1
|
Gao F, Du W, Guo C, Geng P, Liu W, Jin X. α7nACh receptor, a promising target to reduce BBB damage by regulating inflammation and autophagy after ischemic stroke. Biomed Pharmacother 2024; 179:117337. [PMID: 39191022 DOI: 10.1016/j.biopha.2024.117337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 08/13/2024] [Accepted: 08/21/2024] [Indexed: 08/29/2024] Open
Abstract
Increased blood-brain barrier (BBB) permeability can lead to cerebral vasogenic edema and hemorrhagic transformation (HT) after reperfusion with tissue plasminogen activator (tPA), the only United States Food and Drug Administration (FDA)-approved treatment for acute ischemia stroke (AIS). The therapeutic benefits of tPA after AIS are partially outweighed by a more than a six-fold increase in the risk of symptomatic intracerebral hemorrhage. Therefore, strategies to protect the integrity of BBB are urgently needed to reduce HT and vasogenic edema after tPA thrombolysis or endovascular thrombectomy. Interestingly, an NIH study showed that smokers treated with tPA had a significantly lower prevalence of brain hemorrhage than nonsmokers, suggesting that cigarette smoking may protect patients treated with tPA from the side effects of cerebral hemorrhage. Importantly, we recently showed that treatment with nicotine reduces AIS-induced BBB damage and that modulating α7nAChR by modulation could reduce ischemia/reperfusion-induced BBB damage, suggesting that α7nAChR could be a potential target to reduce BBB after AIS. In this review, we first provide an overview of stroke and the impact of α7nAChR activation on BBB damage. Next, we discuss the features and mechanism of BBB destruction after AIS. We then discuss the effect of nicotine effect on BBB integrity as well as the mechanism underlying those effects. Finally, we discuss the side effects and potential strategies for modulating α7nAChR to reduce AIS-induced BBB damage.
Collapse
Affiliation(s)
- Fengying Gao
- Department of Emergency, Shanxi Provincial People's Hospital, Taiyuan 030001, China
| | - Weihong Du
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Chun Guo
- School of Biosciences, University of Sheffield, Firth Court, Western Bank, Sheffield, UK
| | - Panpan Geng
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Wencao Liu
- Department of Emergency, Shanxi Provincial People's Hospital, Taiyuan 030001, China.
| | - Xinchun Jin
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
2
|
Boyle BR, Berghella AP, Blanco-Suarez E. Astrocyte Regulation of Neuronal Function and Survival in Stroke Pathophysiology. ADVANCES IN NEUROBIOLOGY 2024; 39:233-267. [PMID: 39190078 DOI: 10.1007/978-3-031-64839-7_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
The interactions between astrocytes and neurons in the context of stroke play crucial roles in the disease's progression and eventual outcomes. After a stroke, astrocytes undergo significant changes in their morphology, molecular profile, and function, together termed reactive astrogliosis. Many of these changes modulate how astrocytes relate to neurons, inducing mechanisms both beneficial and detrimental to stroke recovery. For example, excessive glutamate release and astrocytic malfunction contribute to excitotoxicity in stroke, eventually causing neuronal death. Astrocytes also provide essential metabolic support and neurotrophic signals to neurons after stroke, ensuring homeostatic stability and promoting neuronal survival. Furthermore, several astrocyte-secreted molecules regulate synaptic plasticity in response to stroke, allowing for the rewiring of neural circuits to compensate for damaged areas. In this chapter, we highlight the current understanding of the interactions between astrocytes and neurons in response to stroke, explaining the varied mechanisms contributing to injury progression and the potential implications for future therapeutic interventions.
Collapse
Affiliation(s)
- Bridget R Boyle
- Department of Neuroscience, Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
- Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA
| | - Andrea P Berghella
- Department of Neuroscience, Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
- Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA
| | - Elena Blanco-Suarez
- Department of Neuroscience, Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.
- Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA.
- Department of Neurological Surgery, Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Collyer E, Blanco-Suarez E. Astrocytes in stroke-induced neurodegeneration: a timeline. FRONTIERS IN MOLECULAR MEDICINE 2023; 3:1240862. [PMID: 39086680 PMCID: PMC11285566 DOI: 10.3389/fmmed.2023.1240862] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/22/2023] [Indexed: 08/02/2024]
Abstract
Stroke is a condition characterized by sudden deprivation of blood flow to a brain region and defined by different post-injury phases, which involve various molecular and cellular cascades. At an early stage during the acute phase, fast initial cell death occurs, followed by inflammation and scarring. This is followed by a sub-acute or recovery phase when endogenous plasticity mechanisms may promote spontaneous recovery, depending on various factors that are yet to be completely understood. At later time points, stroke leads to greater neurodegeneration compared to healthy controls in both clinical and preclinical studies, this is evident during the chronic phase when recovery slows down and neurodegenerative signatures appear. Astrocytes have been studied in the context of ischemic stroke due to their role in glutamate re-uptake, as components of the neurovascular unit, as building blocks of the glial scar, and synaptic plasticity regulators. All these roles render astrocytes interesting, yet understudied players in the context of stroke-induced neurodegeneration. With this review, we provide a summary of previous research, highlight astrocytes as potential therapeutic targets, and formulate questions about the role of astrocytes in the mechanisms during the acute, sub-acute, and chronic post-stroke phases that may lead to neurorestoration or neurodegeneration.
Collapse
Affiliation(s)
| | - Elena Blanco-Suarez
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
4
|
Hernández IH, Villa-González M, Martín G, Soto M, Pérez-Álvarez MJ. Glial Cells as Therapeutic Approaches in Brain Ischemia-Reperfusion Injury. Cells 2021; 10:1639. [PMID: 34208834 PMCID: PMC8305833 DOI: 10.3390/cells10071639] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/24/2021] [Accepted: 06/26/2021] [Indexed: 02/07/2023] Open
Abstract
Ischemic stroke is the second cause of mortality and the first cause of long-term disability constituting a serious socioeconomic burden worldwide. Approved treatments include thrombectomy and rtPA intravenous administration, which, despite their efficacy in some cases, are not suitable for a great proportion of patients. Glial cell-related therapies are progressively overcoming inefficient neuron-centered approaches in the preclinical phase. Exploiting the ability of microglia to naturally switch between detrimental and protective phenotypes represents a promising therapeutic treatment, in a similar way to what happens with astrocytes. However, the duality present in many of the roles of these cells upon ischemia poses a notorious difficulty in disentangling the precise pathways to target. Still, promoting M2/A2 microglia/astrocyte protective phenotypes and inhibiting M1/A1 neurotoxic profiles is globally rendering promising results in different in vivo models of stroke. On the other hand, described oligodendrogenesis after brain ischemia seems to be strictly beneficial, although these cells are the less studied players in the stroke paradigm and negative effects could be described for oligodendrocytes in the next years. Here, we review recent advances in understanding the precise role of mentioned glial cell types in the main pathological events of ischemic stroke, including inflammation, blood brain barrier integrity, excitotoxicity, reactive oxygen species management, metabolic support, and neurogenesis, among others, with a special attention to tested therapeutic approaches.
Collapse
Affiliation(s)
- Ivó H Hernández
- Genomic Instability Group, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
- Center for Molecular Biology "Severo Ochoa" (CBMSO) UAM/CSIC, 28049 Madrid, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain
| | - Mario Villa-González
- Center for Molecular Biology "Severo Ochoa" (CBMSO) UAM/CSIC, 28049 Madrid, Spain
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Gerardo Martín
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Manuel Soto
- Center for Molecular Biology "Severo Ochoa" (CBMSO) UAM/CSIC, 28049 Madrid, Spain
- Departamento de Biología Molecular, Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - María José Pérez-Álvarez
- Center for Molecular Biology "Severo Ochoa" (CBMSO) UAM/CSIC, 28049 Madrid, Spain
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| |
Collapse
|
5
|
High mobility group box 1 promotes the differentiation of spinal ependymal cells into astrocytes rather than neurons. Neuroreport 2021; 32:399-406. [PMID: 33661806 DOI: 10.1097/wnr.0000000000001609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Spinal ependymal cells are involved in proliferation, differentiation and migration after spinal cord injury (SCI) and represent an endogenous source of repair cells for treating SCI. However, 95% of activated ependymal cells eventually differentiate into astrocytes after SCI and ultimately contribute more than half of the new astrocytes that form glial scars in vivo. The factors that regulate the fate of ependymal cells after SCI remain unclear. High mobility group box 1 (HMGB1) is regarded as an important proinflammatory factor in nerve injury, and recent studies have shown that HMGB1 can regulate the fate of stem cells after injury. In this study, we investigated whether HMGB1 released from reactive astrocytes after SCI regulates the proliferation and differentiation of ependymal cells in vitro. Ependymal cells extracted and cultured from the spinal cord of mice were separately treated with astrocyte culture medium (ACM), IL-1β, ACM (IL-1β) and the HMGB1 protein, and the proliferation and differentiation of ependymal cells were detected. Additionally, an HMGB1-neutralizing antibody (anti-HMGB1) was added to further verify the regulatory effect of HMGB1 on ependymal cells. The results showed that HMGB1 released from reactive astrocytes promoted ependymal cell differentiation into astrocytes and inhibited ependymal cell differentiation into neurons in vitro; however, the effect disappeared after the addition of anti-HMGB1. HMGB1 had no significant effect on ependymal cell proliferation. Our findings demonstrate that HMGB1 can regulate the differentiation of ependymal cells after SCI. These results provide a new strategy for the treatment of SCI.
Collapse
|
6
|
Atkinson SP. A Preview of Selected Articles. Stem Cells Transl Med 2019. [PMCID: PMC6877768 DOI: 10.1002/sctm.19-0384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
7
|
Cheng X, Yeung PKK, Zhong K, Zilundu PLM, Zhou L, Chung SK. Astrocytic endothelin-1 overexpression promotes neural progenitor cells proliferation and differentiation into astrocytes via the Jak2/Stat3 pathway after stroke. J Neuroinflammation 2019; 16:227. [PMID: 31733648 PMCID: PMC6858703 DOI: 10.1186/s12974-019-1597-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 09/23/2019] [Indexed: 12/17/2022] Open
Abstract
Background Endothelin-1 (ET-1) is synthesized and upregulated in astrocytes under stroke. We previously demonstrated that transgenic mice over-expressing astrocytic ET-1 (GET-1) displayed more severe neurological deficits characterized by a larger infarct after transient middle cerebral artery occlusion (tMCAO). ET-1 is a known vasoconstrictor, mitogenic, and a survival factor. However, it is unclear whether the observed severe brain damage in GET-1 mice post stroke is due to ET-1 dysregulation of neurogenesis by altering the stem cell niche. Methods Non-transgenic (Ntg) and GET-1 mice were subjected to tMCAO with 1 h occlusion followed by long-term reperfusion (from day 1 to day 28). Neurological function was assessed using a four-point scale method. Infarct area and volume were determined by 2,3,5-triphenyltetra-zolium chloride staining. Neural stem cell (NSC) proliferation and migration in subventricular zone (SVZ) were evaluated by immunofluorescence double labeling of bromodeoxyuridine (BrdU), Ki67 and Sox2, Nestin, and Doublecortin (DCX). NSC differentiation in SVZ was evaluated using the following immunofluorescence double immunostaining: BrdU and neuron-specific nuclear protein (NeuN), BrdU and glial fibrillary acidic protein (GFAP). Phospho-Stat3 (p-Stat3) expression detected by Western-blot and immunofluorescence staining. Results GET-1 mice displayed a more severe neurological deficit and larger infarct area after tMCAO injury. There was a significant increase of BrdU-labeled progenitor cell proliferation, which co-expressed with GFAP, at SVZ in the ipsilateral side of the GET-1 brain at 28 days after tMCAO. p-Stat3 expression was increased in both Ntg and GET-1 mice in the ischemia brain at 7 days after tMCAO. p-Stat3 expression was significantly upregulated in the ipsilateral side in the GET-1 brain than that in the Ntg brain at 7 days after tMCAO. Furthermore, GET-1 mice treated with AG490 (a JAK2/Stat3 inhibitor) sh owed a significant reduction in neurological deficit along with reduced infarct area and dwarfed astrocytic differentiation in the ipsilateral brain after tMCAO. Conclusions The data indicate that astrocytic endothelin-1 overexpression promotes progenitor stem cell proliferation and astr ocytic differentiation via the Jak2/Stat3 pathway.
Collapse
Affiliation(s)
- Xiao Cheng
- Department of Neurology, Guangdong Provincial Hospital of Traditional Chinese Medicine, 111 Dade Road, Guangzhou, 510120, China. .,School of Biomedical Sciences, The University of Hong Kong, HKSAR, China. .,The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 12 Jichang Road, Guangzhou, 510405, China. .,Guangdong Provincial Chinese Emergency Key Laboratory, Guangzhou, 510120, China. .,State Key Laboratory of Dampness Syndrome of Traditional Chinese Medicine, Guangzhou, 510120, China.
| | - Patrick K K Yeung
- School of Biomedical Sciences, The University of Hong Kong, HKSAR, China
| | - Ke Zhong
- Department of Anatomy, Zhong Shan School of Medicine, Sun Yat-Sen University, Guangdong Province, Guangzhou, China
| | - Prince L M Zilundu
- Department of Anatomy, Zhong Shan School of Medicine, Sun Yat-Sen University, Guangdong Province, Guangzhou, China
| | - Lihua Zhou
- Department of Anatomy, Zhong Shan School of Medicine, Sun Yat-Sen University, Guangdong Province, Guangzhou, China
| | - Sookja K Chung
- Faculty of Medicine, Macau University of Science and Technology, Macau, China. .,School of Biomedical Sciences, The University of Hong Kong, HKSAR, China.
| |
Collapse
|
8
|
Zhang X, Shen X, Dong J, Liu WC, Song M, Sun Y, Shu H, Towse CL, Liu W, Liu CF, Jin X. Inhibition of Reactive Astrocytes with Fluorocitrate Ameliorates Learning and Memory Impairment Through Upregulating CRTC1 and Synaptophysin in Ischemic Stroke Rats. Cell Mol Neurobiol 2019; 39:1151-1163. [PMID: 31270712 DOI: 10.1007/s10571-019-00709-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 06/19/2019] [Indexed: 12/12/2022]
Abstract
Ischemic stroke often causes motor and cognitive deficits. Deregulated glia gap junction communication, which is reflected by increased protein levels of glial fibrillary acidic protein (GFAP) and connexin 43 (Cx43), has been observed in ischemic hippocampus and has been associated with cognitive impairment in animal stroke models. Here, we tested the hypothesis that reactive astrocytes-mediated loss of synaptophysin (SYP) and CREB-regulated transcription coactivator 1 (CRTC1) contribute to dysfunction in glia gap junction communication and memory impairment after ischemic stroke. Male Sprague-Dawley rats were subjected to a 90-min middle cerebral artery occlusion (MCAO) with 7-day reperfusion. Fluorocitrate (1 nmol), the reversible inhibitor of the astrocytic tricarboxylic acid cycle, was injected into the right lateral ventricle of MCAO rats once every 2 days starting immediately before reperfusion. The Morris water maze was used to assess memory in conjunction with western blotting and immunostaining to detect protein expression and distribution in the hippocampus. Our results showed that ischemic stroke caused significant memory impairment accompanied by increased protein levels of GFAP and Cx43 in hippocampal tissue. In addition, the levels of several key memory-related important proteins including SYP, CRTC1, myelin basic protein and high-mobility group-box-1 were significantly reduced in the hippocampal tissue. Of note, inhibition of reactive astrocytes with fluorocitrate was shown to significantly reverse the above noted changes induced by ischemic stroke. Taken together, our findings demonstrate that inhibiting reactive astrocytes with fluorocitrate immediately before reperfusion may protect against ischemic stroke-induced memory impairment through the upregulation of CRTC1 and SYP.
Collapse
Affiliation(s)
- Xinyu Zhang
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, Jiangsu Key Laboratory of Neuropsychiatric Diseases, The Second Affiliated Hospital of Soochow University, Institute of Neuroscience, Soochow University, Suzhou, China
| | - Xianzhi Shen
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, Jiangsu Key Laboratory of Neuropsychiatric Diseases, The Second Affiliated Hospital of Soochow University, Institute of Neuroscience, Soochow University, Suzhou, China
| | - Jiali Dong
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, Jiangsu Key Laboratory of Neuropsychiatric Diseases, The Second Affiliated Hospital of Soochow University, Institute of Neuroscience, Soochow University, Suzhou, China
| | - Wen-Cao Liu
- Department of Emergency, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Min Song
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, Jiangsu Key Laboratory of Neuropsychiatric Diseases, The Second Affiliated Hospital of Soochow University, Institute of Neuroscience, Soochow University, Suzhou, China
| | - Yanyun Sun
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, Jiangsu Key Laboratory of Neuropsychiatric Diseases, The Second Affiliated Hospital of Soochow University, Institute of Neuroscience, Soochow University, Suzhou, China
| | - Hui Shu
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, Jiangsu Key Laboratory of Neuropsychiatric Diseases, The Second Affiliated Hospital of Soochow University, Institute of Neuroscience, Soochow University, Suzhou, China
| | - Clare-Louise Towse
- School of Chemistry and Biosciences, University of Bradford, Bradford, BD7 1DP, UK
| | - Wenlan Liu
- The Central Laboratory, Shenzhen Second People's Hospital, Shenzhen University 1st Affiliated Hospital, Shenzhen University School of Medicine, Shenzhen, 518035, China.
| | - Chun-Feng Liu
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, Jiangsu Key Laboratory of Neuropsychiatric Diseases, The Second Affiliated Hospital of Soochow University, Institute of Neuroscience, Soochow University, Suzhou, China.
| | - Xinchun Jin
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, Jiangsu Key Laboratory of Neuropsychiatric Diseases, The Second Affiliated Hospital of Soochow University, Institute of Neuroscience, Soochow University, Suzhou, China.
| |
Collapse
|
9
|
Dai Y, Sun F, Zhu H, Liu Q, Xu X, Gong P, Jiang R, Jin G, Qin J, Chen J, Zhang X, Shi W. Effects and Mechanism of Action of Neonatal Versus Adult Astrocytes on Neural Stem Cell Proliferation After Traumatic Brain Injury. Stem Cells 2019; 37:1344-1356. [PMID: 31287930 DOI: 10.1002/stem.3060] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 06/06/2019] [Accepted: 06/16/2019] [Indexed: 01/13/2023]
Abstract
Due to the limited capacity of brain tissue to self-regenerate after traumatic brain injury (TBI), the mobilization of endogenous neural stem cells (NSCs) is a popular research topic. In the clinic, the neurogenic abilities of adults versus neonates vary greatly, which is likely related to functional differences in NSCs. Recent studies have demonstrated that the molecules secreted from astrocytes play important roles in NSC fate determination. In this study, conditioned media (CM) derived from neonatal or adult rat astrocytes, which were unstimulated or stimulated by lipopolysaccharide (LPS), were prepared to treat NSCs. Our results revealed that neonatal rat astrocytes can significantly promote the proliferation of NSCs, compared with adult rat astrocytes, regardless of whether or not they were stimulated by LPS. Furthermore, we used mass spectrometry to detect the constituents of the CM from each group. We analyzed and screened for a protein, Tenascin-C (TNC), which was highly expressed in the neonatal group but poorly expressed in the adult group. We found that TNC can bind to the NSC surface epidermal growth factor receptor and promote proliferation through the PI3K-AKT pathway in vitro. Additionally, we confirmed in vivo that TNC can promote damage repair in a rat model of TBI, through enhancing the proliferation of endogenous NSCs. We believe that these findings provide a mechanistic understanding of why neonates show better neuroregenerative abilities than adults. This also provides a potential future therapeutic target, TNC, for injury repair after TBI. Stem Cells 2019;37:1344-1356.
Collapse
Affiliation(s)
- Yong Dai
- Jiangsu Clinical Medicine Centre of Tissue Engineering and Nerve Injury Repair, Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, People's Republic of China.,Department of Neurosurgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Feifan Sun
- Jiangsu Clinical Medicine Centre of Tissue Engineering and Nerve Injury Repair, Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, People's Republic of China
| | - Hui Zhu
- Jiangsu Clinical Medicine Centre of Tissue Engineering and Nerve Injury Repair, Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, People's Republic of China
| | - Qianqian Liu
- Jiangsu Clinical Medicine Centre of Tissue Engineering and Nerve Injury Repair, Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, People's Republic of China
| | - Xide Xu
- Jiangsu Clinical Medicine Centre of Tissue Engineering and Nerve Injury Repair, Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, People's Republic of China
| | - Peipei Gong
- Jiangsu Clinical Medicine Centre of Tissue Engineering and Nerve Injury Repair, Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, People's Republic of China
| | - Rui Jiang
- Jiangsu Clinical Medicine Centre of Tissue Engineering and Nerve Injury Repair, Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, People's Republic of China
| | - Guohua Jin
- Department of Anatomy and Neurobiology, School of Medicine, Nantong University, Nantong, People's Republic of China
| | - Jianbing Qin
- Department of Anatomy and Neurobiology, School of Medicine, Nantong University, Nantong, People's Republic of China
| | - Jian Chen
- Jiangsu Clinical Medicine Centre of Tissue Engineering and Nerve Injury Repair, Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, People's Republic of China
| | - Xinghua Zhang
- Department of Anatomy and Neurobiology, School of Medicine, Nantong University, Nantong, People's Republic of China
| | - Wei Shi
- Jiangsu Clinical Medicine Centre of Tissue Engineering and Nerve Injury Repair, Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, People's Republic of China
| |
Collapse
|
10
|
Liu M, Wu Y, Liu Y, Chen Z, He S, Zhang H, Wu L, Tu F, Zhao Y, Liu C, Chen X. Basic Fibroblast Growth Factor Protects Astrocytes Against Ischemia/Reperfusion Injury by Upregulating the Caveolin-1/VEGF Signaling Pathway. J Mol Neurosci 2018; 64:211-223. [PMID: 29299743 DOI: 10.1007/s12031-017-1023-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 12/20/2017] [Indexed: 10/18/2022]
Abstract
A previous in vivo study demonstrated that intracerebroventricular injection of basic fibroblast growth factor (bFGF) in middle cerebral artery occlusion rats increased the expression of caveolin-1 (cav-1) and vascular endothelial growth factor (VEGF) in cerebral ischemia penumbra. Because astrocytes are the largest population in the brain, the aim of this in vitro study was to investigate the influence of bFGF on cav-1 and VEGF expression in rat astrocytes following oxygen glucose deprivation/reoxygenation (OGD/R). For this, an ischemic model in vitro of oxygen glucose deprivation lasting for 6 h, followed by 24 h of reoxygenation was used. Primary astrocytes from newborn rats were pre-treated with siRNA targeting bFGF before OGD/R. Cell viability was measured by a CCK-8 assay. The protein and mRNA expressions of bFGF, cav-1, and VEGF were evaluated by western blotting, immunofluorescence staining, and reverse transcription-quantitative polymerase chain reaction. The results showed that OGD/R reduced cell viability, which was decreased further following bFGF knockdown; however, restoring bFGF improved cell survival. A cav-1 inhibitor abrogated the effect of bFGF on cell viability. The expression levels of bFGF mRNA, bFGF protein, cav-1 mRNA, cav-1 protein, and VEGF protein were higher in OGD/R astrocytes. bFGF knockdown markedly decreased the expression levels of cav-1 mRNA, cav-1 protein, and VEGF protein, which were effectively reversed by exogenous bFGF treatment. Moreover, exogenous bFGF treatment significantly increased the expression levels of cav-1 mRNA, cav-1 protein, and VEGF protein in OGD/R astrocytes; however, a cav-1 inhibitor abolished the effect of bFGF on VEGF protein expression. These results suggested that bFGF may protect astrocytes against ischemia/reperfusion injury by upregulating caveolin-1/VEGF signaling pathway.
Collapse
Affiliation(s)
- Meixia Liu
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, No. 109, Xueyuanxi Road, Wenzhou, Zhejiang, 325027, China
| | - Yudan Wu
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, No. 109, Xueyuanxi Road, Wenzhou, Zhejiang, 325027, China
| | - Yidian Liu
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, No. 109, Xueyuanxi Road, Wenzhou, Zhejiang, 325027, China
| | - Zhenzhen Chen
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, No. 109, Xueyuanxi Road, Wenzhou, Zhejiang, 325027, China
| | - Shujuan He
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, No. 109, Xueyuanxi Road, Wenzhou, Zhejiang, 325027, China
| | - Huimei Zhang
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, No. 109, Xueyuanxi Road, Wenzhou, Zhejiang, 325027, China
| | - Liang Wu
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, No. 109, Xueyuanxi Road, Wenzhou, Zhejiang, 325027, China
| | - Fengxia Tu
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, No. 109, Xueyuanxi Road, Wenzhou, Zhejiang, 325027, China
| | - Yun Zhao
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, No. 109, Xueyuanxi Road, Wenzhou, Zhejiang, 325027, China
| | - Chan Liu
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, No. 109, Xueyuanxi Road, Wenzhou, Zhejiang, 325027, China
| | - Xiang Chen
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, No. 109, Xueyuanxi Road, Wenzhou, Zhejiang, 325027, China.
| |
Collapse
|
11
|
Sun L, Li M, Ma X, Feng H, Song J, Lv C, He Y. Inhibition of HMGB1 reduces rat spinal cord astrocytic swelling and AQP4 expression after oxygen-glucose deprivation and reoxygenation via TLR4 and NF-κB signaling in an IL-6-dependent manner. J Neuroinflammation 2017; 14:231. [PMID: 29178911 PMCID: PMC5702193 DOI: 10.1186/s12974-017-1008-1] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 11/20/2017] [Indexed: 12/11/2022] Open
Abstract
Background Spinal cord astrocyte swelling is an important component to spinal cord edema and is associated with poor functional recovery as well as therapeutic resistance after spinal cord injury (SCI). High mobility group box-1 (HMGB1) is a mediator of inflammatory responses in the central nervous system and plays a critical role after SCI. Given this, we sought to identify both the role and underlying mechanisms of HMGB1 in cellular swelling and aquaporin 4 (AQP4) expression in cultured rat spinal cord astrocytes after oxygen-glucose deprivation/reoxygenation (OGD/R). Methods The post-natal day 1–2 Sprague-Dawley rat spinal cord astrocytes were cultured in vitro, and the OGD/R model was induced. We first investigated the effects of OGD/R on spinal cord astrocytic swelling and HMGB1 and AQP4 expression, as well as HMGB1 release. We then studied the effects of HMGB1 inhibition on cellular swelling, HMGB1 and AQP4 expression, and HMGB1 release. The roles of both toll-like receptor 4 (TLR4)/nuclear factor-kappa B (NF-κB) signaling pathway and interleukin-6 (IL-6) in reducing cellular swelling resulting from HMGB1 inhibition in spinal cord astrocytes after OGD/R were studied. Intergroup data were compared using one-way analysis of variance (ANOVA) followed by Dunnett’s test. Results The OGD/R increased spinal cord astrocytic swelling and HMGB1 and AQP4 expression, as well as HMGB1 release. Inhibition of HMGB1 using either HMGB1 shRNA or ethyl pyruvate resulted in reduced cellular volume, mitochondrial and endoplasmic reticulum swelling, and lysosome number and decreased upregulation of both HMGB1 and AQP4 in spinal cord astrocytes, as well as HMGB1 release. The HMGB1 effects on spinal cord astrocytic swelling and AQP4 upregulation after OGD/R were mediated—at least in part—via activation of TLR4, myeloid differentiation primary response gene 88 (MyD88), and NF-κB. These activation effects can be repressed by TLR4 inhibition using CLI-095 or C34, or by NF-κB inhibition using BAY 11-7082. Furthermore, either OGD/R or HMGB1 inhibition resulted in changes in IL-6 release. IL-6 was also shown to mediate AQP4 expression in spinal cord astrocytes. Conclusions HMGB1 upregulates AQP4 expression and promotes cell swelling in cultured spinal cord astrocytes after OGD/R, which is mediated through HMGB1/TLR4/MyD88/NF-κB signaling and in an IL-6-dependent manner.
Collapse
Affiliation(s)
- Lin Sun
- Department of Orthopedics, Shanxi Academy of Medical Sciences, Shanxi Da Yi Hospital, Shanxi Da Yi Hospital affiliated to Shanxi Medical University, Taiyuan, 030032, China.
| | - Man Li
- Department of Neurology, Second Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, 030001, China
| | - Xun Ma
- Department of Orthopedics, Shanxi Academy of Medical Sciences, Shanxi Da Yi Hospital, Shanxi Da Yi Hospital affiliated to Shanxi Medical University, Taiyuan, 030032, China
| | - Haoyu Feng
- Department of Orthopedics, Shanxi Academy of Medical Sciences, Shanxi Da Yi Hospital, Shanxi Da Yi Hospital affiliated to Shanxi Medical University, Taiyuan, 030032, China
| | - Junlai Song
- Department of Orthopedics, Shanxi Academy of Medical Sciences, Shanxi Da Yi Hospital, Shanxi Da Yi Hospital affiliated to Shanxi Medical University, Taiyuan, 030032, China
| | - Cong Lv
- Department of Orthopedics, Shanxi Academy of Medical Sciences, Shanxi Da Yi Hospital, Shanxi Da Yi Hospital affiliated to Shanxi Medical University, Taiyuan, 030032, China
| | - Yajun He
- Department of Orthopedics, Shanxi Academy of Medical Sciences, Shanxi Da Yi Hospital, Shanxi Da Yi Hospital affiliated to Shanxi Medical University, Taiyuan, 030032, China
| |
Collapse
|
12
|
Shu T, Liu C, Pang M, Wang J, Liu B, Zhou W, Wang X, Wu T, Wang Q, Rong L. Effects and mechanisms of matrix metalloproteinase2 on neural differentiation of induced pluripotent stem cells. Brain Res 2017; 1678:407-418. [PMID: 29137974 DOI: 10.1016/j.brainres.2017.11.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 11/03/2017] [Accepted: 11/08/2017] [Indexed: 01/22/2023]
Abstract
Induced pluripotent stem cells (iPSCs) possess the potential to differentiate into neural lineage cells. Matrix metalloproteinase 2 (MMP2), an endopeptidase in the extracellular matrix, has been shown to protect neural cells from injury. However, the mechanisms and effects of MMP2 on neural differentiation of iPSCs remain poorly understood. Here, we demonstrated a role for MMP2 in the differentiation of iPSCs to neurons via the AKT pathway. Treatment of iPSCs with MMP2 promoted their proliferation and differentiation into neural stem cells (NSCs), and then into neurons. The transcript and protein expression of Nestin and microtubule-associated protein 2 (MAP2) increased. Moreover, MMP2 markedly induced the expression of phospho-AKT (pAKT) during these differentiation stages. Consistently, silencing MMP2 using siRNA attenuated the expression of Nestin, MAP2 and pAKT, compared with the control group. In addition, the increasing levels of Nestin, MAP2 and pAKT in the MMP2 group were declined by pretreatment with the phosphoinositide 3-kinase (PI3K)/AKT inhibitor, LY294002. Furthermore, the study detected that TrkA and TrkB were perhaps the potential receptors for these effects of MMP2 on neural differentiation through PI3K/AKT signaling pathway. Taken together, these results suggest that MMP2 induces the differentiation of iPSCs into neurons by regulating the AKT signaling pathway.
Collapse
Affiliation(s)
- Tao Shu
- Department of Spine Surgery, The 3rd Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | - Chang Liu
- Department of Spine Surgery, The 3rd Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | - Mao Pang
- Department of Spine Surgery, The 3rd Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | - Juan Wang
- Department of Gynaecology, Common Splendor International Health Management, Guangzhou, Guangdong 510000, China
| | - Bin Liu
- Department of Spine Surgery, The 3rd Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | - Wei Zhou
- Department of Orthopedics, The 3rd Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| | - Xuan Wang
- Department of Spine Surgery, The 3rd Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | - Tao Wu
- Department of Emergency, Guangdong Provincial Corps Hospital of Chinese People's Armed Police Forces, Guangzhou Medical University, Guangzhou, Guangdong 510000, China
| | - Qiyou Wang
- Department of Spine Surgery, The 3rd Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, China.
| | - Limin Rong
- Department of Spine Surgery, The 3rd Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, China.
| |
Collapse
|
13
|
Becerra-Calixto A, Cardona-Gómez GP. The Role of Astrocytes in Neuroprotection after Brain Stroke: Potential in Cell Therapy. Front Mol Neurosci 2017; 10:88. [PMID: 28420961 PMCID: PMC5376556 DOI: 10.3389/fnmol.2017.00088] [Citation(s) in RCA: 159] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 03/14/2017] [Indexed: 12/11/2022] Open
Abstract
Astrocytes are commonly involved in negative responses through their hyperreactivity and glial scar formation in excitotoxic and/or mechanical injuries. But, astrocytes are also specialized glial cells of the nervous system that perform multiple homeostatic functions for the survival and maintenance of the neurovascular unit. Astrocytes have neuroprotective, angiogenic, immunomodulatory, neurogenic, and antioxidant properties and modulate synaptic function. This makes them excellent candidates as a source of neuroprotection and neurorestoration in tissues affected by ischemia/reperfusion, when some of their deregulated genes can be controlled. Therefore, this review analyzes pro-survival responses of astrocytes that would allow their use in cell therapy strategies.
Collapse
Affiliation(s)
| | - Gloria P. Cardona-Gómez
- Cellular and Molecular Neurobiology Area, Group of Neuroscience of Antioquia, School of Medicine, Sede de Investigación Universitaria (SIU), University of AntioquiaMedellín, Colombia
| |
Collapse
|
14
|
Yu D, Fan C, Zhang W, Wen Z, Hu L, Yang L, Feng Y, Yin KJ, Mo X. Neuroprotective effect of nicorandil through inhibition of apoptosis by the PI3K/Akt1 pathway in a mouse model of deep hypothermic low flow. J Neurol Sci 2015; 357:119-25. [PMID: 26279331 DOI: 10.1016/j.jns.2015.07.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 06/16/2015] [Accepted: 07/07/2015] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Nicorandil exerts a protective effect on ischemia-reperfusion (I/R) injury in the brain and kidney through anti-apoptotic mechanisms. However, the mechanism by which nicorandil protects against I/R injury induced by deep hypothermic low flow (DHLF) remains unclear. METHODS We used a cerebral I/R model induced by DHLF to determine the neuroprotective effects and possible mechanisms of nicorandil. RESULTS Hematoxylin-eosin (HE) staining and in situ terminal deoxynucleotidyl transferase UTP nick end labeling (TUNEL) assay were used to detect changes in cell morphology and the number of apoptotic cells in hippocampus, respectively. The apoptotic regulators including Bcl-2, Bax, Akt, and p-Akt (the active, phosphorylated form of Akt) were examined by Western blot (WB). Histopathological findings showed that nicorandil significantly alleviated morphological damage in hippocampal and reduced the number of TUNEL-positive nuclei induced by DHLF. Nicorandil also increased the expression of Bcl-2 and decreased the expression of Bax, while increasing p-Akt level. Consistent with these results, nicorandil-mediated neuroprotection was reduced in the Akt1+/- mutant mice and inhibited by LY294002, a PI3K inhibitor. CONCLUSIONS These findings showed that nicorandil provides a neuroprotective role in DHLF-induced I/R injury by inhibiting apoptosis via activation of the PI3K/Akt1 signaling pathway.
Collapse
Affiliation(s)
- Di Yu
- Department of Cardiothoracic Surgery, Nanjing Children's Hospital, Nanjing Medical University, Nanjing 210008, China
| | - Changfeng Fan
- Department of Cardiothoracic Surgery, Nanjing Children's Hospital, Nanjing Medical University, Nanjing 210008, China
| | - Weiyan Zhang
- Department of Cardiothoracic Surgery, Nanjing Children's Hospital, Nanjing Medical University, Nanjing 210008, China
| | - Zhongyuan Wen
- Department of Cardiothoracic Surgery, Nanjing Children's Hospital, Nanjing Medical University, Nanjing 210008, China
| | - Liang Hu
- Department of Cardiothoracic Surgery, Nanjing Children's Hospital, Nanjing Medical University, Nanjing 210008, China
| | - Lei Yang
- Department of Cardiothoracic Surgery, Nanjing Children's Hospital, Nanjing Medical University, Nanjing 210008, China
| | - Yu Feng
- Department of Cardiothoracic Surgery, Nanjing Children's Hospital, Nanjing Medical University, Nanjing 210008, China
| | - Ke-Jie Yin
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Xuming Mo
- Department of Cardiothoracic Surgery, Nanjing Children's Hospital, Nanjing Medical University, Nanjing 210008, China.
| |
Collapse
|
15
|
Bai J, Lyden PD. Revisiting Cerebral Postischemic Reperfusion Injury: New Insights in Understanding Reperfusion Failure, Hemorrhage, and Edema. Int J Stroke 2015; 10:143-52. [DOI: 10.1111/ijs.12434] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 11/14/2014] [Indexed: 01/11/2023]
Abstract
Cerebral postischemic reperfusion injury is defined as deterioration of ischemic brain tissue that parallels and antagonizes the benefits of restoring cerebral circulation after therapeutic thrombolysis for acute ischemic stroke. To understand the paradox of injury caused by treatment, we first emphasize the phenomenon in which recanalization of an occluded artery does not lead to tissue reperfusion. Additionally, no-reflow after recanalization may be due to injury of the neurovascular unit, distal microthrombosis, or both, and certainly worsens outcome. We examine the mechanism of molecular and sub-cellular damage in the neurovascular unit, notably oxidative stress, mitochondrial dysfunction, and apoptosis. At the level of the neurovascular unit, which mediates crosstalk between the damaged brain and systemic responses in blood, we summarize emerging evidence demonstrating that individual cell components play unique and cumulative roles that lead to damage of the blood–brain barrier and neurons. Furthermore, we review the latest developments in establishing a link between the immune system and microvascular dysfunction during ischemic reperfusion. Progress in assessing reperfusion injury has also been made, and we review imaging studies using various magnetic resonance imaging modalities. Lastly, we explore potential treatment approaches, including ischemic preconditioning, postconditioning, pharmacologic agents, and hypothermia.
Collapse
Affiliation(s)
- Jilin Bai
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Patrick D. Lyden
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|