Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Innate Immun ; 12(1): 103-115, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-30928973

RESUMEN

The negative immune regulator Tollip inhibits the proinflammatory response to rhinovirus (RV) infection, a contributor to airway neutrophilic inflammation and asthma exacerbations, but the underlying molecular mechanisms are poorly understood. Tollip may inhibit IRAK1, a signaling molecule downstream of ST2, the receptor of IL-33. This study was carried out to determine whether Tollip downregulates ST2 signaling via inhibition of IRAK1, but promotes soluble ST2 (sST2) production, thereby limiting excessive IL-8 production in human airway epithelial cells during RV infection in a type 2 cytokine milieu (e.g., IL-13 and IL-33 stimulation). Tollip- and IRAK1-deficient primary human tracheobronchial epithelial (HTBE) cells and Tollip knockout (KO) HTBE cells were generated using the shRNA knockdown and CRISPR/Cas9 approaches, respectively. Cells were stimulated with IL-13, IL-33, and/or RV16. sST2, activated IRAK1, and IL-8 were measured. A Tollip KO mouse model was utilized to test if Tollip regulates the airway inflammatory response to RV infection in vivo under IL-13 and IL-33 treatment. Following IL-13, IL-33, and RV treatment, Tollip-deficient (vs. -sufficient) HTBE cells produced excessive IL-8, accompanied by decreased sST2 production but increased IRAK1 activation. IL-8 production following IL-13/IL-33/RV exposure was markedly attenuated in IRAK1-deficient HTBE cells, as well as in Tollip KO HTBE cells treated with an IRAK1 inhibitor or a recombinant sST2 protein. Tollip KO (vs. wild-type) mice developed exaggerated airway neutrophilic responses to RV in the context of IL-13 and IL-33 treatment. Collectively, these data demonstrate that Tollip restricts excessive IL-8 production in type 2 cytokine-exposed human airways during RV infection by promoting sST2 production and inhibiting IRAK1 activation. sST2 and IRAK1 may be therapeutic targets for attenuating excessive neutrophilic airway inflammation in asthma, especially during RV infection.


Asunto(s)
Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neutrófilos/inmunología , Infecciones por Picornaviridae/metabolismo , Mucosa Respiratoria/metabolismo , Rhinovirus/fisiología , Células Th2/inmunología , Células Cultivadas , Citocinas/metabolismo , Humanos , Inmunomodulación , Quinasas Asociadas a Receptores de Interleucina-1/genética , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Activación Neutrófila , Infecciones por Picornaviridae/inmunología , ARN Interferente Pequeño/genética , Mucosa Respiratoria/patología , Transducción de Señal
2.
JCI Insight ; 4(8)2019 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-30996135

RESUMEN

Bacterial permeability family member A1 (BPIFA1), also known as short palate, lung, and nasal epithelium clone 1 (SPLUNC1), is a protein involved in the antiinflammatory response. The goal of this study was to determine whether BPIFA1 expression in asthmatic airways is regulated by genetic variations, altering epithelial responses to type 2 cytokines (e.g., IL-13). Nasal epithelial cells from patients with mild to severe asthma were collected from the National Heart, Lung, and Blood Institute Severe Asthma Research Program centers, genotyped for rs750064, and measured for BPIFA1. To determine the function of rs750064, cells were cultured at air-liquid interface and treated with IL-13 with or without recombinant human BPIFA1 (rhBPIFA1). Noncultured nasal cells with the rs750064 CC genotype had significantly less BPIFA1 mRNA expression than the CT and TT genotypes. Cultured CC versus CT and TT cells without stimulation maintained less BPIFA1 expression. With IL-13 treatment, CC genotype cells secreted more eotaxin-3 than CT and TT genotype cells. Also, rhBPIFA1 reduced IL-13-mediated eotaxin-3. BPIFA1 mRNA levels negatively correlated with serum IgE and fractional exhaled nitric oxide. Baseline FEV1% levels were lower in the asthma patients with the CC genotype (n = 1,016). Our data suggest that less BPIFA1 in asthma patients with the CC allele may predispose them to greater eosinophilic inflammation, which could be attenuated by rhBPIFA1 protein therapy.


Asunto(s)
Asma/genética , Células Epiteliales/inmunología , Regulación de la Expresión Génica/inmunología , Glicoproteínas/genética , Fosfoproteínas/genética , Transducción de Señal/inmunología , Adolescente , Adulto , Anciano , Alelos , Asma/diagnóstico , Asma/tratamiento farmacológico , Asma/inmunología , Células Cultivadas , Quimiocina CCL26/inmunología , Quimiocina CCL26/metabolismo , Niño , Eosinófilos/inmunología , Células Epiteliales/patología , Femenino , Volumen Espiratorio Forzado , Predisposición Genética a la Enfermedad , Glicoproteínas/metabolismo , Glicoproteínas/farmacología , Glicoproteínas/uso terapéutico , Humanos , Interleucina-13/inmunología , Interleucina-13/metabolismo , Masculino , Persona de Mediana Edad , Mucosa Nasal/citología , Mucosa Nasal/inmunología , Fosfoproteínas/metabolismo , Fosfoproteínas/farmacología , Fosfoproteínas/uso terapéutico , Polimorfismo de Nucleótido Simple , Cultivo Primario de Células , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Índice de Severidad de la Enfermedad , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
3.
Toxicol Sci ; 166(2): 345-353, 2018 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-30169750

RESUMEN

Since the start of Afghanistan combat operations in 2001, there has been an increase in complaints of respiratory illnesses in deployed soldiers with no previous history of lung disorders. It is postulated that deployment-related respiratory illnesses are the result of inhalation of desert particulate matter (PM) potentially acting in combination with exposure to other pro-inflammatory compounds. Why some, but not all, soldiers develop respiratory diseases remains unclear. Our goal was to investigate if human airway epithelial cells primed with IL-13, a type 2 inflammatory cytokine, demonstrate stronger pro-inflammatory responses to Afghanistan desert PM (APM). Primary human brushed bronchial epithelial cells from non-deployed, healthy subjects were exposed to APM, both with and without IL-13 pretreatment. APM exposure in conjunction with IL-13 resulted in significantly increased expression of IL-8, a pro-inflammatory cytokine involved in neutrophil recruitment and activation. Furthermore, expression of TLR2 mRNA was increased after combined IL-13 and APM exposure. siRNA-mediated TLR2 knockdown dampened IL-8 production after exposure to APM with IL-13. APM with IL-13 treatment increased IRAK-1 (a downstream signaling molecule of TLR2 signaling) activation, while IRAK-1 knockdown effectively eliminated the IL-8 response to APM and IL-13. Our data suggest that APM exposure may promote neutrophilic inflammation in airways with a type 2 cytokine milieu.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Interleucina-13/farmacología , Material Particulado/envenenamiento , Afganistán , Anciano , Enfermedades Bronquiales/inducido químicamente , Enfermedades Bronquiales/metabolismo , Células Cultivadas , Citocinas , Células Epiteliales/inmunología , Femenino , Voluntarios Sanos , Humanos , Inflamación/inducido químicamente , Inflamación/metabolismo , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Interleucina-13/metabolismo , Interleucina-8/metabolismo , Masculino , Persona de Mediana Edad , ARN Interferente Pequeño , Transducción de Señal , Receptor Toll-Like 2/metabolismo
4.
Methods Mol Biol ; 1799: 371-380, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29956165

RESUMEN

The CRISPR-Cas9 technology is a powerful tool that enables site-specific genome modification (gene editing) and is increasingly used in research to generate gene knockout or knock-in in a variety of cells and organisms. This chapter provides a brief overview of this technology and describes a general methodology applicable to human airway biology research.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Sistema Respiratorio/metabolismo , Línea Celular , Clonación Molecular , Técnicas de Inactivación de Genes , Humanos , Mutación INDEL , Lentivirus/genética , ARN Guía de Kinetoplastida , Mucosa Respiratoria/metabolismo
5.
BMC Immunol ; 17(1): 27, 2016 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-27501951

RESUMEN

BACKGROUND: The lung is exposed to airborne fungal spores, and fungi that colonize the oral cavity such as Candida albicans, but does not develop disease to opportunistic fungal pathogens unless the immune system is compromised. The Group IVA cytosolic phospholipase A2 (cPLA2α) is activated in response to Candida albicans infection resulting in the release of arachidonic acid for eicosanoid production. Although eicosanoids such as prostaglandins and leukotrienes modulate inflammation and immune responses, the role of cPLA2α and eicosanoids in regulating C. albicans lung infection is not understood. METHODS: The responses of cPLA2α(+/+) and cPLA2α(-/-) Balb/c mice to intratracheal instillation of C. albicans were compared. After challenge, we evaluated weight loss, organ fungal burden, and the recruitment of cells and the levels of cytokines and eicosanoids in bronchoalveolar lavage fluid. The ability of macrophages and neutrophils from cPLA2α(+/+) and cPLA2α(-/-) mice to recognize and kill C. albicans was also compared. RESULTS: After C. albicans instillation, cPLA2α(+/+) mice recovered a modest weight loss by 48 h and completely cleared fungi from the lung by 12 h with no dissemination to the kidneys. In cPLA2α(-/-) mice, weight loss continued for 72 h, C. albicans was not completely cleared from the lung and disseminated to the kidneys. cPLA2α(-/-) mice exhibited greater signs of inflammation including higher neutrophil influx, and elevated levels of albumin and pro-inflammatory cytokines/chemokines (IL1α, IL1ß, TNFα, IL6, CSF2, CXCL1, CCL20) in bronchoalveolar lavage fluid. The amounts of cysteinyl leukotrienes, thromboxane B2 and prostaglandin E2 were significantly lower in bronchoalveolar lavage fluid from C. albicans-infected cPLA2α(-/-) mice compared to cPLA2α(+/+) mice. Alveolar macrophages and neutrophils from uninfected cPLA2α(-/-) mice exhibited less killing of C. albicans in vitro than cells from cPLA2α(+/+) mice. In addition alveolar macrophages from cPLA2α(-/-) mice isolated 6 h after instillation of GFP-C. albicans contained fewer internalized fungi than cPLA2α(+/+) macrophages. CONCLUSIONS: The results demonstrate that cPLA2α contributes to immune surveillance and host defense in the lung to prevent infection by the commensal fungus C. albicans and to dampen inflammation.


Asunto(s)
Candida albicans/inmunología , Candidiasis/inmunología , Enfermedades Pulmonares Fúngicas/inmunología , Pulmón/inmunología , Macrófagos Alveolares/fisiología , Neutrófilos/fisiología , Fosfolipasas A2/metabolismo , Animales , Ácido Araquidónico/metabolismo , Movimiento Celular , Células Cultivadas , Citocinas/metabolismo , Eicosanoides/metabolismo , Inmunidad Innata , Mediadores de Inflamación/metabolismo , Pulmón/microbiología , Macrófagos Alveolares/microbiología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Neutrófilos/microbiología , Fosfolipasas A2/genética , Fosfolipasas A2/inmunología
6.
Methods Mol Biol ; 1442: 219-30, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27464698

RESUMEN

This chapter describes two procedures commonly used to examine airway function in mice. Airway function can be assessed in vivo using noninvasive or invasive methods. Noninvasive methods can be used to monitor respiratory function in mice without the involvement of restraint, anesthesia, or surgery. The methods allow for multiple animals to be monitored simultaneously and can be used in longitudinal studies requiring repeated measurements on the same animals. Invasive methods are used to assess airway function under anesthesia, in mechanically ventilated mice. Although used as terminal procedure, the invasive methods are most appropriate for direct assessment of lower airway dysfunction.


Asunto(s)
Pulmón/fisiología , Modelos Animales , Pruebas de Función Respiratoria/métodos , Anestesia , Animales , Ratones , Pruebas de Función Respiratoria/instrumentación
7.
Respir Res ; 16: 117, 2015 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-26410792

RESUMEN

BACKGROUND: Ozone pollution has adverse effects on respiratory health in children and adults. This study was carried out in the mouse model to investigate the influence of age and to define the role of toll-like receptor four (TLR4) in the lung response to ozone exposure during postnatal development. METHODS: Female mice (1 to 6 weeks of age) were exposed for 3 h to ozone (1 part per million) or filtered air. Analyses were carried out at six and 24 h after completion of exposure, to assess the effects on lung permeability, airway neutrophilia, expression of antioxidants and chemokines, and mucus production. The role of TLR4 was defined by examining TLR4 expression in the lung during development, and by investigating the response to ozone in tlr4-deficient mice. RESULTS: Metallothionein-1, calcitonin gene-related product, and chemokine C-X-C ligand (CXCL) five were consistent markers induced by ozone throughout development. Compared with adults, neonates expressed lower levels of pulmonary TLR4 and responded with increased mucus production, and developed an attenuated response to ozone characterized by reduced albumin leakage and neutrophil influx into the airways, and lower expression of CXCL1 and CXCL2 chemokines. Examination of the responses in tlr4-deficient mice indicated that ozone-mediated airway neutrophilia, but not albumin leakage or mucus production were dependent on TLR4. CONCLUSIONS: Collectively, the data demonstrate that the response to ozone is determined by age and is partially dependent on TLR4 signaling. The reduced responsiveness of the neonatal lung to ozone may be due at least in part to insufficient pulmonary TLR4 expression.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Pulmón/efectos de los fármacos , Ozono/toxicidad , Receptor Toll-Like 4/efectos de los fármacos , Factores de Edad , Animales , Antioxidantes/metabolismo , Biomarcadores/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Quimiocinas/metabolismo , Femenino , Exposición por Inhalación , Pulmón/irrigación sanguínea , Pulmón/metabolismo , Ratones Endogámicos BALB C , Ratones Noqueados , Moco/metabolismo , Neuropéptidos/metabolismo , Infiltración Neutrófila/efectos de los fármacos , Albúmina Sérica/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
8.
Proc Natl Acad Sci U S A ; 112(16): 5111-6, 2015 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-25820174

RESUMEN

Silencing of interleukin-32 (IL-32) in a differentiated human promonocytic cell line impairs killing of Mycobacterium tuberculosis (MTB) but the role of IL-32 in vivo against MTB remains unknown. To study the effects of IL-32 in vivo, a transgenic mouse was generated in which the human IL-32γ gene is expressed using the surfactant protein C promoter (SPC-IL-32γTg). Wild-type and SPC-IL-32γTg mice were infected with a low-dose aerosol of a hypervirulent strain of MTB (W-Beijing HN878). At 30 and 60 d after infection, the transgenic mice had 66% and 85% fewer MTB in the lungs and 49% and 68% fewer MTB in the spleens, respectively; the transgenic mice also exhibited greater survival. Increased numbers of host-protective innate and adaptive immune cells were present in SPC-IL-32γTg mice, including tumor necrosis factor-alpha (TNFα) positive lung macrophages and dendritic cells, and IFN-gamma (IFNγ) and TNFα positive CD4(+) and CD8(+) T cells in the lungs and mediastinal lymph nodes. Alveolar macrophages from transgenic mice infected with MTB ex vivo had reduced bacterial burden and increased colocalization of green fluorescent protein-labeled MTB with lysosomes. Furthermore, mouse macrophages made to express IL-32γ but not the splice variant IL-32ß were better able to limit MTB growth than macrophages capable of producing both. The lungs of patients with tuberculosis showed increased IL-32 expression, particularly in macrophages of granulomas and airway epithelial cells but also B cells and T cells. We conclude that IL-32γ enhances host immunity to MTB.


Asunto(s)
Interleucinas/metabolismo , Mycobacterium tuberculosis/patogenicidad , Tuberculosis/inmunología , Tuberculosis/prevención & control , Inmunidad Adaptativa/inmunología , Animales , Antígenos Ly/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Células Cultivadas , Humanos , Inmunidad Innata/inmunología , Interferón gamma , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Macrófagos Alveolares/inmunología , Ratones Transgénicos , Mutación/genética , Subfamilia B de Receptores Similares a Lectina de Células NK/metabolismo , Proteína C Asociada a Surfactante Pulmonar/metabolismo , Sitios de Empalme de ARN/genética , Linfocitos T Reguladores/inmunología , Transfección , Transgenes , Tuberculosis/microbiología , Factor de Necrosis Tumoral alfa/metabolismo , Virulencia/inmunología
9.
J Immunol ; 193(5): 2238-47, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25070841

RESUMEN

Glucocorticoid-induced TNFR family-related protein (GITR)-mediated activation of JNK was shown to regulate the suppressive activity of CD4(+)CD25(+) naturally occurring T regulatory cells (nTregs) in wild-type (WT) hosts. In this study, CD4(+)CD25(+) T cells were shown to be capable of becoming pathogenic effector cells in sensitized and challenged CD8(-/-) recipient mice. Only GITR-expressing CD4(+)CD25(+) T cells, but neither GITR knocked-in CD4(+)CD25(-) T cells nor GITR-silenced CD4(+)CD25(+) T cells, enhanced development of lung allergic responses. Inhibition of JNK in WT nTregs or nTregs from GITR(-/-)and JNK2(-/-) mice failed to enhance lung allergic responses in sensitized and challenged CD8(-/-) recipient mice. The failure to enhance responses was associated with increased bronchoalveolar lavage fluid levels of IL-10 and TGF-ß and decreased levels of IL-5, IL-6, and IL-13. In contrast, nTregs from JNK1(-/-) mice, similar to WT nTregs, were fully effective in enhancing responses. Thus, GITR stimulation of nTregs and signaling through JNK2, but not JNK1, triggered the loss of regulatory function while concomitantly gaining pathogenic CD4(+) T effector cell function responsible for exacerbating asthma-like immunopathology.


Asunto(s)
Asma/inmunología , Pulmón/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Proteína Quinasa 9 Activada por Mitógenos/inmunología , Linfocitos T Reguladores/inmunología , Animales , Asma/genética , Asma/patología , Proteína Relacionada con TNFR Inducida por Glucocorticoide/genética , Proteína Relacionada con TNFR Inducida por Glucocorticoide/inmunología , Interleucina-10/genética , Interleucina-10/inmunología , Pulmón/patología , Sistema de Señalización de MAP Quinasas/genética , Ratones , Ratones Noqueados , Proteína Quinasa 8 Activada por Mitógenos/genética , Proteína Quinasa 8 Activada por Mitógenos/inmunología , Proteína Quinasa 9 Activada por Mitógenos/genética , Linfocitos T Reguladores/patología , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/inmunología
10.
Am J Respir Cell Mol Biol ; 51(1): 143-54, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24521403

RESUMEN

Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract illnesses in infants worldwide. Both RSV-G and RSV-F glycoproteins play pathogenic roles during infection with RSV. The objective of this study was to compare the effects of anti-RSV-G and anti-RSV-F monoclonal antibodies (mAbs) on airway hyperresponsiveness (AHR) and inflammation after primary or secondary RSV infection in mice. In the primary infection model, mice were infected with RSV at 6 weeks of age. Anti-RSV-G or anti-RSV-F mAbs were administered 24 hours before infection or Day +2 postinfection. In a secondary infection model, mice were infected (primary) with RSV at 1 week (neonate) and reinfected (secondary) 5 weeks later. Anti-RSV-G and anti-RSV-F mAbs were administered 24 hours before the primary infection. Both mAbs had comparable effects in preventing airway responses after primary RSV infection. When given 2 days after infection, anti-RSV-G-treated mice showed significantly decreased AHR and airway inflammation, which persisted in anti-RSV-F-treated mice. In the reinfection model, anti-RSV-G but not anti-RSV-F administered during primary RSV infection in neonates resulted in decreased AHR, eosinophilia, and IL-13 but increased levels of IFN-γ in bronchoalveolar lavage on reinfection. These results support the use of anti-RSV-G in the prevention and treatment of RSV-induced disease.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Bronquiolitis Viral/prevención & control , Inflamación/prevención & control , Hipersensibilidad Respiratoria/prevención & control , Infecciones por Virus Sincitial Respiratorio/prevención & control , Proteínas Virales de Fusión/inmunología , Animales , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/uso terapéutico , Bronquiolitis Viral/etiología , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Inflamación/etiología , Ratones , Ratones Endogámicos BALB C , Hipersensibilidad Respiratoria/etiología , Infecciones por Virus Sincitial Respiratorio/complicaciones , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Virus Sincitiales Respiratorios/patogenicidad
11.
Ann Allergy Asthma Immunol ; 112(3): 211-216.e1, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24428972

RESUMEN

BACKGROUND: Numbers of CD8(+) T cells expressing the leukotriene B4 (LTB4) receptor, BLT1, have been correlated with asthma severity. OBJECTIVE: To examine the activation and numbers of BLT1-expressing peripheral blood CD4(+) and CD8(+) T cells from patients with steroid-sensitive (SS) and steroid-resistant (SR) asthma. METHODS: CD4(+) and CD8(+) T cells isolated from peripheral blood of healthy human subjects and patients with SS and SR asthma were stimulated in culture with anti-CD3/anti-CD28 followed by analysis of BLT1 surface expression and cytokine production. Activation of CD8(+) T cells after ligation of BLT1 by LTB4 was monitored by changes in intracellular Ca(2+) concentrations. RESULTS: The number of BLT1-expressing cells was larger in patients with asthma than in controls and larger on activated CD8(+) than on CD4(+) T cells. Addition of LTB4 to activated CD8(+) T cells resulted in increases in intracellular Ca(2+) concentrations. Expansion of activated CD4(+) T cells, unlike CD8(+) T cells, was significantly decreased in the presence of corticosteroid. In patients with SS asthma, numbers of BLT1-expressing CD8(+) T cells were lower in the presence of corticosteroid, unlike in those with SR asthma in whom cell expansion was maintained. Levels of interleukin-13 were highest in cultured CD8(+) T cells, whereas interleukin-10 levels were higher in CD4(+) T cells from controls and patients with SS asthma. Interferon-γ levels were lowest in patients with SR asthma. CONCLUSION: Differences in BLT1 expression, steroid sensitivity, and cytokine production were demonstrated in T lymphocytes from patients with SS and SR asthma. The LTB4-BLT1 pathway in CD8(+) cells may play an important role in asthma and serve as an important target in the treatment of patients with SR asthma.


Asunto(s)
Asma/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Dexametasona/farmacología , Receptores de Leucotrieno B4/metabolismo , Corticoesteroides/farmacología , Adulto , Anticuerpos Monoclonales/inmunología , Antígenos CD28/inmunología , Complejo CD3/inmunología , Calcio/metabolismo , Células Cultivadas , Medios de Cultivo Condicionados/química , Femenino , Humanos , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Interleucina-13/metabolismo , Activación de Linfocitos/inmunología , Masculino
12.
Toxicol Sci ; 138(1): 175-90, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24336422

RESUMEN

Ozone pollution is associated with adverse effects on respiratory health in adults and children but its effects on the neonatal lung remain unknown. This study was carried out to define the effect of acute ozone exposure on the neonatal lung and to profile the transcriptome response. Newborn mice were exposed to ozone or filtered air for 3h. Total RNA was isolated from lung tissues at 6 and 24h after exposure and was subjected to microarray gene expression analysis. Compared to filtered air-exposed littermates, ozone-exposed newborn mice developed a small but significant neutrophilic airway response associated with increased CXCL1 and CXCL5 expression in the lung. Transcriptome analysis indicated that 455 genes were down-regulated and 166 genes were up-regulated by at least 1.5-fold at 6h post-ozone exposure (t-test, p < .05). At 24h, 543 genes were down-regulated and 323 genes were up-regulated in the lungs of ozone-exposed, compared to filtered air-exposed, newborn mice (t-test, p < .05). After controlling for false discovery rate, 50 genes were identified as significantly down-regulated and only a few (RORC, GRP, VREB3, and CYP2B6) were up-regulated at 24h post-ozone exposure (q < .05). Gene ontology enrichment analysis revealed that cell cycle-associated functions including cell division/proliferation were the most impacted pathways, which were negatively regulated by ozone exposure, an adverse effect that was associated with reduced bromo-deoxyuridine incorporation. These results demonstrate that acute ozone exposure alters cell proliferation in the developing neonatal lung through a global suppression of cell cycle function.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Pulmón/efectos de los fármacos , Ozono/toxicidad , Transcriptoma/efectos de los fármacos , Animales , Animales Recién Nacidos , Proliferación Celular/efectos de los fármacos , Quimiocina CXCL1/genética , Quimiocina CXCL5/genética , Regulación hacia Abajo , Perfilación de la Expresión Génica , Exposición por Inhalación , Pulmón/crecimiento & desarrollo , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Neutrófilos/patología , Tráquea/efectos de los fármacos , Tráquea/ultraestructura , Regulación hacia Arriba
13.
Virology ; 446(1-2): 199-206, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24074582

RESUMEN

Human rhinovirus (HRV) is the most common viral etiology in acute exacerbations of asthma. However, the exact mechanisms underlying HRV infection in allergic airways are poorly understood. IL-13 increases interleukin-1 receptor associated kinase M (IRAK-M) and subsequently inhibits airway innate immunity against bacteria. However, the role of IRAK-M in lung HRV infection remains unclear. Here, we provide the first evidence that IRAK-M over-expression promotes lung epithelial HRV-16 replication and autophagy, but inhibits HRV-16-induced IFN-ß and IFN-λ1 expression. Inhibiting autophagy reduces HRV-16 replication. Exogenous IFN-ß and IFN-λ1 inhibit autophagy and HRV-16 replication. Our data indicate the enhancing effect of IRAK-M on epithelial HRV-16 infection, which is partly through the autophagic pathway. Impaired anti-viral interferon production may serve as a direct or an indirect (e.g., autophagy) mechanism of enhanced HRV-16 infection by IRAK-M over-expression. Targeting autophagic pathway or administrating anti-viral interferons may prevent or attenuate viral (e.g., HRV-16) infections in allergic airways.


Asunto(s)
Autofagia , Células Epiteliales/virología , Interacciones Huésped-Patógeno , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Rhinovirus/fisiología , Replicación Viral , Células HeLa , Humanos , Interferón beta/antagonistas & inhibidores , Interferones , Interleucinas/antagonistas & inhibidores
14.
J Allergy Clin Immunol ; 130(5): 1175-1186.e9, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23036746

RESUMEN

BACKGROUND: Recent studies revealed a critical role for thymic stromal lymphopoietin (TSLP) released from epithelial cells and OX40 ligand (OX40L) expressed on dendritic cells (DCs) in T(H)2 priming and polarization. OBJECTIVES: We sought to determine the importance of the TSLP-OX40L axis in neonatal respiratory syncytial virus (RSV) infection. METHODS: Mice were initially infected with RSV as neonates or adults and reinfected 5 weeks later. Anti-OX40L or anti-TSLP were administered during primary or secondary infection. Outcomes included assessment of airway function and inflammation and expression of OX40L, TSLP, and IL-12. RESULTS: OX40L was expressed mainly on CD11c(+)MHC class II (MHCII)(+)CD11b(+) DCs but not CD103(+) DCs. Treatment of neonates with OX40L antibody during primary RSV infection prevented the subsequent enhancement of airway hyperresponsiveness and the development of airway eosinophilia and mucus hyperproduction on reinfection. Administration of anti-TSLP before neonatal RSV infection reduced the accumulation of lung DCs, decreased OX40L expression on lung DCs, and attenuated the enhancement of airway responses after reinfection. CONCLUSIONS: In mice initially infected as neonates, TSLP expression induced by RSV infection is an important upstream event that controls OX40L expression, lung DC migration, and T(H)2 polarization, accounting for the enhanced response on reinfection.


Asunto(s)
Hiperreactividad Bronquial/inmunología , Citocinas/metabolismo , Células Dendríticas/inmunología , Ligando OX40/metabolismo , Eosinofilia Pulmonar/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Animales , Animales Recién Nacidos , Anticuerpos Bloqueadores/administración & dosificación , Hiperreactividad Bronquial/etiología , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Citocinas/genética , Citocinas/inmunología , Células Dendríticas/efectos de los fármacos , Humanos , Inmunidad/efectos de los fármacos , Interleucina-12/metabolismo , Ratones , Ratones Endogámicos BALB C , Moco/metabolismo , Ligando OX40/genética , Ligando OX40/inmunología , Eosinofilia Pulmonar/etiología , Infecciones por Virus Sincitial Respiratorio/complicaciones , Linfopoyetina del Estroma Tímico
15.
J Biol Chem ; 287(21): 17100-17108, 2012 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-22461627

RESUMEN

Naturally occurring Foxp3(+)CD4(+)CD25(+) T regulatory cell (nTreg)-mediated suppression of lung allergic responses is abrogated following ligation of glucocorticoid-induced tumor necrosis receptor (GITR) family-related protein. In vitro stimulation of nTregs with GITR ligand increased phosphorylation of c-Jun N-terminal kinase (JNK) but not extracellular signal-regulated protein kinase (ERK) or p38 MAPK. SP600125, a known JNK inhibitor, prevented GITR-mediated phosphorylation of JNK. Activation of JNK was associated with increases in the upstream mitogen-activated protein kinase kinase 7 (MKK7) and the downstream transcription factor NF-κß. Phosphorylated c-Jun (p-c-Jun), indicative of the activation of JNK, was detected in the immunoprecipitates of nTregs from wild-type but not JNK- or GITR-deficient mice. Treatment with an inhibitor of JNK phosphorylation resulted in complete reversal of all GITR-induced changes in nTreg phenotype and function, with full restoration of suppression of in vivo lung allergic responses and in vitro proliferation of activated CD4(+)CD25(-) T cells. Thus, regulation of JNK phosphorylation plays a central role in T regulatory cell function with therapeutic implications for the treatment of asthma and autoimmune diseases.


Asunto(s)
Proteína Relacionada con TNFR Inducida por Glucocorticoide/metabolismo , Tolerancia Inmunológica/fisiología , MAP Quinasa Quinasa 4/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Linfocitos T Reguladores/metabolismo , Animales , Antracenos/farmacología , Asma/genética , Asma/inmunología , Asma/metabolismo , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Proliferación Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Activación Enzimática/inmunología , Femenino , Proteína Relacionada con TNFR Inducida por Glucocorticoide/genética , Proteína Relacionada con TNFR Inducida por Glucocorticoide/inmunología , Tolerancia Inmunológica/efectos de los fármacos , Pulmón/inmunología , Pulmón/metabolismo , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/inmunología , MAP Quinasa Quinasa 7/genética , MAP Quinasa Quinasa 7/inmunología , MAP Quinasa Quinasa 7/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Noqueados , FN-kappa B/genética , FN-kappa B/inmunología , FN-kappa B/metabolismo , Fosforilación/efectos de los fármacos , Fosforilación/genética , Fosforilación/inmunología , Inhibidores de Proteínas Quinasas/farmacología , Linfocitos T Reguladores/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
16.
Curr Opin Immunol ; 23(6): 801-7, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21840186

RESUMEN

Although national asthma guidelines help organize standards for asthma care, current asthma management is still primarily symptom based. Recent reports provide insights on how to improve asthma management through steps to better understand the natural history of asthma, individualize asthma care, reduce asthma exacerbations, manage inner city asthma, and some potential new ways to use available medications to improve asthma control. Despite many significant gains in managing asthma, we must now find improved strategies to prevent asthma exacerbations, alter the natural history of the disease, and to reduce health disparities in asthma care. Perhaps new directions in personalized medicine including a systems biology approach, along with improved health care access and communication will lead to better methods to alleviate the burden of asthma. This review will discuss the benefits and limitations of the current approach to asthma management, new studies that could impact new directions in asthma management, and new insights related to mechanisms of asthma and allergic airways inflammation that could eventually lead to improved asthma control.


Asunto(s)
Asma/inmunología , Asma/terapia , Animales , Susceptibilidad a Enfermedades , Humanos , Medicina de Precisión
17.
J Immunol ; 186(1): 113-20, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21115736

RESUMEN

Naturally occurring CD4(+)CD25(+)Foxp3(+) T regulatory cells (nTregs) regulate lung allergic responses through production of IL-10 and TGF-ß. nTregs from CD8(-/-) mice failed to suppress lung allergic responses and were characterized by reduced levels of Foxp3, IL-10, and TGF-ß, and high levels of IL-6. Administration of anti-IL-6 or anti-IL-6R to wild-type recipients prior to transfer of CD8(-/-) nTregs restored suppression. nTregs from IL-6(-/-) mice were suppressive, but lost this capability if incubated with IL-6 prior to transfer. The importance of CD8 in regulating the production of IL-6 in nTregs was demonstrated by the loss of suppression and increases in IL-6 following transfer of nTregs from wild-type donors depleted of CD8(+) cells. Transfer of nTregs from CD8(-/-) donors reconstituted with CD8(+) T cells was suppressive, and accordingly, IL-6 levels were reduced. These data identify the critical role of CD8-T regulatory cell interactions in regulating the suppressive phenotype of nTregs through control of IL-6 production.


Asunto(s)
Antígenos CD8/fisiología , Inmunofenotipificación , Terapia de Inmunosupresión , Interleucina-6/biosíntesis , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Animales , Antígenos CD8/genética , Células Cultivadas , Femenino , Factores de Transcripción Forkhead/deficiencia , Factores de Transcripción Forkhead/genética , Interleucina-10/antagonistas & inhibidores , Interleucina-6/deficiencia , Interleucina-6/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Interleucina-6/antagonistas & inhibidores , Receptores de Interleucina-6/fisiología , Hipersensibilidad Respiratoria/genética , Factor de Crecimiento Transformador beta/antagonistas & inhibidores
18.
J Allergy Clin Immunol ; 126(2): 306-16, 316.e1-12, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20624645

RESUMEN

BACKGROUND: Although implicated in the disease, the specific contributions of FcepsilonRI and IL-13 to the pathogenesis of peanut-induced intestinal allergy are not well defined. OBJECTIVES: We sought to determine the contributions of FcepsilonRI, IL-13, and mast cells to the development of intestinal mucosal responses in a murine model of peanut-induced intestinal allergy. METHODS: Sensitized wild-type (WT), FcepsilonRI-deficient (FcepsilonRI(-/-)), and mast cell-deficient (Kit(W-sh/W-sh)) mice received peanut orally every day for 1 week. Bone marrow-derived mast cells (BMMCs) from WT, FcepsilonRI(-/-), IL-4(-/-), IL-13(-/-), and IL-4/IL-13(-/-) mice were differentiated and transferred into WT, FcepsilonRI(-/-), and Kit(W-sh/W-sh) recipients. BMMCs from WT and UBI-GFP/BL6 mice were differentiated and transferred into WT and Kit(W-sh/W-sh) mice. Blockade of IL-13 was achieved by using IL-13 receptor alpha2 (IL-13Ralpha2)-IgG fusion protein. RESULTS: FcepsilonRI(-/-) mice showed decreased intestinal inflammation (mast cell and eosinophil numbers) and goblet cell metaplasia and reduced levels of IL4, IL6, IL13, and IL17A mRNA expression in the jejunum. Transfer of WT BMMCs to FcepsilonRI(-/-) recipients restored their ability to develop intestinal allergic responses unlike transfer of FcepsilonRI(-/-), IL-13(-/-), or IL-4/IL-13(-/-) BMMCs. FcepsilonRI(-/-) mice exhibited lower IL-13 levels and treatment of WT mice with IL-13 receptor alpha2 prevented peanut-induced intestinal allergy and inflammation. CONCLUSIONS: These data indicate that the development of peanut-induced intestinal allergy is mediated through a mast cell-dependent IgE-FcepsilonRI-IL-13 pathway. Targeting IL-13 might be a potential treatment for IgE-mediated peanut-induced allergic responses in the intestine.


Asunto(s)
Arachis/efectos adversos , Inmunoglobulina E/inmunología , Interleucina-13/inmunología , Mastocitos/inmunología , Hipersensibilidad al Cacahuete/inmunología , Receptores de IgE/inmunología , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Inmunidad Mucosa/genética , Inmunidad Mucosa/inmunología , Inmunoglobulina E/genética , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/farmacología , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Interleucina-13/antagonistas & inhibidores , Interleucina-13/genética , Subunidad alfa2 del Receptor de Interleucina-13/genética , Interleucina-17/genética , Interleucina-17/inmunología , Interleucina-4/genética , Interleucina-4/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Intestinos/inmunología , Intestinos/patología , Mastocitos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Hipersensibilidad al Cacahuete/tratamiento farmacológico , Hipersensibilidad al Cacahuete/genética , Hipersensibilidad al Cacahuete/patología , Receptores de IgE/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología
19.
J Immunol ; 185(1): 253-62, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20525882

RESUMEN

Invariant NKT cells (iNKT cells) play a pivotal role in the development of allergen-induced airway hyperresponsiveness (AHR) and inflammation. However, it is unclear what role they play in the initiation (sensitization) phase as opposed to the effector (challenge) phase. The role of iNKT cells during sensitization was examined by determining the response of mice to intratracheal transfer of OVA-pulsed or OVA-alpha-galactosylceramide (OVA/alphaGalCer)-pulsed bone marrow-derived dendritic cells (BMDCs) prior to allergen challenge. Wild-type (WT) recipients of OVA-BMDCs developed AHR, increased airway eosinophilia, and increased levels of Th2 cytokines in bronchoalveolar lavage fluid, whereas recipients of OVA/alphaGalCer BMDCs failed to do so. In contrast, transfer of these same OVA/alphaGalCer BMDCs into IFN-gamma-deficient (IFN-gamma(-/-)) mice enhanced the development of these lung allergic responses, which was reversed by exogenous IFN-gamma treatment following OVA-BMDC transfer. Further, Jalpha18-deficient recipients, which lack iNKT cells, developed the full spectrum of lung allergic responses following reconstitution with highly purified WT liver or spleen iNKT cells and transfer of OVA-BMDCs, whereas reconstituted recipients of OVA/alphaGalCer BMDCs failed to do so. Transfer of iNKT cells from IFN-gamma(-/-) mice restored the development of these responses in Jalpha18-deficient recipients following OVA-BMDC transfer; the responses were enhanced following OVA/alphaGalCer BMDC transfer. iNKT cells from these IFN-gamma(-/-) mice produced higher levels of IL-13 in vitro compared with WT iNKT cells. These data identify IFN-gamma as playing a critical role in dictating the consequences of iNKT cell activation in the initiation phase of the development of AHR and airway inflammation.


Asunto(s)
Alérgenos/administración & dosificación , Interferón gamma/biosíntesis , Células T Asesinas Naturales/inmunología , Ovalbúmina/inmunología , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/prevención & control , Traslado Adoptivo , Alérgenos/inmunología , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Hiperreactividad Bronquial/inmunología , Hiperreactividad Bronquial/patología , Hiperreactividad Bronquial/prevención & control , Células Cultivadas , Técnicas de Cocultivo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/trasplante , Femenino , Galactosilceramidas/administración & dosificación , Galactosilceramidas/inmunología , Galactosilceramidas/metabolismo , Mediadores de Inflamación/administración & dosificación , Mediadores de Inflamación/metabolismo , Interferón gamma/deficiencia , Interferón gamma/fisiología , Intubación Intratraqueal , Ligandos , Hígado/inmunología , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células T Asesinas Naturales/metabolismo , Células T Asesinas Naturales/patología , Ovalbúmina/administración & dosificación , Hipersensibilidad Respiratoria/patología
20.
Am J Respir Crit Care Med ; 182(4): 455-63, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20442434

RESUMEN

RATIONALE: Respiratory syncytial virus (RSV) bronchiolitis in infants may be followed by the development of asthma-like symptoms. Age at first infection dictates consequences upon reinfection. Reinfection of mice initially exposed as neonates to RSV enhanced development of airway hyperresponsiveness (AHR), eosinophilic inflammation, and mucus hyperproduction. RSV lower respiratory tract disease is associated with activation of the leukotriene pathway. OBJECTIVES: To determine the effects of montelukast (MK), a cysteinyl leukotriene (cysLT) receptor antagonist, in primary and secondary RSV-infected newborn and adult mice. METHODS: BALB/c mice were infected with RSV at 1 week (neonate) or 6 to 8 weeks (adult) of age and reinfected 5 weeks later. MK was administered 1 day before the initial infection and through Day 6 after infection. Seven days after primary or secondary infection, airway function was assessed by lung resistance to increasing doses of inhaled methacholine; lung inflammation, goblet cell metaplasia, and cytokine levels in bronchoalveolar lavage fluid were monitored. MEASUREMENTS AND MAIN RESULTS: RSV infection induced cysLT release in bronchoalveolar lavage fluid. MK decreased RSV-induced AHR, airway inflammation, and increased IFN-gamma production in primary infected adult and neonatal mice. MK, administered during initial infection of neonates but not during secondary infection, prevented subsequent enhancement of AHR, airway eosinophilia, and mucus hyperproduction upon reinfection. CONCLUSIONS: MK attenuated the initial responses to primary RSV infection in both age groups and altered the consequences of RSV reinfection in mice initially infected as neonates. These data support an important role for cysLT in RSV-induced AHR and inflammation.


Asunto(s)
Acetatos/farmacología , Antiasmáticos/farmacología , Inflamación/prevención & control , Quinolinas/farmacología , Hipersensibilidad Respiratoria/prevención & control , Infecciones por Virus Sincitial Respiratorio/complicaciones , Virus Sincitiales Respiratorios/efectos de los fármacos , Animales , Animales Recién Nacidos , Bronquiolitis Viral/complicaciones , Bronquiolitis Viral/prevención & control , Bronquiolitis Viral/virología , Líquido del Lavado Bronquioalveolar/virología , Ciclopropanos , Cisteína/antagonistas & inhibidores , Modelos Animales de Enfermedad , Inflamación/complicaciones , Interferón gamma/efectos de los fármacos , Antagonistas de Leucotrieno/farmacología , Leucotrienos , Ratones , Ratones Endogámicos BALB C , Recurrencia , Hipersensibilidad Respiratoria/etiología , Sulfuros
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...