39 results on '"Durbin, Joan"'
Search Results
2. The role of STAT1 in viral sensitization to LPS.
- Author
-
Durbin, Joan, Doughty, Leslie, Nguyen, Ken, Caligiuri, Michael, Van Deusen, Jeff, and Biron, Christine
- Subjects
- *
ENDOTOXINS , *MICROBIAL polysaccharides , *BACTERIAL cell walls , *PYROGENS , *TRANSFER factor (Immunology) , *LYMPHOCYTIC choriomeningitis virus - Abstract
The phenomenon of endotoxin sensitization by virus infection is well documented but not yet well understood. Infection by virtually any viral agent will quickly induce expression of type I interferons (IFN-α/β), and type II IFN-γ production will follow as NK cells and T cells are activated. It has been well established that type II IFN pretreatment can intensify the effects of endotoxin. We have recently demonstrated that type I IFN induction by lymphocytic choriomeningitis virus (LCMV) infection dramatically increases TNF-α production following LPS treatment, and that this sensitization by type I IFN is STAT1 dependent. Taken together these data suggest that the STAT1-mediated, MyD88-independent, arm of the LPS signaling pathway plays an important role in endotoxin toxicity, and that this pathway mediates a major component of virus-enhanced LPS sensitization. [ABSTRACT FROM AUTHOR]
- Published
- 2003
- Full Text
- View/download PDF
3. Anatomy of the Nasal Cavity in the Chinchilla.
- Author
-
Jurcisek, Joseph A., Durbin, Joan E., Kusewitt, Donna F., and Bakaletz, Lauren O.
- Subjects
- *
OTITIS media , *INTRANASAL medication , *IMMUNIZATION , *NASAL cavity , *ANATOMY , *CHINCHILLAS , *LYMPHOID tissue , *VACCINATION - Abstract
There is currently great interest worldwide in developing noninvasive methods for the delivery of vaccines for upper respiratory tract diseases, including middle ear infection (otitis media, OM). One such noninvasive approach believed to have great potential for the prevention of diseases of the airway is to deliver vaccines by the intranasal (i.n.) route. Induction of a local, mucosal immune response in the upper respiratory tract, and particularly in the nasopharynx, would be a highly efficacious approach to prevention of OM. The chinchilla is the preferred rodent host for studying OM. However, although the anatomy of the chinchilla vomeronasal organ, inner ear, middle ear and Eustachian tube have been well-studied, to date there have been no reports in the literature of a similar complete analysis of the nasopharynx and nasal cavities of the chinchilla. In order to develop a relevant animal model of i.n. delivery as a potential immunization approach for the prevention of OM and to use these models for preclinical assessments of various vaccine candidates, it was important that we better understand the anatomy of the chinchilla nasal cavities and nasopharynx. Our anatomical studies revealed that the naso- and maxilloturbinates of the chinchilla nasal cavity more closely resemble the simple turbinates found in other rodents rather than the branched or complex turbinates seen in dogs, cats, and rabbits thus facilitating the i.n. delivery of vaccine candidates. The chinchilla nasal mucosa also contains numerous lymphoid aggregates like that of other rodents. Our findings thus suggest that we will be able to deliver i.n. vaccines effectively to chinchillas and that these vaccines will likely be able to induce specific immune responses.Copyright © 2003 S. Karger AG, Basel [ABSTRACT FROM AUTHOR]
- Published
- 2003
- Full Text
- View/download PDF
4. Differential viral induction of distinct interferon-a genes by positive feedback through interferon regulatory factor-7.
- Author
-
Marie, Isabelle, Durbin, Joan e., and Levy, David E.
- Subjects
- *
INTERFERONS , *VIRUS diseases , *CELLS , *PROTEIN synthesis , *PHOSPHORYLATION , *GENETIC transcription , *GENETIC regulation - Abstract
Interferon (IFN) genes are among the earliest transcriptional responses to virus infection of mammalian cells. Although the regulation of the IFNβ gene has been well characterized, the induction of the large family of IFNα genes has remained obscure. We report that the IFNα genes can be divided into two groups: an immediate-early response gene (IFNα4) which is induced rapidly and without the need for ongoing protein synthesis; and a set of genes that display delayed induction, consisting of at least IFN2, 5, 6 and 8, which are induced more slowly and require cellular protein synthesis. One protein that must be synthesized for induction of the delayed gene set is IFN itself, presumably IFNα4 or IFNβ, which stimulates the JakStat pathway through the IFN receptor, resulting in activation of the transcription factor interferon-stimulated gene factor 3 (ISGF3). Among the IFN-stimulated genes induced through this positive feedback loop is the IFN regulatory factor (IRF) protein, IRF7. Induction of IRF7 protein in response to IFN and its subsequent activation by phosphorylation in response to virus-specific signals, involving two C-terminal serine residues, are required for induction of the delayed IFN gene set. [ABSTRACT FROM AUTHOR]
- Published
- 1998
- Full Text
- View/download PDF
5. Targeted disruption of the mouse Stat1 gene results in compromised innate immunity to viral disease.
- Author
-
Durbin, Joan E. and Hackenmiller, Renee
- Subjects
- *
IMMUNOLOGY , *VIRUS diseases , *MICE , *GENETICS - Abstract
Reports on a study that disrupted the Stat1 gene in mice which resulted in compromised innate immunity to viral disease. Phenotype of Stat1 ES cells; Extraordinary sensitivity to viral pathogens in the absence of STAT1; Indications that STAT1 appears to be specific for interferon pathways which are essential for viability in the face of viral disease.
- Published
- 1996
- Full Text
- View/download PDF
6. An intranasal recombinant NDV-BRSV Fopt vaccine is safe and reduces lesion severity in a colostrum-deprived calf model of RSV infection.
- Author
-
Sacco, Randy E., Mena, Ignacio, Palmer, Mitchell V., Durbin, Russell K., García-Sastre, Adolfo, and Durbin, Joan E.
- Subjects
- *
LUNGS , *ANIMAL diseases , *NEWCASTLE disease virus , *RESPIRATORY diseases , *VACCINE effectiveness , *RESPIRATORY syncytial virus , *IMMUNOGLOBULINS , *WOUND healing - 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. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
7. Interferon induction and function at the mucosal surface.
- Author
-
Durbin, Russell K., Kotenko, Sergei V., and Durbin, Joan E.
- Subjects
- *
VIRUS diseases , *INTERFERONS , *MUCOUS membranes , *TRANSCRIPTION factors , *INFLUENZA A virus , *IMMUNE response - 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/β) 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/β 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. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
8. Stat1 Phosphorylation Determines Ras Oncogenicity by Regulating p27Kip1.
- Author
-
Shuo Wang, Raven, Jennifer F., Durbin, Joan E., and Koromilas, Antonis E.
- Subjects
- *
GENE silencing , *COMPLEMENT inhibition , *PHOSPHORYLATION , *TRANSCRIPTION factors , *INTERFERONS , *ONCOGENIC viruses , *ANTINEOPLASTIC agents , *TYROSINE , *CANCER treatment - Abstract
Inactivation of p27Kip1 is implicated in tumorigenesis and has both prognostic and treatment-predictive values for many types of human cancer. The transcription factor Stat1 is essential for innate immunity and tumor immunosurveillance through its ability to act downstream of interferons. Herein, we demonstrate that Stat1 functions as a suppressor of Ras transformation independently of an interferon response. Inhibition of Ras transformation and tumorigenesis requires the phosphorylation of Stat1 at tyrosine 701 but is independent of Stat1 phosphorylation at serine 727. Stat1 induces p27Kip1 expression in Ras transformed cells at the transcriptional level through mechanisms that depend on Stat1 phosphorylation at tyrosine 701 and activation of Stat3. The tumor suppressor properties of Stat1 in Ras transformation are reversed by the inactivation of p27Kip1. Our work reveals a novel functional link between Stat1 and p27Kip1, which act in coordination to suppress the oncogenic properties of activated Ras. It also supports the notion that evaluation of Stat1 phosphorylation in human tumors may prove a reliable prognostic factor for patient outcome and a predictor of treatment response to anticancer therapies aimed at activating Stat1 and its downstream effectors. [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
9. Activation induced differential regulation of stem cell antigen-1 (Ly-6A/E) expression in murine B cells
- Author
-
Chen, Hui-Chen, Frissora, Frank, Durbin, Joan E., and Muthusamy, Natarajan
- Subjects
- *
STEM cells , *ANTIGENS , *B cells , *MESSENGER RNA , *RNA - Abstract
Expression of stem cell antigen-1 (Ly-6A/E) is developmentally regulated in murine B cells. However, little is known about its modulation during B cell activation. We report here the differential regulation of Ly-6A/E expression in response to diverse activation signals in mature B cells. Stimulation of resting B cells through the antigen receptor (BCR) inhibited, Ly-6A/E surface expression in dose dependent manner. Activation induced downregulation of Ly-6A/E is specific to BCR mediated signaling events as stimulation of B cells with anti-CD40, lipopolysaccharide or interferon-γ induced upregulation of Ly-6A/E surface expression. The activation induced differential modulation of Ly-6A/E expression is mediated at the mRNA levels. A role for BCR signaling in inhibition of Ly-6A/E expression was further confirmed using STAT-1−/− B cells, which expressed constitutive, but not inducible Ly-6A/E. The BCR induced inhibition of Ly-6A/E RNA and surface expression was mimicked by ionomycin, but not phorbol myristate acetate, indicating a role for calcium but not protein kinase C dependent signaling events. Inhibition of calcineurin reversed the BCR or ionomycin inhibited Ly-6A/E expression. Interestingly, in vitro differentiation analysis of Ly-6A/E+ and Ly-6A/E− splenic B cells revealed the Ly-6A/E+ cells to be the major source of antibody production, suggesting a potential role for Ly-6A/E in B cell differentiation. These studies provide the first evidence for activation induced differential modulation and differentiation of Ly-6A/E+ B cells. [Copyright &y& Elsevier]
- Published
- 2003
- Full Text
- View/download PDF
10. Transmission phenotype of Mycobacterium tuberculosis strains is mechanistically linked to induction of distinct pulmonary pathology.
- Author
-
Verma, Sheetal, Bhatt, Kamlesh, Lovey, Arianne, Ribeiro-Rodrigues, Rodrigo, Durbin, Joan, Jones-López, Edward C., Palaci, Moises, Vinhas, Solange A., Alland, David, Dietze, Reynaldo, Ellner, Jerrold J., and Salgame, Padmini
- Subjects
- *
MYCOBACTERIUM tuberculosis , *ACTINOBACTERIA , *IMMUNOLOGY , *TUBERCULIN , *PATHOLOGY - Abstract
In a study of household contacts (HHC), households were categorized into High (HT) and Low (LT) transmission groups based on the proportion of HHC with a positive tuberculin skin test. The Mycobacterium tuberculosis (Mtb) strains from HT and LT index cases of the households were designated Mtb-HT and Mtb-LT, respectively. We found that C3HeB/FeJ mice infected with Mtb-LT strains exhibited significantly higher bacterial burden compared to Mtb-HT strains and also developed diffused inflammatory lung pathology. In stark contrast, a significant number of mice infected with Mtb-HT strains developed caseating granulomas, a lesion type with high potential to cavitate. None of the Mtb-HT infected animals developed diffused inflammatory lung pathology. A link was observed between increased in vitro replication of Mtb-LT strains and their ability to induce significantly high lipid droplet formation in macrophages. These results support that distinct early interactions of Mtb-HT and Mtb-LT strains with macrophages and subsequent differential trajectories in pathological disease may be the mechanism underlying their transmission potential. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
11. A viral-vectored RSV vaccine induces long-lived humoral immunity in cotton rats.
- Author
-
Grieves, Jessica L., Yin, Zhiwei, Garcia-Sastre, Adolfo, Mena, Ignacio, Peeples, Mark E., Risman, Heidi P., Federman, Hannah, Sandoval, Marvin J., Durbin, Russell K., and Durbin, Joan E.
- Subjects
- *
RESPIRATORY syncytial virus infection vaccines , *AIRWAY (Anatomy) , *HUMORAL immunity , *INFANT diseases , *COTTON rats as laboratory animals - 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. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
12. Distinct Roles of Type I and Type III Interferons in Intestinal Immunity to Homologous and Heterologous Rotavirus Infections.
- Author
-
Lin, Jian-Da, Feng, Ningguo, Sen, Adrish, Balan, Murugabaskar, Tseng, Hsiang-Chi, McElrath, Constance, Smirnov, Sergey V., Peng, Jianya, Yasukawa, Linda L., Durbin, Russell K., Durbin, Joan E., Greenberg, Harry B., and Kotenko, Sergei V.
- Subjects
- *
TYPE I interferons , *ROTAVIRUS diseases , *CELLULAR signal transduction , *IMMUNOREGULATION , *GASTROENTEROLOGY , *VIRAL replication - Abstract
Type I (IFN-α/β) 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. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
13. Respiratory syncytial virus infection enhances Pseudomonas aeruginosa biofilm growth through dysregulation of nutritional immunity.
- Author
-
Hendricks, Matthew R., Lashua, Lauren P., Fischer, Douglas K., Flitter, Becca A., Eichinger, Katherine M., Durbin, Joan E., Sarkar, Saumendra N., Coyne, Carolyn B., Empey, Kerry M., and Bomberger, Jennifer M.
- Subjects
- *
RESPIRATORY syncytial virus infections , *CYSTIC fibrosis , *PSEUDOMONAS aeruginosa , *BIOFILMS , *EPITHELIUM - 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 co-culture 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. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
14. Inflammatory impact of IFN-γ in CD8+ T cell-mediated lung injury is mediated by both Stat1-dependent and -independent pathways.
- Author
-
Ramana, Chilakamarti V., DeBerge, Matthew P., Kumar, Aseem, Alia, Christopher S., Durbin, Joan E., and Enelow, Richard I.
- Subjects
- *
INTERFERONS , *LUNG diseases , *IMMUNE response , *CHEMOKINES , *STAT proteins , *CELLULAR signal transduction , *PHYSIOLOGY - 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. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
15. T Cell-Derived IL-17 Mediates Epithelial Changes in the Airway and Drives Pulmonary Neutrophilia.
- Author
-
Fogli, Laura K., Sundrud, Mark S., Goel, Swati, Bajwa, Sofia, Jensen, Kari, Derudder, Emmanuel, Sun, Amy, Coffre, Maryaline, Uyttenhove, Catherine, Van Snick, Jacques, Schmidt-Supprian, Marc, Rao, Anjana, Grunig, Gabriele, Durbin, Joan, Casola, Stefano S., Rajewsky, Klaus, and Koralov, Sergei B.
- Subjects
- *
ASTHMA treatment , *T helper cells , *INTERLEUKIN-17 , *NEUTROPHILS , *STAT proteins , *CHEMOKINES , *IMMUNOREGULATION - Abstract
Th17 cells are a proinflammatory subset of effector T cells that have been implicated in the pathogenesis of asthma. Their production of the cytokine IL-17 is known to induce local recruitment of neutrophils, but the direct impact of IL-17 on the lung epithelium is poorly understood. In this study, we describe a novel mouse model of spontaneous IL-17-driven lung inflammation that exhibits many similarities to asthma in humans. We have found that STAT3 hyperactivity in T lymphocytes causes an expansion of Th17 cells, which home preferentially to the lungs. IL-17 secretion then leads to neutrophil infiltration and lung epithelial changes, in turn leading to a chronic inflammatory state with increased mucus production and decreased lung function. We used this model to investigate the effects of IL-17 activity on airway epithelium and identified CXCL5 and MIP-2 as important factors in neutrophil recruitment. The neutralization of IL-17 greatly reduces pulmonary neutrophilia, underscoring a key role for IL-17 in promoting chronic airway inflammation. These findings emphasize the role of IL-17 in mediating neutrophil-driven pulmonary inflammation and highlight a new mouse model that may be used for the development of novel therapies targeting Th17 cells in asthma and other chronic pulmonary diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
16. Stat1 is a suppressor of ErbB2/Neu-mediated cellular transformation and mouse mammary gland tumor formation.
- Author
-
Raven, Jennifer F., Williams, Virginie, Shuo Wang, Tremblay, Michel L., Muller, William J., Durbin, Joan E., and Koromilas, Antonis E.
- Published
- 2011
- Full Text
- View/download PDF
17. Lambda Interferon Is the Predominant Interferon Induced by Influenza A Virus Infection In Vivo.
- Author
-
Jewell, Nancy A., Cline, Troy, Mertz, Sara E., Smirnov, Sergey V., Flanõ, Emilio, Schindler, Christian, Grieves, Jessica L., Durbin, Russell K., Kotenko, Sergei V., and Durbin, Joan E.
- Subjects
- *
INTERFERONS , *INFLUENZA A virus , *VIRUS diseases , *HOSTS (Biology) , *LABORATORY mice - Abstract
The type I alpha/beta interferons (IFN-α/β) 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/β 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/β signaling in combating systemic disease. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
18. Signal transducer and activator of transcription 1 in T cells plays an indispensable role in immunity to Leishmania major by mediating Th1 cell homing to the site of infection.
- Author
-
Barbi, Joseph, Snider, Heidi M., Bhardwaj, Neeti, Lezama-Dávila, Claudio M., Durbin, Joan E., and Satoskar, Abhay R.
- Subjects
- *
GENETIC transcription , *LEISHMANIASIS , *INTERFERONS , *T cells , *CD4 antigen - Abstract
The signal transducer and activator of transcription 1 (STAT1) signaling pathway mediates the biological functions of IFN-γ. 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. Rang2-/- 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 Thyl.1+ and STAT1-/- Thyl.2+ T cells showed that STAT1 in CD4+ T cells was not required for Thl 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. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
19. Recombinant parainfluenza virus 5 (PIV5) expressing the influenza A virus hemagglutinin provides immunity in mice to influenza A virus challenge
- Author
-
Tompkins, S. Mark, Lin, Yuan, Leser, George P., Kramer, Kari A., Haas, Debra L., Howerth, Elizabeth W., Xu, Jie, Kennett, Mary J., Durbin, Russell K., Durbin, Joan E., Tripp, Ralph, Lamb, Robert A., and He, Biao
- Subjects
- *
PARAINFLUENZA viruses , *SIMIAN viruses , *RNA viruses , *INFLUENZA A virus - Abstract
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. [Copyright &y& Elsevier]
- Published
- 2007
- Full Text
- View/download PDF
20. IL-13 is associated with reduced illness and replication in primary respiratory syncytial virus infection in the mouse
- Author
-
Zhou, Weisong, Hashimoto, Koichi, Moore, Martin L., Elias, Jack A., Zhu, Zhou, Durbin, Joan, Colasurdo, Giuseppe, Rutigliano, John A., Chiappetta, Constance L., Goleniewska, Kasia, O'Neal, Jamye F., Graham, Barney S., and Stokes Peebles, R.
- Subjects
- *
THERAPEUTIC use of cytokines , *CELLULAR immunity , *RESPIRATORY syncytial virus , *MICE - Abstract
Abstract: The role of IL-13 in respiratory syncytial virus (RSV) immunopathogenesis is incompletely described. To assess the effect of IL-13 on primary RSV infection, transgenic mice which either overexpress IL-13 in the lung (IL-13 OE) or non-transgenic littermates (IL-13 NT) were challenged intranasally with RSV. IL-13 OE mice had significantly decreased peak viral titers four days after infection compared to non-transgenic littermates. In addition, IL-13 OE mice had significantly lower RSV-induced weight loss and reduced lung IFN-γ protein expression compared with IL-13 NT mice. In contrast, primary RSV challenge of IL-13 deficient mice resulted in a small, but statistically significant increase in viral titers on day four after infection, no difference in RSV-induced weight loss compared to wild type mice, and augmented IFN-γ production on day 6 after infection. In STAT1-deficient (STAT1 KO) mice, where primary RSV challenge produced high levels of IL-13 production in the lungs, treatment with an IL-13 neutralizing protein resulted in greater peak viral titers both four and six days after RSV and greater RSV-induced weight loss compared to mice treated with a control protein. These results suggest that IL-13 modulates illness from RSV-infection. [Copyright &y& Elsevier]
- Published
- 2006
- Full Text
- View/download PDF
21. Protection against Respiratory Syncytial Virus by a Recombinant Newcastle Disease Virus Vector.
- Author
-
Martinez-Sobrido, Luis, Gitiban, Negin, Fernandez-Sesma, Ana, Cros, Jerome, Mertz, Sara E., Jewell, Nancy A., Hammond, Sue, Flano, Emilio, Durbin, Russell K., Garcia-Sastre, Adolfo, and Durbin, Joan E.
- Subjects
- *
RESPIRATORY syncytial virus , *PARAMYXOVIRUSES , *NEWCASTLE disease , *NEWCASTLE disease virus , *VIRUS diseases in poultry , *RESPIRATORY infections - 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-α/β) 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-α/β receptor. [ABSTRACT FROM AUTHOR]
- Published
- 2006
- Full Text
- View/download PDF
22. Chinchilla and Murine Models of Upper Respiratory Tract Infections with Respiratory Syncytial Virus.
- Author
-
Gitiban, Negin, Jurcisek, Joseph A., Harris, Randall H., Mertz, Sara E., Durbin, Russell K., Bakaletz, Lauren O., and Durbin, Joan E.
- Subjects
- *
RESPIRATORY infections , *RESPIRATORY syncytial virus , *CHINCHILLAS , *IMMUNITY , *EAR diseases , *OTITIS media - 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. [ABSTRACT FROM AUTHOR]
- Published
- 2005
- Full Text
- View/download PDF
23. VSV strains with defects in their ability to shutdown innate immunity are potent systemic anti-cancer agents
- Author
-
Stojdl, David F., Lichty, Brian D., tenOever, Benjamin R., Paterson, Jennifer M., Power, Anthony T., Knowles, Shane, Marius, Ricardo, Reynard, Jennifer, Poliquin, Laurent, Atkins, Harold, Brown, Earl G., Durbin, Russell K., Durbin, Joan E., Hiscott, John, and Bell, John C.
- Subjects
- *
CANCER , *METASTASIS , *PATIENTS , *CANCER cells , *XENOGRAFTS , *OVARIAN cancer , *COLON cancer - 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. [Copyright &y& Elsevier]
- Published
- 2003
- Full Text
- View/download PDF
24. The antitumor effects of IFN-α are abrogated in a STAT1-deficient mouse.
- Author
-
Lesinski, Gregory B., Anghelina, Mirela, Zimmerer, Jason, Bakalakos, Timothy, Badgwell, Brian, Parihar, Robin, Yan Hu, Beckell, Brian, Abood, Gerard, Chaudhury, Abhik Ray, Magro, Cynthia, Durbin, Joan, and Carson III, William E.
- Subjects
- *
MELANOMA , *TUMORS , *CELL lines , *NEUROENDOCRINE tumors , *LYMPHOBLASTOID cell lines - Abstract
Presents a study that investigated the contribution of signal transducer of transcription (STAT)1 signaling within the tumor by generating a STAT1-deficient melanoma cell line, AGS-1. Methodology; Characterization of a STAT1-deficient murine melanoma cell line AGS-1; Reconstitution of STAT1 signaling within the AGS-1cell line.
- Published
- 2003
- Full Text
- View/download PDF
25. The antitumor effects of IFN-alpha are abrogated in a STAT1-deficient mouse.
- Author
-
Lesinski, Gregory B, Anghelina, Mirela, Zimmerer, Jason, Bakalakos, Timothy, Badgwell, Brian, Parihar, Robin, Hu, Yan, Becknell, Brian, Abood, Gerard, Chaudhury, Abhik Ray, Magro, Cynthia, Durbin, Joan, and Carson, William E 3rd
- Subjects
- *
ANIMAL experimentation , *ANTINEOPLASTIC agents , *CARRIER proteins , *CELL division , *CELL lines , *CELLULAR signal transduction , *COMPARATIVE studies , *DOSE-effect relationship in pharmacology , *FLOW cytometry , *GENES , *GENETIC techniques , *IMMUNOBLOTTING , *IMMUNOHISTOCHEMISTRY , *KILLER cells , *RESEARCH methodology , *MEDICAL cooperation , *MICE , *PROTEINS , *RESEARCH , *RETROVIRUSES , *SPLEEN , *TIME , *DNA-binding proteins , *EVALUATION research , *CANCER cell culture , *PHARMACODYNAMICS - Abstract
IFN-alpha activates the signal transducer and activator of transcription (STAT) family of proteins; however, it is unknown whether IFN-alpha exerts its antitumor actions primarily through a direct effect on malignant cells or by stimulating the immune system. To investigate the contribution of STAT1 signaling within the tumor, we generated a STAT1-deficient melanoma cell line, AGS-1. We reconstituted STAT1 into AGS-1 cells by retroviral gene transfer. The resulting cell line (AGS-1STAT1) showed normal regulation of IFN-alpha-stimulated genes (e.g., H2k, ISG-54) as compared with AGS-1 cells infected with the empty vector (AGS-1MSCV). However, mice challenged with the AGS-1, AGS-1STAT1, and AGS-1MSCV cell lines exhibited nearly identical survival in response to IFN-alpha treatment, indicating that restored STAT1 signaling within the tumor did not augment the antitumor activity of IFN-alpha. In contrast, STAT1-/- mice could not utilize exogenous IFN-alpha to inhibit the growth of STAT1+/+ melanoma cells in either an intraperitoneal tumor model or in the adjuvant setting. The survival of tumor-bearing STAT1-/- mice was identical regardless of treatment (IFN-alpha or PBS). Additional cell depletion studies demonstrated that NK cells mediated the antitumor effects of IFN-alpha. Thus, STAT1-mediated gene regulation within immune effectors was necessary for mediating the antitumor effects of IFN-alpha in this experimental system. [ABSTRACT FROM AUTHOR]
- Published
- 2003
- Full Text
- View/download PDF
26. Recovery of Paramyxovirus Simian Virus 5 with a V Protein Lacking the Conserved Cysteine-rich Domain: The Multifunctional V Protein Blocks both Interferon-β Induction and Interferon Signaling
- Author
-
He, Biao, Paterson, Reay G., Stock, Nicola, Durbin, Joan E., Durbin, Russell K., Goodbourn, Stephen, Randall, Richard E., and Lamb, Robert A.
- Subjects
- *
PARAMYXOVIRUSES , *SIMIAN viruses - 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 (rSV5VΔC). In Vero cells rSV5VΔC forms large plaques and grows at a rate and titer similar to those of rSV5. In BHK or CV-1 cells rSV5VΔC forms small plaques and grows poorly. However, even when grown in Vero cells rSV5VΔC 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), rSV5VΔC 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, rSV5VΔC 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-β. The transcription factor IRF-3 which is required for transcription of the IFN-β gene is not translocated from the cytoplasm to the nucleus in rSV5-infected cells. In contrast, in rSV5VΔC-infected cells IRF-3 is localized predominantly in the nucleus and IFN-β 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. [Copyright &y& Elsevier]
- Published
- 2002
- Full Text
- View/download PDF
27. Interferon α/β-mediated inhibition and promotion of interferon γ: STAT1 resolves a paradox.
- Author
-
Nguyen, Khuong B., Cousens, Leslie P., Doughty, Lesley A., Pien, Gary C., Durbin, Joan E., and Biron, Christine A.
- Subjects
- *
INTERFERONS , *VIRUS diseases , *TRANSCRIPTION factors - Abstract
Induction of high systemic levels of type 1 interferons (IFNs) IFN-α and IFN-β is a hallmark of many viral infections. In addition to their potent antiviral effects, these cytokines mediate a number of immunoregulatory functions and can promote IFN-γ expression in T cells. However, during viral infections of mice IFN-γ production is not always observed at the same time as systemic IFN-α/β production and when, elicited at these times, is IFN-α/β?independent. We demonstrate that type 1 interferons not only fail to induce, but also act to inhibit, IFN-γ expression by both NK and T cells. The mechanism of inhibition is dependent upon the IFN-α/β receptor and the signal transducer and activator of transcription 1 (STAT1). In the absence of STAT1, not only are the IFN-α/β?mediated inhibitory effects completely abrogated, but the cytokines themselves can induce IFN-γ expression. These results indicate that endogenous biochemical pathways are in place to negatively regulate NK and T cell IFN-γ expression elicited by IFN-α/β or other stimuli, at times of innate responses to viral infections. They also show that type 1 interferon signaling can occur through STAT1-dependent and independent mechanisms and suggest that efficient induction of IFN-γ expression by IFN-α/β requires STAT1 regulation. Such immunoregulatory pathways may be critical for shaping the endogenous innate and virus-specific adaptive immune responses to viral infections. [ABSTRACT FROM AUTHOR]
- Published
- 2000
- Full Text
- View/download PDF
28. Type III IFNs: Beyond antiviral protection.
- Author
-
Kotenko, Sergei V., Rivera, Amariliz, Parker, Dane, and Durbin, Joan E.
- Subjects
- *
INTERFERON inducers , *IMMUNE response - 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. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
29. Correction: Distinct Roles of Type I and Type III Interferons in Intestinal Immunity to Homologous and Heterologous Rotavirus Infections.
- Author
-
Lin, Jian-Da, Feng, Ningguo, Sen, Adrish, Balan, Murugabaskar, Tseng, Hsiang-Chi, McElrath, Constance, Smirnov, Sergey V., Peng, Jianya, Yasukawa, Linda L., Durbin, Russell K., Durbin, Joan E., Greenberg, Harry B., and Kotenko, Sergei V.
- Subjects
- *
INTERFERONS , *ROTAVIRUS diseases , *IMMUNITY - Abstract
A correction to the article "Distinct Roles of Type I and Type III Interferons in Intestinal Immunity to Homologous and Heterologous Rotavirus Infections" is presented.
- Published
- 2016
- Full Text
- View/download PDF
30. ID: 115: Role of type I and type III interferons in intestinal immunity to homologous and heterologous rotavirus infection.
- Author
-
Lin, Jian-Da, Feng, Ningguo, Sen, Adrish, Balan, Murugabaskar, Tseng, Hsiang-Chi, McElrath, Constance, Durbin, Russell K., Greenberg, Harry B., Durbin, Joan E., and Kotenko, Sergei V.
- Subjects
- *
ROTAVIRUS diseases , *INTERLEUKIN-18 , *CYTOKINES , *GENOMES , *INTERFERONS - Abstract
Types I and III IFNs exert common antiviral activities through distinct receptors, and their relative importance for antiviral protection of different organ remains to be fully explored. Mouse strains deficient in type-specific IFN signaling were used to dissect distinct and overlapping contributions of type I and III IFNs to protection against homologous murine (EW-strain) and heterologous simian (RRV-strain) rotavirus infection of the intestine. EW-RV replicated to similar levels in IFN- λ R, IFN- α R, IFN- α / λ R, IFN- α / γ R and STAT1 deficient suckling mice between 2–7 days post-infection (dpi); and only a moderate increase in fecal shedding was observed in IFN- λ R and STAT1 knock-out (KO) mice at 8–9 dpi. In contrast, Rag2 KO mice showed chronic viral shedding, demonstrating that the homologous EW-RV efficiently antagonizes murine IFN responses, with viral clearance dependent upon adaptive immune responses. Unlike EW-RV, replication of the heterologous RRV was significantly increased in gastro-intestinal tract of either IFN- λ R or IFN- α R deficient mice, and was further enhanced in doubly deficient IFN- α / λ R KO animals, demonstrating the sensitivity of RRV to IFN-mediated antiviral protection, and revealing important, and cooperative roles for both type I and type III IFNs in limiting RRV replication. Similarly, both type I and III IFNs helped to limit extra-intestinal RRV spread, whereas type I IFNs were essential for controlling RRV replication in mesenteric lymph nodes. Overall, these results demonstrate a murine host-specific inhibition of IFN’s antiviral activity by EW-RV, but cooperative action by type I and type III IFNs in limiting simian RRV growth and spread in mice. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
31. 224: Resistance to respiratory syncytial virus replication is STAT1 and STAT2 dependent, but I interferon independent.
- Author
-
Cline, Troy, Lin, Jian-Da, Kotenko, Sergei V., Durbin, Russell K., and Durbin, Joan E.
- Subjects
- *
RESPIRATORY syncytial virus , *VIRAL replication , *GENETIC code , *INTERFERONS , *INFLUENZA A virus , *DISEASE susceptibility , *LABORATORY mice - Abstract
Despite the known ability of type I IFNs to inhibit virus replication, we and others noted that mice lacking the IFN-αβ Receptor (IFNAR−/−) did not show increased sensitivity to either influenza A virus (IAV) or respiratory syncytial virus (RSV) infection. An explanation for this seemingly contradictory result was offered by the discovery of type III or IFN-λ, a unique family of cytokines that bind a distinct receptor but can also promote interferon stimulated gene factor 3 (ISGF3) formation and the transcriptional upregulation of IFN-stimulated genes. The presence of robust IFN-λ induction in IAV-infected IFNAR−/− mice can explain their resistance to infection, but a distinct protective mechanism is at work in RSV-infected IFNAR−/− animals. In the absence of the IFNAR, no type III IFN RNA or protein can be detected in RSV-infected animals, nor is there any evidence of ISGF3 formation or upregulation of the ISGF3 dependent gene, Mx1. Nonetheless, we observe enhanced virus susceptibility to RSV in mice lacking either STAT1 or STAT2. We therefore conclude from our studies that (1) triggering of IFN-λ synthesis is pathogen dependent: induction of IFN-λ by RSV, but not by IAV, is dependent on functional type I IFN signaling, and (2) resistance to RSV is mediated by a novel STAT1/STAT2 containing complex. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
32. Plasticity and Virus Specificity of the Airway Epithelial Cell Immune Response during Respiratory Virus Infection.
- Author
-
Ioannidis, Ioannis, McNally, Beth, Willette, Meredith, Peeples, Mark E., Chaussabel, Damien, Durbin, Joan E., Ramilo, Octavio, Mejias, Asuncion, and Flañoa, Emilio
- Subjects
- *
EPITHELIAL cells , *IMMUNE response , *RESPIRATORY infections , *AIRWAY (Anatomy) , *OLIGONUCLEOTIDES , *PROTEIN microarrays , *GENE expression in viruses , *INFLUENZA viruses - 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. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
33. PS1-114 Immune stimulation by a newcastle disease virus vectored vaccine
- Author
-
Grieves, Jessica, Durbin, Russell, Garcia-Saster, Adolfo, and Durbin, Joan
- Published
- 2011
- Full Text
- View/download PDF
34. CS03-2. Differential negative regulation of type I and type III interferons by UBP43 underlies extended antiviral effects of interferon-lambdas in epithelial cells
- Author
-
Smirnov, Sergey V., Burkart, Christoph, Li, Wei, Lewis-Antes, Anita, Durbin, Joan, Zhang, Dong-Er, and Kotenko, Sergei V.
- Published
- 2011
- Full Text
- View/download PDF
35. SS7-1 Differential negative regulation of type I and type III interferons underlies extended antiviral effects of interferon-lambdas
- Author
-
Smirnov, Sergey V., Burkart, Christoph, Li, Wei, Lewis-Antes, Anita, Durbin, Joan, Zhang, Dong-Er, and Kotenko, Sergei V.
- Published
- 2010
- Full Text
- View/download PDF
36. Differential Type I Interferon Induction by Respiratory Syncytial Virus and Influenza A Virus In Vivo.
- Author
-
Jewell, Nancy A., Vaghefi, Negin, Mertz, Sara E., Akter, Parvis, Peebles Jr., R. Stokes, Bakaletz, Lauren O., Durbin, Russell K., Flaño, Emilio, and Durbin, Joan E.
- Subjects
- *
INTERFERONS , *VIRUSES , *INFLUENZA A virus , *CYTOKINES , *NUCLEIC acids , *VIRUS diseases , *EPITHELIAL cells - 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- α/β 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-α/β 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-α/β in response to either infection, but in pDC-depleted animals only type I IFN induction by influenza virus was impaired. [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
37. Type I Interferon Inhibition and Dendritic Cell Activation during Gammaherpesvirus Respiratory Infection.
- Author
-
Weslow-Schmidt, Janet L., Jewell, Nancy A., Mertz, Sara E., Simas, J. Pedro, Durbin, Joan E., and Flaño, Emilio
- Subjects
- *
INTERFERONS , *DENDRITIC cells , *RESPIRATORY infections , *HERPESVIRUSES , *ANTIVIRAL agents - Abstract
The respiratory tract is a major mucosal site for microorganism entry into the body, and type I interferon (IFN) and dendritic cells constitute a first line of defense against viral infections. We have analyzed the interaction between a model DNA virus, plasmacytoid dendritic cells, and type I IFN during lung infection of mice. Our data show that murine gammaherpesvirus 68 (γ HV68) inhibits type I IFN secretion by dendritic cells and that plasmacytoid dendritic cells are necessary for conventional dendritic cell maturation in response to γ HV68. Following γ HV68 intranasal inoculation, the local and systemic IFN- α/β response is below detectable levels, and plasmacytoid dendritic cells are activated and recruited into the lung with a tissue distribution that differs from that of conventional dendritic cells. Our results suggest that plasmacytoid dendritic cells and type I IFN have important but independent roles during the early response to a respiratory γ HV68 infection. γ HV68 infection inhibits type I IFN production by dendritic cells and is a poor inducer of IFN- α/β in vivo, which may serve as an immune evasion strategy. [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
38. 65 Differential Type I Interferon Induction by Respiratory Viruses In Vivo
- Author
-
Jewell, Nancy A., Vaghefi, Negin, Mertz, Sara E., Akter, Parvis, Peebles, R. Stokes, Bakaletz, Lauren O., Durbin, Russell K., Flaño, Emilio, and Durbin, Joan E.
- Published
- 2007
- Full Text
- View/download PDF
39. 24 Interferon-independent, STAT1-mediated Inhibition of Respiratory Syncytial Virus Replication
- Author
-
Cline, Troy D., Mertz, Sara E., Durbin, Russell K., and Durbin, Joan E.
- Published
- 2007
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.