1
|
Muñoz-Ballester C, Leitzel O, Golf S, Phillips CM, Zeitz MJ, Pandit R, Smyth JW, Lamouille S, Robel S. Astrocytic connexin43 phosphorylation contributes to seizure susceptibility after mild Traumatic Brain Injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.12.623104. [PMID: 39605358 PMCID: PMC11601309 DOI: 10.1101/2024.11.12.623104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Astrocytes play a crucial role in maintaining brain homeostasis through functional gap junctions (GJs) primarily formed by connexin43 (Cx43). These GJs facilitate electrical and metabolic coupling between astrocytes, allowing the passage of ions, glucose, and metabolites. Dysregulation of Cx43 has been implicated in various pathologies, including traumatic brain injury (TBI) and acquired epilepsy. We previously identified a subset of atypical astrocytes after mild TBI that exhibit reduced Cx43 expression and coupling and are correlated with the development of spontaneous seizures. Given that mild TBI affects millions globally and can lead to long-term complications, including post-traumatic epilepsy, understanding the molecular events post-TBI is critical for developing therapeutic strategies. In the present study, we assessed the heterogeneity of Cx43 protein expression after mild TBI. In accordance with our previous findings, a subset of astrocytes lost Cx43 expression. As previously reported after TBI, we also found a significant increase in total Cx43 protein expression after mild TBI, predominantly in the soluble form, suggesting that while junctional Cx43 protein levels remained stable, hemichannels and cytoplasmic Cx43 were increased. We then investigated the phosphorylation of Cx43 at serine 368 after TBI, which is known to influence GJ assembly and function. Phosphorylation of Cx43 at serine 368 is elevated following TBI and Cx43S368A mutant mice, lacking this phosphorylation, exhibited reduced susceptibility to seizures induced by pentylenetetrazol (PTZ). These findings suggest that TBI-induced Cx43 phosphorylation enhances seizure susceptibility, while inhibiting this modification presents a potential therapeutic avenue for mitigating neuronal hyperexcitability and seizure development.
Collapse
Affiliation(s)
- Carmen Muñoz-Ballester
- Cell, Developmental and Integrative Biology Department,
University of Alabama at Birmingham, Birmingham, 21353, AL
| | - Owen Leitzel
- Cell, Developmental and Integrative Biology Department,
University of Alabama at Birmingham, Birmingham, 21353, AL
| | - Samantha Golf
- Cell, Developmental and Integrative Biology Department,
University of Alabama at Birmingham, Birmingham, 21353, AL
| | - Chelsea M Phillips
- Fralin Biomedical Research Institute at Virginia Tech Carilion,
Roanoke, 24016, VA
| | - Michael J Zeitz
- Fralin Biomedical Research Institute at Virginia Tech Carilion,
Roanoke, 24016, VA
| | - Rahul Pandit
- Cell, Developmental and Integrative Biology Department,
University of Alabama at Birmingham, Birmingham, 21353, AL
| | - James W. Smyth
- Fralin Biomedical Research Institute at Virginia Tech Carilion,
Roanoke, 24016, VA
- Department of Biological Sciences, College of Science, Virginia
Tech, Blacksburg, 24061, VA
- Virginia Tech Carilion School of Medicine, Roanoke, 24016,
VA
| | - Samy Lamouille
- Fralin Biomedical Research Institute at Virginia Tech Carilion,
Roanoke, 24016, VA
- Department of Biological Sciences, College of Science, Virginia
Tech, Blacksburg, 24061, VA
- Virginia Tech Carilion School of Medicine, Roanoke, 24016,
VA
| | - Stefanie Robel
- Cell, Developmental and Integrative Biology Department,
University of Alabama at Birmingham, Birmingham, 21353, AL
- Department of Physical Medicine and Rehabilitation, University of
Alabama at Birmingham, Birmingham, 35212, AL
| |
Collapse
|
2
|
Ucar EA, Ozkan E, Shomalizadeh N, Sekerdağ-Kilic E, Akpunar F, Sapanci S, Kesibi J, Ozler C, Bilgez AS, Gursoy-Ozdemir Y. Carbenoxolone mitigates extensive fibrosis formation in PLP-induced EAE model and multiple sclerosis serum-exposed pericyte culture. Front Cell Neurosci 2024; 18:1403974. [PMID: 38746079 PMCID: PMC11091252 DOI: 10.3389/fncel.2024.1403974] [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: 03/20/2024] [Accepted: 04/16/2024] [Indexed: 05/16/2024] Open
Abstract
Introduction Multiple sclerosis (MS) is one of the most common causes of disability in young adults. Nearly, 85% of MS cases start with attacks and remissions, classified as relapsing-remitting multiple sclerosis (RRMS). With repeating attacks, MS causes brain-spinal cord atrophy and enhanced disability as disease progresses. PLP-induced EAE is one of the most established models for pathophysiology and treatment of RRMS. Recent studies demonstrated the possible role of pericytes in perivascular and intra-lesional fibrosis in PLP-induced EAE, whose importance remains elusive. Hence, we have investigated the possible role of pericytes in fibrosis formation and amelioration with a hemichannel blocker, Carbenoxolone (CBX). Methods PLP-induced experimental autoimmune encephalitis (EAE) model is used and the effect of CBX is investigated. Clinical scores were recorded and followed. Perivascular Collagen 1 and 3 accumulations were demonstrated as markers of fibrosis in the spinal cord. To delineate the role of pericytes, human brain vascular pericytes (HBVP) were incubated with the sera of MS patients to induce in-vitro MS model and the fibrosis formation was investigated. Results In the PLP induced in-vivo model, both intracerebroventricular and intraperitoneal CBX have significantly mitigated the disease progression followed by clinical scores, demyelination, and fibrosis. Moreover, CBX significantly mitigated MS-serum-induced fibrosis in the HBVP cell culture. Discussion The study demonstrated two important findings. First, CBX decreases fibrosis formation in both in-vivo and in-vitro MS models. Secondly, it improves neurological scores and decreases demyelination in the EAE model. Therefore, CBX can be potential novel therapeutic option in treating Multiple Sclerosis.
Collapse
Affiliation(s)
- Ege Anil Ucar
- Research Center for Translational Medicine (KUTTAM), Koҫ University, Istanbul, Türkiye
- School of Medicine, Koç University, Istanbul, Türkiye
| | - Esra Ozkan
- Research Center for Translational Medicine (KUTTAM), Koҫ University, Istanbul, Türkiye
- Department of Neurology, Koç University, Istanbul, Türkiye
| | - Narges Shomalizadeh
- Research Center for Translational Medicine (KUTTAM), Koҫ University, Istanbul, Türkiye
| | - Emine Sekerdağ-Kilic
- Research Center for Translational Medicine (KUTTAM), Koҫ University, Istanbul, Türkiye
| | - Fatmanur Akpunar
- Research Center for Translational Medicine (KUTTAM), Koҫ University, Istanbul, Türkiye
| | - Selin Sapanci
- Research Center for Translational Medicine (KUTTAM), Koҫ University, Istanbul, Türkiye
| | - Judy Kesibi
- Research Center for Translational Medicine (KUTTAM), Koҫ University, Istanbul, Türkiye
| | - Ceyda Ozler
- Research Center for Translational Medicine (KUTTAM), Koҫ University, Istanbul, Türkiye
| | - Alara Su Bilgez
- Research Center for Translational Medicine (KUTTAM), Koҫ University, Istanbul, Türkiye
| | - Yasemin Gursoy-Ozdemir
- Research Center for Translational Medicine (KUTTAM), Koҫ University, Istanbul, Türkiye
- School of Medicine, Koç University, Istanbul, Türkiye
- Department of Neurology, Koç University, Istanbul, Türkiye
| |
Collapse
|
3
|
Yamasaki R. Connexins Control Glial Inflammation in Various Neurological Diseases. Int J Mol Sci 2023; 24:16879. [PMID: 38069203 PMCID: PMC10706219 DOI: 10.3390/ijms242316879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/25/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Connexins (Cxs) form gap junctions through homotypic/heterotypic oligomerization. Cxs are initially synthesized in the endoplasmic reticulum, then assembled as hexamers in the Golgi apparatus before being integrated into the cell membrane as hemichannels. These hemichannels remain closed until they combine to create gap junctions, directly connecting neighboring cells. Changes in the intracellular or extracellular environment are believed to trigger the opening of hemichannels, creating a passage between the inside and outside of the cell. The size of the channel pore depends on the Cx isoform and cellular context-specific effects such as posttranslational modifications. Hemichannels allow various bioactive molecules, under ~1 kDa, to move in and out of the host cell in the direction of the electrochemical gradient. In this review, we explore the fundamental roles of Cxs and their clinical implications in various neurological dysfunctions, including hereditary diseases, ischemic brain disorders, degenerative conditions, demyelinating disorders, and psychiatric illnesses. The influence of Cxs on the pathomechanisms of different neurological disorders varies depending on the circumstances. Hemichannels are hypothesized to contribute to proinflammatory effects by releasing ATP, adenosine, glutamate, and other bioactive molecules, leading to neuroglial inflammation. Modulating Cxs' hemichannels has emerged as a promising therapeutic approach.
Collapse
Affiliation(s)
- Ryo Yamasaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| |
Collapse
|
4
|
Glial Cell Metabolic Profile Upon Iron Deficiency: Oligodendroglial and Astroglial Casualties of Bioenergetic Adjustments. Mol Neurobiol 2023; 60:1949-1963. [PMID: 36595194 DOI: 10.1007/s12035-022-03149-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 11/24/2022] [Indexed: 01/04/2023]
Abstract
Iron deficiency (ID) represents one of the most prevalent nutritional deficits, affecting almost two billion people worldwide. Gestational iron deprivation induces hypomyelination due to oligodendroglial maturation deficiencies and is thus a useful experimental model to analyze oligodendrocyte (OLG) requirements to progress to a mature myelinating state. A previous proteomic study in the adult ID brain by our group demonstrated a pattern of dysregulated proteins involved in the tricarboxylic acid cycle and mitochondrial dysfunction. The aim of the present report was to assess bioenergetics metabolism in primary cultures of OLGs and astrocytes (ASTs) from control and ID newborns, on the hypothesis that the regulation of cell metabolism correlates with cell maturation. Oxygen consumption and extracellular acidification rates were measured using a Seahorse extracellular flux analyzer. ID OLGs and ASTs both exhibited decreased spare respiratory capacity, which indicates that ID effectively induces mitochondrial dysfunction. A decrease in glycogen granules was observed in ID ASTs, and an increase in ROS production was detected in ID OLGs. Immunolabeling of structural proteins showed that mitochondrial number and size were increased in ID OLGs, while an increased number of smaller mitochondria was observed in ID ASTs. These results reflect an unfavorable bioenergetic scenario in which ID OLGs fail to progress to a myelinating state, and indicate that the regulation of cell metabolism may impact cell fate decisions and maturation.
Collapse
|
5
|
Chen YH, Jin SY, Yang JM, Gao TM. The Memory Orchestra: Contribution of Astrocytes. Neurosci Bull 2023; 39:409-424. [PMID: 36738435 PMCID: PMC10043126 DOI: 10.1007/s12264-023-01024-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 01/03/2023] [Indexed: 02/05/2023] Open
Abstract
For decades, memory research has centered on the role of neurons, which do not function in isolation. However, astrocytes play important roles in regulating neuronal recruitment and function at the local and network levels, forming the basis for information processing as well as memory formation and storage. In this review, we discuss the role of astrocytes in memory functions and their cellular underpinnings at multiple time points. We summarize important breakthroughs and controversies in the field as well as potential avenues to further illuminate the role of astrocytes in memory processes.
Collapse
Affiliation(s)
- Yi-Hua Chen
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Shi-Yang Jin
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jian-Ming Yang
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Tian-Ming Gao
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
6
|
Murray CJ, Vecchiarelli HA, Tremblay MÈ. Enhancing axonal myelination in seniors: A review exploring the potential impact cannabis has on myelination in the aged brain. Front Aging Neurosci 2023; 15:1119552. [PMID: 37032821 PMCID: PMC10073480 DOI: 10.3389/fnagi.2023.1119552] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/22/2023] [Indexed: 04/11/2023] Open
Abstract
Consumption of cannabis is on the rise as public opinion trends toward acceptance and its consequent legalization. Specifically, the senior population is one of the demographics increasing their use of cannabis the fastest, but research aimed at understanding cannabis' impact on the aged brain is still scarce. Aging is characterized by many brain changes that slowly alter cognitive ability. One process that is greatly impacted during aging is axonal myelination. The slow degradation and loss of myelin (i.e., demyelination) in the brain with age has been shown to associate with cognitive decline and, furthermore, is a common characteristic of numerous neurological diseases experienced in aging. It is currently not known what causes this age-dependent degradation, but it is likely due to numerous confounding factors (i.e., heightened inflammation, reduced blood flow, cellular senescence) that impact the many cells responsible for maintaining overall homeostasis and myelin integrity. Importantly, animal studies using non-human primates and rodents have also revealed demyelination with age, providing a reliable model for researchers to try and understand the cellular mechanisms at play. In rodents, cannabis was recently shown to modulate the myelination process. Furthermore, studies looking at the direct modulatory impact cannabis has on microglia, astrocytes and oligodendrocyte lineage cells hint at potential mechanisms to prevent some of the more damaging activities performed by these cells that contribute to demyelination in aging. However, research focusing on how cannabis impacts myelination in the aged brain is lacking. Therefore, this review will explore the evidence thus far accumulated to show how cannabis impacts myelination and will extrapolate what this knowledge may mean for the aged brain.
Collapse
Affiliation(s)
- Colin J. Murray
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- *Correspondence: Colin J. Murray,
| | | | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Départment de Médicine Moléculaire, Université Laval, Québec City, QC, Canada
- Axe Neurosciences, Center de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
- Neurology and Neurosurgery Department, McGill University, Montréal, QC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
- Institute for Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada
- Marie-Ève Tremblay,
| |
Collapse
|
7
|
Nishimura Y, Masaki K, Matsuse D, Yamaguchi H, Tanaka T, Matsuo E, Hayashida S, Watanabe M, Matsushita T, Sadashima S, Sasagasako N, Yamasaki R, Isobe N, Iwaki T, Kira J. Early and extensive alterations of glial connexins, distal oligodendrogliopathy type demyelination, and nodal/paranodal pathology are characteristic of multiple system atrophy. Brain Pathol 2022; 33:e13131. [PMID: 36368713 PMCID: PMC10154368 DOI: 10.1111/bpa.13131] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 10/24/2022] [Indexed: 11/13/2022] Open
Abstract
The pathological hallmark of multiple system atrophy (MSA) is aberrant accumulation of phosphorylated α-synuclein in oligodendrocytes, forming glial cytoplasmic inclusions (GCIs). Extensive demyelination occurs particularly in the olivopontocerebellar and striatonigral pathways, but its precise mechanism remains elusive. Glial connexins (Cxs), which form gap junction channels between astrocytes and oligodendrocytes, play critical roles in myelin maintenance, and have not been studied in MSA. Therefore, we immunohistochemically investigated glial Cx changes in the cerebellar afferent fibers in 15 autopsied patients with MSA. We classified demyelinating lesions into three stages based on Klüver-Barrera staining: early (Stage I), intermediate (Stage II), and late (Stage III) stages showing subtle, moderate, and severe myelin reduction, respectively. Myelin-associated glycoprotein, but not myelin oligodendrocyte glycoprotein, was preferentially decreased in Stage I, suggesting distal oligodendrogliopathy type demyelination. Accumulation of phosphorylated α-synuclein in oligodendrocytes was frequently seen in Stage I but less frequently observed in Stages II and III. Tubulin polymerization-promoting protein (TPPP/p25α)-positive oligodendrocytes were preserved in Stage I but successively decreased in Stages II and III. Even at Stage I, Cx32 was nearly absent from myelin, despite the relative preservation of other nodal proteins, such as neurofascin, claudin-11/oligodendrocyte-specific protein, and contactin-associated protein 1, which successively decreased in the later stages. Cx32 was re-distributed in the oligodendrocyte cytoplasm and co-localized with GCIs. Cx47 gradually decreased at the oligodendrocyte surface in a stage-dependent manner but was not co-localized with GCIs. Astrocytic Cx43 was down-regulated in Stage I but up-regulated in Stages II and III, reflecting astrogliosis. Cx43/Cx47 gap junctions significantly decreased from Stage I to III. Activated microglia/macrophages and T cells infiltrated in Stage I rather than Stages II and III. Therefore, early and extensive alterations of glial Cxs, particularly Cx32 loss, occur in MSA and may accelerate distal oligodendrogliopathy type demyelination and nodal/paranodal dysfunction through disruption of inter-glial communication.
Collapse
Affiliation(s)
- Yuji Nishimura
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Katsuhisa Masaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Dai Matsuse
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Hiroo Yamaguchi
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Tatsunori Tanaka
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences Kyushu University Fukuoka Japan
- Sumitomo Pharma Osaka Japan
| | - Eriko Matsuo
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Shotaro Hayashida
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Mitsuru Watanabe
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Takuya Matsushita
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Shoko Sadashima
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences Kyushu University Fukuoka Japan
- Department of Neuropathology, Neurological Institute, Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Naokazu Sasagasako
- Department of Neurology, Neuro‐Muscular Center National Omuta Hospital Fukuoka Japan
| | - Ryo Yamasaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Noriko Isobe
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Toru Iwaki
- Department of Neuropathology, Neurological Institute, Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Jun‐ichi Kira
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences Kyushu University Fukuoka Japan
- Translational Neuroscience Center, Graduate School of Medicine, and School of Pharmacy at Fukuoka International University of Health and Welfare Ookawa Japan
- Department of Neurology, Brain and Nerve Center Fukuoka Central Hospital Fukuoka Japan
| |
Collapse
|
8
|
Wang B, Li X, Li H, Xiao L, Zhou Z, Chen K, Gui L, Hou X, Fan R, Chen K, Wu W, Li H, Hu X. Clinical, Radiological and Pathological Characteristics Between Cerebral Small Vessel Disease and Multiple Sclerosis: A Review. Front Neurol 2022; 13:841521. [PMID: 35812110 PMCID: PMC9263123 DOI: 10.3389/fneur.2022.841521] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
Cerebral small vessel disease (CSVD) and multiple sclerosis (MS) are a group of diseases associated with small vessel lesions, the former often resulting from the vascular lesion itself, while the latter originating from demyelinating which can damage the cerebral small veins. Clinically, CSVD and MS do not have specific signs and symptoms, and it is often difficult to distinguish between the two from the aspects of the pathology and imaging. Therefore, failure to correctly identify and diagnose the two diseases will delay early intervention, which in turn will affect the long-term functional activity for patients and even increase their burden of life. This review has summarized recent studies regarding their similarities and difference of the clinical manifestations, pathological features and imaging changes in CSVD and MS, which could provide a reliable basis for the diagnosis and differentiation of the two diseases in the future.
Collapse
Affiliation(s)
- Bijia Wang
- Department of Neurology, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xuegang Li
- Department of Neurosurgery, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Haoyi Li
- Department of Neurosurgery, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Li Xiao
- Department of Neurology, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhenhua Zhou
- Department of Neurology, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Kangning Chen
- Department of Neurology, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Li Gui
- Department of Neurology, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xianhua Hou
- Department of Neurology, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Rong Fan
- Department of Neurology, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Kang Chen
- Department of Radiology, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Wenjing Wu
- Department of Radiology, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Haitao Li
- Department of Radiology, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- *Correspondence: Haitao Li
| | - Xiaofei Hu
- Department of Radiology, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- Xiaofei Hu
| |
Collapse
|
9
|
He T, Yang GY, Zhang Z. Crosstalk of Astrocytes and Other Cells during Ischemic Stroke. LIFE (BASEL, SWITZERLAND) 2022; 12:life12060910. [PMID: 35743941 PMCID: PMC9228674 DOI: 10.3390/life12060910] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/04/2022] [Accepted: 06/07/2022] [Indexed: 12/27/2022]
Abstract
Stroke is a leading cause of death and long-term disability worldwide. Astrocytes structurally compose tripartite synapses, blood–brain barrier, and the neurovascular unit and perform multiple functions through cell-to-cell signaling of neurons, glial cells, and vasculature. The crosstalk of astrocytes and other cells is complicated and incompletely understood. Here we review the role of astrocytes in response to ischemic stroke, both beneficial and detrimental, from a cell–cell interaction perspective. Reactive astrocytes provide neuroprotection through antioxidation and antiexcitatory effects and metabolic support; they also contribute to neurorestoration involving neurogenesis, synaptogenesis, angiogenesis, and oligodendrogenesis by crosstalk with stem cells and cell lineage. In the meantime, reactive astrocytes also play a vital role in neuroinflammation and brain edema. Glial scar formation in the chronic phase hinders functional recovery. We further discuss astrocyte enriched microRNAs and exosomes in the regulation of ischemic stroke. In addition, the latest notion of reactive astrocyte subsets and astrocytic activity revealed by optogenetics is mentioned. This review discusses the current understanding of the intimate molecular conversation between astrocytes and other cells and outlines its potential implications after ischemic stroke. “Neurocentric” strategies may not be sufficient for neurological protection and recovery; future therapeutic strategies could target reactive astrocytes.
Collapse
Affiliation(s)
- Tingting He
- Department of Neurology, Shanghai Tenth People’s Hospital, Tongji University, Shanghai 200072, China;
- Neuroscience and Neuroengineering Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Guo-Yuan Yang
- Neuroscience and Neuroengineering Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
- Correspondence: (G.-Y.Y.); (Z.Z.); Tel.: +86-21-62933186 (G.-Y.Y.); Fax: +86-21-62932302 (G.-Y.Y.)
| | - Zhijun Zhang
- Neuroscience and Neuroengineering Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
- Correspondence: (G.-Y.Y.); (Z.Z.); Tel.: +86-21-62933186 (G.-Y.Y.); Fax: +86-21-62932302 (G.-Y.Y.)
| |
Collapse
|
10
|
Baló's concentric sclerosis - A rare entity within the spectrum of demyelinating diseases. J Neurol Sci 2021; 428:117570. [PMID: 34261000 DOI: 10.1016/j.jns.2021.117570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/19/2021] [Accepted: 07/05/2021] [Indexed: 11/24/2022]
Abstract
Baló's concentric sclerosis (BCS) is a rare, inflammatory demyelinating disease of the central nervous system (CNS). Historically, BCS was thought to be uniformly fatal and diagnosis was based on postmortem findings. With advances in modern neuroimaging, BCS is currently defined by the presence of concentric layered patterns composed of alternating rings of varying intensity. They are best appreciated on gadolinium-enhanced T1-weighted sequences and predominantly occur in the supratentorial cerebral white matter with sparing of cortical U-fibers. The lamellar pattern of the lesions likely reflects bands of demyelination and relative myelin preservation with minimal axonal loss. While BCS falls within the spectrum of atypical demyelinating diseases, there is ongoing debate over whether BCS is a phenotypical variant of multiple sclerosis (MS) or a separate entity. Corticosteroids comprise first-line therapy but there is ongoing controversy regarding appropriate maintenance therapy. First-line MS disease-modifying therapies such as interferon beta-1a are appropriate for patients who fulfill diagnostic criteria for relapsing-remitting MS. Fingolimod should likely be avoided as Baló-like lesions have been reported during its administration or after withdrawal. Monoclonal antibodies such as natalizumab and rituximab are potentially effective at reducing BCS relapses, but alemtuzumab may be relatively ineffective because humoral immunity does not play a central role in BCS pathogenesis.
Collapse
|
11
|
Rao SB, Skauli N, Jovanovic N, Katoozi S, Frigeri A, Froehner SC, Adams ME, Ottersen OP, Amiry-Moghaddam M. Orchestrating aquaporin-4 and connexin-43 expression in brain: Differential roles of α1- and β1-syntrophin. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2021; 1863:183616. [PMID: 33872576 DOI: 10.1016/j.bbamem.2021.183616] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/22/2021] [Accepted: 03/25/2021] [Indexed: 01/09/2023]
Abstract
Aquaporin-4 (AQP4) water channels and gap junction proteins (connexins) are two classes of astrocytic membrane proteins critically involved in brain water and ion homeostasis. AQP4 channels are anchored by α1-syntrophin to the perivascular astrocytic endfoot membrane domains where they control water flux at the blood-brain interface while connexins cluster at the lateral aspects of the astrocytic endfeet forming gap junctions that allow water and ions to dissipate through the astrocyte syncytium. Recent studies have pointed to an interdependence between astrocytic AQP4 and astrocytic gap junctions but the underlying mechanism remains to be explored. Here we use a novel transgenic mouse line to unravel whether β1-syntrophin (coexpressed with α1-syntrophin in astrocytic plasma membranes) is implicated in the expression of AQP4 isoforms and formation of gap junctions in brain. Our results show that while the effect of β1-syntrophin deletion is rather limited, double knockout of α1- and β1-syntrophin causes a downregulation of the novel AQP4 isoform AQP4ex and an increase in the number of astrocytic gap junctions. The present study highlight the importance of syntrophins in orchestrating specialized functional domains of brain astrocytes.
Collapse
Affiliation(s)
- Shreyas B Rao
- Laboratory of Molecular Neuroscience, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Post box 1105, Blindern, 0317 Oslo, Norway.
| | - Nadia Skauli
- Laboratory of Molecular Neuroscience, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Post box 1105, Blindern, 0317 Oslo, Norway.
| | - Nenad Jovanovic
- Laboratory of Molecular Neuroscience, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Post box 1105, Blindern, 0317 Oslo, Norway
| | - Shirin Katoozi
- Laboratory of Molecular Neuroscience, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Post box 1105, Blindern, 0317 Oslo, Norway
| | - Antonio Frigeri
- School of Medicine, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari Aldo Moro, Bari, Italy.
| | - Stanley C Froehner
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195-7290, USA.
| | - Marvin E Adams
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195-7290, USA.
| | - Ole Petter Ottersen
- Laboratory of Molecular Neuroscience, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Post box 1105, Blindern, 0317 Oslo, Norway.
| | - Mahmood Amiry-Moghaddam
- Laboratory of Molecular Neuroscience, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Post box 1105, Blindern, 0317 Oslo, Norway.
| |
Collapse
|
12
|
Kaul D, Schwab SG, Mechawar N, Matosin N. How stress physically re-shapes the brain: Impact on brain cell shapes, numbers and connections in psychiatric disorders. Neurosci Biobehav Rev 2021; 124:193-215. [PMID: 33556389 DOI: 10.1016/j.neubiorev.2021.01.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/20/2021] [Accepted: 01/31/2021] [Indexed: 12/16/2022]
Abstract
Severe stress is among the most robust risk factors for the development of psychiatric disorders. Imaging studies indicate that life stress is integral to shaping the human brain, especially regions involved in processing the stress response. Although this is likely underpinned by changes to the cytoarchitecture of cellular networks in the brain, we are yet to clearly understand how these define a role for stress in human psychopathology. In this review, we consolidate evidence of macro-structural morphometric changes and the cellular mechanisms that likely underlie them. Focusing on stress-sensitive regions of the brain, we illustrate how stress throughout life may lead to persistent remodelling of the both neurons and glia in cellular networks and how these may lead to psychopathology. We support that greater translation of cellular alterations to human cohorts will support parsing the psychological sequalae of severe stress and improve our understanding of how stress shapes the human brain. This will remain a critical step for improving treatment interventions and prevention outcomes.
Collapse
Affiliation(s)
- Dominic Kaul
- Illawarra Health and Medical Research Institute, Northfields Ave, Wollongong 2522, Australia; Molecular Horizons, School of Chemistry and Molecular Biosciences, University of Wollongong, Northfields Ave, Wollongong 2522, Australia
| | - Sibylle G Schwab
- Illawarra Health and Medical Research Institute, Northfields Ave, Wollongong 2522, Australia; Molecular Horizons, School of Chemistry and Molecular Biosciences, University of Wollongong, Northfields Ave, Wollongong 2522, Australia
| | - Naguib Mechawar
- Douglas Mental Health University Institute, 6875 LaSalle blvd, Verdun, Qc, H4H 1R3, Canada
| | - Natalie Matosin
- Illawarra Health and Medical Research Institute, Northfields Ave, Wollongong 2522, Australia; Molecular Horizons, School of Chemistry and Molecular Biosciences, University of Wollongong, Northfields Ave, Wollongong 2522, Australia; Max Planck Institute of Psychiatry, Kraepelinstrasse 2-10, 80804 Munich, Germany.
| |
Collapse
|
13
|
Lohrberg M, Winkler A, Franz J, van der Meer F, Ruhwedel T, Sirmpilatze N, Dadarwal R, Handwerker R, Esser D, Wiegand K, Hagel C, Gocht A, König FB, Boretius S, Möbius W, Stadelmann C, Barrantes-Freer A. Lack of astrocytes hinders parenchymal oligodendrocyte precursor cells from reaching a myelinating state in osmolyte-induced demyelination. Acta Neuropathol Commun 2020; 8:224. [PMID: 33357244 PMCID: PMC7761156 DOI: 10.1186/s40478-020-01105-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 12/13/2020] [Indexed: 12/12/2022] Open
Abstract
Demyelinated lesions in human pons observed after osmotic shifts in serum have been referred to as central pontine myelinolysis (CPM). Astrocytic damage, which is prominent in neuroinflammatory diseases like neuromyelitis optica (NMO) and multiple sclerosis (MS), is considered the primary event during formation of CPM lesions. Although more data on the effects of astrocyte-derived factors on oligodendrocyte precursor cells (OPCs) and remyelination are emerging, still little is known about remyelination of lesions with primary astrocytic loss. In autopsy tissue from patients with CPM as well as in an experimental model, we were able to characterize OPC activation and differentiation. Injections of the thymidine-analogue BrdU traced the maturation of OPCs activated in early astrocyte-depleted lesions. We observed rapid activation of the parenchymal NG2+ OPC reservoir in experimental astrocyte-depleted demyelinated lesions, leading to extensive OPC proliferation. One week after lesion initiation, most parenchyma-derived OPCs expressed breast carcinoma amplified sequence-1 (BCAS1), indicating the transition into a pre-myelinating state. Cells derived from this early parenchymal response often presented a dysfunctional morphology with condensed cytoplasm and few extending processes, and were only sparsely detected among myelin-producing or mature oligodendrocytes. Correspondingly, early stages of human CPM lesions also showed reduced astrocyte numbers and non-myelinating BCAS1+ oligodendrocytes with dysfunctional morphology. In the rat model, neural stem cells (NSCs) located in the subventricular zone (SVZ) were activated while the lesion was already partially repopulated with OPCs, giving rise to nestin+ progenitors that generated oligodendroglial lineage cells in the lesion, which was successively repopulated with astrocytes and remyelinated. These nestin+ stem cell-derived progenitors were absent in human CPM cases, which may have contributed to the inefficient lesion repair. The present study points to the importance of astrocyte-oligodendrocyte interactions for remyelination, highlighting the necessity to further determine the impact of astrocyte dysfunction on remyelination inefficiency in demyelinating disorders including MS.
Collapse
|
14
|
Mozafari S, Deboux C, Laterza C, Ehrlich M, Kuhlmann T, Martino G, Baron-Van Evercooren A. Beneficial contribution of induced pluripotent stem cell-progeny to Connexin 47 dynamics during demyelination-remyelination. Glia 2020; 69:1094-1109. [PMID: 33301181 PMCID: PMC7984339 DOI: 10.1002/glia.23950] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/26/2020] [Accepted: 11/30/2020] [Indexed: 12/17/2022]
Abstract
Oligodendrocytes are extensively coupled to astrocytes, a phenomenon ensuring glial homeostasis and maintenance of central nervous system myelin. Molecular disruption of this communication occurs in demyelinating diseases such as multiple sclerosis. Less is known about the vulnerability and reconstruction of the panglial network during adult demyelination‐remyelination. Here, we took advantage of lysolcithin‐induced demyelination to investigate the expression dynamics of the oligodendrocyte specific connexin 47 (Cx47) and to some extent that of astrocyte Cx43, and whether this dynamic could be modulated by grafted induced pluripotent stem cell (iPSC)‐neural progeny. Our data show that disruption of Cx43‐Cx47 mediated hetero‐cellular gap‐junction intercellular communication following demyelination is larger in size than demyelination. Loss of Cx47 expression is timely rescued during remyelination and accelerated by the grafted neural precursors. Moreover, mouse and human iPSC‐derived oligodendrocytes express Cx47, which co‐labels with astrocyte Cx43, indicating their integration into the panglial network. These data suggest that in rodents, full lesion repair following transplantation occurs by panglial reconstruction in addition to remyelination. Targeting panglial elements by cell therapy or pharmacological compounds may help accelerating or stabilizing re/myelination in myelin disorders.
Collapse
Affiliation(s)
- Sabah Mozafari
- INSERM, U1127, Paris, France.,CNRS, UMR 7225, Paris, France.,Sorbonne Université UPMC Paris 06, UM-75, Paris, France.,ICM-GH Pitié-Salpêtrière, Paris, France
| | - Cyrille Deboux
- INSERM, U1127, Paris, France.,CNRS, UMR 7225, Paris, France.,Sorbonne Université UPMC Paris 06, UM-75, Paris, France.,ICM-GH Pitié-Salpêtrière, Paris, France
| | - Cecilia Laterza
- Institute of Experimental Neurology-DIBIT 2, Division of Neuroscience, IRCCS San Raffaele Hospital and Vita San Raffaele University, Milan, Italy.,Industrial Engineering Department, University of Padova, Padova, Italy
| | - Marc Ehrlich
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany.,Institute of Neuropathology, University Hospital Münster, Münster, Germany
| | - Tanja Kuhlmann
- Institute of Neuropathology, University Hospital Münster, Münster, Germany
| | - Gianvito Martino
- Institute of Experimental Neurology-DIBIT 2, Division of Neuroscience, IRCCS San Raffaele Hospital and Vita San Raffaele University, Milan, Italy
| | - Anne Baron-Van Evercooren
- INSERM, U1127, Paris, France.,CNRS, UMR 7225, Paris, France.,Sorbonne Université UPMC Paris 06, UM-75, Paris, France.,ICM-GH Pitié-Salpêtrière, Paris, France
| |
Collapse
|
15
|
Lapato AS, Thompson SM, Parra K, Tiwari-Woodruff SK. Astrocyte Glutamate Uptake and Water Homeostasis Are Dysregulated in the Hippocampus of Multiple Sclerosis Patients With Seizures. ASN Neuro 2020; 12:1759091420979604. [PMID: 33297722 PMCID: PMC7734542 DOI: 10.1177/1759091420979604] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
While seizure disorders are more prevalent among multiple sclerosis (MS) patients than the population overall and prognosticate earlier death & disability, their etiology remains unclear. Translational data indicate perturbed expression of astrocytic molecules contributing to homeostatic neuronal excitability, including water channels (AQP4) and synaptic glutamate transporters (EAAT2), in a mouse model of MS with seizures (MS+S). However, astrocytes in MS+S have not been examined. To assess the translational relevance of astrocyte dysfunction observed in a mouse model of MS+S, demyelinated lesion burden, astrogliosis, and astrocytic biomarkers (AQP4/EAAT2/ connexin-CX43) were evaluated by immunohistochemistry in postmortem hippocampi from MS & MS+S donors. Lesion burden was comparable in MS & MS+S cohorts, but astrogliosis was elevated in MS+S CA1 with a concomitant decrease in EAAT2 signal intensity. AQP4 signal declined in MS+S CA1 & CA3 with a loss of perivascular AQP4 in CA1. CX43 expression was increased in CA3. Together, these data suggest that hippocampal astrocytes from MS+S patients display regional differences in expression of molecules associated with glutamate buffering and water homeostasis that could exacerbate neuronal hyperexcitability. Importantly, mislocalization of CA1 perivascular AQP4 seen in MS+S is analogous to epileptic hippocampi without a history of MS, suggesting convergent pathophysiology. Furthermore, as neuropathology was concentrated in MS+S CA1, future study is warranted to determine the pathophysiology driving regional differences in glial function in the context of seizures during demyelinating disease.
Collapse
Affiliation(s)
- Andrew S Lapato
- Division of Biomedical Sciences, UCR School of Medicine, Riverside, California, United States.,Center for Glial-Neuronal Interaction, UCR School of Medicine, Riverside, California, United States
| | - Sarah M Thompson
- Division of Biomedical Sciences, UCR School of Medicine, Riverside, California, United States
| | - Karen Parra
- Division of Biomedical Sciences, UCR School of Medicine, Riverside, California, United States
| | - Seema K Tiwari-Woodruff
- Division of Biomedical Sciences, UCR School of Medicine, Riverside, California, United States.,Center for Glial-Neuronal Interaction, UCR School of Medicine, Riverside, California, United States.,Department of Neuroscience, UCR School of Medicine, Riverside, California, United States
| |
Collapse
|
16
|
Mozafari S, Starost L, Manot-Saillet B, Garcia-Diaz B, Xu YKT, Roussel D, Levy MJF, Ottoboni L, Kim KP, Schöler HR, Kennedy TE, Antel JP, Martino G, Angulo MC, Kuhlmann T, Baron-Van Evercooren A. Multiple sclerosis iPS-derived oligodendroglia conserve their properties to functionally interact with axons and glia in vivo. SCIENCE ADVANCES 2020; 6:6/49/eabc6983. [PMID: 33277253 PMCID: PMC7821889 DOI: 10.1126/sciadv.abc6983] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 10/22/2020] [Indexed: 05/04/2023]
Abstract
Remyelination failure in multiple sclerosis (MS) is associated with a migration/differentiation block of oligodendroglia. The reason for this block is highly debated. It could result from disease-related extrinsic or intrinsic regulators in oligodendroglial biology. To avoid confounding immune-mediated extrinsic effect, we used an immune-deficient mouse model to compare induced pluripotent stem cell-derived oligodendroglia from MS and healthy donors following engraftment in the developing CNS. We show that the MS-progeny behaves and differentiates into oligodendrocytes to the same extent as controls. They generate equal amounts of myelin, with bona fide nodes of Ranvier, and promote equal restoration of their host slow conduction. MS-progeny expressed oligodendrocyte- and astrocyte-specific connexins and established functional connections with donor and host glia. Thus, MS oligodendroglia, regardless of major immune manipulators, are intrinsically capable of myelination and making functional axo-glia/glia-glia connections, reinforcing the view that the MS oligodendrocyte differentiation block is not from major intrinsic oligodendroglial deficits.
Collapse
Affiliation(s)
- Sabah Mozafari
- INSERM, U1127, F-75013 Paris, France
- CNRS, UMR 7225, F-75013 Paris, France
- Sorbonne Université UPMC Paris 06, UM-75, F-75005, Paris, France
- ICM-GH Pitié-Salpêtrière, F-75013, Paris, France
| | - Laura Starost
- Institute of Neuropathology, University Hospital Münster, 48149 Münster, Germany
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Blandine Manot-Saillet
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM, Université de Paris, U1266, F-75014 Paris, France
- GHU PARIS Psychiatrie & Neurosciences, Paris, France
| | - Beatriz Garcia-Diaz
- INSERM, U1127, F-75013 Paris, France
- CNRS, UMR 7225, F-75013 Paris, France
- Sorbonne Université UPMC Paris 06, UM-75, F-75005, Paris, France
- ICM-GH Pitié-Salpêtrière, F-75013, Paris, France
| | - Yu Kang T Xu
- McGill Program in Neuroengineering, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Delphine Roussel
- INSERM, U1127, F-75013 Paris, France
- CNRS, UMR 7225, F-75013 Paris, France
- Sorbonne Université UPMC Paris 06, UM-75, F-75005, Paris, France
- ICM-GH Pitié-Salpêtrière, F-75013, Paris, France
| | - Marion J F Levy
- INSERM, U1127, F-75013 Paris, France
- CNRS, UMR 7225, F-75013 Paris, France
- Sorbonne Université UPMC Paris 06, UM-75, F-75005, Paris, France
- ICM-GH Pitié-Salpêtrière, F-75013, Paris, France
| | - Linda Ottoboni
- Institute of Experimental Neurology-DIBIT 2, Division of Neuroscience, IRCCS San Raffaele Hospital and Vita San Raffaele University, Milan, Italy
| | - Kee-Pyo Kim
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Hans R Schöler
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Timothy E Kennedy
- McGill Program in Neuroengineering, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Jack P Antel
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Gianvito Martino
- Institute of Experimental Neurology-DIBIT 2, Division of Neuroscience, IRCCS San Raffaele Hospital and Vita San Raffaele University, Milan, Italy
| | - Maria Cecilia Angulo
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM, Université de Paris, U1266, F-75014 Paris, France
- GHU PARIS Psychiatrie & Neurosciences, Paris, France
| | - Tanja Kuhlmann
- Institute of Neuropathology, University Hospital Münster, 48149 Münster, Germany
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Anne Baron-Van Evercooren
- INSERM, U1127, F-75013 Paris, France.
- CNRS, UMR 7225, F-75013 Paris, France
- Sorbonne Université UPMC Paris 06, UM-75, F-75005, Paris, France
- ICM-GH Pitié-Salpêtrière, F-75013, Paris, France
| |
Collapse
|
17
|
Mozafari S, Baron-Van Evercooren A. Human stem cell-derived oligodendrocytes: From humanized animal models to cell therapy in myelin diseases. Semin Cell Dev Biol 2020; 116:53-61. [PMID: 33082116 DOI: 10.1016/j.semcdb.2020.09.011] [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: 06/17/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 12/15/2022]
Abstract
Oligodendrocytes are main targets in demyelinating and dysmyelinating diseases of the central nervous system (CNS), but are also involved in accidental, neurodegenerative and psychiatric disorders. The underlying pathology of these diseases is not fully understood and treatments are still lacking. The recent discovery of the induced pluripotent stem cell (iPSC) technology has open the possibility to address the biology of human oligodendroglial cells both in the dish and in vivo via engraftment in animal models, and paves the way for the development of treatment for myelin disorders. In this review, we make a short overview of the different sources human oligodendroglial cells, and animal models available for pre-clinical cell therapy. We discuss the anatomical and functional benefit of grafted iPSC-progenitors over their brain counterparts, their use in disease modeling and the missing gaps that still prevent to study their biology in the most integrated way, and to translate iPSC-stem cell based therapy to the clinic.
Collapse
Affiliation(s)
- Sabah Mozafari
- Institut du Cerveau et de la Moelle Epinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM, U1127, CNRS, UMR 7225, Sorbonne Université UM75, F-75013 Paris, France; CNRS, UMR 7225, Paris, France; Sorbonne Universités, Université Pierre et MarieCurie Paris 06, UM-75, Paris, France
| | - Anne Baron-Van Evercooren
- Institut du Cerveau et de la Moelle Epinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM, U1127, CNRS, UMR 7225, Sorbonne Université UM75, F-75013 Paris, France; CNRS, UMR 7225, Paris, France; Sorbonne Universités, Université Pierre et MarieCurie Paris 06, UM-75, Paris, France.
| |
Collapse
|
18
|
Cohen-Salmon M, Slaoui L, Mazaré N, Gilbert A, Oudart M, Alvear-Perez R, Elorza-Vidal X, Chever O, Boulay AC. Astrocytes in the regulation of cerebrovascular functions. Glia 2020; 69:817-841. [PMID: 33058289 DOI: 10.1002/glia.23924] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 12/18/2022]
Abstract
Astrocytes are the most numerous type of neuroglia in the brain and have a predominant influence on the cerebrovascular system; they control perivascular homeostasis, the integrity of the blood-brain barrier, the dialogue with the peripheral immune system, the transfer of metabolites from the blood, and blood vessel contractility in response to neuronal activity. These regulatory processes occur in a specialized interface composed of perivascular astrocyte extensions that almost completely cover the cerebral blood vessels. Scientists have only recently started to study how this interface is formed and how it influences cerebrovascular functions. Here, we review the literature on the astrocytes' role in the regulation of the cerebrovascular system. We cover the anatomy and development of the gliovascular interface, the known gliovascular functions, and molecular factors, the latter's implication in certain pathophysiological situations, and recent cutting-edge experimental tools developed to examine the astrocytes' role at the vascular interface. Finally, we highlight some open questions in this field of research.
Collapse
Affiliation(s)
- Martine Cohen-Salmon
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Leila Slaoui
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Noémie Mazaré
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Alice Gilbert
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Marc Oudart
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Rodrigo Alvear-Perez
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Xabier Elorza-Vidal
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Oana Chever
- Normandie University, UNIROUEN, INSERM, DC2N, IRIB, Rouen, France
| | - Anne-Cécile Boulay
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| |
Collapse
|
19
|
Fumagalli A, Heuninck J, Pizzoccaro A, Moutin E, Koenen J, Séveno M, Durroux T, Junier MP, Schlecht-Louf G, Bachelerie F, Schütz D, Stumm R, Smit MJ, Guérineau NC, Chaumont-Dubel S, Marin P. The atypical chemokine receptor 3 interacts with Connexin 43 inhibiting astrocytic gap junctional intercellular communication. Nat Commun 2020; 11:4855. [PMID: 32978390 PMCID: PMC7519114 DOI: 10.1038/s41467-020-18634-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 09/02/2020] [Indexed: 12/12/2022] Open
Abstract
The atypical chemokine receptor 3 (ACKR3) plays a pivotal role in directing the migration of various cellular populations and its over-expression in tumors promotes cell proliferation and invasiveness. The intracellular signaling pathways transducing ACKR3-dependent effects remain poorly characterized, an issue we addressed by identifying the interactome of ACKR3. Here, we report that recombinant ACKR3 expressed in HEK293T cells recruits the gap junction protein Connexin 43 (Cx43). Cx43 and ACKR3 are co-expressed in mouse brain astrocytes and human glioblastoma cells and form a complex in embryonic mouse brain. Functional in vitro studies show enhanced ACKR3 interaction with Cx43 upon ACKR3 agonist stimulation. Furthermore, ACKR3 activation promotes β-arrestin2- and dynamin-dependent Cx43 internalization to inhibit gap junctional intercellular communication in primary astrocytes. These results demonstrate a functional link between ACKR3 and gap junctions that might be of pathophysiological relevance. The atypical chemokine receptor 3 (ACKR3) is known to regulate cell migration, but the underlying mechanisms are unclear. Here, the authors show, from an interactome analysis, ACKR3 association with the gap junction protein Connexin 43 in vivo and ACKR3-mediated inhibition of astrocyte gap junctional communication.
Collapse
Affiliation(s)
- Amos Fumagalli
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Joyce Heuninck
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Anne Pizzoccaro
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Enora Moutin
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Joyce Koenen
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, 92140, Clamart, France.,Amsterdam Institute for Molecules Medicines and Systems, Division of Medicinal Chemistry, Faculty of Sciences, VU University Amsterdam, 1081, HV, Amsterdam, The Netherlands
| | - Martial Séveno
- Biocampus Montpellier, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Thierry Durroux
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Marie-Pierre Junier
- CNRS UMR8246, Inserm U1130, Neuroscience Paris Seine-IBPS, Sorbonne Universités, Paris, France
| | - Géraldine Schlecht-Louf
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, 92140, Clamart, France
| | - Francoise Bachelerie
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, 92140, Clamart, France
| | - Dagmar Schütz
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747, Jena, Germany
| | - Ralf Stumm
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747, Jena, Germany
| | - Martine J Smit
- Amsterdam Institute for Molecules Medicines and Systems, Division of Medicinal Chemistry, Faculty of Sciences, VU University Amsterdam, 1081, HV, Amsterdam, The Netherlands
| | - Nathalie C Guérineau
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Séverine Chaumont-Dubel
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Philippe Marin
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France.
| |
Collapse
|
20
|
Megalencephalic Leukoencephalopathy with Subcortical Cysts Disease-Linked MLC1 Protein Favors Gap-Junction Intercellular Communication by Regulating Connexin 43 Trafficking in Astrocytes. Cells 2020; 9:cells9061425. [PMID: 32521795 PMCID: PMC7348769 DOI: 10.3390/cells9061425] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/29/2020] [Accepted: 06/03/2020] [Indexed: 01/06/2023] Open
Abstract
Astrocytes, the most numerous cells of the central nervous system, exert critical functions for brain homeostasis. To this purpose, astrocytes generate a highly interconnected intercellular network allowing rapid exchange of ions and metabolites through gap junctions, adjoined channels composed of hexamers of connexin (Cx) proteins, mainly Cx43. Functional alterations of Cxs and gap junctions have been observed in several neuroinflammatory/neurodegenerative diseases. In the rare leukodystrophy megalencephalic leukoencephalopathy with subcortical cysts (MLC), astrocytes show defective control of ion/fluid exchanges causing brain edema, fluid cysts, and astrocyte/myelin vacuolation. MLC is caused by mutations in MLC1, an astrocyte-specific protein of elusive function, and in GlialCAM, a MLC1 chaperon. Both proteins are highly expressed at perivascular astrocyte end-feet and astrocyte-astrocyte contacts where they interact with zonula occludens-1 (ZO-1) and Cx43 junctional proteins. To investigate the possible role of Cx43 in MLC pathogenesis, we studied Cx43 properties in astrocytoma cells overexpressing wild type (WT) MLC1 or MLC1 carrying pathological mutations. Using biochemical and electrophysiological techniques, we found that WT, but not mutated, MLC1 expression favors intercellular communication by inhibiting extracellular-signal-regulated kinase 1/2 (ERK1/2)-mediated Cx43 phosphorylation and increasing Cx43 gap-junction stability. These data indicate MLC1 regulation of Cx43 in astrocytes and Cx43 involvement in MLC pathogenesis, suggesting potential target pathways for therapeutic interventions.
Collapse
|
21
|
Xia CY, Xu JK, Pan CH, Lian WW, Yan Y, Ma BZ, He J, Zhang WK. Connexins in oligodendrocytes and astrocytes: Possible factors for demyelination in multiple sclerosis. Neurochem Int 2020; 136:104731. [PMID: 32201280 DOI: 10.1016/j.neuint.2020.104731] [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: 01/13/2020] [Revised: 03/18/2020] [Accepted: 03/18/2020] [Indexed: 12/13/2022]
Abstract
Increasing evidences support that glial connexins are involved in the demyelination pathology of multiple sclerosis (MS), a chronic inflammatory demyelinating disorder. Here, we review the data from patients with MS and animal models of MS that implicate connexins in demyelination. Connexins expressed in oligodendrocytes and astrocytes show diverse changes at the different phases of MS. Loss of oligodendrocyte or astrocyte connexins contributes to demyelination and exaggerates the pathology of MS. Channel-dependent and -independent connexins are involved in the pathology of demyelination, which is related with myelin integrity, metabolic homeostasis, the brain-blood barrier, the immune cell infiltration, and the inflammatory response. A comprehensive understanding of connexin function in demyelination may provide new therapeutic targets for MS.
Collapse
Affiliation(s)
- Cong-Yuan Xia
- Department of Pharmacy & Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, People's Republic of China
| | - Jie-Kun Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Chen-Hao Pan
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Wen-Wen Lian
- Department of Pharmacy & Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, People's Republic of China
| | - Yu Yan
- Department of Pharmacy & Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, People's Republic of China
| | - Bing-Zhi Ma
- Department of Pharmacy & Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, People's Republic of China
| | - Jun He
- Department of Pharmacy & Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, People's Republic of China.
| | - Wei-Ku Zhang
- Department of Pharmacy & Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, People's Republic of China.
| |
Collapse
|
22
|
Astrocyte and Oligodendrocyte Cross-Talk in the Central Nervous System. Cells 2020; 9:cells9030600. [PMID: 32138223 PMCID: PMC7140446 DOI: 10.3390/cells9030600] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 02/27/2020] [Accepted: 02/28/2020] [Indexed: 12/23/2022] Open
Abstract
Over the last decade knowledge of the role of astrocytes in central nervous system (CNS) neuroinflammatory diseases has changed dramatically. Rather than playing a merely passive role in response to damage it is clear that astrocytes actively maintain CNS homeostasis by influencing pH, ion and water balance, the plasticity of neurotransmitters and synapses, cerebral blood flow, and are important immune cells. During disease astrocytes become reactive and hypertrophic, a response that was long considered to be pathogenic. However, recent studies reveal that astrocytes also have a strong tissue regenerative role. Whilst most astrocyte research focuses on modulating neuronal function and synaptic transmission little is known about the cross-talk between astrocytes and oligodendrocytes, the myelinating cells of the CNS. This communication occurs via direct cell-cell contact as well as via secreted cytokines, chemokines, exosomes, and signalling molecules. Additionally, this cross-talk is important for glial development, triggering disease onset and progression, as well as stimulating regeneration and repair. Its critical role in homeostasis is most evident when this communication fails. Here, we review emerging evidence of astrocyte-oligodendrocyte communication in health and disease. Understanding the pathways involved in this cross-talk will reveal important insights into the pathogenesis and treatment of CNS diseases.
Collapse
|
23
|
Uncoupling of the Astrocyte Syncytium Differentially Affects AQP4 Isoforms. Cells 2020; 9:cells9020382. [PMID: 32046059 PMCID: PMC7072498 DOI: 10.3390/cells9020382] [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: 12/31/2019] [Revised: 01/29/2020] [Accepted: 02/04/2020] [Indexed: 11/25/2022] Open
Abstract
The water channel protein aquaporin-4 (AQP4) and the gap junction forming proteins connexin-43 (Cx43) and connexin-30 (Cx30) are astrocytic proteins critically involved in brain water and ion homeostasis. While AQP4 is mainly involved in water flux across the astrocytic endfeet membranes, astrocytic gap junctions provide syncytial coupling allowing intercellular exchange of water, ions, and other molecules. We have previously shown that mice with targeted deletion of Aqp4 display enhanced gap junctional coupling between astrocytes. Here, we investigate whether uncoupling of the astrocytic syncytium by deletion of the astrocytic connexins Cx43 and Cx30 affects AQP4 membrane localization and expression. By using quantitative immunogold cytochemistry, we show that deletion of astrocytic connexins leads to a substantial reduction of perivascular AQP4, concomitant with a down-regulation of total AQP4 protein and mRNA. Isoform expression analysis shows that while the level of the predominant AQP4 M23 isoform is reduced in Cx43/Cx30 double deficient hippocampal astrocytes, the levels of M1, and the alternative translation AQP4ex isoform protein levels are increased. These findings reveal a complex interdependence between AQP4 and connexins, which are both significantly involved in homeostatic functions and astrogliopathologies.
Collapse
|
24
|
Cocozzelli AG, White TW. Connexin 43 Mutations Lead to Increased Hemichannel Functionality in Skin Disease. Int J Mol Sci 2019; 20:ijms20246186. [PMID: 31817921 PMCID: PMC6940829 DOI: 10.3390/ijms20246186] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/29/2019] [Accepted: 12/04/2019] [Indexed: 12/22/2022] Open
Abstract
Gap junctional channels are specialized components of the cellular membrane that allow the intercellular passage of small metabolites, ions, and second messengers to maintain homeostasis. They are comprised of members of the connexin gene family that encode a wide array of proteins that are expressed in nearly every tissue type. Cx43 is perceived to be the most broadly expressed connexin in humans, with several genetic skin diseases being linked to Cx43 mutations specifically. These mutations, in large, produce a gain of functional hemichannels that contribute to the phenotypes of Erythrokeratoderma Variabilis et Progressiva (EKVP), Palmoplantar Keratodemra Congenital Alopecia-1 (PPKCA1), and others that produce large conductance and increased permselectivity in otherwise quiescent structures. Gaining functional hemichannels can have adverse effects in the skin, inducing apoptosis via Ca2+ overload or increased ATP permeability. Here, we review the link between Cx43 and skin disease. We aim to provide insight into the mechanisms regulating the normal and pathophysiological gating of these essential proteins, as well as address current therapeutic strategies. We also demonstrate that transient transfection of neuro-2a (N2a) cells with mutant Cx43 cDNA resulted in increased hemichannel activity compared to wild-type Cx43 and untransfected cells, which is consistent with other studies in the current literature.
Collapse
|
25
|
Astrocytes in multiple sclerosis and experimental autoimmune encephalomyelitis: Star-shaped cells illuminating the darkness of CNS autoimmunity. Brain Behav Immun 2019; 80:10-24. [PMID: 31125711 DOI: 10.1016/j.bbi.2019.05.029] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/16/2019] [Accepted: 05/20/2019] [Indexed: 12/18/2022] Open
Abstract
Neuropathology in the human autoimmune disease multiple sclerosis (MS) is considered to be mediated by autoreactive leukocytes, such as T cells, B cells, and macrophages. However, the inflammation and tissue damage in MS and its animal model experimental autoimmune encephalomyelitis (EAE) is also critically regulated by astrocytes, the most abundant cell population in the central nervous system (CNS). Under physiological conditions, astrocytes are integral to the development and function of the CNS, whereas in CNS autoimmunity, astrocytes influence the pathogenesis, progression, and recovery of the diseases. In this review, we summarize recent advances in astrocytic functions in the context of MS and EAE, which are categorized into two opposite aspects, one being detrimental and the other beneficial. Inhibition of the detrimental functions and/or enhancement of the beneficial functions of astrocytes might be favorable for the treatment of MS.
Collapse
|
26
|
Wang A, Xu C. The role of connexin43 in neuropathic pain induced by spinal cord injury. Acta Biochim Biophys Sin (Shanghai) 2019; 51:555-561. [PMID: 31056639 DOI: 10.1093/abbs/gmz038] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Indexed: 12/12/2022] Open
Abstract
Neuropathic pain is caused by the damage or dysfunction of the nervous system. In many neuropathic pain models, there is an increase in the number of gap junction (GJ) channels, especially the upregulation of the expression of connexin43 (Cx43), leading to the secretion of various types of cytokines and involvement in the formation of neuropathic pain. GJs are widely distributed in mammalian organs and tissues, and Cx43 is the most abundant connexin (Cx) in mammals. Astrocytes are the most abundant glial cell type in the central nervous system (CNS), which mainly express Cx43. More importantly, GJs play an important role in regulating cell metabolism, signaling, and function. Many existing literatures showed that Cx43 plays an important role in the nervous system, especially in the CNS under normal and pathological conditions. However, many internal mechanisms have not yet been thoroughly explored. In this review, we summarized the current understanding of the role and association of Cx and pannexin channels in neuropathic pain, especially after spinal cord injury, as well as some of our own insights and thoughts which suggest that Cx43 may become an emerging therapeutic target for future neuropathic pain, bringing new hope to patients.
Collapse
Affiliation(s)
- Anhui Wang
- Department of Physiology, Basic Medical College of Nanchang University, Nanchang, China
| | - Changshui Xu
- Department of Physiology, Basic Medical College of Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Autonomic Nervous Function and Disease, Nanchang, China
| |
Collapse
|
27
|
Rothbauer M, Charwat V, Bachmann B, Sticker D, Novak R, Wanzenböck H, Mathies RA, Ertl P. Monitoring transient cell-to-cell interactions in a multi-layered and multi-functional allergy-on-a-chip system. LAB ON A CHIP 2019; 19:1916-1921. [PMID: 31070645 DOI: 10.1039/c9lc00108e] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
We have developed a highly integrated lab-on-a-chip containing embedded electrical microsensors, μdegassers and pneumatically-actuated micropumps to monitor allergic hypersensitivity. Rapid antigen-mediated histamine release (e.g. s to min) and resulting muscle contraction (<30 min) is detected by connecting an immune compartment containing sensitized basophile cells to a vascular co-culture model.
Collapse
Affiliation(s)
- Mario Rothbauer
- Faculty of Technical Chemistry, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria. and Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Verena Charwat
- Department of Biotechnology, University of Agricultural Resources and Life Sciences, Muthgasse 18, 1090 Vienna, Austria
| | - Barbara Bachmann
- Faculty of Technical Chemistry, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria. and Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria and AUVA Research Centre, Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, 1200 Vienna, Austria
| | - Drago Sticker
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Richard Novak
- Department of Chemistry, University of California at Berkeley, Lewis Hall, Berkeley, California, USA
| | - Heinz Wanzenböck
- Faculty of Electrical Engineering, Vienna University of Technology, Gußhausstr. 25-25a, 1040 Vienna, Austria
| | - Richard A Mathies
- Department of Chemistry, University of California at Berkeley, Lewis Hall, Berkeley, California, USA
| | - Peter Ertl
- Faculty of Technical Chemistry, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria. and Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| |
Collapse
|
28
|
The contribution of astrocytes to the neuroinflammatory response in multiple sclerosis and experimental autoimmune encephalomyelitis. Acta Neuropathol 2019; 137:757-783. [PMID: 30847559 DOI: 10.1007/s00401-019-01980-7] [Citation(s) in RCA: 163] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 02/21/2019] [Accepted: 02/23/2019] [Indexed: 02/06/2023]
Abstract
Neuroinflammation is the coordinated response of the central nervous system (CNS) to threats to its integrity posed by a variety of conditions, including autoimmunity, pathogens and trauma. Activated astrocytes, in concert with other cellular elements of the CNS and immune system, are important players in the modulation of the neuroinflammatory response. During neurological disease, they produce and respond to cellular signals that often lead to dichotomous processes, which can promote further damage or contribute to repair. This occurs also in multiple sclerosis (MS), where astrocytes are now recognized as key components of its immunopathology. Evidence supporting this role has emerged not only from studies in MS patients, but also from animal models, among which the experimental autoimmune encephalomyelitis (EAE) model has proved especially instrumental. Based on this premise, the purpose of the present review is to summarize the current knowledge of astrocyte behavior in MS and EAE. Following a brief description of the pathological characteristics of the two diseases and the main functional roles of astrocytes in CNS physiology, we will delve into the specific responses of this cell population, analyzing MS and EAE in parallel. We will define the temporal and anatomical profile of astroglial activation, then focus on key processes they participate in. These include: (1) production and response to soluble mediators (e.g., cytokines and chemokines), (2) regulation of oxidative stress, and (3) maintenance of BBB integrity and function. Finally, we will review the state of the art on the available methods to measure astroglial activation in vivo in MS patients, and how this could be exploited to optimize diagnosis, prognosis and treatment decisions. Ultimately, we believe that integrating the knowledge obtained from studies in MS and EAE may help not only better understand the pathophysiology of MS, but also uncover new signals to be targeted for therapeutic intervention.
Collapse
|
29
|
Amini Harandi A, Esfandani A, Pakdaman H, Abbasi M, Sahraian MA. Balo’s concentric sclerosis: an update and comprehensive literature review. Rev Neurosci 2018; 29:873-882. [DOI: 10.1515/revneuro-2017-0096] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 03/10/2018] [Indexed: 12/17/2022]
Abstract
Abstract
Balo’s concentric sclerosis (BCS) is considered a variant of multiple sclerosis characterized by concentric lamella of alternating demyelinated and partially myelinated tissues. It is a rare and a relatively acute condition. Attacks may proceed rapidly over weeks or months, typically without remission, like Marburg’s variant, resulting in death or severe disability. However, the majority of cases have a more benign, self-limiting course with spontaneous remission. Magnetic resonance imaging is a primary imaging modality in the diagnosis of BCS. Treatment with intense immunosuppression may be indicated in patients with more aggressive form. New reports reveal more evidence regarding the pathophysiology and treatment strategies.
Collapse
Affiliation(s)
- Ali Amini Harandi
- Brain Mapping Research Center , Shahid Beheshti University of Medical Sciences , Tehran 1333635445 , Iran
| | - Akram Esfandani
- Brain Mapping Research Center , Shahid Beheshti University of Medical Sciences , Tehran 1333635445 , Iran
| | - Hossein Pakdaman
- Brain Mapping Research Center , Shahid Beheshti University of Medical Sciences , Tehran 1333635445 , Iran
| | - Mehdi Abbasi
- Brain Mapping Research Center , Shahid Beheshti University of Medical Sciences , Tehran 1333635445 , Iran
| | - Mohammad Ali Sahraian
- MS Research Center, Neuroscience Institute , Tehran University of Medical Sciences , Tehran 1136746911 , Iran
| |
Collapse
|
30
|
Correale J, Ysrraelit MC, Benarroch EE. Metabolic coupling of axons and glial cells. Neurology 2018; 90:737-744. [DOI: 10.1212/wnl.0000000000005339] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
31
|
Jullienne A, Fukuda AM, Ichkova A, Nishiyama N, Aussudre J, Obenaus A, Badaut J. Modulating the water channel AQP4 alters miRNA expression, astrocyte connectivity and water diffusion in the rodent brain. Sci Rep 2018; 8:4186. [PMID: 29520011 PMCID: PMC5843607 DOI: 10.1038/s41598-018-22268-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 02/20/2018] [Indexed: 12/27/2022] Open
Abstract
Aquaporins (AQPs) facilitate water diffusion through the plasma membrane. Brain aquaporin-4 (AQP4) is present in astrocytes and has critical roles in normal and disease physiology. We previously showed that a 24.9% decrease in AQP4 expression after in vivo silencing resulted in a 45.8% decrease in tissue water mobility as interpreted from magnetic resonance imaging apparent diffusion coefficients (ADC). Similar to previous in vitro studies we show decreased expression of the gap junction protein connexin 43 (Cx43) in vivo after intracortical injection of siAQP4 in the rat. Moreover, siAQP4 induced a loss of dye-coupling between astrocytes in vitro, further demonstrating its effect on gap junctions. In contrast, silencing of Cx43 did not alter the level of AQP4 or water mobility (ADC) in the brain. We hypothesized that siAQP4 has off-target effects on Cx43 expression via modification of miRNA expression. The decreased expression of Cx43 in siAQP4-treated animals was associated with up-regulation of miR224, which is known to target AQP4 and Cx43 expression. This could be one potential molecular mechanism responsible for the effect of siAQP4 on Cx43 expression, and the resultant decrease in astrocyte connectivity and dramatic effects on ADC values and water mobility.
Collapse
Affiliation(s)
- Amandine Jullienne
- Basic Sciences Department, Loma Linda University, Loma Linda, CA, 92354, USA
- Department of Physiology, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Andrew M Fukuda
- Basic Sciences Department, Loma Linda University, Loma Linda, CA, 92354, USA
- Department of Physiology, Loma Linda University, Loma Linda, CA, 92354, USA
| | | | - Nina Nishiyama
- Department of Physiology, Loma Linda University, Loma Linda, CA, 92354, USA
| | | | - André Obenaus
- Basic Sciences Department, Loma Linda University, Loma Linda, CA, 92354, USA
- Department of Pediatrics, University of California Irvine, Irvine, CA, 92697, USA
| | - Jérôme Badaut
- Basic Sciences Department, Loma Linda University, Loma Linda, CA, 92354, USA.
- Department of Physiology, Loma Linda University, Loma Linda, CA, 92354, USA.
- CNRS-UMR 5287, University of Bordeaux, 33076, Bordeaux, France.
| |
Collapse
|
32
|
Affiliation(s)
- Ryo Yamasaki
- Department of Neurology; Neurological Institute; Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| |
Collapse
|
33
|
Philips T, Rothstein JD. Oligodendroglia: metabolic supporters of neurons. J Clin Invest 2017; 127:3271-3280. [PMID: 28862639 DOI: 10.1172/jci90610] [Citation(s) in RCA: 222] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Oligodendrocytes are glial cells that populate the entire CNS after they have differentiated from oligodendrocyte progenitor cells. From birth onward, oligodendrocytes initiate wrapping of neuronal axons with a multilamellar lipid structure called myelin. Apart from their well-established function in action potential propagation, more recent data indicate that oligodendrocytes are essential for providing metabolic support to neurons. Oligodendrocytes transfer energy metabolites to neurons through cytoplasmic "myelinic" channels and monocarboxylate transporters, which allow for the fast delivery of short-carbon-chain energy metabolites like pyruvate and lactate to neurons. These substrates are metabolized and contribute to ATP synthesis in neurons. This Review will discuss our current understanding of this metabolic supportive function of oligodendrocytes and its potential impact in human neurodegenerative disease and related animal models.
Collapse
|
34
|
Olabarria M, Goldman JE. Disorders of Astrocytes: Alexander Disease as a Model. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2017; 12:131-152. [PMID: 28135564 DOI: 10.1146/annurev-pathol-052016-100218] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Astrocytes undergo important phenotypic changes in many neurological disorders, including strokes, trauma, inflammatory diseases, infectious diseases, and neurodegenerative diseases. We have been studying the astrocytes of Alexander disease (AxD), which is caused by heterozygous mutations in the GFAP gene, which is the gene that encodes the major astrocyte intermediate filament protein. AxD is a primary astrocyte disease because GFAP expression is specific to astrocytes in the central nervous system (CNS). The accumulation of extremely large amounts of GFAP causes many molecular changes in astrocytes, including proteasome inhibition, stress kinase activation, mechanistic target of rapamycin (mTOR) activation, loss of glutamate and potassium buffering capacity, loss of astrocyte coupling, and changes in cell morphology. Many of these changes appear to be common to astrocyte reactions in other neurological disorders. Using AxD to illuminate common mechanisms, we discuss the molecular pathology of AxD astrocytes and compare that to astrocyte pathology in other disorders.
Collapse
Affiliation(s)
- Markel Olabarria
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032; ,
| | - James E Goldman
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032; ,
| |
Collapse
|
35
|
Lapato AS, Tiwari-Woodruff SK. Connexins and pannexins: At the junction of neuro-glial homeostasis & disease. J Neurosci Res 2017; 96:31-44. [PMID: 28580666 DOI: 10.1002/jnr.24088] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 04/08/2017] [Accepted: 05/01/2017] [Indexed: 12/15/2022]
Abstract
In the central nervous system (CNS), connexin (Cx)s and pannexin (Panx)s are an integral component of homeostatic neuronal excitability and synaptic plasticity. Neuronal Cx gap junctions form electrical synapses across biochemically similar GABAergic networks, allowing rapid and extensive inhibition in response to principle neuron excitation. Glial Cx gap junctions link astrocytes and oligodendrocytes in the pan-glial network that is responsible for removing excitotoxic ions and metabolites. In addition, glial gap junctions help constrain excessive excitatory activity in neurons and facilitate astrocyte Ca2+ slow wave propagation. Panxs do not form gap junctions in vivo, but Panx hemichannels participate in autocrine and paracrine gliotransmission, alongside Cx hemichannels. ATP and other gliotransmitters released by Cx and Panx hemichannels maintain physiologic glutamatergic tone by strengthening synapses and mitigating aberrant high frequency bursting. Under pathological depolarizing and inflammatory conditions, gap junctions and hemichannels become dysregulated, resulting in excessive neuronal firing and seizure. In this review, we present known contributions of Cxs and Panxs to physiologic neuronal excitation and explore how the disruption of gap junctions and hemichannels lead to abnormal glutamatergic transmission, purinergic signaling, and seizures.
Collapse
Affiliation(s)
- Andrew S Lapato
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, 92521.,Center for Glial-Neuronal Interactions, University of California Riverside, Riverside, CA, 92521
| | - Seema K Tiwari-Woodruff
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, 92521.,Center for Glial-Neuronal Interactions, University of California Riverside, Riverside, CA, 92521.,Neuroscience Graduate Program, University of California Riverside, Riverside, CA, 92521
| |
Collapse
|
36
|
Th1 cells downregulate connexin 43 gap junctions in astrocytes via microglial activation. Sci Rep 2016; 6:38387. [PMID: 27929069 PMCID: PMC5143974 DOI: 10.1038/srep38387] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 11/08/2016] [Indexed: 11/28/2022] Open
Abstract
We previously reported early and extensive loss of astrocytic connexin 43 (Cx43) in acute demyelinating lesions of multiple sclerosis (MS) patients. Because it is widely accepted that autoimmune T cells initiate MS lesions, we hypothesized that infiltrating T cells affect Cx43 expression in astrocytes, which contributes to MS lesion formation. Primary mixed glial cell cultures were prepared from newborn mouse brains, and microglia were isolated by anti-CD11b antibody-conjugated magnetic beads. Next, we prepared astrocyte-rich cultures and astrocyte/microglia-mixed cultures. Treatment of primary mixed glial cell cultures with interferon (IFN) γ, interleukin (IL)-4, or IL-17 showed that only IFNγ or IL-17 at high concentrations reduced Cx43 protein levels. Upon treatment of astrocyte-rich cultures and astrocyte/microglia-mixed cultures with IFNγ, Cx43 mRNA/protein levels and the function of gap junctions were reduced only in astrocyte/microglia-mixed cultures. IFNγ-treated microglia-conditioned media and IL-1β, which was markedly increased in IFNγ-treated microglia-conditioned media, reduced Cx43 protein levels in astrocyte-rich cultures. Finally, we confirmed that Th1 cell-conditioned medium decreased Cx43 protein levels in mixed glial cell cultures. These findings suggest that Th1 cell-derived IFNγ activates microglia to release IL-1β that reduces Cx43 gap junctions in astrocytes. Thus, Th1-dominant inflammatory states disrupt astrocytic intercellular communication and may exacerbate MS.
Collapse
|
37
|
Ornelas IM, McLane LE, Saliu A, Evangelou AV, Khandker L, Wood TL. Heterogeneity in oligodendroglia: Is it relevant to mouse models and human disease? J Neurosci Res 2016; 94:1421-1433. [PMID: 27557736 PMCID: PMC5513674 DOI: 10.1002/jnr.23900] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Revised: 07/29/2016] [Accepted: 08/04/2016] [Indexed: 01/09/2023]
Abstract
There are many lines of evidence indicating that oligodendrocyte progenitor cells and oligodendrocyte populations in the central nervous system (CNS) are heterogeneous based on their developmental origins as well as from morphological and molecular criteria. Whether these distinctions reflect functional heterogeneity is less clear and has been the subject of considerable debate. Recent findings, particularly from knockout mouse models, have provided new evidence for regional variations in myelination phenotypes, particularly between brain and spinal cord. These data raise the possibility that oligodendrocytes in these regions have different functional capacities and/or ability to compensate for loss of a specific gene. The goal of this review is to briefly revisit the evidence for oligodendrocyte heterogeneity and then to present data from transgenic and demyelinating mouse models suggesting functional heterogeneity in myelination, demyelination, and remyelination in the CNS and, finally, to discuss the implications of these findings for human diseases. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Isis M Ornelas
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Lauren E McLane
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Aminat Saliu
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Angelina V Evangelou
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Luipa Khandker
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Teresa L Wood
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey.
| |
Collapse
|
38
|
Filous AR, Silver J. "Targeting astrocytes in CNS injury and disease: A translational research approach". Prog Neurobiol 2016; 144:173-87. [PMID: 27026202 PMCID: PMC5035184 DOI: 10.1016/j.pneurobio.2016.03.009] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 02/03/2016] [Accepted: 03/03/2016] [Indexed: 12/31/2022]
Abstract
Astrocytes are a major constituent of the central nervous system. These glia play a major role in regulating blood-brain barrier function, the formation and maintenance of synapses, glutamate uptake, and trophic support for surrounding neurons and glia. Therefore, maintaining the proper functioning of these cells is crucial to survival. Astrocyte defects are associated with a wide variety of neuropathological insults, ranging from neurodegenerative diseases to gliomas. Additionally, injury to the CNS causes drastic changes to astrocytes, often leading to a phenomenon known as reactive astrogliosis. This process is important for protecting the surrounding healthy tissue from the spread of injury, while it also inhibits axonal regeneration and plasticity. Here, we discuss the important roles of astrocytes after injury and in disease, as well as potential therapeutic approaches to restore proper astrocyte functioning.
Collapse
Affiliation(s)
- Angela R Filous
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 216-368-4615, United States.
| | - Jerry Silver
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 216-368-4615, United States.
| |
Collapse
|
39
|
Boulay AC, Cisternino S, Cohen-Salmon M. Immunoregulation at the gliovascular unit in the healthy brain: A focus on Connexin 43. Brain Behav Immun 2016; 56:1-9. [PMID: 26674996 DOI: 10.1016/j.bbi.2015.11.017] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 10/26/2015] [Accepted: 11/28/2015] [Indexed: 01/18/2023] Open
Abstract
In the brain, immune cell infiltration is normally kept at a very low level and a unique microenvironment strictly restricts immune reactions and inflammation. Even in such quiescent environment, a constant immune surveillance is at work allowing the brain to rapidly react to threats. To date, knowledge about the factors regulating the brain-immune system interrelationship in healthy conditions remains elusive. Interestingly, astrocytes, the most abundant glial cells in the brain, may participate in many aspects of this unique homeostasis, in particular due to their close interaction with the brain vascular system and expression of a specific molecular repertoire. Indeed, astrocytes maintain the blood-brain barrier (BBB) integrity, interact with immune cells, and participate in the regulation of intracerebral liquid movements. We recently showed that Connexin 43 (Cx43), a gap junction protein highly expressed by astrocytes at the BBB interface, is an immunoregulating factor. The absence of astroglial Cx43 leads to a transient endothelial activation, a continuous immune recruitment as well as the development of a specific humoral autoimmune response against the von Willebrand factor A domain-containing protein 5a, an extracellular matrix protein expressed by astrocytes. In this review, we propose to gather current knowledge on how astrocytes may influence the immune system in the healthy brain, focusing on their roles at the gliovascular interface. We will also consider pathological situations involving astrocyte-specific autoimmunities. Finally, we will discuss the specific role of astroglial Cx43 and the physiological consequences of immune regulations taking place on inflammation, cognition and behavior in the absence of Cx43.
Collapse
Affiliation(s)
- Anne-Cécile Boulay
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Centre National de la Recherche Scientifique CNRS, Unité Mixte de Recherche 7241, Institut National de la Santé et de la Recherche Médicale INSERM, U1050, Neuroglial Interactions in Cerebral Physiopathology, 75231 Paris Cedex 05, France; University Pierre et Marie Curie, ED, N°158, 75005 Paris, France; MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, 75005 Paris, France
| | - Salvatore Cisternino
- Variabilité de réponse aux psychotropes, INSERM, U1144, Paris F-75006, France; Université Paris Descartes, Faculté de Pharmacie, UMR-S 1144, 75006 Paris, France; Université Paris Diderot, UMR-S 1144, 75013 Paris, France
| | - Martine Cohen-Salmon
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Centre National de la Recherche Scientifique CNRS, Unité Mixte de Recherche 7241, Institut National de la Santé et de la Recherche Médicale INSERM, U1050, Neuroglial Interactions in Cerebral Physiopathology, 75231 Paris Cedex 05, France; University Pierre et Marie Curie, ED, N°158, 75005 Paris, France; MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, 75005 Paris, France.
| |
Collapse
|
40
|
Kim Y, Davidson JO, Gunn KC, Phillips AR, Green CR, Gunn AJ. Role of Hemichannels in CNS Inflammation and the Inflammasome Pathway. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2016; 104:1-37. [DOI: 10.1016/bs.apcsb.2015.12.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|