1
|
Zhu W, Shao P, Ren Y, Liu W, Han X, Zeng K, Dai C, Liu F. Integrating network pharmacology and experimental validation to investigate the mechanism of Gualou Xiebai Banxia decoction against myocardial ischemia. Front Cardiovasc Med 2025; 12:1512791. [PMID: 40329968 PMCID: PMC12052758 DOI: 10.3389/fcvm.2025.1512791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 03/27/2025] [Indexed: 05/08/2025] Open
Abstract
Background Gualou Xiebai Banxia Decoction (GXBD), a traditional Chinese medicine, is used to treat myocardial ischemia (MI). However, the molecular mechanisms underlying its effects remain unclear. This study integrated network pharmacology and experimental validation to investigate the efficacy and potential mechanisms of GXBD in MI treatment. Methods Network pharmacology was used to predict the mechanism of action of GXBD in MI. The predicted results were verified using ECG, echocardiography, HE staining, TTC staining, DHE, JC-1, immunofluorescence, and Western blot analysis in an isoproterenol (ISO)-induced MI rat model. Results Network pharmacology identified 33 active components in GXBD and 139 potential targets against MI, with the PI3K/AKT signaling pathway playing a key role. Compared to the model group, GXBD improved the activities of BNP, CK-MB, and LDH, ameliorated the general condition and cardiac function, and repaired heart damage in MI rats. GXBD decreased MDA and ROS levels, increased SOD and GSH-Px levels, and protected cardiac tissues from oxidative stress. Moreover, GXBD increased ATP content, mitochondrial membrane potential, and the levels of p-PI3K, p-AKT, nuclear NRF2, and MFN2, while decreasing the levels of cytoplasmic NRF2 and DRP1. Conclusion This study suggested that GXBD alleviates myocardial ischemia by ameliorating mitochondrial dysfunction through the PI3K/AKT/NRF2 signaling pathway.
Collapse
Affiliation(s)
- Wanjun Zhu
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Ping Shao
- Benxi National Engineering Research Center for the Pharmaceutics of Traditional Chinese Medicines Co., Ltd., Benxi, Liaoning, China
| | - Ying Ren
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Wenxuan Liu
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Xiaorui Han
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Kexin Zeng
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Chunmei Dai
- Institute of Materia Medica, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Feifei Liu
- The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| |
Collapse
|
2
|
Che W, Jin Y, Chang S, Sun Y, Hou A, Wang C. Prediction of myocardial ischemia-reperfusion injury post-PCI: role of sST2 levels in STEMI patients. BMC Cardiovasc Disord 2025; 25:280. [PMID: 40217168 PMCID: PMC11987365 DOI: 10.1186/s12872-025-04708-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 03/25/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND Myocardial ischemia-reperfusion injury (MIRI) after ST-segment elevation myocardial infarction (STEMI) significantly impacts clinical outcomes. However, only a few studies have examined its clinical predictors and prognostic biomarkers. METHODS Patients with STEMI who received percutaneous coronary intervention (PCI) at Liaoning Provincial People's Hospital between 2020 and 2021 were enrolled in the study. Based on a thorough evaluation of clinical features, which included data obtained from intraoperative angiography and inpatient monitoring, patients were divided into the MIRI group and the non-MIRI group. Upon admission, serum solube growth stimulation expressed gene 2 protein (sST2) levels were assessed, and follow-up examinations were conducted for the patients. RESULTS Patients with MIRI who were admitted to the hospital present elevated serum sST2 levels (P < 0.001). Serum sST2 was recognized as a standalone risk factor contributing to the occurrence of MIRI in STEMI patients who are undergoing PCI (P < 0.001). Throughout the follow-up duration, 37 (17.0%) patients encountered major adverse cardiovascular and cerebrovascular incidents (MACCEs), which included eight (3.6%) deaths from all causes. The Kaplan-Meier assessment revealed that individuals in the MIRI group had an unfavorable prognosis (all log-rank P < 0.05). Both univariable and multivariable Cox regression models were established using MIRI patients as the study cohort. The findings indicated that sST2 levels exceeding 68.98 ng/mL served as one of independent risk factors for predicting MACCEs (all P < 0.001). The model was evaluated using the C-index, AUC, calibration plot, and Decision Curve Analysis (DCA) curve. CONCLUSION Elevated levels of serum sST2 may accurately predict the onset of MIRI following PCI in STEMI patients. Specifically, a serum sST2 concentration > 68.98 ng/mL is a prominent independent risk predictor for overall mortality and MACCE in individuals experiencing MIRI.
Collapse
Affiliation(s)
- Wenjing Che
- Department of Cardiology, The People'S Hospital of China Medical University, the People'S Hospital of Liaoning Province, Shenhe, Shenyang, Liaoning Province, 110016, China.
| | - Yubin Jin
- Department of Cardiology, The People'S Hospital of China Medical University, the People'S Hospital of Liaoning Province, Shenhe, Shenyang, Liaoning Province, 110016, China
| | - Shumin Chang
- Department of Cardiology, The People'S Hospital of China Medical University, the People'S Hospital of Liaoning Province, Shenhe, Shenyang, Liaoning Province, 110016, China
| | - Yihan Sun
- Department of Cardiology, The People'S Hospital of China Medical University, the People'S Hospital of Liaoning Province, Shenhe, Shenyang, Liaoning Province, 110016, China
| | - Aijie Hou
- Department of Cardiology, The People'S Hospital of China Medical University, the People'S Hospital of Liaoning Province, Shenhe, Shenyang, Liaoning Province, 110016, China.
| | - Chengfu Wang
- Department of Cardiology, The People'S Hospital of China Medical University, the People'S Hospital of Liaoning Province, Shenhe, Shenyang, Liaoning Province, 110016, China
| |
Collapse
|
3
|
Fouad MA, Tadros MG, Michel HE. Etanercept ameliorates chronic mild stress-induced depressive-like behavior in rats: Crosstalk between MAPK and STAT3 pathways and norepinephrine and serotonin transporters. Eur J Pharmacol 2024; 978:176801. [PMID: 38945285 DOI: 10.1016/j.ejphar.2024.176801] [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: 04/09/2024] [Revised: 06/13/2024] [Accepted: 06/28/2024] [Indexed: 07/02/2024]
Abstract
Depression is a serious medical illness characterized by persistent feelings of sadness, hopelessness, and lack of interest in daily activities. It can interfere with daily functioning and quality of life. Despite decades of research, the pathophysiology of depression remains incompletely understood. The correlation between depression and inflammation has recently attracted considerable attention. This study investigated the potential antidepressant effect of etanercept, a tumor necrosis factor-alpha (TNF-α) inhibitor, utilizing a chronic mild stress (CMS) model in rats. Male Wistar rats were divided into two groups; one following a non-stressed protocol and the other a stressed protocol for 5 weeks. From the beginning of the third week, rats were treated either with saline daily or with etanercept twice a week (0.3 mg/kg, i.p.) or with fluoxetine daily (10 mg/kg, i.p) as a reference. Etanercept exhibited comparable effects to those of fluoxetine in counteracting CMS-induced behavioral manifestation in the forced swimming and splash tests. Etanercept also restored serotonin and norepinephrine levels to control values in the prefrontal cortex (PFC). Moreover, the current study verified the antioxidant and anti-inflammatory effects of etanercept. Interestingly, etanercept halted the expression of both norepinephrine and serotonin transporters in stressed rats. This could be attributed to abrogation of the p38 mitogen-activated protein kinase (p38 MAPK) and signal transducer and activator of transcription 3 (STAT-3) pathways in the PFC. The findings of the present study contribute to the understanding of the potential of etanercept as an antidepressant and provide insights into the neurobiological mechanisms underlying its therapeutic effects.
Collapse
Affiliation(s)
- Mariam A Fouad
- Laboratory Evaluation Administration, CA of Biological and Innovative Products and Clinical Studies, Egyptian Drug Authority, Giza, Egypt
| | - Mariane G Tadros
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Haidy E Michel
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
4
|
Pasala PK, Raghupathi NK, Yaraguppi DA, Challa RR, Vallamkonda B, Ahmad SF, Chennamsetty Y, Kumari PK, DSNBK P. Potential preventative impact of aloe-emodin nanoparticles on cerebral stroke-associated myocardial injury by targeting myeloperoxidase: In supporting with In silico and In vivo studies. Heliyon 2024; 10:e33154. [PMID: 39022073 PMCID: PMC11253067 DOI: 10.1016/j.heliyon.2024.e33154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/06/2024] [Accepted: 06/14/2024] [Indexed: 07/20/2024] Open
Abstract
The present study examined the potential neuroprotective effects of aloe-emodin (AE) nanoparticles on the cerebral stroke-associated target protein myeloperoxidase (MPO). We investigated the binding interactions between AE and MPO through molecular docking and molecular dynamics simulations. Molecular docking results indicated that AE exhibited a binding energy of -6.9 kcal/mol, whereas it was -7.7 kcal/mol for 2-{[3,5-bis(trifluoromethyl)benzyl]amino}-n-hydroxy-6-oxo-1,6-dihydropyrimidine-5-carboxamide (CCl). Furthermore, molecular dynamics studies demonstrated that AE possesses a stronger binding affinity (-57.137 ± 13.198 kJ/mol) than does CCl (-22.793 ± 30.727 kJ/mol), suggesting that AE has a more substantial inhibitory effect on MPO than does CCl. Despite the therapeutic potential of AE for neurodegenerative disorders, its bioavailability is limited within the body. A proposed hypothesis to enhance the bioavailability of AE is its conversion into aloe-emodin nanoparticles (AENP). The AENPs synthesized through a fabrication method were spherical with a consistent diameter of 104.4 ± 7.9 nm and a polydispersity index ranging from 0.525 to 0.586. In rats experiencing cerebral stroke, there was a notable increase in cerebral infarction size; abnormalities in electrocardiogram (ECG) and electroencephalogram (EEG) patterns; a decrease in brain and cardiac antioxidant activities; and an increase in myeloperoxidase levels compared to those in normal rats. Compared with AE treatment, AENP treatment significantly ameliorated cerebral infarction, normalized ECG and EEG patterns, enhanced brain and cardiac antioxidant activities, and reduced MPO levels in stroke rats. Histopathological evaluations revealed pronounced alterations in the rat hippocampus, with pyknotic nuclei, disarray and loosely packed cells, deterioration of cardiac muscle fibers, and extensive damage to cardiac myocytes, in contrast to those in normal rats. AENP treatment mitigated these pathological changes more effectively than AE treatment in both brain and cardiac cells. These findings support that AENP provides considerable protection against stroke-associated myocardial infarction.
Collapse
Affiliation(s)
- Praveen Kumar Pasala
- Department of Pharmacology, Raghavendra Institute of Pharmaceutical Education and Research, JNTUA, Anantapuramu, Andhra Pradesh, 515721, India
| | - Niranjan Kumar Raghupathi
- Department of Pharmacology, Santhiram College of Pharmacy, JNTUA, Nandyal, 518112, Andhra Pradesh, India
| | - Deepak A. Yaraguppi
- Department of Biotechnology, KLE Technological University, Hubli, Karnataka, 580031, India
| | - Ranadheer Reddy Challa
- Department of Formulation and Development, Quotient Sciences, 3080 McCann Farm Dr, Garnet Valley, PA, 19060, USA
| | - Bhaskar Vallamkonda
- Department of Pharmaceutical Analysis, Odin Pharmaceutical LLC, Somerset, NJ, 08873, USA
| | - Sheikh F. Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Yeswanth Chennamsetty
- Department of Pharmacology, Santhiram College of Pharmacy, JNTUA, Nandyal, 518112, Andhra Pradesh, India
| | - P.V. Kamala Kumari
- Department of Pharmaceutics, Vignan Institute of Pharmaceutical Technology, Duvvada, Visakhapatnam, India
| | - Prasanth DSNBK
- School of Pharmacy & Technology Management, SVKM's Narsee Monjee Institute of Management Studies (NMIMS), Polepally SEZ, TSIIC, Jadcherla, Mahbubnagar, Hyderabad, 509301, India
| |
Collapse
|
5
|
Dhapola R, Beura SK, Sharma P, Singh SK, HariKrishnaReddy D. Oxidative stress in Alzheimer's disease: current knowledge of signaling pathways and therapeutics. Mol Biol Rep 2024; 51:48. [PMID: 38165499 DOI: 10.1007/s11033-023-09021-z] [Citation(s) in RCA: 49] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/23/2023] [Indexed: 01/03/2024]
Abstract
Alzheimer's disease's pathophysiology is still a conundrum. Growing number of evidences have elucidated the involvement of oxidative stress in the pathology of AD rendering it a major target for therapeutic development. Reactive oxygen species (ROS) generated by altered mitochondrial function, dysregulated electron transport chain and other sources elevate aggregated Aβ and neurofibrillary tangles which further stimulating the production of ROS. Oxidative stress induced damage to lipids, proteins and DNA result in neuronal death which leads to AD. In addition, oxidative stress induces apoptosis that is triggered by the modulation of ERK1/2 and Nrf2 pathway followed by increased GSK-3β expression and decreased PP2A activity. Oxidative stress exaggerates disease condition by interfering with various signaling pathways like RCAN1, CREB/ERK, Nrf2, PP2A, NFκB and PI3K/Akt. Studies have reported the role of TNF-α in oxidative stress stimulation that has been regulated by drugs like etanercept increasing the level of anti-oxidants. Other drugs like pramipexole, memantine, carvedilol, and melatonin have been reported to activate CREB/RCAN1 and Nrf2 pathways. In line with this, epigallocatechin gallate and genistein also target Nrf2 and CREB pathway leading to activation of downstream pathways like ARE and Keap1 which ameliorate oxidative stress condition. Donepezil and resveratrol reduce oxidative stress and activate AMPK pathway along with PP2A activation thus promoting tau dephosphorylation and neuronal survival. This study describes in detail the role of oxidative stress in AD, major signaling pathways involving oxidative stress induced AD and drugs under development targeting these pathways which may aid in therapeutic advances for AD.
Collapse
Affiliation(s)
- Rishika Dhapola
- Advanced Pharmacology and Neuroscience Laboratoty, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India
| | - Samir K Beura
- Department of Zoology, School of Basic and Applied Science, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India
| | - Prajjwal Sharma
- Advanced Pharmacology and Neuroscience Laboratoty, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India
| | - Sunil K Singh
- Department of Zoology, School of Basic and Applied Science, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India.
| | - Dibbanti HariKrishnaReddy
- Advanced Pharmacology and Neuroscience Laboratoty, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India.
| |
Collapse
|
6
|
Zhao C, Liu T, Wei H, Li J. Serum oxidative stress factors predict myocardial ischemia reperfusion injury after percutaneous coronary intervention in patients with acute myocardial infarction and type 2 diabetes mellitus. ADVANCES IN INTERVENTIONAL CARDIOLOGY 2023; 19:333-342. [PMID: 38187486 PMCID: PMC10767562 DOI: 10.5114/aic.2023.133475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 09/14/2023] [Indexed: 01/09/2024] Open
Abstract
Introduction Serum oxidative stress factors may be considered to be essential parameters for indicating cell oxidative damage. Aim We designed this study to investigate the clinical diagnostic value of serum oxidative stress factors (superoxide dismutase (SOD), malondialdehyde (MDA), and myeloperoxidase (MPO)) combined with ischemia-modified albumin (IMA) and heat shock protein 70 (HSP70) for myocardial ischemia-reperfusion injury (MIRI) after percutaneous coronary intervention (PCI) in elderly patients with acute myocardial infarction (AMI) and type 2 diabetes mellitus (T2DM). Material and methods From November 2020 to August 2021, 94 patients with AMI + T2DM and 86 patients with AMI were enrolled in the study; they were sub-grouped into the MIRI and non-MIRI groups following the occurrence of MIRI within 48 h after PCI. SOD, MDA, MPO, IMA, and HSP70 levels were determined. The clinical values of the combined serum oxidative stress factors, IMA, and HSP70 levels to predict MIRI events were analyzed. Results There was a higher probability of MIRI events in the AMI + T2DM group than the AMI group (p < 0.05). The ROC curve for the combined prediction of SOD, MDA, MPO, IMA, and HSP70 for the occurrence of MIRI events was higher in both the AMI and the AMI + T2DM groups than for predictive factors alone (all p < 0.05). Conclusions Combined prediction of SOD, MDA, MPO, IMA, and HSP70 has the highest diagnostic value for predicting MIRI events after PCI in AMI patients, especially in patients with AMI combined with T2DM.
Collapse
Affiliation(s)
- Chunyu Zhao
- Department of General Practice, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Tianze Liu
- Clinical Medical College of Jiamusi University, Jiamusi, Heilongjiang, China
| | - Hong Wei
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Jianing Li
- Department of General Practice, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
7
|
Yao Y, Lin L, Tang W, Shen Y, Chen F, Li N, Wang B. Pretreatment with geniposide mitigates myocardial ischemia/reperfusion injury by modulating inflammatory response through TLR4/NF-κB pathway. Eur J Histochem 2023; 67:3742. [PMID: 37682077 PMCID: PMC10518652 DOI: 10.4081/ejh.2023.3742] [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: 03/31/2023] [Accepted: 08/11/2023] [Indexed: 09/09/2023] Open
Abstract
Geniposide (GEN), a medical herb, is known for its therapeutic applications in cardiovascular diseases, though its efficacy in treating myocardial ischemia/reperfusion injury (MI/RI) is yet to be fully elucidated. This study is an endeavor to explore the potential protective mechanism of GEN against MI/RI. To simulate the MI/RI condition, the left anterior descending artery was occluded for 30 min, followed by a reperfusion period of 120 min in a rat model. Three dosages (50, 100, or 150 mg/kg) of GEN were intraperitoneally injected to the Sprague-Dawley rats once a day, for seven days before the ligation of the artery. The rats were categorized into sham group, MI/RI group, and three different dosages GEN-treated groups. As the results showed, the pretreatment with GEN mitigated myocardial injury, reduced infarct volume, inhibited apoptosis, enhanced superoxide dismutase activity, and decreased malondialdehyde and myeloperoxidase activity, as well as serum creatine kinase-MB and lactate dehydrogenase levels. Moreover, GEN ameliorated MI/RI by downregulating protein expression of toll-like receptor 4, myeloid differentiation primary response 88, and p-nuclear factor-κB. In conclusion, the pretreatment of GEN may be considered as a potential therapeutic option for MI/RI.
Collapse
Affiliation(s)
- Yanmei Yao
- Department of General Medicine, The Affiliated Hospital, Hangzhou Normal University, Hangzhou, Zhejiang.
| | - Leqing Lin
- Department of Critical Care Medicine, The Affiliated Hospital, Hangzhou Normal University, Hangzhou, Zhejiang.
| | - Wenxue Tang
- Department of Critical Care Medicine, The Affiliated Hospital, Hangzhou Normal University, Hangzhou, Zhejiang.
| | - Yueliang Shen
- Department of Pathophysiology, Zhejiang University Medical College, Hangzhou, Zhejiang.
| | - Fayu Chen
- Department of General Medicine, The Affiliated Hospital, Hangzhou Normal University, Hangzhou, Zhejiang.
| | - Ning Li
- Department of Hematology and Oncology, The Affiliated Hospital, Hangzhou Normal University, Hangzhou, Zhejiang.
| | - Baiyong Wang
- Department of Critical Care Medicine, The Affiliated Hospital, Hangzhou Normal University, Hangzhou, Zhejiang.
| |
Collapse
|
8
|
Hassan NF, Hassan AH, El-Ansary MR. Cytokine modulation by etanercept ameliorates metabolic syndrome and its related complications induced in rats administered a high-fat high-fructose diet. Sci Rep 2022; 12:20227. [PMID: 36418417 PMCID: PMC9684438 DOI: 10.1038/s41598-022-24593-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 11/17/2022] [Indexed: 11/25/2022] Open
Abstract
The aim of the present study was to investigate the effect of etanercept (ETA)-an anti-tumor necrosis factor α (TNF-α) monoclonal antibody-on metabolic disorders such as obesity, hypertension, dyslipidemia, and insulin resistance associated with the metabolic syndrome (MS). MS was induced in rats via high-fat high-fructose (HFHF) administration for 8 weeks. Rats were divided into three groups: negative control, HFHF model, and ETA-treated groups [HFHF + ETA (0.8 mg/kg/twice weekly, subcutaneously) administered in the last 4 weeks]. ETA effectively diminished the prominent features of MS via a significant reduction in the percent body weight gain along with the modulation of adipokine levels, resulting in a significant elevation of serum adiponectin consistent with TNF-α and serum leptin level normalization. Moreover, ETA enhanced dyslipidemia and the elevated blood pressure. ETA managed the prominent features of MS and its associated complications via the downregulation of the hepatic inflammatory pathway that induces nonalcoholic steatohepatitis (NASH)-from the expression of Toll-like receptor 4, nuclear factor kappa B, and TNF-α until that of transforming growth factor-in addition to significant improvements in glucose utilization, insulin sensitivity, and liver function parameter activity and histopathological examination. ETA was effective for the treatment of all prominent features of MS and its associated complications, such as type II diabetes mellitus and NASH.
Collapse
Affiliation(s)
- Noha F. Hassan
- grid.440876.90000 0004 0377 3957Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt
| | - Azza H. Hassan
- grid.7776.10000 0004 0639 9286Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Mona R. El-Ansary
- grid.440876.90000 0004 0377 3957Department of Biochemistry, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt
| |
Collapse
|
9
|
Chen S, Li A, Wu J, Huang Y, Zou T, Tailaiti T, Wang J. Dexmedetomidine reduces myocardial ischemia-reperfusion injury in young mice through MIF/AMPK/GLUT4 axis. BMC Anesthesiol 2022; 22:289. [PMID: 36104681 PMCID: PMC9472426 DOI: 10.1186/s12871-022-01825-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 08/30/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Reperfusion of ischemic tissue has adverse impact on the myocardium. Dexmedetomidine (Dex) is a α2-adrenergic receptor (α2-AR) agonist with sedative and analgesic effects. Macrophage migration inhibition factor (MIF) is a pressure-regulating cytokine and is responsible for inflammatory and immune diseases. This study aims to reveal the consequences of Dex on myocardial ischemia-reperfusion injury (IRI) in young mice. METHODS Fifty mice were raised and examined. At the end of the experiment, all mice were euthanized. The anterior descending department of the left coronary artery in mice was under ischemia for 60 min, then the ligation line was released and reperfused for 120 min to establish the IRI model. Mice were randomly divided into Sham, control, treatment using 4,5-dihydro-3-(4-hydroxyphenyl)-5-isoxazoleacetic acid (ISO-1), Dex treatment, and Dex combined ISO-1 treatment groups. Interleukin (IL)-6, IL-10 and tumor necrosis factor (TNF-α) were determined by enzyme-linked immunosorbent assay (ELISA). Reactive oxygen species (ROS) and ATP levels were recorded. The expressions of MIF, P-adenosine monophosphate-activated kinase α (AMPKα), glucose transporter (GLUT)4, Bax and Bcl-2 were detected by Western Blot (WB). Hematoxylin and Eosin (H&E) staining was used to study cell morphology. Apoptosis was detected by terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL) assay. Echocardiography was carried out at the end of reperfusion, and the infarct size was calculated by Electron microscopy. RESULTS I/R + Dex group showed significantly increased IL-6 and TNF-α levels and reduced myocardial cell necrosis and apoptosis. H&E staining showed alleviated myocardial disorder, myocardial cell swelling, myocardial fiber fracture, and inflammatory cell infiltration in I/R + Dex group. Myocardial cell necrosis and apoptosis were significantly reduced in I/R + Dex group. ATP level in myocardial tissue of mice in I/R group was substantially decreased, while that in Dex group was increased. WB results showed that MIF, P-AMPK α, GLUT4 and Bcl-2 levels were increased and Bax levels were decreased in I/R + Dex group. CONCLUSION Dex may exert myocardial protection in young mice through MIF/AMPK/GLUT4 axis.
Collapse
Affiliation(s)
- Siyu Chen
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University, No.137, Liyushan Road, Xinshi District, Urumqi, Xinjiang, 830054, Uygur Autonomous Region, China
| | - Aimei Li
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University, No.137, Liyushan Road, Xinshi District, Urumqi, Xinjiang, 830054, Uygur Autonomous Region, China
| | - Jianjiang Wu
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University, No.137, Liyushan Road, Xinshi District, Urumqi, Xinjiang, 830054, Uygur Autonomous Region, China
| | - Yidan Huang
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University, No.137, Liyushan Road, Xinshi District, Urumqi, Xinjiang, 830054, Uygur Autonomous Region, China
| | - Tiantian Zou
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University, No.137, Liyushan Road, Xinshi District, Urumqi, Xinjiang, 830054, Uygur Autonomous Region, China
| | - Taiwangu Tailaiti
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University, No.137, Liyushan Road, Xinshi District, Urumqi, Xinjiang, 830054, Uygur Autonomous Region, China
| | - Jiang Wang
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University, No.137, Liyushan Road, Xinshi District, Urumqi, Xinjiang, 830054, Uygur Autonomous Region, China.
| |
Collapse
|
10
|
Protective effects of dexpanthenol in carbon tetrachloride-induced myocardial toxicity in rats. Tissue Cell 2022; 77:101824. [DOI: 10.1016/j.tice.2022.101824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/29/2022] [Accepted: 05/13/2022] [Indexed: 11/19/2022]
|
11
|
Kouyoumdjian A, Tchervenkov J, Paraskevas S. TFNR2 in Ischemia-Reperfusion Injury, Rejection, and Tolerance in Transplantation. Front Immunol 2022; 13:903913. [PMID: 35874723 PMCID: PMC9300818 DOI: 10.3389/fimmu.2022.903913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 06/10/2022] [Indexed: 11/28/2022] Open
Abstract
Tumor necrosis factor receptor 2 (TNFR2) has been shown to play a crucial role in CD4+ T regulatory cells (CD4+Tregs) expansion and suppressive function. Increasing evidence has also demonstrated its role in a variety of immune regulatory cell subtypes such as CD8+ T regulatory cells (CD8+ Tregs), B regulatory cells (Bregs), and myeloid-derived suppressor cells (MDSCs). In solid organ transplantation, regulatory immune cells have been associated with decreased ischemia-reperfusion injury (IRI), improved graft survival, and improved overall outcomes. However, despite TNFR2 being studied in the context of autoimmune diseases, cancer, and hematopoietic stem cell transplantation, there remains paucity of data in the context of solid organ transplantation and islet cell transplantation. Interestingly, TNFR2 signaling has found a clinical application in islet transplantation which could guide its wider use. This article reviews the current literature on TNFR2 expression in immune modulatory cells as well as IRI, cell, and solid organ transplantation. Our results highlighted the positive impact of TNFR2 signaling especially in kidney and islet transplantation. However, further investigation of TNFR2 in all types of solid organ transplantation are required as well as dedicated studies on its therapeutic use during induction therapy or treatment of rejection.
Collapse
Affiliation(s)
- Araz Kouyoumdjian
- Division of Experimental Surgery, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
- Division of General Surgery, Department of Surgery, McGill University, Montreal, QC, Canada
- *Correspondence: Araz Kouyoumdjian,
| | - Jean Tchervenkov
- Division of Experimental Surgery, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
- Division of General Surgery, Department of Surgery, McGill University, Montreal, QC, Canada
| | - Steven Paraskevas
- Division of Experimental Surgery, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
- Division of General Surgery, Department of Surgery, McGill University, Montreal, QC, Canada
| |
Collapse
|
12
|
Antar SA, El-Gammal MA, Hazem RM, Moustafa YM. Etanercept Mitigates Cadmium Chloride-induced Testicular Damage in Rats "An Insight into Autophagy, Apoptosis, Oxidative Stress and Inflammation". ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:28194-28207. [PMID: 34993805 DOI: 10.1007/s11356-021-18401-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/25/2021] [Indexed: 06/14/2023]
Abstract
RATIONALE Cadmium (Cd) is an environmental and occupational toxin that represents a serious health hazard to humans and other animals. One of the negative consequences of cadmium exposure is testicular injury. OBJECTIVE This study aimed to investigate the therapeutic effect of etanercept against cadmium chloride-induced testicular damage and the probable underlying mechanisms of its action. METHODS A total of sixty rats were divided into six groups: control, cadmium chloride (CdCl2) (7 mg/ kg i.p.), and CdCl2 treated with etanercept (5,10 and 15 mg/kg s.c.) and etanercept only (15 mg/kg s.c.). CdCl2 was administrated as a single dose, while etanercept was administered every 3 days for 3 weeks. RESULTS CdCl2 reduced serum testosterone, testicular glutathione (GSH), catalase (CAT), and superoxide dismutase (SOD). However, it elevated the levels of malondialdehyde (MDA) and microtubule-associated protein light chain 3B (LC3B) in the testes. Cadmium caused pathogenic alterations as well as increased levels of inflammatory biomarkers such as tumor necrosis factor-alpha (TNF-α) and nuclear factor-kappa B (NF-κB). Besides, the gene expressions of caspase-3 and inducible nitric oxide synthase (i-NOS) and Beclin-1 protein increased with CdCl2 exposure. Interestingly, etanercept relieved the previous toxic effects induced by CdCl2 in a dose-dependent manner as evidenced by inhibition of oxidative stress, inflammatory markers, Beclin-1, LC3B, and caspase-3 accompanied by improvement in histopathological changes. CONCLUSION Etanercept provides a potential therapeutic approach to treat testicular tissue against the damaging effects of Cd by reducing oxidative stress, inflammation, apoptosis, and autophagy.
Collapse
Affiliation(s)
- Samar A Antar
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Horus University, New Damietta, 34518, Egypt.
| | - Mohamad A El-Gammal
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Horus University, New Damietta, 34518, Egypt
| | - Reem M Hazem
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt
| | - Yasser M Moustafa
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Badr University, Badr University Cairo, 11829, Egypt
| |
Collapse
|
13
|
Duan H, Li M, Liu J, Sun J, Wu C, Chen Y, Guo X, Liu X. An Integrated Approach Based on Network Analysis Combined With Experimental Verification Reveals PI3K/Akt/Nrf2 Signaling Is an Important Way for the Anti-Myocardial Ischemia Activity of Yi-Qi-Tong-Luo Capsule. Front Pharmacol 2022; 13:794528. [PMID: 35250556 PMCID: PMC8889021 DOI: 10.3389/fphar.2022.794528] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/24/2022] [Indexed: 12/11/2022] Open
Abstract
Background: Yiqi-Tongluo Capsule (YTC) is a Chinese traditional patent medicine that has been used in the treatment of myocardial ischemia (MI). However, its molecular mechanisms against MI have not been clear. Methods: Network analysis and experimental verification were used to explore the potential molecular mechanisms of YTC for MI treatment. Firstly, the main components in the capsules and the potential targets of these components were predicted by online databases. The MI related genes were collected from Genecards and Online Mendelian Inheritance in Man (OMIM) databases. The drug targets and disease targets were intersected, and then the protein-protein interaction (PPI) and Drug-Molecular-Target-Disease Network (DMTD) were constructed, and GO enrichment analysis and KEGG pathway enrichment analysis were performed. Based on the H2O2-stimulated H9c2 cells, flow cytometry, western blot (WB) and immunofluorescence experiments were performed to verify the network analysis prediction. Results: A total of 100 active components and 165 targets of YTC were predicted, in which there were 109 targets intersected with the targets of MI. GO and KEGG analysis showed that these potential targets were related to a variety of biological processes and molecular mechanisms, including oxidative stress and PI3K/AKT pathway. Astragaloside IV (AS IV) and paeoniflorin (PAE) might be the main active components in YTC. The results of cell counting kit-8 (CCK-8) showed that YTC alleviated the damage of H2O2 to H9c2 cells. The results of flow cytometry, DAPI staining and JC-1 probe showed that YTC alleviated H2O2 induced apoptosis in H9c2 cells. In addition, YTC reduced the level of intracellular superoxide anion, increased the activities of superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GSH-Px), and reduced the content of malondialdehyde (MDA) in H2O2-induced H9c2 cells. The results of immunofluorescence and WB showed that the phosphorylation of PI3K and Akt were increased, the expression of Bcl-2 was up-regulated and the expression of cleaved caspase-3 and Bax were down-regulated. Besides, the nuclear translocation of Nrf2 were increased. Conclusion: In conclusion, the results of this study showed that YTC might alleviate MI by suppressing apoptosis induced by oxidative stress via the PI3K/Akt/Nrf2 signal pathway.
Collapse
Affiliation(s)
- Huxinyue Duan
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Meiyan Li
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jia Liu
- Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Guangyuan Hospital of Traditional Chinese Medicine, Guangyuan, China.,Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiayi Sun
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chunjie Wu
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu Chen
- Guangyuan Hospital of Traditional Chinese Medicine, Guangyuan, China
| | - Xiaohui Guo
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xinglong Liu
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
14
|
Bai Q, Li Y, Song K, Huang J, Qin L. Knockdown of XIST up-regulates 263294miR-340-5p to relieve myocardial ischaemia-reperfusion injury via inhibiting cyclin D1. ESC Heart Fail 2021; 9:1050-1060. [PMID: 34970865 PMCID: PMC8934972 DOI: 10.1002/ehf2.13766] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 11/24/2021] [Accepted: 12/02/2021] [Indexed: 12/19/2022] Open
Abstract
Aim Long non‐coding RNAs (lncRNAs) are known to participate in various human diseases, while the role of X inactive‐specific transcript (XIST) binding microRNA‐340‐5p (miR‐340‐5p) remains seldom studied. We aim to identify the role of the XIST/miR‐340‐5p/cyclin D1 (CCND1) axis in the myocardial ischaemia–reperfusion injury (MIRI). Methods and results The mouse MIRI models were established. The expression of XIST, miR‐340‐5p, and CCND1 in mouse myocardial tissues in MIRI mice was assessed. The MIRI mice were respectively treated with altered XIST, miR‐340‐5p, or CCND1. The changes of myocardial enzyme activity were assessed, and the cardiac function was evaluated. Myocardial pathological changes, cardiomyocyte apoptosis and related apoptotic factors, oxidative stress and inflammatory factors were observed in myocardial tissues in mice with MIRI. The binding relationships between XIST and miR‐340‐5p, and between miR‐340‐5p and CCND1 were confirmed. XIST and CCND1 were up‐regulated while miR‐340‐5p was down‐regulated in MIRI mice. Silenced XIST could elevated miR‐340‐5p expression and reduced CCND1 expression, so as to promoted cardiac function and suppressed myocardial enzyme activity, ameliorated pathological changes, decelerated cardiomyocyte apoptosis by elevating Bcl‐2 but reducing the levels of Bax and Caspase‐3, attenuated inflammatory response by repressing IL‐6 and TNF‐α levels, and mitigated oxidative stress by reducing MDA contents and increasing CAT, GSH‐Px, and SOD levels in MIRI mice. XIST sponged miR‐340‐5p and miR‐340‐5p targeted CCND1. Conclusions Knockdown of XIST up‐regulates miR‐340‐5p to relieve MIRI via inhibiting CCND1.
Collapse
Affiliation(s)
- Qijun Bai
- Department of Cardiovascular Medicine Ward II, Zhengzhou Central Hospital Affiliated to Zhengzhou University, 16 North Tongbai Road, Zhongyuan District, Zhengzhou, Henan, 450000, China
| | - Yan Li
- Department of Cardiovascular Medicine Ward II, Zhengzhou Central Hospital Affiliated to Zhengzhou University, 16 North Tongbai Road, Zhongyuan District, Zhengzhou, Henan, 450000, China
| | - Kunpeng Song
- Department of Cardiovascular Medicine Ward II, Zhengzhou Central Hospital Affiliated to Zhengzhou University, 16 North Tongbai Road, Zhongyuan District, Zhengzhou, Henan, 450000, China
| | - Jie Huang
- Department of Geriatric Medicine, Zhengzhou Central Hospital, Zhengzhou, Henan, China
| | - Li Qin
- Department of Cardiovascular Medicine Ward II, Zhengzhou Central Hospital Affiliated to Zhengzhou University, 16 North Tongbai Road, Zhongyuan District, Zhengzhou, Henan, 450000, China
| |
Collapse
|
15
|
Patel PM, Connolly MR, Coe TM, Calhoun A, Pollok F, Markmann JF, Burdorf L, Azimzadeh A, Madsen JC, Pierson RN. Minimizing Ischemia Reperfusion Injury in Xenotransplantation. Front Immunol 2021; 12:681504. [PMID: 34566955 PMCID: PMC8458821 DOI: 10.3389/fimmu.2021.681504] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 08/12/2021] [Indexed: 12/21/2022] Open
Abstract
The recent dramatic advances in preventing "initial xenograft dysfunction" in pig-to-non-human primate heart transplantation achieved by minimizing ischemia suggests that ischemia reperfusion injury (IRI) plays an important role in cardiac xenotransplantation. Here we review the molecular, cellular, and immune mechanisms that characterize IRI and associated "primary graft dysfunction" in allotransplantation and consider how they correspond with "xeno-associated" injury mechanisms. Based on this analysis, we describe potential genetic modifications as well as novel technical strategies that may minimize IRI for heart and other organ xenografts and which could facilitate safe and effective clinical xenotransplantation.
Collapse
Affiliation(s)
- Parth M. Patel
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Margaret R. Connolly
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Taylor M. Coe
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Anthony Calhoun
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Surgery, Division of Cardiac Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Franziska Pollok
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Anesthesiology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - James F. Markmann
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Surgery, Division of Transplantation, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Lars Burdorf
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Surgery, Division of Cardiac Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Agnes Azimzadeh
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Surgery, Division of Cardiac Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Joren C. Madsen
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Surgery, Division of Cardiac Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Richard N. Pierson
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Surgery, Division of Cardiac Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
16
|
Bikhet M, Iwase H, Yamamoto T, Jagdale A, Foote JB, Ezzelarab M, Anderson DJ, Locke JE, Eckhoff DE, Hara H, Cooper DKC. What Therapeutic Regimen Will Be Optimal for Initial Clinical Trials of Pig Organ Transplantation? Transplantation 2021; 105:1143-1155. [PMID: 33534529 DOI: 10.1097/tp.0000000000003622] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
We discuss what therapeutic regimen might be acceptable/successful in the first clinical trial of genetically engineered pig kidney or heart transplantation. As regimens based on a calcineurin inhibitor or CTLA4-Ig have proved unsuccessful, the regimen we administer to baboons is based on induction therapy with antithymocyte globulin, an anti-CD20 mAb (Rituximab), and cobra venom factor, with maintenance therapy based on blockade of the CD40/CD154 costimulation pathway (with an anti-CD40 mAb), with rapamycin, and a corticosteroid. An anti-inflammatory agent (etanercept) is administered for the first 2 wk, and adjuvant therapy includes prophylaxis against thrombotic complications, anemia, cytomegalovirus, and pneumocystis. Using this regimen, although antibody-mediated rejection certainly can occur, we have documented no definite evidence of an adaptive immune response to the pig xenograft. This regimen could also form the basis for the first clinical trial, except that cobra venom factor will be replaced by a clinically approved agent, for example, a C1-esterase inhibitor. However, none of the agents that block the CD40/CD154 pathway are yet approved for clinical use, and so this hurdle remains to be overcome. The role of anti-inflammatory agents remains unproven. The major difference between this suggested regimen and those used in allotransplantation is the replacement of a calcineurin inhibitor with a costimulation blockade agent, but this does not appear to increase the complications of the regimen.
Collapse
Affiliation(s)
- Mohamed Bikhet
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL
| | - Hayato Iwase
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL
| | - Takayuki Yamamoto
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL
| | - Abhijit Jagdale
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL
| | - Jeremy B Foote
- Department of Microbiology and Animal Resources Program, University of Alabama at Birmingham, Birmingham, AL
| | - Mohamed Ezzelarab
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Douglas J Anderson
- Division of Transplantation, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL
| | - Jayme E Locke
- Division of Transplantation, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL
| | - Devin E Eckhoff
- Division of Transplantation, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL
| | - Hidetaka Hara
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL
| | - David K C Cooper
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
17
|
Huang J, Liu Y, Wang M, Wang R, Ling H, Yang Y. FoxO4 negatively modulates USP10 transcription to aggravate the apoptosis and oxidative stress of hypoxia/reoxygenation-induced cardiomyocytes by regulating the Hippo/YAP pathway. J Bioenerg Biomembr 2021; 53:541-551. [PMID: 34251583 DOI: 10.1007/s10863-021-09910-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/01/2021] [Indexed: 12/25/2022]
Abstract
Acute myocardial infarction (AMI) is the main cause of death in the whole world. This study aimed to investigate whether forkhead box O4 (FoxO4) could negatively modulate ubiquitin specific peptidase 10 (USP10) transcription to aggravate the apoptosis and oxidative stress of hypoxia/reoxygenation (H/R)-induced cardiomyocytes through Hippo/YAP pathway. mRNA expression as well as protein expressions of USP10 and FoxO4 in H9C2 cells after H/R induction or transfection were respectively detected by Reverse transcription-quantitative (RT-q) PCR analysis and Western blot. The viability and apoptosis of H9C2 cells after H/R induction or transfection were respectively detected by CCK-8 and TUNEL assays. The expressions of lactate dehydrogenase (LDH), superoxide dismutase (SOD), glutathione peroxidase (GSH-Px) and malondialdehyde (MDA) in H9C2 cells after H/R induction or transfection were analyzed using appropriate kits and intracellular reactive oxygen species (ROS) levels were detected using a ROS Assay Kit. Dual luciferase reporter assay and Chromatin Immunoprecipitation (ChIP) have adopted to confirm the combination of USP10 and FoxO4. Western blot was also used to analyze the expression of apoptosis-related proteins and Hippo/YAP pathway-related proteins. As a result, USP10 expression was decreased in H/R-induced H9C2 cells in a time-dependent manner. USP10 overexpression increased the viability and suppressed the apoptosis and oxidative stress of H/R-induced H9C2 cells. In addition, FoxO4 modulated USP10 transcription. FoxO4 expression was increased in H9C2 cells induced by H/R. FoxO4 overexpression could reverse the protective effects of USP10 overexpression on H/R-induced H9C2 cells by regulating the Hippo/YAP signaling pathway. In conclusion, FoxO4 negatively modulated USP10 transcription to aggravate the apoptosis and oxidative stress of H/R-induced H9C2 cells via blocking Hippo/YAP pathway.
Collapse
Affiliation(s)
- Jingwen Huang
- Department of Nursing, Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Yu Liu
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Qiaokou District, Wuhan, 430030, Hubei Province, China
| | - Mei Wang
- Department of Nursing, Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Rong Wang
- Department of Nursing, Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Huifen Ling
- Department of Nursing, Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Yan Yang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Qiaokou District, Wuhan, 430030, Hubei Province, China.
| |
Collapse
|
18
|
Li A, Zhang X, Luo Q. Neohesperidin alleviated pathological damage and immunological imbalance in rat myocardial ischemia-reperfusion injury via inactivation of JNK and NF-κB p65. Biosci Biotechnol Biochem 2021; 85:251-261. [PMID: 33604646 DOI: 10.1093/bbb/zbaa064] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 10/05/2020] [Indexed: 12/13/2022]
Abstract
Neohesperidin (NEO) exerts antiviral, antioxidant, anti-inflammation, and antitumor effects in some diseases. The purpose of this study was to investigate the effect and mechanism of NEO on myocardial ischemia-reperfusion (I/R) injury. Results indicated that NEO suppressed the levels of serum inflammatory cytokines, myocardial damage markers, and oxidative stress markers, and increased the levels of antioxidant in myocardial I/R rats. NEO also inhibited cell apoptosis. Besides, NEO also inhibited the phosphorylation of c-Jun N-terminal kinases (JNK) and nuclear factor kappa B (NF-κB) p65. Furthermore, the protective effects of NEO on myocardial tissue damage, inflammatory cytokines, myocardial injury markers, oxidative stress markers, cell apoptosis, spleen, thymus and liver indices, and phagocytic indices were reversed by JNK activator and NF-κB activator, respectively. In conclusion, NEO alleviates myocardial damage, oxidative stress, cell apoptosis, and immunological imbalance in I/R injury via the inactivation of JNK and NF-κB, making NEO a potential agent for myocardial I/R therapy.
Collapse
Affiliation(s)
- Aihua Li
- Internal Medicine-Cardiovascular Department, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Xin Zhang
- Internal Medicine-Cardiovascular Department, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Qiuping Luo
- Internal Medicine-Cardiovascular Department, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu Province, China
| |
Collapse
|
19
|
Hsu CC, Li Y, Hsu CT, Cheng JT, Lin MH, Cheng KC, Chen SW. Etanercept Ameliorates Cardiac Fibrosis in Rats with Diet-Induced Obesity. Pharmaceuticals (Basel) 2021; 14:320. [PMID: 33916242 PMCID: PMC8067047 DOI: 10.3390/ph14040320] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/24/2021] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
Diet-induced obesity (DIO) is considered the main risk factor for cardiovascular diseases. Increases in the plasma levels of tumor necrosis factor alpha (TNF-α) is associated with DIO. Etanercept, a TNF-α inhibitor, has been shown to alleviate cardiac hypertrophy. To investigate the effect of etanercept on cardiac fibrosis in DIO model, rats on high fat diet (HFD) were subdivided into two groups: the etanercept group and vehicle group. Cardiac injury was identified by classic methods, while fibrosis was characterized by histological analysis of the hearts. Etanercept treatment at 0.8 mg/kg/week twice weekly by subcutaneous injection effectively alleviates the cardiac fibrosis in HFD-fed rats. STAT3 activation seems to be induced in parallel with fibrosis-related gene expression in the hearts of HFD-fed rats. Decreased STAT3 activation plays a role in the etanercept-treated animals. Moreover, fibrosis-related genes are activated by palmitate in parallel with STAT3 activation in H9c2 cells. Etanercept may inhibit the effects of palmitate, but it is less effective than a direct inhibitor of STAT3. Direct inhibition of STAT3 activation by etanercept seems unlikely. Etanercept has the ability to ameliorate cardiac fibrosis through reduction of STAT3 activation after the inhibition of TNF-α and/or its receptor.
Collapse
Affiliation(s)
- Chia-Chen Hsu
- Department of Exercise and Health Sciences, University of Taipei, Taipei City 110, Taiwan;
- Department of Otorhinolaryngology, Taipei City Hospital, Taipei City 110, Taiwan
- Graduate Institute of Gerontology and Health Care Management, Chang Gung University of Science and Technology, Guishan, Taoyuan City 613, Taiwan;
| | - Yingxiao Li
- Department of Nursing, Tzu Chi University of Science and Technology, Hualien City 970, Taiwan;
| | - Chao-Tien Hsu
- Department of Pathology, I-Shou University Medical Center, Yanchao District, Kaohsiung City 824, Taiwan;
| | - Juei-Tang Cheng
- Department of Medical Research, Chi-Mei Medical Center, Tainan City 710, Taiwan;
| | - Mang-Hung Lin
- Graduate Institute of Gerontology and Health Care Management, Chang Gung University of Science and Technology, Guishan, Taoyuan City 613, Taiwan;
| | - Kai-Chun Cheng
- Department of Pharmacy, College of Pharmacy, Tajen University, Pingtung 907, Taiwan
| | - Shang-Wen Chen
- Division of Cardiology, Chi-Mei Medical Center Liouying, Tainan City 736, Taiwan
| |
Collapse
|
20
|
Gianazza E, Brioschi M, Martinez Fernandez A, Casalnuovo F, Altomare A, Aldini G, Banfi C. Lipid Peroxidation in Atherosclerotic Cardiovascular Diseases. Antioxid Redox Signal 2021; 34:49-98. [PMID: 32640910 DOI: 10.1089/ars.2019.7955] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Significance: Atherosclerotic cardiovascular diseases (ACVDs) continue to be a primary cause of mortality worldwide in adults aged 35-70 years, occurring more often in countries with lower economic development, and they constitute an ever-growing global burden that has a considerable socioeconomic impact on society. The ACVDs encompass diverse pathologies such as coronary artery disease and heart failure (HF), among others. Recent Advances: It is known that oxidative stress plays a relevant role in ACVDs and some of its effects are mediated by lipid oxidation. In particular, lipid peroxidation (LPO) is a process under which oxidants such as reactive oxygen species attack unsaturated lipids, generating a wide array of oxidation products. These molecules can interact with circulating lipoproteins, to diffuse inside the cell and even to cross biological membranes, modifying target nucleophilic sites within biomolecules such as DNA, lipids, and proteins, and resulting in a plethora of biological effects. Critical Issues: This review summarizes the evidence of the effect of LPO in the development and progression of atherosclerosis-based diseases, HF, and other cardiovascular diseases, highlighting the role of protein adduct formation. Moreover, potential therapeutic strategies targeted at lipoxidation in ACVDs are also discussed. Future Directions: The identification of valid biomarkers for the detection of lipoxidation products and adducts may provide insights into the improvement of the cardiovascular risk stratification of patients and the development of therapeutic strategies against the oxidative effects that can then be applied within a clinical setting.
Collapse
Affiliation(s)
- Erica Gianazza
- Proteomics Unit, Monzino Cardiology Center IRCCS, Milan, Italy
| | - Maura Brioschi
- Proteomics Unit, Monzino Cardiology Center IRCCS, Milan, Italy
| | | | | | | | - Giancarlo Aldini
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Cristina Banfi
- Proteomics Unit, Monzino Cardiology Center IRCCS, Milan, Italy
| |
Collapse
|
21
|
Zhang XY, Huang Z, Li QJ, Zhong GQ, Meng JJ, Wang DX, Tu RH. Role of HSP90 in suppressing TLR4-mediated inflammation in ischemic postconditioning. Clin Hemorheol Microcirc 2020; 76:51-62. [PMID: 32651307 DOI: 10.3233/ch-200840] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Myocardial inflammation mediated by toll-like receptor 4 (TLR4) plays an active role in myocardial ischemia/reperfusion (I/R) injury. Studies show that heat shock protein 90 (HSP90) is involved in ischemic postconditioning (IPostC) cardioprotection. This study investigates the roles of TLR4 and HSP90 in IPostC. METHODS Rats were subjected to 30 min ischemia, then 2 h reperfusion. IPostC was applied by three cycles of 30 s reperfusion, then 30 s reocclusion at reperfusion onset. Sixty rats were randomly divided into four groups: sham, I/R, IPostC, and geldanamycin (GA, HSP90 inhibitor, 1 mg/kg) plus IPostC (IPostC + GA). RESULTS IPostC significantly reduced I/R-induced infarct size (40.2±2.1% versus 28.4±2.4%; P < 0.05); the release of cardiac Troponin T, creatine kinase-MB, and lactate dehydrogenase (191.5±3.1 versus 140.6±3.3 pg/ml, 3394.6±132.7 versus 2880.7±125.5 pg/ml, 2686.2±98.6 versus 1848.8±90.1 pg/ml, respectively; P < 0.05); and cardiomyocyte apoptosis (40.3±2.2% versus 27.0±1.6%; P < 0.05). Further, local and circulating IL-1β, IL-6, TNF-α, and ICAM-1 levels decreased; TLR4 expression and nuclear factor-KB (NF-κB) signaling decreased; and cardiac HSP90 expression increased. Blocking HSP90 function with GA inhibited IPostC protection and anti-inflammation, suggesting that IPostC has a HSP90-dependent anti-inflammatory effect. CONCLUSION HSP90 may play a role in IPostC-mediated cardioprotection by inhibiting TLR4 activation, local and systemic inflammation, and NF-kB signaling.
Collapse
Affiliation(s)
- Xin-Yue Zhang
- Department of Cardiology, First Affiliated Hospital, Guang Xi Medical University, Nanning, China
| | - Zheng Huang
- Department of Cardiology, First Affiliated Hospital, Guang Xi Medical University, Nanning, China
| | - Qing-Jie Li
- Department of Cardiology, Second Affiliated Hospital, Guang Xi Medical University, Nanning, China
| | - Guo-Qiang Zhong
- Department of Cardiology, First Affiliated Hospital, Guang Xi Medical University, Nanning, China.,Guang Xi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Disease Control and Prevention, Nanning, China.,Guang Xi Clinical Research Center for Cardio-cerebrovascular Diseases, Nanning, China
| | - Jian-Jun Meng
- Geriatric Healthcare Center, First Affiliated Hospital, Guang Xi Medical University, Nanning, China
| | - Dong-Xiao Wang
- Department of Cardiology, First Affiliated Hospital, Guang Xi Medical University, Nanning, China
| | - Rong-Hui Tu
- Guang Xi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Disease Control and Prevention, Nanning, China.,Guang Xi Clinical Research Center for Cardio-cerebrovascular Diseases, Nanning, China.,Department of Geriatric Cardiology, First Affiliated Hospital, Guang Xi Medical University, Nanning, China
| |
Collapse
|
22
|
Zaghmi A, Drouin-Ouellet J, Brambilla D, Gauthier MA. Treating brain diseases using systemic parenterally-administered protein therapeutics: Dysfunction of the brain barriers and potential strategies. Biomaterials 2020; 269:120461. [PMID: 33218788 DOI: 10.1016/j.biomaterials.2020.120461] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/23/2020] [Accepted: 10/18/2020] [Indexed: 12/12/2022]
Abstract
The parenteral administration of protein therapeutics is increasingly gaining importance for the treatment of human diseases. However, the presence of practically impermeable blood-brain barriers greatly restricts access of such pharmaceutics to the brain. Treating brain disorders with proteins thus remains a great challenge, and the slow clinical translation of these therapeutics may be largely ascribed to the lack of appropriate brain delivery system. Exploring new approaches to deliver proteins to the brain by circumventing physiological barriers is thus of great interest. Moreover, parallel advances in the molecular neurosciences are important for better characterizing blood-brain interfaces, particularly under different pathological conditions (e.g., stroke, multiple sclerosis, Parkinson's disease, and Alzheimer's disease). This review presents the current state of knowledge of the structure and the function of the main physiological barriers of the brain, the mechanisms of transport across these interfaces, as well as alterations to these concomitant with brain disorders. Further, the different strategies to promote protein delivery into the brain are presented, including the use of molecular Trojan horses, the formulation of nanosystems conjugated/loaded with proteins, protein-engineering technologies, the conjugation of proteins to polymers, and the modulation of intercellular junctions. Additionally, therapeutic approaches for brain diseases that do not involve targeting to the brain are presented (i.e., sink and scavenging mechanisms).
Collapse
Affiliation(s)
- A Zaghmi
- Institut National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, QC, J3X 1S2, Canada
| | - J Drouin-Ouellet
- Faculty of Pharmacy, Université de Montréal, CP 6128, succ. Centre-ville, Montréal, QC, H3C 3J7, Canada
| | - D Brambilla
- Faculty of Pharmacy, Université de Montréal, CP 6128, succ. Centre-ville, Montréal, QC, H3C 3J7, Canada
| | - M A Gauthier
- Institut National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, QC, J3X 1S2, Canada.
| |
Collapse
|
23
|
Liu C, Kang LN, Chen F, Mu D, Shen S, Wang K, Hu JX, Xie J, Xu B. Immediate Intracoronary Delivery of Human Umbilical Cord Mesenchymal Stem Cells Reduces Myocardial Injury by Regulating the Inflammatory Process Through Cell-Cell Contact with T Lymphocytes. Stem Cells Dev 2020; 29:1331-1345. [PMID: 32762286 DOI: 10.1089/scd.2019.0264] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Inflammatory response regulation is a mechanism through which human umbilical cord mesenchymal stem cells (HUCMSCs) improve myocardial ischemia reperfusion injury (IRI); however, the timing of HUCMSC delivery to achieve maximum effectiveness is controversial. To investigate the effects of HUCMSC delivery on the acute inflammatory stage of IRI, we transplanted HUCMSCs or HUCMSCs with cyclosporin A (CsA) through the coronary artery simultaneously during ischemia reperfusion in pigs. Ferumoxytol-labeled HUCMSCs (HUCMSC), HUCMSCs with cyclosporin A (HUCMSC+CsA), and PBS (control) groups were investigated to evaluate the homing of transplanted cells and changes in infarct features, cardiac activity, and inflammatory response at three time points post-transplantation. Animals were sacrificed 2 weeks later for histological analysis of the hearts. We detected Prussian blue-dyed granules distributed around T lymphocyte clusters in the infarct area in the HUCMSC group. Infarct size and collagen deposition in the infarct area were lower in the HUCMSC group than in the control and HUCMSC+CsA groups. Cardiac function was mildly impaired in both the control and HUCMSC groups, whereas added CsA had a more severe impact. The levels of proinflammatory markers were lower in the HUCMSC group than in the control group at 24-h follow-up, and the difference was more significant after adding CsA. There were more CD3+ T lymphocytes and Foxp3+ Tregs in the HUCMSC group infarct area than in the other two groups. Proliferation rate of T lymphocytes was higher in the HUCMSC group than in the other two groups. Indirect co-culture experiments in vitro showed that MSCs promoted the generation of CD4+CD25+ Foxp3+Tregs through a paracrine mechanism. These results indicate that immediate intracoronary delivery of HUCMSCs after ischemia reperfusion can reduce acute myocardial IRI and promote myocardial repair, mainly through T lymphocyte interactions to regulate the intense inflammatory response during the acute inflammatory stage.
Collapse
Affiliation(s)
- Chen Liu
- Department of Cardiology, Affiliated Hospital of Yangzhou University, Yangzhou City, People's Republic of China
| | - Li-Na Kang
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School, Nanjing City, People's Republic of China
| | - Fu Chen
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School, Nanjing City, People's Republic of China
| | - Dan Mu
- Department of Radiology, Drum Tower Hospital, Nanjing University Medical School, Nanjing City, People's Republic of China
| | - Song Shen
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School, Nanjing City, People's Republic of China
| | - Kun Wang
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School, Nanjing City, People's Republic of China
| | - Jia-Xin Hu
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School, Nanjing City, People's Republic of China
| | - Jun Xie
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School, Nanjing City, People's Republic of China
| | - Biao Xu
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School, Nanjing City, People's Republic of China
| |
Collapse
|
24
|
Hu Y, Liu X, Zhang T, Chen C, Dong X, Can Y, Liu P. Behavioral and Biochemical Effects of KXS on Postmyocardial Infarction Depression. Front Pharmacol 2020; 11:561817. [PMID: 32973539 PMCID: PMC7481476 DOI: 10.3389/fphar.2020.561817] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 08/13/2020] [Indexed: 01/01/2023] Open
Abstract
Background Depression and coronary heart disease (CHD) often occur together in clinical practice. As a traditional Chinese medicine, Kai-Xin-San (KXS) has been widely used for the treatment of emotion-related disorders. In the present study, we aimed to explore whether KXS had both antidepressive effects and cardioprotective functions in a rat model of myocardial ischemia (MI) with depression. Methods A total of 50 SD rats were randomly assigned into five groups as follows: normal control (control group), celiac injection of isopropyl adrenaline (ISO) (MI group), depression (depression group), MI+ depression (model group) and MI+ depression treated with intragastric administration of 370 mg/kg KXS (KXS group). MI was induced by subcutaneous injection of 85 mg/kg ISO. Depression was developed by a 7-week chronic mild stress (CMS) challenge. Behavioral test was conducted before and during the experiment. Echocardiography and biochemical analysis were carried out after 7 weeks of CMS challenge. Results After 7 weeks of experiment, depression-like behaviors were observed in all the groups except for control and KXS groups, and KXS treatment dramatically increased open-field test scores and sucrose consumption (P < 0.01 vs. model group). Echocardiography and biochemical analysis showed that KXS treatment could improve levels of ejection fraction (EF) and fractional shortening (FS), which were reduced by depression and ISO challenge. Meanwhile, KXS treatment significantly decreased the levels of creation kinase MB (CK-MB) and lactate dehydrogenase (LDH), which were increased in the model group. The activities of superoxide dismutase (SOD), glutathione peroxidase (GSH-PX), catalase (CAT) were increased, while the malondialdehyde (MDA) activity was significantly decreased in the KXS group. Moreover, KXS treatment reduced the levels of C-reactive protein (CRP), interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) in myocardial tissue compared with the model group. Conclusions KXS had antidepressant-like activity and offered cardioprotective effects against ISO-induced myocardial infarction with depression.
Collapse
Affiliation(s)
- Yuan Hu
- Medical Supplier Center, Department of Pharmacy, PLA General Hospital, Beijing, China
| | - Xu Liu
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Tianyi Zhang
- Medical Supplier Center, Department of Pharmacy, PLA General Hospital, Beijing, China
| | - Chao Chen
- Medical Supplier Center, Department of Pharmacy, PLA General Hospital, Beijing, China
| | - Xianzhe Dong
- Medical Supplier Center, Department of Pharmacy, PLA General Hospital, Beijing, China
| | - Yan Can
- Department of Basic Theory of Chinese Medicine, School of Pre-clinical Medicine, Guangzhou University of Chinese Medicine, Higher Education Mega Center, Guangzhou, China.,The Research Centre of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Higher Education Mega Center, Guangzhou, China
| | - Ping Liu
- Medical Supplier Center, Department of Pharmacy, PLA General Hospital, Beijing, China
| |
Collapse
|
25
|
Zhou P, Hua F, Wang X, Huang JL. Therapeutic potential of IKK-β inhibitors from natural phenolics for inflammation in cardiovascular diseases. Inflammopharmacology 2020; 28:19-37. [PMID: 31894515 DOI: 10.1007/s10787-019-00680-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 12/16/2019] [Indexed: 12/19/2022]
Abstract
Cardiovascular disease (CVDs) is a chronic disease with the highest morbidity and mortality in the world. Previous studies have suggested that preventing inflammation serves an efficient role in protection against cardiovascular diseases. Modulation of IKK-β activity can be used to treat and control CVDs associated with chronic inflammation, which targets the phosphorylation of IκB following the release of the RelA complex, and then translocates to the nucleus, eventually triggering the transcription of several genes that induce chemokines, cytokines, and adhesion molecules. Most importantly, the IκB kinase (IKK) complex is involved in transcriptional activation by phosphorylating the inhibitory molecule IkBα, enabling activation of NF-κB. Phenolic compounds possess cardioprotective potential that may be related to modulating inflammatory responses involved in CVDs. The SystemsDock analysis was used to explore whether 38 active compounds inhibit IKK-β activity based on literature. Docking results showed that the top docking score of three chemical compounds were icariin, salvianolic acid B, and plantainoside D in all compounds. Icariin, salvianolic acid B, and plantainoside D are the most promising IKKβ inhibitors. These phytochemicals could be helpful to find the lead compounds on designing and developing novel cardioprotective agents.
Collapse
Affiliation(s)
- Peng Zhou
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, People's Republic of China. .,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, People's Republic of China. .,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, 230012, People's Republic of China.
| | - Fang Hua
- Pharmacy School, Anhui Xinhua University, Hefei, 230088, People's Republic of China.,Natural Products Laboratory, International Joint Lab of Tea Chemistry and Health Effects, State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei, 230036, People's Republic of China
| | - Xiang Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, People's Republic of China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, People's Republic of China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, 230012, People's Republic of China
| | - Jin-Ling Huang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, People's Republic of China. .,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, People's Republic of China. .,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, 230012, People's Republic of China.
| |
Collapse
|
26
|
Chen Q, Liu Y, Ding X, Li Q, Qiu F, Wang M, Shen Z, Zheng H, Fu G. Bone marrow mesenchymal stem cell-secreted exosomes carrying microRNA-125b protect against myocardial ischemia reperfusion injury via targeting SIRT7. Mol Cell Biochem 2019; 465:103-114. [PMID: 31858380 PMCID: PMC6955239 DOI: 10.1007/s11010-019-03671-z] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 11/30/2019] [Indexed: 12/15/2022]
Abstract
MicroRNA-125b (miR-125b) reduces myocardial infarct area and restrains myocardial ischemia reperfusion injury (I/R). In this study, we aimed to investigate the effect of bone marrow mesenchymal stem cell (BMSC)-derived exosomes carrying miR-125b on I/R rats. The myocardial I/R model in rats was constructed by ligation of the left anterior descending coronary artery (LAD). Rats were randomly divided into I/R and Sham group. Lv-cel-miR-67 (control) or Lv-miR-125b was transfected into BMSCs. Exosomes were extracted from transfected BMSCs, and separately named BMSC-Exo-67, BMSC-Exo-125b, and BMSC-Exo. MTT assay and flow cytometry were used to detect the viability and apoptosis of I/R myocardium cells, respectively. The expression of cell apoptosis proteins and the levels of inflammatory factors were examined by Western blot and ELISA assay, respectively. The target relationship between miR-125b and SIRT7 was predicted by using StarBase3.0, and was confirmed by using dual-luciferase reporter gene assay. qRT-PCR, immunohistochemistry staining, and Western blot were used to evaluate the expression of SIRT7 in myocardium tissues in I/R rats. BMSC-derived exosomes were successfully isolated and identified by TEM and positive expression of CD9 and CD63. The expression of miR-125b was down-regulated in I/R myocardium tissues and cells. BMSC-Exo-125b significantly up-regulated miR-125b in I/R myocardium cells. The intervention of BMSC-Exo-125b significantly increased the cell viability, decreased the apoptotic ratio, down-regulated Bax and caspase-3, up-regulated Bcl-2, and decreased the levels of IL-1β, IL-6, and TNF-α in I/R myocardium cells. SIRT7 was a target of miR-125b, and BMSC-Exo-125b significantly down-regulated SIRT7 in myocardium cells. In addition, the injection of BMSC-Exo-125b alleviated the pathological damages and down-regulated SIRT7 in myocardium tissues of I/R rats. BMSC-derived exosomes carrying miR-125b protected against myocardial I/R by targeting SIRT7.
Collapse
Affiliation(s)
- Qi Chen
- Department of Cardiology, School of Medicine, Sir Run Run Shaw Hospital, Biomedical Research Center, Zhejiang University, No. 3, East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Yu Liu
- Department of Cardiology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, No. 321, Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Xueyan Ding
- Department of Cardiology, Zhejiang Provincial People's Hospital, No. 158, Shangtang Road, Hangzhou, 310014, Zhejiang, China
| | - Qinfeng Li
- Department of Cardiology, School of Medicine, Sir Run Run Shaw Hospital, Biomedical Research Center, Zhejiang University, No. 3, East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Fuyu Qiu
- Department of Cardiology, School of Medicine, Sir Run Run Shaw Hospital, Biomedical Research Center, Zhejiang University, No. 3, East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Meihui Wang
- Department of Cardiology, School of Medicine, Sir Run Run Shaw Hospital, Biomedical Research Center, Zhejiang University, No. 3, East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Zhida Shen
- Department of Cardiology, School of Medicine, Sir Run Run Shaw Hospital, Biomedical Research Center, Zhejiang University, No. 3, East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Hao Zheng
- Department of Cardiology, Zhejiang Provincial People's Hospital, No. 158, Shangtang Road, Hangzhou, 310014, Zhejiang, China.
| | - Guosheng Fu
- Department of Cardiology, School of Medicine, Sir Run Run Shaw Hospital, Biomedical Research Center, Zhejiang University, No. 3, East Qingchun Road, Hangzhou, 310016, Zhejiang, China.
| |
Collapse
|
27
|
Peng N, Jin L, He A, Deng C, Wang X. Effect of sulphoraphane on newborn mouse cardiomyocytes undergoing ischaemia/reperfusion injury. PHARMACEUTICAL BIOLOGY 2019; 57:753-759. [PMID: 31686558 PMCID: PMC6844446 DOI: 10.1080/13880209.2019.1680705] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 07/23/2019] [Accepted: 10/11/2019] [Indexed: 05/30/2023]
Abstract
Context: Sulphoraphane (SFN) is an isothiocyanate, having antioxidant activity, antitumor, and therapeutic effects on cardiovascular disease.Objective: This study explores the mechanisms of SFN preconditioning on ischaemia/reperfusion injury (IRI).Materials and methods: Cardiomyocytes were divided into four groups as follows: control group (normoxic condition), SFN group (5 μmol/L), hypoxia/reoxygenation (H/R) group (1 h, 3 h) and SFN + H/R group. Cell viability was determined by MTT method. Levels of creatine kinase (CK), nitric oxide (NO), superoxide dismutase (SOD) and maleic dialdehyde (MDA) were determined by colorimetric method. Cell apoptosis, levels of reactive oxygen species (ROS) and mitochondrial membrane potential (MMP) were determined by flow cytometry. Levels of Bax, Bcl-2, C caspase-3, NF-E2-related factor 2 (Nrf2) and haem oxygenase-1 (HO-1) were detected by Western blot.Results: H/R model inhibited cell viability, increased the levels of LDH, CK, Bax and C caspase-3, and decreased the levels of NO, Bcl-2, while the effect of H/R was partially reversed by SFN. SFN treatment reduced ROS, MDA (from 4.9 nM to 2.8 nM) production, elevated SOD level (from 39.5 U/mL to 61.7 U/mL) and improved MMP damage. Under the effect of SFN, up-regulation of nuclear Nrf2 expression and down-regulation of cytosolic Nrf2 expression were observed, which led to Nrf2 nuclear translocation and enhanced the expression of HO-1.Conclusion: These results suggested that SFN had a protective effect on cardiomyocytes undergoing IRI, and its mechanism may be realized via activating the Nrf2/HO-1 pathway, thereby inhibiting apoptosis. This might provide a new approach for the treatment of ischaemic heart disease.
Collapse
Affiliation(s)
- Na Peng
- Department of Cardiology, Jingmen No. 1 People’s Hospital, Jingmen, China
| | - Luping Jin
- Department of Cardiology, Jingmen No. 1 People’s Hospital, Jingmen, China
| | - Aizhen He
- Department of Cardiology, Jingmen No. 1 People’s Hospital, Jingmen, China
| | - Changjin Deng
- Department of Cardiology, Jingmen No. 1 People’s Hospital, Jingmen, China
| | - Xiaoqin Wang
- Department of Cardiology, Jingmen No. 1 People’s Hospital, Jingmen, China
| |
Collapse
|
28
|
Jiang X, Yu M, Zhu T, Lou L, Chen X, Li Q, Wei D, Sun R. Kcnq1ot1/miR-381-3p/ETS2 Axis Regulates Inflammation in Mouse Models of Acute Respiratory Distress Syndrome. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 19:179-189. [PMID: 31841990 PMCID: PMC6920288 DOI: 10.1016/j.omtn.2019.10.036] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 10/26/2019] [Accepted: 10/28/2019] [Indexed: 12/29/2022]
Abstract
Inflammatory mediators play a key role in the pathogenesis of acute respiratory distress syndrome (ARDS). In this study, we aimed to explore the involvement of the Kcnq1 opposite strand/antisense transcript 1 (Kcnq1ot1)/miR-381-3p/E26 transformation-specific proto-oncogene 2 (ETS2) axis in inflammation of lipopolysaccharide (LPS)-induced ARDS. Microarray analysis revealed ETS2 as an upregulated gene in ARDS. Then, a LPS-induced ARDS mouse model was constructed, with a series of gain- or loss-of-function experiments conducted to evaluate the lung function and neutrophil extracellular trap (NET) formation in lung tissue and determine the neutrophil number, myeloperoxidase (MPO) activity, and inflammatory factor levels in bronchoalveolar lavage fluid (BALF). As the results revealed, downregulated expression of ETS2 resulted in improved lung function, decreased NETs, MPO activity, and levels of interleukin (IL)-6 and tumor necrosis factor alpha (TNF-α), as well as increased IL-10 level. Then, the assays of dual-luciferase reporter, RNA-binding protein immunoprecipitation (RIP), and RNA pull-down were performed to validate that Kcnq1ot1 promoted ETS2 expression by competitively binding to miR-381-3p. Meanwhile, it was also found that Kcnq1ot1 silencing reversed the promotive effect of EST2 on ARDS. Our results provide evidence that Kcnq1ot1 silencing may reduce the inflammatory response in LPS-induced ARDS via inhibition of miR-381-30-dependent ETS2, thereby presenting new molecular understanding for the development of ARDS.
Collapse
Affiliation(s)
- Xiaohui Jiang
- Department of Critical Care Medicine, Chun'an First People's Hospital (Zhejiang Provincial People's Hospital, Chun'an Branch), Hangzhou 311700, P.R. China; Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, P.R. China.
| | - Meihong Yu
- Department of Critical Care Medicine, Chun'an First People's Hospital (Zhejiang Provincial People's Hospital, Chun'an Branch), Hangzhou 311700, P.R. China
| | - Taiping Zhu
- Department of Critical Care Medicine, Chun'an First People's Hospital (Zhejiang Provincial People's Hospital, Chun'an Branch), Hangzhou 311700, P.R. China
| | - Lulu Lou
- Internal Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, P.R. China
| | - Xu Chen
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, P.R. China
| | - Qian Li
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, P.R. China
| | - Danhong Wei
- Department of Neuroscience Care Unit, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, P.R. China
| | - Renhua Sun
- Department of Critical Care Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, P.R. China
| |
Collapse
|
29
|
Mousa AA, El-Gansh HAI, Eldaim MAA, Mohamed MAEG, Morsi AH, El Sabagh HS. Protective effect of Moringa oleifera leaves ethanolic extract against thioacetamide-induced hepatotoxicity in rats via modulation of cellular antioxidant, apoptotic and inflammatory markers. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:32488-32504. [PMID: 31617137 DOI: 10.1007/s11356-019-06368-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 08/29/2019] [Indexed: 05/16/2023]
Abstract
The current study was conducted to evaluate the ameliorative and protective potentials of Moringea oleifera leaves ethanolic extract (MOLE) against thioacetamide (TAA) toxicity. A total of 58 male albino rats were randomly assigned into six experimental groups. G1, rats received distilled water. G2, rats were injected with a single dose of TAA (200 mg/kg BW) i.p. G3, rats were given MOLE (300 mg/kg BW) orally for 26 days. G4, rats were injected TAA as in G2 and treated with MOLE as G3. G5, rats were kept for 26 days without treatment then on day 27 injected with TAA as in G2. G6, rats were given MOLE for 26 days then on day 27 injected with TAA. Phytochemical analysis of MOLE indicated the presence of kaempferol, kaempferol malonylglucoside, kaempferol hexoside, kaempferol -3-O-glucoside, kaempferol-3-O-acetyl-glucoside, cyanidin -3-O-hexoside, ellagic acid, quercetin, quercetin-3-O-glucoside, and apigenin glucoside. Intoxication of rats with TAA significantly elevated activities of serum AST, ALT, and ALP; concentrations of malondialdehyde, nitric oxide, and hepatic tissue protein expression of caspase 3 and COX2 with alteration of the histological structures of hepatic tissues, while it decreased serum levels of total protein, albumin, and hepatic tissue contents of reduced glutathione. Also, TAA intoxication resulted in 62.5% mortality in rats of G5. Treatment of TAA intoxicated rats (G4) with MOLE ameliorated the toxic effects of TAA on hepatic tissue structure and function. It decreased serum activities of AST, ALT, and ALP; enhanced hepatic GSH concentration; reduced pathological alterations and lipid peroxidation; and downregulated caspase 3 and COX2 proteins expression in hepatic tissue. In addition, MOLE protected rats of G6 from TAA-induced hepatic tissues injury and dysfunction, and increased survival rate of rats. In conclusion, MOLE had both ameliorating and protecting potentials against TAA-induced rats liver damage through regulation of antioxidant, anti-apoptotic, and inflammatory biomarkers. Graphical abstract.
Collapse
Affiliation(s)
- Ahmed Abdelmoniem Mousa
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Menoufia, 32897, Egypt
| | - Hala Ali Ibrahim El-Gansh
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Menoufia, 32897, Egypt
| | - Mabrouk Attia Abd Eldaim
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Menoufia University, Shebin Elkom, Menoufia, Egypt.
| | | | - Azza Hassan Morsi
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Hesham Saad El Sabagh
- Department of Toxicology and Forensic Medicine, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Menoufia, 32897, Egypt
| |
Collapse
|
30
|
Wang R, Wang M, Zhou J, Ye T, Xie X, Ni D, Ye J, Han Q, Di C, Guo L, Sun G, Sun X. Shuxuening injection protects against myocardial ischemia-reperfusion injury through reducing oxidative stress, inflammation and thrombosis. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:562. [PMID: 31807543 DOI: 10.21037/atm.2019.09.40] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background Shuxuening injection (SXNI) has a good effect on cardiovascular and cerebrovascular diseases. Here, our study aims to investigate whether SXNI have the protective effect on myocardial ischemia-reperfusion injury (MIRI) and elucidate the mechanism of SXNI's cardiac protection. Methods In this experiment, the coronary arteries of Sprague-Dawley (SD) rats were ligated for the induction of a MIRI model. TTC staining and haematoxylin-eosin (HE), as well as troponin I (TnI), lactate dehydrogenase (LDH), aspartate aminotransferase (AST), creatine kinase (CK) and CK-MB levels, were used to detect the protective effect of SXNI. In rat cardiac tissue, superoxide dismutase, catalase, glutathione and malondialdehyde (MDA) activities and glucose-regulated protein 78 (GRP78), calreticulin (CRT), CCAAT/enhancer binding protein homologous protein (CHOP) and caspase-12 expression levels were detected. In rat serum, the levels of inflammatory factors, including high-sensitivity C-reactive protein, monocyte chemoattractant protein-1, tumour necrosis factor-α, interleukin-6 (IL-6) and IL-1β, were measured by Elisa. In the rat arterial tissue, Toll-like receptor 4 (TLR4)/nuclear factor kappa B (NF-κB) expression was measured by western blot. In the rat plasma, ELISA was used to assay the levels of coagulation and plasmin system indicators, including platelet activating factor, endothelin, tissue factor (TF), plasminogen inhibitor, thromboxane B2, plasma fibrinogen. Results The results showed that SXNI can reduce the infarct size of myocardial tissue, decrease the myocardial enzyme and TnI levels and decrease the degree of myocardial damage compared with the model group. Additionally, SXNI can increase the activity of antioxidant enzymes, reduce the MDA level and decrease the GRP78, CRT, CHOP and caspase-12 expression levels. SXNI also decreased the levels of inflammatory cytokines in rat serum, lowered the level of procoagulant molecules in plasma and reduced the TLR4/NF-κB expression. Conclusions SXNI has protective effect on MIRI mainly by inhibiting oxidative stress and endoplasmic reticulum stress (ERS), thereby regulating TLR4/NF-κB pathway to reduce inflammation, and lowing procoagulant-related factors levels to reduce the risk of thrombosis.
Collapse
Affiliation(s)
- Ruiying Wang
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Beijing 100193, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China.,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.,Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Min Wang
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Beijing 100193, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China.,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.,Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Jiahui Zhou
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Beijing 100193, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China.,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.,Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Tianyuan Ye
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Beijing 100193, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China.,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.,Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Xueheng Xie
- Harbin University of Commerce, Harbin 150076, China
| | - Dong Ni
- Jilin Agricultural University, Changchun 130118, China
| | - Jingxue Ye
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Beijing 100193, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China.,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.,Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Qiaoling Han
- Shiyao Yinhu Pharmaceutical Co., Ltd., Yuncheng 044000, China
| | - Caixia Di
- Shiyao Yinhu Pharmaceutical Co., Ltd., Yuncheng 044000, China
| | - Liang Guo
- Shiyao Yinhu Pharmaceutical Co., Ltd., Yuncheng 044000, China
| | - Guibo Sun
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Beijing 100193, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China.,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.,Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Xiaobo Sun
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Beijing 100193, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China.,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.,Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
| |
Collapse
|
31
|
Kong T, Liu M, Ji B, Bai B, Cheng B, Wang C. Role of the Extracellular Signal-Regulated Kinase 1/2 Signaling Pathway in Ischemia-Reperfusion Injury. Front Physiol 2019; 10:1038. [PMID: 31474876 PMCID: PMC6702336 DOI: 10.3389/fphys.2019.01038] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 07/29/2019] [Indexed: 12/11/2022] Open
Abstract
Extracellular signal-regulated kinase 1/2 (ERK1/2), an important member of the mitogen-activated protein kinase family, is found in many organisms, and it participates in intracellular signal transduction. Various stimuli induce phosphorylation of ERK1/2 in vivo and in vitro. Phosphorylated ERK1/2 moves to the nucleus, activates many transcription factors, regulates gene expression, and controls various physiological processes, finally inducing repair processes or cell death. With the aging of the population around the world, the occurrence of ischemia-reperfusion injury (IRI), especially in the brain, heart, kidney, and other important organs, is becoming increasingly serious. Abnormal activation of the ERK1/2 signaling pathway is closely related to the development and the metabolic mechanisms of IRI. However, the effects of this signaling pathway and the underlying mechanism differ between various models of IRI. This review summarizes the ERK1/2 signaling pathway and the molecular mechanism underlying its role in models of IRI in the brain, heart, liver, kidneys, and other organs. This information will help to deepen the understanding of ERK1/2 signals and deepen the exploration of IRI treatment based on the ERK1/2 study.
Collapse
Affiliation(s)
- Tingting Kong
- Cheeloo College of Medicine, Shandong University, Jinan, China.,School of Mental Health, Neurobiology Key Laboratory of Jining Medical University in Colleges of Shandong, Jining Medical University, Jining, China
| | - Minghui Liu
- School of Mental Health, Neurobiology Key Laboratory of Jining Medical University in Colleges of Shandong, Jining Medical University, Jining, China
| | - Bingyuan Ji
- School of Mental Health, Neurobiology Key Laboratory of Jining Medical University in Colleges of Shandong, Jining Medical University, Jining, China
| | - Bo Bai
- School of Mental Health, Neurobiology Key Laboratory of Jining Medical University in Colleges of Shandong, Jining Medical University, Jining, China
| | - Baohua Cheng
- School of Mental Health, Neurobiology Key Laboratory of Jining Medical University in Colleges of Shandong, Jining Medical University, Jining, China
| | - Chunmei Wang
- School of Mental Health, Neurobiology Key Laboratory of Jining Medical University in Colleges of Shandong, Jining Medical University, Jining, China
| |
Collapse
|
32
|
Wang R, Wen X, Huang C, Liang Y, Mo Y, Xue L. Association between inflammation-based prognostic scores and in-hospital outcomes in elderly patients with acute myocardial infarction. Clin Interv Aging 2019; 14:1199-1206. [PMID: 31308643 PMCID: PMC6615021 DOI: 10.2147/cia.s214222] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 06/12/2019] [Indexed: 11/23/2022] Open
Abstract
Objective Emerging evidence suggests that systemic inflammation is a predictor of poor prognosis in acute myocardial infarction (AMI). In this study, we sought to assess whether inflammation-based prognostic scores are associated with in-hospital outcomes in elderly patients with AMI. Methods In this retrospective study, patients who were over 75-years-old and met the diagnostic criteria for AMI were consecutively recruited from January 1, 2016, to March 31, 2019. Logistic regression and receiver-operating characteristic (ROC) analyses were performed to evaluate the predictive value of the inflammation-based Glasgow Prognostic Score (GPS), Prognostic Index (PI) and Prognostic Nutritional Index (PNI). Results A total of 273 patients were enrolled. The incidence of major cardiovascular adverse events (MACEs) and mortality during hospitalization increased significantly with increasing GPS and PI scores. Multiple logistic regression showed that the GPS was independently associated with MACEs (score 1, RR: 6.711, 95% CI: 1.409–31.968; score 2, RR: 14.063, 95% CI: 3.018–65.535) and mortality (score 1, RR: 8.656, 95% CI: 1.068–70.126; score 2, RR: 10.549, 95% CI: 1.317–84.465). The PI was also independently predictive of MACEs (score 2, RR: 5.132, 95% CI: 1.451–18.148). No significant difference was observed in the PNI between patients with different in-hospital outcomes. When in-hospital MACEs were used as an endpoint, the area under the curve (AUC) of the GPS was 0.740 (95% CI 0.678–0.802), and the AUC of the PI was 0.703 (95% CI 0.634–0.773). When mortality was used as an endpoint, the AUC of the GPS was 0.677 (95% CI 0.602–0.753), and the AUC of the PI was 0.667 (95% CI 0.577–0.757). Conclusion The severity of systemic inflammation is a strong predictor of poor prognosis in elderly patients with AMI. Among these three inflammation-based prognostic scores, the GPS has a better predictive value than the PI and PNI for in-hospital MACEs and mortality.
Collapse
Affiliation(s)
- Rui Wang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Xiaodan Wen
- Department of Geriatrics, Guangdong Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Cheng Huang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Yingcong Liang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Yujing Mo
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Ling Xue
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| |
Collapse
|
33
|
Eken MK, Ersoy GS, Kaygusuz EI, Devranoğlu B, Takır M, Çilingir ÖT, Çevik Ö. Etanercept protects ovarian reserve against ischemia/reperfusion injury in a rat model. Arch Med Sci 2019; 15:1104-1112. [PMID: 31360205 PMCID: PMC6657239 DOI: 10.5114/aoms.2017.72406] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 03/29/2017] [Indexed: 01/07/2023] Open
Abstract
INTRODUCTION Etanercept has been widely used in autoimmune diseases for blocking tumor necrosis factor α (TNF-α), which is an inflammatory cytokine. The anti-apoptotic and anti-inflammatory effects of etanercept against ischemia/reperfusion (I/R) injury have been shown for several tissues in rat studies, but to the best of our knowledge, there are no reports on its protective effects following similar injury in ovarian tissue. The aim of this study was to investigate whether etanercept has beneficial effects on ovarian I/R injury, as well as on ovarian reserve. MATERIAL AND METHODS Twenty-four rats were randomly divided into four groups (n = 6/group): sham (laparotomy only); sham + etanercept; I/R; and I/R + etanercept. Ischemia was induced for 3 h by twisting the ovary, and 24 h after detorsion the ovarian tissues were collected to evaluate histopathologic changes, glutathione (GSH), malondialdehyde (MDA), myeloperoxidase (MPO), and superoxide dismutase (SOD) concentrations for oxidative stress, 8-hydroxy-2'-deoxyguanosine (8-OHdG) for DNA damage, caspase-3 activity for apoptosis and ovarian follicle counts. To measure anti-Mullerian hormone (AMH), serum samples were drawn before and after surgery. RESULTS Tissue GSH and SOD levels were significantly higher, while MDA and MPO levels were significantly lower in the I/R + etanercept group than in the I/R group (p < 0.05, p < 0.01, respectively). Tissue 8-OHdG and caspase-3 activity were significantly lower in the I/R+etanercept group than in the I/R group (p < 0.05, p < 0.01, respectively). Preoperative and postoperative AMH levels were compared and there was a significant reduction in the I/R and I/R + etanercept groups (p < 0.001, p < 0.001). The reduction of AMH in the I/R + etanercept group was significantly lower than in the I/R group. The primordial, preantral and small antral follicle numbers were also significantly higher in the I/R + etanercept group compared to the I/R group (p < 0.001, p < 0.001, p < 0.005, respectively). CONCLUSIONS Etanercept attenuated inflammation and related oxidative stress and also helped to preserve ovarian reserve following ovarian I/R damage.
Collapse
Affiliation(s)
- Meryem Kurek Eken
- Obstetrics and Gynecology Department, Medical Faculty, Adnan Menderes University, Aydın, Turkey
| | - Gulcin Sahin Ersoy
- Obstetric and Gynecology Department, Kartal Dr. Lütfi Kırdar Training and Research Hospital, Istanbul, Turkey
| | - Ecmel Işık Kaygusuz
- Pathology Department, Zeynep Kamil Training and Research Hospital, Istanbul, Turkey
| | - Belgin Devranoğlu
- Infertility Department, Zeynep Kamil Training and Research Hospital, Istanbul, Turkey
| | - Mümtaz Takır
- Internal Medicine Department, Division of Endocrinology and Metabolism, Istanbul Medeniyet University, Istanbul, Turkey
| | - Özlem Tuğçe Çilingir
- Department of Histology and Embryology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Özge Çevik
- Department of Biochemistry, Medical Faculty, Adnan Menderes University, Aydın, Turkey
| |
Collapse
|
34
|
Zhang J, Xu H, Gong L, Liu L. Retracted
: MicroRNA‐132 protects H9c2 cells against oxygen and glucose deprivation‐evoked injury by targeting FOXO3A. J Cell Physiol 2019; 235:176-184. [DOI: 10.1002/jcp.28956] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 05/20/2019] [Accepted: 05/22/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Jingze Zhang
- Department of Neurosurgery The Second Hospital of Jilin University Changchun Jilin China
| | - Haiming Xu
- Department of Cardiology China‐Japan Union Hospital of Jilin University Changchun Jilin China
| | - Licheng Gong
- Department of Cardiology China‐Japan Union Hospital of Jilin University Changchun Jilin China
| | - Long Liu
- Department of Cardiology China‐Japan Union Hospital of Jilin University Changchun Jilin China
| |
Collapse
|
35
|
Abstract
The advent of biologic therapy has enhanced our ability to augment disease in an increasingly targeted manner. The use of biologics in cardiovascular disease (CVD) has steadily increased over the past several decades. Much of the early data on biologics and CVD were derived from their use in rheumatologic populations. Atherosclerosis, myocardial infarction, and heart failure have been closely linked to the inflammatory response. Accordingly, cytokines such as tumor necrosis factor (TNF)-alpha and interleukin (IL)-1 have been targeted. Noninflammatory mediators, such as proprotein convertase subtilisin kexin type 9 (PCSK9), have been selected for therapeutic intervention as well. Furthermore, RNA interference (RNAi) therapy has emerged and may serve as another targeted therapeutic mechanism. Herein, we will review the history, obstacles, and advances in using biologic therapy for CVD.
Collapse
|
36
|
Chen XJ, Ren SM, Dong JZ, Qiu CG, Chen YW, Tao HL. Ginkgo biloba extract-761 protects myocardium by regulating Akt/Nrf2 signal pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:647-655. [PMID: 30858695 PMCID: PMC6387611 DOI: 10.2147/dddt.s191537] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Objective The aim of this study was to investigate the protective effect and mechanism of Ginkgo biloba extract-761 (EGb 761) in the rat with myocardial ischemia–reperfusion injury (MIRI). Materials and methods Forty Sprague Dawley rats were randomly divided into following four groups: sham group, I/R group and EGb 761 groups (20 and 40 mg/kg). MIRI model was established after 14 days of administration. The myocardial infarct size and myocardial histology were measured and compared. Meanwhile, the levels of creatine kinase-MB (CK-MB), lactate dehydrogenase (LDH), troponin T (TnT), TNF-α, IL-6, IL-1β, superoxide dismutase (SOD), malondialdehyde (MDA) and glutathione peroxidase (GSH-Px) were evaluated. Western blot was used to detect the expression of Caspase-3, Bax, Bcl-2, HO-1, Nrf2, Akt, p-Akt and nuclear protein Nrf2. Results The levels of infarct size, CK-MB, LDH, TnT, TNF-α, IL-6 and IL-1β in the EGb 761 groups were significantly lower than those in the ischemia/reperfusion (I/R) group. The content of MDA was lower in the myocardium, whereas the activities of SOD and GSH-Px were higher than those in the I/R group. The expressions of Caspase-3 and Bax in the EGb 761 groups were significantly lower than those in the I/R group, whereas the expressions of Bcl-2, p-Akt and HO-1 and nuclear protein Nrf2 in the EGb 761 groups were higher than those in the I/R group. Conclusion EGb 761 might inhibit the apoptosis of myocardial cells and protect the myocardium by activating the Akt/Nrf2 pathway, increasing the expression of HO-1, decreasing oxidative stress and repressing inflammatory reaction.
Collapse
Affiliation(s)
- Xiao-Jie Chen
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China,
| | - Shu-Min Ren
- Department of Genetics and Prenatal Diagnosis, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jian-Zeng Dong
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China,
| | - Chun-Guang Qiu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China,
| | - Ying-Wei Chen
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China,
| | - Hai-Long Tao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China,
| |
Collapse
|
37
|
Li L, Liu T, Li X, Liu X, Liu L, Li S, Li Z, Zhou Y, Liu F. Protein chip and bioinformatic analyses of differentially expressed proteins involved in the effect of hydrogen-rich water on myocardial ischemia-reperfusion injury. Int J Med Sci 2019; 16:1254-1259. [PMID: 31588191 PMCID: PMC6775260 DOI: 10.7150/ijms.35984] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 06/30/2019] [Indexed: 12/17/2022] Open
Abstract
Background: The differentially expressed proteins (DEPs) involved in the effect of hydrogen-rich water on myocardial ischemia reperfusion injury (MIRI) and their biological processes and signaling pathway were analyzed. Methods: 20 Wistar rats were randomly and equally divided into a control and a hydrogen-rich group. Hearts were removed and fixed in a Langendorff device. The control group was perfused with K-R solution, and the hydrogen-rich water group was perfused with K-R solution + hydrogen-rich water. Protein was extracted from the ventricular tissues, and GSR-CAA-67 was used to identify the DEPs between two groups. DEPs were analyzed through bioinformatic methods. Results: Compared with the control group, in the treatment group, the expression of 25 proteins was obviously decreased (P<0.05). For the DEPs, 359 biological processes, including the regulation of signaling pathways, immune reaction and formation of cardiovascular endothelial cells, were selected by GO enrichment analysis. Five signaling pathways were selected by KEGG pathway enrichment analysis. Conclusions: 25 proteins that are involved in hydrogen-water reducing MIRI were selected by high-throughput GSR-CAA-67. The biological processes and metabolic pathways involved in the DEPs were summarized, providing theoretical evidence for the clinical application of hydrogen-rich water.
Collapse
Affiliation(s)
- Liangtong Li
- School of Medicine, Hebei University, Baoding, 071000, China.,Central Laboratory of Affiliated Hospital of Hebei University, Baoding 071000, China
| | - Tongtong Liu
- Affiliated Hospital of Hebei University, Baoding, 071000, China
| | - Xiangzi Li
- School of Medicine, Hebei University, Baoding, 071000, China
| | - Xuanchen Liu
- School of Medicine, Hebei University, Baoding, 071000, China
| | - Li Liu
- School of Medicine, Hebei University, Baoding, 071000, China
| | - Shaochun Li
- School of Medicine, Hebei University, Baoding, 071000, China
| | - Zhilin Li
- School of Chemistry, Hebei University, Baoding 071000, China
| | - Yujuan Zhou
- School of Medicine, Hebei University, Baoding, 071000, China
| | - Fulin Liu
- Affiliated Hospital of Hebei University, Baoding, 071000, China
| |
Collapse
|
38
|
Jia Y, Li D, Cao Y, Cheng Y, Xiao L, Gao Y, Zhang L, Zeng Z, Wan Z, Zeng R. Inflammation-based Glasgow Prognostic Score in patients with acute ST-segment elevation myocardial infarction: A prospective cohort study. Medicine (Baltimore) 2018; 97:e13615. [PMID: 30558040 PMCID: PMC6319978 DOI: 10.1097/md.0000000000013615] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 11/16/2018] [Indexed: 02/05/2023] Open
Abstract
The inflammation-based Glasgow Prognostic Score (GPS), which involves C-reactive protein and serum albumin levels, has been reported to be a strong independent predictor of mortality in many cancers. This study aimed to investigate whether the GPS is associated with mortality in patients with acute ST-segment elevation myocardial infarction (STEMI) undergoing primary percutaneous coronary intervention (pPCI).In this study, 406 consecutive patients with STEMI at our emergency department (ED) who were undergoing pPCI were prospectively enrolled and assigned a GPS of 0, 1, or 2. Kaplan-Meier survival and multivariable Cox regression analyses were used to evaluate the associations between the GPS and long-term mortality.Twenty-three patients (5.7%) died at the hospital, and 37 (9.7%) died during follow-up (14.4 [9.3-17.6] months). Compared with patients with a lower GPS, those with a higher GPS had significantly higher in-hospital mortality (GPS = 0 vs GPS = 1 vs GPS = 2: 3.3% vs 6.3% vs 28.0%, P < .001), follow-up mortality (4.6% vs 14.3% vs 55.6%, P < .001), and cumulative mortality (9.6% vs 21.1% vs 71.1%, P < .001). Multivariable Cox regression analysis revealed that in patients with a GPS of 1 and 2 (versus 0), the multivariable adjusted hazard ratios (HR) for all-cause mortality were 2.068 (95% CI: 1.082-3.951, P = .028) and 8.305 (95% CI: 4.017-17.171, P < .001), respectively, after controlling for all of the confounding factors. Subgroup analysis showed that a higher GPS was associated with an increased risk of cumulative mortality in the different subgroups.The GPS on admission may be useful for stratifying the risk of adverse outcomes in patients with STEMI undergoing pPCI in the ED.
Collapse
Affiliation(s)
- Yu Jia
- Department of Emergency Medicine, West China Hospital
- Disaster Medicine Center
- Laboratory of Emergency Medicine
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| | - Dongze Li
- Department of Emergency Medicine, West China Hospital
- Disaster Medicine Center
- Laboratory of Emergency Medicine
| | - Yu Cao
- Department of Emergency Medicine, West China Hospital
- Disaster Medicine Center
- Laboratory of Emergency Medicine
| | - Yisong Cheng
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| | - Lei Xiao
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| | - Yongli Gao
- Department of Emergency Medicine, West China Hospital
- Disaster Medicine Center
- Laboratory of Emergency Medicine
| | - Lin Zhang
- Department of Anaesthesia and Intensive Care, Chinese University of Hong Kong, Hong Kong, China
| | - Zhi Zeng
- Department of Emergency Medicine, West China Hospital
| | - Zhi Wan
- Department of Emergency Medicine, West China Hospital
- Disaster Medicine Center
- Laboratory of Emergency Medicine
| | - Rui Zeng
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
39
|
6-Gingerol Protects Heart by Suppressing Myocardial Ischemia/Reperfusion Induced Inflammation via the PI3K/Akt-Dependent Mechanism in Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:6209679. [PMID: 30519268 PMCID: PMC6241357 DOI: 10.1155/2018/6209679] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 09/19/2018] [Accepted: 10/25/2018] [Indexed: 01/04/2023]
Abstract
Our previous study has demonstrated that 6-Gingerol (6-G) could alleviate myocardial ischemia/reperfusion injury (MIRI). However, the molecular mechanism underlying the process of myocardial ischemia/reperfusion (I/R) injury alleviation by 6-G remains unelucidated. The objective of the present study is to further investigate the potential mechanism for 6-G to alleviate MIRI in rats. Thirty-two Sprague-Dawley rats were randomly divided into four groups: the Sham group, the I/R group, the 6-G + I/R group, and the LY294002 (LY) + 6-G + I/R group. For the rats in each of the groups, data were collected for cardiogram, cardiac function, area of myocardial infarction, myocardial pathology, myocardial enzyme, marker of inflammatory response, and PI3K/Akt signaling pathway. We found that the pretreatment of 6-G with 6 mg/kg could shrink the ST section of cardiogram, improve the cardiac function, reduce the area of myocardial infarction and the degree of cardiac pathological injury, lower the level of myocardial enzyme, and inhibit the inflammatory response. In addition, our results also indicated that 6-G could upregulate the expression of PI3K and p-Akt and that LY294002, a blocking agent of PI3K/Akt signaling pathway, could nullify the protecting role of 6-G. Our experimental results showed that 6-G could inhibit I/R-induced inflammatory response through the activation of the PI3K/Akt signaling pathway.
Collapse
|
40
|
Sun J, Yu X, Huangpu H, Yao F. Ginsenoside Rb3 protects cardiomyocytes against hypoxia/reoxygenation injury via activating the antioxidation signaling pathway of PERK/Nrf2/HMOX1. Biomed Pharmacother 2018; 109:254-261. [PMID: 30396083 DOI: 10.1016/j.biopha.2018.09.002] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 09/03/2018] [Accepted: 09/03/2018] [Indexed: 01/14/2023] Open
Abstract
OBJECTIVES This study aimed to investigate the pharmacological function and underlying regulation mechanisms of Ginsenoside-Rb3 (G-Rb3) in cardioprotection. METHODS Cultured H9C2 cells were pre-treated with gradient concentrations of G-Rb3, and subsequently challenged with hypoxia/reoxygenation (H/R) treatment. The generation of intracellular reactive oxygen species (ROS) and cellular antioxidatant capacity were quantified. Cell apoptosis was measured by flow cytometry. Myocardial ischemia reperfusion injury (MIRI) rat models constructed by coronary artery ligation surgery were orally administrated with G-Rb3 for 5 consecutive days, and then infarction area, apoptosis ratio and total antioxidant capacity (T-AOC) of myocardial tissues were measured. PERK phosphorylation inhibitor GSK2656157 and Nrf2 translocation inhibitor ML385 were co-treated with G-Rb3 to further verify the signaling pathway mediated by G-Rb3. RESULTS H/R treatment induced prominent ROS deposition and elevated cell apoptosis ratio in H9C2 cells. G-Rb3 pretreatment suppressed intracellular ROS accumulation and enhanced T-AOC, partially rescuing cardiomyocytes from oxidative stress and apoptosis induced by H/R. In vivo, the cardiac infarction area of MIRI model rats was reduced by G-Rb3 treatment via improved total antioxidant levels. In the further functional and mechanistic studies, G-Rb3 was found to induce PERK phosphorylation and nuclear translocation of transcriptional factor Nrf2, promoting the expression of antioxidative genes such as HMOX1. Inhibitors GSK2656157 and ML385 reversed the effects of G-Rb3. CONCLUSION Our studies revealed a novel mechanism of G-Rb3 to attenuates oxidative stress via activating the antioxidation signaling pathway of PERK/Nrf2/HMOX1 in vivo and in vitro, which may help us to enrich the theoretical knewledge of Ginsenoside-Rb3 in cardiopretection.
Collapse
Affiliation(s)
- Jing Sun
- Second Department of Cardiovascular, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, No. 26, Heping Road, Xiangfang District, Harbin 150040, Heilongjiang Province, PR China
| | - Xiaohong Yu
- Second Department of Cardiovascular, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, No. 26, Heping Road, Xiangfang District, Harbin 150040, Heilongjiang Province, PR China
| | - Haiquan Huangpu
- Second Department of Cardiovascular, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, No. 26, Heping Road, Xiangfang District, Harbin 150040, Heilongjiang Province, PR China
| | - Fengzhen Yao
- Heilongjiang University of Chinese Medicine, No. 24, Heping Road, Xiangfang District, Harbin 150040, Heilongjiang Province, PR China.
| |
Collapse
|
41
|
Fang HC, Wu BQ, Hao YL, Luo Y, Zhao HL, Zhang WY, Zhang ZL, Liang JJ, Liu W, Chen XH. KRT1 gene silencing ameliorates myocardial ischemia-reperfusion injury via the activation of the Notch signaling pathway in mouse models. J Cell Physiol 2018; 234:3634-3646. [PMID: 30191968 DOI: 10.1002/jcp.27133] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 07/05/2018] [Indexed: 12/16/2022]
Abstract
Myocardial ischemia and reperfusion injury (MIRI) includes major drawbacks, such as excessive formation of free radicals and also overload of calcium, which lead to cell death, tissue scarring, and remodeling. The current study aims to explore whether KRT1 silencing may ameliorate MIRI via the Notch signaling pathway in mouse models. Myocardial tissues were used for the determination of the positive rate of KRT1 protein expression, apoptosis of myocardial cells, creatine kinase (CK) and lactate dehydrogenase (LDH) expression, expression of related biomarkers as well as myocardial infarction area. The transfected myocardial cells were treated with KRT1-siRNA, Jagged1, and DAPT (inhibitor of Notch-1 signaling pathway). The expression of KRT1, NICD, Hes1, Bcl-2, and Bax protein was detected. The MTT assay was applied for cell proliferation and flow cytometry was used for cell apoptosis. Mice with MIRI had a higher positive rate of KRT1 protein expression, apoptosis of myocardial cells, CK and LDH expression, myocardial infarction area, increased expression of MDA, NO, SDH, IL-1, IL-6, TNF-α, CRP, KRT1, Bax protein, CK, and LDH, and decreased expression of SOD, NICD, Hes1, and Bcl-2. The downregulation of KRT1 led to decreased expression of KRT1 and Bax protein, increased expression of NICD, Hes1, and Bcl-2, decreased cell apoptosis, and improved cell proliferation. The inhibition of the Notch signaling pathway leads to reduced expression of Bax, increased expression of NICD, Hes1, and Bcl 2, and also decreased cell apoptosis and increased cell proliferation. Our data conclude that KRT1 silencing is able to make MIRI better by activating the Notch signaling pathway in mice.
Collapse
Affiliation(s)
- Hong-Cheng Fang
- Shenzhen Baoan Shajing People's Hospital of Guangzhou Medical University, Shenzhen, China
| | - Bao-Quan Wu
- Department of Geriatrics and Cardiovascular Medicine, Shenzhen Sun Yat-Sen Cardiovascular Hospital, Shenzhen, China
| | - Yun-Ling Hao
- Department of Geriatrics and Cardiovascular Medicine, Shenzhen Sun Yat-Sen Cardiovascular Hospital, Shenzhen, China
| | - Ying Luo
- Department of Geriatrics and Cardiovascular Medicine, Shenzhen Sun Yat-Sen Cardiovascular Hospital, Shenzhen, China
| | - Hong-Lei Zhao
- Department of Geriatrics and Cardiovascular Medicine, Shenzhen Sun Yat-Sen Cardiovascular Hospital, Shenzhen, China
| | - Wen-Ying Zhang
- Department of Geriatrics and Cardiovascular Medicine, Shenzhen Sun Yat-Sen Cardiovascular Hospital, Shenzhen, China
| | - Zhi-Ling Zhang
- Department of Geriatrics and Cardiovascular Medicine, Shenzhen Sun Yat-Sen Cardiovascular Hospital, Shenzhen, China
| | - Jin-Jie Liang
- Department of Geriatrics and Cardiovascular Medicine, Shenzhen Sun Yat-Sen Cardiovascular Hospital, Shenzhen, China
| | - Wei Liu
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xie-Hui Chen
- Department of Geriatrics and Cardiovascular Medicine, Shenzhen Sun Yat-Sen Cardiovascular Hospital, Shenzhen, China
| |
Collapse
|
42
|
Yang J, Yang C, Yang J, Ding J, Li X, Yu Q, Guo X, Fan Z, Wang H. RP105 alleviates myocardial ischemia reperfusion injury via inhibiting TLR4/TRIF signaling pathways. Int J Mol Med 2018; 41:3287-3295. [PMID: 29512709 PMCID: PMC5881694 DOI: 10.3892/ijmm.2018.3538] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 01/12/2018] [Indexed: 02/06/2023] Open
Abstract
The Toll-like receptor 4 (TLR4) signal pathway- induced inflammation is considered to be a crucial link to myocardial ischemia reperfusion injury (MIRI). Our previous study proved that radioprotective 105 kDa protein (RP105), a negative regulator of TLR4, performed a protective role in MIRI by anti-apoptosis approach. However, the mechanism of RP105 cardioprotection of anti-inflammation is still unclear. This study aimed to explore the underlying mechanism of RP105 anti-inflammation effect in MIRI. We established a rat model of MIRI induced by ligation of the left anterior descending coronary artery for 30 min followed by 2 h reperfusion. Animals were pre-infected with Ad-EGFP-RP105, Ad-EGFP or saline at the apex of the heart. All rats were sacrificed to collect blood samples and myocardial tissue and assessed by immunofluorescence, blood biochemical analysis, Evans blue/triphenyltetrazolium chloride (TTC), hematoxylin and eosin (H&E) staining, enzyme-linked immuno sorbent assay (ELISA), western blot analysis, quantitative PCR and electrophoretic mobility shift assay (EMSA). RP105 overexpression with adenovirus vectors reduced serum myocardial enzyme (CK-MB and LDH) activities, decreased myocardial infarct size, mitigated inflammatory factors interferon-β and tumor necrosis factor-α during MIRI. We also found that Ad-RP105 group exerted distinct repression of TLR4/TRIF signal pathway related proteins and mRNAs (TRIF, TBK-1, IRF3 and p-IRF3) with a low transcriptional activity of IRF3. These findings first expounded that RP105 could alleviate the ischemia reperfusion induced inflammatory status in heart via inhibiting TLR4/TRIF signaling pathway and provided a theoretical foundation of RP105 gene in MIRI.
Collapse
Affiliation(s)
- Jun Yang
- Department of Cardiology, The First College of Clinical Medical Sciences
| | - Chaojun Yang
- Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| | - Jian Yang
- Department of Cardiology, The First College of Clinical Medical Sciences
| | - Jiawang Ding
- Department of Cardiology, The First College of Clinical Medical Sciences
| | - Xinxin Li
- Department of Cardiology, The First College of Clinical Medical Sciences
| | - Qinqin Yu
- Department of Cardiology, The First College of Clinical Medical Sciences
| | - Xin Guo
- Department of Cardiology, The First College of Clinical Medical Sciences
| | - Zhixing Fan
- Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| | - Huibo Wang
- Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| |
Collapse
|
43
|
Zhang H, Zhao Y, Xia Z, Du H, Gao Y, Xue D, Zhu Z, Chai Y. Metabolic profiles revealed anti-ischemia-reperfusion injury of Yangxinshi tablet in Rats. JOURNAL OF ETHNOPHARMACOLOGY 2018; 214:124-133. [PMID: 28889959 DOI: 10.1016/j.jep.2017.09.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 09/05/2017] [Accepted: 09/06/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Myocardial ischemia-reperfusion (I/R) injury is a serious injury that is resulted from the recovery of blood supply after myocardial ischemia. Yangxinshi tablet is a compound Chinese herbal preparation and often used to alleviate the myocardial ischemia in clinical, but its protective mechanism of anti-myocardial ischemia reperfusion injury remains unclear. The objective of this study was to evaluate the anti-I/R injury effect of Yangxinshi tablet on a myocardial I/R rat model and to identify serum biomarker metabolites associated with I/R based on ultra-high performance liquid chromatography coupled with quadrupole-time-of-flight mass spectrometry (UHPLC-QTOF/MS) metabolomic method, and explore the metabolic mechanism of anti-I/R injury of Yangxinshi tablet. MATERIALS AND METHODS Unsupervised principle component analysis highlighted significant differences in the metabolome of the myocardial I/R, healthy control and drug-treated rats. Partial least squares-discriminant analysis revealed 25 metabolites as the most potential biomarker metabolites discriminating the myocardial I/R rats and control rats. Most of the metabolites were primarily involved in oxidative stress, energy metabolism, fatty acid metabolism, amino acid metabolism. These metabolites were validated by assessing the efficacy after intragastric administration of Yangxinshit ablet to the myocardial I/R rat model. RESULTS Based on metabolomic results, the action mechanism of anti-I/R injury of Yangxinshi tablet was concluded as follows: (1) enhance the ability of scavenging free radicals and reactive oxygen species in vivo; (2) provide energy for myocardium via accelerating the intracellular carnitine transportion to accelerate the oxidation of fatty acid and (3) attenuate ceramide to reduce cardiomyocyte apoptosis. CONCLUSIONS Yangxinshi tablet has cardio-protection effects on I/R rats via regulation of multiple metabolic pathways involving in oxidative stress, energy metabolism, fatty acid, and amino acid metabolisms. This study will be meaningful for its clinical application and valuable for further exploring the action mechanism of Yangxinshi tablet.
Collapse
Affiliation(s)
- Hai Zhang
- Department of Pharmacy, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, China
| | - Yahong Zhao
- Department of Chinese Materia Medica, Central Research Institute, Shanghai Pharmaceuticals Holding Co.Ltd., Shanghai 201203, China
| | - Zhengxiang Xia
- Department of Pharmacy, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, China
| | - Hongli Du
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai 200433, China
| | - Yue Gao
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Dan Xue
- Department of Chinese Materia Medica, Central Research Institute, Shanghai Pharmaceuticals Holding Co.Ltd., Shanghai 201203, China
| | - Zhenyu Zhu
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China.
| | - Yifeng Chai
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
44
|
Yu Y, Jin L, Zhuang Y, Hu Y, Cang J, Guo K. Cardioprotective effect of rosuvastatin against isoproterenol-induced myocardial infarction injury in rats. Int J Mol Med 2018; 41:3509-3516. [PMID: 29568858 DOI: 10.3892/ijmm.2018.3572] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 01/25/2018] [Indexed: 11/05/2022] Open
Abstract
Rosuvastatin, a member of 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors, exerts various pharmacological activities. This study evaluated the cardioprotective effect of rosuvastatin on isoproterenol-induced myocardial infarction injury in rats. A rat model of myocardial infarction injury was induced by isoproterenol (ISO) for 2 consecutive days, rosuvastatin was administered for 8 weeks. The levels of myocardial infarct size, aspartate transaminase (AST), alanine transaminase (ALT), creatine kinase-MB (CK-MB), lactate dehydrogenase (LDH) activities, as well as malondialdehyde (MDA) levels, superoxide dismutase (SOD), glutathione peroxidase (GPX), catalase (CAT) activities and reduced glutathione (GSH) concentrations were determined. Hematoxylin and eosin staining was used to observe cardiac histological changes. Interleukin-1β (IL-1β) and IL-18 levels in heart tissues were detected with ELISA kits. The mRNA and protein levels of NOD-like receptor superfamily, pyrin domain containing 3 (NLRP3) inflammasome were measured by qRT-PCR and western blot analysis, respectively. Our results showed that treatment with rosuvastatin reduced myocardial infract area, ameliorated histopathological alterations in myocardium, and decreased activities of myocardial injury marker enzymes in ISO-induced rats. In addition, rosuvastatin remarkably restored ISO-induced elevation of lipid peroxidation and decrease of antioxidants, significantly reduced myocardial pro-inflammatory cytokines concentrations in this animal model. Furthermore, rosuvastatin significantly inhibited the activation of NLRP3 inflammasome in this animal model. This study demonstrates that rosuvastatin significantly alleviates ISO-induced myocardial infarction injury. The mechanism is associated with attenuation of oxidative stress and inflammation, via the inhibition of NLRP3 inflammasome.
Collapse
Affiliation(s)
- Ying Yu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Lin Jin
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Yamin Zhuang
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Yan Hu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Jing Cang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Kefang Guo
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
45
|
Abad C, Castaño-Ruiz M, Clavo B, Urso S. Daño por isquemia-reperfusión miocárdico en cirugía cardiaca con circulación extracorpórea. Aspectos bioquímicos. CIRUGIA CARDIOVASCULAR 2018. [DOI: 10.1016/j.circv.2017.09.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
46
|
Hartman CL, Duerr MA, Albert CJ, Neumann WL, McHowat J, Ford DA. 2-Chlorofatty acids induce Weibel-Palade body mobilization. J Lipid Res 2018; 59:113-122. [PMID: 29167411 PMCID: PMC5748502 DOI: 10.1194/jlr.m080200] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 11/02/2017] [Indexed: 01/23/2023] Open
Abstract
Endothelial dysfunction is a hallmark of multiple inflammatory diseases. Leukocyte interactions with the endothelium have significant effects on vascular wall biology and pathophysiology. Myeloperoxidase (MPO)-derived oxidant products released from leukocytes are potential mediators of inflammation and endothelial dysfunction. 2-Chlorofatty acids (2-ClFAs) are produced as a result of MPO-derived HOCl targeting plasmalogen phospholipids. Chlorinated lipids have been shown to be associated with multiple inflammatory diseases, but their impact on surrounding endothelial cells has not been examined. This study tested the biological properties of the 2-ClFA molecular species 2-chlorohexadecanoic acid (2-ClHA) on endothelial cells. A synthetic alkyne analog of 2-ClHA, 2-chlorohexadec-15-ynoic acid (2-ClHyA), was used to examine the subcellular localization of 2-ClFA in human coronary artery endothelial cells. Click chemistry experiments revealed that 2-ClHyA localizes to Weibel-Palade bodies. 2-ClHA and 2-ClHyA promote the release of P-selectin, von Willebrand factor, and angiopoietin-2 from endothelial cells. Functionally, 2-ClHA and 2-ClHyA cause neutrophils to adhere to and platelets to aggregate on the endothelium, as well as increase permeability of the endothelial barrier which has been tied to the release of angiopoietin-2. These findings suggest that 2-ClFAs promote endothelial cell dysfunction, which may lead to broad implications in inflammation, thrombosis, and blood vessel stability.
Collapse
Affiliation(s)
- Celine L Hartman
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO 63104
- Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO 63104
| | - Mark A Duerr
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO 63104
- Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO 63104
| | - Carolyn J Albert
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO 63104
- Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO 63104
| | - William L Neumann
- Department of Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University-Edwardsville, Edwardsville, IL 62026
| | - Jane McHowat
- Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO 63104
- Department of Pathology, Saint Louis University School of Medicine, St. Louis, MO 63104
| | - David A Ford
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO 63104
- Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO 63104
| |
Collapse
|
47
|
Chen C, Chen W, Li Y, Dong Y, Teng X, Nong Z, Pan X, Lv L, Gao Y, Wu G. Hyperbaric oxygen protects against myocardial reperfusion injury via the inhibition of inflammation and the modulation of autophagy. Oncotarget 2017; 8:111522-111534. [PMID: 29340072 PMCID: PMC5762340 DOI: 10.18632/oncotarget.22869] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 08/27/2017] [Indexed: 01/01/2023] Open
Abstract
Our previous study demonstrated that hyperbaric oxygen (HBO) preconditioning protected against myocardial ischemia reperfusion injury (MIRI) and improved myocardial infarction. However, HBO’s effect on MIRI-induced inflammation and autophagy remains unclear. In this study, we investigate the potential impact and underlying mechanism of HBO preconditioning on an MIRI-induced inflammatory response and autophagy using a ligation of the left anterior descending (LAD) coronary artery rat model. Our results showed that HBO restored myocardial enzyme levels and decreased the apoptosis of cardiomyocytes, which were induced by MIRI. Moreover, HBO significantly suppressed MIRI-induced inflammatory cytokines. This effect was associated with the inhibition of the TLR4-nuclear factor kappa-B (NF-κB) pathway. Interestingly, lower expression levels of microtubule-associated protein 1 light chain 3B (LC3B) and Beclin-1 were observed in the HBO-treatment group. Furthermore, we observed that HBO reduced excessive autophagy by activating the mammalian target of the rapamycin (mTOR) pathway, as evidenced by higher expression levels of threonine protein kinase (Akt) and phosphorylated-mTOR. In conclusion, HBO protected cardiomocytes during MIRI by attenuating inflammation and autophagy. Our results provide a new mechanistic insight into the cardioprotective role of HBO against MIRI.
Collapse
Affiliation(s)
- Chunxia Chen
- Department of Hyperbaric Oxygen, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P. R. China
| | - Wan Chen
- Department of Emergency, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P. R. China
| | - Yaoxuan Li
- Department of Neurology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P. R. China
| | - Yanling Dong
- Department of Neurology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P. R. China
| | - Xiaoming Teng
- Department of Neurology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P. R. China
| | - Zhihuan Nong
- Department of Pharmacology, Guangxi Institute of Chinese Medicine and Pharmaceutical Science, Nanning, Guangxi 530022, P. R. China
| | - Xiaorong Pan
- Department of Hyperbaric Oxygen, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P. R. China
| | - Liwen Lv
- Department of Emergency, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P. R. China
| | - Ying Gao
- Department of Biology and Tennessee Center for Botanical Medicine Research, Middle Tennessee State University, Murfreesboro, TN 37132, USA
| | - Guangwei Wu
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P. R. China
| |
Collapse
|
48
|
Umbelliferone Alleviates Myocardial Ischemia: the Role of Inflammation and Apoptosis. Inflammation 2017; 41:464-473. [DOI: 10.1007/s10753-017-0702-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
49
|
Wang L, Xue Y, Ma H, Shi H, Wang L, Cui X. Prazosin protects myocardial cells against anoxia-reoxygenation injury via the extracellular signal‑regulated kinase signaling pathway. Mol Med Rep 2017; 17:2145-2152. [PMID: 29207167 PMCID: PMC5783458 DOI: 10.3892/mmr.2017.8175] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 07/11/2017] [Indexed: 11/09/2022] Open
Abstract
Ischemic heart disease (including coronary arterial atherosclerosis, or vascular cavity stenosis or occlusion) remains the leading cause of disease-associated mortality worldwide. Prazosin, a receptor blocker of postsynaptic adrenaline, is essential in expanding peripheral arteries, which decreases peripheral vascular resistance, and regulates anti-hypertensive action. However, the mechanisms underlying the effects of prazosin have not been fully elucidated. The aim of the present study was to investigate the protective effects of prazosin on myocardial cells against anoxia-reoxygenation injury in a mouse model. The regulatory effects of prazosin on blood lipid levels and blood pressure were investigated in experimental mice. Furthermore, inflammation responses and oxidative stress in myocardial cells were analyzed in mice treated with prazosin. Apoptotic myocardial cells were investigated in experimental mice treated with prazosin. In addition, apoptotic gene expression levels were evaluated in myocardial cells. Extracellular signal-regulated kinase (ERK) expression and phosphorylation was investigated in myocardial cells in mice with anoxia-reoxygenation injury following prazosin treatment. The activity and expression levels of nuclear factor of activated T cells (NF-AT), activator protein 1 (AP-1) and necrosis factor (NF)-κB were observed in myocardial cells. Furthermore, histological analyses were performed to investigate the benefits of prazosin treatment on anoxia-reoxygenation injury. The results of the present study identified that prazosin decreased the expression levels of inflammatory factors, interleukin (IL)-6, tumor necrosis factor (TNF)-α, IL-10 and IL-1 in the serum of mice exhibiting hypoxia/reoxygenation injury. Oxidative stress was observed to be improved and the apoptosis rate was decreased in myocardial cells in anoxia-reoxygenation injury model mice treated with prazosin. ERK expression and phosphorylation was upregulated, and expression levels of NF-AT, AP-1 and NF-κB were downregulated in the myocardial cells of mice treated with prazosin. Blood lipid levels and blood pressure of the anoxia-reoxygenation injury model mice were markedly improved following treatment with prazosin. Histological analysis indicated that the area, circumference fragmentation and segmentation of myocardial cells were significantly improved following prazosin treatment. Thus, these results indicate that prazosin treatment decreases inflammation responses, oxidative stress, and apoptosis of myocardial cells via regulation of the ERK signaling pathway. The findings indicate that prazosin may present as a potential therapeutic agent for the treatment of hypoxia/reoxygenation injury.
Collapse
Affiliation(s)
- Lixin Wang
- Department of Cardiovascular Surgery, The General Hospital of Chinese People's Armed Police Force, Beijing 100039, P.R. China
| | - Yan Xue
- Department of Cardiovascular Surgery, The General Hospital of Chinese People's Armed Police Force, Beijing 100039, P.R. China
| | - Hao Ma
- Department of Cardiovascular Surgery, The General Hospital of Chinese People's Armed Police Force, Beijing 100039, P.R. China
| | - Haiyan Shi
- Department of Cardiovascular Surgery, The General Hospital of Chinese People's Armed Police Force, Beijing 100039, P.R. China
| | - Ling Wang
- Department of Cardiovascular Surgery, The General Hospital of Chinese People's Armed Police Force, Beijing 100039, P.R. China
| | - Xiaozheng Cui
- Department of Cardiovascular Surgery, The General Hospital of Chinese People's Armed Police Force, Beijing 100039, P.R. China
| |
Collapse
|
50
|
Wang L, Ma H, Xue Y, Shi H, Ma T, Cui X. Berberine inhibits the ischemia-reperfusion injury induced inflammatory response and apoptosis of myocardial cells through the phosphoinositide 3-kinase/RAC-α serine/threonine-protein kinase and nuclear factor-κB signaling pathways. Exp Ther Med 2017; 15:1225-1232. [PMID: 29403554 PMCID: PMC5780743 DOI: 10.3892/etm.2017.5575] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Accepted: 07/14/2017] [Indexed: 12/24/2022] Open
Abstract
Myocardial ischemia-reperfusion injury is one of the most common cardiovascular diseases, and can lead to serious damage and dysfunction of the myocardial tissue. Previous studies have demonstrated that berberine exhibits ameliorative effects on cardiovascular disease. The present study further investigated the efficacy and potential mechanism underlying the effects of berberine on ischemia-reperfusion injury in a mouse model. Inflammatory markers were measured in the serum and levels of inflammatory proteins in myocardial cells were investigated after treatment with berberine. In addition, the apoptosis of myocardial cells was investigated after berberine treatment. Apoptosis-associated gene expression levels and apoptotic signaling pathways were analyzed in myocardial cells after treatment with berberine. The phosphoinositide 3-kinase (PI3K)/RAC-α serine/threonine-protein kinase (AKT) and nuclear factor (NF)-κB signaling pathways were also analyzed in myocardial cells after treatment with berberine. Histological analysis was used to analyze the potential benefits of berberine in ischemia-reperfusion injury. The present study identified that inflammatory responses and inflammatory factors were decreased in the myocardial cells of the mouse model of ischemia-reperfusion injury. Mechanism analysis demonstrated that berberine inhibited apoptotic protease-activating factor 1, caspase-3 and caspase-9 expression in myocardial cells. The expression of Bcl2-associated agonist of cell death, Bcl-2-like protein 1 and cellular tumor antigen p53 was upregulated. Expression of NF-κB p65, inhibitor of NF-κB kinase subunit β (IKK-β), NF-κB inhibitor α (IκBα), and NF-κB activity, were inhibited in myocardial cells in the mouse model of ischemia-reperfusion injury. In conclusion, the results of the present study indicate that berberine inhibits inflammatory responses through the NF-κB signaling pathway and suppresses the apoptosis of myocardial cells via the PI3K/AKT signaling pathway in a mouse model of ischemia-reperfusion injury. These results suggest that berberine is a potential drug for the treatment of patients with ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Lixin Wang
- Department of Cardiovascular Surgery, The General Hospital of The Chinese People's Armed Police Forces, Beijing 100039, P.R. China
| | - Hao Ma
- Department of Cardiovascular Surgery, The General Hospital of The Chinese People's Armed Police Forces, Beijing 100039, P.R. China
| | - Yan Xue
- Department of Cardiovascular Surgery, The General Hospital of The Chinese People's Armed Police Forces, Beijing 100039, P.R. China
| | - Haiyan Shi
- Department of Cardiovascular Surgery, The General Hospital of The Chinese People's Armed Police Forces, Beijing 100039, P.R. China
| | - Teng Ma
- Department of Cardiovascular Surgery, The General Hospital of The Chinese People's Armed Police Forces, Beijing 100039, P.R. China
| | - Xiaozheng Cui
- Department of Cardiovascular Surgery, The General Hospital of The Chinese People's Armed Police Forces, Beijing 100039, P.R. China
| |
Collapse
|