Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Infect Immun ; 83(10): 3857-64, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26195556

ABSTRACT

The polymorphic commensal fungus Candida albicans causes life-threatening disease via bloodstream and intra-abdominal infections in immunocompromised and transplant patients. Although host immune evasion is a common strategy used by successful human fungal pathogens, C. albicans provokes recognition by host immune cells less capable of destroying it. To accomplish this, C. albicans white cells secrete a low-molecular-weight chemoattractive stimulant(s) of macrophages, a phagocyte that they are able to survive within and eventually escape from. C. albicans opaque cells do not secrete this chemoattractive stimulant(s). We report here a physiological mechanism that contributes to the differences in the interaction of C. albicans white and opaque cells with macrophages. E,E-Farnesol, which is secreted by white cells only, is a potent stimulator of macrophage chemokinesis, whose activity is enhanced by yeast cell wall components and aromatic alcohols. E,E-farnesol results in up to an 8.5-fold increase in macrophage migration in vitro and promotes a 3-fold increase in the peritoneal infiltration of macrophages in vivo. Therefore, modulation of farnesol secretion to stimulate host immune recognition by macrophages may help explain why this commensal is such a successful pathogen.


Subject(s)
Candida albicans/physiology , Candidiasis/microbiology , Farnesol/immunology , Macrophages/cytology , Quorum Sensing , Animals , Candida albicans/genetics , Candida albicans/immunology , Candidiasis/immunology , Cell Movement , Cells, Cultured , Chemotactic Factors/immunology , Female , Humans , Macrophages/immunology , Mice , Mice, Inbred C57BL
2.
J Virol ; 86(19): 10841-51, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22837198

ABSTRACT

During Theiler's murine encephalomyelitis virus (TMEV) infection of macrophages, it is thought that high interleukin-6 (IL-6) levels contribute to the demyelinating disease found in chronically infected SJL/J mice but absent in B10.S mice capable of clearing the infection. Therefore, IL-6 expression was measured in TMEV-susceptible SJL/J and TMEV-resistant B10.S macrophages during their infection with TMEV DA strain or responses to lipopolysaccharide (LPS) or poly(I · C). Unexpectedly, IL-6 production was greater in B10.S macrophages than SJL/J macrophages during the first 24 h after stimulation with TMEV, LPS, or poly(I · C). Further experiments showed that in B10.S, SJL/J, and RAW264.7 macrophage cells, IL-6 expression was dependent on extracellular signal-regulated kinase (ERK) mitogen-activated protein kinase (MAPK) and enhanced by exogenous IL-12. In SJL/J and RAW264.7 macrophages, exogenous IL-6 resulted in decreased TMEV replication, earlier activation of STAT1 and STAT3, production of nitric oxide, and earlier upregulation of several antiviral genes downstream of STAT1. However, neither inhibition of IL-6-induced nitric oxide nor knockdown of STAT1 diminished the early antiviral effect of exogenous IL-6. In addition, neutralization of endogenous IL-6 from SJL/J macrophages with Fab antibodies did not exacerbate early TMEV infection. Therefore, endogenous IL-6 expression after TMEV infection is dependent on ERK MAPK, enhanced by IL-12, but too slow to decrease viral replication during early infection. In contrast, exogenous IL-6 enhances macrophage control of TMEV infection through preemptive antiviral nitric oxide production and antiviral STAT1 activation. These results indicate that immediate-early production of IL-6 could protect macrophages from TMEV infection.


Subject(s)
Nitric Oxide/chemistry , STAT1 Transcription Factor/metabolism , Theilovirus/immunology , Virus Replication , Animals , Cell Line , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Interleukin-6/metabolism , Lipopolysaccharides/chemistry , MAP Kinase Signaling System , Macrophages/cytology , Mice , Nitric Oxide/metabolism , RNA Interference , STAT3 Transcription Factor/metabolism
3.
J Mol Biol ; 433(1): 166583, 2021 01 08.
Article in English | MEDLINE | ID: mdl-32598936

ABSTRACT

B lymphocytes have well-established effector roles during viral infections, including production of antibodies and functioning as antigen-presenting cells for CD4+ and CD8+ T cells. B cells have also been shown to regulate immune responses and induce regulatory T cells (Tregs). In the Friend virus (FV) model, Tregs are known to inhibit effector CD8+ T-cell responses and contribute to virus persistence. Recent work has uncovered a role for B cells in the induction and activation of Tregs during FV infection. In addition to inducing Tregs, B cell antibody production and antigen-presenting cell activity is a target of Treg suppression. This review focuses on the dynamic interactions between B cells and Tregs during FV infection.


Subject(s)
B-Lymphocytes/immunology , Friend murine leukemia virus/immunology , Host-Pathogen Interactions/immunology , Retroviridae Infections/veterinary , Rodent Diseases/immunology , Rodent Diseases/virology , T-Lymphocytes, Regulatory/immunology , Animals , Antibody Formation/immunology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , B-Lymphocytes/metabolism , Cell Communication/immunology , Rodent Diseases/metabolism , T-Lymphocytes, Regulatory/metabolism
4.
FEBS Lett ; 595(21): 2665-2674, 2021 11.
Article in English | MEDLINE | ID: mdl-34591979

ABSTRACT

Toll-like receptor 3 (TLR3) recognizes viral double-stranded RNA (or the synthetic dsRNA analog poly I:C) and induces a signal transduction pathway that results in activation of transcription factors that induce expression of antiviral genes including type I interferon (IFN-I). Secreted IFN-I positively feeds back to amplify antiviral gene expression. In this report, we study the role of MEK/ERK MAP kinase in modulating antiviral gene expression downstream of TLR3. We find MEK/ERK is a negative regulator of antiviral gene expression by limiting expression of IFN-ß. However, MEK/ERK does not limit antiviral responses downstream of the type I interferon receptor. These findings provide insights into regulatory mechanisms of antiviral gene expression and reveal potential targets for modulating antiviral immunity.


Subject(s)
Antiviral Agents , Extracellular Signal-Regulated MAP Kinases , Interferon-beta , Animals , Mice , Poly I-C , RAW 264.7 Cells
5.
Assay Drug Dev Technol ; 18(6): 261-268, 2020.
Article in English | MEDLINE | ID: mdl-32799543

ABSTRACT

Malignant melanoma is an aggressive cancer with a poor prognosis despite numerous advances in therapeutic strategies. Quercetin is a plant-derived flavonoid suggested to have potent anticancer properties. Quercetin has no demonstrable toxicity in humans, further supporting the possibility of using quercetin therapeutically. We chose to investigate quercetin efficacy against B16 murine melanoma cells and identify the mechanisms of anticancer activity. Treatment of B16 melanoma cells with 50 µg/mL quercetin resulted in a 75% reduction in viability from 6 through 48 h post-treatment. The reduction in cancer cell viability was comparable to or greater than what was observed with etoposide, an established chemotherapeutic. Specifically, we found Quercetin reduced the proliferation of B16 melanoma cells at 48 h as much or more than etoposide. Although quercetin reduced the proportion of cells in the S and G2/M stages of the cell cycle, this could largely be explained by an increase in the subG1 population in quercetin-treated cells (suggesting apoptosis). Quercetin-induced apoptosis was confirmed by flow cytometry analysis of Annexin V+ cells. Collectively, our findings demonstrate quercetin reduces proliferation and induces apoptosis of B16 melanoma cells in vitro.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Melanoma/drug therapy , Quercetin/pharmacology , Skin Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Melanoma/pathology , Mice , Quercetin/chemistry , Skin Neoplasms/pathology , Tumor Cells, Cultured , Melanoma, Cutaneous Malignant
6.
Microbiol Resour Announc ; 8(41)2019 Oct 10.
Article in English | MEDLINE | ID: mdl-31601674

ABSTRACT

We sequenced the genome of a bacterial species recently isolated from fresh water at Dripping Springs, NM, and identified it as Chryseobacterium viscerum This species had previously been isolated only from dead or diseased fish. This report shows that C. viscerum can be found in nature as a free-living species not associated with diseased fish.

7.
mBio ; 10(1)2019 01 22.
Article in English | MEDLINE | ID: mdl-30670616

ABSTRACT

Friend virus (FV) is a naturally occurring mouse retrovirus that infects dividing cells of the hematopoietic lineage, including antigen-presenting cells (APCs). The infection of APCs by viruses often induces their dysfunction, and it has been shown that FV infection reduces the ability of dendritic cells (DCs) to prime critical CD8+ T cell responses. Nonetheless, mice mount vigorous CD8+ T cell responses, so we investigated whether B cells might serve as alternative APCs during FV infection. Direct ex vivo analysis of B cells from FV-infected mice revealed that infected but not uninfected B cells upregulated expression of the costimulatory molecules CD80, CD86, and CD40, as well as major histocompatibility complex class II (MHC-II) molecules. Furthermore, in vitro studies showed that, compared to uninfected B cells from the same mice, the FV-infected B cells had significantly enhanced APC function, as measured by their capacity to prime CD8+ T cell activation and proliferation. Thus, in contrast to DCs, infection of B cells with FV enhanced their APC capacity and ability to stimulate the CD8+ T cell responses essential for virus control. FV infections also induce the activation and expansion of regulatory T cells (Tregs), so it was of interest to determine the impact of Tregs on B cell activation. The upregulation of costimulatory molecule expression and APC function of B cells was even more strongly enhanced by in vivo depletion of regulatory T cells than infection. Thus, Tregs exert potent homeostatic suppression of B cell activation that is partially overcome by FV infection.IMPORTANCE The primary role of B cells in immunity is considered the production of pathogen-specific antibodies, but another, less-well-studied, function of B cells is to present foreign antigens to T cells to stimulate their activation and proliferation. Dendritic cells (DCs) are considered the most important antigen-presenting cells (APCs) for CD8+ T cells, but DCs lose APC function when infected with Friend virus (FV), a model retrovirus of mice. Interestingly, B cells were better able to stimulate CD8+ T cell responses when they were infected with FV. We also found that the activation status of B cells under homeostatic conditions was potently modulated by regulatory T cells. This study illustrates an important link between B cell and T cell responses and illustrates an additional mechanism by which regulatory T cells suppress critical T cell responses during viral infections.


Subject(s)
Antigen Presentation , B-Lymphocytes/immunology , Friend murine leukemia virus/immunology , T-Lymphocytes, Regulatory/immunology , Animals , B-Lymphocytes/chemistry , B7-1 Antigen/analysis , B7-2 Antigen/analysis , CD40 Antigens/analysis , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Histocompatibility Antigens Class II/analysis , Leukemia, Experimental/immunology , Leukemia, Experimental/virology , Lymphocyte Activation , Mice , Retroviridae Infections/immunology , Retroviridae Infections/virology , Tumor Virus Infections/immunology , Tumor Virus Infections/virology
8.
PLoS One ; 13(4): e0195402, 2018.
Article in English | MEDLINE | ID: mdl-29614127

ABSTRACT

Recent vaccine studies with experimental antigens have shown that regulatory T cells (Tregs) constrain the magnitude of B cell responses. This homeostatic Treg-mediated suppression is thought to reduce the potential of germinal center (GC) responses to generate autoreactive antibodies. However, essentially opposite results were observed in live influenza infections where Tregs promoted B cell and antibody responses. Thus, it remains unclear whether Tregs dampen or enhance B cell responses, especially during live viral infections. Here, we use mice infected with Friend retrovirus (FV), which induces a robust expansion of Tregs. Depletion of Tregs led to elevated activation, proliferation, and class switching of B cells. In addition, Treg depletion enhanced the production of virus-specific and virus-neutralizing antibodies and reduced FV viremia. Thus, in contrast to influenza infection, Tregs either directly or indirectly suppress B cells during mouse retroviral infection indicating that the ultimate effect of Tregs on B cell responses is specific to the particular infectious agent.


Subject(s)
Antibodies, Viral/metabolism , Friend murine leukemia virus/immunology , Leukemia, Experimental/immunology , Retroviridae Infections/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Virus Infections/immunology , Animals , B-Lymphocytes/immunology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Immunoglobulin G/metabolism , Mice, Transgenic , Spleen/immunology
9.
mBio ; 8(4)2017 08 01.
Article in English | MEDLINE | ID: mdl-28765225

ABSTRACT

Regulatory T cells (Tregs) are immunosuppressive cells of the immune system that control autoimmune reactivity. Tregs also respond during immune reactions to infectious agents in order to limit immunopathological damage from potent effectors such as CD8+ cytolytic T lymphocytes. We have used the Friend virus (FV) model of retroviral infection in mice to investigate how viral infections induce Tregs. During acute FV infection, there is significant activation and expansion of thymus-derived (natural) Tregs that suppress virus-specific CD8+ T cell responses. Unlike conventional T cells, the responding Tregs are not virus specific, so the mechanisms that induce their expansion are of great interest. We now show that B cells provide essential signals for Treg expansion during FV infection. Treg responses are greatly diminished in B cell-deficient mice but can be restored by adoptive transfers of B cells at the time of infection. The feeble Treg responses in B cell-deficient mice are associated with enhanced virus-specific CD8+ T cell responses and accelerated virus control during the first 2 weeks of infection. In vitro experiments demonstrated that B cells promote Treg activation and proliferation through a glucocorticoid-induced receptor superfamily member 18 (GITR) ligand-dependent mechanism. Thus, B cells play paradoxically opposing roles during FV infection. They provide proliferative signals to immunsosuppressive Tregs, which slows early virus control, and they also produce virus-specific antibodies, which are essential for long-term virus control.IMPORTANCE When infectious agents invade a host, numerous immunological mechanisms are deployed to limit their replication, neutralize their spread, and destroy the host cells harboring the infection. Since immune responses also have a strong capacity to damage host cells and tissues, their magnitude, potency, and duration are under regulatory control. Regulatory T cells are an important component of this control, and the mechanisms that induce them to respond and exert immunosuppressive regulation are of great interest. In the current report, we show that B cells, the cells responsible for making pathogen-specific antibodies, are also involved in promoting the expansion of regulatory T cells during a retroviral infection. In vitro studies demonstrated that they do so via stimulation of the Tregs through interactions between cell surface molecules: GITR interactions with its ligand (GITRL) on B cells and GITR on regulatory T cells. These findings point the way toward therapeutics to better treat infections and autoimmune diseases.


Subject(s)
B-Lymphocytes/immunology , Cell Proliferation , Friend murine leukemia virus/immunology , Retroviridae Infections/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Virus Infections/immunology , Adoptive Transfer , Animals , Antibodies, Viral/biosynthesis , Antibodies, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Glucocorticoid-Induced TNFR-Related Protein/genetics , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Mice , Mice, Inbred C57BL , Receptors, Tumor Necrosis Factor/metabolism , T-Lymphocytes, Regulatory/physiology
10.
Virology ; 488: 88-95, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26609939

ABSTRACT

Amyloid fibrils from semen-derived peptide (SEVI) enhance HIV-1 infectivity in vitro but the ability of SEVI to mediate enhancement of HIV infection in vivo has not been tested. In this study we used immunodeficient mice reconstituted with human immune systems to test for in vivo enhancement of HIV-1 transmission. This mouse model supports mucosal transmission of HIV-1 via the intrarectal route leading to productive infection. In separate experiments with humanized mouse cohorts reconstituted with two different donor immune systems, high dose HIV-1JR-CSF that had been incubated with SEVI amyloid fibrils at physiologically relevant concentrations did not show an increased incidence of infection compared to controls. In addition, SEVI failed to enhance rectal transmission with a reduced concentration of HIV-1. Although we confirmed potent SEVI-mediated enhancement of HIV infectivity in vitro, this model showed no evidence that it plays a role in the much more complex situation of in vivo transmission.


Subject(s)
Amyloid/metabolism , HIV Infections/transmission , Rectum/virology , Semen/chemistry , Animals , CD4-Positive T-Lymphocytes/virology , Cells, Cultured , Disease Models, Animal , Female , Humans , Incidence , Male , Mice, Inbred C57BL , Mice, SCID
11.
Microbes Infect ; 17(6): 426-39, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25777301

ABSTRACT

The role of interferon regulatory factor 3 (IRF3) in the innate immune response to infection has been well studied. However, less is known about IRF3 signaling in shaping the adaptive T cell response. To determine the role of IRF3 in the generation and maintenance of effective anti-viral T cell responses, mice deficient in IRF3 were infected with a potentially persistent virus, Theiler's murine encephalomyelitis virus (TMEV) or with a model acute infection, influenza A virus (IAV). IRF3 was required to prevent TMEV persistence and induce robust TMEV specific effector T cell responses at the site of infection. This defect was more pronounced in the memory phase with an apparent lack of TMEV-specific memory T cells expressing granzyme B (GrB) in IRF3 deficient mice. In contrast, IRF3 had no effect on antigen specific T cell responses at the effector stage during IAV infection. However, memory T cell responses to IAV were also impaired in IRF3 deficient mice. Furthermore, addition of cytokines during peptide restimulation could not restore GrB expression in IRF3 deficient memory T cells. Taken together, IRF3 plays an important role in the maintenance of effective anti-viral T cell memory responses.


Subject(s)
Granzymes/metabolism , Interferon Regulatory Factor-3/deficiency , T-Lymphocytes/immunology , Theilovirus/immunology , Animals , Granzymes/immunology , Mice , Signal Transduction/immunology , T-Lymphocytes/metabolism , Theilovirus/metabolism
12.
Cancer Lett ; 346(1): 122-8, 2014 Apr 28.
Article in English | MEDLINE | ID: mdl-24368188

ABSTRACT

Interferon Response Factor 3 (IRF3) induces several NK-cell activating factors, is activated by poly-I:C, an experimental cancer therapeutic, but is suppressed during many viral infections. IRF3 Knockout (KO) mice exhibited enhanced B16 melanoma growth, impaired intratumoral NK cell infiltration, but not an impaired poly-I:C therapeutic effect due to direct suppression of B16 growth. IRF3 was responsible for poly-I:C decrease in TIM-3 expression by intratumoral dendritic cells, induction of NK-cell Granzyme B and IFN-γ, and induction of macrophage IL-12, IL-15, IL-6, and IRF3-dependent NK-activating molecule (INAM). Thus, IRF3 is a key factor controlling melanoma growth through NK-cell activities, especially during poly-I:C therapy.


Subject(s)
Interferon Regulatory Factor-3/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Animals , Cell Proliferation , Flow Cytometry , Interferon Inducers/pharmacology , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Poly I-C/pharmacology
13.
FEBS Lett ; 587(18): 3014-20, 2013 Sep 17.
Article in English | MEDLINE | ID: mdl-23892079

ABSTRACT

Understanding nitric oxide (NO) in innate anti-viral immunity and immune-mediated pathology is hampered by incomplete details of its transcriptional and signaling factors. We found in macrophages that IRF3, ERK MAP-kinases, and PKR are essential to NO production in response to RNA-virus mimic, poly I:C, a TLR3 agonist. ERK's role in NO induction may be through phosphorylation of serine-171 of IRF3 and expression of NO-inducing cytokines, IL-6 and IFN-ß. However, these cytokines induced less NO in IRF3 knockout or knockdown macrophages. These findings show that ERK and IRF3 coordinate induction of NO by macrophages in response to stimulation of TLR3.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Interferon Regulatory Factor-3/genetics , Macrophages, Peritoneal/drug effects , Poly I-C/pharmacology , Amino Acid Sequence , Animals , Cell Line , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/genetics , Gene Expression Regulation , Interferon Regulatory Factor-3/deficiency , Interferon-beta/biosynthesis , Interleukin-6/biosynthesis , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/metabolism , Mice , Mice, Knockout , Molecular Sequence Data , Nitric Oxide/biosynthesis , Protein Kinase Inhibitors/pharmacology , Signal Transduction
14.
Virus Res ; 178(2): 226-33, 2013 Dec 26.
Article in English | MEDLINE | ID: mdl-24140628

ABSTRACT

IRF3 is an innate anti-viral factor whose role in limiting Theiler's murine encephalomyelitis virus (TMEV) infection and preventing TMEV-induced disease is unclear. Acute disease and innate immune responses of macrophages were examined in IRF3 knockout mice compared with C57Bl/6 mice following in vitro or intracranial infection with either TMEV GDVII or DA. IRF3 deficiency augmented viral infection, as well as morbidity and mortality following intracranial infection with neurovirulent TMEV GDVII. In contrast, IRF3 deficiency prevented hippocampal injury following intracranial infection with persistent TMEV DA. The extent of TMEV infection in macrophages from C57Bl/6 mice was significantly less than that in IRF3 deficient macrophages, which was associated with poor IFN-ß and IL-6 expression in response to TMEV. Reestablishing IRF3 expression in IRF3 deficient macrophages increased control of TMEV replication and increased expression of IFN-ß and IL-6. In addition, IRF3 deficient macrophages failed to exhibit IL-6 antiviral effects, which was associated with inability to sustain IL-6-induced STAT1 activation compared with C57BL/6 macrophages. Altogether, IRF3 contributes to early control of TMEV replication through induction of IL-6 and IFN-ß and support of IL-6 antiviral effects, but contributes to TMEV-induced hippocampal injury.


Subject(s)
Cardiovirus Infections/immunology , Cardiovirus Infections/pathology , Host-Pathogen Interactions , Interferon Regulatory Factor-3/metabolism , Interleukin-6/immunology , Theilovirus/immunology , Animals , Cardiovirus Infections/virology , Hippocampus/immunology , Hippocampus/pathology , Hippocampus/virology , Interferon Regulatory Factor-3/deficiency , Interferon-beta/biosynthesis , Interferon-beta/immunology , Interleukin-6/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Severity of Illness Index , Survival Analysis , Theilovirus/physiology
15.
Virology ; 418(1): 40-8, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21810534

ABSTRACT

Persistent viral infections can lead to disease such as myocarditis. Theiler's murine encephalomyelitis virus (TMEV) infects macrophages of SJL/J (H-2s) mice establishing persistent infections leading to demyelinating disease. In contrast macrophages from B10.S (H-2s) mice clear TMEV. Activation of the transcription factor IRF3 induces IFNß, ISG56, and apoptosis for viral clearance, but also inflammatory cytokines, such as IL-23 and IL6, which contribute to disease. Here we identify polymorphisms in the IRF3 of SJL/J versus B10.S mice that are located in DNA binding, nuclear localization, and autoinhibitory domains. SJL-IRF3 expression in RAW264.7 macrophage cells with or without TMEV infection decreased IL-23p19 promoter activity compared with B10S-IRF3. In contrast SJL-IRF3 increased IL-6, ISG56 and IFNß in response to TMEV. B10S-IRF3 expression augmented apoptotic caspase activation and decreased viral RNA in TMEV-infected macrophages while SJL-IRF3 increased viral replication with less caspase activation. Therefore IRF3 polymorphisms contribute to viral persistence and altered cytokine expression.


Subject(s)
Host-Pathogen Interactions , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Macrophages/immunology , Macrophages/virology , Polymorphism, Genetic , Theilovirus/immunology , Theilovirus/pathogenicity , Animals , Apoptosis , Cell Line , Cytokines/biosynthesis , Female , Mice
SELECTION OF CITATIONS
SEARCH DETAIL