Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Sci Rep ; 12(1): 22552, 2022 12 29.
Article in English | MEDLINE | ID: mdl-36581658

ABSTRACT

Human respiratory syncytial virus (HRSV) is a major cause of severe lower respiratory tract disease in infants and the elderly, yet no safe, effective vaccine is commercially available. Closely related bovine RSV (BRSV) causes respiratory disease in young calves, with many similar features to those seen in HRSV. We previously showed that a Newcastle disease virus (NDV)-vectored vaccine expressing the F glycoprotein of HRSV reduced viral loads in lungs of mice and cotton rats and protected from HRSV. However, clinical signs and pathogenesis of disease in laboratory animals following HRSV infection differs from that observed in human infants. Thus, we examined whether a similar vaccine would protect neonatal calves from BRSV infection. Codon-optimized rNDV vaccine (rNDV-BRSV Fopt) was constructed and administered to colostrum-deprived calves. The rNDV-BRSV Fopt vaccine was well-tolerated and there was no evidence of vaccine-enhanced disease in the upper airways or lungs of these calves compared to the non-vaccinated calves. We found two intranasal doses reduces severity of gross and microscopic lesions and decreases viral load in the lungs. Furthermore, serum neutralizing antibodies were generated in vaccinated calves. Finally, reduced lung CXC chemokine levels were observed in vaccinated calves after BRSV challenge. In summary, we have shown that rNDV-BRSV Fopt vaccine is safe in colostrum-deprived calves, and is effective in reducing lung lesions, and decreasing viral load in upper respiratory tract and lungs after challenge.


Subject(s)
Cattle Diseases , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Respiratory Syncytial Virus, Bovine , Respiratory Syncytial Virus, Human , Female , Pregnancy , Animals , Cattle , Humans , Aged , Newcastle disease virus , Colostrum , Respiratory Syncytial Virus Vaccines/genetics , Antibodies, Viral , Cattle Diseases/prevention & control
2.
Nat Commun ; 12(1): 2624, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33976143

ABSTRACT

The etiology of ulcerative colitis is poorly understood and is likely to involve perturbation of the complex interactions between the mucosal immune system and the commensal bacteria of the gut, with cytokines acting as important cross-regulators. Here we use IFN receptor-deficient mice in a dextran sulfate sodium (DSS) model of acute intestinal injury to study the contributions of type I and III interferons (IFN) to the initiation, progression and resolution of acute colitis. We find that mice lacking both types of IFN receptors exhibit enhanced barrier destruction, extensive loss of goblet cells and diminished proliferation of epithelial cells in the colon following DSS-induced damage. Impaired mucosal healing in double IFN receptor-deficient mice is driven by decreased amphiregulin expression, which IFN signaling can up-regulate in either the epithelial or hematopoietic compartment. Together, these data underscore the pleiotropic functions of IFNs and demonstrate that these critical antiviral cytokines also support epithelial regeneration following acute colonic injury.


Subject(s)
Colitis, Ulcerative/immunology , Interferons/metabolism , Intestinal Mucosa/pathology , Re-Epithelialization/immunology , Animals , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/pathology , Dextran Sulfate/administration & dosage , Dextran Sulfate/toxicity , Disease Models, Animal , Epithelial Cells , Female , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Male , Mice , Mice, Knockout , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Receptors, Interferon/genetics , Receptors, Interferon/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Specific Pathogen-Free Organisms
3.
J Exp Med ; 217(5)2020 05 04.
Article in English | MEDLINE | ID: mdl-32289152

ABSTRACT

With the first reports on coronavirus disease 2019 (COVID-19), which is caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the scientific community working in the field of type III IFNs (IFN-λ) realized that this class of IFNs could play an important role in this and other emerging viral infections. In this Viewpoint, we present our opinion on the benefits and potential limitations of using IFN-λ to prevent, limit, and treat these dangerous viral infections.


Subject(s)
Betacoronavirus/physiology , Coronavirus Infections/metabolism , Interferons/metabolism , Pneumonia, Viral/metabolism , COVID-19 , Humans , Pandemics , SARS-CoV-2 , Virus Internalization
4.
Semin Immunol ; 43: 101303, 2019 06.
Article in English | MEDLINE | ID: mdl-31771761

ABSTRACT

The unexpected discovery of a novel family of antiviral mediators, type III IFNs or IFN-λs, challenged the widely accepted primacy of type I IFNs in antiviral immunity, and it is now well recognized that the IFN-λ-based antiviral system plays a major role in antiviral protection of epithelial barriers. The recent characterization of previously unknown IFN-λ-mediated activities has prompted further reassessment of the role of type I IFNs in innate and adaptive immune and inflammatory responses. Since type I and type III IFNs are co-produced in response to a variety of stimuli, it is likely that many physiological processes are simultaneously and coordinately regulated by these cytokines in pathological conditions, and likely at steady state, as baseline expression of both IFN types is maintained by microbiota. In this review, we discuss emerging differences in the production and signaling of type I and type III IFNs, and summarize results of recent studies describing the involvement of type III IFNs in anti-bacterial and anti-fungal, as well as antiviral, defenses.


Subject(s)
Bacterial Infections/immunology , Interferon Type I/metabolism , Interferons/metabolism , Microbiota/immunology , Mycoses/immunology , Virus Diseases/immunology , Animals , Humans , Immunity , Inflammation , Signal Transduction , Interferon Lambda
5.
Vaccine ; 36(26): 3842-3852, 2018 06 18.
Article in English | MEDLINE | ID: mdl-29779923

ABSTRACT

Human respiratory syncytial virus (RSV) is the leading cause of lower airway disease in infants worldwide and repeatedly infects immunocompetent individuals throughout life. Severe lower airway RSV infection during infancy can be life-threatening, but is also associated with important sequelae including development of asthma and recurrent wheezing in later childhood. The basis for the inadequate, short-lived adaptive immune response to RSV infection is poorly understood, but it is widely recognized that RSV actively antagonizes Type I interferon (IFN) production. In addition to the induction of the anti-viral state, IFN production during viral infection is critical for downstream development of robust, long-lived immunity. Based on the hypothesis that a vaccine that induced robust IFN production would be protective, we previously constructed a Newcastle disease virus-vectored vaccine that expresses the F glycoprotein of RSV (NDV-F) and demonstrated that vaccinated mice had reduced lung viral loads and an enhanced IFN-γ response after RSV challenge. Here we show that vaccination also protected cotton rats from RSV challenge and induced long-lived neutralizing antibody production, even in RSV immune animals. Finally, pulmonary eosinophilia induced by RSV infection of unvaccinated cotton rats was prevented by vaccination. Overall, these data demonstrate enhanced protective immunity to RSV F when this protein is presented in the context of an abortive NDV infection.


Subject(s)
Immunity, Humoral , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Viruses/immunology , Animals , Disease Models, Animal , Female , Interferon-gamma/metabolism , Lung/virology , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Virus Vaccines/isolation & purification , Respiratory Syncytial Viruses/isolation & purification , Sigmodontinae , Time Factors , Viral Load
6.
Sci Immunol ; 2(16)2017 10 06.
Article in English | MEDLINE | ID: mdl-28986419

ABSTRACT

Type III interferons (IFN-λs) are the most recently found members of the IFN cytokine family and engage IFNLR1 and IL10R2 receptor subunits to activate innate responses against viruses. We have identified IFN-λs as critical instructors of antifungal neutrophil responses. Using Aspergillus fumigatus (Af) as a model to study antifungal immune responses, we found that depletion of CCR2+ monocytes compromised the ability of neutrophils to control invasive fungal growth. Using an unbiased approach, we identified type I and III IFNs as critical regulators of the interplay between monocytes and neutrophils responding to Af We found that CCR2+ monocytes are an important early source of type I IFNs that prime optimal expression of IFN-λ. Type III IFNs act directly on neutrophils to activate their antifungal response, and mice with neutrophil-specific deletion of IFNLR1 succumb to invasive aspergillosis. Dysfunctional neutrophil responses in CCR2-depleted mice were rescued by adoptive transfer of pulmonary CCR2+ monocytes or by exogenous administration of IFN-α and IFN-λ. Thus, CCR2+ monocytes promote optimal activation of antifungal neutrophils by initiating a coordinated IFN response. We have identified type III IFNs as critical regulators of neutrophil activation and type I IFNs as early stimulators of IFN-λ expression.


Subject(s)
Immunity, Innate , Interferons/immunology , Invasive Fungal Infections/immunology , Adaptive Immunity , Animals , Aspergillus fumigatus/growth & development , Aspergillus fumigatus/immunology , Aspergillus fumigatus/pathogenicity , Cell Line , Humans , Interferon Type I/immunology , Interferon Type I/metabolism , Interferon-alpha/administration & dosage , Interferon-alpha/immunology , Interferons/administration & dosage , Interferons/genetics , Interferons/metabolism , Invasive Fungal Infections/microbiology , Mice , Monocytes/immunology , Neutrophils/immunology , Receptors, CCR2/deficiency , Receptors, CCR2/immunology , Receptors, Cytokine/genetics , Receptors, Cytokine/metabolism , Receptors, Interferon/deficiency , Receptors, Interferon/genetics , Receptors, Interferon/immunology , Interferon Lambda
7.
J Biol Chem ; 292(18): 7295-7303, 2017 05 05.
Article in English | MEDLINE | ID: mdl-28289095

ABSTRACT

Type I interferons (IFN-α/ß) and the more recently identified type III IFNs (IFN-λ) function as the first line of defense against virus infection and regulate the development of both innate and adaptive immune responses. Type III IFNs were originally identified as a novel ligand-receptor system acting in parallel with type I IFNs, but subsequent studies have provided increasing evidence for distinct roles for each IFN family. In addition to their compartmentalized antiviral actions, these two systems appear to have multiple levels of cross-regulation and act coordinately to achieve effective antimicrobial protection with minimal collateral damage to the host.


Subject(s)
Adaptive Immunity , Immunity, Innate , Interleukins/immunology , Virus Diseases/immunology , Animals , Humans , Interferon Type I/immunology , Interferons
9.
PLoS Pathog ; 12(4): e1005600, 2016 04.
Article in English | MEDLINE | ID: mdl-27128797

ABSTRACT

Type I (IFN-α/ß) and type III (IFN-λ) interferons (IFNs) exert shared antiviral activities through distinct receptors. However, their relative importance for antiviral protection of different organ systems against specific viruses remains to be fully explored. We used mouse strains deficient in type-specific IFN signaling, STAT1 and Rag2 to dissect distinct and overlapping contributions of type I and type III IFNs to protection against homologous murine (EW-RV strain) and heterologous (non-murine) simian (RRV strain) rotavirus infections in suckling mice. Experiments demonstrated that murine EW-RV is insensitive to the action of both types of IFNs, and that timely viral clearance depends upon adaptive immune responses. In contrast, both type I and type III IFNs can control replication of the heterologous simian RRV in the gastrointestinal (GI) tract, and they cooperate to limit extra-intestinal simian RRV replication. Surprisingly, intestinal epithelial cells were sensitive to both IFN types in neonatal mice, although their responsiveness to type I, but not type III IFNs, diminished in adult mice, revealing an unexpected age-dependent change in specific contribution of type I versus type III IFNs to antiviral defenses in the GI tract. Transcriptional analysis revealed that intestinal antiviral responses to RV are triggered through either type of IFN receptor, and are greatly diminished when receptors for both IFN types are lacking. These results also demonstrate a murine host-specific resistance to IFN-mediated antiviral effects by murine EW-RV, but the retention of host efficacy through the cooperative action by type I and type III IFNs in restricting heterologous simian RRV growth and systemic replication in suckling mice. Collectively, our findings revealed a well-orchestrated spatial and temporal tuning of innate antiviral responses in the intestinal tract where two types of IFNs through distinct patterns of their expression and distinct but overlapping sets of target cells coordinately regulate antiviral defenses against heterologous or homologous rotaviruses with substantially different effectiveness.


Subject(s)
Interferon Type I/immunology , Interferon-gamma/immunology , Intestines/immunology , Rotavirus Infections/immunology , Animals , Animals, Newborn , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Humans , Immunoblotting , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Polymerase Chain Reaction , Rotavirus
10.
Proc Natl Acad Sci U S A ; 113(6): 1642-7, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26729873

ABSTRACT

Clinical observations link respiratory virus infection and Pseudomonas aeruginosa colonization in chronic lung disease, including cystic fibrosis (CF) and chronic obstructive pulmonary disease. The development of P. aeruginosa into highly antibiotic-resistant biofilm communities promotes airway colonization and accounts for disease progression in patients. Although clinical studies show a strong correlation between CF patients' acquisition of chronic P. aeruginosa infections and respiratory virus infection, little is known about the mechanism by which chronic P. aeruginosa infections are initiated in the host. Using a coculture model to study the formation of bacterial biofilm formation associated with the airway epithelium, we show that respiratory viral infections and the induction of antiviral interferons promote robust secondary P. aeruginosa biofilm formation. We report that the induction of antiviral IFN signaling in response to respiratory syncytial virus (RSV) infection induces bacterial biofilm formation through a mechanism of dysregulated iron homeostasis of the airway epithelium. Moreover, increased apical release of the host iron-binding protein transferrin during RSV infection promotes P. aeruginosa biofilm development in vitro and in vivo. Thus, nutritional immunity pathways that are disrupted during respiratory viral infection create an environment that favors secondary bacterial infection and may provide previously unidentified targets to combat bacterial biofilm formation.


Subject(s)
Biofilms/growth & development , Immunity , Nutritional Physiological Phenomena , Pseudomonas aeruginosa/physiology , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Viruses/physiology , Animals , Antiviral Agents/pharmacology , Bronchi/pathology , Bronchoalveolar Lavage Fluid , Cystic Fibrosis/microbiology , Cystic Fibrosis/pathology , Epithelial Cells/drug effects , Epithelial Cells/microbiology , Epithelial Cells/virology , Homeostasis/drug effects , Humans , Interferon-beta/pharmacology , Iron/pharmacology , Mice , Microbial Interactions/drug effects , Models, Biological , Pseudomonas aeruginosa/drug effects , Respiratory Syncytial Viruses/drug effects , Signal Transduction/drug effects , Transferrin/metabolism
11.
Comp Med ; 65(4): 315-26, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26310461

ABSTRACT

Infection with respiratory syncytial virus (RSV) generally presents as a mild, upper airway disease in human patients but may cause severe lower airway disease in the very young and very old. Progress toward understanding the mechanisms of RSV pathogenesis has been hampered by a lack of relevant rodent models. Mice, the species most commonly used in RSV research, are resistant to upper respiratory infection and do not recapitulate the pattern of virus spread in the human host. To address the need for better rodent models of RSV infection, we have characterized the acute and chronic pathology of RSV infection of a relatively permissive host, cotton rats (Sigmodon hispidus). We demonstrate that virus delivered to the upper airway results in widespread RSV replication in the ciliated respiratory epithelial cells of the nasal cavity and, to a lesser extent, of the lung. Although acute inflammation is relatively mild and rapidly eliminated after viral clearance, chronic, eosinophilic lung pathology persists. These data support the use of cotton rats as a robust rodent model of human RSV disease, including the association between RSV pneumonia and subsequent development of allergic asthma.


Subject(s)
Asthma/virology , Lung/virology , Pneumonia, Viral/virology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/pathogenicity , Sigmodontinae/virology , Animals , Asthma/immunology , Asthma/pathology , Bronchiolitis/virology , Bronchoalveolar Lavage Fluid/virology , Disease Models, Animal , Inhalation Exposure , Lung/immunology , Lung/pathology , Nasal Mucosa/virology , Pneumonia, Viral/immunology , Pneumonia, Viral/pathology , Pulmonary Eosinophilia/virology , Respiratory Mucosa/virology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Viruses/immunology , Time Factors , Virus Replication
12.
Curr Opin Virol ; 13: 117-22, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26176495

ABSTRACT

The study of human respiratory syncytial virus pathogenesis and immunity has been hampered by its exquisite host specificity, and the difficulties encountered in adapting this virus to a murine host. The reasons for this obstacle are not well understood, but appear to reflect, at least in part, the inability of the virus to block the interferon response in any but the human host. This review addresses some of the issues encountered in mouse models of respiratory syncytial virus infection, and describes the advantages and disadvantages of alternative model systems.


Subject(s)
Disease Models, Animal , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/physiology , Animals , Humans , Respiratory Syncytial Virus, Human/genetics
13.
Am J Physiol Lung Cell Mol Physiol ; 308(7): L650-7, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25617378

ABSTRACT

Influenza infection results in considerable pulmonary pathology, a significant component of which is mediated by CD8(+) T cell effector functions. To isolate the specific contribution of CD8(+) T cells to lung immunopathology, we utilized a nonviral murine model in which alveolar epithelial cells express an influenza antigen and injury is initiated by adoptive transfer of influenza-specific CD8(+) T cells. We report that IFN-γ production by adoptively transferred influenza-specific CD8(+) T cells is a significant contributor to acute lung injury following influenza antigen recognition, in isolation from its impact on viral clearance. CD8(+) T cell production of IFN-γ enhanced lung epithelial cell expression of chemokines and the subsequent recruitment of inflammatory cells into the airways. Surprisingly, Stat1 deficiency in the adoptive-transfer recipients exacerbated the lung injury that was mediated by the transferred influenza-specific CD8(+) T cells but was still dependent on IFN-γ production by these cells. Loss of Stat1 resulted in sustained activation of Stat3 signaling, dysregulated chemokine expression, and increased infiltration of the airways by inflammatory cells. Taken together, these data identify important roles for IFN-γ signaling and Stat1-independent IFN-γ signaling in regulating CD8(+) T cell-mediated acute lung injury. This is the first study to demonstrate an anti-inflammatory effect of Stat1 on CD8(+) T cell-mediated lung immunopathology without the complication of differences in viral load.


Subject(s)
Acute Lung Injury/immunology , CD8-Positive T-Lymphocytes/immunology , Interferon-gamma/physiology , STAT1 Transcription Factor/metabolism , Acute Lung Injury/virology , Animals , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Mice, Inbred BALB C , Mice, Knockout , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , STAT1 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction
14.
Immunol Rev ; 255(1): 25-39, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23947345

ABSTRACT

Interferons (IFNs) are produced in response to virus infection and induce an antiviral state in virtually all cell types. In addition to upregulating the transcription of genes that inhibit virus replication, type I (or -α/ß) IFNs also act to orchestrate the adaptive immune response to virus infection. Recently a new family of antiviral cytokines, the type III (or -λ) IFNs, has been identified that activate the same antiviral pathways via a distinct receptor. Although the identical transcription factor, IFN-stimulated gene factor 3 is activated by either IFN-α/ß or IFN-λ signaling, differences in the induction and action of these two cytokine families are beginning to be appreciated. In this article, we review this emerging body of literature on the differing roles these cytokines play in host defense of the mucosal surface. Although many viruses enter the body through the respiratory and gastrointestinal tracts, we have focused the discussion on influenza A virus, respiratory syncytial virus, and rotavirus, three ubiquitous human pathogens that target the epithelial lining and are associated with a major disease burden.


Subject(s)
Interferons/immunology , Interferons/metabolism , Mucous Membrane/immunology , Mucous Membrane/metabolism , Animals , Gene Expression Regulation , Humans , Janus Kinases/metabolism , Ligands , Mucous Membrane/virology , Phosphorylation , Protein Biosynthesis , STAT Transcription Factors/metabolism , Signal Transduction , Toll-Like Receptors/metabolism , Virus Diseases/genetics , Virus Diseases/immunology , Virus Diseases/metabolism , Viruses/immunology
15.
J Virol ; 86(10): 5422-36, 2012 May.
Article in English | MEDLINE | ID: mdl-22398282

ABSTRACT

Airway epithelial cells (AECs) provide the first line of defense in the respiratory tract and are the main target of respiratory viruses. Here, using oligonucleotide and protein arrays, we analyze the infection of primary polarized human AEC cultures with influenza virus and respiratory syncytial virus (RSV), and we show that the immune response of AECs is quantitatively and qualitatively virus specific. Differentially expressed genes (DEGs) specifically induced by influenza virus and not by RSV included those encoding interferon B1 (IFN-B1), type III interferons (interleukin 28A [IL-28A], IL-28B, and IL-29), interleukins (IL-6, IL-1A, IL-1B, IL-23A, IL-17C, and IL-32), and chemokines (CCL2, CCL8, and CXCL5). Lack of type I interferon or STAT1 signaling decreased the expression and secretion of cytokines and chemokines by the airway epithelium. We also observed strong basolateral polarization of the secretion of cytokines and chemokines by human and murine AECs during infection. Importantly, the antiviral response of human AECs to influenza virus or to RSV correlated with the infection signature obtained from peripheral blood mononuclear cells (PBMCs) isolated from patients with acute influenza or RSV bronchiolitis, respectively. IFI27 (also known as ISG12) was identified as a biomarker of respiratory virus infection in both AECs and PBMCs. In addition, the extent of the transcriptional perturbation in PBMCs correlated with the clinical disease severity. Our results demonstrate that the human airway epithelium mounts virus-specific immune responses that are likely to determine the subsequent systemic immune responses and suggest that the absence of epithelial immune mediators after RSV infection may contribute to explaining the inadequacy of systemic immunity to the virus.


Subject(s)
Epithelial Cells/immunology , Influenza A virus/immunology , Influenza, Human/virology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/immunology , Respiratory Tract Infections/virology , Animals , Cells, Cultured , Cytokines/immunology , Epithelial Cells/virology , Female , Humans , Infant , Influenza A virus/physiology , Influenza, Human/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/physiology , Respiratory Tract Infections/immunology , Species Specificity
16.
Cell Cycle ; 10(5): 794-804, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21311224

ABSTRACT

The anti-tumor function of Stat1 as a regulator of innate immunity and tumor immune surveillance has been long studied and is well understood; however, less clear is its tumor-site specific role. Although Stat1 phosphorylated at tyrosine (Y) 701 and serine (S) 727 is essential for interferon (IFN) signalling, its function in signalling induced in breast cancer cells is not understood. Herein, we show that Stat1 Y701 phosphorylation is increased in human breast tumor cells with elevated levels of ErbB2/HER-2 and in cells transfected with ErbB2/Neu. However, pharmacological inhibition of ErbB2/HER-2 results in the inhibition of Stat1 Y701 phosphorylation indicating an atypical role of phosphorylated Stat1 in the inhibition of ErbB2/HER-2 signalling. Consistent with this notion, we found that Stat1 suppresses tumor development by an activated form of ErbB2/Neu in mouse embryonic fibroblasts in xenograft tumor assays; however, this anti-tumor function of Stat1 does not rely on Y701 and S727 phosphorylation. Experiments with transgenic mice demonstrated that Stat1 acts to suppress Neu-mediated breast tumorigenesis through immune regulatory and tumor-site specific mechanisms. Our data reveal a previous uncharacterized anti-tumor activity of Stat1 in ErbB2/Neu-mediated cell transformation and breast oncogenesis with possible implications in the diagnosis and treatment of ErbB2-positive breast cancers.


Subject(s)
Cell Transformation, Neoplastic , Mammary Neoplasms, Experimental/metabolism , Receptor, ErbB-2/metabolism , STAT1 Transcription Factor/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line , Chlorocebus aethiops , Disease Progression , Female , Gefitinib , Humans , Lapatinib , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Phosphorylation , Quinazolines/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Signal Transduction , Transplantation, Heterologous
17.
Curr Opin Virol ; 1(6): 476-86, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22323926

ABSTRACT

The type I and III interferon (IFN) families consist of cytokines rapidly induced during viral infection that confer antiviral protection on target cells and are critical components of innate immune responses and the transition to effective adaptive immunity. The regulation of their expression involves an intricate and stringently regulated signaling cascade, initiated by recognition most often of viral nucleic acid in cytoplasmic and endosomal compartments and involving a series of protein conformational rearrangements and interactions regulated by helicase action, ubiquitin modification, and protein aggregation, culminating in kinase activation and phosphorylation of critical transcription factors and their regulators. The many IFN subtypes induced by viruses confer amplification, diversification, and cell-type specificity to the host response to infection, providing fertile ground for development of antiviral therapeutics and vaccines.


Subject(s)
Immunity, Innate/immunology , Interferon Type I/immunology , Virus Diseases/immunology , Viruses/immunology , Animals , Gene Expression Regulation, Viral , Humans , Interferon Type I/biosynthesis , Interferon Type I/genetics , Signal Transduction , Transcription Factors/immunology , Ubiquitin/immunology , Virus Diseases/virology
18.
J Virol ; 84(21): 11515-22, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20739515

ABSTRACT

The type I alpha/beta interferons (IFN-α/ß) are known to play an important role in host defense against influenza A virus infection, but we have now discovered that the recently identified type III IFNs (IFN-λ) constitute the major response to intranasal infection with this virus. Type III IFNs were present at much higher levels than type I IFNs in the lungs of infected mice, and the enhanced susceptibility of STAT2-/- animals demonstrated that only signaling through the IFN-α/ß or IFN-λ pathways was sufficient to mediate protection. This finding offers a possible explanation for the similar levels of antiviral protection found in wild-type (WT) mice and in animals lacking a functional type I IFN receptor (IFNAR-/-) but also argues that our current understanding of type III IFN induction is incomplete. While murine IFN-λ production is thought to depend on signaling through the type I IFN receptor, we demonstrate that intranasal influenza A virus infection leads to the robust type III IFN induction in the lungs of both WT and IFNAR-/- mice. This is consistent with previous studies showing that IFNAR-mediated protection is redundant for mucosal influenza virus infection and with data showing that the type III IFN receptor is expressed primarily by epithelial cells. However, the overlapping effects of these two cytokine families are limited by their differential receptor expression, with a requirement for IFN-α/ß signaling in combating systemic disease.


Subject(s)
Cytokines/genetics , Interferons/genetics , Orthomyxoviridae Infections/immunology , Transcriptional Activation , Animals , Epithelial Cells/metabolism , Humans , Influenza A virus , Mice , Mice, Inbred BALB C , Mice, Knockout , Receptor, Interferon alpha-beta/deficiency
19.
Comp Med ; 60(3): 225-32, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20579438

ABSTRACT

Although most viral infections of the upper respiratory tract can predispose to bacterial otitis media, human respiratory syncytial virus (HRSV) is the predominant viral copathogen of this highly prevalent pediatric polymicrobial disease. Rigorous study of the specific mechanisms by which HRSV predisposes to otitis media has been hindered by lack of a relevant animal model. We recently reported that the chinchilla, the preferred rodent host for studying otitis media, is semipermissive for upper-airway HRSV infection. In the current study, we defined the anatomy and kinetics of HRSV infection and spread in the upper airway of chinchilla hosts. Chinchillas were challenged intranasally with a fluorescent-protein-expressing HRSV. Upper-airway tissues were recovered at multiple time points after viral challenge and examined by confocal microscopy and immunohistochemistry. HRSV replication was observed from the rostral- to caudalmost regions of the nasal cavity as well as throughout the Eustachian tube in a time-dependent manner. Although fluorescence was not observed and virus was not detected in nasopharyngeal lavage fluids 14 d after infection, the latest time point examined in this study, occasional clusters of immunopositive cells were present, suggesting that the nasal cavity may serve as a reservoir for HRSV. These data provide important new information concerning the time course of HRSV infection of the uppermost airway and suggest that chinchillas may be useful for modeling the HRSV-induced changes that predispose to secondary bacterial infection.


Subject(s)
Chinchilla , Disease Models, Animal , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human/pathogenicity , Respiratory System , Animals , Chinchilla/anatomy & histology , Chinchilla/virology , Female , Humans , Male , Mice , Nasal Cavity/anatomy & histology , Nasal Cavity/virology , Nasal Lavage Fluid/virology , Otitis Media/veterinary , Otitis Media/virology , Respiratory Syncytial Virus Infections/veterinary , Respiratory Syncytial Virus Infections/virology , Respiratory System/anatomy & histology , Respiratory System/virology , Viral Load
20.
FASEB J ; 23(11): 3990-9, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19641143

ABSTRACT

The signal transducer and activator of transcription 1 (STAT1) signaling pathway mediates the biological functions of IFN-gamma. We have previously shown that the STAT1 pathway is indispensable for host resistance against Leishmania major infection. In this study, we examined the role of STAT1 in lymphocytes and specifically CD4(+) and CD8(+) T cells in mediating immunity against L. major by transferring T cells from wild-type (WT) and STAT1(-/-) C57BL/6 mice into Rag2(-/-) C57BL/6 mice. Rag2(-/-) mice reconstituted with unfractionated STAT1(-/-) splenocytes (B cells and T cells) failed to mount an efficient Th1 response after L. major infection, produced more IL-4, and developed large lesions full of parasites. In contrast, Rag2(-/-) mice reconstituted with WT (STAT1(+/+)) splenocytes mounted a Th1 response and developed self-resolving lesions. Studies using Rag2(-/-) recipients that received a combination of purified CD4(+) and CD8(+) T cells from WT or STAT1(-/-) mice revealed that STAT1 deficiency in CD4(+) T cells, but not in CD8(+) T cells, leads to development of chronic, nonhealing lesions and systemic dissemination of parasites into the spleen after L. major infection. Further studies using Rag2(-/-) recipients of WT Thy1.1(+) and STAT1(-/-) Thy1.2(+) T cells showed that STAT1 in CD4(+) T cells was not required for Th1 differentiation during L. major infection. However, it was critical for up-regulation of CXCR3 on CD4(+) T cells and their migration to the regional lymph node and the cutaneous site of infection. Together, these studies indicate that the STAT1 pathway in CD4(+) T cells plays a critical role in immunity against L. major by controlling the migration of Th1 cells to the site of infection rather than their generation. Further, they reveal an essential role for CD4(+) T cell STAT1 in preventing systemic dissemination of L. major infection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Leishmania major/immunology , Leishmaniasis, Cutaneous/immunology , STAT1 Transcription Factor/physiology , Th1 Cells/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , DNA-Binding Proteins/deficiency , Interferon-gamma/physiology , Leishmaniasis, Cutaneous/pathology , Mice , Mice, Inbred C57BL , STAT1 Transcription Factor/deficiency , Signal Transduction/immunology , Th2 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...