1
|
Zhang D, Qin H, Chen W, Xiang J, Jiang M, Zhang L, Zhou K, Hu Y. Utilizing network pharmacology, molecular docking, and animal models to explore the therapeutic potential of the WenYang FuYuan recipe for cerebral ischemia-reperfusion injury through AGE-RAGE and NF-κB/p38MAPK signaling pathway modulation. Exp Gerontol 2024; 191:112448. [PMID: 38697555 DOI: 10.1016/j.exger.2024.112448] [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: 02/21/2024] [Revised: 04/13/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
BACKGROUND Stroke is a debilitating condition with high morbidity, disability, and mortality that significantly affects the quality of life of patients. In China, the WenYang FuYuan recipe is widely used to treat ischemic stroke. However, the underlying mechanism remains unknown, so exploring the potential mechanism of action of this formula is of great practical significance for stroke treatment. OBJECTIVE This study employed network pharmacology, molecular docking, and in vivo experiments to clarify the active ingredients, potential targets, and molecular mechanisms of the WenYang FuYuan recipe in cerebral ischemia-reperfusion injury, with a view to providing a solid scientific foundation for the subsequent study of this recipe. MATERIALS AND METHODS Active ingredients of the WenYang FuYuan recipe were screened using the traditional Chinese medicine systems pharmacology database and analysis platform. Network pharmacology approaches were used to explore the potential targets and mechanisms of action of the WenYang FuYuan recipe for the treatment of cerebral ischemia-reperfusion injury. The Middle Cerebral Artery Occlusion/Reperfusion 2 h Sprague Dawley rat model was prepared, and TTC staining and modified neurological severity score were applied to examine the neurological deficits in rats. HE staining and Nissl staining were applied to examine the pathological changes in rats. Immunofluorescence labeling and Elisa assay were applied to examine the expression levels of certain proteins and associated factors, while qRT-PCR and Western blotting were applied to examine the expression levels of linked proteins and mRNAs in disease-related signaling pathways. RESULTS We identified 62 key active ingredients in the WenYang FuYuan recipe, with 222 highly significant I/R targets, forming 138 pairs of medication components and component-targets, with the top five being Quercetin, Kaempferol, Luteolin, β-sitosterol, and Stigmasterol. The key targets included TP53, RELA, TNF, STAT1, and MAPK14 (p38MAPK). Targets related to cerebral ischemia-reperfusion injury were enriched in chemical responses, enzyme binding, endomembrane system, while enriched pathways included lipid and atherosclerosis, fluid shear stress and atherosclerosis, AGE-RAGE signaling in diabetic complications. In addition, the main five active ingredients and targets in the WenYang FuYuan recipe showed high binding affinity (e.g. Stigmasterol and MAPK14, total energy <-10.5 Kcal/mol). In animal experiments, the WenYang FuYuan recipe reduced brain tissue damage, increased the number of surviving neurons, and down-regulated S100β and RAGE protein expression. Moreover, the relative expression levels of key targets such as TP53, RELA and p38MAPK mRNA were significantly down-regulated in the WenYang FuYuan recipe group, and serum IL-6 and TNF-a factor levels were reduced. After WenYang FuYuan recipe treatment, the AGE-RAGE signaling pathway and downstream NF-kB/p38MAPK signaling pathway-related proteins were significantly modulated. CONCLUSION This study utilized network pharmacology, molecular docking, and animal experiments to identify the potential mechanism of the WenYang FuYuan recipe, which may be associated with the regulation of the AGE-RAGE signaling pathway and the inhibition of target proteins and mRNAs in the downstream NF-kB/p38MAPK pathway.
Collapse
Affiliation(s)
- Ding Zhang
- Guangxi University of Chinese Medicine, Nanning, China
| | - Hongling Qin
- Guangxi University of Chinese Medicine First Affiliated Hospital, Nanning, China
| | - Wei Chen
- Guangxi University of Chinese Medicine First Affiliated Hospital, Nanning, China
| | - Junjun Xiang
- Guangxi University of Chinese Medicine, Nanning, China
| | - Minghe Jiang
- Guangxi University of Chinese Medicine, Nanning, China
| | - Ling Zhang
- Guangxi University of Chinese Medicine, Nanning, China
| | - Keqing Zhou
- Guangxi University of Chinese Medicine, Nanning, China
| | - Yueqiang Hu
- Guangxi University of Chinese Medicine First Affiliated Hospital, Nanning, China.
| |
Collapse
|
2
|
Rakhimbaeva GS, Abdurakhmonova KBK. ICAM-1 and CRP as biomarkers of 3-month outcome in acute ischaemic stroke. BMJ Neurol Open 2023; 5:e000516. [PMID: 38145240 PMCID: PMC10749038 DOI: 10.1136/bmjno-2023-000516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 12/06/2023] [Indexed: 12/26/2023] Open
Abstract
Background It is clear that, inflammation deteriorates cerebral injury during the acute phase of stroke. While this process is going on, intercellular adhesion molecule-1 (ICAM-1) has a crucial role to play in mediating migration of immune cells into the damaged area. Furthermore, C reactive protein (CRP) is an essential inflammatory molecule in human organism. This research aims to investigate the association between ICAM-1, highly sensitive CRP(hs-CRP) and the prognosis of acute ischaemic stroke (AIS). Methods 118 patients with AIS who were treated at Tashkent Medical Academy were participants in this research project. Blood samples were collected from patients on an empty stomach within 24 hours of admission. Modified Rankin Scale (mRS) was used in order to assess the functional prognosis in 3 months following the case of stroke in patients. The inadequate prognosis is described as mRS≥3. Each biomarker's potential to predict has also been evaluated with receiver operating characteristic analysis. Results ICAM-1 was identified to be an independent predictor of 3-month outcome (OR 1.05, 95 % CI 0.848 to 1.625; p=0.02) (area under the curve (AUC)=0.82 %). Independent associations with functional outcome were also found to be true for hs-CRP (OR 1.22, 95 % CI 0.78 to 1.86; p=0.03) (AUC=0.74 %). Conclusions The outcomes of a 3-month study carried out on patients with AIS showed ICAM-1 and hs-CRP to be independent predictors.
Collapse
|
3
|
Cai X, Song S, Hu J, Wang L, Shen D, Zhu Q, Yang W, Luo Q, Hong J, Li N. Systemic Inflammation Response Index as a Predictor of Stroke Risk in Elderly Patients with Hypertension: A Cohort Study. J Inflamm Res 2023; 16:4821-4832. [PMID: 37901383 PMCID: PMC10612501 DOI: 10.2147/jir.s433190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 10/10/2023] [Indexed: 10/31/2023] Open
Abstract
Objective This study aimed to evaluate the relationship between the systemic inflammation response index (SIRI) and the risk of stroke and its subtypes in elderly patients with hypertension and to explore its predictive accuracy and any potential effect modifiers. Methods The study included 4749 participants with no history of stroke at baseline. Cox regression was used to estimate adjusted hazard ratios (HR) and 95% confidence intervals (CIs). Interaction tests and subgroup analyses were conducted. The predictive performance of various inflammatory indicators for stroke was compared using the area under the curve (AUC), continuous net reclassification improvement (NRI), and integrated discrimination improvement (IDI). Results During a median follow-up period of 3.2 years, 640 strokes were recorded, of which 526 were ischemic and the remainder hemorrhagic. After adjustment for confounders, compared to the reference group, the HRs (95% CI) of stroke were 1.28 (95% CI, 1.01-1.64) and 1.46 (95% CI, 1.14-1.88) for participants in the second and third tertiles, respectively. We observed interactions between SIRI and homocysteine levels (< 15 vs. ≥ 15 μmol/L) (p for interaction = 0.014) on ischemic stroke risk. Furthermore, the AUC, NRI, and IDI analyses demonstrated that SIRI exhibited better predictive value for stroke risk when compared to other indicators. Similar results were observed for both ischemic and hemorrhagic strokes. Conclusion Elevated SIRI levels were significantly associated with the risk of stroke and its subtypes in elderly patients with hypertension, suggesting its potential as a promising indicator for stroke risk in this population. However, larger prospective studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Xintian Cai
- Graduate School, Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
| | - Shuaiwei Song
- Graduate School, Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
| | - Junli Hu
- Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region, Hypertension Research Laboratory, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, Xinjiang, People’s Republic of China
| | - Lei Wang
- Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region, Hypertension Research Laboratory, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, Xinjiang, People’s Republic of China
| | - Di Shen
- Graduate School, Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
| | - Qing Zhu
- Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region, Hypertension Research Laboratory, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, Xinjiang, People’s Republic of China
| | - Wenbo Yang
- Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region, Hypertension Research Laboratory, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, Xinjiang, People’s Republic of China
| | - Qin Luo
- Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region, Hypertension Research Laboratory, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, Xinjiang, People’s Republic of China
| | - Jing Hong
- Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region, Hypertension Research Laboratory, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, Xinjiang, People’s Republic of China
| | - Nanfang Li
- Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region, Hypertension Research Laboratory, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, Xinjiang, People’s Republic of China
| |
Collapse
|
4
|
Li Y, Ying W. Methylene blue reduces the serum levels of interleukin-6 and inhibits STAT3 activation in the brain and the skin of lipopolysaccharide-administered mice. Front Immunol 2023; 14:1181932. [PMID: 37325623 PMCID: PMC10266349 DOI: 10.3389/fimmu.2023.1181932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/19/2023] [Indexed: 06/17/2023] Open
Abstract
It is valuable to search for novel and economical agents for inhibiting STAT3 activation and blocking increases in IL-6 levels, due to the important roles of STAT3 and IL-6 in inflammation. Since Methylene Blue (MB) has shown therapeutical potential for multiple diseases, it has become increasingly important to investigate the mechanisms underlying the effects of MB on inflammation. Using a mouse model of lipopolysaccharide (LPS)-induced inflammation, we investigated the mechanisms underlying the effects of MB on inflammation, obtaining the following findings: First, MB administration attenuated the LPS-induced increases in the serum levels of IL-6; second, MB administration attenuated LPS-induced STAT3 activation of the brain; and third, MB administration attenuated LPS-induced STAT3 activation of the skin. Collectively, our study has suggested that MB administration can decrease the levels of IL-6 and STAT3 activation - two important factors in inflammation. Since MB is a clinically used and relatively economical drug, our findings have suggested therapeutic potential of MB for multiple inflammation-associated diseases due to its effects on STAT3 activation and IL-6 levels.
Collapse
Affiliation(s)
| | - Weihai Ying
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
5
|
Chen X, Li P, Huang R, Zhang J, Ouyang X, Tan D. Ulinastatin affects focal cerebral ischemia-reperfusion injury via SOCS1-mediated JAK2/STAT3 signalling pathway. Clin Exp Pharmacol Physiol 2023; 50:107-116. [PMID: 36222378 DOI: 10.1111/1440-1681.13731] [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: 01/18/2022] [Revised: 09/28/2022] [Accepted: 10/07/2022] [Indexed: 12/14/2022]
Abstract
Cerebral ischemia results in loss of cerebral blood flow, which contributes to neuronal damage, neurocognitive impairment, as well as learning and memory difficulties. Although reperfusion is necessary to restore the blood supply to the brain, it also leads to several detrimental effects on the brain. The purpose of this study was to assess the effects of ulinastatin (UTI) on preventing focal cerebral ischemia/reperfusion-induced injury (FCIRI). First, a rat model of FCIRI was established and treated with UTI. The effects of UTI on FCIRI in rats were evaluated using Morris water maze assay, triphenyl tetrazolium chloride staining, TUNEL, western blot assay, and enzyme-linked immunosorbent assay analysis. UTI was found to improve the learning memory ability, reduce infarction area, inhibit apoptosis and decrease inflammation in FCIRI rats. Messenger RNA microarray analysis of hippocampal tissues revealed that suppressor of cytokine signalling-1 (SOCS1) was the downstream target of UTI in FCIRI. SOCS1 depletion impaired the protective effect of UTI on FCIRI in rats. SOCS1 blocked the activation of the JAK2/STAT3 pathway. JAK2 inhibitor caused the JAK2/STAT3 pathway deficit, hence reversing the effect of sh-SOCS1 on FCIRI in rats. Taken together, our results demonstrate that UTI alleviated FCIRI in rats, which was, to some extent, related to SOCS1-mediated JAK2/STAT3 pathway.
Collapse
Affiliation(s)
- Xiaoxi Chen
- Department of Anesthesiology, Hengyang City Central Hospital, Hengyang, China
| | - Peng Li
- Department of Anesthesiology, Hengyang City Central Hospital, Hengyang, China
| | - Renming Huang
- Department of Anesthesiology, Hengyang City Central Hospital, Hengyang, China
| | - Juan Zhang
- Department of Anesthesiology, Hengyang City Central Hospital, Hengyang, China
| | - Xingzhi Ouyang
- Department of Anesthesiology, Hengyang City Central Hospital, Hengyang, China
| | - Dianxiang Tan
- Department of Anesthesiology, Hengyang City Central Hospital, Hengyang, China
| |
Collapse
|
6
|
Chen RH, Du WD, Wang Q, Li ZF, Wang DX, Yang SL, Feng YL. Effects of Acanthopanax senticosus (Rupr. & Maxim.) Harms on cerebral ischemia-reperfusion injury revealed by metabolomics and transcriptomics. JOURNAL OF ETHNOPHARMACOLOGY 2021; 264:113212. [PMID: 32768643 DOI: 10.1016/j.jep.2020.113212] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cerebral ischemia-reperfusion (CIR) injury is one of the main diseases leading to death and disability. Acanthopanax senticosus (Rupr. & Maxim.) Harms (AS), also known as Panax ginseng, has neuroprotective effects on anti-CIR injury. However, the underlying molecular mechanism of its therapeutic effects is not clear. AIM OF THE STUDY To systematically study and explore the mechanism of Acanthopanax senticosus (Rupr. & Maxim.) Harms extract (ASE) in the treatment of CIR injury based on metabolomics and transcriptomics. MATERIALS AND METHODS The pharmacological basis of ASE in the treatment of CIR was evaluated, and samples were used in plasma metabolomics and brain tissue transcriptomics to reveal potential biomarkers. Finally, according to online database, we analyzed biomarkers identified by the two technologies, explained reasons for the therapeutic effect of ASE, and identify therapeutic targets. RESULTS A total of 53 differential metabolites (DMs) were identified in plasma and 3138 differentially expressed genes (DEGs) were identified in brain tissue from three groups of rats, including sham, ischemia-reperfusion (I/R), and ASE groups. Enrichment analysis showed that Nme6, Tk1, and Pold1 that are involved in the production of deoxycytidine and thymine were significantly up-regulated and Dck was significantly down-regulated by the intervention with ASE. These findings indicated that ASE participates in the pyrimidine metabolism by significantly regulating the balance between dCTP and dTTP. In addition, ASE repaired and promoted the lipid metabolism in rats, which might be due to the significant expression of Dgkz, Chat, and Gpcpd1. CONCLUSIONS The findings of this study suggest that ASE regulates the significant changes in gene expression in metabolites pyrimidine, and lipid metabolism in CIR rats and plays an active role in the treatment of CIR injury through multiple targets and pathways.
Collapse
Affiliation(s)
- Ren-Hao Chen
- Jiangxi University of Traditional Chinese Medicine, Nanchang, 330002, China
| | - Wei-Dong Du
- Jiangxi University of Traditional Chinese Medicine, Nanchang, 330002, China
| | - Qi Wang
- State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Nanchang, 330006, China
| | - Zhi-Feng Li
- Jiangxi University of Traditional Chinese Medicine, Nanchang, 330002, China; Nanchang Key Laboratory of Active Ingredients of Traditional Chinese Medicine and Natural Medicine, Nanchang, 330006, China.
| | - Dong-Xu Wang
- Jiangxi University of Traditional Chinese Medicine, Nanchang, 330002, China
| | - Shi-Lin Yang
- State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Nanchang, 330006, China
| | - Yu-Lin Feng
- State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Nanchang, 330006, China.
| |
Collapse
|
7
|
Shen L, Zhang T, Yang Y, Lu D, Xu A, Li K. FPS-ZM1 Alleviates Neuroinflammation in Focal Cerebral Ischemia Rats via Blocking Ligand/RAGE/DIAPH1 Pathway. ACS Chem Neurosci 2021; 12:63-78. [PMID: 33300334 DOI: 10.1021/acschemneuro.0c00530] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Receptor for advanced glycation end products (RAGEs), a multiligand receptor belonging to the cell-surface immunoglobulin superfamily, has been reported to play a crucial role in neuroinflammation and neurodegenerative diseases. Here, we tested our hypothesis that the RAGE-specific antagonist FPS-ZM1 is neuroprotective against ischemic brain injury. Distal middle cerebral artery occlusion (MCAO) or sham operation was performed on anesthetized Sprague-Dawley male rats (n = 60), which were then treated with FPS-ZM1 or vehicle (four groups in total = Vehicle + MCAO, FPS-ZM1 + MCAO, Vehicle + sham, and FPS-ZM1 + sham). After 1 week, neurological function was evaluated, and then, brain tissues were collected for 2,3,5-triphenyltetrazolium chloride staining, Nissl staining, TUNEL staining, Western blotting, and immunohistochemical analyses. FPS-ZM1 treatment after MCAO markedly attenuated neurological deficits and reduced the infarct area. More interestingly, FPS-ZM1 inhibited ischemia-induced astrocytic activation and microgliosis and decreased the elevated levels of proinflammatory cytokines. Furthermore, FPS-ZM1 blocked the increase in the level of RAGE and, notably, of DIAPH1, the key cytoplasmic hub for RAGE-ligand-mediated activation of cellular signaling. Accordingly, FPS-ZM1 also reversed the MCAO-induced increase in phosphorylation of NF-κB targets that are potentially downstream from RAGE/DIAPH1. Our findings reveal that FPS-ZM1 treatment reduces neuroinflammation in rats with focal cerebral ischemia and further suggest that the ligand/RAGE/DIAPH1 pathway contributes to this FPS-ZM1-mediated alleviation of neuroinflammation.
Collapse
Affiliation(s)
- Lingling Shen
- Department of Neurology and Stroke Centre, the Fist Affiliated Hospital of Jinan University, Guangzhou 510632, China
- Clinical Neuroscience Institute of Jinan University, Guangzhou 510632, China
| | - Tianyuan Zhang
- Department of Neurology and Stroke Centre, the Fist Affiliated Hospital of Jinan University, Guangzhou 510632, China
- Clinical Neuroscience Institute of Jinan University, Guangzhou 510632, China
| | - Yu Yang
- Department of Neurology and Stroke Centre, the Fist Affiliated Hospital of Jinan University, Guangzhou 510632, China
- Clinical Neuroscience Institute of Jinan University, Guangzhou 510632, China
| | - Dan Lu
- Department of Neurology and Stroke Centre, the Fist Affiliated Hospital of Jinan University, Guangzhou 510632, China
- Clinical Neuroscience Institute of Jinan University, Guangzhou 510632, China
| | - Anding Xu
- Department of Neurology and Stroke Centre, the Fist Affiliated Hospital of Jinan University, Guangzhou 510632, China
- Clinical Neuroscience Institute of Jinan University, Guangzhou 510632, China
| | - Keshen Li
- Clinical Neuroscience Institute of Jinan University, Guangzhou 510632, China
| |
Collapse
|
8
|
Liu SS, Wu F, Jin YM, Chang WQ, Xu TM. HDAC11: a rising star in epigenetics. Biomed Pharmacother 2020; 131:110607. [PMID: 32841898 DOI: 10.1016/j.biopha.2020.110607] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/31/2020] [Accepted: 08/02/2020] [Indexed: 02/08/2023] Open
Abstract
Epigenetic mechanisms, such as acetylation, methylation, and succinylation, play pivotal roles in the regulation of multiple normal biological processes, including neuron regulation, hematopoiesis, bone cell maturation, and metabolism. In addition, epigenetic mechanisms are closely associated with the pathological processes of various diseases, such as metabolic diseases, autoimmune diseases and cancers. Epigenetic changes may precede genetic mutation, so research on epigenetic changes and regulation may be important for the early detection and diagnosis of disease. Histone deacetylase11 (HDAC11) is the newest member of the histone deacetylase (HDAC) family and the only class IV histone deacetylase. HDAC11 has different expression levels and biological functions in different systems of the human body and is among the top 1 to 4% of genes overexpressed in cancers, such as breast cancer, hepatocellular carcinoma and renal pelvis urothelial carcinoma. This article analyzes the role and mechanism of HDAC11 in disease, especially in tumorigenesis, in an attempt to provide new ideas for clinical and basic research.
Collapse
Affiliation(s)
- Shan-Shan Liu
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, 130041, Jilin Province, China.
| | - Fei Wu
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, 130041, Jilin Province, China.
| | - Yue-Mei Jin
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, 130041, Jilin Province, China.
| | - Wei-Qin Chang
- Department of Surgery, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun, 130041, Jilin Province, China.
| | - Tian-Min Xu
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, 130041, Jilin Province, China.
| |
Collapse
|
9
|
Sun Z, Yang T, Wang Y, Li C, Yang Y, Wang D, Guo J, Shi T, Wang Y, Qu Y, Wei Q, Feng C. Propionic acid abrogates the deleterious effects of cerebral ischemic reperfusion injury through nuclear factor-κb signaling in mice. Pharmacogn Mag 2020. [DOI: 10.4103/pm.pm_306_19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
10
|
Chrysin prevents cognitive and hippocampal long-term potentiation deficits and inflammation in rat with cerebral hypoperfusion and reperfusion injury. Life Sci 2019; 226:202-209. [DOI: 10.1016/j.lfs.2019.04.027] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 04/05/2019] [Accepted: 04/12/2019] [Indexed: 01/05/2023]
|
11
|
Raffaeli G, Ghirardello S, Passera S, Mosca F, Cavallaro G. Oxidative Stress and Neonatal Respiratory Extracorporeal Membrane Oxygenation. Front Physiol 2018; 9:1739. [PMID: 30564143 PMCID: PMC6288438 DOI: 10.3389/fphys.2018.01739] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 11/19/2018] [Indexed: 12/16/2022] Open
Abstract
Oxidative stress is a frequent condition in critically ill patients, especially if exposed to extracorporeal circulation, and it is associated with worse outcomes and increased mortality. The inflammation triggered by the contact of blood with a non-endogenous surface, the use of high volumes of packed red blood cells and platelets transfusion, the risk of hyperoxia and the impairment of antioxidation systems contribute to the increase of reactive oxygen species and the imbalance of the redox system. This is responsible for the increased production of superoxide anion, hydrogen peroxide, hydroxyl radicals, and peroxynitrite resulting in increased lipid peroxidation, protein oxidation, and DNA damage. The understanding of the pathophysiologic mechanisms leading to redox imbalance would pave the way for the future development of preventive approaches. This review provides an overview of the clinical impact of the oxidative stress during neonatal extracorporeal support and concludes with a brief perspective on the current antioxidant strategies, with the aim to focus on the potential oxidative stress-mediated cell damage that has been implicated in both short and long-term outcomes.
Collapse
Affiliation(s)
- Genny Raffaeli
- NICU, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Stefano Ghirardello
- NICU, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Sofia Passera
- NICU, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Fabio Mosca
- NICU, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Giacomo Cavallaro
- NICU, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
12
|
Wanve M, Kaur H, Sarmah D, Saraf J, Pravalika K, Vats K, Kalia K, Borah A, Yavagal DR, Dave KR, Bhattacharya P. Therapeutic spectrum of interferon-β in ischemic stroke. J Neurosci Res 2018; 97:116-127. [PMID: 30320448 PMCID: PMC7167007 DOI: 10.1002/jnr.24333] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 09/05/2018] [Accepted: 09/05/2018] [Indexed: 12/16/2022]
Abstract
Ischemic stroke is devastating and a major cause of morbidity and mortality worldwide. To date, only clot retrieval devices and/or intravenous tissue plasminogen activators (tPA) have been approved by the US-FDA for the treatment of acute ischemic stroke. Therefore, there is an urgent need to develop an effective treatment for stroke that can have limited shortcomings and broad spectrum of applications. Interferon-beta (IFN-β), an endogenous cytokine and a key anti-inflammatory agent, contributes toward obviating deleterious stroke outcomes. Therefore, exploring the role of IFN-β may be a promising alternative approach for stroke intervention in the future. In the present review, we have discussed about IFN-β along with its different mechanistic roles in ischemic stroke. Furthermore, therapeutic approaches targeting the inflammatory cascade with IFN-β therapy that may be helpful in improving stroke outcome are also discussed.
Collapse
Affiliation(s)
- Madhuri Wanve
- Department of Pharmacology and ToxicologyNational Institute of Pharmaceutical Education and Research (NIPER)GandhinagarIndia
| | - Harpreet Kaur
- Department of Pharmacology and ToxicologyNational Institute of Pharmaceutical Education and Research (NIPER)GandhinagarIndia
| | - Deepaneeta Sarmah
- Department of Pharmacology and ToxicologyNational Institute of Pharmaceutical Education and Research (NIPER)GandhinagarIndia
| | - Jackson Saraf
- Department of Pharmacology and ToxicologyNational Institute of Pharmaceutical Education and Research (NIPER)GandhinagarIndia
| | - Kanta Pravalika
- Department of Pharmacology and ToxicologyNational Institute of Pharmaceutical Education and Research (NIPER)GandhinagarIndia
| | - Kanchan Vats
- Department of Pharmacology and ToxicologyNational Institute of Pharmaceutical Education and Research (NIPER)GandhinagarIndia
| | - Kiran Kalia
- Department of Pharmacology and ToxicologyNational Institute of Pharmaceutical Education and Research (NIPER)GandhinagarIndia
| | - Anupom Borah
- Cellular and Molecular Neurobiology Laboratory, Department of Life Science and BioinformaticsAssam UniversitySilcharIndia
| | - Dileep R. Yavagal
- Department of Neurology and NeurosurgeryUniversity of Miami Miller School of MedicineMiamiFlorida
| | - Kunjan R. Dave
- Department of Neurology and NeurosurgeryUniversity of Miami Miller School of MedicineMiamiFlorida
| | - Pallab Bhattacharya
- Department of Pharmacology and ToxicologyNational Institute of Pharmaceutical Education and Research (NIPER)GandhinagarIndia
| |
Collapse
|
13
|
Memory deficits and hippocampal inflammation in cerebral hypoperfusion and reperfusion in male rats: Neuroprotective role of vanillic acid. Life Sci 2018; 211:126-132. [PMID: 30195619 DOI: 10.1016/j.lfs.2018.08.065] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 08/20/2018] [Accepted: 08/27/2018] [Indexed: 11/20/2022]
Abstract
Ischemic stroke is one of the leading causes of neurological deterioration and mortality worldwide. Neuroprotective strategies are being investigated to minimize cognitive deficits after ischemic events. Here we investigated the neuroprotective potential of vanillic acid (VA) in an animal model of transient bilateral common carotid artery occlusion and reperfusion (BCCAO/R). Adult male Wistar rats (250-300 g) were randomly divided in 4 groups and submitted to either cerebral hypoperfusion-reperfusion or a sham surgery after two-weeks of pretreatment with VA and/or normal saline. To induce the animal model of hypoperfusion, bilateral common carotid arteries were occluded (2VO model) for 30 min, followed by 72 h of reperfusion. Subsequently, their cognitive performance was evaluated in a Morris water maze (MWM) test, and also hippocampi were removed for ELISA assays and TUNEL staining test. The results showed that 2VO significantly reduced the spatial memory performance in MWM. As well as, BCCAO/R increased the level of IL-6, TNF-α and TUNEL positive cells, and also decreased the contents of IL-10 in the hippocampus of vehicle- pretreated groups as compared to the sham-operated groups. Furthermore, 14 consecutive days pretreatment with VA significantly restored the spatial memory, decreased the levels of IL-6, TNF-α and TUNEL positive cells and also increased the IL-10 levels in the hippocampi of the BCCAO/R rats. VA alone did not show any change neither in the status of various cytokines nor behavioral and TUNEL staining tests over sham values. Our data confirm that VA could potentially serve as a novel, promising, and accessible neuroprotective agent against cerebrovascular insufficiency states and vascular dementia.
Collapse
|
14
|
Zhang YY, Huang NN, Fan YC, Li YS, Zhao J, Wang D, Zhang F, Li XH. Peripheral tumor necrosis factor-a-induced protein 8-like 2 mRNA level for predicting 3-month mortality of patients with acute ischemic stroke. J Neurol 2018; 265:2573-2586. [PMID: 30171409 DOI: 10.1007/s00415-018-9036-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 08/20/2018] [Accepted: 08/23/2018] [Indexed: 12/31/2022]
|
15
|
Thioredoxin-Interacting Protein (TXNIP) in Cerebrovascular and Neurodegenerative Diseases: Regulation and Implication. Mol Neurobiol 2018; 55:7900-7920. [PMID: 29488135 DOI: 10.1007/s12035-018-0917-z] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Accepted: 01/21/2018] [Indexed: 02/07/2023]
Abstract
Neurological diseases, including acute attacks (e.g., ischemic stroke) and chronic neurodegenerative diseases (e.g., Alzheimer's disease), have always been one of the leading cause of morbidity and mortality worldwide. These debilitating diseases represent an enormous disease burden, not only in terms of health suffering but also in economic costs. Although the clinical presentations differ for these diseases, a growing body of evidence suggests that oxidative stress and inflammatory responses in brain tissue significantly contribute to their pathology. However, therapies attempting to prevent oxidative damage or inhibiting inflammation have shown little success. Identification and targeting endogenous "upstream" mediators that normalize such processes will lead to improve therapeutic strategy of these diseases. Thioredoxin-interacting protein (TXNIP) is an endogenous inhibitor of the thioredoxin (TRX) system, a major cellular thiol-reducing and antioxidant system. TXNIP regulating redox/glucose-induced stress and inflammation, now is known to get upregulated in stroke and other brain diseases, and represents a promising therapeutic target. In particular, there is growing evidence that glucose strongly induces TXNIP in multiple cell types, suggesting possible physiological roles of TXNIP in glucose metabolism. Recently, a significant body of literature has supported an essential role of TXNIP in the activation of the NOD-like receptor protein (NLRP3)-inflammasome, a well-established multi-molecular protein complex and a pivotal mediator of sterile inflammation. Accordingly, TXNIP has been postulated to reside centrally in detecting cellular damage and mediating inflammatory responses to tissue injury. The majority of recent studies have shown that pharmacological inhibition or genetic deletion of TXNIP is neuroprotective and able to reduce detrimental aspects of pathology following cerebrovascular and neurodegenerative diseases. Conspicuously, the mainstream of the emerging evidences is highlighting TXNIP link to damaging signals in endothelial cells. Thereby, here, we keep the trend to present the accumulative data on CNS diseases dealing with vascular integrity. This review aims to summarize evidence supporting the significant contribution of regulatory mechanisms of TXNIP with the development of brain diseases, explore pharmacological strategies of targeting TXNIP, and outline obstacles to be considered for efficient clinical translation.
Collapse
|
16
|
Zhang YY, Huang NN, Zhao YX, Li YS, Wang D, Fan YC, Li XH. Elevated Tumor Necrosis Factor-a-induced Protein 8-like 2 mRNA from Peripheral Blood Mononuclear Cells in Patients with Acute Ischemic Stroke. Int J Med Sci 2018; 15:1713-1722. [PMID: 30588195 PMCID: PMC6299423 DOI: 10.7150/ijms.27817] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 10/18/2018] [Indexed: 12/23/2022] Open
Abstract
Background: Tumor necrosis factor-a-induced protein 8-like 2 (TIPE2) is a novel regulator of immunity and protects against experimental stroke. However, the expression and function of TIPE2 in patients with acute ischemic stroke has not been well demonstrated. Methods: A total of 182 consecutive patients with acute ischemic stroke and 40 healthy controls were included during November 2015 to June 2016. The mRNA levels of TIPE2, interleukin(IL)-1β, IL-10, IL-6, nuclear factor(NF)-κβ, activator protein(AP)-1, interferon(IFN)-γ and tumor necrosis factor(TNF)-α from peripheral blood mononuclear cells were determined using real time quantitative reverse transcriptase polymerase chain reaction. The severity of stroke was assessed using the National Institutes of Health Stroke Scale (NIHSS) score. Results: The median mRNA levels of TIPE2, TNF-α, AP-1, IFN-γ and NF-κβ in patients with acute ischemic stroke were significantly higher than healthy controls (all P<0.001, respectively). Of note, TIPE2 mRNA showed an increasing trend on a time-dependent manner after the onset of stroke. Furthermore, TIPE2 mRNA was negatively associated with lesion volumes (r=-0.23, P<0.01), NIHSS(r=-0.15, P<0.05), TNF-α(r=-0.33,P<0.001), AP-1(r=-0.28,P<0.001), IFN-γ (r=-0.16, P<0.05) and NF-κβ (r=-0.13, P<0.05), but positively associated with IL-6(r=0.14, P<0.05) and IL-10(r=-0.31, P<0.001). Hierarchy cluster analysis showed that TIPE2 mRNA has nearest membership with TNF-α, followed by IL-6, NF-κβ, AP-1, IL-10, IL-1β and IFN-γ. In addition, TIPE2 mRNA in survivals (n=149) was significantly higher than nonsurvivals (n=33) (P<0.001), and showed a great odd ratio (0.52, 95% confidence interval: 0.349-0.760, P<0.001) on 3-month mortality. Conclusions: TIPE2 mRNA contributed to the immune response of stroke and might be a potential biomarker for the mortality of acute ischemic stroke.
Collapse
Affiliation(s)
- Yuan-Yuan Zhang
- Department of Neurology, Jinan Central Hospital affiliated to Shandong University, Jinan 250013, China
| | - Na-Na Huang
- Department of Neurology, Jinan Central Hospital affiliated to Shandong University, Jinan 250013, China
| | - Yan-Xin Zhao
- Department of Neurology, Jinan Central Hospital affiliated to Shandong University, Jinan 250013, China
| | - Yan-Shuang Li
- Department of Neurology, Jinan Central Hospital affiliated to Shandong University, Jinan 250013, China
| | - Dong Wang
- Department of Neurology, Jinan Central Hospital affiliated to Shandong University, Jinan 250013, China
| | - Yu-Chen Fan
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Xiao-Hong Li
- Department of Neurology, Jinan Central Hospital affiliated to Shandong University, Jinan 250013, China
| |
Collapse
|
17
|
Wang YM, Liu ZY, Ai YH, Zhang LN, Zou Y, Peng QY. Blocking the CD38/cADPR pathway plays a double-edged role in LPS stimulated microglia. Neuroscience 2017; 361:34-42. [PMID: 28807785 DOI: 10.1016/j.neuroscience.2017.08.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 08/02/2017] [Accepted: 08/03/2017] [Indexed: 01/04/2023]
Abstract
Whether the CD38/cyclic ADP-ribose (cADPR) pathway plays a protective or detrimental role in neuroinflammation remains controversial. This study aimed to determine the role of CD38 in neuroinflammation using lipopolysaccharide (LPS)-stimulated BV2 microglial cells and co-cultured Neuro-2a (N2a) cells. In monoculture experiments, BV2 cells were divided into control, CD38 interference (CD38Ri), negative control (NC), LPS, CD38Ri+LPS, NC+LPS and 8-Br-cADPR+LPS groups. In co-culture experiments, N2a cells were co-cultured with BV2 cells for 48h. Nicotinamide adenine dinucleotide (NAD+), cADPR and intracellular Ca2+ levels and CD38 expression increased significantly in LPS-stimulated BV2 cells. CD38 knockdown or 8-Br-cADPR treatment significantly reduced NAD+, cADPR and intracellular Ca2+ levels. CD38 knockdown increased iNOS and NO levels in BV2 cells without LPS treatment; however, CD38 knockdown or 8-Br-cADPR treatment reduced iNOS and NO levels in BV2 cells with LPS treatment. CD38 knockdown increased the ratio of TUNEL-positive cells and cleaved Caspase 3/Caspase 3 ratio, and decreased the Bcl-2/Bax ratio in BV2 cells without LPS treatment; however, CD38 knockdown reduced the TUNEL positivity in BV2 cells with LPS treatment. CD38 knockdown or 8-Br-cADPR inhibited TNF-α, IL-6 (interleukin-6) and IL-1β levels in LPS-stimulated BV2 cells. Co-culture with CD38 knockdown or 8-Br-cADPR-treated BV2 cells did not influence apoptosis or iNOS expression in N2a cells. In conclusion, our results indicate that blocking the CD38/cADPR pathway reduces intracellular Ca2+, NO and the secretion of proinflammatory cytokines. CD38 knockdown exerted a detrimental effect in apoptosis and NO production in normal microglia, but played a protective role in apoptosis and NO production in LPS-stimulated microglia.
Collapse
Affiliation(s)
- Yi-Min Wang
- Department of Critical Care Medicine, Xiang-Ya Hospital, Central South University, Changsha, Hunan Province, China
| | - Zhi-Yong Liu
- Department of Critical Care Medicine, Xiang-Ya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yu-Hang Ai
- Department of Critical Care Medicine, Xiang-Ya Hospital, Central South University, Changsha, Hunan Province, China
| | - Li-Na Zhang
- Department of Critical Care Medicine, Xiang-Ya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yu Zou
- Department of Anesthesia, Xiang-Ya Hospital, Central South University, Changsha, Hunan Province, China
| | - Qian-Yi Peng
- Department of Critical Care Medicine, Xiang-Ya Hospital, Central South University, Changsha, Hunan Province, China.
| |
Collapse
|
18
|
Xu Z, Liu Y, Yang D, Yuan F, Ding J, Wang L, Qu M, Yang G, Tian H. Glibenclamide–sulfonylurea receptor 1 antagonist alleviates LPS-induced BV2 cell activation through the p38/MAPK pathway. RSC Adv 2017. [DOI: 10.1039/c7ra03042h] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We investigated the anti-neuroinflammatory activity and mechanism of glibenclamide, sulfonylurea receptor 1 (Sur1) antagonist, against LPS-induced microglial activationin vitro.
Collapse
Affiliation(s)
- Zhiming Xu
- Department of Neurosurgery
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital
- Shanghai
- China
| | - Yingliang Liu
- Department of Neurosurgery
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital
- Shanghai
- China
| | - Dianxu Yang
- Department of Neurosurgery
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital
- Shanghai
- China
| | - Fang Yuan
- Department of Neurosurgery
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital
- Shanghai
- China
| | - Jun Ding
- Department of Neurosurgery
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital
- Shanghai
- China
| | - Liping Wang
- Department of Neurology
- Ruijin Hospital
- Shanghai Jiao Tong University School of Medicine
- Shanghai
- China
| | - Meijie Qu
- Department of Neurology
- Ruijin Hospital
- Shanghai Jiao Tong University School of Medicine
- Shanghai
- China
| | - Guoyuan Yang
- Department of Neurology
- Ruijin Hospital
- Shanghai Jiao Tong University School of Medicine
- Shanghai
- China
| | - Hengli Tian
- Department of Neurosurgery
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital
- Shanghai
- China
| |
Collapse
|
19
|
Iizumi T, Takahashi S, Mashima K, Minami K, Izawa Y, Abe T, Hishiki T, Suematsu M, Kajimura M, Suzuki N. A possible role of microglia-derived nitric oxide by lipopolysaccharide in activation of astroglial pentose-phosphate pathway via the Keap1/Nrf2 system. J Neuroinflammation 2016; 13:99. [PMID: 27143001 PMCID: PMC4855896 DOI: 10.1186/s12974-016-0564-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 04/26/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Toll-like receptor 4 (TLR4) plays a pivotal role in the pathophysiology of stroke-induced inflammation. Both astroglia and microglia express TLR4, and endogenous ligands produced in the ischemic brain induce inflammatory responses. Reactive oxygen species (ROS), nitric oxide (NO), and inflammatory cytokines produced by TLR4 activation play harmful roles in neuronal damage after stroke. Although astroglia exhibit pro-inflammatory responses upon TLR4 stimulation by lipopolysaccharide (LPS), they may also play cytoprotective roles via the activation of the pentose phosphate pathway (PPP), reducing oxidative stress by glutathione peroxidase. We investigated the mechanisms by which astroglia reduce oxidative stress via the activation of PPP, using TLR4 stimulation and hypoxia in concert with microglia. METHODS In vitro experiments were performed using cells prepared from Sprague-Dawley rats. Coexisting microglia in the astroglial culture were chemically eliminated using L-leucine methyl ester (LME). Cells were exposed to LPS (0.01 μg/mL) or hypoxia (1 % O2) for 12-15 h. PPP activity was measured using [1-(14)C]glucose and [6-(14)C]glucose. ROS and NO production were measured using 2',7'-dichlorodihydrofluorescein diacetate and diaminofluorescein-FM diacetate, respectively. The involvement of nuclear factor-erythroid-2-related factor 2 (Nrf2), a cardinal transcriptional factor under stress conditions that regulates glucose 6-phosphate dehydrogenase, the rate-limiting enzyme of PPP, was evaluated using immunohistochemistry. RESULTS Cultured astroglia exposed to LPS elicited 20 % increases in PPP flux, and these actions of astroglia appeared to involve Nrf2. However, the chemical depletion of coexisting microglia eliminated both increases in PPP and astroglial nuclear translocation of Nrf2. LPS induced ROS and NO production in the astroglial culture containing microglia but not in the microglia-depleted astroglial culture. LPS enhanced astroglial ROS production after glutathione depletion. U0126, an upstream inhibitor of mitogen-activated protein kinase, eliminated LPS-induced NO production, whereas ROS production was unaffected. U0126 also eliminated LPS-induced PPP activation in astroglial-microglial culture, indicating that microglia-derived NO mediated astroglial PPP activation. Hypoxia induced astroglial PPP activation independent of the microglia-NO pathway. Elimination of ROS and NO production by sulforaphane, a natural Nrf2 activator, confirmed the astroglial protective mechanism. CONCLUSIONS Astroglia in concert with microglia may play a cytoprotective role for countering oxidative stress in stroke.
Collapse
Affiliation(s)
- Takuya Iizumi
- Department of Neurology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan
| | - Shinichi Takahashi
- Department of Neurology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan.
| | - Kyoko Mashima
- Department of Neurology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan
| | - Kazushi Minami
- Department of Neurology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan
| | - Yoshikane Izawa
- Department of Neurology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan
| | - Takato Abe
- Department of Neurology, Osaka City University Graduate School of Medicine, Osaka-shi, 545-8585, Osaka , Japan
| | - Takako Hishiki
- Department of Biochemistry, Keio University School of Medicine, Shinjuku-ku, 160-8582, Tokyo, Japan.,Clinical and Translational Research Center, Keio University School of Medicine, Shinjuku-ku, 160-8582, Tokyo, Japan.,JST Exploratory Research for Advanced Technology (ERATO) Suematsu Gas Biology Project, Shinjuku-ku, 160-8582, Tokyo , Japan
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, Shinjuku-ku, 160-8582, Tokyo, Japan.,JST Exploratory Research for Advanced Technology (ERATO) Suematsu Gas Biology Project, Shinjuku-ku, 160-8582, Tokyo , Japan
| | - Mayumi Kajimura
- Department of Biochemistry, Keio University School of Medicine, Shinjuku-ku, 160-8582, Tokyo, Japan.,JST Exploratory Research for Advanced Technology (ERATO) Suematsu Gas Biology Project, Shinjuku-ku, 160-8582, Tokyo , Japan
| | - Norihiro Suzuki
- Department of Neurology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan
| |
Collapse
|
20
|
Fang B, Li XQ, Bao NR, Tan WF, Chen FS, Pi XL, Zhang Y, Ma H. Role of autophagy in the bimodal stage after spinal cord ischemia reperfusion injury in rats. Neuroscience 2016; 328:107-16. [PMID: 27109922 DOI: 10.1016/j.neuroscience.2016.04.019] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 04/13/2016] [Accepted: 04/15/2016] [Indexed: 10/21/2022]
Abstract
Autophagy plays an important role in spinal cord ischemia reperfusion (I/R) injury, but its neuroprotective or neurodegenerative role remains controversial. The extent and persistence of autophagy activation may be the critical factor to explain the opposing effects. In this study, the different roles and action mechanisms of autophagy in the early and later stages after I/R injury were investigated in rats. Thespinal cord I/R injury was induced by 14-min occlusion of the aortic arch, after which rats were treated with autophagic inhibitor (3-methyladenine, 3-MA) or agonist (rapamycin) immediately or 48h following the injury. Autophagy markers, microtubule-associated protein light chain 3-II (LC3-II) and Beclin 1 increased and peaked at the early stage (8h) and the later stage (72h) after spinal cord I/R injury. Beclin 1 was mostly expressed in neurons, but was also expressed to an extent in astrocytes, microglia and vascular endothelial cells. 8h after injury, rats treated with 3-MA showed a decrease in the hind-limb Basso-Beattie-Bresnahan (BBB) motor function scores, surviving motor neurons, and B-cell lymphoma-2 (Bcl-2) expression, and increase in the terminal deoxynucleotidyl transferase (TdT)-mediated dUTP-biotin nick end labeling (TUNEL)-positive cells, Bcl-2-associated X protein (Bax), tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) expression, and activation of microglia, while those treated with rapamycin showed opposing effects. However, 72h after injury, rats treated with 3-MA improved the BBB scores, and the surviving motor neurons, and reduced the autophagic cell death, while those treated with rapamycin had adverse effects. These findings provide the first evidence that early activated autophagy alleviates spinal cord I/R injury via inhibiting apoptosis and inflammation; however later excessively elevated autophagy aggravates I/R injury through inducing autophagic cell death.
Collapse
Affiliation(s)
- Bo Fang
- Department of Anesthesiology, First Affiliated Hospital, China Medical University, North Nanjing Street, No. 155, Shenyang, Liaoning 110001, PR China.
| | - Xiao-Qian Li
- Department of Anesthesiology, First Affiliated Hospital, China Medical University, North Nanjing Street, No. 155, Shenyang, Liaoning 110001, PR China.
| | - Na-Ren Bao
- Department of Anesthesiology, First Affiliated Hospital, China Medical University, North Nanjing Street, No. 155, Shenyang, Liaoning 110001, PR China.
| | - Wen-Fei Tan
- Department of Anesthesiology, First Affiliated Hospital, China Medical University, North Nanjing Street, No. 155, Shenyang, Liaoning 110001, PR China.
| | - Feng-Shou Chen
- Department of Anesthesiology, First Affiliated Hospital, China Medical University, North Nanjing Street, No. 155, Shenyang, Liaoning 110001, PR China.
| | - Xiao-Li Pi
- Department of Anesthesiology, First Affiliated Hospital, China Medical University, North Nanjing Street, No. 155, Shenyang, Liaoning 110001, PR China.
| | - Ying Zhang
- Department of Anesthesiology, First Affiliated Hospital, China Medical University, North Nanjing Street, No. 155, Shenyang, Liaoning 110001, PR China.
| | - Hong Ma
- Department of Anesthesiology, First Affiliated Hospital, China Medical University, North Nanjing Street, No. 155, Shenyang, Liaoning 110001, PR China.
| |
Collapse
|
21
|
Nemeth CL, Miller AH, Tansey MG, Neigh GN. Inflammatory mechanisms contribute to microembolism-induced anxiety-like and depressive-like behaviors. Behav Brain Res 2016; 303:160-7. [DOI: 10.1016/j.bbr.2016.01.057] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 01/21/2016] [Accepted: 01/24/2016] [Indexed: 12/17/2022]
|
22
|
Lu H, Liu X, Zhang N, Zhu X, Liang H, Sun L, Cheng Y. Neuroprotective Effects of Brain-Derived Neurotrophic Factor and Noggin-Modified Bone Mesenchymal Stem Cells in Focal Cerebral Ischemia in Rats. J Stroke Cerebrovasc Dis 2016; 25:410-8. [DOI: 10.1016/j.jstrokecerebrovasdis.2015.10.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 09/09/2015] [Accepted: 10/17/2015] [Indexed: 11/24/2022] Open
|
23
|
Rocamonde B, Paradells S, Garcia Esparza MA, Vives MS, Sauro S, Ramos CM, Pradas MM, Soria JM. Combined application of polyacrylate scaffold and lipoic acid treatment promotes neural tissue reparation after brain injury. Brain Inj 2016; 30:208-16. [PMID: 26745450 DOI: 10.3109/02699052.2015.1091505] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
PRIMARY OBJECTIVE The aim of this study was to investigate the reparative potential of a polymeric scaffold designed for brain tissue repair in combination with lipoic acid. RESEARCH DESIGN Histological, cytological and structural analysis of a combined treatment after a brain cryo-injury model in rats. METHODS AND PROCEDURES Adult Wistar rats were subjected to cryogenic brain injury. A channelled-porous scaffold of ethyl acrylate and hydroxyethylacrylate, p(EA-co-HEA) was grafted into cerebral penumbra alone or combined with intraperitoneal LA administration. Histological and cytological evaluation was performed after 15 and 60 days and structural magnetic resonance (MRI) assessment was performed at 2 and 6 months after the surgery. MAIN OUTCOMES AND RESULTS The scaffold was suitable for the establishment of different cellular types. The results obtained suggest that this strategy promotes blood vessels formation, decreased microglial response and neuron migration, particularly when LA was administrated. CONCLUSIONS These evidences demonstrated that the combination of a channelled polymer scaffold with LA administration may represent a potential treatment for neural tissue repair after brain injury.
Collapse
Affiliation(s)
- Brenda Rocamonde
- a Facultad Ciencias de la Salud, Universidad CEU-Cardenal Herrera , Valencia , Spain
| | - Sara Paradells
- a Facultad Ciencias de la Salud, Universidad CEU-Cardenal Herrera , Valencia , Spain
| | | | - Mavi Sánchez Vives
- b Institut D'Investigacions Biomèdiques August Pi i Sunyer-IDIBAPS , Barcelona , Spain
| | - Salvatore Sauro
- a Facultad Ciencias de la Salud, Universidad CEU-Cardenal Herrera , Valencia , Spain
| | - Cristina Martínez Ramos
- c Centro de Biomateriales e Ingeniería Tisular, Universidad Politécnica de Valencia , Valencia , Spain
| | - Manuel Monleón Pradas
- c Centro de Biomateriales e Ingeniería Tisular, Universidad Politécnica de Valencia , Valencia , Spain
| | - José Miguel Soria
- a Facultad Ciencias de la Salud, Universidad CEU-Cardenal Herrera , Valencia , Spain.,d Instituto de Ciencias Biomédicas, Universidad CEU-Cardenal Herrera , Moncada , Valencia , Spain
| |
Collapse
|
24
|
Kuo P, Scofield BA, Yu I, Chang F, Ganea D, Yen J. Interferon-β Modulates Inflammatory Response in Cerebral Ischemia. J Am Heart Assoc 2016; 5:e002610. [PMID: 26747000 PMCID: PMC4859377 DOI: 10.1161/jaha.115.002610] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Accepted: 11/04/2015] [Indexed: 01/17/2023]
Abstract
BACKGROUND Stroke is a leading cause of death in the world. In >80% of strokes, the initial acute phase of ischemic injury is due to the occlusion of a blood vessel resulting in severe focal hypoperfusion, excitotoxicity, and oxidative damage. Interferon-β (IFNβ), a cytokine with immunomodulatory properties, was approved by the US Food and Drug Administration for the treatment of relapsing-remitting multiple sclerosis for more than a decade. Its anti-inflammatory properties and well-characterized safety profile suggest that IFNβ has therapeutic potential for the treatment of ischemic stroke. METHODS AND RESULTS We investigated the therapeutic effect of IFNβ in the mouse model of transient middle cerebral artery occlusion/reperfusion. We found that IFNβ not only reduced infarct size in ischemic brains but also lessened neurological deficits in ischemic stroke animals. Further, multiple molecular mechanisms by which IFNβ modulates ischemic brain inflammation were identified. IFNβ reduced central nervous system infiltration of monocytes/macrophages, neutrophils, CD4(+) T cells, and γδ T cells; inhibited the production of inflammatory mediators; suppressed the expression of adhesion molecules on brain endothelial cells; and repressed microglia activation in the ischemic brain. CONCLUSIONS Our results demonstrate that IFNβ exerts a protective effect against ischemic stroke through its anti-inflammatory properties and suggest that IFNβ is a potential therapeutic agent, targeting the reperfusion damage subsequent to the treatment with tissue plasminogen activator.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents/pharmacology
- Brain/drug effects
- Brain/immunology
- Brain/metabolism
- Brain/pathology
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Cell Adhesion Molecules/metabolism
- Cell Line
- Chemotaxis, Leukocyte/drug effects
- Disease Models, Animal
- Endothelial Cells/drug effects
- Endothelial Cells/immunology
- Endothelial Cells/metabolism
- Infarction, Middle Cerebral Artery/immunology
- Infarction, Middle Cerebral Artery/metabolism
- Infarction, Middle Cerebral Artery/pathology
- Infarction, Middle Cerebral Artery/prevention & control
- Inflammation Mediators/metabolism
- Interferon-beta/pharmacology
- Macrophages/drug effects
- Macrophages/immunology
- Macrophages/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Microglia/drug effects
- Microglia/immunology
- Microglia/metabolism
- Neuroprotective Agents/pharmacology
- Neutrophil Infiltration/drug effects
- Neutrophils/drug effects
- Neutrophils/immunology
- Neutrophils/metabolism
- Receptor, Interferon alpha-beta/deficiency
- Receptor, Interferon alpha-beta/genetics
Collapse
Affiliation(s)
- Ping‐Chang Kuo
- Department of Microbiology and ImmunologyIndiana University School of MedicineFort WayneIN
| | - Barbara A. Scofield
- Department of Microbiology and ImmunologyIndiana University School of MedicineFort WayneIN
| | - I‐Chen Yu
- Department of Anatomy and Cell BiologyIndiana University School of MedicineFort WayneIN
| | - Fen‐Lei Chang
- Department of NeurologyIndiana University School of MedicineFort WayneIN
| | - Doina Ganea
- Department of Microbiology and ImmunologyTemple University School of MedicinePhiladelphiaPA
| | - Jui‐Hung Yen
- Department of Microbiology and ImmunologyIndiana University School of MedicineFort WayneIN
| |
Collapse
|
25
|
Chisholm NC, Sohrabji F. Astrocytic response to cerebral ischemia is influenced by sex differences and impaired by aging. Neurobiol Dis 2016; 85:245-253. [PMID: 25843666 PMCID: PMC5636213 DOI: 10.1016/j.nbd.2015.03.028] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 03/16/2015] [Accepted: 03/26/2015] [Indexed: 12/21/2022] Open
Abstract
Ischemic stroke occurs more often among the elderly, and within this demographic, women are at an increased risk for stroke and have poorer functional recovery than men. This is also well replicated in animal studies where aging females are shown to have more extensive brain tissue loss as compared to adult females. Astrocytes provide nutrients for neurons, regulate glutamate levels, and release neurotrophins and thus play a key role in the events that occur following ischemia. In addition, astrocytes express receptors for gonadal hormones and synthesize several neurosteroids suggesting that the sex differences in stroke outcome may be mediated through astrocytes. This review discusses key astrocytic responses to ischemia including, reactive gliosis, excitotoxicity, and neuroinflammation. In light of the age and sex differences in stroke outcomes, this review highlights how aging and gonadal hormones influence these responses. Lastly, astrocyte specific changes in gene expression and epigenetic modifications during aging and following ischemia are discussed as possible molecular mechanisms for impaired astrocytic functioning.
Collapse
Affiliation(s)
- Nioka C Chisholm
- Department of Neuroscience and Experimental Therapeutics, Texas A & M Health Science Center, College of Medicine, Bryan, TX 77807, USA
| | - Farida Sohrabji
- Department of Neuroscience and Experimental Therapeutics, Texas A & M Health Science Center, College of Medicine, Bryan, TX 77807, USA.
| |
Collapse
|
26
|
Chen H, Wang C, Wei X, Ding X, Ying W. Malate-Aspartate Shuttle Inhibitor Aminooxyacetate Acid Induces Apoptosis and Impairs Energy Metabolism of Both Resting Microglia and LPS-Activated Microglia. Neurochem Res 2015; 40:1311-8. [PMID: 25998884 DOI: 10.1007/s11064-015-1589-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 03/19/2015] [Accepted: 04/22/2015] [Indexed: 01/08/2023]
Abstract
NADH shuttles mediate the transfer of the reducing equivalents of cytosolic NADH into mitochondria. Cumulating evidence has suggested that malate-aspartate shuttle (MAS), one of the two types of NADH shuttles, plays significant roles in such biological processes as glutamate synthesis in neurons. However, there has been no information regarding the roles of NADH shuttle in the survival and energy metabolism of microglia. In current study, using microglial BV2 cells as a cellular model, we determined the roles of MAS in the survival and energy metabolism of microglia by using aminooxyacetate acid (AOAA)-a widely used MAS inhibitor. Our study has suggested that AOAA can effectively inhibit the MAS activity of the cells. We also found that AOAA can induce both early- and late-stage apoptosis of resting microglia and lipopolysaccharides (LPS)-activated microglia. AOAA also induced mitochondrial depolarization, increases in the cytosolic Ca(2+) concentrations, and decreases in the intracellular ATP levels. Moreover, our study has excluded the possibility that the major nonspecific effect of AOAA-inhibition of GABA transaminase-is involved in theses effects of AOAA. Collectively, our study has provided first information suggesting significant roles of MAS in the survival and energy metabolism in both resting microglia and LPS-activated microglia.
Collapse
Affiliation(s)
- Heyu Chen
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai, 200030, People's Republic of China
| | | | | | | | | |
Collapse
|
27
|
Neuroprotective Effect of Sodium Butyrate against Cerebral Ischemia/Reperfusion Injury in Mice. BIOMED RESEARCH INTERNATIONAL 2015; 2015:395895. [PMID: 26064905 PMCID: PMC4439479 DOI: 10.1155/2015/395895] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 04/24/2015] [Indexed: 11/28/2022]
Abstract
Sodium butyrate (NaB) is a dietary microbial fermentation product of fiber and serves as an important neuromodulator in the central nervous system. In this study, we further investigated that NaB attenuated cerebral ischemia/reperfusion (I/R) injury in vivo and its possible mechanisms. NaB (5, 10 mg/kg) was administered intragastrically 3 h after the onset of reperfusion in bilateral common carotid artery occlusion (BCCAO) mice. After 24 h of reperfusion, neurological deficits scores were estimated. Morphological examination was performed by electron microscopy and hematoxylin-eosin (H&E) staining. The levels of oxidative stress and inflammatory cytokines were assessed. Apoptotic neurons were measured by TUNEL; apoptosis-related protein caspase-3, Bcl-2, Bax, the phosphorylation Akt (p-Akt), and BDNF were assayed by western blot and immunohistochemistry. The results showed that 10 mg/kg NaB treatment significantly ameliorated neurological deficit and histopathology changes in cerebral I/R injury. Moreover, 10 mg/kg NaB treatment markedly restored the levels of MDA, SOD, IL-1β, TNF-α, and IL-8. 10 mg/kg NaB treatment also remarkably inhibited the apoptosis, decreasing the levels of caspase-3 and Bax and increasing the levels of Bcl-2, p-Akt, and BDNF. This study suggested that NaB exerts neuroprotective effects on cerebral I/R injury by antioxidant, anti-inflammatory, and antiapoptotic properties and BDNF-PI3K/Akt pathway is involved in antiapoptotic effect.
Collapse
|
28
|
Vella J, Zammit C, Di Giovanni G, Muscat R, Valentino M. The central role of aquaporins in the pathophysiology of ischemic stroke. Front Cell Neurosci 2015; 9:108. [PMID: 25904843 PMCID: PMC4389728 DOI: 10.3389/fncel.2015.00108] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 04/10/2015] [Indexed: 11/16/2022] Open
Abstract
Stroke is a complex and devastating neurological condition with limited treatment options. Brain edema is a serious complication of stroke. Early edema formation can significantly contribute to infarct formation and thus represents a promising target. Aquaporin (AQP) water channels contribute to water homeostasis by regulating water transport and are implicated in several disease pathways. At least 7 AQP subtypes have been identified in the rodent brain and the use of transgenic mice has greatly aided our understanding of their functions. AQP4, the most abundant channel in the brain, is up-regulated around the peri-infarct border in transient cerebral ischemia and AQP4 knockout mice demonstrate significantly reduced cerebral edema and improved neurological outcome. In models of vasogenic edema, brain swelling is more pronounced in AQP4-null mice than wild-type providing strong evidence of the dual role of AQP4 in the formation and resolution of both vasogenic and cytotoxic edema. AQP4 is co-localized with inwardly rectifying K(+)-channels (Kir4.1) and glial K(+) uptake is attenuated in AQP4 knockout mice compared to wild-type, indicating some form of functional interaction. AQP4-null mice also exhibit a reduction in calcium signaling, suggesting that this channel may also be involved in triggering pathological downstream signaling events. Associations with the gap junction protein Cx43 possibly recapitulate its role in edema dissipation within the astroglial syncytium. Other roles ascribed to AQP4 include facilitation of astrocyte migration, glial scar formation, modulation of inflammation and signaling functions. Treatment of ischemic cerebral edema is based on the various mechanisms in which fluid content in different brain compartments can be modified. The identification of modulators and inhibitors of AQP4 offer new therapeutic avenues in the hope of reducing the extent of morbidity and mortality in stroke.
Collapse
Affiliation(s)
| | | | | | | | - Mario Valentino
- Department of Physiology and Biochemistry, University of MaltaMsida, Malta
| |
Collapse
|
29
|
Habib P, Beyer C. Regulation of brain microglia by female gonadal steroids. J Steroid Biochem Mol Biol 2015; 146:3-14. [PMID: 24607811 DOI: 10.1016/j.jsbmb.2014.02.018] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 02/24/2014] [Indexed: 12/31/2022]
Abstract
Microglial cells are the primary mediators of the CNS immune defense system and crucial for shaping inflammatory responses. They represent a highly dynamic cell population which is constantly moving and surveying their environment. Acute brain damage causes a local attraction and activation of this immune cell type which involves neuron-to-glia and glia-to-glia interactions. The prevailing view attributes microglia a "negative" role such as defense and debris elimination. More topical studies also suggest a protective and "positive" regulatory function. Estrogens and progestins exert anti-inflammatory and neuroprotective effects in the CNS in acute and chronic brain diseases. Recent work revealed that microglial cells express subsets of classical and non-classical estrogen and progesterone receptors in a highly dynamic way. In this review article, we would like to stress the importance of microglia for the spreading of neural damage during hypoxia, their susceptibility to functional modulation by sex steroids, the potency of sex hormones to switch microglia from a pro-inflammatory M1 to neuroprotective M2 phenotype, and the regulation of pro- and anti-inflammatory properties including the inflammasome. We will further discuss the possibility that the neuroprotective action of sex steroids in the brain involves an early and direct modulation of local microglia cell function. This article is part of a Special Issue entitled 'Sex steroids and brain disorders'.
Collapse
Affiliation(s)
- Pardes Habib
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany
| | - Cordian Beyer
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany.
| |
Collapse
|
30
|
Zhang Q, Zhao H, Wang L, Zhang Q, Wang H. Effects of wind-dispelling drugs and deficiency-nourishing drugs of Houshiheisan compound prescription on astrocyte activation and inflammatory factor expression in the corpus striatum of cerebral ischemia rats. Neural Regen Res 2015; 7:1851-7. [PMID: 25624810 PMCID: PMC4298897 DOI: 10.3969/j.issn.1673-5374.2012.24.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Accepted: 06/30/2012] [Indexed: 11/23/2022] Open
Abstract
This study explored protective effects of Houshiheisan and its compound prescription of wind-dispelling drugs and deficiency-nourishing drugs on cerebral ischemia in terms of astrocyte activation and inflammatory factor expression. Results suggested that Houshiheisan lessened neuronal degeneration in the corpus striatum on the ischemic side of rats following cerebral ischemia/reperfusion injury, contributed to astrocyte activation and glial fibrillary acidic protein expression in the corpus striatum and decreased the levels of interleukin-2, interleukin-6, interleukin-1β and tumor necrosis factor-α. Factor analysis results demonstrated that deficiency-nourishing drugs were more beneficial in protecting neurons and upregulating glial fibrillary acidic protein expression than wind-dispelling drugs. However, wind-dispelling drugs were more effective in increasing the number of glial fibrillary acidic protein-positive cells and reducing inflammatory factor expression than deficiency-nourishing drugs. These indicate that different ingredients of Houshiheisan suppress cerebral ischemic injury by promoting astrocyte activation and diminishing inflammatory factor expression.
Collapse
Affiliation(s)
- Qiuxia Zhang
- College of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Hui Zhao
- College of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Lei Wang
- College of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Qi Zhang
- College of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Haizheng Wang
- College of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| |
Collapse
|
31
|
Yan FL, Zhang JH. Role of the sympathetic nervous system and spleen in experimental stroke-induced immunodepression. Med Sci Monit 2014; 20:2489-96. [PMID: 25434807 PMCID: PMC4260620 DOI: 10.12659/msm.890844] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background The mechanism of stroke-induced immunodepression syndrome (SIDS) remains uncertain. Some studies suggest that hyperactivation of the sympathetic nervous system (SNS) may be the key factor underlying SIDS. Catecholamines impair early lymphocyte response, which can increase the risk of stroke-associated infection (SAI). Material/Methods Our study focused on dynamic changes of metanephrine (MN), normetanephrine (NMN), cytokines, and spleen volume in the rat middle cerebral artery occlusion (MCAO) model. Results After MCAO, there is hyperactivation of SNS and pro-/anti-inflammatory imbalance, indicating systemic immunodepression. In addition, rat spleen size was reduced. Correlation analysis indicated that MCAO-induced spleen size reduction correlated with the changes in MN, NMN, and cytokines. Blocking SNS with propranolol can partly reverse the immunodepression and the reduction in spleen volume. Conclusions Taken together, these findings suggest that acute ischemic stroke induces over-activation of the SNS, which lowers the threshold of infection and increases the risk of infection.
Collapse
Affiliation(s)
- Fu-Ling Yan
- Department of Neurology, Zhong-Da Hospital, Southeast University, Nanjing, China (mainland)
| | - Jin-Hua Zhang
- Department of General Medicine, Zhong-Da Hospital, Southeast University, Nanjing, China (mainland)
| |
Collapse
|
32
|
The association of inflammatory markers with cerebral vasoreactivity and carotid atherosclerosis in transient ischaemic attack. Clin Biochem 2014; 47:182-6. [DOI: 10.1016/j.clinbiochem.2014.07.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 07/08/2014] [Accepted: 07/11/2014] [Indexed: 11/22/2022]
|
33
|
Chen S, Yin ZJ, Jiang C, Ma ZQ, Fu Q, Qu R, Ma SP. Asiaticoside attenuates memory impairment induced by transient cerebral ischemia–reperfusion in mice through anti-inflammatory mechanism. Pharmacol Biochem Behav 2014; 122:7-15. [DOI: 10.1016/j.pbb.2014.03.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 03/04/2014] [Accepted: 03/06/2014] [Indexed: 01/08/2023]
|
34
|
Metallothionein-II inhibits lipid peroxidation and improves functional recovery after transient brain ischemia and reperfusion in rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:436429. [PMID: 24719677 PMCID: PMC3956286 DOI: 10.1155/2014/436429] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 01/04/2014] [Accepted: 01/17/2014] [Indexed: 11/18/2022]
Abstract
After transient cerebral ischemia and reperfusion (I/R), damaging mechanisms, such as excitotoxicity and oxidative stress, lead to irreversible neurological deficits. The induction of metallothionein-II (MT-II) protein is an endogenous mechanism after I/R. Our aim was to evaluate the neuroprotective effect of MT-II after I/R in rats. Male Wistar rats were transiently occluded at the middle cerebral artery for 2 h, followed by reperfusion. Rats received either MT (10 μg per rat i.p.) or vehicle after ischemia. Lipid peroxidation (LP) was measured 22 h after reperfusion in frontal cortex and hippocampus; also, neurological deficit was evaluated after ischemia, using the Longa scoring scale. Infarction area was analyzed 72 hours after ischemia. Results showed increased LP in frontal cortex (30.7%) and hippocampus (26.4%), as compared to control group; this effect was fully reversed by MT treatment. Likewise, we also observed a diminished neurological deficit assessed by the Longa scale in those animals treated with MT compared to control group values. The MT-treated group showed a significant (P < 0.05) reduction of 39.9% in the infarction area, only at the level of hippocampus, as compared to control group. Results suggest that MT-II may be a novel neuroprotective treatment to prevent ischemia injury.
Collapse
|
35
|
Mitome-Mishima Y, Yamamoto M, Yatomi K, Nonaka S, Miyamoto N, Urabe T, Arai H, Oishi H. Endothelial cell proliferation in swine experimental aneurysm after coil embolization. PLoS One 2014; 9:e89047. [PMID: 24551215 PMCID: PMC3925181 DOI: 10.1371/journal.pone.0089047] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Accepted: 01/13/2014] [Indexed: 11/18/2022] Open
Abstract
After coil embolization, recanalization in cerebral aneurysms adversely influences long-term prognosis. Proliferation of endothelial cells on the coil surface may reduce the incidence of recanalization and further improve outcomes after coil embolization. We aimed to map the expression of proliferating tissue over the aneurysmal orifice and define the temporal profile of tissue growth in a swine experimental aneurysm model. We compared the outcomes after spontaneous thrombosis with those of coil embolization using histological and morphological techniques. In aneurysms that we not coiled, spontaneous thrombosis was observed, and weak, easily detachable proliferating tissue was evident in the aneurysmal neck. In contrast, in the coil embolization group, histological analysis showed endothelial-like cells lining the aneurysmal opening. Moreover, immunohistochemical and morphological analysis suggested that these cells were immature endothelial cells. Our results indicated the existence of endothelial cell proliferation 1 week after coil embolization and showed immature endothelial cells in septal tissue between the systemic circulation and the aneurysm. These findings suggest that endothelial cells are lead to and proliferate in the former aneurysmal orifice. This is the first examination to evaluate the temporal change of proliferating tissue in a swine experimental aneurysm model.
Collapse
Affiliation(s)
- Yumiko Mitome-Mishima
- Department of Neurosurgery, Juntendo University School of Medicine, Tokyo, Japan
- * E-mail:
| | - Munetaka Yamamoto
- Department of Neurosurgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Kenji Yatomi
- Department of Neurosurgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Senshu Nonaka
- Department of Neurosurgery, Juntendo University Urayasu Hospital, Chiba, Japan
| | - Nobukazu Miyamoto
- Department of Neurology, Juntendo University Urayasu Hospital, Chiba, Japan
| | - Takao Urabe
- Department of Neurology, Juntendo University Urayasu Hospital, Chiba, Japan
| | - Hajime Arai
- Department of Neurosurgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Hidenori Oishi
- Department of Neurosurgery, Juntendo University School of Medicine, Tokyo, Japan
- Department of Neuroendovascular Therapy, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
36
|
Kalin JH, Bergman JA. Development and therapeutic implications of selective histone deacetylase 6 inhibitors. J Med Chem 2013; 56:6297-313. [PMID: 23627282 DOI: 10.1021/jm4001659] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This Perspective provides an in depth look at the numerous disease states in which histone deacetylase 6 (HDAC6) has been implicated. The physiological pathways, protein-protein interactions, and non-histone substrates relating to different pathological conditions are discussed with regard to HDAC6. Furthermore, the compounds and methods used to modulate HDAC6 activity are profiled. The latter half of this Perspective analyzes reported HDAC6 selective inhibitors in terms of structure, potency, and selectivity over the other HDAC isoforms with the intent of providing a comprehensive overview of the molecular tools available. Potential obstacles and future directions of HDAC6 research are also presented.
Collapse
Affiliation(s)
- Jay H Kalin
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, Illinois 60612, United States.
| | | |
Collapse
|
37
|
Hamasaki MY, Hirata MH, Hirata RDC, Himelfarb ST, Campos LMG, Nogueira MI. [Analysis of the mRNA expression of the S100β protein in adipocytes of patients with diabetes mellitus, type 2]. ACTA ACUST UNITED AC 2013; 56:435-40. [PMID: 23108748 DOI: 10.1590/s0004-27302012000700005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Accepted: 08/30/2012] [Indexed: 11/22/2022]
Abstract
OBJECTIVE This study aims to explore the possible relationship between the expression level of S100β protein mRNA with diabetes mellitus type 2 in adipocytes from patients with this disease in comparison with normoglycemic individuals. MATERIALS AND METHODS Samples of adipose tissue of eight patients from the coronary section of the Institute Dante Pazzanese of Cardiology (IDPC), four in Group Diabetes and four of Normoglycemic group, were evaluated by RT-PCR real time. RESULTS An increase around 15 times values, between the threshold cycle (ΔCt), of mRNA expression of S100β protein in adipocytes of the diabetes group was observed in comparison to the control group (p = 0.015). CONCLUSION Our results indicate, for the first time, that there is coexistence of increased expression of the S100β and the type 2 diabetes mellitus gene.
Collapse
Affiliation(s)
- Mike Yoshio Hamasaki
- Laboratório de Neurociências, Departamento de Anatomia, Instituto de Ciências Biomédicas III, Universidade de São Paulo, São Paulo, SP, Brasil
| | | | | | | | | | | |
Collapse
|
38
|
Teng P, Li Y, Cheng W, Zhou L, Shen Y, Wang Y. Neuroprotective effects of Lycium barbarum polysaccharides in lipopolysaccharide-induced BV2 microglial cells. Mol Med Rep 2013; 7:1977-81. [PMID: 23620217 DOI: 10.3892/mmr.2013.1442] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Accepted: 04/22/2013] [Indexed: 11/06/2022] Open
Abstract
Polysaccharides extracted from Lycium barbarum (LBPs) possess a wide variety of biological activities. However, their neuroprotective effects have not yet been fully elucidated. The aim of the present study was to investigate the inhibitory effects of LBPs on the production of lipopolysaccharide (LPS)‑induced proinflammatory mediators in BV2 microglia. BV2 mouse microglial cells were cultured and an MTT assay was performed to determine whether LBPs had an effect on the apoptosis of LPS-stimulated BV2 cells. Our data showed that LPS induced the activation of nuclear factor‑κB (NF‑κB) and its upstream protein caspase 3, upregulated the expression of an additional apoptosis‑inducing factor, heat shock protein 60 (HSP60), in BV2 microglial cells and increased the release of TNF-α and HSP60 in the culture media. Following treatment with LBPs, the activated NF‑κB and caspase 3 were significantly suppressed. Furthermore, the enhanced expression of HSP60 was reduced and the LPS-induced release of TNF-α and HSP60 were inhibited. These results suggest that LBPs may have therapeutic potential for the treatment of neurodegenerative diseases that are accompanied by microglial activation.
Collapse
Affiliation(s)
- Peng Teng
- Department of Neurobiology, Basic Medical College, Center of Scientific Technology, Cranial Cerebral Disease Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | | | | | | | | | | |
Collapse
|
39
|
Li Y, Nie H, Wu D, Zhang J, Wei X, Ying W. Poly(ADP-ribose) polymerase mediates both cell death and ATP decreases in SIRT2 inhibitor AGK2-treated microglial BV2 cells. Neurosci Lett 2013; 544:36-40. [PMID: 23570735 DOI: 10.1016/j.neulet.2013.03.032] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2012] [Revised: 03/11/2013] [Accepted: 03/18/2013] [Indexed: 10/27/2022]
Abstract
Sirtuin 2 (SIRT2), a sirtuin family protein, is a tubulin deacetylase. Recent studies have indicated that SIRT2 plays a key role in programmed necrosis, and the SIRT2 inhibitor AGK2 can decrease the cell death both in a cellular model of Parkinson's disease and in an animal model of myocardial ischemia-reperfusion. However, there has been little information regarding the role of SIRT2 in microglial survival and functions, which play critical roles in multiple neurological disorders. Our current study found that AGK2 at 10 μM - a widely used AGK2 concentration - can induce both late-stage apoptosis and necrosis, as well as a decrease in the intracellular ATP levels of microglial BV2 cells. Our study also showed that both the AGK2-induced cell death and the AGK2-induced ATP decline are mediated by poly(ADP-ribose) polymerase (PARP) activation. Collectively, our study has provided the first evidence suggesting a significant role of SIRT2 in the basal survival of microglia, as well as a mechanism accounting for the effects of SIRT2 on intracellular ATP levels.
Collapse
Affiliation(s)
- Yexin Li
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | | | | | | | | | | |
Collapse
|
40
|
Chen YT, Zang XF, Pan J, Zhu XL, Chen F, Chen ZB, Xu Y. Expression patterns of histone deacetylases in experimental stroke and potential targets for neuroprotection. Clin Exp Pharmacol Physiol 2013; 39:751-8. [PMID: 22651689 DOI: 10.1111/j.1440-1681.2012.05729.x] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
1. Histone deacetylase (HDAC) inhibitors exert neuroprotection in both cellular and animal models of ischaemic stroke. However, which HDAC isoform (or isoforms) mediates this beneficial effect has not yet been determined. 2. In the present study, gene levels of the HDAC isoforms were determined in the mouse cortex using reverse transcription-polymerase chain reaction (RT-PCR), whereas changes in the expression of individual zinc-dependent HDAC family members were evaluated by western blotting, 3, 12, 24 and 48 h after cerebral ischaemia induced by transient middle cerebral artery occlusion in male Kunming mice. 3. The HDAC isoforms HDAC1-11 were all expressed in the mouse cortex and differentially affected by cerebral ischaemia. Notably, there was a substantial increase in HDAC3, HDAC6 and HDAC11 expression during the early phases of experimental stroke, indicating their contribution to stroke pathogenesis. Furthermore, induction of HDAC3 and HDAC6 in cortical neurons by ischaemic stroke was confirmed in vivo and in vitro using double-labelled immunostaining and RT-PCR, respectively. Therefore, small hairpin (sh) RNAs were used to selectively knock down HDAC3 or HDAC6. This knockdown appreciably promoted the survival of cortical neurons subjected to oxygen and glucose deprivation. 4. The findings of the present study demonstrate the expression patterns of HDAC isoforms during experimental ischaemic stroke. Furthermore, HDAC3 and HDAC6 were identified as potential mediators in the neurotoxicity of ischaemic stroke, suggesting that specific therapeutic approaches may be considered according to HDAC subtype.
Collapse
Affiliation(s)
- Yan-Ting Chen
- Department of Neurology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | | | | | | | | | | | | |
Collapse
|
41
|
Jiang Y, Zhu J, Wu L, Xu G, Dai J, Liu X. Tetracycline inhibits local inflammation induced by cerebral ischemia via modulating autophagy. PLoS One 2012; 7:e48672. [PMID: 23144925 PMCID: PMC3492486 DOI: 10.1371/journal.pone.0048672] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 09/28/2012] [Indexed: 11/24/2022] Open
Abstract
Background Tetracycline exerts neuroprotection via suppressing the local inflammation induced by cerebral ischemia. However, the underlying mechanism is not completely clear. Methodology/Principal Findings The mRNA and protein expressions of tumor necrosis factor α and interleukin 6 and the number of activated microglia were measured to detect the inflammatory process in the ischemic hemisphere. The key proteins of nuclear factor kappa B pathway and the binding activity of nuclear factor kappa B were also measured. Two key components of autophagy, Beclin 1 and LC3, were detected by western blotting. Pretreatment with tetracycline inhibited the mRNA and protein expressions of tumor necrosis factor α and interleukin 6 and decreased the numbers of activated and phagocytotic microglia. Tetracycline down regulated the total and phosphorylated expressions of IKK, IκB and p65 (P<0.05). The autophagy inhibitor, 3-methyladenine, inhibited inflammation and activation of nuclear factor kappa B pathway. The levels of Beclin 1 and LC3 were decreased by 3-methyladenine and tetracycline. Conclusions/Significance Our data suggested that pretreatment of tetracycline may inhibit autophagy in the ischemic stroke brain and then suppress the inflammatory process via inhibiting the activation of nuclear factor kappa B pathway.
Collapse
Affiliation(s)
- Yongjun Jiang
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Jiangsu Province, China
| | - Juehua Zhu
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Jiangsu Province, China
| | - Li Wu
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Jiangsu Province, China
| | - Gelin Xu
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Jiangsu Province, China
| | - Jianwu Dai
- Key laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, PR China
- * E-mail: (XL); (JD)
| | - Xinfeng Liu
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Jiangsu Province, China
- * E-mail: (XL); (JD)
| |
Collapse
|
42
|
Aggravated inflammation and increased expression of cysteinyl leukotriene receptors in the brain after focal cerebral ischemia in AQP4-deficient mice. Neurosci Bull 2012; 28:680-92. [PMID: 23132680 DOI: 10.1007/s12264-012-1281-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2012] [Accepted: 04/06/2012] [Indexed: 10/27/2022] Open
Abstract
OBJECTIVE Aquaporin-4 (AQP4), the main water channel protein in the brain, plays a critical role in water homeostasis and brain edema. Here, we investigated its role in the inflammatory responses after focal cerebral ischemia. METHODS In AQP4-knockout (KO) and wild-type mice, focal cerebral ischemia was induced by 30 min of middle cerebral arterial occlusion (MCAO). Ischemic neuronal injury and cellular inflammatory responses, as well as the expression and localization of cysteinyl leukotriene CysLT(2) and CysLT(1) receptors, were determined at 24 and 72 h after MCAO. RESULTS AQP4-KO mice showed more neuronal loss, more severe microglial activation and neutrophil infiltration, but less astrocyte proliferation in the brain after MCAO than wild-type mice. In addition, the protein levels of both CysLT(1) and CysLT(2) receptors were up-regulated in the ischemic brain, and the up-regulation was more pronounced in AQP4-KO mice. The CysLT(1) and CysLT(2) receptors were primarily localized in neurons, microglia and neutrophils; those localized in microglia and neutrophils were enhanced in AQP4-KO mice. CONCLUSION AQP4 may play an inhibitory role in postischemic inflammation.
Collapse
|
43
|
Zhang Y, Wei X, Liu L, Liu S, Wang Z, Zhang B, Fan B, Yang F, Huang S, Jiang F, Chen YH, Yi F. TIPE2, a novel regulator of immunity, protects against experimental stroke. J Biol Chem 2012; 287:32546-55. [PMID: 22859306 DOI: 10.1074/jbc.m112.348755] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The inflammatory responses accompanying stroke are recognized to contribute to secondary ischemic injury. TIPE2 is a very recently identified negative regulator of inflammation that maintains immune homeostasis. However, it is unknown whether TIPE2 is expressed in the brain and contributes to the regulation of cerebral diseases. In this study, we explored the potential roles of TIPE2 in cerebral ischemia/reperfusion injury. TIPE2(-/-) mice were used to assess whether TIPE2 provides neuroprotection following cerebral ischemia/reperfusion induced by middle cerebral artery occlusion (MCAO), and in vitro primary cerebral cell cultures were used to investigate the expression and regulation of TIPE2. Our results show that genetic ablation of the Tipe2 gene significantly increased the cerebral volume of infarction and neurological dysfunction in mice subjected to MCAO. Flow cytometric analysis revealed more infiltrating macrophages, neutrophils, and lymphocytes in the ischemic hemisphere of TIPE2(-/-) mice. The responses to inflammatory cytokines and chemokines were significantly increased in TIPE2(-/-) mouse brain after MCAO. We further observed that TIPE2 was highly induced in WT mice after cerebral ischemia and was expressed mainly in microglia/macrophages, but not in neurons and astrocytes. Finally, we found that regulation of TIPE2 expression was associated with NADPH oxidase activity. These findings demonstrate, for the first time, that TIPE2 is involved in the pathogenesis of stroke and suggest that TIPE2 plays an essential role in a signal transduction pathway that links the inflammatory immune response to specific conditions after cerebral ischemia. Targeting TIPE2 may be a new therapeutic strategy for stroke treatment.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Pharmacology, Shandong University School of Medicine, Jinan 250012, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Malaterre J, McPherson CS, Denoyer D, Lai E, Hagekyriakou J, Lightowler S, Shudo K, Ernst M, Ashley DM, Short JL, Wheeler G, Ramsay RG. Enhanced lithium-induced brain recovery following cranial irradiation is not impeded by inflammation. Stem Cells Transl Med 2012. [PMID: 23197851 DOI: 10.5966/sctm.2011-0046] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Radiation-induced brain injury occurs in many patients receiving cranial radiation therapy, and these deleterious effects are most profound in younger patients. Impaired neurocognitive functions in both humans and rodents are associated with inflammation, demyelination, and neural stem cell dysfunction. Here we evaluated the utility of lithium and a synthetic retinoid receptor agonist in reducing damage in a model of brain-focused irradiation in juvenile mice. We found that lithium stimulated brain progenitor cell proliferation and differentiation following cranial irradiation while also preventing oligodendrocyte loss in the dentate gyrus of juvenile mice. In response to inflammation induced by radiation, which may have encumbered the optimal reparative action of lithium, we used the anti-inflammatory synthetic retinoid Am80 that is in clinical use in the treatment of acute promyelocytic leukemia. Although Am80 reduced the number of cyclooxygenase-2-positive microglial cells following radiation treatment, it did not enhance lithium-induced neurogenesis recovery, and this alone was not significantly different from the effect of lithium on this proinflammatory response. Similarly, lithium was superior to Am80 in supporting the restoration of new doublecortin-positive neurons following irradiation. These data suggest that lithium is superior in its restorative effects to blocking inflammation alone, at least in the case of Am80. Because lithium has been in routine clinical practice for 60 years, these preclinical studies indicate that this drug might be beneficial in reducing post-therapy late effects in patients receiving cranial radiotherapy and that blocking inflammation in this context may not be as advantageous as previously suggested.
Collapse
Affiliation(s)
- Jordane Malaterre
- Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Hashioka S, Klegeris A, McGeer PL. The histone deacetylase inhibitor suberoylanilide hydroxamic acid attenuates human astrocyte neurotoxicity induced by interferon-γ. J Neuroinflammation 2012; 9:113. [PMID: 22647614 PMCID: PMC3410763 DOI: 10.1186/1742-2094-9-113] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Accepted: 05/30/2012] [Indexed: 02/07/2023] Open
Abstract
Backgrounds Increasing evidence shows that the histone deacetylase inhibitor suberoylanilide hydroxamic acid (SAHA) possesses potent anti-inflammatory and immunomodulatory properties. It is tempting to evaluate the potential of SAHA as a therapeutic agent in various neuroinflammatory and neurodegenerative disorders. Methods We examined the effects of SAHA on interferon (IFN)-γ-induced neurotoxicity of human astrocytes and on IFN-γ-induced phosphorylation of signal transducer and activator of transcription (STAT) 3 in human astrocytes. We also studied the effects of SAHA on the astrocytic production of two representative IFN-γ-inducible inflammatory molecules, namely IFN-γ-inducible T cell α chemoattractant (I-TAC) and intercellular adhesion molecule-1 (ICAM-1). Results SAHA significantly attenuated the toxicity of astrocytes activated by IFN-γ towards SH-SY5Y human neuronal cells. In the IFN-γ-activated astrocytes, SAHA reduced the STAT3 phosphorylation. SAHA also inhibited the IFN-γ-induced astrocytic production of I-TAC, but not ICAM-1. These results indicate that SAHA suppresses IFN-γ-induced neurotoxicity of human astrocytes through inhibition of the STAT3 signaling pathway. Conclusion Due to its anti-neurotoxic and anti-inflammatory properties, SAHA appears to have the therapeutic or preventive potential for a wide range of neuroinflammatory disorders associated with activated astrocytes.
Collapse
Affiliation(s)
- Sadayuki Hashioka
- Kinsmen Laboratory of Neurological Research, Department of Psychiatry, the University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
| | | | | |
Collapse
|
46
|
Ma Y, Jiang J, Wang L, Nie H, Xia W, Liu J, Ying W. CD38 is a key enzyme for the survival of mouse microglial BV2 cells. Biochem Biophys Res Commun 2012; 418:714-9. [PMID: 22293203 DOI: 10.1016/j.bbrc.2012.01.084] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 01/14/2012] [Indexed: 12/20/2022]
Abstract
CD38 is a multifunctional enzyme that can not only generate cyclic adenosine diphosphate-ribose (cADPR) - a key Ca(2+) -mobilizing second messenger - by consuming NAD(+), but also hydrolyze extracellular NAD(+). There have been only a small number of studies on the functions of CD38 in the CNS. Brain inflammation plays critical roles in ischemic brain injury and multiple other neurological diseases, in which microglia activation is a key event. In this study we determined the roles of CD38 in the basal survival of mouse BV2 microglia cells by applying CD38 siRNA. Our study found that silencing of CD38 led to significantly decreased survival of the cells. We also found that decreased CD38 levels can lead to apoptosis of the microglial cells, as assessed by flow cytometry-based Annexin V/7-AAD assay, caspase-3 immunostaining and Hoechst staining assays. Our study has further indicated that the CD38 silencing-induced apoptosis is mainly caspase 3-dependent. Collectively, our study has provided the first evidence suggesting that CD38 plays a critical role in the basal survival of microglia, and decreased CD38 can lead to caspase 3-dependent apoptosis of the cells. These results suggest that CD38 may become a therapeutic target for modulating microglial survival in neurological diseases.
Collapse
Affiliation(s)
- Yingxin Ma
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | | | | | | | | | | | | |
Collapse
|
47
|
Wang S, Wang L, Zhang X, Wang C, Zheng G, Wu W, Zhang X, Dong Z, Du L. Effects of subcutaneous low molecular weight heparin and intravenous unfractionated heparin on serum S100 concentrations in patients with cerebrovascular diseases. Clin Chem Lab Med 2012; 50:525-8. [DOI: 10.1515/cclm.2011.794] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Accepted: 10/26/2011] [Indexed: 11/15/2022]
|
48
|
Simonyi A, Serfozo P, Lehmidi TM, Cui J, Gu Z, Lubahn DB, Sun AY, Sun GY. The neuroprotective effects of apocynin. Front Biosci (Elite Ed) 2012; 4:2183-93. [PMID: 22202030 DOI: 10.2741/535] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The recognition of health benefits of phytomedicines and herbal supplements lead to an increased interest to understand the cellular and molecular basis of their biological activities. Apocynin (4-hydroxy-3-methoxy-acetophenone) is a constituent of the Himalayan medicinal herb Picrorhiza kurroa which is regarded as an inhibitor of nicotinamide adenine dinucleotide phosphate (NADPH)-oxidase, a superoxide-producing enzyme. NADPH oxidase appears to be especially important in the modulation of redox-sensitive signaling pathways and also has been implicated in neuronal dysfunction and degeneration, and neuroinflammmation in diseases ranging from stroke, Alzheimer's and Parkinson's diseases to psychiatric disorders. In this review, we aim to give an overview of current literature on the neuroprotective effects of apocynin in the prevention and treatment of neurodegenerative disorders. Particular attention is given to in vivo studies.
Collapse
Affiliation(s)
- Agnes Simonyi
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Zhu J, Jiang Y, Wu L, Lu T, Xu G, Liu X. Suppression of local inflammation contributes to the neuroprotective effect of ginsenoside Rb1 in rats with cerebral ischemia. Neuroscience 2011; 202:342-51. [PMID: 22173011 DOI: 10.1016/j.neuroscience.2011.11.070] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Revised: 11/08/2011] [Accepted: 11/30/2011] [Indexed: 11/15/2022]
Abstract
Local inflammation accounts for the progression of cerebral ischemic insult. Ginsenoside Rb1 (GRb1) is a natural product extracted from Panax ginseng C.A. Meyer. It has been reported to have beneficial effects in cerebral ischemia and to inhibit the inflammatory cascade in sepsis. In this study, to determine whether modulating local inflammation contributed to the neuroprotection of GRb1, male Sprague-Dawley rats were treated with GRb1 or vehicle intranasally for 1 week before being subjected to temporary occlusion of the right middle cerebral artery and reperfusion. Neuroprotection of GRb1 was evaluated with a focus on the key elements of central nervous system (CNS) inflammation, such as inflammatory cells, proinflammatory cytokines, and transcriptional factor. GRb1 reduced infarction volume by 57% (n=6, P<0.01) and significantly alleviated the neurological deficit (n=12, modified neurological severity scores [mNSS]: 6.6±1.1 vs. 8.6±1.1, P<0.05). GRb1 depressed the activation of microglia in the penumbra by 15%-27% from 24 h to 72 h after reperfusion and its further convention into phagocytic microglia/macrophages. In GRb1 group, the peak mRNA level of tumor necrosis factor α (TNF-α) mRNA was decreased by 35% 12 h after reperfusion, whereas the protein level was significantly reduced by 43%-57%. Downregulation by GRb1 of both interleukin (IL)-6 gene and protein after GRb1 administration was also observed. GRb1 partially inhibited the activation of nuclear factor-κB (NF-κB) pathway from 6 h to 72 h after ischemia and reperfusion onset, as determined by the expression of total and phosphorylated NF-κB/p65, inhibitor protein of κB (IκB)-α, and IκB-kinase complex (IKK)-α. All these results indicate that suppression of local inflammation after cerebral ischemia might be one mechanism that contributes to the neuroprotection of GRb1.
Collapse
Affiliation(s)
- J Zhu
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, 305 East Zhongshan Road, Nanjing 210002, Jiangsu Province, China
| | | | | | | | | | | |
Collapse
|
50
|
Yan EB, Hellewell SC, Bellander BM, Agyapomaa DA, Morganti-Kossmann MC. Post-traumatic hypoxia exacerbates neurological deficit, neuroinflammation and cerebral metabolism in rats with diffuse traumatic brain injury. J Neuroinflammation 2011; 8:147. [PMID: 22034986 PMCID: PMC3215944 DOI: 10.1186/1742-2094-8-147] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Accepted: 10/28/2011] [Indexed: 11/29/2022] Open
Abstract
Background The combination of diffuse brain injury with a hypoxic insult is associated with poor outcomes in patients with traumatic brain injury. In this study, we investigated the impact of post-traumatic hypoxia in amplifying secondary brain damage using a rat model of diffuse traumatic axonal injury (TAI). Rats were examined for behavioral and sensorimotor deficits, increased brain production of inflammatory cytokines, formation of cerebral edema, changes in brain metabolism and enlargement of the lateral ventricles. Methods Adult male Sprague-Dawley rats were subjected to diffuse TAI using the Marmarou impact-acceleration model. Subsequently, rats underwent a 30-minute period of hypoxic (12% O2/88% N2) or normoxic (22% O2/78% N2) ventilation. Hypoxia-only and sham surgery groups (without TAI) received 30 minutes of hypoxic or normoxic ventilation, respectively. The parameters examined included: 1) behavioural and sensorimotor deficit using the Rotarod, beam walk and adhesive tape removal tests, and voluntary open field exploration behavior; 2) formation of cerebral edema by the wet-dry tissue weight ratio method; 3) enlargement of the lateral ventricles; 4) production of inflammatory cytokines; and 5) real-time brain metabolite changes as assessed by microdialysis technique. Results TAI rats showed significant deficits in sensorimotor function, and developed substantial edema and ventricular enlargement when compared to shams. The additional hypoxic insult significantly exacerbated behavioural deficits and the cortical production of the pro-inflammatory cytokines IL-6, IL-1β and TNF but did not further enhance edema. TAI and particularly TAI+Hx rats experienced a substantial metabolic depression with respect to glucose, lactate, and glutamate levels. Conclusion Altogether, aggravated behavioural deficits observed in rats with diffuse TAI combined with hypoxia may be induced by enhanced neuroinflammation, and a prolonged period of metabolic dysfunction.
Collapse
Affiliation(s)
- Edwin B Yan
- National Trauma Research Institute, The Alfred Hospital, 89 Commercial Road, Melbourne 3004, Australia
| | | | | | | | | |
Collapse
|