Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 596
Filter
1.
Int Immunopharmacol ; 122: 110674, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37481846

ABSTRACT

We previously demonstrated that experimental traumatic occlusion (ETO) induces a long-lasting nociceptive response. These findings were associated with altered neuronal patterns and suggestive satellite glial cell activation. This study aimed to elucidate the activation of satellite glial cells following ETO in the trigeminal ganglion. Moreover, we explored the involvement of resident and infiltrating cells in trigeminal ganglion in ETO. Finally, we investigated the overexpression of purinergic signaling and the CX3CL1/CX3CR1 axis. RT-qPCR and electrophoresis showed overexpression of GFAP in the trigeminal ganglion (TG), and immunohistochemistry corroborated these findings, demonstrating SGCs activation. ELISA reveals enhanced levels of TNF-α and IL-1ß in TG after 28 d of ETO. In trigeminal ganglia, ETO groups improved the release of CX3CL1, and immunohistochemistry showed higher CX3CR1+ -immunoreactive cells in ETO groups. Immunohistochemistry and electrophoresis of the P2X7 receptor were found in ETO groups. The mRNA levels of IBA1 are upregulated in the 0.7-mm ETO group, while immunohistochemistry showed higher IBA1+ -immunoreactive cells in both ETO groups. The expression of CD68 by electrophoresis and immunohistochemistry was observed in the ETO groups. For last, ELISA revealed increased levels of IL-6, IL-12, and CCL2 in the TG of ETO groups. Furthermore, the mRNA expression revealed augmented transcription factors and cytokines associated with lymphocyte activation, such as RORγt, IL-17, Tbet, IFNγ, FOXP3, and IL-10. The findings of this study suggested that ETO activates SGCs in TG, and purinergic signaling and CX3CL1/CX3CR1 axis were upregulated. We uncovered the involvement of a distinct subtype of macrophages, named sensory neuron-associated macrophage activation (sNMAs), and detected an expanded number of infiltrated macrophages onto TG. These findings indicate that ETO induces chronic/persistent immune response.


Subject(s)
Lymphocyte Activation , Macrophage Activation , Nociceptive Pain , Oligodendroglia , Trigeminal Ganglion , Trigeminal Ganglion/injuries , Nociceptive Pain/immunology , CX3C Chemokine Receptor 1/metabolism , Chemokine CX3CL1/metabolism , Animals , Rats , Glial Fibrillary Acidic Protein/metabolism , Male , Rats, Wistar , Oligodendroglia/immunology , Receptors, Purinergic P2X/metabolism
2.
Cell Rep ; 37(13): 110158, 2021 12 28.
Article in English | MEDLINE | ID: mdl-34965428

ABSTRACT

Non-neuronal responses in neurodegenerative disease have received increasing attention as important contributors to disease pathogenesis and progression. Here we utilize single-cell RNA sequencing to broadly profile 13 cell types in three different mouse models of Alzheimer disease (AD), capturing the effects of tau-only, amyloid-only, or combined tau-amyloid pathology. We highlight microglia, oligodendrocyte, astrocyte, and T cell responses and compare them across these models. Notably, we identify two distinct transcriptional states for oligodendrocytes emerging differentially across disease models, and we determine their spatial distribution. Furthermore, we explore the impact of Trem2 deletion in the context of combined pathology. Trem2 knockout mice exhibit severely blunted microglial responses to combined tau and amyloid pathology, but responses from non-microglial cell types (oligodendrocytes, astrocytes, and T cells) are relatively unchanged. These results delineate core transcriptional states that are engaged in response to AD pathology, and how they are influenced by a key AD risk gene, Trem2.


Subject(s)
Alzheimer Disease/pathology , Amyloid/chemistry , Astrocytes/pathology , Membrane Glycoproteins/physiology , Oligodendroglia/pathology , Receptors, Immunologic/physiology , T-Lymphocytes/immunology , tau Proteins/metabolism , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Animals , Astrocytes/immunology , Astrocytes/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligodendroglia/immunology , Oligodendroglia/metabolism
3.
Cells ; 10(4)2021 04 13.
Article in English | MEDLINE | ID: mdl-33924474

ABSTRACT

Multiple sclerosis (MS) is a chronic inflammatory and neurodegenerative disease of the central nervous system (CNS) affecting more than two million people worldwide. In MS, oligodendrocytes and myelin sheaths are destroyed by autoimmune-mediated inflammation, while remyelination is impaired. Recent investigations of post-mortem tissue suggest that Fibroblast growth factor (FGF) signaling may regulate inflammation and myelination in MS. FGF2 expression seems to correlate positively with macrophages/microglia and negatively with myelination; FGF1 was suggested to promote remyelination. In myelin oligodendrocyte glycoprotein (MOG)35-55-induced experimental autoimmune encephalomyelitis (EAE), systemic deletion of FGF2 suggested that FGF2 may promote remyelination. Specific deletion of FGF receptors (FGFRs) in oligodendrocytes in this EAE model resulted in a decrease of lymphocyte and macrophage/microglia infiltration as well as myelin and axon degeneration. These effects were mediated by ERK/Akt phosphorylation, a brain-derived neurotrophic factor, and downregulation of inhibitors of remyelination. In the first part of this review, the most important pharmacotherapeutic principles for MS will be illustrated, and then we will review recent advances made on FGF signaling in MS. Thus, we will suggest application of FGFR inhibitors, which are currently used in Phase II and III cancer trials, as a therapeutic option to reduce inflammation and induce remyelination in EAE and eventually MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/genetics , Fibroblast Growth Factor 2/genetics , Microglia/immunology , Multiple Sclerosis/genetics , Myelin Sheath/immunology , Receptor, Fibroblast Growth Factor, Type 2/genetics , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/immunology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Fibroblast Growth Factor 2/deficiency , Gene Expression Regulation , Humans , Immunologic Factors/therapeutic use , Mice, Knockout , Microglia/drug effects , Microglia/pathology , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Myelin Sheath/drug effects , Myelin Sheath/pathology , Myelin-Oligodendrocyte Glycoprotein/administration & dosage , Oligodendroglia/drug effects , Oligodendroglia/immunology , Oligodendroglia/pathology , Peptide Fragments/administration & dosage , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 2/immunology , Remyelination/drug effects , Remyelination/genetics , Remyelination/immunology , Signal Transduction
4.
Elife ; 102021 03 23.
Article in English | MEDLINE | ID: mdl-33752802

ABSTRACT

The inflammatory environment of demyelinated lesions in multiple sclerosis (MS) patients contributes to remyelination failure. Inflammation activates a cytoprotective pathway, the integrated stress response (ISR), but it remains unclear whether enhancing the ISR can improve remyelination in an inflammatory environment. To examine this possibility, the remyelination stage of experimental autoimmune encephalomyelitis (EAE), as well as a mouse model that incorporates cuprizone-induced demyelination along with CNS delivery of the proinflammatory cytokine IFN-γ were used here. We demonstrate that either genetic or pharmacological ISR enhancement significantly increased the number of remyelinating oligodendrocytes and remyelinated axons in the inflammatory lesions. Moreover, the combined treatment of the ISR modulator Sephin1 with the oligodendrocyte differentiation enhancing reagent bazedoxifene increased myelin thickness of remyelinated axons to pre-lesion levels. Taken together, our findings indicate that prolonging the ISR protects remyelinating oligodendrocytes and promotes remyelination in the presence of inflammation, suggesting that ISR enhancement may provide reparative benefit to MS patients.


Subject(s)
Central Nervous System/immunology , Cuprizone/adverse effects , Demyelinating Diseases/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Remyelination/physiology , Animals , Axons/immunology , Demyelinating Diseases/chemically induced , Disease Models, Animal , Female , Inflammation/genetics , Inflammation/immunology , Interferon-gamma/genetics , Interferon-gamma/metabolism , Male , Mice , Oligodendroglia/immunology , Remyelination/genetics
5.
Cell Death Dis ; 12(2): 166, 2021 02 08.
Article in English | MEDLINE | ID: mdl-33558485

ABSTRACT

A leading cause of preterm birth is the exposure to systemic inflammation (maternal/fetal infection), which leads to neuroinflammation and white matter injury (WMI). A wide range of cytokines and chemokines are expressed and upregulated in oligodendrocytes (OLs) in response to inflammation and numerous reports show that OLs express several receptors for immune related molecules, which enable them to sense inflammation and to react. However, the role of OL immune response in WMI is unclear. Here, we focus our study on toll-like receptor-3 (TLR3) that is activated by double-strand RNA (dsRNA) and promotes neuroinflammation. Despite its importance, its expression and role in OLs remain unclear. We used an in vivo mouse model, which mimics inflammation-mediated WMI of preterm born infants consisting of intraperitoneal injection of IL-1ß from P1 to P5. In the IL-1ß-treated animals, we observed the upregulation of Tlr3, IL-1ß, IFN-ß, Ccl2, and Cxcl10 in both PDGFRα+ and O4+ sorted cells. This upregulation was higher in O4+ immature OLs (immOLs) as compared to PDGFRα+ OL precursor cells (OPCs), suggesting a different sensitivity to neuroinflammation. These observations were confirmed in OL primary cultures: cells treated with TLR3 agonist Poly(I:C) during differentiation showed a stronger upregulation of Ccl2 and Cxcl10 compared to cells treated during proliferation and led to decreased expression of myelin genes. Finally, OLs were able to modulate microglia phenotype and function depending on their maturation state as assessed by qPCR using validated markers for immunomodulatory, proinflammatory, and anti-inflammatory phenotypes and by phagocytosis and morphological analysis. These results show that during inflammation the response of OLs can play an autonomous role in blocking their own differentiation: in addition, the immune activation of OLs may play an important role in shaping the response of microglia during inflammation.


Subject(s)
Cell Differentiation , Cell Proliferation , Encephalitis/metabolism , Leukoencephalopathies/metabolism , Oligodendroglia/metabolism , Toll-Like Receptor 3/metabolism , White Matter/metabolism , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Encephalitis/genetics , Encephalitis/immunology , Encephalitis/pathology , Female , Inflammation Mediators/metabolism , Leukoencephalopathies/genetics , Leukoencephalopathies/immunology , Leukoencephalopathies/pathology , Male , Mice , Microglia/immunology , Microglia/metabolism , Microglia/pathology , Oligodendroglia/drug effects , Oligodendroglia/immunology , Oligodendroglia/pathology , Poly I-C/pharmacology , Pregnancy , Premature Birth , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Signal Transduction , Toll-Like Receptor 3/agonists , White Matter/drug effects , White Matter/immunology , White Matter/pathology
6.
Int J Neurosci ; 131(12): 1221-1230, 2021 Dec.
Article in English | MEDLINE | ID: mdl-32571126

ABSTRACT

Efficient communication between the glial cells and neurons is a bi-directional process that is essential for conserving normal functioning in the central nervous system (CNS). Neurons dynamically regulate other brain cells in the healthy brain, yet little is known about the first pathways involving oligodendrocytes and neurons. Oligodendrocytes are the myelin-forming cells in the CNS that are needed for the propagation of action potentials along axons and additionally serve to support neurons by neurotrophic factors (NFTs). In demyelinating diseases, like multiple sclerosis (MS), oligodendrocytes are thought to be the victims. Axonal damage begins early and remains silent for years, and neurological disability develops when a threshold of axonal loss is reached, and the compensatory mechanisms are depleted. Three hypotheses have been proposed to explain axonal damage: 1) the damage is caused by an inflammatory process; 2) there is an excessive accumulation of intra-axonal calcium levels; and, 3) demyelinated axons evolve to a degenerative process resulting from the lack of trophic support provided by myelin or myelin-forming cells. Although MS was traditionally considered to be a white matter disease, the demyelination process also occurs in the cerebral cortex. Recent data supports the notion that initial response is triggered by CNS injury. Thus, the understanding of the role of neuron-glial neurophysiology would help provide us with further explanations. We should take in account the suggestion that MS is in part an autoimmune disease that involves genetic and environmental factors, and the pathological response leads to demyelination, axonal loss and inflammatory infiltrates.


Subject(s)
Electrophysiological Phenomena/physiology , Immunity/physiology , Multiple Sclerosis/immunology , Multiple Sclerosis/physiopathology , Oligodendroglia/physiology , Animals , Electrophysiological Phenomena/immunology , Humans , Immunity/immunology , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Oligodendroglia/immunology , Oligodendroglia/metabolism , Oligodendroglia/pathology
7.
Front Immunol ; 11: 572186, 2020.
Article in English | MEDLINE | ID: mdl-33117365

ABSTRACT

Multiple Sclerosis (MS) is traditionally considered an autoimmune-mediated demyelinating disease, the pathoetiology of which is unknown. However, the key question remains whether autoimmunity is the initiator of the disease (outside-in) or the consequence of a slow and as yet uncharacterized cytodegeneration (oligodendrocytosis), which leads to a subsequent immune response (inside-out). Experimental autoimmune encephalomyelitis has been used to model the later stages of MS during which the autoimmune involvement predominates. In contrast, the cuprizone (CPZ) model is used to model early stages of the disease during which oligodendrocytosis and demyelination predominate and are hypothesized to precede subsequent immune involvement in MS. Recent studies combining a boost, or protection, to the immune system with disruption of the blood brain barrier have shown CPZ-induced oligodendrocytosis with a subsequent immune response. In this Perspective, we review these recent advances and discuss the likelihood of an inside-out vs. an outside-in pathoetiology of MS.


Subject(s)
Central Nervous System/physiology , Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , Oligodendroglia/immunology , Animals , Autoimmunity , Cuprizone , Demyelinating Diseases , Disease Models, Animal , Disease Progression , Humans , Mice
8.
Trends Immunol ; 41(9): 794-804, 2020 09.
Article in English | MEDLINE | ID: mdl-32800704

ABSTRACT

An interaction network exists among cells within the brain, maintaining brain homeostasis and ensuring its functional plasticity. In addition to neurons, participating cells include astrocytes, oligodendrocytes, and microglia. Peripheral immune cells, such as monocytes and lymphocytes, have also been found to play an important role in supporting the brain in health and assisting in its repair. Here, we describe the multiple immune-specific modes of cellular dialogue among cells within the mammalian brain and their crosstalk with the periphery in both health and disease. We further suggest that interventions directed at boosting the peripheral immune response can restore the balance between the brain and the immune system and can rewire their communication to modify chronic neurodegenerative diseases.


Subject(s)
Alzheimer Disease , Brain , Immunotherapy , Alzheimer Disease/therapy , Animals , Astrocytes/immunology , Brain/cytology , Brain/immunology , Cell Communication/immunology , Humans , Microglia/immunology , Oligodendroglia/immunology
9.
Proc Natl Acad Sci U S A ; 117(27): 15902-15910, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32571951

ABSTRACT

Neurotropic strains of mouse hepatitis virus (MHV), a coronavirus, cause acute and chronic demyelinating encephalomyelitis with similarities to the human disease multiple sclerosis. Here, using a lineage-tracking system, we show that some cells, primarily oligodendrocytes (OLs) and oligodendrocyte precursor cells (OPCs), survive the acute MHV infection, are associated with regions of demyelination, and persist in the central nervous system (CNS) for at least 150 d. These surviving OLs express major histocompatibility complex (MHC) class I and other genes associated with an inflammatory response. Notably, the extent of inflammatory cell infiltration was variable, dependent on anatomic location within the CNS, and without obvious correlation with numbers of surviving cells. We detected more demyelination in regions with larger numbers of T cells and microglia/macrophages compared to those with fewer infiltrating cells. Conversely, in regions with less inflammation, these previously infected OLs more rapidly extended processes, consistent with normal myelinating function. Together, these results show that OLs are inducers as well as targets of the host immune response and demonstrate how a CNS infection, even after resolution, can induce prolonged inflammatory changes with CNS region-dependent impairment in remyelination.


Subject(s)
Central Nervous System/immunology , Coronavirus Infections/complications , Demyelinating Diseases/etiology , Oligodendroglia/immunology , Animals , Coronavirus Infections/immunology , Histocompatibility Antigens Class I/metabolism , Luminescent Proteins , Male , Mice , Murine hepatitis virus , Oligodendroglia/metabolism , Red Fluorescent Protein
10.
Sci Immunol ; 5(48)2020 06 05.
Article in English | MEDLINE | ID: mdl-32503879

ABSTRACT

Pathogenic autoantibodies causing encephalopathy modify the expression of the glucose transporter in oligodendrocytes.


Subject(s)
Autoantibodies/immunology , Oligodendroglia/immunology , Sugars/immunology , Glucose Transport Proteins, Facilitative/genetics , Glucose Transport Proteins, Facilitative/immunology , Humans , Oligodendroglia/pathology
11.
Pharmacol Res ; 159: 104997, 2020 09.
Article in English | MEDLINE | ID: mdl-32534098

ABSTRACT

Myelin loss is the hallmark of the demyelinating disease multiple sclerosis (MS) and plays a significant role in multiple neurodegenerative diseases. A common factor in all neuropathologies is the central role of microglia, the intrinsic immune cells of the central nervous system (CNS). Microglia are activated in pathology and can have both pro- and anti-inflammatory functions. Here, we examined the effects of the flavonoid agathisflavone on microglia and remyelination in the cerebellar slice model following lysolecithin induced demyelination. Notably, agathisflavone enhances remyelination and alters microglial activation state, as determined by their morphology and cytokine profile. Furthermore, these effects of agathisflavone on remyelination and microglial activation were inhibited by blockade of estrogen receptor α. Thus, our results identify agathisflavone as a novel compound that may act via ER to regulate microglial activation and enhance remyelination and repair.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Biflavonoids/pharmacology , Cerebellum/drug effects , Microglia/drug effects , Myelin Sheath/metabolism , Neuroimmunomodulation/drug effects , Oligodendroglia/drug effects , Remyelination/drug effects , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cerebellum/immunology , Cerebellum/metabolism , Cerebellum/pathology , Cytokines/metabolism , Estrogen Receptor alpha/drug effects , Estrogen Receptor alpha/metabolism , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Lysophosphatidylcholines/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/immunology , Microglia/metabolism , Microglia/pathology , Oligodendroglia/immunology , Oligodendroglia/metabolism , Oligodendroglia/pathology , Phenotype , Tissue Culture Techniques
12.
Proc Natl Acad Sci U S A ; 117(16): 9082-9093, 2020 04 21.
Article in English | MEDLINE | ID: mdl-32253301

ABSTRACT

Current multiple sclerosis (MS) medications are mainly immunomodulatory, having little or no effect on neuroregeneration of damaged central nervous system (CNS) tissue; they are thus primarily effective at the acute stage of disease, but much less so at the chronic stage. An MS therapy that has both immunomodulatory and neuroregenerative effects would be highly beneficial. Using multiple in vivo and in vitro strategies, in the present study we demonstrate that ursolic acid (UA), an antiinflammatory natural triterpenoid, also directly promotes oligodendrocyte maturation and CNS myelin repair. Oral treatment with UA significantly decreased disease severity and CNS inflammation and demyelination in experimental autoimmune encephalomyelitis (EAE), an animal model of MS. Importantly, remyelination and neural repair in the CNS were observed even after UA treatment was started on day 60 post immunization when EAE mice had full-blown demyelination and axonal damage. UA treatment also enhanced remyelination in a cuprizone-induced demyelination model in vivo and brain organotypic slice cultures ex vivo and promoted oligodendrocyte maturation in vitro, indicating a direct myelinating capacity. Mechanistically, UA induced promyelinating neurotrophic factor CNTF in astrocytes by peroxisome proliferator-activated receptor γ(PPARγ)/CREB signaling, as well as by up-regulation of myelin-related gene expression during oligodendrocyte maturation via PPARγ activation. Together, our findings demonstrate that UA has significant potential as an oral antiinflammatory and neural repair agent for MS, especially at the chronic-progressive stage.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Immunomodulation/drug effects , Multiple Sclerosis/drug therapy , Remyelination/drug effects , Triterpenes/pharmacology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Cell Differentiation/drug effects , Corpus Callosum/drug effects , Corpus Callosum/pathology , Cuprizone/toxicity , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Gene Expression Regulation/drug effects , Humans , Male , Mice , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Myelin Sheath/drug effects , Myelin Sheath/pathology , Oligodendroglia/drug effects , Oligodendroglia/immunology , Oligodendroglia/pathology , PPAR gamma/metabolism , Triterpenes/therapeutic use , Ursolic Acid
13.
Cells ; 9(4)2020 04 14.
Article in English | MEDLINE | ID: mdl-32295179

ABSTRACT

Oligodendrocytes are the myelinating cells of the central nervous system (CNS). These cells arise during the embryonic development by the specification of the neural stem cells to oligodendroglial progenitor cells (OPC); newly formed OPC proliferate, migrate, differentiate, and mature to myelinating oligodendrocytes in the perinatal period. It is known that progesterone promotes the proliferation and differentiation of OPC in early postnatal life through the activation of the intracellular progesterone receptor (PR). Progesterone supports nerve myelination after spinal cord injury in adults. However, the role of progesterone in embryonic OPC differentiation as well as the specific PR isoform involved in progesterone actions in these cells is unknown. By using primary cultures obtained from the embryonic mouse spinal cord, we showed that embryonic OPC expresses both PR-A and PR-B isoforms. We found that progesterone increases the proliferation, differentiation, and myelination potential of embryonic OPC through its PR by upregulating the expression of oligodendroglial genes such as neuron/glia antigen 2 (NG2), sex determining region Y-box9 (SOX9), myelin basic protein (MBP), 2',3'-cyclic-nucleotide 3'-phosphodiesterase (CNP1), and NK6 homeobox 1 (NKX 6.1). These effects are likely mediated by PR-B, as they are blocked by the silencing of this isoform. The results suggest that progesterone contributes to the process of oligodendrogenesis during prenatal life through specific activation of PR-B.


Subject(s)
Central Nervous System/embryology , Neurogenesis/genetics , Oligodendroglia/immunology , Protein Isoforms/metabolism , Receptors, Progesterone/metabolism , Animals , Cell Differentiation , Female , Mice
14.
Front Immunol ; 11: 294, 2020.
Article in English | MEDLINE | ID: mdl-32174916

ABSTRACT

Ischemic stroke, which accounts for 75-80% of all strokes, is the predominant cause of morbidity and mortality worldwide. The post-stroke immune response has recently emerged as a new breakthrough target in the treatment strategy for ischemic stroke. Glial cells, including microglia, astrocytes, and oligodendrocytes, are the primary components of the peri-infarct environment in the central nervous system (CNS) and have been implicated in post-stroke immune regulation. However, increasing evidence suggests that glial cells exert beneficial and detrimental effects during ischemic stroke. Microglia, which survey CNS homeostasis and regulate innate immune responses, are rapidly activated after ischemic stroke. Activated microglia release inflammatory cytokines that induce neuronal tissue injury. By contrast, anti-inflammatory cytokines and neurotrophic factors secreted by alternatively activated microglia are beneficial for recovery after ischemic stroke. Astrocyte activation and reactive gliosis in ischemic stroke contribute to limiting brain injury and re-establishing CNS homeostasis. However, glial scarring hinders neuronal reconnection and extension. Neuroinflammation affects the demyelination and remyelination of oligodendrocytes. Myelin-associated antigens released from oligodendrocytes activate peripheral T cells, thereby resulting in the autoimmune response. Oligodendrocyte precursor cells, which can differentiate into oligodendrocytes, follow an ischemic stroke and may result in functional recovery. Herein, we discuss the mechanisms of post-stroke immune regulation mediated by glial cells and the interaction between glial cells and neurons. In addition, we describe the potential roles of various glial cells at different stages of ischemic stroke and discuss future intervention targets.


Subject(s)
Ischemic Stroke/immunology , Neuroglia/immunology , Astrocytes/immunology , Blood-Brain Barrier , Complement Pathway, Classical , Humans , Immunity, Innate , Inflammation , Microglia/immunology , Nerve Tissue Proteins/physiology , Neuroimmunomodulation , Neurons/immunology , Oligodendroglia/immunology , Receptors, Cell Surface/physiology , Signal Transduction , T-Lymphocytes/immunology
15.
PLoS One ; 15(2): e0229362, 2020.
Article in English | MEDLINE | ID: mdl-32078657

ABSTRACT

In the mature rodent brain, Sonic Hedgehog (Shh) signaling regulates stem and progenitor cell maintenance, neuronal and glial circuitry and brain repair. However, the sources and distribution of Shh mediating these effects are still poorly characterized. Here, we report in the adult mouse brain, a broad expression pattern of Shh recognized by the specific monoclonal C9C5 antibody in a subset (11-12%) of CC1+ mature oligodendrocytes that do not express carbonic anhydrase II. These cells express also Olig2 and Sox10, two oligodendrocyte lineage-specific markers, but not PDGFRα, a marker of oligodendrocyte progenitors. In agreement with oligodendroglial cells being a source of Shh in the adult mouse brain, we identify Shh transcripts by single molecule fluorescent in situ hybridization in a subset of cells expressing Olig2 and Sox10 mRNAs. These findings also reveal that Shh expression is more extensive than originally reported. The Shh-C9C5-associated signal labels the oligodendroglial cell body and decorates by intense puncta the processes. C9C5+ cells are distributed in a grid-like manner. They constitute small units that could deliver locally Shh to its receptor Patched expressed in GFAP+ and S100ß+ astrocytes, and in HuC/D+ neurons as shown in PtcLacZ/+ reporter mice. Postnatally, C9C5 immunoreactivity overlaps the myelination peak that occurs between P10 and P20 and is down regulated during ageing. Thus, our data suggest that C9C5+CC1+ oligodendroglial cells are a source of Shh in the mouse postnatal brain.


Subject(s)
Antibodies, Monoclonal/immunology , Brain/metabolism , Hedgehog Proteins/immunology , Hedgehog Proteins/metabolism , Neurons/metabolism , Oligodendroglia/metabolism , Animals , Brain/immunology , Cells, Cultured , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Neurons/immunology , Oligodendroglia/immunology , Patched Receptors/immunology , Patched Receptors/metabolism
16.
Nat Rev Neurosci ; 21(3): 139-152, 2020 03.
Article in English | MEDLINE | ID: mdl-32042145

ABSTRACT

Glial cells are abundant in the CNS and are essential for brain development and homeostasis. These cells also regulate tissue recovery after injury and their dysfunction is a possible contributing factor to neurodegenerative and psychiatric disease. Recent evidence suggests that microglia, which are also the brain's major resident immune cells, provide disease-modifying regulation of the other major glial populations, namely astrocytes and oligodendrocytes. In addition, peripheral immune cells entering the CNS after injury and in disease may directly affect microglial, astrocyte and oligodendrocyte function, suggesting an integrated network of immune cell-glial cell communication.


Subject(s)
Brain/immunology , Central Nervous System Diseases/immunology , Immune System , Neuroglia/immunology , Animals , Astrocytes/immunology , Humans , Lymphocytes/immunology , Macrophages/immunology , Microglia/immunology , Neutrophils/immunology , Oligodendroglia/immunology
17.
Proc Natl Acad Sci U S A ; 117(4): 2160-2169, 2020 01 28.
Article in English | MEDLINE | ID: mdl-31932428

ABSTRACT

In multiple sclerosis plaques, oligodendroglial connexin (Cx) 47 constituting main gap junction channels with astroglial Cx43 is persistently lost. As mice with Cx47 single knockout exhibit no demyelination, the roles of Cx47 remain undefined. We aimed to clarify the effects of oligodendroglia-specific Cx47 inducible conditional knockout (icKO) on experimental autoimmune encephalomyelitis (EAE) induced by myelin oligodendrocyte glycoprotein peptide (MOG35-55) in PLP/CreERT;Cx47fl/fl mice at 14 d after tamoxifen injection. Cx47 icKO mice demonstrated exacerbation of acute and chronic relapsing EAE with more pronounced demyelination than Cx47 flox (fl)/fl littermates. CD3+ T cells more abundantly infiltrated the spinal cord in Cx47 icKO than in Cx47 fl/fl mice throughout the acute to chronic phases. CXCR3-CCR6+CD4+ and IL17+IFNγ-CD4+ helper T (Th) 17 cells isolated from spinal cord and brain tissues were significantly increased in Cx47 icKO mice compared with Cx47 fl/fl mice, while MOG35-55-specific proliferation and proinflammatory cytokine production of splenocytes were unaltered. Microarray analysis of isolated microglia revealed stronger microglial activation toward proinflammatory and injury-response phenotypes with increased expressions of chemokines that can attract Th17 cells, including Ccl2, Ccl3, Ccl4, Ccl7, and Ccl8, in Cx47 icKO mice compared with Cx47 fl/fl mice. In Cx47 icKO mice, NOS2+ and MHC class II+ microglia were more enriched immunohistochemically, and A1-specific astroglial gene expressions and astroglia immunostained for C3, a representative A1 astrocyte marker, were significantly increased at the acute phase, compared with Cx47 fl/fl mice. These findings suggest that oligodendroglia-specific Cx47 ablation induces severe inflammation upon autoimmune demyelination, underscoring a critical role for Cx47 in regulating neuroinflammation.


Subject(s)
Connexins/immunology , Multiple Sclerosis/immunology , Oligodendroglia/immunology , Animals , Chemokines/genetics , Chemokines/immunology , Connexins/genetics , Demyelinating Diseases , Disease Models, Animal , Female , Humans , Mice , Mice, Knockout , Multiple Sclerosis/genetics , Myelin Sheath/genetics , Myelin Sheath/immunology , Spinal Cord/immunology , Th17 Cells/immunology
18.
Int Immunopharmacol ; 81: 106038, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31757678

ABSTRACT

A failure of neurodevelopmental differentiation at the level of oligodendroglial-astrocytic biprogenitors (O2A) is shown to be involved in the pathogenesis of both multiple sclerosis (MS) and glioblastoma multiforme (GBM). In this review article, we suggest that certain antigens of Hepatitis B Virus (HBV) and HBV-Vaccine (HBV-V) could act as immune stimulants in GBM treatment based on several lines of evidence. HBV-Vs may cause rare but prominent neuroimmune side effects including demyelination and multiple sclerosis, which may be associated with HBV-proteins creating antigenic mimicry of oligodendroglial progenitors. The combined prevalance of HBV and Hepatitis C Virus-carrier state is less in patients with brain tumors compared to healthy subjects. Furthermore, within the population of patients with brain tumors, the prevalence is even about two times lesser in GBM in comparison to those with a diagnosis of meningioma. Although indirectly, this epidemiological data may indicate that the immune response triggered against hepadnavirus antigens would eliminate aberrantly differentiating O2A progenitor cells giving rise to GBMs. Moreover, Hepatitis B surface antigen-antibody variable domain is among the top 100 differentially expressed transcripts in fresh frozen and formalin-fixed paraffin-embeded specimens obtained from pediatric GBM tissues in comparison to the control brain tissues. However, the provided evidence is still premature and we think that HBV-V warrants investigation first by epidemiological studies and then by animal experiments to determine whether it reduces the risk of GBM and whether it could slow GBM growth via immune stimulation.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Brain/immunology , Glioblastoma/therapy , Hepatitis B Antibodies/immunology , Hepatitis B Vaccines/administration & dosage , Animals , Astrocytes/immunology , Brain/cytology , Brain/pathology , Carrier State/epidemiology , Carrier State/immunology , Cell Differentiation/immunology , Child , Disease Models, Animal , Glioblastoma/epidemiology , Glioblastoma/immunology , Glioblastoma/pathology , Hepatitis B/epidemiology , Hepatitis B/immunology , Hepatitis B Surface Antigens/immunology , Hepatitis B Vaccines/immunology , Hepatitis B virus/immunology , Humans , Immunogenicity, Vaccine , Myelin Sheath/immunology , Neural Stem Cells/immunology , Oligodendroglia/cytology , Oligodendroglia/immunology , Prevalence
19.
Neurosci Lett ; 715: 134601, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31693930

ABSTRACT

Chronic demyelination and the concomitant loss of trophic support and increased energy demands in axons are thought to contribute to neurodegeneration in a number of neurological diseases such as multiple sclerosis (MS). Adult oligodendrocyte precursor cells (OPCs) play an important role in these demyelinating diseases by generating new myelinating oligodendrocytes that may help limit axonal degeneration. Thus, promoting the differentiation of OPCs and functional integration of newly generated oligodendrocytes is a crucial avenue for the next generation of therapies. Evidence to date suggests that the immune system may both positively and negatively impact OPC differentiation and endogenous remyelination in disease. Inflammatory cytokines not only suppress OPC differentiation but may also directly affect other functions of OPCs. Recent studies have demonstrated that OPCs and oligodendrocytes in both human multiple sclerosis lesions and mouse models of demyelination can express an immunogenic transcriptional signature and upregulate antigen presenting genes. In inflammatory demyelinating mouse models OPCs are capable of presenting antigen and activating CD8 + T cells. Here we review the evidence for this new role of oligodendroglia as antigen presenting cells and how these inflammatory OPCs (iOPCs) and inflammatory oligodendrocytes (iOLs) may influence myelin repair and other disease processes.


Subject(s)
Cell Lineage/physiology , Immunity, Cellular/physiology , Oligodendroglia/immunology , Animals , Cytokines/immunology , Cytokines/metabolism , Humans , Microglia/immunology , Microglia/metabolism , Oligodendroglia/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
20.
Brain Behav Immun ; 84: 132-146, 2020 02.
Article in English | MEDLINE | ID: mdl-31785393

ABSTRACT

The pleotropic cytokine tumor necrosis factor (TNF) is involved in the pathophysiology of multiple sclerosis (MS). In various models of MS, including experimental autoimmune encephalomyelitis (EAE), the membrane-bound form of TNF (tmTNF), which signals primarily via TNFR2, mediates protective and reparative effects, whereas the soluble form (solTNF), which signals primarily via TNFR1, promotes pro-inflammatory and detrimental functions. In this study, we investigated the role of TNFR2 expressed in oligodendrocytes in the early phase of EAE pathogenesis. We demonstrated that mice with specific ablation of oligodendroglial TNFR2 displayed early onset and higher peak of motor dysfunction when subjected to EAE, in advance of which accelerated infiltration of immune cells was observed as early as 10 days post EAE induction. The immune cell influx was preceded by microglial activation and increased blood brain barrier permeability. Lack of oligodendroglial TNFR2 accelerated the expression of inflammatory cytokines as well as expression and activation of the inflammasome. Gene expression profiling of oligodendrocytes sorted from the spinal cord 14 days post EAE induction showed robust upregulation of inflammatory genes, some of which were elevated in cells lacking TNFR2 compared to controls. Together, our data demonstrate that oligodendrocytes are directly involved in inflammation and immune modulation in CNS disease and this function is regulated, at least in part, by TNFR2.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Oligodendroglia/immunology , Receptors, Tumor Necrosis Factor, Type II/metabolism , Signal Transduction , Animals , Female , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...