Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Virol ; 88(24): 14090-104, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25253351

ABSTRACT

UNLABELLED: Suppressors of cytokine signaling (SOCS) proteins are intracellular proteins that inhibit cytokine signaling in a variety of cell types. A number of viral infections have been associated with SOCS upregulation; however, not much is known about the mechanisms regulating SOCS expression during viral infection. In this study, we used two pathologically distinct intracerebral (i.c.) infection models to characterize temporal and spatial aspects of SOCS expression in the virus-infected central nervous system (CNS), and by employing various knockout mouse models, we sought to identify regulatory mechanisms that may underlie a virus induced upregulation of SOCS in the CNS. We found that i.c. infection with either lymphocytic choriomeningitis virus (LCMV) or yellow fever virus (YF) results in gradual upregulation of SOCS1/3 mRNA expression peaking at day 7 postinfection (p.i.). In the LCMV model, SOCS mRNA was expressed in brain resident cells, including astrocytes and some neurons, and for SOCS1 in particular this upregulation was almost entirely mediated by gamma interferon (IFN-γ) produced by infiltrating T cells. After infection with YF, we also found SOCS expression to be upregulated in brain resident cells with a peak on day 7 p.i., but in this model, the upregulation was only partially dependent on IFN-γ and T cells, indicating that at least one other mediator was involved in the upregulation of SOCS following YF infection. We conclude that virus-induced inflammation of the CNS is associated with upregulation of SOCS1/3 mRNA expression in brain resident cells and that at least two distinctive pathways can lead to this upregulation. IMPORTANCE: In the present report, we have studied the induction of SOCS1 and SOCS3 expression in the context of virus-induced CNS infection. We found that both a noncytolytic and a cytolytic virus induce marked upregulation of SOCS1 and -3 expression. Notably, the kinetics of the observed upregulation follows that of activity within proinflammatory signaling pathways and, interestingly, type II interferon (IFN), which is also a key inducer of inflammatory mediators, seems to be essential in initiating this counterinflammatory response. Another key observation is that not only cells of the immune system but also CNS resident cells are actively involved in both the pro- and the counterinflammatory immune circuits; thus, for example, astrocytes upregulate both C-X-C-motif chemokine 10 (CXCL10) and SOCS when exposed to type II IFN in vivo.


Subject(s)
Arenaviridae Infections/pathology , Encephalitis, Viral/pathology , Flavivirus Infections/pathology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Suppressor of Cytokine Signaling Proteins/biosynthesis , Animals , Arenaviridae Infections/immunology , Arenaviridae Infections/virology , Encephalitis, Viral/immunology , Encephalitis, Viral/virology , Flavivirus Infections/immunology , Flavivirus Infections/virology , Gene Expression Profiling , Interferon-gamma/immunology , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/analysis , T-Lymphocytes/immunology , Time Factors , Up-Regulation
2.
J Virol ; 86(13): 7384-92, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22514347

ABSTRACT

Interferon (IFN) regulatory factors (IRFs) are a family of transcription factors involved in regulating type I IFN genes and other genes participating in the early antiviral host response. To better understand the mechanisms involved in virus-induced central nervous system (CNS) inflammation, we studied the influence of IRF1, -3, -7, and -9 on the transcriptional activity of key genes encoding antiviral host factors in the CNS of mice infected with lymphocytic choriomeningitis virus (LCMV). A key finding is that neither IRF3 nor IRF7 is absolutely required for induction of a type I IFN response in the LCMV-infected CNS, whereas concurrent elimination of both factors markedly reduces the virus-induced host response. This is unlike the situation in the periphery, where deficiency of IRF7 almost eliminates the LCMV-induced production of the type I IFNs. This difference is seemingly related to the local environment, as peripheral production of type I IFNs is severely reduced in intracerebrally (i.c.) infected IRF7-deficient mice, which undergo a combined infection of the CNS and peripheral organs, such as spleen and lymph nodes. Interestingly, despite the redundancy of IRF7 in initiating the type I IFN response in the CNS, the response is not abolished in IFN-ß-deficient mice, as might have been expected. Collectively, these data demonstrate that the early type I IFN response to LCMV infection in the CNS is controlled by a concerted action of IRF3 and -7. Consequently this work provides strong evidence for differential regulation of the type I IFN response in the CNS versus the periphery during viral infection.


Subject(s)
Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-7/immunology , Interferon Type I/biosynthesis , Lymphocytic choriomeningitis virus/immunology , Nervous System/immunology , Nervous System/virology , Animals , Female , Interferon Regulatory Factor-3/deficiency , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-7/deficiency , Interferon Regulatory Factor-7/metabolism , Male , Mice , Mice, Inbred C57BL
3.
APMIS ; 117(5-6): 338-55, 2009 May.
Article in English | MEDLINE | ID: mdl-19400861

ABSTRACT

The host counters a viral infection through a complex response made up of components belonging to both the innate and the adaptive immune system. In this report, we review the mechanisms underlying this response, how it is induced and how it is co-ordinated. As cell-cell communication represents the very essence of immune system physiology, a key to a rapid, efficient and optimally regulated immune response is the ability of the involved cells to rapidly shift between a stationary and a mobile state, combined with stringent regulation of cell migration during the mobile state. Through the co-ordinated recruitment of different cell types intended to work in concert, cellular co-operation is optimized particularly under conditions that may involve rare cells. Consequently, a major focus is placed on presenting an overview of the co-operative events and the associated cell migration, which is essential in mounting an efficient host response and co-ordinating innate and adaptive immunity during a primary viral infection.


Subject(s)
Host-Pathogen Interactions/immunology , Immunity, Innate , Leukocytes/immunology , Virus Diseases/immunology , Animals , Antibodies, Viral/biosynthesis , Antibodies, Viral/immunology , Cell Communication , Chemotaxis, Leukocyte , Cytokines/physiology , Dendritic Cells/cytology , Dendritic Cells/immunology , Humans , Immunity, Cellular , Killer Cells, Natural/immunology , Leukocytes/cytology , Lymphocyte Subsets/cytology , Lymphocyte Subsets/immunology , Macrophages/immunology , Monocytes/immunology , Receptors, Cytokine/physiology , Receptors, Pattern Recognition/physiology
4.
J Immunol ; 176(7): 4235-43, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16547260

ABSTRACT

IFN-gamma-inducible protein 10/CXCL10 is a chemokine associated with type 1 T cell responses, regulating the migration of activated T cells through binding to the CXCR3 receptor. Expression of both CXCL10 and CXCR3 are observed during immunopathological diseases of the CNS, and this receptor/ligand pair is thought to play a central role in regulating T cell-mediated inflammation in this organ site. In this report, we investigated the role of CXCL10 in regulating CD8(+) T cell-mediated inflammation in the virus-infected brain. This was done through analysis of CXCL10-deficient mice infected intracerebrally with lymphocytic choriomeningitis virus, which in normal immunocompetent mice induces a fatal CD8(+) T cell-mediated meningoencephalitis. We found that a normal antiviral CD8(+) T cell response was generated in CXCL10-deficient mice, and that lack of CXCL10 had no influence on the accumulation of mononuclear cells in the cerebrospinal fluid. However, analysis of the susceptibility of CXCL10-deficient mice to lymphocytic choriomeningitis virus-induced meningitis revealed that these mice just like CXCR3-deficient mice were partially resistant to this disease, whereas wild-type mice invariably died. Furthermore, despite marked up-regulation of the two remaining CXCR3 ligands: CXCL9 and 11, we found a reduced accumulation of CD8(+) T cells in the brain parenchyma around the time point when wild-type mice succumb as a result of CD8(+) T cell-mediated inflammation. Thus, taken together these results indicate a central role for CXCL10 in regulating the accumulation of effector T cells at sites of CNS inflammation, with no apparent compensatory effect of other CXCR3 ligands.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chemokines, CXC/metabolism , Immunologic Surveillance/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Meningitis, Viral/immunology , Receptors, Chemokine/metabolism , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Chemokine CXCL10 , Chemokine CXCL11 , Chemokine CXCL9 , Chemokines, CXC/deficiency , Chemokines, CXC/genetics , Chemokines, CXC/immunology , Gene Expression Regulation, Viral , Kinetics , Ligands , Lymphocytic Choriomeningitis/metabolism , Lymphocytic Choriomeningitis/pathology , Lymphocytic Choriomeningitis/virology , Meningitis, Viral/metabolism , Meningitis, Viral/pathology , Meningitis, Viral/virology , Mice , Mice, Knockout , Receptors, CXCR3 , Receptors, Chemokine/immunology
5.
J Immunol ; 175(3): 1767-75, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-16034118

ABSTRACT

T cells play a key role in the control of viral infection in the CNS but may also contribute to immune-mediated cell damage. To study the redundancy of the chemokine receptors CXCR3 and CCR5 in regulating virus-induced CD8+ T cell-mediated inflammation in the brain, CXCR3/CCR5 double-deficient mice were generated and infected intracerebrally with noncytolytic lymphocytic choriomeningitis virus. Because these chemokine receptors are mostly expressed by overlapping subsets of activated CD8+ T cells, it was expected that absence of both receptors would synergistically impair effector T cell invasion and therefore protect mice against the otherwise fatal CD8+ T cell-mediated immune attack. Contrary to expectations, the accumulation of mononuclear cells in cerebrospinal fluid was only slightly delayed compared with mice with normal expression of both receptors. Even more surprising, CXCR3/CCR5 double-deficient mice were more susceptible to intracerebral infection than CXCR3-deficient mice. Analysis of effector T cell generation revealed an accelerated antiviral CD8+ T cell response in CXCR3/CCR5 double-deficient mice. Furthermore, while the accumulation of CD8+ T cells in the neural parenchyma was significantly delayed in both CXCR3- and CXCR3/CCR5-deficient mice, more CD8+ T cells were found in the parenchyma of double-deficient mice when these were analyzed around the time when the difference in clinical outcome becomes manifest. Taken together, these results indicate that while CXCR3 plays an important role in controlling CNS inflammation, other receptors but not CCR5 also contribute significantly. Additionally, our results suggest that CCR5 primarily functions as a negative regulator of the antiviral CD8+ T cell response.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lymphocytic Choriomeningitis/cerebrospinal fluid , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Receptors, CCR5/deficiency , Receptors, CCR5/genetics , Receptors, Chemokine/deficiency , Receptors, Chemokine/genetics , Animals , Brain Chemistry/genetics , Brain Chemistry/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cell Aggregation/genetics , Cell Aggregation/immunology , Chemotaxis, Leukocyte/genetics , Genetic Predisposition to Disease , Injections, Intraventricular , Lymphocyte Activation/genetics , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/pathology , Lymphocytic choriomeningitis virus/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neurons/immunology , Neurons/metabolism , Neurons/pathology , RNA, Messenger/biosynthesis , Receptors, CCR5/biosynthesis , Receptors, CXCR3 , Receptors, Chemokine/biosynthesis , Virus Activation/immunology
6.
J Neurosci ; 24(20): 4849-58, 2004 May 19.
Article in English | MEDLINE | ID: mdl-15152045

ABSTRACT

T-cells play an important role in controlling viral infections inside the CNS. To study the role of the chemokine receptor CXCR3 in the migration and positioning of virus-specific effector T-cells within the brain, CXCR3-deficient mice were infected intracerebrally with lymphocytic choriomeningitis virus (LCMV). Analysis of the induction phase of the antiviral CD8+ T-cell response did not reveal any immune defects in CXCR3-deficient mice. Yet, when mice were challenged with LCMV intracerebrally, most CXCR3-deficient mice survived the infection, whereas wild-type mice invariably died from CD8+ T-cell-mediated immunopathology. Quantitative analysis of the cellular infiltrate in CSF of infected mice revealed modest, if any, decrease in the number of mononuclear cells recruited to the meninges in the absence of CXCR3. However, immunohistological analysis disclosed a striking impairment of CD8+ T-cells from CXCR3-deficient mice to migrate from the meninges into the outer layers of the brain parenchyma despite similar localization of virus-infected target cells. Reconstitution of CXCR3-deficient mice with wild-type CD8+ T-cells completely restored susceptibility to LCMV-induced meningitis. Thus, taken together, our results strongly point to a critical role for CXCR3 in the positioning of effector T-cells at sites of viral inflammation in the brain.


Subject(s)
Central Nervous System/immunology , Central Nervous System/metabolism , Immunologic Surveillance/immunology , Lymphocytic Choriomeningitis/immunology , Receptors, Chemokine/biosynthesis , T-Lymphocytes/immunology , Animals , Biomarkers/analysis , Brain/immunology , Brain/metabolism , Brain/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/transplantation , Cell Count , Cell Movement/genetics , Cell Movement/immunology , Central Nervous System/pathology , Flow Cytometry , Genetic Predisposition to Disease , Hyaluronan Receptors/biosynthesis , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/pathology , Lymphocytic Choriomeningitis/pathology , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/pathogenicity , Meninges/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , RNA, Messenger/biosynthesis , Receptors, CXCR3 , Receptors, Chemokine/deficiency , Receptors, Chemokine/genetics , Survival Rate , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation
7.
Blood ; 99(4): 1237-45, 2002 Feb 15.
Article in English | MEDLINE | ID: mdl-11830471

ABSTRACT

The CC chemokine receptor CCR5 is an important coreceptor for human immunodeficiency virus (HIV), and there is a major thrust to develop anti-CCR5-based therapies for HIV-1. However, it is not known whether CCR5 is critical for a normal antiviral T-cell response. This study investigated the immune response to lymphocytic choriomeningitis virus in mice lacking CCR5 (CCR5(-/-) mice). This infection is a classical model for studying antiviral immunity, and influx of CCR5-expressing CD8(+) T cells and macrophages is essential for both virus control and associated immunopathology. Results showed that the virus-induced clonal expansion of antigen-specific T cells was augmented in CCR5(-/-) mice especially with regard to the CD4(+) subset. Despite absence of CCR5, intracerebral infection invariably resulted in lethal T cell-mediated meningitis, and quantitative and qualitative analysis of the inflammatory exudate cells did not reveal any significant differences between gene-targeted mice and wild-type controls. CCR5 was also found to be redundant regarding the ability to eliminate virus from internal organs. Using delayed-type hypersensitivity to evaluate CD8(+) T cell-mediated inflammation, no significant influence of CCR5 was found, not even when viral peptide was used as local trigger instead of live virus. Finally, long-term CD8(+) T cell-mediated immune surveillance was efficiently sustained in CCR5(-/-) mice. Taken together, these results indicate that expression of CCR5 is not critical for T cell-mediated antiviral immunity, and this molecule may therefore constitute a logic and safe target for anti-HIV therapies.


Subject(s)
Receptors, CCR5/immunology , Virus Diseases/immunology , Animals , Antigens, Viral/immunology , Chemotaxis, Leukocyte/immunology , Disease Models, Animal , Immunity , Lymphocyte Activation/immunology , Lymphocytic Choriomeningitis/immunology , Meningitis, Viral/etiology , Meningitis, Viral/immunology , Meningitis, Viral/pathology , Mice , Mice, Knockout , Receptors, CCR5/genetics , Receptors, CCR5/physiology , T-Lymphocyte Subsets/immunology
SELECTION OF CITATIONS
SEARCH DETAIL