Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
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.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
Curr Protoc Cell Biol ; Chapter 26: Unit 26.3, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19499505

ABSTRACT

This unit describes protocols for infecting the mouse respiratory tract, and assaying virus replication and host response in the lung. Respiratory infections are the leading cause of acute illness worldwide, affecting mostly infants and children in developing countries. The purpose of this unit is to provide a basic strategy and protocols to study the pathogenesis and immunology of respiratory virus infection using the mouse as an animal model. The procedures include: (1) basic techniques for mouse infection, tissue sampling, and preservation, (2) determination of viral titers, isolation and analysis of lymphocytes and dendritic cells using flow-cytometry, and (3) lung histology, immunohistochemistry, and in situ hybridization.


Subject(s)
Antigens, Viral/immunology , Lung/immunology , Respiratory Tract Infections/immunology , Respiratory Tract Infections/virology , Virology/methods , Viruses/growth & development , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , Cell Separation , Dendritic Cells/immunology , Dendritic Cells/pathology , Dendritic Cells/virology , Disease Models, Animal , Flow Cytometry , Immunohistochemistry , Lung/pathology , Lung/virology , Mice , Virulence , Virus Replication , Viruses/immunology , Viruses/pathogenicity
11.
Am J Respir Crit Care Med ; 178(9): 969-76, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18689466

ABSTRACT

RATIONALE: Pulmonary infections can impair alveolar fluid clearance (AFC), contributing to formation of lung edema. Effects of influenza A virus (IAV) on AFC are unknown. OBJECTIVES: To determine effects of IAV infection on AFC, and to identify intercellular signaling mechanisms underlying influenza-mediated inhibition of AFC. METHODS: BALB/c mice were infected intranasally with influenza A/WSN/33 (10,000 or 2,500 focus-forming units per mouse). AFC was measured in anesthetized, ventilated mice by instilling 5% bovine serum albumin into the dependent lung. MEASUREMENTS AND MAIN RESULTS: Infection with high-dose IAV resulted in a steady decline in arterial oxygen saturation and increased lung water content. AFC was significantly inhibited starting 1 hour after infection, and remained suppressed through Day 6. AFC inhibition at early time points (1-4 h after infection) did not require viral replication, whereas AFC inhibition later in infection was replication-dependent. Low-dose IAV infection impaired AFC for 10 days, but induced only mild hypoxemia. High-dose IAV infection increased bronchoalveolar lavage fluid ATP and UTP levels. Impaired AFC at Day 2 resulted primarily from reduced amiloride-sensitive AFC, mediated by increased activation of the pyrimidine-P2Y purinergic receptor axis. However, an additional component of AFC impairment was due to activation of A(1) adenosine receptors and stimulation of increased cystic fibrosis transmembrane regulator-mediated anion secretion. Finally, IAV-mediated inhibition of AFC at Day 2 could be reversed by addition of beta-adrenergic agonists to the AFC instillate. CONCLUSIONS: AFC inhibition may be an important feature of early IAV infection. Its blockade may reduce the severity of pulmonary edema and hypoxemia associated with influenza pneumonia.


Subject(s)
Bronchoalveolar Lavage Fluid/virology , Influenza A virus/metabolism , Orthomyxoviridae Infections/metabolism , Animals , Biological Transport, Active , Body Weight , Cytokines/metabolism , Disease Models, Animal , Female , Lung/physiopathology , Male , Mice , Mice, Inbred BALB C , Oxygen/blood , Permeability , Time Factors
12.
J Virol ; 81(18): 9790-800, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17626092

ABSTRACT

Type I interferon (IFN) induction is an immediate response to virus infection, and very high levels of these cytokines are produced when the Toll-like receptors (TLRs) expressed at high levels by plasmacytoid dendritic cells (pDCs) are triggered by viral nucleic acids. Unlike many RNA viruses, respiratory syncytial virus (RSV) does not appear to activate pDCs through their TLRs and it is not clear how this difference affects IFN-alpha/beta induction in vivo. In this study, we investigated type I IFN production triggered by RSV or influenza A virus infection of BALB/c mice and found that while both viruses induced IFN-alpha/beta production by pDCs in vitro, only influenza virus infection could stimulate type I IFN synthesis by pDCs in vivo. In situ hybridization studies demonstrated that the infected respiratory epithelium was a major source of IFN-alpha/beta in response to either infection, but in pDC-depleted animals only type I IFN induction by influenza virus was impaired.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Interferon-alpha/immunology , Interferon-beta/immunology , Orthomyxoviridae Infections/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , Animals , Cattle , Cell Line , Dendritic Cells/immunology , Dendritic Cells/pathology , Dendritic Cells/virology , Mice , Mice, Inbred BALB C , Mice, Knockout , Orthomyxoviridae Infections/pathology , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , Respiratory Mucosa/virology , Respiratory Syncytial Virus Infections/pathology
13.
Virology ; 362(1): 139-50, 2007 May 25.
Article in English | MEDLINE | ID: mdl-17254623

ABSTRACT

Parainfluenza virus type 5 (PIV5), formerly known as simian virus 5 (SV5), is a non-segmented negative strand RNA virus that offers several advantages as a vaccine vector. PIV5 infects many cell types causing little cytopathic effect, it replicates in the cytoplasm of infected cells, and does not have a DNA phase in its life cycle thus avoiding the possibility of introducing foreign genes into the host DNA genome. Importantly, PIV5 can infect humans but it is not associated with any known human illness. PIV5 grows well in tissue culture cells, including Vero cells, which have been approved for vaccine production, and the virus can be obtained easily from the media. To test the feasibility of using PIV5 as a live vaccine vector, the hemagglutinin (HA) gene from influenza A virus strain A/Udorn/72 (H3N2) was inserted into the PIV5 genome as an extra gene between the hemagglutinin-neuraminidase (HN) gene and the large (L) polymerase gene. Recombinant PIV5 containing the HA gene of Udorn (rPIV5-H3) was recovered and it replicated similarly to wild type PIV5, both in vitro and in vivo. The HA protein expressed by rPIV5-H3-infected cells was incorporated into the virions and addition of the HA gene did not increase virus virulence in mice. The efficacy of rPIV5-H3 as a live vaccine was examined in 6-week-old BALB/c mice. The results show that a single dose inoculation provides broad and considerable immunity against influenza A virus infection.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A virus/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Parainfluenza Virus 5/immunology , Animals , Antibodies, Viral/blood , Body Weight , Cattle , Cell Line , Cells, Cultured , Chlorocebus aethiops , Disease Models, Animal , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/immunology , Influenza A virus/genetics , Influenza Vaccines/genetics , Lung/pathology , Lung/virology , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/pathology , Parainfluenza Virus 5/genetics , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
14.
J Virol ; 80(3): 1130-9, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16414990

ABSTRACT

Respiratory syncytial virus (RSV) is a major cause of severe lower respiratory tract disease in infants and the elderly, but no safe and effective RSV vaccine is yet available. For reasons that are not well understood, RSV is only weakly immunogenic, and reinfection occurs throughout life. This has complicated the search for an effective live attenuated viral vaccine, and past trials with inactivated virus preparations have led to enhanced immunopathology following natural infection. We have tested the hypothesis that weak stimulation of innate immunity by RSV correlates with ineffective adaptive responses by asking whether expression of the fusion glycoprotein of RSV by Newcastle disease virus (NDV) would stimulate a more robust immune response to RSV than primary RSV infection. NDV is a potent inducer of both alpha/beta interferon (IFN-alpha/beta) production and dendritic cell maturation, while RSV is not. When a recombinant NDV expressing the RSV fusion glycoprotein was administered to BALB/c mice, they were protected from RSV challenge, and this protection correlated with a robust anti-F CD8+ T-cell response. The effectiveness of this vaccine construct reflects the differential abilities of NDV and RSV to promote dendritic cell maturation and is retained even in the absence of a functional IFN-alpha/beta receptor.


Subject(s)
Newcastle disease virus/genetics , Respiratory Syncytial Viruses/immunology , Respiratory Syncytial Viruses/pathogenicity , Animals , Base Sequence , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Cell Line , Chlorocebus aethiops , DNA, Viral/genetics , Dendritic Cells/cytology , Dendritic Cells/immunology , Genetic Vectors , Humans , Interferon-alpha/biosynthesis , Interferon-beta/biosynthesis , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Knockout , Receptor, Interferon alpha-beta , Receptors, Interferon/deficiency , Receptors, Interferon/genetics , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/genetics , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus Vaccines/pharmacology , Respiratory Syncytial Viruses/genetics , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/pharmacology , Vero Cells , Viral Fusion Proteins/genetics , Viral Fusion Proteins/immunology
15.
J Immunol ; 174(11): 7234-41, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15905569

ABSTRACT

Respiratory syncytial virus (RSV) is the major cause of severe lower airway disease in infants and young children, but no safe and effective RSV vaccine is yet available. The difficulties involved in RSV vaccine development were recognized in an early vaccine trial, when children immunized with a formalin-inactivated virus preparation experienced enhanced illness after natural infection. Subsequent research in animal models has shown that the vaccine-enhanced disease is mediated at least in part by memory cells producing Th2 cytokines. Previously we had observed enhanced, eosinophilic lung pathology during primary infection of IFN-deficient STAT1(-/-) mice that are incapable of generating Th1 CD4(+) cells. To determine whether these effects depended only on Th2 cytokine secretion or involved other aspects of IFN signaling, we infected a series of 129SvEv knockout mice lacking the IFN-alphabetaR (IFN-alphabetaR(-/-)), the IFN-gammaR (IFN-gammaR(-/-)), or both receptors (IFN-alphabetagammaR(-/-)). Although both the IFN-gammaR(-/-) and the IFN-alphabetagammaR(-/-) animals generated strong Th2 responses to RSV-F protein epitopes, predominantly eosinophilic lung disease was limited to mice lacking both IFNRs. Although the absolute numbers of eosinophils in BAL fluids were similar between the strains, very few CD8(+) T cells could be detected in lungs of IFN-alphabetagammaR(-/-) animals, leaving eosinophils as the predominant leukocyte. Thus, although CD4(+) Th2 cell differentiation is necessary for the development of allergic-type inflammation after infection and appears to be unaffected by type I IFNs, innate IFNs clearly have an important role in determining the nature and severity of RSV disease.


Subject(s)
Interferons/physiology , Lung/immunology , Lung/pathology , Respiratory Syncytial Viruses/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/virology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Line , Chemokines/biosynthesis , Cytokines/biosynthesis , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Immunity, Innate/genetics , Interferons/deficiency , Interferons/genetics , Interferons/metabolism , Lung/metabolism , Lung/virology , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Mice, Inbred Strains , Mice, Knockout , Receptor, Interferon alpha-beta , Receptors, Interferon/deficiency , Receptors, Interferon/genetics , Receptors, Interferon/physiology , Respiratory Syncytial Virus Infections/genetics , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/pathology , STAT1 Transcription Factor , Signal Transduction/genetics , Signal Transduction/immunology , Trans-Activators/deficiency , Trans-Activators/genetics , Trans-Activators/physiology , Virus Replication/immunology , Interferon gamma Receptor
16.
J Virol ; 79(10): 6035-42, 2005 May.
Article in English | MEDLINE | ID: mdl-15857989

ABSTRACT

Respiratory syncytial virus (RSV) is a major cause of lower respiratory tract infections in infants and the elderly. While the primary infection is the most serious, reinfection of the upper airway throughout life is the rule. Although relatively little is known about either RSV infection of the upper respiratory tract or host mucosal immunity to RSV, recent literature suggests that RSV is the predominant viral pathogen predisposing to bacterial otitis media (OM). Herein, we describe mouse and chinchilla models of RSV infection of the nasopharynx and Eustachian tube. Both rodent hosts were susceptible to RSV infection of the upper airway following intranasal challenge; however, the chinchilla proved to be more permissive than the mouse. The chinchilla model will likely be extremely useful to test the role of RSV in bacterial OM and the efficacy of RSV vaccine candidates designed to provide mucosal and cytotoxic T-lymphocyte immunity. Ultimately, we hope to investigate the relative ability of these candidates to potentially protect against viral predisposal to bacterial OM.


Subject(s)
Disease Models, Animal , Nasopharyngeal Diseases/virology , Otitis Media/virology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses , Animals , Chinchilla , Disease Susceptibility , Eustachian Tube/pathology , Eustachian Tube/virology , Female , Fluorescent Antibody Technique, Indirect , Immunohistochemistry , Male , Mice , Mice, Inbred BALB C , Nasopharynx/virology , Otitis Media/pathology , Respiratory Mucosa/virology , Respiratory Syncytial Viruses/isolation & purification , Species Specificity
17.
J Virol ; 78(19): 10303-9, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15367596

ABSTRACT

The role of prostanoids in modulating respiratory syncytial virus (RSV) infection is unknown. We found that RSV infection in mice increases production of prostaglandin I(2) (PGI(2)). Mice that overexpress PGI(2) synthase selectively in bronchial epithelium are protected against RSV-induced weight loss and have decreased peak viral replication and gamma interferon levels in the lung compared to nontransgenic littermates. In contrast, mice deficient in the PGI(2) receptor IP have exacerbated RSV-induced weight loss with delayed viral clearance and increased levels of gamma interferon in the lung compared to wild-type mice. These results suggest that signaling through IP has antiviral effects while protecting against RSV-induced illness and that PGI(2) is a potential therapeutic target in the treatment of RSV.


Subject(s)
6-Ketoprostaglandin F1 alpha/analogs & derivatives , Epoprostenol/metabolism , Receptors, Epoprostenol/metabolism , Respiratory Syncytial Virus Infections/physiopathology , Respiratory Syncytial Viruses/pathogenicity , Signal Transduction , 6-Ketoprostaglandin F1 alpha/urine , Animals , Antibodies, Viral/blood , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Disease Models, Animal , Female , Gene Deletion , Interferon-alpha/biosynthesis , Interferon-beta/biosynthesis , Interferon-gamma/analysis , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Lung/chemistry , Lung/pathology , Lung/virology , Male , Mice , Mice, Transgenic , Pulmonary Edema/pathology , Pulmonary Edema/prevention & control , Pulmonary Surfactant-Associated Protein A/biosynthesis , Pulmonary Surfactant-Associated Protein B/biosynthesis , Receptors, Epoprostenol/genetics , Receptors, Epoprostenol/immunology , Respiratory Mucosa , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Viruses/growth & development , Respiratory Syncytial Viruses/immunology , Weight Loss
18.
Viral Immunol ; 17(3): 370-80, 2004.
Article in English | MEDLINE | ID: mdl-15357903

ABSTRACT

Respiratory syncytial virus (RSV), a member of the Paramyxoviridae family, is a major clinical problem causing yearly epidemics of severe lower airway disease in both infants and the elderly. Attempts at vaccination have been frustrated by both the poor immunogenicity of this virus, and the severe immunopathology observed in early vaccine trials. Primary infection generally occurs in infancy, with approximately 5% of infected infants requiring hospitalization. Equally problematic is the apparent link between severe RSV disease and the later development of allergy and asthma. While there is no evidence that natural infection promotes Th2 predominance, development of enhanced eosinophilic disease in children receiving inactivated virus administered with a commonly used adjuvant demonstrated how easily the balance between immune-mediated protection and immune-mediated pathology can be perturbed. In this review we have focused on studies carried out in the mouse model aimed at determining the correlates of RSV protection and explaining the mechanism of vaccine enhanced immunopathology.


Subject(s)
Respiratory Syncytial Virus Infections/immunology , Animals , CX3C Chemokine Receptor 1 , Humans , Immunity, Innate , Interferon-gamma/biosynthesis , Interleukin-13/biosynthesis , Killer Cells, Natural/immunology , Membrane Glycoproteins/physiology , Membrane Proteins/physiology , Receptors, Cell Surface/physiology , Receptors, Chemokine/physiology , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Viruses/immunology , Respiratory Syncytial Viruses/pathogenicity , T-Lymphocytes, Cytotoxic/immunology , Toll-Like Receptors , Viral Proteins/physiology
19.
Cancer Cell ; 4(4): 263-75, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14585354

ABSTRACT

Ideally, an oncolytic virus will replicate preferentially in malignant cells, have the ability to treat disseminated metastases, and ultimately be cleared by the patient. Here we present evidence that the attenuated vesicular stomatitis strains, AV1 and AV2, embody all of these traits. We uncover the mechanism by which these mutants are selectively attenuated in interferon-responsive cells while remaining highly lytic in 80% of human tumor cell lines tested. AV1 and AV2 were tested in a xenograft model of human ovarian cancer and in an immune competent mouse model of metastatic colon cancer. While highly attenuated for growth in normal mice, both AV1 and AV2 effected complete and durable cures in the majority of treated animals when delivered systemically.


Subject(s)
Immunity, Innate/physiology , Interferon-beta/metabolism , Vesicular stomatitis Indiana virus/metabolism , Active Transport, Cell Nucleus , Animals , Colonic Neoplasms/therapy , Colonic Neoplasms/virology , Female , Humans , Immunity, Innate/immunology , Interferon-beta/immunology , Lung Neoplasms/therapy , Lung Neoplasms/virology , Mice , Mice, Knockout , Models, Biological , Mutation , Neoplasms, Experimental/virology , Oligonucleotide Array Sequence Analysis , Ovarian Neoplasms/therapy , Ovarian Neoplasms/virology , Signal Transduction , Vesicular stomatitis Indiana virus/genetics , Viral Matrix Proteins/metabolism , Virus Replication/genetics , Virus Replication/physiology
20.
Virology ; 303(1): 15-32, 2002 Nov 10.
Article in English | MEDLINE | ID: mdl-12482655

ABSTRACT

The V protein of the Paramyxovirus simian virus 5 (SV5) is a multifunctional protein containing an N-terminal 164 residue domain that is shared with the P protein and a distinct C-terminal domain that is cysteine-rich and which is highly conserved among Paramyxoviruses. We report the recovery from Vero cells [interferon (IFN) nonproducing cells] of a recombinant SV5 (rSV5) that lacks the V protein C-terminal specific domain (rSV5VDeltaC). In Vero cells rSV5VDeltaC forms large plaques and grows at a rate and titer similar to those of rSV5. In BHK or CV-1 cells rSV5VDeltaC forms small plaques and grows poorly. However, even when grown in Vero cells rSV5VDeltaC reverts to pseudo-wild-type virus in four to five passages, indicating the importance of the V protein for successful replication of SV5. Whereas rSV5 grows in many cell types with minimal cytopathic effect (CPE), rSV5VDeltaC causes extensive CPE in the same cell types. To overcome the antiviral state induced by IFN, many viruses have evolved mechanisms to counteract the effects of IFN by blocking the production of IFN and abrogating IFN signaling. Whereas rSV5 blocks IFN signaling by mediating the degradation of STAT1, rSV5VDeltaC does not cause the degradation of STAT1 and IFN signaling occurs through formation of the ISGF3 transcription complex. Furthermore, we find that rSV5 infection of cells prevents production of IFN-beta. The transcription factor IRF-3 which is required for transcription of the IFN-beta gene is not translocated from the cytoplasm to the nucleus in rSV5-infected cells. In contrast, in rSV5VDeltaC-infected cells IRF-3 is localized predominantly in the nucleus and IFN-beta is produced. By using ectopic expression of IRF-3, it was shown that after dsRNA treatment and expression of the V protein IRF-3 remained in the cytoplasm, whereas after dsRNA treatment and expression of the P protein (which lacks the C-terminal cysteine-rich domain) IRF-3 was localized predominantly in the nucleus. Thus, SV5 blocks two distinct pathways of the innate immune response, both of which require the presence of the C-terminal specific cysteine-rich domain of the multifunctional SV5 V protein.


Subject(s)
Interferon-beta/biosynthesis , Respirovirus/isolation & purification , Viral Structural Proteins/physiology , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cysteine/chemistry , Cytopathogenic Effect, Viral , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HeLa Cells , Humans , Interferon Regulatory Factor-3 , Interferon-beta/antagonists & inhibitors , Mice , Mice, Inbred BALB C , Mice, Knockout , Molecular Sequence Data , Mutation , Protein Structure, Tertiary , Recombinant Proteins/genetics , Respirovirus/genetics , Respirovirus/pathogenicity , Respirovirus Infections/virology , STAT1 Transcription Factor , Signal Transduction , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Viral Structural Proteins/chemistry , Viral Structural Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...