1
|
Meng F, Cui J, Wang P, Wang J, Sun J, Li L. The Phenotype Changes of Astrocyte During Different Ischemia Conditions. Brain Sci 2024; 14:1256. [PMID: 39766455 PMCID: PMC11674399 DOI: 10.3390/brainsci14121256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/10/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
OBJECTIVES Dementia is becoming a major health problem in the world, and chronic brain ischemia is an established important risk factor in predisposing this disease. Astrocytes, as one major part of the blood-brain barrier (BBB), are activated during chronic cerebral blood flow hypoperfusion. Reactive astrocytes have been classified into phenotype pro-inflammatory type A1 or neuroprotective type A2. However, the specific subtype change of astrocyte and the mechanisms of chronic brain ischemia are still unknown. METHODS In order to depict the phenotype changes and their possible roles during this process, a rat bilateral common carotid artery occlusion model (BCAO) was employed in the present study. Meanwhile, the signaling pathways that possibly regulate these changes were investigated as well. RESULTS After four-week occlusion, astrocytes in the cortex of BCAO rats were shown to be the A2 phenotype, identified by the significant up-regulation of S100a10 accompanied by the down-regulation of Connexin 43 (CX43) protein. Next, we established in vitro hypoxia models, which were set up by stimulating primary astrocyte cultures from rat cortex with cobalt chloride, low glucose, or/and fibrinogen. Consistent with in vivo data, the cultured astrocytes also transformed into the A2 phenotype with the up-regulation of S100a10 and the down-regulation of CX43. In order to explore the mechanism of CX43 protein changes, C6 astrocyte cells were handled in both hypoxia and low-glucose stimulus, in which decreased pERK and pJNK expression were found. CONCLUSIONS In conclusion, our data suggest that in chronic cerebral ischemia conditions, the gradual ischemic insults could promote the transformation of astrocytes into A2 type instead of A1 type, and the phosphorylation of CX43 was negatively regulated by the phosphorylation of ERK and JNK. Also, our data could provide some new evidence of how to leverage the endogenous astrocytes phenotype changes during CNS injury by promoting them to be "protector" and not "culprit".
Collapse
Affiliation(s)
- Fei Meng
- Cardiac Valve Center, Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 101100, China;
| | - Jing Cui
- Department of Pathology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (J.C.); (J.S.)
| | - Peng Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Department of Cardiology, Qilu Hospital, Shandong University, Jinan 250012, China;
| | - Junhui Wang
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada;
| | - Jing Sun
- Department of Pathology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (J.C.); (J.S.)
| | - Liang Li
- Department of Pathology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (J.C.); (J.S.)
| |
Collapse
|
2
|
Donadoni M, Cakir S, Bellizzi A, Swingler M, Sariyer IK. Modeling HIV-1 infection and NeuroHIV in hiPSCs-derived cerebral organoid cultures. J Neurovirol 2024; 30:362-379. [PMID: 38600307 PMCID: PMC11464638 DOI: 10.1007/s13365-024-01204-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/13/2024] [Accepted: 03/22/2024] [Indexed: 04/12/2024]
Abstract
The human immunodeficiency virus (HIV) epidemic is an ongoing global health problem affecting 38 million people worldwide with nearly 1.6 million new infections every year. Despite the advent of combined antiretroviral therapy (cART), a large percentage of people with HIV (PWH) still develop neurological deficits, grouped into the term of HIV-associated neurocognitive disorders (HAND). Investigating the neuropathology of HIV is important for understanding mechanisms associated with cognitive impairment seen in PWH. The major obstacle for studying neuroHIV is the lack of suitable in vitro human culture models that could shed light into the HIV-CNS interactions. Recent advances in induced pluripotent stem cell (iPSC) culture and 3D brain organoid systems have allowed the generation of 2D and 3D culture methods that possess a potential to serve as a model of neurotropic viral diseases, including HIV. In this study, we first generated and characterized several hiPSC lines from healthy human donor skin fibroblast cells. hiPSCs were then used for the generation of microglia-containing human cerebral organoids (hCOs). Once fully characterized, hCOs were infected with HIV-1 in the presence and absence of cART regimens and viral infection was studied by cellular, molecular/biochemical, and virological assays. Our results revealed that hCOs were productively infected with HIV-1 as evident by viral p24-ELISA in culture media, RT-qPCR and RNAscope analysis of viral RNA, as well as ddPCR analysis of proviral HIV-1 in genomic DNA samples. More interestingly, replication and gene expression of HIV-1 were also greatly suppressed by cART in hCOs as early as 7 days post-infections. Our results suggest that hCOs derived from hiPSCs support HIV-1 replication and gene expression and may serve as a unique platform to better understand neuropathology of HIV infection in the brain.
Collapse
Affiliation(s)
- Martina Donadoni
- Department of Microbiology, Immunology and Inflammation, Center for Neurovirology and Gene Editing, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Senem Cakir
- Department of Microbiology, Immunology and Inflammation, Center for Neurovirology and Gene Editing, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Anna Bellizzi
- Department of Microbiology, Immunology and Inflammation, Center for Neurovirology and Gene Editing, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Michael Swingler
- Department of Microbiology, Immunology and Inflammation, Center for Neurovirology and Gene Editing, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Ilker K Sariyer
- Department of Microbiology, Immunology and Inflammation, Center for Neurovirology and Gene Editing, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Shen K, Shi Y, Wang X, Leung SW. Cellular Components of the Blood-Brain Barrier and Their Involvement in Aging-Associated Cognitive Impairment. Aging Dis 2024:AD.202.0424. [PMID: 39122454 DOI: 10.14336/ad.202.0424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 07/01/2024] [Indexed: 08/12/2024] Open
Abstract
Human life expectancy has been significantly extended, which poses major challenges to our healthcare and social systems. Aging-associated cognitive impairment is attributed to endothelial dysfunction in the cardiovascular system and neurological dysfunction in the central nervous system. The central nervous system is considered an immune-privileged tissue due to the exquisite protection provided by the blood-brain barrier. The present review provides an overview of the structure and function of blood-brain barrier, extending the cell components of blood-brain barrier from endothelial cells and pericytes to astrocytes, perivascular macrophages and oligodendrocyte progenitor cells. In particular, the pathological changes in the blood-brain barrier in aging, with special focus on the underlying mechanisms and molecular changes, are presented. Furthermore, the potential preventive/therapeutic strategies against aging-associated blood-brain barrier disruption are discussed.
Collapse
Affiliation(s)
- Kaiyuan Shen
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yi Shi
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xin Wang
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Susan Ws Leung
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
4
|
Hermanova Z, Valihrach L, Kriska J, Maheta M, Tureckova J, Kubista M, Anderova M. The deletion of AQP4 and TRPV4 affects astrocyte swelling/volume recovery in response to ischemia-mimicking pathologies. Front Cell Neurosci 2024; 18:1393751. [PMID: 38818517 PMCID: PMC11138210 DOI: 10.3389/fncel.2024.1393751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 05/01/2024] [Indexed: 06/01/2024] Open
Abstract
Introduction Astrocytic Transient receptor potential vanilloid 4 (TRPV4) channels, together with Aquaporin 4 (AQP4), are suspected to be the key players in cellular volume regulation, and therefore may affect the development and severity of cerebral edema during ischemia. In this study, we examined astrocytic swelling/volume recovery in mice with TRPV4 and/or AQP4 deletion in response to in vitro ischemic conditions, to determine how the deletion of these channels can affect the development of cerebral edema. Methods We used three models of ischemia-related pathological conditions: hypoosmotic stress, hyperkalemia, and oxygenglucose deprivation (OGD), and observed their effect on astrocyte volume changes in acute brain slices of Aqp4-/-, Trpv4-/- and double knockouts. In addition, we employed single-cell RT-qPCR to assess the effect of TRPV4 and AQP4 deletion on the expression of other ion channels and transporters involved in the homeostatic functioning of astrocytes. Results Quantification of astrocyte volume changes during OGD revealed that the deletion of AQP4 reduces astrocyte swelling, while simultaneous deletion of both AQP4 and TRPV4 leads to a disruption of astrocyte volume recovery during the subsequent washout. Of note, astrocyte exposure to hypoosmotic stress or hyperkalemia revealed no differences in astrocyte swelling in the absence of AQP4, TRPV4, or both channels. Moreover, under ischemia-mimicking conditions, we identified two distinct subpopulations of astrocytes with low and high volumetric responses (LRA and HRA), and their analyses revealed that mainly HRA are affected by the deletion of AQP4, TRPV4, or both channels. Furthermore, gene expression analysis revealed reduced expression of the ion transporters KCC1 and ClC2 as well as the receptors GABAB and NMDA in Trpv4-/- mice. The deletion of AQP4 instead caused reduced expression of the serine/cysteine peptidase inhibitor Serpina3n. Discussion Thus, we showed that in AQP4 or TRPV4 knockouts, not only the specific function of these channels is affected, but also the expression of other proteins, which may modulate the ischemic cascade and thus influence the final impact of ischemia.
Collapse
Affiliation(s)
- Zuzana Hermanova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine CAS, Prague, Czechia
- Second Faculty of Medicine, Charles University, Prague, Czechia
| | - Lukas Valihrach
- Department of Cellular Neurophysiology, Institute of Experimental Medicine CAS, Prague, Czechia
- Laboratory of Gene Expression, Institute of Biotechnology CAS, Vestec, Czechia
| | - Jan Kriska
- Department of Cellular Neurophysiology, Institute of Experimental Medicine CAS, Prague, Czechia
| | - Mansi Maheta
- Laboratory of Gene Expression, Institute of Biotechnology CAS, Vestec, Czechia
| | - Jana Tureckova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine CAS, Prague, Czechia
| | - Mikael Kubista
- Laboratory of Gene Expression, Institute of Biotechnology CAS, Vestec, Czechia
| | - Miroslava Anderova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine CAS, Prague, Czechia
| |
Collapse
|
5
|
Shiadeh SMJ, Goretta F, Svedin P, Jansson T, Mallard C, Ardalan M. Long-term impact of maternal obesity on the gliovascular unit and ephrin signaling in the hippocampus of adult offspring. J Neuroinflammation 2024; 21:39. [PMID: 38308309 PMCID: PMC10837922 DOI: 10.1186/s12974-024-03030-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/24/2024] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND Children born to obese mothers are at increased risk of developing mood disorders and cognitive impairment. Experimental studies have reported structural changes in the brain such as the gliovascular unit as well as activation of neuroinflammatory cells as a part of neuroinflammation processing in aged offspring of obese mothers. However, the molecular mechanisms linking maternal obesity to poor neurodevelopmental outcomes are not well established. The ephrin system plays a major role in a variety of cellular processes including cell-cell interaction, synaptic plasticity, and long-term potentiation. Therefore, in this study we determined the impact of maternal obesity in pregnancy on cortical, hippocampal development, vasculature and ephrin-A3/EphA4-signaling, in the adult offspring in mice. METHODS Maternal obesity was induced in mice by a high fat/high sugar Western type of diet (HF/HS). We collected brain tissue (prefrontal cortex and hippocampus) from 6-month-old offspring of obese and lean (control) dams. Hippocampal volume, cortical thickness, myelination of white matter, density of astrocytes and microglia in relation to their activity were analyzed using 3-D stereological quantification. mRNA expression of ephrin-A3, EphA4 and synaptic markers were measured by qPCR in the brain tissue. Moreover, expression of gap junction protein connexin-43, lipocalin-2, and vascular CD31/Aquaporin 4 were determined in the hippocampus by immunohistochemistry. RESULTS Volume of hippocampus and cortical thickness were significantly smaller, and myelination impaired, while mRNA levels of hippocampal EphA4 and post-synaptic density (PSD) 95 were significantly lower in the hippocampus in the offspring of obese dams as compared to offspring of controls. Further analysis of the hippocampal gliovascular unit indicated higher coverage of capillaries by astrocytic end-feet, expression of connexin-43 and lipocalin-2 in endothelial cells in the offspring of obese dams. In addition, offspring of obese dams demonstrated activation of microglia together with higher density of cells, while astrocyte cell density was lower. CONCLUSION Maternal obesity affects brain size, impairs myelination, disrupts the hippocampal gliovascular unit and decreases the mRNA expression of EphA4 and PSD-95 in the hippocampus of adult offspring. These results indicate that the vasculature-glia cross-talk may be an important mediator of altered synaptic plasticity, which could be a link between maternal obesity and neurodevelopmental/neuropsychiatric disorders in the offspring.
Collapse
Affiliation(s)
- Seyedeh Marziyeh Jabbari Shiadeh
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Medicine, Translational Neuropsychiatry Unit, Aarhus University, Aarhus, Denmark
| | - Fanny Goretta
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Pernilla Svedin
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Thomas Jansson
- Division of Reproductive Sciences, Department of OB/GYN, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Carina Mallard
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Maryam Ardalan
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
- Department of Clinical Medicine, Translational Neuropsychiatry Unit, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
6
|
Tian X, Yang W, Jiang W, Zhang Z, Liu J, Tu H. Multi-Omics Profiling Identifies Microglial Annexin A2 as a Key Mediator of NF-κB Pro-inflammatory Signaling in Ischemic Reperfusion Injury. Mol Cell Proteomics 2024; 23:100723. [PMID: 38253182 PMCID: PMC10879806 DOI: 10.1016/j.mcpro.2024.100723] [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: 06/01/2023] [Revised: 12/19/2023] [Accepted: 12/23/2023] [Indexed: 01/24/2024] Open
Abstract
Cerebral stroke is one of the leading causes of mortality and disability worldwide. Restoring the cerebral circulation following a period of occlusion and subsequent tissue oxygenation leads to reperfusion injury. Cerebral ischemic reperfusion (I/R) injury triggers immune and inflammatory responses, apoptosis, neuronal damage, and even death. However, the cellular function and molecular mechanisms underlying cerebral I/R-induced neuronal injury are incompletely understood. By integrating proteomic, phosphoproteomic, and transcriptomic profiling in mouse hippocampi after cerebral I/R, we revealed that the differentially expressed genes and proteins mainly fall into several immune inflammatory response-related pathways. We identified that Annexin 2 (Anxa2) was exclusively upregulated in microglial cells in response to cerebral I/R in vivo and oxygen-glucose deprivation and reoxygenation (OGD/R) in vitro. RNA-seq analysis revealed a critical role of Anxa2 in the expression of inflammation-related genes in microglia via the NF-κB signaling. Mechanistically, microglial Anxa2 is required for nuclear translocation of the p65 subunit of NF-κB and its transcriptional activity upon OGD/R in BV2 microglial cells. Anxa2 knockdown inhibited the OGD/R-induced microglia activation and markedly reduced the expression of pro-inflammatory factors, including TNF-α, IL-1β, and IL-6. Interestingly, conditional medium derived from Anxa2-depleted BV2 cell cultures with OGD/R treatment alleviated neuronal death in vitro. Altogether, our findings revealed that microglia Anxa2 plays a critical role in I/R injury by regulating NF-κB inflammatory responses in a non-cell-autonomous manner, which might be a potential target for the neuroprotection against cerebral I/R injury.
Collapse
Affiliation(s)
- Xibin Tian
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, China
| | - Wuyan Yang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, China
| | - Wei Jiang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, China
| | - Zhen Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, China
| | - Junqiang Liu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, China
| | - Haijun Tu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, China; Shenzhen Research Institute, Hunan University, Shenzhen, Guangdong, China.
| |
Collapse
|
7
|
Gupta S, Khan J, Ghosh S. Molecular mechanism of cognitive impairment associated with Parkinson's disease: A stroke perspective. Life Sci 2024; 337:122358. [PMID: 38128756 DOI: 10.1016/j.lfs.2023.122358] [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: 06/20/2023] [Revised: 12/03/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023]
Abstract
Parkinson's disease (PD) is a common neurological illness that causes several motor and non-motor symptoms, most characteristically limb tremors and bradykinesia. PD is a slowly worsening disease that arises due to progressive neurodegeneration of specific areas of the brain, especially the substantia nigra of the midbrain. Even though PD has continuously been linked to a higher mortality risk in numerous epidemiologic studies, there have been significant discoveries regarding the connection between PD and stroke. The incidence of strokes such as cerebral infarction and hemorrhage is substantially associated with the development of PD. Moreover, cognitive impairments, primarily dementia, have been associated with stroke and PD. However, the underlying molecular mechanism of this phenomenon is still obscure. This concise review focuses on the relationship between stroke and PD, emphasizing the molecular mechanism of cognition deficit and memory loss evident in PD and stroke. Furthermore, we are also highlighting some potential drug molecules that can target both PD and stroke.
Collapse
Affiliation(s)
- Sanju Gupta
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur (IIT-Jodhpur), Rajasthan 342037, India
| | - Juhee Khan
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur (IIT-Jodhpur), Rajasthan 342037, India
| | - Surajit Ghosh
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur (IIT-Jodhpur), Rajasthan 342037, India.
| |
Collapse
|
8
|
Kim S, Kubelka NK, LaPorte HM, Krishnamoorthy VR, Singh M. Estradiol and 3β-diol protect female cortical astrocytes by regulating connexin 43 Gap Junctions. Mol Cell Endocrinol 2023; 578:112045. [PMID: 37595662 PMCID: PMC10592012 DOI: 10.1016/j.mce.2023.112045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 08/03/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023]
Abstract
While estrogens have been described to protect or preserve neuronal function in the face of insults such as oxidative stress, the prevailing mechanistic model would suggest that these steroids exert direct effects on the neurons. However, there is growing evidence that glial cells, such as astrocytes, are key cellular mediators of protection. Noting that connexin 43 (Cx43), a protein highly expressed in astrocytes, plays a key role in mediating inter-cellular communication, we hypothesized that Cx43 is a target of estradiol (E2), and the estrogenic metabolite of DHT, 3β-diol. Additionally, we sought to determine if either or both of these hormones attenuate oxidative stress-induced cytotoxicity by eliciting a reduction in Cx43 expression or inhibition of Cx43 channel permeability. Using primary cortical astrocytes, we found that E2 and 3β-diol were each protective against the mixed metabolic/oxidative insult, iodoacetic acid (IAA). Moreover, these effects were blocked by estrogen receptor antagonists. However, E2 and 3β-diol did not alter Cx43 mRNA levels in astrocytes but did inhibit IAA-induced Cx43 gap junction opening/permeability. Taken together, these data implicate astrocyte Cx43 gap junction as an understudied mediator of the cytoprotective effects of estrogens in the brain. Given the wide breadth of disease states associated with Cx43 function/dysfunction, further understanding the relationship between gonadal steroids and Cx43 channels may contribute to a better understanding of the biological basis for sex differences in various diseases.
Collapse
Affiliation(s)
- Seongcheol Kim
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, United States
| | - Nicholas Knesek Kubelka
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, United States
| | - Heather M LaPorte
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, United States
| | - Vignesh R Krishnamoorthy
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, United States
| | - Meharvan Singh
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, United States.
| |
Collapse
|
9
|
Lei L, Wang YT, Hu D, Gai C, Zhang Y. Astroglial Connexin 43-Mediated Gap Junctions and Hemichannels: Potential Antidepressant Mechanisms and the Link to Neuroinflammation. Cell Mol Neurobiol 2023; 43:4023-4040. [PMID: 37875763 DOI: 10.1007/s10571-023-01426-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 10/14/2023] [Indexed: 10/26/2023]
Abstract
Major depression disorder (MDD) is a neuropsychiatric disorder associated with a high suicide rate and a higher disability rate than any other disease. Evidence suggests that the pathological mechanism of MDD is related to astrocyte dysfunction. Depression is mainly associated with the expression of connexin 43 (Cx43) and the function of Cx43-mediated gap junctions and hemichannels in astrocytes. Moreover, neuroinflammation has been a hotspot in research on the pathology of depression, and Cx43-mediated functions are thought to be involved in neuroinflammation-related depression. However, the specific mechanism of Cx43-mediated functions in neuroinflammation-related depression pathology remains unclear. Therefore, this review summarizes and discusses Cx43 expression, the role of gap junction intercellular communication, and its relationship with neuroinflammation in depression. This review also focuses on the effects of antidepressant drugs (e.g., monoamine antidepressants, psychotropic drugs, and N-methyl-D-aspartate receptor antagonists) on Cx43-mediated function and provides evidence for Cx43 as a novel target for the treatment of MDD. The pathogenesis of MDD is related to astrocyte dysfunction, with reduced Cx43 expression, GJ dysfunction, decreased GJIC and reduced BDNF expression in the depressed brain. The effect of Cx43 on neuroinflammation-related depression involving inflammatory cytokines, glutamate excitotoxicity, and HPA axis dysregulation. Antidepressant drugs targeting Cx43 can effectively relieve depressive symptoms.
Collapse
Affiliation(s)
- Lan Lei
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Sunshine Southern Avenue, Fang-Shan District, Beijing, 102488, China
| | - Ya-Ting Wang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Sunshine Southern Avenue, Fang-Shan District, Beijing, 102488, China
| | - Die Hu
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Sunshine Southern Avenue, Fang-Shan District, Beijing, 102488, China
| | - Cong Gai
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Sunshine Southern Avenue, Fang-Shan District, Beijing, 102488, China
| | - Yi Zhang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Sunshine Southern Avenue, Fang-Shan District, Beijing, 102488, China.
| |
Collapse
|
10
|
Sun N, Cui WQ, Min XM, Zhang GM, Liu JZ, Wu HY. A new perspective on hippocampal synaptic plasticity and post-stroke depression. Eur J Neurosci 2023; 58:2961-2984. [PMID: 37518943 DOI: 10.1111/ejn.16093] [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: 04/11/2023] [Revised: 07/01/2023] [Accepted: 07/03/2023] [Indexed: 08/01/2023]
Abstract
Post-stroke depression, a common complication after stroke, severely affects the recovery and quality of life of patients with stroke. Owing to its complex mechanisms, post-stroke depression treatment remains highly challenging. Hippocampal synaptic plasticity is one of the key factors leading to post-stroke depression; however, the precise molecular mechanisms remain unclear. Numerous studies have found that neurotrophic factors, protein kinases and neurotransmitters influence depressive behaviour by modulating hippocampal synaptic plasticity. This review further elaborates on the role of hippocampal synaptic plasticity in post-stroke depression by summarizing recent research and analysing possible molecular mechanisms. Evidence for the correlation between hippocampal mechanisms and post-stroke depression helps to better understand the pathological process of post-stroke depression and improve its treatment.
Collapse
Affiliation(s)
- Ning Sun
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wen-Qiang Cui
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiao-Man Min
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guang-Ming Zhang
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jia-Zheng Liu
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hong-Yun Wu
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
11
|
Chu H, Dong J, Tang Y, Huang C, Guo Q. Connexin 43 Promotes Neurogenesis via Regulating Aquaporin-4 after Cerebral Ischemia. Neurotox Res 2023; 41:349-361. [PMID: 37074591 DOI: 10.1007/s12640-023-00646-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/27/2023] [Accepted: 04/02/2023] [Indexed: 04/20/2023]
Abstract
We aimed to test the effects of connexin43 (Cx43) on ischemic neurogenesis and examined whether it was dependent on aquaporin-4 (AQP4). We detected the expression of Cx43 and AQP4 in the ipsilateral subventricular zone (SVZ) and peri-infarct cortex after middle cerebral artery occlusion (MCAO). Also, we examined neurogenesis in the above regions via co-labeling of 5-bromo-2-deoxyuridine (BrdU)/neuronal nuclear antigen (NeuN) and BrdU/doublecortin (DCX). The effects of Cx43 and AQP4 were investigated by using two transgenic animals: heterozygous Cx43 (Cx43±) mice and AQP4 knockout (AQP4-/-) mice, and connexin mimetic peptide (CMP), a selective Cx43 blocker. We demonstrated AQP4 and Cx43 were co-expressed in the astrocytes after MCAO and the expression was highly increased in ipsilateral SVZ and peri-infarct cortex. Cx43± mice had larger infarction volumes and worse neurological function. Both BrdU/NeuN and BrdU/DCX co-labeled cells in the two regions were reduced in Cx43± and AQP4-/- mice compared to wild-type (WT) mice, suggesting Cx43 and AQP4 participated in neurogenesis of neural stem cells. Moreover, CMP decreased AQP4 expression and inhibited neurogenesis in WT mice, while the latter failed to be observed in AQP4-/- mice. Besides, higher levels of IL-1β and TNF-α were detected in the SVZ and peri-infarct cortex of AQP4-/- and Cx43± mice than those in WT mice. In conclusion, our data suggest that Cx43 elicits neuroprotective effects after cerebral ischemia through promoting neurogenesis in the SVZ to regenerate the injured neurons, which is AQP4 dependent and associated with down-regulation of inflammatory cytokines IL-1β and TNF-α.
Collapse
Affiliation(s)
- Heling Chu
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, 200233, Shanghai, China
| | - Jing Dong
- Department of Internal Neurology, Qingdao Municipal Hospital, Qingdao, China
| | - Yuping Tang
- Department of Neurology, Huashan Hospital, Fudan University, No. 12 Mid. Wulumuqi Road, Shanghai, 200040, China.
| | - Chuyi Huang
- Health Management Center, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, No. 160 Pujian Road, Shanghai, 200120, China.
| | - Qihao Guo
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, 200233, Shanghai, China.
| |
Collapse
|
12
|
Michinaga S, Hishinuma S, Koyama Y. Roles of Astrocytic Endothelin ET B Receptor in Traumatic Brain Injury. Cells 2023; 12:cells12050719. [PMID: 36899860 PMCID: PMC10000579 DOI: 10.3390/cells12050719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 02/08/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
Traumatic brain injury (TBI) is an intracranial injury caused by accidents, falls, or sports. The production of endothelins (ETs) is increased in the injured brain. ET receptors are classified into distinct types, including ETA receptor (ETA-R) and ETB receptor (ETB-R). ETB-R is highly expressed in reactive astrocytes and upregulated by TBI. Activation of astrocytic ETB-R promotes conversion to reactive astrocytes and the production of astrocyte-derived bioactive factors, including vascular permeability regulators and cytokines, which cause blood-brain barrier (BBB) disruption, brain edema, and neuroinflammation in the acute phase of TBI. ETB-R antagonists alleviate BBB disruption and brain edema in animal models of TBI. The activation of astrocytic ETB receptors also enhances the production of various neurotrophic factors. These astrocyte-derived neurotrophic factors promote the repair of the damaged nervous system in the recovery phase of patients with TBI. Thus, astrocytic ETB-R is expected to be a promising drug target for TBI in both the acute and recovery phases. This article reviews recent observations on the role of astrocytic ETB receptors in TBI.
Collapse
Affiliation(s)
- Shotaro Michinaga
- Department of Pharmacodynamics, Meiji Pharmaceutical University, 2-522-1 Noshio, Tokyo 204-8588, Japan
| | - Shigeru Hishinuma
- Department of Pharmacodynamics, Meiji Pharmaceutical University, 2-522-1 Noshio, Tokyo 204-8588, Japan
| | - Yutaka Koyama
- Laboratory of Pharmacology, Kobe Pharmaceutical University, 4-19-1 Motoyama-Kita Higashinada, Kobe 668-8558, Japan
- Correspondence: ; Tel.: +81-78-441-7572
| |
Collapse
|
13
|
Jiang H, Zhang Y, Wang ZZ, Chen NH. Connexin 43: An Interface Connecting Neuroinflammation to Depression. Molecules 2023; 28:molecules28041820. [PMID: 36838809 PMCID: PMC9961786 DOI: 10.3390/molecules28041820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023] Open
Abstract
Major depressive disorder (MDD) is a leading chronic mental illness worldwide, characterized by anhedonia, pessimism and even suicidal thoughts. Connexin 43 (Cx43), mainly distributed in astrocytes of the brain, is by far the most widely and ubiquitously expressed connexin in almost all vital organs. Cx43 forms gap junction channels in the brain, which mediate energy exchange and effectively maintain physiological homeostasis. Increasing evidence suggests the crucial role of Cx43 in the pathogenesis of MDD. Neuroinflammation is one of the most common pathological features of the central nervous system dysfunctions. Inflammatory factors are abnormally elevated in patients with depression and are closely related to nearly all links of depression. After activating the inflammatory pathway in the brain, the release and uptake of glutamate and adenosine triphosphate, through Cx43 in the synaptic cleft, would be affected. In this review, we have summarized the association between Cx43 and neuroinflammation, the cornerstones linking inflammation and depression, and Cx43 abnormalities in depression. We also discuss the significant association of Cx43 in inflammation and depression, which will help to explore new antidepressant drug targets.
Collapse
Affiliation(s)
- Hong Jiang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical, Science and Peking Union Medical College, Beijing 100050, China
| | - Yi Zhang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Zhen-Zhen Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical, Science and Peking Union Medical College, Beijing 100050, China
- Correspondence: (Z.-Z.W.); (N.-H.C.); Tel.: +86-10-6316-5182 (Z.-Z.W.); +86-10-63165177 (N.-H.C.)
| | - Nai-Hong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical, Science and Peking Union Medical College, Beijing 100050, China
- Correspondence: (Z.-Z.W.); (N.-H.C.); Tel.: +86-10-6316-5182 (Z.-Z.W.); +86-10-63165177 (N.-H.C.)
| |
Collapse
|
14
|
Xue H, Wu M, Wang Y, Zhao Y, Zhang M, Zhang H. The circadian rhythms regulated by Cx43-signaling in the pathogenesis of Neuromyelitis Optica. Front Immunol 2023; 13:1021703. [PMID: 36726988 PMCID: PMC9885795 DOI: 10.3389/fimmu.2022.1021703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 12/29/2022] [Indexed: 01/18/2023] Open
Abstract
Introduction Neuromyelitis Optica (NMO) is an inflammatory demyelinating disease of the central nervous system (CNS). NMO manifests as selective and severe attacks on axons and myelin of the optic nerve and spinal cord, resulting in necrotic cavities. The circadian rhythms are well demonstrated to profoundly impact cellular function, behavior, and disease. This study is aimed to explore the role and molecular basis of circadian rhythms in NMO. Methods We used an Aquaporin 4(AQP4) IgG-induced NMO cell model in isolated astrocytes. The expression of Cx43 and Bmal1 were detected by real-time PCR and Western Blot. TAT-Gap19 and DQP-1105 were used to inhibit Cx43 and glutamate receptor respectively. The knockdown of Bmal1 were performed with the shRNA containing adenovirus. The levels of glutamate, anterior visual pathway (AVP), and vasoactive intestinal peptide (VIP) were quantified by ELISA kits. Results We found that Bmal1 and Clock, two essential components of the circadian clock, were significantly decreased in NMO astrocytes, which were reversed by Cx43 activation (linoleic acid) or glutamate. Moreover, the expression levels of Bmal1 and Clock were also decreased by Cx43 blockade (TAT-Gap19) or glutamate receptor inhibition (DQP-1105). Furthermore, adenovirus-mediated Bmal1 knockdown by shRNA (Ad-sh-Bmal1) dramatically decreased the levels of glutamate, AVP, and VIP from neurons, and significantly down-regulated the protein level of Cx43 in NMO astrocytes with Cx43 activation (linoleic acid) or glutamate treatment. However, Bmal1 knockdown did not alter these levels in normal astrocytes with Cx43 blockade (TAT-Gap19) or glutamate receptor inhibition (DQP-1105). Discussion Collectively, these results suggest that Cx43-glutamate signaling would be a critical upstream regulator that contributes to the NMO-induced rhythmic damage in SCN astrocytes.
Collapse
Affiliation(s)
- Huiru Xue
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China,First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Minghui Wu
- First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yongle Wang
- First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yunfei Zhao
- First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Meini Zhang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China,*Correspondence: Meini Zhang, ; Hui Zhang,
| | - Hui Zhang
- First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China,Department of Medical Imaging, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China,*Correspondence: Meini Zhang, ; Hui Zhang,
| |
Collapse
|
15
|
MiR-182 Inhibition Protects Against Experimental Stroke in vivo and Mitigates Astrocyte Injury and Inflammation in vitro via Modulation of Cortactin Activity. Neurochem Res 2022; 47:3682-3696. [PMID: 35951202 PMCID: PMC10069410 DOI: 10.1007/s11064-022-03718-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 07/08/2022] [Accepted: 07/27/2022] [Indexed: 10/15/2022]
Abstract
Ischemic stroke remains a devastating cerebrovascular disease that accounts for a high proportion of mortality and disability worldwide. MicroRNAs (miRNAs) are a class of small non-coding RNAs that are responsible for regulation of post-transcriptional gene expression, and growing evidence supports a role for miRNAs in stroke injury and recovery. The current study examined the role of miR-182 in experimental stroke using both in vitro and in vivo models of ischemic injury. Brain levels of miR-182 significantly increased after transient middle cerebral artery occlusion (MCAO) in mice and in primary astrocyte cultures subjected to combined oxygen-glucose deprivation/reperfusion (OGD/R) injury. In vivo, stroke volume and neurological score were significantly improved by pre-treatment with miR-182 antagomir. Astrocyte cultures stressed with OGD/R resulted in mitochondrial fragmentation and downregulation of cortactin, an actin-binding protein. Inhibition of miR-182 significantly preserved cortactin expression, reduced mitochondrial fragmentation and improved astrocyte survival after OGD/R. In parallel, lipopolysaccharide (LPS)-induced nitric-oxide release in astrocyte cultures was significantly reduced by miR-182 inhibition, translating to reduced injury in primary neuronal cultures subjected to conditioned medium from LPS-treated astrocytes. These findings identify miR-182 and/or cortactin as potential clinical targets to preserve mitochondrial structure and mitigate neuroinflammation and cell death after ischemic stroke.
Collapse
|
16
|
EphrinA4 mimetic peptide impairs fear conditioning memory reconsolidation in lateral amygdala. Sci Rep 2022; 12:17731. [PMID: 36273074 PMCID: PMC9588004 DOI: 10.1038/s41598-022-21519-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 09/28/2022] [Indexed: 01/18/2023] Open
Abstract
Fear memory may undergo a process after memory reactivation called reconsolidation. To examine the roles of ephrinA4 in fear memory reconsolidation an inhibitory ephrinA4 mimetic peptide (pep-ephrinA4), that targets the EphA binding site and inhibits EphA activation, was used. Pep-ephrinA4 was microinjected into the lateral amygdala (LA) of fear-conditioned rats 24 h after training and 30 min before tone CS memory retrieval. Memory retrieval was unaffected by pep-ephrinA4. However, the animals were impaired in fear memory tested 1 h or 24 h afterward when compared to controls. Fear-conditioned animals injected with pep-ephrinA4 into LA immediately after long-term memory retrieval were unaffected when tested 24 h afterward. Microinjection into LA of a peptide originated from an ephrinA4 site that does not interact with EphA did not affect fear memory reconsolidation. Rats that were administrated with pep-ephrinA4 systemically 24 h after fear conditioning and 30 min before CS memory retrieval were impaired in long-term fear conditioning memory tested 24 h afterward when compared to the control peptide. These results show that ephrinA4 binding sites are needed for long-term fear memory reconsolidation in LA and may serve as a target for the treatment of fear-related disorders by blocking reconsolidation.
Collapse
|
17
|
Liu Y, Chu S, Hu Y, Yang S, Li X, Zheng Q, Ai Q, Ren S, Wang H, Gong L, Xu X, Chen NH. Exogenous Adenosine Antagonizes Excitatory Amino Acid Toxicity in Primary Astrocytes. Cell Mol Neurobiol 2021; 41:687-704. [PMID: 32632892 DOI: 10.1007/s10571-020-00876-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 05/12/2020] [Indexed: 12/29/2022]
Abstract
Excitatory toxicity is still a hot topic in the study of ischemic stroke, and related research has focused mainly on neurons. Adenosine is an important neuromodulator that is known as a "biosignature" in the central nervous system (CNS). The protective effect of exogenous adenosine on neurons has been confirmed, but its mechanism remains elusive. In this study, astrocytes were pretreated with adenosine, and the effects of an A2a receptor (A2aR) inhibitor (SCH58261) and A2b receptor (A2bR) inhibitor (PSB1115) on excitatory glutamate were investigated. An oxygen glucose deprivation/reoxygenation (OGD/R) and glutamate model was generated in vitro. Post-model assessment included expression levels of glutamate transporters (glt-1), gap junction protein (Cx43) and glutamate receptor (AMPAR), Na+-K+-ATPase activity, and diffusion distance of dyes. Glutamate and glutamine contents were determined at different time points. The results showed that (1) adenosine could improve the function of Na+-K+-ATPase, upregulate the expression of glt-1, and enhance the synthesis of glutamine in astrocytes. This effect was associated with A2aR activation but not with A2bR activation. (2) Adenosine could inhibit the expression of gap junction protein (Cx43) and reduce glutamate diffusion. Inhibition of A2aR attenuated adenosine inhibition of gap junction intercellular communication (GJIC) in the OGD/R model, while it enhanced adenosine inhibition of GJIC in the glutamate model, depending on the glutamate concentration. (3) Adenosine could cause AMPAR gradually entered the nucleus from the cytoplasm, thereby reducing the expression of AMPAR on the cell membrane. Taken together, the results indicate that adenosine plays a role of anti-excitatory toxicity effect in protection against neuronal death and the functional recovery of ischemic stroke mainly by targeting astrocytes, which are closely related to A2aR. The present study provided a scientific basis for adenosine prevention and ischemic stroke treatment, thereby providing a new approach for alleviating ischemic stroke.
Collapse
Affiliation(s)
- Yingjiao Liu
- College of Pharmacy, Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Shifeng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Yaomei Hu
- College of Pharmacy, Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, China
| | - Songwei Yang
- College of Pharmacy, Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, China
| | - Xun Li
- College of Pharmacy, Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Qinglian Zheng
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510631, China
| | - Qidi Ai
- College of Pharmacy, Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, China
| | - Siyu Ren
- College of Pharmacy, Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, China
| | - Huiqin Wang
- College of Pharmacy, Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, China
| | - Limin Gong
- College of Pharmacy, Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, China
| | - Xin Xu
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510631, China
| | - Nai-Hong Chen
- College of Pharmacy, Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, China.
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510631, China.
| |
Collapse
|
18
|
Koyama Y, Tsuboi S, Mizogui F. Endothelin-1 decreases the expression of Ephrin-A and B subtypes in cultured rat astrocytes through ET B receptors. Neurosci Lett 2021; 741:135393. [PMID: 33279571 DOI: 10.1016/j.neulet.2020.135393] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 09/08/2020] [Accepted: 09/14/2020] [Indexed: 11/24/2022]
Abstract
Ephrin family proteins are cell surface molecules that regulate several cellular functions through cell-cell interactions. During nervous tissue repair after injury, the expression of ephrin subtypes in astrocytes is altered, affecting the axonal elongation and migration of neuronal precursors. However, the mechanism regulating the expression of ephrin subtypes in astrocytes has not been investigated. Herein, we studied the effects of endothelin-1 (ET-1) on the expression of ephrin subtypes in cultured rat astrocytes. Our results showed that ET-1 (100 nM) treatment for 1-24 h reduced the expression of ephrin-A2, -A4, -B2, and -B3 mRNA and protein in astrocytes, whereas the expression of ephrin-A1, -A3, -A5, and -B1 mRNA were not affected. Sarafotoxin S6c, a selective ETB receptor agonist, decreased the expression of ephrin-A2, -A4, -B2, and -B3 in cultured astrocytes. The decrease in ephrin-A2, -A4, -B2, and -B3 expression by ET-1 treatment was reduced in the presence of BQ788, an ETB receptor antagonist, while FR139317, an ETA receptor antagonist, had no effects. These results suggest that ET-1 is a signaling molecule that downregulates ephrin-A2, -A4, -B2, and -B3 expression in astrocytes.
Collapse
Affiliation(s)
- Yutaka Koyama
- Laboratory of Pharmacology, Kobe Pharmaceutical University, 4-19-1 Motoyama-Kita Higashinada, Kobe 668-8558 Japan.
| | - Sayaka Tsuboi
- Laboratory of Pharmacology, Kobe Pharmaceutical University, 4-19-1 Motoyama-Kita Higashinada, Kobe 668-8558 Japan
| | - Fuka Mizogui
- Laboratory of Pharmacology, Kobe Pharmaceutical University, 4-19-1 Motoyama-Kita Higashinada, Kobe 668-8558 Japan
| |
Collapse
|
19
|
Wang J, Bai T, Wang N, Li H, Guo X. Neuroprotective potential of imatinib in global ischemia-reperfusion-induced cerebral injury: possible role of Janus-activated kinase 2/signal transducer and activator of transcription 3 and connexin 43. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2020; 24:11-18. [PMID: 31908570 PMCID: PMC6940502 DOI: 10.4196/kjpp.2020.24.1.11] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 06/18/2019] [Accepted: 06/20/2019] [Indexed: 12/20/2022]
Abstract
The present study was aimed to explore the neuroprotective role of imatinib in global ischemia-reperfusion-induced cerebral injury along with possible mechanisms. Global ischemia was induced in mice by bilateral carotid artery occlusion for 20 min, which was followed by reperfusion for 24 h by restoring the blood flow to the brain. The extent of cerebral injury was assessed after 24 h of global ischemia by measuring the locomotor activity (actophotometer test), motor coordination (inclined beam walking test), neurological severity score, learning and memory (object recognition test) and cerebral infarction (triphenyl tetrazolium chloride stain). Ischemia-reperfusion injury produced significant cerebral infarction, impaired the behavioral parameters and decreased the expression of connexin 43 and phosphorylated signal transducer and activator of transcription 3 (p-STAT3) in the brain. A single dose administration of imatinib (20 and 40 mg/kg) attenuated ischemia-reperfusion-induced behavioral deficits and the extent of cerebral infarction along with the restoration of connexin 43 and p-STAT3 levels. However, administration of AG490, a selective Janus-activated kinase 2 (JAK2)/STAT3 inhibitor, abolished the neuroprotective actions of imatinib and decreased the expression of connexin 43 and p-STAT3. It is concluded that imatinib has the potential of attenuating global ischemia-reperfusion-induced cerebral injury, which may be possibly attributed to activation of JAK2/STAT3 signaling pathway along with the increase in the expression of connexin 43.
Collapse
Affiliation(s)
- Jieying Wang
- Department of Pediatrics, Shaanxi Provincial People's Hospital, The Affiliated Hospital of Xi'an Medical University, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710068, Shaanxi, China
| | - Taomin Bai
- Department of Pediatrics, Shaanxi Provincial People's Hospital, The Affiliated Hospital of Xi'an Medical University, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710068, Shaanxi, China
| | - Nana Wang
- Central Laboratory, Shaanxi Provincial People's Hospital, The Affiliated Hospital of Xi'an Medical University, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710068, Shaanxi, China
| | - Hongyan Li
- Department of Pediatrics, Shaanxi Provincial People's Hospital, The Affiliated Hospital of Xi'an Medical University, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710068, Shaanxi, China
| | - Xiangyang Guo
- Department of Pediatrics, Shaanxi Provincial People's Hospital, The Affiliated Hospital of Xi'an Medical University, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710068, Shaanxi, China.,Department of Neurology, Xijing Hospital, Air Force Medical University, Xi'an 710032, Shaanxi, China
| |
Collapse
|
20
|
Yang K, Zhou Y, Zhou L, Yan F, Guan L, Liu H, Liu W. Synaptic Plasticity After Focal Cerebral Ischemia Was Attenuated by Gap26 but Enhanced by GAP-134. Front Neurol 2020; 11:888. [PMID: 32982919 PMCID: PMC7479336 DOI: 10.3389/fneur.2020.00888] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 07/13/2020] [Indexed: 12/15/2022] Open
Abstract
Objective: Synaptic plasticity is critical for neurorehabilitation after focal cerebral ischemia. Connexin 43 (Cx43), the main component of the gap junction, has been shown to be pivotal for synaptic plasticity. The objective of this study was to investigate the role of the Cx43 inhibitor (Gap26) and gap junction modifier (GAP-134) in neurorehabilitation and to study their contribution to synaptic plasticity after focal ischemia. Methods: Time course expression of both total and phosphorylated Cx43 (p-Cx43) were detected by western blotting at 3, 7, and 14 d after focal ischemia. Gap26 and GAP-134 were administered starting from 3 d post focal ischemia. Neurological performances were evaluated by balance beam walking test and Y-maze test at 1, 3, and 7 d. Golgi staining and transmission electron microscope (TEM) detection were conducted at 7 d for observing dendritic spine numbers and synaptic ultrastructure, respectively. Immunofluorescent staining was used at 7 d for detection of synaptic plasticity markers, including synaptophysin (SYN) and growth-associated protein-43 (GAP-43). Results: Expression levels of both total Cx43 and p-Cx43 were increased after focal cerebral ischemia, peaking at 7 d. Compared with the MCAO group, Gap26 worsened the neurological behavior and decreased the dendritic spine number while GAP-134 improved the neurobehavior and increased the number of dendritic spines. Moreover, Gap26 further destroyed the synaptic structure, concomitant with downregulated SYN and GAP-43, whereas GAP-134 alleviated synaptic destruction and upregulated SYN and GAP-43. Conclusion: These findings suggested that Cx43 or the gap junction was involved in synaptic plasticity, thereby promoting neural recovery after ischemic stroke. Treatments enhancing gap junctions may be potential promising therapeutic measures for neurorehabilitation after ischemic stroke.
Collapse
Affiliation(s)
- Kailing Yang
- Department of Physiology, School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China.,The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ying Zhou
- Department of Physiology, School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China.,The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lequan Zhou
- Department of Physiology, School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China.,The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fuman Yan
- Department of Physiology, School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China.,The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Li Guan
- Department of Physiology, School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China.,The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haimei Liu
- Department of Physiology, School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China.,The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Liu
- Department of Physiology, School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China.,The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
21
|
Zhao Y, Yang J, Li C, Zhou G, Wan H, Ding Z, Wan H, Zhou H. Role of the neurovascular unit in the process of cerebral ischemic injury. Pharmacol Res 2020; 160:105103. [PMID: 32739425 DOI: 10.1016/j.phrs.2020.105103] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 12/17/2022]
Abstract
Cerebral ischemic injury exhibits both high morbidity and mortality worldwide. Traditional research of the pathogenesis of cerebral ischemic injury has focused on separate analyses of the involved cell types. In recent years, the neurovascular unit (NVU) mechanism of cerebral ischemic injury has been proposed in modern medicine. Hence, more effective strategies for the treatment of cerebral ischemic injury may be provided through comprehensive analysis of brain cells and the extracellular matrix. However, recent studies that have investigated the function of the NVU in cerebral ischemic injury have been insufficient. In addition, the metabolism and energy conversion of the NVU depend on interactions among multiple cell types, which make it difficult to identify the unique contribution of each cell type. Therefore, in the present review, we comprehensively summarize the regulatory effects and recovery mechanisms of four major cell types (i.e., astrocytes, microglia, brain-microvascular endothelial cells, and neurons) in the NVU under cerebral ischemic injury, as well as discuss the interactions among these cell types in the NVU. Furthermore, we discuss the common signaling pathways and signaling factors that mediate cerebral ischemic injury in the NVU, which may help to provide a theoretical basis for the comprehensive elucidation of cerebral ischemic injury.
Collapse
Affiliation(s)
- Yu Zhao
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Jiehong Yang
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Chang Li
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Guoying Zhou
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Haofang Wan
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Zhishan Ding
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China
| | - Haitong Wan
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China.
| | - Huifen Zhou
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, PR China.
| |
Collapse
|
22
|
Liang Z, Wang X, Hao Y, Qiu L, Lou Y, Zhang Y, Ma D, Feng J. The Multifaceted Role of Astrocyte Connexin 43 in Ischemic Stroke Through Forming Hemichannels and Gap Junctions. Front Neurol 2020; 11:703. [PMID: 32849190 PMCID: PMC7411525 DOI: 10.3389/fneur.2020.00703] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 06/09/2020] [Indexed: 12/13/2022] Open
Abstract
Ischemic stroke is a multi-factorial cerebrovascular disease with high worldwide morbidity and mortality. In the past few years, multiple studies have revealed the underlying mechanism of ischemia/reperfusion injury, including calcium overload, amino acid toxicity, oxidative stress, and inflammation. Connexin 43 (Cx43), the predominant connexin protein in astrocytes, has been recently proven to display non-substitutable roles in the pathology of ischemic stroke development and progression through forming gap junctions and hemichannels. Under normal conditions, astrocytic Cx43 could be found in hemichannels or in the coupling with other hemichannels on astrocytes, neurons, or oligodendrocytes to form the neuro-glial syncytium, which is involved in metabolites exchange between communicated cells, thus maintaining the homeostasis of the CNS environment. In ischemic stroke, the phosphorylation of Cx43 might cause the degradation of gap junctions and the opening of hemichannels, contributing to the release of inflammatory mediators. However, the remaining gap junctions could facilitate the exchange of protective and harmful metabolites between healthy and injured cells, protecting the injured cells to some extent or damaging the healthy cells depending on the balance of the exchange of protective and harmful metabolites. In this study, we review the changes in astrocytic Cx43 expression and distribution as well as the influence of these changes on the function of astrocytes and other cells in the CNS, providing new insight into the pathology of ischemic stroke injury; we also discuss the potential of astrocytic Cx43 as a target for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Zhen Liang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Xu Wang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Yulei Hao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Lin Qiu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Yingyue Lou
- Department of Plastic and Reconstructive Surgery, The First Hospital of Jilin University, Changchun, China
| | - Yaoting Zhang
- Department of Cardiology, The First Hospital of Jilin University, Changchun, China
| | - Di Ma
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jiachun Feng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
23
|
Elgebaly MM. Ephrin-Eph Signaling as a Novel Neuroprotection Path in Ischemic Stroke. J Mol Neurosci 2020; 70:2001-2006. [PMID: 32488844 DOI: 10.1007/s12031-020-01603-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 05/19/2020] [Indexed: 12/20/2022]
Abstract
The search for novel neuroprotection strategies in ischemic stroke continues, as revascularization using tissue-plasminogen activator is the only pharmacological method currently available to patients. The purpose of this review article is to summarize research findings regarding the erythropoietin-producing hepatocellular receptor pathway as an emerging novel molecular target for neuroprotection in ischemic stroke. Ephrin-Eph interactions represent a new strategy in neuroprotection. Potential therapeutic targets include the different cellular locations within the neurovascular unit (e.g. astrocytes and neurons) and the different ephrin receptor subtypes. In particular, ephrin-B2/EphB4 receptor stimulation seems to exert neuroprotective effects, while stimulation of other ligands/receptors results in deleterious effects, during the post-ischemic stroke recovery phase. Neuroprotection, assessed by either a decrease in neurovascular unit injury markers or improvement in motor function tests, can be achieved by modulating the activity of different ephrin-Eph receptor subtypes. These novel molecular targets provide multiple potential neuroprotective therapeutic benefits, with meaningful clinical outcomes.
Collapse
Affiliation(s)
- Mostafa M Elgebaly
- College of Pharmacy, Pharmaceutical Sciences Department, Larkin University, 18301 N Miami ave, Miami, FL, 33169, USA.
| |
Collapse
|
24
|
Wang X, Yang XL, Kong WL, Zeng ML, Shao L, Jiang GT, Cheng JJ, Kong S, He XH, Liu WH, Chen TX, Peng BW. TRPV1 translocated to astrocytic membrane to promote migration and inflammatory infiltration thus promotes epilepsy after hypoxic ischemia in immature brain. J Neuroinflammation 2019; 16:214. [PMID: 31722723 PMCID: PMC6852893 DOI: 10.1186/s12974-019-1618-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 10/16/2019] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Neonatal hypoxic-ischemic brain damage (HIBD), a leading cause of neonatal mortality, has intractable sequela such as epilepsy that seriously affected the life quality of HIBD survivors. We have previously shown that ion channel dysfunction in the central nervous system played an important role in the process of HIBD-induced epilepsy. Therefore, we continued to validate the underlying mechanisms of TRPV1 as a potential target for epilepsy. METHODS Neonatal hypoxic ischemia and oxygen-glucose deprivation (OGD) were used to simulate HIBD in vivo and in vitro. Primarily cultured astrocytes were used to assess the expression of TRPV1, glial fibrillary acidic protein (GFAP), cytoskeletal rearrangement, and inflammatory cytokines by using Western blot, q-PCR, and immunofluorescence. Furthermore, brain electrical activity in freely moving mice was recorded by electroencephalography (EEG). TRPV1 current and neuronal excitability were detected by whole-cell patch clamp. RESULTS Astrocytic TRPV1 translocated to the membrane after OGD. Mechanistically, astrocytic TRPV1 activation increased the inflow of Ca2+, which promoted G-actin polymerized to F-actin, thus promoted astrocyte migration after OGD. Moreover, astrocytic TRPV1 deficiency decreased the production and release of pro-inflammatory cytokines (TNF, IL-6, IL-1β, and iNOS) after OGD. It could also dramatically attenuate neuronal excitability after OGD and brain electrical activity in HIBD mice. Behavioral testing for seizures after HIBD revealed that TRPV1 knockout mice demonstrated prolonged onset latency, shortened duration, and decreased seizure severity when compared with wild-type mice. CONCLUSIONS Collectively, TRPV1 promoted astrocyte migration thus helped the infiltration of pro-inflammatory cytokines (TNF, IL-1β, IL-6, and iNOS) from astrocytes into the vicinity of neurons to promote epilepsy. Our study provides a strong rationale for astrocytic TRPV1 to be a therapeutic target for anti-epileptogenesis after HIBD.
Collapse
Affiliation(s)
- Xin Wang
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xing-Liang Yang
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Wei-Lin Kong
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Meng-Liu Zeng
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Lin Shao
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Guang-Tong Jiang
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Jing-Jing Cheng
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Shuo Kong
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xiao-Hua He
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Wan-Hong Liu
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Tao-Xiang Chen
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Bi-Wen Peng
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
25
|
Reeves C, Pradim-Jardim A, Sisodiya SM, Thom M, Liu JYW. Spatiotemporal dynamics of PDGFRβ expression in pericytes and glial scar formation in penetrating brain injuries in adults. Neuropathol Appl Neurobiol 2019; 45:609-627. [PMID: 30636077 PMCID: PMC6767497 DOI: 10.1111/nan.12539] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 10/17/2018] [Indexed: 12/30/2022]
Abstract
Aims Understanding the spatiotemporal dynamics of reactive cell types following brain injury is important for future therapeutic interventions. We have previously used penetrating cortical injuries following intracranial recordings as a brain repair model to study scar‐forming nestin‐expressing cells. We now explore the relationship between nestin‐expressing cells, PDGFRβ+ pericytes and Olig2+ glia, including their proliferation and functional maturation. Methods In 32 cases, ranging from 3 to 461 days post injury (dpi), immunohistochemistry for PDGFRβ, nestin, GFAP, Olig2, MCM2, Aquaporin 4 (Aq4), Glutamine Synthetase (GS) and Connexin 43 (Cx43) was quantified for cell densities, labelling index (LI) and cellular co‐expression at the injury site compared to control regions. Results PDGFRβ labelling highlighted both pericytes and multipolar parenchymal cells. PDGFRβ LI and PDGFRβ+/MCM2+ cells significantly increased in injury Zones at 10–13 dpi with migration of pericytes away from vessels with increased co‐localization of PDGRFβ with nestin compared to control regions (P < 0.005). Olig2+/MCM2+ cell populations peaked at 13 dpi with significantly higher cell densities at injury sites than in control regions (P < 0.01) and decreasing with dpi (P < 0.05). Cx43 LI was reduced in acute injuries but increased with dpi (P < 0.05) showing significant cellular co‐localization with nestin and GFAP (P < 0.005 and P < 0.0001) but not PDGFRβ. Conclusions These findings indicate that PDGFRβ+ and Olig2+ cells contribute to the proliferative fraction following penetrating brain injuries, with evidence of pericyte migration. Dynamic changes in Cx43 in glial cell types with dpi suggest functional alterations during temporal stages of brain repair.
Collapse
Affiliation(s)
- C Reeves
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, Queen Square, London.,Department of Neuropathology, UCL Queen Square Institute of Neurology, Queen Square, London
| | - A Pradim-Jardim
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, Queen Square, London.,Department of Neurology and Neurosurgery, Universidade Federal de Sao Paulo, UNIFESP, Sao Paulo/SP, Brazil
| | - S M Sisodiya
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, Queen Square, London.,Chalfont Centre for Epilepsy, Chesham Lane, Chalfont St Peter, Bucks, SL9 0RJ, UK
| | - M Thom
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, Queen Square, London.,Department of Neuropathology, UCL Queen Square Institute of Neurology, Queen Square, London
| | - J Y W Liu
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, Queen Square, London.,Department of Neuropathology, UCL Queen Square Institute of Neurology, Queen Square, London.,School of life Sciences, University of Westminster, London, W1W 6UW, UK
| |
Collapse
|
26
|
Beckmann A, Grißmer A, Wolf S, Recktenwald J, Meier C. Oxygen-Glucose Deprivation in Mouse Astrocytes is Associated with Ultrastructural Changes in Connexin 43 Gap Junctions. Neuroscience 2018; 397:67-79. [PMID: 30513376 DOI: 10.1016/j.neuroscience.2018.11.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 11/26/2018] [Accepted: 11/28/2018] [Indexed: 01/09/2023]
Abstract
In the intact brain, astrocytes play an important role in a number of physiological functions like spatial buffering of potassium, maintenance of calcium homeostasis, neurotransmitter release, regulation of the cerebral blood flow, and many more. As pathophysiological events upon hypoxic-ischemic brain injury include excitotoxicity by glutamate release as well as oxidative stress, astrocytes and their gap junction-based syncytium are of major relevance for regulating the extent of resulting brain damage. The gap junction protein Connexin (Cx) 43 contributes mainly to the astrocytic intercellular communication. As little is known about the ultrastructural assemblage of Cx43 and its changes in response to hypoxic events, we chose temporary oxygen and glucose deprivation with subsequent reoxygenation (OGD-R) as a metabolic inhibition model of hypoxia in primary murine astrocytes. Gap junction morphology and assembly/disintegration were analyzed at the ultrastructural level using freeze-fracture replica immunolabeling. The exposure of cultured astrocytes to short-term OGD-R resulted in the activation of ERK1/2 (p44/p42), downregulation of Cx43 protein expression, and the rearrangement of Cx43 particles within the cell membrane and within gap junctions. These changes in gap junction morphology were associated with phosphorylation of Cx43 at Serine 368. Analysis of the nearest-neighbor distance within gap junction plaques revealed the loosening of Cx43 particle clusters. Together with the observation of additional connexons being present in the vicinity of gap junction plaques after OGD-R treatment, our study indicates that changes in gap junction assembly are associated with the early phase of hypoxic cell damage.
Collapse
Affiliation(s)
- Anja Beckmann
- Department of Anatomy and Cell Biology, Saarland University, 66421 Homburg, Saar, Germany
| | - Alexander Grißmer
- Department of Anatomy and Cell Biology, Saarland University, 66421 Homburg, Saar, Germany
| | - Sandra Wolf
- Department of Anatomy and Cell Biology, Saarland University, 66421 Homburg, Saar, Germany
| | - Johanna Recktenwald
- Department of Anatomy and Cell Biology, Saarland University, 66421 Homburg, Saar, Germany
| | - Carola Meier
- Department of Anatomy and Cell Biology, Saarland University, 66421 Homburg, Saar, Germany.
| |
Collapse
|
27
|
Szeto V, Chen NH, Sun HS, Feng ZP. The role of K ATP channels in cerebral ischemic stroke and diabetes. Acta Pharmacol Sin 2018; 39:683-694. [PMID: 29671418 PMCID: PMC5943906 DOI: 10.1038/aps.2018.10] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 02/19/2018] [Indexed: 12/18/2022] Open
Abstract
ATP-sensitive potassium (KATP) channels are ubiquitously expressed on the plasma membrane of cells in multiple organs, including the heart, pancreas and brain. KATP channels play important roles in controlling and regulating cellular functions in response to metabolic state, which are inhibited by ATP and activated by Mg-ADP, allowing the cell to couple cellular metabolic state (ATP/ADP ratio) to electrical activity of the cell membrane. KATP channels mediate insulin secretion in pancreatic islet beta cells, and controlling vascular tone. Under pathophysiological conditions, KATP channels play cytoprotective role in cardiac myocytes and neurons during ischemia and/or hypoxia. KATP channel is a hetero-octameric complex, consisting of four pore-forming Kir6.x and four regulatory sulfonylurea receptor SURx subunits. These subunits are differentially expressed in various cell types, thus determining the sensitivity of the cells to specific channel modifiers. Sulfonylurea class of antidiabetic drugs blocks KATP channels, which are neuroprotective in stroke, can be one of the high stoke risk factors for diabetic patients. In this review, we discussed the potential effects of KATP channel blockers when used under pathological conditions related to diabetics and cerebral ischemic stroke.
Collapse
Affiliation(s)
- Vivian Szeto
- Departments of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada M5S 1A8
| | - Nai-hong Chen
- Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Hong-shuo Sun
- Departments of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada M5S 1A8
- Surgery
- Pharmacology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada M5S 1A8
| | - Zhong-ping Feng
- Departments of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada M5S 1A8
| |
Collapse
|
28
|
Yin X, Feng L, Ma D, Yin P, Wang X, Hou S, Hao Y, Zhang J, Xin M, Feng J. Roles of astrocytic connexin-43, hemichannels, and gap junctions in oxygen-glucose deprivation/reperfusion injury induced neuroinflammation and the possible regulatory mechanisms of salvianolic acid B and carbenoxolone. J Neuroinflammation 2018; 15:97. [PMID: 29587860 PMCID: PMC5872583 DOI: 10.1186/s12974-018-1127-3] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 03/12/2018] [Indexed: 02/07/2023] Open
Abstract
Background Glia-mediated neuroinflammation is related to brain injury exacerbation after cerebral ischemia/reperfusion (I/R) injury. Astrocytic hemichannels or gap junctions, which were mainly formed by connexin-43, have been implicated in I/R damage. However, the exact roles of astrocytic hemichannels and gap junction in neuroinflammatory responses induced by I/R injury remain unknown. Methods Primary cultured astrocytes were subjected to OGD/R injury, an in vitro model of I/R injury. Salvianolic acid B (SalB) or carbenoxolone (CBX) were applied for those astrocytes. Besides, Cx43 mimetic peptides Gap19 or Gap26 were also applied during OGD/R injury; Cx43 protein levels were determined by western blot and cytoimmunofluorescene staining, hemichannel activities by Ethidium bromide uptake and ATP concentration detection, and gap junction intercellular communication (GJIC) permeability by parachute assay. Further, astrocyte-conditioned medium (ACM) was collected and incubated with microglia. Meanwhile, ATP or apyrase were applied to explore the role of ATP during OGD/R injury. Microglial activation, M1/M2 phenotypes, and M1/M2-related cytokines were detected. Also, microglia-conditioned medium (MEM) was collected and incubated with astrocytes to further investigate its influence on astrocytic hemichannel activity and GJIC permeability. Lastly, effects of ACM and MCM on neuronal viability were detected by flow cytometry. Results We found that OGD/R induced abnormally opened hemichannels with increased ATP release and EtBr uptake but reduced GJIC permeability. WB tests showed decreased astrocytic plasma membrane’s Cx43, while showing an increase in cytoplasma. Treating OGD/R-injured microglia with ATP or OGD/R-ACM induced further microglial activation and secondary pro-inflammatory cytokine release, with the M1 phenotype predominating. Conversely, astrocytes incubated with OGD/R-MCM exhibited increased hemichannel opening but reduced GJIC coupling. Both SalB and CBX inhibited abnormal astrocytic hemichannel opening and ATP release and switched the activated microglial phenotype from M1 to M2, thus providing effective neuroprotection. Application of Gap19 or Gap26 showed similar results with CBX. We also found that OGD/R injury caused both plasma membrane p-Cx43(Ser265) and p-Src(Tyr416) significantly upregulated; application of SalB may be inhibiting Src kinase and attenuating Cx43 internalization. Meanwhile, CBX treatment induced obviously downregulation of p-Cx43(Ser368) and p-PKC(Ser729) protein levels in plasma membrane. Conclusions We propose a vicious cycle exists between astrocytic hemichannel and microglial activation after OGD/R injury, which would aggravate neuroinflammatory responses and neuronal damage. Astrocytic Cx43, hemichannels, and GJIC play critical roles in OGD/R injury-induced neuroinflammatory responses; treatment differentially targeting astrocytic Cx43, hemichannels, and GJIC may provide novel avenues for therapeutics during cerebral I/R injury. Electronic supplementary material The online version of this article (10.1186/s12974-018-1127-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiang Yin
- Department of Neurology and Neuroscience Center, the First Hospital of Jilin University, Changchun, Jilin Province, 130021, People's Republic of China
| | - Liangshu Feng
- Department of Neurology and Neuroscience Center, the First Hospital of Jilin University, Changchun, Jilin Province, 130021, People's Republic of China
| | - Di Ma
- Department of Neurology and Neuroscience Center, the First Hospital of Jilin University, Changchun, Jilin Province, 130021, People's Republic of China
| | - Ping Yin
- Department of Neurology and Neuroscience Center, the First Hospital of Jilin University, Changchun, Jilin Province, 130021, People's Republic of China
| | - Xinyu Wang
- Department of Neurology and Neuroscience Center, the First Hospital of Jilin University, Changchun, Jilin Province, 130021, People's Republic of China
| | - Shuai Hou
- Department of Neurology and Neuroscience Center, the First Hospital of Jilin University, Changchun, Jilin Province, 130021, People's Republic of China
| | - Yulei Hao
- Department of Neurology and Neuroscience Center, the First Hospital of Jilin University, Changchun, Jilin Province, 130021, People's Republic of China
| | - Jingdian Zhang
- Department of Neurology and Neuroscience Center, the First Hospital of Jilin University, Changchun, Jilin Province, 130021, People's Republic of China
| | - Meiying Xin
- Department of Neurology and Neuroscience Center, the First Hospital of Jilin University, Changchun, Jilin Province, 130021, People's Republic of China
| | - Jiachun Feng
- Department of Neurology and Neuroscience Center, the First Hospital of Jilin University, Changchun, Jilin Province, 130021, People's Republic of China.
| |
Collapse
|
29
|
Malik S, Theis M, Eugenin EA. Connexin43 Containing Gap Junction Channels Facilitate HIV Bystander Toxicity: Implications in NeuroHIV. Front Mol Neurosci 2017; 10:404. [PMID: 29259541 PMCID: PMC5723329 DOI: 10.3389/fnmol.2017.00404] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 11/21/2017] [Indexed: 02/02/2023] Open
Abstract
Human immunodeficiency virus-1 (HIV-1) infection compromises the central nervous system (CNS) in a significant number of infected individuals, resulting in neurological dysfunction that ranges from minor cognitive deficits to frank dementia. While macrophages/microglia are the predominant CNS cells infected by HIV, our laboratory and others have shown that HIV-infected astrocytes, although present in relatively low numbers with minimal to undetectable viral replication, play key role in NeuroAIDS pathogenesis. Our laboratory has identified that HIV "hijacks" connexin (Cx) containing channels, such as gap junctions (GJs) and hemichannels (HCs), to spread toxicity and apoptosis to uninfected cells even in the absence of active viral replication. In this study, using a murine model with an astrocyte-directed deletion of Cx43 gene (hGFAP-cre Cx43fl/fl) and control Cx43fl/fl mice, we examined whether few HIV-infected human astrocytoma cells (U87-CD4-CCR5), microinjected into the mouse cortex, can spread toxicity and apoptosis through GJ-mediated mechanisms, into the mouse cells, which are resistant to HIV infection. In the control Cx43fl/fl mice, microinjection of HIV-infected U87-CD4-CCR5 cells led to apoptosis in 84.28 ± 6.38% of mouse brain cells around the site of microinjection, whereas hGFAP-cre Cx43fl/fl mice exhibited minimal apoptosis (2.78 ± 1.55%). However, simultaneous injection of GJ blocker, 18α-glycyrrhetinic acid, and Cx43 blocking peptide along with microinjection of HIV-infected cells prevented apoptosis in Cx43fl/fl mice, demonstrating the Cx43 is essential for HIV-induced bystander toxicity. In conclusion, our findings demonstrate that Cx43 expression, and formation of GJs is essential for bystander apoptosis during HIV infection. These findings reveal novel potential therapeutic targets to reduce astrocyte-mediated bystander toxicity in HIV-infected individuals because despite low to undetectable viral replication in the CNS, Cx channels hijacked by HIV amplify viral neuropathogenesis.
Collapse
Affiliation(s)
- Shaily Malik
- Public Health Research Institute (PHRI), Newark, NJ, United States.,Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Martin Theis
- Institute of Cellular Neurosciences, University of Bonn, Bonn, Germany
| | - Eliseo A Eugenin
- Public Health Research Institute (PHRI), Newark, NJ, United States.,Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| |
Collapse
|
30
|
Meneses CS, Müller HY, Herzberg DE, Uberti B, Bustamante HA, Werner MP. Immunofluorescence characterization of spinal cord dorsal horn microglia and astrocytes in horses. PeerJ 2017; 5:e3965. [PMID: 29085760 PMCID: PMC5661433 DOI: 10.7717/peerj.3965] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 10/06/2017] [Indexed: 12/12/2022] Open
Abstract
The role of glial cells in pain modulation has recently gathered attention. The objective of this study was to determine healthy spinal microglia and astrocyte morphology and disposition in equine spinal cord dorsal horns using Iba-1 and GFAP/Cx-43 immunofluorescence labeling, respectively. Five adult horses without visible wounds or gait alterations were selected. Spinal cord segments were obtained post-mortem for immunohistochemical and immunocolocalization assays. Immunodetection of spinal cord dorsal horn astrocytes was done using a polyclonal goat antibody raised against Glial Fibrillary Acidic Protein (GFAP) and a polyclonal rabbit antibody against Connexin 43 (Cx-43). For immunodetection of spinal cord dorsal horn microglia, a polyclonal rabbit antibody against a synthetic peptide corresponding to the C-terminus of ionized calcium-binding adaptor molecule 1 (Iba-1) was used. Epifluorescence and confocal images were obtained for the morphological and organizational analysis. Evaluation of shape, area, cell diameter, cell process length and thickness was performed on dorsal horn microglia and astrocyte. Morphologically, an amoeboid spherical shape with a mean cell area of 92.4 + 34 µm2 (in lamina I, II and III) was found in horse microglial cells, located primarily in laminae I, II and III. Astrocyte primary stem branches (and cellular bodies to a much lesser extent) are mainly detected using GFAP. Thus, double GFAP/Cx-43 immunolabeling was needed in order to accurately characterize the morphology, dimension and cell density of astrocytes in horses. Horse and rodent astrocytes seem to have similar dimensions and localization. Horse astrocyte cells have an average diameter of 56 + 14 µm, with a main process length of 28 + 8 µm, and thickness of 1.4 + 0.3 µm, mainly situated in laminae I, II and III. Additionally, a close association between end-point astrocyte processes and microglial cell bodies was found. These results are the first characterization of cell morphology and organizational aspects of horse spinal glia. Iba-1 and GFAP/Cx-43 can successfully immune-label microglia and astrocytes respectively in horse spinal cords, and thus reveal cell morphology and corresponding distribution within the dorsal horn laminae of healthy horses. The conventional hyper-ramified shape that is normally visible in resting microglial cells was not found in horses. Instead, horse microglial cells had an amoeboid spherical shape. Horse protoplasmic astroglia is significantly smaller and structurally less complex than human astrocytes, with fewer main GFAP processes. Instead, horse astrocytes tend to be similar to those found in rodent’s model, with small somas and large cell processes. Microglia and astrocytes were found in the more superficial regions of the dorsal horn, similarly to that previously observed in humans and rodents. Further studies are needed to demonstrate the molecular mechanisms involved in the neuron-glia interaction in horses.
Collapse
Affiliation(s)
| | - Heine Yacob Müller
- Veterinary Sciences Graduate School, Universidad Austral de Chile, Valdivia, Chile
| | | | - Benjamín Uberti
- Veterinary Clinical Sciences Department, Universidad Austral de Chile, Valdivia, Chile
| | | | | |
Collapse
|
31
|
Kovacs GG, Yousef A, Kaindl S, Lee VM, Trojanowski JQ. Connexin-43 and aquaporin-4 are markers of ageing-related tau astrogliopathy (ARTAG)-related astroglial response. Neuropathol Appl Neurobiol 2017; 44:491-505. [PMID: 28755467 DOI: 10.1111/nan.12427] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Revised: 07/14/2017] [Accepted: 07/24/2017] [Indexed: 12/21/2022]
Abstract
AIMS Ageing-related tau astrogliopathy (ARTAG) appears in subependymal, subpial, perivascular, white matter (WM) and grey matter (GM) locations. Physical effects, blood-brain barrier dysfunction and blood- or vessel-related factors have been considered as aetiology. As connexin-43 (Cx43) and aquaporin-4 (AQP4) are related to these, we hypothesized that their immunoreactivity (IR) varies with ARTAG in a location-specific manner. METHODS We performed a morphometric immunohistochemical study measuring the densities of IR of Cx43, AQP4, AT8 (phospho-tau) and glial fibrillar acidic protein (GFAP). We analysed the amygdala and hippocampus in age-matched cases with (n = 19) and without (n = 20) ARTAG in each of the locations it aggregates. RESULTS We show a dramatic increase (>6-fold; P < 0.01) of Cx43 density of IR in ARTAG cases correlating strongly with AT8 density of IR, irrespective of the presence of neuronal tau pathology or reactive gliosis measured by GFAP density of IR, in the GM. In contrast, AQP4 density of IR was increased only in the WM and GM, and was associated with increased AT8 density of IR only in WM and perivascular areas. DISCUSSION Our study reveals distinctive astroglial responses in each of the locations associated with ARTAG. Our observations support the concept that factors related to brain-fluid interfaces and water-ion imbalances most likely play a role in the generation of ARTAG. As Cx43 is crucial for maintaining neuronal homeostasis, the ARTAG-dependent increase of Cx43 density of IR suggests that the development of ARTAG in the GM most likely indicates an early response to the degeneration of neurons.
Collapse
Affiliation(s)
- G G Kovacs
- Institute of Neurology, Medical University of Vienna, Vienna, Austria.,Center for Neurodegenerative Disease Research, Institute on Aging and Department of Pathology and Laboratory Medicine of the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - A Yousef
- Center for Neurodegenerative Disease Research, Institute on Aging and Department of Pathology and Laboratory Medicine of the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - S Kaindl
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - V M Lee
- Center for Neurodegenerative Disease Research, Institute on Aging and Department of Pathology and Laboratory Medicine of the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - J Q Trojanowski
- Center for Neurodegenerative Disease Research, Institute on Aging and Department of Pathology and Laboratory Medicine of the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
32
|
Choi DH, Ahn JH, Choi IA, Kim JH, Kim BR, Lee J. Effect of task-specific training on Eph/ephrin expression after stroke. BMB Rep 2017; 49:635-640. [PMID: 27756445 PMCID: PMC5346325 DOI: 10.5483/bmbrep.2016.49.11.172] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Indexed: 11/30/2022] Open
Abstract
Recent evidence indicates that the ephrin receptors and ephrin ligands (Eph/ephrin) expression modulate axonal reorganization and synaptic plasticity in stroke recovery. To investigate the effect of task-specific training (TST) on Eph/ephrin expression in the corticospinal tract (CST) after stroke, we compared Eph/ephrin expression in the peri-infarct cortex, pyramid, and spinal cord of a photothrombotic stroke model of rat brains treated with or without TST. The TST treatment showed significantly better recovery in the behavioral tests compared with no treatment. The significant upregulation of ephrin-A1 and ephrin-A5 observed in activated astrocytes of the CST at 2 weeks’ post-stroke was decreased by TST. At 5 weeks, post-stroke, the elevated ephrin-A5 levels were decreased in the ipsilateral pyramid and spinal cord by TST. Glial fibrillary acidic protein was upregulated concomitantly with the altered ephrin expression after stroke, and the expression of these proteins was attenuated by TST. These data suggest that TST alters the expression of ephrin ligands in the CST after stroke.
Collapse
Affiliation(s)
- Dong-Hee Choi
- Departments of Medical Science, Konkuk University School of Medicine; Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, Seoul 05029, Korea
| | - Jin-Hee Ahn
- Departments of Medical Science, Konkuk University School of Medicine, Seoul 05029, Korea
| | - In-Ae Choi
- Departments of Medical Science, Konkuk University School of Medicine, Seoul 05029, Korea
| | - Ji-Hye Kim
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, Seoul 05029, Korea
| | - Bo-Ram Kim
- Department of Rehabilitation Medicine, Konkuk University School of Medicine, Seoul 05029, Korea
| | - Jongmin Lee
- Rehabilitation Medicine, Konkuk University School of Medicine; Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, Seoul 05029, Korea
| |
Collapse
|
33
|
Santos JD, Batista RI, Magalhães LC, Paula Jr. AR, Souza SS, Salamone DF, Bhat MH, Teixeira DI, Freitas VJ, Melo LM. Overexpression of hyaluronan synthase 2 and gonadotropin receptors in cumulus cells of goats subjected to one-shot eCG/FSH hormonal treatment for ovarian stimulation. Anim Reprod Sci 2016; 170:15-24. [DOI: 10.1016/j.anireprosci.2016.03.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 03/06/2016] [Accepted: 03/15/2016] [Indexed: 12/21/2022]
|
34
|
Abstract
Astrocytes are the most explored non-neuronal cells in the brain under neurophysiological and neurodegenerative conditions. Extensive research has been done to understand their specific role during neuropathological conditions but still the existing findings could not conclude their mechanism of action and their specific role in neurodegenerative conditions. This review discusses their physiological and pathological roles, their activation, morphological alterations and their probable use in search of new therapeutic targets for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Sarika Singh
- a 1 Toxicology Division, CSIR-CDRI , Lucknow , India.,b 2 Department of Biochemistry and Biophysics , University of California , San Francisco, San Francisco , CA , USA
| | - Neeraj Joshi
- a 1 Toxicology Division, CSIR-CDRI , Lucknow , India.,b 2 Department of Biochemistry and Biophysics , University of California , San Francisco, San Francisco , CA , USA
| |
Collapse
|