201
|
Zhang A, Song Y, Zhang Z, Jiang S, Chang S, Cai Z, Liu F, Zhang X, Ni G. Effects of autophagy inhibitor 3-Methyladenine on ischemic stroke: A protocol for systematic review and meta-analysis. Medicine (Baltimore) 2021; 100:e23873. [PMID: 33530181 PMCID: PMC7850754 DOI: 10.1097/md.0000000000023873] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 11/24/2020] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Ischemic stroke is a huge threat to human health globally. Rescuing neurons in the ischemic penumbra (IP) is pivotal after the onset of ischemic stroke, and autophagy is essential to the survival of IP neurons and the development of related pathological processes. As the most common autophagy inhibitor, 3-Methyladenine (3-MA) is widely used in studies related to the mechanism of neuronal autophagy in ischemic stroke; however, there is no consensus has been reached on its effects of neuroprotection or neurodamage, which hinders the development and clinical application of autophagy-targeted therapy strategies for the treatment of ischemic stroke. METHODS We will search the following electronic bibliographic databases: PubMed, EMBASE, Scopus, Science Direct, and Web of Science. Participant intervention comparator outcomes of this study are as flowing: P, animal models of ischemic stroke; I, received 3-MA treatment merely; C, received only vehicle or sham treatment, or no treatment; O, Primary outcomes are infarct volume; neuro-behavioral scores. Secondary outcomes are cerebral blood flow, blood-brain barrier permeability, cerebral hemorrhage, brain water content. Review Manager 5.3 and Stata 15.1 will be used in data analysis. The characteristics of the studies, the experimental model, and the main results will be described, the quality assessment and the risk of bias assessment will be conducted. A narrative synthesis will be made for the included studies. Besides, if sufficient qualitative data is available, a meta-analysis will be conducted. I2 statistics will be used to assess heterogeneity. DISCUSSION This systematic review and meta-analysis of the autophagy inhibitor 3-MAs effects on animal models of ischemic stroke can help us to understand whether inhibiting autophagy brings protection or damage to IP neurons; in addition, it also helps to clarify the specific role of autophagy in cerebral infarction. Therefore, this study can provide evidence for the future development of therapy strategies targeting autophagy and bring more hope to patients with ischemic stroke. PROSPERO REGISTRATION NUMBER CRD42020194262.
Collapse
|
202
|
β-1, 3-galactosyltransferase 2 deficiency exacerbates brain injury after transient focal cerebral ischemia in mice. Brain Res Bull 2021; 169:104-111. [PMID: 33482286 DOI: 10.1016/j.brainresbull.2021.01.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/08/2021] [Accepted: 01/12/2021] [Indexed: 11/22/2022]
Abstract
Glycosyltransferases are enzymes that catalyze the formation of a variety of glycoconjugates. Glycoconjugates play vital roles in the nervous system. β-1, 3-Galactosyltransferase 2 (B3galt2) is one of the major types of glycosyltransferases, which has not been reported in ischemia induced-brain injury. The purpose of this study was to explore the role of B3galt2 exerts and its underlying mechanism in cerebral ischemia in mice. Wild-type (WT) and heterozygous B3galt2 knockout (B3galt2-/+) mice were subjected to 90 min transient focal cerebral ischemia by middle cerebral artery occlusion (MCAO). The brain samples were analyzed at 24 h after reperfusion. The B3galt2 level in the peri-infarct penumbra was quantified. The cerebral infarct volume, neurological deficits, apoptosis and the levels of Reelin and Dab1 were assessed. Compared with control mice, B3galt2-/+ mice not only showed severe brain damage, neurologic functional deficits, but also showed severe neuronal apoptosis in the cortical penumbra after ischemia/reperfusion (I/R). The Caspase-3 activity was increased and the levels of Reelin and Dab1 were decreased in B3galt2-/+ mice. Recombinant human Reelin (rh-Reelin) administered intracerebroventricularly before MCAO significantly reduced infarct volume, and prevented neuronal loss in B3galt2-/+ mice after I/R. Our results suggest B3galt2 deficiency exacerbates ischemic brain damage in acute ischemic stroke in mice, and this was reversed by giving rh-Reelin. B3galt2 might play a beneficial role for neurons survival in the penumbra through modulation of Reelin pathway.
Collapse
|
203
|
Li Y, Tang Y, Yang GY. Therapeutic application of exosomes in ischaemic stroke. Stroke Vasc Neurol 2021; 6:483-495. [PMID: 33431513 PMCID: PMC8485240 DOI: 10.1136/svn-2020-000419] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/28/2020] [Accepted: 09/18/2020] [Indexed: 02/07/2023] Open
Abstract
Ischaemic stroke is a leading cause of long-term disability in the world, with limited effective treatments. Increasing evidence demonstrates that exosomes are involved in ischaemic pathology and exhibit restorative therapeutic effects by mediating cell–cell communication. The potential of exosome therapy for ischaemic stroke has been actively investigated in the past decade. In this review, we mainly discuss the current knowledge of therapeutic applications of exosomes from different cell types, different exosomal administration routes, and current advances of exosome tracking and targeting in ischaemic stroke. We also briefly summarised the pathology of ischaemic stroke, exosome biogenesis, exosome profile changes after stroke as well as registered clinical trials of exosome-based therapy.
Collapse
Affiliation(s)
- Yongfang Li
- Department of Neurology, Ruijin Hospital, School of medcine, Shanghai Jiao Tong University, Shanghai, China
| | - Yaohui Tang
- Neuroscience and Neuroengineering Center, Medx Research Institute, Shanghai Jiao Tong University School of Biomedical Engineering, Shanghai, China
| | - Guo-Yuan Yang
- Department of Neurology, Ruijin Hospital, School of medcine, Shanghai Jiao Tong University, Shanghai, China .,Neuroscience and Neuroengineering Center, Medx Research Institute, Shanghai Jiao Tong University School of Biomedical Engineering, Shanghai, China
| |
Collapse
|
204
|
Xiang W, Long Z, Zeng J, Zhu X, Yuan M, Wu J, Wu Y, Liu L. Mechanism of Radix Rhei Et Rhizome Intervention in Cerebral Infarction: A Research Based on Chemoinformatics and Systematic Pharmacology. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:6789835. [PMID: 34531920 PMCID: PMC8440083 DOI: 10.1155/2021/6789835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 08/13/2021] [Indexed: 02/05/2023]
Abstract
OBJECTIVE To explore the therapeutic targets, network modules, and coexpressed genes of Radix Rhei Et Rhizome intervention in cerebral infarction (CI), and to predict significant biological processes and pathways through network pharmacology. To explore the differential proteins of Radix Rhei Et Rhizome intervention in CI, conduct bioinformatics verification, and initially explain the possible therapeutic mechanism of Radix Rhei Et Rhizome intervention in CI through proteomics. METHODS The TCM database was used to predict the potential compounds of Radix Rhei Et Rhizome, and the PharmMapper was used to predict its potential targets. GeneCards and OMIM were used to search for CI-related genes. Cytoscape was used to construct a protein-protein interaction (PPI) network and to screen out core genes and detection network modules. Then, DAVID and Metascape were used for enrichment analysis. After that, in-depth analysis of the proteomics data was carried out to further explore the mechanism of Radix Rhei Et Rhizome intervention in CI. RESULTS (1) A total of 14 Radix Rhei Et Rhizome potential components and 425 potential targets were obtained. The core components include sennoside A, palmidin A, emodin, toralactone, and so on. The potential targets were combined with 297 CI genes to construct a PPI network. The targets shared by Radix Rhei Et Rhizome and CI include ALB, AKT1, MMP9, IGF1, CASP3, etc. The biological processes that Radix Rhei Et Rhizome may treat CI include platelet degranulation, cell migration, fibrinolysis, platelet activation, hypoxia, angiogenesis, endothelial cell apoptosis, coagulation, and neuronal apoptosis. The signaling pathways include Ras, PI3K-Akt, TNF, FoxO, HIF-1, and Rap1 signaling pathways. (2) Proteomics shows that the top 20 proteins in the differential protein PPI network were Syp, Syn1, Mbp, Gap43, Aif1, Camk2a, Syt1, Calm1, Calb1, Nsf, Nefl, Hspa5, Nefh, Ncam1, Dcx, Unc13a, Mapk1, Syt2, Dnm1, and Cltc. Differential protein enrichment results show that these proteins may be related to synaptic vesicle cycle, vesicle-mediated transport in synapse, presynaptic endocytosis, synaptic vesicle endocytosis, axon guidance, calcium signaling pathway, and so on. CONCLUSION This study combined network pharmacology and proteomics to explore the main material basis of Radix Rhei Et Rhizome for the treatment of CI such as sennoside A, palmidin A, emodin, and toralactone. The mechanism may be related to the regulation of biological processes (such as synaptic vesicle cycle, vesicle-mediated transport in synapse, presynaptic endocytosis, and synaptic vesicle endocytosis) and signaling pathways (such as Ras, PI3K-Akt, TNF, FoxO, HIF-1, Rap1, and axon guidance).
Collapse
Affiliation(s)
- Wang Xiang
- The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi Province, China
| | - Zhiyong Long
- Shantou University Medical College, Shantou University, Shantou, Guangdong, China
| | - Jinsong Zeng
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Xiaofei Zhu
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Mengxia Yuan
- Shantou University Medical College, Shantou University, Shantou, Guangdong, China
| | - Jiamin Wu
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yonghe Wu
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Liang Liu
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| |
Collapse
|
205
|
Lu J, Luo Y, Mei S, Fang Y, Zhang J, Chen S. The Effect of Melatonin Modulation of Non-coding RNAs on Central Nervous System Disorders: An Updated Review. Curr Neuropharmacol 2020; 19:3-23. [PMID: 32359338 PMCID: PMC7903498 DOI: 10.2174/1570159x18666200503024700] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 04/06/2020] [Accepted: 04/25/2020] [Indexed: 01/19/2023] Open
Abstract
Melatonin is a hormone produced in and secreted by the pineal gland. Besides its role in regulating circadian rhythms, melatonin has a wide range of protective functions in the central nervous system (CNS) disorders. The mechanisms underlying this protective function are associated with the regulatory effects of melatonin on related genes and proteins. In addition to messenger ribonucleic acid (RNA) that can be translated into protein, an increasing number of non-coding RNAs in the human body are proven to participate in many diseases. This review discusses the current progress of research on the effects of melatonin modulation of non-coding RNAs (ncRNAs), including microRNA, long ncRNA, and circular RNA. The role of melatonin in regulating common pathological mechanisms through these ncRNAs is also summarized. Furthermore, the ncRNAs, currently shown to be involved in melatonin signaling in CNS diseases, are discussed. The information compiled in this review will open new avenues for future research into melatonin mechanisms and provide a further understanding of ncRNAs in the CNS.
Collapse
Affiliation(s)
- Jianan Lu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Yujie Luo
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Shuhao Mei
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Yuanjian Fang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Sheng Chen
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| |
Collapse
|
206
|
Abdelwahab SA, Elsebay SAG, Ibrahim MFG, Abdel Hafez SMN. Cerebral and cerebellar histological changes in the rat animal model of rotenone induced parkinsonism can be ameliorated by bone marrow derived stem cell conditioned media. J Chem Neuroanat 2020; 111:101892. [PMID: 33220428 DOI: 10.1016/j.jchemneu.2020.101892] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 11/29/2022]
Abstract
Parkinson disease is the second most common neurodegenerative disease affecting elderly patients. It occurs due to the progressive degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc). We continue our work in this model focusing on other brain areas affected with this disorder; cerebral cortex and cerebellum (areas other than substantia nigra) for better understanding the motor and behavior effect of the Parkinson disease as a forward steep for its treatment and medical control. This work aims to evaluate the therapeutic effect of stem cell-conditioned medium in the Parkinsonism model. In this study, Parkinsonism model was induced in rats by daily subcutaneous injection of 0.5 mg/Kg of rotenone for 28 days. Thirty rats were divided randomly into 3 groups; control, Parkinson, and conditioned medium (CM) treated groups. Cerebral Cortex and Cerebellum were obtained for histological, immunohistochemical and biochemical studies. In the Parkinsonism group, marked histological changes were observed. These findings were nearly ameliorated in CM treated group as confirmed by the biochemical, histological, and immunohistochemical (anti-alpha synculein, anti GFAP and anti nestin) studies. It could be concluded that CM had a good therapeutic effect on Parkinsonism induced damage in both the cerebral cortex and cerebellum.
Collapse
|
207
|
LncRNA FOXD3-AS1 knockdown protects against cerebral ischemia/reperfusion injury via miR-765/BCL2L13 axis. Biomed Pharmacother 2020; 132:110778. [PMID: 33068927 DOI: 10.1016/j.biopha.2020.110778] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/11/2020] [Accepted: 09/17/2020] [Indexed: 12/15/2022] Open
Abstract
AIMS Long non-coding RNAs (lncRNAs) FOXD3-AS1 was reported to be increased in cardiomyocyte ischemic injury. However, its role and underlying molecular mechanism in ischemic stroke remain unknown. This study was to investigate the role of FOXD3-AS1 in cerebral ischemia/reperfusion injury. METHODS The expression of FOXD3-AS1 and miR-765 were measured with qRT-PCR. The shared putative miR-765 binding sites both in BCL2L13 and FOXD3-AS1 were identified with bioinformatics, luciferase reporter assay and RNA immunoprecipitation. Apoptosis and its related proteins were detected by TUNEL assay, Hoechst 33,258 staining, flow cytometry and western blot. Infarct volume and the neurological status were evaluated with TTC staining and neurologic deficit score, respectively. RESULTS The up-regulation of FOXD3-AS1 and down-regulation of miR-765 were found in both mouse brains after cerebral ischemia/reperfusion (I/R) and neuroblastoma cells of neuro-2A (N2a) after oxygen-glucose deprivation/reoxygenation (OGD/R). Moreover, the overexpression of miR-765 reduced N2a cell apoptosis caused by OGD/R. MiR-765 could target BCL2L13 directly. In addition, we found that FOXD3-AS1 bound to miR-765 directly, acting as a ceRNA to modulate the expression of BCL2L13. Overexpression of FOXD3-AS1 antagonized the inhibitory impact of miR-765 on the expression of BCL2L13 and the apoptosis of N2a cells treated with OGD/R, while FOXD3-AS1 knockdown promoted the inhibitory impact of miR-765 on the expression of BCL2L13 and the apoptosis of N2a cells treated with OGD/R. Furthermore, we found that neurological deficits and brain injury induced by I/R in vivo were attenuated by FOXD3-AS1 knockdown. CONCLUSIONS We verified a critical signaling pathway of FOXD3-AS1/miR-765/BCL2L13 in regulating cerebral ischemia/reperfusion injury.
Collapse
|
208
|
Yan W, Sun W, Fan J, Wang H, Han S, Li J, Yin Y. Sirt1-ROS-TRAF6 Signaling-Induced Pyroptosis Contributes to Early Injury in Ischemic Mice. Neurosci Bull 2020; 36:845-859. [PMID: 32253651 PMCID: PMC7410906 DOI: 10.1007/s12264-020-00489-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 10/04/2019] [Indexed: 01/01/2023] Open
Abstract
Stroke is an acute cerebro-vascular disease with high incidence and poor prognosis, most commonly ischemic in nature. In recent years, increasing attention has been paid to inflammatory reactions as symptoms of a stroke. However, the role of inflammation in stroke and its underlying mechanisms require exploration. In this study, we evaluated the inflammatory reactions induced by acute ischemia and found that pyroptosis occurred after acute ischemia both in vivo and in vitro, as determined by interleukin-1β, apoptosis-associated speck-like protein, and caspase-1. The early inflammation resulted in irreversible ischemic injury, indicating that it deserves thorough investigation. Meanwhile, acute ischemia decreased the Sirtuin 1 (Sirt1) protein levels, and increased the TRAF6 (TNF receptor associated factor 6) protein and reactive oxygen species (ROS) levels. In further exploration, both Sirt1 suppression and TRAF6 activation were found to contribute to this pyroptosis. Reduced Sirt1 levels were responsible for the production of ROS and increased TRAF6 protein levels after ischemic exposure. Moreover, N-acetyl-L-cysteine, an ROS scavenger, suppressed the TRAF6 accumulation induced by oxygen-glucose deprivation via suppression of ROS bursts. These phenomena indicate that Sirt1 is upstream of ROS, and ROS bursts result in increased TRAF6 levels. Further, the activation of Sirt1 during the period of ischemia reduced ischemia-induced injury after 72 h of reperfusion in mice with middle cerebral artery occlusion. In sum, these results indicate that pyroptosis-dependent machinery contributes to the neural injury during acute ischemia via the Sirt1-ROS-TRAF6 signaling pathway. We propose that inflammatory reactions occur soon after oxidative stress and are detrimental to neuronal survival; this provides a promising therapeutic target against ischemic injuries such as a stroke.
Collapse
Affiliation(s)
- Weijie Yan
- Department of Neurobiology, Ministry of Education Key Laboratory for Neurodegenerative Disorders, Capital Medical University, Beijing, 100069, China
| | - Wei Sun
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Jiahui Fan
- Department of Neurobiology, Ministry of Education Key Laboratory for Neurodegenerative Disorders, Capital Medical University, Beijing, 100069, China
| | - Haiqing Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Song Han
- Department of Neurobiology, Ministry of Education Key Laboratory for Neurodegenerative Disorders, Capital Medical University, Beijing, 100069, China
| | - Junfa Li
- Department of Neurobiology, Ministry of Education Key Laboratory for Neurodegenerative Disorders, Capital Medical University, Beijing, 100069, China
| | - Yanling Yin
- Department of Neurobiology, Ministry of Education Key Laboratory for Neurodegenerative Disorders, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
209
|
Wu S, Yang T, Cen K, Zou Y, Shi X, Zhou D, Gao Y, Chai L, Zhao Y, Sun Y, Zhu L. In Vitro Evaluation of the Neuroprotective Effect of Panax notoginseng Saponins by Activating the EGFR/PI3K/AKT Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2020; 2020:1403572. [PMID: 32802113 PMCID: PMC7415117 DOI: 10.1155/2020/1403572] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/27/2020] [Accepted: 05/08/2020] [Indexed: 12/22/2022]
Abstract
OBJECTIVE This study investigated whether Panax notoginseng saponins (PNS) extracted from Panax notoginseng (Bruk.) F. H. Chen played a neuroprotective role by affecting the EGFR/PI3K/AKT pathway in oxygen-glucose deprived (OGD) SH-SY5Y cells. MATERIALS AND METHODS Different groups of OGD SH-SY5Y cells were treated with varying doses of PNS, PNS + AG1478 (a specific inhibitor of EGFR), or AG1478 for 16 hours. CCK8, Annexin V-FITC/PI apoptosis analysis, and LDH release analysis were used to determine cell viability, apoptosis rate, and amounts of LDH. Quantitative real-time PCR (q-RT-PCR) and western blotting were used to measure mRNA and proteins levels of p-EGFR/EGFR, p-PI3K/PI3K, and p-AKT/AKT in SH-SY5Y cells subjected to OGD. RESULTS PNS significantly enhanced cell viability, reduced apoptosis, and weakened cytotoxicity by inhibiting the release of LDH. The mRNA expression profiles of EGFR, PI3K, and AKT showed no difference between model and other groups. Additionally, ratios of p-EGFR, p-PI3K, and p-AKT to EGFR, PI3K, and AKT proteins expression, respectively, all increased significantly. CONCLUSIONS These findings indicate that PNS enhanced neuroprotective effects by activating the EGFR/PI3K/AKT pathway and elevating phosphorylation levels in OGD SH-SY5Y cells.
Collapse
Affiliation(s)
- Shuang Wu
- Key Laboratory of Chinese Internal Medicine of Educational Ministry and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Tiantian Yang
- Key Laboratory of Chinese Internal Medicine of Educational Ministry and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Kai Cen
- Department of Stomatology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yihuai Zou
- Department of Neurology and Stroke Center, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaowei Shi
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Dongrui Zhou
- Key Laboratory of Chinese Internal Medicine of Educational Ministry and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yonghong Gao
- Key Laboratory of Chinese Internal Medicine of Educational Ministry and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Limin Chai
- Key Laboratory of Chinese Internal Medicine of Educational Ministry and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yizhou Zhao
- Key Laboratory of Chinese Internal Medicine of Educational Ministry and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yikun Sun
- Key Laboratory of Chinese Internal Medicine of Educational Ministry and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Lingqun Zhu
- Key Laboratory of Chinese Internal Medicine of Educational Ministry and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
210
|
Zhang DX, Zheng WC, Bai Y, Bai J, Fu L, Wang XP, Zhang LM. CORM-3 improves emotional changes induced by hemorrhagic shock via the inhibition of pyroptosis in the amygdala. Neurochem Int 2020; 139:104784. [PMID: 32652269 DOI: 10.1016/j.neuint.2020.104784] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/30/2020] [Accepted: 06/06/2020] [Indexed: 12/20/2022]
Abstract
Hemorrhagic shock and resuscitation (HSR) may lead to long-term neurological dysfunction, such as depression and anxiety. Carbon monoxide (CO) has emerged as an excellent neuroprotective agent against caspase-1-associated pyroptosis, following HSR. We evaluated the effects and determined the mechanism through which CO protects against emotional changes in a model of HSR, in rats. We subjected rats to treatments with an exogenous, CO-releasing compound (CORM-3, 4 mg/kg), in vivo, after HSR. We measured sucrose preference and performed tail suspension and open field tests 7 days after HSR, assessed brain magnetic resonance imaging 12 h after HSR and evaluated pyroptosis, and neuronal and astrocyte death in the amygdala 12 h post-HSR. We also measured changes in behavior and pathology, following an injection of recombinant murine interleukin (IL)-18 into the amygdala. HSR-treated rats displayed increased depression-like and anxiety-like behaviors, increased amygdalar injury, as indicated by T2-weighted magnetic resonance imaging (MRI) and cerebral blood flow with arterial spin labeling (CBFASL), associated with both neuronal and astrocytic death and pyroptosis, and upregulated IL-18 expression was observed in astrocytes. CORM-3 administration after resuscitation, via a femoral vein injection, provided neuroprotection against HSR, and this neuroprotective effect could be partially reversed by the injection of recombinant murine IL-18 into the amygdala. Therefore, CORM-3 alleviated HSR-induced neuronal pyroptosis and emotional changes, through the downregulation of IL-18 in astrocytes.
Collapse
Affiliation(s)
- Dong-Xue Zhang
- Department of Gerontology, Cangzhou Central Hospital, Cangzhou, China
| | - Wei-Chao Zheng
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, China
| | - Yang Bai
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, China
| | - Jing Bai
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, China
| | - Lan Fu
- Department of Radiodiagnosis, Cangzhou Central Hospital, Cangzhou, China
| | - Xu-Peng Wang
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, China
| | - Li-Min Zhang
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, China.
| |
Collapse
|
211
|
Neuroprotective role of glutathione peroxidase 4 in experimental subarachnoid hemorrhage models. Life Sci 2020; 257:118050. [PMID: 32634425 DOI: 10.1016/j.lfs.2020.118050] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 06/29/2020] [Accepted: 07/01/2020] [Indexed: 01/18/2023]
Abstract
BACKGROUND AND PURPOSE Early brain injury is an essential pathological process after subarachnoid hemorrhage (SAH), with many cell death modalities. Ferroptosis is a newly discovered regulated cell death caused by the iron-dependent accumulation of lipid peroxidation, which can be prevented by glutathione peroxidase 4 (GPX4). Our study aimed to investigate the role of GPX4 in neuronal cell death after experimental SAH. METHODS In vivo experimental SAH was induced by injecting autologous arterial blood into the prechiasmatic cistern in male Sprague-Dawley rats. Meanwhile, the in vitro SAH model was performed with primary rat cortical neurons cultured in medium containing hemoglobin (Hb). Adenovirus was used to overexpress GPX4 before experimental SAH. GPX4 expression was detected by western blot and immunofluorescence experiments. Malondialdehyde (MDA) was measured to evaluate the level of lipid peroxidation. Nissl staining was employed to assess cell death in vivo, whereas lactate dehydrogenase (LDH) release was used to evaluate cell damage in vitro. The brain water content and neurological deficits were evaluated to determine brain injury. RESULTS Endogenous GPX4 was mainly expressed in neurons, and its expression decreased at 24 h after experimental SAH. Overexpression of GPX4 significantly reduced lipid peroxidation and cell death in the experimental SAH models both in vivo and in vitro. Moreover, overexpression of GPX4 ameliorated brain edema and neurological deficits at 24 h after SAH. CONCLUSIONS The decrease of GPX4 expression potentially plays an important role in ferroptosis during early brain injury after SAH. Overexpression of GPX4 has a neuroprotective effect after SAH.
Collapse
|
212
|
Abstract
Patients with stroke have a high risk of infection which may be predicted by age, procalcitonin, interleukin-6, C-reactive protein, National Institute of Health stroke scale (NHSS) score, diabetes, etc. These prediction methods can reduce unfavourable outcome by preventing the occurrence of infection.We aim to identify early predictors for urinary tract infection in patients after stroke.In 186 collected acute stroke patients, we divided them into urinary tract infection group, other infection type groups, and non-infected group. Data were recorded at admission. Independent risk factors and infection prediction model were determined using Logistic regression analyses. Likelihood ratio test was used to detect the prediction effect of the model. Receiver operating characteristic curve and the corresponding area under the curve were used to measure the predictive accuracy of indicators for urinary tract infection.Of the 186 subjects, there were 35 cases of urinary tract infection. Elevated interleukin-6, higher NIHSS, and decreased hemoglobin may be used to predict urinary tract infection. And the predictive model for urinary tract infection (including sex, NIHSS, interleukin-6, and hemoglobin) have the best predictive effect.This study is the first to discover that decreased hemoglobin at admission may predict urinary tract infection. The prediction model shows the best accuracy.
Collapse
Affiliation(s)
- Ya-ming Li
- Department of Neurology, Jiading District Central Hospital affiliated to Shanghai University of Medicine & Health Sciences
| | - Jian-hua Xu
- Department of Neurology, Jiading District Central Hospital affiliated to Shanghai University of Medicine & Health Sciences
| | - Yan-xin Zhao
- Department of Neurology, Tenth People's Hospital affiliated to Tongji University, Shanghai, China
| |
Collapse
|
213
|
Zhang ZY, Fang YJ, Luo YJ, Lenahan C, Zhang JM, Chen S. The role of medical gas in stroke: an updated review. Med Gas Res 2020; 9:221-228. [PMID: 31898607 PMCID: PMC7802415 DOI: 10.4103/2045-9912.273960] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Medical gas is a large class of bioactive gases used in clinical medicine and basic scientific research. At present, the role of medical gas in neuroprotection has received growing attention. Stroke is a leading cause of death and disability in adults worldwide, but current treatment is still very limited. The common pathological changes of these two types of stroke may include excitotoxicity, free radical release, inflammation, cell death, mitochondrial disorder, and blood-brain barrier disruption. In this review, we will discuss the pathological mechanisms of stroke and the role of two medical gases (hydrogen and hydrogen sulfide) in stroke, which may potentially provide a new insight into the treatment of stroke.
Collapse
Affiliation(s)
- Ze-Yu Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Yuan-Jian Fang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Yu-Jie Luo
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Cameron Lenahan
- Burrell College of Osteopathic Medicine, Las Cruces, NM; Center for Neuroscience Research, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Jian-Ming Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Sheng Chen
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
214
|
Wang MM, Zhang M, Feng YS, Xing Y, Tan ZX, Li WB, Dong F, Zhang F. Electroacupuncture Inhibits Neuronal Autophagy and Apoptosis via the PI3K/AKT Pathway Following Ischemic Stroke. Front Cell Neurosci 2020; 14:134. [PMID: 32477073 PMCID: PMC7242565 DOI: 10.3389/fncel.2020.00134] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 04/21/2020] [Indexed: 12/14/2022] Open
Abstract
Electroacupuncture (EA) is a safe and effective therapy for ischemic stroke in both clinical and laboratory settings. However, the underlying mechanism behind EA treatment for stroke remains unclear. Here, we aimed to evaluate whether EA treatment at the acupoints of Zusanli (ST36) and Quchi (LI11) exerted a neuroprotective effect on ischemic stroke rats by modulating autophagy and apoptosis via the PI3K/AKT/mTOR signaling pathway. EA was performed at 24 h following brain ischemia/reperfusion (I/R) for 30 min per day for 3 days. Our results indicated that EA treatment significantly decreased neurological deficits and cerebral infarct volume in ischemic stroke rats. Also, EA intervention markedly reduced neuronal apoptosis by suppressing the activation of cleaved caspase-3 (CCAS3) at 72 h following I/R, as shown by a Western blot analysis. Furthermore, EA treatment after ischemic stroke suppressed the ischemia activated expression level of LC3II/I and Atg7 and increased the ischemia inhibited expression level of PI3K, phosphorylation of mTOR, phosphorylation of AKT, P62 and LAMP1, hence mediating the autophagy level of the neurocyte, which was reversed by the PI3K inhibitor Dactolisib. In summary, our results indicate that the protective effects of EA treatment at points of Quchi (LI11) and Zusanli (ST36) in rats following cerebral I/R injury was associated with the inhibition of neuronal apoptosis and autophagy via activating the PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Man-Man Wang
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Min Zhang
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, China
| | - Ya-Shuo Feng
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ying Xing
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zi-Xuan Tan
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Wen-Bin Li
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, China
| | - Fang Dong
- Department of Clinical Laboratory Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Feng Zhang
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China.,Hebei Provincial Orthopedic Biomechanics Key Laboratory, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
215
|
Liu H, Guo W, Guo H, Zhao L, Yue L, Li X, Feng D, Luo J, Wu X, Cui W, Qu Y. Bakuchiol Attenuates Oxidative Stress and Neuron Damage by Regulating Trx1/TXNIP and the Phosphorylation of AMPK After Subarachnoid Hemorrhage in Mice. Front Pharmacol 2020; 11:712. [PMID: 32499702 PMCID: PMC7243250 DOI: 10.3389/fphar.2020.00712] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 04/30/2020] [Indexed: 12/14/2022] Open
Abstract
Subarachnoid hemorrhage (SAH) is a fatal cerebrovascular condition with complex pathophysiology that reduces brain perfusion and causes cerebral functional impairments. An increasing number of studies indicate that early brain injury (EBI), which occurs within the first 72 h of SAH, plays a crucial role in the poor prognosis of SAH. Bakuchiol (Bak) has been demonstrated to have multiorgan protective effects owing to its antioxidative and anti-inflammatory properties. The present study was designed to investigate the effects of Bak on EBI after SAH and its underlying mechanisms. In this study, 428 adult male C57BL/6J mice weighing 20 to 25 g were observed to investigate the effects of Bak administration in an SAH animal model. The neurological function and brain edema were assessed. Content of MDA/3-NT/8-OHdG/superoxide anion and the activity of SOD and GSH-Px were tested. The function of the blood-brain barrier (BBB) and the protein levels of claudin-5, occludin, zonula occludens-1, and matrix metalloproteinase-9 were observed. TUNEL staining and Fluoro-Jade C staining were conducted to evaluate the death of neurons. Ultrastructural changes of the neurons were observed under the transmission electron microscope. Finally, the roles of Trx, TXNIP, and AMPK in the protective effect of Bak were investigated. The data showed that Bak administration 1) increased the survival rate and alleviated neurological functional deficits; 2) alleviated BBB disruption and brain edema; 3) attenuated oxidative stress by reducing reactive oxygen species, MDA, 3-NT, 8-OHdG, gp91phox, and 4-HNE; increased the activities of SOD and GSH-Px; and alleviated the damage to the ultrastructure of mitochondria; 4) inhibited cellular apoptosis by regulating the protein levels of Bcl-2, Bax, and cleaved caspase-3; and 5) upregulated the protein levels of Trx1 as well as the phosphorylation of AMPK and downregulated the protein levels of TXNIP. Moreover, the protective effects of Bak were partially reversed by PX-12 and compound C. To summarize, Bak attenuates EBI after SAH by alleviating BBB disruption, oxidative stress, and apoptosis via regulating Trx1/TXNIP expression and the phosphorylation of AMPK. Its powerful protective effects might make Bak a promising novel drug for the treatment of EBI after SAH.
Collapse
Affiliation(s)
- Haixiao Liu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China.,Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Wei Guo
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Hao Guo
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Lei Zhao
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Liang Yue
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xia Li
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Dayun Feng
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jianing Luo
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xun Wu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Wenxing Cui
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yan Qu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
216
|
Fang Y, Gao S, Wang X, Cao Y, Lu J, Chen S, Lenahan C, Zhang JH, Shao A, Zhang J. Programmed Cell Deaths and Potential Crosstalk With Blood-Brain Barrier Dysfunction After Hemorrhagic Stroke. Front Cell Neurosci 2020; 14:68. [PMID: 32317935 PMCID: PMC7146617 DOI: 10.3389/fncel.2020.00068] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 03/06/2020] [Indexed: 12/13/2022] Open
Abstract
Hemorrhagic stroke is a life-threatening neurological disease characterized by high mortality and morbidity. Various pathophysiological responses are initiated after blood enters the interstitial space of the brain, compressing the brain tissue and thus causing cell death. Recently, three new programmed cell deaths (PCDs), necroptosis, pyroptosis, and ferroptosis, were also found to be important contributors in the pathophysiology of hemorrhagic stroke. Additionally, blood-brain barrier (BBB) dysfunction plays a crucial role in the pathophysiology of hemorrhagic stroke. The primary insult following BBB dysfunction may disrupt the tight junctions (TJs), transporters, transcytosis, and leukocyte adhesion molecule expression, which may lead to brain edema, ionic homeostasis disruption, altered signaling, and immune infiltration, consequently causing neuronal cell death. This review article summarizes recent advances in our knowledge of the mechanisms regarding these new PCDs and reviews their contributions in hemorrhagic stroke and potential crosstalk in BBB dysfunction. Numerous studies revealed that necroptosis, pyroptosis, and ferroptosis participate in cell death after subarachnoid hemorrhage (SAH) and intracerebral hemorrhage (ICH). Endothelial dysfunction caused by these three PCDs may be the critical factor during BBB damage. Also, several signaling pathways were involved in PCDs and BBB dysfunction. These new PCDs (necroptosis, pyroptosis, ferroptosis), as well as BBB dysfunction, each play a critical role after hemorrhagic stroke. A better understanding of the interrelationship among them might provide us with better therapeutic targets for the treatment of hemorrhagic stroke.
Collapse
Affiliation(s)
- Yuanjian Fang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shiqi Gao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoyu Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yang Cao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianan Lu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Sheng Chen
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Cameron Lenahan
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, United States.,Burrell College of Osteopathic Medicine, Las Cruces, NM, United States.,Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, United States.,Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA, United States.,Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda, CA, United States.,Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Brain Research Institute, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, China
| |
Collapse
|
217
|
Cell Death Pathways in Ischemic Stroke and Targeted Pharmacotherapy. Transl Stroke Res 2020; 11:1185-1202. [PMID: 32219729 DOI: 10.1007/s12975-020-00806-z] [Citation(s) in RCA: 201] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 03/04/2020] [Accepted: 03/10/2020] [Indexed: 02/07/2023]
Abstract
Ischemic stroke is one of the significant causes of morbidity and mortality, affecting millions of people across the globe. Cell injury in the infarct region is an inevitable consequence of focal cerebral ischemia. Subsequent reperfusion exacerbates the harmful effect and increases the infarct volume. These cellular injuries follow either a regulated pathway involving tightly structured signaling cascades and molecularly defined effector mechanisms or a non-regulated pathway, also known as accidental cell death, where the process is biologically uncontrolled. Classical cell death pathways are long established and well reported in several articles that majorly define apoptotic cell death. A recent focus on cell death study also considers investigation on non-classical pathways that are tightly regulated, may or may not involve caspases, but non-apoptotic. Pathological cell death is a cardinal feature of different neurodegenerative diseases. Although ischemia cannot be classified as a neurodegenerative disease, it is a cerebrovascular event where the infarct region exhibits aberrant cell death. Over the past few decades, several therapeutic options have been implicated for ischemic stroke. However, their use has been hampered owing to the number of limitations that they possess. Ischemic penumbral neurons undergo apoptosis and become dysfunctional; however, they are salvageable. Thus, understanding the role of different cell death pathways is crucial to aid in the modern treatment of protecting apoptotic neurons.
Collapse
|
218
|
Liu S, Dai Y, Zhou C, Zhu T. Parecoxib exhibits anti-inflammatory and neuroprotective effects in a rat model of transient global cerebral ischemia. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH-PART A 2020; 83:203-214. [PMID: 32216542 DOI: 10.1080/15287394.2020.1745722] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Transient global cerebral ischemia (tGCI) induces inflammation leading to secondary brain injury. Data suggested that cyclooxygenase-2 (COX-2) is involved in the occurrence and development of inflammatory reaction after reperfusion; however, the effectiveness of a highly selective COX-2 inhibitor, parecoxib, to counteract tGCI remains to be determined. Thus, the aim of this study was to investigate the potential protective actions of parecoxib in a rat model of tGCI and the role inflammation plays in this disorder. Adult male Sprague-Dawley rats were administered parecoxib 10 or 20 mg/kg intraperitoneally (ip) at 5 min, 24 or 48 hr after tGCI. Control rats received an equal volume of 0.9% saline. The rat model of tGCI was established using the method of bilateral common carotid artery occlusion combined with arterial hypotension. The following parameters were measured: Neurological Severity Score, morphological changes in the hippocampal CA1 region, Evans blue (EB) extravasation, brain water content, levels of matrix metalloproteinase-9 (MMP-9), zonula occludens-1 (ZO-1), neuronal apoptosis, the protein expression of Bcl-2, Bax, COX-2, prostaglandin E2 (PGE2), interleukin-1β (IL-1β), and tumor necrosis factor-α (TNF-α). Parecoxib treatment significantly improved neurological function and morphological defects in the hippocampal CA1 region, reduced levels of COX-2, PGE2, IL-1β, and TNF-α. In addition, parecoxib attenuated brain edema and BBB destruction as evidenced by increased ZO-1 expression and decreased MMP-9 expression. Further, parecoxib reduced neuronal apoptosis via diminished protein expression of Bax and enhanced expression of Bcl-2.
Collapse
Affiliation(s)
- Shaoxing Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,Department of Anesthesiology, Chengdu Second People's Hospital, Chengdu, Sichuan, China
| | - Yue'e Dai
- Department of Anesthesiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Chen Zhou
- The Laboratory of Anesthesia and Critical Care Medicine, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tao Zhu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
219
|
Fu L, Zhang DX, Zhang LM, Song YC, Liu FH, Li Y, Wang XP, Zheng WC, Wang XD, Gui CX, Kong XJ, Kang LQ. Exogenous carbon monoxide protects against mitochondrial DNA‑induced hippocampal pyroptosis in a model of hemorrhagic shock and resuscitation. Int J Mol Med 2020; 45:1176-1186. [PMID: 32124959 PMCID: PMC7053849 DOI: 10.3892/ijmm.2020.4493] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 01/29/2020] [Indexed: 11/05/2022] Open
Abstract
Carbon monoxide‑releasing molecule‑3 (CORM‑3), which is an exogenous carbon monoxide (CO) compound, slowly releases CO under physiological conditions; this exerts neuroprotective effects against incomplete ischemia/reperfusion injury. The objective of the present study was to investigate whether the administration of CORM‑3 protects against nucleotide‑binding oligomerization domain‑like receptor pyrin domain‑3 (NLRP3) inflammasome formation and neuronal pyroptosis in the hippocampus following hemorrhagic shock and resuscitation (HSR). To establish this, an HSR model was created. Hemorrhagic shock was induced in adult male Sprague‑Dawley rats under sevoflurane anesthesia by bleeding using a heparinized syringe to maintain a mean arterial pressure of 30±5 mmHg for 60 min. Resuscitation was performed by reperfusion of the blood and, if necessary, administering sterile saline to achieve the baseline arterial pressure. Following resuscitation, CORM‑3 (4 mg/kg) was injected via the femoral vein. Neuronal pyroptosis in the hippocampus, mitochondrial morphology, mitochondrial DNA (mtDNA), brain magnetic resonance imaging, expression levels of NLRP3 and the interaction of pro‑caspase‑1 and apoptosis‑associated speck‑like protein containing a CARD domain (ASC) were examined 12 h after HSR; locomotor activity was assessed 7 days after HSR. Compared with HSR‑treated rats, CORM‑3 administration resulted in a lower level of neuronal pyroptosis in the hippocampus, improved mitochondrial morphology, a lower mtDNA level, steadier levels of metabolites, decreased expression levels of NLRP3 and pro‑caspase‑1 interacting with ASC and enhanced locomotor activity. In conclusion, treatment with CORM‑3 ameliorated impairments of locomotor and exploratory activities in a rat model of HSR. The mechanism may be associated with the inhibition of mitochondrial DNA‑induced pyroptosis via improvements in cell metabolism.
Collapse
Affiliation(s)
- Lan Fu
- Graduate School, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Dong-Xue Zhang
- Department of Gerontology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Li-Min Zhang
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Yan-Cheng Song
- Department of Radiodiagnosis, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Feng-Hai Liu
- Department of Radiodiagnosis, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Yan Li
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Xu-Peng Wang
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Wei-Chao Zheng
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Xiao-Dong Wang
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Chun-Xiao Gui
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Xiang-Jun Kong
- Central Laboratory, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Li-Qing Kang
- Department of Radiodiagnosis, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| |
Collapse
|
220
|
Wang GY, Wang TZ, Zhang YY, Li F, Yu BY, Kou JP. NMMHC IIA Inhibition Ameliorates Cerebral Ischemic/Reperfusion-Induced Neuronal Apoptosis Through Caspase-3/ROCK1/MLC Pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:13-25. [PMID: 32021088 PMCID: PMC6954088 DOI: 10.2147/dddt.s230882] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/19/2019] [Indexed: 12/26/2022]
Abstract
Purpose Our previous studies have indicated that non-muscle myosin heavy chain IIA (NMMHC IIA) is involved in H2O2-induced neuronal apoptosis, which is associated with the positive feedback loop of caspase-3/ROCK1/MLC pathway. However, the neuroprotective effect of NMMHC IIA inhibition with an adeno-associated virus (AAV) vector after transient middle cerebral artery occlusion (MCAO) and its role in caspases-3/ROCK1/MLC pathway remain blurred. Methods Green fluorescent protein (GFP) and a small hairpin RNA targeting Myh9 (encoding NMMHC IIA) were cloned and packaged into the AAV9 vector. AAV-shMyh9 or control vector were injected into C57BL/6J mice four weeks prior to 60 min MCAO. Twenty-four hours after reperfusion, functional and histological analyses of the mice were performed. Results In this study, AAV-shMyh9 was used to down-regulate NMMHC IIA expression in mice. We found that down-regulation of NMMHC IIA could improve neurological scores and histological injury in ischemic mice. Ischemic attack also activated neuronal apoptosis, and this effect was partially attenuated when NMMHC IIA was inhibited by AAV-shMyh9. In addition, AAV-shMyh9 significantly reduced cerebral ischemic/reperfusion (I/R)-induced NMMHC IIA-actin interaction, caspase-3 cleavage, Rho-associated kinase1 (ROCK1) activation and myosin light-chains (MLC) phosphorylation. Conclusion Consequently, we showed that AAV-shMyh9 inhibits I/R-induced neuronal apoptosis linked with caspase-3/ROCK1/MLC/NMMHC IIA-actin cascade, which has also been confirmed to be a positive feedback loop. These findings put some insights into the neuroprotective effect of AAV-shMyh9 associated with the regulation of NMMHC IIA-related pathway under ischemic attack and provide a therapeutic strategy for ischemic stroke.
Collapse
Affiliation(s)
- Guang-Yun Wang
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Tie-Zheng Wang
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Yuan-Yuan Zhang
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Fang Li
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Bo-Yang Yu
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Resource and Development of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Jun-Ping Kou
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| |
Collapse
|
221
|
Park J, Lai MKP, Arumugam TV, Jo DG. O-GlcNAcylation as a Therapeutic Target for Alzheimer's Disease. Neuromolecular Med 2020; 22:171-193. [PMID: 31894464 DOI: 10.1007/s12017-019-08584-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 12/13/2019] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia and the number of elderly patients suffering from AD has been steadily increasing. Despite worldwide efforts to cope with this disease, little progress has been achieved with regard to identification of effective therapeutics. Thus, active research focusing on identification of new therapeutic targets of AD is ongoing. Among the new targets, post-translational modifications which modify the properties of mature proteins have gained attention. O-GlcNAcylation, a type of PTM that attaches O-linked β-N-acetylglucosamine (O-GlcNAc) to a protein, is being sought as a new target to treat AD pathologies. O-GlcNAcylation has been known to modify the two important components of AD pathological hallmarks, amyloid precursor protein, and tau protein. In addition, elevating O-GlcNAcylation levels in AD animal models has been shown to be effective in alleviating AD-associated pathology. Although studies investigating the precise mechanism of reversal of AD pathologies by targeting O-GlcNAcylation are not yet complete, it is clearly important to examine O-GlcNAcylation regulation as a target of AD therapeutics. This review highlights the mechanisms of O-GlcNAcylation and its role as a potential therapeutic target under physiological and pathological AD conditions.
Collapse
Affiliation(s)
- Jinsu Park
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Korea
- Department of Health Science and Technology, Sungkyunkwan University, Seoul, 06351, Korea
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Thiruma V Arumugam
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Korea.
- Department of Physiology, Yong Loo Lin School Medicine, National University of Singapore, Singapore, 117593, Singapore.
- Department of Physiology, Anatomy & Microbiology, School of Life Sciences, La Trobe University, Bundoora, VIC, Australia.
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Korea.
- Department of Health Science and Technology, Sungkyunkwan University, Seoul, 06351, Korea.
- Biomedical Institute for Convergence, Sungkyunkwan University, Suwon, 16419, Korea.
| |
Collapse
|
222
|
Santos PT, O'Brien CE, Chen MW, Hopkins CD, Adams S, Kulikowicz E, Singh R, Koehler RC, Martin LJ, Lee JK. Proteasome Biology Is Compromised in White Matter After Asphyxic Cardiac Arrest in Neonatal Piglets. J Am Heart Assoc 2019; 7:e009415. [PMID: 30371275 PMCID: PMC6474957 DOI: 10.1161/jaha.118.009415] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Neurological deficits in hypoxic‐ischemic encephalopathy, even with therapeutic hypothermia, are partially attributed to white matter injury. We theorized that proteasome insufficiency contributes to white matter injury. Methods and Results Neonatal piglets received hypoxia‐ischemia (HI) or sham procedure with normothermia, hypothermia, or hypothermia+rewarming. Some received a proteasome activator drug (oleuropein) or white matter–targeted, virus‐mediated proteasome knockdown. We measured myelin oligodendrocyte glycoprotein, proteasome subunit 20S (P20S), proteasome activity, and carbonylated and ubiquitinated protein levels in white matter and cerebral cortex. HI reduced myelin oligodendrocyte glycoprotein levels regardless of temperature, and myelin oligodendrocyte glycoprotein loss was associated with increased ubiquitinated and carbonylated protein levels. Ubiquitinated and carbonyl‐damaged proteins increased in white matter 29 hours after HI during hypothermia to exceed levels at 6 to 20 hours. In cortex, ubiquitinated proteins decreased. Ubiquitinated and carbonylated protein accumulation coincided with lower P20S levels in white matter; P20S levels also decreased in cerebral cortex. However, proteasome activity in white matter lagged behind that in cortex 29 hours after HI during hypothermia. Systemic oleuropein enhanced white matter P20S and protected the myelin, whereas proteasome knockdown exacerbated myelin oligodendrocyte glycoprotein loss and ubiquitinated protein accumulation after HI. At the cellular level, temperature and HI interactively affected macroglial P20S enrichment in subcortical white matter. Rewarming alone increased macroglial P20S immunoreactivity, but this increase was blocked by HI. Conclusions Oxidized and ubiquitinated proteins accumulate with HI‐induced white matter injury. Proteasome insufficiency may drive this injury. Hypothermia did not prevent myelin damage, protect the proteasome, or preserve oxidized and ubiquitinated protein clearance after HI.
Collapse
Affiliation(s)
- Polan T Santos
- 1 Department of Anesthesiology and Critical Care Medicine Johns Hopkins University Baltimore MD
| | - Caitlin E O'Brien
- 1 Department of Anesthesiology and Critical Care Medicine Johns Hopkins University Baltimore MD
| | - May W Chen
- 2 Division of Neonatology Department of Pediatrics Johns Hopkins University Baltimore MD
| | - C Danielle Hopkins
- 1 Department of Anesthesiology and Critical Care Medicine Johns Hopkins University Baltimore MD
| | - Shawn Adams
- 1 Department of Anesthesiology and Critical Care Medicine Johns Hopkins University Baltimore MD
| | - Ewa Kulikowicz
- 1 Department of Anesthesiology and Critical Care Medicine Johns Hopkins University Baltimore MD
| | - Rashmi Singh
- 1 Department of Anesthesiology and Critical Care Medicine Johns Hopkins University Baltimore MD
| | - Raymond C Koehler
- 1 Department of Anesthesiology and Critical Care Medicine Johns Hopkins University Baltimore MD
| | - Lee J Martin
- 3 Department of Pathology Johns Hopkins University Baltimore MD
| | - Jennifer K Lee
- 1 Department of Anesthesiology and Critical Care Medicine Johns Hopkins University Baltimore MD
| |
Collapse
|
223
|
Wu L, Zeng S, Cao Y, Huang Z, Liu S, Peng H, Zhi C, Ma S, Hu K, Yuan Z. Inhibition of HDAC4 Attenuated JNK/c-Jun-Dependent Neuronal Apoptosis and Early Brain Injury Following Subarachnoid Hemorrhage by Transcriptionally Suppressing MKK7. Front Cell Neurosci 2019; 13:468. [PMID: 31708743 PMCID: PMC6823346 DOI: 10.3389/fncel.2019.00468] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 10/01/2019] [Indexed: 12/31/2022] Open
Abstract
The c-Jun N-terminal kinase (JNK)/c-Jun cascade-dependent neuronal apoptosis has been identified as a central element for early brain injury (EBI) following subarachnoid hemorrhage (SAH), but the molecular mechanisms underlying this process are still thoroughly undefined to date. In this study, we found that pan-histone deacetylase (HDAC) inhibition by TSA, SAHA, VPA, and M344 led to a remarkable decrease in the phosphorylation of JNK and c-Jun, concomitant with a significant abrogation of apoptosis caused by potassium deprivation in cultured cerebellar granule neurons (CGNs). Further investigation showed that these effects resulted from HDAC inhibition-induced transcriptional suppression of MKK7, a well-known upstream kinase of JNK. Using small interference RNAs (siRNAs) to silence the respective HDAC members, HDAC4 was screened to be required for MKK7 transcription and JNK/c-Jun activation. LMK235, a specific HDAC4 inhibitor, dose-dependently suppressed MKK7 transcription and JNK/c-Jun activity. Functionally, HDAC4 inhibition via knockdown or LMK235 significantly rescued CGN apoptosis induced by potassium deprivation. Moreover, administration of LMK235 remarkably ameliorated the EBI process in SAH rats, associated with an obvious reduction in MKK7 transcription, JNK/c-Jun activity, and neuronal apoptosis. Collectively, the findings provide new insights into the molecular mechanism of neuronal apoptosis regarding HDAC4 in the selective regulation of MKK7 transcription and JNK/c-Jun activity. HDAC4 inhibition could be a potential alternative to prevent MKK7/JNK/c-Jun axis-mediated nervous disorders, including SAH-caused EBI.
Collapse
Affiliation(s)
- Liqiang Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province, Ministry of Education of China, Institute of Neuroscience of Guangzhou Medical University, Guangzhou, China
| | - Shulian Zeng
- Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province, Ministry of Education of China, Institute of Neuroscience of Guangzhou Medical University, Guangzhou, China
| | - Yali Cao
- Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province, Ministry of Education of China, Institute of Neuroscience of Guangzhou Medical University, Guangzhou, China
| | - Ziyan Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province, Ministry of Education of China, Institute of Neuroscience of Guangzhou Medical University, Guangzhou, China
| | - Sisi Liu
- Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province, Ministry of Education of China, Institute of Neuroscience of Guangzhou Medical University, Guangzhou, China
| | - Huaidong Peng
- Department of Pharmacy, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Cheng Zhi
- Department of Pathology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shanshan Ma
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Guangzhou, China
| | - Kunhua Hu
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Guangzhou, China
| | - Zhongmin Yuan
- Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province, Ministry of Education of China, Institute of Neuroscience of Guangzhou Medical University, Guangzhou, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Guangzhou, China
| |
Collapse
|
224
|
Grimsey NL, Savinainen JR, Attili B, Ahamed M. Regulating membrane lipid levels at the synapse by small-molecule inhibitors of monoacylglycerol lipase: new developments in therapeutic and PET imaging applications. Drug Discov Today 2019; 25:330-343. [PMID: 31622747 DOI: 10.1016/j.drudis.2019.10.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 09/17/2019] [Accepted: 10/09/2019] [Indexed: 12/14/2022]
Abstract
Monoacylglycerol lipase (MAGL) is a major endocannabinoid hydrolyzing enzyme and can be regulated to control endogenous lipid levels in the brain. This review highlights the pharmacological roles and in vivo PET imaging of MAGL in brain.
Collapse
Affiliation(s)
- Natasha L Grimsey
- Department of Pharmacology and Clinical Pharmacology, and Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Juha R Savinainen
- Institute of Biomedicine, Faculty of Health Sciences, The University of Eastern Finland, Finland
| | - Bala Attili
- Department of Radiology, The University of Cambridge, UK
| | - Muneer Ahamed
- ARC Centre for Innovation in Biomedical Imaging Technology, Centre for Advanced Imaging, The University of Queensland, Australia.
| |
Collapse
|
225
|
Hu L, Zhang H, Wang B, Ao Q, Shi J, He Z. MicroRNA-23b alleviates neuroinflammation and brain injury in intracerebral hemorrhage by targeting inositol polyphosphate multikinase. Int Immunopharmacol 2019; 76:105887. [PMID: 31536904 DOI: 10.1016/j.intimp.2019.105887] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/21/2019] [Accepted: 09/05/2019] [Indexed: 12/27/2022]
Abstract
Neuroinflammation plays a critical role in the pathogenesis of intracerebral hemorrhage (ICH), contributing to detrimental brain injury and neurological function deficits. MicroRNA-23b (miR-23b) exerts anti-inflammatory effects in many diseases and is downregulated in patients with ICH. This study aimed to evaluate the involvement of miR-23b in ICH models in vivo and in vitro, using basal ganglia injection of collagenase type VII in rats and hemin stimulation for cells, respectively. Exogenous overexpression of miR-23b by transfection with lentivirus-miR-23b (LV-miR-23b) or miR-23b mimics was evaluated by RT-qPCR. In this study, we found miR-23b was downregulated in the ICH models and its overexpression effectively alleviated neurological deficits, brain edema, hematoma area, and neuronal apoptosis in ICH rats. Western blotting for neuroinflammation markers and immunofluorescence staining for microglial activation demonstrated that miR-23b could alleviate neuroinflammation in ICH in vivo. We also performed an in vitro mechanism study using BV2 microglial cells and HT22 neuronal cell lines to explore how miR-23b modulates neuroinflammation and neuronal protection after ICH. We found that miR-23b significantly decreased hemin-stimulated inflammation response in BV2 cells and attenuated co-cultured HT22 neuronal cell death. Additionally, we verified that miR-23b suppressed inflammation in BV2 cells by targeting inositol polyphosphate multikinase (IPMK) and that autophagy regulation through the Akt/mTOR pathway was involved in miR-23b-regulated inflammation after ICH. Our study illustrated that miR-23b played a protective role in ICH through inhibiting neuroinflammation by targeting IPMK; this mechanism may be related to the regulation of the Akt/mTOR autophagy pathway, making it a potential target for ICH treatment.
Collapse
Affiliation(s)
- Liuting Hu
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, 110000,People's Republic of China
| | - Heyu Zhang
- Dapartment of Neurology, The First Affiliated Hospital Sun Yat-sen University,Guangzhou 510080,People's Republic of China
| | - Bingyang Wang
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, 110000,People's Republic of China
| | - Qiang Ao
- Department of Tissue Engineering, China Medical University, Shenyang 110122, People's Republic of China
| | - Jing Shi
- Department of Neurology, Dandong Central Hospital, Dandong 118002,People's Republic of China
| | - Zhiyi He
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, 110000,People's Republic of China.
| |
Collapse
|
226
|
Alkuraishy HM, Al-Gareeb AI, Waheed HJ. Lipoprotein-Associated Phospholipase A2 is Linked with Poor Cardio-Metabolic Profile in Patients with Ischemic Stroke: A Study of Effects of Statins. J Neurosci Rural Pract 2019; 9:496-503. [PMID: 30271040 PMCID: PMC6126307 DOI: 10.4103/jnrp.jnrp_97_18] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Objectives The objective of the study is to investigate the effects of statins on the lipoprotein-associated phospholipase A2 (Lp-PLA2) mass in patients with ischemic stroke. Materials and Methods A total number of 59 patient ages 43-69 years with cerebral stroke compared to 39 healthy controls that matching the age and body weight. The patients were divided into 32 patients on statins therapy assigned as statins users and 27 patients, not on statins therapy assigned as nonstatins users. Anthropometric and biochemical measurements were done including lipid profile and inflammatory biomarkers. Results Stroke patients on statins therapy showed a comparable low of Lp-PLA (29.82 ± 3.19 IU/mL) to nonstatins user stroke patients (15.58 ± 5.73 IU/mL). Lp-PLA2 mass levels were positively correlated with body mass index, blood pressure changes, total cholesterol, triglyceride, and very low-density lipoprotein and stroke risk (SR) percentage. Conclusions Patients on statins with ischemic stroke had low levels of Lp-PLA2 mass levels compared to nonstatins user with ischemic stroke. Lp-PLA2 mass levels were higher in men than women and correlated with lipid profile and SR in patients with ischemic stroke.
Collapse
Affiliation(s)
- Hayder M Alkuraishy
- Department of Clinical Pharmacology and Therapeutic, Medical Faculty College of Medicine, Al-Mustansiriya University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Therapeutic, Medical Faculty College of Medicine, Al-Mustansiriya University, Baghdad, Iraq
| | - Huda J Waheed
- Department of Clinical Pharmacology and Therapeutic, Medical Faculty College of Medicine, Al-Mustansiriya University, Baghdad, Iraq
| |
Collapse
|
227
|
Zhang LM, Zhang DX, Fu L, Li Y, Wang XP, Qi MM, Li CC, Song PP, Wang XD, Kong XJ. Carbon monoxide-releasing molecule-3 protects against cortical pyroptosis induced by hemorrhagic shock and resuscitation via mitochondrial regulation. Free Radic Biol Med 2019; 141:299-309. [PMID: 31265876 DOI: 10.1016/j.freeradbiomed.2019.06.031] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 06/28/2019] [Accepted: 06/29/2019] [Indexed: 01/14/2023]
Abstract
OBJECTIVE Carbon monoxide (CO) releasing molecule (CORM)-3, a water-soluble CORM, has protective effects against inflammatory and ischemia/reperfusion injury. We determined the effect of CORM-3 against neuronal pyroptosis in a model of hemorrhagic shock and resuscitation (HSR) in rats via mitochondrial regulation. METHODS Rats were treated with CORM-3 (4 mg/kg) in vitro after HSR. We measured cortical CO content 3-24 h after HSR; assessed neuronal pyroptosis, mitochondrial morphology, ROS production, and mitochondrial membrane potential at 12 h after HSR; and evaluated brain magnetic resonance imaging at 24 h after HSR and learning ability 30 days after HSR. We also measured soluble guanylate-cyclase (sGC)-cyclic guanosine monophosphate (cGMP) signaling pathway activity using a blocker of sGC, NS2028, and 125I-cGMP assay. RESULTS Among rats that underwent HSR, CORM-3-treated rats had more CO in the cortical tissue than sham- and iCORM-3-treated rats. CORM-3-treated rats had significantly less neuronal pyroptosis in the cortical tissue; higher sGC activity and cGMP content; lower ROS production; better mitochondrial morphology, function, and membrane potential; and enhanced learning/memory ability than HSR-treated rats. However, these neuroprotective effects of CORM-3 were partially inhibited by NS2028. CONCLUSION CORM-3 may alleviate neuronal pyroptosis and improve neurological recovery in HSR through mitochondrial regulation mediated by the sGC-cGMP pathway. Thus, CO administration could be a promising therapeutic strategy for hemorrhagic shock.
Collapse
Affiliation(s)
- Li-Min Zhang
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, China.
| | - Dong-Xue Zhang
- Department of Gerontology, Cangzhou Central Hospital, Cangzhou, China
| | - Lan Fu
- Department of Radiodiagnosis, Cangzhou Central Hospital, Cangzhou, China
| | - Yan Li
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, China
| | - Xu-Peng Wang
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, China
| | - Man-Man Qi
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, China
| | - Chen-Chen Li
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, China
| | - Pan-Pan Song
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, China
| | - Xiao-Dong Wang
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, China
| | - Xiang-Jun Kong
- Central Laboratory, Cangzhou Central Hospital, Cangzhou, China
| |
Collapse
|
228
|
Burmistrova O, Olias-Arjona A, Lapresa R, Jimenez-Blasco D, Eremeeva T, Shishov D, Romanov S, Zakurdaeva K, Almeida A, Fedichev PO, Bolaños JP. Targeting PFKFB3 alleviates cerebral ischemia-reperfusion injury in mice. Sci Rep 2019; 9:11670. [PMID: 31406177 PMCID: PMC6691133 DOI: 10.1038/s41598-019-48196-z] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 07/26/2019] [Indexed: 12/12/2022] Open
Abstract
The glycolytic rate in neurons is low in order to allow glucose to be metabolized through the pentose-phosphate pathway (PPP), which regenerates NADPH to preserve the glutathione redox status and survival. This is controlled by 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3 (PFKFB3), the pro-glycolytic enzyme that forms fructose-2,6-bisphosphate, a powerful allosteric activator of 6-phosphofructo-1-kinase. In neurons, PFKFB3 protein is physiologically inactive due to its proteasomal degradation. However, upon an excitotoxic stimuli, PFKFB3 becomes stabilized to activate glycolysis, thus hampering PPP mediated protection of redox status leading to neurodegeneration. Here, we show that selective inhibition of PFKFB3 activity by the small molecule AZ67 prevents the NADPH oxidation, redox stress and apoptotic cell death caused by the activation of glycolysis triggered upon excitotoxic and oxygen-glucose deprivation/reoxygenation models in mouse primary neurons. Furthermore, in vivo administration of AZ67 to mice significantly alleviated the motor discoordination and brain infarct injury in the middle carotid artery occlusion ischemia/reperfusion model. These results show that pharmacological inhibition of PFKFB3 is a suitable neuroprotective therapeutic strategy in excitotoxic-related disorders such as stroke.
Collapse
Affiliation(s)
| | - Ana Olias-Arjona
- Institute of Functional Biology and Genomics (IBFG), Universidad de Salamanca, CSIC, Salamanca, Spain
| | - Rebeca Lapresa
- Institute of Functional Biology and Genomics (IBFG), Universidad de Salamanca, CSIC, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Hospital Universitario de Salamanca, Universidad de Salamanca, Salamanca, Spain
| | - Daniel Jimenez-Blasco
- Institute of Functional Biology and Genomics (IBFG), Universidad de Salamanca, CSIC, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Hospital Universitario de Salamanca, Universidad de Salamanca, Salamanca, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | | | | | | | | | - Angeles Almeida
- Institute of Functional Biology and Genomics (IBFG), Universidad de Salamanca, CSIC, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Hospital Universitario de Salamanca, Universidad de Salamanca, Salamanca, Spain
| | | | - Juan P Bolaños
- Institute of Functional Biology and Genomics (IBFG), Universidad de Salamanca, CSIC, Salamanca, Spain.
- Institute of Biomedical Research of Salamanca (IBSAL), Hospital Universitario de Salamanca, Universidad de Salamanca, Salamanca, Spain.
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain.
| |
Collapse
|
229
|
Evzelman MA, Mityaeva EV, Lashkhiia IB, Kamchatnov PR. Acute cerebral ischemia and inflammation. Zh Nevrol Psikhiatr Im S S Korsakova 2019; 119:73-80. [DOI: 10.17116/jnevro201911912273] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
230
|
He HW, Zhang YL, Yu BQ, Ye G, You W, So KF, Li X. Soluble Nogo receptor 1 fusion protein protects neural progenitor cells in rats with ischemic stroke. Neural Regen Res 2019; 14:1755-1764. [PMID: 31169193 PMCID: PMC6585563 DOI: 10.4103/1673-5374.257531] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Soluble Nogo66 receptor-Fc protein (sNgR-Fc) enhances axonal regeneration following central nervous system injury. However, the underlying mechanisms remain unclear. In this study, we investigated the effects of sNgR-Fc on the proliferation and differentiation of neural progenitor cells. The photothrombotic cortical injury model of ischemic stroke was produced in the parietal cortex of Sprague-Dawley rats. The rats with photothrombotic cortical injury were randomized to receive infusion of 400 μg/kg sNgR-Fc (sNgR-Fc group) or an equal volume of phosphate-buffered saline (photothrombotic cortical injury group) into the lateral ventricle for 3 days. The effects of sNgR-Fc on the proliferation and differentiation of endogenous neural progenitor cells were examined using BrdU staining. Neurological function was evaluated with the Morris water maze test. To further examine the effects of sNgR-Fc treatment on neural progenitor cells, photothrombotic cortical injury was produced in another group of rats that received transplantation of neural progenitor cells from the hippocampus of embryonic Sprague-Dawley rats. The animals were then given an infusion of phosphate-buffered saline (neural progenitor cells group) or sNgR-Fc (sNgR-Fc + neural progenitor cells group) into the lateral ventricle for 3 days. sNgR-Fc enhanced the proliferation of cultured neural progenitor cells in vitro as well as that of endogenous neural progenitor cells in vivo, compared with phosphate-buffered saline, and it also induced the differentiation of neural progenitor cells into neurons. Compared with the photothrombotic cortical injury group, escape latency in the Morris water maze and neurological severity score were greatly reduced, and distance traveled in the target quadrant was considerably increased in the sNgR-Fc group, indicating a substantial improvement in neurological function. Furthermore, compared with phosphate-buffered saline infusion, sNgR-Fc infusion strikingly improved the survival and differentiation of grafted neural progenitor cells. Our findings show that sNgR-Fc regulates neural progenitor cell proliferation, migration and differentiation. Therefore, sNgR-Fc is a potential novel therapy for stroke and neurodegenerative diseases, The protocols were approved by the Committee on the Use of Live Animals in Teaching and Research of the University of Hong Kong (approval No. 4560-17) in November, 2015.
Collapse
Affiliation(s)
- Hai-Wei He
- School of Medicine, South China University of Technology; Department of Emergency Medicine, Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong Province, China
| | - Yue-Lin Zhang
- Department of Emergency Medicine, Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong Province, China
| | - Bao-Qi Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Key Laboratory of Remodelling- related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Gen Ye
- Department of Emergency Medicine, Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong Province, China
| | - Wei You
- Department of Emergency Medicine, Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong Province, China
| | - Kwok-Fai So
- School of Biomedical Sciences, The State Key Laboratory of Brain and Cognitive sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Xin Li
- School of Medicine, South China University of Technology; Department of Emergency Medicine, Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong Province, China
| |
Collapse
|
231
|
Jiang Q, Stone CR, Geng X, Ding Y. Hypoxia-inducible factor-1 α and RIP3 triggers NLRP3 inflammasome in ischemic stroke. Brain Circ 2018; 4:191-192. [PMID: 30693347 PMCID: PMC6329214 DOI: 10.4103/bc.bc_35_18] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 12/06/2018] [Accepted: 12/06/2018] [Indexed: 01/06/2023] Open
Affiliation(s)
- Qian Jiang
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Christopher R Stone
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Xiaokun Geng
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China.,Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan, USA.,Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
232
|
Shao BZ, Cao Q, Liu C. Targeting NLRP3 Inflammasome in the Treatment of CNS Diseases. Front Mol Neurosci 2018; 11:320. [PMID: 30233319 PMCID: PMC6131647 DOI: 10.3389/fnmol.2018.00320] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 08/17/2018] [Indexed: 12/12/2022] Open
Abstract
Central nervous system (CNS) is one of the largest killers of people’s health all over the world. The overactivation of the immune and inflammatory responses is considered as an important factor, contributing to the pathogenesis and progression of CNS disorders. Among all kinds of immune and inflammatory reaction, the inflammasome, a complex of proteins, has been drawn increasingly attention to by researchers. The initiation and activation of the inflammasome is involved in the onset of various kinds of diseases. The NLRP3 inflammasome, the most studied member of the inflammasome, is closely associated with many kinds of CNS disorders. Here in this review, the roles of the NLRP3 inflammasome in the pathogenesis and progression of several well-known CNS diseases would be discussed, including cerebrovascular diseases, neurodegenerative diseases, multiple sclerosis, depression as well as other CNS disorders. In addition, several therapeutic strategies targeting on the NLRP3 inflammasome for the treatment of CNS disorders would be described in this review.
Collapse
Affiliation(s)
- Bo-Zong Shao
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| | - Qi Cao
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| | - Chong Liu
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| |
Collapse
|
233
|
Kim YO, Kim HJ, Abu-Taweel GM, Oh J, Sung GH. Neuroprotective and therapeutic effect of Cordyceps militaris on ischemia-induced neuronal death and cognitive impairments. Saudi J Biol Sci 2018; 26:1352-1357. [PMID: 31762595 PMCID: PMC6864366 DOI: 10.1016/j.sjbs.2018.08.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 08/14/2018] [Accepted: 08/16/2018] [Indexed: 12/12/2022] Open
Abstract
Cordyceps militaris is a type of fungus consumed by people all over the world and renowned for their nutritional benefits and herbal formulas to promote health and longevity. In the present study investigation was carried out to explore the therapeutic properties and neuroprotective effect of the C. militaris on ischemic brain neuronal injury, impairment of memory and learning in experimental rats induced by a global cerebral ischemia-reperfusion injury in WISTAR rats. Vascular Dementia with transient global brain injuries induced by a four-vessel occlusion (4-VO) in WISTAR rats. Further, donepezil (5 mg/kg) and C. militaris was (100 and 300 mg/kg, p.o.) were orally administered for 7 days in 4-VO WISTAR rats. C. militaris has the ability to improve memory impairments due to global cerebral ischemia and scopolamine-induced memory deterioration. Our present findings suggest that C. militaris may be a potential candidate for the neuroprotection of hippocampus and the recovery of various vascular dementia or neuroinflammatory disorders.
Collapse
Affiliation(s)
- Young Ock Kim
- Institute for Healthcare and Life Science, International St. Mary's Hospital and College of Medicine, Catholic Kwandong University, Incheon 22711, Republic of Korea
| | - Hak Jae Kim
- Department of Clinical Pharmacology, College of Medicine, Soonchunhyang University Cheonan Chungam, 31151, Republic of Korea
| | - Gasem Mohammad Abu-Taweel
- Department of Basic Sciences, College of Education, Imam Abdulrahman Bin Faisal University, P.O. Box 2375, Dammam 31451, Saudi Arabia
| | - Junsang Oh
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea.,Department of Microbiology, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do 25601, Republic of Korea
| | - Gi-Ho Sung
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea.,Department of Microbiology, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do 25601, Republic of Korea
| |
Collapse
|