101
|
Liu Y, Fan H, Li X, Liu J, Qu X, Wu X, Liu M, Liu Z, Yao R. Trpv4 regulates Nlrp3 inflammasome via SIRT1/PGC-1α pathway in a cuprizone-induced mouse model of demyelination. Exp Neurol 2020; 337:113593. [PMID: 33387462 DOI: 10.1016/j.expneurol.2020.113593] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 11/28/2020] [Accepted: 12/27/2020] [Indexed: 12/29/2022]
Abstract
Increasing evidence has demonstrated that the Nod-like receptor pyrin domain containing 3 (Nlrp3) inflammasome overactivated during demyelinating disorders. It has been implicated that transient receptor potential type 4 (Trpv4) is regarded as a polymodal ionotropic receptor that plays an important role in a multitude of pathological conditions, including inflammation. The aim of this study was to investigate whether the Trpv4 channel regulates Nlrp3 inflammasome in the corpus callosum of mice with demyelination. Our results showed that CPZ treatment significantly increased the expression of Trpv4, activated Nlrp3 inflammasome, reduced peroxisome proliferator-activated receptor gamma coactivator 1α (PGC-1α) and decreased mitochondrial function. siRNA-mediated Nlrp3 knockdown inhibited glial activation and alleviated demyelination. Whereas knockdown of Trpv4 by siRNA markedly ameliorated Nlrp3 inflammasome activation and restored mitochondrial function as well as reducing the level of reactive oxygen species (ROS). Meanwhile, glial activation, demyelination and behavioral impairment induced by CPZ were also alleviated by siRNA-mediated Trpv4 knockdown. Furthermore, immunoprecipitation and use of a lysine acetylation assay showed that Sirtuin1 (SIRT1) mediated the PGC-1α deacetylation, which is involved in Nlrp3 inflammasome activation. These findings suggest that Trpv4 regulates mitochondrial function through the SIRT1/PGC-1α pathway, which further trigger Nlrp3 inflammasome activation in the CPZ-induced demyelination in mice.
Collapse
Affiliation(s)
- Yanan Liu
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou 221004, PR China; Department of Human Anatomy, Xuzhou Medical University, Xuzhou 221004, PR China
| | - Hongbin Fan
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou 221004, PR China; Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, PR China
| | - Xinyu Li
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou 221004, PR China; Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, PR China
| | - Jing Liu
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou 221004, PR China; Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, PR China
| | - Xuebin Qu
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou 221004, PR China
| | - Xiuxiang Wu
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou 221004, PR China
| | - Meiying Liu
- Department of Human Anatomy, Xuzhou Medical University, Xuzhou 221004, PR China
| | - Zhian Liu
- Department of Human Anatomy, Xuzhou Medical University, Xuzhou 221004, PR China
| | - Ruiqin Yao
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou 221004, PR China.
| |
Collapse
|
102
|
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
|
103
|
Werkman IL, Dubbelaar ML, van der Vlies P, de Boer-Bergsma JJ, Eggen BJL, Baron W. Transcriptional heterogeneity between primary adult grey and white matter astrocytes underlie differences in modulation of in vitro myelination. J Neuroinflammation 2020; 17:373. [PMID: 33308248 PMCID: PMC7733297 DOI: 10.1186/s12974-020-02045-3] [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: 08/23/2020] [Accepted: 11/25/2020] [Indexed: 12/31/2022] Open
Abstract
Background Multiple sclerosis (MS) is an inflammation-mediated demyelinating disease of the central nervous system that eventually results in secondary axonal degeneration due to remyelination failure. Successful remyelination is orchestrated by astrocytes (ASTRs) and requires sequential activation, recruitment, and maturation of oligodendrocyte progenitor cells (OPCs). In both MS and experimental models, remyelination is more robust in grey matter (GM) than white matter (WM), which is likely related to local differences between GM and WM lesions. Here, we investigated whether adult gmASTRs and wmASTRs per se and in response to MS relevant Toll-like receptor (TLR) activation differently modulate myelination. Methods Differences in modulation of myelination between adult gmASTRs and wmASTRs were examined using an in vitro myelinating system that relies on a feeding layer of ASTRs. Transcriptional profiling and weighted gene co-expression network analysis were used to analyze differentially expressed genes and gene networks. Potential differential modulation of OPC proliferation and maturation by untreated adult gmASTRs and wmASTRs and in response to TLR3 and TLR4 agonists were assessed. Results Our data reveal that adult wmASTRs are less supportive to in vitro myelination than gmASTRs. WmASTRs more abundantly express reactive ASTR genes and genes of a neurotoxic subtype of ASTRs, while gmASTRs have more neuro-reparative transcripts. We identified a gene network module containing cholesterol biosynthesis enzyme genes that positively correlated with gmASTRs, and a network module containing extracellular matrix-related genes that positively correlated with wmASTRs. Adult wmASTRs and gmASTRs responding to TLR3 agonist Poly(I:C) distinctly modulate OPC behavior, while exposure to TLR4 agonist LPS of both gmASTRs and wmASTRs results in a prominent decrease in myelin membrane formation. Conclusions Primary adult gmASTRs and wmASTRs are heterogeneous at the transcriptional level, differed in their support of in vitro myelination, and their pre-existing phenotype determined TLR3 agonist responses. These findings point to a role of ASTR heterogeneity in regional differences in remyelination efficiency between GM and WM lesions. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-020-02045-3.
Collapse
Affiliation(s)
- Inge L Werkman
- Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713, AV, Groningen, the Netherlands
| | - Marissa L Dubbelaar
- Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713, AV, Groningen, the Netherlands
| | - Pieter van der Vlies
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Jelkje J de Boer-Bergsma
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Bart J L Eggen
- Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713, AV, Groningen, the Netherlands
| | - Wia Baron
- Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713, AV, Groningen, the Netherlands.
| |
Collapse
|
104
|
das Neves SP, Sousa JC, Sousa N, Cerqueira JJ, Marques F. Altered astrocytic function in experimental neuroinflammation and multiple sclerosis. Glia 2020; 69:1341-1368. [PMID: 33247866 DOI: 10.1002/glia.23940] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/14/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) that affects about 2.5 million people worldwide. In MS, the patients' immune system starts to attack the myelin sheath, leading to demyelination, neurodegeneration, and, ultimately, loss of vital neurological functions such as walking. There is currently no cure for MS and the available treatments only slow the initial phases of the disease. The later-disease mechanisms are poorly understood and do not directly correlate with the activity of immune system cells, the main target of the available treatments. Instead, evidence suggests that disease progression and disability are better correlated with the maintenance of a persistent low-grade inflammation inside the CNS, driven by local glial cells, like astrocytes and microglia. Depending on the context, astrocytes can (a) exacerbate inflammation or (b) promote immunosuppression and tissue repair. In this review, we will address the present knowledge that exists regarding the role of astrocytes in MS and experimental animal models of the disease.
Collapse
Affiliation(s)
- Sofia Pereira das Neves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga, Portugal
| | - João Carlos Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga, Portugal.,Clinical Academic Center, Braga, Portugal
| | - João José Cerqueira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga, Portugal.,Clinical Academic Center, Braga, Portugal
| | - Fernanda Marques
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga, Portugal
| |
Collapse
|
105
|
Bozic I, Savic D, Lavrnja I. Astrocyte phenotypes: Emphasis on potential markers in neuroinflammation. Histol Histopathol 2020; 36:267-290. [PMID: 33226087 DOI: 10.14670/hh-18-284] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Astrocytes, the most abundant glial cells in the central nervous system (CNS), have numerous integral roles in all CNS functions. They are essential for synaptic transmission and support neurons by providing metabolic substrates, secreting growth factors and regulating extracellular concentrations of ions and neurotransmitters. Astrocytes respond to CNS insults through reactive astrogliosis, in which they go through many functional and molecular changes. In neuroinflammatory conditions reactive astrocytes exert both beneficial and detrimental functions, depending on the context and heterogeneity of astrocytic populations. In this review we profile astrocytic diversity in the context of neuroinflammation; with a specific focus on multiple sclerosis (MS) and its best-described animal model experimental autoimmune encephalomyelitis (EAE). We characterize two main subtypes, protoplasmic and fibrous astrocytes and describe the role of intermediate filaments in the physiology and pathology of these cells. Additionally, we outline a variety of markers that are emerging as important in investigating astrocytic biology in both physiological conditions and neuroinflammation.
Collapse
Affiliation(s)
- Iva Bozic
- Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Danijela Savic
- Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Irena Lavrnja
- Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
106
|
Misrielal C, Mauthe M, Reggiori F, Eggen BJL. Autophagy in Multiple Sclerosis: Two Sides of the Same Coin. Front Cell Neurosci 2020; 14:603710. [PMID: 33328897 PMCID: PMC7714924 DOI: 10.3389/fncel.2020.603710] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 10/26/2020] [Indexed: 01/08/2023] Open
Abstract
Multiple sclerosis (MS) is a complex auto-immune disorder of the central nervous system (CNS) that involves a range of CNS and immune cells. MS is characterized by chronic neuroinflammation, demyelination, and neuronal loss, but the molecular causes of this disease remain poorly understood. One cellular process that could provide insight into MS pathophysiology and also be a possible therapeutic avenue, is autophagy. Autophagy is an intracellular degradative pathway essential to maintain cellular homeostasis, particularly in neurons as defects in autophagy lead to neurodegeneration. One of the functions of autophagy is to maintain cellular homeostasis by eliminating defective or superfluous proteins, complexes, and organelles, preventing the accumulation of potentially cytotoxic damage. Importantly, there is also an intimate and intricate interplay between autophagy and multiple aspects of both innate and adaptive immunity. Thus, autophagy is implicated in two of the main hallmarks of MS, neurodegeneration, and inflammation, making it especially important to understand how this pathway contributes to MS manifestation and progression. This review summarizes the current knowledge about autophagy in MS, in particular how it contributes to our understanding of MS pathology and its potential as a novel therapeutic target.
Collapse
Affiliation(s)
- Chairi Misrielal
- Molecular Neurobiology, Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Mario Mauthe
- Molecular Cell Biology, Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Fulvio Reggiori
- Molecular Cell Biology, Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Bart J L Eggen
- Molecular Neurobiology, Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
107
|
Zhang C, Cheng N, Qiao B, Zhang F, Wu J, Liu C, Li Y, Du J. Age-related decline of interferon-gamma responses in macrophage impairs satellite cell proliferation and regeneration. J Cachexia Sarcopenia Muscle 2020; 11:1291-1305. [PMID: 32725722 PMCID: PMC7567146 DOI: 10.1002/jcsm.12584] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 03/18/2020] [Accepted: 04/07/2020] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Impaired muscle regeneration and increased muscle fibrosis are observed in aged muscle accompanied by progressive loss of muscle mass (sarcopenia). However, the underlying mechanism is still unclear. METHODS The differentiated expressed genes in young and aged muscles after acute injury by cardiotoxin were identified by RNA-sequence analysis. Single-cell RNA-sequence analysis was used to identify cell clusters and functions in young muscle after acute injury, and flow cytometry analysis and sorting were used to validate the function. The proliferation and differentiation functions of satellite cells were accessed by immunostaining with 5-ethynyl-2'-deoxyuridine and embryonic myosin heavy chain (eMyHC), respectively. Muscle regeneration ability was accessed by histopathological and molecular biological methods. RESULTS Gene expression patterns associated with responses to interferon-gamma (IFN-γ) (15 genes; false discovery rate < 0.001) were significantly down-regulated during muscle regeneration in aged mice (P = 2.25e-7). CD8+ T cells were the main source of increased IFN-γ after injury, adoptive transfer of wild-type CD8+ T cells to IFN-γ-deficient young mice resulted in 78% increase in cross-sectional areas (CSAs) of regenerated myofibres (P < 0.05) and 63% decrease in muscle fibrosis (P < 0.05) after injury. Single-cell RNA-sequence analysis identified a novel subset of macrophages [named as IFN-responsive macrophages (IFNRMs)] that specifically expressed IFN-responsive genes (Ifit3, Isg15, Irf7, etc.) in young mice at 3 days after injury, and the number of this macrophage subset was ~20% lower in aged mice at the same time (P < 0.05). IFNRMs secreted cytokine C-X-C motif chemokine 10 (CXCL10) that promoted the proliferation and differentiation of satellite cells via its receptor, CXCR3. Intramuscular recombinant CXCL10 treatment in aged mice rejuvenated the proliferation of satellite cells (80% increase in Ki-67+ Pax7+ cells, P < 0.01) and resulted in 27% increase in CSA of regenerated myofibres (P < 0.01) and 29% decrease in muscle fibrosis (P < 0.05). CONCLUSIONS Our study indicates that decline in IFN-γ response in a novel subset of macrophage contributes to satellite cells dysfunctions in aged skeletal muscles and demonstrates that this mechanism can be targeted to restore age-associated myogenesis.
Collapse
Affiliation(s)
- Congcong Zhang
- Beijing Anzhen Hospital, Capital Medical University; Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovation Center for Cardiovascular Disorders; Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Naixuan Cheng
- Beijing Anzhen Hospital, Capital Medical University; Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovation Center for Cardiovascular Disorders; Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Bokang Qiao
- Beijing Anzhen Hospital, Capital Medical University; Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovation Center for Cardiovascular Disorders; Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Fan Zhang
- Beijing Anzhen Hospital, Capital Medical University; Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovation Center for Cardiovascular Disorders; Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Jian Wu
- Section of Physiology and Biochemistry of Sports, Capital University of Physical Education and Sports, Beijing, China
| | - Chang Liu
- Beijing Anzhen Hospital, Capital Medical University; Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovation Center for Cardiovascular Disorders; Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Yulin Li
- Beijing Anzhen Hospital, Capital Medical University; Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovation Center for Cardiovascular Disorders; Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Jie Du
- Beijing Anzhen Hospital, Capital Medical University; Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovation Center for Cardiovascular Disorders; Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| |
Collapse
|
108
|
Yoo SW, Agarwal A, Smith MD, Khuder SS, Baxi EG, Thomas AG, Rojas C, Moniruzzaman M, Slusher BS, Bergles DE, Calabresi PA, Haughey NJ. Inhibition of neutral sphingomyelinase 2 promotes remyelination. SCIENCE ADVANCES 2020; 6:6/40/eaba5210. [PMID: 33008902 PMCID: PMC7852391 DOI: 10.1126/sciadv.aba5210] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Accepted: 08/20/2020] [Indexed: 05/05/2023]
Abstract
Myelination requires a highly organized synthesis of multiple lipid species that regulate myelin curvature and compaction. For reasons that are not understood, central nervous system remyelinated axons often have thin myelin sheaths with a disorganized structure susceptible to secondary demyelination. We found that expression of the sphingomyelin hydrolase neutral sphingomyelinase 2 (nSMase2) during the differentiation of oligodendrocyte progenitor cells (OPCs) to myelinating oligodendrocytes changes their response to inflammatory cytokines. OPCs do not express nSMase2 and exhibit a protective/regenerative response to tumor necrosis factor-α and interleukin-1β. Oligodendrocytes express nSMase2 and exhibit a stress response to cytokine challenge that includes an overproduction of ceramide, a sphingolipid that forms negative curvatures in membranes. Pharmacological inhibition or genetic deletion of nSMase2 in myelinating oligodendrocytes normalized the ceramide content of remyelinated fibers and increased thickness and compaction. These results suggest that inhibition of nSMase2 could improve the quality of myelin and stabilize structure.
Collapse
Affiliation(s)
- Seung-Wan Yoo
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Amit Agarwal
- The Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Matthew D Smith
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Saja S Khuder
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Emily G Baxi
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ajit G Thomas
- The Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Camilo Rojas
- Department of Comparative Medicine and Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Mohammed Moniruzzaman
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Barbara S Slusher
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- The Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Comparative Medicine and Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Psychiatry, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Dwight E Bergles
- The Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Johns Hopkins University Kavli Neuroscience Discovery Institute, Baltimore, MD 21287, USA
| | - Peter A Calabresi
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- The Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Norman J Haughey
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
- Department of Psychiatry, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
109
|
Compensatory Neuroprotective Response of Thioredoxin Reductase against Oxidative-Nitrosative Stress Induced by Experimental Autoimmune Encephalomyelitis in Rats: Modulation by Theta Burst Stimulation. Molecules 2020; 25:molecules25173922. [PMID: 32867364 PMCID: PMC7503723 DOI: 10.3390/molecules25173922] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/05/2020] [Accepted: 08/08/2020] [Indexed: 12/12/2022] Open
Abstract
Cortical theta burst stimulation (TBS) structured as intermittent (iTBS) and continuous (cTBS) could prevent the progression of the experimental autoimmune encephalomyelitis (EAE). The interplay of brain antioxidant defense systems against free radicals (FRs) overproduction induced by EAE, as well as during iTBS or cTBS, have not been entirely investigated. This study aimed to examine whether oxidative-nitrogen stress (ONS) is one of the underlying pathophysiological mechanisms of EAE, which may be changed in terms of health improvement by iTBS or cTBS. Dark Agouti strain female rats were tested for the effects of EAE and TBS. The rats were randomly divided into the control group, rats specifically immunized for EAE and nonspecifically immuno-stimulated with Complete Freund's adjuvant. TBS or sham TBS was applied to EAE rats from 14th-24th post-immunization day. Superoxide dismutase activity, levels of superoxide anion (O2•-), lipid peroxidation, glutathione (GSH), nicotinamide adenine dinucleotide phosphate (NADPH), and thioredoxin reductase (TrxR) activity were analyzed in rat spinal cords homogenates. The severity of EAE clinical coincided with the climax of ONS. The most critical result refers to TrxR, which immensely responded against the applied stressors of the central nervous system (CNS), including immunization and TBS. We found that the compensatory neuroprotective role of TrxR upregulation is a positive feedback mechanism that reduces the harmfulness of ONS. iTBS and cTBS both modulate the biochemical environment against ONS at a distance from the area of stimulation, alleviating symptoms of EAE. The results of our study increase the understanding of FRs' interplay and the role of Trx/TrxR in ONS-associated neuroinflammatory diseases, such as EAE. Also, our results might help the development of new ideas for designing more effective medical treatment, combining neuropsychological with noninvasive neurostimulation-neuromodulation techniques to patients living with MS.
Collapse
|
110
|
Yazdankhah M, Shang P, Ghosh S, Hose S, Liu H, Weiss J, Fitting CS, Bhutto IA, Zigler JS, Qian J, Sahel JA, Sinha D, Stepicheva NA. Role of glia in optic nerve. Prog Retin Eye Res 2020; 81:100886. [PMID: 32771538 DOI: 10.1016/j.preteyeres.2020.100886] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/09/2020] [Accepted: 07/20/2020] [Indexed: 12/13/2022]
Abstract
Glial cells are critically important for maintenance of neuronal activity in the central nervous system (CNS), including the optic nerve (ON). However, the ON has several unique characteristics, such as an extremely high myelination level of retinal ganglion cell (RGC) axons throughout the length of the nerve (with virtually all fibers myelinated by 7 months of age in humans), lack of synapses and very narrow geometry. Moreover, the optic nerve head (ONH) - a region where the RGC axons exit the eye - represents an interesting area that is morphologically distinct in different species. In many cases of multiple sclerosis (demyelinating disease of the CNS) vision problems are the first manifestation of the disease, suggesting that RGCs and/or glia in the ON are more sensitive to pathological conditions than cells in other parts of the CNS. Here, we summarize current knowledge on glial organization and function in the ON, focusing on glial support of RGCs. We cover both well-established concepts on the important role of glial cells in ON health and new findings, including novel insights into mechanisms of remyelination, microglia/NG2 cell-cell interaction, astrocyte reactivity and the regulation of reactive astrogliosis by mitochondrial fragmentation in microglia.
Collapse
Affiliation(s)
- Meysam Yazdankhah
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Peng Shang
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sayan Ghosh
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Stacey Hose
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Haitao Liu
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Joseph Weiss
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Christopher S Fitting
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Imran A Bhutto
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - J Samuel Zigler
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jiang Qian
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - José-Alain Sahel
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Institut de la Vision, INSERM, CNRS, Sorbonne Université, F-75012, Paris, France
| | - Debasish Sinha
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Nadezda A Stepicheva
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
111
|
Soch A, Sominsky L, Younesi S, De Luca SN, Gunasekara M, Bozinovski S, Spencer SJ. The role of microglia in the second and third postnatal weeks of life in rat hippocampal development and memory. Brain Behav Immun 2020; 88:675-687. [PMID: 32360602 DOI: 10.1016/j.bbi.2020.04.082] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/28/2020] [Accepted: 04/28/2020] [Indexed: 12/31/2022] Open
Abstract
Microglia are resident immune cells of the central nervous system (CNS). In adulthood they are involved in surveillance and responses to pathogens and injury and prenatally they play a role in brain development. However, the role of microglia during the early postnatal period and how they impact development long-term remains poorly understood. Here, to investigate the specific role of microglia in postnatal development, we used a Cx3cr1-Dtr transgenic Wistar rat model to acutely ablate microglia from either postnatal day (P) 7 or 14. We specifically assessed how transient microglial ablation affected astrocytes and neurons acutely, during the juvenile period, and in adulthood. Hippocampal microglial numbers remained low at P21 in the P7-ablated animals and complexity remained reduced after P14-ablation. This protracted effect on these key immune cells led to a small but significant increase in CA1 mature neuron numbers and a significant increase in astrocyte density in the subgranular dentate gyrus in adults that had their microglia ablated at P14. However, these histological differences were small, and spatial and recognition memory in novel objection and place recognition tests were not affected. Overall, our data reveal for the first time that the transient depletion of microglia during the neonatal period impacts briefly on the brain but that the long-lasting effects are minimal. Neonatal microglia may be dispensable in the establishment of hippocampal brain function. These data also imply that novel therapeutic anti-inflammatories that cross the blood-brain barrier to inhibit microglia are unlikely to have long-term negative consequences if administered in the neonatal period.
Collapse
Affiliation(s)
- Alita Soch
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Vic., Australia
| | - Luba Sominsky
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Vic., Australia
| | - Simin Younesi
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Vic., Australia
| | - Simone N De Luca
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Vic., Australia
| | - Maneesha Gunasekara
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Vic., Australia
| | - Steven Bozinovski
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Vic., Australia
| | - Sarah J Spencer
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Vic., Australia; ARC Centre of Excellence for Nanoscale Biophotonics, RMIT University, Melbourne, Vic., Australia.
| |
Collapse
|
112
|
Rawji KS, Gonzalez Martinez GA, Sharma A, Franklin RJ. The Role of Astrocytes in Remyelination. Trends Neurosci 2020; 43:596-607. [DOI: 10.1016/j.tins.2020.05.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/05/2020] [Accepted: 05/26/2020] [Indexed: 12/21/2022]
|
113
|
Yolk sac-derived Pdcd11-positive cells modulate zebrafish microglia differentiation through the NF-κB-Tgfβ1 pathway. Cell Death Differ 2020; 28:170-183. [PMID: 32709934 PMCID: PMC7853042 DOI: 10.1038/s41418-020-0591-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 07/07/2020] [Accepted: 07/09/2020] [Indexed: 12/22/2022] Open
Abstract
Microglia are the primary immune cells in the central nervous system, which plays a vital role in neuron development and neurodegenerative diseases. Microglial precursors in peripheral hematopoietic tissues colonize the central nervous system during early embryogenesis. However, how intrinsic and extrinsic signals integrate to regulate microglia’s differentiation remains undefined. In this study, we identified the cerebral white matter hyperintensities susceptibility gene, programmed cell death protein 11 (PDCD11), as an essential factor regulating microglia differentiation. In zebrafish, pdcd11 deficiency prevents the differentiation of the precursors to mature brain microglia. Although, the inflammatory featured macrophage brain colonization is augmented. At 22 h post fertilization, the Pdcd11-positive cells on the yolk sac are distinct from macrophages and neutrophils. Mechanistically, PDCD11 exerts its physiological role by differentially regulating the functions of nuclear factor-kappa B family members, P65 and c-Rel, suppressing P65-mediated expression of inflammatory cytokines, such as tnfα, and enhancing the c-Rel-dependent appearance of tgfβ1. The present study provides novel insights in understanding microglia differentiation during zebrafish development.
Collapse
|
114
|
Kipp M. Does Siponimod Exert Direct Effects in the Central Nervous System? Cells 2020; 9:cells9081771. [PMID: 32722245 PMCID: PMC7463861 DOI: 10.3390/cells9081771] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 12/11/2022] Open
Abstract
The modulation of the sphingosine 1-phosphate receptor is an approved treatment for relapsing multiple sclerosis because of its anti-inflammatory effect of retaining lymphocytes in lymph nodes. Different sphingosine 1-phosphate receptor subtypes are expressed in the brain and spinal cord, and their pharmacological effects may improve disease development and neuropathology. Siponimod (BAF312) is a novel sphingosine 1-phosphate receptor modulator that has recently been approved for the treatment of active secondary progressive multiple sclerosis (MS). In this review article, we summarize recent evidence suggesting that the active role of siponimod in patients with progressive MS may be due to direct interaction with central nervous system cells. Additionally, we tried to summarize our current understanding of the function of siponimod and discuss the effects observed in the case of MS.
Collapse
Affiliation(s)
- Markus Kipp
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, 18057 Rostock, Germany
| |
Collapse
|
115
|
The ameliorative effects of a phenolic derivative of Moringa oleifera leave against vanadium-induced neurotoxicity in mice. IBRO Rep 2020; 9:164-182. [PMID: 32803016 PMCID: PMC7417907 DOI: 10.1016/j.ibror.2020.07.004] [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: 03/12/2020] [Accepted: 07/03/2020] [Indexed: 12/29/2022] Open
Abstract
Vanadium, a transition series metal released during some industrial activities, induces oxidative stress and lipid peroxidation. Ameliorative effect of a pure compound from the methanolic extract of Moringa oleifera leaves, code-named MIMO2, in 14-day old mice administered with vanadium (as sodium metavanadate 3 mg/kg) for 2 weeks was assessed. Results from body weight monitoring, muscular strength, and open field showed slight reduction in body weight and locomotion deficit in vanadium-exposed mice, ameliorated with MIMO2 co-administration. Degeneration of the Purkinje cell layer and neuronal death in the hippocampal CA1 region were observed in vanadium-exposed mice and both appeared significantly reduced with MIMO2 co-administration. Demyelination involving the midline of the corpus callosum, somatosensory and retrosplenial cortices was also reduced with MIMO2. Microglia activation and astrogliosis observed through immunohistochemistry were also alleviated. Immunohistochemistry for myelin, axons and oligodendrocyte lineage cells were also carried out and showed that in vanadium-treated mice brains, oligodendrocyte progenitor cells increased NG2 immunolabelling with hypertrophy and bushy, ramified appearance of their processes. MIMO2 displayed ameliorative and antioxidative effects in vanadium-induced neurotoxicity in experimental murine species. This is likely the first time MIMO2 is being used in vivo in an animal model.
Collapse
|
116
|
Werkman IL, Lentferink DH, Baron W. Macroglial diversity: white and grey areas and relevance to remyelination. Cell Mol Life Sci 2020; 78:143-171. [PMID: 32648004 PMCID: PMC7867526 DOI: 10.1007/s00018-020-03586-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 06/20/2020] [Accepted: 06/23/2020] [Indexed: 02/06/2023]
Abstract
Macroglia, comprising astrocytes and oligodendroglial lineage cells, have long been regarded as uniform cell types of the central nervous system (CNS). Although regional morphological differences between these cell types were initially described after their identification a century ago, these differences were largely ignored. Recently, accumulating evidence suggests that macroglial cells form distinct populations throughout the CNS, based on both functional and morphological features. Moreover, with the use of refined techniques including single-cell and single-nucleus RNA sequencing, additional evidence is emerging for regional macroglial heterogeneity at the transcriptional level. In parallel, several studies revealed the existence of regional differences in remyelination capacity between CNS grey and white matter areas, both in experimental models for successful remyelination as well as in the chronic demyelinating disease multiple sclerosis (MS). In this review, we provide an overview of the diversity in oligodendroglial lineage cells and astrocytes from the grey and white matter, as well as their interplay in health and upon demyelination and successful remyelination. In addition, we discuss the implications of regional macroglial diversity for remyelination in light of its failure in MS. Since the etiology of MS remains unknown and only disease-modifying treatments altering the immune response are available for MS, the elucidation of macroglial diversity in grey and white matter and its putative contribution to the observed difference in remyelination efficiency between these regions may open therapeutic avenues aimed at enhancing endogenous remyelination in either area.
Collapse
Affiliation(s)
- Inge L Werkman
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV, Groningen, the Netherlands
- Department of Biology, University of Virginia, Charlottesville, VA, 22904, USA
| | - Dennis H Lentferink
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV, Groningen, the Netherlands
| | - Wia Baron
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV, Groningen, the Netherlands.
| |
Collapse
|
117
|
Shelestak J, Singhal N, Frankle L, Tomor R, Sternbach S, McDonough J, Freeman E, Clements R. Increased blood-brain barrier hyperpermeability coincides with mast cell activation early under cuprizone administration. PLoS One 2020; 15:e0234001. [PMID: 32511268 PMCID: PMC7279587 DOI: 10.1371/journal.pone.0234001] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 05/15/2020] [Indexed: 12/20/2022] Open
Abstract
The cuprizone induced animal model of demyelination is characterized by demyelination in many regions of the brain with high levels of demyelination in the corpus callosum as well as changes in neuronal function by 4–6 weeks of exposure. The model is used as a tool to study demyelination and subsequent degeneration as well as therapeutic interventions on these effects. Historically, the cuprizone model has been shown to contain no alterations to blood-brain barrier integrity, a key feature in many diseases that affect the central nervous system. Cuprizone is generally administered for 4–6 weeks to obtain maximal demyelination and degeneration. However, emerging evidence has shown that the effects of cuprizone on the brain may occur earlier than measurable gross demyelination. This study sought to investigate changes to blood-brain barrier permeability early in cuprizone administration. Results showed an increase in blood-brain barrier permeability and changes in tight junction protein expression as early as 3 days after beginning cuprizone treatment. These changes preceded glial morphological activation and demyelination known to occur during cuprizone administration. Increases in mast cell presence and activity were measured alongside the increased permeability implicating mast cells as a potential source for the blood-brain barrier disruption. These results provide further evidence of blood-brain barrier alterations in the cuprizone model and a target of therapeutic intervention in the prevention of cuprizone-induced pathology. Understanding how mast cells become activated under cuprizone and if they contribute to blood-brain barrier alterations may give further insight into how and when the blood-brain barrier is affected in CNS diseases. In summary, cuprizone administration causes an increase in blood-brain barrier permeability and this permeability coincides with mast cell activation.
Collapse
Affiliation(s)
- John Shelestak
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, Ohio, United States of America
- * E-mail:
| | - Naveen Singhal
- Department of Biochemistry, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, India
| | - Lana Frankle
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, Ohio, United States of America
| | - Riely Tomor
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, Ohio, United States of America
| | - Sarah Sternbach
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, Ohio, United States of America
| | - Jennifer McDonough
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, Ohio, United States of America
| | - Ernest Freeman
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, Ohio, United States of America
| | - Robert Clements
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, Ohio, United States of America
| |
Collapse
|
118
|
Delayed Demyelination and Impaired Remyelination in Aged Mice in the Cuprizone Model. Cells 2020; 9:cells9040945. [PMID: 32290524 PMCID: PMC7226973 DOI: 10.3390/cells9040945] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/03/2020] [Accepted: 04/07/2020] [Indexed: 12/22/2022] Open
Abstract
To unravel the failure of remyelination in multiple sclerosis (MS) and to test promising remyelinating treatments, suitable animal models like the well-established cuprizone model are required. However, this model is only standardized in young mice. This does not represent the typical age of MS patients. Furthermore, remyelination is very fast in young mice, hindering the examination of effects of remyelination-promoting agents. Thus, there is the need for a better animal model to study remyelination. We therefore aimed to establish the cuprizone model in aged mice. 6-month-old C57BL6 mice were fed with different concentrations of cuprizone (0.2–0.6%) for 5–6.5 weeks. De- and remyelination in the medial and lateral parts of the corpus callosum were analyzed by immunohistochemistry. Feeding aged mice 0.4% cuprizone for 6.5 weeks resulted in the best and most reliable administration scheme with virtually complete demyelination of the corpus callosum. This was accompanied by a strong accumulation of microglia and near absolute loss of mature oligodendrocytes. Subsequent remyelination was initially robust but remained incomplete. The remyelination process in mature adult mice better represents the age of MS patients and offers a better model for the examination of regenerative therapies.
Collapse
|
119
|
de Jong CGHM, Gabius HJ, Baron W. The emerging role of galectins in (re)myelination and its potential for developing new approaches to treat multiple sclerosis. Cell Mol Life Sci 2020; 77:1289-1317. [PMID: 31628495 PMCID: PMC7113233 DOI: 10.1007/s00018-019-03327-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 09/27/2019] [Accepted: 09/30/2019] [Indexed: 12/12/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory, demyelinating and neurodegenerative disease of the central nervous system with unknown etiology. Currently approved disease-modifying treatment modalities are immunomodulatory or immunosuppressive. While the applied drugs reduce the frequency and severity of the attacks, their efficacy to regenerate myelin membranes and to halt disease progression is limited. To achieve such therapeutic aims, understanding biological mechanisms of remyelination and identifying factors that interfere with remyelination in MS can give respective directions. Such a perspective is given by the emerging functional profile of galectins. They form a family of tissue lectins, which are potent effectors in processes as diverse as adhesion, apoptosis, immune mediator release or migration. This review focuses on endogenous and exogenous roles of galectins in glial cells such as oligodendrocytes, astrocytes and microglia in the context of de- and (re)myelination and its dysregulation in MS. Evidence is arising for a cooperation among family members so that timed expression and/or secretion of galectins-1, -3 and -4 result in modifying developmental myelination, (neuro)inflammatory processes, de- and remyelination. Dissecting the mechanisms that underlie the distinct activities of galectins and identifying galectins as target or tool to modulate remyelination have the potential to contribute to the development of novel therapeutic strategies for MS.
Collapse
Affiliation(s)
- Charlotte G H M de Jong
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Hans-Joachim Gabius
- Institute of Physiological Chemistry, Faculty of Veterinary Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Wia Baron
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands.
| |
Collapse
|
120
|
Ronzano R, Thetiot M, Lubetzki C, Desmazieres A. Myelin Plasticity and Repair: Neuro-Glial Choir Sets the Tuning. Front Cell Neurosci 2020; 14:42. [PMID: 32180708 PMCID: PMC7059744 DOI: 10.3389/fncel.2020.00042] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 02/12/2020] [Indexed: 12/11/2022] Open
Abstract
The plasticity of the central nervous system (CNS) in response to neuronal activity has been suggested as early as 1894 by Cajal (1894). CNS plasticity has first been studied with a focus on neuronal structures. However, in the last decade, myelin plasticity has been unraveled as an adaptive mechanism of importance, in addition to the previously described processes of myelin repair. Indeed, it is now clear that myelin remodeling occurs along with life and adapts to the activity of neuronal networks. Until now, it has been considered as a two-part dialog between the neuron and the oligodendroglial lineage. However, other glial cell types might be at play in myelin plasticity. In the present review, we first summarize the key structural parameters for myelination, we then describe how neuronal activity modulates myelination and finally discuss how other glial cells could participate in myelinic adaptivity.
Collapse
Affiliation(s)
- Remi Ronzano
- Institut du Cerveau et de la Moelle épinière, Sorbonne Universités UPMC Université Paris 06, CNRS UMR7225-Inserm U1127, Paris, France
| | - Melina Thetiot
- Institut du Cerveau et de la Moelle épinière, Sorbonne Universités UPMC Université Paris 06, CNRS UMR7225-Inserm U1127, Paris, France
- Unit Zebrafish Neurogenetics, Department of Developmental & Stem Cell Biology, Institut Pasteur, CNRS, Paris, France
| | - Catherine Lubetzki
- Institut du Cerveau et de la Moelle épinière, Sorbonne Universités UPMC Université Paris 06, CNRS UMR7225-Inserm U1127, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France
| | - Anne Desmazieres
- Institut du Cerveau et de la Moelle épinière, Sorbonne Universités UPMC Université Paris 06, CNRS UMR7225-Inserm U1127, Paris, France
| |
Collapse
|
121
|
Wang S, Deng J, Fu H, Guo Z, Zhang L, Tang P. Astrocytes directly clear myelin debris through endocytosis pathways and followed by excessive gliosis after spinal cord injury. Biochem Biophys Res Commun 2020; 525:S0006-291X(20)30337-5. [PMID: 32070495 DOI: 10.1016/j.bbrc.2020.02.069] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 02/10/2020] [Indexed: 12/18/2022]
Abstract
Persisted myelin debris inhibit axon regeneration and contribute to further tissue damage after spinal cord injury (SCI). The traditional view is that myelin debris is mainly cleared by microglia and macrophages, while astrocytes cannot directly engulf myelin debris because they are absent from lesion core. Here, we definitely showed that astrocytes could directly uptake myelin debris both in vitro and in vivo to effectively complement the clearance function. Therefore, it can be shown that astrocytes can exert myelin clearance effect directly and indirectly after spinal cord injury. The damaged myelin debris was transported to lysosomes for degradation through endocytosis pathways, finally resulting in excessive gliosis. This process may be a potential target for regulating neural tissue repair and excessive glia scar formation after SCI.
Collapse
Affiliation(s)
- Song Wang
- School of Medicine, Nankai University, Tianjin, China; Department of Orthopedics, The General Hospital of People's Liberation Army, Beijing, China
| | - Junhao Deng
- Department of Orthopedics, The General Hospital of People's Liberation Army, Beijing, China
| | - Haitao Fu
- Department of Orthopaedics, The Affiliated Hospital of Qingdao University, Shandong, China
| | - Zhongkui Guo
- School of Medicine, Nankai University, Tianjin, China; Department of Orthopedics, The General Hospital of People's Liberation Army, Beijing, China
| | - Licheng Zhang
- Department of Orthopedics, The General Hospital of People's Liberation Army, Beijing, China.
| | - Peifu Tang
- School of Medicine, Nankai University, Tianjin, China; Department of Orthopedics, The General Hospital of People's Liberation Army, Beijing, China.
| |
Collapse
|
122
|
Liu Y, Wu C, Hou Z, Fu X, Yuan L, Sun S, Zhang H, Yang D, Yao X, Yang J. Pseudoginsenoside-F11 Accelerates Microglial Phagocytosis of Myelin Debris and Attenuates Cerebral Ischemic Injury Through Complement Receptor 3. Neuroscience 2020; 426:33-49. [DOI: 10.1016/j.neuroscience.2019.11.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 02/08/2023]
|
123
|
Abstract
Microglia are resident macrophages of the CNS that are involved in its development, homeostasis and response to infection and damage. Microglial activation is a common feature of neurological disorders, and although in some instances this activation can be damaging, protective and regenerative functions of microglia have been revealed. The most prominent example of the regenerative functions is a role for microglia in supporting regeneration of myelin after injury, a process that is critical for axonal health and relevant to numerous disorders in which loss of myelin integrity is a prevalent feature, such as multiple sclerosis, Alzheimer disease and motor neuron disease. Although drugs that are intended to promote remyelination are entering clinical trials, the mechanisms by which remyelination is controlled and how microglia are involved are not completely understood. In this Review, we discuss work that has identified novel regulators of microglial activation - including molecular drivers, population heterogeneity and turnover - that might influence their pro-remyelination capacity. We also discuss therapeutic targeting of microglia as a potential approach to promoting remyelination.
Collapse
|
124
|
Yin JJ, He Y, An J, Miao Q, Sui RX, Wang Q, Yu JZ, Xiao BG, Ma CG. Dynamic Balance of Microglia and Astrocytes Involved in the Remyelinating Effect of Ginkgolide B. Front Cell Neurosci 2020; 13:572. [PMID: 31969806 PMCID: PMC6960131 DOI: 10.3389/fncel.2019.00572] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 12/12/2019] [Indexed: 01/04/2023] Open
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disorder in the central nervous system (CNS), in which remyelination failure results in persistent neurologic impairment. Ginkgolide B (GB), a major terpene lactone and active component of Ginkgo biloba, has neuroprotective effects in several models of neurological diseases. Here, our results show, by using an in vivo cuprizone (CPZ)-induced demyelinating model, administration of GB improved behavior abnormalities, promoted myelin generation, and significantly regulated the dynamic balance of microglia and astrocytes by inhibiting the expression of TLR4, NF-κB and iNOS as well as IL-1β and TNF-α, and up-regulating the expression of Arg-1 and neurotrophic factors. GB treatment also induced the generation of oligodendrocyte precursor cells (OPCs). In vitro cell experiments yielded the results similar to those of the in vivo model. The dynamic balance by decreasing microglia-mediated neuroinflammation and promoting astrocyte-derived neurotrophic factors should contribute to endogenous remyelination. Despite GB treatment may represent a novel strategy for promoting myelin recovery, the precise mechanism of GB targeting microglia and astrocytes remains to be further explored.
Collapse
Affiliation(s)
- Jun-Jun Yin
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Yan He
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Jun An
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Qiang Miao
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Ruo-Xuan Sui
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Qing Wang
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Jie-Zhong Yu
- Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Institute of Brain Science, Shanxi Datong University, Datong, China
| | - Bao-Guo Xiao
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Cun-Gen Ma
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan, China.,Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Institute of Brain Science, Shanxi Datong University, Datong, China
| |
Collapse
|
125
|
Blocking the Thrombin Receptor Promotes Repair of Demyelinated Lesions in the Adult Brain. J Neurosci 2020; 40:1483-1500. [PMID: 31911460 DOI: 10.1523/jneurosci.2029-19.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 12/15/2019] [Accepted: 12/17/2019] [Indexed: 01/14/2023] Open
Abstract
Myelin loss limits neurological recovery and myelin regeneration and is critical for restoration of function. We recently discovered that global knock-out of the thrombin receptor, also known as Protease Activated Receptor 1 (PAR1), accelerates myelin development. Here we demonstrate that knocking out PAR1 also promotes myelin regeneration. Outcomes in two unique models of myelin injury and repair, that is lysolecithin or cuprizone-mediated demyelination, showed that PAR1 knock-out in male mice improves replenishment of myelinating cells and remyelinated nerve fibers and slows early axon damage. Improvements in myelin regeneration in PAR1 knock-out mice occurred in tandem with a skewing of reactive astrocyte signatures toward a prorepair phenotype. In cell culture, the promyelinating effects of PAR1 loss of function are consistent with possible direct effects on the myelinating potential of oligodendrocyte progenitor cells (OPCs), in addition to OPC-indirect effects involving enhanced astrocyte expression of promyelinating factors, such as BDNF. These findings highlight previously unrecognized roles of PAR1 in myelin regeneration, including integrated actions across the oligodendrocyte and astroglial compartments that are at least partially mechanistically linked to the powerful BDNF-TrkB neurotrophic signaling system. Altogether, findings suggest PAR1 may be a therapeutically tractable target for demyelinating disorders of the CNS.SIGNIFICANCE STATEMENT Replacement of oligodendroglia and myelin regeneration holds tremendous potential to improve function across neurological conditions. Here we demonstrate Protease Activated Receptor 1 (PAR1) is an important regulator of the capacity for myelin regeneration across two experimental murine models of myelin injury. PAR1 is a G-protein-coupled receptor densely expressed in the CNS, however there is limited information regarding its physiological roles in health and disease. Using a combination of PAR1 knock-out mice, oligodendrocyte monocultures and oligodendrocyte-astrocyte cocultures, we demonstrate blocking PAR1 improves myelin production by a mechanism related to effects across glial compartments and linked in part to regulatory actions toward growth factors such as BDNF. These findings set the stage for development of new clinically relevant myelin regeneration strategies.
Collapse
|
126
|
Watson AES, Goodkey K, Footz T, Voronova A. Regulation of CNS precursor function by neuronal chemokines. Neurosci Lett 2020; 715:134533. [DOI: 10.1016/j.neulet.2019.134533] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 09/16/2019] [Accepted: 10/01/2019] [Indexed: 02/07/2023]
|
127
|
Kriszta G, Nemes B, Sándor Z, Ács P, Komoly S, Berente Z, Bölcskei K, Pintér E. Investigation of Cuprizone-Induced Demyelination in mGFAP-Driven Conditional Transient Receptor Potential Ankyrin 1 (TRPA1) Receptor Knockout Mice. Cells 2019; 9:cells9010081. [PMID: 31905673 PMCID: PMC7017039 DOI: 10.3390/cells9010081] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/19/2019] [Accepted: 12/20/2019] [Indexed: 12/24/2022] Open
Abstract
Transient receptor potential ankyrin 1 (TRPA1) receptors are non-selective cation channels responsive to a variety of exogenous irritants and endogenous stimuli including products of oxidative stress. It is mainly expressed by primary sensory neurons; however, expression of TRPA1 by astrocytes and oligodendrocytes has recently been detected in the mouse brain. Genetic deletion of TRPA1 was shown to attenuate cuprizone-induced oligodendrocyte apoptosis and myelin loss in mice. In the present study we aimed at investigating mGFAP-Cre conditional TRPA1 knockout mice in the cuprizone model. These animals were generated by crossbreeding GFAP-Cre+/− and floxed TRPA1 (TRPA1Fl/Fl) mice. Cuprizone was administered for 6 weeks and demyelination was followed by magnetic resonance imaging (MRI). At the end of the treatment, demyelination and glial activation was also investigated by histological methods. The results of the MRI showed that demyelination was milder at weeks 3 and 4 in both homozygous (GFAP-Cre+/− TRPA1Fl/Fl) and heterozygous (GFAP-Cre+/− TRPA1Fl/−) conditional knockout animals compared to Cre−/− control mice. However, by week 6 of the treatment the difference was not detectable by either MRI or histological methods. In conclusion, TRPA1 receptors on astrocytes may transiently contribute to the demyelination induced by cuprizone, however, expression and function of TRPA1 receptors by other cells in the brain (oligodendrocytes, microglia, neurons) warrant further investigation.
Collapse
Affiliation(s)
- Gábor Kriszta
- Department of Pharmacology and Pharmacotherapy, University of Pécs Medical School, Pécs H-7624, Hungary; (G.K.); (B.N.); (Z.S.); (K.B.)
- Molecular Pharmacology Research Group and Center for Neuroscience, János Szentágothai Research Center, University of Pécs, Pécs H-7624, Hungary
- Research Group for Experimental Diagnostic Imaging, University of Pécs Medical School, Pécs H-7624, Hungary;
| | - Balázs Nemes
- Department of Pharmacology and Pharmacotherapy, University of Pécs Medical School, Pécs H-7624, Hungary; (G.K.); (B.N.); (Z.S.); (K.B.)
| | - Zoltán Sándor
- Department of Pharmacology and Pharmacotherapy, University of Pécs Medical School, Pécs H-7624, Hungary; (G.K.); (B.N.); (Z.S.); (K.B.)
| | - Péter Ács
- Department of Neurology, University of Pécs Medical School, Pécs H-7623, Hungary; (P.Á.); (S.K.)
| | - Sámuel Komoly
- Department of Neurology, University of Pécs Medical School, Pécs H-7623, Hungary; (P.Á.); (S.K.)
| | - Zoltán Berente
- Research Group for Experimental Diagnostic Imaging, University of Pécs Medical School, Pécs H-7624, Hungary;
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, Pécs H-7624, Hungary
| | - Kata Bölcskei
- Department of Pharmacology and Pharmacotherapy, University of Pécs Medical School, Pécs H-7624, Hungary; (G.K.); (B.N.); (Z.S.); (K.B.)
- Molecular Pharmacology Research Group and Center for Neuroscience, János Szentágothai Research Center, University of Pécs, Pécs H-7624, Hungary
| | - Erika Pintér
- Department of Pharmacology and Pharmacotherapy, University of Pécs Medical School, Pécs H-7624, Hungary; (G.K.); (B.N.); (Z.S.); (K.B.)
- Molecular Pharmacology Research Group and Center for Neuroscience, János Szentágothai Research Center, University of Pécs, Pécs H-7624, Hungary
- Correspondence:
| |
Collapse
|
128
|
Molina-Gonzalez I, Miron VE. Astrocytes in myelination and remyelination. Neurosci Lett 2019; 713:134532. [PMID: 31589903 DOI: 10.1016/j.neulet.2019.134532] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 09/13/2019] [Accepted: 10/01/2019] [Indexed: 02/07/2023]
Abstract
Astrocytes are known to play critical roles in central nervous system development, homeostasis, and response to injury. In addition to well-defined functions in synaptic signalling and blood-brain barrier control, astrocytes are now emerging as important contributors to white matter health. Here, we review the roles of astrocytes in myelin formation and regeneration (remyelination), focusing on both direct interactions with oligodendrocyte lineage cells, and indirect influences via crosstalk with central nervous system resident macrophages, microglia.
Collapse
Affiliation(s)
- Irene Molina-Gonzalez
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Veronique E Miron
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
129
|
An J, Yin JJ, He Y, Sui RX, Miao Q, Wang Q, Yu JZ, Yu JW, Shi FD, Ma CG, Xiao BG. Temporal and Spatial Dynamics of Astroglial Reaction and Immune Response in Cuprizone-Induced Demyelination. Neurotox Res 2019; 37:587-601. [DOI: 10.1007/s12640-019-00129-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 10/03/2019] [Accepted: 10/25/2019] [Indexed: 12/14/2022]
|
130
|
Nyamoya S, Steinle J, Chrzanowski U, Kaye J, Schmitz C, Beyer C, Kipp M. Laquinimod Supports Remyelination in Non-Supportive Environments. Cells 2019; 8:cells8111363. [PMID: 31683658 PMCID: PMC6912710 DOI: 10.3390/cells8111363] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 01/20/2023] Open
Abstract
Inflammatory demyelination, which is a characteristic of multiple sclerosis lesions, leads to acute functional deficits and, in the long term, to progressive axonal degeneration. While remyelination is believed to protect axons, the endogenous-regenerative processes are often incomplete or even completely fail in many multiple sclerosis patients. Although it is currently unknown why remyelination fails, recurrent demyelination of previously demyelinated white matter areas is one contributing factor. In this study, we investigated whether laquinimod, which has demonstrated protective effects in active multiple sclerosis patients, protects against recurrent demyelination. To address this, male mice were intoxicated with cuprizone for up to eight weeks and treated with either a vehicle solution or laquinimod at the beginning of week 5, where remyelination was ongoing. The brains were harvested and analyzed by immunohistochemistry. At the time-point of laquinimod treatment initiation, oligodendrocyte progenitor cells proliferated and maturated despite ongoing demyelination activity. In the following weeks, myelination recovered in the laquinimod- but not vehicle-treated mice, despite continued cuprizone intoxication. Myelin recovery was paralleled by less severe microgliosis and acute axonal injury. In this study, we were able to demonstrate that laquinimod, which has previously been shown to protect against cuprizone-induced oligodendrocyte degeneration, exerts protective effects during oligodendrocyte progenitor differentiation as well. By this mechanism, laquinimod allows remyelination in non-supportive environments. These results should encourage further clinical studies in progressive multiple sclerosis patients.
Collapse
Affiliation(s)
- Stella Nyamoya
- Institute of Anatomy, Rostock University Medical Center, 18057 Rostock, Germany.
- Institute of Neuroanatomy and JARA-BRAIN, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany.
| | - Julia Steinle
- Institute of Neuroanatomy and JARA-BRAIN, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany.
| | - Uta Chrzanowski
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, 80336 Munich, Germany.
| | - Joel Kaye
- AyalaPharma, VP Research & Nonclinical Development, Rehovot 7670104, Israel.
| | - Christoph Schmitz
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, 80336 Munich, Germany.
| | - Cordian Beyer
- Institute of Neuroanatomy and JARA-BRAIN, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany.
| | - Markus Kipp
- Institute of Neuroanatomy and JARA-BRAIN, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany.
- Centre for Transdisciplinary Neurosciences, Rostock University Medical Center, 18057 Rostock, Germany.
| |
Collapse
|
131
|
Zhao Z, Bao XQ, Zhang Z, Liu H, Zhang D. Phloroglucinol derivative compound 21 attenuates cuprizone-induced multiple sclerosis mice through promoting remyelination and inhibiting neuroinflammation. SCIENCE CHINA-LIFE SCIENCES 2019; 63:905-914. [PMID: 31637574 DOI: 10.1007/s11427-019-9821-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 09/03/2019] [Indexed: 12/20/2022]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune demyelinating disease in the central nervous system. The myelin loss is mainly caused by dysfunction of oligodendrocytes and inflammatory responses of microglia and astrocytes further aggravate the demyelination. Current therapies for MS focus on suppressing the overactivated immune response but cannot halt the disease progress, so effective drugs are urgently needed. Compound 21 is a phloroglucinol derivative that has been proved to have an outstanding anti-inflammatory effect. The purpose of the present study is to investigate whether this novel compound is effective in MS. The cuprizone-induced model was used in this study to mimic the pathological progress of MS. The results showed that Compound 21 significantly improved the neurological dysfunction and motor coordination impairment. Luxol Fast Blue staining and myelin basic protein immunostaining demonstrated that Compound 21 remarkably promoted remyelination. In addition, Compound 21 significantly promoted oligodendrocytes differentiation. Furthermore, we found that Compound 21 decreased microglia and astrocytes activities and the subsequent neuroinflammatory response, indicating that the anti-inflammatory effect of Compound 21 was also involved in its neuro-protection. All the data prove that Compound 21 exerts protective effect on MS through promoting remyelination and suppressing neuroinflammation, indicating that Compound 21 might be a potential drug candidate for MS treatment.
Collapse
Affiliation(s)
- Zhe Zhao
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Xiu-Qi Bao
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Zihong Zhang
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Hui Liu
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Dan Zhang
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
132
|
Baaklini CS, Rawji KS, Duncan GJ, Ho MFS, Plemel JR. Central Nervous System Remyelination: Roles of Glia and Innate Immune Cells. Front Mol Neurosci 2019; 12:225. [PMID: 31616249 PMCID: PMC6764409 DOI: 10.3389/fnmol.2019.00225] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 09/04/2019] [Indexed: 12/31/2022] Open
Abstract
In diseases such as multiple sclerosis (MS), inflammation can injure the myelin sheath that surrounds axons, a process known as demyelination. The spontaneous regeneration of myelin, called remyelination, is associated with restoration of function and prevention of axonal degeneration. Boosting remyelination with therapeutic intervention is a promising new approach that is currently being tested in several clinical trials. The endogenous regulation of remyelination is highly dependent on the immune response. In this review article, we highlight the cell biology of remyelination and its regulation by innate immune cells. For the purpose of this review, we discuss the roles of microglia, and also astrocytes and oligodendrocyte progenitor cells (OPCs) as they are being increasingly recognized to have immune cell functions.
Collapse
Affiliation(s)
- Charbel S. Baaklini
- Department of Medicine, Division of Neurology, Neuroscience and Mental Health Institute, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Khalil S. Rawji
- Wellcome Trust-Medical Research Council, Cambridge Stem Cell Institute, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Greg J. Duncan
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health and Science University, Portland, OR, United States
| | - Madelene F. S. Ho
- Department of Medicine, Division of Neurology, Neuroscience and Mental Health Institute, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada
| | | |
Collapse
|
133
|
Pars K, Gingele M, Kronenberg J, Prajeeth CK, Skripuletz T, Pul R, Jacobs R, Gudi V, Stangel M. Fumaric Acids Do Not Directly Influence Gene Expression of Neuroprotective Factors in Highly Purified Rodent Astrocytes. Brain Sci 2019; 9:brainsci9090241. [PMID: 31546798 PMCID: PMC6769695 DOI: 10.3390/brainsci9090241] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 09/12/2019] [Accepted: 09/16/2019] [Indexed: 12/30/2022] Open
Abstract
(1) Background: Dimethylfumarate (DMF) has been approved for the treatment of relapsing remitting multiple sclerosis. However, the mode of action of DMF and its assumed active primary metabolite monomethylfumarate (MMF) is still not fully understood. Former reports suggest a neuroprotective effect of DMF mediated via astrocytes by reducing pro-inflammatory activation of these glial cells. We investigated potential direct effects of DMF and MMF on neuroprotective factors like neurotrophic factors and growth factors in astrocytes to elucidate further possible mechanisms of the mode of action of fumaric acids; (2) Methods: highly purified cultures of primary rat astrocytes were pre-treated in vitro with DMF or MMF and incubated with lipopolysaccharides (LPS) or a mixture of interferon gamma (IFN-γ) plus interleukin 1 beta (IL-1β) in order to simulate an inflammatory environment. The gene expression of neuroprotective factors such as neurotrophic factors (nuclear factor E2-related factor 2 (NGF), brain-derived neurotrophic factor (BDNF), glial cell-derived neurotrophic factor (GDNF)) and growth factors (fibroblast growth factor 2 (FGF2), platelet-derived growth factor subunit A (PDGFa), ciliary neurotrophic factor (CNTF)) as well as cytokines (tumor necrosis factor alpha (TNFα), interleukin 6 (IL-6), IL-1β, inducible nitric oxide synthase (iNOS)) was examined by determining the transcription level with real-time quantitative polymerase chain reaction (qPCR); (3) Results: The stimulation of highly purified astrocytes with either LPS or cytokines changed the expression profile of growth factors and pro- inflammatory factors. However, the expression was not altered by either DMF nor MMF in unstimulated or stimulated astrocytes; (4) Conclusions: There was no direct influence of fumaric acids on neuroprotective factors in highly purified primary rat astrocytes. This suggests that the proposed potential neuroprotective effect of fumaric acid is not mediated by direct stimulation of neurotrophic factors in astrocytes but is rather mediated by other pathways or indirect mechanisms via other glial cells like microglia as previously demonstrated.
Collapse
Affiliation(s)
- Kaweh Pars
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover, Medical School, 30559 Hannover, Germany.
- Department of Neurology, European Medical School, University Oldenburg, 26129 Oldenburg, Germany.
| | - Marina Gingele
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover, Medical School, 30559 Hannover, Germany.
| | - Jessica Kronenberg
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover, Medical School, 30559 Hannover, Germany.
- Center for Systems Neuroscience, University of Veterinary Medicine, 30559 Hannover, Germany.
| | - Chittappen K Prajeeth
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover, Medical School, 30559 Hannover, Germany.
| | - Thomas Skripuletz
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover, Medical School, 30559 Hannover, Germany.
| | - Refik Pul
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover, Medical School, 30559 Hannover, Germany.
- Department of Neurology, University Clinic Essen, 45147 Essen, Germany.
| | - Roland Jacobs
- Department of Clinical Immunology and Rheumatology, Hannover Medical School, 30559 Hannover, Germany.
| | - Viktoria Gudi
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover, Medical School, 30559 Hannover, Germany.
| | - Martin Stangel
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover, Medical School, 30559 Hannover, Germany.
- Center for Systems Neuroscience, University of Veterinary Medicine, 30559 Hannover, Germany.
| |
Collapse
|
134
|
Mandolesi G, Bullitta S, Fresegna D, De Vito F, Rizzo FR, Musella A, Guadalupi L, Vanni V, Stampanoni Bassi M, Buttari F, Viscomi MT, Centonze D, Gentile A. Voluntary running wheel attenuates motor deterioration and brain damage in cuprizone-induced demyelination. Neurobiol Dis 2019; 129:102-117. [DOI: 10.1016/j.nbd.2019.05.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 10/23/2018] [Accepted: 05/13/2019] [Indexed: 12/27/2022] Open
|
135
|
Galloway DA, Gowing E, Setayeshgar S, Kothary R. Inhibitory milieu at the multiple sclerosis lesion site and the challenges for remyelination. Glia 2019; 68:859-877. [PMID: 31441132 DOI: 10.1002/glia.23711] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 06/26/2019] [Accepted: 07/26/2019] [Indexed: 12/14/2022]
Abstract
Regeneration of myelin, following injury, can occur within the central nervous system to reinstate proper axonal conductance and provide trophic support. Failure to do so renders the axons vulnerable, leading to eventual degeneration, and neuronal loss. Thus, it is essential to understand the mechanisms by which remyelination or failure to remyelinate occur, particularly in the context of demyelinating and neurodegenerative disorders. In multiple sclerosis, oligodendrocyte progenitor cells (OPCs) migrate to lesion sites to repair myelin. However, during disease progression, the ability of OPCs to participate in remyelination diminishes coincident with worsening of the symptoms. Remyelination is affected by a broad range of cues from intrinsic programming of OPCs and extrinsic local factors to the immune system and other systemic elements including diet and exercise. Here we review the literature on these diverse inhibitory factors and the challenges they pose to remyelination. Results spanning several disciplines from fundamental preclinical studies to knowledge gained in the clinic will be discussed.
Collapse
Affiliation(s)
- Dylan A Galloway
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, Canada
| | - Elizabeth Gowing
- Neurosciences Department, Faculty of Medicine, Centre de recherche du CHUM, Université de Montreal, Montreal, Quebec, Canada
| | - Solmaz Setayeshgar
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Rashmi Kothary
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Medicine, Department of Biochemistry, Microbiology and Immunology, and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
136
|
Sen MK, Mahns DA, Coorssen JR, Shortland PJ. Behavioural phenotypes in the cuprizone model of central nervous system demyelination. Neurosci Biobehav Rev 2019; 107:23-46. [PMID: 31442519 DOI: 10.1016/j.neubiorev.2019.08.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 08/01/2019] [Accepted: 08/12/2019] [Indexed: 12/14/2022]
Abstract
The feeding of cuprizone (CPZ) to animals has been extensively used to model the processes of demyelination and remyelination, with many papers adopting a narrative linked to demyelinating conditions like multiple sclerosis (MS), the aetiology of which is unknown. However, no current animal model faithfully replicates the myriad of symptoms seen in the clinical condition of MS. CPZ ingestion causes mitochondrial and endoplasmic reticulum stress and subsequent apoptosis of oligodendrocytes leads to central nervous system demyelination and glial cell activation. Although there are a wide variety of behavioural tests available for characterizing the functional deficits in animal models of disease, including that of CPZ-induced deficits, they have focused on a narrow subset of outcomes such as motor performance, cognition, and anxiety. The literature has not been systematically reviewed in relation to these or other symptoms associated with clinical MS. This paper reviews these tests and makes recommendations as to which are the most important in order to better understand the role of this model in examining aspects of demyelinating diseases like MS.
Collapse
Affiliation(s)
- Monokesh K Sen
- School of Medicine, Western Sydney University, New South Wales, Australia
| | - David A Mahns
- School of Medicine, Western Sydney University, New South Wales, Australia
| | - Jens R Coorssen
- Departments of Health Sciences and Biological Sciences, Faculties of Applied Health Sciences and Mathematics & Science, Brock University, Ontario, Canada.
| | - Peter J Shortland
- Science and Health, Western Sydney University, New South Wales, Australia.
| |
Collapse
|
137
|
Allnoch L, Baumgärtner W, Hansmann F. Impact of Astrocyte Depletion upon Inflammation and Demyelination in a Murine Animal Model of Multiple Sclerosis. Int J Mol Sci 2019; 20:ijms20163922. [PMID: 31409036 PMCID: PMC6719128 DOI: 10.3390/ijms20163922] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 08/07/2019] [Accepted: 08/10/2019] [Indexed: 12/17/2022] Open
Abstract
Astrocytes play a key role in demyelinating diseases, like multiple sclerosis (MS), although many of their functions remain unknown. The aim of this study was to investigate the impact of astrocyte depletion upon de- and remyelination, inflammation, axonal damage, and virus distribution in Theiler`s murine encephalomyelitis (TME). Groups of two to six glial fibrillary acidic protein (GFAP)-thymidine-kinase transgenic SJL mice and SJL wildtype mice were infected with TME virus (TMEV) or mock (vehicle only). Astrocyte depletion was induced by the intraperitoneal administration of ganciclovir during the early and late phase of TME. The animals were clinically investigated while using a scoring system and a rotarod performance test. Necropsies were performed at 46 and 77 days post infection. Cervical and thoracic spinal cord segments were investigated using hematoxylin and eosin (H&E), luxol fast blue-cresyl violet (LFB), immunohistochemistry targeting Amigo2, aquaporin 4, CD3, CD34, GFAP, ionized calcium-binding adapter molecule 1 (Iba1), myelin basic protein (MBP), non-phosphorylated neurofilaments (np-NF), periaxin, S100A10, TMEV, and immunoelectron microscopy. The astrocyte depleted mice showed a deterioration of clinical signs, a downregulation and disorganization of aquaporin 4 in perivascular astrocytes accompanied by vascular leakage. Furthermore, astrocyte depleted mice showed reduced inflammation and lower numbers of TMEV positive cells in the spinal cord. The present study indicates that astrocyte depletion in virus triggered CNS diseases contributes to a deterioration of clinical signs that are mediated by a dysfunction of perivascular astrocytes.
Collapse
Affiliation(s)
- Lisa Allnoch
- Department of Pathology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany.
- Center for Systems Neuroscience, 30559 Hannover, Germany.
| | - Florian Hansmann
- Department of Pathology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
- Center for Systems Neuroscience, 30559 Hannover, Germany
| |
Collapse
|
138
|
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: 47] [Impact Index Per Article: 7.8] [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
|
139
|
Rajizadeh MA, Sheibani V, Bejeshk MA, Mohtashami Borzadaran F, Saghari H, Esmaeilpour K. The effects of high intensity exercise on learning and memory impairments followed by combination of sleep deprivation and demyelination induced by etidium bromide. Int J Neurosci 2019; 129:1166-1178. [DOI: 10.1080/00207454.2019.1640695] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Mohammad Amin Rajizadeh
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
- Department of Physiology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Vahid Sheibani
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
- Department of Physiology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Abbas Bejeshk
- Department of Physiology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Hasan Saghari
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Khadijeh Esmaeilpour
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
140
|
Unal DB, Caliari SR, Lampe KJ. Engineering biomaterial microenvironments to promote myelination in the central nervous system. Brain Res Bull 2019; 152:159-174. [PMID: 31306690 DOI: 10.1016/j.brainresbull.2019.07.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 06/09/2019] [Accepted: 07/08/2019] [Indexed: 01/01/2023]
Abstract
Promoting remyelination and/or minimizing demyelination are key therapeutic strategies under investigation for diseases and injuries like multiple sclerosis (MS), spinal cord injury, stroke, and virus-induced encephalopathy. Myelination is essential for efficacious neuronal signaling. This myelination process is originated by oligodendrocyte progenitor cells (OPCs) in the central nervous system (CNS). Resident OPCs are capable of both proliferation and differentiation, and also migration to demyelinated injury sites. OPCs can then engage with these unmyelinated or demyelinated axons and differentiate into myelin-forming oligodendrocytes (OLs). However this process is frequently incomplete and often does not occur at all. Biomaterial strategies can now be used to guide OPC and OL development with the goal of regenerating healthy myelin sheaths in formerly damaged CNS tissue. Growth and neurotrophic factors delivered from such materials can promote proliferation of OPCs or differentiation into OLs. While cell transplantation techniques have been used to replace damaged cells in wound sites, they have also resulted in poor transplant cell viability, uncontrollable differentiation, and poor integration into the host. Biomaterial scaffolds made from extracellular matrix (ECM) mimics that are naturally or synthetically derived can improve transplanted cell survival, support both transplanted and endogenous cell populations, and direct their fate. In particular, stiffness and degradability of these scaffolds are two parameters that can influence the fate of OPCs and OLs. The future outlook for biomaterials research includes 3D in vitro models of myelination / remyelination / demyelination to better mimic and study these processes. These models should provide simple relationships of myelination to microenvironmental biophysical and biochemical properties to inform improved therapeutic approaches.
Collapse
Affiliation(s)
- Deniz B Unal
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA 22903, United States
| | - Steven R Caliari
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA 22903, United States; Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22903, United States
| | - Kyle J Lampe
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA 22903, United States.
| |
Collapse
|
141
|
Espitia Pinzon N, van Mierlo H, de Jonge JC, Brevé JJP, Bol JGJM, Drukarch B, van Dam AM, Baron W. Tissue Transglutaminase Promotes Early Differentiation of Oligodendrocyte Progenitor Cells. Front Cell Neurosci 2019; 13:281. [PMID: 31312122 PMCID: PMC6614186 DOI: 10.3389/fncel.2019.00281] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 06/11/2019] [Indexed: 01/09/2023] Open
Abstract
Demyelinated lesions of the central nervous system are characteristic for multiple sclerosis (MS). Remyelination is not very effective, particular at later stages of the disease, which results in a chronic neurodegenerative character with worsening of symptoms. Previously, we have shown that the enzyme Tissue Transglutaminase (TG2) is downregulated upon differentiation of oligodendrocyte progenitor cells (OPCs) into myelin-forming oligodendrocytes and that TG2 knock-out mice lag behind in remyelination after cuprizone-induced demyelination. Here, we examined whether astrocytic or oligodendroglial TG2 affects OPCs in a cell-specific manner to modulate their differentiation, and therefore myelination. Our findings indicate that human TG2-expressing astrocytes did not modulate OPC differentiation and myelination. In contrast, persistent TG2 expression upon OPC maturation or exogenously added recombinant TG2 accelerated OPC differentiation and myelin membrane formation. Continuous exposure of recombinant TG2 to OPCs at different consecutive developmental stages, however, decreased OPC differentiation and myelin membrane formation, while it enhanced myelination in dorsal root ganglion neuron-OPC co-cultures. In MS lesions, TG2 is absent in OPCs, while human OPCs show TG2 immunoreactivity during brain development. Exposure to the MS-relevant pro-inflammatory cytokine IFN-γ increased TG2 expression in OPCs and prolonged expression of endogenous TG2 upon differentiation. However, despite the increased TG2 levels, OPC maturation was not accelerated, indicating that TG2-mediated OPC differentiation may be counteracted by other pathways. Together, our data show that TG2, either endogenously expressed, or exogenously supplied to OPCs, accelerates early OPC differentiation. A better understanding of the role of TG2 in the OPC differentiation process during MS is of therapeutic interest to overcome remyelination failure.
Collapse
Affiliation(s)
- Nathaly Espitia Pinzon
- Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Hanneke van Mierlo
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Jenny C de Jonge
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - John J P Brevé
- Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - John G J M Bol
- Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Benjamin Drukarch
- Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Anne-Marie van Dam
- Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Wia Baron
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
142
|
Luo D, Lin R, Luo L, Li Q, Chen T, Qiu R, Li Y. Glial Plasticity in the Trigeminal Root Entry Zone of a Rat Trigeminal Neuralgia Animal Model. Neurochem Res 2019; 44:1893-1902. [PMID: 31209727 DOI: 10.1007/s11064-019-02824-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 05/12/2019] [Accepted: 06/05/2019] [Indexed: 12/16/2022]
Abstract
The trigeminal root entry zone (TREZ) is the transitional zone of central and peripheral tissue compartments in the trigeminal root. Microvascular compression on the TREZ is the main etiology of most idiopathic trigeminal neuralgia (TN) patients. However, the pathogenesis of TN is still uncertain. To investigate the glial plasticity changes in oligodendrocytes, Schwann cells, astrocytes and microglia/macrophages in the TREZ in TN, immunohistochemical staining and Western blot methods were performed in rats with TN induced by compression injury. The results showed that mechanical compression injury in the trigeminal nerve of the TN rats induced glial plasticity in the TREZ, which dynamically changed the glial interface of the CNS-PNS transitional zone. Additionally, glial fibrillary acidic protein (GFAP)-immunoreactive astrocyte processes significantly proliferated and extended distally from the central region to the peripheral side of the TREZ after nerve compression injury in the TN group. Moreover, the expression of p75 in Schwann cells was upregulated on the peripheral side of the TREZ, and activated Iba-1-immunoreactive microglia/macrophages were observed on both sides of the TREZ. A significantly higher number of Schwann cells, astrocytes and microglia/macrophages were found in the TN group than in the sham operation group (p < 0.05). In conclusion, mechanical compression injury in the TN rats activated various glial cells, including oligodendrocytes, astrocytes, Schwann cells and microglia/macrophages, in the CNS-PNS transitional zone of TREZ. Changes in glial cell plasticity in the TREZ after compression injury might be involved in TN pathogenesis.
Collapse
Affiliation(s)
- DaoShu Luo
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China.,Key Laboratory of Brain Aging and Neurodegenerative Diseases of Fujian Province, Fuzhou, 350122, China
| | - Ren Lin
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China.,Key Laboratory of Brain Aging and Neurodegenerative Diseases of Fujian Province, Fuzhou, 350122, China
| | - LiLi Luo
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - QiuHua Li
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Ting Chen
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - RongHui Qiu
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - YunQing Li
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China. .,Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, The Fourth Military Medical University, No. 169, West Changle Road, Xi'an, 710032, China.
| |
Collapse
|
143
|
Silverman SM, Ma W, Wang X, Zhao L, Wong WT. C3- and CR3-dependent microglial clearance protects photoreceptors in retinitis pigmentosa. J Exp Med 2019; 216:1925-1943. [PMID: 31209071 PMCID: PMC6683998 DOI: 10.1084/jem.20190009] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/04/2019] [Accepted: 05/09/2019] [Indexed: 12/24/2022] Open
Abstract
Silverman et al. demonstrate that complement activation features prominently in retinitis pigmentosa in close association with activated microglia. This response mediates adaptive neuroprotection for photoreceptors by facilitating a C3-CR3–dependent clearance of apoptotic photoreceptors by microglial phagocytosis. Complement activation has been implicated as contributing to neurodegeneration in retinal and brain pathologies, but its role in retinitis pigmentosa (RP), an inherited and largely incurable photoreceptor degenerative disease, is unclear. We found that multiple complement components were markedly up-regulated in retinas with human RP and the rd10 mouse model, coinciding spatiotemporally with photoreceptor degeneration, with increased C3 expression and activation localizing to activated retinal microglia. Genetic ablation of C3 accelerated structural and functional photoreceptor degeneration and altered retinal inflammatory gene expression. These phenotypes were recapitulated by genetic deletion of CR3, a microglia-expressed receptor for the C3 activation product iC3b, implicating C3-CR3 signaling as a regulator of microglia–photoreceptor interactions. Deficiency of C3 or CR3 decreased microglial phagocytosis of apoptotic photoreceptors and increased microglial neurotoxicity to photoreceptors, demonstrating a novel adaptive role for complement-mediated microglial clearance of apoptotic photoreceptors in RP. These homeostatic neuroinflammatory mechanisms are relevant to the design and interpretation of immunomodulatory therapeutic approaches to retinal degenerative disease.
Collapse
Affiliation(s)
- Sean M Silverman
- Section on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, MD
| | - Wenxin Ma
- Section on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, MD
| | - Xu Wang
- Section on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, MD
| | - Lian Zhao
- Section on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, MD
| | - Wai T Wong
- Section on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
144
|
Roboon J, Hattori T, Ishii H, Takarada-Iemata M, Le TM, Shiraishi Y, Ozaki N, Yamamoto Y, Sugawara A, Okamoto H, Higashida H, Kitao Y, Hori O. Deletion of CD38 Suppresses Glial Activation and Neuroinflammation in a Mouse Model of Demyelination. Front Cell Neurosci 2019; 13:258. [PMID: 31244614 PMCID: PMC6563778 DOI: 10.3389/fncel.2019.00258] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 05/23/2019] [Indexed: 01/09/2023] Open
Abstract
CD38 is an enzyme that catalyzes the synthesis of cyclic adenosine diphosphate-ribose from nicotinamide adenine dinucleotide (NAD+). We recently reported that this molecule regulates the maturation and differentiation of glial cells such as astrocytes and oligodendrocytes (OLs) in the developing brain. To analyze its role in the demyelinating situation, we employed cuprizone (CPZ)-induced demyelination model in mice, which is characterized by oligodendrocyte-specific apoptosis, followed by the strong glial activation, demyelination, and repopulation of OLs. By using this model, we found that CD38 was upregulated in both astrocytes and microglia after CPZ administration. Experiments using wild-type and CD38 knockout (KO) mice, together with those using cultured glial cells, revealed that CD38 deficiency did not affect the initial decrease of the number of OLs, while it attenuated CPZ-induced demyelination, and neurodegeneration. Importantly, the clearance of the degraded myelin and oligodendrocyte repopulation were also reduced in CD38 KO mice. Further experiments revealed that these observations were associated with reduced levels of glial activation and inflammatory responses including phagocytosis, most likely through the enhanced level of NAD+ in CD38-deleted condition. Our results suggest that CD38 and NAD+ in the glial cells play a critical role in the demyelination and subsequent oligodendrocyte remodeling through the modulation of glial activity and neuroinflammation.
Collapse
Affiliation(s)
- Jureepon Roboon
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Tsuyoshi Hattori
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Hiroshi Ishii
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Mika Takarada-Iemata
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Thuong Manh Le
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yoshitake Shiraishi
- Department of Functional Anatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Noriyuki Ozaki
- Department of Functional Anatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yasuhiko Yamamoto
- Department of Biochemistry and Molecular Vascular Biology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Akira Sugawara
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroshi Okamoto
- Department of Biochemistry and Molecular Vascular Biology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan.,Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Haruhiro Higashida
- Research Center for Child Mental Development, Kanazawa University, Kanazawa, Japan
| | - Yasuko Kitao
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Osamu Hori
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
145
|
Kim JY, Yoon JY, Sugiura Y, Lee SK, Park JD, Song GJ, Yang HJ. Dendropanax morbiferus leaf extract facilitates oligodendrocyte development. ROYAL SOCIETY OPEN SCIENCE 2019; 6:190266. [PMID: 31312492 PMCID: PMC6599778 DOI: 10.1098/rsos.190266] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 06/04/2019] [Indexed: 06/10/2023]
Abstract
Treatment of multiple sclerosis is effective when anti-inflammatory, neuroprotective and regenerative strategies are combined. Dendropanax morbiferus (DM) has anti-inflammatory, anti-oxidative properties, which may be beneficial for multiple sclerosis. However, there have been no reports on the effects of DM on myelination, which is critical for regenerative processes. To know whether DM benefits myelination, we checked differentiation and myelination of oligodendrocytes (OLs) in various primary culture systems treated with DM leaf EtOH extracts or control. DM extracts increased the OL membrane size in the mixed glial and pure OL precursor cell (OPC) cultures and changed OL-lineage gene expression patterns in the OPC cultures. Western blot analysis of DM-treated OPC cultures showed upregulation of MBP and phosphorylation of ERK1/2. In myelinating cocultures, DM extracts enhanced OL differentiation, followed by increased axonal contacts and myelin gene upregulations such as Myrf, CNP and PLP. Phytochemical analysis by LC-MS/MS identified multiple components from DM extracts, containing bioactive molecules such as quercetin, cannabidiol, etc. Our results suggest DM extracts enhance OL differentiation, followed by an increase in membrane size and axonal contacts, thereby indicating enhanced myelination. In addition, we found that DM extracts contain multiple bioactive components, warranting further studies in relation to finding effective components for enhancing myelination.
Collapse
Affiliation(s)
- Ji-Young Kim
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul 120-749, Republic of Korea
| | - Ju-Young Yoon
- Department of Integrative Biosciences, University of Brain Education, Cheonan 31228, Republic of Korea
| | - Yuki Sugiura
- Department of Biochemistry and Integrative Medical Biology, School of Medicine, Keio University, Tokyo 160-8582, Japan
| | - Soo-Kyoung Lee
- Department of Health Science and Daily Sports, Global Cyber University, Cheonan 31228, Republic of Korea
| | - Jae-Don Park
- Cheju Halla University, Jeju 63092, Republic of Korea
| | - Gyun-Jee Song
- Department of Medical Science, International St Mary's Hospital, Catholic Kwandong University, Gangneung, Republic of Korea
| | - Hyun-Jeong Yang
- Department of Integrative Biosciences, University of Brain Education, Cheonan 31228, Republic of Korea
- Korea Institute of Brain Science, Seoul, Republic of Korea
| |
Collapse
|
146
|
Neural Stem Cells of the Subventricular Zone Contribute to Neuroprotection of the Corpus Callosum after Cuprizone-Induced Demyelination. J Neurosci 2019; 39:5481-5492. [PMID: 31138656 DOI: 10.1523/jneurosci.0227-18.2019] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 03/20/2019] [Accepted: 03/21/2019] [Indexed: 12/13/2022] Open
Abstract
Myelin loss occurring in demyelinating diseases, including multiple sclerosis, is the leading cause of long-lasting neurological disability in adults. While endogenous remyelination, driven by resident oligodendrocyte precursor cells (OPCs), might partially compensate myelin loss in the early phases of demyelinating disorders, this spontaneous reparative potential fails at later stages. To investigate the cellular mechanisms sustaining endogenous remyelination in demyelinating disorders, we focused our attention on endogenous neural precursor cells (eNPCs) located within the subventricular zone (SVZ) since this latter area is considered one of the primary sources of new OPCs in the adult forebrain. First, we fate mapped SVZ-eNPCs in cuprizone-induced demyelination and found that SVZ endogenous neural stem/precursor cells are recruited during the remyelination phase to the corpus callosum (CC) and are capable of forming new oligodendrocytes. When we ablated SVZ-derived eNPCs during cuprizone-induced demyelination in female mice, the animals displayed reduced numbers of oligodendrocytes within the lesioned CC. Although this reduction in oligodendrocytes did not impact the ensuing remyelination, eNPC-ablated mice experienced increased axonal loss. Our results indicate that, in toxic models of demyelination, SVZ-derived eNPCs contribute to support axonal survival.SIGNIFICANCE STATEMENT One of the significant challenges in MS research is to understand the detrimental mechanisms leading to the failure of CNS tissue regeneration during disease progression. One possible explanation is the inability of recruited oligodendrocyte precursor cells (OPCs) to complete remyelination and to sustain axonal survival. The contribution of endogenous neural precursor cells (eNPCs) located in the subventricular zone (SVZ) to generate new OPCs in the lesion site has been debated. Using transgenic mice to fate map and to selectively kill SVZ-derived eNPCs in the cuprizone demyelination model, we observed migration of SVZ-eNPCs after injury and their contribution to oligodendrogenesis and axonal survival. We found that eNPCs are dispensable for remyelination but protect partially from increased axonal loss.
Collapse
|
147
|
Templeton N, Kivell B, McCaughey-Chapman A, Connor B, La Flamme AC. Clozapine administration enhanced functional recovery after cuprizone demyelination. PLoS One 2019; 14:e0216113. [PMID: 31071102 PMCID: PMC6508663 DOI: 10.1371/journal.pone.0216113] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 04/15/2019] [Indexed: 11/19/2022] Open
Abstract
The atypical antipsychotic agent, clozapine, is used to treat a variety of neurological disorders including schizophrenia and Parkinson's disease and readily crosses the blood brain barrier to interact with a wide range of neuroreceptors including those for dopamine and serotonin. Recent work has shown that clozapine can reduce neuroinflammation in experimental autoimmune encephalomyelitis, a neuroinflammatory model of multiple sclerosis (MS) and mediates its effects in the central nervous system. To further characterise the protection provided by clozapine, the cuprizone model of demyelination was used to assess the effect of clozapine treatment on the cellular events surrounding demyelination and remyelination. Using this model of non-immune demyelination, we found that clozapine administration was unable to prevent demyelination, but when administered post demyelination, was able to enhance the rate of functional recovery. The more rapid improvement of clozapine-treated mice correlated with a decreased level of astrocyte and microglial activation but only modestly enhanced remyelination. Together, these studies highlight the potential of clozapine to support enhanced functional recovery after demyelination, such as that occurring during MS.
Collapse
Affiliation(s)
- Nikki Templeton
- Centre for Biodiscovery, School of Biological Sciences and School of Chemical and Physical Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - Bronwyn Kivell
- Centre for Biodiscovery, School of Biological Sciences and School of Chemical and Physical Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - Amy McCaughey-Chapman
- Department of Pharmacology and Clinical Pharmacology, Centre for Brain Research, School of Medical Sciences, FMHS, the University of Auckland, Auckland, New Zealand
| | - Bronwen Connor
- Department of Pharmacology and Clinical Pharmacology, Centre for Brain Research, School of Medical Sciences, FMHS, the University of Auckland, Auckland, New Zealand
| | - Anne Camille La Flamme
- Centre for Biodiscovery, School of Biological Sciences and School of Chemical and Physical Sciences, Victoria University of Wellington, Wellington, New Zealand
- Malaghan Institute for Medical Research, Wellington, New Zealand
- * E-mail:
| |
Collapse
|
148
|
Zhou YQ, Liu DQ, Chen SP, Sun J, Zhou XR, Xing C, Ye DW, Tian YK. The Role of CXCR3 in Neurological Diseases. Curr Neuropharmacol 2019; 17:142-150. [PMID: 29119926 PMCID: PMC6343204 DOI: 10.2174/1570159x15666171109161140] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 06/22/2017] [Accepted: 11/07/2017] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Neurological diseases have become an obvious challenge due to insufficient therapeutic intervention. Therefore, novel drugs for various neurological disorders are in desperate need. Recently, compelling evidence has demonstrated that chemokine receptor CXCR3, which is a G protein-coupled receptor in the CXC chemokine receptor family, may play a pivotal role in the development of neurological diseases. The aim of this review is to provide evidence for the potential of CXCR3 as a therapeutic target for neurological diseases. METHODS English journal articles that focused on the invovlement of CXCR3 in neurological diseases were searched via PubMed up to May 2017. Moreover, reference lists from identified articles were included for overviews. RESULTS The expression level of CXCR3 in T cells was significantly elevated in several neurological diseases, including multiple sclerosis (MS), glioma, Alzheimer's disease (AD), chronic pain, human T-lymphotropic virus type 1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) and bipolar disorder. CXCR3 antagonists showed therapeutic effects in these neurological diseases. CONCLUSION These studies provided hard evidence that CXCR3 plays a vital role in the pathogenesis of MS, glioma, AD, chronic pain, HAM/TSP and bipolar disorder. CXCR3 is a crucial molecule in neuroinflammatory and neurodegenerative diseases. It regulates the activation of infiltrating cells and resident immune cells. However, the exact functions of CXCR3 in neurological diseases are inconclusive. Thus, it is important to understand the topic of chemokines and the scope of their activity in neurological diseases.
Collapse
Affiliation(s)
- Ya-Qun Zhou
- Anesthesiology Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dai-Qiang Liu
- Anesthesiology Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shu-Ping Chen
- Anesthesiology Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Sun
- Anesthesiology Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xue-Rong Zhou
- Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cui Xing
- Department of Obstetrics & Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Da-Wei Ye
- Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Ke Tian
- Anesthesiology Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
149
|
Galloway DA, Phillips AEM, Owen DRJ, Moore CS. Phagocytosis in the Brain: Homeostasis and Disease. Front Immunol 2019; 10:790. [PMID: 31040847 PMCID: PMC6477030 DOI: 10.3389/fimmu.2019.00790] [Citation(s) in RCA: 206] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 03/26/2019] [Indexed: 12/28/2022] Open
Abstract
Microglia are resident macrophages of the central nervous system and significantly contribute to overall brain function by participating in phagocytosis during development, homeostasis, and diseased states. Phagocytosis is a highly complex process that is specialized for the uptake and removal of opsonized and non-opsonized targets, such as pathogens, apoptotic cells, and cellular debris. While the role of phagocytosis in mediating classical innate and adaptive immune responses has been known for decades, it is now appreciated that phagocytosis is also critical throughout early neural development, homeostasis, and initiating repair mechanisms. As such, modulating phagocytic processes has provided unexplored avenues with the intent of developing novel therapeutics that promote repair and regeneration in the CNS. Here, we review the functional consequences that phagocytosis plays in both the healthy and diseased CNS, and summarize how phagocytosis contributes to overall pathophysiological mechanisms involved in brain injury and repair.
Collapse
Affiliation(s)
- Dylan A Galloway
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Alexandra E M Phillips
- Division of Brain Sciences, Department of Medicine Hammersmith Hospital, Imperial College London, London, United Kingdom
| | - David R J Owen
- Division of Brain Sciences, Department of Medicine Hammersmith Hospital, Imperial College London, London, United Kingdom
| | - Craig S Moore
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada
| |
Collapse
|
150
|
Delayed Astrogliosis Associated with Reduced M1 Microglia Activation in Matrix Metalloproteinase 12 Knockout Mice during Theiler's Murine Encephalomyelitis. Int J Mol Sci 2019; 20:ijms20071702. [PMID: 30959793 PMCID: PMC6480673 DOI: 10.3390/ijms20071702] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/20/2019] [Accepted: 04/02/2019] [Indexed: 12/30/2022] Open
Abstract
Theiler’s murine encephalomyelitis (TME) represents a versatile animal model for studying the pathogenesis of demyelinating diseases such as multiple sclerosis. Hallmarks of TME are demyelination, astrogliosis, as well as inflammation. Previous studies showed that matrix metalloproteinase 12 knockout (Mmp12−/−) mice display an ameliorated clinical course associated with reduced demyelination. The present study aims to elucidate the impact of MMP12 deficiency in TME with special emphasis on astrogliosis, macrophages infiltrating the central nervous system (CNS), and the phenotype of microglia/macrophages (M1 or M2). SJL wild-type and Mmp12−/− mice were infected with TME virus (TMEV) or vehicle (mock) and euthanized at 28 and 98 days post infection (dpi). Immunohistochemistry or immunofluorescence of cervical and thoracic spinal cord for detecting glial fibrillary acidic protein (GFAP), ionized calcium-binding adaptor molecule 1 (Iba1), chemokine receptor 2 (CCR2), CD107b, CD16/32, and arginase I was performed and quantitatively evaluated. Statistical analyses included the Kruskal–Wallis test followed by Mann–Whitney U post hoc tests. TMEV-infected Mmp12−/− mice showed transiently reduced astrogliosis in association with a strong trend (p = 0.051) for a reduced density of activated/reactive microglia/macrophages compared with wild-type mice at 28 dpi. As astrocytes are an important source of cytokine production, including proinflammatory cytokines triggering or activating phagocytes, the origin of intralesional microglia/macrophages as well as their phenotype were determined. Only few phagocytes in wild-type and Mmp12−/− mice expressed CCR2, indicating that the majority of phagocytes are represented by microglia. In parallel to the reduced density of activated/reactive microglia at 98 dpi, TMEV-infected Mmp12−/− showed a trend (p = 0.073) for a reduced density of M1 (CD16/32- and CD107b-positive) microglia, while no difference regarding the density of M2 (arginase I- and CD107b-positive) cells was observed. However, a dominance of M1 cells was detected in the spinal cord of TMEV-infected mice at all time points. Reduced astrogliosis in Mmp12−/− mice was associated with a reduced density of activated/reactive microglia and a trend for a reduced density of M1 cells. This indicates that MMP12 plays an important role in microglia activation, polarization, and migration as well as astrogliosis and microglia/astrocyte interaction.
Collapse
|