Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters











Publication year range
1.
Nature ; 633(8028): 174-181, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39198639

ABSTRACT

Sterile inflammation after myocardial infarction is classically credited to myeloid cells interacting with dead cell debris in the infarct zone1,2. Here we show that cardiomyocytes are the dominant initiators of a previously undescribed type I interferon response in the infarct borderzone. Using spatial transcriptomics analysis in mice and humans, we find that myocardial infarction induces colonies of interferon-induced cells (IFNICs) expressing interferon-stimulated genes decorating the borderzone, where cardiomyocytes experience mechanical stress, nuclear rupture and escape of chromosomal DNA. Cardiomyocyte-selective deletion of Irf3 abrogated IFNIC colonies, whereas mice lacking Irf3 in fibroblasts, macrophages, neutrophils or endothelial cells, Ccr2-deficient mice or plasmacytoid-dendritic-cell-depleted mice did not. Interferons blunted the protective matricellular programs and contractile function of borderzone fibroblasts, and increased vulnerability to pathological remodelling. In mice that died after myocardial infarction, IFNIC colonies were immediately adjacent to sites of ventricular rupture, while mice lacking IFNICs were protected from rupture and exhibited improved survival3. Together, these results reveal a pathological borderzone niche characterized by a cardiomyocyte-initiated innate immune response. We suggest that selective inhibition of IRF3 activation in non-immune cells could limit ischaemic cardiomyopathy while avoiding broad immunosuppression.


Subject(s)
Interferon Type I , Myocardial Infarction , Animals , Female , Humans , Male , Mice , Dendritic Cells/immunology , Endothelial Cells/metabolism , Fibroblasts/metabolism , Gene Expression Profiling , Immunity, Innate , Interferon Regulatory Factor-3/antagonists & inhibitors , Interferon Regulatory Factor-3/deficiency , Interferon Regulatory Factor-3/metabolism , Interferon Type I/metabolism , Interferon Type I/immunology , Macrophages/metabolism , Macrophages/immunology , Mice, Inbred C57BL , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Neutrophils/metabolism , Receptors, CCR2/metabolism , Receptors, CCR2/deficiency , Receptors, CCR2/genetics
3.
J Gen Virol ; 100(1): 46-62, 2019 01.
Article in English | MEDLINE | ID: mdl-30451651

ABSTRACT

Interferon (IFN) regulatory factors (IRFs) are important determinants of the innate response to infection. We evaluated the role(s) of combined and individual IRF deficiencies in the outcome of infection of C57BL/6 mice with Sindbis virus, an alphavirus that infects neurons and causes encephalomyelitis. The brain and spinal cord levels of Irf7, but not Irf3 mRNAs, were increased after infection. IRF3/5/7-/- and IRF3/7-/- mice died within 3-4 days with uncontrolled virus replication, similar to IFNα receptor-deficient mice, while all wild-type (WT) mice recovered. IRF3-/- and IRF7-/- mice had brain levels of IFNα that were lower, but brain and spinal cord levels of IFNß and IFN-stimulated gene mRNAs that were similar to or higher than WT mice without detectable serum IFN or increases in Ifna or Ifnb mRNAs in the lymph nodes, indicating that the differences in outcome were not due to deficiencies in the central nervous system (CNS) type I IFN response. IRF3-/- mice developed persistent neurological deficits and had more spinal cord inflammation and higher CNS levels of Il1b and Ifnγ mRNAs than WT mice, but all mice survived. IRF7-/- mice died 5-8 days after infection with rapidly progressive paralysis and differed from both WT and IRF3-/- mice in the induction of higher CNS levels of IFNß, tumour necrosis factor (TNF) α and Cxcl13 mRNA, delayed virus clearance and more extensive cell death. Therefore, fatal disease in IRF7-/- mice is likely due to immune-mediated neurotoxicity associated with failure to regulate the production of inflammatory cytokines such as TNFα in the CNS.


Subject(s)
Alphavirus Infections/physiopathology , Encephalomyelitis/physiopathology , Host-Pathogen Interactions , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-7/metabolism , Sindbis Virus/growth & development , Animals , Brain/pathology , Disease Models, Animal , Gene Expression Profiling , Interferon Regulatory Factor-3/deficiency , Interferon Regulatory Factor-7/deficiency , Mice, Inbred C57BL , Mice, Knockout , Spinal Cord/pathology , Survival Analysis
4.
Elife ; 72018 06 19.
Article in English | MEDLINE | ID: mdl-29914621

ABSTRACT

Type I interferon (IFN-I) responses are critical for the control of RNA virus infections, however, many viruses, including Dengue (DENV) and Chikungunya (CHIKV) virus, do not directly activate plasmacytoid dendritic cells (pDCs), robust IFN-I producing cells. Herein, we demonstrated that DENV and CHIKV infected cells are sensed by pDCs, indirectly, resulting in selective IRF7 activation and IFN-I production, in the absence of other inflammatory cytokine responses. To elucidate pDC immunomodulatory functions, we developed a mouse model in which IRF7 signaling is restricted to pDC. Despite undetectable levels of IFN-I protein, pDC-restricted IRF7 signaling controlled both viruses and was sufficient to protect mice from lethal CHIKV infection. Early pDC IRF7-signaling resulted in amplification of downstream antiviral responses, including an accelerated natural killer (NK) cell-mediated type II IFN response. These studies revealed the dominant, yet indirect role of pDC IRF7-signaling in directing both type I and II IFN responses during arbovirus infections.


Subject(s)
Chikungunya Fever/immunology , Dengue/immunology , Host-Pathogen Interactions/immunology , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-7/immunology , Interferon Type I/immunology , Animals , Chikungunya Fever/genetics , Chikungunya Fever/mortality , Chikungunya Fever/pathology , Chikungunya virus/growth & development , Chikungunya virus/immunology , Chikungunya virus/pathogenicity , Dendritic Cells/immunology , Dendritic Cells/virology , Dengue/genetics , Dengue/mortality , Dengue/pathology , Dengue Virus/growth & development , Dengue Virus/immunology , Dengue Virus/pathogenicity , Disease Models, Animal , Female , Gene Expression Regulation , Humans , Interferon Regulatory Factor-3/deficiency , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-7/deficiency , Interferon Regulatory Factor-7/genetics , Interferon Type I/genetics , Interferon-gamma/genetics , Interferon-gamma/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/virology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Viral/antagonists & inhibitors , RNA, Viral/genetics , RNA, Viral/immunology , Signal Transduction , Spleen/immunology , Spleen/virology , Survival Analysis
5.
PLoS Pathog ; 13(12): e1006748, 2017 12.
Article in English | MEDLINE | ID: mdl-29244871

ABSTRACT

Chikungunya virus (CHIKV) and Ross River virus (RRV) are mosquito-transmitted alphaviruses that cause debilitating acute and chronic musculoskeletal disease. Monocytes are implicated in the pathogenesis of these infections; however, their specific roles are not well defined. To investigate the role of inflammatory Ly6ChiCCR2+ monocytes in alphavirus pathogenesis, we used CCR2-DTR transgenic mice, enabling depletion of these cells by administration of diptheria toxin (DT). DT-treated CCR2-DTR mice displayed more severe disease following CHIKV and RRV infection and had fewer Ly6Chi monocytes and NK cells in circulation and muscle tissue compared with DT-treated WT mice. Furthermore, depletion of CCR2+ or Gr1+ cells, but not NK cells or neutrophils alone, restored virulence and increased viral loads in mice infected with an RRV strain encoding attenuating mutations in nsP1 to levels detected in monocyte-depleted mice infected with fully virulent RRV. Disease severity and viral loads also were increased in DT-treated CCR2-DTR+;Rag1-/- mice infected with the nsP1 mutant virus, confirming that these effects are independent of adaptive immunity. Monocytes and macrophages sorted from muscle tissue of RRV-infected mice were viral RNA positive and had elevated expression of Irf7, and co-culture of Ly6Chi monocytes with RRV-infected cells resulted in induction of type I IFN gene expression in monocytes that was Irf3;Irf7 and Mavs-dependent. Consistent with these data, viral loads of the attenuated nsP1 mutant virus were equivalent to those of WT RRV in Mavs-/- mice. Finally, reconstitution of Irf3-/-;Irf7-/- mice with CCR2-DTR bone marrow rescued mice from severe infection, and this effect was reversed by depletion of CCR2+ cells, indicating that CCR2+ hematopoietic cells are capable of inducing an antiviral response. Collectively, these data suggest that MAVS-dependent production of type I IFN by monocytes is critical for control of acute alphavirus infection and that determinants in nsP1, the viral RNA capping protein, counteract this response.


Subject(s)
Alphavirus Infections/immunology , Alphavirus Infections/virology , Monocytes/immunology , Monocytes/virology , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , Antigens, Ly/metabolism , Chikungunya virus/immunology , Chikungunya virus/pathogenicity , Diphtheria Toxin/pharmacology , Heparin-binding EGF-like Growth Factor/genetics , Heparin-binding EGF-like Growth Factor/immunology , Humans , Inflammation/virology , Interferon Regulatory Factor-3/deficiency , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-7/deficiency , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/immunology , Interferon Type I/biosynthesis , Interferon Type I/genetics , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Monocytes/drug effects , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Ross River virus/genetics , Ross River virus/immunology , Ross River virus/pathogenicity , Viral Load , Virulence/genetics , Virulence/immunology
6.
Eur J Immunol ; 46(11): 2614-2628, 2016 11.
Article in English | MEDLINE | ID: mdl-27546168

ABSTRACT

Very few transcription factors have been identified that are required by antigen-presenting cells (APCs) to induce T helper type 2 (Th2) responses. Because lung CD11b+ conventional dendritic cells (CD11b+ cDCs) are responsible for priming Th2 responses in house-dust mite (HDM)-induced airway allergy, we used them as a model to identify transcriptional events regulating the pro-Th2 activity of cDCs. Transcriptomic profiling of lung CD11b+ cDCs exposed to HDM in vivo revealed first that HDM triggers an antiviral defence-like response, and second that the majority of HDM-induced transcriptional changes depend on the transcription factor Interferon Response Factor-3 (Irf3). Validating the functional relevance of these observations, Irf3-deficient CD11b+ cDCs displayed reduced pro-allergic activity. Indeed, Irf3-deficient CD11b+ cDCs induced less Th2, more regulatory T cell, and similar Th1 differentiation in naïve CD4+ T cells compared to their wild-type counterparts. The altered APC activity of Irf3 CD11b+ cDCs was associated with reduced expression of CD86 and was phenocopied by blocking CD86 activity in wild-type CD11b+ cDCs. Altogether, these results establish Irf3, known mostly for its role in antiviral responses, as a transcription factor involved in the induction of Th2 responses through the promotion of pro-Th2 costimulation in CD11b+ DCs.


Subject(s)
Allergens/immunology , Antigens, Dermatophagoides/immunology , Dendritic Cells/immunology , Interferon Regulatory Factor-3/metabolism , Lung/immunology , Th2 Cells/immunology , Transcription Factors/immunology , Administration, Intranasal , Animals , Antigens, Dermatophagoides/administration & dosage , CD11b Antigen/genetics , CD11b Antigen/immunology , Cell Differentiation , Interferon Regulatory Factor-3/deficiency , Interferon Regulatory Factor-3/genetics , Mice , Mice, Knockout , Microarray Analysis , Phenotype
7.
PLoS One ; 11(5): e0155243, 2016.
Article in English | MEDLINE | ID: mdl-27182740

ABSTRACT

Chikungunya virus is an arbovirus spread predominantly by Aedes aegypti and Ae. albopictus mosquitoes, and causes debilitating arthralgia and arthritis. While these are common manifestations during acute infection and it has been suggested they can recur in patients chronically, gaps in knowledge regarding the pathogenesis still exist. Two established mouse models were utilized (adult IRF 3/7 -/- -/- and wild-type C57BL/6J mice) to evaluate disease manifestations in bones and joints at various timepoints. Novel lesions in C57BL/6J mice consisted of periostitis (91%) and foci of cartilage of necrosis (50% of mice at 21 DPI). Additionally, at 21 DPI, 50% and 75% of mice exhibited periosteal bone proliferation affecting the metatarsal bones, apparent via histology and µCT, respectively. µCT analysis did not reveal any alterations in trabecular bone volume measurements in C57BL/6J mice. Novel lesions demonstrated in IRF 3/7 -/- -/- mice at 5 DPI included focal regions of cartilage necrosis (20%), periosteal necrosis (66%), and multifocal ischemic bone marrow necrosis (100%). Contralateral feet in 100% of mice of both strains had similar, though milder lesions. Additionally, comparison of control IRF 3/7 -/- -/- and wild-type C57BL/6J mice demonstrated differences in cortical bone. These experiments demonstrate novel manifestations of disease similar to those occurring in humans, adding insight into disease pathogenesis, and representing new potential targets for therapeutic interventions. Additionally, results demonstrate the utility of µCT in studies of bone and joint pathology and illustrate differences in bone dynamics between mouse strains.


Subject(s)
Bone Diseases/etiology , Bone Diseases/pathology , Chikungunya Fever/complications , Chikungunya Fever/virology , Chikungunya virus , Joint Diseases/etiology , Joint Diseases/pathology , Animals , Biopsy , Bone Diseases/diagnosis , Disease Models, Animal , Disease Progression , Female , Inflammation/etiology , Inflammation/pathology , Interferon Regulatory Factor-3/deficiency , Interferon Regulatory Factor-7/deficiency , Joint Diseases/diagnosis , Male , Mice , Mice, Knockout , Necrosis/etiology , Necrosis/pathology , Phenotype , X-Ray Microtomography
8.
PLoS One ; 10(10): e0139481, 2015.
Article in English | MEDLINE | ID: mdl-26447467

ABSTRACT

Chikungunya virus (CHIKV) is a reemerging, ordinarily mosquito-transmitted, alphavirus that occasionally produces hemorrhagic manifestations, such as nose bleed and bleeding gums, in human patients. Interferon response factor 3 and 7 deficient (IRF3/7-/-) mice, which are deficient for interferon α/ß responses, reliably develop hemorrhagic manifestations after CHIKV infection. Here we show that infectious virus was present in the oral cavity of CHIKV infected IRF3/7-/- mice, likely due to hemorrhagic lesions in the olfactory epithelium that allow egress of infected blood into the nasal, and subsequently, oral cavities. In addition, IRF3/7-/- mice were more susceptible to infection with CHIKV via intranasal and oral routes, with IRF3/7-/- mice also able to transmit virus mouse-to-mouse without an arthropod vector. Cynomolgus macaques often show bleeding gums after CHIKV infection, and analysis of saliva from several infected monkeys also revealed the presence of viral RNA and infectious virus. Furthermore, saliva samples collected from several acute CHIKV patients with hemorrhagic manifestations were found to contain viral RNA and infectious virus. Oral fluids can therefore be infectious during acute CHIKV infections, likely due to hemorrhagic manifestations in the oral/nasal cavities.


Subject(s)
Chikungunya Fever/pathology , Chikungunya virus/genetics , Saliva/virology , Adolescent , Animals , Chikungunya Fever/transmission , Chikungunya Fever/virology , Chikungunya virus/isolation & purification , Child , Disease Models, Animal , Female , Haplorhini , Humans , Interferon Regulatory Factor-3/deficiency , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-7/deficiency , Interferon Regulatory Factor-7/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Olfactory Mucosa/pathology , Olfactory Mucosa/virology , RNA, Viral/metabolism , Viral Load
10.
J Exp Med ; 212(9): 1371-9, 2015 Aug 24.
Article in English | MEDLINE | ID: mdl-26216125

ABSTRACT

Herpes simplex encephalitis (HSE) in children has previously been linked to defects in type I interferon (IFN) production downstream of Toll-like receptor 3. Here, we describe a novel genetic etiology of HSE by identifying a heterozygous loss-of-function mutation in the IFN regulatory factor 3 (IRF3) gene, leading to autosomal dominant (AD) IRF3 deficiency by haploinsufficiency, in an adolescent female patient with HSE. IRF3 is activated by most pattern recognition receptors recognizing viral infections and plays an essential role in induction of type I IFN. The identified IRF3 R285Q amino acid substitution results in impaired IFN responses to HSV-1 infection and particularly impairs signaling through the TLR3-TRIF pathway. In addition, the R285Q mutant of IRF3 fails to become phosphorylated at S386 and undergo dimerization, and thus has impaired ability to activate transcription. Finally, transduction with WT IRF3 rescues the ability of patient fibroblasts to express IFN in response to HSV-1 infection. The identification of IRF3 deficiency in HSE provides the first description of a defect in an IFN-regulating transcription factor conferring increased susceptibility to a viral infection in the CNS in humans.


Subject(s)
Encephalitis, Herpes Simplex/genetics , Fibroblasts/metabolism , Haploinsufficiency , Herpesvirus 1, Human/metabolism , Interferon Regulatory Factor-3/deficiency , Mutation, Missense , Adolescent , Amino Acid Substitution , Encephalitis, Herpes Simplex/metabolism , Encephalitis, Herpes Simplex/pathology , Female , Fibroblasts/pathology , Fibroblasts/virology , Herpesvirus 1, Human/genetics , Humans , Interferon Regulatory Factor-3/metabolism , Interferon Type I/biosynthesis , Interferon Type I/genetics , Phosphorylation , Protein Multimerization/genetics
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.
J Immunol ; 194(7): 3236-45, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25710914

ABSTRACT

Type I IFN production is an important host immune response against viral and bacterial infections. However, little is known about the ligands and corresponding host receptors that trigger type I IFN production during bacterial infections. We used a model intracellular pathogen, Francisella novicida, to begin characterizing the type I IFN response to bacterial pathogens. F. novicida replicates in the cytosol of host cells and elicits a robust type I IFN response that is largely TLR independent, but is dependent on the adapter molecule STING, suggesting that the type I IFN stimulus during F. novicida infection is cytosolic. In this study, we report that the cytosolic DNA sensors, cyclic GMP-AMP synthase (cGAS) and Ifi204, are both required for the STING-dependent type I IFN response to F. novicida infection in both primary and immortalized murine macrophages. We created cGAS, Ifi204, and Sting functional knockouts in RAW264.7 macrophages and demonstrated that cGAS and Ifi204 cooperate to sense dsDNA and activate the STING-dependent type I IFN pathway. In addition, we show that dsDNA from F. novicida is an important type I IFN stimulating ligand. One outcome of cGAS-STING signaling is the activation of the absent in melanoma 2 inflammasome in response to F. novicida infection. Whereas the absent in melanoma 2 inflammasome is beneficial to the host during F. novicida infection, type I IFN signaling by STING and IFN regulatory factor 3 is detrimental to the host during F. novicida infection. Collectively, our studies indicate that cGAS and Ifi204 cooperate to sense cytosolic dsDNA and F. novicida infection to produce a strong type I IFN response.


Subject(s)
Francisella/immunology , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/metabolism , Interferon Type I/metabolism , Nuclear Proteins/metabolism , Nucleotidyltransferases/metabolism , Phosphoproteins/metabolism , Animals , Bone Marrow Cells/metabolism , Cell Line , Cytosol/immunology , Cytosol/metabolism , DNA/immunology , Disease Models, Animal , Gene Expression , Gene Expression Regulation , Gram-Negative Bacterial Infections/genetics , Gram-Negative Bacterial Infections/mortality , Inflammasomes/metabolism , Interferon Regulatory Factor-3/deficiency , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Macrophages/immunology , Macrophages/metabolism , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Nuclear Proteins/genetics , Nucleotidyltransferases/genetics , Phosphoproteins/genetics , Protein Binding , RNA Interference , Signal Transduction
13.
J Neuroinflammation ; 11: 130, 2014 Jul 28.
Article in English | MEDLINE | ID: mdl-25069698

ABSTRACT

BACKGROUND: Experimental autoimmune encephalomyelitis (EAE) is an animal model of autoimmune inflammatory demyelination that is mediated by Th1 and Th17 cells. The transcription factor interferon regulatory factor 3 (IRF3) is activated by pathogen recognition receptors and induces interferon-ß production. METHODS: To determine the role of IRF3 in autoimmune inflammation, we immunised wild-type (WT) and irf3(-/-) mice to induce EAE. Splenocytes from WT and irf3(-/-) mice were also activated in vitro in Th17-polarising conditions. RESULTS: Clinical signs of disease were significantly lower in mice lacking IRF3, with reduced Th1 and Th17 cells in the central nervous system. Peripheral T-cell responses were also diminished, including impaired proliferation and Th17 development in irf3(-/-) mice. Myelin-reactive CD4+ cells lacking IRF3 completely failed to transfer EAE in Th17-polarised models as did WT cells transferred into irf3(-/-) recipients. Furthermore, IRF3 deficiency in non-CD4+ cells conferred impairment of Th17 development in antigen-activated cultures. CONCLUSION: These data show that IRF3 plays a crucial role in development of Th17 responses and EAE and warrants investigation in human multiple sclerosis.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Interferon Regulatory Factor-3/deficiency , Animals , CD4-Positive T-Lymphocytes/pathology , Cell Proliferation/drug effects , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Flow Cytometry , Interferon Regulatory Factor-3/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin-Oligodendrocyte Glycoprotein/toxicity , Peptide Fragments/toxicity , Phorbol Esters/pharmacology , Spinal Cord/pathology , T-Lymphocytes/drug effects , Th17 Cells/drug effects , Th17 Cells/metabolism , Transfection
14.
PLoS Negl Trop Dis ; 8(6): e2933, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24901990

ABSTRACT

The new world arenavirus Junín virus (JUNV) is the causative agent of Argentine hemorrhagic fever, a lethal human infectious disease. Adult laboratory mice are generally resistant to peripheral infection by JUNV. The mechanism underlying the mouse resistance to JUNV infection is largely unknown. We have reported that interferon receptor knockout mice succumb to JUNV infection, indicating the critical role of interferon in restricting JUNV infection in mice. Here we report that the pathogenic and vaccine strains of JUNV were highly sensitive to interferon in murine primary cells. Treatment with low concentrations of interferon abrogated viral NP protein expression in murine cells. The replication of both JUNVs was enhanced in IRF3/IRF7 deficient cells. In addition, the vaccine strain of JUNV displayed impaired growth in primary murine cells. Our data suggested a direct and potent role of host interferon response in restricting JUNV replication in mice. The defect in viral growth for vaccine JUNV might also partially explain its attenuation in mice.


Subject(s)
Antiviral Agents/pharmacology , Interferons/immunology , Interferons/pharmacology , Junin virus/drug effects , Junin virus/immunology , Animals , Cells, Cultured , Interferon Regulatory Factor-3/deficiency , Interferon Regulatory Factor-7/deficiency , Interferons/deficiency , Junin virus/growth & development , Junin virus/physiology , Mice, Inbred C57BL , Mice, Knockout , Viral Proteins/biosynthesis , Virus Replication/drug effects
15.
Mol Immunol ; 57(2): 100-10, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24096085

ABSTRACT

Measles virus (MV) infects CD150Tg/Ifnar (IFN alpha receptor)(-/-) mice but not CD150 (a human MV receptor)-transgenic (Tg) mice. We have shown that bone marrow-derived dendritic cells (BMDCs) from CD150Tg/Ifnar(-/-) mice are permissive to MV in contrast to those from simple CD150Tg mice, which reveals a crucial role of type I interferon (IFN) in natural tropism against MV. Yet, the mechanism whereby BMDCs produce initial type I IFN has not been elucidated in MV infection. RNA virus infection usually allows cells to generate double-stranded RNA and induce activation of IFN regulatory factor (IRF) 3/7 transcription factors, leading to the production of type I IFN through the retinoic acid-inducible gene I (RIG-I)/melanoma differentiation-associated gene 5 (MDA5)-mitochondrial antiviral signaling protein (MAVS) pathway. In mouse experimental BMDCs models, we found CD150Tg/Mavs(-/-)BMDCs, but not CD150Tg/Irf3(-/-)/Irf7(-/-)BMDCs, permissive to MV. IFN-α/ß were not induced in MV-infected CD150Tg/Mavs(-/-)BMDCs, while IFN-ß was subtly induced in CD150Tg/Irf3(-/-)/Irf7(-/-)BMDCs. In vivo systemic infection was therefore established by transfer of MV-infected CD150Tg/Mavs(-/-) BMDCs to CD150Tg/Ifnar(-/-) mice. These data indicate that MAVS-dependent, IRF3/7-independent IFN-ß induction triggers the activation of the IFNAR pathway so as to restrict the spread of MV by infected BMDCs. Hence, MAVS participates in the initial induction of type I IFN in BMDCs and IFNAR protects against MV spreading. We also showed the importance of IL-10-producing CD4(+) T cells induced by MV-infected BMDCs in vitro, which may account for immune modulation due to the functional aberration of DCs.


Subject(s)
Dendritic Cells/immunology , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-7/genetics , Interferon-beta/metabolism , Measles/immunology , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Vesicular Transport/deficiency , Adaptor Proteins, Vesicular Transport/genetics , Animals , Antigens, CD/metabolism , Bone Marrow Cells/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Line , Chlorocebus aethiops , DEAD Box Protein 58 , DEAD-box RNA Helicases/metabolism , Dendritic Cells/cytology , Interferon Regulatory Factor-3/deficiency , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-7/deficiency , Interferon Regulatory Factor-7/metabolism , Interferon-Induced Helicase, IFIH1 , Interferon-beta/biosynthesis , Interleukin-10/metabolism , Measles virus/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Interferon alpha-beta/genetics , Receptors, Cell Surface/metabolism , Signal Transduction/immunology , Signaling Lymphocytic Activation Molecule Family Member 1 , Vero Cells
16.
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
17.
J Immunol ; 191(8): 4194-201, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24043884

ABSTRACT

We investigated the roles of IFN regulatory factor (IRF)-3 and IRF-7 in innate antiviral immunity against dengue virus (DENV). Double-deficient Irf-3(-/-)7(-/-) mice infected with the DENV2 strain S221 possessed 1,000-150,000 fold higher levels of viral RNA than wild-type and single-deficient mice 24 h postinfection (hpi); however, they remained resistant to lethal infection. IFN-α/ß was induced similarly in wild-type and Irf-3(-/-) mice post-DENV infection, whereas in the Irf-7(-/-) and Irf-3(-/-)7(-/-) mice, significantly low levels of IFN-α/ß expression was observed within 24 hpi. IFN-stimulated gene induction was also delayed in Irf-3(-/-)7(-/-) mice relative to wild-type and single-deficient mice. In particular, Cxcl10 and Ifnα2 were rapidly induced independently of both IRF-3 and IRF-7 in the Irf-3(-/-)7(-/-) mice with DENV infection. Higher levels of serum IFN-γ, IL-6, CXCL10, IL-8, IL-12 p70, and TNF were also observed in Irf-3(-/-)7(-/-) mice 24 hpi, at which time point viral titers peaked and started to be cleared. Ab-mediated blockade experiments revealed that IFN-γ, CXCL10, and CXCR3 function to restrict DENV replication in Irf-3(-/-)7(-/-) mice. Additionally, the IFN-stimulated genes Cxcl10, Ifit1, Ifit3, and Mx2 can be induced via an IRF-3- and IRF-7-independent pathway that does not involve IFN-γ signaling for protection against DENV. Collectively, these results demonstrate that IRF-3 and IRF-7 are redundant, albeit IRF-7 plays a more important role than IRF-3 in inducing the initial IFN-α/ß response; only the combined actions of IRF-3 and IRF-7 are necessary for efficient control of early DENV infection; and the late, IRF-3- and IRF-7-independent pathway contributes to anti-DENV immunity.


Subject(s)
Dengue Virus/immunology , Dengue/immunology , Immunity, Innate , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-7/metabolism , Interferon-alpha/blood , Interferon-beta/blood , Adaptor Proteins, Signal Transducing , Aedes , Animals , Carrier Proteins/metabolism , Cell Line , Chemokine CXCL10/biosynthesis , Chemokine CXCL10/blood , Chemokine CXCL10/immunology , Interferon Regulatory Factor-3/deficiency , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-7/deficiency , Interferon Regulatory Factor-7/genetics , Interferon-gamma/blood , Interferon-gamma/immunology , Interleukin-12/blood , Interleukin-6/blood , Interleukin-8/blood , Intracellular Signaling Peptides and Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Myxovirus Resistance Proteins/metabolism , Proteins/metabolism , RNA, Viral/blood , RNA-Binding Proteins , Receptors, CXCR3/immunology , Signal Transduction , Tumor Necrosis Factors/blood , Viral Load , Virus Replication/immunology
18.
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
19.
Cell Rep ; 3(6): 1840-6, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23770239

ABSTRACT

Although molecular components that execute noninflammatory apoptotic cell death are well defined, molecular pathways that trigger necrotic cell death remain poorly characterized. Here, we show that in response to infection with adenovirus or Listeria monocytogenes, macrophages in vivo undergo rapid proinflammatory necrotic death that is controlled by interferon-regulatory factor 3 (IRF3). The transcriptional activity of IRF3 is, surprisingly, not required for the induction of necrosis, and it proceeds normally in mice deficient in all known regulators of necrotic death or IRF3 activation, including RIPK3, caspases 1, 8, or 11, STING, and IPS1/MAVS. Although L. monocytogenes triggers necrosis to promote the infection, IRF3-dependent necrosis is required for reducing pathogen burden in the models of disseminated infection with adenovirus. Therefore, our studies implicate IRF3 as a principal and nonredundant component of a physiologically regulated necrotic cell-death pathway that operates as an effective innate immune mechanism of host protection against disseminated virus infection.


Subject(s)
Adenovirus Infections, Human/immunology , Adenoviruses, Human/immunology , Interferon Regulatory Factor-3/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Macrophages/microbiology , Macrophages/pathology , Adenovirus Infections, Human/pathology , Animals , Caspases/metabolism , Immunity, Innate/immunology , Interferon Regulatory Factor-3/deficiency , Interferon Regulatory Factor-3/genetics , Listeriosis/pathology , Macrophages/immunology , Macrophages/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Necrosis/immunology , Necrosis/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction
20.
J Immunol ; 189(6): 2860-8, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22896628

ABSTRACT

Type I IFNs are induced by pathogens to protect the host from infection and boost the immune response. We have recently demonstrated that this IFN response is not restricted to pathogens, as the Gram-positive bacterium Lactobacillus acidophilus, a natural inhabitant of the intestine, induces high levels of IFN-ß in dendritic cells. In the current study, we investigate the intracellular pathways involved in IFN-ß upon stimulation of dendritic cells with L. acidophilus and reveal that this IFN-ß induction requires phagosomal uptake and processing but bypasses the endosomal receptors TLR7 and TLR9. The IFN-ß production is fully dependent on the TIR adapter molecule MyD88, partly dependent on IFN regulatory factor (IRF)1, but independent of the TIR domain-containing adapter inducing IFN-ß MyD88 adapter-like, IRF and IRF7. However, our results suggest that IRF3 and IRF7 have complementary roles in IFN-ß signaling. The IFN-ß production is strongly impaired by inhibitors of spleen tyrosine kinase (Syk) and PI3K. Our results indicate that L. acidophilus induces IFN-ß independently of the receptors typically used by bacteria, as it requires MyD88, Syk, and PI3K signaling and phagosomal processing to activate IRF1 and IRF3/IRF7 and thereby the release of IFN-ß.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/microbiology , Interferon Regulatory Factor-1/physiology , Interferon Regulatory Factor-3/physiology , Interferon Regulatory Factor-7/physiology , Interferon-beta/metabolism , Lactobacillus acidophilus/immunology , Myeloid Differentiation Factor 88/physiology , Animals , Cells, Cultured , Dendritic Cells/metabolism , Endosomes/immunology , Endosomes/metabolism , Endosomes/microbiology , Interferon Regulatory Factor-1/deficiency , Interferon Regulatory Factor-3/antagonists & inhibitors , Interferon Regulatory Factor-3/deficiency , Interferon Regulatory Factor-7/antagonists & inhibitors , Interferon Regulatory Factor-7/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/deficiency , Phagosomes/immunology , Phagosomes/metabolism , Protein Processing, Post-Translational/immunology , Signal Transduction/genetics , Signal Transduction/immunology
SELECTION OF CITATIONS
SEARCH DETAIL