1
|
Hassanpour M, Salybkov AA, Kobayashi S, Asahara T. Anti-inflammatory Prowess of endothelial progenitor cells in the realm of biology and medicine. NPJ Regen Med 2024; 9:27. [PMID: 39349482 PMCID: PMC11442670 DOI: 10.1038/s41536-024-00365-z] [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: 12/15/2023] [Accepted: 08/23/2024] [Indexed: 10/02/2024] Open
Abstract
Endothelial inflammation plays a crucial role in vascular-related diseases, a leading cause of global mortality. Among various cellular players, endothelial progenitor cells (EPCs) emerge as non-differentiated endothelial cells circulating in the bloodstream. Recent evidence highlights the transformative role of EPCs in shifting from an inflammatory/immunosuppressive crisis to an anti-inflammatory/immunomodulatory response. Despite the importance of these functions, the regulatory mechanisms governing EPC activities and their physiological significance in vascular regenerative medicine remain elusive. Surprisingly, the current literature lacks a comprehensive review of EPCs' effects on inflammatory processes. This narrative review aims to fill this gap by exploring the cutting-edge role of EPCs against inflammation, from molecular intricacies to broader medical perspectives. By examining how EPCs modulate inflammatory responses, we aim to unravel their anti-inflammatory significance in vascular regenerative medicine, deepening insights into EPCs' molecular mechanisms and guiding future therapeutic strategies targeting vascular-related diseases.
Collapse
Affiliation(s)
- Mehdi Hassanpour
- Shonan Research, Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
- Center for Cell therapy & Regenerative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
| | - Amankeldi A Salybkov
- Shonan Research, Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
- Center for Cell therapy & Regenerative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
| | - Shuzo Kobayashi
- Shonan Research, Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
| | - Takayuki Asahara
- Shonan Research, Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan.
- Center for Cell therapy & Regenerative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan.
| |
Collapse
|
2
|
Gao X, Ma C, Liang S, Chen M, He Y, Lei W. PANoptosis: Novel insight into regulated cell death and its potential role in cardiovascular diseases (Review). Int J Mol Med 2024; 54:74. [PMID: 38963054 PMCID: PMC11254103 DOI: 10.3892/ijmm.2024.5398] [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: 01/25/2024] [Accepted: 05/08/2024] [Indexed: 07/05/2024] Open
Abstract
PANoptosis, a complex form of proinflammatory programmed cell death, including apoptosis, pyroptosis and necroptosis, has been an emerging concept in recent years that has been widely reported in cancer, infectious diseases and neurological disorders. Cardiovascular diseases (CVDs) are an important global health problem, posing a serious threat to individuals' lives. An increasing body of research shows that inflammation has a pivotal role in CVDs, which provides an important theoretical basis for PANoptosis to promote the progression of CVDs. To date, only sporadic studies on PANoptosis in CVDs have been reported and its role in the field of CVDs has not been fully explored. Elucidating the various modes of cardiomyocyte death, the specific molecular mechanisms and the links among the various modes of death under various stressful stimuli is of notable clinical significance for a deeper understanding of the pathophysiology of CVDs. The present review summarizes the molecular mechanisms of apoptosis, pyroptosis, necroptosis and PANoptosis and their prospects in the field of CVDs.
Collapse
Affiliation(s)
- Xinyu Gao
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Cuixue Ma
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Shan Liang
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Meihong Chen
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Yuan He
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Wei Lei
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
- Department of Precision Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| |
Collapse
|
3
|
Lv S, Zhao X, Ma C, Zhao D, Sun T, Fu W, Wei Y, Li W. Advancements in the study of acute lung injury resulting from intestinal ischemia/reperfusion. Front Med (Lausanne) 2024; 11:1399744. [PMID: 38933104 PMCID: PMC11199783 DOI: 10.3389/fmed.2024.1399744] [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: 03/12/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
Intestinal ischemia/reperfusion is a prevalent pathological process that can result in intestinal dysfunction, bacterial translocation, energy metabolism disturbances, and subsequent harm to distal tissues and organs via the circulatory system. Acute lung injury frequently arises as a complication of intestinal ischemia/reperfusion, exhibiting early onset and a grim prognosis. Without appropriate preventative measures and efficacious interventions, this condition may progress to acute respiratory distress syndrome and elevate mortality rates. Nonetheless, the precise mechanisms and efficacious treatments remain elusive. This paper synthesizes recent research models and pertinent injury evaluation criteria within the realm of acute lung injury induced by intestinal ischemia/reperfusion. The objective is to investigate the roles of pathophysiological mechanisms like oxidative stress, inflammatory response, apoptosis, ferroptosis, and pyroptosis; and to assess the strengths and limitations of current therapeutic approaches for acute lung injury stemming from intestinal ischemia/reperfusion. The goal is to elucidate potential targets for enhancing recovery rates, identify suitable treatment modalities, and offer insights for translating fundamental research into clinical applications.
Collapse
Affiliation(s)
- Shihua Lv
- Key Laboratory of Anesthesia and Intensive Care Research, Harbin, China
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xudong Zhao
- Department of Hepatopancreatobiliary, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Can Ma
- Key Laboratory of Anesthesia and Intensive Care Research, Harbin, China
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dengming Zhao
- Key Laboratory of Anesthesia and Intensive Care Research, Harbin, China
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tian Sun
- Key Laboratory of Anesthesia and Intensive Care Research, Harbin, China
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenchao Fu
- Key Laboratory of Anesthesia and Intensive Care Research, Harbin, China
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yuting Wei
- Key Laboratory of Anesthesia and Intensive Care Research, Harbin, China
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenzhi Li
- Key Laboratory of Anesthesia and Intensive Care Research, Harbin, China
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
4
|
Zhou HM, Yue SJ, Wang WX, Zhang Q, Xu DQ, Li JJ, Tang YP, Yang XY. Exploring the effective compounds and potential mechanisms of Shengxian Decoction against coronary heart disease by UPLC-Q-TOF/MS and network pharmacology analysis. Heliyon 2024; 10:e29558. [PMID: 38681620 PMCID: PMC11046127 DOI: 10.1016/j.heliyon.2024.e29558] [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: 09/21/2023] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 05/01/2024] Open
Abstract
As a well-known classical Chinese medicine prescription, Shengxian Decoction (SXD) has been applied for a century to treat cardiovascular diseases, especially coronary heart disease (CHD), but the potentially effective compounds and underlying mechanisms remain unclear. With ultra-performance liquid chromatography-quadrupole-time of flight-mass spectrometry (UPLC-Q-TOF/MS) and network pharmacology analysis, the potential effective compounds of SXD and their pharmacological mechanisms against CHD were identified and revealed. 57 effective compounds with favorable pharmacokinetic characteristics and biological activities were screened through UPLC-Q-TOF/MS analysis, database and literature mining, interacting with 96 CHD-related targets to support potential synergistic therapeutic actions. Systematic analysis of the PPI network and microarray data further revealed six core targets, including TNF, IL-1β, IL-6, TP53, VEGFA and PTGS2, which were mainly involved in fluid shear stress and atherosclerosis, lipid and atherosclerosis, PI3K-Akt signaling pathway et al. Moreover, the proposed contribution indexes of effective compounds indicated these compounds, including isoferulic acid, quercetin, calycosin, ferulic acid, kaempferol, calycosin 7-O-glycoside, formononetin, astragaloside IV and saikosaponin D, as the core compounds of SXD. The molecular docking results confirmed that those core compound-target pairs exhibited strong binding energy. Furthermore, we validated that SXD significantly alleviated myocardial tissue injury in CHD rats and reversed H/R-induced decreases in H9c2 cell viability by attenuating the production of TNF, IL-6 and IL-1β, and reducing cardiomyocyte apoptosis via down-regulating the TP53, caspase3 and cytochrome C mRNA expression levels as well as caspase3, caspase9 and cytochrome C protein expression levels according to RT-qPCR and Western blot results. Our findings explained the pharmacological mechanisms underlying the effectiveness of SXD in the treatment of CHD, and laid a foundation for future basic and clinical research of SXD.
Collapse
Affiliation(s)
- Hao-ming Zhou
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi Province, China
| | - Shi-jun Yue
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi Province, China
- International Joint Research Center on Resource Utilization and Quality Evaluation of Traditional Chinese Medicine of Hebei Province, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Wen-xiao Wang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi Province, China
- International Joint Research Center on Resource Utilization and Quality Evaluation of Traditional Chinese Medicine of Hebei Province, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Qiao Zhang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi Province, China
| | - Ding-qiao Xu
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi Province, China
| | - Jia-jia Li
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi Province, China
| | - Yu-ping Tang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi Province, China
| | - Xin-yu Yang
- Department of Pharmacy, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
- Beijing Key Laboratory of Bio-characteristic Profiling for Evaluation of Rational Drug Use, Beijing, 100038, China
| |
Collapse
|
5
|
Hassanpour M, Salybekov AA, Kobayashi S, Asahara T. CD34 positive cells as endothelial progenitor cells in biology and medicine. Front Cell Dev Biol 2023; 11:1128134. [PMID: 37138792 PMCID: PMC10150654 DOI: 10.3389/fcell.2023.1128134] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 04/03/2023] [Indexed: 05/05/2023] Open
Abstract
CD34 is a cell surface antigen expressed in numerous stem/progenitor cells including hematopoietic stem cells (HSCs) and endothelial progenitor cells (EPCs), which are known to be rich sources of EPCs. Therefore, regenerative therapy using CD34+ cells has attracted interest for application in patients with various vascular, ischemic, and inflammatory diseases. CD34+ cells have recently been reported to improve therapeutic angiogenesis in a variety of diseases. Mechanistically, CD34+ cells are involved in both direct incorporation into the expanding vasculature and paracrine activity through angiogenesis, anti-inflammatory, immunomodulatory, and anti-apoptosis/fibrosis roles, which support the developing microvasculature. Preclinical, pilot, and clinical trials have well documented a track record of safety, practicality, and validity of CD34+ cell therapy in various diseases. However, the clinical application of CD34+ cell therapy has triggered scientific debates and controversies in last decade. This review covers all preexisting scientific literature and prepares an overview of the comprehensive biology of CD34+ cells as well as the preclinical/clinical details of CD34+ cell therapy for regenerative medicine.
Collapse
Affiliation(s)
- Mehdi Hassanpour
- Shonan Research Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
- Center for Cell Therapy and Regenerative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
| | - Amankeldi A. Salybekov
- Shonan Research Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
- Center for Cell Therapy and Regenerative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
| | - Shuzo Kobayashi
- Shonan Research Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
| | - Takayuki Asahara
- Shonan Research Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
- Center for Cell Therapy and Regenerative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
- *Correspondence: Takayuki Asahara,
| |
Collapse
|
6
|
Exosomes Derived from BMSCs Ameliorate Intestinal Ischemia-Reperfusion Injury by Regulating miR-144-3p-Mediated Oxidative Stress. Dig Dis Sci 2022; 67:5090-5106. [PMID: 35624329 DOI: 10.1007/s10620-022-07546-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 02/10/2022] [Indexed: 01/05/2023]
Abstract
BACKGROUND Intestinal ischemia-reperfusion (I/R) injury is a critical pathophysiological process involved in many acute and critical diseases, and it may seriously threaten the lives of patients. Exosomes derived from bone marrow mesenchymal stem cells (BMSC-exos) may be an effective therapeutic approach for I/R injury. AIMS This study aimed to investigate the role and possible mechanism of BMSC-exos in intestinal I/R injury in vivo and in vitro based on the miR-144-3p and PTEN/Akt/Nrf2 pathways. METHODS BMSC-exos were isolated from mouse BMSCs by super centrifugation methods. The effects of BMSC-exos on I/R intestinal injury, intestinal cell apoptosis, oxidative stress and the PTEN/Akt/Nrf2 pathway were explored in vivo and in vitro. Furthermore, the relationship between miR-144-3p and PTEN was confirmed by a dual-luciferase reporter assay. The miR-144-3p mimic and inhibitor were used to further clarify the role of miR-144-3p in the mitigation of intestinal I/R by BMSC-exos. RESULTS BMSC-exos effectively alleviated intestinal pathological injury, reduced intestinal cell apoptosis, relieved oxidative stress and regulated the PTEN/Akt/Nrf2 pathway in vivo and in vitro. In addition, miR-144-3p was significantly reduced in the oxygen and glucose deprivation/reperfusion cell model, and miR-144-3p could directly target PTEN to regulate its expression. Additional studies showed that changing the expression of miR-144-3p in BMSC-exos significantly affected the regulation of intestinal injury, intestinal cell apoptosis, oxidative stress and the PTEN/Akt/Nrf2 pathway in I/R, suggesting that miR-144-3p in BMSC-exos plays an important role in regulating the PTEN/Akt/Nrf2 during intestinal I/R. CONCLUSIONS BMSC-exos carrying miR-144-3p alleviated intestinal I/R injury by regulating oxidative stress.
Collapse
|
7
|
Mahdavi FS, Mardi S, Mohammadi S, Ansari S, Yaslianifard S, Fallah P, Mozhgani SH. MicroRNA-146: Biomarker and Mediator of Cardiovascular Disease. DISEASE MARKERS 2022; 2022:7767598. [PMID: 39281713 PMCID: PMC11401689 DOI: 10.1155/2022/7767598] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 09/15/2022] [Indexed: 09/18/2024]
Abstract
Cardiovascular diseases (CVDs) are the prime cause of morbidity and mortality worldwide. Although noticeable progress has been made in the diagnosis, prognosis, and treatment, there is still a critical demand for new diagnostic biomarkers and novel therapeutic interventions to reduce this disease incidence. Many investigations have been conducted on the regulatory effects of microRNAs in cardiovascular diseases. miRNA circulating serum level changes are correlated with several CVDs. In addition, there is growing evidence representing the potential role of miRNAs as diagnostic biomarkers or potential therapeutic targets for CVD. Preliminary studies identified the prominent role of miR-146 in host defense, innate immunity, and different immunological diseases by regulating cytokine production and innate immunity modification in bacterial infections. However, more recently, it was also associated with CVD development. miR-146 has received much attention, with positive results in most studies. Research demonstrated the crucial role of this molecule in the pathogenesis of cardiac disease and related mechanisms. As a result, many potential applications of miR-146 are expected. In this paper, we provide an overview of recent studies highlighting the role of miR-146 in CVD, focusing on CAD (coronary artery disease), cardiomyopathy, and MI (myocardial infarction) in particular and discussing its current scientific state, and use a prognostic biomarker as a therapeutic agent for cardiovascular diseases.
Collapse
Affiliation(s)
- Fatemeh Sadat Mahdavi
- Student Research Committee, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Shayan Mardi
- Student Research Committee, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Sareh Mohammadi
- Cardiovascular Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Sarina Ansari
- Student Research Committee, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Somayeh Yaslianifard
- Department of Microbiology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Parviz Fallah
- Department of Laboratory Science, School of Allied Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Sayed-Hamidreza Mozhgani
- Department of Microbiology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
- Noncommunicable Disease Research Center, Alborz University of Medical Sciences, Karaj, Iran
| |
Collapse
|
8
|
Integrated Gut-Heart Axis and Network Pharmacology to Reveal the Mechanisms of the Huoxue Wentong Formula Against Myocardial Ischemia. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:9538512. [PMID: 35600966 PMCID: PMC9117028 DOI: 10.1155/2022/9538512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 04/18/2022] [Indexed: 11/20/2022]
Abstract
Background Myocardial ischemia (MI) is a major public health problem with high mortality and morbidity worldwide. Huoxue Wentong formula (HX), a traditional Chinese medicine (TCM) formula, exhibits unambiguous effects on treating MI and preventing cardiovascular diseases. However, the molecular mechanism of the therapeutic effects of HX on MI remains largely unknown. Objective This study combined microbiology, metabolomics, and network pharmacology to explore the relationship between the gut microbiota and its metabolites in MI rats and the efficacy of HX. Methods First, the MI rat model was established by ligation of left anterior descending. Echocardiography, Masson's staining, and hematoxylin and eosin staining were used to evaluate the effect of HX on MI. Then, fecal metabolomics and 16S rRNA sequencing were used to obtain the microbial and metabolic characteristics of HX on MI. After that, network pharmacology was used to predict the target and action pathway of HX in treating MI. Finally, the relationship between fecal metabolites and target was explored through bioinformatics. Results HX can improve the cardiac function and ameliorated myocardial fibrosis in MI rats. Moreover, HX can affect the gut microbiota community and metabolites of MI rats, especially Bacteroides, Deferribacteres, Ruminococcus_sp._zagget7, Acidobacteria, daidzein, L-lactic acid, and malate. Network pharmacology found that HX can function through tumor necrosis factor (TNF), tumor protein p53 (TP53), interleukin 6 (IL6), vascular endothelial growth factor A (VEGFA), fos proto-oncogene (FOS), bcl2-associated X (BAX), myeloperoxidase (MPO), PI3K-Akt signaling pathways, and HIF-1 signaling pathway. The mechanism study showed that the anti-MI effect of HX was related to valine, leucine, and isoleucine biosynthesis, fatty acid biosynthesis, and arachidonic acid metabolism. Conclusion This study demonstrates that HX treated MI rats in a multitarget and multipathway manner. Its mechanism is related to the change of gut microbiota and the regulation of valine, leucine and isoleucine biosynthesis, fatty acid biosynthesis, and arachidonic acid metabolism.
Collapse
|
9
|
Particulate Matter-Induced Acute Coronary Syndrome: MicroRNAs as Microregulators for Inflammatory Factors. Mediators Inflamm 2021; 2021:6609143. [PMID: 34931116 PMCID: PMC8684514 DOI: 10.1155/2021/6609143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 11/18/2021] [Indexed: 12/03/2022] Open
Abstract
The most prevalent cause of mortality and morbidity worldwide is acute coronary syndrome (ACS) and its consequences. Exposure to particulate matter (PM) from air pollution has been shown to impair both. Various plausible pathogenic mechanisms have been identified, including microRNAs (miRNAs), an epigenetic regulator for gene expression. Endogenous miRNAs, average 22-nucleotide RNAs (ribonucleic acid), regulate gene expression through mRNA cleavage or translation repression and can influence proinflammatory gene expression posttranscriptionally. However, little is known about miRNA responses to fine PM (PM2.5, PM10, ultrafine particles, black carbon, and polycyclic aromatic hydrocarbon) from air pollution and their potential contribution to cardiovascular consequences, including systemic inflammation regulation. For the past decades, microRNAs (miRNAs) have emerged as novel, prospective diagnostic and prognostic biomarkers in various illnesses, including ACS. We wanted to outline some of the most important studies in the field and address the possible utility of miRNAs in regulating particulate matter-induced ACS (PMIA) on inflammatory factors in this review.
Collapse
|
10
|
Jin Z, Zhang W, Luo Y, Li X, Qing L, Zuo Q, Fang J, Wu W. Protective effect of Qingre Huoxue decoction against myocardial infarction via PI3K/Akt autophagy pathway based on UPLC-MS, network pharmacology, and in vivo evidence. PHARMACEUTICAL BIOLOGY 2021; 59:1607-1618. [PMID: 34818128 PMCID: PMC8635559 DOI: 10.1080/13880209.2021.2001542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/22/2021] [Accepted: 10/28/2021] [Indexed: 06/13/2023]
Abstract
CONTEXT Qingre Huoxue (QRHX) decoction, a traditional Chinese medicine, has been widely used to prevent and treat myocardial infarction (MI). OBJECTIVE This study elucidates the possible mechanisms of QRHX in preventing or treating MI in a rat model. MATERIALS AND METHODS The chemical constituents of QRHX were identified by UPLC-MS. Sprague-Dawley rats were randomly divided into the Sham (normal saline), Model (normal saline), QRHX-L, QRHX-M and QRHX-H group (n = 10 per group). QRHX decoction was administered by gavage to the rats for 14 days (5, 10 and 20 g/kg/day). The left anterior descending ligation method was performed to develop MI in Model and QRHX groups, and the same surgical procedures excluding ligation sutures were performed for the sham group. Finally, we evaluated cardiac function, myocardial fibrosis degree, serum inflammatory factors, autophagy levels and verified the signalling pathways in vivo. RESULTS A total of 68 active components of QRHX corresponding to 223 active targets were obtained and 2558 MI-related disease targets were collected. After integration, 123 QRHX anti-MI targets were obtained, and 70 signalling pathways, such as PI3K/Akt, were identified by enrichment analysis. In vivo experiments suggest that QRHX could reduce the degree of myocardial fibrosis, downregulate serum inflammatory factors, and promote autophagy in MI rats. DISCUSSION AND CONCLUSIONS QRHX plays a protective role in the myocardium by mediating PI3K/Akt signalling pathway to activate autophagy and inhibiting inflammatory factor expression. These findings provide a scientific basis for further research and validation of QRHX as a potential therapeutic for MI.
Collapse
Affiliation(s)
- Zheng Jin
- ZhuJiang Hospital, Southern Medical University, Guangzhou, China
| | - Wenbo Zhang
- Research Center of Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuan Luo
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiushen Li
- Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, China
| | - Lijin Qing
- Department of Cardiovascular, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qiang Zuo
- First Affiliated Hospital, Anhui University of Chinese Medicine, Hefei, China
| | - Junfeng Fang
- Department of Emergency, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Wu
- Department of Cardiovascular, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
11
|
Lai TC, Lee TL, Chang YC, Chen YC, Lin SR, Lin SW, Pu CM, Tsai JS, Chen YL. MicroRNA-221/222 Mediates ADSC-Exosome-Induced Cardioprotection Against Ischemia/Reperfusion by Targeting PUMA and ETS-1. Front Cell Dev Biol 2020; 8:569150. [PMID: 33344446 PMCID: PMC7744807 DOI: 10.3389/fcell.2020.569150] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 11/11/2020] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular disease is a major health problem in industrialized and developing countries and is the leading cause of death and disability. Myocardial ischemia/reperfusion (I/R) causes cardiomyocyte damage such as apoptosis and hypertrophy. The purpose of this study was to investigate the effects of exosomes from adipose-derived stem cells (ADSC-Exo) on hearts from I/R mice and to explore the underlying mechanisms. ADSC-Exo significantly decreased I/R-induced cardiomyocyte apoptosis and hypertrophy, as detected by TdT-mediated dUTP nick end-labeling (TUNEL) and wheat germ agglutinin (WGA) staining, respectively. In addition, the expression of apoptosis-related proteins p-p53 and PUMA and hypertrophy-related proteins ETS-1 and ANP were significantly reduced in the cardiomyocytes of ADSC-Exo-treated I/R mice compared to those of control mice. Both PUMA and ETS-1 are reported to be target genes for miR-221/222. I/R operation significantly reduced miR-221/222 expression, while ADSC-Exo treatment increased miR-221/222 expression, as detected by RT-qPCR. We also observed that cardiac I/R operation markedly increased cell apoptosis and hypertrophy in miR-221/222 knockout (KO) mice, while ADSC-Exo reduced the effects of I/R operation. Furthermore, ADSC-Exo protected H9c2 cardiomyocytes from H2O2-induced damage by reducing apoptosis and hypertrophy in vitro. H2O2 treatment significantly reduced miR-221/222 expression, while ADSC-Exo treatment reversed this effect in H9c2 cells. ADSC-Exo treatment decreased H2O2-induced PUMA and ETS-1 expression. Compared with control treatment, I/R treatment significantly reduced p-AKT and increased p-p65, while ADSC-Exo and miR-221/222 mimics attenuated these effects. The AKT activator SC79 and p65 inhibitor Bay 11-7082 reduced H2O2-induced cell apoptosis and hypertrophy. Based on these findings, ADSC-Exo prevents cardiac I/R injury through the miR-221/miR-222/PUMA/ETS-1 pathway. Therefore, ADSC-Exo is an effective inhibitor of I/R-induced heart injury.
Collapse
Affiliation(s)
- Tsai-Chun Lai
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tzu-Lin Lee
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Chun Chang
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Chen Chen
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shu-Rung Lin
- Department of Bioscience Technology, College of Science, Chung Yuan Christian University, Taoyuan, Taiwan.,Center for Nanotechnology and Center for Biomedical Technology, Chung Yuan Christian University, Taoyuan, Taiwan
| | - Shu-Wha Lin
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chi-Ming Pu
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Division of Plastic Surgery, Department of Surgery, Cathay General Hospital, Taipei, Taiwan
| | - Jaw-Shiun Tsai
- Department of Family Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Center for Complementary and Integrated Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Yuh-Lien Chen
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
12
|
Chen Y, Yin Y, Jiang H. miR-30e-5p Alleviates Inflammation and Cardiac Dysfunction After Myocardial Infarction Through Targeting PTEN. Inflammation 2020; 44:769-779. [PMID: 33180227 DOI: 10.1007/s10753-020-01376-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/16/2020] [Accepted: 10/25/2020] [Indexed: 12/28/2022]
Abstract
Accumulating studies show that microRNAs are candidate biomarkers and therapeutic targets for cardiovascular diseases including myocardial infarction (MI). Bioinformatics analysis suggested that compared with Sprague-Dawley (SD) rats without MI, miR-30e-5p expression in the left ventricle tissue of SD rats with MI was significantly downregulated, suggesting miR-30e-5p may participate in the pathogenesis of MI. In this study, H9c2 cardiomyocytes were exposed to hypoxia to establish a hypoxic cell model. SD rats with left anterior descending coronary artery ligation were used for the MI animal model. Quantitative real-time polymerase chain reaction (qRT-PCR) was performed to evaluate the miR-30e-5p and PTEN mRNA expressions in cells and tissues. Western blot was used for detecting the expression of PTEN protein. miR-30e-5p expression in H9c2 cells was then inhibited or overexpressed, and its effects on viability and apoptosis were examined by cell counting kit-8 (CCK-8) assay and TUNEL assay, respectively. ELISA was used to detect inflammatory factors. The regulatory relationship between PTEN and miR-30e-5p was investigated by bioinformatics analysis, qRT-PCR, Western blot, and dual-luciferase reporter assay. It was found that miR-30e-5p expression was significantly downregulated in animal models and H9c2 cells under hypoxia. Overexpression of miR-30e-5p led to a dramatic increase of cell viability, accompanied by the decrease of IL-1β, TNF-α, IL-6, LDH, CK-MB, and cTnI. Furthermore, PTEN was identified as a target of miR-30e-5p, and PTEN overexpression reversed the effects of miR-30e -5p on H9c2 cells. To conclude, we confirm that miR-30e-5p alleviates inflammation and myocardial injury induced by MI via suppressing PTEN.
Collapse
Affiliation(s)
- Yongli Chen
- Department of Cardiology, Tianjin Chest Hospital, 261 Taierzhuang South Road, Jinnan District, Tianjin, 300222, China
| | - Yan Yin
- Department of Respiratory, Tianjin Chest Hospital, Tianjin, 300222, China
| | - Hua Jiang
- Department of Cardiology, Tianjin Chest Hospital, 261 Taierzhuang South Road, Jinnan District, Tianjin, 300222, China.
| |
Collapse
|
13
|
Yang W, Li S, Zhao Y, Sun Y, Huang Y, Cao X, Xing C, Yang F, Liu W, Zhao X, Dong X, Bai H, Shang X. Changes in the expression of interleukin-10 in myocardial infarction and its relationship with macrophage activation and cell apoptosis. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:643. [PMID: 32566580 PMCID: PMC7290626 DOI: 10.21037/atm-20-3349] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Currently, the role of interleukin-10 (IL-10) as an anti-inflammatory factor in the occurrence and development of heart disease is still unclear. This study aimed to observe the dynamic changes in the expression of IL-10 in serum and myocardial tissues and to investigate the relationship of IL-10 expression with macrophage activation and cardiomyocyte apoptosis during the occurrence of myocardial infarction (MI). Methods Mice models with MI were prepared by ligating the anterior descending branch of the coronary artery. The animals were classified into the sham operation group (the control group), and the day 1, 7, 14, and 28 MI groups based. Results On days 7 and 14, the cells with positive IL-10 expression were largely distributed in the infarct areas, while cells with positive IL-10 expression were decreased on day 28. Serum IL-10 was significantly positively correlated with IL-10 protein expression in myocardial tissues. Moreover, Bcl-2 and Bax protein expression in myocardial tissues, along with the ratio of Bcl-2/Bax proteins, were gradually elevated with prolonged time of infarction. The expression of arginase protein increased gradually too. There were positive correlations between IL-10 and arginase expressions, and between the expressions of Bcl-2 and Bax proteins. Conclusions After the occurrence of MI, the expression of IL-10 first increased and then decreased in serum and myocardial tissues, with this likely affecting macrophage activation, phenotypic transformation, and the occurrence of cardiomyocyte apoptosis.
Collapse
Affiliation(s)
- Wenqi Yang
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Shuming Li
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan 065299, China
| | - Yang Zhao
- Department of Medical imageology, Hebei North University, Zhangjiakou 075132, China
| | - Ying Sun
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan 065299, China
| | - Yuling Huang
- Department of Cardiology, North China University of Science and Technology Affiliated Hospital, Tangshan 065299, China
| | - Xinying Cao
- Department of Cardiology, North China University of Science and Technology Affiliated Hospital, Tangshan 065299, China
| | - Cainai Xing
- Department of Cardiology, North China University of Science and Technology Affiliated Hospital, Tangshan 065299, China
| | - Fang Yang
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan 065299, China
| | - Wenbo Liu
- Department of Cardiology, North China University of Science and Technology Affiliated Hospital, Tangshan 065299, China
| | - Xuan Zhao
- Department of Cardiology, North China University of Science and Technology Affiliated Hospital, Tangshan 065299, China
| | - Xuejing Dong
- Department of Cardiology, North China University of Science and Technology Affiliated Hospital, Tangshan 065299, China
| | - Hua Bai
- Department of Cardiology, North China University of Science and Technology Affiliated Hospital, Tangshan 065299, China
| | - Xiaoming Shang
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang 050017, China
| |
Collapse
|
14
|
Li S, Wang Y, Zhao C, Zhang M, Wang W, Yu X, Huang J, Wang Z, Zhu B, Yin C, Cai H. Akt inhibitor deguelin aggravates inflammation and fibrosis in myocarditis. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2019; 22:1275-1282. [PMID: 32128091 PMCID: PMC7038425 DOI: 10.22038/ijbms.2019.35518.8473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 05/12/2019] [Indexed: 11/16/2022]
Abstract
OBJECTIVES Myocarditis is characterized by inflammatory cell infiltration in myocardial stroma. Attenuation of tumor necrosis factor (TNF)-α and interleukin (IL)-1β is a reliable mark for improving the prognosis. Protein kinase B (Akt) plays an important role in the development and progression of myocarditis. The specific role of the natural inhibitor of Akt, Deguelin, on myocarditis has not been reported. In this study, we used deguelin to investigate the effects of natural Akt inhibitor on myocarditis in experimental autoimmune myocarditis (EAM) rats. MATERIALS AND METHODS EAM rat models were made by using Lewis rats and Deguelin was injected intraperitoneally on day 3, 6, 9, 12 and 15 after successful modeling. On day 18, rats were sacrificed and the heart weight (HW)/ body weight (BW) ratio were measured. The pathological changes, pathological scores and fibrosis area were evaluated after H.&E. and Masson's trichrome staining. The mRNA levels of TNF-α and IL-1β were measured by RT-qPCR, while the protein expressions of TNF-α and IL-1β were detected by immunohistochemical staining and Western bolt. The protein expressions of Akt, Akt1, phosphorylated (p-) Akt and nuclear factor (NF)-κB were detected by Western bolt. RESULTS We found that the TNF-α and IL-1β levels, inflammatory scores and fibrosis areas were markedly increased after 18 days deguelin administration. CONCLUSION Akt inhibition with deguelin may aggravate myocarditis of EAM rats.
Collapse
Affiliation(s)
- Shanshan Li
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Yue Wang
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Chunming Zhao
- Human anatomy and Histology and Embryology, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Meixiang Zhang
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Wei Wang
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Xiaowei Yu
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Jiao Huang
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Zhao Wang
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Bo Zhu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - Chengqian Yin
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - Hongxing Cai
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| |
Collapse
|
15
|
Qin Q, Cui L, Zhou Z, Zhang Z, Wang Y, Zhou C. Inhibition of microRNA-141-3p Reduces Hypoxia-Induced Apoptosis in H9c2 Rat Cardiomyocytes by Activating the RP105-Dependent PI3K/AKT Signaling Pathway. Med Sci Monit 2019; 25:7016-7025. [PMID: 31532760 PMCID: PMC6765341 DOI: 10.12659/msm.916361] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 04/16/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Oxidative stress in myocardial ischemia results in cardiomyocyte apoptosis. The expression of microRNA-141-3p (miR-141-3p) and the 105 kD toll-like receptor protein (TLR), RP105, have been identified in cardiomyocytes in vitro. This study aimed to investigate the effects of hypoxia in H9c2 rat cardiomyoblasts with and without the inhibition of miR-141-3p and to investigate the expression of RP105 and the PI3K/AKT signaling pathway. MATERIAL AND METHODS H9c2 rat cardiomyoblasts were cultured in conditions of hypoxia and treated with a specific miR-141-3p-inhibitor. RP105 short-interfering RNA (siRNA) was constructed, and LY294002 was used to inhibit the PI3KA/AKT pathway. The fluorescent probe, dihydroethidium (DHE), was used to detect reactive oxygen species (ROS). Flow cytometry evaluated ROS and apoptosis. Quantitative real-time polymerase chain reaction (RT-qPCR) and Western blot studied the expression of the PI3K/AKT pathway genes and proteins. Bioinformatics and dual-luciferase reporter assays were used to identify the targets for miR-141-3p. RESULTS A predictive TargetScan algorithm showed that the RP105 gene was a potential target of miR-141-3p. Expression of miR-141-3p was significantly increased in hypoxic H9c2 cells, and inhibition of miR-141-3p increased cell viability and reduced apoptosis. Also, miR-141-3p was shown to target 3'-UTR of RP105. Down-regulation of RP105 associated with hypoxia and its downstream PI3K/AKT pathway were significantly increased following miR-141-3p inhibition. The protective effect of miR-141-3p inhibition in hypoxic H9c2 cells was abolished by the absence of RP105 and inhibition of PI3K/AKT. CONCLUSIONS Inhibition of miR-141-3p reduced hypoxia-induced apoptosis in H9c2 cardiomyocytes in vitro by activating the RP105-dependent PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Qiaoji Qin
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, P.R. China
| | - Liqiang Cui
- Department of Anesthesiology, Qingdao Chengyang People’s Hospital, Qingdao, Shandong, P.R. China
| | - Zhenggang Zhou
- Department of Spine Surgery, Qingdao Chengyang People’s Hospital, Qingdao, Shandong, P.R. China
| | - Zhirong Zhang
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, P.R. China
| | - Yini Wang
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, P.R. China
| | - Changyong Zhou
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, P.R. China
| |
Collapse
|