1
|
Luo Y, Dong W, Yuan L, Zhu YA, Zhang DD, Ni H, Zhu W. The Role of Thrombo-inflammation in Ischemic Stroke: Focus on the Manipulation and Clinical Application. Mol Neurobiol 2025; 62:2362-2375. [PMID: 39107669 DOI: 10.1007/s12035-024-04397-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 07/22/2024] [Indexed: 01/28/2025]
Abstract
Stroke leaves a great economic burden due to its high morbidity and mortality. Rapid revascularization of targeted vessel(s) is the effective treatment for ischemic stroke, but subsequent ischemia-reperfusion (I/R) injury is a common complication following revascularization, leading to microcirculation dysfunction and infarct volume increase. Thrombo-inflammation, the interaction between thrombosis and inflammation, plays a critical role in the pathophysiology of ischemic stroke. In the context of I/R injury, thrombo-inflammation consists of platelet activation, endothelial injury, and inflammatory cell infiltration. Numerous studies are devoted to exploring methods of regulating thrombo-inflammation to mitigate I/R injury post-stroke, including blocking activations of platelets and neutrophils. Drugs such as antiplatelet medications, anticoagulants, and glucocorticoids have been confirmed to have the potential to regulate thrombo-inflammation. Furthermore, several recently developed drugs have also shown promises in relieving I/R injury by manipulating thrombo-inflammation. However, the majority of these studies are still in the preclinical stage. Herein, in this review, we will address the mechanisms of thrombo-inflammation in ischemic stroke, related research advances, and particularly the clinical feasibility of thrombo-inflammation as a therapeutic strategy against I/R injury.
Collapse
Affiliation(s)
- Yuanfei Luo
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Weichen Dong
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Linying Yuan
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yunqing Amelia Zhu
- Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, ON, M5B 1W8, Canada
| | - Dachuan Dustin Zhang
- Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, ON, M5B 1W8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A1, Canada
| | - Heyu Ni
- Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, ON, M5B 1W8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A1, Canada
- Canadian Blood Services Centre for Innovation, Toronto, ON, M5G 2M1, Canada
- CCOA Therapeutics Inc., Toronto, ON, M5B 1W8, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A1, Canada
- Department of Medicine, University of Toronto, Toronto, ON, M5S 1A1, Canada
| | - Wusheng Zhu
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
2
|
Zhao Y, He M, Cui L, Zhang M, Zhao T, Yang X, Xu Y, Dong J, He K, Zhang H, Chen L. Systematic screening of protein-coding gene expression identified VWF as a potential key regulator in anthracycline-based chemotherapy-exacerbated metastasis of breast cancer. BMC Cancer 2024; 24:1243. [PMID: 39379897 PMCID: PMC11462902 DOI: 10.1186/s12885-024-12999-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 09/27/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Breast cancer is the most commonly diagnosed cancer worldwide. Although major treatments represented by chemotherapy have shown effectiveness at the initial period, recurrence and metastasis still occur later after treatments. The alternation of the tumor microenvironment by chemotherapy is confirmed as a trigger of the elevated proliferation and migration of the remaining tumor cells. METHODS Using bioinformatic methods, differential gene expression analysis was used to determine DEGs between post-chemotherapy and pre-chemotherapy samples of breast cancer patients, followed by survival analysis and ELISA analysis of the potential key genes. An in vitro model of 2 breast cancer cells lines was used to demonstrate the role of VWF in the evasion and migration of breast cancer cells, using cell migration, evasion and wound healing assays, PCR and molecular docking analysis. RESULTS 19 hub genes were further identified using GO and KEGG pathway analyses and WGCNA. The 5 secreted protein-coding genes with reported carcinogenesis effects (VWF, SVEP1, DPT, ADIPOQ, and LPL) were further analyzed in breast cancer patients and VWF was identified as a potential key regulator in the anthracycline-based chemotherapy-exacerbated metastasis. It was further confirmed that anthracycline-based chemotherapeutics doxorubicin exacerbated VWF upregulation and the evasion and migration of breast cancer cells. Based on molecular docking analysis and previous study, berberine was used as an inhibitor of VWF, and showed an effective inhibition of the doxorubicin-exacerbated VWF upregulation, migration and evasion in breast cancer. CONCLUSIONS Doxorubicin-exacerbated evasion and migration through VWF upregulation. Berberine as an inhibitor of VWF was able to reversed the doxorubicin-exacerbated VWF upregulation and evasion and migration in breast cancer cells.
Collapse
Affiliation(s)
- Yawei Zhao
- Department of Pharmacology, Nanomedicine Engineering Laboratory of Jilin Province, College of Basic Medical Sciences, Jilin University, No. 126 Xinmin Street, Changchun, 130021, China
| | - Meihui He
- Department of Pharmacology, Nanomedicine Engineering Laboratory of Jilin Province, College of Basic Medical Sciences, Jilin University, No. 126 Xinmin Street, Changchun, 130021, China
- Changchun Medical College, Changchun, 130031, China
| | - Lianzhi Cui
- Clinical Laboratory, Jilin Cancer Hospital, Changchun, 130012, China
| | - Min Zhang
- Department of Pharmacology, Nanomedicine Engineering Laboratory of Jilin Province, College of Basic Medical Sciences, Jilin University, No. 126 Xinmin Street, Changchun, 130021, China
| | - Tianyu Zhao
- Department of Pharmacology, Nanomedicine Engineering Laboratory of Jilin Province, College of Basic Medical Sciences, Jilin University, No. 126 Xinmin Street, Changchun, 130021, China
| | - Xuehan Yang
- Department of Pharmacology, Nanomedicine Engineering Laboratory of Jilin Province, College of Basic Medical Sciences, Jilin University, No. 126 Xinmin Street, Changchun, 130021, China
| | - Yang Xu
- Department of Pharmacology, Nanomedicine Engineering Laboratory of Jilin Province, College of Basic Medical Sciences, Jilin University, No. 126 Xinmin Street, Changchun, 130021, China
| | - Jianhua Dong
- Department of Pharmacology, Nanomedicine Engineering Laboratory of Jilin Province, College of Basic Medical Sciences, Jilin University, No. 126 Xinmin Street, Changchun, 130021, China
| | - Kan He
- Department of Pharmacology, Nanomedicine Engineering Laboratory of Jilin Province, College of Basic Medical Sciences, Jilin University, No. 126 Xinmin Street, Changchun, 130021, China
| | - Hansi Zhang
- Department of Pharmacology, Nanomedicine Engineering Laboratory of Jilin Province, College of Basic Medical Sciences, Jilin University, No. 126 Xinmin Street, Changchun, 130021, China.
| | - Li Chen
- Department of Pharmacology, Nanomedicine Engineering Laboratory of Jilin Province, College of Basic Medical Sciences, Jilin University, No. 126 Xinmin Street, Changchun, 130021, China.
- School of Nursing, Jilin University, Changchun, 130021, China.
| |
Collapse
|
3
|
Shahraki J, Tabrizian K, Rezaee R, Tashakori B, Dadrezaei S, Ghorani V, Bagheri G, Jahantigh H, Hashemzaei M. Hesperidin neuroprotective effects against carbon monoxide-induced toxicity in male rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7673-7681. [PMID: 38700797 DOI: 10.1007/s00210-024-03132-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 04/29/2024] [Indexed: 08/04/2024]
Abstract
Carbon monoxide (CO) is produced via incomplete combustion of fossil fuels and it may cause long-term neurological sequel upon exposure. Hesperidin (HES), a flavanone glycoside found in citrus plants, exerts diverse beneficial health effects. The present study mechanistically examined the neuroprotective effects of HES in CO-poisoned rats. Thirty male Wistar rats (five groups of six animals) were exposed to 3000 ppm CO for 1 h. Immediately after the exposure and on the next 4 consecutive days (totally five doses), rats intraperitoneally received either normal saline (the control group) or different doses of HES (25, 50, and 100 mg/kg). A sham group that was not exposed to CO was also considered. After evaluation of spatial learning and memory using a Morris water maze (MWM), animals were sacrificed and oxidative stress status in blood samples, and Akt, Bax, Bcl2, and brain-derived neurotrophic factor (BDNF) expression in brain samples were assessed. Western blot analysis indicated increased Akt but decreased Bax/Bcl2 levels in the HES 100 mg/kg, and induced BDNF levels in all HES-treated groups. MWM results showed that HES significantly decreased memory loss. The current findings indicate that HES could alleviate neurological impairments induced by CO in rats.
Collapse
Affiliation(s)
- Jafar Shahraki
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Zabol University of Medical Sciences, Zabol, Iran
| | - Kaveh Tabrizian
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Zabol University of Medical Sciences, Zabol, Iran
| | - Ramin Rezaee
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Behnam Tashakori
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Zabol University of Medical Sciences, Zabol, Iran
| | - Seyedehzahra Dadrezaei
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Zabol University of Medical Sciences, Zabol, Iran
| | - Vahideh Ghorani
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Clinical Research Development Unit, Faculty of Medicine, Imam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Hosseinali Jahantigh
- Department of Pathology, Amiralmomenin Hospital, Zabol University of Medical Sciences, Zabol, Iran
| | - Mahmoud Hashemzaei
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Zabol University of Medical Sciences, Zabol, Iran.
| |
Collapse
|
4
|
Tao D, Xia X, Zhang X, Yang R, Yang Y, Zhang L, Shi Y, Lv D, Chen P, He B, Shen Z. Integrated network pharmacology, molecular docking and pharmacodynamic study reveals protective effects and mechanisms of corilagin against cerebral ischemia-induced injury. Exp Neurol 2024; 374:114697. [PMID: 38266765 DOI: 10.1016/j.expneurol.2024.114697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 01/11/2024] [Accepted: 01/18/2024] [Indexed: 01/26/2024]
Abstract
BACKGROUND Stroke is one of the leading causes of death and long-term disability worldwide. Previous studies have found that corilagin has antioxidant, anti-inflammatory, anti-atherosclerotic and other pharmacological activities and has a protective effect against cardiac and cerebrovascular injury. OBJECTIVES The aim of this study was to investigate the protective effects of corilagin against ischemic stroke and to elucidate the underlying molecular mechanisms using network pharmacology, molecular docking, and animal and cell experiments. METHODS We investigated the potential of corilagin to ameliorate cerebral ischemia-reperfusion injury using in vivo rat middle cerebral artery occlusion/reperfusion (MCAO/R) and in vitro oxygen-glucose deprivation/reoxygenation (OGD/R) models. RESULTS Our results suggest that corilagin may exert its anti-ischemic stroke effect by interacting with 92 key targets, including apoptosis-associated proteins (Bcl-2, Bax, caspase-3) and PI3K/Akt signaling pathway-related proteins. In vivo and in vitro experiments showed that corilagin treatment improved neurological deficits, attenuated cerebral infarct volume, and mitigated neuronal damage in MCAO/R rats. Corilagin treatment also enhanced the survival of PC12 cells exposed to OGD/R, reduced the rate of LDH leakage, inhibited cell apoptosis, and activated the PI3K/Akt signaling pathway. Importantly, the effects of corilagin on the PI3K/Akt signaling pathway and apoptosis-associated proteins were reversed by the PI3K-specific inhibitor LY294002. CONCLUSIONS These results indicate that the molecular mechanism of the anti-ischemic effect of corilagin involves inhibiting neuronal apoptosis and activating the PI3K/Akt signaling pathway. These findings provide a theoretical and experimental basis for the further development and application of corilagin as a potential anti-ischemic stroke agent.
Collapse
Affiliation(s)
- Daiju Tao
- School of Pharmaceutical Science, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming 650500, PR China; College of Modern biomedical industry, Kunming Medical University, Kunming 650500, PR China
| | - Xin Xia
- School of Pharmaceutical Science, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming 650500, PR China; People's Hospital of Yilong County, Sichuan Province 637600, PR China
| | - Xiaochao Zhang
- School of Pharmaceutical Science, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming 650500, PR China; College of Modern biomedical industry, Kunming Medical University, Kunming 650500, PR China
| | - Renhua Yang
- School of Pharmaceutical Science, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming 650500, PR China; College of Modern biomedical industry, Kunming Medical University, Kunming 650500, PR China
| | - Yuan Yang
- School of Pharmaceutical Science, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming 650500, PR China; College of Modern biomedical industry, Kunming Medical University, Kunming 650500, PR China
| | - Li Zhang
- School of Pharmaceutical Science, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming 650500, PR China; College of Modern biomedical industry, Kunming Medical University, Kunming 650500, PR China
| | - Yunke Shi
- School of Pharmaceutical Science, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming 650500, PR China; College of Modern biomedical industry, Kunming Medical University, Kunming 650500, PR China
| | - Di Lv
- School of Pharmaceutical Science, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming 650500, PR China; College of Modern biomedical industry, Kunming Medical University, Kunming 650500, PR China
| | - Peng Chen
- School of Pharmaceutical Science, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming 650500, PR China; College of Modern biomedical industry, Kunming Medical University, Kunming 650500, PR China.
| | - Bo He
- School of Pharmaceutical Science, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming 650500, PR China; College of Modern biomedical industry, Kunming Medical University, Kunming 650500, PR China.
| | - Zhiqiang Shen
- School of Pharmaceutical Science, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming 650500, PR China; College of Modern biomedical industry, Kunming Medical University, Kunming 650500, PR China.
| |
Collapse
|
5
|
Li L, Stegner D. Immunothrombosis versus thrombo-inflammation: platelets in cerebrovascular complications. Res Pract Thromb Haemost 2024; 8:102344. [PMID: 38433977 PMCID: PMC10907225 DOI: 10.1016/j.rpth.2024.102344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/22/2023] [Accepted: 01/30/2024] [Indexed: 03/05/2024] Open
Abstract
A State-of-the Art lecture titled "Thrombo-Neuroinflammatory Disease" was presented at the International Society on Thrombosis and Haemostasis Congress in 2023. First, we would like to advocate for discrimination between immunothrombosis and thrombo-inflammation, as immunothrombosis describes an overshooting inflammatory reaction that results in detrimental thrombotic activity. In contrast, thrombo-inflammation describes the interplay of platelets and coagulation with the immunovascular system, resulting in the recruitment of immune cells and loss of barrier function (hence, hallmarks of inflammation). Both processes can be observed in the brain, with cerebral venous thrombosis being a prime example of immunothrombosis, while infarct progression in response to ischemic stroke is a paradigmatic example of thrombo-inflammation. Here, we review the pathomechanisms underlying cerebral venous thrombosis and ischemic stroke from a platelet-centric perspective and discuss translational implications. Finally, we summarize relevant new data on this topic presented during the 2023 International Society on Thrombosis and Haemostasis Congress.
Collapse
Affiliation(s)
- Lexiao Li
- Julius-Maximilians-Universität Würzburg, Rudolf Virchow Center for Integrative and Translational Bioimaging, Würzburg, Germany
- University Hospital Würzburg, Institute of Experimental Biomedicine, Würzburg, Germany
| | - David Stegner
- Julius-Maximilians-Universität Würzburg, Rudolf Virchow Center for Integrative and Translational Bioimaging, Würzburg, Germany
- University Hospital Würzburg, Institute of Experimental Biomedicine, Würzburg, Germany
| |
Collapse
|
6
|
Xu H, Dong J, Li Y, Zhang L, Yin J, Zhu C, Wang X, Ren K, Zhang H, Zhao D. Neuritin has a neuroprotective role in the rat model of acute ischemia stroke by inhibiting neuronal apoptosis and NLRP3 inflammasome. J Stroke Cerebrovasc Dis 2023; 32:107391. [PMID: 37832268 DOI: 10.1016/j.jstrokecerebrovasdis.2023.107391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 09/22/2023] [Accepted: 09/24/2023] [Indexed: 10/15/2023] Open
Abstract
OBJECTIVES This study explored the anti-inflammatory, anti-neuronal apoptosis, and neuroprotective effects of Neuritin in rat models of acute ischemia stroke (AIS). METHODS AIS was induced in male Sprague Dawley rats by middle cerebral artery occlusion (MCAO). Rats were divided into sham, MCAO, MCAO+neuritin, MCAO + neuritin + PBS, MCAO + neuritin+MCC950, and MCAO + neuritin + MSU groups. Neurological score assessment, brain water content measurement, HE staining, TTC staining, TUNEL staining, ELISA, and Western blot were performed. RESULTS Neuritin significantly improved the neurobehavioral score, infarct size, brain water content, apoptosis, and neuroinflammatory response compared with the MCAO and MCAO + PBS groups within 24 h after AIS. Moreover, Neuritin inhibited the protein expression of NLRP3 inflammasome, and reduced the expression of IL-18 and IL-1B, thereby reducing the inflammatory response. Meanwhile, the neuroprotection, anti-inflammation, and anti-apoptosis effects of Neuritin were enhanced by MCC950 but partly counteracted by MSU. CONCLUSION Neuritin may reduce brain injury after AIS by inhibiting the expression of NLRP3 inflammasome and then inhibiting the inflammatory response.
Collapse
Affiliation(s)
- Hui Xu
- Department of Neurosurgery, the First Affiliated Hospital of Medical College, Shihezi University, Shihezi 832000, China
| | - Jiangtao Dong
- Department of Neurosurgery, the First Affiliated Hospital of Medical College, Shihezi University, Shihezi 832000, China
| | - Yang Li
- Department of Neurosurgery, the First Affiliated Hospital of Medical College, Shihezi University, Shihezi 832000, China
| | - Lei Zhang
- Department of Neuromedicine, Beitun Hospital, the Tenth Division of Xinjiang Production and Construction Corps, Beitun 836000, China
| | - Jiangwen Yin
- Department of Anesthesiology, First Affiliated Hospital of Medical College, Shihezi University, Shihezi 832000, China
| | - Chao Zhu
- Department of Neurosurgery, the First Affiliated Hospital of Medical College, Shihezi University, Shihezi 832000, China
| | - Xu Wang
- Department of Neurosurgery, the First Affiliated Hospital of Medical College, Shihezi University, Shihezi 832000, China
| | - Kunhao Ren
- Department of Neurosurgery, the First Affiliated Hospital of Medical College, Shihezi University, Shihezi 832000, China
| | - Hao Zhang
- Department of Neurosurgery, the First Affiliated Hospital of Medical College, Shihezi University, Shihezi 832000, China
| | - Dong Zhao
- Department of Neurosurgery, the First Affiliated Hospital of Medical College, Shihezi University, Shihezi 832000, China.
| |
Collapse
|
7
|
Hong QL, Ding YH, Chen JY, Shi SS, Liang RS, Tu XK. Schisandrin B Protects against Ischemic Brain Damage by Regulating PI3K/AKT Signaling in Rats. Chin J Integr Med 2023; 29:885-894. [PMID: 37357242 DOI: 10.1007/s11655-023-3596-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2022] [Indexed: 06/27/2023]
Abstract
OBJECTIVE To explore the effect and mechanism of schisandrin B (Sch B) in the treatment of cerebral ischemia in rats. METHODS The cerebral ischemia models were induced by middle cerebral artery occlusion (MCAO) and reperfusion. Sprague-Dawley rats were divided into 6 groups using a random number table, including sham, MCAO, MCAO+Sch B (50 mg/kg), MCAO+Sch B (100 mg/kg), MCAO+Sch B (100 mg/kg)+LY294002, and MCAO+Sch B (100 mg/kg)+wortmannin groups. The effects of Sch B on pathological indicators, including neurological deficit scores, cerebral infarct volume, and brain edema, were subsequently studied. Tissue apoptosis was identified by terminal transferase-mediated dUTP nick end-labeling (TUNEL) staining. The protein expressions involved in apoptosis, inflammation response and oxidative stress were examined by immunofluorescent staining, biochemical analysis and Western blot analysis, respectively. The effect of Sch B on phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling was also explored. RESULTS Sch B treatment decreased neurological deficit scores, cerebral water content, and infarct volume in MCAO rats (P<0.05 or P<0.01). Neuronal nuclei and TUNEL staining indicated that Sch B also reduced apoptosis in brain tissues, as well as the Bax/Bcl-2 ratio and caspase-3 expression (P<0.01). Sch B regulated the production of myeloperoxidase, malondialdehyde, nitric oxide and superoxide dismutase, as well as the release of cytokine interleukin (IL)-1 β and IL-18, in MCAO rats (P<0.05 or P<0.01). Sch B promoted the phosphorylation of PI3K and AKT. Blocking the PI3K/AKT signaling pathway with LY294002 or wortmannin reduced the protective effect of Sch B against cerebral ischemia (P<0.05 or P<0.01). CONCLUSIONS Sch B reduced apoptosis, inflammatory response, and oxidative stress of MCAO rats by modulating the PI3K/AKT pathway. Sch B had a potential for treating cerebral ischemia.
Collapse
Affiliation(s)
- Quan-Long Hong
- Department of Neurology, The First Hospital of Quanzhou Affiliated to Fujian Medical University, Quanzhou, Fujian Province, 362000, China
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Yi-Hang Ding
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Jing-Yi Chen
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Song-Sheng Shi
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Ri-Sheng Liang
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Xian-Kun Tu
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China.
| |
Collapse
|
8
|
Ermutlu İ, Fesli R, Arıkök AT, Ergüder Bİ, Kertmen H, Gürer B. Neuroprotective Effects of Niacin on Ischemia/Reperfusion Injury of the Rabbit Spinal Cord. World Neurosurg 2023; 177:e644-e656. [PMID: 37400055 DOI: 10.1016/j.wneu.2023.06.117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 06/23/2023] [Accepted: 06/24/2023] [Indexed: 07/05/2023]
Abstract
OBJECTIVE Previous studies have shown niacin has neuroprotective effects on the central nervous system. However, its specific effect on spinal cord ischemia/reperfusion injury has not yet been explored. This study aims to evaluate whether niacin can contribute neuroprotective effects on spinal cord ischemia/reperfusion injury. METHODS Rabbits were randomized into 4 groups of 8 animals: group I (control), group II (ischemia), group III (30 mg/kg methylprednisolone, intraperitoneal), and group IV (500 mg/kg niacin, intraperitoneal). The rabbits in group IV were premedicated with niacin for 7 days prior to inducing ischemia/reperfusion injury. The control group was subjected only to a laparotomy, while the remaining groups underwent spinal cord ischemia through a 20-minute occlusion of the aorta caudal to the left renal artery. Following the procedure, levels of catalase, malondialdehyde, xanthine oxidase, myeloperoxidase, and caspase-3 were analyzed. Ultrastructural, histopathological, and neurological evaluations were also performed. RESULTS Spinal cord ischemia/reperfusion injury resulted in increased levels of xanthine oxidase, malondialdehyde, myeloperoxidase, and caspase-3, with a concomitant decrease in catalase levels. Treatment with methylprednisolone and niacin led to decreased levels of xanthine oxidase, malondialdehyde, myeloperoxidase, and caspase-3 and an increase in catalase. Both methylprednisolone and niacin treatments demonstrated improvements in histopathological, ultrastructural, and neurological assessments. CONCLUSIONS Our findings suggest that niacin has antiapoptotic, anti-inflammatory, antioxidant, and neuroprotective effects at least equal to methylprednisolone in ischemia/reperfusion injury of the spinal cord. This study is the first to report the neuroprotective impact of niacin on spinal cord ischemia/reperfusion injury. Further research is warranted to elucidate the role of niacin in this context.
Collapse
Affiliation(s)
- İlçim Ermutlu
- Department of Neurosurgery, Cizre Selahattin Cizrelioğlu City Hospital, Şırnak, Turkey
| | - Ramazan Fesli
- Department of Neurosurgery, Medicalpark Hospital, Mersin, Turkey
| | - Ata Türker Arıkök
- Department of Pathology, Ministry of Health Dışkapı Yıldırım Beyazıt Education and Research Hospital, Ankara, Turkey
| | - Berrin İmge Ergüder
- Department of Biochemistry, Ankara University, Faculty of Medicine, Ankara, Turkey
| | - Hayri Kertmen
- Department of Neurosurgery, Ministry of Health Dışkapı Yıldırım Beyazıt Education and Research Hospital, Ankara, Turkey
| | - Bora Gürer
- Department of Neurosurgery, İstinye University, Faculty of Medicine, Istanbul, Turkey.
| |
Collapse
|
9
|
Duan Q, Wu J. Dihydroartemisinin ameliorates cerebral I/R injury in rats via regulating VWF and autophagy-mediated SIRT1/FOXO1 pathway. Open Med (Wars) 2023; 18:20230698. [PMID: 37415610 PMCID: PMC10320570 DOI: 10.1515/med-2023-0698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 03/06/2023] [Accepted: 03/22/2023] [Indexed: 07/08/2023] Open
Abstract
Dihydroartemisinin (DHA) has been found to inhibit the expression of von Willebrand factor (VWF), a marker of endothelial cell injury, but its mechanism in cerebral ischemia/reperfusion (I/R) injury remains obscure. In this study, I/R model was constructed through middle cerebral artery occlusion (MCAO) in rats, followed by DHA administration. The effect of DHA on rat cerebral I/R injury was investigated by 2,3,5-triphenyltetrazolium chloride staining, hematoxylin and eosin staining, TUNEL staining, and Western blot. Brain microvascular endothelial cells (BMVECs) isolated from newborn rats were exposed to oxygen-glucose deprivation/reoxygenation (OGD/R), and then treated with DHA. The results showed that MCAO treatment induced infarction, nerve cell apoptosis, and brain tissue impairment in rats, which was mitigated by DHA. OGD/R inhibited viability and accelerated apoptosis of BMVECs, which was alleviated by DHA. I/R procedures or OGD/R up-regulated expressions of VWF, ATG7, Beclin1, and LC3-II/LC3-I ratio, while down-regulating Occludin, Claudin-5, ZO-1, P62, SIRT1, and FOXO1 expressions in vivo and in vitro; however, these effects of I/R procedures or OGD/R were offset by DHA. VWF overexpression reversed the above effects of DHA on OGD/R-induced BMVECs. In summary, DHA ameliorates cerebral I/R injury in rats by reducing VWF level and activating autophagy-mediated SIRT1/FOXO1 signaling pathway.
Collapse
Affiliation(s)
- Qi Duan
- Emergency Department, Nantong Rich Hospital, Nantong, Jiangsu, 226006, China
| | - Junxia Wu
- Emergency Department, The Sixth People’s Hospital of Nantong, No. 500 Yonghe Road, Gangzha District, Nantong, Jiangsu, 226000, China
| |
Collapse
|
10
|
Amalia M, Puteri MU, Saputri FC, Sauriasari R, Widyantoro B. Platelet Glycoprotein-Ib (GPIb) May Serve as a Bridge between Type 2 Diabetes Mellitus (T2DM) and Atherosclerosis, Making It a Potential Target for Antiplatelet Agents in T2DM Patients. Life (Basel) 2023; 13:1473. [PMID: 37511848 PMCID: PMC10381765 DOI: 10.3390/life13071473] [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: 05/29/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a persistent metabolic condition that contributes to the development of cardiovascular diseases. Numerous studies have provided evidence that individuals with T2DM are at a greater risk of developing cardiovascular diseases, typically two to four times more likely than those without T2DM, mainly due to an increased risk of atherosclerosis. The rupture of an atherosclerotic plaque leading to pathological thrombosis is commonly recognized as a significant factor in advancing cardiovascular diseases caused by TD2M, with platelets inducing the impact of plaque rupture in established atherosclerosis and predisposing to the primary expansion of atherosclerosis. Studies suggest that individuals with T2DM have platelets that display higher baseline activation and reactivity than those without the condition. The expression enhancement of several platelet receptors is known to regulate platelet activation signaling, including platelet glycoprotein-Ib (GPIb). Furthermore, the high expression of platelet GP1b has been reported to increase the risk of platelet adhesion, platelet-leucocyte interaction, and thrombo-inflammatory pathology. However, the study exploring the role of GP1b in promoting platelet activation-induced cardiovascular diseases in T2DM patients is still limited. Therefore, we summarize the important findings regarding pathophysiological continuity between T2DM, platelet GPIb, and atherosclerosis and highlight the potential therapy targeting GPIb as a novel antiplatelet agent for preventing further cardiovascular incidents in TD2M patients.
Collapse
Affiliation(s)
- Muttia Amalia
- Doctoral Program, Faculty of Pharmacy, Universitas Indonesia, Kampus UI Depok, Depok 16424, Indonesia
| | - Meidi Utami Puteri
- Laboratory of Pharmacology-Toxicology, Faculty of Pharmacy, Universitas Indonesia, Kampus UI Depok, Depok 16424, Indonesia
| | - Fadlina Chany Saputri
- Laboratory of Pharmacology-Toxicology, Faculty of Pharmacy, Universitas Indonesia, Kampus UI Depok, Depok 16424, Indonesia
| | - Rani Sauriasari
- Faculty of Pharmacy, Universitas Indonesia, Kampus UI Depok, Depok 16424, Indonesia
| | - Bambang Widyantoro
- National Cardiovascular Center Harapan Kita, Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Indonesia, Jakarta 11420, Indonesia
| |
Collapse
|
11
|
Wang YH, Long HP, Zhang SX, Liu J, Zhao HQ, Yi J, Linga J. Network pharmacology-based and pharmacological evaluation of the effects of Curcumae Radix on cerebral ischemia–Reperfusion injury. WORLD JOURNAL OF TRADITIONAL CHINESE MEDICINE 2023. [DOI: 10.4103/2311-8571.370154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
|
12
|
Cell Death Mechanisms in Cerebral Ischemia-Reperfusion Injury. Neurochem Res 2022; 47:3525-3542. [PMID: 35976487 DOI: 10.1007/s11064-022-03697-8] [Citation(s) in RCA: 114] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/11/2022] [Accepted: 07/14/2022] [Indexed: 10/15/2022]
Abstract
Ischemic stroke is one of the major causes of morbidity and mortality, affecting millions of people worldwide. Inevitably, the interruption of cerebral blood supply after ischemia may promote a cascade of pathophysiological processes. Moreover, the subsequent restoration of blood flow and reoxygenation may further aggravate brain tissue injury. Although recombinant tissue plasminogen activator (rt-PA) is the only approved therapy for restoring blood perfusion, the reperfusion injury and the narrow therapeutic time window restrict its application for most stroke patients. Increasing evidence indicates that multiple cell death mechanisms are relevant to cerebral ischemia-reperfusion injury, including apoptosis, necrosis, necroptosis, autophagy, pyroptosis, ferroptosis, and so on. Therefore, it is crucial to comprehend various cell death mechanisms and their interactions. In this review, we summarize the various signaling pathways underlying cerebral ischemia-reperfusion injury and elaborate on the crosstalk between the different mechanisms.
Collapse
|
13
|
Khan H, Kaur Grewal A, Gurjeet Singh T. Mitochondrial dynamics related neurovascular approaches in cerebral ischemic injury. Mitochondrion 2022; 66:54-66. [DOI: 10.1016/j.mito.2022.08.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/14/2022] [Accepted: 08/02/2022] [Indexed: 12/30/2022]
|
14
|
Anfibatide alleviates inflammation and apoptosis via inhibiting NF-kappaB/NLRP3 axis in ischemic stroke. Eur J Pharmacol 2022; 926:175032. [PMID: 35584710 DOI: 10.1016/j.ejphar.2022.175032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 04/25/2022] [Accepted: 05/11/2022] [Indexed: 12/29/2022]
Abstract
Recent evidence suggests that Nod-like receptor protein-3 (NLRP3) inflammasome is a key mediator of inflammatory response and can induce the activation of apoptosis signaling pathways in ischemic stroke. In this research, we assessed the effects of anfibatide (ANF) on inflammatory and apoptosis in cerebral ischemic injury and the potential mechanisms. Middle cerebral artery occlusion (MCAO) model was established on male Sprague-Dawley rats to induce cerebral ischemia/reperfusion (I/R) injury in vivo. Primary cortical neurons (PCN) cells were exposed to oxygen-glucose deprivation and reintroduction (OGD/R) to mimic cerebral I/R injury in vitro. The results showed that ANF markedly alleviated infarct volume, neurological deficit and neurobehavioral impairment in MCAO/R rats, enhanced cell viability and decreased LDH release in PCN after OGD/R. The number of TUNEL-positive cells, Bax, cleaved-caspase-3, p-IκBα, p-p65, NLRP3, ASC, cleaved caspase-1, IL-β and IL-18 proteins expression were significantly upregulated in the cortex of MCAO/R rats and PCN exposed to OGD/R, NLRP3 and caspase-1 mRNA levels were also evidently elevated. Bcl-2 protein expression significantly decreased in the cortex of MCAO/R rats. Treatment with ANF obviously inhibited the expression of p-IκBα, p-p65, NLRP3, ASC, cleaved caspase-1, Bax and cleaved-caspase-3, promoted the expression of Bcl-2, then decreased the TUNEL-positive cell number and the level of inflammatory cytokines (IL-β and IL-18) in cerebral ischemia reperfusion in vito and in vitro. Our findings suggest that ANF exerts effects of alleviating inflammation and apoptosis through inhibiting NF-kappaB/NLRP3 axis. ANF is a potential candidate for treating cerebral I/R injury.
Collapse
|
15
|
Protective effects of fraxin on cerebral ischemia-reperfusion injury by mediating neuroinflammation and oxidative stress through PPAR-γ/NF-κB pathway. Brain Res Bull 2022; 187:49-62. [PMID: 35772607 DOI: 10.1016/j.brainresbull.2022.06.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 05/24/2022] [Accepted: 06/25/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND Inflammation and oxidative stress are associated with the pathogenesis of cerebral ischemia-reperfusion (I/R) injury. Fraxin, one of the primary active ingredients of Cortex Fraxini, may have potent anti-inflammatory activity. This study intended to investigate the function and mechanism of fraxin in a middle cerebral artery occlusion (MCAO) model. METHODS A middle cerebral artery occlusion (MCAO) rat model was engineered. Both in-vivo and in-vitro models were dealt with Fraxin. The profiles of inflammation-concerned cytokines, proteins and oxidative stress factors were determined by RT-PCR, western blot, and enzyme-linked immunosorbent assay (ELISA), and neuronal apoptosis and reactive oxygen species (ROS) levels were measured. The neurological functions of rats were evaluated by Morris water maze and modified neurological severity scores (mNSS). RESULTS The data revealed that fraxin abated the OGD/R-mediated release of inflammatory and oxidative stress mediators, enhanced "M2″-like BV2 microglia polarization, and mitigated HT22 cell apoptosis. Mechanistically, fraxin boosted PPAR-γ expression, activated the Nrf2/HO-1 pathway, and suppressed NF-κB, IKK-β,p38 MAPK, ERK1/2 and Keap1 in a dose-dependent manner. Furthermore, attenuating PPAR-γ through pharmacological treatment with GW9662 (a PPAR-γ antagonist) mainly weakened the neuroprotective and anti-inflammatory functions of fraxin. CONCLUSION Fraxin could considerably ameliorate cerebral I/R damage by repressing oxidative stress, inflammatory response, and cell apoptosis through abrogating the PPARγ/ NF-κB pathway.
Collapse
|
16
|
De Meyer SF, Langhauser F, Haupeltshofer S, Kleinschnitz C, Casas AI. Thromboinflammation in Brain Ischemia: Recent Updates and Future Perspectives. Stroke 2022; 53:1487-1499. [PMID: 35360931 DOI: 10.1161/strokeaha.122.038733] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Despite decades of promising preclinical validation and clinical translation, ischemic stroke still remains as one of the leading causes of death and disability worldwide. Within its complex pathophysiological signatures, thrombosis and inflammation, that is, thromboinflammation, are highly interconnected processes leading to cerebral vessel occlusion, inflammatory responses, and severe neuronal damage following the ischemic event. Hence, we here review the most recent updates on thromboinflammatory-dependent mediators relevant after stroke focusing on recent discoveries on platelet modulation, a potential regulation of the innate and adaptive immune system in thromboinflammation, utterly providing a thorough up-to-date overview of all therapeutic approaches currently undergoing clinical trial.
Collapse
Affiliation(s)
- Simon F De Meyer
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Belgium (S.F.D.M.)
| | - Friederike Langhauser
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Germany (F.L., S.H., C.K., A.I.C.)
| | - Steffen Haupeltshofer
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Germany (F.L., S.H., C.K., A.I.C.)
| | - Christoph Kleinschnitz
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Germany (F.L., S.H., C.K., A.I.C.)
| | - Ana I Casas
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Germany (F.L., S.H., C.K., A.I.C.).,Department of Pharmacology and Personalised Medicine, Faculty of Health, Medicine, and Life Sciences, Maastricht University, the Netherlands (A.I.C.)
| |
Collapse
|
17
|
Tsai YT, Huang HC, Kao ST, Chang TT, Cheng CY. Neuroprotective Effects of Alpinia oxyphylla Miq against Mitochondria-Related Apoptosis by the Interactions between Upregulated p38 MAPK Signaling and Downregulated JNK Signaling in the Subacute Phase of Cerebral Ischemia-Reperfusion in Rats. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2022; 50:2057-2083. [DOI: 10.1142/s0192415x22500884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Apoptosis in the penumbra region is the major cell death mechanism occurring during ischemia–reperfusion injury’s early phase. Here, we evaluated how the Alpinia oxyphylla Miq (AOM) affects mitochondria-related apoptosis 3 days after transient middle cerebral artery occlusion (MCAo) and examined the mechanisms underlying the regulation of MAPK-mediated mitochondria-related apoptotic signaling in the peri-infarct cortex in rats. The rats were administered the AOM extract intraperitoneally at doses of 0.2[Formula: see text]g/kg (AOM-0.2[Formula: see text]g), 0.4[Formula: see text]g/kg (AOM-0.4[Formula: see text]g), or 0.8[Formula: see text]g/kg (AOM-0.8[Formula: see text]g) at MCAo initiation. The AOM-0.4[Formula: see text]g and AOM-0.8[Formula: see text]g significantly ameliorated apoptotic cell death and considerably downregulated cytochrome c (cyto c) and cleaved caspase-3 immunoreactivity 3 days after reperfusion. Simultaneously, they significantly downregulated cytosolic p-JNK/JNK, cathepsin B/actin, cyto c/actin, Smac/DIABLO/actin, cleaved caspase-3/actin, and AIF/actin and mitochondrial p53/HSP60 and Bax/HSP60 fractions but upregulated cytosolic p-p38 MAPK/p38 MAPK, p-p90RSK/actin, p-Bad/Bad, p-CREB/actin, and XIAP/actin and cytosolic and mitochondrial Bcl-2/Bax and Bcl-xL/Bax fractions in the peri-infarct cortex. Pretreatment with SB203580 — a p38 MAPK inhibitor — completely abrogated the effects of AOM-0.8[Formula: see text]g on the aforementioned protein expression, whereas treatment with SP600125 — a JNK inhibitor — exerted protective effects similar to those of AOM-0.8[Formula: see text]g. Treatment with 0.4 or 0.8[Formula: see text]g/kg AOM has neuroprotective effects against mitochondria-related apoptosis by suppressing cyto c, Smac/DIABLO, and AIF release from the mitochondria to cytosol. The anti-mitochondria related apoptotic effects of the AOM extract are attributable to the interactions between upregulated p38 MAPK/p90RSK-mediated p-Bad and CREB signaling and downregulated JNK/cathepsin B-mediated Bax and p53 signaling in the peri-infarct cortex 3 days after transient MCAo.
Collapse
Affiliation(s)
- Yueh-Ting Tsai
- School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University Taichung 40402, Taiwan
- Department of Traditional Chinese Medicine, Kuang Tien General Hospital, Taichung, Taiwan
| | - Hui-Chi Huang
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Chinese Medicine, China Medical University Taichung 40402, Taiwan
| | - Shung-Te Kao
- School of Chinese Medicine, College of Chinese Medicine, China Medical University Taichung 40402, Taiwan
| | - Tung-Ti Chang
- School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University Taichung 40402, Taiwan
| | - Chin-Yi Cheng
- School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University Taichung 40402, Taiwan
- Department of Chinese Medicine, Hui-Sheng Hospital, Taichung 42056, Taiwan
- Department of Chinese Medicine, China Medical University Hospital, Taichung 42056, Taiwan
| |
Collapse
|
18
|
MicroRNA-21-5p Reduces Hypoxia/Reoxygenation-Induced Neuronal Cell Damage through Negative Regulation of CPEB3. Anal Cell Pathol (Amst) 2021; 2021:5543212. [PMID: 34900520 PMCID: PMC8660214 DOI: 10.1155/2021/5543212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 09/04/2021] [Accepted: 11/07/2021] [Indexed: 11/26/2022] Open
Abstract
Objectives To explore the role of microRNA-21-5p (miR-21-5p) in hypoxia/reoxygenation- (H/R-) induced HT22 cell damage. Methods The hypoxia/reoxygenation (H/R) model was established in mouse neuronal cells HT22. Cell Counting Kit-8 (CCK-8) and qRT-PCR were used to determine the effects of H/R treatment on cell viability and miR-21-5p expression. HT22 cells were transfected with miR-21-5p mimic or negative control (NC) followed by the induction of H/R; cell viability, apoptosis, and SOD, MDA, and LDH activities were detected. Besides, the apoptosis-related proteins including BAX, BCL2, cleaved caspase-3, and caspase-3 as well as proteins of EGFR/PI3K/AKT signaling pathways were measured by Western blot. To verify the target relation between cytoplasmic polyadenylation element binding protein 3 (CPEB3) and miR-21-5p, luciferase reporter gene experiment was performed. After cotransfection with miR-21-5p mimic and CPEB3 plasmids, the reversal effects of CPEB3 on miR-21-5p in H/R damage were studied. Results H/R treatment could significantly reduce the cell viability (P < 0.05) and miR-21-5p levels (P < 0.05) in HT22 cells. After overexpressing miR-21-5p, cell viability was increased (P < 0.05) under H/R treatment, and the apoptosis rate and the levels of apoptosis-related proteins were suppressed (all P < 0.05). Furthermore, SOD activity was increased (P < 0.05), while MDA and LDH activity was decreased (both P < 0.05). Besides, miR-21-5p could restore the activation of the EGFR/PI3K/AKT signaling pathway inhibited by H/R treatment (all P < 0.05). The luciferase reporter gene experiment verified that CPEB3 is the target of miR-21-5p (P < 0.05). When coexpressing miR-21-5p mimic and CPEB3 in the cells, the protective effects of miR-21-5p under H/R were reversed (all P < 0.05), and the activation of the EGFR/PI3K/AKT pathway was also inhibited (all P < 0.05). Conclusion This study showed that miR-21-5p may regulate the EGFR/PI3K/AKT signaling pathway by targeting CPEB3 to reduce H/R-induced cell damage and apoptosis.
Collapse
|
19
|
Su G, Qu Y, Li G, Deng M. Sevoflurane protects against cerebral ischemia/reperfusion injury via microrna-30c-5p modulating homeodomain-interacting protein kinase 1. Bioengineered 2021; 12:11858-11871. [PMID: 34709114 PMCID: PMC8810137 DOI: 10.1080/21655979.2021.1999551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/22/2021] [Accepted: 10/23/2021] [Indexed: 12/21/2022] Open
Abstract
Sevoflurane (SEV) has been reported to be an effective neuroprotective agent for cerebral ischemia/reperfusion injury (CIRI). However, the precise molecular mechanisms of Sev preconditioning in CIRI remain largely unknown. Therefore, CIRI model was established via middle cerebral artery occlusion method. SEV was applied before modeling. after successful modeling, lentivirus was injected into the lateral ventricle of the brain. Neurological impairment score was performed in each group, and histopathologic condition, infarct volume, apoptosis, inflammation, oxidative stress, microRNA (miR)-30 c-5p and homeodomain-interacting protein kinase 1 (HIPK1) were detected. Mouse hippocampal neuronal cell line HT22 cells were pretreated with SEV, and the in vitro model was stimulated via oxygen-glucose deprivation and reoxygenation. The corresponding plasmids were transfected, and the cell growth was detected, including inflammation and oxidative stress, etc. The targeting of miR-30 c-5p with HIPK1 was examined. The results clarified that reduced miR-30 c-5p and elevated HIPK1 were manifested in CIRI. SEV could improve CIRI and modulate the miR-30 c-5p-HIPK1 axis in vitro and in vivo, and miR-30 c-5p could target HIPK1. Depressed miR-30 c-5p could eliminate the protection of SEV in vitro and in vivo. Repression of HIPK1 reversed the effect of reduced miR-30 c-5p on CIRI. Therefore, it is concluded that SEV is available to depress CIRI via targeting HIPK1 through upregulated miR-30 c-5p.
Collapse
Affiliation(s)
- Guoning Su
- Department of Anesthesia, Affiliated Hospital of Yunnan University, KunMing City, China
| | - Yan Qu
- Department of Anesthesia, Affiliated Hospital of Yunnan University, KunMing City, China
| | - Gang Li
- Department of Anesthesia, Affiliated Hospital of Yunnan University, KunMing City, China
| | - Min Deng
- Department of Anesthesia, Affiliated Hospital of Yunnan University, KunMing City, China
| |
Collapse
|
20
|
Buhlman LM, Krishna G, Jones TB, Thomas TC. Drosophila as a model to explore secondary injury cascades after traumatic brain injury. Biomed Pharmacother 2021; 142:112079. [PMID: 34463269 PMCID: PMC8458259 DOI: 10.1016/j.biopha.2021.112079] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/11/2021] [Accepted: 08/17/2021] [Indexed: 12/14/2022] Open
Abstract
Drosophilae are emerging as a valuable model to study traumatic brain injury (TBI)-induced secondary injury cascades that drive persisting neuroinflammation and neurodegenerative pathology that imposes significant risk for long-term neurological deficits. As in mammals, TBI in Drosophila triggers axonal injury, metabolic crisis, oxidative stress, and a robust innate immune response. Subsequent neurodegeneration stresses quality control systems and perpetuates an environment for neuroprotection, regeneration, and delayed cell death via highly conserved cell signaling pathways. Fly injury models continue to be developed and validated for both whole-body and head-specific injury to isolate, evaluate, and modulate these parallel pathways. In conjunction with powerful genetic tools, the ability for longitudinal evaluation, and associated neurological deficits that can be tested with established behavioral tasks, Drosophilae are an attractive model to explore secondary injury cascades and therapeutic intervention after TBI. Here, we review similarities and differences between mammalian and fly pathophysiology and highlight strategies for their use in translational neurotrauma research.
Collapse
Affiliation(s)
- Lori M Buhlman
- Biomedical Sciences Program, Midwestern University, Glendale, AZ, USA.
| | - Gokul Krishna
- Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA; Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA
| | - T Bucky Jones
- Department of Anatomy, Midwestern University, Glendale, AZ, USA
| | - Theresa Currier Thomas
- Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA; Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA; Phoenix VA Health Care System, Phoenix, AZ, USA.
| |
Collapse
|
21
|
Cheng CY, Chiang SY, Kao ST, Huang SC. Alpinia oxyphylla Miq extract reduces cerebral infarction by downregulating JNK-mediated TLR4/T3JAM- and ASK1-related inflammatory signaling in the acute phase of transient focal cerebral ischemia in rats. Chin Med 2021; 16:82. [PMID: 34419138 PMCID: PMC8379872 DOI: 10.1186/s13020-021-00495-2] [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: 05/10/2021] [Accepted: 08/13/2021] [Indexed: 12/03/2022] Open
Abstract
Background Post-ischemic inflammation is a crucial component in stroke pathology in the early phase of cerebral ischemia–reperfusion (I/R) injury. Inflammation caused by microglia, astrocytes, and necrotic cells, produces pro-inflammatory mediators and exacerbates cerebral I/R injury. This study evaluated the effects of the Alpinia oxyphylla Miq [Yi Zhi Ren (YZR)] extract on cerebral infarction at 1 day after 90 min of transient middle cerebral artery occlusion (MCAo) and investigated the molecular mechanisms underlying the regulation of c-Jun N-terminal kinase (JNK)-mediated inflammatory cascades in the penumbral cortex. Rats were intraperitoneally injected with the YZR extract at the doses of 0.2 g/kg (YZR-0.2 g), 0.4 g/kg (YZR-0.4 g), or 0.8 g/kg (YZR-0.8 g) at MCAo onset. Results YZR-0.4 g and YZR-0.8 g treatments markedly reduced cerebral infarction, attenuated neurological deficits, and significantly downregulated the expression of phospho-apoptosis signal-regulating kinase 1 (p-ASK1)/ASK1, tumor necrosis factor receptor-associated factor 3 (TRAF3), TRAF3-interacting JNK-activating modulator (T3JAM), ionized calcium-binding adapter molecule 1 (Iba1), p-JNK/JNK, inducible nitric oxide synthase, cyclooxygenase-2, tumor necrosis factor-α, toll-like receptor 4 (TLR4), glial fibrillary acidic protein (GFAP), nuclear factor-kappa B (NF-κB), and interleukin-6 in the penumbral cortex at 1 day after reperfusion. SP600125 (SP), a selective JNK inhibitor, had the same effects. Furthermore, Iba1- and GFAP-positive cells were colocalized with TLR4, and colocalization of GFAP-positive cells was found with NF-κB in the nuclei. Conclusion YZR-0.4 g and YZR-0.8 g treatments exerted beneficial effects on cerebral ischemic injury by downregulating JNK-mediated signaling in the peri-infarct cortex. Moreover, the anti-infarction effects of YZR extract treatments were partially attributed to the downregulation of JNK-mediated TLR4/T3JAM- and ASK1-related inflammatory signaling pathways in the penumbral cortex at 1 day after reperfusion.
Collapse
Affiliation(s)
- Chin-Yi Cheng
- School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan.,Department of Chinese Medicine, Hui-Sheng Hospital, Taichung, 42056, Taiwan
| | - Su-Yin Chiang
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan
| | - Shung-Te Kao
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan
| | - Shang-Chih Huang
- Department of Neurology, China Medical University Hospital, Taichung City, 40447, Taiwan.
| |
Collapse
|
22
|
Javadinia SS, Abbaszadeh-Goudarzi K, Mahdian D, Hosseini A, Ghalenovi M, Javan R. A review of the protective effects of quercetin-rich natural compounds for treating ischemia-reperfusion injury. Biotech Histochem 2021; 97:237-246. [PMID: 34157912 DOI: 10.1080/10520295.2021.1937701] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Ischemia-reperfusion (IR) injury causes dysfunction of tissues and organs, and oxidative stress plays an important role. During IR, reactive oxygen species (ROS) are increased. Antioxidants are used to decrease ROS associated with IR. We review the protective effects of quercetin-rich natural antioxidants against IR. We searched PubMed, ScienceDirect, Scopus and Cochrane databases using the keywords: ischemic reperfusion, quercetin, antioxidant and herbal medicine. The effects of quercetin during IR have been reported for animal models in vitro and in vivo. Quercetin-rich plants including Abelmoschus esculentus, coriander, Hypericum perforatum, onion, Psidium guajava, buckwheat and Rosa laevigata Michx have been used to reduce oxidative stress damage to various organs during IR.
Collapse
Affiliation(s)
- Sara Sadat Javadinia
- Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Kazem Abbaszadeh-Goudarzi
- Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran.,Leishmaniasis Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Davood Mahdian
- Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran.,Department of Pharmacology, School of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Azar Hosseini
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mina Ghalenovi
- Faculty of Midwifery, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Roghayeh Javan
- Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| |
Collapse
|
23
|
Li BX, Dai X, Xu XR, Adili R, Neves MAD, Lei X, Shen C, Zhu G, Wang Y, Zhou H, Hou Y, Ni T, Pasman Y, Yang Z, Qian F, Zhao Y, Gao Y, Liu J, Teng M, Marshall AH, Cerenzia EG, Li ML, Ni H. In vitro assessment and phase I randomized clinical trial of anfibatide a snake venom derived anti-thrombotic agent targeting human platelet GPIbα. Sci Rep 2021; 11:11663. [PMID: 34083615 PMCID: PMC8175443 DOI: 10.1038/s41598-021-91165-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 05/18/2021] [Indexed: 12/29/2022] Open
Abstract
The interaction of platelet GPIbα with von Willebrand factor (VWF) is essential to initiate platelet adhesion and thrombosis, particularly under high shear stress conditions. However, no drug targeting GPIbα has been developed for clinical practice. Here we characterized anfibatide, a GPIbα antagonist purified from snake (Deinagkistrodon acutus) venom, and evaluated its interaction with GPIbα by surface plasmon resonance and in silico modeling. We demonstrated that anfibatide interferds with both VWF and thrombin binding, inhibited ristocetin/botrocetin- and low-dose thrombin-induced human platelet aggregation, and decreased thrombus volume and stability in blood flowing over collagen. In a single-center, randomized, and open-label phase I clinical trial, anfibatide was administered intravenously to 94 healthy volunteers either as a single dose bolus, or a bolus followed by a constant rate infusion of anfibatide for 24 h. Anfibatide inhibited VWF-mediated platelet aggregation without significantly altering bleeding time or coagulation. The inhibitory effects disappeared within 8 h after drug withdrawal. No thrombocytopenia or anti-anfibatide antibodies were detected, and no serious adverse events or allergic reactions were observed during the studies. Therefore, anfibatide was well-tolerated among healthy subjects. Interestingly, anfibatide exhibited pharmacologic effects in vivo at concentrations thousand-fold lower than in vitro, a phenomenon which deserves further investigation.Trial registration: Clinicaltrials.gov NCT01588132.
Collapse
Affiliation(s)
- Benjamin Xiaoyi Li
- Lee's Pharmaceutical Holdings Limited, 1/F, Building 20E, Phase 3, Hong Kong Science Park, Shatin, N.T. Hong Kong SAR, China. .,Zhaoke Pharmaceutical Co. Limited, Hefei, China.
| | - Xiangrong Dai
- Lee's Pharmaceutical Holdings Limited, 1/F, Building 20E, Phase 3, Hong Kong Science Park, Shatin, N.T. Hong Kong SAR, China.,Zhaoke Pharmaceutical Co. Limited, Hefei, China
| | - Xiaohong Ruby Xu
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada.,Toronto Platelet Immunobiology Group, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Reheman Adili
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada.,Toronto Platelet Immunobiology Group, Toronto, Canada
| | - Miguel Antonio Dias Neves
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada.,Toronto Platelet Immunobiology Group, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.,Canadian Blood Services Centre for Innovation, Toronto, Canada
| | - Xi Lei
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada.,Toronto Platelet Immunobiology Group, Toronto, Canada
| | - Chuanbin Shen
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada.,Toronto Platelet Immunobiology Group, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Guangheng Zhu
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada.,Toronto Platelet Immunobiology Group, Toronto, Canada
| | - Yiming Wang
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada.,Toronto Platelet Immunobiology Group, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.,Canadian Blood Services Centre for Innovation, Toronto, Canada
| | - Hui Zhou
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada.,Toronto Platelet Immunobiology Group, Toronto, Canada
| | - Yan Hou
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada.,Toronto Platelet Immunobiology Group, Toronto, Canada
| | - Tiffany Ni
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada.,Toronto Platelet Immunobiology Group, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Yfke Pasman
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada.,Toronto Platelet Immunobiology Group, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.,Canadian Blood Services Centre for Innovation, Toronto, Canada
| | | | - Fang Qian
- Zhaoke Pharmaceutical Co. Limited, Hefei, China
| | - Yanan Zhao
- Wannan Medical College First Affiliated Hospital, Yijishan Hospital, Wuhu, China
| | - Yongxiang Gao
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Jing Liu
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Maikun Teng
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Alexandra H Marshall
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada.,Toronto Platelet Immunobiology Group, Toronto, Canada
| | - Eric G Cerenzia
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada.,Toronto Platelet Immunobiology Group, Toronto, Canada.,Department of Physiology, University of Toronto, Toronto, Canada
| | - Mandy Lokyee Li
- Lee's Pharmaceutical Holdings Limited, 1/F, Building 20E, Phase 3, Hong Kong Science Park, Shatin, N.T. Hong Kong SAR, China
| | - Heyu Ni
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada. .,Toronto Platelet Immunobiology Group, Toronto, Canada. .,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada. .,Canadian Blood Services Centre for Innovation, Toronto, Canada. .,Department of Physiology, University of Toronto, Toronto, Canada. .,Department of Medicine, University of Toronto, Toronto, Canada. .,St. Michael's Hospital, Room 421, LKSKI-Keenan Research Centre, 209 Victoria Street, Toronto, ON, M5B 1W8, Canada.
| |
Collapse
|
24
|
Liu H, Zhao Z, Wu T, Zhang Q, Lu F, Gu J, Jiang T, Xue J. Inhibition of autophagy-dependent pyroptosis attenuates cerebral ischaemia/reperfusion injury. J Cell Mol Med 2021; 25:5060-5069. [PMID: 33938129 PMCID: PMC8178262 DOI: 10.1111/jcmm.16483] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/03/2021] [Accepted: 02/20/2021] [Indexed: 12/17/2022] Open
Abstract
Autophagy is closely associated with cerebral ischaemia/reperfusion injury, but the underlying mechanisms are unknown. We investigated whether Spautin-1 ameliorates cerebral ischaemia/reperfusion injury by inhibiting autophagy and whether its derived pyroptosis is involved in this process. We explored the mechanism of Spautin-1 in cerebral ischaemia/reperfusion. To answer these questions, healthy male Sprague-Dawley rats were exposed to middle cerebral artery occlusion for 60 minutes followed by reperfusion for 24 hours. We found that cerebral ischaemia/reperfusion increased the expression levels of autophagy and pyroptosis-related proteins. Treatment with Spautin-1 reduced the infarct size and water content and restored some neurological functions. In vitro experiments were performed using oxygen-glucose deprivation/reoxygenation to model PC12 cells. The results showed that PC12 cells showed a significant decrease in cell viability and a significant increase in ROS and autophagy levels. Spautin-1 treatment reduced autophagy and ROS accumulation and attenuated NLRP3 inflammasome-dependent pyroptosis. However, these beneficial effects were greatly blocked by USP13 overexpression, which significantly counteracted the inhibition of autophagy and NLRP3 inflammasome-dependent ferroptosis by Spautin-1. Together, these results suggest that Spautin-1 may ameliorate cerebral ischaemia-reperfusion injury via the autophagy/pyroptosis pathway. Thus, inhibition of autophagy may be considered as a promising therapeutic approach for cerebral ischaemia-reperfusion injury.
Collapse
Affiliation(s)
- Hui Liu
- Department of Neurology and Institute of NeurologyThe Affiliated Changshu Hospital of Xuzhou Medical SchoolSuzhouJiangsuChina
| | - Zongbo Zhao
- Department of Neurology and Institute of NeurologyThe Affiliated Changshu Hospital of Xuzhou Medical SchoolSuzhouJiangsuChina
| | - Tao Wu
- Department of Neurology and Institute of NeurologyThe Affiliated Changshu Hospital of Xuzhou Medical SchoolSuzhouJiangsuChina
| | - Qiu Zhang
- Department of Neurology and Institute of NeurologyThe Affiliated Changshu Hospital of Xuzhou Medical SchoolSuzhouJiangsuChina
| | - Fenying Lu
- Department of GastroenterologyThe Affiliated Changshu Hospital of Xuzhou Medical SchoolSuzhouJiangsuChina
| | - Jie Gu
- Department of Key LaboratoryThe Affiliated Changshu Hospital of Xuzhou Medical SchoolSuzhouJiangsuChina
| | - Tingwang Jiang
- Department of Key LaboratoryThe Affiliated Changshu Hospital of Xuzhou Medical SchoolSuzhouJiangsuChina
- Jiangsu Key Laboratory of Brain Disease BioinformationXuzhou Medical UniversityXuzhouJiangsuChina
| | - Jianzhong Xue
- Department of Neurology and Institute of NeurologyThe Affiliated Changshu Hospital of Xuzhou Medical SchoolSuzhouJiangsuChina
| |
Collapse
|
25
|
Yang C, Xu Y, Zhang W, Ma M, Wang S, Chai L, Guo H, Hu L. Salvianolate lyophilized injection regulates the autophagy-lysosomal pathway in cerebral ischaemia/reperfusion rats. JOURNAL OF ETHNOPHARMACOLOGY 2021; 271:113898. [PMID: 33556476 DOI: 10.1016/j.jep.2021.113898] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 12/24/2020] [Accepted: 01/28/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Activation of autophagy has been implicated in cerebral ischiemia/reperfusion (I/R) injury. Salvianolate lyophilized injection (SLI) has been widely used in the clinical treatment of cerebrovascular disease in China. Whether SLI has any influence on the activation of autophagy in cerebral I/R injury remains elusive. AIM OF THE STUDY The aim of this study were to assess whether SLI attenuates I/R-induced brain injury and evaluate its associated mechanisms. MATERIALS AND METHODS Focal cerebral ischaemia was induced by middle cerebral artery occlusion (MCAO). SLI (21 mg/kg) was injected intravenously at the beginning of the reperfusion period and 24 and 48 h after ischaemia. The effects of SLI on brain injury were detected according to infarct volume, neurological score, brain oedema, and HE and TUNEL staining at 72 h post-MCAO. Western blotting was used to detect alterations in the autophagy-relevant proteins LC3, Beclin-1, mTOR, p62, Lamp-1, and CTSD in the ipsilateral cortex at 24 or 72 h post-MCAO. RESULTS We first demonstrated that SLI significantly alleviated the infarct volume, neurological deficits, and brain oedema, and reduced the number of TUNEL-positive cells in rats with cerebral I/R injury. Next, we found that SLI has a bidirectional regulatory effect on autophagy: early-stage (24 h) cerebral ischaemia promotes the activation of autophagy and developmental-stage (72 h) cerebral ischaemia has an inhibitory effect. SLI enhanced I/R-induced autophagy as evidenced by the increased expression level of the autophagy marker protein LC3Ⅱ, as well as the decreased expression of mTOR and the autophagy substrate protein p62, but there was no change in lysosomal activity at 24 h after I/R-induced injury. Moreover, SLI also inhibited excessive activation of autophagy at 72 h after I/R-induced injury, which manifested as downregulating LC3Ⅱ expression, upregulating mTOR and p62 expression, and inhibiting lysosomal activity. CONCLUSION SLI has a protective effect on cerebral ischaemia/reperfusion injury, which may be mediated by the autophagy-lysosome pathway.
Collapse
Affiliation(s)
- Changshuo Yang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, #10 Boyanghu Road, Jinghai District, Tianjin, 301617, China
| | - Yangyang Xu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, #10 Boyanghu Road, Jinghai District, Tianjin, 301617, China
| | - Wenqi Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, #10 Boyanghu Road, Jinghai District, Tianjin, 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Mengmeng Ma
- Beijing Northen Hospital of Weaponry Industry, #10 CheDaoGou, HaiDian District, Beijing, 100089, China
| | - Shaoxia Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, #10 Boyanghu Road, Jinghai District, Tianjin, 301617, China
| | - Lijuan Chai
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, #10 Boyanghu Road, Jinghai District, Tianjin, 301617, China
| | - Hong Guo
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, #10 Boyanghu Road, Jinghai District, Tianjin, 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Limin Hu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, #10 Boyanghu Road, Jinghai District, Tianjin, 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
26
|
Yu G, Sun W, Wang W, Le C, Liang D, Shuai L. Overexpression of microRNA-202-3p in bone marrow mesenchymal stem cells improves cerebral ischemia-reperfusion injury by promoting angiogenesis and inhibiting inflammation. Aging (Albany NY) 2021; 13:11877-11888. [PMID: 33893248 PMCID: PMC8109138 DOI: 10.18632/aging.202889] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 02/27/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND Cerebral ischemia-reperfusion injury (CIRI) can cause brain tissue inflammation, neuronal degeneration, and apoptosis. There is increasing evidence that microRNAs (miRNA) exert neuroprotective effects by regulating the inflammatory process during cerebral ischemia-reperfusion injury. Additionally, it is increasingly acknowledged that neuroinflammation is regulated by Toll-like receptor 4 (TLR4). However, it is unclear whether miRNA can exert its neuroprotective effects by regulating TLR4-mediated inflammation. METHODS The effects of BMSCs over-expressing miR-202-3p on CIRI, angiogenesis in midbrain tissue, and the release of inflammatory factors (IFs) in the serum were measured using in vivo rat models. We also used SH-SY5Y cells to establish an ischemia-reperfusion in vitro cell model. The interaction between miR-202-3p and TLR4 was analyzed by overexpressing miR-202-3p and knocking down TLR4. Knockdown of TLR4 was performed using siRNA. RESULTS Overexpression of miR-202-3p in BMSCs could significantly improve brain function and reduce brain damage. Simultaneously, miR-202-3p could significantly promote angiogenesis, increase the expression of vWF and VEGF, and reduce the expression of IFs. When the expression of TLR4 was significantly reduced in SH-SY5Y cells, the expression of IFs increased. Therefore, miRNA-202-3p may interact with TLR4 to modulate inflammation. CONCLUSION Our data indicated that miR-202-3p potentially exerts its neuroprotective effects and protects against CIRI by regulating TLR4-mediated inflammation.
Collapse
Affiliation(s)
- Guohua Yu
- Department of Rehabilitation, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Weiming Sun
- Department of Rehabilitation, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Wansong Wang
- Department of Rehabilitation, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Changhao Le
- Department of Rehabilitation, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Dehuan Liang
- Department of Rehabilitation, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Lang Shuai
- Department of Rehabilitation, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| |
Collapse
|
27
|
Tan L, Liu L, Yao J, Piao C. miR-145-5p attenuates inflammatory response and apoptosis in myocardial ischemia-reperfusion injury by inhibiting (NADPH) oxidase homolog 1. Exp Anim 2021; 70:311-321. [PMID: 33658472 PMCID: PMC8390312 DOI: 10.1538/expanim.20-0160] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Myocardial ischemia-reperfusion (I/R) injury is a common complication following reperfusion therapy that involves a series of immune or apoptotic reactions. Studies have revealed the potential roles of miRNAs in I/R injury. Herein, we established a myocardial I/R model in rats and a hypoxia/reoxygenation (H/R) model in H9c2 cells and investigated the effect of miR-145-5p on myocardial I/R injury. After 3 h or 24 h of reperfusion, left ventricular end-systolic pressure (LVESP), ejection fraction (EF), and fractional shortening (FS) were obviously decreased, and left ventricular end-diastolic pressure (LVEDP) was increased. Meanwhile, I/R induced an increase in myocardial infarction area. Moreover, a decrease in miR-145-5p and increase in (NADPH) oxidase homolog 1 (NOH-1) were observed following I/R injury. With this in mind, we performed a luciferase reporter assay and demonstrated that miR-145-5p directly bound to NOH-1 3' untranslated region (UTR). Furthermore, miR-145-5p mimics decreased the levels of tumor necrosis factor (TNF)-α, IL-1β, and IL-6 via oxygen and glucose deprivation/reperfusion (OGD/R) stimulation. Upregulation of miR-145-5p increased cell viability and reduced apoptosis accompanied by downregulation of Bax, cleaved caspase-3, cleaved poly(ADP-ribose) polymerase (PARP) and upregulation of Bcl2. In addition, miR-145-5p overexpression increased superoxide dismutase (SOD) activity and reduced reactive oxygen species (ROS) and malondialdehyde (MDA) content under OGD/R stress. Notably, NOH-1 could significantly abrogate the above effects, suggesting that it is involved in miR-145-5p-regulated I/R injury. In summary, our findings indicated that miR-145-5p/NOH-1 has a protective effect on myocardial I/R injury by inhibiting the inflammatory response and apoptosis.
Collapse
Affiliation(s)
- Lili Tan
- Department of Cardiology, The Second Affiliated Hospital of Shenyang Medical College, 20 Beijiu Road, Shenyang 110002, P.R. China
| | - Limin Liu
- Department of Cardiology, The Second Affiliated Hospital of Shenyang Medical College, 20 Beijiu Road, Shenyang 110002, P.R. China
| | - Jian Yao
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Shenyang Medical College, 20 Beijiu Road, Shenyang 110002, P.R. China
| | - Chenghao Piao
- Department of Cardiology, The Second Affiliated Hospital of Shenyang Medical College, 20 Beijiu Road, Shenyang 110002, P.R. China
| |
Collapse
|
28
|
Lu F, Wang L, Chen Y, Zhong X, Huang Z. In vitro cultured calculus bovis attenuates cerebral ischaemia-reperfusion injury by inhibiting neuronal apoptosis and protecting mitochondrial function in rats. JOURNAL OF ETHNOPHARMACOLOGY 2020; 263:113168. [PMID: 32730869 DOI: 10.1016/j.jep.2020.113168] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 05/18/2020] [Accepted: 07/07/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In vitro cultured calculus bovis (ICCB), which is produced based on the formation mechanism of bovine gallstones, is used to replace the natural bezoar. It has been used in the clinic to treat brain diseases, including stroke, Alzheimer's disease and depression. AIM OF STUDY ICCB is used to treat encephalopathy in the clinic. We explored the effects of ICCB on cerebral ischaemia-reperfusion injury (CIRI) and the potential associated mechanisms. MATERIALS AND METHODS Rats were subjected to middle cerebral artery occlusion for 90 min, followed by 24 h of reperfusion, after being given different concentrations of ICCB once a day for 3 days. Subsequently, the neurological scores, brain oedema and volume of cerebral infarction were measured, and the histopathological changes in the cortex neurons were observed by haematoxylin and eosin staining (H&E). Apoptosis was determined by terminal deoxynucleotidyl transferase-mediated dUTP nick end labelling (TUNEL). Ultrastructural changes in the mitochondria of the cortex were assessed by transmission electron microscopy (TEM). The apoptosis-related proteins Bax, Bcl-2, caspase-9, caspase-3, Mito-Cyt C and Cyto-Cyt C were detected by Western blotting. RESULTS Compared with those in the control group, the neurological scores, the volumes of cerebral infarction, and the brain water contents were significantly decreased in the ICCB groups at doses of 50 and 100 mg/kg. The ICCB treatment effectively decreased the neuronal apoptosis resulting from the CIRI-induced neuron injury. In addition, the histopathological damage and the mitochondria ultrastructure injury were partially improved in the CIRI rats after ICCB treatment. Western blotting analysis indicated that ICCB significantly decreased the expression of Bax, caspase-9, caspase-3 and Cyto-Cyt C protein levels while increasing the expression of Bcl-2 and Mito-Cyt C protein levels. CONCLUSION The ICCB protected against CIRI by suppressing the mitochondria-mediated apoptotic signalling pathway.
Collapse
Affiliation(s)
- Feibiao Lu
- Institute of Traditional Chinese Medicine Resources, College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 311400, PR China
| | - Lingfeng Wang
- Institute of Traditional Chinese Medicine Resources, College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 311400, PR China
| | - Yanyue Chen
- Institute of Traditional Chinese Medicine Resources, College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 311400, PR China
| | - Xiaoming Zhong
- Institute of Traditional Chinese Medicine Resources, College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 311400, PR China.
| | - Zhen Huang
- Institute of Traditional Chinese Medicine Resources, College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 311400, PR China.
| |
Collapse
|
29
|
Chen X, Yao Z, Peng X, Wu L, Wu H, Ou Y, Lai J. Eupafolin alleviates cerebral ischemia/reperfusion injury in rats via blocking the TLR4/NF‑κB signaling pathway. Mol Med Rep 2020; 22:5135-5144. [PMID: 33173992 PMCID: PMC7646971 DOI: 10.3892/mmr.2020.11637] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 09/11/2020] [Indexed: 12/20/2022] Open
Abstract
Eupatorium perfoliatum L. (E. perfoliatium) has been used traditionally for treating fever, malaria and inflammation‑associated diseases. Eupafolin, the extract of E. perfoliatium, was also reported to suppress inflammation. The present study aimed to investigate the protective effects of eupafolin on cerebral ischemia/reperfusion (I/R) injury in rats and its possible underlying mechanisms. Cerebral I/R injury was induced in rats by middle cerebral artery occlusion (MCAO) for 1.5 h, followed by reperfusion. The rats were randomly assigned into six groups: Control, model, 10 mg/kg eupafolin, 20 mg/kg eupafolin, 50 mg/kg eupafolin and 20 mg/kg nimodipine. Eupafolin and nimodipine were intragastrically administrated to the rats 1 week before MCAO induction. Following reperfusion for 24 h, the neurological deficit was scored, and brain samples were harvested for evaluating encephaledema, infarct volume, oxidative stress, apoptosis, inflammation and the expression of TLR4/NF‑κB signaling. The results revealed that eupafolin decreased the neurological score, relieved encephaledema and decreased infarct volume. Eupafolin also attenuated oxidative stress, neuronal apoptosis and inflammation, with decreases in lactate dehydrogenase, malondialdehyde, TUNEL‑positive cells, Bax and caspase‑3, along with TNF‑α, IL‑1β and IL‑6, but increases in superoxide dismutase and Bcl‑2 levels. Furthermore, eupafolin may decrease the expression of TLR4 downstream proteins and proteins involved in the NF‑κB pathway. Treatment with TLR4 agonist‑LPS significantly blunted the protective effect of eupafolin on encephaledema and cerebral infarct. Meanwhile, 20 mg/kg eupafolin showed nearly equivalent effects to the positive‑control drug nimodipine. In conclusion, eupafolin protected against cerebral I/R injury in rats and the underlying mechanism may be associated with the suppression of apoptosis and inflammation via inhibiting the TLR4/ NF‑κB signaling pathway.
Collapse
Affiliation(s)
- Xingwang Chen
- Department of Intensive Care Unit, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, Guangdong 518104, P.R. China
| | - Zhijun Yao
- Department of Intensive Care Unit, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, Guangdong 518104, P.R. China
| | - Xian Peng
- Department of Intensive Care Unit, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, Guangdong 518104, P.R. China
| | - Long Wu
- Department of Intensive Care Unit, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, Guangdong 518104, P.R. China
| | - Huachu Wu
- Department of Intensive Care Unit, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, Guangdong 518104, P.R. China
| | - Yuantong Ou
- Department of Intensive Care Unit, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, Guangdong 518104, P.R. China
| | - Jianbo Lai
- Department of Intensive Care Unit, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, Guangdong 518104, P.R. China
| |
Collapse
|
30
|
Wang J, Zhang W, Ma B, Zhang H, Fan Z, Li M, Li X. A novel biscoumarin derivative dephosphorylates ERK and alleviates apoptosis induced by mitochondrial oxidative damage in ischemic stroke mice. Life Sci 2020; 264:118499. [PMID: 33141045 DOI: 10.1016/j.lfs.2020.118499] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 09/15/2020] [Accepted: 09/21/2020] [Indexed: 11/17/2022]
Abstract
AIM We previously reported the protective effects of biscoumarin derivatives against oxidative stress, but effects of the derivative on mitochondrial oxidative damage induced apoptosis in ischemic stroke remains unknown. METHODS Primary neurons were subjected to oxygen and glucose deprivation (OGD) for the in vitro simulation of ischemic stroke, and an ischemic stroke model was established in mice by operation of middle cerebral artery occlusion (MCAO). RESULTS The results indicated that the nontoxic concentration range of biscoumarin derivative Comp. B in neurons was from 0 to 30 μg/ml and the optimal protective concentration was 20 μg/ml. Treatment with Comp. B increased the cell survival rate and alleviated mitochondrial oxidative damage and apoptosis in OGD-treated neurons. Comp. B reduced the ratio of Bax/Bcl-2, inhibited the phosphorylation of ERK, and thus alleviated apoptosis in OGD-treated neurons. Further research demonstrated that the dephosphorylation effect on ERK of Comp. B is a key factor in alleviating apoptosis in neurons induced by OGD injury. Furthermore, Comp. B reduced the infarct volume, improved neurobehavioural score, and alleviated morphological changes and brain apoptosis in MCAO mice. CONCLUSION The novel biscoumarin derivative Comp. B alleviates mitochondrial oxidative damage and apoptosis in ischemic stroke mice. These findings might provide new insights that will aid in elucidating the effect of biscoumarin derivative against cerebral ischemic reperfusion injury and support the new development of Comp. B as a potential treatment for ischemic stroke.
Collapse
Affiliation(s)
- Jun Wang
- Department of Digestive Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Wentong Zhang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China
| | - Bo Ma
- Department of Pharmacology, The Fourth Military Medical University, Xi'an, China
| | - Hongchen Zhang
- Department of Neurosurgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zhaoyang Fan
- Department of Pharmacology, The Fourth Military Medical University, Xi'an, China
| | - Mingkai Li
- Department of Pharmacology, The Fourth Military Medical University, Xi'an, China.
| | - Xia Li
- Department of Neurosurgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
31
|
Jiang Y, Wang A, Bai H, Ye M. [Protective effect of serine methyltransferase against hepatic ischemia-reperfusion injury in mice]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2020; 40:506-512. [PMID: 32895129 DOI: 10.12122/j.issn.1673-4254.2020.04.09] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To investigate the protective effect of serine hydroxymethyl transferase 2 (SHMT2) against hepatic ischemia-reperfusion injury in mice. METHODS Sixty C57BL/6 mice were divided equally into sham-operated group, saline adeno-associated virus group (AVV-GFP), and adeno-associated virus silencing group (AAV-SHMT2). The adeno-associated virus and normal saline were injected into the tail vein of the mice 2 weeks before establishment of a 70% ischemia-reperfusion model in the liver. qPCR, Western blotting, immunofluorescence and immunohistochemistry were used to detect the changes of AST/ALT concentration, SHMT2, JNK, NF-κB, caspase-3 and downstream inflammatory factors in the mice, and HE staining was used to observe the pathological damage of the liver tissue in each group; the cell apoptosis in the liver was detected using TUNEL assay. RESULTS The expression of SHMT2 increased with time after hepatic ischemia-reperfusion and reached the highest level at 24 h (the relative expression was 1.5, P < 0.05). At 24 h after hepatic ischemia-reperfusion, the levels of AST/ALT in AAV-SHMT2 group (588/416 U/L) were significantly higher than those in the control group (416/345 U/L) and the empty vector group (387/321 U/L) (P < 0.05). Compared with those in the control group and the empty vector group, the level of SHMT2 was significantly decreased in AAV-SHMT2 group (with a relative expression of 0.24, P < 0.05), the levels of p-JNK and p-p65 were significantly increased (relative expression of 0.80 and 0.97, respectively, P < 0.05), and the levels TNF-α and IL-1β were consistently elevated (relative expression levels of 1.6 and 1.2, respectively, P < 0.05). No significant differences were found in these parameters between the empty vector group and the control group (P>0.05). CONCLUSIONS SHMT2 may alleviate liver cell apoptosis in mice with hepatic ischemia-reperfusion injury by inhibiting the activation of JNK pathway and excessive activation of NF-κB pathway to reduce hepatic damage.
Collapse
Affiliation(s)
- Yu Jiang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Ankang Wang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - He Bai
- Department of General Surgery, First Affiliated Hospital of Xi'an Medical College, Xi'an 710000, China
| | - Mingxin Ye
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
32
|
Zhang E, Chen Q, Wang J, Li D, Wan Z, Ju X. Protective role of microRNA-27a upregulation and HSP90 silencing against cerebral ischemia-reperfusion injury in rats by activating PI3K/AKT/mTOR signaling pathway. Int Immunopharmacol 2020; 86:106635. [PMID: 32634698 DOI: 10.1016/j.intimp.2020.106635] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 05/12/2020] [Accepted: 05/22/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVE MicroRNAs (miRNAs) have been reported in cerebral ischemia-reperfusion injury, yet the function of miR-27a in it has seldom been mentioned. This study aims to assess the mechanisms of miR-27a in rats with cerebral ischemia-reperfusion injury. METHODS The cerebral ischemia-reperfusion models of rat pups were established by bilateral carotid artery occlusion. Rats were treated with miR-27a agomir, silenced HSP90 expression plasmids or PI3K/AKT/mTOR pathway agonist. Oxidative stress indices, inflammatory factors, brain tissue water content, cerebral infarct volume, neurological function score and neuronal apoptosis in rats with cerebral ischemia-reperfusion injury were measured. MiR-27a and HSP90 expression and PI3K/AKT/mTOR phosphorylation levels in the brain tissues of rats were also detected. RESULTS MiR-27a expression and PI3K/AKT/mTOR phosphorylation levels were downregulated while HSP90 expression was upregulated in cerebral ischemia-reperfusion injury rats. Elevated miR-27a or reduced HSP90 diminished water content, neuronal apoptosis and infarct volume, suppressed oxidative stress and inflammatory response, as well as improved neurological deficits and pathological damages. Moreover, elevated miR-27a or silenced HSP90 upregulated PI3K/AKT/mTOR phosphorylation levels in cerebral ischemia-reperfusion injury rats. HSP90 silencing or PI3K/AKT/mTOR pathway agonist reversed the unfavorable effects of low miR-27a expression on cerebral ischemia-reperfusion injury rats. CONCLUSION To conclude, our study demonstrates that elevated miR-27a or decreased HSP90 attenuates oxidative stress and inflammatory response, and improves neurological function in cerebral ischemia-reperfusion injury rats by activating PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Ensheng Zhang
- Department of Pediatrics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China; Department of Pediatrics, Maternal and Child Health Care Hospital of Shandong Province, Cheeloo College of Medicine, Jinan 250014, Shandong, China
| | - Qian Chen
- Department of Pediatrics, Maternal and Child Health Care Hospital of Shandong Province, Cheeloo College of Medicine, Jinan 250014, Shandong, China
| | - Jing Wang
- Department of Urology, First Affiliated Hospital of Shandong First Medical University, Jinan 250014, Shandong, China
| | - Dong Li
- Department of Pediatrics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
| | - Zhenxia Wan
- Department of Pediatrics, Maternal and Child Health Care Hospital of Shandong Province, Cheeloo College of Medicine, Jinan 250014, Shandong, China
| | - Xiuli Ju
- Department of Pediatrics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China.
| |
Collapse
|
33
|
Xia P, Zhang F, Yuan Y, Chen C, Huang Y, Li L, Wang E, Guo Q, Ye Z. ALDH 2 conferred neuroprotection on cerebral ischemic injury by alleviating mitochondria-related apoptosis through JNK/caspase-3 signing pathway. Int J Biol Sci 2020; 16:1303-1323. [PMID: 32210721 PMCID: PMC7085232 DOI: 10.7150/ijbs.38962] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 12/29/2019] [Indexed: 12/14/2022] Open
Abstract
Past studies have indicated that the dysregulation of Aldehyde dehydrogenase 2 (ALDH2) is related to the pathogenesis of acute stroke. However, the underlying mechanisms of ALDH2-mediated acute stroke are still not well understood. Thus, our study was designed to explore the influence of ALDH2 in acute stroke and determine whether its related mechanisms are involved in regulating mitochondria-associated apoptosis modulating JNK/caspase-3 pathway. In vitro analysis on the gain and loss of ALDH2 and JNK function were performed to explore its influence on OGD/R injury and relevant signaling pathways. Our findings suggested that ALDH2 expression was significantly down-regulated in rats suffering from acute stroke and also in primary cortical cultured neurons and PC12 cells upon OGD/R stimulation. ALDH2 overexpression markedly decreased infarct size and improved neurological outcomes. Furthermore, ALDH2 overexpression significantly suppressed stroke-induced mitochondria-associated apoptosis and inhibited p-JNK activation and p-JNK/caspase-3 complex formation. Similarly, in in vitro OGD/R models, ALDH2 reintroduction not only promoted cellular viability and moderated LDH release, but also inhibited mitochondria-related apoptosis. Moreover, JNK inhibition relieved OGD/R-induced cellular injury and apoptosis while JNK activation aggravated them. Furthermore, ALDH2 overexpression and JNK inhibition significantly reduced caspase-3 activation and transcription which was triggered by OGD/R damage. Caspase-3 activation and transcription also re-elevated during activation of JNK in ALDH2-reintroduced cells. Finally, ChIP assay revealed that p-JNK was bound to caspase-3 promoter. Collectively, ALDH2 overexpression led to a significant reduction in mitochondria-related apoptosis via JNK-mediated caspase-3 activation and transcription in both in vitro and in vivo cerebral ischemia models.
Collapse
Affiliation(s)
- Pingping Xia
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410078, Hunan Province, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, 410008, Hunan, P. R. China
| | - Fan Zhang
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410078, Hunan Province, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, 410008, Hunan, P. R. China
| | - Yajing Yuan
- Department of Anesthesiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin 300060, China
| | - Cheng Chen
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410078, Hunan Province, China
| | - Yan Huang
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410078, Hunan Province, China
| | - Longyan Li
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410078, Hunan Province, China
| | - E Wang
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410078, Hunan Province, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, 410008, Hunan, P. R. China
| | - Qulian Guo
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410078, Hunan Province, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, 410008, Hunan, P. R. China
| | - Zhi Ye
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410078, Hunan Province, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, 410008, Hunan, P. R. China
| |
Collapse
|
34
|
Denorme F, Vanhoorelbeke K, De Meyer SF. von Willebrand Factor and Platelet Glycoprotein Ib: A Thromboinflammatory Axis in Stroke. Front Immunol 2019; 10:2884. [PMID: 31921147 PMCID: PMC6928043 DOI: 10.3389/fimmu.2019.02884] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 11/25/2019] [Indexed: 01/23/2023] Open
Abstract
von Willebrand factor (VWF) and platelets are key mediators of normal hemostasis. At sites of vascular injury, VWF recruits platelets via binding to the platelet receptor glycoprotein Ibα (GPIbα). Over the past decades, it has become clear that many hemostatic factors, including VWF and platelets, are also involved in inflammatory processes, forming intriguing links between hemostasis, thrombosis, and inflammation. The so-called “thrombo-inflammatory” nature of the VWF-platelet axis becomes increasingly recognized in different cardiovascular pathologies, making it a potential therapeutic target to interfere with both thrombosis and inflammation. In this review, we discuss the current evidence for the thrombo-inflammatory activity of VWF with a focus on the VWF-GPIbα axis and discuss its implications in the setting of ischemic stroke.
Collapse
Affiliation(s)
- Frederik Denorme
- Laboratory for Thrombosis Research, KU Leuven, Kortrijk, Belgium
| | | | - Simon F De Meyer
- Laboratory for Thrombosis Research, KU Leuven, Kortrijk, Belgium
| |
Collapse
|
35
|
Qin Y, Zhang Q, Liu Y. Analysis of knowledge bases and research focuses of cerebral ischemia-reperfusion from the perspective of mapping knowledge domain. Brain Res Bull 2019; 156:15-24. [PMID: 31843561 DOI: 10.1016/j.brainresbull.2019.12.004] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 12/03/2019] [Indexed: 11/17/2022]
Abstract
Cerebral ischemia-reperfusion (IR) has attracted wide attention as a serious clinical problem. So far, the field has accumulated a large amount of scientific research literature. To clarify the temporal and spatial distribution characteristics of research resources, knowledge bases and research focuses, a visual analysis was performed on 5814 articles cited in the WoS databases from 2004 to 2019. This analysis was based on bibliometrics and mapping knowledge domain (MKD) analysis with VOSviewer, and CiteSpace 5.4.R4. The results can be elaborated from four aspects. First, the volume of publications in this area is on the rise. Second, the United States and China are the active regions. The USA is the central region of cerebral ischemia-reperfusion research. Third, the knowledge bases of IR have focused on five major areas of "Suitable small-animal models", "A framework with further study", "Molecular signaling targets by oxidative stress", "Finding new potential targets for therapy" and "Protective effect of multiple transient ischemia". Fourth, the research focuses consist of three representative areas: "Oxidative stress closelyd with cerebral ischemia-reperfusion", "Neuronal apoptosis and neuronal protection", and "Neuroprotective effect of the blood-brain barrier".
Collapse
Affiliation(s)
- Yi Qin
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China; Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Qing Zhang
- No.4 Hospital Beijing University of Chinese Medicine, Zaozhuang, Shandong 277000
| | - Yaru Liu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
36
|
Gong Y, Chen J, Jin Y, Wang C, Zheng M, He L. GW9508 ameliorates cognitive impairment via the cAMP-CREB and JNK pathways in APPswe/PS1dE9 mouse model of Alzheimer's disease. Neuropharmacology 2019; 164:107899. [PMID: 31809762 DOI: 10.1016/j.neuropharm.2019.107899] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 11/14/2019] [Accepted: 11/30/2019] [Indexed: 12/12/2022]
Abstract
GPR40 was utilized as the drug target to the treatment of diabetes, but the function and mechanisms ameliorating the Alzheimer's disease (AD) remain unknown. In present study, the typical APP/PS1 mouse model was applied to explore the function and mechanism of GPR40 in AD. GPR40 agonist GW9508 and antagonist GW1100 were respectively given by i.c.v. injection to activate/inhibit the GPR40 in the brain of APP/PS1 mice which illustrated the function and mechanism of GPR40 in ameliorating AD symptoms. Morris water maze test, step-through test, Y-maze spontaneous alternation test, open field test and new object recognition test were used to test the cognitive function and memory ability of mice, while molecular biology experiments such as Western blot, immunofluorescence, JC-1 were used to detect the corresponding changes of signal pathways. The results revealed that treatment with GW9508 could significantly ameliorate cognitive deficits of APP/PS1 mice, upregulate the expression levels of cAMP, p-CREB and neurotrophic factors in vivo, while GW9508 also ameliorate Aβ1-42-induced neuron damage and downregulate the expression levels of pathological protein such as p-JNK, JNK and apoptosis-related proteins such as IL-6, IL-1β, TNF-α and caspase-3 in vitro. Meanwhile, high-content screening also showed that GW9508 promoted the cellular differentiation of SH-SY5Y cells, while GW1100 reversed the effects of GW9508. These results suggested that GPR40 was an underlying therapeutic target for the treatment of AD and GPR40 agonist could be explored as the emerging AD therapeutic drug.
Collapse
Affiliation(s)
- Yuhang Gong
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Jingjing Chen
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Yongzeng Jin
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Chen Wang
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Menglin Zheng
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Ling He
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
37
|
Anfibatide Preserves Blood–Brain Barrier Integrity by Inhibiting TLR4/RhoA/ROCK Pathway After Cerebral Ischemia/Reperfusion Injury in Rat. J Mol Neurosci 2019; 70:71-83. [DOI: 10.1007/s12031-019-01402-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 08/27/2019] [Indexed: 12/13/2022]
|
38
|
Echinocystic acid, a natural plant extract, alleviates cerebral ischemia/reperfusion injury via inhibiting the JNK signaling pathway. Eur J Pharmacol 2019; 861:172610. [PMID: 31425684 DOI: 10.1016/j.ejphar.2019.172610] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 08/09/2019] [Accepted: 08/14/2019] [Indexed: 01/19/2023]
Abstract
Echinocystic acid (EA) was found to possess antiviral, anti-inflammatory and antioxidation activities. A recent study showed the antiapoptotic effects of EA on acute myocardial infarction. In this study, we demonstrated the potential neuroprotective effects of EA on cerebral ischemia/reperfusion (I/R) injury in mice. Intraperitoneal injection of EA 1 h before ischemia significantly reduced the cerebral infarct volume and neurological deficit after 60 min of ischemia and 24 h of reperfusion. The neuroprotective effects of EA occurred in a dose-dependent manner. Then, we explored the mechanisms of neuroprotection by EA. This compound exerted antiapoptotic activity by upregulating the level of Bcl-2 and simultaneously downregulating the levels of cleaved caspase-3 and Bax. Furthermore, EA also possessed anti-inflammatory activity and prevented the excessive phosphorylation of NF-κB (p-P65) and the increase in IL-1β and IL-6 levels. Finally, our data indicated that EA treatment decreased the level of phosphorylated JNK in vivo, and the JNK activator anisomycin (AN) reversed the neuroprotective effects of EA, indicating that the JNK pathway is involved in the antiapoptotic and anti-inflammatory mechanisms of EA. In summary, our findings suggest that EA provides neuroprotective effects through its antiapoptotic and anti-inflammatory activities by inhibiting the JNK signaling pathway in cerebral I/R injury. Due to its safety and lack of toxicity, EA is a potential candidate for the treatment of ischemic stroke in future clinical trials.
Collapse
|
39
|
The Mechanism of MAPK Signal Transduction Pathway Involved with Electroacupuncture Treatment for Different Diseases. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:8138017. [PMID: 31467579 PMCID: PMC6699341 DOI: 10.1155/2019/8138017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 06/07/2019] [Accepted: 07/03/2019] [Indexed: 12/13/2022]
Abstract
The mitogen-activated protein kinase (MAPK) signal transduction pathway plays an important role in the regulation of various diseases, such as cardiovascular and cerebrovascular diseases, and takes part in anti-inflammatory effects, analgesic effects, protection against injury, and maintenance of gastrointestinal functions. Electroacupuncture therapy is an external therapy used in traditional Chinese medicine. By adding external electrical stimulation to traditional acupuncture, the stimulus gets doubled and the therapeutic efficacy gets enhanced accordingly. It combines the benefits of both acupuncture and electrical stimulation. In recent years, some studies have explored the molecular mechanisms of MAPK signal pathways involved with electroacupuncture treatment. Based on these recent studies, this article summarizes the mechanisms of MAPK signal transduction pathways involved with electroacupuncture treatment. This adds great value to the studies of molecular mechanisms of electroacupuncture treatment and also provides an effective reference for its clinical use.
Collapse
|
40
|
Bai H, Wen J, Gong JP, Wu H, Yuan FC, Cao D, Wu YK, Lai X, Wang MH. Blockade of the Notch1/Jagged1 pathway in Kupffer cells aggravates ischemia-reperfusion injury of orthotopic liver transplantation in mice. Autoimmunity 2019; 52:176-184. [PMID: 31322442 DOI: 10.1080/08916934.2019.1637424] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Liver ischemia-reperfusion injury (IRI) represents a risk factor for early graft dysfunction and an obstacle to expanding donor pool in orthotopic liver transplantation (OLT). Kupffer cells (KCs) are the largest antigen-presenting cell (APC) group and the primary modulators of inflammation in liver tissues. The vital role of Notch1/Jagged1 pathway in mouse OLT model has been reported, however, its potential therapeutic mechanism is unknown. Here, we made use of short hairpin RNA-Jagged1 and AAV-Jagged1 to explore the effects of Notch1/Jagged1 pathway in OLT. In vitro, blockade of Notch1/Jagged1 pathway downregulated the expression of Hairy and enhancer of split-1 (Hes1) gene, which in turn increased the proinflammatory effects of KCs. Moreover, the anti-inflammatory effects of Notch1/Jagged1 pathway were induced by inhibiting Hes1/gene of phosphate and tension/protein kinase B/Toll-like receptor 4/nuclear factor kappa B (Hes1/PTEN/AKT/TLR4/NF-κB) axis in KCs. In vivo, we used a well-established mouse model of OLT to mimic clinical transplantation. Mice were stochastically divided into 6 groups: Sham group (n = 15); Normal saline (NS) group (n = 15); Adeno-associated virus-green fluorescent protein (AAV-GFP) group (n = 15); AAV-Jagged1 group (n = 15); Clodronate liposome (CL) group (n = 15); CL+AAV-Jagged1 group (n = 15) . After OLT the liver damage in AAV-Jagged1 group were significantly accentuated compared to the AAV-GFP group. While blockade of Jagged1 aftet clearence of KCs by CL would not lead to further liver injuries. Taken together, our study demonstrated that blockade of Notch1/Jagged1 pathway aggravates inflammation induced by lipopolysaccharide (LPS) via Hes1/PTEN/AKT/TLR4/NF-κB in KCs, and the blockade of Notch1/Jagged1 pathway in donor liver increased neutrophil/macrophage infiltration and hepatocellular apoptosis, which suggested the function of Notch1/Jagged1 pathway in mouse OLT and highlighted the protective function of Notch1/Jagged1 pathway in liver transplantation.
Collapse
Affiliation(s)
- He Bai
- Department of Hepatobiliary Surgery, Second Affiliated Hospital of Chongqing Medical University , Chongqing , People's Republic of China
| | - Jian Wen
- Department of Hepatobiliary Surgery, Second Affiliated Hospital of Chongqing Medical University , Chongqing , People's Republic of China
| | - Jian-Ping Gong
- Department of Hepatobiliary Surgery, Second Affiliated Hospital of Chongqing Medical University , Chongqing , People's Republic of China
| | - Hao Wu
- Department of Hepatobiliary Surgery, Second Affiliated Hospital of Chongqing Medical University , Chongqing , People's Republic of China
| | - Fang-Chao Yuan
- Department of Hepatobiliary Surgery, Second Affiliated Hospital of Chongqing Medical University , Chongqing , People's Republic of China
| | - Ding Cao
- Department of Hepatobiliary Surgery, Second Affiliated Hospital of Chongqing Medical University , Chongqing , People's Republic of China
| | - Ya-Kun Wu
- Department of Hepatobiliary Surgery, Suining Central Hospital , Sichuan , People's Republic of China
| | - Xing Lai
- Department of Hepatobiliary and Thyroid Breast Surgery, Tongnan District People's Hospital , Chongqing , People's Republic of China
| | - Meng-Hao Wang
- Department of Hepatobiliary Surgery, Second Affiliated Hospital of Chongqing Medical University , Chongqing , People's Republic of China
| |
Collapse
|
41
|
Wang Y, Wang D, Jin Z. miR‑27a suppresses TLR4‑induced renal ischemia‑reperfusion injury. Mol Med Rep 2019; 20:967-976. [PMID: 31173204 PMCID: PMC6625210 DOI: 10.3892/mmr.2019.10333] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 03/11/2019] [Indexed: 12/20/2022] Open
Abstract
Ischemia reperfusion injury (IRI) is one of the primary causes of acute renal injury and even acute renal failure. An increasing body of evidence has indicated that the aberrant expression of microRNAs (miRNA/miR) is closely associated with the progression of IRI. In the process of renal IRI, inflammatory reactions, cell adhesion and the death of renal tubular epithelial cells serve important roles. The present study investigated the expression of renal miRNAs following renal IRI in an attempt to identify the miRNAs that exert pivotal functions in renal IRI. The present study revealed that miR-27a, which was downregulated in IRI, and the 3′-untranslated region (UTR) of Toll-like receptor 4 (TLR4) have associated binding sites. The levels of TLR4, miR-27a and specific associated proteins in the renal tissues of gestational rats were determined by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis, immunohistochemistry and western blotting. The associations between miR-27a and TLR4 were analyzed, and the regulatory effect of miR-27a on TLR4 was detected via a luciferase reporter gene assay, western blotting and RT-qPCR in vivo and in vitro. In addition, the present study demonstrated that miR-27a suppressed the expression of TLR4 by binding to the 3′UTR of TLR4. The overexpression of miR-27a downregulated the expression of TLR4, which in turn inhibited the inflammation, cell adhesion and cell death in IRI. Therefore, miR-27a inhibited inflammation in IRI by decreasing the expression of TLR4.
Collapse
Affiliation(s)
- Yang Wang
- Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Dan Wang
- Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Zhen Jin
- Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
42
|
Eble JA. Structurally Robust and Functionally Highly Versatile-C-Type Lectin (-Related) Proteins in Snake Venoms. Toxins (Basel) 2019; 11:toxins11030136. [PMID: 30823637 PMCID: PMC6468738 DOI: 10.3390/toxins11030136] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 02/19/2019] [Accepted: 02/20/2019] [Indexed: 12/31/2022] Open
Abstract
Snake venoms contain an astounding variety of different proteins. Among them are numerous C-type lectin family members, which are grouped into classical Ca2+- and sugar-binding lectins and the non-sugar-binding snake venom C-type lectin-related proteins (SV-CLRPs), also called snaclecs. Both groups share the robust C-type lectin domain (CTLD) fold but differ in a long loop, which either contributes to a sugar-binding site or is expanded into a loop-swapping heterodimerization domain between two CLRP subunits. Most C-type lectin (-related) proteins assemble in ordered supramolecular complexes with a high versatility of subunit numbers and geometric arrays. Similarly versatile is their ability to inhibit or block their target molecules as well as to agonistically stimulate or antagonistically blunt a cellular reaction triggered by their target receptor. By utilizing distinct interaction sites differentially, SV-CLRPs target a plethora of molecules, such as distinct coagulation factors and receptors of platelets and endothelial cells that are involved in hemostasis, thrombus formation, inflammation and hematogenous metastasis. Because of their robust structure and their high affinity towards their clinically relevant targets, SV-CLRPs are and will potentially be valuable prototypes to develop new diagnostic and therapeutic tools in medicine, provided that the molecular mechanisms underlying their versatility are disclosed.
Collapse
Affiliation(s)
- Johannes A Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstr. 15, 48149 Münster, Germany.
| |
Collapse
|
43
|
Shvedova M, Anfinogenova Y, Atochina-Vasserman EN, Schepetkin IA, Atochin DN. c-Jun N-Terminal Kinases (JNKs) in Myocardial and Cerebral Ischemia/Reperfusion Injury. Front Pharmacol 2018; 9:715. [PMID: 30026697 PMCID: PMC6041399 DOI: 10.3389/fphar.2018.00715] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 06/13/2018] [Indexed: 12/18/2022] Open
Abstract
In this article, we review the literature regarding the role of c-Jun N-terminal kinases (JNKs) in cerebral and myocardial ischemia/reperfusion injury. Numerous studies demonstrate that JNK-mediated signaling pathways play an essential role in cerebral and myocardial ischemia/reperfusion injury. JNK-associated mechanisms are involved in preconditioning and post-conditioning of the heart and the brain. The literature and our own studies suggest that JNK inhibitors may exert cardioprotective and neuroprotective properties. The effects of modulating the JNK-depending pathways in the brain and the heart are reviewed. Cardioprotective and neuroprotective mechanisms of JNK inhibitors are discussed in detail including synthetic small molecule inhibitors (AS601245, SP600125, IQ-1S, and SR-3306), ion channel inhibitor GsMTx4, JNK-interacting proteins, inhibitors of mixed-lineage kinase (MLK) and MLK-interacting proteins, inhibitors of glutamate receptors, nitric oxide (NO) donors, and anesthetics. The role of JNKs in ischemia/reperfusion injury of the heart in diabetes mellitus is discussed in the context of comorbidities. According to reviewed literature, JNKs represent promising therapeutic targets for protection of the brain and the heart against ischemic stroke and myocardial infarction, respectively. However, different members of the JNK family exert diverse physiological properties which may not allow for systemic administration of non-specific JNK inhibitors for therapeutic purposes. Currently available candidate JNK inhibitors with high therapeutic potential are identified. The further search for selective JNK3 inhibitors remains an important task.
Collapse
Affiliation(s)
- Maria Shvedova
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Yana Anfinogenova
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
- RASA Center in Tomsk, Tomsk Polytechnic University, Tomsk, Russia
| | - Elena N. Atochina-Vasserman
- RASA Center in Tomsk, Tomsk Polytechnic University, Tomsk, Russia
- RASA Center, Kazan Federal University, Kazan, Russia
| | - Igor A. Schepetkin
- RASA Center in Tomsk, Tomsk Polytechnic University, Tomsk, Russia
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Dmitriy N. Atochin
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
- RASA Center in Tomsk, Tomsk Polytechnic University, Tomsk, Russia
| |
Collapse
|
44
|
MicRNA-320 facilitates the brain parenchyma injury via regulating IGF-1 during cerebral I/R injury in mice. Biomed Pharmacother 2018; 102:86-93. [DOI: 10.1016/j.biopha.2018.03.036] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Revised: 02/27/2018] [Accepted: 03/09/2018] [Indexed: 01/01/2023] Open
|
45
|
Abd El-Aal SA, Abd El-Fattah MA, El-Abhar HS. CoQ10 Augments Rosuvastatin Neuroprotective Effect in a Model of Global Ischemia via Inhibition of NF-κB/JNK3/Bax and Activation of Akt/FOXO3A/Bim Cues. Front Pharmacol 2017; 8:735. [PMID: 29081748 PMCID: PMC5645536 DOI: 10.3389/fphar.2017.00735] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 09/29/2017] [Indexed: 12/12/2022] Open
Abstract
Statins were reported to lower the Coenzyme Q10 (CoQ10) content upon their inhibition of HMG-CoA reductase enzyme and both are known to possess neuroprotective potentials; therefore, the aim is to assess the possible use of CoQ10 as an adds-on therapy to rosuvastatin to improve its effect using global I/R model. Rats were allocated into sham, I/R, rosuvastatin (10 mg/kg), CoQ10 (10 mg/kg) and their combination. Drugs were administered orally for 7 days before I/R. Pretreatment with rosuvastatin and/or CoQ10 inhibited the hippocampal content of malondialdehyde, nitric oxide, and boosted glutathione and superoxide dismutase. They also opposed the upregulation of gp91phox, and p47phox subunits of NADPH oxidase. Meanwhile, both agents reduced content/expression of TNF-α, iNOS, NF-κBp65, ICAM-1, and MPO. Besides, all regimens abated cytochrome c, caspase-3 and Bax, but increased Bcl-2 in favor of cell survival. On the molecular level, they increased p-Akt and its downstream target p-FOXO3A, with the inhibition of the nuclear content of FOXO3A to downregulate the expression of Bim, a pro-apoptotic gene. Additionally, both treatments downregulate the JNK3/c-Jun signaling pathway. The effect of the combination regimen overrides that of either treatment alone. These effects were reflected on the alleviation of the hippocampal damage in CA1 region inflicted by I/R. Together, these findings accentuate the neuroprotective potentials of both treatments against global I/R by virtue of their rigorous multi-pronged actions, including suppression of hippocampal oxidative stress, inflammation, and apoptosis with the involvement of the Akt/FOXO3A/Bim and JNK3/c-Jun/Bax signaling pathways. The study also nominates CoQ10 as an adds-on therapy with statins.
Collapse
Affiliation(s)
- Sarah A Abd El-Aal
- Department of Pharmacology and Toxicology, October 6 University, Cairo, Egypt
| | - Mai A Abd El-Fattah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Hanan S El-Abhar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
46
|
Mršić-Pelčić J, Pilipović K, Pelčić G, Vitezić D, Župan G. Decrease in Oxidative Stress Parameters after Post-Ischaemic Recombinant Human Erythropoietin Administration in the Hippocampus of Rats Exposed to Focal Cerebral Ischaemia. Basic Clin Pharmacol Toxicol 2017. [DOI: 10.1111/bcpt.12833] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Jasenka Mršić-Pelčić
- Department of Pharmacology; Faculty of Medicine; University of Rijeka; Rijeka Croatia
| | - Kristina Pilipović
- Department of Pharmacology; Faculty of Medicine; University of Rijeka; Rijeka Croatia
| | - Goran Pelčić
- Clinics for Ophthalmology; Clinical Hospital Centre Rijeka; Rijeka Croatia
| | - Dinko Vitezić
- Department of Pharmacology; Faculty of Medicine; University of Rijeka; Rijeka Croatia
| | - Gordana Župan
- Department of Pharmacology; Faculty of Medicine; University of Rijeka; Rijeka Croatia
| |
Collapse
|