Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Rev Med Virol ; 31(1): 1-10, 2021 01.
Article in English | MEDLINE | ID: mdl-32845042

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic is a rapidly evolving global emergency that continues to strain healthcare systems. Emerging research describes a plethora of patient factors-including demographic, clinical, immunologic, hematological, biochemical, and radiographic findings-that may be of utility to clinicians to predict COVID-19 severity and mortality. We present a synthesis of the current literature pertaining to factors predictive of COVID-19 clinical course and outcomes. Findings associated with increased disease severity and/or mortality include age > 55 years, multiple pre-existing comorbidities, hypoxia, specific computed tomography findings indicative of extensive lung involvement, diverse laboratory test abnormalities, and biomarkers of end-organ dysfunction. Hypothesis-driven research is critical to identify the key evidence-based prognostic factors that will inform the design of intervention studies to improve the outcomes of patients with COVID-19 and to appropriately allocate scarce resources.


Subject(s)
COVID-19 , Severity of Illness Index , Adult , Aging , Biomarkers , COVID-19/mortality , COVID-19/pathology , COVID-19/transmission , Child , Comorbidity , Humans , Hypoxia/pathology , Prognosis , SARS-CoV-2/pathogenicity
3.
PLoS One ; 7(8): e42850, 2012.
Article in English | MEDLINE | ID: mdl-22880122

ABSTRACT

Various cell types in both lymphoid and non-lymphoid tissues produce the anti-inflammatory cytokine interleukin (IL)-10 during murine cytomegalovirus (MCMV) infection. The functions of IL-10 in the liver during acute infection and the cells that generate this cytokine at this site have not been extensively investigated. In this study, we demonstrate that the production of IL-10 in the liver is elevated in C57BL/6 mice during late acute MCMV infection. Using IL-10 green fluorescence protein (GFP) reporter knock-in mice, designated IL-10-internal ribosomal entry site (IRES)-GFP-enhanced reporter (tiger), NK cells are identified as major IL-10 expressing cells in the liver after infection, along with T cells and other leukocytes. In the absence of IL-10, mice exhibit marked elevations in proinflammatory cytokines and in the numbers of mononuclear cells and lymphocytes infiltrating the liver during this infection. IL-10-deficiency also enhances liver injury without improving viral clearance from this site. Collectively, the results indicate that IL-10-producing cells in the liver provide protection from collateral injury by modulating the inflammatory response associated with MCMV infection.


Subject(s)
Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/pathology , Inflammation/pathology , Interleukin-10/metabolism , Liver/immunology , Liver/pathology , Muromegalovirus/physiology , Acute Disease , Alanine Transaminase/blood , Animals , CD8-Positive T-Lymphocytes , Chemokines/biosynthesis , Cytomegalovirus Infections/blood , Cytomegalovirus Infections/virology , Inflammation/blood , Inflammation/complications , Inflammation/microbiology , Inflammation Mediators/metabolism , Interferon-gamma/metabolism , Interleukin-10/biosynthesis , Interleukin-10/deficiency , Leukocytes/metabolism , Liver/virology , Mice , Mice, Inbred C57BL , Tumor Necrosis Factor-alpha/metabolism , Viral Load
4.
PLoS One ; 7(6): e39161, 2012.
Article in English | MEDLINE | ID: mdl-22723955

ABSTRACT

Antiviral defense in the liver during acute infection with the hepatotropic virus murine cytomegalovirus (MCMV) involves complex cytokine and cellular interactions. However, the mechanism of viral sensing in the liver that promotes these cytokine and cellular responses has remained unclear. Studies here were undertaken to investigate the role of nucleic acid-sensing Toll-like receptors (TLRs) in initiating antiviral immunity in the liver during infection with MCMV. We examined the host response of UNC93B1 mutant mice, which do not signal properly through TLR3, TLR7 and TLR9, to acute MCMV infection to determine whether liver antiviral defense depends on signaling through these molecules. Infection of UNC93B1 mutant mice revealed reduced production of systemic and liver proinflammatory cytokines including IFN-α, IFN-γ, IL-12 and TNF-α when compared to wild-type. UNC93B1 deficiency also contributed to a transient hepatitis later in acute infection, evidenced by augmented liver pathology and elevated systemic alanine aminotransferase levels. Moreover, viral clearance was impaired in UNC93B1 mutant mice, despite intact virus-specific CD8+ T cell responses in the liver. Altogether, these results suggest a combined role for nucleic acid-sensing TLRs in promoting early liver antiviral defense during MCMV infection.


Subject(s)
Herpesviridae Infections/immunology , Herpesviridae Infections/metabolism , Immunity, Innate , Inflammation/immunology , Liver/immunology , Membrane Transport Proteins/metabolism , Muromegalovirus/immunology , Animals , Cytokines/biosynthesis , Cytokines/immunology , Herpesviridae Infections/genetics , Herpesviridae Infections/virology , Liver/virology , Membrane Transport Proteins/genetics , Mice , Mice, Inbred C57BL , Mutation , T-Lymphocytes/immunology , Toll-Like Receptors/metabolism
5.
J Immunol ; 183(4): 2810-7, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19620305

ABSTRACT

Monocytes/macrophages are critical early innate immune responders during murine CMV (MCMV) infection. It has been established that inflammatory monocyte/macrophages are released from the bone marrow and into the peripheral blood before entry into infected tissue sites. We previously reported a role for IFN-alpha/beta in promotion of CCR2-mediated recruitment of monocyte/macrophages into the liver in response to MCMV infection. However, the mechanisms that support the migration of monocyte/macrophages from the bone marrow and into the peripheral blood under conditions of MCMV infection have not been elucidated. Herein, we demonstrate an accumulation of monocyte/macrophages in the bone marrow of MCMV-infected CCR2-deficient mice, whereas circulating monocyte/macrophages are profoundly diminished. The CCR2 ligands MCP-1, MCP-3, and MCP-5 are detected in bone marrow and in serum from MCMV-infected mice. Furthermore, bone marrow leukocytes from naive mice produce high levels of MCP-1 and MCP-5, and moderate levels of MCP-3, when stimulated with recombinant IFN-alpha in culture. We identify bone marrow F4/80(+) cells as major producers of MCP-1, MCP-3, and MCP-5. Moreover, induction of CCR2 ligands is dependent on IFN-alpha/beta-mediated signals and MCMV infection. Taken together, the results reveal a critical role for inflammatory cytokines in stimulating production of CCR2-binding chemokines from F4/80(+) cells in the bone marrow, and they suggest that local production of chemokines supports monocyte/macrophage egress from the bone marrow into the blood during a virus infection.


Subject(s)
Bone Marrow Cells/immunology , Herpesviridae Infections/immunology , Inflammation Mediators/physiology , Interferon-alpha/physiology , Interferon-beta/physiology , Muromegalovirus/immunology , Receptors, CCR2/biosynthesis , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cells, Cultured , Chemokine CCL2/biosynthesis , Chemokine CCL2/metabolism , Chemotaxis, Leukocyte/immunology , Herpesviridae Infections/metabolism , Herpesviridae Infections/pathology , Ligands , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Monocytes/metabolism , Monocytes/pathology , Receptors, CCR2/deficiency , Receptors, CCR2/metabolism , Recombinant Proteins/pharmacology , Signal Transduction/immunology
6.
J Immunol ; 179(9): 6176-83, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17947693

ABSTRACT

Chemokine responses critical for inflammation and promotion of effective innate control of murine CMV (MCMV) in liver have been shown to be dependent on immunoregulatory functions elicited by IFN-alphabeta. However, it remains to be determined whether upstream factors that promote viral sensing resulting in the rapid secretion of IFN-alphabeta in liver differ from those described in other tissues. Because plasmacytoid dendritic cells (pDCs) are known producers of high levels of systemic IFN-alpha in response to MCMV, this study examines the in vivo contribution of pDCs to IFN-alpha production in the liver, and whether production of the cytokine and ensuing inflammatory events are dependent on TLR9, MyD88, or both. We demonstrate that whereas MyD88 deficiency markedly impaired secretion of IFN-alpha, production of the cytokine was largely independent of TLR9 signaling, in the liver. MyD88 and TLR9 were needed for IFN-alpha production in the spleen. Moreover, hepatic but not splenic pDCs produced significant amounts of intracellular IFN-alpha in the absence of TLR9 function during infection. Furthermore, production of CCL2, CCL3, and IFN-gamma, as well as the accumulation of macrophages and NK cells, was not affected in the absence of functional TLR9 in the liver. In contrast, these responses were dramatically reduced in MyD88(-/-) mice. Additionally, MyD88(-/-) but not TLR9(-/-) mice exhibited increased sensitivity to virus infection in liver. Collectively, our results define contrasting compartmental functions for TLR9 and MyD88, and suggest that the infected tissue site uniquely contributes to the process of virus sensing and regulation of localized antiviral responses.


Subject(s)
Cytomegalovirus Infections/metabolism , Cytomegalovirus Infections/virology , Liver Diseases/metabolism , Liver Diseases/virology , Muromegalovirus/physiology , Myeloid Differentiation Factor 88/metabolism , Toll-Like Receptor 9/metabolism , Animals , Cytomegalovirus Infections/genetics , Dendritic Cells/metabolism , Inflammation/metabolism , Interferon-alpha/biosynthesis , Interferon-beta/biosynthesis , Liver Diseases/pathology , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Spleen/metabolism , Toll-Like Receptor 9/deficiency , Toll-Like Receptor 9/genetics
7.
Am J Pathol ; 171(1): 87-96, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17591956

ABSTRACT

Although studies blocking the Fas pathway indicate it can decrease organ damage while improving septic (cecal ligation and puncture, CLP) mouse survival, little is known about how Fas-Fas ligand (FasL) interactions mediate this protection at the tissue level. Here, we report that although Fas expression on splenocytes and hepatocytes is up-regulated by CLP and is inhibited by in vivo short interfering RNA, FasL as well as the frequency of CD8(+) T cells are differentially altered by sepsis in the spleen (no change in FasL, decreased percentage of CD8(+) and CD4(+) T cells) versus the liver (increased FasL expression on CD8(+) T cells and increase in percentage/number). Adoptive transfer of CLP FasL(+/+) versus FasL(-/-) mouse liver CD8(+) T cells to severe combined immunodeficient or RAG1(-/-) recipient mice indicated that these cells could induce inflammation. The FasL-mediated cytotoxic capacity of these septic mouse liver CD8(+) T cells was shown by their ability to damage directly cultured hepatocytes. Finally, although CD8(-/-) mice exhibited a reduction in both CLP-induced liver active caspase-3 staining and blood interleukin-6 levels, only FasL(-/-) (but not CD8(-/-)) protected the septic mouse spleen from increasing apoptosis. Thus, although truncating Fas-FasL signaling ameliorates many untoward effects of sepsis, the pathological mode of action is distinct at the tissue level.


Subject(s)
Apoptosis , CD8-Positive T-Lymphocytes/physiology , Fas Ligand Protein/physiology , Hepatitis/metabolism , Sepsis/immunology , fas Receptor/physiology , Adoptive Transfer , Animals , Hepatitis/immunology , Inflammation/immunology , Liver/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, SCID , Sepsis/pathology , Signal Transduction , Spleen/immunology , Spleen/metabolism
8.
J Virol ; 81(3): 1241-50, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17108043

ABSTRACT

Innate inflammatory events promoting antiviral defense in the liver against murine cytomegalovirus (MCMV) infection have been characterized. However, the mechanisms that regulate the selective recruitment of inflammatory T lymphocytes to the liver during MCMV infection have not been defined. The studies presented here demonstrate the expression of monokine induced by gamma interferon (IFN-gamma; Mig/CXCL9) and IFN-gamma-inducible protein 10 (IP-10/CXCL10) in liver leukocytes and correlate their production with the infiltration of MCMV-specific CD8 T cells into the liver. Antibody-mediated neutralization of CXCL9 and CXCL10 and studies using mice deficient in CXCR3, the primary known receptor for these chemokines, revealed that CXCR3-dependent mechanisms promote the infiltration of virus-specific CD8 T cells into the liver during acute infection with MCMV. Furthermore, CXCR3 functions augmented the hepatic accumulation of CD8 T-cell IFN-gamma responses to MCMV. Evaluation of protective functions demonstrated enhanced pathology that overlapped with transient increases in virus titers in CXCR3-deficient mice. However, ultimate viral clearance and survival were not compromised. Thus, CXCR3-mediated signals support the accumulation of MCMV-specific CD8 T cells that contribute to, but are not exclusively required for, protective responses in a virus-infected tissue site.


Subject(s)
Cytomegalovirus Infections/immunology , Lymphocyte Activation , Muromegalovirus/immunology , Receptors, Chemokine/biosynthesis , T-Lymphocytes/immunology , Animals , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/virology , Liver/pathology , Mice , Mice, Inbred C57BL , Muromegalovirus/drug effects , Muromegalovirus/growth & development , Receptors, CXCR3 , T-Lymphocytes/metabolism
9.
FASEB J ; 20(7): 896-905, 2006 May.
Article in English | MEDLINE | ID: mdl-16675847

ABSTRACT

Leukocyte trafficking to the central nervous system (CNS), regulated in part by chemokines, determines severity of the demyelinating diseases multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE). To examine chemokine receptor CX3CR1 in EAE, we studied CX3CR1(GFP/GFP) mice, in which CX3CR1 targeting by insertion of Green Fluorescent Protein (GFP) allowed tracking of CX3CR1+ cells in CX3CR1(+/GFP) animals and cells destined to express CX3CR1 in CX3CR1(GFP/GFP) knockouts. NK cells were markedly reduced in the inflamed CNS of CX3CR1-deficient mice with EAE, whereas recruitment of T cells, NKT cells and monocyte/macrophages to the CNS during EAE did not require CX3CR1. Impaired recruitment of NK cells in CX3CR1(GFP/GFP) mice was associated with increased EAE-related mortality, nonremitting spastic paraplegia and hemorrhagic inflammatory lesions. The absence of CD1d did not affect the severity of EAE in CX3CR1(GFP/GFP) mice, arguing against a role for NKT cells. Accumulation of NK cells in livers of wild-type (WT) and CX3CR1(GFP/GFP) mice with cytomegalovirus hepatitis was equivalent, indicating that CX3CL1 mediated chemoattraction of NK cells was relatively specific for the CNS. These results are the first to define a chemokine that governs NK cell migration to the CNS, and the findings suggest novel therapeutic manipulation of CX3CR1+ NK cells.


Subject(s)
Central Nervous System/metabolism , Chemokines, CX3C/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Membrane Proteins/metabolism , Animals , Antigens, CD1/metabolism , Antigens, CD1d , Brain Stem/pathology , Central Nervous System/pathology , Chemokine CX3CL1 , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Expression Regulation , Hemorrhage/pathology , Killer Cells, Natural/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Paraparesis, Spastic/physiopathology , Spinal Cord/pathology
10.
J Immunol ; 174(3): 1549-56, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15661915

ABSTRACT

IFN-alpha/beta-mediated functions promote production of MIP-1alpha (or CCL3) by mediating the recruitment of MIP-1alpha-producing macrophages to the liver during early infection with murine CMV. These responses are essential for induction of NK cell inflammation and IFN-gamma delivery to support effective control of local infection. Nevertheless, it remains to be established if additional chemokine functions are regulated by IFN-alpha/beta and/or play intermediary roles in supporting macrophage trafficking. The chemokine MCP-1 (or CCL2) plays a distinctive role in the recruitment of macrophages by predominantly stimulating the CCR2 chemokine receptor. Here, we examine the roles of MCP-1 and CCR2 during murine CMV infection in liver. MCP-1 production preceded that of MIP-1alpha during infection and was dependent on IFN-alpha/beta effects for induction. Resident F4/80(+) liver leukocytes were identified as primary IFN-alpha/beta responders and major producers of MCP-1. Moreover, MCP-1 deficiency was associated with a dramatic reduction in the accumulation of macrophages and NK cells, as well as decreased production of MIP-1alpha and IFN-gamma in liver. These responses were also markedly impaired in mice with a targeted disruption of CCR2. Furthermore, MCP-1- and CCR2-deficient mice exhibited increased viral titers and elevated expression of the liver enzyme alanine aminotransferase in serum. These mice also had widespread virus-induced liver pathology and succumbed to infection. Collectively, these results establish MCP-1 and CCR2 interactions as factors promoting early liver inflammatory responses and define a mechanism for innate cytokines in regulation of chemokine functions critical for effective localized antiviral defenses.


Subject(s)
Chemokine CCL2/physiology , Cytomegalovirus Infections/immunology , Interferon Type I/physiology , Liver/immunology , Liver/pathology , Receptors, Chemokine/physiology , Animals , Cell Movement/genetics , Cell Movement/immunology , Chemokine CCL2/biosynthesis , Chemokine CCL2/deficiency , Chemokine CCL2/genetics , Chemokine CCL3 , Chemokine CCL4 , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/pathology , Cytomegalovirus Infections/virology , Female , Humans , Immunity, Innate , Inflammation/immunology , Inflammation/virology , Interferon Type I/administration & dosage , Interferon Type I/therapeutic use , Kinetics , Liver/metabolism , Liver/virology , Macrophage Inflammatory Proteins/biosynthesis , Macrophage Inflammatory Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muromegalovirus/immunology , Receptors, CCR2 , Receptors, Chemokine/biosynthesis , Receptors, Chemokine/deficiency , Receptors, Chemokine/genetics , Recombinant Proteins , Viral Load
11.
Viral Immunol ; 16(3): 291-306, 2003.
Article in English | MEDLINE | ID: mdl-14583145

ABSTRACT

The recruitment of immune effector cells to localized sites of infection is crucial for the effective delivery of innate immune mechanisms. Under the conditions of infections with murine cytomegalovirus (MCMV), a herpesvirus with pathogenic potential, early immune functions are essential in the control of virus replication and virus-induced pathology. Our studies have demonstrated that the chemokine macrophage inflammatory protein-1alpha (MIP-1alpha) is critical for natural killer (NK) cell inflammation and delivery of interferon (IFN)-gamma to mediate downstream protective responses against MCMV infection in liver. Moreover, IFN-alpha/beta-dependent mechanisms promote MIP-1alpha production and subsequently the accumulation of NK cells in liver. Taken together, the studies highlighted in this review define a unique in vivo pathway mediated by innate cytokines in regulating chemokine responses that are essential in the promotion of NK cell inflammation for localized antiviral defense. In addition, the downstream consequences of these events in enhancing endogenous adaptive immune responses will also be discussed. Overall, the innate cytokine/chemokine networks that are described emphasize the emerging importance of chemokine functions for protective immune responses during infection with viruses.


Subject(s)
Chemokines/biosynthesis , Cytokines/biosynthesis , Inflammation/immunology , Animals , Chemokine CCL3 , Chemokine CCL4 , Herpesviridae Infections/immunology , Humans , Immunity, Innate , Interferon Type I/biosynthesis , Killer Cells, Natural/immunology , Liver/immunology , Macrophage Inflammatory Proteins/biosynthesis , Macrophage Inflammatory Proteins/deficiency , Mice , Models, Immunological , Muromegalovirus/immunology , Muromegalovirus/pathogenicity
12.
Immunity ; 19(1): 59-70, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12871639

ABSTRACT

Dendritic cells (DCs) present microbial antigens to T cells and provide inflammatory signals that modulate T cell differentiation. While the role of DCs in adaptive immunity is well established, their involvement in innate immune defenses is less well defined. We have identified a TNF/iNOS-producing (Tip)-DC subset in spleens of Listeria monocytogenes-infected mice that is absent from CCR2-deficient mice. The absence of Tip-DCs results in profound TNF and iNOS deficiencies and an inability to clear primary bacterial infection. CD8 and CD4 T cell responses to L. monocytogenes antigens are preserved in CCR2-deficient mice, indicating that Tip-DCs are not essential for T cell priming. Tip-DCs, as the predominant source of TNF and iNOS during L. monocytogenes infection, orchestrate and mediate innate immune defense against this intracellular bacterial pathogen.


Subject(s)
Dendritic Cells/immunology , Immunity, Innate , Listeriosis/immunology , Nitric Oxide Synthase/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Antigens, Differentiation/analysis , CD11b Antigen/analysis , CD11c Antigen/analysis , Listeria monocytogenes , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II , Receptors, CCR2 , Receptors, Chemokine/physiology
13.
J Exp Med ; 197(7): 885-98, 2003 Apr 07.
Article in English | MEDLINE | ID: mdl-12682109

ABSTRACT

Differentiation of dendritic cells (DCs) into particular subsets may act to shape innate and adaptive immune responses, but little is known about how this occurs during infections. Plasmacytoid dendritic cells (PDCs) are major producers of interferon (IFN)-alpha/beta in response to many viruses. Here, the functions of these and other splenic DC subsets are further analyzed after in vivo infection with murine cytomegalovirus (MCMV). Viral challenge induced PDC maturation, their production of high levels of innate cytokines, and their ability to activate natural killer (NK) cells. The conditions also licensed PDCs to efficiently activate CD8 T cells in vitro. Non-plasmacytoid DCs induced T lymphocyte activation in vitro. As MCMV preferentially infected CD8alpha+ DCs, however, restricted access to antigens may limit plasmacytoid and CD11b+ DC contribution to CD8 T cell activation. IFN-alpha/beta regulated multiple DC responses, limiting viral replication in all DC and IL-12 production especially in the CD11b+ subset but promoting PDC accumulation and CD8alpha+ DC maturation. Thus, during defense against a viral infection, PDCs appear specialized for initiation of innate, and as a result of their production of IFN-alpha/beta, regulate other DCs for induction of adaptive immunity. Therefore, they may orchestrate the DC subsets to shape endogenous immune responses to viruses.


Subject(s)
Dendritic Cells/immunology , Herpesviridae Infections/immunology , Interferon-alpha/physiology , Interferon-beta/physiology , Muromegalovirus , Animals , Antigen Presentation , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Chemokines/biosynthesis , Cytokines/biosynthesis , Dendritic Cells/physiology , Killer Cells, Natural/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL
14.
J Immunol ; 169(8): 4279-87, 2002 Oct 15.
Article in English | MEDLINE | ID: mdl-12370359

ABSTRACT

NK cell cytotoxicity, IFN-gamma expression, proliferation, and accumulation are rapidly induced after murine CMV infections. Under these conditions, the responses were shown to be elicited in overlapping populations. Nevertheless, there were distinct signaling molecule requirements for induction of functions within the subsets. IL-12/STAT4 was critical for NK cell IFN-gamma expression, whereas IFN-alphabeta/STAT1 were required for induction of cytotoxicity. The accumulation/survival of proliferating NK cells was STAT4-independent but required IFN-alphabeta/STAT1 induction of IL-15. Taken together, the results define the coordinated interactions between the cytokines IFN-alphabeta, IL-12, and IL-15 for activation of protective NK cell responses during viral infections, and emphasize these factors' nonredundant functions under in vivo physiological conditions.


Subject(s)
Interferon-alpha/physiology , Interleukin-12/physiology , Interleukin-15/physiology , Killer Cells, Natural/immunology , Killer Cells, Natural/virology , Animals , Cytotoxicity, Immunologic/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Herpesviridae Infections/genetics , Herpesviridae Infections/immunology , Injections, Intraperitoneal , Interferon-alpha/administration & dosage , Interferon-alpha/deficiency , Interferon-alpha/genetics , Interferon-beta/deficiency , Interferon-beta/genetics , Interferon-beta/physiology , Interferon-gamma/biosynthesis , Interleukin-12/administration & dosage , Interleukin-12/deficiency , Interleukin-12/genetics , Interleukin-15/biosynthesis , Killer Cells, Natural/metabolism , Lymphocyte Activation/genetics , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Muromegalovirus/immunology , STAT1 Transcription Factor , STAT4 Transcription Factor , Signal Transduction/genetics , Signal Transduction/immunology , Trans-Activators/deficiency , Trans-Activators/genetics , Trans-Activators/physiology
15.
J Clin Invest ; 110(3): 321-30, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12163451

ABSTRACT

Macrophage inflammatory protein 1alpha (MIP-1alpha, CCL3) is critical for liver NK cell inflammation and delivery of IFN-gamma to mediate downstream protective responses against murine cytomegalovirus (MCMV) infections. This system was used to evaluate the upstream contribution of the type 1 IFNs, IFN-alpha/beta, in promotion of MIP-1alpha production. Mice deficient in IFN-alpha/beta functions, as a result of mutation in the receptor for these cytokines (IFN-alpha/betaR(-)), were profoundly deficient in MIP-1alpha expression and accumulation of NK cells and macrophages in the liver and had increased sensitivity to MCMV infection. The cytokines themselves were responsible for the immunoregulatory effects, since administration of recombinant IFN-alpha (rIFN-alpha) to immunocompetent mice also induced these changes. IFN-alpha/beta was required for NK cell accumulation during infection, and MIP-1alpha was required for NK cell accumulation in response to administered rIFN-alpha. In vivo trafficking assays demonstrated a requirement for IFN-alpha/betaR signaling for leukocyte localization in, and delivery of MIP-1alpha-producing macrophages to, the liver. These results extend characterization of the cytokine and chemokine cascade required for protection against viral infections in tissues by defining IFN-alpha/beta-dependent mechanisms promoting MIP-1alpha production and the resulting hepatic accumulation of NK cells.


Subject(s)
Interferon-alpha/immunology , Interferon-beta/immunology , Killer Cells, Natural/immunology , Liver/immunology , Macrophage Inflammatory Proteins/immunology , Macrophages/immunology , Animals , Biological Transport , Cell Movement , Chemokine CCL3 , Chemokine CCL4 , Female , Herpesviridae Infections/immunology , Humans , Interferon Type I/administration & dosage , Interferon Type I/immunology , Interferon-alpha/biosynthesis , Interferon-beta/biosynthesis , Killer Cells, Natural/cytology , Liver/pathology , Macrophage Inflammatory Proteins/biosynthesis , Macrophage Inflammatory Proteins/genetics , Macrophages/cytology , Male , Membrane Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Muromegalovirus/immunology , Receptor, Interferon alpha-beta , Receptors, Interferon/genetics , Receptors, Interferon/immunology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/immunology , Recombinant Proteins
16.
J Virol ; 76(9): 4520-5, 2002 May.
Article in English | MEDLINE | ID: mdl-11932417

ABSTRACT

Viruses and viral components can be potent inducers of alpha/beta interferons (IFN-alpha/beta). In culture, IFN-alpha/beta prime for their own expression, in response to viruses, through interferon regulatory factor 7 (IRF-7) induction. The studies presented here evaluated the requirements for functional IFN receptors and the IFN signaling molecule STAT1 in IFN-alpha/beta induction during infections of mice with lymphocytic choriomeningitis virus (LCMV). At 24 h after infection, levels of induced IFN-alpha/beta in serum were reduced 90 to 95% in IFN-alpha/beta receptor-deficient (IFN-alpha/betaR(-/-)) and STAT1(-/-) mice compared to those in wild-type mice. However, at 48 h, these mice showed elevated expression in the serum whereas IFN-alpha/beta levels were still reduced >75% in IFN-alpha/betagammaR(-/-) mice even though the viral burden was heavy. Levels of IFN-beta, IFN-alpha4, and non-IFN-alpha4 subtype mRNA expression correlated with IFN-alpha/beta bioactivity, and all IFN-alpha/beta subtypes were coincidentally detectable. IRF-7 mRNA was induced under conditions of IFN-alpha/beta production, including late production in IFN-alpha/betaR(-/-) mice. These data demonstrate that the presence of the virus alone is not sufficient to induce IFN-alpha/beta during LCMV infection in vivo. Instead, autocrine amplification through the IFN-alpha/betaR is necessary for optimal induction. In the absence of a functional IFN-alpha/betaR, however, alternative mechanisms, independent of STAT1 but requiring a functional IFN-gammaR, take over.


Subject(s)
DNA-Binding Proteins/metabolism , Interferon-alpha/metabolism , Interferon-beta/metabolism , Lymphocytic choriomeningitis virus/immunology , Receptors, Interferon/metabolism , Trans-Activators/metabolism , Animals , DNA-Binding Proteins/genetics , Interferon Regulatory Factor-7 , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Membrane Proteins , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Interferon alpha-beta , Receptors, Interferon/genetics , STAT1 Transcription Factor , Trans-Activators/genetics , Interferon gamma Receptor
17.
J Exp Med ; 195(4): 517-28, 2002 Feb 18.
Article in English | MEDLINE | ID: mdl-11854364

ABSTRACT

Interferon (IFN)-alpha/beta and interleukin (IL)-12 are cytokines critical in defense against viruses, but their cellular sources and mechanisms of regulation for in vivo expression remain poorly characterized. The studies presented here identified a novel subset of dendritic cells (DCs) as major producers of the cytokines during murine cytomegalovirus (MCMV) but not lymphocytic choriomeningitis virus (LCMV) infections. These DCs differed from those activated by Toxoplasma antigen but were related to plasmacytoid cells, as assessed by their CD8alpha(+)Ly6G/C(+)CD11b(-) phenotype. Another DC subset (CD8alpha(2)Ly6G/C(-)CD11b(+)) also contributed to IL-12 production in MCMV-infected immunocompetent mice, modestly. However, it dramatically increased IL-12 expression in the absence of IFN-alpha/beta functions. Conversely, IFN-alpha/beta production was greatly reduced under these conditions. Thus, a cross-regulation of DC subset cytokine responses was defined, whereby secretion of type I IFNs by CD8alpha(+) DCs resulted in responses limiting IL-12 expression by CD11b(+) DCs but enhancing overall IFN-alpha/beta production. Taken together, these data indicate that CD8alpha(+)Ly6G/C(+)CD11b(-) DCs play important roles in limiting viral replication and regulating immune responses, through cytokine production, in some but not all viral infections. They also illustrate the plasticity of cellular sources for innate cytokines in vivo and provide new insights into the roles of IFNs in shaping immune responses to viruses.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Expression Regulation , Interferon-alpha/immunology , Interferon-beta/immunology , Interleukin-12/immunology , Lymphocytic choriomeningitis virus/immunology , Muromegalovirus/immunology , Animals , Antigens, Viral/immunology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dendritic Cells/cytology , Flow Cytometry , Immunohistochemistry , Interferon-alpha/biosynthesis , Interferon-alpha/genetics , Interferon-beta/biosynthesis , Interferon-beta/genetics , Interleukin-12/biosynthesis , Interleukin-12/genetics , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Receptors, Interferon/metabolism , STAT1 Transcription Factor , Signal Transduction , Spleen/cytology , Spleen/immunology , T-Lymphocytes/immunology , Trans-Activators/genetics , Trans-Activators/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...