51
|
The Effect of Angelica sinensis Polysaccharide on Neuronal Apoptosis in Cerebral Ischemia-Reperfusion Injury via PI3K/AKT Pathway. INT J POLYM SCI 2021. [DOI: 10.1155/2021/7829341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
In the present study, the protective effects and mechanism of Angelica sinensis polysaccharide (ASP) were investigated in rats with cerebral ischemia-reperfusion injury (CIRI). Rats were randomly divided into sham group, CIRI group, ASP treatment group, and ASP and LY294002 treatment group. H&E results confirmed the successful induction of CIRI in Sprague-Dawley rats. Compared with the sham group, the neurological function score, percentage of myocardial infarction area, neuronal apoptosis, oxidative stress, and inflammation in the CIRI group were significantly increased. Compared with the CIRI group, the ASP group’s neurological function score, percentage of myocardial infarction area, neuronal apoptosis, oxidative stress, and inflammation were significantly reduced. However, compared with the ASP group, LY294002 inhibited the effect of ASP in CIRI rats. CIRI downregulated the PI3K/AKT pathway and upregulated the apoptosis level. And ASP activated the PI3K/AKT pathway and Bcl-2 protein expression, while it inhibited caspase-3 and Bax expression. LY294002 could significantly inhibit the protective effect of ASP on nerve injury and the expression and phosphorylation of PI3K and Akt protein in CIRI rats. ASP could effectively improve nerve function and nerve cell apoptosis of CIRI rats by activating the PI3K/AKT signaling pathway.
Collapse
|
52
|
Tejchman K, Kotfis K, Sieńko J. Biomarkers and Mechanisms of Oxidative Stress-Last 20 Years of Research with an Emphasis on Kidney Damage and Renal Transplantation. Int J Mol Sci 2021; 22:ijms22158010. [PMID: 34360776 PMCID: PMC8347360 DOI: 10.3390/ijms22158010] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 07/22/2021] [Accepted: 07/23/2021] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress is an imbalance between pro- and antioxidants that adversely influences the organism in various mechanisms and on many levels. Oxidative damage occurring concomitantly in many cellular structures may cause a deterioration of function, including apoptosis and necrosis. The damage leaves a molecular “footprint”, which can be detected by specific methodology, using certain oxidative stress biomarkers. There is an intimate relationship between oxidative stress, inflammation, and functional impairment, resulting in various diseases affecting the entire human body. In the current narrative review, we strengthen the connection between oxidative stress mechanisms and their active compounds, emphasizing kidney damage and renal transplantation. An analysis of reactive oxygen species (ROS), antioxidants, products of peroxidation, and finally signaling pathways gives a lot of promising data that potentially will modify cell responses on many levels, including gene expression. Oxidative damage, stress, and ROS are still intensively exploited research subjects. We discuss compounds mentioned earlier as biomarkers of oxidative stress and present their role documented during the last 20 years of research. The following keywords and MeSH terms were used in the search: oxidative stress, kidney, transplantation, ischemia-reperfusion injury, IRI, biomarkers, peroxidation, and treatment.
Collapse
Affiliation(s)
- Karol Tejchman
- Department of General and Transplantation Surgery, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.T.); (J.S.)
| | - Katarzyna Kotfis
- Department of Anesthesiology, Intensive Therapy and Acute Intoxications, Pomeranian Medical University, 70-111 Szczecin, Poland
- Correspondence: ; Tel.: +48914661144
| | - Jerzy Sieńko
- Department of General and Transplantation Surgery, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.T.); (J.S.)
| |
Collapse
|
53
|
Wang P, Wang S, Chen H, Deng X, Zhang L, Xu H, Yang H. TCMIP v2.0 Powers the Identification of Chemical Constituents Available in Xinglou Chengqi Decoction and the Exploration of Pharmacological Mechanisms Acting on Stroke Complicated With Tanre Fushi Syndrome. Front Pharmacol 2021; 12:598200. [PMID: 34335236 PMCID: PMC8320350 DOI: 10.3389/fphar.2021.598200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 06/21/2021] [Indexed: 11/13/2022] Open
Abstract
Xinglou Chengqi (XLCQ) decoction, composed of three botanical drugs and one inorganic drug, is used in clinics during the treatment of acute stroke complicated with Tanre Fushi (TRFS) syndrome in China. However, its active ingredients and the molecular mechanism have not been clarified. So, we aimed to preliminarily characterize its chemical constituents and investigate its pharmacological mechanisms using an integrative pharmacology strategy, including component analysis, network prediction, and experimental verification. We employed UPLC-QTOF-MS/MS to describe the chemical profile of XLCQ, Integrative Pharmacology-based Network Computational Research Platform of Traditional Chinese Medicine (TCMIP v2.0, http://www.tcmip.cn/), to assist in identifying the chemical components and predict the putative molecular mechanism against acute stroke complicated with TRFS, and LPS-stimulated BV-2 cells to verify the anti-neuroinflammatory effects of luteolin, apigenin, and chrysoeriol. Altogether, 197 chemical compounds were identified or tentatively characterized in the water extraction of XLCQ, 22 of them were selected as the key active constituents that may improve the pathological state by regulating 27 corresponding targets that are mainly involved in inflammation/immune-related pathways, and furthermore, luteolin, apigenin, and chrysoeriol exhibited good anti-neuroinflammatory effects from both protein and mRNA levels. In summary, it is the first time to employ an integrative pharmacology strategy to delineate 22 constituents that may improve the pathological state of stroke with TRFS by regulating 27 corresponding targets, which may offer a highly efficient way to mine the scientific connotation of traditional Chinese medicine prescriptions. This study might be a supplement for the deficiency of the basic research of XLCQ.
Collapse
Affiliation(s)
- Ping Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shuang Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.,College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Hong Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.,College of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiaofang Deng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Luoqi Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Haiyu Xu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.,Shaanxi Institute of International Trade and Commerce, Xianyang, China
| | - Hongjun Yang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
54
|
Hajinejad M, Sahab-Negah S. Neuroinflammation: The next target of exosomal microRNAs derived from mesenchymal stem cells in the context of neurological disorders. J Cell Physiol 2021; 236:8070-8081. [PMID: 34189724 DOI: 10.1002/jcp.30495] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/28/2021] [Accepted: 06/17/2021] [Indexed: 12/16/2022]
Abstract
Among different types of mechanisms involved in neurological disorders, neuroinflammation links initial insults to secondary injuries and triggers some chronic outcomes, for example, neurodegenerative disorders. Thus, anti-inflammatory substances can be targeted as a novel therapeutic option for translational and clinical research to improve brain disease outcomes. In this review, we propose to introduce a new insight into the anti-inflammatory effects of mesenchymal stem cells (MSCs) as the most frequent source for stem cell therapy in neurological diseases. Our insight incorporates a bystander effect of these stem cells in modulating inflammation and microglia/macrophage polarization through exosomes. Exosomes are nano-sized membrane vesicles that carry cell-specific constituents, including protein, lipid, DNA, and RNA. microRNAs (miRNAs) have recently been detected in exosomes that can be taken up by other cells and affect the behavior of recipient cells. In this article, we outline and highlight the potential use of exosomal miRNAs derived from MSCs for inflammatory pathways in the context of neurological disorders. Furthermore, we suggest that focusing on exosomal miRNAs derived from MSCs in the course of neuroinflammatory pathways in the future could reveal their functions for diverse neurological diseases, including brain injuries and neurodegenerative diseases. It is hoped that this study will contribute to a deep understanding of stem cell bystander effects through exosomal miRNAs.
Collapse
Affiliation(s)
- Mehrdad Hajinejad
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Anatomy and Cell Biology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sajad Sahab-Negah
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.,Department of Neuroscience, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
55
|
Zhuo Y, Yuan R, Chen X, He J, Chen Y, Zhang C, Sun K, Yang S, Liu Z, Gao H. Tanshinone I exerts cardiovascular protective effects in vivo and in vitro through inhibiting necroptosis via Akt/Nrf2 signaling pathway. Chin Med 2021; 16:48. [PMID: 34183021 PMCID: PMC8240219 DOI: 10.1186/s13020-021-00458-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 06/16/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Tanshinone I (TI) is a primary component of Salvia miltiorrhiza Bunge (Danshen), which confers a favorable role in a variety of pharmacological activities including cardiovascular protection. However, the exact mechanism of the cardiovascular protection activity of TI remains to be illustrated. In this study, the cardiovascular protective effect and its mechanism of TI were investigated. METHODS In this study, tert-butyl hydroperoxide (t-BHP)-stimulated H9c2 cells model was employed to investigate the protective effect in vitro. The cell viability was determined by 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl tetrazolium bromide (MTT) assay and lactate dehydrogenase (LDH) kit. The reactive-oxygen-species (ROS) level and mitochondrial membrane potential (MMP) were investigated by the flow cytometry and JC-1 assay, respectively. While in vivo experiment, the cardiovascular protective effect of TI was determined by using myocardial ischemia-reperfusion (MI/R) model including hematoxylin-eosin (H&E) staining assay and determination of superoxide dismutase (SOD) and malondialdehyde (MDA). Tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) release were detected by Enzyme-linked immunosorbent assay (ELISA). Receptor interacting protein kinase 1 (RIP1), receptor interacting protein kinase 3 (RIP3), receptor interacting protein kinase 3 (MLKL), protein kinase B (Akt), Nuclear factor erythroid 2 related factor 2 (Nrf2), Heme oxygenase-1 (HO-1) and NAD(P)H: quinone oxidoreductase-1 (NQO-1) were determined by western blotting. RESULTS Our data demonstrated that TI pretreatment attenuated t-BHP and MI/R injury-induced necroptosis by inhibiting the expression of p-RIP1, p-RIP3, and p-MLKL. TI activated the Akt/Nrf2 pathway to promote the expression of antioxidant-related proteins such as phosphorylation of Akt, nuclear factor erythroid 2 related factor 2 (Nrf2), quinone oxidoreductase-1 (NQO-1) and heme oxygenase-1 (HO-1) expression in t-BHP-stimulated H9c2 cells. TI relieved oxidative stress by mitigating ROS generation and reversing MMP loss. In vivo experiment, TI made electrocardiograph (ECG) recovery better and lessened the degree of myocardial tissue damage. The counts of white blood cell (WBC), neutrophil (Neu), lymphocyte (Lym), and the release of TNF-α and IL-6 were reversed by TI treatment. SOD level was increased, while MDA level was decreased by TI treatment. CONCLUSION Collectively, our findings indicated that TI exerted cardiovascular protective activities in vitro and in vivo through suppressing RIP1/RIP3/MLKL and activating Akt/Nrf2 signaling pathways, which could be developed into a cardiovascular protective agent.
Collapse
Affiliation(s)
- Youqiong Zhuo
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530200, China
| | - Renyikun Yuan
- State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, China
| | - Xinxin Chen
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530200, China
| | - Jia He
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530200, China
| | - Yangling Chen
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530200, China
| | - Chenwei Zhang
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530200, China
| | - Kaili Sun
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530200, China
| | - Shilin Yang
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530200, China
| | - Zhenjie Liu
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530200, China
| | - Hongwei Gao
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China.
- Guangxi Engineering Technology Research Center of Advantage Chinese Patent Drug and Ethnic Drug Development, Nanning, 530200, China.
| |
Collapse
|
56
|
Lei Y, Yang M, Li H, Xu R, Liu J. miR‑130b regulates PTEN to activate the PI3K/Akt signaling pathway and attenuate oxidative stress‑induced injury in diabetic encephalopathy. Int J Mol Med 2021; 48:141. [PMID: 34080640 PMCID: PMC8175068 DOI: 10.3892/ijmm.2021.4974] [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: 09/14/2020] [Accepted: 04/29/2021] [Indexed: 12/11/2022] Open
Abstract
Diabetic encephalopathy (DE) is one of the main chronic complications of diabetes, and is characterized by cognitive defects. MicroRNAs (miRNAs/miRs) are widely involved in the development of diabetes-related complications. The present study evaluated the role of miR-130b in DE and investigated its mechanisms of action. PC12 cells and hippocampal cells were exposed to a high glucose environment to induce cell injuries to mimic the in vitro model of DE. Cells were transfected with miR-130b mimic, miR-130b inhibitor and small interfering RNA (si)-phosphatase and tensin homolog (PTEN) to evaluate the protective effect of the miR-130b/PTEN axis against oxidative stress in high glucose-stimulated cells involving Akt activity. Furthermore, the effect of agomir-130b was also assessed on rats with DE. The expression of miR-130b was reduced in the DE models in vivo and in vitro. The administration of miR-130b mimic increased the viability of high glucose-stimulated cells, prevented apoptosis, increased the activity of superoxide dismutase (SOD), decreased the malondialdehyde (MDA) content, activated Akt protein levels and inhibited the mitochondria-mediated apoptotic pathway. The administration of miR-130b inhibitor exerted opposite effects, while si-PTEN reversed the effects of miR-130b inhibitor. In vivo, the administration of agomir-130b attenuated cognitive disorders and neuronal damage, increased SOD activity, reduced the MDA content, activated Akt protein levels and inhibited the mitochondria-mediated apoptosis pathway in rats with DE. On the whole, these results suggest that miR-130b activates the PI3K/Akt signaling pathway to exert protective effects against oxidative stress injury via the regulation of PTEN in rats with DE.
Collapse
Affiliation(s)
- Yonghua Lei
- Department of Traditional Chinese Medicine, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Ming Yang
- Department of Traditional Chinese Medicine, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Hong Li
- Department of Endocrinology, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P.R. China
| | - Rongjuan Xu
- Department of Endocrinology, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P.R. China
| | - Junbao Liu
- Department of Traditional Chinese Medicine, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan 450003, P.R. China
| |
Collapse
|
57
|
Jiao F, Varghese K, Wang S, Liu Y, Yu H, Booz GW, Roman RJ, Liu R, Fan F. Recent Insights Into the Protective Mechanisms of Paeoniflorin in Neurological, Cardiovascular, and Renal Diseases. J Cardiovasc Pharmacol 2021; 77:728-734. [PMID: 34001724 PMCID: PMC8169546 DOI: 10.1097/fjc.0000000000001021] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/05/2021] [Indexed: 12/12/2022]
Abstract
ABSTRACT The monoterpene glycoside paeoniflorin (PF) is the principal active constituent of the traditional Chinese herbal medicines, Radix Paeoniae Alba and Radix Paeoniae Rubra, which have been used for millennia to treat cardiovascular diseases (eg, hypertension, bleeding, and atherosclerosis) and neurological ailments (eg, headaches, vertigo, dementia, and pain). Recent evidence has revealed that PF exerts inhibitory effects on inflammation, fibrosis, and apoptosis by targeting several intracellular signaling cascades. In this review, we address the current knowledge about the pharmacokinetic properties of PF and its molecular mechanisms of action. We also present results from recent preclinical studies supporting the utility of PF for the treatment of pain, cerebral ischemic injury, and neurodegenerative diseases, such as Alzheimer's and Parkinson's diseases. Moreover, new evidence suggests a general protective role of PF in heart attack, diabetic kidney, and atherosclerosis. Mechanistically, PF exerts multiple anti-inflammatory actions by targeting toll-like receptor-mediated signaling in both parenchymal and immune cells (in particular, macrophages and dendritic cells). A better understanding of the molecular actions of PF may lead to the expansion of its therapeutic uses.
Collapse
Affiliation(s)
- Feng Jiao
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Department of Neurosurgery, Peking University People’s Hospital, Beijing, 100044, China
| | - Kevin Varghese
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Shaoxun Wang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Yedan Liu
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Hongwei Yu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | - George W. Booz
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Richard J. Roman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Ruen Liu
- Department of Neurosurgery, Peking University People’s Hospital, Beijing, 100044, China
| | - Fan Fan
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
58
|
Wang H, Zhou G, Zhuang M, Wang W, Fu X. Utilizing network pharmacology and molecular docking to explore the underlying mechanism of Guizhi Fuling Wan in treating endometriosis. PeerJ 2021; 9:e11087. [PMID: 33859874 PMCID: PMC8020871 DOI: 10.7717/peerj.11087] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 02/19/2021] [Indexed: 12/12/2022] Open
Abstract
Background Guizhi Fuling Wan (GZFLW) is a widely used classical Chinese herbal formulae prescribed for the treatment of endometriosis (EMs). This study aimed to predict the key targets and mechanisms of GZFLW in the treatment of EMs by network pharmacology and molecular docking. Methods Firstly, related compounds and targets of GZFLW were identified through the TCMSP, BATMAN-TCM and CASC database. Then, the EMs target database was built by GeneCards. The overlapping targets between GZFLW and EMs were screened out, and then data of the PPI network was obtained by the STRING Database to analyze the interrelationship of these targets. Furthermore, a topological analysis was performed to screen the hub targets. After that, molecular docking technology was used to confirm the binding degree of the main active compounds and hub targets. Finally, the DAVID database and Metascape database were used for GO and KEGG enrichment analysis. Results A total of 89 GZFLW compounds and 284 targets were collected. One hundred one matching targets were picked out as the correlative targets of GZFLW in treating EMs. Among these, 25 significant hub targets were recognized by the PPI network. Coincidently, molecular docking simulation indicated that the hub targets had a good bonding activity with most active compounds (69.71%). Furthermore, 116 items, including the inflammatory reaction, RNA polymerase, DNA transcription, growth factor activity, and steroid-binding, were selected by GO enrichment analysis. Moreover, the KEGG enrichment analysis results included 100 pathways focused on the AGE-RAGE pathway, HIF pathway, PI3K Akt pathway, MAPK pathway, and TP53 pathway, which exposed the potential mechanisms of GZFLW in treating EMs. Also, the MTT colorimetric assay indicated that the cell proliferation could be inhibited by GZFLW. Compared with the control group, the protein levels of P53, BAX, and caspase3 in the drug groups were all increased in Western blotting results. The results of flow cytometry showed that the percentage of apoptotic cells in the GZFLW group was significantly higher than that in the control group. Conclusion Through the exploration of network pharmacology and molecular docking technology, GZFLW has a therapeutic effect on EMs through multi-target mechanism. This study provided a good foundation for further experimental research.
Collapse
Affiliation(s)
- Haoxian Wang
- Medical College, China Three Gorges University, Yichang, China
| | - Gang Zhou
- College of Traditional Chinese Medicine, Three Gorges University & Yichang Hospital of Traditional Chinese Medicine, Yichang, China
| | - Mingyan Zhuang
- Maternity and Child Health Care Hospital, Three Gorges University, Yichang, China
| | - Wei Wang
- College of Traditional Chinese Medicine, Three Gorges University & Yichang Hospital of Traditional Chinese Medicine, Yichang, China
| | - Xianyun Fu
- Medical College, China Three Gorges University, Yichang, China
| |
Collapse
|
59
|
Zhang Z, Yang W. Paeoniflorin protects PC12 cells from oxygen-glucose deprivation/reoxygenation-induced injury via activating JAK2/STAT3 signaling. Exp Ther Med 2021; 21:572. [PMID: 33850544 PMCID: PMC8027733 DOI: 10.3892/etm.2021.10004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 02/26/2021] [Indexed: 12/23/2022] Open
Abstract
Ischemic stroke is the most common type of stroke, and it has become a major health issue as it is characterized by high mortality and morbidity rates. Paeoniflorin (PF) is a natural compound and the main active ingredient of Radix Paeoniae. The aim of the present study was to investigate the role of PF in oxygen-glucose deprivation/reoxygenation (OGD/R)-induced injury of PC12 cells and its association with the Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) pathway. An in vitro model of OGD/R injury was established in PC12 cells. Subsequently, Cell Counting Kit-8 assay and ELISA were used to evaluate cell viability and the secretion of inflammatory factors, respectively, in PC12 cells subjected to OGD/R and treated with PF. The levels of oxidative stress indicators and inflammatory factors were measured using corresponding commercial kits. In addition, the apoptosis rate of PC12 cells subjected to OGD/R and treated with PF was determined by flow cytometry, and the expression of apoptosis-related proteins was analyzed by western blotting. Additionally, the expression levels of JAK2/STAT3 pathway-related proteins were also evaluated. The cell viability, levels of oxidative stress, inflammation and apoptosis were also measured in OGD/R-induced PC12 cell injury models following co-treatment of cells with PF and FLLL32, a specific inhibitor of JAK2/STAT3 signaling. Cell viability was reduced, while oxidative stress and inflammation were increased after OGD/R-induced injury. However, the treatment of cells with PF significantly enhanced cell viability, and alleviated oxidative stress, inflammation and apoptosis of OGD/R-treated PC12 cells. Furthermore, PF activated the JAK2/STAT3 signaling pathway. Following FLLL32 intervention, the effects of PF on oxidative stress, inflammation and apoptosis of OGD/R-treated PC12 cells were reversed. In conclusion, the findings of the present study suggested that PF may protect PC12 cells from OGD/R-induced injury via activating the JAK2/STAT3 signaling pathway, thus providing novel insight into the mechanism through which PF may alleviate ischemic stroke and indicating a potential strategy for ischemic stroke treatment.
Collapse
Affiliation(s)
- Zhuo Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Weimin Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| |
Collapse
|
60
|
Notoginsenoside R1 alleviates TEGDMA-induced mitochondrial apoptosis in preodontoblasts through activation of Akt/Nrf2 pathway-dependent mitophagy. Toxicol Appl Pharmacol 2021; 417:115482. [PMID: 33689844 DOI: 10.1016/j.taap.2021.115482] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/25/2021] [Accepted: 03/03/2021] [Indexed: 02/07/2023]
Abstract
Incomplete polymerization or biodegradation of dental resin materials results in the release of resin monomers such as triethylene glycol dimethacrylate (TEGDMA), causing severe injury of dental pulp cells. To date, there has been no efficient treatment option for this complication, in part due to the lack of understanding of the mechanism underlying these phenomena. Here, for the first time, we found that notoginsenoside R1 (NR1), a bioactive ingredient extracted from Panax notoginseng, exerted an obvious protective effect on TEGDMA-induced mitochondrial apoptosis in the preodontoblast mDPC6T cell line. In terms of the mechanism of action, NR1 enhanced the level of phosphorylated Akt (protein kinase B), resulting in the activation of a transcriptional factor, nuclear factor erythroid 2-related factor 2 (Nrf2), and eventually upregulating cellular ability to resist TEGDMA-related toxicity. Inhibiting the Akt/Nrf2 pathway by pharmaceutical inhibitors significantly decreased NR1-mediated cellular antioxidant properties and aggravated mitochondrial oxidative damage in TEGDMA-treated cells. Interestingly, NR1 also promoted mitophagy, which was identified as the potential downstream of the Akt/Nrf2 pathway. Blocking the Akt/Nrf2 pathway inhibited mitophagy and abolished the protection of NR1 on cells exposed to TEGDMA. In conclusion, these findings reveal that the activation of Akt/Nrf2 pathway-mediated mitophagy by NR1 might be a promising approach for preventing resin monomer-induced dental pulp injury.
Collapse
|
61
|
Wang BN, Wu CB, Chen ZM, Zheng PP, Liu YQ, Xiong J, Xu JY, Li PF, Mamun AA, Ye LB, Zheng ZL, Wu YQ, Xiao J, Wang J. DL-3-n-butylphthalide ameliorates diabetes-associated cognitive decline by enhancing PI3K/Akt signaling and suppressing oxidative stress. Acta Pharmacol Sin 2021; 42:347-360. [PMID: 33462377 PMCID: PMC8027654 DOI: 10.1038/s41401-020-00583-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 11/16/2020] [Indexed: 02/07/2023] Open
Abstract
DL-3-n-Butylphthalide (DL-NBP), a small molecular compound extracted from the seeds of Apium graveolens Linn (Chinese celery), has been shown to exert neuroprotective effects due to its anti-inflammatory, anti-oxidative and anti-apoptotic activities. DL-NBP not only protects against ischemic cerebral injury, but also ameliorates vascular cognitive impairment in dementia patients including AD and PD. In the current study, we investigated whether and how DL-NBP exerted a neuroprotective effect against diabetes-associated cognitive decline (DACD) in db/db mice, a model of type-2 diabetes. db/db mice were orally administered DL-NBP (20, 60, 120 mg· kg-1· d-1) for 8 weeks. Then the mice were subjected to behavioral test, their brain tissue was collected for morphological and biochemical analyses. We showed that oral administration of DL-NBP significantly ameliorated the cognitive decline with improved learning and memory function in Morris water maze testing. Furthermore, DL-NBP administration attenuated diabetes-induced morphological alterations and increased neuronal survival and restored the levels of synaptic protein PSD95, synaptophysin and synapsin-1 as well as dendritic density in the hippocampus, especially at a dose of 60 mg/kg. Moreover, we revealed that DL-NBP administration suppressed oxidative stress by upregulating Nrf2/HO-1 signaling, and increased brain-derived neurotrophic factor (BDNF) expression by activating PI3K/Akt/CREB signaling in the hippocampus. These beneficial effects of DL-NBP were observed in high glucose-treated PC12 cells. Our results suggest that DL-NBP may be a potential pharmacologic agent for the treatment of DACD.
Collapse
Affiliation(s)
- Bei-Ni Wang
- Department of Hand Surgery and Peripheral Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Cheng-Biao Wu
- Research Center, Affiliated Xiangshan Hospital, Wenzhou Medical University, Ningbo, 315700, China
| | - Zi-Miao Chen
- Department of Hand Surgery and Peripheral Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Pei-Pei Zheng
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Ya-Qian Liu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Jun Xiong
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Jing-Yu Xu
- The Institute of Life Sciences, Engineering Laboratory of Zhejiang province for Pharmaceutical Development of Growth Factors, Biomedical Collaborative Innovation Center of Wenzhou, Wenzhou University, Wenzhou, 325035, China
| | - Pei-Feng Li
- Department of Hand Surgery and Peripheral Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Abdullah Al Mamun
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Li-Bing Ye
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Zhi-Long Zheng
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Yan-Qing Wu
- The Institute of Life Sciences, Engineering Laboratory of Zhejiang province for Pharmaceutical Development of Growth Factors, Biomedical Collaborative Innovation Center of Wenzhou, Wenzhou University, Wenzhou, 325035, China.
| | - Jian Xiao
- Department of Hand Surgery and Peripheral Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, China.
| | - Jian Wang
- Department of Hand Surgery and Peripheral Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
62
|
Wang Y, Liu F, Liu P. 23-Hydroxytormentic acid reduces cerebral ischemia/reperfusion damage in rats through anti-apoptotic, antioxidant, and anti-inflammatory mechanisms. Naunyn Schmiedebergs Arch Pharmacol 2021; 394:1045-1054. [PMID: 33394135 DOI: 10.1007/s00210-020-02038-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 12/08/2020] [Indexed: 11/25/2022]
Abstract
23-Hydroxytormentic acid (23-HTA) is an important herbal medicine purified from immature fruits of African Rubus aceae (Rosaceae). This study was carried out to examine the protection properties and potential mechanisms of 23-HTA against cerebral ischemia/reperfusion (I/R) damage. Rats underwent middle cerebral artery occlusion/reperfusion (MCAO/R) 2/24 h. All animals were euthanized 24 h after reperfusion. Rats were injected with various concentrations of 23-HTA intraperitoneally. Evaluations of infarct volumes, neurological deficit, and brain water contents were carried out to assess the outcome of 23-HTA treatment. The results showed that 23-HTA reduced infarct volumes, brain water content, and neurological deficit in a dosage-dependent manner. 23-HTA can also significantly reduce the numbers of TUNEL-positive cells, the expression levels of Bax, caspase-3, lipid peroxidation, Sod 1, Sod 2, catalase, and pro-inflammatory cytokines TNF and IL-1β and increase the expression levels of Bcl-2 and p-Akt. 23-HTA has a neuroprotective effect due to its anti-apoptotic, antioxidant, and anti-inflammatory effects.
Collapse
Affiliation(s)
- Yamin Wang
- Department of Neurology, The 80th Army Hospital of the Chinese People's Liberation Army, No.256 Beigong West Street, Weicheng District, Weifang, 261041, Shandong, China
| | - Fengrong Liu
- Department of Neurology, The 80th Army Hospital of the Chinese People's Liberation Army, No.256 Beigong West Street, Weicheng District, Weifang, 261041, Shandong, China
| | - Peng Liu
- Department of Neurology, The 80th Army Hospital of the Chinese People's Liberation Army, No.256 Beigong West Street, Weicheng District, Weifang, 261041, Shandong, China.
| |
Collapse
|
63
|
Abstract
Bcl-xL is a pro-survival protein of the Bcl2 family found in the mitochondrial membrane. Bcl-xL supports growth, development, and maturation of neurons, and it also prevents neuronal death during neurotoxic stimulation. This article reviews the mechanisms and upstream signaling that regulate the activity and abundance of Bcl-xL. Our team and others have reported that oxidative stress is a key regulator of intracellular Bcl-xL balance in neurons. Oxidative stress regulates synthesis, degradation, and activity of Bcl-xL and therefore neuronal function. During apoptosis, pro-apoptotic Bcl2 proteins such as Bax and Bak translocate to and oligomerize in the mitochondrial membrane. Formation of oligomers causes release of cytochrome c and activation of caspases that lead to neuronal death. Bcl-xL binds directly to pro-apoptotic Bcl2 proteins to block apoptotic signaling. Although anti-apoptotic roles of Bcl-xL have been well documented, an increasing number of studies in recent decades show that protein binding partners of Bcl-xL are not limited to Bcl2 proteins. Bcl-xL forms a complex with F1Fo ATP synthase, DJ-1, DRP1, IP3R, and the ryanodine receptor. These proteins support physiological processes in neurons such as growth and development and prevent neuronal damage by regulating mitochondrial ATP production, synapse formation, synaptic vesicle recycling, neurotransmission, and calcium signaling. However, under conditions of oxidative stress, Bcl-xL undergoes proteolytic cleavage thus lowering the abundance of functional Bcl-xL in neurons. Additionally, oxidative stress alters formation of Bcl-xL-mediated multiprotein complexes by regulating post-translational phosphorylation. Finally, oxidative stress regulates transcription factors that target the Bcl-x gene and alter accessibility of microRNA to mRNA influencing mRNA levels of Bcl-xL. In this review, we discussed how Bcl-xL supports the normal physiology of neurons, and how oxidative stress contributes to pathology by manipulating the dynamics of Bcl-xL production, degradation, and activity.
Collapse
Affiliation(s)
- Han-A Park
- Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, Tuscaloosa, AL, USA
| | - Katheryn Broman
- Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, Tuscaloosa, AL, USA
| | - Elizabeth A Jonas
- Department of Internal Medicine, Section of Endocrinology, Yale University, New Haven, CT, USA
| |
Collapse
|
64
|
Zhang B, Shen J, Zhong Z, Zhang L. PKM2 Aggravates Cerebral Ischemia Reperfusion-Induced Neuroinflammation via TLR4/MyD88/TRAF6 Signaling Pathway. Neuroimmunomodulation 2021; 28:29-37. [PMID: 33744886 DOI: 10.1159/000509710] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 06/24/2020] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES Cerebral ischemia-reperfusion (I/R) injury is the leading cause of ischemic stroke. Pyruvate Kinase isozymes M2 (PKM2), as a critical glycolytic enzyme during glycolysis, is involved in neuronal apoptosis in rats with hypoxic-ischemic encephalopathy. This study focused on functional investigation and potential molecular mechanism toward PKM2 in cerebral I/R injury. METHODS Cerebral I/R injury model was established by middle cerebral artery occlusion (MCAO) in vivo or oxygen-glucose deprivation and reoxygenation (OGD/R) in vitro. qRT-PCR and Western blot were used to detect the expression of PKM2 in I/R injury models. The effects of PKM2 on I/R injury were determined via triphenyl tetrazolium chloride staining and evaluation of neurological deficits. Cell Counting Kit-8 was employed to detect cell viability, and ELISA was conducted to detect pro-inflammatory cytokines. The underlying mechanism involved in regulation of PKM2 on I/R injury was investigated via ELISA and Western blot. RESULTS PKM2 was upregulated after cerebral I/R injury. Knockdown of PKM2 alleviated MCAO-induced infarction and neurological dysfunction. Moreover, PKM2 knockdown also alleviated OGD/R-induced neuronal cell injury and inflammatory response. Mechanistically, PKM2 knockdown-induced neuroprotection was accompanied by inhibition of high-mobility group box 1 (HMGB1), reflected by inactivation of TLR4/MyD88 (myeloid differentiation factor 88)/TRAF6 (TNF receptor-associated factor 6) signaling pathway. CONCLUSIONS Knockdown of PKM2 attenuated cerebral I/R injury through HMGB1-mediated TLR4/MyD88/TRAF6 expression change, providing a potential target for cerebral I/R injury treatment.
Collapse
Affiliation(s)
- Baocheng Zhang
- Department of Intensive Care Unit, Jinshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Jie Shen
- Department of Intensive Care Unit, Jinshan Hospital Affiliated to Fudan University, Shanghai, China,
| | - Zhiyue Zhong
- Department of Intensive Care Unit, Jinshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Lin Zhang
- Department of Intensive Care Unit, Jinshan Hospital Affiliated to Fudan University, Shanghai, China
| |
Collapse
|
65
|
Liu W, Xie G, Yuan G, Xie D, Lian Z, Lin Z, Ye J, Zhou W, Zhou W, Li H, Wang X, Feng H, Liu Y, Yao G. 6'-O-Galloylpaeoniflorin Attenuates Osteoclasto-genesis and Relieves Ovariectomy-Induced Osteoporosis by Inhibiting Reactive Oxygen Species and MAPKs/c-Fos/NFATc1 Signaling Pathway. Front Pharmacol 2021; 12:641277. [PMID: 33897430 PMCID: PMC8058459 DOI: 10.3389/fphar.2021.641277] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/19/2021] [Indexed: 02/05/2023] Open
Abstract
Emerging evidence suggests bright prospects of some natural antioxidants in the treatment of osteoporosis. 6'-O-Galloylpaeoniflorin (GPF), an antioxidant isolated from peony roots (one of very widely used Oriental medicines, with various anti-inflammatory, antitumor, and antioxidant activities), shows a series of potential clinical applications. However, its effects on osteoporosis remain poorly investigated. The current study aimed to explore whether GPF can attenuate osteoclastogenesis and relieve ovariectomy-induced osteoporosis via attenuating reactive oxygen species (ROS), and investigate the possible mechanism. After the culture of primary murine bone marrow-derived macrophages/monocytes were induced by the use of macrophage colony-stimulating factor (M-CSF) and the receptor activator of NF-κB ligand (RANKL) and then treated with GPF. Cell proliferation and viability were assessed by Cell Counting Kit-8 (CCK-8) assay. Thereafter, the role of GPF in the production of osteoclasts and the osteogenic resorption of mature osteoclasts were evaluated by tartrate-resistant acid phosphatase (TRAP) staining, podosome belt formation, and resorption pit assay. Western blotting and qRT-PCR examination were performed to evaluate proteins' generation and osteoclast-specific gene levels, respectively. The ROS generation in cells was measured in vitro by 2',7'-Dichlorodi-hydrofluorescein diacetate (DCFH-DA). Ovariectomy-induced osteoporosis mouse administered with GPF or vehicle was performed to explore the in vivo potential of GPF, then a micro-CT scan was performed in combination with histological examination for further analysis. GPF suppressed the formation of osteoclasts and podosome belts, as well as bone resorption when induced by RANKL through affecting intracellular ROS activity, MAPKs signaling pathway, and subsequent NFATc1 translocation and expression, as well as osteoclast-specific gene expression in vitro. In vivo study suggested that exposure to GPF prevented osteoporosis-related bone loss in the ovariectomized mice. These findings indicate that GPF attenuates osteoclastogenesis and relieves ovariectomy-induced osteoporosis by inhibiting ROS and MAPKs/c-Fos/NFATc1 signaling pathway. This suggested that GPF may be potentially used to treat bone diseases like periodontitis, rheumatoid arthritis, and osteoporosis associated with osteoclasts.
Collapse
Affiliation(s)
- Wenjie Liu
- Department of Orthopedics, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Gang Xie
- Department of Orthopedics, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Guixin Yuan
- Department of Orthopedics, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Dantao Xie
- Department of Orthopedics, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Zhen Lian
- Department of Orthopedics, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Zihong Lin
- Department of Orthopedics, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Jiajie Ye
- Department of Orthopedics, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Wenyun Zhou
- Department of Orthopedics, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Weijun Zhou
- Department of Orthopedics, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Henghui Li
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Xinjia Wang
- Department of Orthopedics, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Haotian Feng
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
- Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning, China
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Ying Liu
- Institute of Translational Medicine, Shanghai University, Shanghai, China
- *Correspondence: Ying Liu, ; Guanfeng Yao,
| | - Guanfeng Yao
- Department of Orthopedics, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
- *Correspondence: Ying Liu, ; Guanfeng Yao,
| |
Collapse
|
66
|
Li J, Yang C, Wang Y. miR‑126 overexpression attenuates oxygen‑glucose deprivation/reperfusion injury by inhibiting oxidative stress and inflammatory response via the activation of SIRT1/Nrf2 signaling pathway in human umbilical vein endothelial cells. Mol Med Rep 2020; 23:165. [PMID: 33355373 PMCID: PMC7789090 DOI: 10.3892/mmr.2020.11804] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 09/09/2020] [Indexed: 12/23/2022] Open
Abstract
MicroRNA‑126 (miR‑126) has been reported to be implicated in the pathogenesis of cerebral ischemia/reperfusion (I/R) injury; however, its role is still unclear and requires further investigation. The objective of the present study was to determine the neuroprotective effect of miR‑126 overexpression against oxygen‑glucose deprivation/reoxygenation (OGD/R)‑induced human umbilical vein endothelial cell (HUVEC) injury, an in vitro model of cerebral I/R injury, and to further explore the role of the NAD‑dependent protein deacetylase sirtuin‑1 (SIRT1)/nuclear factor erythroid 2‑related factor 2 (Nrf2) signaling pathway in this process. The results of the present study revealed that miR‑126 expression was markedly reduced in HUVECs subjected to OGD/R treatment. Functional experiments demonstrated that transfection with miR‑126 mimics attenuated OGD/R‑induced down‑regulation of cell viability, and reversed OGD/R‑induced up‑regulation of lactate dehydrogenase release, apoptosis and caspase‑3 activity in HUVECs. Notably, OGD/R reduced SIRT1 and heme oxygenase‑1 expression, and induced the nuclear translocation of Nrf2, as demonstrated by the increase in cytoplasmic Nrf2 expression and the decrease in nuclear Nrf2 expression. Following transfection with miR‑126 mimics, these effects of OGD/R were reversed, indicating that miR‑126 overexpression promoted the SIRT1/Nrf2 signaling pathway. Additionally, miR‑126 mimics attenuated OGD/R‑induced cytotoxicity and apoptosis, which was blocked by inhibition of the SIRT1/Nrf2 signaling pathway followed by transfection with SIRT1‑small interfering RNA (siRNA). Furthermore, miR‑126 mimics decreased ROS generation and malondialdehyde content, and increased superoxide dismutase and glutathione peroxidase activity in HUVECs exposed to OGD/R, and these effects of miR‑126 mimics were also blocked by SIRT1‑siRNA. Additionally, the miR‑126 mimics‑induced the decreases in the levels of pro‑inflammatory cytokines, including tumor necrosis factor‑α, interleukin (IL)‑1β and IL‑6, and the miR‑126 mimics‑induced increase in anti‑inflammatory cytokines, including IL‑10, were reversed by SIRT1‑siRNA. Overall, these results suggested that miR‑126 overexpression attenuated OGD/R‑induced neurotoxicity to HUVECs by alleviating oxidative stress and the inflammatory response via promotion of the SIRT1/Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Jixin Li
- Department of Neurology, The First People's Hospital, Taizhou, Zhejiang 317000, P.R. China
| | - Caili Yang
- Department of Neurology, The First People's Hospital, Taizhou, Zhejiang 317000, P.R. China
| | - Yan Wang
- Department of Neurology, The Second People's Hospital, Taizhou, Zhejiang 317016, P.R. China
| |
Collapse
|
67
|
Chen X, Zhang J, Xia L, Wang L, Li H, Liu H, Zhou J, Feng Z, Jin H, Yang J, Yang Y, Wu B, Zhang L, Chen G, Wang G. β-Arrestin-2 attenuates hepatic ischemia-reperfusion injury by activating PI3K/Akt signaling. Aging (Albany NY) 2020; 13:2251-2263. [PMID: 33323551 PMCID: PMC7880335 DOI: 10.18632/aging.202246] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 09/24/2020] [Indexed: 12/12/2022]
Abstract
Hepatic ischemia-reperfusion injury (IRI) remains a common complication during liver transplantation (LT), partial hepatectomy and hemorrhagic shock in patients. As a member of the G protein-coupled receptors adaptors, ARRB2 has been reported to be involved in a variety of physiological and pathological processes. However, whether β-arrestin-2 affects the pathogenesis of hepatic IRI remains unknown. The goal of the present study was to determine whether ARRB2 protects against hepatic IR injury and elucidate the underlying mechanisms. To this end, 70% hepatic IR models were established in ARRB2 knockdown mice and wild-type littermates, with blood and liver samples collected at 1, 6 and 12 h after reperfusion to evaluate liver injury. The effect of ARBB2 on PI3K/Akt signaling during IR injury was evaluated in vivo, and PI3K/Akt pathway regulation by ARRB2 was further assessed in vitro. Our results showed that ARRB2 knockdown aggravates hepatic IR injury by promoting the apoptosis of hepatocytes and inhibiting their proliferation. In addition, ARRB2 deficiency inhibited PI3K/Akt pathway activation, while the administration of the PI3K/Akt inhibitor PX866 resulted in severe IR injury in mice. Furthermore, the liver-protecting effect of ARRB2 was shown to depend on PI3K/Akt pathway activation. In summary, our results suggest that β-Arrestin-2 protects against hepatic IRI by activating PI3K/Akt signaling, which may provide a novel therapeutic strategy for treating liver ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Xiaolong Chen
- Department of Hepatic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, Guangdong Province, P. R. China.,Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, Guangdong Province, P. R. China
| | - Junbin Zhang
- Department of Hepatic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, Guangdong Province, P. R. China.,Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, Guangdong Province, P. R. China
| | - Long Xia
- Department of Hepatic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, Guangdong Province, P. R. China.,Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, Guangdong Province, P. R. China
| | - Li Wang
- Department of Hepatic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, Guangdong Province, P. R. China.,Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, Guangdong Province, P. R. China
| | - Hui Li
- Department of Hepatic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, Guangdong Province, P. R. China.,Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, Guangdong Province, P. R. China
| | - Huilin Liu
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, Guangdong Province, P. R. China.,Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, Guangdong Province, P. R. China
| | - Jing Zhou
- Department of Pathology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, Guangdong Province, P. R. China
| | - Zhiying Feng
- Department of Pathology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, Guangdong Province, P. R. China
| | - Hai Jin
- Department of Medical Ultrasonics, Guangzhou First People's Hospital, The Second Affiliated Hospital of South China University of Technology, Guangzhou 510630, Guangdong Province, P. R. China
| | - JianXu Yang
- Department of Intensive Care Unit, Henan Provincial People's Hospital, Zhengzhou 450003, Henan Province, P. R. China
| | - Yang Yang
- Department of Hepatic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, Guangdong Province, P. R. China.,Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, Guangdong Province, P. R. China
| | - Bin Wu
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, Guangdong Province, P. R. China.,Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, Guangdong Province, P. R. China
| | - Lei Zhang
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, Guangdong Province, P. R. China.,Department of Biliary-Pancreatic Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, Guangdong Province, P. R. China
| | - Guihua Chen
- Department of Hepatic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, Guangdong Province, P. R. China.,Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, Guangdong Province, P. R. China
| | - Genshu Wang
- Department of Hepatic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, Guangdong Province, P. R. China.,Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, Guangdong Province, P. R. China
| |
Collapse
|
68
|
Wang M, Li Q, Zhang Y, Liu H. Total Glucosides of Peony Protect Cardiomyocytes against Oxidative Stress and Inflammation by Reversing Mitochondrial Dynamics and Bioenergetics. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6632413. [PMID: 33354278 PMCID: PMC7735829 DOI: 10.1155/2020/6632413] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/30/2020] [Accepted: 11/13/2020] [Indexed: 01/14/2023]
Abstract
Total glucosides of peony (TGP) are used to treat rheumatoid arthritis and systemic lupus erythematosus. We explored the protective effects of TGP on cardiomyocyte oxidative stress and inflammation in the presence of hydrogen peroxide by focusing on mitochondrial dynamics and bioenergetics. Our study demonstrated that hydrogen peroxide significantly repressed cardiomyocyte viability and promoted cell apoptosis through induction of the mitochondrial death pathway. TGP treatment sustained cardiomyocyte viability, reduced cardiomyocyte apoptosis, and decreased inflammation and oxidative stress. Molecular investigation indicated that hydrogen peroxide caused mitochondrial dynamics disruption and bioenergetics reduction in cardiomyocytes, but this alteration could be normalized by TGP. We found that disruption of mitochondrial dynamics abolished the regulatory effects of TGP on mitochondrial bioenergetics; TGP modulated mitochondrial dynamics through the AMP-activated protein kinase (AMPK) pathway; and inhibition of AMPK alleviated the protective effects of TGP on mitochondria. Our results showed that TGP treatment reduces cardiomyocyte oxidative stress and inflammation in the presence of hydrogen peroxide by correcting mitochondrial dynamics and enhancing mitochondrial bioenergetics. Additionally, the regulatory effects of TGP on mitochondrial function seem to be mediated through the AMPK pathway. These findings are promising for myocardial injury in patients with rheumatoid arthritis and systemic lupus erythematosus.
Collapse
Affiliation(s)
- Mengmeng Wang
- Department of Rheumatism and Immunology, Tianjin First Central hospital, Tianjin, China
| | - Qiang Li
- Department of Pharmacy, Tianjin Union Medical Center, Tianjin, China
| | - Ying Zhang
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Hao Liu
- Department of Pharmacy, Nankai University, Tianjin, China
| |
Collapse
|
69
|
Shen S, Ma L, Shao F, Jin L, Bian Z. Long Non-Coding RNA (lncRNA) NEAT1 Aggravates Cerebral Ischemia-Reperfusion Injury by Suppressing the Inhibitory Effect of miR-214 on PTEN. Med Sci Monit 2020; 26:e924781. [PMID: 32815529 PMCID: PMC7453753 DOI: 10.12659/msm.924781] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Background Cerebral ischemia-reperfusion injury is a form of serious nervous system injury. Activation of the PI3K/Akt pathway can effectively relieve cerebral ischemia-reperfusion injury. miR-214 can target and inhibit the expression of PTEN, thereby alleviating its inhibitory effect on the PI3K/Akt pathway. Moreover, lncRNA NEAT1 was reported to affect proliferation and metastasis of tumor cells by targeting and suppressing the expression of miR-214. However, whether lncRNA NEAT1 affects the cerebral ischemia-reperfusion-induced damage by regulating the miR-214/PTEN/PI3K/Akt pathway is unclear. Material/Methods The miR-214 agomir and miR-214 antagomir were designed and injected into the encephalocele of MCAO rats. Next, the production of oxidative stress kinase and apoptosis of brain cells were detected using commercial kits. The levels of PTEN, PI3K, Akt, p-Akt, and VEGF in brain tissues were determined. Next, the targeting effect of lncRNA NEAT1 and miR-214 was determined with luciferase reporter assay. Results Overexpression of miR-214 relieved the apoptosis and oxidative stress of brain tissues. Overexpression of miR-214 promoted the expression of PI3K, Akt, p-Akt, and VEGF by inhibiting the production of PTEN. However, overexpression of lncRNA NEAT1 repressed the remission effect of miR-214 on cerebral ischemia-reperfusion-induced damage and inhibited the production of PI3K, Akt, p-Akt, and VEGF by rescuing the levels of PTEN. Conclusions lncRNA NEAT1 aggravates cerebral ischemia-reperfusion injury by abolishing the activation effect of miR-214 on the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Shouyin Shen
- Department of Geriatrics, The Third People's Hospital of Nantong City, Nantong, Jiangsu, China (mainland)
| | - Liang Ma
- Department of Geriatrics, The Third People's Hospital of Nantong City, Nantong, Jiangsu, China (mainland)
| | - Feng Shao
- Department of Emergency, The Third People's Hospital of Nantong City, Nantong, Jiangsu, China (mainland)
| | - Li Jin
- Department of Emergency, The Third People's Hospital of Nantong City, Nantong, Jiangsu, China (mainland)
| | - Zhaolian Bian
- Department of Gastroenterology, The Third People's Hospital of Nantong City, Nantong, Jiangsu, China (mainland)
| |
Collapse
|
70
|
Tang C, Hu Y, Lyu H, Gao J, Jiang J, Qin X, Wu Y, Wang J, Chai X. Neuroprotective effects of 1-O-hexyl-2,3,5-trimethylhydroquinone on ischaemia/reperfusion-induced neuronal injury by activating the Nrf2/HO-1 pathway. J Cell Mol Med 2020; 24:10468-10477. [PMID: 32677362 PMCID: PMC7521305 DOI: 10.1111/jcmm.15659] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 04/29/2020] [Accepted: 06/29/2020] [Indexed: 01/01/2023] Open
Abstract
1-O-Hexyl-2,3,5-trimethylhydroquinone (HTHQ), a lipophilic phenolic agent, has an antioxidant activity and reactive oxygen species (ROS) scavenging property. However, the role of HTHQ on cerebral ischaemic/reperfusion (I/R) injury and the underlying mechanisms remain poorly understood. In the present study, we demonstrated that HTHQ treatment ameliorated cerebral I/R injury in vivo, as demonstrated by the decreased infarct volume ration, neurological deficits, oxidative stress and neuronal apoptosis. HTHQ treatment increased the levels of nuclear factor erythroid 2-related factor 2 (Nrf2) and its downstream antioxidant protein, haeme oxygenase-1 (HO-1). In addition, HTHQ treatment decreases oxidative stress and neuronal apoptosis of PC12 cells following hypoxia and reperfusion (H/R) in vitro. Moreover, we provided evidence that PC12 cells were more vulnerable to H/R-induced oxidative stress after si-Nrf2 transfection, and the HTHQ-mediated protection was lost in PC12 cells transfected with siNrf2. In conclusion, these results suggested that HTHQ possesses neuroprotective effects against oxidative stress and apoptosis after cerebral I/R injury via activation of the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Chaoliang Tang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yida Hu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Haiyan Lyu
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jie Gao
- Department of Anesthesia, Critical Care & Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jiazhen Jiang
- Department of Emergency, Huashan Hospital North, Fudan University, Shanghai, China
| | - Xiude Qin
- Department of Neurology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Yuanbo Wu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jiawu Wang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xiaoqing Chai
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
71
|
Hannan MA, Dash R, Sohag AAM, Haque MN, Moon IS. Neuroprotection Against Oxidative Stress: Phytochemicals Targeting TrkB Signaling and the Nrf2-ARE Antioxidant System. Front Mol Neurosci 2020; 13:116. [PMID: 32714148 PMCID: PMC7346762 DOI: 10.3389/fnmol.2020.00116] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 06/04/2020] [Indexed: 12/14/2022] Open
Abstract
Oxidative stress (OS) plays a critical role in the pathophysiology of several brain-related disorders, including neurodegenerative diseases and ischemic stroke, which are the major causes of dementia. The Nrf2-ARE (nuclear factor erythroid 2-related factor 2/antioxidant responsive element antioxidant) system, the primary cellular defense against OS, plays an essential role in neuroprotection by regulating the expressions of antioxidant molecules and enzymes. However, simultaneous events resulting in the overproduction of reactive oxygen species (ROS) and deregulation of the Nrf2-ARE system damage essential cell components and cause loss of neuron structural and functional integrity. On the other hand, TrkB (tropomyosin-related kinase B) signaling, a classical neurotrophin signaling pathway, regulates neuronal survival and synaptic plasticity, which play pivotal roles in memory and cognition. Also, TrkB signaling, specifically the TrkB/PI3K/Akt (TrkB/phosphatidylinositol 3 kinase/protein kinase B) pathway promotes the activation and nuclear translocation of Nrf2, and thus, confers neuroprotection against OS. However, the TrkB signaling pathway is also known to be downregulated in brain disorders due to lack of neurotrophin support. Therefore, activations of TrkB and the Nrf2-ARE signaling system offer a potential approach to the design of novel therapeutic agents for brain disorders. Here, we briefly overview the development of OS and the association between OS and the pathogenesis of neurodegenerative diseases and brain injury. We propose the cellular antioxidant defense and TrkB signaling-mediated cell survival systems be considered pharmacological targets for the treatment of neurodegenerative diseases, and review the literature on the neuroprotective effects of phytochemicals that can co-activate these neuronal defense systems.
Collapse
Affiliation(s)
- Md. Abdul Hannan
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju, South Korea
- Department of Biochemistry and Molecular Biology, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Raju Dash
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju, South Korea
| | - Abdullah Al Mamun Sohag
- Department of Biochemistry and Molecular Biology, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Md. Nazmul Haque
- Department of Fisheries Biology and Genetics, Patuakhali Science and Technology University, Patuakhali, Bangladesh
| | - Il Soo Moon
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju, South Korea
| |
Collapse
|
72
|
Zeng Z, Liu HM, Zhang YY, Chen R, Sun T, Li W, Sun Q, Xia ZY, Meng QT. Aggravated intestinal ischemia‑reperfusion injury is associated with activated mitochondrial autophagy in a mouse model of diabetes. Mol Med Rep 2020; 22:1892-1900. [PMID: 32582983 PMCID: PMC7411361 DOI: 10.3892/mmr.2020.11270] [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: 08/14/2019] [Accepted: 04/06/2020] [Indexed: 01/19/2023] Open
Abstract
Numerous studies have reported that diabetes is associated with an increased susceptibility to cardiac ischemia- reperfusion injury; however, the mechanism underlying the role of diabetes during intestinal ischemia-reperfusion (IIR) has yet to be elucidated. The present study evaluated the intestinal pathological alterations and possible underlying mechanisms in a mouse model of type 1 diabetes mellitus with IIR. The effects of diabetes were investigated by assessing the histopathology, oxidative stress, inflammatory cytokine levels in intestine tissues and blood plasma, and protein expression levels of phosphatase and tensin homolog-induced putative kinase (PINK1), Parkin and the ratio of light chain 3B (LC3B) II/I. The results demonstrated that diabetes increased the Chiu's intestinal injury score, concentration of interleukin (IL)-1β, IL-6 and tumor necrosis factor (TNF)-α, and levels of oxidative stress. Furthermore, the alterations were more pronounced in the diabetes with IIR group. The expression levels of PINK1 and Parkin, as well as the ratio of LC3BII/I, were significantly upregulated in the IIR group compared with the Sham group. Diabetes activated PINK1 and Parkin, and increased the expression of LC3BII. Furthermore, transmission electron microscopy revealed that mitochondrial destruction and the number of autophagosomes was increased in the diabetic groups compared with the non-diabetic groups. Collectively, the results of the present study suggest that diabetes increased intestinal vulnerability to IIR by enhancing inflammation and oxidative stress. Furthermore, IIR was associated with overactivation of mitochondrial autophagy; therefore, the increased vulnerability to IIR-induced intestine damage due to diabetes may be associated with PINK1/Parkin-regulated mitochondrial autophagy.
Collapse
Affiliation(s)
- Zi Zeng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Hui-Min Liu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yun-Yan Zhang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Rong Chen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Tao Sun
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Wei Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qian Sun
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Zhong-Yuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qing-Tao Meng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
73
|
Effects and Mechanisms of Five Psoralea Prenylflavonoids on Aging-Related Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2128513. [PMID: 32655760 PMCID: PMC7320294 DOI: 10.1155/2020/2128513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 05/12/2020] [Accepted: 05/28/2020] [Indexed: 01/09/2023]
Abstract
During the aging process, senescent cells gradually accumulate in the organs; they secrete proinflammatory cytokines and other factors, collectively known as the senescence-associated secretory phenotype (SASP). SASP secretions contribute to “inflammaging,” which is a state of chronic, systemic, sterility, low-grade inflammatory microenvironment and a key risk factor in the development of aging-related diseases. Fructus psoraleae is a traditional Chinese medical herb best known for delaying aging and treating osteoporosis. Prenylflavonoids from fructus psoraleae are the main bioactive compounds responsible for its pharmacological applications, such as beaching, bavachinin, bavachalcone, isobavachalcone, and neobavaisoflavone. In previous decades, there have been some promising studies on the pharmacology of fructus psoraleae. Here, we focus on the anti-inflammatory and antiaging diseases of five psoralea prenylflavonoids, such as cardiovascular protection, diabetes and obesity intervention, neuroprotection, and osteoporosis, and discuss the mechanism of these active ingredients for better understanding the material basis and drug application of fructus psoraleae in Chinese medicine.
Collapse
|
74
|
Gentiopicroside Ameliorates Oxidative Stress and Lipid Accumulation through Nuclear Factor Erythroid 2-Related Factor 2 Activation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2940746. [PMID: 32655764 PMCID: PMC7317617 DOI: 10.1155/2020/2940746] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 04/10/2020] [Indexed: 12/15/2022]
Abstract
The activation of nuclear factor erythroid 2-related factor 2 (Nrf2) is closely related to the alleviation of nonalcoholic fatty liver disease (NAFLD) by regulating oxidative stress and lipid homeostasis. Gentiopicroside (GPS), an iridoid glycoside found in the Gentianaceae, possesses anti-inflammatory and antioxidant effects. However, the protective effects of GPS on lipid accumulation and oxidative damage have not been investigated thoroughly in free fatty acid- (FFA-) induced HepG2 cells and tyloxapol- (Ty-) induced hyperlipidemia mice. Cell counting kit-8 assays, Oil Red O staining, Western blotting analysis, extraction of nuclear and cytosolic proteins, and biochemical index assay were employed to explore the mechanisms by which GPS exerts a protective effect on FFA-induced HepG2 cells and Ty-induced hyperlipidemia mouse model. This paper demonstrates that GPS could effectively alleviate NAFLD by elevating cell viability, reducing fatty deposition, downregulating TG, and activating nucleus Nrf2 in FFA-induced HepG2 cells. Meanwhile, GPS significantly regulated the activation of phosphatidylinositol 3-kinase (PI3K)/AKT signaling pathway, Nrf2 antioxidant pathway, peroxisome proliferator-activated receptor α (PPARα), and GPS-inhibited sterol regulatory element-binding protein-1c (SREBP-1c) expression in FFA-stimulated lipid accumulation of HepG2 cells and Ty-treated mice. Interestingly, we highlight that PI3K/AKT inhibitor (LY294002) markedly increased the expression of Nrf2 antioxidant pathway, PPARα, and downregulated SREBP-1c in FFA-stimulated HepG2 cells. For these reasons, we found that the deletion of Nrf2 could lose the protective effects of GPS on the Nrf2 antioxidant pathway and PPARα activation and SREBP-1c inactivation in FFA-stimulated HepG2 cells and Ty-treated mice. GPS treatment had no effect on abnormal lipogenesis and antioxidant enzymes in Ty-induced Nrf2−/− mice. This work gives a new explanation that GPS may be a useful therapeutic strategy for NAFLD through upregulation of the Nrf2 antioxidant pathway, which can alleviate oxidative damage and lipid accumulation.
Collapse
|
75
|
Wang YY, Chang CY, Lin SY, Wang JD, Wu CC, Chen WY, Kuan YH, Liao SL, Wang WY, Chen CJ. Quercetin protects against cerebral ischemia/reperfusion and oxygen glucose deprivation/reoxygenation neurotoxicity. J Nutr Biochem 2020; 83:108436. [PMID: 32599520 DOI: 10.1016/j.jnutbio.2020.108436] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 03/27/2020] [Accepted: 05/20/2020] [Indexed: 01/01/2023]
Abstract
Beyond nutrition effect, quercetin is applied as a complement or an alternative for promoting human health and treating diseases. However, its complicated neuroprotective mechanisms have not yet been fully elucidated. This study provides evidence of an alternative target for quercetin, and sheds light on the mechanisms of its neuroprotection against cerebral ischemia/reperfusion (I/R) injury in Sprague-Dawley rats. Oral pretreatment using quercetin has alleviated cerebral I/R-induced neurological deficits, brain infarction, blood-brain barrier disruption, oxidative stress, TNF-α and IL-1β mRNA expression, along with apoptotic caspase 3 activity. The neuroprotective, anti-oxidative, anti-inflammatory, and anti-apoptotic effects of quercetin were replicated in rat hippocampal slice cultures and neuron/glia cultures which suffered from oxygen-glucose deprivation and reoxygenation (OGDR). Biochemical studies revealed a reduction of extracellular signal-regulated kinase (ERK) and Akt phosphorylation, along with an increase in protein tyrosine and serine/threonine phosphatase activity in cerebral I/R rat cortical tissues and OGDR hippocampal slice and neuron/glia cultures. Quercetin alleviated the changes in ERK/Akt phosphorylation and protein phosphatase activities. Inhibition of ERK or Akt alone was enough to cause apoptotic cell death and cytotoxicity in hippocampal slice cultures and neuron/glia cultures, while activators of ERK or Akt alleviated OGDR-induced cytotoxicity. Taken together, our results demonstrate that quercetin alleviated the increment of protein tyrosine and serine/threonine phosphatase activity, along with the reduction of ERK and Akt phosphorylation, which may play pivotal roles in the expansion of brain injury after cerebral I/R.
Collapse
Affiliation(s)
- Ya-Yu Wang
- Department of Family Medicine, Taichung Veterans General Hospital, Taichung City, Taiwan; Institute of Clinical Medicine, National Yang Ming University, Taipei City, Taiwan.
| | - Cheng-Yi Chang
- Department of Surgery, Feng Yuan Hospital, Taichung City, Taiwan.
| | - Shih-Yi Lin
- Institute of Clinical Medicine, National Yang Ming University, Taipei City, Taiwan; Center for Geriatrics and Gerontology, Taichung Veterans General Hospital, Taichung City, Taiwan.
| | - Jiaan-Der Wang
- Children's Medical Center, Taichung Veterans General Hospital, Taichung City, Taiwan; Department of Industrial Engineering and Enterprise Information, Tunghai University, Taichung City, Taiwan.
| | - Chih-Cheng Wu
- Department of Anesthesiology, Taichung Veterans General Hospital, Taichung City, Taiwan; Department of Financial Engineering, Providence University, Taichung City, Taiwan; Department of Data Science and Big Data Analytics, Providence University, Taichung City, Taiwan.
| | - Wen-Ying Chen
- Department of Veterinary Medicine, National Chung-Hsing University, Taichung City, Taiwan.
| | - Yu-Hsiang Kuan
- Department of Pharmacology, Chung Shan Medical University, Taichung City, Taiwan.
| | - Su-Lan Liao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung City, Taiwan.
| | - Wen-Yi Wang
- Department of Nursing, Hung-Kuang University, Taichung City, Taiwan.
| | - Chun-Jung Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung City, Taiwan; Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung City, Taiwan.
| |
Collapse
|
76
|
Chen H, He Y, Chen S, Qi S, Shen J. Therapeutic targets of oxidative/nitrosative stress and neuroinflammation in ischemic stroke: Applications for natural product efficacy with omics and systemic biology. Pharmacol Res 2020; 158:104877. [PMID: 32407958 DOI: 10.1016/j.phrs.2020.104877] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/11/2022]
Abstract
Oxidative/nitrosative stress and neuroinflammation are critical pathological processes in cerebral ischemia-reperfusion injury, and their intimate interactions mediate neuronal damage, blood-brain barrier (BBB) damage and hemorrhagic transformation (HT) during ischemic stroke. We review current progress towards understanding the interactions of oxidative/nitrosative stress and inflammatory responses in ischemic brain injury. The interactions between reactive oxygen species (ROS)/reactive nitrogen species (RNS) and innate immune receptors such as TLR2/4, NOD-like receptor, RAGE, and scavenger receptors are crucial pathological mechanisms that amplify brain damage during cerebral ischemic injury. Furthermore, we review the current progress of omics and systematic biology approaches for studying complex network regulations related to oxidative/nitrosative stress and inflammation in the pathology of ischemic stroke. Targeting oxidative/nitrosative stress and neuroinflammation could be a promising therapeutic strategy for ischemic stroke treatment. We then review recent advances in discovering compounds from medicinal herbs with the bioactivities of simultaneously regulating oxidative/nitrosative stress and pro-inflammatory molecules for minimizing ischemic brain injury. These compounds include sesamin, baicalin, salvianolic acid A, 6-paradol, silymarin, apocynin, 3H-1,2-Dithiole-3-thione, (-)-epicatechin, rutin, Dl-3-N-butylphthalide, and naringin. We finally summarize recent developments of the omics and systematic biology approaches for exploring the molecular mechanisms and active compounds of Traditional Chinese Medicine (TCM) formulae with the properties of antioxidant and anti-inflammation for neuroprotection. The comprehensive omics and systematic biology approaches provide powerful tools for exploring therapeutic principles of TCM formulae and developing precision medicine for stroke treatment.
Collapse
Affiliation(s)
- Hansen Chen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong Special Administrative Region; The University of Hong Kong-Shenzhen Institute of Research and Innovation (HKU-SIRI), China
| | - Yacong He
- School of Chinese Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Shuang Chen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Suhua Qi
- School of Medical Technology, Xuzhou Medical University, Xuzhou, 221002, China
| | - Jiangang Shen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong Special Administrative Region; The University of Hong Kong-Shenzhen Institute of Research and Innovation (HKU-SIRI), China; School of Medical Technology, Xuzhou Medical University, Xuzhou, 221002, China.
| |
Collapse
|
77
|
Adipose-derived mesenchymal stem cells protect against CMS-induced depression-like behaviors in mice via regulating the Nrf2/HO-1 and TLR4/NF-κB signaling pathways. Acta Pharmacol Sin 2020; 41:612-619. [PMID: 31796867 PMCID: PMC7468309 DOI: 10.1038/s41401-019-0317-6] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 10/09/2019] [Indexed: 12/27/2022] Open
Abstract
Increasing studies show that inflammatory processes may be involved in depressive disorders. Nuclear factor erythroid-2 related factor 2 (Nrf2) modulates tissue microglial M1 phenotypic changes to the M2 phenotype, which is implicated in protection against inflammatory diseases. We have reported that the adipose-derived mesenchymal stem cells (ADSCs) display anti-inflammatory activity. In this study we explored whether the mechanism of anti-inflammatory activity of ADSCs was related to Nrf2. ADSCs were isolated from mouse fat pads and intravenously administered to chronic mild stress (CMS)-exposed C57BL/6 mice at the dose of 1 × 106 once a week for 3 weeks. We showed that ADSC administration significantly remedied CMS-induced depressive-like behaviors in sucrose preference test, tail suspension test, and forced swim test accompanied by suppressing microglial activation and the expression of inflammatory factors including MCP-1, TNF-α, IL-1β, and IL-6. Furthermore, ADSC administration promoted both the expression of BDNF and TrkB, and promoted Nrf2/HO-1 signaling but suppressed TLR4/NF-κB signaling in brain tissue. In order to elucidate the role of Nrf2/HO-1 signaling in ADSC-caused neuroprotection, Nrf2-modified ADSCs were cocultured with BV2 microglial cells, then exposed to lipopolysaccharide (LPS). Downregulation of Nrf2 in ADSCs decreased the protective effects of ADSCs against LPS-induced microglial activation and M1 polarization. Nrf2 overexpression in ADSCs markedly suppressed LPS-induced TLR4 and NF-κB expression in microglial cells. These results suggest a possible antidepressive mechanism correlated with microglial polarization for anti-inflammatory agents, which may provide a new microglia-targeted strategy for depression therapy.
Collapse
|
78
|
Protection of the Geum japonicum Thunb. var. chinense extracts against oxygen-glucose deprivation and re-oxygenation induced astrocytes injury via BDNF/PI3K/Akt/CREB pathway. Biomed Pharmacother 2020; 127:110123. [PMID: 32361162 DOI: 10.1016/j.biopha.2020.110123] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/25/2020] [Accepted: 03/25/2020] [Indexed: 11/21/2022] Open
Abstract
Geum japonicum Tunb. var. chinense (GJ) is a traditional Chinese medicine usually used for the alleviation of dizziness and headache. Previous studies have reported that the GJ extracts could alleviate cerebral I/R injury by reducing apoptosis in vivo. To further elucidate the positive role and underlying mechanism of the GJ extracts in cerebral I/R injury, the current study investigated the effects of the GJ extracts on oxygen-glucose deprivation and re-oxygenation (OGD/R)-induced astrocytes injury in light of BDNF/PI3K/Akt/CREB signaling pathway with seropharmacological method. In the present study, the LC-MS profiling of the GJ extracts, obtain by reflux extraction, led to the identification of three possible active components were 5-desgalloylstachyurin, tellimagrandin II (TG II) and 3,4,5-Trihydroxybenzaldehyde (THBA). Drug-containing serum was collected from rats given different doses of the GJ extracts (0, 1.75 g/kg, 7 g/kg). Data indicated that the GJ extracts could increase the cell viability and decrease apoptosis and the expression of glial fibrillary acidic protein (GFAP) in OGD/R-induced astrocytes. In addition, the detection of apoptosis-related factors showed that the GJ extracts could obviously increase the expression of Bcl-2 and reduce the expression of Bax, Caspase-3 and cleaved-Caspase-3. Furthermore, the GJ extracts markedly increased the expression of BDNF, TrkB, PI3K, p-Akt and p-CREB. All these effects of the GJ extracts could be significantly reversed by LY294002, an inhibitor of PI3K. These data indicated that the GJ extracts could protect astrocytes against OGD/R-induced injury by inhibiting astrocytes reactivity and apoptosis, owing to the activation of the BDNF/PI3K/Akt/CREB pathway. The results support the application of the GJ extracts in the treatment of ischemic stroke and other ischemic encephalopathy.
Collapse
|
79
|
Gao J, Song L, Xia H, Peng L, Wen Z. 6'-O-galloylpaeoniflorin regulates proliferation and metastasis of non-small cell lung cancer through AMPK/miR-299-5p/ATF2 axis. Respir Res 2020; 21:39. [PMID: 32014006 PMCID: PMC6998290 DOI: 10.1186/s12931-020-1277-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 01/03/2020] [Indexed: 12/20/2022] Open
Abstract
Background Recent studies have shown 6'-O-galloylpaeoniflorin (GPF), a nature product extracted from the roots of paeoniflorin exerts anti-oxidant and anti-inflammatory activities. However, the effects of GPF on the proliferation and invasion in non-small cell lung cancer (NSCLC) cells have not been clarified. Methods MTT assay was performed to determine the cytotoxicity of GPF treatment on NSCLC cells. Colony formation assay, cell scratch test and transwell assay were performed to determine the proliferation and invasion of NSCLC cells in vitro, respectively. An A549 cell xenograft mouse model was performed to confirm the growth of NSCLC cells in vivo. Western blotting was used to measure the levels of activating transcription factor 2 (ATF2), AMP-activated protein kinase (AMPK) and phosph-AMPK (p-AMPK). Luciferase assay was used to validate the binding of miR-299-5p on the 3' untranslated region (UTR) of ATF2. Results Administration of GPF (50 or 100 μM) was significantly cytotoxic to A549 cells and H1299 cells, as well as inhibited the clonality, invasion and metastasis of NSCLC cells in vitro. GPF treatment also inhibited the tumor growth of NSCLC cell mouse xenografts in vivo. Exotic expression of miR-299-5p significantly inhibited the growth of NSCLC cells in vitro and in vivo. Downregulation of miR-299-5p expression attenuated the inhibition of the proliferation and metastasis of non-small cell lung cancer cells by GPF treatment. miR-299-5p significantly decreased ATF2 mRNA and protein levels in A549 cells (p < 0.05). Overexpression of ATF2 blocked the inhibitory effect of miR-299-5p on the proliferation and invasiveness of A549 cells. Conclusions GPF regulates miR-299-5p/ATF2 axis in A549 cells via the AMPK signalling pathway, thereby inhibiting the proliferation and metastasis of non-small cell lung cancer cells.
Collapse
Affiliation(s)
- Jinying Gao
- Department of Respiratory Medicine, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Lei Song
- Department of Respiratory Medicine, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Huan Xia
- Department of Respiratory Medicine, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Liping Peng
- Department of Respiratory Medicine, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, First Hospital of Jilin University, Changchun, Jilin Province, China.
| | - Zhongmei Wen
- Department of Respiratory Medicine, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, First Hospital of Jilin University, Changchun, Jilin Province, China.
| |
Collapse
|
80
|
CGRP Reduces Apoptosis of DRG Cells Induced by High-Glucose Oxidative Stress Injury through PI3K/AKT Induction of Heme Oxygenase-1 and Nrf-2 Expression. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2053149. [PMID: 31885775 PMCID: PMC6899316 DOI: 10.1155/2019/2053149] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 10/22/2019] [Indexed: 01/26/2023]
Abstract
Dorsal root ganglion (DRG) neurons, which are sensitive to oxidative stress due to their anatomical and structural characteristics, play a complex role in the initiation and progression of diabetic bladder neuropathy. We investigated the hypothesis that the antioxidant and antiapoptotic effects of CGRP may be partly related to the expression of Nrf2 and HO-1, via the phosphatidylinositol 3-kinase (PI3K)/AKT pathway, thus reducing apoptosis and oxidative stress responses. This study shows that CGRP activates the PI3K/AKT pathway, thereby inducing increased expression of Nrf2 and HO-1 and resulting in the decrease of reactive oxygen species and malondialdehyde levels and reduced neuronal apoptosis. These effects were suppressed by LY294002, an inhibitor of the PI3K/AKT pathway. Therefore, regulation of Nrf2 and HO-1 expression by the PI3K/AKT pathway plays an important role in the regulation of the antioxidant and antiapoptotic responses in DRG cells in a high-glucose culture model.
Collapse
|
81
|
Yang H, Lv H, Li H, Ci X, Peng L. Oridonin protects LPS-induced acute lung injury by modulating Nrf2-mediated oxidative stress and Nrf2-independent NLRP3 and NF-κB pathways. Cell Commun Signal 2019; 17:62. [PMID: 31186013 PMCID: PMC6558832 DOI: 10.1186/s12964-019-0366-y] [Citation(s) in RCA: 159] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 05/14/2019] [Indexed: 12/24/2022] Open
Abstract
Background Oxidative stress and the resulting inflammation are essential pathological processes in acute lung injury (ALI). Nuclear factor erythroid 2-related factor 2 (Nrf2), a vital transcriptional factor, possesses antioxidative potential and has become a primary target to treat many diseases. Oridonin (Ori), isolated from the plant Rabdosia Rrubescens, is a natural substance that possesses antioxidative and anti-inflammatory effects. Our aim was to study whether the anti-inflammatory and antioxidant effects of Ori on LPS-induced ALI were mediated by Nrf2. Methods MTT assays, Western blotting analysis, a mouse model, and hematoxylin-eosin (H & E) staining were employed to explore the mechanisms by which Ori exerts a protective effect on LPS-induced lung injury in RAW264.7 cells and in a mouse model. Results Our results indicated that Ori increased the expression of Nrf2 and its downstream genes (HO-1, GCLM), which was mediated by the activation of Akt and MAPK. Additionally, Ori inhibited LPS-induced activation of the pro-inflammatory pathways NLRP3 inflammasome and NF-κB pathways. These two pathways were also proven to be Nrf2-independent by the use of a Nrf2 inhibitor. In keeping with these findings, Ori alleviated LPS-induced histopathological changes, the enhanced production of myeloperoxidase and malondialdehyde, and the depleted expression of GSH and superoxide dismutase in the lung tissue of mice. Furthermore, the expression of LPS-induced NLRP3 inflammasome and NF-κB pathways was more evident in Nrf2-deficient mice but could still be reversed by Ori. Conclusions Our results demonstrated that Ori exerted protective effects on LPS-induced ALI via Nrf2-independent anti-inflammatory and Nrf2-dependent antioxidative activities.
Collapse
Affiliation(s)
- Huahong Yang
- Institute of Translational Medicine, The First Hospital of Jilin University, Dongminzhu road 519, Changchun, Jilin, 130001, People's Republic of China.,Department of Respiratory Medicine, The First Hospital of Jilin University, Xinmin road 71, Changchun, Jilin, 130001, People's Republic of China
| | - Hongming Lv
- Institute of Translational Medicine, The First Hospital of Jilin University, Dongminzhu road 519, Changchun, Jilin, 130001, People's Republic of China
| | - Haijun Li
- Institute of Translational Medicine, The First Hospital of Jilin University, Dongminzhu road 519, Changchun, Jilin, 130001, People's Republic of China
| | - Xinxin Ci
- Institute of Translational Medicine, The First Hospital of Jilin University, Dongminzhu road 519, Changchun, Jilin, 130001, People's Republic of China. .,Department of Respiratory Medicine, The First Hospital of Jilin University, Xinmin road 71, Changchun, Jilin, 130001, People's Republic of China.
| | - Liping Peng
- Department of Respiratory Medicine, The First Hospital of Jilin University, Xinmin road 71, Changchun, Jilin, 130001, People's Republic of China.
| |
Collapse
|
82
|
Wang H, Wei W, Lan X, Liu N, Li Y, Ma H, Sun T, Peng X, Zhuang C, Yu J. Neuroprotective Effect of Swertiamain on Cerebral Ischemia/Reperfusion Injury by Inducing the Nrf2 Protective Pathway. ACS Chem Neurosci 2019; 10:2276-2286. [PMID: 30753053 DOI: 10.1021/acschemneuro.8b00605] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Oxidative stress plays a vital role in the development of cerebral ischemic/reperfusion (I/R). Targeting oxidative stress is proposed to be an effective strategy to treat cerebral I/R injury. Gentiana macrophylla Pall is reported to have a potential protective effect against stroke. Swertiamarin (Swe), an active secoiridoid glycoside compound isolated from Gentiana macrophylla Pall, has been reported to possess antioxidative potential. This study is to explore whether Swe could prevent brain from I/R injury, and the related mechanisms of oxidative stress are also elucidated using mice middle cerebral artery occlusion (MCAO) model and primary hippocampal neurons oxygen-glucose deprivation/reperfusion (OGD/R) model. Swe (25, 100, or 400 mg/kg) was pretreated intraperitoneally for 7 days until establishment of the MCAO model, while hippocampal neurons were maintained in Swe (0.1, 1, or 10 μM) in the entire process of reoxygenation. The results indicated that Swe pretreatment markedly decreased infarct volume, apoptotic neurons, and oxidative damage and promoted neurologic recovery in vivo. It also decreased reactive oxygen species (ROS) and increased cell viability in vitro. Western blot analyses and immunofluorescence staining demonstrated that Swe pretreatment promoted Nrf2 nuclear translocation from Keap1-Nrf2 complex and enhanced the expressions of NAD(P)H: quinone oxidoreductase-1 (NQO1) and heme oxygenase-1 (HO-1) both in vivo and in vitro, while the expressions could be reversed by a Nrf2 inhibitor. The binding mode of Keap1 with Swe was also proposed by covalent molecular docking. Collectively, Swe could be considered as a promising protective agent against cerebral I/R injury through suppressing oxidative stress by activation of the Nrf2 protective pathway.
Collapse
Affiliation(s)
- Hui Wang
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Wei Wei
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Xiaobing Lan
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Ning Liu
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- Ningxia Hui Medicine Modern Engineering Research Center and Collaborative Innovation Center, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Yuxiang Li
- College of Nursing, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Hanxiang Ma
- Department of Anesthesiology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Tao Sun
- Ningxia Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Xiaodong Peng
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Chunlin Zhuang
- Ningxia Hui Medicine Modern Engineering Research Center and Collaborative Innovation Center, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, P.R. China
| | - Jianqiang Yu
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- Ningxia Hui Medicine Modern Engineering Research Center and Collaborative Innovation Center, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- Ningxia Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| |
Collapse
|
83
|
Huang WY, Jiang C, Ye HB, Jiao JT, Cheng C, Huang J, Liu J, Zhang R, Shao JF. miR-124 upregulates astrocytic glutamate transporter-1 via the Akt and mTOR signaling pathway post ischemic stroke. Brain Res Bull 2019; 149:231-239. [PMID: 31004734 DOI: 10.1016/j.brainresbull.2019.04.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 03/29/2019] [Accepted: 04/15/2019] [Indexed: 10/27/2022]
Abstract
High-concentration glutamic acid (Glu) induced by ischemic stroke can be inhibited by glutamate transporter-1 (GLT-1), which is the main mechanism for preventing excessive extracellular glutamate accumulation in the central nervous system. Upregulation of miR-124 could reduce the infarct area and promote the recovery of neurological function after ischemic stroke. A previous study investigated whether miR-124 could regulate GLT-1 expression in normal culture conditions. However, the role of miR-124 in the regulation of GLT-1 expression and further mechanisms after ischemic stroke remain unclear. In this study, the effects of miR-124 on GLT-1 expression in astrocytes after ischemic stroke were explored using an in vitro model of ischemic stroke (oxygen-glucose deprivation/reperfusion, OGD/reperfusion). The expression of GLT-1 was significantly decreased with lower expression of miR-124 in astrocytes injured by OGD/reperfusion. When miR-124 expression was improved, the expression of GLT-1 was notably increased in astrocytes injured by OGD/reperfusion. The results revealed that GLT-1 expression in astrocytes had a relationship with miR-124 after OGD/reperfusion. However, a direct interaction could not be confirmed with a luciferase reporter assay. Further results demonstrated that an inhibitor of Akt could decrease the increased protein expression of GLT-1 induced by miR-124 mimics, and an inhibitor of mTOR could increase the reduced protein expression of GLT-1 caused by a miR-124 inhibitor in astrocytes injured by different OGD/reperfusion conditions. These results indicated that miR-124 could regulate GLT-1 expression in astrocytes after OGD/reperfusion through the Akt and mTOR pathway.
Collapse
Affiliation(s)
- Wei-Yi Huang
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, People's Republic of China
| | - Chen Jiang
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, People's Republic of China
| | - Han-Bin Ye
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, People's Republic of China
| | - Jian-Tong Jiao
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, People's Republic of China
| | - Chao Cheng
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, People's Republic of China
| | - Jin Huang
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, People's Republic of China
| | - Jin Liu
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, People's Republic of China
| | - Rui Zhang
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, People's Republic of China
| | - Jun-Fei Shao
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, People's Republic of China.
| |
Collapse
|
84
|
Liu Q, Li Z, Liu Y, Xiao Q, Peng X, Chen Q, Deng R, Gao Z, Yu F, Zhang Y. Hydromorphine postconditioning protects isolated rat heart against ischemia-reperfusion injury via activating P13K/Akt/eNOS signaling. Cardiovasc Ther 2019; 36:e12481. [PMID: 30597772 DOI: 10.1111/1755-5922.12481] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 12/28/2018] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Myocardial ischemia/reperfusion injury (myocardial I/R injury) has a high disability rate and mortality. Novel treatments for myocardial I/R injury are necessary. AIM In order to explore the protective effect of hydromorphine on myocardial I/R injury, we illuminate the underlying mechanism of the protective effect. RESULTS Hydromorphine significantly reduced myocardial infarct size (IFN/AAR), CKMB (Creatine Kinase MB) and TN-T (Troponin T) release, and improved cardiac function compared with I/R group. However, these advantageous effects were partly suppressed in the presence of hydromorphine. Myocardial I/R injury significantly decreased the phosphorylation of Akt and eNOS, and down-regulated total nitric oxide and nitrotyrosine content, while these inhibitory effects were partly abolished by hydromorphine. Conversely, the activated effects of hydromorphine on the phosphorylation of Akt and eNOS, and NO release were totally reversed by LY294002, which, used individually, show the same influence on reperfusion injury. CONCLUSIONS These findings suggest that hydromorphine postconditioning may protect isolated rat heart against reperfusion injury via activating P13K/Akt/eNOS signaling.
Collapse
Affiliation(s)
- Qing Liu
- Department of Anesthesiology, Affiliated traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Zhengfen Li
- Department of Anesthesiology, Affiliated traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Yuexin Liu
- Department of Anesthesiology, Affiliated traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Qiuxia Xiao
- Department of Anesthesiology, Affiliated traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Xuan Peng
- Department of Anesthesiology, Affiliated traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Qi Chen
- Department of Anesthesiology, Affiliated traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Rui Deng
- Department of Anesthesiology, Affiliated traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Zhiwei Gao
- Department of Anesthesiology, Affiliated traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Fengxu Yu
- Department of Cardio-thoracic Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Ying Zhang
- Department of Anesthesiology, Affiliated traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
85
|
Cong P, Tong C, Liu Y, Shi L, Shi X, Zhao Y, Xiao K, Jin H, Liu Y, Hou M. CD28 Deficiency Ameliorates Thoracic Blast Exposure-Induced Oxidative Stress and Apoptosis in the Brain through the PI3K/Nrf2/Keap1 Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8460290. [PMID: 31885821 PMCID: PMC6915017 DOI: 10.1155/2019/8460290] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 09/25/2019] [Accepted: 10/24/2019] [Indexed: 12/22/2022]
Abstract
Blast exposure is a worldwide public health concern, but most related research has been focused on direct injury. Thoracic blast exposure-induced neurotrauma is a type of indirect injuries where research is lacking. As CD28 stimulates T cell activation and survival and contributes to inflammation initiation, it may play a role in thoracic blast exposure-induced neurotrauma. However, it has not been investigated. To explore the effects of CD28 on thoracic blast exposure-induced brain injury and its potential molecular mechanisms, a mouse model of thoracic blast exposure-induced brain injury was established. Fifty C57BL/6 wild-type (WT) and fifty CD28 knockout (CD28-/-) mice were randomly divided into five groups (one control group and four model groups), with ten mice (from each of the two models) for each group. Lung and brain tissue and serum samples were collected at 12 h, 24 h, 48 h, and 1 week after thoracic blast exposure. Histopathological changes were detected by hematoxylin-eosin staining. The expressions of inflammatory-related factors were detected by ELISA. Oxidative stress in the brain tissue was evaluated by determining the generation of reactive oxygen species (ROS) and the expressions of thioredoxin (TRX), malondialdehyde (MDA), SOD-1, and SOD-2. Apoptosis in the brain tissue was evaluated by TUNEL staining and the levels of Bax, Bcl-xL, Bad, Cytochrome C, and caspase-3. In addition, proteins of related pathways were also studied by western blotting and immunofluorescence. We found that CD28 deficiency significantly reduced thoracic blast exposure-induced histopathological changes and decreased the levels of inflammatory-related factors, including IL-1β, TNF-α, and S100β. In the brain tissue, CD28 deficiency also significantly attenuated thoracic blast exposure-induced generation of ROS and expressions of MDA, TRX, SOD-1, and SOD-2; lowered the number of apoptotic cells and the expression of Bax, cleaved caspase-3, Cytochrome C, and Bad; and maintained Bcl-xL expression. Additionally, CD28 deficiency significantly ameliorated thoracic blast exposure-induced increases of p-PI3K and Keap1 and the decrease of Nrf2 expression in the brain. Our results indicate that CD28 deficiency has a protective effect on thoracic blast exposure-induced brain injury that might be associated with the PI3K/Nrf2/Keap1 signaling pathway.
Collapse
Affiliation(s)
- Peifang Cong
- 1College of Medicine and Biological Information Engineering, Northeastern University, No. 195, Chuangxin Road, Hunnan District, Shenyang l10016, China
- 2Emergency Medicine Department of General Hospital of Northern theater command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, No. 83, Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Changci Tong
- 2Emergency Medicine Department of General Hospital of Northern theater command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, No. 83, Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Ying Liu
- 2Emergency Medicine Department of General Hospital of Northern theater command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, No. 83, Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Lin Shi
- 2Emergency Medicine Department of General Hospital of Northern theater command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, No. 83, Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Xiuyun Shi
- 2Emergency Medicine Department of General Hospital of Northern theater command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, No. 83, Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Yan Zhao
- 3Institute of Metal Research, Chinese Academy of Sciences, No. 72, Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Keshen Xiao
- 3Institute of Metal Research, Chinese Academy of Sciences, No. 72, Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Hongxu Jin
- 2Emergency Medicine Department of General Hospital of Northern theater command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, No. 83, Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Yunen Liu
- 2Emergency Medicine Department of General Hospital of Northern theater command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, No. 83, Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Mingxiao Hou
- 1College of Medicine and Biological Information Engineering, Northeastern University, No. 195, Chuangxin Road, Hunnan District, Shenyang l10016, China
| |
Collapse
|
86
|
Xu L, Xing Q, Huang T, Zhou J, Liu T, Cui Y, Cheng T, Wang Y, Zhou X, Yang B, Yang GL, Zhang J, Zang X, Ma S, Guan F. HDAC1 Silence Promotes Neuroprotective Effects of Human Umbilical Cord-Derived Mesenchymal Stem Cells in a Mouse Model of Traumatic Brain Injury via PI3K/AKT Pathway. Front Cell Neurosci 2018; 12:498. [PMID: 30662396 PMCID: PMC6328439 DOI: 10.3389/fncel.2018.00498] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 12/03/2018] [Indexed: 01/09/2023] Open
Abstract
Stem cell transplantation is a promising therapy for traumatic brain injury (TBI), but low efficiency of survival and differentiation of transplanted stem cells limits its clinical application. Histone deacetylase 1 (HDAC1) plays important roles in self-renewal of stem cells as well as the recovery of brain disorders. However, little is known about the effects of HDAC1 on the survival and efficacy of human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) in vivo. In this study, our results showed that HDAC1 silence promoted hUC-MSCs engraftment in the hippocampus and increased the neuroprotective effects of hUC-MSCs in TBI mouse model, which was accompanied by improved neurological function, enhanced neurogenesis, decreased neural apoptosis, and reduced oxidative stress in the hippocampus. Further mechanistic studies revealed that the expressions of phosphorylated PTEN (p-PTEN), phosphorylated Akt (p-Akt), and phosphorylated GSK-3β (p-GSK-3β) were upregulated. Intriguingly, the neuroprotective effects of hUC-MSCs with HDAC1 silence on behavioral performance of TBI mice was markedly attenuated by LY294002, an inhibitor of the PI3K/AKT pathway. Taken together, our findings suggest that hUC-MSCs transplantation with HDAC1 silence may provide a potential strategy for treating TBI in the future.
Collapse
Affiliation(s)
- Ling Xu
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
- Henan Provincial People’s Hospital, Zhengzhou, China
| | - Qu Xing
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Tuanjie Huang
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Jiankang Zhou
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Tengfei Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Yuanbo Cui
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
- Translational Medicine Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Tian Cheng
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yaping Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Xinkui Zhou
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Bo Yang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | | | - Jiewen Zhang
- Henan Provincial People’s Hospital, Zhengzhou, China
| | - Xingxing Zang
- Department of Microbiology and Immunology, Einstein College of Medicine, Bronx, NY, United States
| | - Shanshan Ma
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
- *Correspondence: Shanshan Ma Fangxia Guan
| | - Fangxia Guan
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
- Henan Provincial People’s Hospital, Zhengzhou, China
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Shanshan Ma Fangxia Guan
| |
Collapse
|