Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
J Gen Virol ; 102(10)2021 10.
Article in English | MEDLINE | ID: mdl-34661517

ABSTRACT

Rabies is a zoonotic disease caused by the rabies virus (RABV). RABV can lead to fatal encephalitis and is still a serious threat in most parts of the world. Interferon regulatory factor 7 (IRF7) is the main transcriptional regulator of type I IFN, and it is crucial for the induction of IFNα/ß and the type I IFN-dependent immune response. In this study, we focused on the role of IRF7 in the pathogenicity and immunogenicity of RABV using an IRF7-/- mouse model. The results showed that the absence of IRF7 made mice more susceptible to RABV, because IRF7 restricted the replication of RABV in the early stage of infection. IRF7 deficiency affected the recruitment of plasmacytoid dendritic cells to the draining lymph nodes (dLNs), reduced the production of type I IFN and expression of IFN-stimulated genes. Furthermore, we found that the ability to produce specific RABV-neutralizing antibody was impaired in IRF7-/- mice. Consistently, IRF7 deficiency affected the recruitment of germinal-centre B cells to dLNs, and the generation of plasma cells and RABV-specific antibody secreting cells. Moreover, the absence of IRF7 downregulated the induction of IFN-γ and reduced type 1 T helper cell (Th1)-dependent antibody production. Collectively, our findings demonstrate that IRF7 promotes humoral immune responses and compromises the pathogenicity of RABV in a mouse model.


Subject(s)
Interferon Regulatory Factor-7/physiology , Rabies virus/immunology , Rabies virus/pathogenicity , Rabies/immunology , Rabies/virology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Antibody-Producing Cells/immunology , B-Lymphocytes/immunology , Cell Line , Dendritic Cells/immunology , Disease Models, Animal , Female , Immunity, Humoral , Interferon Regulatory Factor-7/deficiency , Interferon Regulatory Factor-7/genetics , Interferons/analysis , Lymph Nodes/immunology , Male , Mice , Mice, Inbred C57BL , Rabies Vaccines/immunology , Th1 Cells/immunology , Viral Load
2.
Infect Immun ; 89(11): e0040721, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34370509

ABSTRACT

During chronic infection with Helicobacter pylori, Schlafen 4-expressing myeloid-derived suppressor cells (SLFN4+ MDSCs) create a microenvironment favoring intestinal metaplasia and neoplastic transformation. SLFN4 can be induced by alpha interferon (IFN-α), which is mainly secreted from plasmacytoid dendritic cells (pDCs). This study tested the hypothesis that Helicobacter pylori infection promotes SLFN4+ MDSC differentiation by inducing pDCs to secrete IFN-α. C57BL/6 mice were gavaged with H. pylori, and infection lasted 2, 4, or 6 months. Mouse pDCs were isolated from bone marrow of wild-type C57BL/6J mice. The results showed that H. pylori infection increased the number of SLFN4+ MDSCs by inducing IFN-α expression in mice. Further mechanistic experiments unraveled that IFN-α induced SLFN4 transcription by binding to the Slfn4 promoter. Furthermore, H. pylori infection stimulated pDCs to secrete IFN-α by activating the TLR9-MyD88-IRF7 pathway. Collectively, Helicobacter pylori infection promotes SLFN4+ MDSC differentiation by inducing secretion of IFN-α from pDCs.


Subject(s)
Carrier Proteins/genetics , Dendritic Cells/immunology , Helicobacter Infections/immunology , Helicobacter pylori , Interferon Type I/biosynthesis , Myeloid-Derived Suppressor Cells/cytology , Animals , Cell Differentiation , Interferon Regulatory Factor-7/physiology , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/physiology , Promoter Regions, Genetic , Toll-Like Receptor 9/physiology
3.
Int J Mol Sci ; 22(16)2021 Aug 18.
Article in English | MEDLINE | ID: mdl-34445616

ABSTRACT

Neovascularization restores blood flow recovery after ischemia in peripheral arterial disease. The main two components of neovascularization are angiogenesis and arteriogenesis. Both of these processes contribute to functional improvements of blood flow after occlusion. However, discriminating between the specific contribution of each process is difficult. A frequently used model for investigating neovascularization is the murine hind limb ischemia model (HLI). With this model, it is difficult to determine the role of angiogenesis, because usually the timing for the sacrifice of the mice is chosen to be optimal for the analysis of arteriogenesis. More importantly, the occurring angiogenesis in the distal calf muscles is probably affected by the proximally occurring arteriogenesis. Therefore, to understand and subsequently intervene in the process of angiogenesis, a model is needed which investigates angiogenesis without the influence of arteriogenesis. In this study we evaluated the in vivo Matrigel plug assay in genetic deficient mice to investigate angiogenesis. Mice deficient for interferon regulatory factor (IRF)3, IRF7, RadioProtective 105 (RP105), Chemokine CC receptor CCR7, and p300/CBP-associated factor (PCAF) underwent the in vivo Matrigel model. Histological analysis of the Matrigel plugs showed an increased angiogenesis in mice deficient of IRF3, IRF7, and RP105, and a decreased angiogenesis in PCAF deficient mice. Our results also suggest an involvement of CCR7 in angiogenesis. Comparing our results with results of the HLI model found in the literature suggests that the in vivo Matrigel plug assay is superior in evaluating the angiogenic response after ischemia.


Subject(s)
Antigens, CD/physiology , Hindlimb/blood supply , Interferon Regulatory Factor-3/physiology , Interferon Regulatory Factor-7/physiology , Ischemia/pathology , Neovascularization, Pathologic/pathology , p300-CBP Transcription Factors/physiology , Animals , Collagen , Drug Combinations , Hindlimb/pathology , Ischemia/metabolism , Laminin , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic/metabolism , Proteoglycans , Recovery of Function
4.
Cell Biol Int ; 45(4): 831-838, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33325089

ABSTRACT

Breast cancer is a kind of malignant tumor that severely threatens women's lives and health worldwide. Tumor-associated macrophages (TAMs) have been reported to mediate tumor progression, while the mechanism still needs further identification. In this study, we found that M2 macrophages promoted increased cell proliferation and migration as well as reduced expression of interferon regulatory factor 7 (IRF7) and increased the expression of miR-1587 in breast cancer cells. Overexpression of IRF7 or miR-1587 knockdown reversed M2 macrophage-induced cell proliferation and migration as well as tumor growth in vivo. Mechanistically, miR-1587 targeted the 3'-untranslated region (3'-UTR) of IRF7 mRNA to regulate its protein expression leading to tumor progression. Collectively, this study revealed that the miR-1587/IRF7 axis mediates M2 macrophage-induced breast cancer progression, and this sheds light on further clinical therapy for breast cancer by targeting TAMs as well as the miR-1587/IRF7 axis.


Subject(s)
Breast Neoplasms , Gene Expression Regulation, Neoplastic , Interferon Regulatory Factor-7/physiology , Macrophages/immunology , MicroRNAs/physiology , Animals , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Humans , Macrophages/cytology , Mice , Mice, Inbred BALB C , Mice, Nude
5.
PLoS Pathog ; 16(10): e1008973, 2020 10.
Article in English | MEDLINE | ID: mdl-33045014

ABSTRACT

The liver is a central regulator of metabolic homeostasis and serum metabolite levels. Hepatocytes are the functional units of the liver parenchyma and not only responsible for turnover of biomolecules but also act as central immune signaling platforms. Hepatotropic viruses infect liver tissue, resulting in inflammatory responses, tissue damage and hepatitis. Combining well-established in vitro and in vivo model systems with transcriptomic analyses, we show that type I interferon signaling initiates a robust antiviral immune response in hepatocytes. Strikingly, we also identify IFN-I as both, sufficient and necessary, to induce wide-spread metabolic reprogramming in hepatocytes. IFN-I specifically rewired tryptophan metabolism and induced hepatic tryptophan oxidation to kynurenine via Tdo2, correlating with altered concentrations of serum metabolites upon viral infection. Infected Tdo2-deficient animals displayed elevated serum levels of tryptophan and, unexpectedly, also vast increases in the downstream immune-suppressive metabolite kynurenine. Thus, Tdo2-deficiency did not result in altered serum homeostasis of the tryptophan to kynurenine ratio during infection, which seemed to be independent of hepatocyte-intrinsic compensation via the IDO-axis. These data highlight that inflammation-induced reprogramming of systemic tryptophan metabolism is tightly regulated in viral hepatitis.


Subject(s)
Antiviral Agents/metabolism , Hepatitis, Viral, Animal/immunology , Hepatocytes/immunology , Inflammation/immunology , Kynurenine/metabolism , Receptor, Interferon alpha-beta/physiology , Tryptophan/metabolism , Animals , Female , Hepatitis Viruses/isolation & purification , Hepatitis, Viral, Animal/metabolism , Hepatitis, Viral, Animal/virology , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Immunity, Innate/immunology , Inflammation/metabolism , Inflammation/pathology , Inflammation/virology , Interferon Regulatory Factor-7/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , STAT1 Transcription Factor/physiology , Tryptophan Oxygenase/physiology
6.
Eur J Immunol ; 50(8): 1142-1153, 2020 08.
Article in English | MEDLINE | ID: mdl-32135578

ABSTRACT

Exposure to diesel exhaust particles (DEPs) is associated with acute inflammatory responses in the lung and exacerbation of respiratory diseases. However, the mechanism by which DEPs trigger the inflammatory responses remains unclear. Here, we demonstrated that the IFN response factors IRF3 and IRF7 played pivotal roles in DEP-induced pulmonary inflammation. DEPs could not directly induce inflammatory cytokine expression in mouse cells, whereas DEPs triggered autophagy both in vitro and in vivo. The DEP-induced autophagy was augmented in the absence of IRF3 and IRF7, but not in the absence of IFNAR. The expression of Raptor was induced by IRF3 and IRF7 in response to DEPs treatment. Furthermore, administration of the mechanistic target of rapamycin (mTOR) inhibitor alleviated the inflammatory responses in the lung during DEP exposure. Our findings define an IFNAR-independent role of increased autophagy in the absence of IRF3 and IRF7 during pulmonary DEP exposure, and provide the basis to develop new therapeutic approaches to counteract the adverse effects of DEPs and possibly other ambient particulate matters.


Subject(s)
Autophagy/physiology , Interferon Regulatory Factor-3/physiology , Interferon Regulatory Factor-7/physiology , Mechanistic Target of Rapamycin Complex 1/physiology , Pneumonia/etiology , Vehicle Emissions/toxicity , Animals , Cytokines/biosynthesis , Mice , Mice, Inbred C57BL , Receptor, Interferon alpha-beta/physiology , Sirolimus/pharmacology
7.
Vet Microbiol ; 221: 74-80, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29981711

ABSTRACT

Chicken melanoma differentiation-associated gene 5 (chMDA5) is a key pattern recognition receptor (PRR) that recognizes RNA viral infections and initiates an antiviral innate immune response in chickens. MicroRNAs (miRNAs) are involved in the regulation of chMDA5 to sense RNA virus infection, but how it exerts antiviral activity against infectious bursal disease virus (IBDV) infection and regulates chMDA5 in chicken cells is unclear. Thus, we measured the expression of chMDA5 in IBDV-infected DT40 cells and found it significantly increased. Overexpression of chMDA5 activated the IFN-ß and Mx promoters via IRF7-dependent pathways and inhibited replication of IBDV in DT40 cells. The opposite effect occurred after chMDA5 knockdown using siRNA. Also, gga-miR-142-5p regulated chMDA5 according to bioinformatic analysis and data from a dual-luciferase reporter system. Overexpression of gga-miR-142-5p reduced the expression of the chMDA5 protein, promoting IBDV replication, and decreased the activity of the IFN-ß and Mx promoters via an IRF7-dependent pathway; however, it had no effect on the NF-κB-dependent pathway in DT40 cells. Thus, gga-miR-142-5p is a negative regulator of chMDA5 and promotes IBDV replication in DT40 cells through an IRF7-dependent pathway.


Subject(s)
Immunity, Innate , Infectious bursal disease virus/physiology , Interferon Regulatory Factor-7/physiology , Virus Replication/physiology , Animals , B-Lymphocytes/physiology , Cell Line , Chickens , RNA Interference
8.
Oncogene ; 36(21): 2969-2980, 2017 05 25.
Article in English | MEDLINE | ID: mdl-28092673

ABSTRACT

Accumulation of myeloid-derived suppressor cells (MDSCs) is one of the major obstacles against achieving appropriate anti-tumor immune responses and successful tumor immunotherapy. Granulocytic MDSCs (G-MDSCs) are common in tumor-bearing hosts. However, the mechanisms regulating the development of MDSCs, especially G-MDSCs, remain poorly understood. In this report, we showed that interferon regulatory factor 7 (IRF7) plays an important role in the development of G-MDSCs, but not monocytic MDSCs. IRF7 deficiency caused significant elevation of G-MDSCs, and therefore enhanced tumor growth and metastasis in mice. IRF7 deletion did not affect the suppressive activity of G-MDSCs. Mechanistic studies showed that S100A9, a negative regulator of myeloid cell differentiation, was transrepressed by the IRF7 protein. S100A9 knockdown almost completely abrogated the effects of IRF7 deletion on G-MDSC development and tumor metastasis. Importantly, IRF7 expression levels negatively correlated with the G-MDSC frequency and tumor metastasis, as well as S100A9 expression, in cancer patients. In summary, our study demonstrated that IRF7 represents a novel regulator of G-MDSC development in cancer, which may have predictive value for tumor progression.


Subject(s)
Calgranulin B/genetics , Cell Differentiation/genetics , Granulocytes/physiology , Interferon Regulatory Factor-7/physiology , Myeloid-Derived Suppressor Cells/physiology , Neoplasms/genetics , Animals , Biomarkers, Tumor/genetics , Cell Differentiation/immunology , Cell Line, Tumor , Down-Regulation/genetics , Gene Expression Regulation, Neoplastic , Humans , Immunotherapy , Interferon Regulatory Factor-7/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/diagnosis , Neoplasms/immunology , Neoplasms/pathology , Prognosis , Signal Transduction/genetics , Tumor Escape/genetics
9.
J Immunol ; 197(6): 2167-76, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27527596

ABSTRACT

Systemic lupus erythematosus (SLE) is a prototypic systemic autoimmune disease characterized by the production of autoantibodies against nuclear components. Recent genetic studies of SLE patients have revealed that IFN regulatory factor (IRF) 7 gene polymorphisms are associated with an increased risk of SLE, but the precise role of IRF7 in SLE development is not fully understood. We investigated the role of IRF7 in the pathogenesis of SLE using a mouse model and saw a curious dissociation of autoantibody production and development of glomerulonephritis. SLE was chemically induced into IRF7-deficient mice, and glomerulonephritis with deposits of IgG and lipogranulomas were observed after 10 mo. However, these mice failed to produce anti-dsDNA, ssDNA, ribonucleoprotein, and Sm autoantibodies. Following the chemical induction, IRF7-deficient mice expressed substantially lower levels of IFN-stimulated genes than did wild-type mice, but NF-κB target genes were equally upregulated in both strains. Therefore, the type I IFN pathway seems critical for the autoantibody production, but the NF-κB activation is sufficient for the development of glomerulonephritis in this model. Our study thus demonstrates a specific requirement for IRF7 in autoantibody production and uncovers a new layer of complexity in the pathogenesis of SLE.


Subject(s)
Autoantibodies/biosynthesis , Interferon Regulatory Factor-7/physiology , Lupus Nephritis/etiology , Animals , Antibodies, Antinuclear/biosynthesis , Apoptosis/drug effects , Dendritic Cells/physiology , Female , Glomerulonephritis/etiology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/physiology , Neutrophils/physiology , Picolines/pharmacology
10.
J Biol Chem ; 291(25): 13206-15, 2016 Jun 17.
Article in English | MEDLINE | ID: mdl-27129230

ABSTRACT

Viral infection or lipopolysaccharide (LPS) treatment induces expression of a large array of genes, the products of which play a critical role in host antipathogen immunity and inflammation. We have previously reported that the expression of ubiquitin-specific protease 25 (USP25) is significantly up-regulated after viral infection or LPS treatment, and this is essential for innate immune signaling. However, the mechanism behind this phenomenon is unclear. In this study, we found that viral infection-induced up-regulation of Usp25 is diminished in cells lacking interferon regulatory factor 7 (IRF7) or interferon α receptor 1 (IFNAR1) but not p65. Sendai virus- or type I interferon-induced up-regulation of Usp25 requires de novo protein synthesis of IRF7. Furthermore, IRF7 directly binds to the two conserved IRF binding sites on the USP25 promoter to drive transcription of Usp25, and mutation of these two sites abolished Sendai virus-induced IRF7-mediated activation of the USP25 promoter. Our study has uncovered a previously unknown mechanism by which viral infection or LPS induces up-regulation of USP25.


Subject(s)
Interferon Regulatory Factor-7/physiology , Interferon Type I/physiology , Ubiquitin Thiolesterase/genetics , Animals , Cells, Cultured , Enzyme Induction/immunology , Herpes Simplex/enzymology , Herpesvirus 1, Human/physiology , Lipopolysaccharides/pharmacology , Mice, Knockout , Promoter Regions, Genetic , Protein Binding , Signal Transduction , Transcription, Genetic , Ubiquitin Thiolesterase/metabolism , Up-Regulation/immunology
11.
Eur J Immunol ; 44(7): 2153-64, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24777946

ABSTRACT

Interleukin-33 (IL-33), an IL-1 family cytokine and nuclear alarmin, is constitutively expressed in epithelial barrier tissues and human blood vessels. However, little is known about the induced expression of IL-33 in monocytes and macrophages, which are major cytokine-producing cells of the innate immune system. Here, we report the induction of IL33 expression in both human monocytes and mouse macrophages from C57BL/6 mice by the acute-phase protein serum amyloid A (SAA). SAA-induced transcriptional activation of the Il33 gene, resulting in nuclear accumulation of the IL-33 protein. TLR2, one of the SAA receptors, was primarily responsible for the induction of IL-33. Progressive deletion of the human IL-33 promoter led to the identification of two potential binding sites for interferon regulatory factor 7 (IRF7), one of which (-277/-257) was found to be important for SAA-stimulated IL-33 promoter activity. IRF7 was recruited to the IL-33 promoter upon SAA stimulation, and silencing IRF7 expression in THP-1 cells abrogated SAA-induced Il33 expression. SAA also promoted an interaction between TNF receptor-associated factor 6 and IRF7. Taken together, these results identify IRF7 as a critical transcription factor for SAA-induced Il33 expression in monocytes and macrophages.


Subject(s)
Interferon Regulatory Factor-7/physiology , Interleukins/genetics , Serum Amyloid A Protein/pharmacology , Animals , Base Sequence , Cells, Cultured , Interleukin-33 , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Promoter Regions, Genetic , RNA, Messenger/analysis , Signal Transduction , TNF Receptor-Associated Factor 6/physiology , Toll-Like Receptor 2/physiology
13.
Hypertension ; 63(4): 713-22, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24396025

ABSTRACT

Cardiac hypertrophy is a complex pathological process that involves multiple factors including inflammation and apoptosis. Interferon regulatory factor 7 (IRF7) is a multifunctional regulator that participates in immune regulation, cell differentiation, apoptosis, and oncogenesis. However, the role of IRF7 in cardiac hypertrophy remains unclear. We performed aortic banding in cardiac-specific IRF7 transgenic mice, IRF7 knockout mice, and the wild-type littermates of these mice. Our results demonstrated that IRF7 was downregulated in aortic banding-induced animal hearts and cardiomyocytes that had been treated with angiotensin II or phenylephrine for 48 hours. Accordingly, heart-specific overexpression of IRF7 significantly attenuated pressure overload-induced cardiac hypertrophy, fibrosis, and dysfunction, whereas loss of IRF7 led to opposite effects. Moreover, IRF7 protected against angiotensin II-induced cardiomyocyte hypertrophy in vitro. Mechanistically, we identified that IRF7-dependent cardioprotection was mediated through IRF7 binding to inhibitor of κB kinase-ß, and subsequent nuclear factor-κB inactivation. In fact, blocking nuclear factor-κB signaling with cardiac-specific inhibitors of κBα(S32A/S36A) super-repressor transgene counteracted the adverse effect of IRF7 deficiency. Conversely, activation of nuclear factor-κB signaling via a cardiac-specific conditional inhibitor of κB kinase-ß(S177E/S181E) (constitutively active) transgene negated the antihypertrophic effect of IRF7 overexpression. Our data demonstrate that IRF7 acts as a novel negative regulator of pathological cardiac hypertrophy by inhibiting nuclear factor-κB signaling and may constitute a potential therapeutic target for pathological cardiac hypertrophy.


Subject(s)
Cardiomegaly/pathology , Cardiomegaly/physiopathology , Interferon Regulatory Factor-7/physiology , Signal Transduction/physiology , Angiotensin II/pharmacology , Animals , Cells, Cultured , Disease Models, Animal , Hypertrophy/chemically induced , Hypertrophy/pathology , Hypertrophy/physiopathology , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/physiology , In Vitro Techniques , Interferon Regulatory Factor-7/deficiency , Interferon Regulatory Factor-7/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Myocytes, Cardiac/physiology , NF-kappa B/antagonists & inhibitors , NF-kappa B/physiology
14.
Blood ; 121(20): 4090-100, 2013 May 16.
Article in English | MEDLINE | ID: mdl-23535064

ABSTRACT

As a result of its interaction with transcription factors, HIV type 1 (HIV-1) Tat can modulate the expression of both HIV and cellular genes. In antigen-presenting cells Tat induces the expression of a subset of interferon (IFN)-stimulated genes (ISGs) in the absence of IFNs. We investigated the genome-wide Tat association with promoters in immature dendritic cells and in monocyte-derived macrophages. Among others, Tat associated with the MAP2K6, MAP2K3, and IRF7 promoters that are functionally part of IL-1 and p38 mitogen-activated protein kinase (MAPK) signaling pathways. The association correlated with their increased gene expression, increased activation of p38 MAPK and of phosphorylated signal transducer and activator of transcription 1 (STAT1), and consequent induction of ISGs. Probing these pathways with RNA interference, pharmacological p38 MAPK inhibition, and in cell lines lacking STAT1s or the type I IFN receptor chain confirmed the role of MAPKKs and IRF7 in Tat-mediated modulation of ISGs and excluded the involvement of IFNs in this modulation. Tat interaction with the 2 MAPKK and IRF7 promoters in HIV-1-infected cells and the resulting persistent activation of ISGs, which include inflammatory cytokines and chemokines, can contribute to the increased immune activation that characterizes HIV infection.


Subject(s)
Antigen-Presenting Cells/metabolism , Gene Products, tat/metabolism , Interferon Regulatory Factor-7/genetics , Interferons/pharmacology , MAP Kinase Kinase 3/genetics , MAP Kinase Kinase 6/genetics , Promoter Regions, Genetic , p38 Mitogen-Activated Protein Kinases/metabolism , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/physiology , Cells, Cultured , Chemokines/genetics , Chemokines/metabolism , Gene Products, tat/physiology , HIV Infections/genetics , HIV Infections/immunology , HIV-1/physiology , Humans , Interferon Regulatory Factor-7/metabolism , Interferon Regulatory Factor-7/physiology , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , MAP Kinase Kinase 3/metabolism , MAP Kinase Kinase 6/metabolism , Protein Binding , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Up-Regulation/drug effects , Up-Regulation/genetics , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/physiology
15.
J Virol ; 86(24): 13515-23, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23035219

ABSTRACT

Human noroviruses (HuNoV) are the major cause of epidemic, nonbacterial gastroenteritis in the world. The short course of HuNoV-induced symptoms has implicated innate immunity in control of norovirus (NoV) infection. Studies using murine norovirus (MNV) confirm the importance of innate immune responses during NoV infection. Type I alpha and beta interferons (IFN-α/ß) limit HuNoV replicon function, restrict MNV replication in cultured cells, and control MNV replication in vivo. Therefore, the cell types and transcription factors involved in antiviral immune responses and IFN-α/ß-mediated control of NoV infection are important to define. We used mice with floxed alleles of the IFNAR1 chain of the IFN-α/ß receptor to identify cells expressing lysozyme M or CD11c as cells that respond to IFN-α/ß to restrict MNV replication in vivo. Furthermore, we show that the transcription factors IRF-3 and IRF-7 work in concert to initiate unique and overlapping antiviral responses to restrict MNV replication in vivo. IRF-3 and IRF-7 restrict MNV replication in both cultured macrophages and dendritic cells, are required for induction of IFN-α/ß in macrophages but not dendritic cells, and are dispensable for the antiviral effects of IFN-α/ß that block MNV replication. These studies suggest that expression of the IFN-α/ß receptor on macrophages/neutrophils and dendritic cells, as well as of IRF-3 and IRF-7, is critical for innate immune responses to NoV infection.


Subject(s)
Interferon Regulatory Factor-3/physiology , Interferon Regulatory Factor-7/physiology , Interferon Type I/physiology , Norovirus/physiology , Virus Replication/physiology , Animals , Base Sequence , Cell Line , DNA Primers , Dendritic Cells/immunology , Immunity, Innate , Macrophages/immunology , Mice , Norovirus/immunology , Reverse Transcriptase Polymerase Chain Reaction
16.
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
17.
Nat Med ; 18(8): 1224-31, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22820642

ABSTRACT

Breast cancer metastasis is a key determinant of long-term patient survival. By comparing the transcriptomes of primary and metastatic tumor cells in a mouse model of spontaneous bone metastasis, we found that a substantial number of genes suppressed in bone metastases are targets of the interferon regulatory factor Irf7. Restoration of Irf7 in tumor cells or administration of interferon led to reduced bone metastases and prolonged survival time. In mice deficient in the interferon (IFN) receptor or in natural killer (NK) and CD8(+) T cell responses, metastasis was accelerated, indicating that Irf7-driven suppression of metastasis was reliant on IFN signaling to host immune cells. We confirmed the clinical relevance of these findings in over 800 patients in which high expression of Irf7-regulated genes in primary tumors was associated with prolonged bone metastasis-free survival. This gene signature may identify patients that could benefit from IFN-based therapies. Thus, we have identified an innate immune pathway intrinsic to breast cancer cells, the suppression of which restricts immunosurveillance to enable metastasis.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Gene Silencing , Interferon Regulatory Factor-7/physiology , Mammary Neoplasms, Experimental/immunology , Neoplasm Proteins/physiology , Tumor Escape/physiology , Animals , Breast Neoplasms/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunologic Surveillance , Interferon Regulatory Factor-7/antagonists & inhibitors , Interferon Regulatory Factor-7/biosynthesis , Interferon Regulatory Factor-7/genetics , Interferon-Stimulated Gene Factor 3, gamma Subunit/antagonists & inhibitors , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Interferon-Stimulated Gene Factor 3, gamma Subunit/physiology , Interferon-alpha/pharmacology , Killer Cells, Natural/immunology , Mammary Neoplasms, Experimental/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Neoplasm Metastasis/physiopathology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Receptors, Interferon/deficiency , Receptors, Interferon/physiology , Recombinant Proteins/metabolism , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/immunology , T-Lymphocyte Subsets/immunology , Tumor Escape/genetics
18.
J Virol ; 86(18): 9888-98, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22761364

ABSTRACT

Chikungunya virus (CHIKV) infections can produce severe disease and mortality. Here we show that CHIKV infection of adult mice deficient in interferon response factors 3 and 7 (IRF3/7(-/-)) is lethal. Mortality was associated with undetectable levels of alpha/beta interferon (IFN-α/ß) in serum, ∼50- and ∼10-fold increases in levels of IFN-γ and tumor necrosis factor (TNF), respectively, increased virus replication, edema, vasculitis, hemorrhage, fever followed by hypothermia, oliguria, thrombocytopenia, and raised hematocrits. These features are consistent with hemorrhagic shock and were also evident in infected IFN-α/ß receptor-deficient mice. In situ hybridization suggested CHIKV infection of endothelium, fibroblasts, skeletal muscle, mononuclear cells, chondrocytes, and keratinocytes in IRF3/7(-/-) mice; all but the latter two stained positive in wild-type mice. Vaccination protected IRF3/7(-/-) mice, suggesting that defective antibody responses were not responsible for mortality. IPS-1- and TRIF-dependent pathways were primarily responsible for IFN-α/ß induction, with IRF7 being upregulated >100-fold in infected wild-type mice. These studies suggest that inadequate IFN-α/ß responses following virus infection can be sufficient to induce hemorrhagic fever and shock, a finding with implications for understanding severe CHIKV disease and dengue hemorrhagic fever/dengue shock syndrome.


Subject(s)
Alphavirus Infections/immunology , Alphavirus Infections/prevention & control , Chikungunya virus/pathogenicity , Interferon Regulatory Factor-3/physiology , Interferon Regulatory Factor-7/physiology , Adaptor Proteins, Vesicular Transport/deficiency , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/physiology , Alphavirus Infections/pathology , Animals , Chikungunya Fever , Chikungunya virus/immunology , Chikungunya virus/physiology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Interferon Regulatory Factor-3/deficiency , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-7/deficiency , Interferon Regulatory Factor-7/genetics , Interferon-alpha/biosynthesis , Interferon-alpha/pharmacology , Interferon-beta/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/physiology , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/physiology , Shock, Hemorrhagic/immunology , Shock, Hemorrhagic/prevention & control , Virus Replication/drug effects
20.
Stroke ; 43(5): 1383-9, 2012 May.
Article in English | MEDLINE | ID: mdl-22403050

ABSTRACT

BACKGROUND AND PURPOSE: Systemic administration of Toll-like receptor (TLR) 4 and TLR9 agonists before cerebral ischemia have been shown to reduce ischemic injury by reprogramming the response of the brain to stroke. Our goal was to explore the mechanism of TLR-induced neuroprotection by determining whether a TLR7 agonist also protects against stroke injury. METHODS: C57Bl/6, TNF(-/-), interferon (IFN) regulatory factor 7(-/-), or type I IFN receptor (IFNAR)(-/-) mice were subcutaneously administered the TLR7 agonist Gardiquimod (GDQ) 72 hours before middle cerebral artery occlusion. Infarct volume and functional outcome were determined after reperfusion. Plasma cytokine responses and induction of mRNA for IFN-related genes in the brain were measured. IFNAR(-/-) mice also were treated with the TLR4 agonist (lipopolysaccharide) or the TLR9 agonist before middle cerebral artery occlusion and infarct volumes measured. RESULTS: The results show that GDQ reduces infarct volume as well as functional deficits in mice. GDQ pretreatment provided robust neuroprotection in TNF(-/-) mice, indicating that TNF was not essential. GDQ induced a significant increase in plasma IFNα levels and both IRF7(-/-) and IFNAR(-/-) mice failed to be protected, implicating a role for IFN signaling in TLR7-mediated protection. CONCLUSIONS: Our studies provide the first evidence that TLR7 preconditioning can mediate neuroprotection against ischemic injury. Moreover, we show that the mechanism of protection is unique from other TLR preconditioning ligands in that it is independent of TNF and dependent on IFNAR.


Subject(s)
Aminoquinolines/therapeutic use , Brain/blood supply , Imidazoles/therapeutic use , Ischemic Preconditioning/methods , Membrane Glycoproteins/agonists , Neuroprotective Agents/therapeutic use , Receptor, Interferon alpha-beta/physiology , Stroke/prevention & control , Toll-Like Receptor 7/agonists , Animals , Brain Infarction/pathology , Interferon Regulatory Factor-7/deficiency , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Signal Transduction/physiology , Stroke/physiopathology , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/physiology
SELECTION OF CITATIONS
SEARCH DETAIL