Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
PLoS One ; 16(10): e0255309, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34618816

RESUMEN

BACKGROUND: Type III interferon, or interferon lambda (IFNλ) is a crucial antiviral cytokine induced by influenza infection. While IFNλ is important for anti-viral host defense, published data demonstrate that IFNλ is pathogenic during influenza/bacterial super-infection. It is known that polymorphisms in specific IFNλ genes affect influenza responses, but the effect of IFNλ subtypes on bacterial super-infection is unknown. METHODS: Using an established model of influenza, Staphylococcus aureus super-infection, we studied IFNλ3-/- and control mice to model a physiologically relevant reduction in IFNλ and to address its role in super-infection. RESULTS: Surprisingly, IFNλ3-/- mice did not have significantly lower total IFNλ than co-housed controls, and displayed no change in viral or bacterial clearance. Importantly, both control and IFNλ3-/- mice displayed a positive correlation between viral burden and total IFNλ in the bronchoalveolar lavage during influenza/bacterial super-infection, suggesting that higher influenza viral burden drives a similar total IFNλ response regardless of IFNλ3 gene integrity. Interestingly, total IFNλ levels positively correlated with bacterial burden, while viral burden and bronchoalveolar lavage cellularity did not. CONCLUSIONS: These data suggest IFNλ2 can compensate for IFNλ3 to mount an effective antiviral and defense, revealing a functional redundancy in these highly similar IFNλ subtypes. Further, the IFNλ response to influenza, as opposed to changes in cellular inflammation or viral load, significantly correlates with susceptibility to bacterial super-infection. Moreover, the IFNλ response is regulated and involves redundant subtypes, suggesting it is of high importance to pulmonary pathogen defense.


Asunto(s)
Interferones/análisis , Interferones/inmunología , Interleucinas/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Animales , Línea Celular , Coinfección/inmunología , Coinfección/microbiología , Perros , Femenino , Interferones/genética , Interleucinas/genética , Células de Riñón Canino Madin Darby , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/patología , Polimorfismo Genético/genética , Infecciones Estafilocócicas/prevención & control , Sobreinfección/inmunología , Sobreinfección/microbiología , Carga Viral/inmunología , Interferón lambda
2.
Front Immunol ; 11: 574027, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33101299

RESUMEN

Asthma is a highly prevalent, chronic respiratory disease that impacts millions of people worldwide and causes thousands of deaths every year. Asthmatics display different phenotypes with distinct genetic components, environmental causes, and immunopathologic signatures, and are broadly characterized into type 2-high or type 2-low (non-type 2) endotypes by linking clinical characteristics, steroid responsiveness, and molecular pathways. Regardless of asthma severity and adequate disease management, patients may experience acute exacerbations of symptoms and a loss of disease control, often triggered by respiratory infections. The interferon (IFN) family represents a group of cytokines that play a central role in the protection against and exacerbation of various infections and pathologies, including asthma. Type I and III IFNs in particular play an indispensable role in the host immune system to fight off pathogens, which seems to be altered in both pediatric and adult asthmatics. Impaired IFN production leaves asthmatics susceptible to infection and with uncontrolled type 2 immunity, promotes airway hyperresponsiveness (AHR), and inflammation which can lead to asthma exacerbations. However, IFN deficiency is not observed in all asthmatics, and alterations in IFN expression may be independent of type 2 immunity. In this review, we discuss the link between type I and III IFNs and asthma both in general and in specific contexts, including during viral infection, co-infection, and bacterial/fungal infection. We also highlight several studies which examine the potential role for type I and III IFNs as asthma-related therapies.


Asunto(s)
Asma/inmunología , Interferones/inmunología , Interferones/uso terapéutico , Asma/tratamiento farmacológico , Asma/etiología , Bacterias/patogenicidad , Hongos/patogenicidad , Humanos , Inmunomodulación , Pulmón/inmunología , Infecciones del Sistema Respiratorio/complicaciones , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Infecciones del Sistema Respiratorio/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Virus/patogenicidad
3.
Mucosal Immunol ; 12(5): 1231-1243, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31296910

RESUMEN

Secondary bacterial pneumonia is a significant complication of severe influenza infection and Staphylococcus aureus and Streptococcus pneumoniae are the primary pathogens of interest. IL-22 promotes S. aureus and S. pneumoniae host defense in the lung through epithelial integrity and induction of antimicrobial peptides and is inhibited by the soluble decoy receptor IL-22-binding protein (IL-22BP). Little is known about the effect of the IL-22/IL-22BP regulatory pathway on lung infection, and it has not been studied in the setting of super-infection. We exposed wild-type and IL-22BP-/- mice to influenza A/PR/8/34 for 6 days prior to infection with S. aureus (USA300) S. pneumoniae. Super-infected IL-22BP-/- mice had decreased bacterial burden and improved survival compared to controls. IL-22BP-/- mice exhibited decreased inflammation, increased lipocalin 2 expression, and deletion of IL-22BP was associated with preserved epithelial barrier function with evidence of improved tight junction stability. Human bronchial epithelial cells treated with IL-22Fc showed evidence of improved tight junctions compared to untreated cells. This study revealed that IL-22BP-/- mice are protected during influenza, bacterial super-infection, suggesting that IL-22BP has a pro-inflammatory role and impairs epithelial barrier function likely through interaction with IL-22.


Asunto(s)
Infecciones Bacterianas/metabolismo , Infecciones Bacterianas/microbiología , Proteínas Portadoras/metabolismo , Interleucinas/metabolismo , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/virología , Sobreinfección , Animales , Infecciones Bacterianas/genética , Infecciones Bacterianas/patología , Carga Bacteriana , Barrera Alveolocapilar/metabolismo , Barrera Alveolocapilar/patología , Barrera Alveolocapilar/virología , Proteínas Portadoras/genética , Modelos Animales de Enfermedad , Expresión Génica , Interleucinas/genética , Recuento de Leucocitos , Masculino , Ratones , Ratones Noqueados , Monocitos/inmunología , Monocitos/metabolismo , Monocitos/patología , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/patología , Permeabilidad , Unión Proteica , Staphylococcus aureus , Streptococcus pneumoniae , Uniones Estrechas , Interleucina-22
4.
Viruses ; 11(6)2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-31159430

RESUMEN

Influenza virus is among the most common causes of respiratory illness worldwide and can be complicated by secondary bacterial pneumonia, a frequent cause of mortality. When influenza virus infects the lung, the innate immune response is activated, and interferons and inflammatory mediators are released. This "cytokine storm" is thought to play a role in influenza-induced lung pathogenesis. Peroxisome proliferator-activated receptor gamma (PPARγ) is a member of the nuclear hormone receptor super-family. PPARγ has numerous functions including enhancing lipid and glucose metabolism and cellular differentiation and suppressing inflammation. Synthetic PPARγagonists (thiazolidinediones or glitazones) have been used clinically in the treatment of type II diabetes. Using data from the National Health and Nutrition Examination Survey (NHANES), diabetic participants taking rosiglitazone had an increased risk of mortality from influenza/pneumonia compared to those not taking the drug. We examined the effect of rosiglitazone treatment during influenza and secondary bacterial (Methicillin resistant Staphylococcus aureus) pneumonia in mice. We found decreased influenza viral burden, decreased numbers of neutrophils and macrophages in bronchoalveolar lavage, and decreased production of cytokines and chemokines in influenza infected, rosiglitazone-treated mice when compared to controls. However, rosiglitazone treatment compromised bacterial clearance during influenza-bacterial super-infection. Both human and mouse data suggest that rosiglitazone treatment worsens the outcome of influenza-associated pneumonia.


Asunto(s)
Infecciones Bacterianas , Coinfección/tratamiento farmacológico , Inflamación/patología , Gripe Humana , Rosiglitazona/efectos adversos , Animales , Infecciones Bacterianas/complicaciones , Infecciones Bacterianas/tratamiento farmacológico , Infecciones Bacterianas/inmunología , Coinfección/inmunología , Coinfección/microbiología , Coinfección/virología , Citocinas/efectos de los fármacos , Citocinas/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/microbiología , Diabetes Mellitus Tipo 2/virología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación/genética , Inflamación/microbiología , Inflamación/virología , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H1N1 del Virus de la Influenza A/crecimiento & desarrollo , Subtipo H1N1 del Virus de la Influenza A/inmunología , Gripe Humana/complicaciones , Gripe Humana/tratamiento farmacológico , Gripe Humana/inmunología , Interferones/efectos de los fármacos , Interferones/metabolismo , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Pulmón/virología , Masculino , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/crecimiento & desarrollo , Staphylococcus aureus Resistente a Meticilina/inmunología , Ratones , PPAR gamma/agonistas , Rosiglitazona/farmacología , Transducción de Señal/efectos de los fármacos , Carga Viral/efectos de los fármacos
5.
Clin Microbiol Rev ; 32(3)2019 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-31142498

RESUMEN

Community-acquired pneumonia (CAP) is a leading cause of morbidity and mortality worldwide. Despite broad literature including basic and translational scientific studies, many gaps in our understanding of host-pathogen interactions remain. In this review, pathogen virulence factors that drive lung infection and injury are discussed in relation to their associated host immune pathways. CAP epidemiology is considered, with a focus on Staphylococcus aureus and Streptococcus pneumoniae as primary pathogens. Bacterial factors involved in nasal colonization and subsequent virulence are illuminated. A particular emphasis is placed on bacterial pore-forming toxins, host cell death, and inflammasome activation. Identified host-pathogen interactions are then examined by linking pathogen factors to aberrant host response pathways in the context of acute lung injury in both primary and secondary infection. While much is known regarding bacterial virulence and host immune responses, CAP management is still limited to mostly supportive care. It is likely that improvements in therapy will be derived from combinatorial targeting of both pathogen virulence factors and host immunomodulation.


Asunto(s)
Infecciones Comunitarias Adquiridas/inmunología , Infecciones Comunitarias Adquiridas/microbiología , Interacciones Huésped-Patógeno/inmunología , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/microbiología , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Humanos , Staphylococcus aureus/patogenicidad , Staphylococcus aureus/fisiología , Streptococcus pneumoniae/patogenicidad , Streptococcus pneumoniae/fisiología
6.
Infect Immun ; 87(5)2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30804099

RESUMEN

Influenza kills 30,000 to 40,000 people each year in the United States and causes 10 times as many hospitalizations. A common complication of influenza is bacterial superinfection, which exacerbates morbidity and mortality from the viral illness. Recently, methicillin-resistant Staphylococcus aureus (MRSA) has emerged as the dominant pathogen found in bacterial superinfection, with Streptococcus pneumoniae a close second. However, clinicians have few tools to treat bacterial superinfection. Current therapy for influenza/bacterial superinfection consists of treating the underlying influenza infection and adding various antibiotics, which are increasingly rendered ineffective by rising bacterial multidrug resistance. Several groups have recently proposed the use of the antiviral cytokine interferon lambda (IFN-λ) as a therapeutic for influenza, as administration of pegylated IFN-λ improves lung function and survival during influenza by reducing the overabundance of neutrophils in the lung. However, our data suggest that therapeutic IFN-λ impairs bacterial clearance during influenza superinfection. Specifically, mice treated with an adenoviral vector to overexpress IFN-λ during influenza infection exhibited increased bacterial burdens upon superinfection with either MRSA or S. pneumoniae Surprisingly, adhesion molecule expression, antimicrobial peptide production, and reactive oxygen species activity were not altered by IFN-λ treatment. However, neutrophil uptake of MRSA and S. pneumoniae was significantly reduced upon IFN-λ treatment during influenza superinfection in vivo Together, these data support the theory that IFN-λ decreases neutrophil motility and function in the influenza-infected lung, which increases the bacterial burden during superinfection. Thus, we believe that caution should be exercised in the possible future use of IFN-λ as therapy for influenza.


Asunto(s)
Antivirales/uso terapéutico , Gripe Humana/complicaciones , Gripe Humana/tratamiento farmacológico , Interferones/uso terapéutico , Staphylococcus aureus/efectos de los fármacos , Streptococcus pneumoniae/efectos de los fármacos , Sobreinfección/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Infecciones Estafilocócicas/patología , Sobreinfección/etiología , Estados Unidos
7.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA