Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
ASN Neuro ; 12: 1759091420979604, 2020.
Article in English | MEDLINE | ID: mdl-33297722

ABSTRACT

While seizure disorders are more prevalent among multiple sclerosis (MS) patients than the population overall and prognosticate earlier death & disability, their etiology remains unclear. Translational data indicate perturbed expression of astrocytic molecules contributing to homeostatic neuronal excitability, including water channels (AQP4) and synaptic glutamate transporters (EAAT2), in a mouse model of MS with seizures (MS+S). However, astrocytes in MS+S have not been examined. To assess the translational relevance of astrocyte dysfunction observed in a mouse model of MS+S, demyelinated lesion burden, astrogliosis, and astrocytic biomarkers (AQP4/EAAT2/ connexin-CX43) were evaluated by immunohistochemistry in postmortem hippocampi from MS & MS+S donors. Lesion burden was comparable in MS & MS+S cohorts, but astrogliosis was elevated in MS+S CA1 with a concomitant decrease in EAAT2 signal intensity. AQP4 signal declined in MS+S CA1 & CA3 with a loss of perivascular AQP4 in CA1. CX43 expression was increased in CA3. Together, these data suggest that hippocampal astrocytes from MS+S patients display regional differences in expression of molecules associated with glutamate buffering and water homeostasis that could exacerbate neuronal hyperexcitability. Importantly, mislocalization of CA1 perivascular AQP4 seen in MS+S is analogous to epileptic hippocampi without a history of MS, suggesting convergent pathophysiology. Furthermore, as neuropathology was concentrated in MS+S CA1, future study is warranted to determine the pathophysiology driving regional differences in glial function in the context of seizures during demyelinating disease.


Subject(s)
Astrocytes/metabolism , Glutamic Acid/metabolism , Hippocampus/metabolism , Homeostasis/physiology , Multiple Sclerosis/metabolism , Seizures/metabolism , Aged , Astrocytes/pathology , Female , Gliosis/metabolism , Gliosis/pathology , Hippocampus/pathology , Humans , Male , Middle Aged , Multiple Sclerosis/epidemiology , Multiple Sclerosis/pathology , Seizures/epidemiology , Seizures/pathology , Water/metabolism
2.
Sci Rep ; 9(1): 503, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30679747

ABSTRACT

Pharmaceutical agents currently approved for the treatment of multiple sclerosis reduce relapse rates, but do not reverse or prevent neurodegeneration nor initiate myelin repair. The highly selective estrogen receptor (ER) ß ligand chloroindazole (IndCl) shows particular promise promoting both remyelination while reducing inflammatory cytokines in the central nervous system of mice with experimental autoimmune encephalomyelitis. To optimize these benefits, we developed and screened seven novel IndCl analogues for their efficacy in promoting primary oligodendrocyte (OL) progenitor cell survival, proliferation, and differentiation in vitro by immunohistochemistry. Two analogues, IndCl-o-chloro and IndCl-o-methyl, induced proliferation and differentiation equivalent to IndCl and were selected for subsequent in vivo evaluation for their impact on clinical disease course, white matter pathology, and inflammation. Both compounds ameliorated disease severity, increased mature OLs, and improved overall myelination in the corpus callosum and white matter tracts of the spinal cord. These effects were accompanied by reduced production of the OL toxic molecules interferon-γ and chemokine (C-X-C motif) ligand, CXCL10 by splenocytes with no discernable effect on central nervous system-infiltrating leukocyte numbers, while IndCl-o-methyl also reduced peripheral interleukin (IL)-17. In addition, expression of the chemokine CXCL1, which is associated with developmental oligodendrogenesis, was upregulated by IndCl and both analogues. Furthermore, callosal compound action potential recordings from analogue-treated mice demonstrated a larger N1 component amplitude compared to vehicle, suggesting more functionally myelinated fibers. Thus, the o-Methyl and o-Chloro IndCl analogues represent a class of ERß ligands that offer significant remyelination and neuroprotection as well as modulation of the immune system; hence, they appear appropriate to consider further for therapeutic development in multiple sclerosis and other demyelinating diseases.


Subject(s)
Corpus Callosum , Estrogen Receptor beta/agonists , Immunologic Factors/pharmacology , Indazoles/pharmacology , Multiple Sclerosis , Remyelination/drug effects , Animals , Cell Proliferation/drug effects , Cell Survival/drug effects , Corpus Callosum/metabolism , Corpus Callosum/pathology , Cytokines/metabolism , Estrogen Receptor beta/metabolism , Female , Ligands , Male , Mice , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Oligodendroglia/metabolism , Oligodendroglia/pathology
3.
Proc Natl Acad Sci U S A ; 115(24): 6291-6296, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29844175

ABSTRACT

Estrogen receptor ß (ERß) ligands promote remyelination in mouse models of multiple sclerosis. Recent work using experimental autoimmune encephalomyelitis (EAE) has shown that ERß ligands induce axon remyelination, but impact peripheral inflammation to varying degrees. To identify if ERß ligands initiate a common immune mechanism in remyelination, central and peripheral immunity and pathology in mice given ERß ligands at peak EAE were assessed. All ERß ligands induced differential expression of cytokines and chemokines, but increased levels of CXCL1 in the periphery and in astrocytes. Oligodendrocyte CXCR2 binds CXCL1 and has been implicated in normal myelination. In addition, despite extensive immune cell accumulation in the CNS, all ERß ligands promoted extensive remyelination in mice at peak EAE. This finding highlights a component of the mechanism by which ERß ligands mediate remyelination. Hence, interplay between the immune system and central nervous system may be responsible for the remyelinating effects of ERß ligands. Our findings of potential neuroprotective benefits arising from the presence of CXCL1 could have implications for improved therapies for multiple sclerosis.


Subject(s)
Axons/metabolism , Chemokine CXCL1/metabolism , Estrogen Receptor beta/metabolism , Myelin Sheath/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Axons/drug effects , Cells, Cultured , Central Nervous System/drug effects , Central Nervous System/metabolism , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Female , Ligands , Mice , Mice, Inbred C57BL , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Myelin Sheath/drug effects , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Receptors, Interleukin-8B/metabolism
4.
J Neurosci Res ; 96(1): 31-44, 2018 01.
Article in English | MEDLINE | ID: mdl-28580666

ABSTRACT

In the central nervous system (CNS), connexin (Cx)s and pannexin (Panx)s are an integral component of homeostatic neuronal excitability and synaptic plasticity. Neuronal Cx gap junctions form electrical synapses across biochemically similar GABAergic networks, allowing rapid and extensive inhibition in response to principle neuron excitation. Glial Cx gap junctions link astrocytes and oligodendrocytes in the pan-glial network that is responsible for removing excitotoxic ions and metabolites. In addition, glial gap junctions help constrain excessive excitatory activity in neurons and facilitate astrocyte Ca2+ slow wave propagation. Panxs do not form gap junctions in vivo, but Panx hemichannels participate in autocrine and paracrine gliotransmission, alongside Cx hemichannels. ATP and other gliotransmitters released by Cx and Panx hemichannels maintain physiologic glutamatergic tone by strengthening synapses and mitigating aberrant high frequency bursting. Under pathological depolarizing and inflammatory conditions, gap junctions and hemichannels become dysregulated, resulting in excessive neuronal firing and seizure. In this review, we present known contributions of Cxs and Panxs to physiologic neuronal excitation and explore how the disruption of gap junctions and hemichannels lead to abnormal glutamatergic transmission, purinergic signaling, and seizures.


Subject(s)
Central Nervous System Diseases/metabolism , Connexins/metabolism , Gap Junctions/metabolism , Homeostasis/physiology , Neuroglia/metabolism , Animals , Calcium Signaling/physiology , Central Nervous System Diseases/pathology , Connexins/chemistry , Gap Junctions/chemistry , Humans , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Neuroglia/chemistry , Neuroglia/pathology
5.
Neuroscience ; 346: 409-422, 2017 03 27.
Article in English | MEDLINE | ID: mdl-28153692

ABSTRACT

Multiple sclerosis (MS) patients are three to six times more likely to develop epilepsy compared to the rest of the population. Seizures are more common in patients with early onset or progressive forms of the disease and prognosticate rapid progression to disability and death. Gray matter atrophy, hippocampal lesions, interneuron loss, and elevated juxtacortical lesion burden have been identified in MS patients with seizures; however, translational studies aimed at elucidating the pathophysiological processes underlying MS epileptogenesis are limited. Here, we report that cuprizone-mediated chronically demyelinated (9-12weeks) mice exhibit marked changes to dorsal hippocampal electroencephalography (EEG) and evidence of overt seizure activity. Immunohistochemical (IHC) analyses within the hippocampal CA1 region revealed extensive demyelination, loss of parvalbumin (PV+) interneurons, widespread gliosis, and changes in aquaporin-4 (AQP4) expression. Our results suggest that chronically demyelinated mice are a valuable model with which we may begin to understand the mechanisms underlying demyelination-induced seizures.


Subject(s)
Demyelinating Diseases/physiopathology , Hippocampus/physiopathology , Multiple Sclerosis/physiopathology , Seizures/physiopathology , Animals , Aquaporin 4/metabolism , Cuprizone/administration & dosage , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Disease Models, Animal , Electroencephalography , Gliosis/metabolism , Hippocampus/drug effects , Hippocampus/pathology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/metabolism , Multiple Sclerosis/chemically induced , Multiple Sclerosis/pathology , Neurons/drug effects , Neurons/pathology , Seizures/chemically induced , Seizures/pathology
6.
Metab Brain Dis ; 30(6): 1515-29, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26246072

ABSTRACT

Inflammation plays a critical role in the pathogenesis of ischemic stroke. This process depends, in part, upon proinflammatory factors released by activated resident central nervous system (CNS) microglia (MG). Previous studies demonstrated that transfer of IL-10(+) B-cells reduced infarct volumes in male C57BL/6 J recipient mice when given 24 h prior to or therapeutically at 4 or 24 h after experimental stroke induced by 60 min middle cerebral artery occlusion (MCAO). The present study assesses possible sex differences in immunoregulation by IL-10(+) B-cells on primary male vs. female MG cultured from naïve and ischemic stroke-induced mice. Thus, MG cultures were treated with recombinant (r)IL-10, rIL-4 or IL-10(+) B-cells after lipopolysaccharide (LPS) activation and evaluated by flow cytometry for production of proinflammatory and anti-inflammatory factors. We found that IL-10(+) B-cells significantly reduced MG production of TNF-α, IL-1ß and CCL3 post-MCAO and increased their expression of the anti-inflammatory M2 marker, CD206, by cell-cell interactions. Moreover, MG from female vs. male mice had higher expression of IL-4 and IL-10 receptors and increased production of IL-4, especially after treatment with IL-10(+) B-cells. These findings indicate that IL-10-producing B-cells play a crucial role in regulating MG activation, proinflammatory cytokine release and M2 phenotype induction, post-MCAO, with heightened sensitivity of female MG to IL-4 and IL-10. This study, coupled with our previous demonstration of increased numbers of transferred IL-10(+) B-cells in the ischemic hemisphere, provide a mechanistic basis for local regulation by secreted IL-10 and IL-4 as well as direct B-cell/MG interactions that promote M2-MG.


Subject(s)
Brain Ischemia/immunology , Brain Ischemia/metabolism , Microglia/immunology , Microglia/metabolism , Stroke/immunology , Stroke/metabolism , Animals , B-Lymphocytes/immunology , Cell Communication , Coculture Techniques , Female , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Interleukin-10/genetics , Interleukin-10/metabolism , Interleukin-4/metabolism , Lectins, C-Type/biosynthesis , Lectins, C-Type/genetics , Male , Mannose Receptor , Mannose-Binding Lectins/biosynthesis , Mannose-Binding Lectins/genetics , Mice , Mice, Inbred C57BL , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Sex Characteristics
7.
Stroke ; 46(10): 2926-34, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26306753

ABSTRACT

BACKGROUND AND PURPOSE: Both pathogenic and regulatory immune processes are involved in the middle cerebral artery occlusion (MCAO) model of experimental stroke, including interactions involving the programmed death 1 (PD-1) receptor and its 2 ligands, PD-L1 and PD-L2. Although PD-1 reduced stroke severity, PD-L1 and PD-L2 appeared to play pathogenic roles, suggesting the use of anti-PD-L monoclonal antibody therapy for MCAO. METHODS: Male C57BL/6 mice were treated with a single dose of anti-PD-L1 monoclonal antibody 4 hours after MCAO and evaluated for clinical, histological and immunologic changes after 96 hours of reperfusion. RESULTS: Blockade of the PD-L1 checkpoint using a single injection of 200 µg anti-PD-L1 monoclonal antibody given intravenously 4 hours after occlusion significantly reduced MCAO infarct volumes and improved neurological outcomes after 96 hours of reperfusion. Treatment partially reversed splenic atrophy and decreased central nervous system infiltrating immune cells concomitant with enhanced appearance of CD8(+) regulatory T cells in the lesioned central nervous system hemisphere. CONCLUSIONS: This study demonstrates for the first time the beneficial therapeutic effects of PD-L1 checkpoint blockade on MCAO, thus validating proposed mechanisms obtained in our previous studies using PD-1- and PD-L-deficient mice. These results provide strong support for the use of available humanized anti-PD-L1 antibodies for treatment of human stroke subjects.


Subject(s)
Antibodies, Monoclonal/pharmacology , B7-H1 Antigen/antagonists & inhibitors , Brain/drug effects , Infarction, Middle Cerebral Artery/immunology , Animals , B7-H1 Antigen/immunology , Brain/immunology , Brain/pathology , Central Nervous System/immunology , Disease Models, Animal , Infarction, Middle Cerebral Artery/pathology , Male , Mice , Mice, Inbred C57BL , Severity of Illness Index , T-Lymphocytes, Regulatory/immunology
8.
J Neuroimmunol ; 285: 129-36, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26198929

ABSTRACT

Women with multiple sclerosis (MS) often experience clinical improvement during pregnancy, indicating that sex hormones might have therapeutic effects in MS. Our previous studies have demonstrated that B cells and PD-L1 are crucial for E2 (17ß-estradiol)-mediated protection against experimental autoimmune encephalomyelitis (EAE). We here demonstrate that the transfer of IL-10(+) B cells into E2-treated PD-L1(-/-) mice after EAE induction could partially restore E2-mediated protection and decrease the frequency of pro-inflammatory cells in the CNS compared to E2/saline treated PD-L1(-/-) mice. Hence, co-administration of IL-10(+) B cells and E2 might have a powerful therapeutic potential for treatment of EAE.


Subject(s)
B-Lymphocytes/metabolism , B7-H1 Antigen/deficiency , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Estradiol/therapeutic use , Interleukin-10/biosynthesis , Animals , B-Lymphocytes/immunology , B-Lymphocytes/transplantation , B7-H1 Antigen/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Estradiol/pharmacology , Female , Interleukin-10/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use
9.
Metab Brain Dis ; 30(5): 1117-27, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25776868

ABSTRACT

Clinical improvement during pregnancy in multiple sclerosis (MS) patients suggests that sex hormones exert potent regulatory effects on autoimmune function. Our previous studies demonstrated that estrogen- (17ß-estradiol; E2) mediated protection against experimental autoimmune encephalomyelitis (EAE), a mouse model for MS, hinges on the B cells, leading to elevated numbers of IL-10 secreting CD1d(hi)CD5(+) B regulatory cells (Bregs) in wild type mice. Our data show that co-administration of E2 and IL-10(+) B cells ameliorates EAE disease severity and limits CNS infiltrating leukocytes in B cell deficient mice. Additionally, treatment with E2 and Bregs reduces demyelination and dramatically decreases the proportion of CD11b(+)CD45(hi) activated microglia/macrophages found in the CNS of immunized animals compared to vehicle, E2 or Breg cells alone. Furthermore, mice given E2 and Bregs exhibit increased numbers of peripheral programmed death-1 positive CD4(+)Foxp3(+) regulatory T cells (Tregs) and up-regulation of programmed death receptor-ligand-1 and CD80 expression on monocytes. Our study suggests IL-10 producing Bregs have powerful therapeutic potential as an agent against EAE when augmented with E2 treatment.


Subject(s)
B-Lymphocytes/transplantation , Central Nervous System , Encephalomyelitis, Autoimmune, Experimental/therapy , Estradiol/administration & dosage , Interleukin-10/administration & dosage , Severity of Illness Index , Animals , B-Lymphocytes/immunology , Central Nervous System/immunology , Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Inflammation/immunology , Inflammation/pathology , Inflammation/therapy , Interleukin-10/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Treatment Outcome
10.
Metab Brain Dis ; 30(4): 911-924, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25537181

ABSTRACT

Clinical stroke induces inflammatory processes leading to cerebral and splenic injury and profound peripheral immunosuppression. IL-10 expression is elevated during major CNS diseases and limits inflammation in the brain. Recent evidence demonstrated that transfer of IL-10(+) B-cells reduced infarct volume in male C57BL/6J (wild-type, WT) recipient mice when given 24 h prior to or 4 h after middle cerebral artery occlusion (MCAO). The purpose of this study was to determine if passively transferred IL-10(+) B-cells can exert therapeutic and immunoregulatory effects when injected 24 h after MCAO induction in B-cell-sufficient male WT mice. The results demonstrated that IL-10(+) B-cell treated mice had significantly reduced infarct volumes in the ipsilateral cortex and hemisphere and improved neurological deficits vs. Vehicle-treated control mice after 60 min occlusion and 96 h of reperfusion. The MCAO-protected B-cell recipient mice had less splenic atrophy and reduced numbers of activated, inflammatory T-cells, decreased infiltration of T-cells and a less inflammatory milieu in the ischemic hemispheres compared with Vehicle-treated control mice. These immunoregulatory changes occurred in concert with the predominant appearance of IL-10-secreting CD8(+)CD122(+) Treg cells in both the spleen and the MCAO-affected brain hemisphere. This study for the first time demonstrates a major neuroprotective role for IL-10(+) B-cells in treating MCAO in male WT mice at a time point well beyond the ~4 h tPA treatment window, leading to the generation of a dominant IL-10(+)CD8(+)CD122(+) Treg population associated with spleen preservation and reduced CNS inflammation.


Subject(s)
B-Lymphocytes/metabolism , B-Lymphocytes/transplantation , CD8-Positive T-Lymphocytes/physiology , Interleukin-10/metabolism , Interleukin-2 Receptor beta Subunit/physiology , Stroke/therapy , Adoptive Transfer/methods , Animals , Central Nervous System/drug effects , Central Nervous System/metabolism , Central Nervous System/pathology , Interleukin-10/administration & dosage , Male , Mice , Mice, Inbred C57BL , Stroke/pathology , Treatment Outcome
11.
Front Cell Neurosci ; 8: 228, 2014.
Article in English | MEDLINE | ID: mdl-25157219

ABSTRACT

Stroke outcome is worsened by the infiltration of inflammatory immune cells into ischemic brains. Our recent study demonstrated that PD-L1- and to a lesser extent PD-L2-deficient mice had smaller brain infarcts and fewer brain-infiltrating cells vs. wild-type (WT) mice, suggesting a pathogenic role for PD-ligands in experimental stroke. We sought to ascertain PD-L1 and PD-L2-expressing cell types that affect T-cell activation, post-stroke in the context of other known co-stimulatory molecules. Thus, cells from male WT and PD-L-deficient mice undergoing 60 min of middle cerebral artery occlusion (MCAO) followed by 96 h of reperfusion were treated with neutralizing antibodies to study co-stimulatory and co-inhibitory interactions between CD80, cytotoxic T-lymphocyte antigen-4 (CTLA-4), PD-1, and PD-Ls that regulate CD8(+) and CD4(+) T-cell activation. We found that antibody neutralization of PD-1 and CTLA-4 signaling post-MCAO resulted in higher proliferation in WT CD8(+) and CD4(+) T-cells, confirming an inhibitory role of PD-1 and CTLA-4 on T-cell activation. Also, CD80/CD28 interactions played a prominent regulatory role for the CD8(+) T-cells and the PD-1/PD-L2 interactions were dominant in controlling the CD4(+) T-cell responses in WT mice after stroke. A suppressive phenotype in PD-L1-deficient mice was attributed to CD80/CTLA-4 and PD-1/PD-L2 interactions. PD-L2 was crucial in modulating CD4(+) T-cell responses, whereas PD-L1 regulated both CD8(+) and CD4(+) T-cells. To establish the contribution of PD-L1 and PD-L2 on regulatory B-cells (Bregs), infarct volumes were evaluated in male PD-L1- and PD-L2-deficient mice receiving IL-10(+) B-cells 4h post-MCAO. PD-L2- but not PD-L1-deficient recipients of IL-10(+) B-cells had markedly reduced infarct volumes, indicating a regulatory role of PD-L2 on Bregs. These results imply that PD-L1 and PD-L2 differentially control induction of T- and Breg-cell responses after MCAO, thus suggesting that selective targeting of PD-L1 and PD-L2 might represent a valuable therapeutic strategy in stroke.

SELECTION OF CITATIONS
SEARCH DETAIL
...