical inflammasome pathways, which independently facilitate caspase-1 or caspase-11/4/5 activation and gasdermin D (GSDMD) cleavage. Cardiac fibroblast and myeloid cell pyroptosis also contributes to the pathogenesis and development of heart diseases. This review summarizes the recent studies on pyroptosis in heart diseases and discusses the associated therapeutic targets.
Collapse
|
26
|
Protective effects of trehalose preconditioning on cardiac and coronary endothelial function through eNOS signaling pathway in a rat model of ischemia-reperfusion injury. Mol Cell Biochem 2022; 477:2403-2414. [PMID: 35578066 DOI: 10.1007/s11010-022-04451-y] [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: 01/31/2022] [Accepted: 04/25/2022] [Indexed: 01/18/2023]
Abstract
Coronary endothelial dysfunction is a major cause of ischemia-reperfusion (I/R) injury. Trehalose, a natural disaccharide, has been reported to ameliorate endothelial dysfunction during aging by activating endothelial nitric oxide synthase (eNOS); however, its role in I/R injury is unknown. This study evaluated the effects of trehalose preconditioning on cardiac and coronary endothelial function after I/R. Langendorff-perfused rat hearts underwent 30 min of global ischemia followed by 80 min of reperfusion with or without trehalose preconditioning. Rate pressure product (RPP) and coronary flow (CF) were measured during reperfusion. Perivascular edema was assessed by hematoxylin and eosin staining. Myocardial oxidative stress and apoptosis were evaluated by immunohistochemistry and TUNEL staining, respectively. eNOS dimerization was determined by western blotting. An eNOS inhibitor was used to examine the role of eNOS. Trehalose preconditioning showed a higher recovery rate after I/R as indicated by high RPP (control vs. trehalose, 28 ± 6% vs. 46 ± 9%; P = 0.017, Cohen's d = 2.3) and CF values (35 ± 10% vs. 55 ± 9%; P = 0.025, d = 1.7). Furthermore, trehalose preconditioning reduced perivascular edema, myocardial oxidative stress, and apoptosis. The eNOS dimerization ratio was increased by trehalose (1.2 ± 0.2 vs. 1.6 ± 0.2; P = 0.023, d = 2.1), which was associated with the recovery of RPP and CF. These effects of trehalose were abolished by the eNOS inhibitor. Trehalose preconditioning showed protective effects on cardiac and coronary endothelial function after I/R through the eNOS signaling pathway.
Collapse
|
27
|
Sabe SA, Feng J, Sellke FW, Abid MR. Mechanisms and clinical implications of endothelium-dependent vasomotor dysfunction in coronary microvasculature. Am J Physiol Heart Circ Physiol 2022; 322:H819-H841. [PMID: 35333122 PMCID: PMC9018047 DOI: 10.1152/ajpheart.00603.2021] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 03/21/2022] [Accepted: 03/21/2022] [Indexed: 12/16/2022]
Abstract
Coronary microvascular disease (CMD), which affects the arterioles and capillary endothelium that regulate myocardial perfusion, is an increasingly recognized source of morbidity and mortality, particularly in the setting of metabolic syndrome. The coronary endothelium plays a pivotal role in maintaining homeostasis, though factors such as diabetes, hypertension, hyperlipidemia, and obesity can contribute to endothelial injury and consequently arteriolar vasomotor dysfunction. These disturbances in the coronary microvasculature clinically manifest as diminished coronary flow reserve, which is a known independent risk factor for cardiac death, even in the absence of macrovascular atherosclerotic disease. Therefore, a growing body of literature has examined the molecular mechanisms by which coronary microvascular injury occurs at the level of the endothelium and the consequences on arteriolar vasomotor responses. This review will begin with an overview of normal coronary microvascular physiology, modalities of measuring coronary microvascular function, and clinical implications of CMD. These introductory topics will be followed by a discussion of recent advances in the understanding of the mechanisms by which inflammation, oxidative stress, insulin resistance, hyperlipidemia, hypertension, shear stress, endothelial cell senescence, and tissue ischemia dysregulate coronary endothelial homeostasis and arteriolar vasomotor function.
Collapse
Affiliation(s)
- Sharif A Sabe
- Cardiovascular Research Center, Rhode Island Hospital, Providence, Rhode Island
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
| | - Jun Feng
- Cardiovascular Research Center, Rhode Island Hospital, Providence, Rhode Island
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
| | - Frank W Sellke
- Cardiovascular Research Center, Rhode Island Hospital, Providence, Rhode Island
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
| | - M Ruhul Abid
- Cardiovascular Research Center, Rhode Island Hospital, Providence, Rhode Island
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
| |
Collapse
|
28
|
Ji M, Cheng J, Zhang D. Oxycodone protects cardiac microvascular endothelial cells against ischemia/reperfusion injury by binding to Sigma-1 Receptor. Bioengineered 2022; 13:9628-9644. [PMID: 35412431 PMCID: PMC9161947 DOI: 10.1080/21655979.2022.2057632] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 01/14/2023] Open
Abstract
Endothelial dysfunction is an important mechanism involved in myocardial ischemia-reperfusion (I/R) injury. We aimed to explore the effects of Oxycodone on myocardial I/R injury in vivo and in vitro to reveal its mechanisms related to Sigma-1 Receptor (SIGMAR1). A rat model of I/R-induced myocardial injury was developed. The ischemic area and myocardial histopathological changes after oxycodone addition were evaluated by TTC staining and H&E staining. LDH, CK-MB and cTnI levels were used to assess myocardial function. Then, the endothelial integrity was reflected by the expressions of ZO-1, Claudin-1 and Occludin. Afterward, ELISA, RT-qPCR, western blot and immunofluorescence assays were adopted for the detection of inflammation-related genes. SIGMAR1 expression in myocardial tissues induced by I/R and cardiac microvascular endothelial cells (CMECs) under hypoxic/reoxygenation (H/R) was determined using RT-qPCR and western blotting. Subsequently, after SIGMAR1 silencing or BD1047 addition (a SIGMAR1 antagonist), cell apoptosis and endothelial integrity were analyzed in the presence of Oxycodone in H/R-stimulated CMECs. Results indicated that Oxycodone decreased the ischemic area and improved myocardial function in myocardial I/R injury rat. Oxycodone improved myocardial histopathological injury and elevated endothelial integrity, evidenced by upregulated ZO-1, Claudin-1 and Occludin expressions. Moreover, inflammatory response was alleviated after Oxycodone administration. Molecular docking suggested that SIGMAR1 could directly bind to Oxycodone. Oxycodone elevated SIGMAR1 expression and SIGMAR1 deletion or BD1047 addition attenuated the impacts of Oxycodone on apoptosis and endothelial integrity of CMECs induced by H/R. Collectively, Oxycodone alleviates myocardial I/R injury in vivo and in vitro by binding to SIGMAR1.
Collapse
Affiliation(s)
- Meihua Ji
- Department of Anesthesiology, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
- Department of Anesthesiology of Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jing Cheng
- Department of Anesthesiology, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
- Department of Anesthesiology of Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Daimin Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
29
|
Liu K, Liu D, Cui W. Protective Effect and Mechanism of Traditional Chinese Medicine on Myocardial Ischemia Reperfusion Injury. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:6121407. [PMID: 35399643 PMCID: PMC8991389 DOI: 10.1155/2022/6121407] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 11/17/2022]
Abstract
After acute myocardial infarction, early restoration of myocardial perfusion by thrombolysis or percutaneous coronary intervention is the most effective way to reduce the size of myocardial infarction and improve clinical outcomes. However, recovery of blood flow to the ischemic myocardium may cause ischemia-reperfusion (I/R) injury, a phenomenon that instead reduces the efficacy of myocardial reperfusion. Traditional Chinese medicine (TCM) has long been used for the treatment of cardiovascular diseases and has shown remarkable efficacy. Many studies have shown that some TCMs and their active components can exert protective effects against myocardial I/R injury through different mechanisms. This review summarized the protective mechanisms and current research advances of TCMs in myocardial I/R injury.
Collapse
Affiliation(s)
- Kuo Liu
- Cardiology Department, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Demin Liu
- Cardiology Department, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Wei Cui
- Cardiology Department, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| |
Collapse
|
30
|
Lai CC, Tang CY, Fu SK, Tseng WC, Tseng KW. Effects of swimming training on myocardial protection in rats. Biomed Rep 2022; 16:19. [PMID: 35251606 PMCID: PMC8850963 DOI: 10.3892/br.2022.1502] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 12/23/2021] [Indexed: 11/07/2022] Open
Abstract
Swimming is important for promoting and maintaining health, as it can increase the efficiency of the cardiovascular system and decrease the occurrence of cardiovascular diseases. The objective of the present study was to examine whether swimming training could decrease myocardial injury in rats caused by myocardial ischemia/reperfusion (I/R). Sprague-Dawley rats were randomized into four groups, namely the Sham, coronary artery occlusion, swimming training and ischemic preconditioning (IPC) groups. Myocardial I/R was induced in anesthetized male Sprague-Dawley rats by a 40-min occlusion followed by a 3-h reperfusion of the left anterior descending coronary artery. The rats were sacrificed after surgery and their hearts were examined. The results demonstrated that the number of TUNEL-positive nuclei and degree of caspase-3 activation were both significantly increased in the myocardium following myocardial I/R in rats, indicating increased cardiomyocyte apoptosis. On the other hand, swimming training decreased the serum levels of creatine phosphokinase, lactate dehydrogenase and cardiac troponin I, and was associated with reduced histological damage and myocardial infarct size. Furthermore, swimming training also reduced TNF-α levels, caspase-3 activation and enhanced Bcl-2 activation, which decreased the number of apoptotic cells in the myocardium. The findings of the present study showed that swimming training and IPC could similarly decrease myocardial injury following myocardial I/R, and may therefore be used as exercise training to effectively prevent myocardial injury.
Collapse
Affiliation(s)
- Chang-Chi Lai
- Department of Exercise and Health Sciences, University of Taipei, Taipei 11153, Taiwan, R.O.C
| | - Chia-Yu Tang
- Department of Physical Education, Chang Gung University, Taoyuan 33302, Taiwan, R.O.C
| | - Szu-Kai Fu
- Graduate Institute of Sports Training, University of Taipei, Taipei 11153, Taiwan, R.O.C
| | - Wei-Chin Tseng
- Department of Exercise and Health Sciences, University of Taipei, Taipei 11153, Taiwan, R.O.C
| | - Kuo-Wei Tseng
- Department of Exercise and Health Sciences, University of Taipei, Taipei 11153, Taiwan, R.O.C
| |
Collapse
|
31
|
Zhang H, Kim H, Park BW, Noh M, Kim Y, Park J, Park JH, Kim JJ, Sim WS, Ban K, Park HJ, Kwon YG. CU06-1004 enhances vascular integrity and improves cardiac remodeling by suppressing edema and inflammation in myocardial ischemia-reperfusion injury. Exp Mol Med 2022; 54:23-34. [PMID: 34997212 PMCID: PMC8814060 DOI: 10.1038/s12276-021-00720-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 11/11/2021] [Accepted: 11/15/2021] [Indexed: 11/13/2022] Open
Abstract
Ischemia-reperfusion (I/R) injury accelerates the cardiomyocytes (CMs) death by oxidative stress, and thereby deteriorates cardiac function. There has been a paradigm shift in the therapeutic perspective more towards the prevention or amelioration of damage caused by reperfusion. Cardiac microvascular endothelial cells (CMECs) are more vulnerable to reperfusion injury and play the crucial roles more than CMs in the pathological process of early I/R injury. In this study, we investigate that CU06-1004, as a vascular leakage blocker, can improve cardiac function by inhibiting CMEC's hyperpermeability and subsequently reducing the neutrophil's plugging and infiltration in infarcted hearts. CU06-1004 was delivered intravenously 5 min before reperfusion and the rats were randomly divided into three groups: (1) vehicle, (2) low-CU06-1004 (1 mg/kg, twice at 24 h intervals), and (3) high-CU06-1004 (5 mg/kg, once before reperfusion). CU06-1004 treatment reduced necrotic size and cardiac edema by enhancing vascular integrity, as demonstrated by the presence of intact junction proteins on CMECs and surrounding pericytes in early I/R injury. It also decreased the expression of vascular cell adhesion molecule 1 (VCAM-1) on CMECs, resulting in reduced infiltration of neutrophils and macrophages. Echocardiography showed that the CU06-1004 treatment significantly improved cardiac function compared with the vehicle group. Interestingly, single high-dose treatment with CU06-1004 provided a greater functional improvement than repetitive low-dose treatment until 8 weeks post I/R. These findings demonstrate that CU06-1004 enhances vascular integrity and improves cardiac function by preventing lethal myocardial I/R injury. It can provide a promising therapeutic option, as potential adjunctive therapy to current reperfusion strategies.
Collapse
Affiliation(s)
- Haiying Zhang
- grid.15444.300000 0004 0470 5454Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 120-749 Republic of Korea ,R&D Department, Curacle Co. Ltd, Seongnam-si, Republic of Korea
| | - Hyeok Kim
- grid.411947.e0000 0004 0470 4224Department of Medical Life Science, College of Medicine, The Catholic University of Korea, Seoul, 06591 Republic of Korea
| | - Bong Woo Park
- grid.411947.e0000 0004 0470 4224Department of Medical Life Science, College of Medicine, The Catholic University of Korea, Seoul, 06591 Republic of Korea
| | - Minyoung Noh
- grid.15444.300000 0004 0470 5454Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 120-749 Republic of Korea
| | - Yeomyeong Kim
- grid.15444.300000 0004 0470 5454Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 120-749 Republic of Korea
| | - Jeongeun Park
- grid.15444.300000 0004 0470 5454Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 120-749 Republic of Korea
| | - Jae-Hyun Park
- grid.411947.e0000 0004 0470 4224Department of Medical Life Science, College of Medicine, The Catholic University of Korea, Seoul, 06591 Republic of Korea
| | - Jin-Ju Kim
- grid.411947.e0000 0004 0470 4224Department of Medical Life Science, College of Medicine, The Catholic University of Korea, Seoul, 06591 Republic of Korea
| | - Woo-Sup Sim
- grid.411947.e0000 0004 0470 4224Department of Medical Life Science, College of Medicine, The Catholic University of Korea, Seoul, 06591 Republic of Korea
| | - Kiwon Ban
- grid.35030.350000 0004 1792 6846Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, 999077 Hong Kong
| | - Hun-Jun Park
- Department of Medical Life Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea. .,Division of Cardiology, Department of Internal Medicine, The Catholic University of Korea, Banpo-daero 222, Seocho-gu, Seoul, 137701, Republic of Korea.
| | - Young-Guen Kwon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 120-749, Republic of Korea.
| |
Collapse
|
32
|
Thitiwuthikiat P, Ta-uea T, Ponghan T, Meebua S, Siriwittayawan D, Nuamchit T. The protective effects of reparixin against endothelial ischemia-reperfusion injury. Int J Health Sci (Qassim) 2022; 16:20-24. [PMID: 35599941 PMCID: PMC9092537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Objectives Myocardial ischemia is a lack of blood supply to myocardial tissue. Rapid reperfusion therapy is required to prevent myocardial infarction. However, ischemia and reperfusion contribute to myocardial and endothelial injury or ischemia-reperfusion injury (IRI). A pro-inflammatory cytokine interleukin-8 (IL-8/CXCL8) plays an important role in the activation of neutrophil accumulation and promotes endothelial dysfunction. Therefore, inhibition of IRI through the regulation of inflammation using a CXCL8 receptor inhibitor reparixin is an attractive target. The aim of this study is to evaluate the effect of reparixin on endothelial cell viability after IRI. Methods Human vascular endothelial cells (EA.hy926) were cultured and pretreated with reparixin at concentrations of 0-1 mg/ml. To simulate ischemia, the cells were exposed to simulated ischemia solution for 60 min. Then, the cells were given complete medium as reperfusion followed by treatment with reparixin and incubated for 24 h. Cell viability was tested using MTT assay. Results Percentages of cell viability of reparixin-treated groups of 0.0625 mg/mL (67.88 ± 7.82% control) and 0.125 mg/mL (84.28 ± 4.68% control) were significantly higher than that of the IR group (44.31 ± 4.64% control) at P < 0.05. The percentage of cell viability in the 0.125 mg/mL reparixin-treated group was not significantly different compared to the control. Conclusion Pretreatment and treatment of endothelial cells with reparixin, which is a CXCL-8 receptor inhibitor, demonstrated a protective effect on cell viability after simulated ischemia-reperfusion. However, further studies to investigate the underlying mechanisms are needed.
Collapse
Affiliation(s)
- Piyanuch Thitiwuthikiat
- Department of Cardio-thoracic Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, Thailand
| | - Tamonlak Ta-uea
- Department of Cardio-thoracic Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, Thailand
| | - Theeraya Ponghan
- Department of Cardio-thoracic Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, Thailand
| | - Saranya Meebua
- Department of Cardio-thoracic Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, Thailand
| | - Duangduan Siriwittayawan
- Department of Cardio-thoracic Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, Thailand
| | - Teonchit Nuamchit
- Department of Cardio-thoracic Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, Thailand,Address for correspondence: Teonchit Nuamchit, Department of Cardio-thoracic Technology, Faculty of Allied Health Sciences, Naresuan University, Thapho, Muang, Phitsanulok 65000, Thailand. E-mail:
| |
Collapse
|
33
|
Qamar A, Zhao J, Xu L, McLeod P, Huang X, Jiang J, Liu W, Haig A, Zhang ZX. Cyclophilin D Regulates the Nuclear Translocation of AIF, Cardiac Endothelial Cell Necroptosis and Murine Cardiac Transplant Injury. Int J Mol Sci 2021; 22:11038. [PMID: 34681708 PMCID: PMC8540562 DOI: 10.3390/ijms222011038] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/23/2021] [Accepted: 10/08/2021] [Indexed: 12/26/2022] Open
Abstract
Ischemia-reperfusion injury (IRI) is an inevitable consequence of organ transplant procedure and associated with acute and chronic organ rejection in transplantation. IRI leads to various forms of programmed cell death, which worsens tissue damage and accelerates transplant rejection. We recently demonstrated that necroptosis participates in murine cardiac microvascular endothelial cell (MVEC) death and murine cardiac transplant rejection. However, MVEC death under a more complex IRI model has not been studied. In this study, we found that simulating IRI conditions in vitro by hypoxia, reoxygenation and treatment with inflammatory cytokines induced necroptosis in MVECs. Interestingly, the apoptosis-inducing factor (AIF) translocated to the nucleus during MVEC necroptosis, which is regulated by the mitochondrial permeability molecule cyclophilin D (CypD). Furthermore, CypD deficiency in donor cardiac grafts inhibited AIF translocation and mitigated graft IRI and rejection (n = 7; p = 0.002). Our studies indicate that CypD and AIF play significant roles in MVEC necroptosis and cardiac transplant rejection following IRI. Targeting CypD and its downstream AIF may be a plausible approach to inhibit IRI-caused cardiac damage and improve transplant survival.
Collapse
Affiliation(s)
- Adnan Qamar
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, B4-231, 339 Windermere Road, London, ON N6A 5A5, Canada; (A.Q.); (J.Z.); (L.X.); (P.M.); (X.H.); (J.J.)
- Department of Pathology, Western University, 1151 Richmond Street, London, ON N6A 3K7, Canada; (W.L.); (A.H.)
| | - Jianqi Zhao
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, B4-231, 339 Windermere Road, London, ON N6A 5A5, Canada; (A.Q.); (J.Z.); (L.X.); (P.M.); (X.H.); (J.J.)
- Department of Pathology, Western University, 1151 Richmond Street, London, ON N6A 3K7, Canada; (W.L.); (A.H.)
- Department of Rheumatology and Immunology, The First Hospital of Jilin University, 3808 Jiefang Road, Changchun 130021, China
| | - Laura Xu
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, B4-231, 339 Windermere Road, London, ON N6A 5A5, Canada; (A.Q.); (J.Z.); (L.X.); (P.M.); (X.H.); (J.J.)
- Department of Pathology, Western University, 1151 Richmond Street, London, ON N6A 3K7, Canada; (W.L.); (A.H.)
| | - Patrick McLeod
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, B4-231, 339 Windermere Road, London, ON N6A 5A5, Canada; (A.Q.); (J.Z.); (L.X.); (P.M.); (X.H.); (J.J.)
| | - Xuyan Huang
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, B4-231, 339 Windermere Road, London, ON N6A 5A5, Canada; (A.Q.); (J.Z.); (L.X.); (P.M.); (X.H.); (J.J.)
| | - Jifu Jiang
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, B4-231, 339 Windermere Road, London, ON N6A 5A5, Canada; (A.Q.); (J.Z.); (L.X.); (P.M.); (X.H.); (J.J.)
| | - Weihua Liu
- Department of Pathology, Western University, 1151 Richmond Street, London, ON N6A 3K7, Canada; (W.L.); (A.H.)
| | - Aaron Haig
- Department of Pathology, Western University, 1151 Richmond Street, London, ON N6A 3K7, Canada; (W.L.); (A.H.)
| | - Zhu-Xu Zhang
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, B4-231, 339 Windermere Road, London, ON N6A 5A5, Canada; (A.Q.); (J.Z.); (L.X.); (P.M.); (X.H.); (J.J.)
- Department of Pathology, Western University, 1151 Richmond Street, London, ON N6A 3K7, Canada; (W.L.); (A.H.)
- Multi-Organ Transplant Program, London Health Sciences Centre, London, ON N6A 5A5, Canada
- Division of Nephrology, Department of Medicine, Western University, London, ON N6A 3K7, Canada
| |
Collapse
|
34
|
Jelemenský M, Kovácsházi C, Ferenczyová K, Hofbauerová M, Kiss B, Pállinger É, Kittel Á, Sayour VN, Görbe A, Pelyhe C, Hambalkó S, Kindernay L, Barančík M, Ferdinandy P, Barteková M, Giricz Z. Helium Conditioning Increases Cardiac Fibroblast Migration Which Effect Is Not Propagated via Soluble Factors or Extracellular Vesicles. Int J Mol Sci 2021; 22:10504. [PMID: 34638845 PMCID: PMC8508629 DOI: 10.3390/ijms221910504] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/21/2021] [Accepted: 09/24/2021] [Indexed: 12/18/2022] Open
Abstract
Helium inhalation induces cardioprotection against ischemia/reperfusion injury, the cellular mechanism of which remains not fully elucidated. Extracellular vesicles (EVs) are cell-derived, nano-sized membrane vesicles which play a role in cardioprotective mechanisms, but their function in helium conditioning (HeC) has not been studied so far. We hypothesized that HeC induces fibroblast-mediated cardioprotection via EVs. We isolated neonatal rat cardiac fibroblasts (NRCFs) and exposed them to glucose deprivation and HeC rendered by four cycles of 95% helium + 5% CO2 for 1 h, followed by 1 h under normoxic condition. After 40 h of HeC, NRCF activation was analyzed with a Western blot (WB) and migration assay. From the cell supernatant, medium extracellular vesicles (mEVs) were isolated with differential centrifugation and analyzed with WB and nanoparticle tracking analysis. The supernatant from HeC-treated NRCFs was transferred to naïve NRCFs or immortalized human umbilical vein endothelial cells (HUVEC-TERT2), and a migration and angiogenesis assay was performed. We found that HeC accelerated the migration of NRCFs and did not increase the expression of fibroblast activation markers. HeC tended to decrease mEV secretion of NRCFs, but the supernatant of HeC or the control NRCFs did not accelerate the migration of naïve NRCFs or affect the angiogenic potential of HUVEC-TERT2. In conclusion, HeC may contribute to cardioprotection by increasing fibroblast migration but not by releasing protective mEVs or soluble factors from cardiac fibroblasts.
Collapse
Affiliation(s)
- Marek Jelemenský
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia; (M.J.); (K.F.); (L.K.); (M.B.)
| | - Csenger Kovácsházi
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; (C.K.); (B.K.); (V.N.S.); (A.G.); (C.P.); (S.H.); (P.F.)
| | - Kristína Ferenczyová
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia; (M.J.); (K.F.); (L.K.); (M.B.)
| | - Monika Hofbauerová
- Institute of Physics, Slovak Academy of Sciences, Dúbravská Cesta 9, 84511 Bratislava, Slovakia;
- Centre for Advanced Materials Application, Slovak Academy of Sciences, Dúbravská Cesta 9, 84511 Bratislava, Slovakia
| | - Bernadett Kiss
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; (C.K.); (B.K.); (V.N.S.); (A.G.); (C.P.); (S.H.); (P.F.)
- MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary
| | - Éva Pállinger
- Department of Genetics, Cell and Immunobiology, Semmelweis University, 1089 Budapest, Hungary;
| | - Ágnes Kittel
- Institute of Experimental Medicine, Eötvös Loránd Research Network, 1083 Budapest, Hungary;
| | - Viktor Nabil Sayour
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; (C.K.); (B.K.); (V.N.S.); (A.G.); (C.P.); (S.H.); (P.F.)
| | - Anikó Görbe
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; (C.K.); (B.K.); (V.N.S.); (A.G.); (C.P.); (S.H.); (P.F.)
- MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary
- Pharmahungary Group, 6722 Szeged, Hungary
| | - Csilla Pelyhe
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; (C.K.); (B.K.); (V.N.S.); (A.G.); (C.P.); (S.H.); (P.F.)
| | - Szabolcs Hambalkó
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; (C.K.); (B.K.); (V.N.S.); (A.G.); (C.P.); (S.H.); (P.F.)
| | - Lucia Kindernay
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia; (M.J.); (K.F.); (L.K.); (M.B.)
| | - Miroslav Barančík
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia; (M.J.); (K.F.); (L.K.); (M.B.)
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; (C.K.); (B.K.); (V.N.S.); (A.G.); (C.P.); (S.H.); (P.F.)
- MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary
- Pharmahungary Group, 6722 Szeged, Hungary
| | - Monika Barteková
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia; (M.J.); (K.F.); (L.K.); (M.B.)
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, 81372 Bratislava, Slovakia
| | - Zoltán Giricz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; (C.K.); (B.K.); (V.N.S.); (A.G.); (C.P.); (S.H.); (P.F.)
- Pharmahungary Group, 6722 Szeged, Hungary
| |
Collapse
|
35
|
Oyama Y, Walker LA, Eckle T. Targeting circadian PER2 as therapy in myocardial ischemia and reperfusion injury. Chronobiol Int 2021; 38:1262-1273. [PMID: 34034593 PMCID: PMC8355134 DOI: 10.1080/07420528.2021.1928160] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/30/2021] [Accepted: 05/04/2021] [Indexed: 01/22/2023]
Abstract
The cycle of day and night dominates life on earth. Therefore, almost all living organisms adopted a molecular clock linked to the light-dark cycles. It is now well established that this molecular clock is crucial for human health and wellbeing. Disruption of the molecular clockwork directly results in a myriad of disorders, including cardiovascular diseases. Further, the onset of many cardiovascular diseases such as acute myocardial infarction exhibits a circadian periodicity with worse outcomes in the early morning hours. Based on these observations, the research community became interested in manipulating the molecular clock to treat cardiovascular diseases. In recent years, several exciting discoveries of pharmacological agents or molecular mechanisms targeting the molecular clockwork have paved the way for circadian medicine's arrival in cardiovascular diseases. The current review will outline the most recent circadian therapeutic advances related to the circadian rhythm protein Period2 (PER2) to treat myocardial ischemia and summarize future research in the respective field.
Collapse
Affiliation(s)
- Yoshimasa Oyama
- Department of Anesthesiology, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
- Department of Anesthesiology and Intensive Care Medicine, Oita University Faculty of Medicine, Oita, Japan
| | - Lori A Walker
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Tobias Eckle
- Department of Anesthesiology, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
- Department of Cell and Developmental Biology, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
36
|
Marracino L, Fortini F, Bouhamida E, Camponogara F, Severi P, Mazzoni E, Patergnani S, D’Aniello E, Campana R, Pinton P, Martini F, Tognon M, Campo G, Ferrari R, Vieceli Dalla Sega F, Rizzo P. Adding a "Notch" to Cardiovascular Disease Therapeutics: A MicroRNA-Based Approach. Front Cell Dev Biol 2021; 9:695114. [PMID: 34527667 PMCID: PMC8435685 DOI: 10.3389/fcell.2021.695114] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 08/09/2021] [Indexed: 12/18/2022] Open
Abstract
Dysregulation of the Notch pathway is implicated in the pathophysiology of cardiovascular diseases (CVDs), but, as of today, therapies based on the re-establishing the physiological levels of Notch in the heart and vessels are not available. A possible reason is the context-dependent role of Notch in the cardiovascular system, which would require a finely tuned, cell-specific approach. MicroRNAs (miRNAs) are short functional endogenous, non-coding RNA sequences able to regulate gene expression at post-transcriptional levels influencing most, if not all, biological processes. Dysregulation of miRNAs expression is implicated in the molecular mechanisms underlying many CVDs. Notch is regulated and regulates a large number of miRNAs expressed in the cardiovascular system and, thus, targeting these miRNAs could represent an avenue to be explored to target Notch for CVDs. In this Review, we provide an overview of both established and potential, based on evidence in other pathologies, crosstalks between miRNAs and Notch in cellular processes underlying atherosclerosis, myocardial ischemia, heart failure, calcification of aortic valve, and arrhythmias. We also discuss the potential advantages, as well as the challenges, of using miRNAs for a Notch-based approach for the diagnosis and treatment of the most common CVDs.
Collapse
Affiliation(s)
- Luisa Marracino
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | | | - Esmaa Bouhamida
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Francesca Camponogara
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Paolo Severi
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Elisa Mazzoni
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Simone Patergnani
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Emanuele D’Aniello
- Cardiovascular Institute, Azienda Ospedaliero-Universitaria di Ferrara, Ferrara, Italy
| | - Roberta Campana
- Cardiovascular Institute, Azienda Ospedaliero-Universitaria di Ferrara, Ferrara, Italy
| | - Paolo Pinton
- Maria Cecilia Hospital, GVM Care & Research, Ravenna, Italy
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Fernanda Martini
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Mauro Tognon
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Gianluca Campo
- Maria Cecilia Hospital, GVM Care & Research, Ravenna, Italy
- Cardiovascular Institute, Azienda Ospedaliero-Universitaria di Ferrara, Ferrara, Italy
| | - Roberto Ferrari
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, Ferrara, Italy
- Maria Cecilia Hospital, GVM Care & Research, Ravenna, Italy
| | | | - Paola Rizzo
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, Ferrara, Italy
- Maria Cecilia Hospital, GVM Care & Research, Ravenna, Italy
| |
Collapse
|
37
|
Ullah K, Wu R. Hypoxia-Inducible Factor Regulates Endothelial Metabolism in Cardiovascular Disease. Front Physiol 2021; 12:670653. [PMID: 34290616 PMCID: PMC8287728 DOI: 10.3389/fphys.2021.670653] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/13/2021] [Indexed: 12/30/2022] Open
Abstract
Endothelial cells (ECs) form a physical barrier between the lumens and vascular walls of arteries, veins, capillaries, and lymph vessels; thus, they regulate the extravasation of nutrients and oxygen from the circulation into the perivascular space and participate in mechanisms that maintain cardiovascular homeostasis and promote tissue growth and repair. Notably, their role in tissue repair is facilitated, at least in part, by their dependence on glycolysis for energy production, which enables them to resist hypoxic damage and promote angiogenesis in ischemic regions. ECs are also equipped with a network of oxygen-sensitive molecules that collectively activate the response to hypoxic injury, and the master regulators of the hypoxia response pathway are hypoxia-inducible factors (HIFs). HIFs reinforce the glycolytic dependence of ECs under hypoxic conditions, but whether HIF activity attenuates or exacerbates the progression and severity of cardiovascular dysfunction varies depending on the disease setting. This review summarizes how HIF regulates the metabolic and angiogenic activity of ECs under both normal and hypoxic conditions and in a variety of diseases that are associated with cardiovascular complications.
Collapse
Affiliation(s)
- Karim Ullah
- Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Rongxue Wu
- Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL, United States
| |
Collapse
|
38
|
Phosphoproteomic response of cardiac endothelial cells to ischemia and ultrasound. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2021; 1869:140683. [PMID: 34119693 DOI: 10.1016/j.bbapap.2021.140683] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 04/30/2021] [Accepted: 06/04/2021] [Indexed: 12/13/2022]
Abstract
Myocardial infarction and subsequent therapeutic interventions activate numerous intracellular cascades in every constituent cell type of the heart. Endothelial cells produce several protective compounds in response to therapeutic ultrasound, under both normoxic and ischemic conditions. How endothelial cells sense ultrasound and convert it to a beneficial biological response is not known. We adopted a global, unbiased phosphoproteomics approach aimed at understanding how endothelial cells respond to ultrasound. Here, we use primary cardiac endothelial cells to explore the cellular signaling events underlying the response to ischemia-like cellular injury and ultrasound exposure in vitro. Enriched phosphopeptides were analyzed with a high mass accuracy liquid chromatrography (LC) - tandem mass spectrometry (MS/MS) proteomic platform, yielding multiple alterations in both total protein levels and phosphorylation events in response to ischemic injury and ultrasound. Application of pathway algorithms reveals numerous protein networks recruited in response to ultrasound including those regulating RNA splicing, cell-cell interactions and cytoskeletal organization. Our dataset also permits the informatic prediction of potential kinases responsible for the modifications detected. Taken together, our findings begin to reveal the endothelial proteomic response to ultrasound and suggest potential targets for future studies of the protective effects of ultrasound in the ischemic heart.
Collapse
|
39
|
Yang H, Su J, Meng W, Chen X, Xu Y, Sun B. MiR-518a-5p Targets GZMB to Extenuate Vascular Endothelial Cell Injury Induced by Hypoxia-Reoxygenation and Thereby Improves Myocardial Ischemia. Int Heart J 2021; 62:658-665. [PMID: 33994508 DOI: 10.1536/ihj.20-619] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
To probe the function of miR-518a-5p/Granzyme B (GZMB) in hypoxia/reoxygenation (H/R) -induced vascular endothelial cell injury.The key genes of myocardial infarction were screened by bioinformatic methods. The upstream micro RNAs (miRNAs) of GZMB were predicted by TargetScan. The binding of miR-518a-5p to GZMB was verified with luciferase reporter assay. The H/R model was constructed with human vascular endothelial cell (HUVEC) in vitro. Cell Counting Kit-8 (CCK8) assay was performed to detect cell proliferation. Western blot was utilized to evaluate the levels of indicated proteins.GZMB was up-regulated in patients with myocardial infarction and identified as the key gene by the bioinformatics analysis. Then the prediction from TargetScan indicated that miR-518a-5p, which is down-regulated in myocardial infarction patients, might be the potential upstream miRNA for GZMB. The following experiments verified that miR-518a-5p could bind to the 3'UTR of GZMB and negatively modulates GZMB expression. More importantly, the miR-518a-5p mimic enhanced cell proliferation and repressed apoptosis of H/R-injured HUVEC cells by inhibiting GZMB expression.We proved that miR-518a-5p could partly attenuate H/R-induced HUVEC cell injury by targeting GZMB, and perhaps the miR-518a-5p/GZMB axis could be potential therapeutic targets for myocardial infarction.
Collapse
Affiliation(s)
- Hui Yang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Harbin Medical University
| | - Jingjing Su
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Harbin Medical University
| | - Weixin Meng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Harbin Medical University
| | - Xiaoya Chen
- Department of Ultrasound, The First Affiliated Hospital of Harbin Medical University
| | - Ying Xu
- Editorial Department, Journal of Harbin Medical University
| | - Bo Sun
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Harbin Medical University
| |
Collapse
|
40
|
H 2S Pretreatment Is Promigratory and Decreases Ischemia/Reperfusion Injury in Human Microvascular Endothelial Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8886666. [PMID: 33953839 PMCID: PMC8068530 DOI: 10.1155/2021/8886666] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/23/2020] [Accepted: 04/01/2021] [Indexed: 12/03/2022]
Abstract
Endothelial cell injury and vascular function strongly correlate with cardiac function following ischemia/reperfusion injury. Several studies indicate that endothelial cells are more sensitive to ischemia/reperfusion compared to cardiomyocytes and are critical mediators of cardiac ischemia/reperfusion injury. H2S is involved in the regulation of cardiovascular system homeostasis and can act as a cytoprotectant during ischemia/reperfusion. Activation of ERK1/2 in endothelial cells after H2S stimulation exerts an enhancement of angiogenesis while its inhibition significantly decreases H2S cardioprotective effects. In this work, we investigated how H2S pretreatment for 24 hours prevents the ischemia/reperfusion injury and promotes angiogenesis on microvascular endothelial cells following an ischemia/reperfusion protocol in vitro, using a hypoxic chamber and ischemic buffer to simulate the ischemic event. H2S preconditioning positively affected cell viability and significantly increased endothelial cell migration when treated with 1 μM H2S. Furthermore, mitochondrial function was preserved when cells were preconditioned. Since ERK1/2 phosphorylation was extremely enhanced in ischemia/reperfusion condition, we inhibited ERK both directly and indirectly to verify how H2S triggers this pathway in endothelial cells. Taken together, our data suggest that H2S treatment 24 hours before the ischemic insult protects endothelial cells from ischemia/reperfusion injury and eventually decreases myocardial injury.
Collapse
|
41
|
Sharma P, Wang X, Ming CLC, Vettori L, Figtree G, Boyle A, Gentile C. Considerations for the Bioengineering of Advanced Cardiac In Vitro Models of Myocardial Infarction. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2003765. [PMID: 33464713 DOI: 10.1002/smll.202003765] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 09/03/2020] [Indexed: 06/12/2023]
Abstract
Despite the latest advances in cardiovascular biology and medicine, myocardial infarction (MI) remains one of the major causes of deaths worldwide. While reperfusion of the myocardium is critical to limit the ischemic damage typical of a MI event, it causes detrimental morphological and functional changes known as "reperfusion injury." This complex scenario is poorly represented in currently available models of ischemia/reperfusion injury, leading to a poor translation of findings from the bench to the bedside. However, more recent bioengineered in vitro models of the human heart represent more clinically relevant tools to prevent and treat MI in patients. These include 3D cultures of cardiac cells, the use of patient-derived stem cells, and 3D bioprinting technology. This review aims at highlighting the major features typical of a heart attack while comparing current in vitro, ex vivo, and in vivo models. This information has the potential to further guide in developing novel advanced in vitro cardiac models of ischemia/reperfusion injury. It may pave the way for the generation of advanced pathophysiological cardiac models with the potential to develop personalized therapies.
Collapse
Affiliation(s)
- Poonam Sharma
- Faculty of Medicine and Health, University of Newcastle, Newcastle, NSW, 2308, Australia
- School of Medicine and Public Health, University of Sydney, Sydney, NSW, 2000, Australia
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, 2065, Australia
- School of Biomedical Engineering/FEIT, University of Technology Sydney, Building 11, Level 10, Room 115, 81 Broadway, Ultimo, NSW, 2007, Australia
| | - Xiaowei Wang
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - Clara Liu Chung Ming
- School of Biomedical Engineering/FEIT, University of Technology Sydney, Building 11, Level 10, Room 115, 81 Broadway, Ultimo, NSW, 2007, Australia
| | - Laura Vettori
- School of Biomedical Engineering/FEIT, University of Technology Sydney, Building 11, Level 10, Room 115, 81 Broadway, Ultimo, NSW, 2007, Australia
| | - Gemma Figtree
- School of Medicine and Public Health, University of Sydney, Sydney, NSW, 2000, Australia
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, 2065, Australia
| | - Andrew Boyle
- Faculty of Medicine and Health, University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Carmine Gentile
- School of Medicine and Public Health, University of Sydney, Sydney, NSW, 2000, Australia
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, 2065, Australia
- School of Biomedical Engineering/FEIT, University of Technology Sydney, Building 11, Level 10, Room 115, 81 Broadway, Ultimo, NSW, 2007, Australia
| |
Collapse
|
42
|
Dagher O, Mury P, Thorin-Trescases N, Noly PE, Thorin E, Carrier M. Therapeutic Potential of Quercetin to Alleviate Endothelial Dysfunction in Age-Related Cardiovascular Diseases. Front Cardiovasc Med 2021; 8:658400. [PMID: 33860002 PMCID: PMC8042157 DOI: 10.3389/fcvm.2021.658400] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/05/2021] [Indexed: 12/12/2022] Open
Abstract
The vascular endothelium occupies a catalog of functions that contribute to the homeostasis of the cardiovascular system. It is a physically active barrier between circulating blood and tissue, a regulator of the vascular tone, a biochemical processor and a modulator of coagulation, inflammation, and immunity. Given these essential roles, it comes to no surprise that endothelial dysfunction is prodromal to chronic age-related diseases of the heart and arteries, globally termed cardiovascular diseases (CVD). An example would be ischemic heart disease (IHD), which is the main cause of death from CVD. We have made phenomenal advances in treating CVD, but the aging endothelium, as it senesces, always seems to out-run the benefits of medical and surgical therapies. Remarkably, many epidemiological studies have detected a correlation between a flavonoid-rich diet and a lower incidence of mortality from CVD. Quercetin, a member of the flavonoid class, is a natural compound ubiquitously found in various food sources such as fruits, vegetables, seeds, nuts, and wine. It has been reported to have a wide range of health promoting effects and has gained significant attention over the years. A growing body of evidence suggests quercetin could lower the risk of IHD by mitigating endothelial dysfunction and its risk factors, such as hypertension, atherosclerosis, accumulation of senescent endothelial cells, and endothelial-mesenchymal transition (EndoMT). In this review, we will explore these pathophysiological cascades and their interrelation with endothelial dysfunction. We will then present the scientific evidence to quercetin's anti-atherosclerotic, anti-hypertensive, senolytic, and anti-EndoMT effects. Finally, we will discuss the prospect for its clinical use in alleviating myocardial ischemic injuries in IHD.
Collapse
Affiliation(s)
- Olina Dagher
- Department of Cardiac Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Surgery, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.,Center for Research, Montreal Heart Institute, Montreal, QC, Canada
| | - Pauline Mury
- Center for Research, Montreal Heart Institute, Montreal, QC, Canada
| | | | - Pierre Emmanuel Noly
- Department of Surgery, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.,Center for Research, Montreal Heart Institute, Montreal, QC, Canada
| | - Eric Thorin
- Department of Surgery, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.,Center for Research, Montreal Heart Institute, Montreal, QC, Canada
| | - Michel Carrier
- Department of Surgery, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.,Center for Research, Montreal Heart Institute, Montreal, QC, Canada
| |
Collapse
|
43
|
Wang R, Wang M, Zhou J, Wu D, Ye J, Sun G, Sun X. Saponins in Chinese Herbal Medicine Exerts Protection in Myocardial Ischemia-Reperfusion Injury: Possible Mechanism and Target Analysis. Front Pharmacol 2021; 11:570867. [PMID: 33597866 PMCID: PMC7883640 DOI: 10.3389/fphar.2020.570867] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 10/28/2020] [Indexed: 12/21/2022] Open
Abstract
Myocardial ischemia is a high-risk disease among middle-aged and senior individuals. After thrombolytic therapy, heart tissue can potentially suffer further damage, which is called myocardial ischemia-reperfusion injury (MIRI). At present, the treatment methods and drugs for MIRI are scarce and cannot meet the current clinical needs. The mechanism of MIRI involves the interaction of multiple factors, and the current research hotspots mainly include oxidative stress, inflammation, calcium overload, energy metabolism disorders, pyroptosis, and ferroptosis. Traditional Chinese medicine (TCM) has multiple targets and few toxic side effects; clinical preparations containing Panax ginseng C. A. Mey., Panax notoginseng (Burk.) F. H. Chen, Aralia chinensis L., cardioprotection, and other Chinese herbal medicines have been used to treat patients with coronary heart disease, angina pectoris, and other cardiovascular diseases. Studies have shown that saponins are the main active substances in TCMs containing Panax ginseng C. A. Mey., Panax notoginseng (Burk.) F. H. Chen, Aralia chinensis L., and Radix astragali. In the present review, we sorted the saponin components with anti-MIRI effects and their regulatory mechanisms. Each saponin can play a cardioprotective role via multiple mechanisms, and the signaling pathways involved in different saponins are not the same. We found that more active saponins in Panax ginseng C. A. Mey. are mainly dammar-type structures and have a strong regulatory effect on energy metabolism. The highly active saponin components of Aralia chinensis L. are oleanolic acid structures, which have significant regulatory effects on calcium homeostasis. Therefore, saponins in Chinese herbal medicine provide a broad application prospect for the development of highly effective and low-toxicity anti-MIRI drugs.
Collapse
Affiliation(s)
- Ruiying Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, China
| | - Min Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, China
| | - Jiahui Zhou
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, China
| | - Daoshun Wu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, China
| | - Jingxue Ye
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guibo Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
44
|
Margraf A, Ludwig N, Zarbock A, Rossaint J. Systemic Inflammatory Response Syndrome After Surgery: Mechanisms and Protection. Anesth Analg 2020; 131:1693-1707. [PMID: 33186158 DOI: 10.1213/ane.0000000000005175] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The immune system is an evolutionary hallmark of higher organisms that defends the host against invading pathogens and exogenous infections. This defense includes the recruitment of immune cells to the site of infection and the initiation of an inflammatory response to contain and eliminate pathogens. However, an inflammatory response may also be triggered by noninfectious stimuli such as major surgery, and, in case of an overshooting, still not comprehensively understood reaction, lead to tissue destruction and organ dysfunction. Unfortunately, in some cases, the immune system may not effectively distinguish between stimuli elicited by major surgery, which ideally should only require a modest inflammatory response, and those elicited by trauma or pathogenic infection. Surgical procedures thus represent a potential trigger for systemic inflammation that causes the secretion of proinflammatory cytokines, endothelial dysfunction, glycocalyx damage, activation of neutrophils, and ultimately tissue and multisystem organ destruction. In this review, we discuss and summarize currently available mechanistic knowledge on surgery-associated systemic inflammation, demarcation toward other inflammatory complications, and possible therapeutic options. These options depend on uncovering the underlying mechanisms and could include pharmacologic agents, remote ischemic preconditioning protocols, cytokine blockade or clearance, and optimization of surgical procedures, anesthetic regimens, and perioperative inflammatory diagnostic assessment. Currently, a large gap between basic science and clinically confirmed data exists due to a limited evidence base of translational studies. We thus summarize important steps toward the understanding of the precise time- and space-regulated processes in systemic perioperative inflammation.
Collapse
Affiliation(s)
- Andreas Margraf
- From the Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | | | | | | |
Collapse
|
45
|
Mentkowski KI, Euscher LM, Patel A, Alevriadou BR, Lang JK. Monocyte recruitment and fate specification after myocardial infarction. Am J Physiol Cell Physiol 2020; 319:C797-C806. [PMID: 32877204 DOI: 10.1152/ajpcell.00330.2020] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Monocytes are critical mediators of the inflammatory response following myocardial infarction (MI) and ischemia-reperfusion injury. They are involved in both initiation and resolution of inflammation and play an integral role in cardiac repair. The antagonistic nature of their function is dependent on their subset heterogeneity and biphasic response following injury. New advancements in single-cell transcriptomics and mass cytometry have allowed us to identify smaller, transcriptionally distinct clusters that may have functional relevance in disease and homeostasis. Additionally, recent insights into the spatiotemporal dynamics of monocytes following ischemic injury and their subsequent interactions with the endothelium and other immune cells reveal a complex interplay between monocytes and the cardiac milieu. In this review, we highlight recent findings on monocyte functional heterogeneity, present new mechanistic insight into monocyte recruitment and fate specification following MI, and discuss promising therapeutic avenues targeting monocytes for the treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Kyle I Mentkowski
- Department of Medicine, Division of Cardiology, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York.,Department of Biomedical Engineering, University at Buffalo, Buffalo, New York
| | - Lindsey M Euscher
- Department of Medicine, Division of Cardiology, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York.,Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York
| | - Akshar Patel
- Department of Biomedical Engineering, University at Buffalo, Buffalo, New York
| | - B Rita Alevriadou
- Department of Biomedical Engineering, University at Buffalo, Buffalo, New York
| | - Jennifer K Lang
- Department of Medicine, Division of Cardiology, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York.,Department of Biomedical Engineering, University at Buffalo, Buffalo, New York.,Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York.,Veterans Affairs Western New York Healthcare System, Buffalo, New York
| |
Collapse
|
46
|
Hao D, Liu R, Gao K, He C, He S, Zhao C, Sun G, Farmer DL, Panitch A, Lam KS, Wang A. Developing an Injectable Nanofibrous Extracellular Matrix Hydrogel With an Integrin αvβ3 Ligand to Improve Endothelial Cell Survival, Engraftment and Vascularization. Front Bioeng Biotechnol 2020; 8:890. [PMID: 32850742 PMCID: PMC7403189 DOI: 10.3389/fbioe.2020.00890] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 07/10/2020] [Indexed: 01/01/2023] Open
Abstract
Endothelial cell (EC) transplantation via injectable collagen hydrogel has received much attention as a potential treatment for various vascular diseases. However, the therapeutic effect of transplanted ECs is limited by their poor viability, which partially occurs as a result of cellular apoptosis triggered by the insufficient cell-extracellular matrix (ECM) engagement. Integrin binding to the ECM is crucial for cell anchorage to the surrounding matrix, cell spreading and migration, and further activation of intracellular signaling pathways. Although collagen contains several different types of integrin binding sites, it still lacks sufficient specific binding sites for ECs. Previously, using one-bead one-compound (OBOC) combinatorial technology, we identified LXW7, an integrin αvβ3 ligand, which possessed a strong binding affinity to and enhanced functionality of ECs. In this study, to improve the EC-matrix interaction, we developed an approach to molecularly conjugate LXW7 to the collagen backbone, via a collagen binding peptide SILY, in order to increase EC specific integrin binding sites on the collagen hydrogel. Results showed that in the in vitro 2-dimensional (2D) culture model, the LXW7-treated collagen surface significantly improved EC attachment and survival and decreased caspase 3 activity in an ischemic-mimicking environment. In the in vitro 3-dimensional (3D) culture model, LXW7-modified collagen hydrogel significantly improved EC spreading, proliferation, and survival. In a mouse subcutaneous implantation model, LXW7-modified collagen hydrogel improved the engraftment of transplanted ECs and supported ECs to form vascular network structures. Therefore, LXW7-functionalized collagen hydrogel has shown promising potential to improve vascularization in tissue regeneration and may be used as a novel tool for EC delivery and the treatment of vascular diseases.
Collapse
Affiliation(s)
- Dake Hao
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, CA, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, United States
| | - Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Kewa Gao
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, CA, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, United States
| | - Chuanchao He
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Siqi He
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, CA, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, United States
| | - Cunyi Zhao
- Department of Biological and Agricultural Engineering, University of California, Davis, Davis, CA, United States
| | - Gang Sun
- Department of Biological and Agricultural Engineering, University of California, Davis, Davis, CA, United States
| | - Diana L. Farmer
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, CA, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, United States
| | - Alyssa Panitch
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, CA, United States
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States
| | - Kit S. Lam
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Aijun Wang
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, CA, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, United States
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States
| |
Collapse
|
47
|
Kavanagh DPJ, Lokman AB, Neag G, Colley A, Kalia N. Imaging the injured beating heart intravitally and the vasculoprotection afforded by haematopoietic stem cells. Cardiovasc Res 2020; 115:1918-1932. [PMID: 31062860 PMCID: PMC6803816 DOI: 10.1093/cvr/cvz118] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 03/20/2019] [Accepted: 05/01/2019] [Indexed: 12/16/2022] Open
Abstract
Aims Adequate microcirculatory perfusion, and not just opening of occluded arteries, is critical to salvage heart tissue following myocardial infarction. However, the degree of microvascular perfusion taking place is not known, limited primarily by an inability to directly image coronary microcirculation in a beating heart in vivo. Haematopoietic stem/progenitor cells (HSPCs) offer a potential therapy but little is known about their homing dynamics at a cellular level and whether they protect coronary microvessels. This study used intravital microscopy to image the anaesthetized mouse beating heart microcirculation following stabilization. Methods and results A 3D-printed stabilizer was attached to the ischaemia–reperfusion injured (IRI) beating heart. The kinetics of neutrophil, platelet and HSPC recruitment, as well as functional capillary density (FCD), was imaged post-reperfusion. Laser speckle contrast imaging (LSCI) was used for the first time to monitor ventricular blood flow in beating hearts. Sustained hyperaemic responses were measured throughout reperfusion, initially indicating adequate flow resumption. Intravital microscopy confirmed large vessel perfusion but demonstrated poor transmission of flow to downstream coronary microvessels. Significant neutrophil adhesion and microthrombus formation occurred within capillaries with the latter occluding them, resulting in patchy perfusion and reduced FCD. Interestingly, ‘patrolling’ neutrophils were also observed in capillaries. Haematopoietic stem/progenitor cells readily trafficked through the heart but local retention was poor. Despite this, remarkable anti-thromboinflammatory effects were observed, consequently improving microvascular perfusion. Conclusion We present a novel approach for imaging multiple microcirculatory perturbations in the beating heart with LSCI assessment of blood flow. Despite deceptive hyperaemic responses, increased microcirculatory flow heterogeneity was seen, with non-perfused areas interspersed with perfused areas. Microthrombi, rather than neutrophils, appeared to be the major causative factor. We further applied this technique to demonstrate local stem cell presence is not a pre-requisite to confer vasculoprotection. This is the first detailed in vivo characterization of coronary microcirculatory responses post-reperfusion injury.
Collapse
Affiliation(s)
- Dean P J Kavanagh
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Adam B Lokman
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Georgiana Neag
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Abigail Colley
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Neena Kalia
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
48
|
Feliu C, Peyret H, Brassart-Pasco S, Oszust F, Poitevin G, Nguyen P, Millart H, Djerada Z. Ticagrelor Prevents Endothelial Cell Apoptosis through the Adenosine Signalling Pathway in the Early Stages of Hypoxia. Biomolecules 2020; 10:biom10050740. [PMID: 32397519 PMCID: PMC7277469 DOI: 10.3390/biom10050740] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/05/2020] [Accepted: 05/07/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Several studies have reported the beneficial effects of anti-platelet drugs in cardioprotection against ischaemia-reperfusion injuries. To date, no studies have focused on the indirect cytoprotective effects of ticagrelor via adenosine receptor on the endothelium. METHOD By evaluating cell viability and cleaved caspase 3 expression, we validated a model of endothelial cell apoptosis induced by hypoxia. In hypoxic endothelial cells treated with ticagrelor, we quantified the extracellular concentration of adenosine, and then we studied the involvement of adenosine pathways in the cytoprotective effect of ticagrelor. RESULTS Our results showed that 10 µM ticagrelor induced an anti-apoptotic effect in our model associated with an increase of extracellular adenosine concentration. Similar experiments were conducted with cangrelor but did not demonstrate an anti-apoptotic effect. We also found that A2B and A3 adenosine receptors were involved in the anti-apoptotic effect of ticagrelor in endothelial cells exposed to 2 h of hypoxia stress. CONCLUSION we described an endothelial cytoprotective mechanism of ticagrelor against hypoxia stress, independent of blood elements. We highlighted a mechanism triggered mainly by the increased extracellular bioavailability of adenosine, which activates A2B and A3 receptors on the endothelium.
Collapse
Affiliation(s)
- Catherine Feliu
- Department of Pharmacology, Hémostase et Remodelage Vasculaire post-Ischémie (HERVI) E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims CEDEX, France; (C.F.); (H.P.); (F.O.); (H.M.)
| | - Hélène Peyret
- Department of Pharmacology, Hémostase et Remodelage Vasculaire post-Ischémie (HERVI) E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims CEDEX, France; (C.F.); (H.P.); (F.O.); (H.M.)
| | - Sylvie Brassart-Pasco
- UMR CNRS/URCA 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Reims University Hospital, SFR CAP-santé, 51, rue Cognacq-Jay, 51095 Reims CEDEX, France;
| | - Floriane Oszust
- Department of Pharmacology, Hémostase et Remodelage Vasculaire post-Ischémie (HERVI) E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims CEDEX, France; (C.F.); (H.P.); (F.O.); (H.M.)
| | - Gaël Poitevin
- Laboratory of Hematology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims CEDEX, France; (G.P.); (P.N.)
| | - Philippe Nguyen
- Laboratory of Hematology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims CEDEX, France; (G.P.); (P.N.)
| | - Hervé Millart
- Department of Pharmacology, Hémostase et Remodelage Vasculaire post-Ischémie (HERVI) E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims CEDEX, France; (C.F.); (H.P.); (F.O.); (H.M.)
| | - Zoubir Djerada
- Department of Pharmacology, Hémostase et Remodelage Vasculaire post-Ischémie (HERVI) E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims CEDEX, France; (C.F.); (H.P.); (F.O.); (H.M.)
- Correspondence: ; Tel.: +33-3-26-83-27-82; Fax: +33-3-26-78-84-56
| |
Collapse
|
49
|
Kelm NQ, Beare JE, Weber GJ, LeBlanc AJ. Thrombospondin-1 mediates Drp-1 signaling following ischemia reperfusion in the aging heart. FASEB Bioadv 2020; 2:304-314. [PMID: 32395703 PMCID: PMC7211039 DOI: 10.1096/fba.2019-00090] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 11/07/2019] [Accepted: 02/19/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Ischemia reperfusion (IR) injury leads to activation of dynamin-related protein (Drp-1), causing mitochondrial fission and generation of reactive oxygen species (ROS), but the molecular mechanisms that activate Drp-1 are not known. The purpose of this study was to establish a link between Thbs-1 and fission protein (Drp-1) through Pgc-1α following IR in advancing age. METHODS Female Fischer-344 rats were divided into four groups: Young Control, Young + IR, Old Control, and Old + IR. Heart function and coronary flow were evaluated at baseline and 72 hours after IR, hearts were explanted and mitochondrial ROS generation was measured using MitoPY1, as well as protein levels of Thbs-1, Pgc-1α, and Drp-1. In vitro, rat aortic endothelial cells (RAEC) were treated with siRNA or plasmid for Pgc-1α to evaluate Pgc-1α effect on Drp-1. RESULTS Mitochondrial ROS generation in heart tissue increased in both age groups following IR. Old animals exhibited diastolic dysfunction at baseline; after IR they displayed reduced systolic function and exacerbated diastolic dysfunction compared to young controls. IR increased Thbs-1 and Drp-1 expression in young and old hearts compared to control. siRNA to Pgc-1α enhanced levels of Drp-1 in RAECs and increased ROS generation after hypoxia, while Pgc-1α plasmid ameliorates Drp-1 expression in the presence of exogenous Thbs-1. CONCLUSION These results highlight a novel signaling pathway by which Thbs-1 regulates mitochondrial fission protein (Drp-1) and ROS generation during hypoxia, and presumably, following IR. Inhibiting Thbs-1 immediately after IR may prevent Drp-1-mediated mitochondrial fission and is likely to improve the diastolic function of the heart by reducing ROS-mediated cardiomyocyte damage in the aged population.
Collapse
Affiliation(s)
- Natia Q. Kelm
- Cardiovascular Innovation InstituteUniversity of LouisvilleLouisvilleKYUSA
| | - Jason E. Beare
- Cardiovascular Innovation InstituteUniversity of LouisvilleLouisvilleKYUSA
- Kentucky Spinal Cord Injury Research CenterUniversity of LouisvilleLouisvilleKYUSA
| | | | - Amanda J. LeBlanc
- Cardiovascular Innovation InstituteUniversity of LouisvilleLouisvilleKYUSA
- Department of PhysiologyUniversity of LouisvilleLouisvilleKYUSA
| |
Collapse
|
50
|
Bartman CM, Eckle T. Circadian-Hypoxia Link and its Potential for Treatment of Cardiovascular Disease. Curr Pharm Des 2020; 25:1075-1090. [PMID: 31096895 DOI: 10.2174/1381612825666190516081612] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/03/2019] [Indexed: 12/29/2022]
Abstract
Throughout the evolutionary time, all organisms and species on Earth evolved with an adaptation to consistent oscillations of sunlight and darkness, now recognized as 'circadian rhythm.' Single-cellular to multisystem organisms use circadian biology to synchronize to the external environment and provide predictive adaptation to changes in cellular homeostasis. Dysregulation of circadian biology has been implicated in numerous prevalent human diseases, and subsequently targeting the circadian machinery may provide innovative preventative or treatment strategies. Discovery of 'peripheral circadian clocks' unleashed widespread investigations into the potential roles of clock biology in cellular, tissue, and organ function in healthy and diseased states. Particularly, oxygen-sensing pathways (e.g. hypoxia inducible factor, HIF1), are critical for adaptation to changes in oxygen availability in diseases such as myocardial ischemia. Recent investigations have identified a connection between the circadian rhythm protein Period 2 (PER2) and HIF1A that may elucidate an evolutionarily conserved cellular network that can be targeted to manipulate metabolic function in stressed conditions like hypoxia or ischemia. Understanding the link between circadian and hypoxia pathways may provide insights and subsequent innovative therapeutic strategies for patients with myocardial ischemia. This review addresses our current understanding of the connection between light-sensing pathways (PER2), and oxygen-sensing pathways (HIF1A), in the context of myocardial ischemia and lays the groundwork for future studies to take advantage of these two evolutionarily conserved pathways in the treatment of myocardial ischemia.
Collapse
Affiliation(s)
- Colleen Marie Bartman
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, Graduate Training Program in Cell Biology, Stem Cells, and Development, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Tobias Eckle
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, Graduate Training Program in Cell Biology, Stem Cells, and Development, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|