1
|
Zhao X, Gu C, Wang M, Huang S, Gong X, Kang M, Zhang T, Shen J, Yang X, Xi Y, Pan J. Human Neural Progenitor Cell-Derived Exosomes Deliver miR- 100 - 5p Targeting NOX4 mRNA to Alleviate Oxidative Stress in Acute Ischemia Injury. Mol Neurobiol 2025:10.1007/s12035-025-04952-z. [PMID: 40402409 DOI: 10.1007/s12035-025-04952-z] [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: 12/29/2023] [Accepted: 04/13/2025] [Indexed: 05/23/2025]
Abstract
The prevention and treatment of acute ischemic stroke have been longstanding challenges. Therapies targeting angiogenesis hold promising potential for ischemic injury repair. Reactive oxygen species (ROS) production, induced by the overexpression of NADPH oxidase 4 (NOX4), is a key factor that inhibits angiogenesis during the acute phase of ischemia/reperfusion. Therefore, targeting NOX4 gene expression can reduce ROS production and promote angiogenesis. In this study, human neural progenitor cell (hNPC)-derived-exosomal miRNAs were identified using high-throughput sequencing and online database, with miR-100-5p showing potential to suppress NOX4 expression. We then incorporated exogenous miR-100-5p into hNPC-derived exosomes through electroporation. These miR-100-5p-loaded exosomes were then applied to in vitro models of mouse brain microvascular endothelial cells (bEND.3) subjected to oxygen glucose deprivation and reperfusion (OGD/R). The results showed that miR-100-5p could significantly reduce NOX4 expression and ROS levels induced by OGD/R in bEND.3 cells. Similarly, in vivo analysis showed that mice models of middle cerebral artery occlusion (MCAO) injected with hNPC-derived exosomes loaded with miR-100-5p exhibited smaller brain infarct size, reduced apoptosis, and improved neurological performance compared to MCAO mice receiving PBS. Our findings demonstrate the successful delivery of miR-100-5p via hNPC-derived exosomes and its protective effect on brain microvascular endothelial cells following ischemia injury.
Collapse
Affiliation(s)
- Xianlei Zhao
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China
- Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, 310000, China
- Center for Genetic Medicine, Zhejiang University Lnternational Institute of Medicine, Yiwu, 322000, China
- Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, 310000, China
- The Women's Hospital and Institute of Genetics, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Chenjie Gu
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Min Wang
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China
- Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, 310000, China
- Center for Genetic Medicine, Zhejiang University Lnternational Institute of Medicine, Yiwu, 322000, China
- Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, 310000, China
- The Women's Hospital and Institute of Genetics, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Sicong Huang
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xinghan Gong
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China
- Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, 310000, China
- Center for Genetic Medicine, Zhejiang University Lnternational Institute of Medicine, Yiwu, 322000, China
- Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, 310000, China
- The Women's Hospital and Institute of Genetics, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Muxue Kang
- The Women's Hospital and Institute of Genetics, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Tiesong Zhang
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jian Shen
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xiaohang Yang
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China
- Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, 310000, China
- Center for Genetic Medicine, Zhejiang University Lnternational Institute of Medicine, Yiwu, 322000, China
- Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, 310000, China
- The Women's Hospital and Institute of Genetics, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yongmei Xi
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China.
- Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, 310000, China.
- Center for Genetic Medicine, Zhejiang University Lnternational Institute of Medicine, Yiwu, 322000, China.
- Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, 310000, China.
- The Women's Hospital and Institute of Genetics, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Jianwei Pan
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| |
Collapse
|
2
|
Liu L, Ma Z, Han Q, Meng W, Ye H, Zhang T, Xia Y, Xiang Z, Ke Y, Guan X, Shi Q, Ataullakhanov FI, Panteleev M. Phenylboronic Ester-Bridged Chitosan/Myricetin Nanomicelle for Penetrating the Endothelial Barrier and Regulating Macrophage Polarization and Inflammation against Ischemic Diseases. ACS Biomater Sci Eng 2023. [PMID: 37327139 DOI: 10.1021/acsbiomaterials.3c00414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
The brain and liver are more susceptible to ischemia and reperfusion (IR) injury (IRI), which triggers the reactive oxygen species (ROS) burst and inflammatory cascade and results in severe neuronal damage or hepatic injury. Moreover, the damaged endothelial barrier contributes to proinflammatory activity and limits the delivery of therapeutic agents such as some macromolecules and nanomedicine despite the integrity being disrupted after IRI. Herein, we constructed a phenylboronic-decorated chitosan-based nanoplatform to deliver myricetin, a multifunctional polyphenol molecule for the treatment of cerebral and hepatic ischemia. The chitosan-based nanostructures are widely studied cationic carriers for endothelium penetration such as the blood-brain barrier (BBB) and sinusoidal endothelial barrier (SEB). The phenylboronic ester was chosen as the ROS-responsive bridging segment for conjugation and selective release of myricetin molecules, which meanwhile scavenged the overexpressed ROS in the inflammatory environment. The released myricetin molecules fulfill a variety of roles including antioxidation through multiple phenolic hydroxyl groups, inhibition of the inflammatory cascade by regulation of the macrophage polarization from M1 to M2, and endothelial injury repairment. Taken together, our present study provides valuable insight into the development of efficient antioxidant and anti-inflammatory platforms for potential application against ischemic disease.
Collapse
Affiliation(s)
- Lei Liu
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Zhifang Ma
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Qiaoyi Han
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Wei Meng
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Hongbo Ye
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Tianci Zhang
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Yu Xia
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Zehong Xiang
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Yue Ke
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Xinghua Guan
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Qiang Shi
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
- Key Laboratory of Polymeric Materials Design and Synthesis for Biomedical Function, Soochow University, Suzhou, Jiangsu 215123, China
| | - Fazly I Ataullakhanov
- Dmitry Rogachev National Research Center of Pediatric Hematology, Oncology and Immunology, Moscow 117198, Russia
- Faculty of Physics, Lomonosov Moscow State University, Leninskie Gory, 1, build. 2, GSP-1, Moscow 119991, Russia
| | - Mikhail Panteleev
- Dmitry Rogachev National Research Center of Pediatric Hematology, Oncology and Immunology, Moscow 117198, Russia
| |
Collapse
|
3
|
Ning K, Gao R. Icariin protects cerebral neural cells from ischemia‑reperfusion injury in an in vitro model by lowering ROS production and intracellular calcium concentration. Exp Ther Med 2023; 25:151. [PMID: 36911386 PMCID: PMC9995791 DOI: 10.3892/etm.2023.11849] [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: 10/12/2022] [Accepted: 01/26/2023] [Indexed: 02/18/2023] Open
Abstract
Ischemia is one of the major causes of stroke. The present study investigated the protection of cultured neural cells by icariin (ICA) against ischemia-reperfusion (I/R) injury and possible mechanisms underlying the protection. Neural cells were isolated from neonatal rats and cultured in vitro. The cells were subjected to oxygen-glucose deprivation and reoxygenation (OGD-R) as an I/R mimic to generate I/R injury, and were post-OGD-R treated with ICA. Following the treatments, cell viability, apoptosis, reactive oxygen species (ROS), lactate dehydrogenase (LDH), superoxide dismutase (SOD) and Ca2+ concentration were assessed using Cell Counting Kit-8 assay, flow cytometry, CyQUANT™ LDH Cytotoxicity Assay, H2DCFDA and SOD colorimetric activity kit. After OGD-R, considerable I/R injury was observed in the neural cells, as indicated by reduced cell viability, increased apoptosis and increased production of ROS and LDH (P<0.05). Cellular Ca2+ concentration was also increased, while SOD activity remained unchanged. Post-OGD-R ICA treatments increased cell viability up to 87.1% (P<0.05) and reduced apoptosis as low as 6.6% (P<0.05) in a concentration-dependent manner. The treatments also resulted in fewer ROS (P<0.05), lower extracellular LDH content (440.5 vs. 230.3 U/l; P<0.05) and reduced Ca2+ increase (P<0.05). These data suggest that ICA protects the neural cells from I/R injury in an in vitro model through antioxidation activity and maintaining cellular Ca2+ homeostasis. This function may be explored as a potential therapeutic strategy for ischemia-related diseases after further in vivo studies.
Collapse
Affiliation(s)
- Ke Ning
- Department of International Medicine, Affiliated Zhongshan Hospital, Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Rong Gao
- Surgical Intensive Care Unit, Affiliated Zhongshan Hospital, Dalian University, Dalian, Liaoning 116001, P.R. China
| |
Collapse
|
4
|
Shen D, Wang H, Zhu H, Jiang C, Xie F, Zhang H, Lv Q, Liu Q, Wang Z, Qi N, Wang H. Pre-clinical efficacy evaluation of human umbilical cord mesenchymal stem cells for ischemic stroke. Front Immunol 2023; 13:1095469. [PMID: 36726973 PMCID: PMC9885855 DOI: 10.3389/fimmu.2022.1095469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 12/29/2022] [Indexed: 01/15/2023] Open
Abstract
Objective This study explored the underlying therapeutic mechanism of human umbilical cord mesenchymal stem cells (hUCMSCs) for ischemic stroke (IS), and determined the optimal administration time windows and dose-effect relationship. Methods The levels of SDF-1α, IL-10, IL-6, TNF-α, BDNF, IL-1β, and VEGF-A factors in serum and brain tissue lysate were measured by ELISA. The pathological status of brain tissues was evaluated by Hematoxylin-Eosin (HE) staining, and apoptosis of nerve cells was detected by tunel. The protein expression of CXCR-4, NeuN, and Nestin in the brain tissues was assessed through immunofluorescence. The balance beam, forelimb muscle strength, and limb placement were tested on MCAO rats at different time points and doses. The infarct area of the rat brain tissues was measured at the end of the experiment. Results The hUCMSC treatment during the acute phase of MCAO significantly reduced the secretion of IL-6, TNF-α, IL-1β but increased IL-10 in serum, and the levels of SDF-α and BDNF in serum and brain tissues lysate were also increased. The pathological results showed that there were more neurons in the treatment group compared to the model group. Immunofluorescence assays showed that the expression of CXCR4、Nestin、NeuN was relatively higher than that in the model group. The d4 and d7 treatment significantly improves the motor function, promotes the recovery of forelimb muscle strength, increases the forelimb placement rate and reduces the scope of cerebral infarction, but the d14 treatment group has less therapeutic effect compared to the d4 and d7 treatment. The 2×107/kg treatment showed the best therapeutic effect, followed by the 1×107/kg treatment, and the worst is 0.5×107/kg treatment from the test of balance beam, forelimb muscle strength, limb placement and the infarct area of the rat brain tissues. Conclusion The hUCMSCs can inhibit the infiltration of inflammatory cells in the brain tissue, and promote the repair of brain tissue structure and function. Early intervention by injecting high-dose of hUCMSCs can significantly improve the recovery of neurological/motor function and reduce the size of cerebral infarction in rats.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Nianmin Qi
- *Correspondence: Hao Wang, ; Nianming Qi,
| | - Hao Wang
- *Correspondence: Hao Wang, ; Nianming Qi,
| |
Collapse
|
5
|
Guo R, Chen P, Fu T, Zhang R, Zhu Y, Jin N, Xu H, Xia Y, Tian X. Activation of Delta-Opioid Receptor Protects ARPE19 Cells against Oxygen-Glucose Deprivation/Reoxygenation-Induced Necroptosis and Apoptosis by Inhibiting the Release of TNF- α. J Ophthalmol 2022; 2022:2285663. [PMID: 36457949 PMCID: PMC9708366 DOI: 10.1155/2022/2285663] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 10/20/2022] [Accepted: 11/04/2022] [Indexed: 09/22/2023] Open
Abstract
PURPOSE Retinal ischemia-reperfusion injury (RIRI) is the basis of the pathology that leads to many retinal diseases and induces necroptosis and apoptosis. Tumor necrosis factor-α (TNF-α) is critically involved in necroptosis and apoptosis. Delta-opioid receptor (DOR) activation inhibits TNF-α release in our previous studies, it might prevent necroptosis and apoptosis by inhibiting the release of TNF-α. However, the role of TNF-α and DOR in necroptosis and apoptosis of retinal pigment epithelial (RPE) cells remains largely unknown. Here, we explored the mechanisms of TNF-α and DOR in necroptosis and apoptosis using an oxygen-glucose deprivation/reoxygenation (OGD/R) model of adult retinal pigment epithelial cell line-19 (ARPE19) cells. MATERIALS AND METHODS ARPE19 cells were exposed to OGD/R conditions to mimic RIRI in vitro. Cell viability was quantified using the Cell Counting Kit-8 (CCK-8) assay. Morphological changes were observed by inverted microscopy. TNF-α protein levels in cell lysates were measured by enzyme-linked immunosorbent assay (ELISA). The DOR agonist TAN-67 and antagonist naltrindole (NTI) were used to pretreat cells for 1 or 2 hours before OGD24/R36 administration. Calcein acetoxymethylester/propidium iodide (Calcein-AM/PI) and Terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) staining were used to detect necroptotic and apoptotic ARPE19 cells, respectively. The protein expression of DOR, p-RIP1 (RIP1), p-RIP3 (RIP3), p-MLKL (MLKL), and cleaved Caspase3 (Caspase3) was measured by western blotting. RESULTS OGD severely damaged ARPE19 cells. Prolonged reoxygenation significantly increased TNF-α level and decreased DOR expression in ARPE19 cells. Pretreatment with the DOR agonist TAN-67 (10 µM) significantly improved ARPE19 cell viability after OGD24/R36 by reducing the number of necroptotic and apoptotic cells. Furthermore, DOR activation significantly inhibited TNF-α release and suppressed the expression of proteins related to necroptosis and apoptosis, including p-RIP1, p-RIP3, p-MLKL, and cleaved Caspase3, after OGD24/R36. This effect was reversed by the DOR antagonist NTI. CONCLUSION These results strongly suggest that DOR activation inhibits necroptosis and apoptosis by decreasing TNF-α release, leading to the prevention of OGD/R-induced injury in ARPE19 cells. This study provides an innovative idea for clinical treatment strategies for retinal damage and vision loss due to RIRI.
Collapse
Affiliation(s)
- Runjie Guo
- Experiment Center of Science and Technology, Laboratory Animal Center, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ping Chen
- Experiment Center of Science and Technology, Laboratory Animal Center, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tiantian Fu
- Experiment Center of Science and Technology, Laboratory Animal Center, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ren Zhang
- Shanghai Chinese Medicine Literature Museum, Shanghai 201203, China
| | - Yuan Zhu
- Shanghai Jinshan District Hospital of Traditional Chinese and Western Medicine, Shanghai 201501, China
| | - Nange Jin
- Department of Vision Sciences, University of Houston College of Optometry, Houston, TX 77204, USA
| | - Hong Xu
- Department of Acupuncture-Moxibustion, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yong Xia
- School of Acupuncture-Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xuesong Tian
- Experiment Center of Science and Technology, Laboratory Animal Center, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
6
|
Hu S, Chen Y, Huang S, Liu M, Liu Y, Huang S. Sodium Danshensu protects against oxygen glucose deprivation/reoxygenation-induced astrocytes injury through regulating NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasome and tuberous sclerosis complex-2 (TSC2)/mammalian target of rapamycin (mTOR) pathways. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1097. [PMID: 36388798 PMCID: PMC9652549 DOI: 10.21037/atm-22-2143] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 08/31/2022] [Indexed: 09/18/2023]
Abstract
BACKGROUND Cerebral ischemic stroke is a serious condition with high incidence, mortality, and associated disability. Currently, effective therapeutic options are available for ischemic stroke are limited. Accumulating evidence indicates that sodium Danshensu, mono sodium compound derived from Salvia miltiorrhiza, plays protective roles in ischemic stroke. However, the underlying protective mechanism of sodium Danshensu in cerebral ischemic stroke remains unknown. METHODS In the current study, we explored the role and mechanism of sodium Danshensu on astrocytes exposed to oxygen-glucose deprivation/reoxygenation (OGD/R), which mimics the process of ischemia-reperfusion. The impact of sodium Danshensu on cell viability and apoptosis after OGD/R were evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-dophenyl tetrazolium bromide (MTT) assay and flow cytometry. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) and western blot were used to detect the expression of target messenger RNA (mRNA) and proteins associated with apoptosis and autophagy. The release of lactate dehydrogenase (LDH) was determined, and the production of proinflammatory cytokines were detected using enzyme-linked immunosorbent assay (ELISA) kits. RESULTS It was found that sodium Danshensu could significantly increase cell viability and decrease LDH release and apoptosis. Besides, it inhibited the production of proinflammatory cytokines, including tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, and IL-6. Sodium Danshensu also dose-dependently decreased protein and mRNA levels of nucleotide binding oligomerization NOD-like receptor pyrin domain containing 3 (NLRP3) and high mobility group box 1 (HMGB1), which play a crucial role in promoting ischemic stroke-induced cell injury. Moreover, sodium Danshensu dose-dependently upregulated Beclin 1 expression, downregulated P62 protein expression, and further increased LC3B-II/LC3B-I ratio through inducing autophagy in astrocytes. Additionally, we noticed that sodium Danshensu dose-dependently increased tuberous sclerosis complex-2 (TSC2) protein expression, while significantly reduced the levels of mammalian target of rapamycin (mTOR) in the presence of OGD/R insult. CONCLUSIONS These findings suggest that sodium Danshensu protects against OGD/R-induced injury by modulating the NLRP3 inflammasome and TSC2/mTOR pathways.
Collapse
Affiliation(s)
- Shengzhao Hu
- Department of Emergency, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yingli Chen
- Department of Hematology, Jiangxi Provincial Children’s Hospital, Nanchang, China
| | - Shipeng Huang
- Department of Emergency, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Min Liu
- Department of Emergency, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ying Liu
- Department of Emergency, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shaofang Huang
- Department of Emergency, the First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
7
|
Wang H, Chen Y, Li P, Chen Y, Yu D, Tan Q, Liu X, Guo Z. Biphasic effects of statins on neuron cell functions under oxygen-glucose deprivation and normal culturing conditions via different mechanisms. Pharmacol Res Perspect 2022; 10:e01001. [PMID: 36029136 PMCID: PMC9419152 DOI: 10.1002/prp2.1001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 07/07/2022] [Accepted: 07/26/2022] [Indexed: 11/11/2022] Open
Abstract
While there is a growing interest in the use of statins, HMG‐CoA reductase inhibitors, to treat neurodegenerative diseases, statins are associated with conflicting effects within the central nervous system (CNS) without clear evidence of the underlying mechanisms. This study systematically investigated effects of four statins (atorvastatin, pitavastatin, cerivastatin, and lovastatin) on neuronal cells under pathological condition using an in vitro model depicting ischemic injury, as well as tested under physiological condition. All four statins at micromolar concentrations display toxic effects on neuron cells under physiological condition. Atorvastatin and cerivastatin but not pitavastatin or lovastatin at nanomolar concentrations display protective effects on neuron cells under ischemic injury condition, via decreased ischemic injury‐induced oxidative stress, oxidative damage, and inflammation. Mechanistically, atorvastatin, pitavastatin, and lovastatin induces neuron cell apoptosis via prenylation‐independent manner. Other mechanisms are involved in the pro‐apoptotic effect of cerivastatin. Prenylation is not involved in the protective effects of statins under ischemic injury condition. Our work provides better understanding on the multiple differential effects of statins on neuron cells under physiological condition and ischemic injury, and elucidate their underlying mechanisms, which may be of relevance to the influence of statins in CNS.
Collapse
Affiliation(s)
- Hui Wang
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, China
| | - Yun Chen
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, China
| | - Ping Li
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, China
| | - Yan Chen
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, China
| | - Danfang Yu
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, China
| | - Qian Tan
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, China
| | - Xiaoli Liu
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, China
| | - Zhenli Guo
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, China
| |
Collapse
|
8
|
Lin H, Zhang Y, Dong S, Cai X, Jiang H, Fan Y, Ying K, Du B, Yu P, Yang W. Targeted Therapy of Ischemic Stroke via Crossing the Blood-Brain Barrier Using Edaravone-Loaded Multiresponsive Microgels. ACS APPLIED BIO MATERIALS 2022; 5:4165-4178. [PMID: 36083038 DOI: 10.1021/acsabm.2c00325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Ischemic stroke, as a prevalent neurological disorder, often results in rapid increases in the production of reactive oxygen species (ROS) and inflammatory factors in the focal ischemic area. Though edaravone is an approved treatment, its effect is limited due to its weak ability to cross the blood-brain barrier (BBB) and short half-life. Other effective pharmacological treatment options are clearly lacking. In this study, PNIVDBrF-3-Eda (also named MG-3-Eda) was prepared using a thermo- and pH dual-responsive PNIVDBrF microgel. These were designed with a positively charged network, as synthesized by simultaneous quaternization cross-linking and surfactant-free emulsion copolymerization, to be loaded with the negatively charged edaravone. We then investigated whether such a targeted delivery of edaravone could provide enhanced neuroprotection. Cytotoxicity assays confirmed that the microgel (<1 mg/mL) exhibited promising cytocompatibility with no remarkable effect on cell viability, cell cycle regulation, or apoptosis levels. In vitro and in vivo experiments demonstrated that the microgels could successfully penetrate the blood-brain barrier where efficient BBB crossing was observed after disruption of the BBB due to ischemic injury. This enabled MG-3-Eda to target the cerebral ischemic area and achieve local release of edaravone. Treatment with MG-3-Eda significantly reduced the cerebral infarct area in transient middle cerebral artery occlusion (tMCAO) mice and significantly improved behavioral scores. MG-3-Eda treatment also prevented the reduction in NF200 expression, a neuronal marker protein, and attenuated microglia activation (as detected by Iba1) in the local ischemic area via local antioxidant and anti-inflammatory effects. A superior neuroprotective effect was noted for MG-3-Eda compared to that for free edaravone. Our results indicate that MG-3-Eda administration represents a clear potential treatment for cerebral ischemia via its targeted delivery of edaravone to ischemic areas where it provides significant local antioxidant and anti-inflammatory effects.
Collapse
Affiliation(s)
- Hongwei Lin
- Department of Neurosurgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Yi Zhang
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Shunni Dong
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science & Engineering, Zhejiang University, Hangzhou 310012, China
| | - Xiaobo Cai
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Hui Jiang
- Department of Toxicology and Department of Medical Oncology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Yang Fan
- Department of Toxicology and Department of Medical Oncology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Kaiyue Ying
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Binyang Du
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science & Engineering, Zhejiang University, Hangzhou 310012, China
| | - Peilin Yu
- Department of Toxicology and Department of Medical Oncology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Wei Yang
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| |
Collapse
|
9
|
Van Breedam E, Ponsaerts P. Promising Strategies for the Development of Advanced In Vitro Models with High Predictive Power in Ischaemic Stroke Research. Int J Mol Sci 2022; 23:ijms23137140. [PMID: 35806146 PMCID: PMC9266337 DOI: 10.3390/ijms23137140] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/21/2022] [Accepted: 06/24/2022] [Indexed: 11/16/2022] Open
Abstract
Although stroke is one of the world’s leading causes of death and disability, and more than a thousand candidate neuroprotective drugs have been proposed based on extensive in vitro and animal-based research, an effective neuroprotective/restorative therapy for ischaemic stroke patients is still missing. In particular, the high attrition rate of neuroprotective compounds in clinical studies should make us question the ability of in vitro models currently used for ischaemic stroke research to recapitulate human ischaemic responses with sufficient fidelity. The ischaemic stroke field would greatly benefit from the implementation of more complex in vitro models with improved physiological relevance, next to traditional in vitro and in vivo models in preclinical studies, to more accurately predict clinical outcomes. In this review, we discuss current in vitro models used in ischaemic stroke research and describe the main factors determining the predictive value of in vitro models for modelling human ischaemic stroke. In light of this, human-based 3D models consisting of multiple cell types, either with or without the use of microfluidics technology, may better recapitulate human ischaemic responses and possess the potential to bridge the translational gap between animal-based in vitro and in vivo models, and human patients in clinical trials.
Collapse
|
10
|
Chen M, Wang F, Fan L, Wang H, Gu S. Long Noncoding RNA TUG1 Aggravates Cerebral Ischemia/Reperfusion Injury by Acting as a ceRNA for miR-3072-3p to Target St8sia2. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9381203. [PMID: 35498127 PMCID: PMC9042630 DOI: 10.1155/2022/9381203] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/22/2022] [Accepted: 03/07/2022] [Indexed: 12/20/2022]
Abstract
Long noncoding RNA taurine-upregulated gene 1 (TUG1) is considered to be involved in postischemic cerebral inflammation, whereas polysialic acid (polySia, PSA), the product of St8sia2, constitutes polysialylated neural adhesion cell molecule (PSA-NCAM) in both mice and humans and that cerebral PSA-NCAM level is elevated in neuronal progenitor cells in response to transient focal ischemia. Herein, we aim to identify novel miRNAs that bridge the functions of St8sia2 and TUG1 in ischemia-associated injuries. In both in vivo (C57BL/6J mouse ischemia/reperfusion, I/R model) and in vitro (mouse neuroblastoma N2A cell oxygen glucose deprivation/reoxygenation, OGD model) settings, we observed upregulated TUG1 and St8sia2 after the induction of ischemic injury, accompanied by reduced miR-3072-3p expression. We performed siRNA-induced TUG1 knockdown combined with the induction of ischemic injury; the results showed that inhibiting TUG1 expression led to the reduced infarct area and improved neurological deficit. Through bioinformatics analysis, miR-3072-3p was found to target both St8sia2 and TUG1, which was subsequently verified by the luciferase reporter system and RNA binding protein immunoprecipitation assay. Also, the addition of miR-3072-3p mimic/inhibitor resulted in reduced/elevated St8sia2 expression at the protein level. Further studies revealed that in both in vivo and in vitro settings, TUG1 bound competitively to miR-3072-3p to regulate St8sia2 expression and promote apoptosis. In summary, targeting the TUG1/miR-3072-3p/St8sia2 regulatory cascade, a novel cascade we identified in cerebral ischemia injury, may render feasible therapeutic possibilities for overcoming cerebral ischemic insults.
Collapse
Affiliation(s)
- Miao Chen
- Department of Emergency, The First Affiliated Hospital of Hainan Medical University, No. 31, Longhua Road, Longhua District, Haikou City, Hainan Province 570102, China
| | - Feng Wang
- Neurology Department, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 200137, China
| | - Limin Fan
- The Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, No. 1239, Siping Road, Shanghai 200092, China
| | - Hairong Wang
- Department of Emergency, Xinhua Hospital Affiliated to Shanghai Jiaotong University, School of Medicine, No. 1665, Kongjiang Road, Shanghai 20092, China
| | - Shuo Gu
- Department of Pediatric Neurosurgery, The First Affiliated Hospital of Hainan Medical University, No. 31, Longhua Road, Longhua District, Haikou City, Hainan Province 570102, China
| |
Collapse
|
11
|
The Assessment of Endovascular Therapies in Ischemic Stroke: Management, Problems and Future Approaches. J Clin Med 2022; 11:jcm11071864. [PMID: 35407472 PMCID: PMC8999747 DOI: 10.3390/jcm11071864] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/18/2022] [Accepted: 03/25/2022] [Indexed: 02/06/2023] Open
Abstract
Ischemic stroke accounts for over 80% of all strokes and is one of the leading causes of mortality and permanent disability worldwide. Intravenous administration of recombinant tissue plasminogen activator (rt-PA) is an approved treatment strategy for acute ischemic stroke of large arteries within 4.5 h of onset, and mechanical thrombectomy can be used for large arteries occlusion up to 24 h after onset. Improving diagnostic work up for acute treatment, reducing onset-to-needle time and urgent radiological access angiographic CT images (angioCT) and Magnetic Resonance Imaging (MRI) are real problems for many healthcare systems, which limits the number of patients with good prognosis in real world compared to the results of randomized controlled trials. The applied endovascular procedures demonstrated high efficacy, but some cellular mechanisms, following reperfusion, are still unknown. Changes in the morphology and function of mitochondria associated with reperfusion and ischemia-reperfusion neuronal death are still understudied research fields. Moreover, future research is needed to elucidate the relationship between continuously refined imaging techniques and the variable structure or physical properties of the clot along with vascular permeability and the pleiotropism of ischemic reperfusion lesions in the penumbra, in order to define targeted preventive procedures promoting long-term health benefits.
Collapse
|
12
|
Cai SC, Yi CA, Hu XS, Tang GY, Yi LM, Li XP. Isoquercitrin Upregulates Aldolase C Through Nrf2 to Ameliorate OGD/R-Induced Damage in SH-SY5Y Cells. Neurotox Res 2021; 39:1959-1969. [PMID: 34773594 DOI: 10.1007/s12640-021-00430-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 10/09/2021] [Accepted: 10/15/2021] [Indexed: 10/19/2022]
Abstract
Isoquercitrin (ISO), an extract from Chinese traditional herb, exhibits potent neuroprotective roles in various disease models. However, its role in stroke is not fully understood. We established oxygen-glucose deprivation and reoxygenation (OGD/R) model in SH-SY5Y cell to study the roles of ISO in stroke. In the experiment, the changes of LDH level and cell viability (MTT) were analyzed. Apoptotic cells stained with anti-Annexin V antibody and propidium iodide (PI) were detected by flow cytometry. The mRNA and protein level of aldolase C (ALDOC) and nuclear factor erythroid 2-related factor (Nrf2) was determined by real-time quantitative polymerase chain reaction (qPCR) and Western blotting assay, respectively. The localization of Nrf2 was investigated by immunofluorescent assay. OGD/R reduced cell viability via inducing cell apoptosis, while ISO treatment reduced the level of apoptosis in OGD/R-treated SH-SY5Y cells ISO rescued OGD/R-treated cells. Mechanistically, the expression of Nrf2 and ALDOC was upregulated upon ISO treatment, while knockdown of ALDOC diminished the activation of autophagy and hence inhibited ISO-mediated protective activity. We further demonstrated that ISO enhanced ALDOC transcription by promoting nuclear translocation of Nrf2, and suppression of Nrf2 decreased the expression of ALDOC. Our data revealed that ISO exhibited neuroprotective activity in OGD/R model through Nrf2-ALDOC-autopagy axis and highlighted the potential application of ISO in stroke treatment.
Collapse
Affiliation(s)
- Shi-Chang Cai
- Department of Human Anatomy, School of Medicine, Hunan University of Medicine, Huaihua, 418000, Hunan Province, People's Republic of China.,School of Medicine, Hunan Provincial Key Laboratory for Synthetic Biology of Traditional Chinese Medicine, Hunan University of Medicine, Huaihua, 418000, Hunan Province, People's Republic of China
| | - Chuan-An Yi
- Medical Morphology Experimental Center of School of Medicine, Hunan University of Medicine, Huaihua, 418000, Hunan Province, People's Republic of China
| | - Xiang-Shang Hu
- Department of Human Anatomy, School of Medicine, Hunan University of Medicine, Huaihua, 418000, Hunan Province, People's Republic of China
| | - Gen-Yun Tang
- School of Medicine, Hunan Provincial Key Laboratory for Synthetic Biology of Traditional Chinese Medicine, Hunan University of Medicine, Huaihua, 418000, Hunan Province, People's Republic of China
| | - Li-Ming Yi
- Department of Human Anatomy, School of Medicine, Hunan University of Medicine, Huaihua, 418000, Hunan Province, People's Republic of China
| | - Xiu-Ping Li
- School of Public Health and Laboratory Medicine, Hunan University of Medicine, No.492 Jinxi South Road, Hecheng District, Huaihua, 418000, Hunan Province, People's Republic of China.
| |
Collapse
|
13
|
Su F, Yang H, Guo A, Qu Z, Wu J, Wang Q. Mitochondrial BK Ca Mediates the Protective Effect of Low-Dose Ethanol Preconditioning on Oxygen-Glucose Deprivation and Reperfusion-Induced Neuronal Apoptosis. Front Physiol 2021; 12:719753. [PMID: 34759831 PMCID: PMC8573145 DOI: 10.3389/fphys.2021.719753] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 09/22/2021] [Indexed: 11/13/2022] Open
Abstract
Ischemia-reperfusion (I/R) injury contributes to the morbidity and mortality of ischemic strokes. As an in vitro model, oxygen-glucose deprivation and reperfusion (OGD/R) exposure induces neuronal injury. Low-dose ethanol preconditioning (EtOH-PC) was reported to alleviate neuronal apoptosis during OGD/R. However, whether the mitochondrial BKCa (mitoBKCa) channel is involved in the neuroprotective effect of EtOH-PC during OGD/R is not clearly defined. This study attempts to explore the mediation of the mitoBKCa channel in the neuroprotective effect of EtOH-PC on OGD/R-induced neuronal apoptosis and the underlying mechanisms. OGD/R model was established using primary cortical neurons that were preincubated with ethanol. Subsequently, the cell viability was measured by CCK-8 assay, and the apoptotic cells were determined by TUNEL assay. Annexin V/7-AAD staining and mitochondrial membrane potential using JC-10 were detected by flow cytometry. Western blot analysis was performed to check the apoptosis-related proteins. In the mixed primary culture, 95% neurofilament-positive cells were cortical neurons. Low-dose EtOH-PC (10 mmol/L) for 24 h significantly attenuated the OGD2h/R24h-induced neuronal apoptosis through activating the BKCa channel. Further investigations suggested that ethanol pretreatment increased the mitochondrial membrane potential (MMP) and downregulated the production of cleaved caspase 3 in OGD/R-injured neurons by activating the mitoBKCa channel. Low-dose ethanol pretreatment significantly attenuated the OGD/R-induced neuronal apoptosis mediated by the mitoBKCa channel which modulated the mitochondrial function by impeding the uncontrolled opening of mitochondrial permeability transition pore (MPTP).
Collapse
Affiliation(s)
- Fang Su
- Department of Neurology, The Fourth Hospital of Harbin Medical University, Harbin, China.,Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Huajun Yang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Anchen Guo
- China National Clinical Research Center for Neurological Diseases, Beijing, China.,Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Zhengyi Qu
- Department of Neurology, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Jianping Wu
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Qun Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| |
Collapse
|
14
|
Amruta N, Bix G. ATN-161 Ameliorates Ischemia/Reperfusion-induced Oxidative Stress, Fibro-inflammation, Mitochondrial damage, and Apoptosis-mediated Tight Junction Disruption in bEnd.3 Cells. Inflammation 2021; 44:2377-2394. [PMID: 34420157 PMCID: PMC8380192 DOI: 10.1007/s10753-021-01509-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/25/2021] [Accepted: 06/27/2021] [Indexed: 12/21/2022]
Abstract
We have previously demonstrated the significance of endothelial cell-expressed α5β1 integrin in ischemic stroke, having shown that α5β1 integrin endothelial cell-selective knockout mice are significantly resistance to ischemic stroke injury via preservation of the tight junction protein claudin-5 and subsequent stabilization of the blood–brain barrier (BBB). In addition, inhibition of α5β1 by the small peptide noncompetitive integrin α5 inhibitor, ATN-161, is beneficial in a mouse model of ischemic stroke through reduction of infarct volume, edema, stabilization of the BBB, and reduced inflammation and immune cell infiltration into the brain. In continuation with our previous findings, we have further evaluated the mechanistic role of ATN-161 in vitro and found that oxygen and glucose deprivation and reperfusion (OGD/R)-induced inflammation, oxidative stress, apoptosis, mitochondrial depolarization, and fibrosis attenuate tight junction integrity via induction of α5, NLRP3, p-FAK, and p-AKT signaling in mouse brain endothelial cells. ATN-161 treatment (10 µM) effectively inhibited OGD/R-induced extracellular matrix (ECM) deposition by reducing integrin α5, MMP-9, and fibronectin expression, as well as reducing oxidative stress by reducing mitochondrial superoxide radicals, intracellular ROS, inflammation by reducing NLRP3 inflammasome, tight junction loss by reducing claudin-5 and ZO-1 expression levels, mitochondrial damage by inhibiting mitochondrial depolarization, and apoptosis via regulation of p-FAK and p-AKT levels. Taken together, our results further support therapeutically targeting α5 integrin with ATN-161, a safe, well-tolerated, and clinically validated peptide, in ischemic stroke.
Collapse
Affiliation(s)
- Narayanappa Amruta
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, Room 1349, 131 S. Robertson, Ste 1300, New Orleans, LA, 70112, USA
| | - Gregory Bix
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, Room 1349, 131 S. Robertson, Ste 1300, New Orleans, LA, 70112, USA. .,Department of Neurology, Tulane University School of Medicine, New Orleans, LA, 70112, USA. .,Tulane Brain Institute, Tulane University, New Orleans, LA, 70112, USA. .,Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, Room 1349, 131 S. Robertson, Ste 1300, New Orleans, LA, 70112, USA.
| |
Collapse
|
15
|
Ohta S. Direct Targets and Subsequent Pathways for Molecular Hydrogen to Exert Multiple Functions: Focusing on Interventions in Radical Reactions. Curr Pharm Des 2021; 27:595-609. [PMID: 32767925 DOI: 10.2174/1381612826666200806101137] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 05/27/2020] [Indexed: 01/10/2023]
Abstract
Molecular hydrogen (H2) was long regarded as non-functional in mammalian cells. We overturned the concept by demonstrating that H2 exhibits antioxidant effects and protects cells against oxidative stress. Subsequently, it has been revealed that H2 has multiple functions in addition to antioxidant effects, including antiinflammatory, anti-allergic functions, and as cell death and autophagy regulation. Additionally, H2 stimulates energy metabolism. As H2 does not readily react with most biomolecules without a catalyst, it is essential to identify the primary targets with which H2 reacts or interacts directly. As a first event, H2 may react directly with strong oxidants, such as hydroxyl radicals (•OH) in vivo. This review addresses the key issues related to this in vivo reaction. •OH may have a physiological role because it triggers a free radical chain reaction and may be involved in the regulation of Ca2+- or mitochondrial ATP-dependent K+-channeling. In the subsequent pathway, H2 suppressed a free radical chain reaction, leading to decreases in lipid peroxide and its end products. Derived from the peroxides, 4-hydroxy-2-nonenal functions as a mediator that up-regulates multiple functional PGC-1α. As the other direct target in vitro and in vivo, H2 intervenes in the free radical chain reaction to modify oxidized phospholipids, which may act as an antagonist of Ca2+-channels. The resulting suppression of Ca2+-signaling inactivates multiple functional NFAT and CREB transcription factors, which may explain H2 multi-functionality. This review also addresses the involvement of NFAT in the beneficial role of H2 in COVID-19, Alzheimer's disease and advanced cancer. We discuss some unsolved issues of H2 action on lipopolysaccharide signaling, MAPK and NF-κB pathways and the Nrf2 paradox. Finally, as a novel idea for the direct targeting of H2, this review introduces the possibility that H2 causes structural changes in proteins via hydrate water changes.
Collapse
Affiliation(s)
- Shigeo Ohta
- Department of Neurology Medicine, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| |
Collapse
|
16
|
Teng H, Li M, Qian L, Yang H, Pang M. Long non‑coding RNA SNHG16 inhibits the oxygen‑glucose deprivation and reoxygenation‑induced apoptosis in human brain microvascular endothelial cells by regulating miR‑15a‑5p/bcl‑2. Mol Med Rep 2020; 22:2685-2694. [PMID: 32945414 PMCID: PMC7453539 DOI: 10.3892/mmr.2020.11385] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 06/03/2020] [Indexed: 12/20/2022] Open
Abstract
MicroRNA (miR) 15a-5p can promote ischemia/reperfusion (I/R)-induced apoptosis of cerebral vascular endothelial cells, which is inhibited by long non-coding RNAs (lncRNAs). The present study investigated the potential of lncRNAs targeting miR-15a-5p to regulate oxygen-glucose deprivation and reoxygenation (OGD-R)-induced apoptosis of human brain microvascular endothelial cells (hBMECs). hBMECs were transfected with or without miR-15a-5p or its mutant, together with p-small nucleolar RNA host gene 16 (SNHG16) or its mutant. Following OGD-R, proliferation, apoptosis and miR-15a-5p, SNHG16 and Bcl-2 expression levels were determined using MTT, flow cytometry, reverse transcription-quantitative PCR or western blotting. The potential interaction of SNHG16 with miR-15a-5p was analyzed by pull-down, luciferase and immunoprecipitation assays. OGD-R induced apoptosis of hBMECs and increased miR-15a-5p expression levels in a time-dependent manner. miR-15a-5p overexpression decreased the proliferation of hBMECs and promoted apoptosis by decreasing Bcl-2 expression levels. SNHG16 was pulled-down by miR-15a-5p and anti-Ago2. miR-15a-5p overexpression significantly decreased SNHG16-regulated luciferase activity and hBMEC survival by increasing apoptosis. SNHG16 overexpression decreased miR-15a-5p expression levels in hBMECs. SNHG16 gradually decreased following OGD-R and its overexpression decreased miR-15a-5p expression levels and promoted the proliferation of hBMECs by decreasing apoptosis. SNHG16 enhanced Bcl-2 expression levels in hBMECs, which was abrogated by miR-15a-5p. Bioinformatics suggest that SNHG16 may antagonize the binding of miR-15a-5p to the 3′UTR of Bcl-2 mRNA. These findings suggest that SNHG16 may protect hBMECs from OGD-R-induced apoptosis by antagonizing the miR-15a-5p/bcl-2 axis. Thus, targeting SNHG16-based mechanisms may provide novel therapeutic strategies for treatment of ischemic stroke.
Collapse
Affiliation(s)
- Hongwei Teng
- Department of Neurosurgery, Binhai County People's Hospital, Yancheng, Jiangsu 224500, P.R. China
| | - Ming Li
- Department of Laboratory Medicine, Binhai County People's Hospital, Yancheng, Jiangsu 224500, P.R. China
| | - Lei Qian
- Department of Laboratory Medicine, Binhai County People's Hospital, Yancheng, Jiangsu 224500, P.R. China
| | - Hua Yang
- Department of Neurosurgery, Binhai County People's Hospital, Yancheng, Jiangsu 224500, P.R. China
| | - Mingzhi Pang
- Department of Neurosurgery, Wuxi No. 2 Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214002, P.R. China
| |
Collapse
|
17
|
Andjelkovic AV, Stamatovic SM, Phillips CM, Martinez-Revollar G, Keep RF. Modeling blood-brain barrier pathology in cerebrovascular disease in vitro: current and future paradigms. Fluids Barriers CNS 2020; 17:44. [PMID: 32677965 PMCID: PMC7367394 DOI: 10.1186/s12987-020-00202-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/15/2020] [Indexed: 12/16/2022] Open
Abstract
The complexity of the blood-brain barrier (BBB) and neurovascular unit (NVU) was and still is a challenge to bridge. A highly selective, restrictive and dynamic barrier, formed at the interface of blood and brain, the BBB is a "gatekeeper" and guardian of brain homeostasis and it also acts as a "sensor" of pathological events in blood and brain. The majority of brain and cerebrovascular pathologies are associated with BBB dysfunction, where changes at the BBB can lead to or support disease development. Thus, an ultimate goal of BBB research is to develop competent and highly translational models to understand mechanisms of BBB/NVU pathology and enable discovery and development of therapeutic strategies to improve vascular health and for the efficient delivery of drugs. This review article focuses on the progress being made to model BBB injury in cerebrovascular diseases in vitro.
Collapse
Affiliation(s)
- Anuska V Andjelkovic
- Department of Pathology, University of Michigan Medical School, 7520 MSRB I, 1150 West Medical Center Dr, Ann Arbor, MI, 48109-5602, USA.
| | - Svetlana M Stamatovic
- Department of Pathology, University of Michigan Medical School, 7520 MSRB I, 1150 West Medical Center Dr, Ann Arbor, MI, 48109-5602, USA
| | - Chelsea M Phillips
- Graduate Program in Neuroscience, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Gabriela Martinez-Revollar
- Department of Pathology, University of Michigan Medical School, 7520 MSRB I, 1150 West Medical Center Dr, Ann Arbor, MI, 48109-5602, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Molecular Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
18
|
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: 40] [Impact Index Per Article: 8.0] [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
|
19
|
Hao D, He C, Ma B, Lankford L, Reynaga L, Farmer DL, Guo F, Wang A. Hypoxic Preconditioning Enhances Survival and Proangiogenic Capacity of Human First Trimester Chorionic Villus-Derived Mesenchymal Stem Cells for Fetal Tissue Engineering. Stem Cells Int 2019; 2019:9695239. [PMID: 31781252 PMCID: PMC6874947 DOI: 10.1155/2019/9695239] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 08/23/2019] [Accepted: 09/04/2019] [Indexed: 12/19/2022] Open
Abstract
Prenatal stem cell-based regenerative therapies have progressed substantially and have been demonstrated as effective treatment options for fetal diseases that were previously deemed untreatable. Due to immunoregulatory properties, self-renewal capacity, and multilineage potential, autologous human placental chorionic villus-derived mesenchymal stromal cells (CV-MSCs) are an attractive cell source for fetal regenerative therapies. However, as a general issue for MSC transplantation, the poor survival and engraftment is a major challenge of the application of MSCs. Particularly for the fetal transplantation of CV-MSCs in the naturally hypoxic fetal environment, improving the survival and engraftment of CV-MSCs is critically important. Hypoxic preconditioning (HP) is an effective priming approach to protect stem cells from ischemic damage. In this study, we developed an optimal HP protocol to enhance the survival and proangiogenic capacity of CV-MSCs for improving clinical outcomes in fetal applications. Total cell number, DNA quantification, nuclear area test, and cell viability test showed HP significantly protected CV-MSCs from ischemic damage. Flow cytometry analysis confirmed HP did not alter the immunophenotype of CV-MSCs. Caspase-3, MTS, and Western blot analysis showed HP significantly reduced the apoptosis of CV-MSCs under ischemic stimulus via the activation of the AKT signaling pathway that was related to cell survival. ELISA results showed HP significantly enhanced the secretion of vascular endothelial growth factor (VEGF) and hepatocyte growth factor (HGF) by CV-MSCs under an ischemic stimulus. We also found that the environmental nutrition level was critical for the release of brain-derived neurotrophic factor (BDNF). The angiogenesis assay results showed HP-primed CV-MSCs could significantly enhance endothelial cell (EC) proliferation, migration, and tube formation. Consequently, HP is a promising strategy to increase the tolerance of CV-MSCs to ischemia and improve their therapeutic efficacy in fetal clinical applications.
Collapse
Affiliation(s)
- Dake Hao
- Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA
| | - Chuanchao He
- Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Bowen Ma
- Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Lee Lankford
- Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Lizette Reynaga
- Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Diana L. Farmer
- Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA
| | - Fuzheng Guo
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA
| | - Aijun Wang
- Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA
- Department of Biomedical Engineering, University of California Davis, Davis, CA 95616, USA
| |
Collapse
|
20
|
Acetyl-11-keto-β-boswellic acid (AKBA) Attenuates Oxidative Stress, Inflammation, Complement Activation and Cell Death in Brain Endothelial Cells Following OGD/Reperfusion. Neuromolecular Med 2019; 21:505-516. [DOI: 10.1007/s12017-019-08569-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 09/04/2019] [Indexed: 12/19/2022]
|
21
|
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: 2.2] [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
|
22
|
George SA, Hoeker G, Calhoun PJ, Entz M, Raisch TB, King DR, Khan M, Baker C, Gourdie RG, Smyth JW, Nielsen MS, Poelzing S. Modulating cardiac conduction during metabolic ischemia with perfusate sodium and calcium in guinea pig hearts. Am J Physiol Heart Circ Physiol 2019; 316:H849-H861. [PMID: 30707595 DOI: 10.1152/ajpheart.00083.2018] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We previously demonstrated that altering extracellular sodium (Nao) and calcium (Cao) can modulate a form of electrical communication between cardiomyocytes termed "ephaptic coupling" (EpC), especially during loss of gap junction coupling. We hypothesized that altering Nao and Cao modulates conduction velocity (CV) and arrhythmic burden during ischemia. Electrophysiology was quantified by optically mapping Langendorff-perfused guinea pig ventricles with modified Nao (147 or 155 mM) and Cao (1.25 or 2.0 mM) during 30 min of simulated metabolic ischemia (pH 6.5, anoxia, aglycemia). Gap junction-adjacent perinexal width ( WP), a candidate cardiac ephapse, and connexin (Cx)43 protein expression and Cx43 phosphorylation at S368 were quantified by transmission electron microscopy and Western immunoblot analysis, respectively. Metabolic ischemia slowed CV in hearts perfused with 147 mM Nao and 2.0 mM Cao; however, theoretically increasing EpC with 155 mM Nao was arrhythmogenic, and CV could not be measured. Reducing Cao to 1.25 mM expanded WP, as expected during ischemia, consistent with reduced EpC, but attenuated CV slowing while delaying arrhythmia onset. These results were further supported by osmotically reducing WP with albumin, which exacerbated CV slowing and increased early arrhythmias during ischemia, whereas mannitol expanded WP, permitted conduction, and delayed the onset of arrhythmias. Cx43 expression patterns during the various interventions insufficiently correlated with observed CV changes and arrhythmic burden. In conclusion, decreasing perfusate calcium during metabolic ischemia enhances perinexal expansion, attenuates conduction slowing, and delays arrhythmias. Thus, perinexal expansion may be cardioprotective during metabolic ischemia. NEW & NOTEWORTHY This study demonstrates, for the first time, that modulating perfusate ion composition can alter cardiac electrophysiology during simulated metabolic ischemia.
Collapse
Affiliation(s)
- Sharon A George
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University , Blacksburg, Virginia.,Center for Heart and Reparative Medicine Research, Virginia Tech Carilion Research Institute , Roanoke, Virginia
| | - Gregory Hoeker
- Center for Heart and Reparative Medicine Research, Virginia Tech Carilion Research Institute , Roanoke, Virginia
| | - Patrick J Calhoun
- Center for Heart and Reparative Medicine Research, Virginia Tech Carilion Research Institute , Roanoke, Virginia.,Department of Biological Sciences, Virginia Polytechnic Institute and State University , Blacksburg, Virginia
| | - Michael Entz
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University , Blacksburg, Virginia.,Center for Heart and Reparative Medicine Research, Virginia Tech Carilion Research Institute , Roanoke, Virginia
| | - Tristan B Raisch
- Center for Heart and Reparative Medicine Research, Virginia Tech Carilion Research Institute , Roanoke, Virginia.,Translational Biology, Medicine, and Health Graduate Program, Virginia Polytechnic Institute and State University , Blacksburg, Virginia
| | - D Ryan King
- Center for Heart and Reparative Medicine Research, Virginia Tech Carilion Research Institute , Roanoke, Virginia.,Translational Biology, Medicine, and Health Graduate Program, Virginia Polytechnic Institute and State University , Blacksburg, Virginia
| | - Momina Khan
- Department of Human Food Nutrition and Exercise, Virginia Polytechnic Institute and State University , Blacksburg, Virginia
| | - Chandra Baker
- Center for Heart and Reparative Medicine Research, Virginia Tech Carilion Research Institute , Roanoke, Virginia
| | - Robert G Gourdie
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University , Blacksburg, Virginia.,Center for Heart and Reparative Medicine Research, Virginia Tech Carilion Research Institute , Roanoke, Virginia
| | - James W Smyth
- Center for Heart and Reparative Medicine Research, Virginia Tech Carilion Research Institute , Roanoke, Virginia.,Department of Biological Sciences, Virginia Polytechnic Institute and State University , Blacksburg, Virginia
| | - Morten S Nielsen
- Department of Biomedical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Steven Poelzing
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University , Blacksburg, Virginia.,Center for Heart and Reparative Medicine Research, Virginia Tech Carilion Research Institute , Roanoke, Virginia.,Translational Biology, Medicine, and Health Graduate Program, Virginia Polytechnic Institute and State University , Blacksburg, Virginia
| |
Collapse
|
23
|
Li J, Zhang S, Liu X, Han D, Xu J, Ma Y. Neuroprotective effects of leonurine against oxygen-glucose deprivation by targeting Cx36/CaMKII in PC12 cells. PLoS One 2018; 13:e0200705. [PMID: 30016355 PMCID: PMC6049927 DOI: 10.1371/journal.pone.0200705] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 07/02/2018] [Indexed: 01/31/2023] Open
Abstract
Leonurine has been reported to play an important role in ameliorating cognitive dysfunction, inhibiting ischemic stroke, and attenuating perihematomal edema and neuroinflammation in intracerebral hemorrhage. However, the exact mechanism and potential molecular targets of this effect remain unclear. Thus, in this study we investigated the neuroprotective effects of leonurine on hypoxia ischemia injury and explored the underlying mechanisms. An in vitro model of oxygen-glucose deprivation (OGD)-induced PC12 cells was established to mimic ischemic-like conditions. Cell viability, apoptosis, Cx36 and pCaMKII/CaMKII expression levels were evaluated after treatment with leonurine. The Cx36-selective antagonist mefloquine and CaMKII Inhibitor KN-93 were used to investigate the neuroprotective effect of leonurine on and the involvement of Cx36/CaMKII in this process. The results revealed that cell viability decreased and cell apoptosis and the protein expression of Cx36 and pCaMKII/CaMKII increased in the OGD-induced PC12 cells. Leonurine significantly increased cell viability and decreased cell apoptosis and the protein expression of Cx36 and pCaMKII/CaMKII in the OGD-induced PC12 cells. The specific inhibitor of Cx36 and CaMKII displayed similar protective effects. Moreover, the inhibition of Cx36 reduced pCaMKII levels and the ratio of pCaMKII/CaMKII in the OGD-induced PC12 cells, and vice versa. Taken together, these results suggest that leonurine might have a protective effect on OGD-induced PC12 cells through targeting the Cx36/CaMKII pathway. Thus, leonurine appears to have potential as a preventive or therapeutic drug against ischemic-induced neuronal injury.
Collapse
Affiliation(s)
- Jiao Li
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Shuang Zhang
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xiaoxi Liu
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Deping Han
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jianqin Xu
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yunfei Ma
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
24
|
Vijayan M, Kumar S, Yin X, Zafer D, Chanana V, Cengiz P, Reddy PH. Identification of novel circulatory microRNA signatures linked to patients with ischemic stroke. Hum Mol Genet 2018; 27:2318-2329. [PMID: 29701837 PMCID: PMC6005038 DOI: 10.1093/hmg/ddy136] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 04/01/2018] [Accepted: 04/09/2018] [Indexed: 12/22/2022] Open
Abstract
MicroRNAs (miRNAs) are involved in growth, development, and occurrence and progression of many diseases. MiRNA-mediated post-transcriptional regulation is poorly understood in vascular biology and pathology. The purpose of this is to determine circulatory miRNAs as early detectable peripheral biomarkers in patients with ischemic stroke (IS). MiRNAs expression levels were measured in IS serum samples and healthy controls using Illumina deep sequencing analysis and identified differentially expressed miRNAs. Differentially expressed miRNAs were further validated using SYBR-green-based quantitative real-time PCR (qRT-PCR) assay in postmortem IS brains, lymphoblastoid IS cell lines, oxygen and glucose deprivation/reoxygenation -treated human and mouse neuroblastoma cells, and mouse models of hypoxia and ischemia (HI)-induced stroke. A total of 4656 miRNAs were differentially expressed in IS serum samples relative to healthy controls. Out of 4656 miRNAs, 272 were found to be significantly deregulated in IS patients. Interestingly, we found several novel and previously unreported miRNAs in IS patients relative to healthy controls. Further analyses revealed that some candidate miRNAs and its target genes were involved in the regulation of the stroke. To the best of our knowledge, this is the first study identified potential novel candidate miRNAs in IS serum samples from the residents of rural West Texas. MiRNAs identified in this study could potentially be used as a biomarker and the development of novel therapeutic approaches for stroke. Further studies are necessary to better understand miRNAs-regulated stroke cellular changes.
Collapse
Affiliation(s)
- Murali Vijayan
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Subodh Kumar
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Xiangling Yin
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Dila Zafer
- Waisman Center and Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Vishal Chanana
- Waisman Center and Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Pelin Cengiz
- Waisman Center and Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - P Hemachandra Reddy
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Garrison Institute on Aging, South West Campus, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Department of Speech, Language and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA and
- Department of Public Health, Graduate School of Biomedical Sciences, Lubbock, TX, USA
| |
Collapse
|
25
|
Soejima E, Ohki T, Kurita Y, Yuan X, Tanaka K, Kakino S, Hara K, Nakayama H, Tajiri Y, Yamada K. Protective effect of 3-hydroxybutyrate against endoplasmic reticulum stress-associated vascular endothelial cell damage induced by low glucose exposure. PLoS One 2018; 13:e0191147. [PMID: 29554103 PMCID: PMC5858752 DOI: 10.1371/journal.pone.0191147] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 01/01/2018] [Indexed: 12/25/2022] Open
Abstract
Aims/Hypothesis The aim of this study was to elucidate the mechanism by which severe hypoglycemia accelerates vascular complications. Furthermore, we assessed the possible protective effect of ketone bodies against the endothelial cell damage caused by glucose deficiency. Methods Human umbilical vein endothelial cells (HUVECs) were cultured at a glucose level of either 0.56 or 5.6 mmol/L with or without 3-hydroxybutyrate (3-HB) supplementation. Cell viability was assessed with a CCK-8 assay and a lactate dehydrogenase (LDH) release assay. The activity of caspases was measured using fluorogenic substrates. The expression of genes associated with endothelial cell function and endoplasmic reticulum (ER) stress was evaluated by real-time quantitative PCR. Protein levels of ER stress-related molecules were assessed by Western blotting. Results Culture of HUVECs in low-glucose medium for 24 or 48 h resulted in reduction of cell viability accompanied by activation of caspase-3/7 and caspase-8. The addition of a pan caspase inhibitor attenuated the cell death. After incubation in the low-glucose medium, we found reduced mRNA and protein levels of endothelial nitric oxide synthase. ER stress responses mediated by phosphorylation of protein kinase RNA-like ER kinase (PERK) and cleavage of activating transcription factor 6 (ATF6) were augmented, but X-box binding protein 1 (Xbp1) splicing was reduced. Most of these responses to glucose deficiency were significantly attenuated by supplementation with 3-HB. Conclusions/Interpretation These observations showed that exposure to low glucose induces ER stress, caspase activation, endothelial cell dysfunction and cell death. The beneficial effects of 3-HB shown in this study suggest that hypoketonemic severe hypoglycemia induced by insulin injections or insulin secretagogue administration may be more harmful than hyperketonemic severe hypoglycemia.
Collapse
Affiliation(s)
- Eri Soejima
- Division of Endocrinology and Metabolism, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Tsuyoshi Ohki
- Division of Endocrinology and Metabolism, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Yayoi Kurita
- Division of Endocrinology and Metabolism, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Xiaohong Yuan
- Division of Endocrinology and Metabolism, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Kayo Tanaka
- Division of Endocrinology and Metabolism, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Satomi Kakino
- Division of Endocrinology and Metabolism, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Kento Hara
- Division of Endocrinology and Metabolism, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Hitomi Nakayama
- Division of Endocrinology and Metabolism, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Yuji Tajiri
- Division of Endocrinology and Metabolism, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Kentaro Yamada
- Division of Endocrinology and Metabolism, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
- * E-mail:
| |
Collapse
|
26
|
Baek A, Kim JH, Pyo S, Jung JH, Park EJ, Kim SH, Cho SR. The Differential Effects of Repetitive Magnetic Stimulation in an In Vitro Neuronal Model of Ischemia/Reperfusion Injury. Front Neurol 2018; 9:50. [PMID: 29487560 PMCID: PMC5816832 DOI: 10.3389/fneur.2018.00050] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 01/18/2018] [Indexed: 01/08/2023] Open
Abstract
Repetitive transcranial magnetic stimulation (rTMS) is a non-invasive therapy that has been implicated in treatment of serious neurological disorders. However, the neurobiological mechanisms underlying the effects of rTMS remain unclear. Therefore, this study examined the differential effects of repetitive magnetic stimulation (rMS) in an in vitro neuronal model of ischemia/reperfusion (I/R) injury, depending on low and high frequency. Neuro-2a cells were differentiated with retinoic acid and established for in vitro neuronal model of I/R injury under a subsequent 3 h of oxygen and glucose deprivation/reoxygenation (OGD/R) condition. After the I/R injury, the differentiated neuronal cells were stimulated with rMS on day 1 and randomly divided into three groups: OGD/R+sham, OGD/R+low-frequency, and OGD/R+high-frequency groups. High-frequency rMS increases cell proliferation through activation of extracellular signal-regulated kinases and AKT-signaling pathway and inhibits apoptosis in OGD/R-injured cells. Furthermore, high-frequency rMS increases Ca2+–calmodulin-dependent protein kinase II (CaMKII)-cAMP-response element binding protein (CREB) signaling pathway, further leading to alternation of brain-derived neurotrophic factor expression and synaptic plasticity in OGD/R injured cells. These results verified the neurobiological mechanisms of frequency-dependent rMS in I/R injury-treated neuronal cells. These mechanisms will help develop more powerful and credible rTMS stimulation treatment protocols.
Collapse
Affiliation(s)
- Ahreum Baek
- Department and Rehabilitation Medicine, Yonsei University Wonju College of Medicine, Wonju, South Korea.,Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Ji Hyun Kim
- Department and Rehabilitation Medicine, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Soonil Pyo
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, South Korea
| | - Joon-Ho Jung
- Department of Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Eun Jee Park
- Department of Rehabilitation Medicine, Graduate School Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Sung Hoon Kim
- Department and Rehabilitation Medicine, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Sung-Rae Cho
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, South Korea.,Yonsei Stem Cell Center, Avison Biomedical Research Center, Seoul, South Korea.,Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
27
|
Zhang JJ, Zhu JJ, Hu YB, Xiang GH, Deng LC, Wu FZ, Wei XJ, Wang YH, Sun LY, Lou XQ, Shao MM, Mao M, Zhang HY, Xu YP, Zhu SP, Xiao J. Transplantation of bFGF-expressing neural stem cells promotes cell migration and functional recovery in rat brain after transient ischemic stroke. Oncotarget 2017; 8:102067-102077. [PMID: 29254225 PMCID: PMC5731935 DOI: 10.18632/oncotarget.22155] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 10/03/2017] [Indexed: 01/19/2023] Open
Abstract
Cerebrovascular disease such as stroke is one of the most common diseases in the aging population, and neural stem cells (NSCs) transplantation may provide an alternative therapy for cerebral ischemia. However, a hostile microenvironment in the ischemic brain offers is challenging for the survival of the transplanted cells. Considering the neuroprotective role of basic fibroblast growth factor (bFGF), the present study investigated whether bFGF gene-modified NSCs could improve the neurological function deficit after transient middle cerebral artery occlusion (MCAO) in adult male Sprague-Dawley rats. These rats were intravenously injected with modified NSCs (5×106/200 μL) or vehicle 24 h after MCAO. Histological analysis was performed on days 7 and 28 after tMCAO. The survival, migration, proliferation, and differentiation of the transplanted modified C17.2 cells in the brain were improved. In addition, the intravenous infusion of NSCs and bFGF gene-modified C17.2 cells improved the functional recovery as compared to the control. Furthermore, bFGF promoted the C17.2 cell growth, survival, and differentiation into mature neurons within the infarct region. These data suggested that bFGF gene-modified NSCs have the potential to be a therapeutic agent in brain ischemia.
Collapse
Affiliation(s)
- Jin-Jing Zhang
- Department of Pharmacy, Affiliated Cixi People's Hospital, Wenzhou Medical University, Ningbo, Zhejiang, 315300, China.,Institute of Molecular Pharmacology, School of Pharmaceutics Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Juan-Juan Zhu
- Department of Geriatrics and Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Yuan-Bo Hu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Guang-Heng Xiang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Lian-Cheng Deng
- Institute of Molecular Pharmacology, School of Pharmaceutics Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Fen-Zan Wu
- Institute of Molecular Pharmacology, School of Pharmaceutics Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.,Department of Neurosurgery, Affiliated Cixi People's Hospital, Wenzhou Medical University, Ningbo, Zhejiang, 315300, China
| | - Xiao-Jie Wei
- Department of Neurosurgery, Affiliated Cixi People's Hospital, Wenzhou Medical University, Ningbo, Zhejiang, 315300, China
| | - Ying-Hao Wang
- Department of Pharmacy, Affiliated Cixi People's Hospital, Wenzhou Medical University, Ningbo, Zhejiang, 315300, China
| | - Liang-Yan Sun
- Department of Pharmacy, Affiliated Cixi People's Hospital, Wenzhou Medical University, Ningbo, Zhejiang, 315300, China
| | - Xiao-Qing Lou
- Department of Pharmacy, Affiliated Cixi People's Hospital, Wenzhou Medical University, Ningbo, Zhejiang, 315300, China
| | - Min-Min Shao
- Department of Pharmacy, Affiliated Cixi People's Hospital, Wenzhou Medical University, Ningbo, Zhejiang, 315300, China
| | - Mao Mao
- Department of Pharmacy, Affiliated Cixi People's Hospital, Wenzhou Medical University, Ningbo, Zhejiang, 315300, China
| | - Hong-Yu Zhang
- Institute of Molecular Pharmacology, School of Pharmaceutics Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yue-Ping Xu
- Department of Pharmacy, Affiliated Cixi People's Hospital, Wenzhou Medical University, Ningbo, Zhejiang, 315300, China
| | - Si-Pin Zhu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Jian Xiao
- Institute of Molecular Pharmacology, School of Pharmaceutics Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| |
Collapse
|
28
|
Xin JW, Jiang YG. Long noncoding RNA MALAT1 inhibits apoptosis induced by oxygen-glucose deprivation and reoxygenation in human brain microvascular endothelial cells. Exp Ther Med 2017; 13:1225-1234. [PMID: 28413461 PMCID: PMC5377418 DOI: 10.3892/etm.2017.4095] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 07/07/2016] [Indexed: 12/15/2022] Open
Abstract
Cerebral ischemia/reperfusion (I/R) injury leads to brain vascular dysfunction, which is characterized by endothelial cell injury or death. Long noncoding (lnc) RNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is reportedly associated with endothelial cell functions and dysfunctions. In the present study, the role of MALAT1 in I/R-induced cerebral vascular endothelial cell apoptosis was explored using oxygen-glucose deprivation and reoxygenation (OGD-R) as an in vitro I/R injury model. Primary human brain microvascular endothelial cells were cultured under OGD-R, and the expression levels of MALAT1 and cell apoptosis were measured at 6, 9, 12, 24 and 36 h post-reoxygenation. The expression levels of MALAT1 and the apoptotic rate of cells exposed to OGD-R exhibited contrasting trends following reoxygenation. Following OGD-R, lentiviral overexpression of MALAT1 increased phosphatidylinositol 3-kinase (PI3K) activities and the activation of Akt phosphorylation, and decreased cell apoptosis and caspase 3 activities, which were successfully abolished by treatment with a PI3K inhibitor, Wortmannin. Conversely, lentiviral knockdown of MALAT1 decreased PI3K activities and the activation of Akt phosphorylation, and increased cell apoptosis and caspase 3 activity. Overexpression and knockdown of MALAT1 exhibited no significant effects on OGD-R-induced reactive oxygen species (ROS) production. In conclusion, to the best of our knowledge, the present study was the first to suggest that lncRNA MALAT1 may protect human brain vascular endothelial cells from OGD-R-induced apoptosis via a PI3K-dependent mechanism. These findings suggest that MALAT1 may be a potential novel therapeutic target for cerebral I/R injury.
Collapse
Affiliation(s)
- Jia-Wei Xin
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Yu-Gang Jiang
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
29
|
He F, Duan X, Dai R, Wang W, Yang C, Lin Q. PROTECTIVE EFFECTS OF ETHYL ACETATE EXTRACTION FROM GASTRODIA ELATA BLUME ON BLOOD-BRAIN BARRIER IN FOCAL CEREBRAL ISCHEMIA REPERFUSION. AFRICAN JOURNAL OF TRADITIONAL, COMPLEMENTARY, AND ALTERNATIVE MEDICINES 2016; 13:199-209. [PMID: 28852737 PMCID: PMC5566145 DOI: 10.21010/ajtcam.v13i4.26] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Background: Damage of the blood brain barrier (BBB) during the process of cerebral ischemic injury is a key factor which influences the therapeutic efficacy to the cerebral ischemic injury. The present study was designed to verify the mechanisms underlying the protective effects of the ethyl acetate (EtOAc) extraction from Gastrodia elata Blume (GEB) on the BBB by developing a model of cerebral ischemia-reperfusion in rats. Material and methods: MCAO/R model in rats was developed through a thread embolism method. The neurological scales, the moisture and the evans blue (EB) contents of brains were detected. Meanwhile, the release of nitric oxide (NO) and activities of NO synthase (NOS) in brain tissues were measured. Western blotting analyses were also performed to assess the protein expressions of AQP-4, Occludin and Claudin-5 in brain tissue. Results: After rats were pretreated with different concentrations of EtOAc extractions from GEB, the neurologic scores, the EB contents in the brain tissues and the moisture of the brains were significantly decreased. Meanwhile, the release of NO, the activities of nNOS and iNOS were notably inhibited. Furthemore, the protein expression of AQP-4 was markedly decreased, but the protein expressions of -5 and Occludin were significantly increased. Conclusion: the EtOAc extracts of GEB may decrease the permeability of BBB when focal cerebral ischemia occurs. The inhibition of the NOS pathways, the attenuation of the protein expression of AQP-4 and the enhancement of the expressions of the tight junction proteins may contribute to the protective effects of the EtOAc extracts from GEB on BBB.
Collapse
Affiliation(s)
- Fangyan He
- The Department of Pharmacology, Yunnan University of Traditional Chinese Medicine, Kunming 650500, China
| | - Xiaohua Duan
- The Key Modern Research Laboratory for Ethno-pharmacognosy of Yunnan Higher School, Yunnan University of Traditional Chinese Medicine, Kunming 650500, China
| | - Rong Dai
- The Department of Pharmacology, Yunnan University of Traditional Chinese Medicine, Kunming 650500, China
| | - Wei Wang
- The Department of Pharmacology, Yunnan University of Traditional Chinese Medicine, Kunming 650500, China
| | - Cui Yang
- Ethnic Drug Screening & Pharmacology Center, Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming 650500, China
| | - Qing Lin
- The Department of Pharmacology, Yunnan University of Traditional Chinese Medicine, Kunming 650500, China
| |
Collapse
|
30
|
Glucose Transporters at the Blood-Brain Barrier: Function, Regulation and Gateways for Drug Delivery. Mol Neurobiol 2016; 54:1046-1077. [PMID: 26801191 DOI: 10.1007/s12035-015-9672-6] [Citation(s) in RCA: 224] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 12/17/2015] [Indexed: 12/31/2022]
Abstract
Glucose transporters (GLUTs) at the blood-brain barrier maintain the continuous high glucose and energy demands of the brain. They also act as therapeutic targets and provide routes of entry for drug delivery to the brain and central nervous system for treatment of neurological and neurovascular conditions and brain tumours. This article first describes the distribution, function and regulation of glucose transporters at the blood-brain barrier, the major ones being the sodium-independent facilitative transporters GLUT1 and GLUT3. Other GLUTs and sodium-dependent transporters (SGLTs) have also been identified at lower levels and under various physiological conditions. It then considers the effects on glucose transporter expression and distribution of hypoglycemia and hyperglycemia associated with diabetes and oxygen/glucose deprivation associated with cerebral ischemia. A reduction in glucose transporters at the blood-brain barrier that occurs before the onset of the main pathophysiological changes and symptoms of Alzheimer's disease is a potential causative effect in the vascular hypothesis of the disease. Mutations in glucose transporters, notably those identified in GLUT1 deficiency syndrome, and some recreational drug compounds also alter the expression and/or activity of glucose transporters at the blood-brain barrier. Approaches for drug delivery across the blood-brain barrier include the pro-drug strategy whereby drug molecules are conjugated to glucose transporter substrates or encapsulated in nano-enabled delivery systems (e.g. liposomes, micelles, nanoparticles) that are functionalised to target glucose transporters. Finally, the continuous development of blood-brain barrier in vitro models is important for studying glucose transporter function, effects of disease conditions and interactions with drugs and xenobiotics.
Collapse
|
31
|
Yin B, Barrionuevo G, Weber SG. Optimized real-time monitoring of glutathione redox status in single pyramidal neurons in organotypic hippocampal slices during oxygen-glucose deprivation and reperfusion. ACS Chem Neurosci 2015; 6:1838-48. [PMID: 26291433 DOI: 10.1021/acschemneuro.5b00186] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
A redox-sensitive Grx1-roGFP2 fusion protein was introduced by transfection into single pyramidal neurons in the CA1 subfield of organotypic hippocampal slice cultures (OHSCs). We assessed changes in the GSH system in neuronal cytoplasm and mitochondria during oxygen-glucose deprivation and reperfusion (OGD/RP), an in vitro model of stroke. Pyramidal cells in a narrow range of depths below the surface of the OHSC were transfected by gene gun or single-cell electroporation with cyto- or mito-Grx1-roGFP2. To mimic the conditions of acute stroke, we developed an optimized superfusion system with the capability of rapid and reproducible exchange of the solution bathing the OHSCs. Measurements of pO2 as a function of tissue depth show that in the region containing the transfected cells, the pO2 is well-controlled. We also found that the pO2 changes on the same time scale as changes in intracranial pressure, cerebral blood flow, and pO2 during acute stroke. Determining the reduction potential, EGSH, from the ratiometric fluorescence signal requires an absolute intensity measurement during calibration of the Grx1-roGFP2. Using the signal from cotransfected tdTomato as an internal standard during calibration improves quantitative measurements of Grx1-roGFP2 redox status and allows EGSH to be determined. EGSH becomes more reducing during OGD and more oxidizing during RP in mitochondria while changes in cytoplasm are not significant compared with controls.
Collapse
Affiliation(s)
- Bocheng Yin
- Department
of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Germán Barrionuevo
- Department
of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Stephen G. Weber
- Department
of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
32
|
Andrabi SS, Parvez S, Tabassum H. Melatonin and Ischemic Stroke: Mechanistic Roles and Action. Adv Pharmacol Sci 2015; 2015:384750. [PMID: 26435711 PMCID: PMC4575994 DOI: 10.1155/2015/384750] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 08/09/2015] [Accepted: 08/19/2015] [Indexed: 11/21/2022] Open
Abstract
Stroke is one of the most devastating neurological disabilities and brain's vulnerability towards it proves to be fatal and socio-economic loss of millions of people worldwide. Ischemic stroke remains at the center stage of it, because of its prevalence amongst the several other types attacking the brain. The various cascades of events that have been associated with stroke involve oxidative stress, excitotoxicity, mitochondrial dysfunction, upregulation of Ca(2+) level, and so forth. Melatonin is a neurohormone secreted by pineal and extra pineal tissues responsible for various physiological processes like sleep and mood behaviour. Melatonin has been implicated in various neurological diseases because of its antioxidative, antiapoptotic, and anti-inflammatory properties. We have previously reviewed the neuroprotective effect of melatonin in various models of brain injury like traumatic brain injury and spinal cord injury. In this review, we have put together the various causes and consequence of stroke and protective role of melatonin in ischemic stroke.
Collapse
Affiliation(s)
- Syed Suhail Andrabi
- Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi 110062, India
| | - Suhel Parvez
- Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi 110062, India
| | - Heena Tabassum
- Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi 110062, India
- Department of Biochemistry, Jamia Hamdard (Hamdard University), New Delhi 110062, India
| |
Collapse
|