Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 9: 2151, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30337919

RESUMEN

Influenza is a common respiratory virus that infects between 5 and 20% of the US population and results in 30,000 deaths annually. A primary cause of influenza-associated death is secondary bacterial pneumonia. We have previously shown that influenza induces type I interferon (IFN)-mediated inhibition of Type 17 immune responses, resulting in exacerbation of bacterial burden during influenza and Staphylococcus aureus super-infection. In this study, we investigated the role of STAT2 signaling during influenza and influenza-bacterial super-infection in mice. Influenza-infected STAT2-/- mice had increased morbidity, viral burden, and inflammation when compared to wild-type mice. Despite an exaggerated inflammatory response to influenza infection, we found increased bacterial control and survival in STAT2 deficient mice during influenza-MRSA super-infection compared to controls. Further, we found that increased bacterial clearance during influenza-MRSA super-infection is not due to rescue of Type 17 immunity. Absence of STAT2 was associated with increased accumulation of M1, M2 and M1/M2 co-expressing macrophages during influenza-bacterial super-infection. Neutralization of IFNγ (M1) and/or Arginase 1 (M2) impaired bacterial clearance in Stat2-/- mice during super-infection, demonstrating that pulmonary macrophages expressing a mixed M1/M2 phenotype promote bacterial control during influenza-bacterial super-infection. Together, these results suggest that the STAT2 signaling is involved in suppressing macrophage activation and bacterial control during influenza-bacterial super-infection. Further, these studies reveal novel mechanistic insight into the roles of macrophage subpopulations in pulmonary host defense.


Asunto(s)
Gripe Humana/inmunología , Macrófagos Alveolares/inmunología , Neumonía Estafilocócica/inmunología , Factor de Transcripción STAT2/metabolismo , Sobreinfección/inmunología , Animales , Trasplante de Médula Ósea , Embrión de Pollo , Modelos Animales de Enfermedad , Femenino , Humanos , Subtipo H1N1 del Virus de la Influenza A/inmunología , Gripe Humana/diagnóstico , Gripe Humana/microbiología , Gripe Humana/mortalidad , Activación de Macrófagos/inmunología , Macrófagos Alveolares/metabolismo , Masculino , Células Madre Mesenquimatosas , Staphylococcus aureus Resistente a Meticilina/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neumonía Estafilocócica/diagnóstico , Neumonía Estafilocócica/microbiología , Neumonía Estafilocócica/mortalidad , Cultivo Primario de Células , Factor de Transcripción STAT2/genética , Factor de Transcripción STAT2/inmunología , Índice de Severidad de la Enfermedad , Transducción de Señal/inmunología , Sobreinfección/diagnóstico , Sobreinfección/microbiología , Sobreinfección/mortalidad , Quimera por Trasplante
2.
Sci Rep ; 7: 43139, 2017 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-28233801

RESUMEN

Atopic asthma is a prevalent respiratory disease that is characterized by inflammation, mucus hypersecretion, and airway hyperresponsiveness. The complexity of this heterogeneous disorder has commanded the need to better define asthma phenotypes based on underlying molecular mechanisms of disease. Although classically viewed as a type 2-regulated disease, type 17 helper T (Th17) cells are known to be influential in asthma pathogenesis, predominantly in asthmatics with neutrophilia and severe refractory disease. Bromodomain and extra-terminal domain (BET) chromatin adaptors serve as immunomodulators by directly regulating Th17 responses and Th17-mediated pathology in murine models of autoimmunity and infection. Based on this, we hypothesized that BET proteins may also play an essential role in neutrophil-dominant allergic airway disease. Using a murine model of neutrophil-dominant allergic airway disease, we demonstrate that BET inhibition limits pulmonary inflammation and alters the Th17-related inflammatory milieu in the lungs. In addition, inhibition of BET proteins improved lung function (specifically quasi-static lung compliance and tissue elastance) and reduced mucus production in airways. Overall, these studies show that BET proteins may have a critical role in asthma pathogenesis by altering type 17 inflammation, and thus interfering with BET-dependent chromatin signaling may provide clinical benefits to patients suffering from asthma.


Asunto(s)
Asma/patología , Neutrófilos/inmunología , Factores de Transcripción/antagonistas & inhibidores , Animales , Modelos Animales de Enfermedad , Inflamación/patología , Pulmón/patología , Ratones , Proteínas del Tejido Nervioso/metabolismo , Receptores de Superficie Celular/metabolismo , Células Th17/inmunología , Factores de Transcripción/metabolismo
3.
Immunohorizons ; 1(6): 81-91, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-29577113

RESUMEN

Influenza is an annual, global health care concern. Secondary bacterial pneumonia is a severe complication associated with primary influenza virus infection, often resulting in critical morbidity and mortality. Our laboratory has identified influenza-induced suppression of anti-bacterial Type 17 immunity as a mechanism for enhanced susceptibility to bacterial super-infection. We have shown that influenza-induced type I interferon impairs Type 17 activation. STAT1 is a transcription factor involved in interferon signaling, shared by type I, II, and III interferon. In this work, we investigated the role of STAT1 signaling during influenza, methicillin-resistant Staphylococcus aureus (MRSA) super-infection. STAT1-/- mice had increased morbidity and airway inflammation compared to control mice during influenza mono-infection. Despite this worsened anti-viral response, STAT1-/- mice were protected from super-infection bacterial burden and mortality compared to controls. Type 17 immune activation was increased in lymphocytes in STAT1-/- mice during super-infection. The elevation in Type 17 immunity was not related to increased IL-23 production, as type I interferon could inhibit IL-23 expression in a STAT1 independent manner. STAT1-/- antigen presenting cells were inherently biased towards Type 17 polarization compared to control cells. Further, STAT1-/- dendritic cells produced attenuated IL-6 and TNFα upon heat-killed S. aureus stimulation compared to control. Overall, these data indicate that STAT1 signaling plays a detrimental role in influenza, MRSA super-infection by controlling the magnitude of Type 17 immune activation.

4.
J Immunol ; 196(3): 963-77, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26729801

RESUMEN

IL-13 and IL-17A, produced mainly by Th2 and Th17 cells, respectively, have an influential role in asthma pathogenesis. We examined the role of IL-13 and IL-17A in mediating airway hyperresponsiveness (AHR), lung inflammation, and mucus metaplasia in a dual Th2/Th17 model of asthma. IL-13 and/or IL-17A were neutralized using mAbs. Th2/Th17 adoptive transfer induced a mixed asthma phenotype characterized by elevated eosinophilia and neutrophilia, tissue inflammation, mucus metaplasia, and AHR that were partially reversible with steroid treatment. Pulmonary inflammation and quasi-static lung compliance were largely unaffected by neutralization of IL-13 and/or IL-17A. However, neutralization of IL-13 alone or in combination with IL-17A significantly attenuated AHR and mucus metaplasia. Further, STAT6 activation was attenuated following IL-13 and IL-13/IL-17A Ab treatment. We next assessed the role of STAT6 in Th2/Th17-mediated allergic airway disease using STAT6(-/-) mice. STAT6(-/-) mice adoptively transferred with Th2/Th17 cells had decreased AHR compared with controls. These data suggest that IL-13 drives AHR and mucus metaplasia in a STAT6-dependent manner, without directly contributing to airway or tissue inflammation. IL-17A independently contributes to AHR, but it only partially mediates inflammation and mucus metaplasia in a mixed Th2/Th17 model of steroid-resistant asthma.


Asunto(s)
Asma/inmunología , Interleucina-13/inmunología , Interleucina-17/inmunología , Hipersensibilidad Respiratoria/inmunología , Traslado Adoptivo , Animales , Asma/patología , Modelos Animales de Enfermedad , Resistencia a Medicamentos , Immunoblotting , Metaplasia/inmunología , Metaplasia/patología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones SCID , Ratones Transgénicos , Moco/inmunología , Reacción en Cadena de la Polimerasa , Hipersensibilidad Respiratoria/patología , Células Th17/inmunología , Células Th2/inmunología , Transcriptoma
5.
Am J Physiol Lung Cell Mol Physiol ; 309(2): L158-67, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-26001778

RESUMEN

Suppression of type 17 immunity by type I interferon (IFN) during influenza A infection has been shown to enhance susceptibility to secondary bacterial pneumonia. Although this mechanism has been described in coinfection with gram-positive bacteria, it is unclear whether similar mechanisms may impair lung defense against gram-negative infections. Furthermore, precise delineation of the duration of type I IFN-associated susceptibility to bacterial infection remains underexplored. Therefore, we investigated the effects of preceding influenza A virus infection on subsequent challenge with the gram-negative bacteria Escherichia coli or Pseudomonas aeruginosa and the temporal association between IFN expression with susceptibility to Staphylococcus aureus challenge in a mouse model of influenza and bacterial coinfection. Here we demonstrate that preceding influenza A virus led to increased lung E. coli and P. aeruginosa bacterial burden, which was associated with suppression of type 17 immunity and attenuation of antimicrobial peptide expression. Enhanced susceptibility to S. aureus coinfection ceased at day 14 of influenza infection, when influenza-associated type I IFN levels had returned to baseline levels, further suggesting a key role for type I IFN in coinfection pathogenesis. These findings further implicate type I IFN-associated suppression of type 17 immunity and antimicrobial peptide production as a conserved mechanism for enhanced susceptibility to both gram-positive and gram-negative bacterial coinfection during influenza infection.


Asunto(s)
Infecciones por Escherichia coli/microbiología , Virus de la Influenza A/patogenicidad , Infecciones por Orthomyxoviridae/microbiología , Neumonía Bacteriana/microbiología , Neumonía/microbiología , Receptor de Interferón alfa y beta/fisiología , Infecciones Estafilocócicas/microbiología , Animales , Péptidos Catiónicos Antimicrobianos/metabolismo , Coinfección/inmunología , Coinfección/microbiología , Coinfección/virología , Susceptibilidad a Enfermedades , Escherichia coli/inmunología , Escherichia coli/patogenicidad , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/virología , Virus de la Influenza A/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Neumonía/inmunología , Neumonía/virología , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/virología , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/virología , Staphylococcus aureus/inmunología , Staphylococcus aureus/patogenicidad
6.
Respir Res ; 16: 10, 2015 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-25651926

RESUMEN

BACKGROUND: Influenza is a common respiratory virus and Staphylococcus aureus frequently causes secondary pneumonia during influenza infection, leading to increased morbidity and mortality. Influenza has been found to attenuate subsequent Type 17 immunity, enhancing susceptibility to secondary bacterial infections. IL-27 is known to inhibit Type 17 immunity, suggesting a potential critical role for IL-27 in viral and bacterial co-infection. METHODS: A murine model of influenza and Staphylococcus aureus infection was used to mimic human viral, bacterial co-infection. C57BL/6 wild-type, IL-27 receptor α knock-out, and IL-10 knock-out mice were infected with Influenza H1N1 (A/PR/8/34) or vehicle for 6 days followed by challenge with Staphylococcus aureus or vehicle for 24 hours. Lung inflammation, bacterial burden, gene expression, and cytokine production were determined. RESULTS: IL-27 receptor α knock-out mice challenged with influenza A had increased morbidity compared to controls, but no change in viral burden. IL-27 receptor α knock-out mice infected with influenza displayed significantly decreased IL-10 production compared to wild-type. IL-27 receptor α knock-out mice co-infected with influenza and S. aureus had improved bacterial clearance compared to wild-type controls. Importantly, there were significantly increased Type 17 responses and decreased IL-10 production in IL-27 receptor α knock-out mice. Dual infected IL-10-/- mice had significantly less bacterial burden compared to dual infected WT mice. CONCLUSIONS: These data reveal that IL-27 regulates enhanced susceptibility to S. aureus pneumonia following influenza infection, potentially through the induction of IL-10 and suppression of IL-17.


Asunto(s)
Coinfección , Subtipo H1N1 del Virus de la Influenza A/inmunología , Interleucinas/inmunología , Pulmón/inmunología , Infecciones por Orthomyxoviridae/inmunología , Neumonía Estafilocócica/inmunología , Staphylococcus aureus/inmunología , Animales , Carga Bacteriana , Modelos Animales de Enfermedad , Interacciones Huésped-Patógeno , Inmunidad Celular , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Interleucina-10/deficiencia , Interleucina-10/genética , Interleucina-17/inmunología , Pulmón/microbiología , Pulmón/virología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/virología , Neumonía Estafilocócica/genética , Neumonía Estafilocócica/microbiología , Receptores de Citocinas/deficiencia , Receptores de Citocinas/genética , Receptores de Interleucina , Staphylococcus aureus/patogenicidad , Células Th17/inmunología , Células Th17/microbiología , Células Th17/virología , Factores de Tiempo , Carga Viral
7.
Artículo en Inglés | MEDLINE | ID: mdl-27011794

RESUMEN

BACKGROUND: Patients with cystic fibrosis (CF) complicated by allergic bronchopulmonary aspergillosis (ABPA) are vitamin D deficient and in vitro treatment with 1,25 (OH)2 vitamin D3 of CD4+ cells from CF patients with ABPA decreases Aspergillus fumigatus(Af)-induced Th2 responses. This Phase I clinical trial investigated the safety and effectiveness of daily vitamin D3 supplementation in CF patients with ABPA to reduce allergic responses and ABPA symptoms, and increase serum vitamin D levels. METHODS: Seven patients ages 12 years and older with a clinical diagnosis of CF and ABPA with current evidence of Af sensitization received 4000 IU vitamin D3 (cholecalciferol) daily for 24 weeks. The primary outcome of the study was safety followed by the Aspergillus induced IL-13 response in CD4+ T cells to test the hypothesis that vitamin D supplementation is safe and reduces Aspergillus induced IL-13 responses in CD4+ T cells. Secondary outcomes included total IgE, Aspergillus-specific IgE, vitamin D levels, FEV1, urinary calcium/creatinine ratio, and cytokine production by Aspergillus-stimulated peripheral blood T cells. RESULTS: Six months of vitamin D3 supplementation resulted in significant increases in serum 25-(OH) vitamin D level, and the treatment was well tolerated without evidence of vitamin D toxicity or hypercalcemia. There were no serious adverse events. Daily vitamin D supplementation led to significantly decreased Aspergillus induced IL-13 responses between the baseline visit and that at 24 weeks (p = 0.04). Aspergillus-specific IgE level was also significantly decreased after 8 (p = 0.035) and 24 weeks of daily vitamin D supplementation (p = 0.04). CONCLUSIONS: 4000 IU vitamin D3 daily over a 24-week period is well tolerated in CF patients with a history ABPA and current evidence of Th2 immunity to Af. . Daily vitamin D supplementation was associated with reduced Aspergillus induced IL-13 responses from peripheral. . CD4+ T cells and Aspergillus-specific IgE levels, as well as increased serum vitamin D levels. This treatment was well tolerated and the study supports further investigation of the use of vitamin D supplementation in Th2 mediated diseases. TRIAL REGISTRATION: This trial was registered at www.clinicaltrials.gov as NCT01222273.

8.
J Infect Dis ; 209(6): 865-75, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24072844

RESUMEN

Influenza A represents a significant cause of morbidity and mortality worldwide. Bacterial complications of influenza A confer the greatest risk to patients. TH17 pathway inhibition has been implicated as a mechanism by which influenza A alters bacterial host defense. Here we show that preceding influenza causes persistent Staphylococcus aureus infection and suppression of TH17 pathway activation in mice. Influenza does not inhibit S. aureus binding and uptake by phagocytic cells but instead attenuates S. aureus induced TH17 related antimicrobial peptides necessary for bacterial clearance in the lung. Importantly, exogenous lipocalin 2 rescued viral exacerbation of S. aureus infection and decreased free iron levels in the bronchoalveolar lavage from mice coinfected with S. aureus and influenza. These findings indicate a novel mechanism by which influenza A inhibits TH17 immunity and increases susceptibility to secondary bacterial pneumonia. Identification of new mechanisms in the pathogenesis of bacterial pneumonia could lead to future therapeutic targets.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/metabolismo , Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/microbiología , Neumonía Estafilocócica/microbiología , Staphylococcus aureus/inmunología , Análisis de Varianza , Animales , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/microbiología , Líquido del Lavado Bronquioalveolar/virología , Coinfección/microbiología , Coinfección/virología , Interacciones Huésped-Patógeno/inmunología , Virus de la Influenza A/patogenicidad , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Neutrófilos/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Neumonía Estafilocócica/inmunología , Neumonía Estafilocócica/virología , Staphylococcus aureus/patogenicidad , Células Th17
9.
PLoS One ; 8(12): e82865, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24324838

RESUMEN

Influenza viruses pose a significant health risk and annually impose a great cost to patients and the health care system. The molecular determinants of influenza severity, often exacerbated by secondary bacterial infection, are largely unclear. We generated a novel outbred mouse model of influenza virus, Staphylococcus aureus, and co-infection utilizing influenza A/CA/07/2009 virus and S. aureus (USA300). Outbred mice displayed a wide range of pathologic phenotypes following influenza virus or co-infection ranging broadly in severity. Influenza viral burden positively correlated with weight loss although lung histopathology did not. Inflammatory cytokines including IL-6, TNF-α, G-CSF, and CXCL10 positively correlated with both weight loss and viral burden. In S. aureus infection, IL-1ß, G-CSF, TNF-α, and IL-6 positively correlated with weight loss and bacterial burden. In co-infection, IL-1ß production correlated with decreased weight loss suggesting a protective role. The data demonstrate an approach to identify biomarkers of severe disease and to understand pathogenic mechanisms in pneumonia.


Asunto(s)
Coinfección , Virus de la Influenza A , Infecciones por Orthomyxoviridae/virología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus , Animales , Animales no Consanguíneos , Carga Bacteriana , Embrión de Pollo , Citocinas/biosíntesis , Citocinas/genética , Modelos Animales de Enfermedad , Expresión Génica , Subtipo H1N1 del Virus de la Influenza A , Pulmón/microbiología , Pulmón/patología , Pulmón/virología , Masculino , Ratones , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/patología , Fenotipo , Neumonía/genética , Neumonía/microbiología , Neumonía/patología , Neumonía/virología , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/metabolismo , Infecciones Estafilocócicas/patología , Carga Viral , Pérdida de Peso
10.
J Immunol ; 191(10): 5153-9, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24089191

RESUMEN

Pneumonia is a leading cause of death worldwide. Staphylococcal aureus can be a cause of severe pneumonia alone or a common pathogen in secondary pneumonia following influenza. Recently, we reported that preceding influenza attenuated the Type 17 pathway, increasing the lung's susceptibility to secondary infection. IL-1ß is known to regulate host defense, including playing a role in Th17 polarization. We examined whether IL-1ß signaling is required for S. aureus host defense and whether influenza infection impacted S. aureus-induced IL-1ß production and subsequent Type 17 pathway activation. Mice were challenged with S. aureus (USA 300), with or without preceding Influenza A/PR/8/34 H1N1 infection. IL-1R1(-/-) mice had significantly higher S. aureus burden, increased mortality, and decreased Type 17 pathway activation following S. aureus challenge. Coinfected mice had significantly decreased IL-1ß production versus S. aureus infection alone at early time points following bacterial challenge. Preceding influenza did not attenuate S. aureus-induced inflammasome activation, but there was early suppression of NF-κB activation, suggesting an inhibition of NF-κB-dependent transcription of pro-IL-1ß. Furthermore, overexpression of IL-1ß in influenza and S. aureus-coinfected mice rescued the induction of IL-17 and IL-22 by S. aureus and improved bacterial clearance. Finally, exogenous IL-1ß did not significantly rescue S. aureus host defense during coinfection in IL-17RA(-/-) mice or in mice in which IL-17 and IL-22 activity were blocked. These data reveal a novel mechanism by which Influenza A inhibits S. aureus-induced IL-1ß production, resulting in attenuation of Type 17 immunity and increased susceptibility to bacterial infection.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/inmunología , Interleucina-1beta/metabolismo , Infecciones por Orthomyxoviridae/inmunología , Neumonía Estafilocócica/inmunología , Staphylococcus aureus/inmunología , Animales , Carga Bacteriana/genética , Carga Bacteriana/inmunología , Coinfección/inmunología , Coinfección/microbiología , Coinfección/virología , Activación Enzimática/inmunología , Inflamasomas/inmunología , Interleucina-17/biosíntesis , Interleucina-1beta/biosíntesis , Interleucinas/biosíntesis , Pulmón/inmunología , Pulmón/microbiología , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Receptores Tipo I de Interleucina-1/deficiencia , Receptores Tipo I de Interleucina-1/genética , Transducción de Señal , Interleucina-22
11.
Am J Pathol ; 182(4): 1286-96, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23490254

RESUMEN

Influenza infection is widespread in the United States and the world. Despite low mortality rates due to infection, morbidity is common and little is known about the molecular events involved in recovery. Influenza infection results in persistent distal lung remodeling, and the mechanism(s) involved are poorly understood. Recently IL-22 has been found to mediate epithelial repair. We propose that IL-22 is critical for recovery of normal lung function and architecture after influenza infection. Wild-type and IL-22(-/-) mice were infected with influenza A PR8/34 H1N1 and were followed up for up to 21 days post infection. IL-22 receptor was localized to the airway epithelium in naive mice but was expressed at the sites of parenchymal lung remodeling induced by influenza infection. IL-22(-/-) mice displayed exacerbated lung injury compared with wild-type mice, which correlated with decreased lung function 21 days post infection. Epithelial metaplasia was observed in wild-type mice but was not evident in IL-22(-/-) animals that were characterized with an increased fibrotic phenotype. Gene expression analysis revealed aberrant expression of epithelial genes involved in repair processes, among changes in several other biological processes. These data indicate that IL-22 is required for normal lung repair after influenza infection. IL-22 represents a novel pathway involved in interstitial lung disease.


Asunto(s)
Epitelio/patología , Epitelio/virología , Interleucinas/metabolismo , Pulmón/patología , Pulmón/virología , Infecciones por Orthomyxoviridae/patología , Cicatrización de Heridas , Animales , Membrana Basal/metabolismo , Membrana Basal/patología , Colágeno/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Células Epiteliales/virología , Epitelio/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Interleucinas/deficiencia , Pulmón/fisiopatología , Metaplasia , Ratones , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/fisiopatología , Infecciones por Orthomyxoviridae/virología , Receptores de Interleucina/metabolismo , Pruebas de Función Respiratoria , Transducción de Señal/genética , Interleucina-22
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...