1
|
Intranasal Methylprednisolone Ameliorates Neuroinflammation Induced by Chronic Toluene Exposure. Pharmaceutics 2022; 14:pharmaceutics14061195. [PMID: 35745768 PMCID: PMC9230943 DOI: 10.3390/pharmaceutics14061195] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/17/2021] [Accepted: 01/10/2022] [Indexed: 01/27/2023] Open
Abstract
Inhalants are chemical substances that induce intoxication, and toluene is the main component of them. Increasing evidence indicates that a dependence on inhalants involves a state of chronic stress associated to the activation of immune cells in the central nervous system and release of proinflammatory mediators, especially in some brain areas such as the nucleus accumbens and frontal cortex, where the circuits of pleasure and reward are. In this study, anti-neuroinflammatory treatment based on a single dose of intranasal methylprednisolone was assessed in a murine model of chronic toluene exposure. The levels of proinflammatory mediators, expression levels of Iba-1 and GFAP, and histological changes in the frontal cortex and nucleus accumbens were evaluated after the treatment. The chronic exposure to toluene significantly increased the levels of TNF-α, IL-6, and NO, the expression of GFAP, and induced histological alterations in mouse brains. The treatment with intranasally administered MP significantly reduced the expression of TNF-α and NO and the expression of GFAP (p < 0.05); additionally, it reversed the central histological damage. These results indicate that intranasally administered methylprednisolone could be considered as a treatment to reverse neuroinflammation and histological damages associated with the use of inhalants.
Collapse
|
2
|
Gao X, Wu D, Dou L, Zhang H, Huang L, Zeng J, Zhang Y, Yang C, Li H, Liu L, Ma B, Yuan Q. Protective effects of mesenchymal stem cells overexpressing extracellular regulating kinase 1/2 against stroke in rats. Brain Res Bull 2019; 149:42-52. [PMID: 31002912 DOI: 10.1016/j.brainresbull.2019.04.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 02/28/2019] [Accepted: 04/09/2019] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Although transplantation of bone marrow-derived mesenchymal stem cells (MSCs) has shown beneficial effects on stroke, lower survival of MSCs limits effects. Extracellular regulating kinase 1/2 signaling (ERK1/2) is crucial for cell survival, differentiation, and proliferation. This study was designed to explore whether MSCs modified by over-expressing ERK1/2 may reinforce beneficial effects on stroke in rats. METHODS rat MSCs transfected with ERK1/2 and empty lentivirus to generate MSCs overexpressing ERK1/2 (ERK/MSCs) and MSCs (as a control), respectively. In vitro, ERK/MSCs were plated and exposed to glutamate-induced condition, and viability of ERK/MSCs was measured. Furthermore, neural induction of ERK/MSCs was investigated in vitro. Cerebral ischemic rats were induced by occluding middle cerebral artery, and then were stereotaxically injected into ipsilateral right lateral ventricle with ERK/MSCs or MSCs 3 days after stroke and survived for 7 or 14 days after injection. RESULTS ERK/MSCs showed better viability in physiological and glutamate-induced neurotoxic conditions compared to MSCs. After neural induction, more neurons were be differentiated from ERK/MSCs than from MSCs. After transplantation, more numbers of grafted cells and improved functional recovery were observed in ERK/MSCs-treated rats compared with MSCs-treated rats. Compared with MSCs treatment, ERK/MSCs treatment significantly increased proliferation of neural stem cells in the subventricle zone (SVZ) and the MAP2/nestin double-labeled cells adjacent to the SVZ, enhanced the numbers of reactive astrocytes while suppressed microglial activation. Besides, TNF-α level was elevated in ERK/MSCs-treated rats. CONCLUSION ERK/MSCs transplantation showed better functional recovery after stroke in rats, likely in part through enhancing survival of MSCs and possibly by modulating the proliferation, neuronal de-differentiation and neuroinflammation.
Collapse
Affiliation(s)
- Xiaoqing Gao
- Department of Neurology, Shanghai Tongji hospital, Tongji University School of Medicine, Shanghai, 200065, China; Department of Anatomy and Neurobiology, Southwest Medical University, Luzhou, 646000, China
| | - Dandan Wu
- Department of Neurology, Shanghai Tongji hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Ling Dou
- Department of Neurology, Shanghai Tongji hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Haibo Zhang
- Department of Neurology, Shanghai Tongji hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Liang Huang
- Department of Neurology, Shanghai Tongji hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Jiaqi Zeng
- Department of Neurology, Shanghai Tongji hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Yiiie Zhang
- Department of Neurology, Shanghai Tongji hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Chaoxian Yang
- Department of Anatomy and Neurobiology, Southwest Medical University, Luzhou, 646000, China
| | - Huanhuan Li
- Department of Neurology, Shanghai Tongji hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Lifen Liu
- Department of Neurology, Shanghai Tongji hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Bin Ma
- Department of Molecular and Biomedical Sciences, School of Veterinary and Life Sciences, Murdoch University, Perth, WA, Australia
| | - Qionglan Yuan
- Department of Neurology, Shanghai Tongji hospital, Tongji University School of Medicine, Shanghai, 200065, China.
| |
Collapse
|
3
|
Haque ME, Gabr RE, George SD, Boren SB, Vahidy FS, Zhang X, Arevalo OD, Alderman S, Narayana PA, Hasan KM, Friedman ER, Sitton CW, Savitz SI. Serial Cerebral Metabolic Changes in Patients With Ischemic Stroke Treated With Autologous Bone Marrow Derived Mononuclear Cells. Front Neurol 2019; 10:141. [PMID: 30858820 PMCID: PMC6397870 DOI: 10.3389/fneur.2019.00141] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 02/04/2019] [Indexed: 12/13/2022] Open
Abstract
Purpose: Cell-based therapy offers new opportunities for the development of novel treatments to promote tissue repair, functional restoration, and cerebral metabolic balance. N-acetylasperate (NAA), Choline (Cho), and Creatine (Cr) are three major metabolites seen on proton magnetic resonance spectroscopy (MRS) that play a vital role in balancing the biochemical processes and are suggested as markers of recovery. In this preliminary study, we serially monitored changes in these metabolites in ischemic stroke patients who were treated with autologous bone marrow-derived mononuclear cells (MNCs) using non-invasive MRS. Materials and Methods: A sub-group of nine patients (3 male, 6 female) participated in a serial MRS study, as part of a clinical trial on autologous bone marrow cell therapy in acute ischemic stroke. Seven to ten million mononuclear cells were isolated from the patient's bone marrow and administered intravenously within 72 h of onset of injury. MRS data were obtained at 1, 3, and 6 months using a whole-body 3.0T MRI. Single voxel point-resolved spectroscopy (PRESS) was obtained within the lesion and contralesional gray matter. Spectral analysis was done using TARQUIN software and absolute concentration of NAA, Cho, and Cr was determined. National Institute of Health Stroke Scale (NIHSS) was serially recoreded. Two-way analysis of variance was performed and p < 0.05 considered statistically significant. Results: All metabolites showed statistically significant or clear trends toward lower ipsilesional concentrations compared to the contralesional side at all time points. Statistically significant reductions were found in ipsilesional NAA at 1M and 3M, Cho at 6M, and Cr at 1M and 6M (p < 0.03), compared to the contralesional side. Temporally, ipsilesional NAA increased between 3M and 6M (p < 0.01). On the other hand, ipsilesional Cho showed continued decline till 6M (p < 0.01). Ipsilesional Cr was stable over time. Contralesional metabolites were relatively stable over time, with only Cr showing a reduction 3M (p < 0.02). There was a significant (p < 0.03) correlation between ipsilesional NAA and NIHSS at 3M follow-up. Conclusion: Serial changes in metabolites suggest that MRS can be applied to monitor therapeutic changes. Post-treatment increasing trends of NAA concentration and significant correlation with NIHSS support a potential therapeutic effect.
Collapse
Affiliation(s)
- Muhammad E Haque
- Institute for Stroke and Cerebrovascular Diseases, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Refaat E Gabr
- Diagnostic and Interventional Imaging, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Sarah D George
- Institute for Stroke and Cerebrovascular Diseases, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Seth B Boren
- Institute for Stroke and Cerebrovascular Diseases, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Farhaan S Vahidy
- Institute for Stroke and Cerebrovascular Diseases, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Xu Zhang
- Biostatistics, Epidemiology, Research Design Component, Center for Clinical and Translational Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Octavio D Arevalo
- Diagnostic and Interventional Imaging, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Susan Alderman
- Institute for Stroke and Cerebrovascular Diseases, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Ponnada A Narayana
- Diagnostic and Interventional Imaging, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Khader M Hasan
- Diagnostic and Interventional Imaging, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Elliott R Friedman
- Diagnostic and Interventional Imaging, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Clark W Sitton
- Diagnostic and Interventional Imaging, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Sean I Savitz
- Institute for Stroke and Cerebrovascular Diseases, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
4
|
He M, Shi X, Yang M, Yang T, Li T, Chen J. Mesenchymal stem cells-derived IL-6 activates AMPK/mTOR signaling to inhibit the proliferation of reactive astrocytes induced by hypoxic-ischemic brain damage. Exp Neurol 2018; 311:15-32. [PMID: 30213506 DOI: 10.1016/j.expneurol.2018.09.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 08/10/2018] [Accepted: 09/07/2018] [Indexed: 01/08/2023]
Abstract
Mesenchymal stem cells (MSCs) treatment is an effective strategy for the functional repair of central nervous system (CNS) insults through the production of bioactive molecules. We have previously demonstrated that the interleukin-6 (IL-6) secreted by MSCs plays an anti-apoptotic role in injured astrocytes and partly promotes functional recovery in neonatal rats with hypoxic-ischemic brain damage (HIBD). However, the mechanisms of IL-6 underlying the proliferation of injured astrocytes have not been fully elucidated. In this study, we investigated the therapeutic effects of MSCs on astrocyte proliferation in neonatal rats subjected to HIBD. A HIBD model was established in Sprague Dawley (SD) rats, and MSCs were administered by intracerebroventricular injection 5 days after HIBD. Rat primary astrocytes were cultured, subjected to oxygen glucose deprivation (OGD) injury and then immediately co-cultured with MSCs in vitro. Immunofluorescence staining, Cell Counting Kit (CCK)-8, flow cytometry, Ca2+ imaging, enzyme-linked immunosorbent assay (ELISA), western blotting, and co-immunoprecipitation (Co-IP) were performed. We found that MSCs transplantation not only promoted the recovery of learning and memory function in HIBD rats but also significantly reduced the number of Ki67+/glial fibrillary acidic protein (GFAP)+ cells in the hippocampi 7-14 days after HIBD. In addition to increasing IL-6 expression in both the hippocampi of HIBD rats and astrocyte culture medium, MSCs treatment in vitro significantly increased the expression levels of glycoprotein (gp) 130 and phosphorylated AMP-activated protein kinase α (p-AMPKα) and decreased the expression levels of p-mammalian target of rapamycin (mTOR) and its downstream targets. Furthermore, MSCs treatment induced a protein-protein interaction between gp130 and p-AMPKα. Suppression of IL-6 expression in MSCs reversed the above regulatory functions of MSCs in hippocampal astrocytes. The utilization of rapamycin further confirmed that mTOR participated in the proliferation of reactive astrocytes. These findings suggest that endogenous IL-6 produced by MSCs in the HIBD microenvironment provides therapeutic advantages by activating AMPK/mTOR signaling, thus reducing the proliferation of reactive astrocytes.
Collapse
Affiliation(s)
- Mulan He
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China; Chongqing Stem Cell Therapy Engineering Technical Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Xia Shi
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China; Chongqing Stem Cell Therapy Engineering Technical Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Miao Yang
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China; Chongqing Stem Cell Therapy Engineering Technical Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Ting Yang
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| | - Tingyu Li
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
| | - Jie Chen
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China; Chongqing Stem Cell Therapy Engineering Technical Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, China.
| |
Collapse
|
5
|
Song M, Lee JH, Bae J, Bu Y, Kim EC. Human Dental Pulp Stem Cells Are More Effective Than Human Bone Marrow-Derived Mesenchymal Stem Cells in Cerebral Ischemic Injury. Cell Transplant 2017; 26:1001-1016. [PMID: 28105979 DOI: 10.3727/096368916x694391] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
We compared the therapeutic effects and mechanism of transplanted human dental pulp stem cells (hDPSCs) and human bone marrow-derived mesenchymal stem cells (hBM-MSCs) in a rat stroke model and an in vitro model of ischemia. Rats were intravenously injected with hDPSCs or hBM-MSCs 24 h after middle cerebral artery occlusion (MCAo), and both groups showed improved functional recovery and reduced infarct volume versus control rats, but the hDPSC group showed greater reduction in infarct volume than the hBM-MSC group. The positive area for the endothelial cell marker was greater in the lesion boundary areas in the hDPSC group than in the hBM-MSC group. Administration of hDPSCs to rats with stroke significantly decreased reactive gliosis, as evidenced by the attenuation of MCAo-induced GFAP+/nestin+ and GFAP+/Musashi-1+ cells, compared with hBM-MSCs. In vivo findings were confirmed by in vitro data illustrating that hDPSCs showed superior neuroprotective, migratory, and in vitro angiogenic effects in oxygen-glucose deprivation (OGD)-injured human astrocytes (hAs) versus hBM-MSCs. Comprehensive comparative bioinformatics analyses from hDPSC- and hBM-MSC-treated in vitro OGD-injured hAs were examined by RNA sequencing technology. In gene ontology and KEGG pathway analyses, significant pathways in the hDPSC-treated group were the MAPK and TGF-β signaling pathways. Thus, hDPSCs may be a better cell therapy source for ischemic stroke than hBM-MSCs.
Collapse
|
6
|
Xin H, Wang F, Li Y, Lu QE, Cheung WL, Zhang Y, Zhang ZG, Chopp M. Secondary Release of Exosomes From Astrocytes Contributes to the Increase in Neural Plasticity and Improvement of Functional Recovery After Stroke in Rats Treated With Exosomes Harvested From MicroRNA 133b-Overexpressing Multipotent Mesenchymal Stromal Cells. Cell Transplant 2016; 26:243-257. [PMID: 27677799 DOI: 10.3727/096368916x693031] [Citation(s) in RCA: 188] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
We previously demonstrated that multipotent mesenchymal stromal cells (MSCs) that overexpress microRNA 133b (miR-133b) significantly improve functional recovery in rats subjected to middle cerebral artery occlusion (MCAO) compared with naive MSCs and that exosomes generated from naive MSCs mediate the therapeutic benefits of MSC therapy for stroke. Here we investigated whether exosomes isolated from miR-133b-overexpressing MSCs (Ex-miR-133b+) exert amplified therapeutic effects. Rats subjected to 2 h of MCAO were intra-arterially injected with Ex-miR-133b+, exosomes from MSCs infected by blank vector (Ex-Con), or phosphate-buffered saline (PBS) and were sacrificed 28 days after MCAO. Compared with the PBS treatment, both exosome treatment groups exhibited significant improvement of functional recovery. Ex-miR-133b+ treatment significantly increased functional improvement and neurite remodeling/brain plasticity in the ischemic boundary area compared with the Ex-Con treatment. Treatment with Ex-miR-133b+ also significantly increased brain exosome content compared with Ex-Con treatment. To elucidate mechanisms underlying the enhanced therapeutic effects of Ex-miR-133b+, astrocytes cultured under oxygen- and glucose-deprived (OGD) conditions were incubated with exosomes harvested from naive MSCs (Ex-Naive), miR-133b downregulated MSCs (Ex-miR-133b-), and Ex-miR-133b+. Compared with the Ex-Naive treatment, Ex-miR-133b+ significantly increased exosomes released by OGD astrocytes, whereas Ex-miR-133b- significantly decreased the release. Also, exosomes harvested from OGD astrocytes treated with Ex-miR-133b+ significantly increased neurite branching and elongation of cultured cortical embryonic rat neurons compared with the exosomes from OGD astrocytes subjected to Ex-Con. Our data suggest that exosomes harvested from miR-133b-overexpressing MSCs improve neural plasticity and functional recovery after stroke with a contribution from a stimulated secondary release of neurite-promoting exosomes from astrocytes.
Collapse
|
7
|
Li Y, Xu XL, Zhao D, Pan LN, Huang CW, Guo LJ, Lu Q, Wang J. TLR3 ligand Poly IC Attenuates Reactive Astrogliosis and Improves Recovery of Rats after Focal Cerebral Ischemia. CNS Neurosci Ther 2016; 21:905-13. [PMID: 26494128 DOI: 10.1111/cns.12469] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 09/06/2015] [Accepted: 09/13/2015] [Indexed: 01/08/2023] Open
Abstract
AIMS Brain ischemia activates astrocytes in a process known as astrogliosis. Although this process has beneficial effects, excessive astrogliosis can impair neuronal recovery. Polyinosinic-polycytidylic acid (Poly IC) has shown neuroprotection against cerebral ischemia-reperfusion injury, but whether it regulates reactive astrogliosis and glial scar formation is not clear. METHODS We exposed cultured astrocytes to oxygen-glucose deprivation/reoxygenation (OGD/R) and used a rat middle cerebral artery occlusion (MCAO)/reperfusion model to investigate the effects of Poly IC. Astrocyte proliferation and proliferation-related molecules were evaluated by immunostaining and Western blotting. Neurological deficit scores, infarct volumes and neuroplasticity were evaluated in rats after transient MCAO. RESULTS In vitro, Poly IC inhibited astrocyte proliferation, upregulated Toll-like receptor 3 (TLR3) expression, upregulated interferon-β, and downregulated interleukin-6 production. These changes were blocked by a neutralizing antibody against TLR3, suggesting that Poly IC function is TLR3-dependent. Moreover, in the MCAO model, Poly IC attenuated reactive astrogliosis, reduced brain infarction volume, and improved neurological function. In addition, Poly IC prevented MCAO-induced reductions in soma size, dendrite length, and number of dendritic bifurcations in cortical neurons of the infarct penumbra. CONCLUSIONS By ameliorating astrogliosis-related damage, Poly IC is a potential therapeutic agent for attenuating neuronal damage and promoting recovery after brain ischemia.
Collapse
Affiliation(s)
- Yang Li
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xu-Lin Xu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,The Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Dan Zhao
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin-Na Pan
- Medical Department of Neurology, The Second Hospital of Nanchang, Nanchang, China
| | - Chun-Wei Huang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lian-Jun Guo
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,The Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Qing Lu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,The Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| |
Collapse
|
8
|
Huang W, Lv B, Zeng H, Shi D, Liu Y, Chen F, Li F, Liu X, Zhu R, Yu L, Jiang X. Paracrine Factors Secreted by MSCs Promote Astrocyte Survival Associated With GFAP Downregulation After Ischemic Stroke via p38 MAPK and JNK. J Cell Physiol 2015; 230:2461-75. [DOI: 10.1002/jcp.24981] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 02/23/2015] [Accepted: 03/02/2015] [Indexed: 12/16/2022]
Affiliation(s)
- Weiyi Huang
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| | - Bingke Lv
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| | - Huijun Zeng
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| | - Dandan Shi
- Department of Anatomy; Key Laboratory of Construction and Detection of Guangdong Province; Southern Medical University; Guangzhou China
| | - Yi Liu
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| | - Fanfan Chen
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| | - Feng Li
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| | - Xinghui Liu
- Department of Anatomy; Key Laboratory of Construction and Detection of Guangdong Province; Southern Medical University; Guangzhou China
| | - Rong Zhu
- Department of Anatomy; Key Laboratory of Construction and Detection of Guangdong Province; Southern Medical University; Guangzhou China
| | - Lei Yu
- Department of Anatomy; Key Laboratory of Construction and Detection of Guangdong Province; Southern Medical University; Guangzhou China
| | - Xiaodan Jiang
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| |
Collapse
|
9
|
Song M, Jue SS, Cho YA, Kim EC. Comparison of the effects of human dental pulp stem cells and human bone marrow-derived mesenchymal stem cells on ischemic human astrocytes in vitro. J Neurosci Res 2015; 93:973-83. [PMID: 25663284 DOI: 10.1002/jnr.23569] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 11/27/2014] [Accepted: 01/12/2015] [Indexed: 12/11/2022]
Abstract
This study assesses the cytoprotective effects of human dental pulp stem cells (hDPSCs) and conditioned medium from hDPSCs (CM-hDPSCs) on ischemic human astrocytes (hAs) in vitro compared with human bone marrow-derived mesenchymal stem cells (hMSCs). Ischemia of hAs was induced by oxygen-glucose deprivation (OGD). CM-hDPSCs and hMSCs were collected after 48 hr of culture. Cell death was determined by 3-[4,5-dimethylthialzol-2-yl]-2,5-diphenyltetrazolium bromide and cellular ATP assays. The expression of glial fibrillary acidic protein (GFAP) and musashi-1 as markers of reactive astrogliosis was examined with immunochemical staining. mRNA expression and reactive oxygen species (ROS) were analyzed by RT-PCR and flow cytometry, respectively. OGD increased cytotoxicity in a time-dependent manner and decreased cellular ATP content concomitantly in hAs. Pretreatment and posttreatment with hDPSCs were associated with greater recovery from OGD-induced cytotoxicity in hAs compared with hMSCs. Similarly, CM-hDPSCs had a greater effect on OGD-induced cytotoxicity in a dose-dependent manner. Pre- and posttreatment with CM-hDPSCs or CM-hMSCs attenuated OGD-induced GFAP, nestin, and musashi-1 expression in hAs. Furthermore, treatment of cells with CM-hDPSCs and hMSCs blocked OGD-induced ROS production and interleukin-1ß upregulation. This study demonstrates for the first time that hDPSCs and CM-hDPSCs confer superior cytoprotection against cell death in an in vitro OGD model compared with hMSCs as shown by cell viability assay. Reactive gliosis, ROS production, and inflammatory mediators might contribute to this protective effect. Therefore, hDPSCs could represent an alternative source of cell therapy for ischemic stroke.
Collapse
Affiliation(s)
- Miyeoun Song
- Department of Oral and Maxillofacial Pathology, Research Center for Tooth and Periodontal Regeneration, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | | | | | | |
Collapse
|
10
|
Xin H, Li Y, Chopp M. Exosomes/miRNAs as mediating cell-based therapy of stroke. Front Cell Neurosci 2014; 8:377. [PMID: 25426026 PMCID: PMC4226157 DOI: 10.3389/fncel.2014.00377] [Citation(s) in RCA: 198] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 10/22/2014] [Indexed: 12/19/2022] Open
Abstract
Cell-based therapy, e.g., multipotent mesenchymal stromal cell (MSC) treatment, shows promise for the treatment of various diseases. The strong paracrine capacity of these cells and not their differentiation capacity, is the principal mechanism of therapeutic action. MSCs robustly release exosomes, membrane vesicles (~30–100 nm) originally derived in endosomes as intraluminal vesicles, which contain various molecular constituents including proteins and RNAs from maternal cells. Contained among these constituents, are small non-coding RNA molecules, microRNAs (miRNAs), which play a key role in mediating biological function due to their prominent role in gene regulation. The release as well as the content of the MSC generated exosomes are modified by environmental conditions. Via exosomes, MSCs transfer their therapeutic factors, especially miRNAs, to recipient cells, and therein alter gene expression and thereby promote therapeutic response. The present review focuses on the paracrine mechanism of MSC exosomes, and the regulation and transfer of exosome content, especially the packaging and transfer of miRNAs which enhance tissue repair and functional recovery. Perspectives on the developing role of MSC mediated transfer of exosomes as a therapeutic approach will also be discussed.
Collapse
Affiliation(s)
- Hongqi Xin
- Department of Neurology, Henry Ford Hospital Detroit, MI, USA
| | - Yi Li
- Department of Neurology, Henry Ford Hospital Detroit, MI, USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital Detroit, MI, USA ; Department of Physics, Oakland University Rochester, MI, USA
| |
Collapse
|
11
|
Sun X, Fang B, Zhao X, Zhang G, Ma H. Preconditioning of mesenchymal stem cells by sevoflurane to improve their therapeutic potential. PLoS One 2014; 9:e90667. [PMID: 24599264 PMCID: PMC3944720 DOI: 10.1371/journal.pone.0090667] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Accepted: 02/05/2014] [Indexed: 01/09/2023] Open
Abstract
Background Bone marrow mesenchymal stem cells (MSCs) have been found to produce beneficial effects on ischemia-reperfusion injury. However, most of the MSCs died when transplanted into the ischemic tissue, which severely limit their therapeutic potential. Methods Using an in vitro model of hypoxia and serum deprivation (H/SD), we investigated the hypothesis that sevoflurane preconditioning could protect MSCs against H/SD-induced apoptosis and improve their migration, proliferation, and therapeutic potential. The H/SD of MSCs and neuron-like PC12 cells were incubated in a serum-free medium and an oxygen concentration below 0.1% for 24 h. Sevoflurane preconditioning was performed through a 2-h incubation of MSCs in an airtight chamber filled with 2 vol% sevoflurane. Apoptosis of MSCs or neuron-like PC12 cells was assessed using Annexin V-FITC/propidium iodide (PI). Furthermore, the mitochondrial membrane potential was assessed using lipophilic cationic probe. The proliferation rate was evaluated through cell cycle analysis. Finally, HIF-1α, HIF-2α, VEGF and p-Akt/Akt levels were measured by western blot. Results Sevoflurane preconditioning minimized the MSCs apoptosis and loss of mitochondrial membrane potential. Furthermore, it increased the migration and expression of HIF-1α, HIF-2α, VEGF, and p-Akt/Akt, reduced by H/SD. In addition, neuron-like PC12 cells were more resistant to H/SD-induced apoptosis when they were co-cultured with sevoflurane preconditioning MSCs. Conclusion These findings suggest that sevoflurane preconditioning produces protective effects on survival and migration of MSCs against H/SD, as well as improving the therapeutic potential of MSCs. These beneficial effects might be mediated at least in part by upregulating HIF-1α, HIF-2α, VEGF, and p-Akt/Akt.
Collapse
Affiliation(s)
- Xuejun Sun
- Department of Anesthesiology, First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| | - Bo Fang
- Department of Anesthesiology, First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| | - Xi Zhao
- Department of Anesthesiology, First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| | - Guangwei Zhang
- Department of Cardiac Surgery, First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| | - Hong Ma
- Department of Anesthesiology, First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
- * E-mail:
| |
Collapse
|
12
|
East E, Johns N, Georgiou M, Golding JP, Loughlin AJ, Kingham PJ, Phillips JB. A 3D in vitro model reveals differences in the astrocyte response elicited by potential stem cell therapies for CNS injury. Regen Med 2013; 8:739-46. [PMID: 24147529 PMCID: PMC3831573 DOI: 10.2217/rme.13.61] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
AIM This study aimed to develop a 3D culture model to test the extent to which transplanted stem cells modulate astrocyte reactivity, where exacerbated glial cell activation could be detrimental to CNS repair success. MATERIALS & METHODS The reactivity of rat astrocytes to bone marrow mesenchymal stem cells, neural crest stem cells (NCSCs) and differentiated adipose-derived stem cells was assessed after 5 days. Schwann cells were used as a positive control. RESULTS NCSCs and differentiated Schwann cell-like adipose-derived stem cells did not increase astrocyte reactivity. Highly reactive responses to bone marrow mesenchymal stem cells and Schwann cells were equivalent. CONCLUSION This approach can screen therapeutic cells prior to in vivo testing, allowing cells likely to trigger a substantial astrocyte response to be identified at an early stage. NCSCs and differentiated Schwann cell-like adipose-derived stem cells may be useful in treating CNS damage without increasing astrogliosis.
Collapse
Affiliation(s)
- Emma East
- Department of Life Health & Chemical Sciences, The Open University, Walton Hall, Milton Keynes, MK7 6AA, UK
| | - Noémie Johns
- Department of Life Health & Chemical Sciences, The Open University, Walton Hall, Milton Keynes, MK7 6AA, UK
| | - Melanie Georgiou
- Department of Life Health & Chemical Sciences, The Open University, Walton Hall, Milton Keynes, MK7 6AA, UK
| | - Jon P Golding
- Department of Life Health & Chemical Sciences, The Open University, Walton Hall, Milton Keynes, MK7 6AA, UK
| | - A Jane Loughlin
- Department of Life Health & Chemical Sciences, The Open University, Walton Hall, Milton Keynes, MK7 6AA, UK
| | - Paul J Kingham
- Department of Integrative Medical Biology, Umeå University, SE 901 87, Umeå, Sweden
| | - James B Phillips
- Department of Life Health & Chemical Sciences, The Open University, Walton Hall, Milton Keynes, MK7 6AA, UK
- Department of Biomaterials & Tissue Engineering, UCL Eastman Dental Institute, 256 Gray’s Inn Road, London WC1X 8LD, UK
| |
Collapse
|
13
|
Kazanis I, Gorenkova N, Zhao JW, Franklin RJM, Modo M, Ffrench-Constant C. The late response of rat subependymal zone stem and progenitor cells to stroke is restricted to directly affected areas of their niche. Exp Neurol 2013; 248:387-97. [PMID: 23830949 PMCID: PMC3782662 DOI: 10.1016/j.expneurol.2013.06.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2013] [Revised: 06/20/2013] [Accepted: 06/24/2013] [Indexed: 01/08/2023]
Abstract
Ischaemia leads to increased proliferation of progenitors in the subependymal zone (SEZ) neurogenic niche of the adult brain and to generation and migration of newborn neurons. Here we investigated the spatiotemporal characteristics of the mitotic activity of adult neural stem and progenitor cells in the SEZ during the sub-acute and chronic post-ischaemic phases. Ischaemia was induced by performing a 1h unilateral middle cerebral artery occlusion (MCAO) and tissue was collected 4/5 weeks and 1 year after the insult. Neural stem cells (NSCs) responded differently from their downstream progenitors to MCAO, with NSCs being activated only transiently whilst progenitors remain activated even at 1 year post-injury. Importantly, mitotic activation was observed only in the affected areas of the niche and specifically in the dorsal half of the SEZ. Analysis of the topography of mitoses, in relation to the anatomy of the lesion and to the position of ependymal cells and blood vessels, suggested an interplay between lesion-derived recruiting signals and the local signals that normally control proliferation in the chronic post-ischaemic phase.
Collapse
Affiliation(s)
- Ilias Kazanis
- MRC Cambridge Centre for Stem Cell Biology and Regenerative Medicine and Department of Veterinary Medicine, University of Cambridge, Cambridge, UK.
| | | | | | | | | | | |
Collapse
|
14
|
Gao Z, Zhu Q, Zhang Y, Zhao Y, Cai L, Shields CB, Cai J. Reciprocal modulation between microglia and astrocyte in reactive gliosis following the CNS injury. Mol Neurobiol 2013; 48:690-701. [PMID: 23613214 DOI: 10.1007/s12035-013-8460-4] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 04/09/2013] [Indexed: 12/16/2022]
Abstract
Reactive gliosis, also known as glial scar formation, is an inflammatory response characterized by the proliferation of microglia and astrocytes as well as astrocytic hypertrophy following injury in the central nervous system (CNS). The glial scar forms a physical and molecular barrier to isolate the injured area from adjacent normal nervous tissue for re-establishing the integrity of the CNS. It prevents the further spread of cellular damage but represents an obstacle to regrowing axons. In this review, we integrated the current findings to elucidate the tightly reciprocal modulation between activated microglia and astrocytes in reactive gliosis and proposed that modification of cellular response to the injury or cellular reprogramming in the glial scar could lead advances in axon regeneration and functional recovery after the CNS injury.
Collapse
Affiliation(s)
- Zhongwen Gao
- Department of Spine Surgery, the First Hospital of Jilin University, 71 Xinmin Street, Changchun, Jilin, 130021, China
| | | | | | | | | | | | | |
Collapse
|
15
|
Gubert F, Zaverucha-do-Valle C, Figueiredo FR, Bargas-Rega M, Paredes BD, Mencalha AL, Abdelhay E, Gutfilen B, Barbosa da Fonseca LM, Mendez-Otero R, Santiago MF. Bone-marrow cell therapy induces differentiation of radial glia-like cells and rescues the number of oligodendrocyte progenitors in the subventricular zone after global cerebral ischemia. Stem Cell Res 2013; 10:241-56. [DOI: 10.1016/j.scr.2012.11.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 10/29/2012] [Accepted: 11/30/2012] [Indexed: 01/17/2023] Open
|
16
|
Bluguermann C, Wu L, Petrigliano F, McAllister D, Miriuka S, Evseenko DA. Novel aspects of parenchymal-mesenchymal interactions: from cell types to molecules and beyond. Cell Biochem Funct 2013; 31:271-80. [PMID: 23315627 DOI: 10.1002/cbf.2950] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 12/06/2012] [Accepted: 12/06/2012] [Indexed: 12/26/2022]
Abstract
Mesenchymal stem or stromal cells (MSCs) were initially isolated from the bone marrow and received their name on the basis of their ability to differentiate into multiple lineages such as bone, cartilage, fat and muscle. However, more recent studies suggest that MSCs residing in perivascular compartments of the small and large blood vessels play a regulatory function supporting physiologic and pathologic responses of parenchymal cells, which define the functional representation of an organ or tissue. MSCs secrete or express factors that reach neighbouring parenchymal cells via either a paracrine effect or a direct cell-to-cell interaction promoting functional activity, survival and proliferation of the parenchymal cells. Previous concept of 'epithelial-stromal' interactions can now be widened. Given that MSC can also support hematopoietic, neuronal and other non-epithelial parenchymal lineages, terms 'parenchymal-stromal' or 'parenchymal-mesenchymal' interactions may better describe the supportive or 'trophic' functions of MSC. Importantly, in many cases, MSCs specifically provide supportive microenvironment for the most primitive stem or progenitor populations and therefore can play a role as 'stem/progenitor niche' forming cells. So far, regulatory roles of MSCs have been reported in many tissues. In this review article, we summarize the latest studies that focused on the supportive function of MSC. This thread of research leads to a new perspective on the interactions between parenchymal and mesenchymal cells and justifies a principally novel approach for regenerative medicine based on co-application of MSC and parenchymal cell for the most efficient tissue repair.
Collapse
Affiliation(s)
- Carolina Bluguermann
- Department of Orthopaedic Surgery, Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California at Los Angeles (UCLA), Los Angeles, CA, USA
| | | | | | | | | | | |
Collapse
|
17
|
Ding Y, Yan Q, Ruan JW, Zhang YQ, Li WJ, Zeng X, Huang SF, Zhang YJ, Wu JL, Fisher D, Dong H, Zeng YS. Electroacupuncture Promotes the Differentiation of Transplanted Bone Marrow Mesenchymal Stem Cells Overexpressing TrkC into Neuron-Like Cells in Transected Spinal Cord of Rats. Cell Transplant 2013; 22:65-86. [DOI: 10.3727/096368912x655037] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Our previous study indicated that electroacupuncture (EA) could increase neurotrophin-3 (NT-3) levels in the injured spinal cord, stimulate the differentiation of transplanted bone marrow mesenchymal stem cells (MSCs), and improve functional recovery in the injured spinal cord of rats. However, the number of neuron-like cells derived from the MSCs is limited. It is known that NT-3 promotes the survival and differentiation of neurons by preferentially binding to its receptor TrkC. In this study, we attempted to transplant TrkC gene-modified MSCs (TrkC-MSCs) into the spinal cord with transection to investigate whether EA treatment could promote NT-3 secretion in the injured spinal cord and to determine whether increased NT-3 could further enhance transplanted MSCs overexpressing TrkC to differentiate into neuron-like cells, resulting in increased axonal regeneration and functional improvement in the injured spinal cord. Our results showed that EA increased NT-3 levels; furthermore, it promoted neuron-phenotype differentiation, synaptogenesis, and myelin formation of transplanted TrkC-MSCs. In addition, TrkC-MSC transplantation combined with EA (the TrkC-MSCs + EA group) treatment promoted the growth of the descending BDA-labeled corticospinal tracts (CSTs) and 5-HT-positive axonal regeneration across the lesion site into the caudal cord. In addition, the conduction of cortical motor-evoked potentials (MEPs) and hindlimb locomotor function increased as compared to controls (treated with the LacZ-MSCs, TrkC-MSCs, and LacZ-MSCs + EA groups). In the TrkC-MSCs + EA group, the injured spinal cord also showed upregulated expression of the proneurogenic factors laminin and GAP-43 and downregulated GFAP and chondroitin sulfate proteoglycans (CSPGs), major inhibitors of axonal growth. Together, our data suggest that TrkC-MSC transplantation combined with EA treatment spinal cord injury not only increased MSC survival and differentiation into neuron-like cells but also promoted CST regeneration across injured sites to the caudal cord and functional improvement, perhaps due to increase of NT-3 levels, upregulation of laminin and GAP-43, and downregulation of GFAP and CSPG proteins.
Collapse
Affiliation(s)
- Ying Ding
- Division of Neuroscience, Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Qing Yan
- Division of Neuroscience, Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jing-Wen Ruan
- Department of Acupuncture of the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yan-Qing Zhang
- Division of Neuroscience, Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Wen-Jie Li
- Division of Neuroscience, Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiang Zeng
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Si-Fan Huang
- Division of Neuroscience, Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yu-Jiao Zhang
- Division of Neuroscience, Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jin-Lang Wu
- Department of Electron Microscope, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Danny Fisher
- Department of Psychiatry and Behavioral Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Hongxin Dong
- Department of Psychiatry and Behavioral Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Yuan-Shan Zeng
- Division of Neuroscience, Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
18
|
Wasielewski B, Jensen A, Roth-Härer A, Dermietzel R, Meier C. Neuroglial activation and Cx43 expression are reduced upon transplantation of human umbilical cord blood cells after perinatal hypoxic-ischemic injury. Brain Res 2012; 1487:39-53. [PMID: 22796290 DOI: 10.1016/j.brainres.2012.05.066] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2012] [Accepted: 05/31/2012] [Indexed: 12/15/2022]
Abstract
Glial cells play a crucial role in the pathomechanism of perinatal hypoxic-ischemic brain injury (HI) and are involved in the maintenance of a chronic state of inflammation that causes delayed neuronal damage. Activation of astrocytes is one factor prolonging brain damage and contributing to the formation of a glial scar that limits neuronal plasticity. In this context, the major astrocytic gap junction protein Connexin 43 (Cx43) has been ascribed various functions including regulation of astrocytic migration and proliferation. Here, we investigate glial responses like microglia/macrophages and astrocytic activation in a rat model of neonatal HI and characterize changes of these parameters upon transplantation of human umbilical cord blood cells (hUCB). As an alleviation of motor function in lesioned rats has previously been described in transplanted animals, we analyze the putative correlation between motor function and glial activation over time. The lesion-induced impairment of motor function, assessed by forelimb use bias, muscle strength and distal spasticity, was alleviated upon transplantation of hUCB short and long term. HI induced an acute inflammatory reaction with activation of microglia/macrophages and reactive astrogliosis associated with perilesional upregulation of Cx43 that slowly declined during the chronic post-ischemic phase. hUCB transplantation accelerated the regression of inflammatory events, narrowed the perilesional astrocytic wall and led to a downregulation of the investigated astrocytic proteins. Thus, in the immature brain, hUCB may indirectly reduce secondary cell death upon hypoxia-ischemia and facilitate post-ischemic plasticity through the attenuation of reactive gliosis. This article is part of a Special Issue entitled Electrical Synapses.
Collapse
Affiliation(s)
- Bianca Wasielewski
- Department of Neuroanatomy and Molecular Brain Research, Ruhr-University Bochum, D-44801 Bochum, Germany
| | | | | | | | | |
Collapse
|
19
|
Scheibe F, Klein O, Klose J, Priller J. Mesenchymal stromal cells rescue cortical neurons from apoptotic cell death in an in vitro model of cerebral ischemia. Cell Mol Neurobiol 2012; 32:567-76. [PMID: 22290155 DOI: 10.1007/s10571-012-9798-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Accepted: 01/06/2012] [Indexed: 01/01/2023]
Abstract
Cell therapy with mesenchymal stromal cells (MSCs) was found to protect neurons from damage after experimental stroke and is currently under investigation in clinical stroke trials. In order to elucidate the mechanisms of MSC-induced neuroprotection, we used the in vitro oxygen–glucose deprivation (OGD) model of cerebral ischemia. Co-culture of primary cortical neurons with MSCs in a transwell co-culture system for 48 h prior to OGD-reduced neuronal cell death by 30-35%. Similar protection from apoptosis was observed with MSC-conditioned media when added 48 h or 30 min prior to OGD, or even after OGD. Western blot analysis revealed increased phosphorylation of STAT3 and Akt in neuronal cultures after treatment with MSC-conditioned media. Inhibition of the PI3K/Akt pathway completely abolished the neuroprotective potential of MSC-conditioned media, suggesting that MSCs can improve neuronal survival by an Akt-dependent anti-apoptotic signaling cascade. Using mass spectrometry, we identified plasminogen activator inhibitor-1 as an active compound in MSC-conditioned media. Thus, paracrine factors secreted by MSCs protect neurons from apoptotic cell death in the OGD model of cerebral ischemia.
Collapse
Affiliation(s)
- Franziska Scheibe
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | |
Collapse
|
20
|
Skardelly M, Gaber K, Burdack S, Scheidt F, Hilbig H, Boltze J, Förschler A, Schwarz S, Schwarz J, Meixensberger J, Schuhmann MU. Long-term benefit of human fetal neuronal progenitor cell transplantation in a clinically adapted model after traumatic brain injury. J Neurotrauma 2011; 28:401-14. [PMID: 21083415 DOI: 10.1089/neu.2010.1526] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Experimental human fetal neural progenitor cell (hfNPC) transplantation has proven to be a promising therapeutic approach after traumatic brain injury (TBI). However, the long-term efficacy and safety, which are both highly important for clinical translation of this approach, have thus far not been investigated. This study investigated the effect of local (L, 1 × 10(5) cells) and systemic (S, 5 × 10(5) cells) administration of PKH-26-labeled pre-differentiated hfNPCs over a period of 12 weeks, beginning 24 h after severe controlled cortical impact TBI in Sprague-Dawley rats. Accelerating rotarod testing revealed a trend toward functional improvement beginning 1 week after transplantation, and persisting until the end of the experiment. The traumatic lesion volume as quantified by magnetic resonance imaging was smaller in both treatment groups compared to control (C) animals (C = 54.50 mm(3), L = 32 mm(3), S = 37.50 mm(3)). Correspondingly, neuronal (NeuN) staining showed increased neuronal survival at the border of the lesion in both transplanted groups (S = 92.4%; L = 87.2%; 72.5%). Histological analysis of the brain compartments revealed transiently increased angiogenesis and reduced astroglial reaction during the first 4 weeks post-transplantation. PKH-26-positive cells were detected exclusively after local transplantation without any evidence of tumor formation. However, graft differentiation was seen only in very rare cases. In conclusion, transplantation of hfNPCs improved the long-term functional outcome after TBI, diminished trauma lesion size, and increased neuronal survival in the border zone of the lesion. This therapeutic effect was not likely due to cell replacement, but was associated with transiently increased angiogenesis and reduced astrogliosis.
Collapse
Affiliation(s)
- Marco Skardelly
- Department of Neurosurgery, University of Leipzig, Leipzig, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Yin Y, Li G. Hypoxia induces T Helper 17 cell upregulation in cultured peripheral blood mononuclear cells from chronic stage patients of severe cerebral infarction. Microbiol Immunol 2011; 55:130-4. [PMID: 21204953 DOI: 10.1111/j.1348-0421.2010.00301.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Studies have shown the IL-17A involvement in human ischemic stroke patients in vivo. Whether the IL-17A expression was originated from Th17 and could be stimulated by hypoxia remained unknown. Here we report the Th17 upregulation in anaerobic cultured PBMC from chronic stage patients of severe cerebral infarction. By using ELISA and FACS we examined IL-1β, IFN-γ, IL-23 and IL-17A protein levels in the supernatants and Th1/Th17 ratios in PBMC. Statistical significance of Th17 but not Th1 upregulation was proved in 6-hr anaerobic cultured patient groups (P < 0.001). Hence, Th17 might be essential in the autoimmune pathogenesis when hypoxia recurs in severe ischemic stroke patients.
Collapse
Affiliation(s)
- YanHong Yin
- Department of Neurology, Chinese People Liberation Army (PLA) General Hospital, 28 Fuxing Road, Haidian District, Beijing 100854, China.
| | | |
Collapse
|
22
|
Horn AP, Bernardi A, Luiz Frozza R, Grudzinski PB, Hoppe JB, de Souza LF, Chagastelles P, de Souza Wyse AT, Bernard EA, Battastini AMO, Campos MM, Lenz G, Nardi NB, Salbego C. Mesenchymal Stem Cell-Conditioned Medium Triggers Neuroinflammation and Reactive Species Generation in Organotypic Cultures of Rat Hippocampus. Stem Cells Dev 2011; 20:1171-81. [DOI: 10.1089/scd.2010.0157] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Ana Paula Horn
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brazil
- Laboratório de Histologia, Instituto de Ciências Biológicas, FURG, Avenida Itália, Carreiros, Rio Grande, RS, Brazil
| | - Andressa Bernardi
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brazil
| | - Rudimar Luiz Frozza
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brazil
| | - Patrícia Bencke Grudzinski
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brazil
| | - Juliana Bender Hoppe
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brazil
| | - Luiz Fernando de Souza
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brazil
| | - Pedro Chagastelles
- Departamento de Genética, Instituto de Biociências, UFRGS, Porto Alegre, RS, Brazil
| | - Angela Terezinha de Souza Wyse
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brazil
| | - Elena Aida Bernard
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brazil
| | - Ana Maria Oliveira Battastini
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brazil
| | - Maria Martha Campos
- Faculdade de Odontologia e Instituto de Toxicologia, PUCRS, Porto Alegre, RS, Brazil
| | - Guido Lenz
- Departamento de Biofísica, Instituto de Biociências, UFRGS, Porto Alegre, RS, Brazil
| | - Nance Beyer Nardi
- Departamento de Genética, Instituto de Biociências, UFRGS, Porto Alegre, RS, Brazil
| | - Christianne Salbego
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brazil
| |
Collapse
|
23
|
Ding Y, Yan Q, Ruan JW, Zhang YQ, Li WJ, Zeng X, Huang SF, Zhang YJ, Wang S, Dong H, Zeng YS. Bone Marrow Mesenchymal Stem Cells and Electroacupuncture Downregulate the Inhibitor Molecules and Promote the Axonal Regeneration in the Transected Spinal Cord of Rats. Cell Transplant 2011; 20:475-91. [DOI: 10.3727/096368910x528102] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Our previous study has reported that electroacupuncture (EA) promotes survival, differentiation of bone marrow mesenchymal stem cells (MSCs), and functional improvement in spinal cord-transected rats. In this study, we further investigated the structural bases of this functional improvement and the potential mechanisms of axonal regeneration in injured spinal cord after MSCs and EA treatment. Five experimental groups, 1) sham control (Sham-control); 2) operated control (Op-control); 3) electroacupuncture treatment (EA); 4) MSCs transplantation (MSCs), and 5) MSCs transplantation combined with electroacupuncture (MSCs + EA), were designed for this study. Western blots and immunohistochemical staining were used to assess the fibrillary acidic protein (GFAP) and chondroitin sulfate proteoglycans (CSPGs) proteins expression. Basso, Beattie, Bresnahan (BBB) locomotion test, cortical motor evoked potentials (MEPs), and anterograde and retrograde tracing were utilized to assess cortical-spinal neuronal projection regeneration and functional recovery. In the MSCs + EA group, increased labeling descending corticospinal tract (CST) projections into the lesion site showed significantly improved BBB scales and enhanced motor evoked potentials after 10 weeks of MSCs transplant and EA treatment. The structural and functional recovery after MSCs + EA treatment may be due to downregulated GFAP and CSPGs protein expression, which prevented axonal degeneration as well as improved axonal regeneration.
Collapse
Affiliation(s)
- Ying Ding
- Division of Neuroscience, Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Qing Yan
- Division of Neuroscience, Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jing-Wen Ruan
- Department of Acupuncture of the first Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yan-Qing Zhang
- Division of Neuroscience, Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Wen-Jie Li
- Division of Neuroscience, Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiang Zeng
- Division of Neuroscience, Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Si-Fan Huang
- Division of Neuroscience, Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yu-Jiao Zhang
- Division of Neuroscience, Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Shirlene Wang
- Department of Psychiatry and Behavioral Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Hongxin Dong
- Department of Psychiatry and Behavioral Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Yuan-Shan Zeng
- Division of Neuroscience, Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Center for Stem Cell Biology and Tissue Engineering, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Institute of Spinal Cord Injury, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
24
|
Lee JK, Seo EM, Lee SS, Park SH, Sim YB, Jung JS, Kim SM, Suh HW. Activation of PPARalpha Attenuates IFNgamma and IL-1beta-induced Cell Proliferation in Astrocytes: Involvement of IL-6 Independent Pathway. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2010; 14:185-9. [PMID: 20631892 DOI: 10.4196/kjpp.2010.14.3.185] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2010] [Revised: 06/11/2010] [Accepted: 06/23/2010] [Indexed: 11/15/2022]
Abstract
The present study demonstrates the effect of fibrates, agonists of PPARalpha on cytokines-induced proliferation in primary cultured astrocytes. Alone or combination treatment with cytokines, such as IL-1beta (10 ng/ml), IFNgamma (10 ng/ml), and TNF-alpha (10 ng/ml) cause a significant increase of cell proliferation in a time-dependent manner. Treatment of astrocytes with bezafibrate and fenofibrate (0, 5, and 10 microM) reduced the IFNgamma and IL-1beta-induced cell proliferation in a dose-dependent manner. To address the involvement of IL-6 on the IFNgamma and IL-1beta-induced cell proliferation, released IL-6 level was measured. IFNgamma and IL-1beta cause an increase of released IL-6 protein level in a time-dependent manner. Furthermore, pretreatment with IL-6 antibody (0, 0.1, 1, 2.5, and 5 ng/ml) dose-dependently inhibited the IFNgamma and IL-1beta-induced cell proliferation. However, bezafibrate and fenofibrate did not affect increased mRNA and protein levels of IL-6 in IFNgamma and IL-1beta-stimulated astrocytes. Taken together, these results clearly suggest that activation of PPARalpha attenuates the IFNgamma and IL-1beta-induced cell proliferation through IL-6 independent pathway.
Collapse
Affiliation(s)
- Jin-Koo Lee
- Institute of Natural Medicine, Hallym University, Chuncheon 200-702, Korea
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Xin H, Li Y, Shen LH, Liu X, Wang X, Zhang J, Pourabdollah-Nejad D S, Zhang C, Zhang L, Jiang H, Zhang ZG, Chopp M. Increasing tPA activity in astrocytes induced by multipotent mesenchymal stromal cells facilitate neurite outgrowth after stroke in the mouse. PLoS One 2010; 5:e9027. [PMID: 20140248 PMCID: PMC2815778 DOI: 10.1371/journal.pone.0009027] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2009] [Accepted: 01/11/2010] [Indexed: 10/31/2022] Open
Abstract
We demonstrate that tissue plasminogen activator (tPA) and its inhibitors contribute to neurite outgrowth in the central nervous system (CNS) after treatment of stroke with multipotent mesenchymal stromal cells (MSCs). In vivo, administration of MSCs to mice subjected to middle cerebral artery occlusion (MCAo) significantly increased activation of tPA and downregulated PAI-1 levels in the ischemic boundary zone (IBZ) compared with control PBS treated mice, concurrently with increases of myelinated axons and synaptophysin. In vitro, MSCs significantly increased tPA levels and concomitantly reduced plasminogen activator inhibitor 1 (PAI-1) expression in astrocytes under normal and oxygen and glucose deprivation (OGD) conditions. ELISA analysis of conditioned medium revealed that MSCs stimulated astrocytes to secrete tPA. When primary cortical neurons were cultured in the conditioned medium from MSC co-cultured astrocytes, these neurons exhibited a significant increase in neurite outgrowth compared to conditioned medium from astrocytes alone. Blockage of tPA with a neutralizing antibody or knock-down of tPA with siRNA significantly attenuated the effect of the conditioned medium on neurite outgrowth. Addition of recombinant human tPA into cortical neuronal cultures also substantially enhanced neurite outgrowth. Collectively, these in vivo and in vitro data suggest that the MSC mediated increased activation of tPA in astrocytes promotes neurite outgrowth after stroke.
Collapse
Affiliation(s)
- Hongqi Xin
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Yi Li
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Li Hong Shen
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Xianshuang Liu
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Xinli Wang
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Jing Zhang
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, United States of America
| | | | - Chunling Zhang
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Li Zhang
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Hao Jiang
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, United States of America
| | - Michael Chopp
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, United States of America
- Department of Physics, Oakland University, Rochester, Michigan, United States of America
- * E-mail:
| |
Collapse
|
26
|
Mandal A, Shahidullah M, Delamere NA, Terán MA. Elevated hydrostatic pressure activates sodium/hydrogen exchanger-1 in rat optic nerve head astrocytes. Am J Physiol Cell Physiol 2009; 297:C111-20. [PMID: 19419999 DOI: 10.1152/ajpcell.00539.2008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Optic nerve head astrocytes become abnormal in eyes that have elevated intraocular pressure, and cultured astrocytes display altered protein expression after being subjected for > or = 1 days to elevated hydrostatic pressure. Here we show that 2-h elevated hydrostatic pressure (15 or 30 mmHg) causes phosphorylation of ERK1/2, ribosomal S6 protein kinase (p90(RSK)), and Na/H exchanger (NHE)1 in cultured rat optic nerve head astrocytes as judged by Western blot analysis. The MEK/ERK inhibitor U0126 abolished phosphorylation of NHE1 and p90(RSK) as well as ERK1/2. To examine NHE1 activity, cytoplasmic pH (pH(i)) was measured with BCECF and, in some experiments, cells were acidified by 5-min exposure to 20 mM ammonium chloride. Although baseline pH(i) was unaltered, the rate of pH(i) recovery from acidification was fourfold higher in pressure-treated astrocytes. In the presence of either U0126 or dimethylamiloride (DMA), an NHE inhibitor, hydrostatic pressure did not change the rate of pH(i) recovery. The findings are consistent with NHE1 activation due to phosphorylation of ERK1/2, p90(RSK), and NHE1 that occurs in response to hydrostatic pressure. These responses may precede long-term changes of protein expression known to occur in pressure-stressed astrocytes.
Collapse
Affiliation(s)
- Amritlal Mandal
- Dept. of Physiology, Univ. of Arizona, Tucson, AZ 85724, USA
| | | | | | | |
Collapse
|
27
|
Shen LH, Gao Q, Li Y, Savant-Bhonsale S, Chopp M. Down-regulation of neurocan expression in reactive astrocytes promotes axonal regeneration and facilitates the neurorestorative effects of bone marrow stromal cells in the ischemic rat brain. Glia 2008; 56:1747-54. [PMID: 18618668 PMCID: PMC2575136 DOI: 10.1002/glia.20722] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The glial scar, a primarily astrocytic structure bordering the infarct tissue inhibits axonal regeneration after stroke. Neurocan, an axonal extension inhibitory molecule, is up-regulated in the scar region after stroke. Bone marrow stromal cells (BMSCs) reduce the thickness of glial scar wall and facilitate axonal remodeling in the ischemic boundary zone. To further clarify the role of BMSCs in axonal regeneration and its underlying mechanism, the current study focused on the effect of BMSCs on neurocan expression in the ischemic brain. Thirty-one adult male Wistar rats were subjected to 2 h of middle cerebral artery occlusion followed by an injection of 3 x 10(6) rat BMSCs (n = 16) or phosphate-buffered saline (n = 15) into the tail vein 24 h later. Animals were sacrificed at 8 days after stroke. Immunostaining analysis showed that reactive astrocytes were the primary source of neurocan, and BMSC-treated animals had significantly lower neurocan and higher growth associated protein 43 expression in the penumbral region compared with control rats, which was confirmed by Western blot analysis of the brain tissue. To further investigate the effects of BMSCs on astrocyte neurocan expression, single reactive astrocytes were collected from the ischemic boundary zone using laser capture microdissection. Neurocan gene expression was significantly down-regulated in rats receiving BMSC transplantation (n = 4/group). Primary cultured astrocytes showed similar alterations; BMSC coculture during reoxygenation abolished the up-regulation of neurocan gene in astrocytes undergoing oxygen-glucose deprivation (n = 3/group). Our data suggest that BMSCs promote axonal regeneration by reducing neurocan expression in peri-infarct astrocytes.
Collapse
Affiliation(s)
- Li Hong Shen
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, U.S.A
| | - Qi Gao
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, U.S.A
| | - Yi Li
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, U.S.A
| | | | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, U.S.A
- Department of Physics, Oakland University, Rochester, Michigan, U.S.A
| |
Collapse
|