1
|
Kim JH, Lee JH, Nan Z, Choi JW, Song JW. Di(2-ethylhexyl) phthalate exposure aggravates hypoxia/reoxygenation injury in cerebral endothelial cells by downregulating epithelial cadherin expression. Toxicol Res (Camb) 2024; 13:tfae163. [PMID: 39371678 PMCID: PMC11447374 DOI: 10.1093/toxres/tfae163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/06/2024] [Accepted: 09/26/2024] [Indexed: 10/08/2024] Open
Abstract
Di-(2-ethylhexyl) phthalate (DEHP) is a widely used plasticizer that has adverse health effects. Most phthalates exhibit reproductive toxicity and are associated with diseases such as cardiovascular disorders. However, the effect of DEHP exposure on acute hypoxia/reperfusion injury remains unknown. Therefore, we assessed whether hypoxia/reperfusion injury is aggravated by exposure to DEHP and investigated plausible underlying mechanisms, including oxidative stress and expression of cyclooxygenase-2 (COX-2)/prostaglandin E2 (PGE2) and endothelial junctional proteins. bEnd.3 cells were exposed to DEHP and subsequently subjected to oxygen-glucose deprivation (OGD). Cell viability was analyzed using 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) proliferation assay. The effect of DEHP/OGD/reoxygenation (R) was evaluated by assessing the levels of NO, reactive oxygen species (ROS), and PGE2. The expression of COX-2, cleaved caspase-3, cleaved PARP, inducible nitric oxide synthase (iNOS), and the endothelial tight junction proteins claudin-5 and ZO-1 was evaluated using quantitative polymerase chain reaction and western blotting. OGD/R decreased cell viability, and DEHP exposure before OGD/R further aggravated cell viability. DEHP/OGD/R significantly increased NO, PGE2, and ROS production following OGD/R. In the DEHP/OGD/R group, iNOS, COX-2, cleaved caspase-3, and cleaved PARP expression increased, and claudin-5 and ZO-1 levels decreased compared with those in the OGD/R group. E-Cadherin expression decreased significantly after DEHP/OGD/R exposure compared with that after OGD/R; this decrease in expression was recovered by treatment with the COX-2 inhibitor indomethacin and antioxidant N-acetylcysteine. Exposure to DEHP exacerbated hypoxia-reoxygenation injury. The enhanced damage upon DEHP exposure was associated with increased oxidative stress and COX-2 expression, leading to E-cadherin downregulation and increased apoptosis.
Collapse
Affiliation(s)
- Jin Hee Kim
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jae Hoon Lee
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Zhengyu Nan
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Ja Woo Choi
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jong Wook Song
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| |
Collapse
|
2
|
Ma W, Zhao X, Wang Q, Wu X, Yang T, Chen Y, Zhu Y, Wang X. SCM-198 ameliorates the quality of postovulatory and maternally aged oocytes by reducing oxidative stress. J Ovarian Res 2024; 17:178. [PMID: 39217393 PMCID: PMC11365136 DOI: 10.1186/s13048-024-01504-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
Oocyte aging is a key constraint on oocyte quality, leading to fertilization failure and abnormal embryonic development. In addition, it is likely to generate unfavorable assisted reproductive technology (ART) outcomes. SCM-198, a synthetic form of leonurine, was found to rescue the rate of oocyte fragmentation caused by postovulatory aging. Therefore, the aim of this study was to conduct a more in-depth investigation of SCM-198 by exploring its relationship with aged oocytes after ovulation or maternal aging and clarifying whether it affects cell quality. The results indicate that, compared to the postovulatory aged group, the 50 µM SCM-198 group significantly improved sperm-egg binding and increased fertilization of aged oocytes, restoring the spindle apparatus/chromosome structure, cortical granule distribution, and ovastacin and Juno protein distribution. The 50 µM SCM-198 group showed significantly normal mitochondrial distribution, low levels of reactive oxygen species (ROS), and a small quantity of early oocyte apoptosis compared to the postovulatory aged group. Above all, in vivo supplementation with SCM-198 effectively eliminated excess ROS and reduced the spindle/chromosome structural defects in aged mouse oocytes. In summary, these findings indicate that SCM-198 inhibits excessive oxidative stress in oocytes and alters oocyte quality both in vitro and in vivo.
Collapse
Affiliation(s)
- Wei Ma
- Center for Reproductive Medicine, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, China
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, 226019, China
| | - Xi Zhao
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, 226019, China
| | - Qingxin Wang
- Center for Reproductive Medicine, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, China
| | - Xue Wu
- Center for Reproductive Medicine, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, China
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, 226019, China
| | - Tingting Yang
- Center for Reproductive Medicine, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, China
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, 226019, China
| | - Yuqi Chen
- Center for Reproductive Medicine, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, China
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, 226019, China
| | - Yizhun Zhu
- State Key Laboratory of Quality Research in Chinese Medicine, School of Pharmacy, Macau University of Science and Technology, Macau, SAR, China.
| | - Xia Wang
- Center for Reproductive Medicine, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, China.
| |
Collapse
|
3
|
Yang J, Yu B, Zheng J. Natural herbal extract roles and mechanisms in treating cerebral ischemia: A systematic review. Front Pharmacol 2024; 15:1424146. [PMID: 39156109 PMCID: PMC11327066 DOI: 10.3389/fphar.2024.1424146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 07/03/2024] [Indexed: 08/20/2024] Open
Abstract
Background Stroke has been the focus of medical research due to its serious consequences and sequelae. Among the tens of millions of new stroke patients every year, cerebral ischemia patients account for the vast majority. While cerebral ischemia drug research and development is still ongoing, most drugs are terminated at preclinical stages due to their unacceptable toxic side effects. In recent years, natural herbs have received considerable attention in the pharmaceutical research and development field due to their low toxicity levels. Numerous studies have shown that natural herbs exert actions that cannot be ignored when treating cerebral ischemia. Methods We reviewed and summarized the therapeutic effects and mechanisms of different natural herbal extracts on cerebral ischemia to promote their application in this field. We used keywords such as "natural herbal extract," "herbal medicine," "Chinese herbal medicine" and "cerebral ischemia" to comprehensively search PubMed, ScienceDirect, ScienceNet, CNKI, and Wanfang databases, after which we conducted a detailed screening and review strategy. Results We included 120 high-quality studies up to 10 January 2024. Natural herbal extracts had significant roles in cerebral ischemia treatments via several molecular mechanisms, such as improving regional blood flow disorders, protecting the blood-brain barrier, and inhibiting neuronal apoptosis, oxidative stress and inflammatory responses. Conclusion Natural herbal extracts are represented by low toxicity and high curative effects, and will become indispensable therapeutic options in the cerebral ischemia treatment field.
Collapse
Affiliation(s)
| | | | - Jian Zheng
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
4
|
Wang G, Li Z, Lin P, Zhang H, Wang Y, Zhang T, Wang H, Li H, Lin L, Zhao Y, Jia L, Chen Y, Ji H, Zhao W, Fu Z, Zhong Z. Knockdown of Smox protects the integrity of the blood-brain barrier through antioxidant effect and Nrf2 pathway activation in stroke. Int Immunopharmacol 2024; 126:111183. [PMID: 37984250 DOI: 10.1016/j.intimp.2023.111183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 10/23/2023] [Accepted: 11/01/2023] [Indexed: 11/22/2023]
Abstract
Once an ischemic stroke occurs, reactive oxygen species (ROS) and oxidative stress degrade the tight connections between cerebral endothelial cells resulting in their damage. The expression of antioxidant genes may be enhanced, and ROS formation may be reduced following Nrf2 activation, which is associated with protection against ischemic stroke. Overexpression of spermine oxidase (Smox) in the neocortex led to increased H2O2 production. However, how Smox impacts the regulation of the blood-brain barrier (BBB) through antioxidants has not been examined yet. We conducted experiments both in the cell level and in the transient middle cerebral artery occlusion (tMCAO) model to evaluate the effect of Smox siRNA lentivirus (si-Smox) knockdown on BBB protection against ischemic stroke. Mice treated with si-Smox showed remarkably decreased BBB breakdown and reduced endothelial inflammation following stroke. The treatment with si-Smox significantly elevated the Bcl-2 to Bax ratio and decreased the production of cleaved caspase-3 in the tMCAO model. Further investigation revealed that the neuroprotective effect was the result of the antioxidant properties of si-Smox, which reduced oxidative stress and enhanced CD31+ cells in the peri-infarct cortical areas. Of significance, si-Smox activated Nrf2 in both bEnd.3 cells and tMCAO animals, and blocking Nrf2 with brusatol diminished the protective effects of si-Smox. The study findings suggest that si-Smox exerts neuroprotective effects and promotes angiogenesis by activating the Nrf2 pathway, thus decreasing oxidative stress and apoptosis caused by tMCAO. As a result, si-Smox may hold potential as a therapeutic candidate for preserving BBB integrity while treating ischemic stroke.
Collapse
Affiliation(s)
- Guangtian Wang
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Zhihui Li
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Peng Lin
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Huishu Zhang
- Teaching Center of Biotechnology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Yanyan Wang
- Teaching Center of Morphology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Tongshuai Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Hui Wang
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Heming Li
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Lexun Lin
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Yuehui Zhao
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Lina Jia
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Yang Chen
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Hong Ji
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Wenran Zhao
- Department of Cell Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Zhongqiu Fu
- Department of Neonatology, Zhuhai Women and Children's Hospital, Zhuhai, Guangdong 519000, China.
| | - Zhaohua Zhong
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China.
| |
Collapse
|
5
|
Kuang X, Chen S, Ye Q. The Role of Histone Deacetylases in NLRP3 Inflammasomesmediated Epilepsy. Curr Mol Med 2024; 24:980-1003. [PMID: 37519210 DOI: 10.2174/1566524023666230731095431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/08/2023] [Accepted: 06/19/2023] [Indexed: 08/01/2023]
Abstract
Epilepsy is one of the most common brain disorders that not only causes death worldwide, but also affects the daily lives of patients. Previous studies have revealed that inflammation plays an important role in the pathophysiology of epilepsy. Activation of inflammasomes can promote neuroinflammation by boosting the maturation of caspase-1 and the secretion of various inflammatory effectors, including chemokines, interleukins, and tumor necrosis factors. With the in-depth research on the mechanism of inflammasomes in the development of epilepsy, it has been discovered that NLRP3 inflammasomes may induce epilepsy by mediating neuronal inflammatory injury, neuronal loss and blood-brain barrier dysfunction. Therefore, blocking the activation of the NLRP3 inflammasomes may be a new epilepsy treatment strategy. However, the drugs that specifically block NLRP3 inflammasomes assembly has not been approved for clinical use. In this review, the mechanism of how HDACs, an inflammatory regulator, regulates the activation of NLRP3 inflammasome is summarized. It helps to explore the mechanism of the HDAC inhibitors inhibiting brain inflammatory damage so as to provide a potential therapeutic strategy for controlling the development of epilepsy.
Collapse
Affiliation(s)
- Xi Kuang
- Hainan Health Vocational College,Haikou, Hainan, 570311, China
| | - Shuang Chen
- Hubei Provincial Hospital of Integrated Chinese and Western Medicine, 430022, Hubei, China
| | - Qingmei Ye
- Hainan General Hospital & Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan, China
| |
Collapse
|
6
|
Shang J, Li W, Zhang H, Wang W, Liu N, Gao D, Wang F, Yan X, Gao C, Sun R, Zhang H, Ma K, Shao F, Zhang J. C-kit controls blood-brain barrier permeability by regulating caveolae-mediated transcytosis after chronic cerebral hypoperfusion. Biomed Pharmacother 2024; 170:115778. [PMID: 38141279 DOI: 10.1016/j.biopha.2023.115778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/13/2023] [Accepted: 10/20/2023] [Indexed: 12/25/2023] Open
Abstract
Blood-brain barrier (BBB) dysfunction plays a pivotal role in the pathology of chronic cerebral hypoperfusion (CCH)-related neurodegenerative diseases. Continuous endothelial cells (EC) that line the blood vessels of the brain are important components of the BBB to strictly control the flow of substances and maintain the homeostatic environment of the brain. However, the molecular mechanisms from the perspective of EC-induced BBB dysfunction after CCH are largely unknown. In this study, the BBB function was assessed using immunostaining and transmission electron microscopy. The EC dysfunction profile was screened by using EC enrichment followed by RNA sequencing. After identified the key EC dysfunction factor, C-kit, we used the C-kit inhibition drug (imatinib) and C-kit down-regulation method (AAV-BR1-C-kit shRNA) to verify the role of C-kit on BBB integrity and EC transcytosis after CCH. Furthermore, we also activated C-kit with stem cell factor (SCF) to observe the effects of C-kit on BBB following CCH. We explored that macromolecular proteins entered the brain mainly through EC transcytosis after CCH and caused neuronal loss. Additionally, we identified receptor tyrosine kinase C-kit as a key EC dysfunction molecule. Furthermore, the pharmacological inhibition of C-kit with imatinib counteracted BBB leakage by reducing caveolae-mediated transcytosis. Moreover, treatment with AAV-BR1-C-kit shRNA, which targets brain EC to inhibit C-kit expression, also ameliorated BBB leakage by reducing caveolae-mediated transcytosis. Furthermore, the SCF increased the permeability of the BBB by actively increasing caveolae-mediated transcytosis. This study provides evidence that C-kit is a key BBB permeability regulator through caveolae-mediated transcytosis in EC after CCH.
Collapse
Affiliation(s)
- Junkui Shang
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Wei Li
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Huiwen Zhang
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Wan Wang
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Ning Liu
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Dandan Gao
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430072, China
| | - Fengyu Wang
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Xi Yan
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Chenhao Gao
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Ruihua Sun
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Haohan Zhang
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Kai Ma
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Fengmin Shao
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China; Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan 450003, China.
| | - Jiewen Zhang
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China.
| |
Collapse
|
7
|
Wang W, Wang Y, Pei H, Li M, Zhu A, Du R, Peng GJ. The mechanism of simultaneous intake of Jujuboside A and B in the regulation of sleep at the hypothalamic level. Aging (Albany NY) 2023; 15:9426-9437. [PMID: 37679031 PMCID: PMC10564420 DOI: 10.18632/aging.204995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 07/06/2023] [Indexed: 09/09/2023]
Abstract
To study the effect of co-administration of Jujuboside A and B (Ju A+B) on sleep, healthy KM mice were given different doses of Ju A+B with a behavioral evaluation of sleep state. Serum levels of key neurotransmitters (5HT, DA, and NE) were measured. The hypothalamus of KM mice was analyzed for differential protein expression using TMT quantitative proteomics, and differential expression protein (DEP) bioinformatics analysis was used to explore potential mechanisms. The result shows that Ju A+B affects sleep by expressing the protein in the hypothalamus. Compared with the control group, the test group showed 10 up-regulated and 139 down-regulated DEPs. The key DEPs were found at the tight junction. Western blot showed reliable alteration of the key DEPs in proteomics. The result of interaction network analysis attributed the potential of Jujuboside to the changes in blood-brain barrier, which provided basic and theoretical data for the efficacy evaluation and mechanism of Jujuboside.
Collapse
Affiliation(s)
- Wei Wang
- College of Humanities and College of Home Economics, Jilin Agricultural University, Changchun 130118, China
| | - Yi Wang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China
| | - Hongyan Pei
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Mingming Li
- College of Humanities and College of Home Economics, Jilin Agricultural University, Changchun 130118, China
| | - Aozhe Zhu
- College of Humanities and College of Home Economics, Jilin Agricultural University, Changchun 130118, China
| | - Rui Du
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Gao Jun Peng
- New Vita Bioengineering Tianjin Co., Ltd, Tianjin 301726, China
| |
Collapse
|
8
|
Chen S, Wang N, Xiong S, Xia X. The correlation between primary open-angle glaucoma (POAG) and gut microbiota: a pilot study towards predictive, preventive, and personalized medicine. EPMA J 2023; 14:539-552. [PMID: 37605653 PMCID: PMC10439875 DOI: 10.1007/s13167-023-00336-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 07/29/2023] [Indexed: 08/23/2023]
Abstract
Background Glaucoma is the leading cause of irreversible blindness worldwide. Emerged evidence has shown that glaucoma is considered an immune system related disorder. The gut is the largest immune organ in the human body and the gut microbiota (GM) plays an irreversible role in maintaining immune homeostasis. But, how the GM influences glaucoma remains unrevealed. This study aimed at investigating the key molecules/pathways mediating the GM and the glaucoma to provide new biomarkers for future predictive, preventive, and personalized medicine. Methods Datasets from the primary open-angle glaucoma (POAG) patients (GSE138125) and datasets for target genes of GM/GM metabolites were downloaded from a public database. For GSE138125, the differentially expressed genes (DEGs) between healthy and POAG samples were identified. And the online Venn diagram tool was used to obtain the DEGs from POAG related to GM. After which GM-related DEGs were analyzed by correlation analysis, pathway enrichment analysis, and protein-protein interaction (PPI) network analysis. Human trabecular meshwork cells were used for validation, and the mRNA level of hub genes was verified by quantitative real-time polymerase chain reaction (RT-qPCR) in the in vitro glaucoma model. Results A total of 16 GM-related DEGs in POAG were identified from the above 2 datasets (9 upregulated genes and 7 downregulated genes). Pathway enrichment analysis indicated that these genes are mostly enriched in immune regulation especially macrophages-related pathways. Then 6 hub genes were identified by PPI network analysis and construction of key modules. Finally, RT-qPCR confirmed that the expression of the hub genes in the in vitro glaucoma model was consistent with the results of bioinformatics analysis of the mRNA chip. Conclusion This bioinformatic study elucidates NFKB1, IL18, KITLG, TLR9, FKBP2, and HDAC4 as hub genes for POAG and GM regulation. Immune response modulated by macrophages plays an important role in POAG and may be potential targets for future predictive, preventive, and personalized diagnosis and treatment. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-023-00336-2.
Collapse
Affiliation(s)
- Si Chen
- Eye Center of Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Nan Wang
- Eye Center of Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Siqi Xiong
- Eye Center of Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Xiaobo Xia
- Eye Center of Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan China
| |
Collapse
|
9
|
Tang Z, Meng S, Song Z, Yang X, Li X, Guo H, Du M, Chen J, Zhu YZ, Wang X. Neutrophil membrane fusogenic nanoliposomal leonurine for targeted ischemic stroke therapy via remodeling cerebral niche and restoring blood-brain barrier integrity. Mater Today Bio 2023; 20:100674. [PMID: 37273794 PMCID: PMC10238753 DOI: 10.1016/j.mtbio.2023.100674] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/19/2023] [Accepted: 05/18/2023] [Indexed: 06/06/2023] Open
Abstract
Ischemic stroke (IS) constitutes the leading cause of global morbidity and mortality. Neuroprotectants are essential to ameliorate the clinical prognosis, but their therapeutic outcomes are tremendously compromised by insufficient delivery to the ischemic lesion and intricate pathogenesis associated with neuronal damage, oxidative stress, inflammation responses, blood-brain barrier (BBB) dysfunction, etc. Herein, a biomimetic nanosystem (Leo@NM-Lipo) composed of neutrophil membrane-fused nanoliposomal leonurine (Leo) is constructed, which can not only efficiently penetrate and repair the disrupted BBB but also robustly remodel the harsh cerebral microenvironment to reverse ischemia-reperfusion (I/R) injury. More specifically, the neutrophil membrane inherits the BBB penetrating, infarct core targeting, inflammation neutralization, and immune evasion properties of neutrophils, while Leo, a naturally occurring neuroprotectant, exerts pleiotropic effects to attenuate brain damage. Remarkably, comprehensive investigations disclose the critical factors influencing the targetability and therapeutic performances of biomimetic nanosystems. Leo@NM-Lipo with a low membrane protein-to-lipid ratio of 1:10 efficiently targets the ischemic lesion and rescues the injured brain by alleviating neuronal apoptosis, oxidative stress, neuroinflammation, and restoring BBB integrity in transient middle cerebral artery occlusion (tMCAO) rats. Taken together, our study provides a neutrophil-mimetic nanoplatform for targeted IS therapy and sheds light on the rational design of biomimetic nanosystems favoring wide medical applications.
Collapse
Affiliation(s)
- Zhuang Tang
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078, China
| | - Shiyu Meng
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078, China
| | - Zhiling Song
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078, China
| | - Xiaoxue Yang
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078, China
| | - Xinzhi Li
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078, China
| | - Hui Guo
- School of Chemical Engineering and Technology, Sun Yat-sen University, Zhuhai 519082, China
| | - Meirong Du
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078, China
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Jun Chen
- School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yi Zhun Zhu
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078, China
| | - Xiaolin Wang
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078, China
| |
Collapse
|
10
|
Zeng Y, Xu Y, Pan Y, Guo H. KLF10 knockdown negatively regulates CTRP3 to improve OGD/R-induced brain microvascular endothelial cell injury and barrier dysfunction through Nrf2/HO-1 signaling pathway. Tissue Cell 2023; 82:102106. [PMID: 37210762 DOI: 10.1016/j.tice.2023.102106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 05/23/2023]
Abstract
Ischemic stroke seriously endangers human health and even death. This study aimed to investigate the role of KLF10/CTRP3 in oxygen-glucose deprivation/reperfusion (OGD/R)-induced brain microvascular endothelial cells injury, as well as the regulatory effects of the Nrf2/HO-1 signaling pathway. OGD/R-induced human microvascular endothelial cells (hBMECs) were used to simulate the model of cerebral ischemia-reperfusion (I/R) injury. The expression of KLF10/CTRP3 in OGD/R-induced hBMECs as well as the transfection efficiency were all detected by RT-qPCR and western blot. The interaction of KLF10 and CTRP3 was confirmed by dual-luciferase reporter assay and chromatin immunoprecipitation (ChIP). The viability, apoptosis and endothelial permeability of OGD/R-induced hBMECs was detected by CCK-8, TUNEL and FITC-Dextran assay kit. The capacity of cell migration was assessed by wound healing assay. The expression of apoptosis related proteins, oxidative stress levels and tight junction proteins was also detected. As a result, the expression of KLF10 was increased in OGD/R-induced hBMECs and downregulation of KLF10 could promote the viability, migration and suppress the apoptosis, oxidative stress and endothelial permeability by downregulating the expression of caspase 3, Bax, cleaved PARP, ROS, MDA, and upregulating the expression of Bcl-2, SOD, GSH-Px, ZO-1, occludin, claudin-5. Nrf2/HO-1 signaling pathway was inhibited in OGD/R-induced hBMECs, which was activated by downregulation of KLF10. KLF10 was demonstrated to be combined with CTRP3 and KLF10 inhibited transcription of CTRP3 in hBMECs. The above changes affected by downregulation of KLF10 could be reversed by the interference with CTRP3. In conclusion, KLF10 knockdown improved OGD/R-induced brain microvascular endothelial cell injury and barrier dysfunction through the activation of Nrf2/HO-1 signaling pathway, which was weakened by the downregulation of CTRP3.
Collapse
Affiliation(s)
- Youchao Zeng
- Department of Neurology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi 56300, Guizhou, China.
| | - Yongsu Xu
- Nursing Department, Affiliated Hospital of Zunyi Medical University, Zunyi 56300, Guizhou, China
| | - Yongjie Pan
- Department of Neurology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi 56300, Guizhou, China
| | - Hong Guo
- Department of Neurology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi 56300, Guizhou, China
| |
Collapse
|
11
|
Aitbaev KA, Murkamilov IT, Fomin VV, Rayimzhanov ZR, Yusupova TF, Yusupov FA. [New data on the pathophysiology of ischemic stroke: epigenetic mechanisms in focus]. Zh Nevrol Psikhiatr Im S S Korsakova 2023; 123:24-29. [PMID: 38148694 DOI: 10.17116/jnevro202312312224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2023]
Abstract
Epigenetics is a branch of molecular biology that studies modifications able to change gene expression without changing the DNA sequence. Epigenetic modulations include DNA methylation, histone modifications, and noncoding RNAs. These heritable and modifiable gene changes can be caused by lifestyle and dietary factors. In recent years, epigenetic changes have been associated with the pathogenesis of a number of diseases, such as diabetes mellitus, obesity, renal pathology and various types of cancer. They were also associated with the pathogenesis of cardiovascular diseases, including ischemic stroke. In this regard, it is important to note that since epigenetic modifications are reversible processes, they can help in the development of new therapeutic approaches to treat human diseases. This mini-review presents the latest data on the influence of epigenetic modifications on the pathogenesis of ischemic stroke obtained both in animal models and in patients.
Collapse
Affiliation(s)
- K A Aitbaev
- Research Institute of Molecular Biology and Medicine, Bishkek, Kyrgyzstan
| | - I T Murkamilov
- Akhunbaev Kyrgyz State Medical Academy, Bishkek, Kyrgyzstan
- Kyrgyz- Russian Slavic University named after the First President of Russia B.N. Yeltsin, Bishkek, Kyrgyzstan
| | - V V Fomin
- Sechenov First Moscow State Medical University, Moscow, Russia
| | - Z R Rayimzhanov
- Burdenko Military Clinical Hospital Ministry of Defense of Russia, Moscow, Russia
| | | | | |
Collapse
|
12
|
Liu A, Hu J, Yeh TS, Wang C, Tang J, Huang X, Chen B, Huangfu L, Yu W, Zhang L. Neuroprotective Strategies for Stroke by Natural Products: Advances and Perspectives. Curr Neuropharmacol 2023; 21:2283-2309. [PMID: 37458258 PMCID: PMC10556387 DOI: 10.2174/1570159x21666230717144752] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 09/09/2023] Open
Abstract
Cerebral ischemic stroke is a disease with high prevalence and incidence. Its management focuses on rapid reperfusion with intravenous thrombolysis and endovascular thrombectomy. Both therapeutic strategies reduce disability, but the therapy time window is short, and the risk of bleeding is high. Natural products (NPs) have played a key role in drug discovery, especially for cancer and infectious diseases. However, they have made little progress in clinical translation and pose challenges to the treatment of stroke. Recently, with the investigation of precise mechanisms in cerebral ischemic stroke and the technological development of NP-based drug discovery, NPs are addressing these challenges and opening up new opportunities in cerebral stroke. Thus, in this review, we first summarize the structure and function of diverse NPs, including flavonoids, phenols, terpenes, lactones, quinones, alkaloids, and glycosides. Then we propose the comprehensive neuroprotective mechanism of NPs in cerebral ischemic stroke, which involves complex cascade processes of oxidative stress, mitochondrial damage, apoptosis or ferroptosis-related cell death, inflammatory response, and disruption of the blood-brain barrier (BBB). Overall, we stress the neuroprotective effect of NPs and their mechanism on cerebral ischemic stroke for a better understanding of the advances and perspective in NPs application that may provide a rationale for the development of innovative therapeutic regimens in ischemic stroke.
Collapse
Affiliation(s)
- Aifen Liu
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Jingyan Hu
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Tzu-Shao Yeh
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong 226019, China
| | - Chengniu Wang
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Jilong Tang
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Xiaohong Huang
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Bin Chen
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Liexiang Huangfu
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Weili Yu
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Lei Zhang
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
- Department of Pharmaceutical Botany, School of Pharmacy, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
13
|
Li X, Wang Y, Zhou X, Wang H, Xu J. Electroacupuncture Pretreatment Alleviates Cerebral Ischemia-reperfusion Injury by Down-regulating Mir-155-5p. Curr Neurovasc Res 2023; 20:480-492. [PMID: 37642006 DOI: 10.2174/1567202620666230828092916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/16/2023] [Accepted: 08/20/2023] [Indexed: 08/31/2023]
Abstract
BACKGROUND Increasing evidence shows that electroacupuncture pretreatment (EP) plays a crucial role in cerebral ischemia-reperfusion (I/R) injury, and cerebral I/R injury is the most serious complication of ischemic stroke treatment. The role of miR-155-5p in cerebral I/R injury has been studied, but the regulation of EP on miR-155-5p has not been reported. METHODS The middle cerebral artery occlusion (MCAO) mice were used to investigate the role of EP in cerebral I/R injury. Longa and modified neurological severity scores (mNSS) were used to evaluate neurological impairment. HE staining and TUNEL staining were used to evaluate brain injury. The expressions of miR-155-5p, Yin Yang 1 (YY1) and p53 were detected by qRT-PCR. The expressions of related proteins were detected by western blot. The binding of YY1 to miR- 155-5p was verified by dual-luciferase reporter assay and chromatin immunoprecipitation (ChIP) assay. Mice brain microvascular endothelial cells (BMECs) were isolated and cultured for in vitro experiments. Oxygen-glucose deprivation/reoxygenation (OGD/R) was used to verify the role of YY1, p53 and miR-155-5p in cerebral I/R injury in vitro. RESULTS MCAO modeling induced brain injury, apoptosis, and increased levels of miR-155-5p, YY1, and p53. EP markedly alleviated brain injury and reduced levels of miR-155-5p, p53, and YY1. miR-155 agomir markedly increased the expression of miR-155-5p and p53. miR-155 antagomir decreased the levels of miR-155-5p and p53. Dual-luciferase reporter and ChIP assay verified that YY1 regulated miR-155-5p expression. YY1 shNRA greatly decreased miR-155-5p and p53. Inhibition of p53 decreased miR-155-5p expression. Both miR-155-5p inhibitor and YY1 shRNA promoted proliferation, inhibited apoptosis, and decreased levels of ICAM-1 and Eselectin of OGD/R-treated BMECs. Inhibition of p53 strengthened the effect of miR-155-5p inhibitor and YY1 shNRA on BMECs. CONCLUSION Electroacupuncture pretreatment alleviates cerebral ischemia-reperfusion injury by regulating the YY1/p53/miR-155-5p axis.
Collapse
Affiliation(s)
- Xuejing Li
- Department of Rehabilitation Medicine, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huai'an, China
| | - Ying Wang
- Department of Rehabilitation Medicine, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huai'an, China
| | - Xiang Zhou
- Department of Rehabilitation Medicine, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huai'an, China
| | - Hui Wang
- Department of Rehabilitation Medicine, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huai'an, China
| | - Jiang Xu
- Department of Rehabilitation Medicine, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huai'an, China
| |
Collapse
|
14
|
袁 巧, 谢 丽, 陈 朝. [Astragalus polysaccharide protects against blood-brain barrier damage in MCAO rats by inhibiting P2X7R channel]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2022; 42:1705-1711. [PMID: 36504064 PMCID: PMC9742771 DOI: 10.12122/j.issn.1673-4254.2022.11.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To investigate the protective effect of astragalus polysaccharide (APS) against blood-brain barrier in a rat model of middle cerebral artery occlusion (MCAO) and the role of P2X7R channel in the protective mechanism. METHODS In rat microglial cell models of oxygen and glucose deprivation (OGD) or ATP treatment, the formation of blood-brain barrier in vitro was assessed using the leak test, and the effect of APS on the permeability of the blood-brain barrier was determined using LC-MS. In 12 SD rats, MCAO model was established followed by treatment with intraperitoneal injection of normal saline (n= 6) or APS (45 mg/kg, n=6) for 3 consecutive days, with another 6 rats without MCAO receiving saline injections as the control group. The permeability of the blood-brain barrier of the rats was evaluated by determining Evans blue (EB) extravasation, and ATP content in the brain tissue was detected using ELISA; the expression levels of matrix metalloproteinase-9 (MMP-9) and P2X7R in the brain tissue were detected with Western blot. RESULTS In the in vitro cell model of OGD or ATP treatment, APS treatment obviously promoted the repair of blood-brain barrier integrity. In the rat models, the EB content in the brain tissue and the blood-brain barrier permeability increased significantly in MCAO+saline group and MCAO+APS group as compared with those in the control group (P < 0.01). Compared with saline treatment, APS treatment significantly decreased EB content in the brain tissue and improved the blood-brain barrier permeability in the MCAO rats (P < 0.05). MCAO caused a significant reduction of ATP content and obviously increased the expression levels of MMP-9 and P2X7R in the brain tissue of the rats (P < 0.01), and these changes were significantly alleviated after APS treatment (P < 0.01 or 0.05). CONCLUSION APS can protect the brain tissue of MCAO rats by stabilizing the internal environment, down-regulating the expression of MMP-9 and improving the permeability of blood-brain barrier under cerebral ischemia and hypoxia, and its mechanism may involve the inhibition of P2X7R channel.
Collapse
Affiliation(s)
- 巧 袁
- 广州中医药大学,广东 广州 510800Guangzhou University of Chinese Medicine, Guangzhou 510800, China
| | - 丽英 谢
- 广州中医药大学附属广州中西医结合医院脑病科,广东 广州 510800Department of Encephalopathy, Guangzhou Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou 510800, China
| | - 朝俊 陈
- 广州中医药大学附属广州中西医结合医院脑病科,广东 广州 510800Department of Encephalopathy, Guangzhou Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou 510800, China
| |
Collapse
|
15
|
Li Z, Chen K, Rose P, Zhu YZ. Natural products in drug discovery and development: Synthesis and medicinal perspective of leonurine. Front Chem 2022; 10:1036329. [PMID: 36324522 PMCID: PMC9618625 DOI: 10.3389/fchem.2022.1036329] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 10/05/2022] [Indexed: 12/03/2022] Open
Abstract
Natural products, those molecules derived from nature, have been used by humans for thousands of years to treat ailments and diseases. More recently, these compounds have inspired chemists to use natural products as structural templates in the development of new drug molecules. One such compound is leonurine, a molecule isolated and characterized in the tissues of Herb leonuri. This molecule has received attention from scientists in recent years due to its potent anti-oxidant, anti-apoptotic, and anti-inflammatory properties. More recently researchers have shown leonurine to be useful in the treatment of cardiovascular and nervous system diseases. Like other natural products such as paclitaxel and artemisinin, the historical development of leonurine as a therapeutic is very interesting. Therefore, this review provided an overview of natural product discovery, through to the development of a potential new drug. Content will summarize known plant sources, the pathway used in the synthesis of leonurine, and descriptions of leonurine’s pharmacological properties in mammalian systems.
Collapse
Affiliation(s)
- Zhaoyi Li
- State Key Laboratory of Quality Research in Chinese Medicine, School of Pharmacy, Macau University of Science and Technology, Taipa, Macau, China
| | - Keyuan Chen
- State Key Laboratory of Quality Research in Chinese Medicine, School of Pharmacy, Macau University of Science and Technology, Taipa, Macau, China
| | - Peter Rose
- School of Biosciences, University of Nottingham, Nottingham, United Kingdom
| | - Yi Zhun Zhu
- State Key Laboratory of Quality Research in Chinese Medicine, School of Pharmacy, Macau University of Science and Technology, Taipa, Macau, China
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
- *Correspondence: Yi Zhun Zhu,
| |
Collapse
|
16
|
Leonurine Reduces Oxidative Stress and Provides Neuroprotection against Ischemic Injury via Modulating Oxidative and NO/NOS Pathway. Int J Mol Sci 2022; 23:ijms231710188. [PMID: 36077582 PMCID: PMC9456230 DOI: 10.3390/ijms231710188] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
Leonurine (Leo) has been found to have neuroprotective effects against cerebral ischemic injury. However, the exact molecular mechanism underlying its neuroprotective ability remains unclear. The aim of the present study was to investigate whether Leo could provide protection through the nitric oxide (NO)/nitric oxide synthase (NOS) pathway. We firstly explored the effects of NO/NOS signaling on oxidative stress and apoptosis in in vivo and in vitro models of cerebral ischemia. Further, we evaluated the protective effects of Leo against oxygen and glucose deprivation (OGD)-induced oxidative stress and apoptosis in PC12 cells. We found that the rats showed anxiety-like behavior, and the morphology and number of neurons were changed in a model of photochemically induced cerebral ischemia. Both in vivo and in vitro results show that the activity of superoxide dismutase (SOD) and glutathione (GSH) contents were decreased after ischemia, and reactive oxygen species (ROS) and malondialdehyde (MDA) levels were increased, indicating that cerebral ischemia induced oxidative stress and neuronal damage. Moreover, the contents of NO, total NOS, constitutive NOS (cNOS) and inducible NOS (iNOS) were increased after ischemia in rat and PC12 cells. Treatment with L-nitroarginine methyl ester (L-NAME), a nonselective NOS inhibitor, could reverse the change in NO/NOS expression and abolish these detrimental effects of ischemia. Leo treatment decreased ROS and MDA levels and increased the activity of SOD and GSH contents in PC12 cells exposed to OGD. Furthermore, Leo reduced NO/NOS production and cell apoptosis, decreased Bax expression and increased Bcl-2 levels in OGD-treated PC12 cells. All the data suggest that Leo protected against oxidative stress and neuronal apoptosis in cerebral ischemia by inhibiting the NO/NOS system. Our findings indicate that Leo could be a potential agent for the intervention of ischemic stroke and highlighted the NO/NOS-mediated oxidative stress signaling.
Collapse
|
17
|
Insights into the Explicit Protective Activity of Herbals in Management of Neurodegenerative and Cerebrovascular Disorders. Molecules 2022; 27:molecules27154970. [PMID: 35956919 PMCID: PMC9370592 DOI: 10.3390/molecules27154970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 07/29/2022] [Accepted: 08/01/2022] [Indexed: 11/16/2022] Open
Abstract
The longstanding progressive neurodegenerative conditions of the central nervous system arise mainly due to deterioration, degradation and eventual neuronal cell loss. As an individual ages, the irreversible neurodegenerative disorders associated with aging also begin to develop, and these have become exceedingly prominent and pose a significant burden mentally, socially and economically on both the individual and their family. These disorders express several symptoms, such as tremors, dystonia, loss of cognitive functions, impairment of motor activity leading to immobility, loss of memory and many more which worsen with time. The treatment employed in management of these debilitating neurodegenerative disorders, such as Parkinson’s disease (which mainly involves the loss of dopaminergic neurons in the nigrostriatal region), Alzheimer’s disease (which arises due to accumulation of Tau proteins causing diffusive atrophy in the brain), Huntington’s disease (which involves damage of striatal and spinal neurons, etc.), have several adverse effects, leading to exploration of several lead targets and molecules existing in herbal drugs. The current review highlights the mechanistic role of natural products in the treatment of several neurodegenerative and cerebrovascular diseases such as Parkinson’s disease, Alzheimer’s disease, ischemic stroke and depression.
Collapse
|
18
|
Gallego-Fabrega C, Muiño E, Cárcel-Márquez J, Llucià-Carol L, Lledós M, Martín-Campos JM, Cullell N, Fernández-Cadenas I. Genome-Wide Studies in Ischaemic Stroke: Are Genetics Only Useful for Finding Genes? Int J Mol Sci 2022; 23:6840. [PMID: 35743317 PMCID: PMC9224543 DOI: 10.3390/ijms23126840] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/31/2022] [Accepted: 06/01/2022] [Indexed: 02/07/2023] Open
Abstract
Ischaemic stroke is a complex disease with some degree of heritability. This means that heritability factors, such as genetics, could be risk factors for ischaemic stroke. The era of genome-wide studies has revealed some of these heritable risk factors, although the data generated by these studies may also be useful in other disciplines. Analysis of these data can be used to understand the biological mechanisms associated with stroke risk and stroke outcome, to determine the causality between stroke and other diseases without the need for expensive clinical trials, or to find potential drug targets with higher success rates than other strategies. In this review we will discuss several of the most relevant studies regarding the genetics of ischaemic stroke and the potential use of the data generated.
Collapse
Affiliation(s)
- Cristina Gallego-Fabrega
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (C.G.-F.); (E.M.); (J.C.-M.); (L.L.-C.); (M.L.); (J.M.M.-C.); (N.C.)
| | - Elena Muiño
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (C.G.-F.); (E.M.); (J.C.-M.); (L.L.-C.); (M.L.); (J.M.M.-C.); (N.C.)
| | - Jara Cárcel-Márquez
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (C.G.-F.); (E.M.); (J.C.-M.); (L.L.-C.); (M.L.); (J.M.M.-C.); (N.C.)
| | - Laia Llucià-Carol
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (C.G.-F.); (E.M.); (J.C.-M.); (L.L.-C.); (M.L.); (J.M.M.-C.); (N.C.)
- Institute for Biomedical Research of Barcelona (IIBB), National Spanish Research Council (CSIC), 08036 Barcelona, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Miquel Lledós
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (C.G.-F.); (E.M.); (J.C.-M.); (L.L.-C.); (M.L.); (J.M.M.-C.); (N.C.)
| | - Jesús M. Martín-Campos
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (C.G.-F.); (E.M.); (J.C.-M.); (L.L.-C.); (M.L.); (J.M.M.-C.); (N.C.)
| | - Natalia Cullell
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (C.G.-F.); (E.M.); (J.C.-M.); (L.L.-C.); (M.L.); (J.M.M.-C.); (N.C.)
| | - Israel Fernández-Cadenas
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (C.G.-F.); (E.M.); (J.C.-M.); (L.L.-C.); (M.L.); (J.M.M.-C.); (N.C.)
- Stroke Pharmacogenomics and Genetics Group, Fundació MútuaTerrassa per la Docència i la Recerca, 08221 Terrassa, Spain
| |
Collapse
|
19
|
Stroke and Etiopathogenesis: What Is Known? Genes (Basel) 2022; 13:genes13060978. [PMID: 35741740 PMCID: PMC9222702 DOI: 10.3390/genes13060978] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/13/2022] [Accepted: 05/17/2022] [Indexed: 02/05/2023] Open
Abstract
Background: A substantial portion of stroke risk remains unexplained, and a contribution from genetic factors is supported by recent findings. In most cases, genetic risk factors contribute to stroke risk as part of a multifactorial predisposition. A major challenge in identifying the genetic determinants of stroke is fully understanding the complexity of the phenotype. Aims: Our narrative review is needed to improve our understanding of the biological pathways underlying the disease and, through this understanding, to accelerate the identification of new drug targets. Methods: We report, the research in the literature until February 2022 in this narrative review. The keywords are stroke, causes, etiopathogenesis, genetic, epigenetic, ischemic stroke. Results: While better risk prediction also remains a long-term goal, its implementation is still complex given the small effect-size of genetic risk variants. Some authors encourage the use of stroke genetic panels for stroke risk assessment and further stroke research. In addition, new biomarkers for the genetic causes of stroke and new targets for gene therapy are on the horizon. Conclusion: We summarize the latest evidence and perspectives of ischemic stroke genetics that may be of interest to the physician and useful for day-to-day clinical work in terms of both prevention and treatment of ischemic stroke.
Collapse
|
20
|
Wang L, Liu Y, Zhang X, Ye Y, Xiong X, Zhang S, Gu L, Jian Z, Wang H. Endoplasmic Reticulum Stress and the Unfolded Protein Response in Cerebral Ischemia/Reperfusion Injury. Front Cell Neurosci 2022; 16:864426. [PMID: 35602556 PMCID: PMC9114642 DOI: 10.3389/fncel.2022.864426] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/07/2022] [Indexed: 12/15/2022] Open
Abstract
Ischemic stroke is an acute cerebrovascular disease characterized by sudden interruption of blood flow in a certain part of the brain, leading to serious disability and death. At present, treatment methods for ischemic stroke are limited to thrombolysis or thrombus removal, but the treatment window is very narrow. However, recovery of cerebral blood circulation further causes cerebral ischemia/reperfusion injury (CIRI). The endoplasmic reticulum (ER) plays an important role in protein secretion, membrane protein folding, transportation, and maintenance of intracellular calcium homeostasis. Endoplasmic reticulum stress (ERS) plays a crucial role in cerebral ischemia pathophysiology. Mild ERS helps improve cell tolerance and restore cell homeostasis; however, excessive or long-term ERS causes apoptotic pathway activation. Specifically, the protein kinase R-like endoplasmic reticulum kinase (PERK), activating transcription factor 6 (ATF6), and inositol-requiring enzyme 1 (IRE1) pathways are significantly activated following initiation of the unfolded protein response (UPR). CIRI-induced apoptosis leads to nerve cell death, which ultimately aggravates neurological deficits in patients. Therefore, it is necessary and important to comprehensively explore the mechanism of ERS in CIRI to identify methods for preserving brain cells and neuronal function after ischemia.
Collapse
Affiliation(s)
- Lei Wang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yan Liu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xu Zhang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yingze Ye
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shudi Zhang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhihong Jian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
- Zhihong Jian,
| | - Hongfa Wang
- Rehabilitation Medicine Center, Department of Anesthesiology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- *Correspondence: Hongfa Wang,
| |
Collapse
|
21
|
Oxidative Injury in Ischemic Stroke: A Focus on NADPH Oxidase 4. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1148874. [PMID: 35154560 PMCID: PMC8831073 DOI: 10.1155/2022/1148874] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 01/08/2022] [Accepted: 01/17/2022] [Indexed: 02/06/2023]
Abstract
Ischemic stroke is a leading cause of disability and mortality worldwide. Thus, it is urgent to explore its pathophysiological mechanisms and find new therapeutic strategies for its successful treatment. The relationship between oxidative stress and ischemic stroke is increasingly appreciated and attracting considerable attention. ROS serves as a source of oxidative stress. It is a byproduct of mitochondrial metabolism but primarily a functional product of NADPH oxidases (NOX) family members. Nicotinamide adenine dinucleotide phosphate oxidase 4 (NOX4) is most closely related to the formation of ROS during ischemic stroke. Its expression is significantly upregulated after cerebral ischemia, making it a promising target for treating ischemic stroke. Several drugs targeting NOX4, such as SCM-198, Iso, G-Rb1, betulinic acid, and electroacupuncture, have shown efficacy as treatments of ischemic stroke. MTfp-NOX4 POC provides a novel insight for the treatment of stroke. Combinations of these therapies also provide new approaches for the therapy of ischemic stroke. In this review, we summarize the subcellular location, expression, and pathophysiological mechanisms of NOX4 in the occurrence and development of ischemic stroke. We also discuss the therapeutic strategies and related regulatory mechanisms for treating ischemic stroke. We further comment on the shortcomings of current NOX4-targeted therapy studies and the direction for improvement.
Collapse
|
22
|
Han D, Li F, Zhang H, Ji C, Shu Q, Wang C, Ni H, Zhu Y, Wang S. Mesencephalic astrocyte-derived neurotrophic factor restores blood-brain barrier integrity of aged mice after ischaemic stroke/reperfusion through anti-inflammation via TLR4/MyD88/NF-κB pathway. J Drug Target 2021; 30:430-441. [PMID: 34747270 DOI: 10.1080/1061186x.2021.2003803] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Ischaemic stroke remains a leading cause of disability and mortality worldwide and ageing-associated inflammation for the aged patients specifically leads to worse post-stroke blood-brain barrier (BBB) disruption than young subjects. Accordingly, suppression of excessive inflammation can alleviate BBB injury, which provides potential therapeutic treatment for ischaemic stroke of the aged. Prior studies revealed that mesencephalic astrocyte-derived neurotrophic factor (MANF) regulated inflammatory response and alleviated liver injury in ageing. However, it is unclear whether MANF confer similar benefit to BBB of aged mice suffered from ischaemic stroke. Transient cerebral ischaemia induced by middle cerebral artery occlusion (MCAO) was conducted in aged mice (18-20 months old). MANF was injected into the right lateral ventricle 2 h after MCAO. BBB integrity, tight junctional proteins, ultrastructure of microvessels, infarct volume, neurological scores, brain water content, pro-inflammatory cytokines and neutrophil infiltration rate were determined 72 h after MCAO. H2O2-induced senescent bEnd.3 cells were applied in the in vitro study to investigate the possible mechanism. First, we confirmed that ischaemic stroke/reperfusion in senescent condition promoted the over-expression of MANF on brain endothelial cells. Then, MANF supplement could suppress the pro-inflammatory factor production, restore BBB integrity and then alleviate infarct volume, neurological scores, brain water content and neutrophil infiltration rate. In addition, MANF maintained BBB integrity after ischaemic stroke of aged condition dependent on TLR4/MyD88/NF-κB pathway via intervention of pro-inflammatory factors production. In summary, the recognition of MANF in the process of BBB breakdown at aged condition may offer novel therapeutic approaches for ischaemic stroke.
Collapse
Affiliation(s)
- Dan Han
- Department of Pharmacy, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.,Nanjing Medical Center for Clinical Pharmacy, Nanjing, China.,School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Fengyang Li
- Department of Pharmacology, Key Laboratory of Chemical Biology, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Haixia Zhang
- Department of Pharmacy, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Cheng Ji
- Department of Pharmacy, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Qing Shu
- Department of Pharmacy, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Cheng Wang
- School of Pharmacy, Changzhou University, Changzhou, China
| | - Huanyu Ni
- Department of Pharmacy, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yun Zhu
- Department of Pharmacy, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Siliang Wang
- Department of Pharmacy, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
23
|
Qiu LN, Tan YR, Luo YJ, Chen XJ. Leonurine protects against influenza A virus infection-induced pneumonia in mice. Pathog Dis 2021; 79:6372906. [PMID: 34543397 DOI: 10.1093/femspd/ftab045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 09/15/2021] [Indexed: 12/24/2022] Open
Abstract
Influenza A virus (H1N1), a swine-origin influenza A virus, causes seasonal epidemics that result in severe illnesses and deaths. Leonurine has been reported to function as an anti-inflammatory agent with protective effects on nervous, urinary and cardiovascular systems. However, the therapeutic effects of leonurine on the pneumonia caused by H1N1 infection remain unclear. Hematoxylin and eosin staining was performed to evaluate the lung injuries of mice infected by H1N1. The amount of immune cells was analyzed by flow cytometry. Enzyme-linked immunosorbent assay was used to evaluate the alteration of multiple cytokines in lung tissues. Real-time quantitative polymerase chain reaction assay was performed to investigate the ribonucleic acid (RNA) levels of certain genes. The protein levels in toll-like receptor 4/nuclear factor kappa-light-chain-enhancer of activated B cells (TLR4/NF-κB) signaling were estimated by western blot assay. Leonurine treatment significantly inhibited the mortality caused by H1N1 infection. Leonurine treatment (60 mg/kg) alleviated the lung injuries caused by virus infection. The inflammatory cell accumulation and cytokine expression were inhibited by the leonurine administration. Leonurine inhibited the mRNA expression of pro-inflammatory cytokines in the lung homogenates at day 5 postinfection. Leonurine regulated the TLR4/NF-κB signaling in the lung homogenates of H1N1-infected mice at day 5 postinfection. Leonurine protects against H1N1 infection-induced pneumonia in mice.
Collapse
Affiliation(s)
- Li-Nan Qiu
- Department of Traditional Chinese Medicine, Daqing Oilfield General Hospital, 9 Zhongkang Street, Daqing 163001, China
| | - Ya-Rong Tan
- Day Clinic Centre, Daqing Oilfield General Hospital, 9 Zhongkang Street, Daqing 163001, China
| | - Yu-Ju Luo
- Department of Medical Imaging, Daqing Oilfield General Hospital, 9 Zhongkang Street, Daqing 163001, China
| | - Xiao-Juan Chen
- Department of Pediatrics, Daqing Oilfield General Hospital, 9 Zhongkang Street, Daqing 163001, China
| |
Collapse
|
24
|
Liu Y, Cheng Y, Zhang W, Tian H. Neuroprotective Effect of a New Free Radical Scavenger HL-008 in an Ischemia-Reperfusion Injury Rat Model. Neuroscience 2021; 465:105-115. [PMID: 33915200 DOI: 10.1016/j.neuroscience.2021.04.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/13/2021] [Accepted: 04/15/2021] [Indexed: 12/28/2022]
Abstract
Oxidative stress plays a critical role in cerebral ischemia-reperfusion injury. We have previously developed a powerful antioxidant, HL-008. This study aimed to investigate the neuroprotective function of HL-008. HL-008 efficacy in vitro and in vivo was evaluated using a PC-12 cell oxidative stress model induced by hydrogen peroxide and a rat model of middle cerebral artery occlusion, respectively. The MTT assay was used to analyze cell viability. 2,3,5-Triphenyltetrazolium chloride and Hematoxylin and Eosin staining, immunofluorescence, western blot, and proteomics were used to evaluate the infarction volume, brain tissue morphology, apoptosis, inflammation, and related pathways. Indicators related to oxidative levels were also detected. HL-008 significantly reduced the cerebral infarction volume induced by ischemia-reperfusion, improved the neurological score, alleviated oxidative stress and inflammation in the brain tissue, reduced glial cell activation, inhibited brain tissue apoptosis by influencing multiple signaling pathways, and had a neuroprotective effect. If HL-008 is successfully developed, it could significantly improve stroke patients' quality of life.
Collapse
Affiliation(s)
- Yahong Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 238, Baidi Road, Tianjin, China
| | - Ying Cheng
- Center for Marine Bioproducts Development, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, SA 5042, Australia
| | - Wei Zhang
- Center for Marine Bioproducts Development, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, SA 5042, Australia.
| | - Hongqi Tian
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 238, Baidi Road, Tianjin, China.
| |
Collapse
|
25
|
Stone NL, England TJ, O'Sullivan SE. Protective Effects of Cannabidivarin and Cannabigerol on Cells of the Blood-Brain Barrier Under Ischemic Conditions. Cannabis Cannabinoid Res 2021; 6:315-326. [PMID: 33998890 PMCID: PMC8380798 DOI: 10.1089/can.2020.0159] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background and Objectives: Preclinical studies have shown cannabidiol is protective in models of ischemic stroke. Based on results from our recent systematic review, we investigated the effects of two promising neuroprotective phytocannabinoids, cannabigerol (CBG) and cannabidivarin (CBDV), on cells of the blood-brain barrier (BBB), namely human brain microvascular endothelial cells (HBMECs), pericytes, and astrocytes. Experimental Approach: Cultures were subjected to oxygen-glucose deprivation (OGD) protocol to model ischemic stroke and cell culture medium was assessed for cytokines and adhesion molecules post-OGD. Astrocyte cell lysates were also analyzed for DNA damage markers. Antagonist studies were conducted where appropriate to study receptor mechanisms. Results: In astrocytes CBG and CBDV attenuated levels of interleukin-6 (IL-6) and lactate dehydrogenase (LDH), whereas CBDV (10 nM-10 μM) also decreased vascular endothelial growth factor (VEGF) secretion. CBDV (300 nM-10 μM) attenuated levels of monocyte chemoattractant protein (MCP)-1 in HBMECs. In astrocytes, CBG decreased levels of DNA damage proteins, including p53, whereas CBDV increased levels of DNA damage markers. Antagonists for CB1, CB2, PPAR-γ, PPAR-α, 5-HT1A, and TRPV1 had no effect on CBG (3 μM) or CBDV (1 μM)-mediated decreases in LDH in astrocytes. GPR55 and GPR18 were partially implicated in the effects of CBDV, but no molecular target was identified for CBG. Conclusions: We show that CBG and CBDV were protective against OG mediated injury in three different cells that constitute the BBB, modulating different hallmarks of ischemic stroke pathophysiology. These data enhance our understanding of the protective effects of CBG and CBDV and warrant further investigation into these compounds in ischemic stroke. Future studies should identify other possible neuroprotective effects of CBG and CBDV and their corresponding mechanisms of action.
Collapse
Affiliation(s)
- Nicole L Stone
- Division of Medical Sciences and Graduate Entry Medicine, School of Medicine, University of Nottingham, Royal Derby Hospital, Derby, United Kingdom
| | - Timothy J England
- Division of Medical Sciences and Graduate Entry Medicine, School of Medicine, University of Nottingham, Royal Derby Hospital, Derby, United Kingdom.,University Hospitals of Derby and Burton NHS Foundation Trust, Royal Derby Hospital, Derby, United Kingdom
| | - Saoirse E O'Sullivan
- University Hospitals of Derby and Burton NHS Foundation Trust, Royal Derby Hospital, Derby, United Kingdom.,CanPharmaConsulting, Nottingham, United Kingdom
| |
Collapse
|
26
|
Yang M, Feng Y, Yan S, Wu Z, Xiao X, Sang J, Ye S, Liu F, Cui W. Evans Blue Might Produce Pathologically Activated Neuroprotective Effects via the Inhibition of the P2X4R/p38 Signaling Pathway. Cell Mol Neurobiol 2021; 41:293-307. [PMID: 32382851 DOI: 10.1007/s10571-020-00852-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 04/16/2020] [Indexed: 12/15/2022]
Abstract
The main pathological features of ischemic stroke include neuronal damage and blood-brain barrier (BBB) dysfunction. Previous studies have shown that Evans Blue, a dye used to probe BBB integrity, could enter the brain only during the pathological status of ischemic stroke, indicating the potential pathologically activated therapeutic use of this chemical to treat ischemic stroke. In this study, we have reported that Evans Blue could produce in vitro neuroprotective effects against iodoacetic acid (IAA)-induced hypoxia neuronal death in HT22 cells. We further found that P2X purinoreceptor 4 (P2X4R), a subtype of ATP-gated cation channel, was expressed in HT22 cells. Evans Blue could prevent IAA-induced increase of P2X4R mRNA and protein expression. Interestingly, shRNA of P2X4R could protect against IAA-induced activation of p38, and SB203580, a specific inhibitor of p38, could reverse IAA-induced neurotoxicity, indicating that p38 is a downstream signaling molecule of P2X4R. Molecular docking analysis further demonstrated the possible interaction between Evans Blue and the ATP binding site of P2X4R. Most importantly, pre-treatment of Evans Blue could largely reduce neurological and behavioral abnormity, and decrease brain infarct volume in middle cerebral artery occlusion/reperfusion (MCAO) rats. All these results strongly suggested that Evans Blue could exert neuroprotective effects via inhibiting the P2X4R/p38 pathway, possibly by acting on the ATP binding site of P2X4R, indicating that Evans Blue might be further developed as a pathologically activated therapeutic drug against ischemic stroke.
Collapse
Affiliation(s)
- Mengxiang Yang
- Translational Medicine Center of Pain, Emotion and Cognition, Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, 315211, China
| | - Yi Feng
- Translational Medicine Center of Pain, Emotion and Cognition, Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, 315211, China
| | - Sicheng Yan
- Translational Medicine Center of Pain, Emotion and Cognition, Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, 315211, China
| | - Zhuoying Wu
- Translational Medicine Center of Pain, Emotion and Cognition, Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, 315211, China
| | - Xiao Xiao
- Translational Medicine Center of Pain, Emotion and Cognition, Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, 315211, China
| | - Jingcheng Sang
- Key Laboratory of Industrial Fermentation Microbiology of Education, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Shazhou Ye
- Translational Medicine Center of Pain, Emotion and Cognition, Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, 315211, China
| | - Fufeng Liu
- Key Laboratory of Industrial Fermentation Microbiology of Education, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Wei Cui
- Translational Medicine Center of Pain, Emotion and Cognition, Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, 315211, China.
- Department of Physiology, Medical School, Ningbo University, Zhejiang, China.
| |
Collapse
|
27
|
Huang L, Xu DQ, Chen YY, Yue SJ, Tang YP. Leonurine, a potential drug for the treatment of cardiovascular system and central nervous system diseases. Brain Behav 2021; 11:e01995. [PMID: 33300684 PMCID: PMC7882174 DOI: 10.1002/brb3.1995] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 11/17/2020] [Accepted: 11/20/2020] [Indexed: 12/13/2022] Open
Abstract
Leonurus japonicus Houtt., a traditional Chinese herbal medicine, is often used as a gynecological medicine with the effect of promoting blood circulation, regulating menstruation, clearing heat, and detoxificating. As the most important alkaloid in L. japonicus, leonurine has a wide range of biological activities, such as antioxidation, anti-inflammation, and anti-apoptosis. Cardiovascular system and central nervous system diseases are arrogant killers that threaten human lives and health around the world, but many drugs for treating them have certain side effects. This paper reviews the potential therapeutic effects of leonurine on cardiovascular system and central nervous system diseases, summarizes the previous research progress, and focuses on its therapeutic effect in various diseases. Although leonurine plays a prominent role in the treatment of cardiovascular system and central nervous system diseases, there are still some shortages, such as low bioavailability, weak transmembrane ability, and poor fat solubility. Therefore, the structure modification of leonurine may solve these problems and provide reference value for the development of new drugs. At present, leonurine is in clinical trial, and it is hoped that our summary will help to provide guidance for its future research on the basic science and clinical application.
Collapse
Affiliation(s)
- Lu Huang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Ding-Qiao Xu
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Yan-Yan Chen
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Shi-Jun Yue
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Yu-Ping Tang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, China
| |
Collapse
|
28
|
Yang J, Wang Z, Liu X, Lu P. Modulation of vascular integrity and neuroinflammation by peroxiredoxin 4 following cerebral ischemia-reperfusion injury. Microvasc Res 2021; 135:104144. [PMID: 33515567 DOI: 10.1016/j.mvr.2021.104144] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 01/16/2021] [Accepted: 01/25/2021] [Indexed: 01/10/2023]
Abstract
Ischemic stroke is a leading cause of morbidity and mortality worldwide, with oxidative stress playing a key role in the injury mechanism of thrombolytic therapy. There is increasing evidence that oxidative stress damages endothelial cells (ECs), degrades tight junction proteins (TJs), and contributes to increased blood-brain barrier (BBB) permeability. It has been demonstrated that the breakdown of BBB could increase the risk of intracerebral hemorrhagic transformation in ischemic stroke. And an episode of cerebral ischemia/reperfusion (I/R) also initiates oxidative stress-mediated inflammatory processes in ECs, which further promotes BBB disruption and the progression of brain injury. Previous studies have revealed that antioxidants could inhibit ROS generation and attenuate BBB disruption after cerebral I/R. Peroxiredoxin 4 (Prx4) is a member of the antioxidant enzymes family (Prx1-6) and has been characterized to be an efficient H2O2 scavenger. It should be noted that Prx4 may be directly involved in the protection of ECs from the effects of ROS and function in ECs as a membrane-associated peroxidase. This paper reviewed the implication of Prx4 on vascular integrity and neuroinflammation following a cerebral I/R injury.
Collapse
Affiliation(s)
- Jiping Yang
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China.
| | - Zairan Wang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China
| | - Xiuying Liu
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China
| | - Pengchao Lu
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China
| |
Collapse
|
29
|
Lin Q, Wang W, Yang L, Duan X. 4-Methoxybenzylalcohol protects brain microvascular endothelial cells against oxygen-glucose deprivation/reperfusion-induced injury via activation of the PI3K/AKT signaling pathway. Exp Ther Med 2021; 21:252. [PMID: 33613705 PMCID: PMC7856387 DOI: 10.3892/etm.2021.9684] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 09/02/2020] [Indexed: 11/07/2022] Open
Abstract
Damage to the blood-brain barrier (BBB) during the process of cerebral ischemic injury is a key factor that affects the treatment of this condition. The present study aimed to assess the potential effects of 4-methoxybenzyl alcohol (4-MA) on brain microvascular endothelial cells (bEnd.3) against oxygen-glucose deprivation/reperfusion (OGD/Rep) using an in vitro model that mimics in vivo ischemia/reperfusion injury. In addition, the present study aimed to explore whether this underlying mechanism was associated with the inhibition of pro-inflammatory factors and the activation status of the PI3K/Akt signaling pathway. bEnd.3 cells were subjected to OGD/Rep-induced injury before being treated with 4-MA, following which cell viability, lactate dehydrogenase (LDH) release and levels of nitric oxidase (NO) were detected by colorimetry, pro-inflammatory factors including tumor necrosis factor-α (TNF-α), interleukin (IL)-1β and IL-6, were detected by ELISA. The expression levels of occluding and claudin-5were evaluated by immunofluorescence staining. The expression levels of AKT, phosphorylated (p)-Akt, endothelial nitric oxide synthase (eNOS) and p-eNOS were also measured by western blot analysis. After bEnd.3 cells were subjected to OGD/Rep-induced injury, cell viability and NO levels were significantly decreased, whilst LDH leakage and inflammatory factor (TNF-α, IL-1β and IL-6) levels were significantly increased. Treatment with 4-MA significantly ameliorated cell viability, LDH release and the levels of NO and pro-inflammatory factors TNF-α, IL-1β and IL-6 as a result of OGD/Rep. Furthermore, treatment with 4-MA upregulated the expression of occludin, claudin-5, Akt and eNOS, in addition to increasing eNOS and AKT phosphorylation in bEnd.3 cells. These results suggest that 4-MA can alleviate OGD/Rep-induced injury in bEnd.3 cells by inhibiting inflammation and by activating the PI3K/AKT signaling pathway as a possible mechanism. Therefore, 4-MA can serve as a potential candidate for treating OGD/Rep-induced injury.
Collapse
Affiliation(s)
- Qing Lin
- Department of Pharmacology, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Weili Wang
- Department of Pharmacology, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Liping Yang
- Yunnan Key Laboratory of Dai and Yi Medicine, University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Xiaohua Duan
- Yunnan Key Laboratory of Dai and Yi Medicine, University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
30
|
Li Y, Xiang L, Wang C, Song Y, Miao J, Miao M. Protection against acute cerebral ischemia/reperfusion injury by Leonuri Herba Total Alkali via modulation of BDNF-TrKB-PI3K/Akt signaling pathway in rats. Biomed Pharmacother 2021; 133:111021. [PMID: 33227709 DOI: 10.1016/j.biopha.2020.111021] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/09/2020] [Accepted: 11/15/2020] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE To observe the brain protective effect of Leonuri Herba Total Alkali (LHA) on cerebral ischemia reperfusion injury in rats, so as to provide basis for clinical research. METHODS Adult male SD rats were randomly assigned into sham group, middle cerebral artery occlusion/reperfusion (MCAO/R) group, and LHA + MCAO/R group (25 mg/kg, 50 mg/kg, and 100 mg/kg). Fourteen days before MCAO/R surgery, the rats in treatment groups were orally administered with LHA in ultrapure water once daily for 14 days, while rats in the sham and MCAO groups were given the same amount of saline in advance. After 1 h of administration on the 14th day, MCAO surgery was subjected. The neurological deficits, brain infarct volume, histopathology, immunofluorescence, inflammation indicators and the gene/protein expressions of BDNF-TrKB-PI3K/Akt signaling pathway in the rat brain tissue were evaluated 24 h after the MCAO/R-injury. RESULTS It was found that rats in LHA pre-administration group showed significantly reduced neurological deficit scores, infarction volume, the serum levels of NSE and S100β. Meanwhile, the content of Evans Blue (EB) in brain tissue from LHA group was decreased, as well as the levels of inflammatory cytokines and their gene levels. Moreover, LHA pre-administration inhibited the expression of CD44, GFAP, FOXO1 and promoted the expression of BDNF and NeuN. In addition, LHA pre-administration could up-regulate the protein expression of TrkB, p-PI3K, p-Akt, Bcl-2, and down-regulate the protein expression of Bax, and increase the level of Bcl-2/Bax. CONCLUSIONS The study demonstrated that LHA pre-administration could regulate the PI3K/Akt pathway by increasing BDNF levels, and play a neuroprotective role in cerebral ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Yan Li
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, PR China
| | - Liling Xiang
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, PR China
| | - Can Wang
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, PR China
| | - Yagang Song
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, PR China
| | - Jinxin Miao
- National International Cooperation Base of Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, PR China.
| | - Mingsan Miao
- National International Cooperation Base of Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, PR China.
| |
Collapse
|
31
|
Icariside II attenuates cerebral ischemia/reperfusion-induced blood-brain barrier dysfunction in rats via regulating the balance of MMP9/TIMP1. Acta Pharmacol Sin 2020; 41:1547-1556. [PMID: 32488170 DOI: 10.1038/s41401-020-0409-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 03/26/2020] [Indexed: 02/06/2023] Open
Abstract
Cerebral ischemia/reperfusion (I/R) results in harmful consequences during ischemic stroke, especially the disruption of the blood-brain barrier (BBB), which leads to severe hemorrhagic transformation through aggravation of edema and brain hemorrhage. Our previous study demonstrated that icariside II (ICS II), which is derived from Herba Epimedii, attenuates cerebral I/R injury by inhibiting the GSK-3β-mediated activation of autophagy both in vitro and in vivo. However, the effect of ICS II on the BBB remains unclear. Thus, in this study, we investigated the regulation of BBB integrity by ICS II after cerebral I/R injury and further explored the underlying mechanism in rats. Cerebral I/R injury was induced by middle cerebral artery occlusion (MCAO), and the treatment groups were administered ICS II at a dose of 16 mg/kg by gavage twice a day for 3 days. The results showed that ICS II effectively prevented BBB disruption, as evidenced by Evans Blue staining. Moreover, ICS II not only significantly reduced the expression of MMP2/9 but also increased TIMP1 and tight junction protein (occludin, claudin 5, and ZO 1) expression. Intriguingly, ICS II may directly bind to both MMP2 and MMP9, as evidenced by molecular docking. In addition, ICS II also inhibited cerebral I/R-induced apoptosis and ameliorated the Bax/Bcl-2 ratio and cleaved-caspase 3 level. Collectively, our findings reveal that ICS II significantly ameliorates I/R-induced BBB disruption and neuronal apoptosis in MCAO rats by regulating the MMP9/TIMP1 balance and inhibiting the caspase 3-dependent apoptosis pathway.
Collapse
|
32
|
Lee BK, Hyun SW, Jung YS. Yuzu and Hesperidin Ameliorate Blood-Brain Barrier Disruption during Hypoxia via Antioxidant Activity. Antioxidants (Basel) 2020; 9:antiox9090843. [PMID: 32916895 PMCID: PMC7555663 DOI: 10.3390/antiox9090843] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 08/31/2020] [Accepted: 09/08/2020] [Indexed: 12/14/2022] Open
Abstract
Yuzu and its main component, hesperidin (HSP), have several health benefits owing to their anti-inflammatory and antioxidant properties. We examined the effects of yuzu and HSP on blood-brain barrier (BBB) dysfunction during ischemia/hypoxia in an in vivo animal model and an in vitro BBB endothelial cell model, and also investigated the underlying mechanisms. In an in vitro BBB endothelial cell model, BBB permeability was determined by measurement of Evans blue extravasation in vivo and in vitro. The expression of tight junction proteins, such as claudin-5 and zonula occludens-1 (ZO-1), was detected by immunochemistry and western blotting, and the reactive oxygen species (ROS) level was measured by 2'7'-dichlorofluorescein diacetate intensity. Yuzu and HSP significantly ameliorated the increase in BBB permeability and the disruption of claudin-5 and ZO-1 in both in vivo and in vitro models. In bEnd.3 cells, yuzu and HSP were shown to inhibit the disruption of claudin-5 and ZO-1 during hypoxia, and the protective effects of yuzu and HSP on claudin-5 degradation seemed to be mediated by Forkhead box O 3a (FoxO3a) and matrix metalloproteinase (MMP)-3/9. In addition, well-known antioxidants, trolox and N-acetyl cysteine, significantly attenuated the BBB permeability increase, disruption of claudin-5 and ZO-1, and FoxO3a activation during hypoxia, suggesting that ROS are important mediators of BBB dysfunction during hypoxia. Collectively, these results indicate that yuzu and HSP protect the BBB against dysfunction via maintaining integrity of claudin-5 and ZO-1, and these effects of yuzu and HSP appear to be a facet of their antioxidant properties. Our findings may contribute to therapeutic strategies for BBB-associated neurodegenerative diseases.
Collapse
Affiliation(s)
- Bo Kyung Lee
- College of Pharmacy, Ajou University, Suwon 16499, Korea, (S.-W.H.)
| | - Soo-Wang Hyun
- College of Pharmacy, Ajou University, Suwon 16499, Korea, (S.-W.H.)
| | - Yi-Sook Jung
- College of Pharmacy, Ajou University, Suwon 16499, Korea, (S.-W.H.)
- Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Korea
- Correspondence: ; Tel.: +82-31-219-3444
| |
Collapse
|
33
|
Yi X, Xu C, Huang P, Zhang L, Qing T, Li J, Wang C, Zeng T, Lu J, Han Z. 1-Trifluoromethoxyphenyl-3-(1-Propionylpiperidin-4-yl) Urea Protects the Blood-Brain Barrier Against Ischemic Injury by Upregulating Tight Junction Protein Expression, Mitigating Apoptosis and Inflammation In Vivo and In Vitro Model. Front Pharmacol 2020; 11:1197. [PMID: 32848796 PMCID: PMC7427473 DOI: 10.3389/fphar.2020.01197] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/23/2020] [Indexed: 02/05/2023] Open
Abstract
We previously have revealed that 1-trifluoromethoxyphenyl-3-(1- propionylpiperidin-4-yl) urea (TPPU), as a soluble epoxide hydrolase (sEH) inhibitor can reduce infarct volume, protect blood-brain barrier (BBB) and brain against ischemic injury in rats. Here, we investigated the potential mechanisms of TPPU on BBB integrity in both in permanent middle cerebral artery occlusion (pMCAO) rat model and in oxygen-glucose deprivation/reperfusion (OGD/R)-induced human brain microvascular endothelial cells (HBMVECs) model. In pMCAO rat, TPPU administration decreased brain edema and Evans blue content, increased tight junction proteins (TJs) expression of claudin-5, occludin, and zonula occludens-1 (ZO-1). In OGD/R model, OGD/R significantly increased permeability and cell apoptosis, downregulated the expression of claudin-5, ZO-1, occludin, and lymphoma (Bcl)-2. Notably, TPPU pretreatment effectively protected the BBB integrity by reducing the permeability, promoting expression of claudin-5, ZO-1, occluding and Bcl-2, mitigating reactive oxygen species (ROS) injury and release of interleukin-1β (IL-1β), IL-6β, and tumor necrosis factor-α (TNF-α), downregulating expression of matrix metalloproteinase-9 (MMP-9), MMP-2, bcl-2-associated X protein (Bax), IL-1β, IL-6β, and TNF-α. Moreover, OGD/R induced the up-regulation of p-p65, p-IκB, and p-p38, which were effectively decreased after TPPU pretreatment in comparison with that of the OGD/R group. Furthermore, pyrrolidinedithiocarbamate (PDTC, a selective inhibitor of NF-κB p65) not only alleviated the OGD/R-induced HBMVECs injury and permeability, but also reduced the expression of TNF-α, IL-6, IL-1β, p-p65, and p-IκB, and the protective effect of PDTC was equivalent to that of TPPU. These results indicate that TPPU protects BBB integrity against ischemic injury by multiple protective mechanisms, at least in part, by reducing ROS, inflammation, apoptosis, and suppressing the nuclear factor-κB (NF-κB) and p38 signaling pathways.
Collapse
Affiliation(s)
- Xingyang Yi
- Department of Neurology, People's Hospital of Deyang City, Deyang, China.,Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Chongxi Xu
- Department of Neurosurgery, West China Hospital of Sichuan University, Chendu, China
| | - Pan Huang
- Department of Neurology, People's Hospital of Deyang City, Deyang, China
| | - Linlei Zhang
- Department of General Intensive Care Unit, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ting Qing
- Department of Neurology, People's Hospital of Deyang City, Deyang, China.,Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jie Li
- Department of Neurology, People's Hospital of Deyang City, Deyang, China
| | - Chun Wang
- Department of Neurology, People's Hospital of Deyang City, Deyang, China
| | - Tao Zeng
- Department of Neurology, People's Hospital of Deyang City, Deyang, China.,Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Jing Lu
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Neurology, Chengdu Fifth People's Hospital, Chengdu, China
| | - Zhao Han
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
34
|
Leng J, Liu W, Li L, Wei FY, Tian M, Liu HM, Guo W. MicroRNA-429/Cxcl1 Axis Protective Against Oxygen Glucose Deprivation/Reoxygenation-Induced Injury in Brain Microvascular Endothelial Cells. Dose Response 2020; 18:1559325820913785. [PMID: 32284700 PMCID: PMC7139192 DOI: 10.1177/1559325820913785] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 02/02/2020] [Accepted: 02/18/2020] [Indexed: 01/12/2023] Open
Abstract
Objective: The objective of the present work was to study the role of Cxcl1 in cerebral
ischemia–reperfusion (I/R) injury and to in-depth explore its pathogenesis. Methods: The expression of Cxcl1 based on the public data was analyzed. Then, we constructed an
oxygen glucose deprivation/reoxygenation (OGD/R) model in vitro using mice brain
microvascular endothelial cells (BMECs) to simulate cerebral I/R in vivo. Results: The results of quantitative real-time polymerase chain reaction assay uncovered that
Cxcl1 showed higher expression while miR-429 showed lower expression in BMECs damaged by
OGD/R, whereas overexpression of Cxcl1 or inhibition of miR-429 expression can
strengthen this effect. Hereafter, through dual luciferase reporter assay, we verified
that miR-429 directly targets Cxcl1 and negatively regulates Cxcl1 expression.
Furthermore, the results also revealed that overexpression of Cxcl1 can reverse the
miR-429-mediated effects. Conclusion: We concluded that miR-429 exerts protective effects against OGD/R-induce injury in
vitro through modulation of Cxcl1 and nuclear factor kinase B pathway, hoping provide a
new view on the pathogenesis of cerebral I/R injury and a feasible potential therapeutic
target.
Collapse
Affiliation(s)
- Jun Leng
- Department of Rehabilitation Medicine, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, People's Republic of China.,Co-first authors and contributed equally to this work
| | - Wei Liu
- Department of Rehabilitation Medicine, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, People's Republic of China.,Co-first authors and contributed equally to this work
| | - Li Li
- Department of Rehabilitation Medicine, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, People's Republic of China
| | - Fang Yue Wei
- Shandong University of Traditional Chinese Medicine Rehabilitation College Rehabilitation Medicine and Physiotherapy, Jinan, Shandong Province, People's Republic of China
| | - Meng Tian
- Competitive sports section 1 of Sports Science Research Center of Shandong Province, Jinan, Shandong Province, People's Republic of China
| | - Hui Min Liu
- Shandong University of Traditional Chinese Medicine Rehabilitation College Rehabilitation Medicine and Physiotherapy, Jinan, Shandong Province, People's Republic of China
| | - Wen Guo
- Shandong University of Traditional Chinese Medicine Rehabilitation College Rehabilitation Medicine and Physiotherapy, Jinan, Shandong Province, People's Republic of China
| |
Collapse
|
35
|
Wang F, Li R, Tu P, Chen J, Zeng K, Jiang Y. Total Glycosides of Cistanche deserticola Promote Neurological Function Recovery by Inducing Neurovascular Regeneration via Nrf-2/Keap-1 Pathway in MCAO/R Rats. Front Pharmacol 2020; 11:236. [PMID: 32256351 PMCID: PMC7089931 DOI: 10.3389/fphar.2020.00236] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 02/20/2020] [Indexed: 12/21/2022] Open
Abstract
Background The traditional Chinese medicine Cistanche deserticola has been reported to be valid for cardiovascular and cerebrovascular diseases. However, its active components for the protection of ischemic stroke are not clear. We aimed to explore the active components of C. deserticola against ischemic stroke as well as its potential mechanisms. Methods We investigated the brain protective effects of extracts from C. deserticola, total glycosides (TGs), polysaccharides (PSs), and oligosaccharides (OSs) in a rat model of middle cerebral artery occlusion-reperfusion (MCAO/R). 2, 3, 5-Triphenyltetrazolium chloride (TTC) staining was used to assess the cerebral infarction volume, and Evans blue assay was adopted to assess the blood-brain barrier (BBB) permeability. Then, the expressions CD31, α-SMA, PDGFRβ, SYN, PSD95, MAP-2, ZO-1, claudin-5, occludin, Keap-1, and Nrf-2 were analyzed using western blotting or immunofluorescence, and the activities MDA, SOD, CAT, and GSH-Px were analyzed using kits. Results TGs treatment remarkably decreased neurological deficit scores and infarction volumes, promoted angiogenesis and neural remodeling, and effectively maintained blood-brain-barrier integrity compared with the model group. Furthermore, TGs significantly decreased MDA levels and increased antioxidant activities (SOD, CAT, and GSH-Px) in brains. Meanwhile, TGs remarkably downregulated Keap-1 expression and facilitated Nrf-2 nuclear translocation. On the contrary, no protective effects were observed for PSs and OSs groups. Conclusion TGs are the main active components of C. deserticola against MCAO/R-induced cerebral injury, and protection is mainly via the Nrf-2/Keap-1 pathway.
Collapse
Affiliation(s)
- Fujiang Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Ruiyan Li
- Department of Pharmacology, Changzhi Medical College, Shanxi, China
| | - Pengfei Tu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jianping Chen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Kewu Zeng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yong Jiang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| |
Collapse
|
36
|
Phoenixin-14 protects human brain vascular endothelial cells against oxygen-glucose deprivation/reoxygenation (OGD/R)-induced inflammation and permeability. Arch Biochem Biophys 2020; 682:108275. [DOI: 10.1016/j.abb.2020.108275] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 01/08/2020] [Accepted: 01/16/2020] [Indexed: 11/19/2022]
|
37
|
Qu XY, Zhang YM, Tao LN, Gao H, Zhai JH, Sun JM, Song YQ, Zhang SX. XingNaoJing injections protect against cerebral ischemia/reperfusion injury and alleviate blood-brain barrier disruption in rats, through an underlying mechanism of NLRP3 inflammasomes suppression. Chin J Nat Med 2020; 17:498-505. [PMID: 31514981 DOI: 10.1016/s1875-5364(19)30071-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Indexed: 12/15/2022]
Abstract
The aim of this study was to explore the neuroprotective effect and mechanism of XingNaoJing injections (XNJ) on cerebral ischemia injury and blood-brain barrier (BBB) disruption. Middle cerebral artery occlusion (MCAO) method was applicated to establish the model of cerebral ischemia/reperfusion (I/R) injury in rats. BBB permeability after I/R injury was assessed with the leaking amount of Evans Blue and the expression of occludin and ZO-1. The expression of NOD-like receptor family, pyrin domain containing (NLRP3) was checked to explore the inhibition of inflammation by XNJ. The results showed that XNJ could significantly increase the survival percent, decrease the infarct area and ameliorate neurological deficits and brain damage after I/R injury. Leaking amount of Evans Blue was reduced by XNJ, and the expression of tight junction protein, occludin and ZO-1 was also up-regulated by XNJ, which showed a role of protection on BBB disruption. The expression of NLRP3 was inhibited after exposure of XNJ, which was associated with inhibition of the inflammatory response. In summary, XNJ could suppress NLRP3 inflammasomes and improve BBB disruption and brain damage in rats after cerebral I/R injury, which provided a beneficial insight to further explore XNJ.
Collapse
Affiliation(s)
- Xiao-Yu Qu
- Department of Pharmacy, the First Hospital of Jilin University, Changchun 130021, China
| | - Yue-Ming Zhang
- Department of Pharmacy, the First Hospital of Jilin University, Changchun 130021, China
| | - Li-Na Tao
- Department of Pharmacy, the First Hospital of Jilin University, Changchun 130021, China
| | - Huan Gao
- Department of Pharmacy, the First Hospital of Jilin University, Changchun 130021, China
| | - Jing-Hui Zhai
- Department of Pharmacy, the First Hospital of Jilin University, Changchun 130021, China
| | - Jing-Meng Sun
- Department of Pharmacy, the First Hospital of Jilin University, Changchun 130021, China
| | - Yan-Qing Song
- Department of Pharmacy, the First Hospital of Jilin University, Changchun 130021, China.
| | - Si-Xi Zhang
- Department of Pharmacy, the First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
38
|
Stanzione R, Cotugno M, Bianchi F, Marchitti S, Forte M, Volpe M, Rubattu S. Pathogenesis of Ischemic Stroke: Role of Epigenetic Mechanisms. Genes (Basel) 2020; 11:genes11010089. [PMID: 31941075 PMCID: PMC7017187 DOI: 10.3390/genes11010089] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/29/2019] [Accepted: 01/09/2020] [Indexed: 12/17/2022] Open
Abstract
Epigenetics is the branch of molecular biology that studies modifications able to change gene expression without altering the DNA sequence. Epigenetic modulations include DNA methylation, histone modifications, and noncoding RNAs. These gene modifications are heritable and modifiable and can be triggered by lifestyle and nutritional factors. In recent years, epigenetic changes have been associated with the pathogenesis of several diseases such as diabetes, obesity, renal pathology, and different types of cancer. They have also been related with the pathogenesis of cardiovascular diseases including ischemic stroke. Importantly, since epigenetic modifications are reversible processes they could assist with the development of new therapeutic approaches for the treatment of human diseases. In the present review article, we aim to collect the most recent evidence concerning the impact of epigenetic modifications on the pathogenesis of ischemic stroke in both animal models and humans.
Collapse
Affiliation(s)
- Rosita Stanzione
- IRCCS Neuromed, Via Atinense, 18, 86077 Pozzilli IS, Italy; (M.C.); (F.B.); (S.M.); (M.F.); (M.V.); (S.R.)
- Correspondence: ; Tel.: +86-5915224
| | - Maria Cotugno
- IRCCS Neuromed, Via Atinense, 18, 86077 Pozzilli IS, Italy; (M.C.); (F.B.); (S.M.); (M.F.); (M.V.); (S.R.)
| | - Franca Bianchi
- IRCCS Neuromed, Via Atinense, 18, 86077 Pozzilli IS, Italy; (M.C.); (F.B.); (S.M.); (M.F.); (M.V.); (S.R.)
| | - Simona Marchitti
- IRCCS Neuromed, Via Atinense, 18, 86077 Pozzilli IS, Italy; (M.C.); (F.B.); (S.M.); (M.F.); (M.V.); (S.R.)
| | - Maurizio Forte
- IRCCS Neuromed, Via Atinense, 18, 86077 Pozzilli IS, Italy; (M.C.); (F.B.); (S.M.); (M.F.); (M.V.); (S.R.)
| | - Massimo Volpe
- IRCCS Neuromed, Via Atinense, 18, 86077 Pozzilli IS, Italy; (M.C.); (F.B.); (S.M.); (M.F.); (M.V.); (S.R.)
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Speranza Rubattu
- IRCCS Neuromed, Via Atinense, 18, 86077 Pozzilli IS, Italy; (M.C.); (F.B.); (S.M.); (M.F.); (M.V.); (S.R.)
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| |
Collapse
|
39
|
Zeng C, Wang D, Chen C, Chen L, Chen B, Li L, Chen M, Xing H. Zafirlukast protects blood-brain barrier integrity from ischemic brain injury. Chem Biol Interact 2019; 316:108915. [PMID: 31816286 DOI: 10.1016/j.cbi.2019.108915] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/22/2019] [Accepted: 12/05/2019] [Indexed: 02/07/2023]
Abstract
Stroke has been considered the second leading cause of death worldwide, and ischemic stroke accounts for the vast majority of stroke cases. Some of the main features of ischemic stroke are increased brain permeability, ischemia/reperfusion injury, oxidative stress, and acute inflammation. Antagonism of cysLT1R has been shown to provide cardiovascular and neural benefits. In the present study, we investigated the effects of the cysLT1R antagonist zafirlukast both in vivo and in vitro using a middle cerebral artery occlusion (MCAO) mouse model and human brain microvascular endothelial cells (HBMVECs). In vivo, we found that zafirlukast pretreatment could reduce MCAO-induced increased brain permeability by rescuing the expression levels of the tight junction proteins occludin and ZO-1. In vitro, we found that zafirlukast could suppress the increase in endothelial monolayer permeability induced by OGD/R via rescue of occludin and ZO-1 expression; additionally, we found that zafirlukast prevented OGD/R-induced degradation of the extracellular matrix via inhibition of MMP-2 and MMP-9 expression. Finally, we found that zafirlukast could also inhibit OGD/R-induced activation of the critical proinflammatory regulator NF-κB by preventing phosphorylation and nuclear translocation of p65 protein. Together, our findings demonstrate a promising role for zafirlukast in preventing damage induced by ischemic stroke and reperfusion injury.
Collapse
Affiliation(s)
- Chaosheng Zeng
- Department of Neurology, The Second Affiliated Hospital of Hainan Medical University, Haikou City, Hainan Province, China
| | - Desheng Wang
- Department of Neurology, The Second Affiliated Hospital of Hainan Medical University, Haikou City, Hainan Province, China
| | - Cong Chen
- Department of Neurology, The Second Affiliated Hospital of Hainan Medical University, Haikou City, Hainan Province, China.
| | - Lin Chen
- Department of Neurology, The Second Affiliated Hospital of Hainan Medical University, Haikou City, Hainan Province, China
| | - Bocan Chen
- Department of Neurology, The Second Affiliated Hospital of Hainan Medical University, Haikou City, Hainan Province, China
| | - Li Li
- Department of Neurology, The Second Affiliated Hospital of Hainan Medical University, Haikou City, Hainan Province, China
| | - Min Chen
- Department of Neurology, The Second Affiliated Hospital of Hainan Medical University, Haikou City, Hainan Province, China
| | - Huaijie Xing
- Department of Neurology, The Second Affiliated Hospital of Hainan Medical University, Haikou City, Hainan Province, China
| |
Collapse
|
40
|
Neuroprotective Effect of SCM-198 through Stabilizing Endothelial Cell Function. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7850154. [PMID: 31827699 PMCID: PMC6885260 DOI: 10.1155/2019/7850154] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/31/2019] [Accepted: 08/14/2019] [Indexed: 01/02/2023]
Abstract
Leonurine, also named SCM-198, which was extracted from Herba leonuri, displayed a protective effect on various cardiovascular and brain diseases, like ischemic stroke. Ischemic stroke which is the leading cause of morbidity and mortality, ultimately caused irreversible neuron damage. This study is aimed at exploring the possible therapeutic potential of SCM-198 in the protection against postischemic neuronal injury and possible underlying mechanisms. A transient middle cerebral artery occlusion (tMCAO) rat model was utilized to measure the protective effect of SCM-198 on neurons. TEM was used to determine neuron ultrastructural changes. The brain slices were stained with Nissl staining solution for Nissl bodies. Fluoro-Jade B (FJB) was used for staining the degenerating neurons. In the oxygen-glucose deprivation and re-oxygenation (OGD/R) model of bEnd.3 cells treated with SCM-198 (0.1, 1, 10 μM). Then, the bEnd.3 cells were cocultured with SH-SY5Y cells. Cell viability, MDA level, CAT activity, and apoptosis were examined to evaluate the cytotoxicity of these treatments. Western blot and immunofluorescent assays were used to examine the expression of protein related to the p-STAT3/NOX4/Bcl-2 signaling pathway. Coimmunoprecipitation was performed to determine the interaction between p-STAT3 and NOX4. In the transient middle cerebral artery occlusion (tMCAO) rat model, we found that treatment with SCM-198 could ameliorate neuron morphology and reduce the degenerating cell and neuron loss. In the in vitro model of bEnd.3 cell oxygen-glucose deprivation and reoxygenation (OGD/R), treatment with SCM-198 restored the activity of catalase (CAT), improved the expression of Cu-Zn superoxide dismutase (SOD1), and decreased the malondialdehyde (MDA) production. SCM-198 treatment prevented OGD/R-induced cell apoptosis as indicated by increased cell viability and decreased the number of TUNEL-positive cells, accompanied with upregulation of Bcl-2 and Bcl-xl protein and downregulation Bax protein. The results were consistent with SH-SY5Y cells which coculture with bEnd.3 cells. The forthcoming study revealed that SCM-198 activated the p-STAT3/NOX4/Bcl-2 signaling pathway. All the data indicated that SCM-198 protected against oxidative stress and neuronal damage in in vivo and in vitro injury models via the p-STAT3/NOX4/Bcl-2 signaling pathway. Our results suggested that SCM-198 could be the potential drug for neuroprotective effect through stabilizing endothelial cell function.
Collapse
|
41
|
Stamatovic SM, Phillips CM, Martinez-Revollar G, Keep RF, Andjelkovic AV. Involvement of Epigenetic Mechanisms and Non-coding RNAs in Blood-Brain Barrier and Neurovascular Unit Injury and Recovery After Stroke. Front Neurosci 2019; 13:864. [PMID: 31543756 PMCID: PMC6732937 DOI: 10.3389/fnins.2019.00864] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/31/2019] [Indexed: 12/13/2022] Open
Abstract
Cessation of blood flow leads to a complex cascade of pathophysiological events at the blood-vascular-parenchymal interface which evolves over time and space, and results in damage to neural cells and edema formation. Cerebral ischemic injury evokes a profound and deleterious upregulation in inflammation and triggers multiple cell death pathways, but it also induces a series of the events associated with regenerative responses, including vascular remodeling, angiogenesis, and neurogenesis. Emerging evidence suggests that epigenetic reprograming could play a pivotal role in ongoing post-stroke neurovascular unit (NVU) changes and recovery. This review summarizes current knowledge about post-stroke recovery processes at the NVU, as well as epigenetic mechanisms and modifiers (e.g., DNA methylation, histone modifying enzymes and microRNAs) associated with stroke injury, and NVU repair. It also discusses novel drug targets and therapeutic strategies for enhancing post-stroke recovery.
Collapse
Affiliation(s)
- Svetlana M. Stamatovic
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Chelsea M. Phillips
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI, United States
| | | | - Richard F. Keep
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Molecular Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Anuska V. Andjelkovic
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
42
|
Li Y, Lin Y, Huang X, Xu C, Liu X, Wang L, Yu M, Li D, Zhu Y, Du M. SCM-198 protects endometrial stromal cells from oxidative damage through Bax/Bcl-2 and ERK signaling pathways. Acta Biochim Biophys Sin (Shanghai) 2019; 51:580-587. [PMID: 31032514 DOI: 10.1093/abbs/gmz035] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Indexed: 01/12/2023] Open
Abstract
Increasing amounts of evidence demonstrated that accumulative reactive oxygen species (ROS) and apoptosis of human endometrial stromal cells (ESCs) are closely associated with endometrial dysfunction induced by oxidative stress, which plays an important role in the pathological process of multiple gynecological and reproduction-related diseases. SCM-198, an alkaloid active component of Leonurus japonicas Houtt, has been reported to have anti-oxidative activity. However, the specific mechanisms of SCM-198 in the prevention of endometrial damage remain unknown. In the present study, we assessed the effect of SCM-198 on hydrogen peroxide (H2O2)-induced oxidative injury in ESCs. ESCs were pretreated with SCM-198 for 4 h and then challenged with H2O2. Morphology changes, apoptosis rate, and intracellular ROS production were measured to assess the level of oxidative injury. Flow cytometry and western blot analysis were performed to detect the expression levels of Bax, Bcl-2, active-caspase-3, and mitogen-activated protein kinases pathways. Classic inflammation cytokines were measured by real-time polymerase chain reactions. Our results showed that SCM-198 attenuated apoptosis and ROS generation of ESCs induced by H2O2. H2O2 induced the apparent apoptotic characteristics, including fragmentation of DNA, upregulation of Bax/Bcl2, activation of caspase-3, and secretion of inflammation cytokines, which were all ameliorated by SCM-198. Furthermore, H2O2-induced apoptosis-related ERK1/2 pathway activation was restrained by SCM-198 pretreatment. These findings suggested that SCM-198 could protect ESCs from oxidative injury, mainly by inhibiting oxidative stress and reducing apoptosis.
Collapse
Affiliation(s)
- Yunyun Li
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Yikong Lin
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Xixi Huang
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Chunfang Xu
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Xinhua Liu
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Li Wang
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Min Yu
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Dajin Li
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Yizhun Zhu
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
- State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau SAR, China
| | - Meirong Du
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| |
Collapse
|
43
|
Yang L, Liu G, Lian K, Qiao Y, Zhang B, Zhu X, Luo Y, Shang Y, Gu XL. Dietary leonurine hydrochloride supplementation attenuates lipopolysaccharide challenge-induced intestinal inflammation and barrier dysfunction by inhibiting the NF-κB/MAPK signaling pathway in broilers. J Anim Sci 2019; 97:1679-1692. [PMID: 30789669 DOI: 10.1093/jas/skz078] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 02/20/2019] [Indexed: 12/20/2022] Open
Abstract
This study was performed to evaluate the beneficial effects of dietary leonurine hydrochloride (LH) supplementation on intestinal morphology and barrier integrity and further illuminate its underlying antioxidant and immunomodulatory mechanisms in lipopolysaccharide (LPS)-treated broilers. A total of 120 1-d-old male broilers (Ross 308) were assigned to 4 treatment groups with 6 replicates of 5 birds per cage. The experiment was designed in a 2 × 2 factorial arrangement with LH (0 or 120 mg/kg) and LPS (injection of saline or 1.5 mg/kg body weight) as treatments. On days 14, 16, 18, and 20 of the trial, broilers were intraperitoneally injected with LPS or physiological saline. Compared with the control group, LPS-challenged broilers showed impaired growth performance (P < 0.05) from day 15 to day 21 of the trial, increased serum diamine oxidase (DAO) and D-lactic acid (D-LA) levels coupled with reduced glutathione (GSH) content and total superoxide dismutase (T-SOD) activity (duodenal and jejunal mucosa), reduced malondialdehyde (MDA) content (duodenal, jejunal, and ileal mucosa), and compromised morphological structure of the duodenum and jejunum. Additionally, LPS challenge increased (P < 0.05) the mRNA expression of proinflammatory cytokine genes and reduced tight junction (TJ) protein expression in the jejunum. However, dietary LH prevented LPS-induced reductions in average daily gain (ADG) and average daily feed intake (ADFI) in broilers. It also alleviated LPS challenge-induced increases in serum DAO levels, MDA content (duodenal and jejunal mucosa), and jejunal crypt depth (P < 0.05) but reduced villus height, GSH content (jejunal mucosa), and T-SOD activity (duodenal and jejunal mucosa) (P < 0.05). Additionally, LH supplementation significantly downregulated the mRNA expression of nuclear factor (NF)-κB, cyclooxygenase-2 (COX-2), and proinflammatory cytokines (TNF-α, IL-1β, and IL-6) and upregulated the mRNA expression of zonula occludens-1 (ZO-1) and Occludin in the jejunal mucosa induced by LPS (P < 0.05). On the other hand, LH administration prevented LPS-induced activation of the p38, extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) mitogen-activated protein kinases (MAPKs) and attenuated IkB alpha (IκBα) phosphorylation and nuclear translocation of NF-κB (p65) in the jejunal mucosa. In conclusion, dietary LH supplementation attenuates intestinal mucosal disruption mainly by accelerating the expression of TJ proteins and inhibiting activation of the NF-κB/MAPK signaling pathway.
Collapse
Affiliation(s)
- Li Yang
- College of Animal Science and Technology, Shihezi University, Xinjiang, People's Republic of China
| | - Gang Liu
- College of Animal Science and Technology, Shihezi University, Xinjiang, People's Republic of China
| | - Kexun Lian
- College of Animal Science and Technology, Shihezi University, Xinjiang, People's Republic of China
| | - Yanjie Qiao
- College of Animal Science and Technology, Shihezi University, Xinjiang, People's Republic of China
| | - Baojun Zhang
- College of Animal Science and Technology, Shihezi University, Xinjiang, People's Republic of China
| | - Xiaoqing Zhu
- College of Animal Science and Technology, Shihezi University, Xinjiang, People's Republic of China
| | - Yan Luo
- College of Animal Science and Technology, Shihezi University, Xinjiang, People's Republic of China
| | - Yunxia Shang
- College of Animal Science and Technology, Shihezi University, Xinjiang, People's Republic of China
| | - Xin-Li Gu
- College of Animal Science and Technology, Shihezi University, Xinjiang, People's Republic of China
| |
Collapse
|
44
|
Xie YZ, Zhang XJ, Zhang C, Yang Y, He JN, Chen YX. Protective effects of leonurine against ischemic stroke in mice by activating nuclear factor erythroid 2-related factor 2 pathway. CNS Neurosci Ther 2019; 25:1006-1017. [PMID: 31087454 PMCID: PMC6698971 DOI: 10.1111/cns.13146] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 04/08/2019] [Accepted: 04/11/2019] [Indexed: 12/14/2022] Open
Abstract
Aims Leonurine has been shown to trigger antioxidant responses during ischemic stroke, and nuclear factor erythroid 2‐related factor 2 (Nrf‐2) imparts protective effects against oxidative injury. The present study has determined that leonurine prevents ischemic injury of brain tissues via Nrf‐2 pathway activation. Methods Male ICR mice and Nrf‐2−/− mice were subjected to permanent middle cerebral artery occlusion (pMCAO) and received leonurine treatment at 2 hours after pMCAO by intraperitoneal injection. Neurological deficit scores as well as infarct volume were assessed to determine the neuroprotective role of leonurine. Nrf‐2 was investigated using Western blotting and real‐time polymerase chain reaction (RT‐PCR) analysis to elucidate the neuroprotective mechanism of leonurine. Commercial kits were employed to determine reactive oxygen species (ROS), superoxide (SOD), catalase (CAT), glutathione peroxidase (GSH‐Px), malonaldehyde (MDA), and glutathione (GSH). Vascular endothelial growth factor (VEGF) was evaluated by Western blotting and RT‐PCR analysis, and VEGF was localized using immunofluorescence. Results The application of leonurine on ICR mice resulted in an improvement in neurological deficit scores and a reduction in infarct volume. Leonurine upregulated nuclear Nrf‐2 protein and increased total Nrf‐2 protein expression and mRNA levels. Leonurine regulated SOD, MDA, CAT, GSH, and GSH‐Px, and it significantly inhibited ROS production in ICR mice. Leonurine improved VEGF expression and increased VEGF expression in neurons, astrocytes, and endothelial cells. However, leonurine had no obvious beneficial effects on Nrf‐2−/− mice. Conclusions Leonurine exerted neuroprotective effects, promoted antioxidant responses, and upregulated VEGF expression by activating the Nrf‐2 pathway.
Collapse
Affiliation(s)
- Yan-Zhao Xie
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, China.,The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiang-Jian Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, China
| | - Cong Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, China
| | - Yang Yang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, China
| | - Jun-Na He
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, China
| | - Yan-Xia Chen
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, China.,Department of Endocrinology, Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
45
|
Zhao H, Zheng T, Yang X, Fan M, Zhu L, Liu S, Wu L, Sun C. Cryptotanshinone Attenuates Oxygen-Glucose Deprivation/ Recovery-Induced Injury in an in vitro Model of Neurovascular Unit. Front Neurol 2019; 10:381. [PMID: 31057477 PMCID: PMC6482155 DOI: 10.3389/fneur.2019.00381] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 03/28/2019] [Indexed: 12/11/2022] Open
Abstract
Cryptotanshinone (CTs), an active component isolated from the root of Salvia miltiorrhiza (SM), has been shown to exert potent neuroprotective property. We here established an oxygen-glucose deprivation/recovery (OGD/R)-injured Neurovascular Unit (NVU) model in vitro to observe the neuroprotective effects of CTs on cerebral ischemia/reperfusion injury (CIRI), and explore the underlying mechanisms. CTs was observed to significantly inhibit the OGD/R-induced neuronal apoptosis, and decease the activation of Caspase-3 and the degradation of poly-ADP-ribose polymerase (PARP), as well as the increase of Bax/Bcl-2 ratio in neurons under OGD/R condition. The inhibitory effects of CTs on neuron apoptosis were associated with the blocking of mitogen-activated protein kinase (MAPK) signaling pathway. CTs also remarkably ameliorated OGD/R-induced reduction of transepithelial electrical resistance (TEER) values and the increase of transendothelial permeability coefficient (Pe) of sodium fluorescein (SF) by upregulating the expression of ZO-1, Claudin-5, and Occludin in brain microvascular endothelial cells (BMECs), which might be related to the down-regulation of matrix metalloproteinase (MMP)-9 expression. Based on these findings, CTs may play a neuroprotective role in OGD/R injure in NVU models in vitro by inhibiting cell apoptosis and alleviating the damage of blood-brain barrier (BBB).
Collapse
Affiliation(s)
- Hongye Zhao
- Department of Physiology and Key Laboratory of Brain Diseases of Liaoning Province, School of Basic Medical Sciences, Dalian Medical University, Dalian, China.,Department of Physiology, School of Basic Medical Sciences, Qiqihar Medical University, Qiqihar, China
| | - Tiezheng Zheng
- Department of Physiology and Key Laboratory of Brain Diseases of Liaoning Province, School of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xiaohan Yang
- Department of Physiology and Key Laboratory of Brain Diseases of Liaoning Province, School of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Ming Fan
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Lingling Zhu
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Shuhong Liu
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Liying Wu
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Changkai Sun
- Department of Physiology and Key Laboratory of Brain Diseases of Liaoning Province, School of Basic Medical Sciences, Dalian Medical University, Dalian, China.,Department of Biomedical Engineering, Faculty of Electronic Information and Electrical Engineering & Research Center for the Control Engineering of Translational Precision Medicine, Dalian University of Technology, Dalian, China
| |
Collapse
|
46
|
Autoimmunity in acute ischemic stroke and the role of blood-brain barrier: the dark side or the light one? Front Med 2019; 13:420-426. [PMID: 30929189 DOI: 10.1007/s11684-019-0688-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Accepted: 12/27/2018] [Indexed: 02/07/2023]
Abstract
This article presents a synopsis of the current data on the mechanisms of blood-brain barrier (BBB) alteration and autoimmune response in acute ischemic stroke. Most researchers confirm the relationship between the severity of immunobiochemical changes and clinical outcome of acute ischemic stroke. Ischemic stroke is accompanied by aseptic inflammation, which alters the brain tissue and exposes the co-stimulatory molecules of the immune system and the neuronal antigens. To date, BBB is not considered the border between the immune system and central nervous system, and the local immune subsystems are found within and behind the BBB. BBB disruption contributes to the leakage of brain autoantigens and induction of secondary autoimmune response to neuronal antigens and long-term inflammation. Glymphatic system function is altered and jeopardized both in hemorrhagic and ischemic stroke types. The receptors of innate immunity (toll-like receptor-2 and toll-like receptor-4) are also involved in acute ischemia-reperfusion injury. Immune response is related to the key processes of blood clotting and fibrinolysis. At the same time, the stroke-induced immune activation may promote reparation phenomena in the brain. Subsequent research on the reduction of the acute ischemic brain injury through the target regulation of the immune response is promising.
Collapse
|
47
|
Ranjbar Taklimie F, Gasterich N, Scheld M, Weiskirchen R, Beyer C, Clarner T, Zendedel A. Hypoxia Induces Astrocyte-Derived Lipocalin-2 in Ischemic Stroke. Int J Mol Sci 2019; 20:ijms20061271. [PMID: 30871254 PMCID: PMC6471434 DOI: 10.3390/ijms20061271] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 02/22/2019] [Accepted: 03/07/2019] [Indexed: 12/18/2022] Open
Abstract
Ischemic stroke causes rapid hypoxic damage to the core neural tissue which is followed by graded chronological tissue degeneration in the peri-infarct zone. The latter process is mainly triggered by neuroinflammation, activation of inflammasomes, proinflammatory cytokines, and pyroptosis. Besides microglia, astrocytes play an important role in the fine-tuning of the inflammatory network in the brain. Lipocalin-2 (LCN2) is involved in the control of innate immune responses, regulation of excess iron, and reactive oxygen production. In this study, we analyzed LCN2 expression in hypoxic rat brain tissue after ischemic stroke and in astrocyte cell cultures receiving standardized hypoxic treatment. Whereas no LCN2-positive cells were seen in sham animals, the number of LCN2-positive cells (mainly astrocytes) was significantly increased after stroke. In vitro studies with hypoxic cultured astroglia revealed that LCN2 expression is significantly increased after only 2 h, then further increased, followed by a stepwise decline. The expression pattern of several proinflammatory cytokines mainly followed that profile in wild type (WT) but not in cultured LCN2-deficient astrocytes. Our data revealed that astrocytes are an important source of LCN2 in the peri-infarct region under hypoxic conditions. However, we must also stress that brain-intrinsic LCN2 after the initial hypoxia period might come from other sources such as invaded immune cells and peripheral organs via blood circulation. In any case, secreted LCN2 might have an influence on peripheral organ functions and the innate immune system during brain hypoxia.
Collapse
Affiliation(s)
| | - Natalie Gasterich
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany.
| | - Miriam Scheld
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany.
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH Aachen University Hospital, 52074 Aachen, Germany.
| | - Cordian Beyer
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany.
| | - Tim Clarner
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany.
| | - Adib Zendedel
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany.
| |
Collapse
|
48
|
New progress in the approaches for blood–brain barrier protection in acute ischemic stroke. Brain Res Bull 2019; 144:46-57. [DOI: 10.1016/j.brainresbull.2018.11.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/10/2018] [Accepted: 11/13/2018] [Indexed: 02/06/2023]
|
49
|
Guo T, Wang Y, Guo Y, Wu S, Chen W, Liu N, Wang Y, Geng D. 1, 25-D 3 Protects From Cerebral Ischemia by Maintaining BBB Permeability via PPAR-γ Activation. Front Cell Neurosci 2018; 12:480. [PMID: 30618630 PMCID: PMC6304345 DOI: 10.3389/fncel.2018.00480] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 11/23/2018] [Indexed: 12/23/2022] Open
Abstract
The blood-brain barrier (BBB) is a physical and biochemical barrier that maintains cerebral homeostasis. BBB dysfunction in an ischemic stroke, results in brain injury and subsequent neurological impairment. The aim of this study was to determine the possible protective effects of 1, 25-dihydroxyvitamin D3 [1, 25(OH)2D3, 1, 25-D3, vit D] on BBB dysfunction, at the early stages of an acute ischemic brain injury. We analyzed the effects of 1, 25-D3 on BBB integrity in terms of histopathological changes, the neurological deficit, infarct size and the expression of brain derived neurotrophic factor (BDNF), in a middle cerebral artery occlusion/reperfusion (MCAO/R) rat model. BBB permeability and the expression of permeability-related proteins in the brain were also evaluated by Evans blue (EB) staining and Western blotting respectively. To determine the possible mechanism underlying the role of 1, 25-D3 in BBB maintenance, after MCAO/R, the rats were treated with the specific peroxisome proliferator-activated receptor gamma (PPARγ) inhibitor GW9662. Supplementation with 1, 25-D3 markedly improved the neurological scores of the rats, decreased the infarct volume, prevented neuronal deformation and upregulated the expression of the tight junction (TJ) and BDNF proteins in their brains. Furthermore, it activated PPARγ but downregulated neuro-inflammatory cytokines such as nuclear factor kappa-B (NF-κB) and tumor necrosis factor-α (TNF-α), after MCAO/R. Taken together, 1, 25-D3 protects against cerebral ischemia by maintaining BBB permeability, upregulating the level of BDNF and inhibiting PPARγ-mediated neuro-inflammation.
Collapse
Affiliation(s)
- Ting Guo
- Department of Neurology, Xuzhou Medical University, Xuzhou, China
| | - Yanqiang Wang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Neurology, The Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Yuanfang Guo
- Department of Respiratory Medicine, Ganyu District People’s Hospital, Lianyungang, China
| | - Shuguang Wu
- Department of Anesthesiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Weiwei Chen
- Department of Neurology, The Central Hospital of Xuzhou, Xuzhou, China
| | - Na Liu
- Department of Neurology, Xuzhou Medical University, Xuzhou, China
| | - Yu Wang
- Department of Neurology, Xuzhou Medical University, Xuzhou, China
| | - Deqin Geng
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
50
|
Hu S, Wu Y, Zhao B, Hu H, Zhu B, Sun Z, Li P, Du S. Panax notoginseng Saponins Protect Cerebral Microvascular Endothelial Cells against Oxygen-Glucose Deprivation/Reperfusion-Induced Barrier Dysfunction via Activation of PI3K/Akt/Nrf2 Antioxidant Signaling Pathway. Molecules 2018; 23:molecules23112781. [PMID: 30373188 PMCID: PMC6278530 DOI: 10.3390/molecules23112781] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 10/16/2018] [Accepted: 10/25/2018] [Indexed: 12/15/2022] Open
Abstract
Oxidative stress plays a critical role in cerebral ischemia/reperfusion (I/R)-induced blood-brain barrier (BBB) disruption. Panax notoginseng saponins (PNS) possess efficient antioxidant activity and have been used in the treatment of cerebral ischemic stroke in China. In this study, we determined the protective effects of PNS on BBB integrity and investigated the underlying mechanism in cerebral microvascular endothelial cells (bEnd.3) exposed to oxygen-glucose deprivation/reperfusion (OGD/R). MTT and LDH release assays revealed that PNS mitigated the OGD/R-induced cell injury in a dose-dependent manner. TEER and paracellular permeability assays demonstrated that PNS alleviated the OGD/R-caused disruption of BBB integrity. Fluorescence probe DCFH-DA showed that PNS suppressed ROS generation in OGD/R-treated cells. Immunofluorescence and western blot analysis indicated that PNS inhibited the degradation of tight junction proteins triggered by OGD/R. Moreover, mechanism investigations suggested that PNS increased the phosphorylation of Akt, the activity of nuclear Nrf2, and the expression of downstream antioxidant enzyme HO-1. All the effects of PNS could be reversed by co-treatment with PI3K inhibitor LY294002. Taken together, these observations suggest that PNS may act as an extrinsic regulator that activates Nrf2 antioxidant signaling depending on PI3K/Akt pathway and protects against OGD/R-induced BBB disruption in vitro.
Collapse
Affiliation(s)
- Shaonan Hu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Yali Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Bo Zhao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Haiyan Hu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Baochen Zhu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Zongxi Sun
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Pengyue Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Shouying Du
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|