1
|
Yue TT, Cao YJ, Cao YX, Li WX, Wang XY, Si CY, Xia H, Zhu MJ, Tang JF, Wang H. Shuxuening Injection Inhibits Apoptosis and Reduces Myocardial Ischemia-Reperfusion Injury in Rats through PI3K/AKT Pathway. Chin J Integr Med 2024; 30:421-432. [PMID: 38153596 DOI: 10.1007/s11655-023-3650-z] [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: 06/14/2023] [Indexed: 12/29/2023]
Abstract
OBJECTIVE To investigate the main components and potential mechanism of Shuxuening Injection (SXNI) in the treatment of myocardial ischemia-reperfusion injury (MIRI) through network pharmacology and in vivo research. METHODS The Traditional Chinese Medicine Systems Pharmacology (TCMSP) and PharmMapper databases were used to extract and evaluate the effective components of Ginkgo biloba leaves, the main component of SXNI. The Online Mendelian Inheritance in Man (OMIM) and GeneCards databases were searched for disease targets and obtain the drug target and disease target intersections. The active ingredient-target network was built using Cytoscape 3.9.1 software. The STRING database, Metascape online platform, and R language were used to obtain the key targets and signaling pathways of the anti-MIRI effects of SXNI. In order to verify the therapeutic effect of different concentrations of SXNI on MIRI in rats, 60 rats were first divided into 5 groups according to random number table method: the sham operation group, the model group, SXNI low-dose (3.68 mg/kg), medium-dose (7.35 mg/kg), and high-dose (14.7 mg/kg) groups, with 12 rats in each group. Then, another 60 rats were randomly divided into 5 groups: the sham operation group, the model group, SXNI group (14.7 mg/kg), SXNI+LY294002 group, and LY294002 group, with 12 rats in each group. The drug was then administered intraperitoneally at body weight for 14 days. The main biological processes were validated using in vivo testing. Evans blue/triphenyltetrazolium chloride (TTC) double staining, hematoxylin-eosin (HE) staining, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay, enzyme-linked immunosorbent assay (ELISA), and Western blot analysis were used to investigate the efficacy and mechanism of SXNI in MIRI rats. RESULTS Eleven core targets and 30 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were selected. Among these, the phosphoinositide 3-kinase (PI3K)/ protein kinase B (AKT) pathway was closely related to SXNI treatment of MIRI. In vivo experiments showed that SXNI reduced the myocardial infarction area in the model group, improved rat heart pathological damage, and reduced the cardiomyocyte apoptosis rate (all P<0.01). After SXNI treatment, the p-PI3K/PI3K and p-AKT/AKT ratios as well as B-cell lymphoma-2 (Bcl-2) protein expression in cardiomyocytes were increased, while the Bax and cleaved caspase 3 protein expression levels were decreased (all P<0.05). LY294002 partially reversed the protective effect of SXNI on MIRI. CONCLUSION SXNI protects against MIRI by activating the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Tong-Tong Yue
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Ying-Jie Cao
- Department of Pharmacy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 451200, China
| | - Ya-Xuan Cao
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Wei-Xia Li
- Department of Pharmacy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 451200, China
| | - Xiao-Yan Wang
- Department of Pharmacy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 451200, China
| | - Chun-Ying Si
- Department of Cardiovascular Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 451200, China
| | - Han Xia
- Department of Cardiovascular Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 451200, China
| | - Ming-Jun Zhu
- Department of Cardiovascular Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 451200, China
| | - Jin-Fa Tang
- Department of Pharmacy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 451200, China
| | - He Wang
- Department of Cardiovascular Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 451200, China.
| |
Collapse
|
2
|
Momeni Z, Danesh S, Ahmadpour M, Eshraghi R, Farkhondeh T, Pourhanifeh MH, Samarghandian S. Protective Roles and Therapeutic Effects of Gallic Acid in the Treatment of Cardiovascular Diseases: Current Trends and Future Directions. Curr Med Chem 2024; 31:3733-3751. [PMID: 37815180 DOI: 10.2174/0109298673259299230921150030] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 07/19/2023] [Accepted: 08/18/2023] [Indexed: 10/11/2023]
Abstract
Cardiovascular diseases (CVDs) are serious life-threatening illnesses and significant problematic issues for public health having a heavy economic burden on all society worldwide. The high incidence of these diseases as well as high mortality rates make them the leading causes of death and disability. Therefore, finding novel and more effective therapeutic methods is urgently required. Gallic acid, an herbal medicine with numerous biological properties, has been utilized in the treatment of various diseases for thousands of years. It has been demonstrated that gallic acid possesses pharmacological potential in regulating several molecular and cellular processes such as apoptosis and autophagy. Moreover, gallic acid has been investigated in the treatment of CVDs both in vivo and in vitro. Herein, we aimed to review the available evidence on the therapeutic application of gallic acid for CVDs including myocardial ischemia-reperfusion injury and infarction, drug-induced cardiotoxicity, hypertension, cardiac fibrosis, and heart failure, with a focus on underlying mechanisms.
Collapse
Affiliation(s)
- Zahra Momeni
- Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Sepideh Danesh
- Research Hub Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Ahmadpour
- Research Hub Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Eshraghi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Tahereh Farkhondeh
- Department of Toxicology and Pharmacology, School of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Hossein Pourhanifeh
- Research Hub Institute, Tehran University of Medical Sciences, Tehran, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Saeed Samarghandian
- University of Neyshabur Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
3
|
L-Borneol 7-O-[β-D-Apiofuranosyl-(1 6)]-β-D-Glucopyranoside Alleviates Myocardial Ischemia-Reperfusion Injury in Rats and Hypoxic/Reoxygenated Injured Myocardial Cells via Regulating the PI3K/AKT/mTOR Signaling Pathway. J Immunol Res 2022; 2022:5758303. [PMID: 35600046 PMCID: PMC9119761 DOI: 10.1155/2022/5758303] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 04/13/2022] [Accepted: 04/26/2022] [Indexed: 12/12/2022] Open
Abstract
Ischemia/reperfusion (I/R) is a primary cause of morbidity and mortality in acute myocardial infarction (AMI). L-Borneol 7-O-[β-D-apiofuranosyl-(1→6)]-β-D-glucopyranoside (LBAG), extracted from the Radix Ophiopogonis, is the main bioactive component that may be exerting cardiovascular protection in AMI. The purpose was to examine the effects of LBAG on myocardial I/R injury (MIRI) in rats and H9c2 cells treated with hypoxia/reoxygenation (H/R). MIRI was induced through the combination of ischemia with reperfusion for 30 min and 24 h, respectively. LBAG was administered 7 days before vascular ligation. Myocardial function was detected by an electrocardiograph, histological, TTC, and TUNEL staining analyses. The influences of LBAG on the content concentration of cardiac enzymes in the serum were measured by ELISA. Moreover, H9c2 cells were exposed to LBAG or combined with AKT inhibitor (perifosine) and then exposed to H/R for simulating the cardiac injury process. Afterward, cell viability, LDH, CD-KM release, apoptosis, and autophagy were evaluated by CCK-8 and ELISA assays, flow cytometry, TUNEL, and immunofluorescence staining, respectively. Additionally, the proteins of apoptosis, autophagy, and PI3K/mTOR pathway were determined by western blotting. In I/R rats, LBAG pretreatment significantly ameliorated cardiac function, as illustrated by reducing the infarct size, myocardial autophagy, and apoptosis levels. In H/R-induced H9c2 cells, LBAG pretreatment significantly decreased cell apoptosis, LC3 II/I, and Beclin 1 levels, elevated the Bcl-2 levels, attenuated LDH, and CD-KM production. Moreover, LBAG pretreatment markedly increased the PI3K/mTOR pathway activation, and the protective influences of LBAG were partly abolished with the AKT inhibitor perifosine treatment. These findings demonstrated the protective functions of LBAG on I/R by regulating apoptosis and autophagy in vitro and in vivo by activating the PI3K/mTOR pathway.
Collapse
|
4
|
Liu J, Tang L, Lu Q, Yu Y, Xu QG, Zhang S, Chen YX, Dai WJ, Li JC. Plasma Quantitative Lipid Profiles: Identification of CarnitineC18:1-OH, CarnitineC18:2-OH and FFA (20:1) as Novel Biomarkers for Pre-warning and Prognosis in Acute Myocardial Infarction. Front Cardiovasc Med 2022; 9:848840. [PMID: 35479277 PMCID: PMC9037999 DOI: 10.3389/fcvm.2022.848840] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/08/2022] [Indexed: 02/05/2023] Open
Abstract
This study was aimed to determine the association between potential plasma lipid biomarkers and early screening and prognosis of Acute myocardial infarction (AMI). In the present study, a total of 795 differentially expressed lipid metabolites were detected based on ultra-performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS). Out of these metabolites, 25 lipid metabolites were identified which showed specifical expression in the AMI group compared with the healthy control (HC) group and unstable angina (UA) group. Then, we applied the least absolute shrinkage and selection operator (LASSO) and support vector machine-recursive feature elimination (SVM-RFE) methods to obtain three lipid molecules, including CarnitineC18:1-OH, CarnitineC18:2-OH and FFA (20:1). The three lipid metabolites and the diagnostic model exhibited well predictive ability in discriminating between AMI patients and UA patients in both the discovery and validation sets with an area under the curve (AUC) of 0.9. Univariate and multivariate logistic regression analyses indicated that the three lipid metabolites may serve as potential biomarkers for diagnosing AMI. A subsequent 1-year follow-up analysis indicated that the three lipid biomarkers also had prominent performance in predicting re-admission of patients with AMI due to cardiovascular events. In summary, we used quantitative lipid technology to delineate the characteristics of lipid metabolism in patients with AMI, and identified potential early diagnosis biomarkers of AMI via machine learning approach.
Collapse
Affiliation(s)
- Jun Liu
- Medical Research Center and Department of Cardiology, Yue Bei People's Hospital, Shantou University Medical College, Shaoguan, China
| | - Liangqiu Tang
- Medical Research Center and Department of Cardiology, Yue Bei People's Hospital, Shantou University Medical College, Shaoguan, China
| | - Qiqi Lu
- Medical Research Center and Department of Cardiology, Yue Bei People's Hospital, Shantou University Medical College, Shaoguan, China
| | - Yi Yu
- Medical Research Center and Department of Cardiology, Yue Bei People's Hospital, Shantou University Medical College, Shaoguan, China
- The Central Laboratory, Yangjiang People's Hospital, Yangjiang, China
| | - Qiu-Gui Xu
- The Central Laboratory, Yangjiang People's Hospital, Yangjiang, China
| | - Shanqiang Zhang
- Medical Research Center and Department of Cardiology, Yue Bei People's Hospital, Shantou University Medical College, Shaoguan, China
| | - Yun-Xian Chen
- Medical Research Center and Department of Cardiology, Yue Bei People's Hospital, Shantou University Medical College, Shaoguan, China
| | - Wen-Jie Dai
- Medical Research Center and Department of Cardiology, Yue Bei People's Hospital, Shantou University Medical College, Shaoguan, China
| | - Ji-Cheng Li
- Medical Research Center and Department of Cardiology, Yue Bei People's Hospital, Shantou University Medical College, Shaoguan, China
- The Central Laboratory, Yangjiang People's Hospital, Yangjiang, China
- Department of Histology and Embryology, Shaoguan University School of Medicine, Shaoguan, China
- Institute of Cell Biology, Zhejiang University, Hangzhou, China
| |
Collapse
|
5
|
Di Cesare F, Tenori L, Meoni G, Gori AM, Marcucci R, Giusti B, Molino-Lova R, Macchi C, Pancani S, Luchinat C, Saccenti E. Lipid and metabolite correlation networks specific to clinical and biochemical covariate show differences associated with sexual dimorphism in a cohort of nonagenarians. GeroScience 2021; 44:1109-1128. [PMID: 34324142 PMCID: PMC9135919 DOI: 10.1007/s11357-021-00404-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/13/2021] [Indexed: 12/26/2022] Open
Abstract
This study defines and estimates the metabolite-lipidic component association networks constructed from an array of 20 metabolites and 114 lipids identified and quantified via NMR spectroscopy in the serum of a cohort of 355 Italian nonagenarians and ultra-nonagenarian. Metabolite-lipid association networks were built for men and women and related to an array of 101 clinical and biochemical parameters, including the presence of diseases, bio-humoral parameters, familiarity diseases, drugs treatments, and risk factors. Different connectivity patterns were observed in lipids, branched chains amino acids, alanine, and ketone bodies, suggesting their association with the sex-related and sex-clinical condition-related intrinsic metabolic changes. Furthermore, our results demonstrate, using a holistic system biology approach, that the characterization of metabolic structures and their dynamic inter-connections is a promising tool to shed light on the dimorphic pathophysiological mechanisms of aging at the molecular level.
Collapse
Affiliation(s)
- Francesca Di Cesare
- Magnetic Resonance Center (CERM), University of Florence, Sesto Fiorentino, Italy
| | - Leonardo Tenori
- Magnetic Resonance Center (CERM), University of Florence, Sesto Fiorentino, Italy.,Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy
| | | | - Anna Maria Gori
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,Atherothrombotic Unit, Careggi University Hospital, Florence, Italy
| | - Rossella Marcucci
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,Atherothrombotic Unit, Careggi University Hospital, Florence, Italy
| | - Betti Giusti
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,Atherothrombotic Unit, Careggi University Hospital, Florence, Italy
| | | | - Claudio Macchi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | | | - Claudio Luchinat
- Magnetic Resonance Center (CERM), University of Florence, Sesto Fiorentino, Italy.,Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy.,Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine (CIRMMP), Sesto Fiorentino, Italy
| | - Edoardo Saccenti
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, Wageningen, the Netherlands.
| |
Collapse
|
6
|
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: 14] [Impact Index Per Article: 3.5] [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
|