Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Sci Rep ; 12(1): 22552, 2022 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-36581658

RESUMEN

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.


Asunto(s)
Enfermedades de los Bovinos , Infecciones por Virus Sincitial Respiratorio , Vacunas contra Virus Sincitial Respiratorio , Virus Sincitial Respiratorio Bovino , Virus Sincitial Respiratorio Humano , Femenino , Embarazo , Animales , Bovinos , Humanos , Anciano , Virus de la Enfermedad de Newcastle , Calostro , Vacunas contra Virus Sincitial Respiratorio/genética , Anticuerpos Antivirales , Enfermedades de los Bovinos/prevención & control
2.
Nat Commun ; 12(1): 2624, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33976143

RESUMEN

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


Asunto(s)
Colitis Ulcerosa/inmunología , Interferones/metabolismo , Mucosa Intestinal/patología , Repitelización/inmunología , Animales , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/patología , Sulfato de Dextran/administración & dosificación , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Células Epiteliales , Femenino , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Masculino , Ratones , Ratones Noqueados , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Receptores de Interferón/genética , Receptores de Interferón/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Organismos Libres de Patógenos Específicos
3.
J Exp Med ; 217(5)2020 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-32289152

RESUMEN

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


Asunto(s)
Betacoronavirus/fisiología , Infecciones por Coronavirus/metabolismo , Interferones/metabolismo , Neumonía Viral/metabolismo , COVID-19 , Humanos , Pandemias , SARS-CoV-2 , Internalización del Virus
4.
Semin Immunol ; 43: 101303, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31771761

RESUMEN

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.


Asunto(s)
Infecciones Bacterianas/inmunología , Interferón Tipo I/metabolismo , Interferones/metabolismo , Microbiota/inmunología , Micosis/inmunología , Virosis/inmunología , Animales , Humanos , Inmunidad , Inflamación , Transducción de Señal , Interferón lambda
5.
PLoS Pathog ; 15(3): e1007613, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30840702

RESUMEN

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.


Asunto(s)
Mycobacterium tuberculosis/metabolismo , Tuberculosis Pulmonar/transmisión , Virulencia/genética , Animales , Modelos Animales de Enfermedad , Transmisión de Enfermedad Infecciosa , Femenino , Granuloma , Pulmón/patología , Macrófagos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/patogenicidad , Fenotipo , Tuberculosis/etiología , Tuberculosis Pulmonar/etiología , Virulencia/fisiología
6.
Vaccine ; 36(26): 3842-3852, 2018 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-29779923

RESUMEN

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.


Asunto(s)
Inmunidad Humoral , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Interferón gamma/metabolismo , Pulmón/virología , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Vacunas contra Virus Sincitial Respiratorio/aislamiento & purificación , Virus Sincitiales Respiratorios/aislamiento & purificación , Sigmodontinae , Factores de Tiempo , Carga Viral
7.
Sci Immunol ; 2(16)2017 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-28986419

RESUMEN

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


Asunto(s)
Inmunidad Innata , Interferones/inmunología , Infecciones Fúngicas Invasoras/inmunología , Inmunidad Adaptativa , Animales , Aspergillus fumigatus/crecimiento & desarrollo , Aspergillus fumigatus/inmunología , Aspergillus fumigatus/patogenicidad , Línea Celular , Humanos , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Interferón-alfa/administración & dosificación , Interferón-alfa/inmunología , Interferones/administración & dosificación , Interferones/genética , Interferones/metabolismo , Infecciones Fúngicas Invasoras/microbiología , Ratones , Monocitos/inmunología , Neutrófilos/inmunología , Receptores CCR2/deficiencia , Receptores CCR2/inmunología , Receptores de Citocinas/genética , Receptores de Citocinas/metabolismo , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Receptores de Interferón/inmunología , Interferón lambda
8.
J Biol Chem ; 292(18): 7295-7303, 2017 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-28289095

RESUMEN

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


Asunto(s)
Inmunidad Adaptativa , Inmunidad Innata , Interleucinas/inmunología , Virosis/inmunología , Animales , Humanos , Interferón Tipo I/inmunología , Interferones
10.
PLoS Pathog ; 12(4): e1005600, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-27128797

RESUMEN

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.


Asunto(s)
Interferón Tipo I/inmunología , Interferón gamma/inmunología , Intestinos/inmunología , Infecciones por Rotavirus/inmunología , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Humanos , Immunoblotting , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena de la Polimerasa , Rotavirus
11.
Proc Natl Acad Sci U S A ; 113(6): 1642-7, 2016 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-26729873

RESUMEN

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


Asunto(s)
Biopelículas/crecimiento & desarrollo , Inmunidad , Fenómenos Fisiológicos de la Nutrición , Pseudomonas aeruginosa/fisiología , Infecciones por Virus Sincitial Respiratorio/patología , Virus Sincitiales Respiratorios/fisiología , Animales , Antivirales/farmacología , Bronquios/patología , Líquido del Lavado Bronquioalveolar , Fibrosis Quística/microbiología , Fibrosis Quística/patología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/microbiología , Células Epiteliales/virología , Homeostasis/efectos de los fármacos , Humanos , Interferón beta/farmacología , Hierro/farmacología , Ratones , Interacciones Microbianas/efectos de los fármacos , Modelos Biológicos , Pseudomonas aeruginosa/efectos de los fármacos , Virus Sincitiales Respiratorios/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transferrina/metabolismo
12.
Comp Med ; 65(4): 315-26, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26310461

RESUMEN

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.


Asunto(s)
Asma/virología , Pulmón/virología , Neumonía Viral/virología , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitiales Respiratorios/patogenicidad , Sigmodontinae/virología , Animales , Asma/inmunología , Asma/patología , Bronquiolitis/virología , Líquido del Lavado Bronquioalveolar/virología , Modelos Animales de Enfermedad , Exposición por Inhalación , Pulmón/inmunología , Pulmón/patología , Mucosa Nasal/virología , Neumonía Viral/inmunología , Neumonía Viral/patología , Eosinofilia Pulmonar/virología , Mucosa Respiratoria/virología , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/patología , Virus Sincitiales Respiratorios/inmunología , Factores de Tiempo , Replicación Viral
13.
Curr Opin Virol ; 13: 117-22, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26176495

RESUMEN

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.


Asunto(s)
Modelos Animales de Enfermedad , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/fisiología , Animales , Humanos , Virus Sincitial Respiratorio Humano/genética
14.
Cell Host Microbe ; 17(4): 429-40, 2015 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-25816775

RESUMEN

Resistance and tolerance are two defense strategies employed by the host against microbial threats. Autophagy-mediated degradation of bacteria has been extensively described as a major resistance mechanism. Here we find that the dominant function of autophagy proteins during infections with the epidemic community-associated methicillin-resistant Staphylococcus aureus USA300 is to mediate tolerance rather than resistance. Atg16L1 hypomorphic mice (Atg16L1(HM)), which have reduced autophagy, were highly susceptible to lethality in both sepsis and pneumonia models of USA300 infection. Autophagy confers protection by limiting the damage caused by α-toxin, particularly to endothelial cells. Remarkably, Atg16L1(HM) mice display enhanced survival rather than susceptibility upon infection with α-toxin-deficient S. aureus. These results identify an essential role for autophagy in tolerance to Staphylococcal disease and highlight how a single virulence factor encoded by a pathogen can determine whether a given host factor promotes tolerance or resistance.


Asunto(s)
Autofagia , Toxinas Bacterianas/inmunología , Proteínas Hemolisinas/inmunología , Tolerancia Inmunológica , Staphylococcus aureus Resistente a Meticilina/inmunología , Animales , Proteínas Relacionadas con la Autofagia , Proteínas Portadoras/genética , Ratones Noqueados , Neumonía Estafilocócica/inmunología , Neumonía Estafilocócica/microbiología , Sepsis/inmunología , Sepsis/microbiología , Análisis de Supervivencia
15.
Am J Physiol Lung Cell Mol Physiol ; 308(7): L650-7, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25617378

RESUMEN

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.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Linfocitos T CD8-positivos/inmunología , Interferón gamma/fisiología , Factor de Transcripción STAT1/metabolismo , Lesión Pulmonar Aguda/virología , Animales , Linfocitos T CD8-positivos/metabolismo , Células Cultivadas , Ratones Endogámicos BALB C , Ratones Noqueados , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/metabolismo , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal
16.
Immunol Rev ; 255(1): 25-39, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23947345

RESUMEN

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.


Asunto(s)
Interferones/inmunología , Interferones/metabolismo , Membrana Mucosa/inmunología , Membrana Mucosa/metabolismo , Animales , Regulación de la Expresión Génica , Humanos , Quinasas Janus/metabolismo , Ligandos , Membrana Mucosa/virología , Fosforilación , Biosíntesis de Proteínas , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Receptores Toll-Like/metabolismo , Virosis/genética , Virosis/inmunología , Virosis/metabolismo , Virus/inmunología
17.
J Immunol ; 191(6): 3100-11, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23966625

RESUMEN

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.


Asunto(s)
Asma/inmunología , Enfermedades del Sistema Inmune/inmunología , Interleucina-17/inmunología , Trastornos Leucocíticos/inmunología , Neutrófilos/inmunología , Mucosa Respiratoria/inmunología , Animales , Asma/metabolismo , Separación Celular , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Interleucina-17/metabolismo , Ratones , Ratones Endogámicos C57BL , Neumonía/inmunología , Neumonía/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Mucosa Respiratoria/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Transfección
18.
J Virol ; 86(10): 5422-36, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22398282

RESUMEN

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.


Asunto(s)
Células Epiteliales/inmunología , Virus de la Influenza A/inmunología , Gripe Humana/virología , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitiales Respiratorios/inmunología , Infecciones del Sistema Respiratorio/virología , Animales , Células Cultivadas , Citocinas/inmunología , Células Epiteliales/virología , Femenino , Humanos , Lactante , Virus de la Influenza A/fisiología , Gripe Humana/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/fisiología , Infecciones del Sistema Respiratorio/inmunología , Especificidad de la Especie
19.
Cell Cycle ; 10(5): 794-804, 2011 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-21311224

RESUMEN

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


Asunto(s)
Transformación Celular Neoplásica , Neoplasias Mamarias Experimentales/metabolismo , Receptor ErbB-2/metabolismo , Factor de Transcripción STAT1/metabolismo , Animales , Antineoplásicos/farmacología , Línea Celular , Chlorocebus aethiops , Progresión de la Enfermedad , Femenino , Gefitinib , Humanos , Lapatinib , Glándulas Mamarias Animales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Transgénicos , Fosforilación , Quinazolinas/farmacología , Receptor ErbB-2/antagonistas & inhibidores , Transducción de Señal , Trasplante Heterólogo
20.
Curr Opin Virol ; 1(6): 476-86, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22323926

RESUMEN

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


Asunto(s)
Inmunidad Innata/inmunología , Interferón Tipo I/inmunología , Virosis/inmunología , Virus/inmunología , Animales , Regulación Viral de la Expresión Génica , Humanos , Interferón Tipo I/biosíntesis , Interferón Tipo I/genética , Transducción de Señal , Factores de Transcripción/inmunología , Ubiquitina/inmunología , Virosis/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...