Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Mol Med ; 21: 233-41, 2015 Jan 06.
Article in English | MEDLINE | ID: mdl-25569805

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE), an animal model of human multiple sclerosis (MS), is mediated by myelin-specific autoreactive T cells that cause inflammation and demyelination in the central nervous system (CNS), with significant contributions from activated microglia and macrophages. The molecular bases for expansion and activation of these cells, plus trafficking to the CNS for peripheral cells, are not fully understood. Allograft inflammatory factor-1 (Aif-1) (also known as ionized Ca(2+) binding adapter-1 [Iba-1]) is induced in leukocytes in MS and EAE; here we provide the first assessment of Aif-1 function in this setting. After myelin oligodendrocyte glycoprotein peptide (MOG35-55) immunization, Aif-1-deficient mice were less likely than controls to develop EAE and had less CNS leukocyte infiltration and demyelination; their spinal cords contained fewer CD4 T cells and microglia and more CD8 T cells. These mice also showed significantly less splenic CD4 T-cell expansion and activation, plus decreased proinflammatory cytokine expression. These findings identify Aif-1 as a potent molecule that promotes expansion and activation of CD4 T cells, plus elaboration of a proinflammatory cytokine milieu, in MOG35-55-induced EAE and as a potential therapeutic target in MS.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Calcium-Binding Proteins/deficiency , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Microfilament Proteins/deficiency , Animals , Demyelinating Diseases/genetics , Demyelinating Diseases/immunology , Demyelinating Diseases/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/diagnosis , Encephalomyelitis, Autoimmune, Experimental/metabolism , Leukocytes/immunology , Leukocytes/metabolism , Leukocytes/pathology , Lymphocyte Activation , Mice , Mice, Knockout , Severity of Illness Index , Spleen/immunology , Spleen/metabolism , Spleen/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
2.
Glia ; 61(4): 453-65, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23322421

ABSTRACT

Among the constituent cell types of the multiple sclerosis (MS) plaque, the astrocyte has been the least considered as a player in the pathogenesis of the lesion. Traditionally, it has been assigned a secondary scarring role with little or no role in lesion formation or repair. However, the recent upsurge of interest in the demyelinating condition neuromyelitis optica (NMO) has resulted in NMO being identified as the first disease of myelin in which primary damage to astrocytes, resulting from a humoral immune response that forms against the water channel aquaporin-4, has been documented. This finding in NMO prompted us to re-examine data and material from cases of MS displaying active lesions. Our reappraisal revealed unambiguous early damage to perivascular astrocyte end-feet and to hypertrophic astrocytes in the adjacent parenchyma, but whether this was a primary event was difficult to evaluate due to concomitant edema and inflammation in these acute lesions. The astrocyte damage was long-lasting since resolving lesions displaying remyelination also showed defects in the integrity of the astrocytic covering around blood vessels. Analysis of our findings and of the astrocytic literature supports multiple roles for the astrocyte in the evolution of changes encountered in MS depending upon lesion stage and lesion topography. At variance with the somewhat inhibitory role of the astrocyte is the abundant and growing evidence for this cell to actively participate in both lesion development and repair. We propose that the unequivocal selective early involvement of the astrocyte in MS lesions may have therapeutic relevance


Subject(s)
Astrocytes/pathology , Multiple Sclerosis/pathology , Animals , Blood-Brain Barrier/pathology , Brain/pathology , Demyelinating Diseases/epidemiology , Demyelinating Diseases/pathology , Humans , Multiple Sclerosis/epidemiology , Neuromyelitis Optica/epidemiology , Neuromyelitis Optica/pathology , Spinal Cord/pathology
3.
J Neurosci Res ; 90(10): 1941-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22715030

ABSTRACT

Heat shock proteins (HSPs) are stress-responsive proteins that serve as important molecules contributing to cellular "protein triage." We and others have reported an increase of selected HSPs in multiple sclerosis (MS) lesions. However, the exact expression pattern of HSP family genes in MS is not known. The aim of our research was to assess global transcriptional changes of all gene members of the HSP families within MS lesions and associated normal-appearing white matter (NAWM). To this end, we used laser capture microdissection (LCM) to isolate defined regions of chronic-active MS lesions (n = 5), one of the most common types of MS lesions. To identify changes in HSP genes in relation to different areas of the plaque, we used genome-wide microarray analysis. We detected a significant change in the transcriptional profile of the demyelinated region compared with NAWM. In particular, overall expression of different HSP genes was upregulated in different areas of chronic-active lesion. These changes were linked to an upregulation of heat shock factor 4 (HSF4). This is the first global analysis of transcriptional changes in HSPs in the central nervous system during MS. The results support a relationship between HSP activation and lesion activity.


Subject(s)
Heat-Shock Proteins/genetics , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism , Aged , Autopsy , Brain/pathology , Computational Biology , DNA-Binding Proteins/biosynthesis , Demyelinating Diseases/genetics , Female , Gene Expression Profiling , Heat Shock Transcription Factors , Humans , Laser Capture Microdissection , Male , Microarray Analysis , Multiple Sclerosis/pathology , RNA/genetics , Transcription Factors/biosynthesis , Transcriptional Activation/genetics , Up-Regulation/physiology
4.
J Neuropathol Exp Neurol ; 71(5): 422-33, 2012 May.
Article in English | MEDLINE | ID: mdl-22487860

ABSTRACT

The abundant axonal microtubule-associated protein tau regulates microtubule and actin dynamics, thereby contributing to normal neuronal function. We examined whether mice deficient in tau (Tau(-/-)) or with high levels of human tau differ from wild-type (WT) mice in their susceptibility to neuroaxonal injury in experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis. After sensitization with MOG35-55, there was no difference in clinical disease course between human tau and WT mice, but Tau mice had more severe clinical disease and significantly more axonal damage in spinal cord white matter than those in WT mice. Axonal damage in gray matter correlated with clinical severity in individual mice. By immunoblot analysis, the early microtubule-associated protein-1b was increased 2-fold in the spinal cords of Tau mice with chronic experimental autoimmune encephalomyelitis versus naive Tau mice. This difference was not detected in comparable WT animals, which suggests that there was compensation for the loss of tau in the deficient mice. In addition, levels of the growth arrest-specific protein 7b, a tau-binding protein that is stabilized when bound to tau, were higher in WT than those in Tau(-/-) spinal cord samples. These data indicate that loss of tau exacerbates experimental autoimmune encephalomyelitis and suggest that maintaining tau integrity might reduce the axonal damage that occurs in inflammatory neurodegenerative diseases such as multiple sclerosis.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/physiopathology , Gene Expression Regulation/genetics , Genetic Predisposition to Disease , Glycoproteins/adverse effects , Neurons/pathology , Peptide Fragments/adverse effects , Spinal Cord/pathology , tau Proteins/deficiency , Age Factors , Animals , Axons/metabolism , Axons/pathology , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Freund's Adjuvant/adverse effects , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin-Oligodendrocyte Glycoprotein , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Myelinated/pathology , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Paralysis/genetics , Phosphorylation/physiology , Spinal Cord/metabolism
5.
J Neuroimmunol ; 245(1-2): 8-14, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22342190

ABSTRACT

We showed previously that mice deficient in astrocyte gap junctions Cx43 and Cx30 exhibit white matter vacuolation and hypomyelination. In this study we tested the hypothesis that loss of astrocytic gap junction proteins leads to exacerbation of the primary demyelinating diseases, using experimental autoimmune encephalomyelitis (EAE) as a model system. To test for this, Cx43 floxed mice were crossed with GFAP:Cre, Cx30 null mice to generate mice lacking astrocytic expression of both Cx43 and Cx30 (dKO). EAE was induced using myelin oligodendrocyte glycoprotein (MOG(35-55)) peptide, and mice were monitored for acute expression of disease. No statistically significant difference in clinical or pathological expression of EAE was observed. Lesion load and susceptibility of different areas of the CNS to inflammation were similar in all genotypes. Moreover, no differences were noted in blood-brain barrier (BBB) permeability, tissue wet weight, axonal pathology, gliosis or demyelination during acute disease. These data show that loss of the astrocytic connexins, Cx43 and Cx30, and the white matter pathology observed in these mice does not statistically affect clinical or pathological expression of EAE and show that astrocyte gap junctions do not regulate autoimmune inflammation and associated BBB disruption in acute EAE.


Subject(s)
Astrocytes/immunology , Connexin 43/deficiency , Connexins/deficiency , Encephalomyelitis, Autoimmune, Experimental/immunology , Gap Junctions/immunology , Genetic Predisposition to Disease , Animals , Astrocytes/metabolism , Astrocytes/pathology , Connexin 30 , Connexin 43/genetics , Connexins/genetics , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Gap Junctions/genetics , Gap Junctions/pathology , Genetic Predisposition to Disease/genetics , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Severity of Illness Index
6.
J Neuroinflammation ; 8: 49, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21569627

ABSTRACT

BACKGROUND: Axl, together with Tyro3 and Mer, constitute the TAM family of receptor tyrosine kinases. In the nervous system, Axl and its ligand Growth-arrest-specific protein 6 (Gas6) are expressed on multiple cell types. Axl functions in dampening the immune response, regulating cytokine secretion, clearing apoptotic cells and debris, and maintaining cell survival. Axl is upregulated in various disease states, such as in the cuprizone toxicity-induced model of demyelination and in multiple sclerosis (MS) lesions, suggesting that it plays a role in disease pathogenesis. To test for this, we studied the susceptibility of Axl-/- mice to experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis. METHODS: WT and Axl-/- mice were immunized with myelin oligodendrocyte glycoprotein (MOG)35-55 peptide emulsified in complete Freund's adjuvant and injected with pertussis toxin on day 0 and day 2. Mice were monitored daily for clinical signs of disease and analyzed for pathology during the acute phase of disease. Immunological responses were monitored by flow cytometry, cytokine analysis and proliferation assays. RESULTS: Axl-/- mice had a significantly more severe acute phase of EAE than WT mice. Axl-/- mice had more spinal cord lesions with larger inflammatory cuffs, more demyelination, and more axonal damage than WT mice during EAE. Strikingly, lesions in Axl-/- mice had more intense Oil-Red-O staining indicative of inefficient clearance of myelin debris. Fewer activated microglia/macrophages (Iba1+) were found in and/or surrounding lesions in Axl-/- mice relative to WT mice. In contrast, no significant differences were noted in immune cell responses between naïve and sensitized animals. CONCLUSIONS: These data show that Axl alleviates EAE disease progression and suggests that in EAE Axl functions in the recruitment of microglia/macrophages and in the clearance of debris following demyelination. In addition, these data provide further support that administration of the Axl ligand Gas6 could be therapeutic for immune-mediated demyelinating diseases.


Subject(s)
Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Inflammation/immunology , Myelin Sheath/pathology , Proto-Oncogene Proteins/immunology , Receptor Protein-Tyrosine Kinases/immunology , Animals , Central Nervous System/immunology , Cytokines/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Glycoproteins/immunology , Inflammation/pathology , Macrophages/cytology , Macrophages/immunology , Mice , Mice, Knockout , Microglia/cytology , Microglia/immunology , Myelin Sheath/metabolism , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments/immunology , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Axl Receptor Tyrosine Kinase
7.
Glia ; 58(8): 964-74, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20169621

ABSTRACT

Notch1 receptor signaling regulates oligodendrocyte progenitor differentiation and myelin formation in development, and during remyelination in the adult CNS. In active multiple sclerosis lesions, Notch1 localizes to oligodendrocyte lineage cells, and its ligand Jagged1 is expressed by reactive astrocytes. Here, we examined induction of Jagged1 in human astrocytes, and its impact on oligodendrocyte differentiation. In human astrocyte cultures, the cytokine TGFbeta1 induced Jagged1 expression and blockade of the TGFbeta1 receptor kinase ALK5 abrogated Jagged1 induction. TGFbeta2 and beta3 had similar effects, but induction was not observed in response to the TGFbeta family member activin A or other cytokines. Downstream, TGFbeta1 activated Smad-dependent signaling, and Smad-independent pathways that included PI3 kinase, p38, and JNK MAP kinase, but only inhibition of the Smad-dependent pathway blocked Jagged1 expression. SiRNA inhibition of Smad3 downregulated induction of Jagged1, and this was potentiated by Smad2 siRNA. Purified oligodendrocyte progenitor cells (OPCs) nucleofected with Notch1 intracellular signaling domain displayed a shift towards proliferation at the expense of differentiation, demonstrating functional relevance of Notch1 signaling in OPCs. Furthermore, human OPCs plated onto Jagged1-expressing astrocytes exhibited restricted differentiation. Collectively, these data illustrate the mechanisms underlying Jagged1 induction in human astrocytes, and suggest that TGFbeta1-induced activation of Jagged1-Notch1 signaling may impact the size and differentiation of the OPC pool in the human CNS.


Subject(s)
Astrocytes/drug effects , Calcium-Binding Proteins/metabolism , Cell Differentiation/physiology , Cell Proliferation , Extracellular Matrix Proteins/pharmacology , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Oligodendroglia/physiology , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Smad3 Protein/metabolism , Stem Cells/physiology , Transforming Growth Factor beta/pharmacology , Analysis of Variance , Animals , Animals, Newborn , Astrocytes/metabolism , Cerebral Cortex/cytology , Enzyme Inhibitors/pharmacology , Extracellular Matrix Proteins/metabolism , Humans , Jagged-1 Protein , RNA, Small Interfering/pharmacology , Rats , Receptor, Transforming Growth Factor-beta Type I , Serrate-Jagged Proteins , Transfection/methods , Transforming Growth Factor beta/metabolism
8.
Proc Natl Acad Sci U S A ; 106(45): 19162-7, 2009 Nov 10.
Article in English | MEDLINE | ID: mdl-19855010

ABSTRACT

In the developing CNS, Notch1 and its ligand, Jagged1, regulate oligodendrocyte differentiation and myelin formation, but their role in repair of demyelinating lesions in diseases such as multiple sclerosis remains unresolved. To address this question, we generated a mouse model in which we targeted Notch1 inactivation to oligodendrocyte progenitor cells (OPCs) using Olig1Cre and a floxed Notch1 allele, Notch1(12f). During CNS development, OPC differentiation was potentiated in Olig1Cre:Notch1(12f/12f) mice. Importantly, in adults, remyelination of demyelinating lesions was also accelerated, at the expense of proliferation within the progenitor population. Experiments in vitro confirmed that Notch1 signaling was permissive for OPC expansion but inhibited differentiation and myelin formation. These studies also revealed that astrocytes exposed to TGF-beta1 restricted OPC maturation via Jagged1-Notch1 signaling. These data suggest that Notch1 signaling is one of the mechanisms regulating OPC differentiation during CNS remyelination. Thus, Notch1 may represent a potential therapeutical avenue for lesion repair in demyelinating disease.


Subject(s)
Cell Differentiation/physiology , Central Nervous System/cytology , Myelin Sheath/physiology , Oligodendroglia/physiology , Receptor, Notch1/metabolism , Signal Transduction/physiology , Animals , Calcium-Binding Proteins/metabolism , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , Membrane Proteins/metabolism , Mice , Oligodendroglia/cytology , Serrate-Jagged Proteins
9.
Virology ; 392(2): 246-59, 2009 Sep 30.
Article in English | MEDLINE | ID: mdl-19646728

ABSTRACT

In the CNS, microglia are the primary targets of HIV infection. In this study, we investigated the effect of activation of the innate antiviral receptors TLR3 and TLR4 on HIV infection of primary human microglia, as well as microglial cell signaling and gene expression. Ligands for both TLR3 and TLR4 potently inhibited HIV replication in microglia through a pathway requiring IRF3. Surprisingly, a remarkably similar pattern of cell signaling and gene expression was observed in TLR3- and TLR4-activated microglia, suggesting a relatively minor role for MyD88 following TLR4 activation in these cells. HIV did not activate IRF3 but rather decreased IRF3 protein, indicating that HIV does not activate TLR3 or RIG-like helicases in microglia. Taken together, these results indicate that activation of TLR3 or TLR4 will elicit antiviral immunity, in addition to inducing proinflammatory responses. We suggest that a balanced expression between inflammatory and innate immune genes might be achieved by IRF3 over-expression.


Subject(s)
HIV Infections/immunology , Interferon Regulatory Factor-3/immunology , Microglia/immunology , Toll-Like Receptor 3/immunology , Toll-Like Receptor 4/immunology , Cells, Cultured , Gene Expression Profiling , HIV/physiology , Humans , Interferon-beta/immunology , Microglia/virology , Oligonucleotide Array Sequence Analysis , RNA Interference , Virus Replication
10.
Curr Top Microbiol Immunol ; 336: 63-81, 2009.
Article in English | MEDLINE | ID: mdl-19688328

ABSTRACT

Protection against viral infections is critically dependent upon the early production of significant levels of type 1 interferons and the expression of interferon-stimulated genes that function as the effectors of innate antiviral immunity. Activation of Toll-like receptors on cells of the immune system is known to play an important role in this process. In this chapter we review evidence for a role of TLRs in innate immune responses against viral infections of the central nervous system. By far the most extensive literature pertains to TLR3. Data from various laboratories have shown that TLR3 is expressed in cells endogenous to the CNS, particularly in astrocytes and microglia. Triggering TLR3 by synthetic dsRNA, poly I:C effectively induces innate antiviral responses as well as boosts adaptive immune responses. Additional experiments show cooperative responses between TLRs (3, 7/8 and 9) in mounting an effective antiviral immune response in the periphery. Perhaps the most exciting data are from patient populations that document the critical role that specific TLRs play in specific CNS infections. Studies also suggest that inappropriate activation of the TLRs can result in a pathogenic outcome rather than a protective one. Since TLR ligands are being actively considered for their antiviral and potential adjuvant effects, this will be an important issue to address in the context of the CNS environment.


Subject(s)
Central Nervous System Infections/immunology , Central Nervous System Infections/virology , Toll-Like Receptor 3/immunology , Virus Diseases/immunology , Animals , Humans , Virus Diseases/virology
11.
J Neurosci ; 29(24): 7743-52, 2009 Jun 17.
Article in English | MEDLINE | ID: mdl-19535586

ABSTRACT

Astrocytes are coupled via gap junctions (GJs) comprising connexin 43 (Cx43) (Gja1) and Cx30 (Gjb6), which facilitate intercellular exchange of ions. Astrocyte connexins also form heterotypic GJs with oligodendrocytic somata and lamellae. Loss of oligodendrocyte gap junctions results in oligodendrocyte and myelin pathology. However, whether loss of astrocyte GJs affects oligodendrocytes and myelin is not known. To address this question, mice with astrocyte-targeted deletion of Cx43 and global loss of Cx30 [double knock-out (dKO)] were studied using Western blotting, immunohistochemistry, electron microscopy, and functional assays. Commencing around postnatal day 23 and persisting into old age, we found widespread pathology of white matter tracts comprising vacuolated oligodendrocytes and intramyelinic edema. In contrast, gray matter pathology was restricted to the CA1 region of the hippocampus, and consisted of edematous astrocytes. No differences were observed in synaptic density or total NeuN(+) cells in the hippocampus, or olig2(+) cells in the corpus callosum. However, in dKO mice, fewer CC1-positive mature oligodendrocytes were detected, and Western blotting indicated reduced myelin basic protein. Pathology was not noted in mice expressing a single allele of either Cx43 or Cx30. When compared with single connexin knock-outs, dKO mice were impaired in sensorimotor (rotarod, balance beam assays) and spatial memory tasks (object recognition assays). We conclude that loss of astrocytic GJs can result in white matter pathology that has functional consequences.


Subject(s)
Astrocytes/metabolism , Connexin 43/deficiency , Connexins/deficiency , Demyelinating Diseases/pathology , Hippocampus/pathology , Phenotype , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/metabolism , Calcium-Binding Proteins , Cell Proliferation , Connexin 30 , DNA-Binding Proteins/metabolism , Demyelinating Diseases/complications , Demyelinating Diseases/genetics , Disease Models, Animal , Gap Junctions/pathology , Glial Fibrillary Acidic Protein/genetics , In Situ Nick-End Labeling/methods , Memory Disorders/genetics , Memory Disorders/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins , Microscopy, Electron/methods , Myelin Basic Protein/metabolism , Myelin Sheath/pathology , Nerve Tissue Proteins/metabolism , Oligodendrocyte Transcription Factor 2
12.
J Clin Invest ; 119(1): 10-3, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19104146

ABSTRACT

MS results from destruction of the protective myelin sheath surrounding axons, which prevents the transmission of nerve impulses. Precursors of oligodendrocytes, the cells capable of myelinating axons, are preserved in demyelinating lesions; however, why these precursors do not differentiate into mature oligodendrocytes and remyelinate axons is unknown. Contactin is a noncanonical Notch receptor ligand that mediates oligodendrocyte differentiation. In this issue of the JCI, Nakahara et al. show that Contactin is abundantly expressed on demyelinated axons in human chronic MS lesions and that Notch1 is activated in oligodendrocyte precursor cells (see the related article beginning on page 169). However, Notch1 intracellular domain coassociates with the nuclear transporter Importin beta but fails to show evidence of nuclear translocation. These cytoplasmic aggregates also contain TAT-interacting protein 30 kDa (TIP30), a proapoptotic factor, which inhibits nuclear transport and, consequently, Notch1-mediated oligodendrocyte differentiation and remyelination. These data target TIP30 as a new pathogenic factor in MS.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Multiple Sclerosis/pathology , Myelin Sheath/metabolism , Receptor, Notch1/metabolism , Regeneration/physiology , Acetyltransferases/metabolism , Contactins , Humans , Multiple Sclerosis/metabolism , Multiple Sclerosis/physiopathology , Oligodendroglia/cytology , Oligodendroglia/metabolism , Receptor, Notch1/genetics , Signal Transduction/physiology , Transcription Factors/metabolism
13.
Brain Res ; 1240: 1-11, 2008 Nov 13.
Article in English | MEDLINE | ID: mdl-18804096

ABSTRACT

Activation of the receptor tyrosine kinase Axl recruits signaling molecules that regulate cell growth and survival. To evaluate Axl's role in brain during cuprizone toxicity, mice were fed cuprizone and evaluated at 3-, 4-, and 6-week cuprizone treatment and 3- and 5-week post-cuprizone withdrawal. At 4-week cuprizone treatment, the corpora callosa of wildtype (WT) mice had robust Oil Red O+ staining indicative of ongoing phagocytosis. Axl-/- mice had minimal Oil Red O+ staining, fewer microglia, and significantly more TUNEL+/ASPA+ mature oligodendrocytes than the WT. At 6-week cuprizone treatment, there was significantly more Oil Red O+ staining in the Axl-/- corpora callosa than in the WT indicating a lag in the clearance of cellular and myelin debris. Relative to WT mice, there were fewer mature oligodendrocytes and significantly more SMI-32+ axons at 3-week post-cuprizone withdrawal, indicative of axonal damage in the Axl-/- corpora callosa. Electron microscopy determined that at 3-week post-cuprizone withdrawal the number of dystrophic axons and axons containing autophagosome-like vacuoles/mouse was increased in the Axl-/- mice relative to the WT mice. In Axl-/- corpora callosa, 5-week post-cuprizone withdrawal, the number of mature oligodendrocytes was comparable to the WT mice, but axons in the Axl-/- mice were SMI-32+, suggesting that Axl-/- mice have delayed clearance of apoptotic oligodendrocytes and myelin debris resulting in prolonged axonal damage and recovery from cuprizone toxicity.


Subject(s)
Axons/pathology , Chelating Agents/toxicity , Corpus Callosum/pathology , Cuprizone/toxicity , Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Apoptosis/drug effects , Axons/drug effects , Corpus Callosum/drug effects , In Situ Nick-End Labeling , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Oligodendroglia/drug effects , Oligodendroglia/pathology , Proto-Oncogene Proteins , Axl Receptor Tyrosine Kinase
14.
J Immunol ; 179(7): 4503-12, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17878346

ABSTRACT

Inflammation leads to induction of tissue stress conditions that might contribute to the generation of mechanisms limiting ongoing immune responses. We have shown previously that peptides derived from brain tissue of mice with experimental autoimmune encephalomyelitis (EAE) complexed with the chaperone heat shock protein 70 (Hsp70-pc) induce an NK-cell-dependent tolerance for subsequent EAE sensitization. We now present data that showed that the MHC class I-related glycoprotein H60 determines Hsp70-pc-induced EAE inhibition. Hsp70-pc led to significant and selective up-regulation of H60 expression in SJL/J mice, and Ab-blocking of H60 expression led to loss of EAE tolerance. Similarly, blocking of the NK cell receptor for H60, NKG2D, also reversed the Hsp70-pc-induced EAE inhibition. In contrast, in C57BL/6 mice H60 was not expressed, and Hsp70-pc-induced tolerance was not detected. The NK cell mediated Hsp70-pc-induced tolerance to EAE was dependent on modulation of dendritic cells function leading to diminished T cell reactivity to PLP. As, no increase of H60 expression on T cells from EAE mice immunized with PLP was detected, and no enhanced loss of CD3+ H60+ over CD3+ H60- cells in Hsp70-pc-induced EAE tolerance was found direct killing of H60+ PLP-reactive cells seems not to be involved in the Hsp70-pc-induced tolerance induction. We have provided evidence that Hsp70-pc-induced tolerance for EAE, mediated by NK cells, involves induction of H60 ligand and its interaction with NKG2D receptor. NK cells tolerization of EAE depends on altered dendritic cells activity leading to enhanced death of Ag reactive cells.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , HSP70 Heat-Shock Proteins/pharmacology , Immune Tolerance/drug effects , Immune Tolerance/immunology , Minor Histocompatibility Antigens/metabolism , Peptides/pharmacology , Receptors, Immunologic/metabolism , Animals , Antibodies/immunology , Antibodies/pharmacology , Antigens/immunology , Cell Proliferation/drug effects , Cells, Cultured , Coculture Techniques , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Female , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , HSP70 Heat-Shock Proteins/chemistry , Interferon-gamma/biosynthesis , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice , Minor Histocompatibility Antigens/immunology , NK Cell Lectin-Like Receptor Subfamily K , Peptides/chemistry , Receptors, Immunologic/immunology , Receptors, Natural Killer Cell , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
15.
J Virol ; 81(18): 9838-50, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17626075

ABSTRACT

Indoleamine 2,3-dioxygenase (IDO) is the first and rate-limiting enzyme in the kynurenine pathway of tryptophan catabolism and has been implicated in neurotoxicity and suppression of the antiviral T-cell response in HIV encephalitis (HIVE). Here we show that the Toll-like receptor 3 (TLR3) ligand poly(I:C) (PIC) induces the expression of IDO in human astrocytes. PIC was less potent than gamma interferon (IFN-gamma) but more potent than IFN-beta in inducing IDO. PIC induction of IDO was mediated in part by IFN-beta but not IFN-gamma, and both NF-kappaB and interferon regulatory factor 3 (IRF3) were required. PIC also upregulated TLR3, thereby augmenting the primary (IFN-beta) and secondary (IDO and viperin) response genes upon subsequent stimulation with PIC. In HIVE, the transcripts for TLR3, IFN-beta, IDO, and viperin were increased and IDO immunoreactivity was detected in reactive astrocytes as well as macrophages and microglia. PIC caused suppression of intracellular replication of human immunodeficiency virus pseudotyped with vesicular stomatitis virus G protein and human cytomegalovirus in a manner dependent on IRF3 and IDO. The involvement of IDO was demonstrated by partial but significant reversal of the PIC-mediated antiviral effect by IDO RNA interference and/or tryptophan supplementation. Importantly, the cytokine interleukin-1 abolished IFN-gamma-induced IDO enzyme activity in a nitric oxide-dependent manner without suppressing protein expression. Our results demonstrate that IDO is an innate antiviral protein induced by double-stranded RNA and suggest a therapeutic utility for PIC in human viral infections. They also show that IDO activity can be dissociated from protein expression, indicating that the local central nervous system cytokine and nitric oxide environment determines IDO function.


Subject(s)
Astrocytes/immunology , Encephalitis, Viral/immunology , HIV Infections/immunology , HIV-1/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Interferon Inducers/pharmacology , Poly I-C/pharmacology , Toll-Like Receptor 3/immunology , Virus Replication/immunology , Astrocytes/enzymology , Astrocytes/virology , Cells, Cultured , Cytokines/immunology , Cytokines/pharmacology , Cytomegalovirus/genetics , Cytomegalovirus/immunology , Cytomegalovirus Infections/enzymology , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/immunology , Encephalitis, Viral/enzymology , Encephalitis, Viral/genetics , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Enzymologic/immunology , HIV Infections/enzymology , HIV Infections/genetics , HIV-1/genetics , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-3/metabolism , Kynurenine/genetics , Kynurenine/immunology , Kynurenine/metabolism , Ligands , Macrophages/enzymology , Macrophages/immunology , Macrophages/virology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Microglia/enzymology , Microglia/immunology , Microglia/virology , NF-kappa B/immunology , NF-kappa B/metabolism , Nitric Oxide/immunology , Nitric Oxide/metabolism , Oxidoreductases Acting on CH-CH Group Donors , Proteins/immunology , Proteins/metabolism , Toll-Like Receptor 3/agonists , Toll-Like Receptor 3/metabolism , Tryptophan/immunology , Tryptophan/metabolism , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Envelope Proteins/metabolism
16.
J Neurosci ; 26(47): 12174-85, 2006 Nov 22.
Article in English | MEDLINE | ID: mdl-17122042

ABSTRACT

Mechanisms that regulate oligodendrocyte survival and myelin formation are an intense focus of research into myelin repair in the lesions of multiple sclerosis (MS). Although demyelination and oligodendrocyte loss are pathological hallmarks of the disease, increased oligodendrocyte numbers and remyelination are frequently observed in early lesions, but these diminish as the disease course progresses. In the current study, we used a microarray-based approach to investigate genes regulating repair in MS lesions, and identified interleukin-11 (IL-11) as an astrocyte-derived factor that potentiates oligodendrocyte survival and maturation, and myelin formation. IL-11 was induced in human astrocyte cultures by the cytokines IL-1beta and TGFbeta1, which are both prominently expressed in MS plaques. In MS tissue samples, IL-11 was expressed by reactive astrocytes, with expression particularly localized at the myelinated border of both active and silent lesions. Its receptor, IL-11R alpha, was expressed by oligodendrocytes. In experiments in human cultures in vitro, IL-11R alpha localized to immature oligodendrocytes, and its expression decreased during maturation. In cultures treated with IL-11, we observed a significant increase in oligodendrocyte number, and this was associated with enhanced oligodendrocyte survival and maturation. Importantly, we also found that IL-11 treatment was associated with significantly increased myelin formation in rodent CNS cocultures. These data are the first to implicate IL-11 in oligodendrocyte viability, maturation, and myelination. We suggest that this pathway may represent a potential therapeutic target for oligodendrocyte protection and remyelination in MS.


Subject(s)
Interleukin-11/pharmacology , Myelin Sheath/drug effects , Oligodendroglia/drug effects , Animals , Blotting, Western , Bromodeoxyuridine/metabolism , Cell Count/methods , Cell Survival/drug effects , Central Nervous System/cytology , Enzyme-Linked Immunosorbent Assay/methods , Fetus , Fluorescent Antibody Technique/methods , Ganglia, Spinal/cytology , Humans , In Situ Nick-End Labeling/methods , Interleukin-1beta/pharmacology , Microarray Analysis/methods , Microscopy, Electron, Transmission/methods , Myelin Sheath/metabolism , Myelin Sheath/ultrastructure , Neurons/drug effects , Neurons/ultrastructure , Oligodendroglia/ultrastructure , RNA, Messenger/biosynthesis , Rats , Reverse Transcriptase Polymerase Chain Reaction/methods , Time Factors , Tissue Culture Techniques , Transforming Growth Factor beta1/pharmacology
17.
J Immunol ; 177(8): 5574-84, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-17015745

ABSTRACT

Loss of blood-brain barrier (BBB) integrity is believed to be an early and significant event in lesion pathogenesis in the inflammatory demyelinating disease multiple sclerosis (MS), and understanding mechanisms involved may lead to novel therapeutic avenues for this disorder. Well-differentiated endothelium forms the basis of the BBB, while astrocytes control the balance between barrier stability and permeability via production of factors that restrict or promote vessel plasticity. In this study, we report that the proinflammatory cytokine IL-1beta, which is prominently expressed in active MS lesions, causes a shift in the expression of these factors to favor plasticity and permeability. The transcription factor, hypoxia inducible factor-1 (HIF-1), plays a significant role in this switch. Using a microarray-based approach, we found that in human astrocytes, IL-1beta induced the expression of genes favoring vessel plasticity, including HIF-1alpha and its target, vascular endothelial growth factor-A (VEGF-A). Demonstrating relevance to MS, we showed that HIF-1alpha and VEGF-A were expressed by reactive astrocytes in active MS lesions, while the VEGF receptor VEGFR2/flk-1 localized to endothelium and IL-1 to microglia/macrophages. Suggesting functional significance, we found that expression of IL-1beta in the brain induced astrocytic expression of HIF-1alpha, VEGF-A, and BBB permeability. In addition, we confirmed VEGF-A to be a potent inducer of BBB permeability and angiogenesis, and demonstrated the importance of IL-1beta-induced HIF-1alpha in its regulation. These results suggest that IL-1beta contributes to BBB permeability in MS via reactivation of the HIF-VEGF axis. This pathway may represent a potential therapeutic target to restrict lesion formation.


Subject(s)
Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Gene Expression Regulation/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Interleukin-1beta/physiology , Multiple Sclerosis/pathology , Neovascularization, Pathologic/pathology , Astrocytes/metabolism , Astrocytes/pathology , Blood-Brain Barrier/pathology , Endothelium/metabolism , Gene Expression Profiling , Humans , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Interleukin-1beta/pharmacology , Multiple Sclerosis/etiology , Neovascularization, Pathologic/drug therapy , Permeability/drug effects , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/physiology , Vascular Endothelial Growth Factor Receptor-2/metabolism
18.
J Immunol ; 177(7): 4735-41, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-16982913

ABSTRACT

TLR3 functions as a viral nucleic acid sentinel activated by dsRNA viruses and virus replication intermediates within intracellular vesicles. To explore the spectrum of genes induced in human astrocytes by TLR3, we used a microarray approach and the analog polyriboinosinic polyribocytidylic acid (pIC) as ligand. As expected for TLR activation, pIC induced a wide array of cytokines and chemokines known for their role in inflammatory responses, as well as up-regulation of the receptor itself. The data also showed activation of a broad spectrum of antiviral response genes. To determine whether pIC induced an antiviral state in astrocytes, a pseudotyped HIV viral particle, vesicular stomatitis virus g-env-HIV-1, was used. pIC significantly abrogated HIV-1 replication, whereas IL-1, which also potently activates astrocytes, did not. One of the most highly up-regulated genes on microarray was the protein viperin/cig5. We found that viperin/cig5 expression was dependent on IFN regulatory factor 3 and NF-kappaB signaling, and that repetitive stimulation with pIC, but not IL-1, further increased expression. Viperin induction could also be substantially inhibited by neutralizing Abs to IFN-beta, as could HIV-1 replication. To explore a role for viperin in IFN-beta-mediated inhibition of HIV-1, we used an RNA interference (RNAi) approach. RNAi directed against viperin, but not a scrambled RNAi, significantly inhibited viperin expression, and also significantly reversed pIC-induced inhibition of HIV-1 replication. We conclude that viperin contributes to the antiviral state induced by TLR3 ligation in astrocytes, supporting a role for astrocytes as part of the innate immune response against infection in the CNS.


Subject(s)
Antiviral Agents/immunology , Astrocytes/immunology , Astrocytes/virology , Poly I-C/immunology , Proteins/immunology , Toll-Like Receptor 3/metabolism , Blotting, Western , Chemokines/metabolism , Fetus , HIV-1/immunology , Humans , Immunohistochemistry , Interferon-beta/immunology , Interleukin-1/immunology , Oligonucleotide Array Sequence Analysis , Oxidoreductases Acting on CH-CH Group Donors , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Vesicular stomatitis Indiana virus/genetics , Vesicular stomatitis Indiana virus/immunology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
19.
Glia ; 54(8): 775-85, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16958087

ABSTRACT

Toll-like receptor 3 (TLR3) is a component of the innate immune response that responds to dsRNA viruses and virus replication intermediates. In this study we show that activation of astrocytes with the dsRNA mimetic polyinosinic-cytidylic acid (pI:C) results in loss of expression of connexin43 (Cx43) mRNA and protein while upregulating the expression of the ionotropic P2 receptor P2X(4)R. Analysis of the signaling pathways involved failed to demonstrate a role for the p38 MAP kinase, ERK, or JNK signaling pathways whereas an inhibitor of the PI3 kinase/Akt pathway effectively blocked the action of pI:C. Using adenoviral vectors containing a super-repressor of NF-kappaB (NF-kappaB SR) construct or a dominant negative interferon regulatory factor 3 (dnIRF3) construct showed that inhibition of both transcription factors also blocked the effects of pI:C. To explore the functional consequences of pI:C activation we used a pore-forming assay for P2X(4)R activity and a scrape loading assay for gap junction intercellular communication (GJIC). No pore-forming activity consistent with functional P2X(4)R expression was detected in either control or activated astrocytes. In contrast, robust Lucifer yellow transfer indicative of GJIC was detected in resting cells that was lost following pI:C activation. The dnIRF3 construct failed to restore GJIC whereas the NF-kappaB SR or the NF-kappaB inhibitor BAY11-7082 and the PI3K inhibitor LY294002 all significantly reversed the effect of pI:C on GJ connectivity. We conclude that activation of the innate immune response in astrocytes is associated with functional loss of GJIC through a pathway involving NF-kappaB and PI3 kinase.


Subject(s)
Astrocytes/immunology , Connexin 43/metabolism , Gap Junctions/immunology , NF-kappa B/immunology , Phosphatidylinositol 3-Kinases/immunology , Toll-Like Receptor 3/metabolism , Astrocytes/cytology , Astrocytes/metabolism , Cell Communication/drug effects , Cell Communication/genetics , Cell Communication/immunology , Cells, Cultured , Down-Regulation/drug effects , Down-Regulation/immunology , Enzyme Inhibitors/pharmacology , Gap Junctions/drug effects , Gap Junctions/metabolism , Humans , Immunity, Innate/drug effects , Immunity, Innate/genetics , Immunity, Innate/immunology , Interferon Inducers/pharmacology , Interferon Regulatory Factor-3/genetics , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Poly I-C/pharmacology , RNA Virus Infections/immunology , RNA Viruses/genetics , RNA, Double-Stranded/genetics , RNA, Double-Stranded/immunology , Receptors, Purinergic P2/immunology , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2X4 , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 3/agonists , Up-Regulation/drug effects , Up-Regulation/genetics , Up-Regulation/immunology
20.
J Neurosci ; 26(21): 5638-48, 2006 May 24.
Article in English | MEDLINE | ID: mdl-16723520

ABSTRACT

Growth arrest-specific protein 6 (gas6) activity is mediated through the receptor tyrosine kinase family members Axl, Rse, and Mer, all of which are expressed in human oligodendrocytes. In this study, we examined whether recombinant human (rh) gas6 protects oligodendrocytes from growth factor (insulin) withdrawal or tumor necrosis factor-alpha (TNFalpha) cytotoxicity. In addition, we examined whether the effect was caspase-dependent, which receptor mediated the protective effect, and whether survival required Akt1 activation. Oligodendrocyte viability was assessed by O4 staining and terminal deoxynucleotidyl transferase-mediated biotinylated UTP nick end labeling. Addition of rhgas6 to insulin-depleted cultures resulted in a significant increase in oligodendrocyte viability. Rhgas6 and caspase inhibitors also reduced active caspase-3 immunoreactivity relative to TNFalpha-only-treated cultures. In cultures treated with TNFalpha (100 ng/ml), the oligodendrocyte survival rate was 18% compared with cultures treated with TNFalpha and rhgas6 (64%) or the caspase inhibitors IETD-fmk [z-Ile-Glu(OMe)-Thr-Asp(OMe)-fluoromethyl ketone] (65%) and zVAD-fmk (N-benzyloxycarbonyl-Val-Ala-Asp-fluoromethyl ketone) (63%). Increased phosphoAkt (Ser473) immunoreactivity was detected 15 min after administration of gas6 and TNFalpha to oligodendrocyte cultures but not in TNFalpha-treated cultures. The gas6 protective effect was abrogated by the Axl decoy receptor Axl-Fc, by the phosphatidylinositol 3 (PI3) kinase inhibitor LY294002 [2-(4-morpholinyl)-8-phenyl-1(4H)-benzopyran-4-one], and in Akt1(-/-) oligodendrocytes. Oligodendrocyte cultures established from wild-type and Rse(-/-) mice, but not from Axl(-/-) mice, were also protected from TNFalpha-induced cell death when maintained in rhgas6. We conclude that gas6 signaling through the Axl receptor and the PI3 kinase/Akt1 survival pathway protects oligodendrocytes from growth factor withdrawal and TNFalpha-mediated cell death.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/administration & dosage , Apoptosis/drug effects , Apoptosis/physiology , Cell Survival/drug effects , Cells, Cultured , Cytoprotection/drug effects , Cytoprotection/physiology , Dose-Response Relationship, Drug , Enzyme Activation , Humans , Oligodendroglia/drug effects , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...