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










Publication year range
1.
Int J Mol Sci ; 21(20)2020 Oct 19.
Article in English | MEDLINE | ID: mdl-33086489

ABSTRACT

The infection of susceptible mice with Theiler's murine encephalomyelitis virus (TMEV) induces a T cell-mediated demyelinating disease. This system has been studied as a relevant infection model for multiple sclerosis (MS). Therefore, defining the type of T cell responses and their functions is critically important for understanding the relevant pathogenic mechanisms. In this study, we adoptively transferred naive VP2-specific TCR-Tg CD4+ T cells into syngeneic susceptible SJL mice and monitored the development of the disease and the activation and proliferation of CD4+ T cells during the early stages of viral infection. The preexisting VP2-specific naive CD4+ T cells promoted the pathogenesis of the disease in a dose-dependent manner. The transferred VP2-specific CD4+ T cells proliferated rapidly in the CNS starting at 2-3 dpi. High levels of FoxP3+CD4+ T cells were found in the CNS early in viral infection (3 dpi) and persisted throughout the infection. Activated VP2-specific FoxP3+CD4+ T cells inhibited the production of IFN-γ, but not IL-17, via the same VP2-specific CD4+ T cells without interfering in proliferation. Thus, the early presence of regulatory T cells in the CNS with viral infection may favor the induction of pathogenic Th17 cells over protective Th1 cells in susceptible mice, thereby establishing the pathogenesis of virus-induced demyelinating disease.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cardiovirus Infections/immunology , Cardiovirus Infections/virology , Central Nervous System/immunology , Central Nervous System/virology , Theilovirus/physiology , Adoptive Transfer , Animals , Cell Proliferation , Central Nervous System/pathology , Cytokines/biosynthesis , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Demyelinating Diseases/virology , Forkhead Transcription Factors/metabolism , Interferon-gamma/metabolism , Interleukin-17/biosynthesis , Mice, Transgenic , Receptors, Antigen, T-Cell/metabolism , Species Specificity
2.
J Virol ; 89(3): 1867-78, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25428872

ABSTRACT

UNLABELLED: Intracerebral infection with Theiler's murine encephalomyelitis virus (TMEV) induces immune-mediated demyelinating disease in susceptible SJL/J mice but not in resistant C57BL/6 mice. Previous studies have indicated that the major histocompatibility complex (MHC) genes play the most prominent role in the development of TMEV-induced demyelinating disease. In this study, we used C57BL/6.S (B6.S) congenic mice, which carry H-2(s) MHC genes instead of H-2(b) MHC genes in conjunction with the C57BL/6 (B6) background genes. Our data show that virus-infected B6.S mice are free from disease and have significantly lower viral loads than susceptible SJL mice, particularly in the spinal cord. A strong protective Th1-type T helper response with virtually no pathogenic Th17 response was detected in B6.S mice, in contrast to the reduced Th1- and robust Th17-type responses in SJL mice. Notably, lower levels of viral infectivity in B6.S antigen-presenting cells (APCs) correlated with the disease resistance and T-cell-type response. In vitro studies using APCs from B6.S and SJL mice show that TLR2, -3, -4, and -7, but not TLR9, signaling can replace viral infection and augment the effect of viral infection in the differentiation of the pathogenic Th17 cell type. Taken together, these results strongly suggest that the viral replication levels in APCs critically affect the induction of protective versus pathogenic Th cell types via the signaling of pattern recognition receptors for innate immune responses. Our current findings further imply that the levels of viral infectivity/replication and TLR-mediated signaling play critical roles in the pathogenesis of chronic viral diseases. IMPORTANCE: This study indicates that innate immune cytokines produced in antigen-presenting cells stimulating the T cell immune responses during early viral infection play a critical role in determining the susceptibility of mice to the development of demyelinating disease. The level of innate immune cytokines reflects the level of initial viral infection in the antigen-presenting cells, and the level determines the development of T cell types, which are either protective or pathogenic. The level of initial viral infection to the cells is controlled by a gene or genes that are not associated with the major histocompatibility antigen complex genes. This finding has an important implication in controlling not only chronic viral infections but also infection-induced autoimmune-like diseases, which are closely associated with the pathogenic type of T cell responses.


Subject(s)
Antigen-Presenting Cells/virology , Cardiovirus Infections/pathology , Cardiovirus Infections/virology , Demyelinating Diseases/pathology , Demyelinating Diseases/virology , Theilovirus/isolation & purification , Viral Load , Animals , Antigen-Presenting Cells/immunology , Cardiovirus Infections/immunology , Demyelinating Diseases/immunology , Female , Genes, MHC Class I , Mice, Inbred C57BL , Th1 Cells/immunology , Th17 Cells/immunology , Theilovirus/immunology , Toll-Like Receptors/immunology
3.
J Virol ; 88(15): 8479-89, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24829345

ABSTRACT

UNLABELLED: Interleukin-6 (IL-6) plays an important role in the development and progression of inflammatory responses, autoimmune diseases, and cancers. Many viral infections, including Theiler's murine encephalomyelitis virus (TMEV), result in the vigorous production of IL-6. However, the role of IL-6 in the development of virus-induced inflammatory responses is unclear. The infection of susceptible mice with TMEV induces the development of chronic demyelinating disease, which is considered a relevant infectious model for multiple sclerosis. In this study, we demonstrate that resistant C57BL/6 mice carrying an IL-6 transgene (IL-6 Tg) develop a TMEV-induced demyelinating disease accompanied by an increase in viral persistence and an elevated Th17 cell response in the central nervous system. Either IL-6 or IL-17 induced the expression of Bcl-2 and Bcl-xL at a high concentration. The upregulated expression of prosurvival molecules in turn inhibited target cell destruction by virus-specific CD8(+) T cells. More interestingly, IL-6 and IL-17 synergistically promoted the expression of these prosurvival molecules, preventing cellular apoptosis at a much lower (<5-fold) concentration. The signals involved in the synergy appear to include the activation of both STAT3 and NF-κB via distinct cytokine-dependent pathways. Thus, the excessive IL-6 promotes the generation of Th17 cells, and the resulting IL-6 and IL-17 synergistically promote viral persistence by protecting virus-infected cells from apoptosis and CD8(+) T cell-mediated target destruction. These results suggest that blocking both IL-6 and IL-17 functions are important considerations for therapies of chronic viral diseases, autoimmune diseases, and cancers. IMPORTANCE: This study indicates that an excessive level of IL-6 cytokine produced following viral infection promotes the development of IL-17-producing pathogenic helper T cells. We demonstrate here for the first time that IL-6 together with IL-17 synergistically enhances the expression of survival molecules to hinder critical host defense mechanisms removing virus-infected cells. This finding has an important implication in controlling not only chronic viral infections but also autoimmune diseases and cancers, which are associated with prolonged cell survival.


Subject(s)
Apoptosis , Immune Evasion , Interleukin-17/metabolism , Interleukin-6/metabolism , T-Lymphocytes, Cytotoxic/immunology , Theilovirus/immunology , Theilovirus/physiology , Animals , Female , Mice , Mice, Inbred C57BL , Mice, Transgenic
4.
J Virol ; 87(21): 11538-51, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23966393

ABSTRACT

Infection with Theiler's murine encephalomyelitis virus (TMEV) in the central nervous system (CNS) of susceptible mice results in an immune-mediated demyelinating disease which is considered a relevant viral model of human multiple sclerosis. We previously demonstrated that the expression of positive costimulatory molecules (CD40, CD80, and CD86) is higher on the microglia of TMEV-resistant C57BL/6 (B6) mice than the microglia of TMEV-susceptible SJL/J (SJL) mice. In this study, we analyzed the expression levels of the negative costimulatory molecules PD-1 and PDL-1 in the CNS of TMEV-infected SJL mice and B6 mice. Our results indicated that TMEV infection induces the expression of both PD-1 and PDL-1 on microglia and macrophages in the CNS but not in the periphery. The expression of PD-1 only on CNS-infiltrating macrophages and not on resident microglia was considerably higher (>4-fold) in TMEV-infected SJL mice than TMEV-infected B6 mice. We further demonstrated that interleukn-6 (IL-6) is necessary to induce the maximal expression of PDL-1 but not PD-1 after TMEV infection using IL-6-deficient mice and IL-6-transgenic mice in conjunction with recombinant IL-6. In addition, cells from type I interferon (IFN) receptor knockout mice failed to upregulate PD-1 and PDL-1 expression after TMEV infection in vitro, indicating that type I IFN signaling is associated with the upregulation. However, other IFN signaling may also participate in the upregulation. Taken together, these results strongly suggest that the expression of PD-1 and PDL-1 in the CNS is primarily upregulated following TMEV infection via type I IFN signaling and the maximal expression of PDL-1 additionally requires IL-6 signaling.


Subject(s)
B7-H1 Antigen/metabolism , Cardiovirus Infections/virology , Central Nervous System/virology , Interleukin-6/immunology , Microglia/virology , Programmed Cell Death 1 Receptor/metabolism , Theilovirus/immunology , Animals , Cardiovirus Infections/immunology , Female , Macrophages/virology , Mice , Mice, Knockout , Mice, Transgenic
5.
J Virol ; 86(24): 13717-28, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23055563

ABSTRACT

Theiler's virus-induced demyelinating disease has been extensively investigated as a model for persistent viral infection and multiple sclerosis (MS). However, the role of CD8(+) T cells in the development of disease remains unclear. To assess the role of virus-specific CD8(+) T cells in the pathogenesis of demyelinating disease, a single amino acid substitution was introduced into the predominant viral epitope (VP3 from residues 159 to 166 [VP3(159-166)]) and/or a subdominant viral epitope (VP3(173-181)) of susceptible SJL/J mice by site-directed mutagenesis. The resulting variant viruses (N160V, P179A, and N160V/P179A) failed to induce CD8(+) T cell responses to the respective epitopes. Surprisingly, mice infected with N160V or N160V/P179A virus, which lacks CD8(+) T cells against VP3(159-166), did not develop demyelinating disease, in contrast to wild-type virus or P179A virus lacking VP3(173-181)-specific CD8(+) T cells. Our findings clearly show that the presence of VP3(159-166)-specific CD8(+) T cells, rather than viral persistence itself, is strongly correlated with disease development. VP3(173-181)-specific CD8(+) T cells in the central nervous system (CNS) of these virus-infected mice expressed higher levels of transforming growth factor ß, forkhead box P3, interleukin-22 (IL-22), and IL-17 mRNA but caused minimal cytotoxicity compared to that caused by VP3(159-166)-specific CD8(+) T cells. VP3(159-166)-specific CD8(+) T cells exhibited high functional avidity for gamma interferon production, whereas VP3(173-181)-specific CD8(+) T cells showed low avidity. To our knowledge, this is the first report indicating that the induction of the IL-17-producing CD8(+) T cell type is largely epitope specific and that this specificity apparently plays a differential role in the pathogenicity of virus-induced demyelinating disease. These results strongly advocate for the careful consideration of CD8(+) T cell-mediated intervention of virus-induced inflammatory diseases.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes/immunology , Multiple Sclerosis/immunology , Virus Diseases/immunology , Animals , Cell Line , Cricetinae , Disease Models, Animal , Mice , Multiple Sclerosis/virology , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Virus Replication
6.
J Neuroinflammation ; 9: 217, 2012 Sep 17.
Article in English | MEDLINE | ID: mdl-22985464

ABSTRACT

BACKGROUND: Theiler's virus infection induces chronic demyelinating disease in mice and has been investigated as an infectious model for multiple sclerosis (MS). IL-1 plays an important role in the pathogenesis of both the autoimmune disease model (EAE) and this viral model for MS. However, IL-1 is known to play an important protective role against certain viral infections. Therefore, it is unclear whether IL-1-mediated signaling plays a protective or pathogenic role in the development of TMEV-induced demyelinating disease. METHODS: Female C57BL/6 mice and B6.129S7-Il1r1tm1Imx/J mice (IL-1R KO) were infected with Theiler's murine encephalomyelitis virus (1 x 106 PFU). Differences in the development of demyelinating disease and changes in the histopathology were compared. Viral persistence, cytokine production, and immune responses in the CNS of infected mice were analyzed using quantitative PCR, ELISA, and flow cytometry. RESULTS: Administration of IL-1ß, thereby rending resistant B6 mice susceptible to TMEV-induced demyelinating disease, induced a high level of Th17 response. Interestingly, infection of TMEV into IL-1R-deficient resistant C57BL/6 (B6) mice also induced TMEV-induced demyelinating disease. High viral persistence was found in the late stage of viral infection in IL-1R-deficient mice, although there were few differences in the initial anti-viral immune responses and viral persistent levels between the WT B6 and IL-1R-deficiecent mice. The initial type I IFN responses and the expression of PDL-1 and Tim-3 were higher in the CNS of TMEV-infected IL-1R-deficient mice, leading to deficiencies in T cell function that permit viral persistence. CONCLUSIONS: These results suggest that the presence of high IL-1 level exerts the pathogenic role by elevating pathogenic Th17 responses, whereas the lack of IL-1 signals promotes viral persistence in the spinal cord due to insufficient T cell activation by elevating the production of inhibitory cytokines and regulatory molecules. Therefore, the balance of IL-1 signaling appears to be extremely important for the protection from TMEV-induced demyelinating disease, and either too much or too little signaling promotes the development of disease.


Subject(s)
Demyelinating Diseases/virology , Interleukin-1beta/physiology , Poliomyelitis/virology , Signal Transduction/immunology , Animals , Demyelinating Diseases/etiology , Demyelinating Diseases/pathology , Disease Models, Animal , Female , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/etiology , Multiple Sclerosis/pathology , Multiple Sclerosis/virology , Poliomyelitis/etiology , Poliomyelitis/pathology , Theilovirus/growth & development , Theilovirus/immunology
7.
Blood ; 119(13): 3128-31, 2012 Mar 29.
Article in English | MEDLINE | ID: mdl-22310910

ABSTRACT

Surprisingly little is known about the interaction of human blood mononuclear cells with viruses. Here, we show that monocytes are the predominant cell type infected when peripheral blood mononuclear cells are exposed to viruses ex vivo. Remarkably, infection with vesicular stomatitis virus, vaccinia virus, and a variety of influenza A viruses (including circulating swine-origin virus) induces monocytes to differentiate within 18 hours into CD16(-)CD83(+) mature dendritic cells with enhanced capacity to activate T cells. Differentiation into dendritic cells does not require cell division and occurs despite the synthesis of viral proteins, which demonstrates that monocytes counteract the capacity of these highly lytic viruses to hijack host cell biosynthetic capacity. Indeed, differentiation requires infectious virus and viral protein synthesis. These findings demonstrate that monocytes are uniquely susceptible to viral infection among blood mononuclear cells, with the likely purpose of generating cells with enhanced capacity to activate innate and acquired antiviral immunity.


Subject(s)
Cell Differentiation , Dendritic Cells/physiology , Monocytes/physiology , Virus Diseases/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Cell Differentiation/immunology , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/virology , Humans , Influenza A virus/immunology , Influenza A virus/physiology , Influenza, Human/blood , Influenza, Human/immunology , Monocytes/immunology , Monocytes/virology , Time Factors , Vaccinia/blood , Vaccinia/immunology , Vaccinia virus/immunology , Vaccinia virus/physiology , Vesicular Stomatitis/blood , Vesicular Stomatitis/immunology , Vesiculovirus/immunology , Vesiculovirus/physiology , Virus Diseases/physiopathology
8.
J Neuroimmunol ; 226(1-2): 27-37, 2010 Sep 14.
Article in English | MEDLINE | ID: mdl-20538350

ABSTRACT

Theiler's murine encephalomyelitis virus (TMEV) establishes a persistent infection in the central nervous system (CNS). To examine the role of type I interferon (IFN-I)-mediated signals in TMEV infection, mice lacking a subunit of the type I IFN receptor (IFN-IR KO mice) were utilized. In contrast to wild type mice, IFN-IR KO mice developed rapid fatal encephalitis accompanied with greater viral load and infiltration of immune cells to the CNS. The proportion of virus-specific CD4(+) and CD8(+) T cell responses in the CNS was significantly lower in IFN-IR KO mice during the early stage of infection. Levels of IFN-γ and IL-17 produced by isolated primed CD4(+) T cells in response to DCs from TMEV-infected IFN-IR KO mice were also lower than those stimulated by DCs from TMEV-infected wild type control mice. The less efficient stimulation of virus-specific T cells by virus-infected antigen-presenting cells is attributable in part to the low level expression of activation markers on TMEV-infected cells from IFN-IR KO mice. However, due to high levels of cellular infiltration and viral loads in the CNS, the overall numbers of virus-specific T cells are higher in IFN-IR KO mice during the later stage of viral infection. These results suggest that IFN-I-mediated signals play important roles in controlling cellular infiltration to the CNS and shaping local T cell immune responses.


Subject(s)
Central Nervous System/immunology , Enterovirus Infections/immunology , Enterovirus Infections/pathology , Neutrophil Infiltration/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Animals , Antigen-Presenting Cells/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation/drug effects , Central Nervous System/virology , Cytokines/immunology , Cytokines/metabolism , Enterovirus Infections/virology , Female , Flow Cytometry/methods , Interferon Type I/deficiency , Interleukin-17/metabolism , Mice , Mice, Knockout , Neutrophil Infiltration/drug effects , Receptor, Interferon alpha-beta/deficiency , Signal Transduction/physiology , T-Lymphocytes/virology , Theilovirus/pathogenicity
9.
J Exp Med ; 206(2): 313-28, 2009 Feb 16.
Article in English | MEDLINE | ID: mdl-19204109

ABSTRACT

Persistent viral infection and its associated chronic diseases are a global health concern. Interleukin (IL) 17-producing Th17 cells have been implicated in the pathogenesis of various autoimmune diseases, and in protection from bacterial or fungal infection. However, the role of Th17 cells in persistent viral infection remains unknown. We report that Th17 cells preferentially develop in vitro and in vivo in an IL-6-dependent manner after Theiler's murine encephalomyelitis virus infection. Th17 cells promote persistent viral infection and induce the pathogenesis of chronic demyelinating disease. IL-17 up-regulates antiapoptotic molecules and, consequently, increases persistent infection by enhancing the survival of virus-infected cells and blocking target cell destruction by cytotoxic T cells. Neutralization of IL-17 augments virus clearance by eliminating virus-infected cells and boosting lytic function by cytotoxic T cells, leading to the prevention of disease development. Thus, these results indicate a novel pathogenic role of Th17 cells via IL-17 in persistent viral infection and its associated chronic inflammatory diseases.


Subject(s)
Cardiovirus Infections/complications , Cardiovirus Infections/immunology , Demyelinating Diseases/etiology , Gene Expression Regulation/immunology , T-Lymphocytes, Helper-Inducer/immunology , Theilovirus/immunology , Animals , Cytotoxicity Tests, Immunologic , Demyelinating Diseases/virology , Flow Cytometry , Interleukin-17/immunology , Interleukin-6/genetics , Mice , Mice, Transgenic
10.
PLoS Pathog ; 3(8): e124, 2007 Aug 24.
Article in English | MEDLINE | ID: mdl-17722981

ABSTRACT

Although persistent viral diseases are a global health concern, the mechanisms of differential susceptibility to such infections among individuals are unknown. Here, we report that differential interactions between dendritic cells (DCs) and virus are critical in determining resistance versus susceptibility in the Theiler murine encephalomyelitis virus-induced demyelinating disease model of multiple sclerosis. This virus induces a chronic demyelinating disease in susceptible mice, whereas the virus is completely cleared in resistant strains of mice. DCs from susceptible mice are more permissive to viral infection, resulting in severe deficiencies in development, expansion, and function, in contrast to DCs from resistant mice. Although protective prior to viral infection, higher levels of type I interferons (IFNs) and IFN-gamma produced by virus-infected DCs from susceptible mice further contribute to the differential inhibition of DC development and function. An increased DC number and/or acquired resistance of DCs to viral infection render susceptible mice resistant to viral persistence and disease progression. Thus, the differential permissiveness of DCs to infectious agents and its subsequent functional and developmental deficiencies determine the outcome of infection- associated diseases. Therefore, arming DCs against viral infection-induced functional decline may provide a useful intervention for chronic infection-associated diseases.


Subject(s)
Cardiovirus Infections/immunology , Demyelinating Diseases/immunology , Dendritic Cells/immunology , Theilovirus/immunology , Animals , Brain/pathology , Brain/virology , Cardiovirus Infections/pathology , Cardiovirus Infections/virology , Cell Count , Cytokines/metabolism , Demyelinating Diseases/pathology , Demyelinating Diseases/virology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Disease Susceptibility/immunology , Female , Gene Expression , Gene Silencing , Interferon Type I/genetics , Interferon Type I/metabolism , Interferon-gamma/genetics , Interferon-gamma/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , RNA, Messenger/metabolism , Spinal Cord/pathology , Spinal Cord/virology , Theilovirus/pathogenicity , Virus Replication/immunology
11.
Virology ; 360(1): 159-71, 2007 Mar 30.
Article in English | MEDLINE | ID: mdl-17095033

ABSTRACT

Theiler's virus infection induces an immune-mediated demyelinating disease, providing a relevant animal model of human multiple sclerosis. VP2(121-130)-specific CD8+ T cells in resistant H-2b mice account for the majority of CNS-infiltrating CD8+ T cells. To further study the role of the CD8(+) T cells, we generated a panel of mutant viruses substituted with L, G, or T at the anchor residue (M130) of the VP2(121-130) epitope. M130L virus (M130L-V) with a substitution of M with L displayed similar properties as wild-type virus (WT-V). However, M130G-V and M130T-V could not establish a persistent infection in the CNS. The level of both virus-specific CD8+ and CD4+ T cell responses is significantly reduced in mice infected with these variant viruses. While all mutant and wild-type viruses replicate comparably in BHK cells, replication of M130G-V and M130T-V in macrophages was significantly lower compared to those infected with WT-V and M130L-V. Interestingly, these mutant viruses deficient in replication in primary mouse cells showed drastically reduced binding ability to the cells. These results suggest that the anchor residue of the predominant CD8+ T cell epitope of TMEV in resistant mice is critical for the virus to infect target cells and this deficiency may result in poor viral persistence leading to correspondingly low T cell responses in the periphery and CNS. Thus, selection of the cellular binding region of the virus as the predominant epitope for CD8+ T cells in resistant mice may provide a distinct advantage in controlling viral persistence by preventing escape mutations.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Capsid Proteins/immunology , Epitopes, T-Lymphocyte/immunology , Immunodominant Epitopes/immunology , Multiple Sclerosis/etiology , Theilovirus/physiology , Animals , Brain/immunology , Brain/virology , CD4-Positive T-Lymphocytes/immunology , Cell Line , Cells, Cultured , Disease Models, Animal , Epitopes, T-Lymphocyte/genetics , Female , H-2 Antigens/immunology , Histocompatibility Antigen H-2D , Lymphocyte Activation , Macrophages/virology , Mice , Mice, Inbred C57BL , Mutation , Spinal Cord/immunology , Spinal Cord/virology , Virus Replication
12.
J Immunol ; 177(2): 934-43, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16818748

ABSTRACT

IFN-gamma is considered to be a Th1 cytokine with immunomodulatory effects on a variety of immune cells. In this study, we determined whether dendritic cell (DC) function was aberrant in IFN-gamma knockout (GKO) mice. The results demonstrated that IFN-gamma deficiency did not interfere with bone marrow-derived DC development and maturation in vitro. However, functional analysis showed that bone marrow-derived DC from GKO mice had altered cytokine secretion, allostimulatory and Ag presentation capacity, chemokine receptor expression, and in vitro chemotaxis. LPS induced the recruitment of DC from different organs into the spleen; epicutaneously sensitized DC with hapten (FITC) accumulated in the draining lymph nodes and CD11c(+) DC levels in the draining lymph nodes from autoantigen (interphotoreceptor retinoid-binding protein) immunized mice were enhanced in GKO mice as compared with wild-type mice. After treatment of GKO mice with i.p. IFN-gamma injection restored IFN-gamma levels in vivo, DC migration decreased in response to LPS or FITC. IFN-gamma altered the adaptive immune responses in vivo, since T cell priming and IL-2 production were increased in interphotoreceptor retinoid-binding protein-immunized GKO mice. Furthermore, in IFN-gamma-treated GKO mice, experimental autoimmune uveitis score enhancement and T cell activation were eliminated. Taken together, IFN-gamma appears to play a negative regulatory role on in vivo DC function, resulting in suppression of Ag-specific T cell priming.


Subject(s)
Cell Movement/immunology , Dendritic Cells/cytology , Down-Regulation/immunology , Haptens/administration & dosage , Interferon-gamma/physiology , T-Lymphocytes/immunology , Animals , Antigen Presentation/genetics , Autoimmune Diseases/prevention & control , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cells, Cultured , Chemotaxis, Leukocyte/genetics , Cytokines/biosynthesis , Dendritic Cells/immunology , Dendritic Cells/metabolism , Down-Regulation/genetics , Fluorescein-5-isothiocyanate/administration & dosage , Haptens/immunology , Interferon-gamma/administration & dosage , Interferon-gamma/deficiency , Interferon-gamma/genetics , Lymphocyte Activation/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptors, Chemokine/biosynthesis , Recombinant Proteins , T-Lymphocytes/metabolism , Uveitis, Anterior/immunology , Uveitis, Anterior/prevention & control
13.
J Neuroimmunol ; 137(1-2): 154-63, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12667660

ABSTRACT

IFN-gamma production is a hallmark of Th1 response, and IFN-gamma has multiple roles in Th1 development, depending on the experimental conditions. In this study, the correlation between IFN-gamma and interleukin 12 receptor beta2 (IL-12R beta 2) expression was investigated in experimental autoimmune uveitis (EAU) susceptible B10.A and resistant BALB/c mice. B10.A mice expressed high IL-12R beta 2 on T cells either in the disease target eyes or draining lymph node cells (LNC), and its expression correlated with the Th1-type response. In contrast, BALB/c mice only expressed minimal IL-12R beta 2 in draining LNC and had lower Th1-response. B10.A mice produced more IFN-gamma and generated a higher number of Th1 cells than that of BALB/c in the draining LNC. Furthermore, IL-12R beta 2 expression and STAT4 signaling were inhibited by anti-IFN-gamma mAb in the cultured draining LNC from B10.A mice, but enhanced by adding exogenous IFN-gamma in the cultured cells from BALB/c mice. The IL-12R beta 2 expression on Th1 cells from draining LNC was increased in the presence of IL-12 and IFN-gamma. In conclusion, IFN-gamma production correlated with IL-12R beta 2 expression on Th1-cells and IFN-gamma had a potential to regulate IL-12R beta 2 expression in vivo. This regulatory mechanism might be involved in EAU induction.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Interferon-gamma/pharmacology , Receptors, Interleukin/biosynthesis , Up-Regulation/physiology , Uveitis/immunology , Uveitis/metabolism , Animals , Cattle , Cells, Cultured , DNA-Binding Proteins/physiology , Encephalomyelitis, Autoimmune, Experimental/genetics , Female , Genetic Predisposition to Disease , Interferon-gamma/physiology , Mice , Mice, Inbred A , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, Interleukin-12 , STAT4 Transcription Factor , Signal Transduction/immunology , Trans-Activators/physiology , Up-Regulation/immunology , Uveitis/genetics
14.
J Immunol ; 170(4): 1728-36, 2003 Feb 15.
Article in English | MEDLINE | ID: mdl-12574336

ABSTRACT

Pertussis toxin (PTX) has been widely used as an adjuvant to induce Th1-mediated organ-specific autoimmune diseases in animal models. However, the cellular and molecular mechanisms remain to be defined. In this study, we showed that dendritic cells (DC) stimulated with PTX (PTX-DC) were able to substitute for PTX to promote experimental autoimmune uveitis (EAU). EAU induced by PTX-DC revealed a typical Th1 response, characterized by high uveitogenic retinal Ag interphotoreceptor retinoid-binding protein (IRBP)-specific IFN-gamma and IL-12 production in the draining lymph nodes, as well as increased levels of anti-IRBP IgG2a and decreased levels of anti-IRBP IgG1 in the serum of IRBP-immunized mice. Furthermore, PTX-DC preferentially induced T cells to produce the Th1 cytokine, IFN-gamma. After being stimulated with PTX, DC exhibited up-regulation of MHC class II, CD80, CD86, CD40, and DEC205. PTX-DC had also increased allostimulatory capacity and IL-12 and TNF-alpha production. Serum IL-12 was increased in naive mice that received PTX-DC i.p. In addition, PTX activated extracellular signal-regulated kinase in DC. Following the inhibition of extracellular signal-regulated kinase signaling, the maturation of PTX-DC was reduced. Subsequently, the ability of PTX-DC to promote IFN-gamma production by T cells in vitro and to induce EAU in vivo was blocked. The results suggest that PTX might exert an adjuvant effect on DC to promote their maturation and the production of proinflammatory cytokines, thereby eliciting a Th1 response.


Subject(s)
Adjuvants, Immunologic/pharmacology , Dendritic Cells/immunology , Pertussis Toxin/pharmacology , Th1 Cells/immunology , Th1 Cells/metabolism , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/physiology , Animals , Cell Differentiation/immunology , Cell Movement/immunology , Cells, Cultured , Coculture Techniques , Dendritic Cells/cytology , Dendritic Cells/enzymology , Dendritic Cells/transplantation , Eye Proteins/administration & dosage , Eye Proteins/immunology , Female , Injections, Intraperitoneal , Injections, Subcutaneous , Interferon-gamma/biosynthesis , Interleukin-12/blood , Interleukin-12/metabolism , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphocyte Culture Test, Mixed , MAP Kinase Signaling System/immunology , Mice , Mice, Inbred A , Mice, Inbred BALB C , Mice, Inbred C57BL , Pertussis Toxin/administration & dosage , Pertussis Toxin/physiology , Retinol-Binding Proteins/administration & dosage , Retinol-Binding Proteins/immunology , Th1 Cells/enzymology , Uveitis/enzymology , Uveitis/etiology , Uveitis/immunology , Uveitis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...