Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Glia ; 66(5): 987-998, 2018 05.
Article in English | MEDLINE | ID: mdl-29380422

ABSTRACT

Previously, we determined microRNA-31 (miR-31) is a noncoding tumor suppressive gene frequently deleted in glioblastoma (GBM); miR-31 suppresses tumor growth, in part, by limiting the activity of NF-κB. Herein, we expand our previous studies by characterizing the role of miR-31 during neural precursor cell (NPC) to astrocyte differentiation. We demonstrate that miR-31 expression and activity is suppressed in NPCs by stem cell factors such as Lin28, c-Myc, SOX2 and Oct4. However, during astrocytogenesis, miR-31 is induced by STAT3 and SMAD1/5/8, which mediate astrocyte differentiation. We determined miR-31 is required for terminal astrocyte differentiation, and that the loss of miR-31 impairs this process and/or prevents astrocyte maturation. We demonstrate that miR-31 promotes astrocyte development, in part, by reducing the levels of Lin28, a stem cell factor implicated in NPC renewal. These data suggest that miR-31 deletions may disrupt astrocyte development and/or homeostasis.


Subject(s)
Astrocytes/metabolism , Cell Differentiation/physiology , MicroRNAs/metabolism , Neural Stem Cells/metabolism , Animals , Cells, Cultured , Fluorescent Antibody Technique , Immunoblotting , In Situ Hybridization , Mice, Inbred C57BL , RNA-Binding Proteins/metabolism , Real-Time Polymerase Chain Reaction , Xenopus laevis
2.
J Immunol ; 198(11): 4244-4254, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28468969

ABSTRACT

CK2 is a highly conserved and pleiotropic serine/threonine kinase that promotes many prosurvival and proinflammatory signaling pathways, including PI3K/Akt/mTOR and JAK/STAT. These pathways are essential for CD4+ T cell activation and polarization, but little is known about how CK2 functions in T cells. In this article, we demonstrate that CK2 expression and kinase activity are induced upon CD4+ T cell activation. Targeting the catalytic activity of CK2 using the next-generation small molecule inhibitor CX-4945 in vitro significantly and specifically inhibited mouse and human Th17 cell differentiation while promoting the generation of Foxp3+ regulatory T cells (Tregs). These findings were associated with suppression of PI3K/Akt/mTOR activation and STAT3 phosphorylation upon CX-4945 treatment. Furthermore, we demonstrate that CX-4945 treatment inhibits the maturation of Th17 cells into inflammatory IFN-γ-coproducing effector cells. The Th17/Treg axis and maturation of Th17 cells are major contributing factors to the pathogenesis of many autoimmune disorders, including multiple sclerosis. Using a murine model of multiple sclerosis, experimental autoimmune encephalomyelitis, we demonstrate that in vivo administration of CX-4945 targets Akt/mTOR signaling in CD4+ T cells and the Th17/Treg axis throughout disease. Importantly, CX-4945 treatment after disease initiation significantly reduced disease severity, which was associated with a significant decrease in the frequency of pathogenic IFN-γ+ and GM-CSF+ Th17 cells in the CNS. Our data implicate CK2 as a regulator of the Th17/Treg axis and Th17 cell maturation and suggest that CK2 could be targeted for the treatment of Th17 cell-driven autoimmune disorders.


Subject(s)
Casein Kinase II/metabolism , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Casein Kinase II/antagonists & inhibitors , Casein Kinase II/genetics , Cell Differentiation , Class I Phosphatidylinositol 3-Kinases , Encephalomyelitis, Autoimmune, Experimental/immunology , Gene Expression Regulation , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Lymphocyte Activation , Mice , Multiple Sclerosis/immunology , Multiple Sclerosis/physiopathology , Naphthyridines/pharmacology , Phenazines , Phosphatidylinositol 3-Kinases/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/physiology , Th1 Cells/immunology , Th17 Cells/physiology
3.
J Neurooncol ; 132(2): 219-229, 2017 04.
Article in English | MEDLINE | ID: mdl-28181105

ABSTRACT

Protein kinase CK2 is a ubiquitously expressed serine/threonine kinase composed of two catalytic subunits (α) and/or (α') and two regulatory (ß) subunits. The expression and kinase activity of CK2 is elevated in many different cancers, including glioblastoma (GBM). Brain tumor initiating cells (BTICs) are a subset of cells that are highly tumorigenic and promote the resistance of GBM to current therapies. We previously reported that CK2 activity promotes prosurvival signaling in GBM. In this study, the role of CK2 signaling in BTIC function was examined. We found that expression of CK2α was increased in CD133+ BTICs compared to CD133- cells within the same GBM xenolines. Treatment with CX-4945, an ATP-competitive inhibitor of CK2, led to reduced expression of Sox2 and Nestin, transcription factors important for the maintenance of stem cells. Similarly, inhibition of CK2 also reduced the frequency of CD133+ BTICs over the course of 7 days, indicating a role for CK2 in BTIC persistence and survival. Importantly, using an in vitro limiting dilution assay, we found that inhibition of CK2 kinase activity with CX-4945 or siRNA knockdown of the CK2 catalytic subunits reduced neurosphere formation in GBM xenolines of different molecular subtypes. Lastly, we found that inhibition of CK2 led to decreased EGFR levels in some xenolines, and combination treatment with CX-4945 and Gefitinib to inhibit CK2 and EGFR, respectively, provided optimal inhibition of viability of cells. Therefore, due to the integration of CK2 in multiple signaling pathways important for BTIC survival, CK2 is a promising target in GBM.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , AC133 Antigen/metabolism , Animals , Casein Kinase II/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Female , Gefitinib , Humans , Mice , Mice, Inbred C57BL , Mice, Nude , Naphthyridines/pharmacology , Phenazines , Pregnancy , Quinazolines/pharmacology , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
4.
J Biol Chem ; 291(30): 15830-40, 2016 07 22.
Article in English | MEDLINE | ID: mdl-27226638

ABSTRACT

Inflammation and endoplasmic reticulum (ER) stress are associated with many neurological diseases. ER stress is brought on by the accumulation of misfolded proteins in the ER, which leads to activation of the unfolded protein response (UPR), a conserved pathway that transmits signals to restore homeostasis or eliminate the irreparably damaged cell. We provide evidence that inhibition or genetic haploinsufficiency of protein kinase R-like endoplasmic reticulum kinase (PERK) can selectively control inflammation brought on by ER stress without impinging on UPR-dependent survival and adaptive responses or normal immune responses. Using astrocytes lacking one or both alleles of PERK or the PERK inhibitor GSK2606414, we demonstrate that PERK haploinsufficiency or partial inhibition led to reduced ER stress-induced inflammation (IL-6, CCL2, and CCL20 expression) without compromising prosurvival responses. In contrast, complete loss of PERK blocked canonical PERK-dependent UPR genes and promoted apoptosis. Reversal of eIF2α-mediated translational repression using ISRIB potently suppressed PERK-dependent inflammatory gene expression, indicating that the selective modulation of inflammatory gene expression by PERK inhibition may be linked to attenuation of eIF2α phosphorylation and reveals a previously unknown link between translational repression and transcription of inflammatory genes. Additionally, ER-stressed astrocytes can drive an inflammatory M1-like phenotype in microglia, and this can be attenuated with inhibition of PERK. Importantly, targeting PERK neither disrupted normal cytokine signaling in astrocytes or microglia nor impaired macrophage phagocytosis or T cell polarization. Collectively, this work suggests that targeting PERK may provide a means for selective immunoregulation in the context of ER stress without disrupting normal immune function.


Subject(s)
Astrocytes/immunology , Endoplasmic Reticulum Stress/immunology , Macrophages/immunology , Microglia/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , eIF-2 Kinase/immunology , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/immunology , Indoles/pharmacology , Inflammation/genetics , Inflammation/immunology , Mice , Mice, Knockout , Phosphorylation/drug effects , Phosphorylation/genetics , Phosphorylation/immunology , Signal Transduction/drug effects , Signal Transduction/genetics , eIF-2 Kinase/antagonists & inhibitors , eIF-2 Kinase/genetics
5.
Oncotarget ; 7(15): 20621-35, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-26967393

ABSTRACT

In glioma, microglia and macrophages are the largest population of tumor-infiltrating cells, referred to as glioma associated macrophages (GAMs). Herein, we sought to determine the role of Suppressor of Cytokine Signaling 3 (SOCS3), a negative regulator of Signal Transducer and Activator of Transcription 3 (STAT3), in GAM functionality in glioma. We utilized a conditional model in which SOCS3 deletion is restricted to the myeloid cell population. We found that SOCS3-deficient bone marrow-derived macrophages display enhanced and prolonged expression of pro-inflammatory M1 cytokines when exposed to glioma tumor cell conditioned medium in vitro. Moreover, we found that deletion of SOCS3 in the myeloid cell population delays intracranial tumor growth and increases survival of mice bearing orthotopic glioma tumors in vivo. Although intracranial tumors from mice with SOCS3-deficient myeloid cells appear histologically similar to control mice, we observed that loss of SOCS3 in myeloid cells results in decreased M2 polarized macrophage infiltration in the tumors. Furthermore, loss of SOCS3 in myeloid cells results in increased CD8+ T-cell and decreased regulatory T-cell infiltration in the tumors. These findings demonstrate a beneficial effect of M1 polarized macrophages on suppressing glioma tumor growth, and highlight the importance of immune cells in the tumor microenvironment.


Subject(s)
Brain Neoplasms/pathology , Glioma/pathology , Myeloid Cells/pathology , Suppressor of Cytokine Signaling 3 Protein/physiology , T-Lymphocytes, Regulatory/pathology , Animals , Apoptosis , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Cell Proliferation , Glioma/genetics , Glioma/immunology , Mice , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Tumor Cells, Cultured , Tumor Microenvironment/immunology
6.
Oncotarget ; 5(15): 6484-96, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25153725

ABSTRACT

Breast cancer is the most common malignancy in women worldwide and remains a major cause of mortality, thus necessitating further therapeutic advancements. In breast cancer, numerous cell signaling pathways are aberrantly activated to produce the myriad phenotypes associated with malignancy; such pathways include the PI3K/Akt/mTOR, NF-κB and JAK/STAT cascades. These pathways are highly interconnected, but one prominent lateral enhancer of each is the remarkably promiscuous kinase CK2. CK2 expression has been shown to be elevated in cancer, thus implicating it in tumorigenesis through its effects on oncogenic signaling cascades. In this study, we identify aberrant expression of CK2 subunits in human breast samples from The Cancer Genome Atlas dataset. Additionally, two specific small molecule inhibitors of CK2, CX-4945 and TBB, were used to examine the role of CK2 in two human breast cancer cell lines, MDA-MB-231 and MCF-7 cells. We show that CK2 inhibition attenuates constitutive PI3K/Akt/mTOR, NF-κB and STAT3 activation and inducible NF-κB and JAK/STAT activation and downstream transcriptional activity. CX-4945 treatment caused a range of phenotypic changes in these cell lines, including decreased viability, cell cycle arrest, apoptosis and loss of migratory capacity. Overall, these results demonstrate the tremendous potential of CK2 as a clinical target in breast cancer.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Casein Kinase II/antagonists & inhibitors , Breast Neoplasms/metabolism , Casein Kinase II/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Female , Humans , MCF-7 Cells , Naphthyridines/pharmacology , Phenazines , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Tumor Cells, Cultured , Up-Regulation
7.
Arthritis Rheumatol ; 66(9): 2368-79, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24838610

ABSTRACT

OBJECTIVE: Fucosylation catalyzed by fucosyltransferases (FUTs) is an important posttranslational modification involved in a variety of biologic processes. This study was undertaken to determine the roles of fucosylation in rheumatoid arthritis (RA) and to assess the efficacy of reestablishing immune homeostasis with the use of 2-deoxy-d-galactose (2-d-gal), a fucosylation inhibitor. METHODS: Quantitative polymerase chain reaction was performed to determine the expression of FUT genes in synovial tissue from RA and osteoarthritis (OA) patients and in fluorescence-activated cell-sorted cells from RA synovial fluid. The in vivo inhibitory effect of 2-d-gal was evaluated in a murine collagen-induced arthritis (CIA) model. The in vitro effects of 2-d-gal on differentiation of inflammatory macrophages, production of cytokines, and antigen uptake, processing, and presentation functions were analyzed. RESULTS: FUTs that are involved in terminal or subterminal fucosylation, but not those involved in core fucosylation or O-fucosylation, were up-regulated in RA compared to OA synovial tissue. The expression of terminal FUTs was highly positively correlated with the expression of TNF (encoding for tumor necrosis factor α). Terminal FUTs were predominantly expressed in M1 macrophages. In vivo, 2-d-gal treatment of mice precluded the development of CIA by reducing inflammatory macrophages and Th17 cells in the draining lymph nodes and decreasing the levels of TNFα, interleukin-6 (IL-6), and antibodies to type II collagen in the serum. In vitro, treatment with 2-d-gal skewed the differentiation of M1 macrophages to IL-10-producing M2 macrophages. Furthermore, 2-d-gal significantly inhibited the antigen-presenting function of M1 macrophages. CONCLUSION: Terminal fucosylation is a novel hallmark of inflammatory macrophages. Inhibition of terminal FUTs reshapes the differentiation and functions of M1 macrophages, leading to resolution of inflammation in arthritis.


Subject(s)
Arthritis, Experimental/drug therapy , Galactose/analogs & derivatives , Macrophages/drug effects , Synovial Membrane/drug effects , Adult , Aged , Animals , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Female , Galactose/pharmacology , Galactose/therapeutic use , Humans , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Middle Aged , Osteoarthritis/metabolism , Osteoarthritis/pathology , Synovial Membrane/metabolism , Synovial Membrane/pathology
8.
J Immunol ; 192(1): 59-72, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24323580

ABSTRACT

Pathogenic Th cells and myeloid cells are involved in the pathogenesis of multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE), an animal model of MS. The JAK/STAT pathway is used by numerous cytokines for signaling and is critical for development, regulation, and termination of immune responses. Dysregulation of the JAK/STAT pathway has pathological implications in autoimmune and neuroinflammatory diseases. Many of the cytokines involved in MS/EAE, including IL-6, IL-12, IL-23, IFN-γ, and GM-CSF, use the JAK/STAT pathway to induce biological responses. Thus, targeting JAKs has implications for treating autoimmune inflammation of the brain. We have used AZD1480, a JAK1/2 inhibitor, to investigate the therapeutic potential of inhibiting the JAK/STAT pathway in models of EAE. AZD1480 treatment inhibits disease severity in myelin oligodendrocyte glycoprotein-induced classical and atypical EAE models by preventing entry of immune cells into the brain, suppressing differentiation of Th1 and Th17 cells, deactivating myeloid cells, inhibiting STAT activation in the brain, and reducing expression of proinflammatory cytokines and chemokines. Treatment of SJL/J mice with AZD1480 delays disease onset of PLP-induced relapsing-remitting disease, reduces relapses and diminishes clinical severity. AZD1480 treatment was also effective in reducing ongoing paralysis induced by adoptive transfer of either pathogenic Th1 or Th17 cells. In vivo AZD1480 treatment impairs both the priming and expansion of T cells and attenuates Ag presentation functions of myeloid cells. Inhibition of the JAK/STAT pathway has clinical efficacy in multiple preclinical models of MS, suggesting the feasibility of the JAK/STAT pathway as a target for neuroinflammatory diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Janus Kinases/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , STAT Transcription Factors/metabolism , Signal Transduction/drug effects , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/drug effects , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Female , Humans , Janus Kinases/antagonists & inhibitors , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Transgenic , Myeloid Cells/drug effects , Myeloid Cells/immunology , Myeloid Cells/metabolism , Pyrazoles/administration & dosage , Pyrimidines/administration & dosage , Th1 Cells/cytology , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/cytology , Th17 Cells/immunology , Th17 Cells/metabolism
9.
Eur J Immunol ; 44(4): 1137-42, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24356888

ABSTRACT

Mechanisms that modulate the generation of Th17 cells are incompletely understood. We report that the activation of casein kinase 2 (CK2) by CD5 is essential for the efficient generation of Th17 cells in vitro and in vivo. In our study, the CD5-CK2 signaling pathway enhanced TCR-induced activation of AKT and promoted the differentiation of Th17 cells by two independent mechanisms: inhibition of glycogen synthase kinase 3 (GSK3) and activation of mTOR. Genetic ablation of the CD5-CK2 signaling pathway attenuated TCR-induced AKT activation and consequently increased activity of GSK3 in Th17 cells. This resulted in increased sensitivity of Th17 cells to IFN-γ-mediated inhibition. In the absence of CD5-CK2 signaling, we observed decreased activity of S6K and attenuated nuclear translocation of RORγt (ROR is retinoic acid receptor related orphan receptor). These results reveal a novel and essential function of the CD5-CK2 signaling pathway and GSK3-IFN-γ axis in regulating Th-cell differentiation and provide a possible means to dampen Th17-type responses in autoimmune diseases.


Subject(s)
CD5 Antigens/immunology , Cell Differentiation/immunology , Interferon-gamma/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Th17 Cells/immunology , Animals , CD5 Antigens/genetics , CD5 Antigens/metabolism , Casein Kinase II/genetics , Casein Kinase II/immunology , Casein Kinase II/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Nucleus/immunology , Cell Nucleus/metabolism , Cells, Cultured , Flow Cytometry , Glycogen Synthase Kinase 3/immunology , Glycogen Synthase Kinase 3/metabolism , Immunohistochemistry , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Interferon/immunology , Receptors, Interferon/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Th17 Cells/metabolism , Interferon gamma Receptor
10.
J Immunol ; 191(6): 2967-77, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23960239

ABSTRACT

Type I IFNs (IFN-α and IFN-ß) and type II IFN (IFN-γ) mediate both regulation and inflammation in multiple sclerosis, neuromyelitis optica, and in experimental autoimmune encephalomyelitis (EAE). However, the underlying mechanism for these Janus-like activities of type I and II IFNs in neuroinflammation remains unclear. Although endogenous type I IFN signaling provides a protective response in neuroinflammation, we find that when IFN-γ signaling is ablated, type I IFNs drive inflammation, resulting in exacerbated EAE. IFN-γ has a disease stage-specific opposing function in EAE. Treatment of mice with IFN-γ during the initiation phase of EAE leads to enhanced severity of disease. In contrast, IFN-γ treatment during the effector phase attenuated disease. This immunosuppressive activity of IFN-γ required functional type I IFN signaling. In IFN-α/ß receptor-deficient mice, IFN-γ treatment during effector phase of EAE exacerbated disease. Using an adoptive transfer EAE model, we found that T cell-intrinsic type I and II IFN signals are simultaneously required to establish chronic EAE by encephalitogenic Th1 cells. However, in Th17 cells loss of either IFN signals leads to the development of a severe chronic disease. The data imply that type I and II IFN signals have independent but nonredundant roles in restraining encephalitogenic Th17 cells in vivo. Collectively, our data show that type I and II IFNs function in an integrated manner to regulate pathogenesis in EAE.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Interferon Type I/immunology , Interferon-gamma/immunology , Adoptive Transfer , Animals , Encephalomyelitis, Autoimmune, Experimental/metabolism , Immunohistochemistry , Interferon Type I/metabolism , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/immunology , Th17 Cells/immunology
11.
J Neurosci ; 33(23): 9592-600, 2013 Jun 05.
Article in English | MEDLINE | ID: mdl-23739956

ABSTRACT

Accumulation of α-synuclein (α-syn) in the brain is a core feature of Parkinson disease (PD) and leads to microglial activation, production of inflammatory cytokines and chemokines, T-cell infiltration, and neurodegeneration. Here, we have used both an in vivo mouse model induced by viral overexpression of α-syn as well as in vitro systems to study the role of the MHCII complex in α-syn-induced neuroinflammation and neurodegeneration. We find that in vivo, expression of full-length human α-syn causes striking induction of MHCII expression by microglia, while knock-out of MHCII prevents α-syn-induced microglial activation, antigen presentation, IgG deposition, and the degeneration of dopaminergic neurons. In vitro, treatment of microglia with aggregated α-syn leads to activation of antigen processing and presentation of antigen sufficient to drive CD4 T-cell proliferation and to trigger cytokine release. These results indicate a central role for microglial MHCII in the activation of both the innate and adaptive immune responses to α-syn in PD and suggest that the MHCII signaling complex may be a target of neuroprotective therapies for the disease.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Dopaminergic Neurons/metabolism , Genes, MHC Class II/physiology , Microglia/metabolism , Nerve Degeneration/metabolism , alpha-Synuclein/biosynthesis , Animals , Animals, Newborn , Cell Proliferation , Cells, Cultured , Dopaminergic Neurons/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Degeneration/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology
12.
PLoS One ; 7(12): e52658, 2012.
Article in English | MEDLINE | ID: mdl-23285134

ABSTRACT

Inhibitors of glycogen synthase kinase 3 (GSK3) are being explored as therapy for chronic inflammatory diseases. We previously demonstrated that the GSK inhibitor lithium is beneficial in experimental autoimmune encephalomyelitis (EAE), the mouse model of multiple sclerosis. In this study we report that lithium suppresses EAE induced by encephalitogenic interferon-γ (IFN-γ)-producing T helper (Th1) cells but not by interleukin (IL)-17-producing T helper (Th17) cells. The therapeutic activity of lithium required functional IFN-γ-signaling, but not the receptor for type I IFN (IFNAR). Inhibitor/s of GSK3 attenuated IFN-γ dependent activation of the transcription factor STAT1 in naïve T cells as well as in encephalitogenic T cells and Th1 cells. The inhibition of STAT1 activation was associated with reduced IFN-γ production and decreased expansion of encephalitogenic Th1 cells. Furthermore, lithium treatment induced Il27 expression within the spinal cords of mice with EAE. In contrast, such treatment of Ifngr(-/-) mice did not induce Il27 and was associated with lack of therapeutic response. Our study reveals a novel mechanism for the efficacy of GSK3 targeting in EAE, through the IFN-γ-STAT1 axis that is independent IFNAR-STAT1 axis. Overall our findings set the framework for the use of GSK3 inhibitors as therapeutic agents in autoimmune neuroinflammation.


Subject(s)
Autoimmunity , Central Nervous System/immunology , Central Nervous System/metabolism , Interferon-gamma/metabolism , Lithium/pharmacology , Signal Transduction/drug effects , Animals , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Interleukins/genetics , Interleukins/metabolism , Male , Mice , Mice, Knockout , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Phosphorylation/drug effects , STAT1 Transcription Factor/metabolism , Th1 Cells/drug effects , Th1 Cells/immunology , Th17 Cells/drug effects , Th17 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...