Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 12(8): e1005861, 2016 08.
Article in English | MEDLINE | ID: mdl-27580079

ABSTRACT

Innate immune responses in general, and type I interferons (T1IFNs) in particular, play an important and often essential role during primary viral infections, by directly combatting the virus and by maximizing the primary adaptive immune response. Several studies have suggested that T1IFNs also contribute very substantially to the secondary (recall) response; they are thought (i) to be required to drive the early attrition of memory T cells, (ii) to support the subsequent expansion of surviving virus-specific memory cells, and (iii) to assist in the suppression and clearance of the infectious agent. However, many of these observations were predicated upon models in which T1IFN signaling was interrupted prior to a primary immune response, raising the possibility that the resulting memory cells might be intrinsically abnormal. We have directly addressed this by using an inducible-Cre model system in which the host remains genetically-intact during the primary response to infection, and in which T1IFN signaling can be effectively ablated prior to secondary viral challenge. We report that, in stark contrast to primary infection, T1IFN signaling is not required during the recall response. IFNαßR-deficient memory CD8+ and CD4+ memory T cells undergo attrition and expansion with kinetics that are indistinguishable from those of receptor-sufficient cells. Moreover, even in the absence of functional T1IFN signaling, the host's immune capacity to rapidly suppress, and then to eradicate, a secondary infection remains intact. Thus, this study shows that T1IFN signaling is dispensable during the recall response to a virus infection. Moreover, two broader implications may be drawn. First, a T cell's requirement for a cytokine is highly dependent on the cell's maturation / differentiation status. Consequently, second, these data underscore the importance of evaluating a gene's impact by modulating its expression or function in a temporally-controllable manner.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Interferon Type I/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Signal Transduction/immunology , Animals , Interferon Type I/genetics , Lymphocytic Choriomeningitis/genetics , Mice , Mice, Transgenic , Signal Transduction/genetics
2.
Virology ; 498: 69-81, 2016 11.
Article in English | MEDLINE | ID: mdl-27564543

ABSTRACT

CD8(+) memory T cells produce IFNγ within hours of secondary infection, but this is quickly terminated in vivo despite the presence of stimulatory viral antigen, suggesting that active suppression occurs. Herein, we investigated the in vivo effector function of CD8(+) memory T cells during successive encounters with viral antigen. CD8(+) T cells in immune mice receiving prior viral or peptide challenge failed to reproduce IFNγ during LCMV rechallenge. Surprisingly, this refractory state was induced even in memory cells that had not encountered their cognate antigen, indicating that the silencing of CD8(+) T cell responses is TCR-independent. Direct injection of IFNγ also suppressed the ability of virus-specific memory cells to respond to subsequent viral challenge. We propose the existence of a negative feedback loop whereby IFNγ, produced by memory CD8(+) T cells to combat viral challenge, acts - directly or indirectly - to limit its further production.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cytokines/biosynthesis , Immunomodulation , Interferon-gamma/metabolism , Receptors, Antigen, T-Cell/metabolism , Animals , Granzymes/metabolism , Immunologic Memory , Immunophenotyping , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/metabolism , Lymphocytic choriomeningitis virus/immunology , Mice , Phenotype , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
3.
Front Cell Neurosci ; 8: 165, 2014.
Article in English | MEDLINE | ID: mdl-24987333

ABSTRACT

Intracranial infection of the neurotropic JHM strain of mouse hepatitis virus (JHMV) into the central nervous system (CNS) of susceptible strains of mice results in an acute encephalomyelitis, accompanied by viral replication in glial cells and robust infiltration of virus-specific T cells that contribute to host defense through cytokine secretion and cytolytic activity. Mice surviving the acute stage of disease develop an immune-mediated demyelinating disease, characterized by viral persistence in white matter tracts and a chronic neuroinflammatory response dominated by T cells and macrophages. Chemokines and their corresponding chemokine receptors are dynamically expressed throughout viral infection of the CNS, influencing neuroinflammation by regulating immune cell infltration and glial biology. This review is focused upon the pleiotropic chemokine receptor CXCR2 and its effects upon neutrophils and oligodendrocytes during JHMV infection and a number of other models of CNS inflammation.

4.
J Immunol ; 193(4): 1873-85, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25015828

ABSTRACT

In vitro studies have shown that naive CD8(+) T cells are unable to express most of their effector proteins until after at least one round of cell division has taken place. We have reassessed this issue in vivo and find that naive CD8(+) T cells mount Ag-specific responses within hours of infection, before proliferation has commenced. Newly activated naive Ag-specific CD8(+) T cells produce a rapid pulse of IFN-γ in vivo and begin to accumulate granzyme B and perforin. Later, in vivo cytolytic activity is detectable, coincident with the initiation of cell division. Despite the rapid development of these functional attributes, no antiviral effect was observed early during infection, even when the cells are present in numbers similar to those of virus-specific memory cells. The evolutionary reason for the pulse of IFN-γ synthesis by naive T cells is uncertain, but the lack of antiviral impact suggests that it may be regulatory.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Interferon-gamma/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/transplantation , Cell Differentiation/immunology , Cell Division/immunology , Granzymes/biosynthesis , Immunologic Memory/immunology , Interferon-gamma/biosynthesis , Lymphocyte Activation/immunology , Lymphocytic Choriomeningitis/virology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Perforin/biosynthesis , T-Box Domain Proteins/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis , Virus Replication/immunology
5.
J Immunol ; 191(8): 4211-22, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24026080

ABSTRACT

CD8(+) memory T cells are abundant and are activated in a near-synchronous manner by infection, thereby providing a unique opportunity to evaluate the coordinate functional and phenotypic changes that occur in vivo within hours of viral challenge. Using two disparate virus challenges of mice, we show that splenic CD8(+) memory T cells rapidly produced IFN-γ in vivo; however, within 18-24 h, IFN-γ synthesis was terminated and remained undetectable for ≥ 48 h. A similar on/off response was observed in CD8(+) memory T cells in the peritoneal cavity. Cessation of IFN-γ production in vivo occurred despite the continued presence of immunostimulatory viral Ag, indicating that the initial IFN-γ response had been actively downregulated and that the cells had been rendered refractory to subsequent in vivo Ag contact. Downregulation of IFN-γ synthesis was accompanied by the upregulation of inhibitory receptor expression on the T cells, and ex vivo analyses using synthetic peptides revealed a concurrent hierarchical loss of cytokine responsiveness (IL-2, then TNF, then IFN-γ) taking place during the first 24 h following Ag contact. Thus, within hours of virus challenge, CD8(+) memory T cells display the standard hallmarks of T cell exhaustion, a phenotype that previously was associated only with chronic diseases and that is generally viewed as a gradually developing and pathological change in T cell function. Our data suggest that, instead, the "exhaustion" phenotype is a rapid and normal physiological T cell response.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Costimulatory and Inhibitory T-Cell Receptors/biosynthesis , Costimulatory and Inhibitory T-Cell Receptors/immunology , Down-Regulation , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Poxviridae Infections/immunology , Spleen/cytology , Spleen/immunology , Tumor Necrosis Factors/biosynthesis , Up-Regulation , Vaccinia virus/genetics , Vaccinia virus/immunology
6.
Virology ; 435(1): 110-7, 2013 Jan 05.
Article in English | MEDLINE | ID: mdl-23217621

ABSTRACT

Inoculation with the neurotropic JHM strain of mouse hepatitis virus (MHV) into the central nervous system (CNS) of susceptible strains of mice results in an acute encephalomyelitis in which virus preferentially replicates within glial cells while excluding neurons. Control of viral replication during acute disease is mediated by infiltrating virus-specific T cells via cytokine secretion and cytolytic activity, however sterile immunity is not achieved and virus persists resulting in chronic neuroinflammation associated with demyelination. CXCR2 is a chemokine receptor that upon binding to specific ligands promotes host defense through recruitment of myeloid cells to the CNS as well as protecting oligodendroglia from cytokine-mediated death in response to MHV infection. These findings highlight growing evidence of the diverse and important role of CXCR2 in regulating neuroinflammatory diseases.


Subject(s)
Central Nervous System/virology , Coronavirus Infections/virology , Demyelinating Diseases/virology , Encephalomyelitis/virology , Murine hepatitis virus/physiology , Oligodendroglia/virology , Receptors, Interleukin-8B/immunology , Acute Disease , Animals , Cell Movement , Central Nervous System/immunology , Central Nervous System/pathology , Coronavirus Infections/immunology , Coronavirus Infections/pathology , Cytokines/biosynthesis , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Encephalomyelitis/immunology , Encephalomyelitis/pathology , Mice , Murine hepatitis virus/pathogenicity , Myeloid Cells/immunology , Myeloid Cells/virology , Oligodendroglia/immunology , Oligodendroglia/pathology , Receptors, Interleukin-8B/genetics , T-Lymphocytes/immunology , T-Lymphocytes/virology , Virus Replication
7.
Future Virol ; 7(4): 349-359, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22582084

ABSTRACT

Inoculation of the neurotropic JHM strain of mouse hepatitis virus (JHMV) into the central nervous system (CNS) of susceptible strains of mice results in wide-spread replication within glial cells accompanied by infiltration of virus-specific T lymphocytes that control virus through cytokine secretion and cytolytic activity. Virus persists within white matter tracts of surviving mice resulting in demyelination that is amplified by inflammatory T cells and macrophages. In response to infection, numerous cytokines/chemokines are secreted by resident cells of the CNS and inflammatory leukocytes that participate in both host defense and disease. Among these are the ELR-positive chemokines that are able to signal through CXC chemokine receptors including CXCR2. Early following JHMV infection, ELR-positive chemokines contribute to host defense by attracting CXCR2-expressing cells including polymorphonuclear cells to the CNS that aid in host defense through increasing the permeability the blood-brain-barrier (BBB). During chronic disease, CXCR2 signaling on oligodendroglia protects these cells from apoptosis and restricts the severity of demyelination. This review covers aspects related to host defense and disease in response to JHMV infection and highlights the different roles of CXCR2 signaling in these processes.

8.
Virology ; 429(1): 74-90, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22551767

ABSTRACT

In vitro studies have shown that enteroviruses employ strategies that may impair the ability of DCs to trigger T cell immunity, but it is unclear how these viruses affect DCs in vivo. Here, we evaluate the effects of wild-type (wt) coxsackievirus B3 on DCs in vitro and in a murine model in vivo. Although CVB3 does not productively infect the vast majority of DCs, virus infection profoundly reduces splenic conventional DC numbers and diminishes their capacity to prime naïve CD8(+) T cells in vitro. In contrast to recombinant CVB3, highly pathogenic wt virus infection significantly diminishes the host's capacity to mount T cell responses, which is temporally associated with the loss of CD8α(+) DCs. Our findings demonstrate that enterovirus infection substantially alters the number, heterogeneity, and stimulatory capacity of DCs in vivo, and these dramatic immunomodulatory effects may weaken the host's capacity to mount antiviral T cell responses.


Subject(s)
Coxsackievirus Infections/immunology , Dendritic Cells/immunology , Enterovirus B, Human/physiology , Animals , Coxsackievirus Infections/virology , Dendritic Cells/virology , Enterovirus B, Human/genetics , Enterovirus B, Human/immunology , Humans , Male , Mice , Mice, Inbred C57BL
9.
Cell Host Microbe ; 11(3): 227-39, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22423963

ABSTRACT

Neutrophils are innate immune cells that counter pathogens by many mechanisms, including release of antimicrobial proteins such as calprotectin to inhibit bacterial growth. Calprotectin sequesters essential micronutrient metals such as zinc, thereby limiting their availability to microbes, a process termed nutritional immunity. We find that while calprotectin is induced by neutrophils during infection with the gut pathogen Salmonella Typhimurium, calprotectin-mediated metal sequestration does not inhibit S. Typhimurium proliferation. Remarkably, S. Typhimurium overcomes calprotectin-mediated zinc chelation by expressing a high affinity zinc transporter (ZnuABC). A S. Typhimurium znuA mutant impaired for growth in the inflamed gut was rescued in the absence of calprotectin. ZnuABC was also required to promote the growth of S. Typhimurium over that of competing commensal bacteria. Thus, our findings indicate that Salmonella thrives in the inflamed gut by overcoming the zinc sequestration of calprotectin and highlight the importance of zinc acquisition in bacterial intestinal colonization.


Subject(s)
Leukocyte L1 Antigen Complex/metabolism , Neutrophils/metabolism , Salmonella Infections, Animal/metabolism , Salmonella typhimurium/physiology , Zinc/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cecum/metabolism , Cecum/microbiology , Cecum/pathology , Diarrhea/immunology , Diarrhea/metabolism , Diarrhea/microbiology , Feces/chemistry , Feces/microbiology , Host-Pathogen Interactions , Immunity, Innate , Inflammation/immunology , Inflammation/metabolism , Inflammation/microbiology , Leukocyte L1 Antigen Complex/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Binding , Salmonella Infections, Animal/immunology , Salmonella Infections, Animal/pathology , Salmonella typhimurium/growth & development , Salmonella typhimurium/metabolism , Transcription, Genetic
10.
Blood ; 117(16): 4349-57, 2011 Apr 21.
Article in English | MEDLINE | ID: mdl-21224471

ABSTRACT

Emergency mobilization of neutrophil granulocytes (neutrophils) from the bone marrow (BM) is a key event of early cellular immunity. The hematopoietic cytokine granulocyte-colony stimulating factor (G-CSF) stimulates this process, but it is unknown how individual neutrophils respond in situ. We show by intravital 2-photon microscopy that a systemic dose of human clinical-grade G-CSF rapidly induces the motility and entry of neutrophils into blood vessels within the tibial BM of mice. Simultaneously, the neutrophil-attracting chemokine KC (Cxcl1) spikes in the blood. In mice lacking the KC receptor Cxcr2, G-CSF fails to mobilize neutrophils and antibody blockade of Cxcr2 inhibits the mobilization and induction of neutrophil motility in the BM. KC is expressed by megakaryocytes and endothelial cells in situ and is released in vitro by megakaryocytes isolated directly from BM. This production of KC is strongly increased by thrombopoietin (TPO). Systemic G-CSF rapidly induces the increased production of TPO in BM. Accordingly, a single injection of TPO mobilizes neutrophils with kinetics similar to G-CSF, and mice lacking the TPO receptor show impaired neutrophil mobilization after short-term G-CSF administration. Thus, a network of signaling molecules, chemokines, and cells controls neutrophil release from the BM, and their mobilization involves rapidly induced Cxcr2-mediated motility controlled by TPO as a pacemaker.


Subject(s)
Bone Marrow Cells/cytology , Granulocyte Colony-Stimulating Factor/immunology , Neutrophils/cytology , Receptors, Interleukin-8B/immunology , Thrombopoietin/immunology , Animals , Bone Marrow/immunology , Bone Marrow Cells/immunology , Bone and Bones/cytology , Cell Line , Cell Movement , Cells, Cultured , Humans , Megakaryocytes/cytology , Megakaryocytes/immunology , Mice , Mice, Inbred C57BL , Neutrophils/immunology
12.
PLoS One ; 5(6): e11340, 2010 Jun 28.
Article in English | MEDLINE | ID: mdl-20596532

ABSTRACT

BACKGROUND: The functional role of ELR-positive CXC chemokines during viral-induced demyelination was assessed. Inoculation of the neuroattenuated JHM strain of mouse hepatitis virus (JHMV) into the CNS of susceptible mice results in an acute encephalomyelitis that evolves into a chronic demyelinating disease, modeling white matter pathology observed in the human demyelinating disease Multiple Sclerosis. METHODOLOGY/PRINCIPAL FINDINGS: JHMV infection induced the rapid and sustained expression of transcripts specific for the ELR+ chemokine ligands CXCL1 and CXCL2, as well as their binding receptor CXCR2, which was enriched within the spinal cord during chronic infection. Inhibiting CXCR2 signaling with neutralizing antiserum significantly (p<0.03) delayed clinical recovery. Moreover, CXCR2 neutralization was associated with an increase in the severity of demyelination that was independent of viral recrudescence or modulation of neuroinflammation. Rather, blocking CXCR2 was associated with increased numbers of apoptotic cells primarily within white matter tracts, suggesting that oligodendrocytes were affected. JHMV infection of enriched oligodendrocyte progenitor cell (OPC) cultures revealed that apoptosis was associated with elevated expression of cleaved caspase 3 and muted Bcl-2 expression. Inclusion of CXCL1 within JHMV infected cultures restricted caspase 3 cleavage and increased Bcl-2 expression that was associated with a significant (p<0.001) decrease in apoptosis. CXCR2 deficient oligodendrocytes were refractory to CXCL1 mediated protection from JHMV-induced apoptosis, readily activating caspase 3 and down regulating Bcl-2. CONCLUSION/SIGNIFICANCE: These findings highlight a previously unappreciated role for CXCR2 signaling in protecting oligodendrocyte lineage cells from apoptosis during inflammatory demyelination initiated by viral infection of the CNS.


Subject(s)
Disease Models, Animal , Multiple Sclerosis/pathology , Murine hepatitis virus/physiology , Oligodendroglia/metabolism , Receptors, Interleukin-8B/metabolism , Signal Transduction , Animals , Apoptosis , Chemokine CXCL1/metabolism , Chemokine CXCL2/metabolism , Ligands , Mice , Mice, Inbred C57BL , Multiple Sclerosis/metabolism , Multiple Sclerosis/virology , Oligodendroglia/pathology , Polymerase Chain Reaction , Up-Regulation
13.
Crit Rev Immunol ; 30(2): 119-30, 2010.
Article in English | MEDLINE | ID: mdl-20370625

ABSTRACT

Mouse hepatitis virus (MHV) is a positive-strand RNA virus that causes an acute encephalomyelitis that later resolves into a chronic fulminating demyelinating disease. Cytokine production, chemokine secretion, and immune cell infiltration into the central nervous system are critical to control viral replication during acute infection. Despite potent antiviral T-lymphocyte activity, sterile immunity is not achieved, and MHV chronically persists within oligodendrocytes. Continued infiltration and activation of the immune system, a result of the lingering viral antigen and RNA within oligodendrocytes, lead directly to the development of an immune-mediated demyelination that bears remarkable similarities, both clinically and histologically, to the human demyelinating disease multiple sclerosis. MHV offers a unique model system for studying host defense during acute viral infection and immune-mediated demyelination during chronic infection.


Subject(s)
Central Nervous System Viral Diseases/immunology , Central Nervous System Viral Diseases/virology , Coronavirus Infections/immunology , Coronavirus Infections/virology , Murine hepatitis virus/immunology , Murine hepatitis virus/pathogenicity , Animals , Central Nervous System Viral Diseases/pathology , Host-Pathogen Interactions , Humans , Multiple Sclerosis/pathology
14.
Curr Immunol Rev ; 5(4): 267-276, 2009 May 04.
Article in English | MEDLINE | ID: mdl-19946572

ABSTRACT

Multiple Sclerosis (MS) is an immune-mediated demyelinating disease of humans. Although causes of MS are enigmatic, underlying elements contributing to disease development include both genetic and environmental factors. Recent epidemiological evidence has pointed to viral infection as a trigger to initiating white matter damage in humans. Mouse hepatitis virus (MHV) is a positive strand RNA virus that, following intracranial infection of susceptible mice, induces an acute encephalomyelitis that later resolves into a chronic fulminating demyelinating disease. Immune cell infiltration into the central nervous system is critical both to quell viral replication and instigate demyelination. Recent efforts by our laboratory and others have focused upon strategies capable of enhancing remyelination in response to viral-induced demyelination, both by dampening chronic inflammation and by surgical engraftment of remyelination - competent neural precursor cells.

15.
PLoS Pathog ; 5(11): e1000648, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19893623

ABSTRACT

The functional role of ELR-positive CXC chemokines in host defense during acute viral-induced encephalomyelitis was determined. Inoculation of the neurotropic JHM strain of mouse hepatitis virus (JHMV) into the central nervous system (CNS) of mice resulted in the rapid mobilization of PMNs expressing the chemokine receptor CXCR2 into the blood. Migration of PMNs to the CNS coincided with increased expression of transcripts specific for the CXCR2 ELR-positive chemokine ligands CXCL1, CXCL2, and CXCL5 within the brain. Treatment of JHMV-infected mice with anti-CXCR2 blocking antibody reduced PMN trafficking into the CNS by >95%, dampened MMP-9 activity, and abrogated blood-brain-barrier (BBB) breakdown. Correspondingly, CXCR2 neutralization resulted in diminished infiltration of virus-specific T cells, an inability to control viral replication within the brain, and 100% mortality. Blocking CXCR2 signaling did not impair the generation of virus-specific T cells, indicating that CXCR2 is not required to tailor anti-JHMV T cell responses. Evaluation of mice in which CXCR2 is genetically silenced (CXCR2-/- mice) confirmed that PMNs neither expressed CXCR2 nor migrated in response to ligands CXCL1, CXCL2, or CXCL5 in an in vitro chemotaxis assay. Moreover, JHMV infection of CXCR2-/- mice resulted in an approximate 60% reduction of PMN migration into the CNS, yet these mice survived infection and controlled viral replication within the brain. Treatment of JHMV-infected CXCR2-/- mice with anti-CXCR2 antibody did not modulate PMN migration nor alter viral clearance or mortality, indicating the existence of compensatory mechanisms that facilitate sufficient migration of PMNs into the CNS in the absence of CXCR2. Collectively, these findings highlight a previously unappreciated role for ELR-positive chemokines in enhancing host defense during acute viral infections of the CNS.


Subject(s)
Chemokines, CXC/immunology , Encephalomyelitis/immunology , Encephalomyelitis/virology , Receptors, Interleukin-8B/immunology , Acute Disease , Animals , Brain Chemistry , Chemokine CXCL1 , Chemokine CXCL2 , Chemokine CXCL5 , Chemokines, CXC/biosynthesis , Mice , Mice, Knockout , Murine hepatitis virus , Neutrophil Infiltration/immunology , Receptors, Interleukin-8B/deficiency , T-Lymphocytes/immunology , Virus Replication
16.
Eur J Immunol ; 36(3): 613-22, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16479546

ABSTRACT

Lymphocyte infiltration into the central nervous system (CNS) following viral infection represents an important component of host defense and is required for control of viral replication. However, the mechanisms governing inflammation in response to viral infection of the CNS are not well understood. Following intracranial (i.c.) infection of susceptible mice with mouse hepatitis virus (MHV), mice develop an acute encephalomyelitis followed by a chronic demyelinating disease. The CXC chemokine ligand 10 (CXCL10) is expressed following MHV infection and signals T cells to migrate into the CNS. The functional contribution of the CXCL10 receptor CXCR3 in host defense and disease in response to MHV infection was evaluated. The majority of CD4+ and CD8+ T cells infiltrating the CNS following MHV infection express CXCR3. Administration of anti-CXCR3 antibody reduced CD4+ T cell infiltration (p

Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Movement/immunology , Central Nervous System Viral Diseases/immunology , Encephalomyelitis/immunology , Murine hepatitis virus/immunology , Acute Disease , Animals , Antibodies, Monoclonal/administration & dosage , Cell Movement/drug effects , Cell Proliferation/drug effects , Central Nervous System Viral Diseases/drug therapy , Central Nervous System Viral Diseases/virology , Chemokine CXCL10 , Chemokines, CXC/immunology , Chronic Disease , Demyelinating Diseases/drug therapy , Demyelinating Diseases/immunology , Demyelinating Diseases/virology , Encephalomyelitis/drug therapy , Encephalomyelitis/virology , Inflammation/immunology , Inflammation/virology , Mice , Mice, Inbred BALB C , Receptors, CXCR3 , Receptors, Chemokine/immunology
SELECTION OF CITATIONS
SEARCH DETAIL