Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Immunobiology ; 223(10): 549-554, 2018 10.
Article in English | MEDLINE | ID: mdl-29960806

ABSTRACT

In this study, we investigated IL-10 and IL-17 specific immunomodulatory potential of S-nitrosoglutathione (GSNO), a physiological nitric oxide carrier molecule, in experimental autoimmune encephalomyelitis (EAE). In active EAE model, GSNO treatment attenuated EAE severity and splenic CD4+ T cells isolated from these mice exhibited decreased IL-17 expression without affecting the IFN-γ expression compared to the cells from untreated EAE mice. Similarly, adoptive transfer of these cells to nave mice resulted in reduction in IL-17 expression in the spinal cords of recipient mice with milder EAE severity. CD4+ T cells isolated from GSNO treated EAE mice, as compared to untreated EAE mice, still expressed lower levels of IL-17 under TH17 skewing conditions, but expressed similar levels of IFN-γ under TH1 skewing condition. Interestingly, under both TH17 and TH1 skewing condition, CD4+ T cells isolated from GSNO treated EAE mice, as compared to untreated EAE mice, expressed higher levels of IL-10 and adoptive transfer of these TH17 and TH1 skewed cells seemingly exhibited milder EAE disease. In addition, adoptive transfer of CD4+ T cells from GSNO treated EAE mice to active EAE mice also ameliorated EAE disease with induction of spinal cord expression of IL-10 and reduction in of IL-17, thus suggesting the participation of IL-10 mechanism in GSNO mediated immunomodulation. GSNO treatment of mice passively immunized with CD4+ T cells either from GSNO treated EAE mice or untreated mice further ameliorated EAE disease, supporting efficacy of GSNO for prophylaxis and therapy in EAE. Overall, these data document a modulatory role of GSNO in IL-17/IL-10 axis of EAE and other autoimmune diseases.


Subject(s)
Adoptive Transfer , Encephalomyelitis, Autoimmune, Experimental/therapy , Immunomodulation , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-17/metabolism , Nitric Oxide Donors/pharmacology , S-Nitrosoglutathione/pharmacology , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/transplantation , Encephalomyelitis, Autoimmune, Experimental/immunology , Mice , Mice, Inbred C57BL , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/transplantation , Th17 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/transplantation
2.
J Neuroimmune Pharmacol ; 5(2): 240-51, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20091246

ABSTRACT

S-nitrosoglutathione (GSNO) is a physiological nitric oxide molecule which regulates biological activities of target proteins via s-nitrosylation leading to attenuation of chronic inflammation. In this study we evaluated the therapeutic efficacy of GSNO in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. Oral administration of GSNO (0.5 or 1.0 mg/kg) reduced disease progression in chronic models (SJL and C57BL/6) of EAE induced with PLP((139-151)) or MOG((35-55)) peptides, respectively. GSNO attenuated EAE disease by reducing the production of IL17 (from Th(i) or Th17 cells) and the infiltration of CD4 T cells into the central nervous system without affecting the levels of Th1 (IFN gamma) and Th2 (IL4) immune responses. Inhibition of IL17 was observed in T cells under normal as well as Th17 skewed conditions. In vitro studies showed that the phosphorylation of STAT3 and expression of ROR gamma, key regulators of IL17 signaling, were reduced while phosphorylation of STAT4 or STAT6 and expression of T-bet or GATA3 remained unaffected, suggesting that GSNO preferentially targets Th17 cells. Collectively, GSNO attenuated EAE via modulation of Th17 cells and its effects are independent of Th1 or Th2 cells functions, indicating that it may have therapeutic potential for Th17-mediated autoimmune diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Nitric Oxide Donors/therapeutic use , S-Nitrosoglutathione/therapeutic use , Animals , Blotting, Western , CD4-Positive T-Lymphocytes/drug effects , Cell Differentiation/drug effects , Female , GATA3 Transcription Factor/metabolism , Glycoproteins/immunology , Interleukin-17/biosynthesis , Mice , Mice, Inbred C57BL , Myelin Proteolipid Protein/immunology , Myelin-Oligodendrocyte Glycoprotein , Nuclear Receptor Subfamily 1, Group F, Member 3/biosynthesis , Peptide Fragments/immunology , Phosphorylation , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Spinal Cord/pathology , Th1 Cells/immunology , Th2 Cells/immunology
3.
J Immunol ; 182(12): 8005-14, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19494326

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE) is a T cell-mediated autoimmune disease of the CNS. Metformin is the most widely used drug for diabetes and mediates its action via activating AMP-activated protein kinase (AMPK). We provide evidence that metformin attenuates the induction of EAE by restricting the infiltration of mononuclear cells into the CNS, down-regulating the expression of proinflammatory cytokines (IFN-gamma, TNF-alpha, IL-6, IL-17, and inducible NO synthase (iNOS)), cell adhesion molecules, matrix metalloproteinase 9, and chemokine (RANTES). Furthermore, the AMPK activity and lipids alterations (total phospholipids and in free fatty acids) were restored by metformin treatment in the CNS of treated EAE animals, suggesting the possible involvement of AMPK. Metformin activated AMPK in macrophages and thereby inhibited biosynthesis of phospholipids as well as neutral lipids and also down-regulated the expression of endotoxin (LPS)-induced proinflammatory cytokines and their mediators (iNOS and cyclooxygenase 2). It also attenuated IFN-gamma and IL-17-induced iNOS and cyclooxygenase 2 expression in RAW267.4 cells, further supporting its anti-inflammatory property. Metformin inhibited T cell-mediated immune responses including Ag-specific recall responses and production of Th1 or Th17 cytokines, while it induced the generation of IL-10 in spleen cells of treated EAE animals. Altogether these findings reveal that metformin may have a possible therapeutic value for the treatment of multiple sclerosis and other inflammatory diseases.


Subject(s)
Central Nervous System/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Metformin/therapeutic use , Multiple Sclerosis/drug therapy , AMP-Activated Protein Kinases/metabolism , Animals , Cells, Cultured , Central Nervous System/immunology , Central Nervous System/metabolism , Chronic Disease , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Lipid Metabolism/drug effects , Mice , Mice, Inbred C57BL , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Th1 Cells/drug effects , Th1 Cells/immunology
4.
Biochem Biophys Res Commun ; 386(1): 16-20, 2009 Aug 14.
Article in English | MEDLINE | ID: mdl-19486896

ABSTRACT

AMP-activated protein kinase (AMPK) is an energy sensing metabolic switch in mammalian cells. Here, we report our novel finding that AMPK is lost in all immune cells of experimental autoimmune encephalomyelitis (EAE), an inflammatory disease of Central Nervous System (CNS). AMPKalpha1 is predominantly expressed in T cells and antigen presenting cells (APCs), which are primarily involved in EAE disease progression. AMPK is lost at protein level in spleen macrophages, total T cells and their subsets (CD4, CD8 and regulatory T cells) isolated from EAE afflicted animals compared to control, without affecting its mRNA levels suggesting that the loss of AMPK protein is the result of posttranscriptional modification. To examine its pathological relevance in inflammatory disease, EAE was induced in wild type (+/+) and AMPKalpha1 null mice (-/-) using MOG(35-55) peptide. AMPKalpha1(-/-) mice exhibited severe EAE disease with profound infiltration of mononuclear cells compared to wild type mice however, AMPKalpha2 is not involved in enhancing the severity of the disease. Spleen cells isolated from AMPKalpha1(-/-) immunized mice exhibited a significant induction in the production of IFNgamma. Our study identifies AMPK as a down regulated target during disease in all immune cells and possibly restoring AMPK may serve as a novel therapeutic target in autoimmune diseases like multiple sclerosis (MS).


Subject(s)
AMP-Activated Protein Kinases/biosynthesis , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/pathology , AMP-Activated Protein Kinases/genetics , Animals , Down-Regulation , Isoenzymes/biosynthesis , Isoenzymes/genetics , Male , Mice , Mice, Mutant Strains , Protein Subunits/biosynthesis , Protein Subunits/genetics , Spleen/enzymology
5.
Prostate ; 69(12): 1334-42, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19517575

ABSTRACT

BACKGROUND: Paired homeobox 2 gene (PAX2) is a transcriptional regulator, aberrantly expressed in prostate cancer cells and its down-regulation promotes cell death in these cells. The molecular mechanisms of tumor progression by PAX2 over-expression are still unclear. However, it has been reported that angiotensin-II (A-II) induces cell growth in prostate cancer via A-II type 1 receptor (AT1R) and is mediated by the phosphorylation of mitogen activated protein kinase (MAPK) as well as signal transducer and activator of transcription 3 (STAT3). METHODS: Here we have demonstrated that A-II up-regulates PAX2 expression in prostate epithelial cells and prostate cancer cell lines resulting in increased cell growth. Furthermore, AT1R receptor antagonist losartan was shown to inhibit A-II induced PAX2 expression in prostate cancer. Moreover, analysis using pharmacological inhibitors against MEK1/2, ERK1/2, JAK-II, and phospho-STAT3 demonstrated that AT1R-mediated stimulatory effect of A-II on PAX2 expression was regulated in part by the phosphorylation of ERK1/2, JAK II, and STAT3 pathways. In addition, we have showed that down-regulation of PAX2 by an AT1R antagonist as well as JAK-II and STAT3 inhibitors suppress prostate cancer cell growth. RESULTS: Collectively, these findings show for the first time that the renin-angiotensin system (RAS) may promote prostate tumorigenesis via up-regulation of PAX2 expression. CONCLUSIONS: Therefore, PAX2 may be a novel therapeutic target for the treatment of carcinomas such as prostate cancer via the down-regulation of its expression by targeting the AT1R signaling pathways.


Subject(s)
Angiotensin II/physiology , PAX2 Transcription Factor/metabolism , Prostate/metabolism , Prostatic Neoplasms/metabolism , Receptor, Angiotensin, Type 2/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II Type 2 Receptor Blockers , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Enzyme Inhibitors/pharmacology , Humans , Losartan/pharmacology , Male , Phosphorylation , Prostate/drug effects , Prostate/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , RNA, Messenger/metabolism , Signal Transduction , Up-Regulation
6.
J Neurochem ; 105(5): 1820-33, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18248608

ABSTRACT

Krabbe disease (KD) is an inherited neurological disorder caused by the deficiency of galactocerebrosidase activity resulting in accumulation of psychosine, which leads to energy depletion, loss of oligodendrocytes, induction of gliosis, and inflammation by astrocytes in CNS. In this study, for the first time, we report the regulation of 'cellular energy switch,' AMP-activated protein kinase (AMPK), by psychosine in oligodendrocytes and astrocytes. Psychosine treatment significantly down-regulated AMPK activity, resulting in increased biosynthesis of lipids including cholesterol and free fatty acid in oligodendrocytes cell line (MO3.13) and primary astrocytes. Pharmacological activator of AMPK, 5-aminoimidazole-4-carboxamide-1-beta-4-ribofuranoside (AICAR) attenuated the psychosine-mediated down-regulation of AMPK and restored altered biosynthesis of lipids. AICAR treatment also down-regulated psychosine induced expression of proinflammatory cytokines and inducible nitric oxide synthase in primary astrocytes. However, AICAR treatment had no effect on psychosine induced-reactive oxygen species generation, arachidonic acid release, and death of oligodendrocytes; suggesting the specific role of AMPK in regulation of psychosine-mediated inflammatory response of astrocytes but not in cell death of oligodendrocytes. This study delineates an explicit role for AMPK in psychosine induced inflammation in astrocytes without directly affecting the cell death of oligodendrocytes. It also suggests that AMPK activating agents act as anti-inflammatory agents and can hold a therapeutic potential in Krabbe disease/twitcher disease, particularly when used in combination with drugs, which protect oligodendrocyte cell loss, such as sPLA2 inhibitor [Giri et al., J. Lipid Res. 47 (2006), 1478].


Subject(s)
Astrocytes/enzymology , Leukodystrophy, Globoid Cell/enzymology , Multienzyme Complexes/physiology , Oligodendroglia/enzymology , Protein Serine-Threonine Kinases/physiology , Psychosine/physiology , AMP-Activated Protein Kinases , Animals , Astrocytes/pathology , Astrocytes/physiology , Cell Death/physiology , Cells, Cultured , Down-Regulation/physiology , Leukodystrophy, Globoid Cell/metabolism , Leukodystrophy, Globoid Cell/pathology , Lipid Metabolism/physiology , Multienzyme Complexes/antagonists & inhibitors , Neural Pathways/physiology , Oligodendroglia/pathology , Oligodendroglia/physiology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Rats
7.
J Ocul Pharmacol Ther ; 23(3): 221-31, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17593005

ABSTRACT

PURPOSE: Experimental autoimmune uveitis (EAU), an animal model of human uveitis, is an organ-specific autoimmune disease mediated by various inflammatory cytokines. In particular, tumor necrosis factor (TNF)-alpha, interleukin (IL)-1beta and interferon (IFN)-gamma are known to play a role in its pathogenesis. S-nitrosothiol S-nitrosoglutathione (GSNO), a slow nitric oxide (NO) donor, was reported to have beneficial effects in inflammatory disease in ischemia-reperfusion injury. The efficacy of GSNO treatment on interphotoreceptor retinoid-binding protein (IRBP)-induced EAU was investigated, using functional, histologic, and immunologic readouts. METHODS: Mice were immunized with a single injection of IRBP(161180) peptide to induce EAU, followed by a daily treatment with GSNO (1 mg/kg). Electroretinogram (ERG) analysis, histopathology, and immunologic responses to IRBP were analyzed. The effects of GSNO treatment on the antigen-specific T-cell recall responses and their cytokine production were determined. RESULTS: A single immunization of IRBP(161180) peptide led to significant structural damage of the retina and concomitant elimination of ERGs. Daily oral GSNO treatment from days 1-14 following immunization was found to be effective against IRBP-induced EAU. Histopathologic and ERG analysis both demonstrated significant retinal protection in GSNO-treated mice. The GSNO treatment of EAU animals significantly attenuated the levels of TNF-alpha, IL-1beta, IFN-gamma, and IL-10 in retinas, as measured by quantitative real-time polymerase chain reaction analysis. The splenocytes isolated from EAU- and GSNO-treated mice had lower antigen-specific T-cell proliferation in response to IRBP protein, and their cytokine production was inhibited. CONCLUSIONS: The oral administration of GSNO significantly suppressed the levels of inflammatory mediators in the retinas of EAU mice. This suppression was associated with the maintenance of normal retinal histology and function. These results clearly demonstrated the therapeutic potential of GSNO in EAU, and provide new insights for the treatment of human uveitis.


Subject(s)
Autoimmune Diseases/prevention & control , Nitric Oxide Donors/pharmacology , S-Nitrosoglutathione/pharmacology , Uveitis/prevention & control , Administration, Oral , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/physiopathology , Cytokines/drug effects , Cytokines/metabolism , Disease Models, Animal , Electroretinography , Eye Proteins , Male , Mice , Photoreceptor Cells , Polymerase Chain Reaction , Retina/drug effects , Retina/physiopathology , Retinol-Binding Proteins , Spleen/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Uveitis/immunology , Uveitis/physiopathology
8.
Glia ; 55(1): 65-77, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17019693

ABSTRACT

S-Nitrosoglutathione (GSNO) is an endogenous nitric oxide carrier and recently, has been documented for its anti-inflammatory effects in rat model of cerebral ischemia (Khan et al. (2005) J Cereb Blood Flow Metab 25:177-192). Here, we explored the neuroprotective effects mediated by GSNO in Lewis rat model of EAE and its mechanism of action using in vitro model of monocyte-endothelial cell interaction. Oral administration of GSNO attenuated the clinical disease course in EAE animals by inhibiting the infiltration of vascular immune cells in the CNS that subsequently led to the reduction in the expression of proinflammatory cytokines and consequently limited demyelination. Based on the inhibition in infiltration of immune cells, we hypothesized that GSNO modulated endothelial cell activation that led to reduce cellular infiltration in the CNS. Using an in vitro model, we established that GSNO inhibited monocyte adhesion to the activated endothelial cell, which was mediated by down regulation of endothelial cell adhesion molecules (CAMs). The mechanism by which GSNO modulated CAMs expression appeared to be via S-nitrosylation of p65, which consequently inhibited nuclear factor kappa B (NF-kappaB) activation in endothelial cells. These observations suggest that GSNO exerts its protective effects in EAE by inhibition of cellular infiltration into the CNS by S-nitrosylation of p65, thereby modulating NF-kappaB-CAMs pathway in endothelial cells.


Subject(s)
Blood-Brain Barrier/drug effects , Central Nervous System/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Endothelial Cells/drug effects , Monocytes/drug effects , S-Nitrosoglutathione/pharmacology , Administration, Oral , Animals , Blood-Brain Barrier/physiology , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Adhesion Molecules/drug effects , Cell Adhesion Molecules/metabolism , Cell Line , Central Nervous System/immunology , Central Nervous System/physiopathology , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/immunology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Endothelial Cells/metabolism , Female , Mice , Monocytes/metabolism , NF-kappa B/metabolism , Nitric Oxide Donors/pharmacology , Nitric Oxide Donors/therapeutic use , Nitro Compounds/chemistry , Nitro Compounds/metabolism , Rats , Rats, Inbred Lew , S-Nitrosoglutathione/therapeutic use , Signal Transduction/drug effects , Signal Transduction/physiology , Transcription Factor RelA/drug effects , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/metabolism
9.
Immunology ; 118(3): 384-91, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16827899

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE) is mediated by myelin-specific CD4+ T helper 1 (Th1) cells, while recovery from the disease is associated with the presence of Th2 cells. Here we used animals with targeted deletion of the T-bet gene to determine its role in the progression of EAE. T-bet regulates the production of interferon-gamma (IFN-gamma) in CD4+ and natural killer cells, and CD4+ T cells from T-bet-deficient mice were unable to differentiate into a Th1 phenotype. Moreover BALB/c mice deficient in T-bet were resistant to the induction of EAE disease, with minimal inflammatory infiltrates in the central nervous system. These mice were resistant to EAE induction even when PLP(180-199) peptide specific effector T cells from BALB/c wild type were transferred to BALB/c T-bet-deficient mice. This resistance to EAE is may be caused by the production of the anti-inflammatory cytokine interleukin-10 (IL-10) from the spleen cells upon ex vivo stimulation with PLP(180-199) peptide and in vivo presence in the central nervous system. There was no difference in the recall responses in spleen cells from T-bet-deficient and wild type mice; however, less secretion of IFN-gamma was observed from primed splenocytes. The expression of IFN-gamma was less in the central nervous system of T-bet-deficient mice whereas IL-10 was significantly higher in T-bet-deficient as compared to wild type mice. These data indicate that T-bet genes play a critical role in maintaining the encephalitogenic nature of CD4+ T cells in autoimmune responses during EAE disease progression.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Transcription Factors/immunology , Adoptive Transfer , Animals , Apoptosis/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Coculture Techniques , Cytokines/biosynthesis , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/genetics , Female , Immunity, Innate , Immunologic Memory/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Spinal Cord/immunology , Spleen/immunology , T-Box Domain Proteins , Transcription Factors/deficiency , Transcription Factors/genetics
10.
J Neurosci Res ; 84(3): 614-25, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16770773

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE) is a model for studying multiple sclerosis (MS), a chronic demyelinating disorder of the CNS. 5-Aminoimidazole-4-carboxamide ribonucleoside (AICAR), an activator of AMP-activated protein kinase (AMPK), has been reported to show antiinflammatory and immunomodulatory effects in various models of inflammation. Recently, we have reported AICAR-mediated attenuation of active and passive EAE in mouse model [Nath et al. (2005) J. Immunol. 175:566-574]. Here we used a rat model of acute EAE to show antiinflammatory effects of AICAR after daily treatment starting at onset of the disease. By maintaining the blood-brain barrier (BBB), AICAR-administered animals showed lower clinical scores compared with untreated EAE animals. AICAR inhibited the infiltration of inflammatory cells across the BBB, resulting in lowered expression of proinflammatory mediators in the CNS and protection from severe demyelination. By using in vitro model of endothelial-leukocyte interaction, we showed that AICAR inhibited adhesion of monocytes to tumor necrosis factor-alpha-activated endothelial cells. One of the mechanisms of this action is through down-regulation of expression of endothelial cell adhesion molecules via modulation of nuclear factor kappaB activation. The data suggest that AICAR attenuates EAE progression by limiting infiltration of leukocytes across the BBB, thereby controlling the consequent inflammatory reaction in the CNS.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Blood-Brain Barrier/drug effects , Chemotaxis, Leukocyte/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Endothelial Cells/drug effects , Monocytes/drug effects , Ribonucleotides/pharmacology , Aminoimidazole Carboxamide/pharmacology , Aminoimidazole Carboxamide/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Blood-Brain Barrier/immunology , Blood-Brain Barrier/metabolism , Cell Adhesion/drug effects , Cell Adhesion/immunology , Cell Adhesion Molecules/drug effects , Cell Adhesion Molecules/immunology , Chemotaxis, Leukocyte/immunology , Disease Models, Animal , Down-Regulation/drug effects , Down-Regulation/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Endothelial Cells/metabolism , Female , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Mice , Monocytes/metabolism , NF-kappa B/immunology , NF-kappa B/metabolism , Rats , Rats, Inbred Lew , Ribonucleotides/therapeutic use , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
11.
J Immunol ; 175(1): 566-74, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15972693

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis, is a Th1-mediated inflammatory demyelinating disease of the CNS. AMP-activated protein kinase was reported recently to have anti-inflammatory activities by negatively regulating NF-kappaB signaling. In this study, we investigated the prophylactic and therapeutic efficacy of an AMP-activated protein kinase activator, 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR), in active and passive EAE induced by active immunization with PLP(139-151) or MOG(35-55) and in adoptive transfer of PLP(139-151)-sensitized T cells, respectively. In vivo treatment with AICAR exerted both prophylactic and therapeutic effects on EAE, attenuating the severity of clinical disease. The anti-inflammatory effects of AICAR were associated with the inhibition of the Ag-specific recall responses and inhibition of the Th1-type cytokines IFN-gamma and TNF-alpha, whereas it induced the production of Th2 cytokines IL-4 and IL-10. Treatment of PLP(139-151)-specific T cells in vitro with AICAR decreased their expression of T-bet in response to IL-12, a Th1 transcription factor, whereas in response to IL-4, it induced the expression and phosphorylation of Th2 transcription factors GATA3 and STAT6, respectively. Moreover, treatment of APCs in vitro with AICAR inhibited their capability to present the proteolipid protein peptide to PLP(139-151)-specific T cells. In an irrelevant Th1-mediated, OT-2 TCR transgenic mouse model, AICAR impaired in vivo Ag-specific expansion of CD4(+) T cells. Together, these findings show for the first time that AICAR is a novel immunomodulator with promising beneficial effects for the treatment of multiple sclerosis and other Th1-mediated inflammatory diseases.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Immunologic Factors/therapeutic use , Ribonucleotides/therapeutic use , AMP-Activated Protein Kinases , Aminoimidazole Carboxamide/therapeutic use , Animals , Antigen Presentation/drug effects , Cytokines/metabolism , DNA-Binding Proteins/biosynthesis , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Enzyme Activation/drug effects , Female , GATA3 Transcription Factor , Glycoproteins/immunology , Humans , Interleukin-10/biosynthesis , Mice , Mice, Inbred C57BL , Multienzyme Complexes/metabolism , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Myelin Proteolipid Protein/immunology , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments/immunology , Protein Serine-Threonine Kinases/metabolism , T-Box Domain Proteins , Th1 Cells/drug effects , Th1 Cells/immunology , Th2 Cells/drug effects , Th2 Cells/immunology , Trans-Activators/biosynthesis , Transcription Factors/antagonists & inhibitors
12.
J Neurochem ; 94(1): 204-14, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15953363

ABSTRACT

Integrity of the blood-brain barrier is essential for the normal functioning of CNS. Its disruption contributes to the pathobiology of various inflammatory neurodegenerative disorders. We have shown that the HMG-CoA reductase inhibitor (lovastatin) attenuated experimental autoimmune encephalomyelitis (EAE, an inflammatory disease of CNS) in rodents by inhibiting the infiltration of mononuclear cells into the CNS. Here, using an in vitro system, we report that lovastatin inhibits endothelial-monocyte cell interaction by down-regulating the expression of vascular cell adhesion molecule-1 and E-selectin by inhibiting the phosphoinositide 3 kinase (PI3-kinase)/protein kinase B (Akt)/nuclear factor-kappa B (NF-kappaB) pathway in endothelial cells. It inhibits tumor necrosis factor alpha (TNFalpha)-induced PI3-kinase, Akt and NF-kappaB activation in these cells. Co-transfection of constitutively active forms of PI3-kinase and Akt reversed the lovastatin-mediated inhibition of TNFalpha-induced adhesion, as well as activation of NF-kappaB, indicating the involvement of the PI3-kinase/Akt pathway in the interaction of adhesion molecules and the process of adhesion. This study reports that lovastatin down-regulates the pathway affecting the expression and interaction of adhesion molecules on endothelial cells, which in turn restricts the migration and infiltration of mononuclear cells thereby attenuating the pathogenesis of inflammatory diseases.


Subject(s)
Cell Communication/drug effects , Endothelial Cells/drug effects , Lovastatin/pharmacology , Monocytes/drug effects , NF-kappa B/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Communication/physiology , Cells, Cultured , Endothelial Cells/enzymology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Mice , Monocytes/enzymology , NF-kappa B/physiology , Phosphatidylinositol 3-Kinases/physiology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-akt , Signal Transduction/physiology
13.
J Immunol ; 172(2): 1273-86, 2004 Jan 15.
Article in English | MEDLINE | ID: mdl-14707106

ABSTRACT

The 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors or statins are newly identified immunomodulators. In vivo treatment of SJL/J mice with lovastatin reduced the duration and clinical severity of active and passive experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis. Lovastatin induced the expression of GATA3 and the phosphorylation of STAT6, whereas it inhibited tyrosine phosphorylation of Janus kinase 2, tyrosine kinase 2, and STAT4. Inhibition of the Janus kinase-STAT4 pathway by lovastatin modulated T0 to Th1 differentiation and reduced cytokine (IFN-gamma and TNF-alpha) production, thus inducing Th2 cytokines (IL-4, IL-5, and IL-10). It inhibited T-bet (T box transcription factor) and NF-kappaB in activated T cells and significantly reduced infiltration of CD4- and MHC class II-positive cells to CNS. Further, it stabilized IL-4 production and GATA-3 expression in differentiated Th2 cells, whereas in differentiated Th1 cells it inhibited the expression of T-bet and reduced the production of IFN-gamma. Moreover, lovastatin-exposed macrophage and BV2 (microglia) in allogeneic MLRs induced the production of the anti-inflammatory cytokine IL-10. These observations indicate that the anti-inflammatory effects of lovastatin are mediated via T cells as well as APCs, because it modulates the polarization patterns of naive T cell activation in an APC-independent system. Together, these findings reveal that lovastatin may have possible therapeutic value involving new targets (in both APCs and T cells) for the treatment of multiple sclerosis and other inflammatory diseases.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Multiple Sclerosis/drug therapy , Multiple Sclerosis/enzymology , Animals , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Line , Cell Migration Inhibition , Cell Movement/drug effects , DNA-Binding Proteins/biosynthesis , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Epitopes, T-Lymphocyte/immunology , Female , GATA3 Transcription Factor , Growth Inhibitors/pharmacology , Growth Inhibitors/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Injections, Intraperitoneal , Interphase/drug effects , Interphase/immunology , Lovastatin/administration & dosage , Lovastatin/pharmacology , Lovastatin/therapeutic use , Mice , Mice, Inbred Strains , Multiple Sclerosis/immunology , Multiple Sclerosis/prevention & control , Myelin Proteolipid Protein/antagonists & inhibitors , Myelin Proteolipid Protein/immunology , Neurons/drug effects , Neurons/immunology , Neurons/pathology , Spinal Cord/drug effects , Spinal Cord/pathology , T-Box Domain Proteins , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/enzymology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , Th1 Cells/drug effects , Th1 Cells/immunology , Th2 Cells/drug effects , Th2 Cells/immunology , Trans-Activators/biosynthesis , Transcription Factors/antagonists & inhibitors , Transcription Factors/biosynthesis , Up-Regulation/drug effects
14.
Blood ; 100(12): 4169-76, 2002 Dec 01.
Article in English | MEDLINE | ID: mdl-12393694

ABSTRACT

Bone marrow (BM)-derived dendritic cells (DCs) cultured in granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin 4 (IL-4) have been used to generate antitumor immune responses. The cytokine Flt3 ligand (Flt3L) also has been shown to generate BM DCs. We sought to determine if DCs generated by using Flt3L then matured with lipopolysaccharide (LPS) could lead to DCs with in vivo anti-acute myelogenous leukemia (anti-AML) activity. LPS and tumor necrosis factor alpha (TNF-alpha) are effective agents for maturing DCs; however, they have potential in vivo toxicities. Cytosine-phosphorothioate-guanine oligodeoxynucleotides (CpGs) are considered relatively nontoxic, potent activators of DC function and maturation in vitro and in vivo. We investigated whether CpGs would be comparable to TNF-alpha or LPS for the maturation of GM-CSF/IL-4-generated DCs. DCs cultured with GM-CSF/IL-4 and matured with TNF-alpha, LPS, or CpG produced a greater allogeneic T-cell response compared with Flt3L/LPS-generated DCs. All 4 distinct DC types were pulsed with AML-lysate and administered before tumor challenge produced an increase in the total number of splenic anti-AML-specific cytotoxic T-lymphocyte precursors and led to significantly (P < or =.0001) improved survival compared with nonvaccinated controls. GM-CSF/IL-4/LPS was superior to Flt3L/LPS for generating anti-AML effects in vivo. Whereas TNF-alpha was comparable to LPS in conferring on GM-CSF/IL-4 DCs anti-AML effects in vivo, CpGs were superior to LPS. These data have important clinical implications and are the first to show that Flt3L-generated DCs can provide antitumor protection and that nontoxic agents such as CpGs and Flt3L may be useful in the clinical development of DC vaccines.


Subject(s)
Adjuvants, Immunologic/pharmacology , Dendritic Cells/cytology , Immunotherapy, Adoptive/methods , Leukemia, Myeloid/therapy , Adjuvants, Immunologic/toxicity , Animals , Antigens, Neoplasm/immunology , Bone Marrow Cells/cytology , Cell Culture Techniques/methods , Cell Differentiation/drug effects , Dendritic Cells/drug effects , Dendritic Cells/immunology , Female , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Immunity, Cellular , Interleukin-4/pharmacology , Leukemia, Myeloid/immunology , Lipopolysaccharides/pharmacology , Lipopolysaccharides/toxicity , Membrane Proteins/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Oligonucleotides/pharmacology , Thionucleotides/pharmacology , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...