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










Base de dados
Intervalo de ano de publicação
1.
J Innate Immun ; 12(1): 103-115, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-30928973

RESUMO

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.


Assuntos
Quinases Associadas a Receptores de Interleucina-1/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neutrófilos/imunologia , Infecções por Picornaviridae/metabolismo , Mucosa Respiratória/metabolismo , Rhinovirus/fisiologia , Células Th2/imunologia , Células Cultivadas , Citocinas/metabolismo , Humanos , Imunomodulação , Quinases Associadas a Receptores de Interleucina-1/genética , Proteína 1 Semelhante a Receptor de Interleucina-1/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Ativação de Neutrófilo , Infecções por Picornaviridae/imunologia , RNA Interferente Pequeno/genética , Mucosa Respiratória/patologia , Transdução de Sinais
2.
JCI Insight ; 4(8)2019 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-30996135

RESUMO

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.


Assuntos
Asma/genética , Células Epiteliais/imunologia , Regulação da Expressão Gênica/imunologia , Glicoproteínas/genética , Fosfoproteínas/genética , Transdução de Sinais/imunologia , Adolescente , Adulto , Idoso , Alelos , Asma/diagnóstico , Asma/tratamento farmacológico , Asma/imunologia , Células Cultivadas , Quimiocina CCL26/imunologia , Quimiocina CCL26/metabolismo , Criança , Eosinófilos/imunologia , Células Epiteliais/patologia , Feminino , Volume Expiratório Forçado , Predisposição Genética para Doença , Glicoproteínas/metabolismo , Glicoproteínas/farmacologia , Glicoproteínas/uso terapêutico , Humanos , Interleucina-13/imunologia , Interleucina-13/metabolismo , Masculino , Pessoa de Meia-Idade , Mucosa Nasal/citologia , Mucosa Nasal/imunologia , Fosfoproteínas/metabolismo , Fosfoproteínas/farmacologia , Fosfoproteínas/uso terapêutico , Polimorfismo de Nucleotídeo Único , Cultura Primária de Células , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Índice de Gravidade de Doença , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
3.
Toxicol Sci ; 166(2): 345-353, 2018 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-30169750

RESUMO

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.


Assuntos
Células Epiteliais/efeitos dos fármacos , Interleucina-13/farmacologia , Material Particulado/intoxicação , Afeganistão , Idoso , Broncopatias/induzido quimicamente , Broncopatias/metabolismo , Células Cultivadas , Citocinas , Células Epiteliais/imunologia , Feminino , Voluntários Saudáveis , Humanos , Inflamação/induzido quimicamente , Inflamação/metabolismo , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Interleucina-13/metabolismo , Interleucina-8/metabolismo , Masculino , Pessoa de Meia-Idade , RNA Interferente Pequeno , Transdução de Sinais , Receptor 2 Toll-Like/metabolismo
4.
Methods Mol Biol ; 1799: 371-380, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29956165

RESUMO

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.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes , Sistema Respiratório/metabolismo , Linhagem Celular , Clonagem Molecular , Técnicas de Inativação de Genes , Humanos , Mutação INDEL , Lentivirus/genética , RNA Guia de Cinetoplastídeos , Mucosa Respiratória/metabolismo
5.
BMC Immunol ; 17(1): 27, 2016 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-27501951

RESUMO

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.


Assuntos
Candida albicans/imunologia , Candidíase/imunologia , Pneumopatias Fúngicas/imunologia , Pulmão/imunologia , Macrófagos Alveolares/fisiologia , Neutrófilos/fisiologia , Fosfolipases A2/metabolismo , Animais , Ácido Araquidônico/metabolismo , Movimento Celular , Células Cultivadas , Citocinas/metabolismo , Eicosanoides/metabolismo , Imunidade Inata , Mediadores da Inflamação/metabolismo , Pulmão/microbiologia , Macrófagos Alveolares/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Neutrófilos/microbiologia , Fosfolipases A2/genética , Fosfolipases A2/imunologia
6.
Methods Mol Biol ; 1442: 219-30, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27464698

RESUMO

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.


Assuntos
Pulmão/fisiologia , Modelos Animais , Testes de Função Respiratória/métodos , Anestesia , Animais , Camundongos , Testes de Função Respiratória/instrumentação
7.
Respir Res ; 16: 117, 2015 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-26410792

RESUMO

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.


Assuntos
Poluentes Atmosféricos/toxicidade , Pulmão/efeitos dos fármacos , Ozônio/toxicidade , Receptor 4 Toll-Like/efeitos dos fármacos , Fatores Etários , Animais , Antioxidantes/metabolismo , Biomarcadores/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Quimiocinas/metabolismo , Feminino , Exposição por Inalação , Pulmão/irrigação sanguínea , Pulmão/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Knockout , Muco/metabolismo , Neuropeptídeos/metabolismo , Infiltração de Neutrófilos/efeitos dos fármacos , Albumina Sérica/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
8.
Proc Natl Acad Sci U S A ; 112(16): 5111-6, 2015 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-25820174

RESUMO

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.


Assuntos
Interleucinas/metabolismo , Mycobacterium tuberculosis/patogenicidade , Tuberculose/imunologia , Tuberculose/prevenção & controle , Imunidade Adaptativa/imunologia , Animais , Antígenos Ly/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Cultivadas , Humanos , Imunidade Inata/imunologia , Interferon gama , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Linfonodos/imunologia , Linfonodos/patologia , Macrófagos Alveolares/imunologia , Camundongos Transgênicos , Mutação/genética , Subfamília B de Receptores Semelhantes a Lectina de Células NK/metabolismo , Proteína C Associada a Surfactante Pulmonar/metabolismo , Sítios de Splice de RNA/genética , Linfócitos T Reguladores/imunologia , Transfecção , Transgenes , Tuberculose/microbiologia , Fator de Necrose Tumoral alfa/metabolismo , Virulência/imunologia
9.
J Immunol ; 193(5): 2238-47, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-25070841

RESUMO

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.


Assuntos
Asma/imunologia , Pulmão/imunologia , Sistema de Sinalização das MAP Quinases/imunologia , Proteína Quinase 9 Ativada por Mitógeno/imunologia , Linfócitos T Reguladores/imunologia , Animais , Asma/genética , Asma/patologia , Proteína Relacionada a TNFR Induzida por Glucocorticoide/genética , Proteína Relacionada a TNFR Induzida por Glucocorticoide/imunologia , Interleucina-10/genética , Interleucina-10/imunologia , Pulmão/patologia , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Camundongos Knockout , Proteína Quinase 8 Ativada por Mitógeno/genética , Proteína Quinase 8 Ativada por Mitógeno/imunologia , Proteína Quinase 9 Ativada por Mitógeno/genética , Linfócitos T Reguladores/patologia , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/imunologia
10.
Am J Respir Cell Mol Biol ; 51(1): 143-54, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24521403

RESUMO

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.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Bronquiolite Viral/prevenção & controle , Inflamação/prevenção & controle , Hipersensibilidade Respiratória/prevenção & controle , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Proteínas Virais de Fusão/imunologia , Animais , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/uso terapêutico , Bronquiolite Viral/etiologia , Citocinas/metabolismo , Ensaio de Imunoadsorção Enzimática , Inflamação/etiologia , Camundongos , Camundongos Endogâmicos BALB C , Hipersensibilidade Respiratória/etiologia , Infecções por Vírus Respiratório Sincicial/complicações , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sinciciais Respiratórios/imunologia , Vírus Sinciciais Respiratórios/patogenicidade
11.
Ann Allergy Asthma Immunol ; 112(3): 211-216.e1, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24428972

RESUMO

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.


Assuntos
Asma/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Dexametasona/farmacologia , Receptores do Leucotrieno B4/metabolismo , Corticosteroides/farmacologia , Adulto , Anticorpos Monoclonais/imunologia , Antígenos CD28/imunologia , Complexo CD3/imunologia , Cálcio/metabolismo , Células Cultivadas , Meios de Cultivo Condicionados/química , Feminino , Humanos , Interferon gama/metabolismo , Interleucina-10/metabolismo , Interleucina-13/metabolismo , Ativação Linfocitária/imunologia , Masculino
12.
Toxicol Sci ; 138(1): 175-90, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24336422

RESUMO

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.


Assuntos
Poluentes Atmosféricos/toxicidade , Pulmão/efeitos dos fármacos , Ozônio/toxicidade , Transcriptoma/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Proliferação de Células/efeitos dos fármacos , Quimiocina CXCL1/genética , Quimiocina CXCL5/genética , Regulação para Baixo , Perfilação da Expressão Gênica , Exposição por Inalação , Pulmão/crescimento & desenvolvimento , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Neutrófilos/patologia , Traqueia/efeitos dos fármacos , Traqueia/ultraestrutura , Regulação para Cima
13.
Virology ; 446(1-2): 199-206, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24074582

RESUMO

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.


Assuntos
Autofagia , Células Epiteliais/virologia , Interações Hospedeiro-Patógeno , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Rhinovirus/fisiologia , Replicação Viral , Células HeLa , Humanos , Interferon beta/antagonistas & inibidores , Interferons , Interleucinas/antagonistas & inibidores
14.
J Allergy Clin Immunol ; 130(5): 1175-1186.e9, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23036746

RESUMO

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.


Assuntos
Hiper-Reatividade Brônquica/imunologia , Citocinas/metabolismo , Células Dendríticas/imunologia , Ligante OX40/metabolismo , Eosinofilia Pulmonar/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sinciciais Respiratórios/imunologia , Animais , Animais Recém-Nascidos , Anticorpos Bloqueadores/administração & dosagem , Hiper-Reatividade Brônquica/etiologia , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Citocinas/genética , Citocinas/imunologia , Células Dendríticas/efeitos dos fármacos , Humanos , Imunidade/efeitos dos fármacos , Interleucina-12/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Muco/metabolismo , Ligante OX40/genética , Ligante OX40/imunologia , Eosinofilia Pulmonar/etiologia , Infecções por Vírus Respiratório Sincicial/complicações , Linfopoietina do Estroma do Timo
15.
J Biol Chem ; 287(21): 17100-17108, 2012 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-22461627

RESUMO

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.


Assuntos
Proteína Relacionada a TNFR Induzida por Glucocorticoide/metabolismo , Tolerância Imunológica/fisiologia , MAP Quinase Quinase 4/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Linfócitos T Reguladores/metabolismo , Animais , Antracenos/farmacologia , Asma/genética , Asma/imunologia , Asma/metabolismo , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Proliferação de Células/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Ativação Enzimática/imunologia , Feminino , Proteína Relacionada a TNFR Induzida por Glucocorticoide/genética , Proteína Relacionada a TNFR Induzida por Glucocorticoide/imunologia , Tolerância Imunológica/efeitos dos fármacos , Pulmão/imunologia , Pulmão/metabolismo , MAP Quinase Quinase 4/genética , MAP Quinase Quinase 4/imunologia , MAP Quinase Quinase 7/genética , MAP Quinase Quinase 7/imunologia , MAP Quinase Quinase 7/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Camundongos Knockout , NF-kappa B/genética , NF-kappa B/imunologia , NF-kappa B/metabolismo , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Fosforilação/imunologia , Inibidores de Proteínas Quinases/farmacologia , Linfócitos T Reguladores/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
16.
Curr Opin Immunol ; 23(6): 801-7, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21840186

RESUMO

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.


Assuntos
Asma/imunologia , Asma/terapia , Animais , Suscetibilidade a Doenças , Humanos , Medicina de Precisão
17.
J Immunol ; 186(1): 113-20, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21115736

RESUMO

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.


Assuntos
Antígenos CD8/fisiologia , Imunofenotipagem , Terapia de Imunossupressão , Interleucina-6/biossíntese , Hipersensibilidade Respiratória/imunologia , Hipersensibilidade Respiratória/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Animais , Antígenos CD8/genética , Células Cultivadas , Feminino , Fatores de Transcrição Forkhead/deficiência , Fatores de Transcrição Forkhead/genética , Interleucina-10/antagonistas & inibidores , Interleucina-6/deficiência , Interleucina-6/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Interleucina-6/antagonistas & inibidores , Receptores de Interleucina-6/fisiologia , Hipersensibilidade Respiratória/genética , Fator de Crescimento Transformador beta/antagonistas & inibidores
18.
J Allergy Clin Immunol ; 126(2): 306-16, 316.e1-12, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20624645

RESUMO

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.


Assuntos
Arachis/efeitos adversos , Imunoglobulina E/imunologia , Interleucina-13/imunologia , Mastócitos/imunologia , Hipersensibilidade a Amendoim/imunologia , Receptores de IgE/imunologia , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Imunidade nas Mucosas/genética , Imunidade nas Mucosas/imunologia , Imunoglobulina E/genética , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/farmacologia , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Interleucina-13/antagonistas & inibidores , Interleucina-13/genética , Subunidade alfa2 de Receptor de Interleucina-13/genética , Interleucina-17/genética , Interleucina-17/imunologia , Interleucina-4/genética , Interleucina-4/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Intestinos/imunologia , Intestinos/patologia , Mastócitos/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Hipersensibilidade a Amendoim/tratamento farmacológico , Hipersensibilidade a Amendoim/genética , Hipersensibilidade a Amendoim/patologia , Receptores de IgE/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacologia
19.
J Immunol ; 185(1): 253-62, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20525882

RESUMO

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.


Assuntos
Alérgenos/administração & dosagem , Interferon gama/biossíntese , Células T Matadoras Naturais/imunologia , Ovalbumina/imunologia , Hipersensibilidade Respiratória/imunologia , Hipersensibilidade Respiratória/prevenção & controle , Transferência Adotiva , Alérgenos/imunologia , Animais , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Hiper-Reatividade Brônquica/imunologia , Hiper-Reatividade Brônquica/patologia , Hiper-Reatividade Brônquica/prevenção & controle , Células Cultivadas , Técnicas de Cocultura , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/transplante , Feminino , Galactosilceramidas/administração & dosagem , Galactosilceramidas/imunologia , Galactosilceramidas/metabolismo , Mediadores da Inflamação/administração & dosagem , Mediadores da Inflamação/metabolismo , Interferon gama/deficiência , Interferon gama/fisiologia , Intubação Intratraqueal , Ligantes , Fígado/imunologia , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células T Matadoras Naturais/metabolismo , Células T Matadoras Naturais/patologia , Ovalbumina/administração & dosagem , Hipersensibilidade Respiratória/patologia
20.
Am J Respir Crit Care Med ; 182(4): 455-63, 2010 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-20442434

RESUMO

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.


Assuntos
Acetatos/farmacologia , Antiasmáticos/farmacologia , Inflamação/prevenção & controle , Quinolinas/farmacologia , Hipersensibilidade Respiratória/prevenção & controle , Infecções por Vírus Respiratório Sincicial/complicações , Vírus Sinciciais Respiratórios/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Bronquiolite Viral/complicações , Bronquiolite Viral/prevenção & controle , Bronquiolite Viral/virologia , Líquido da Lavagem Broncoalveolar/virologia , Ciclopropanos , Cisteína/antagonistas & inibidores , Modelos Animais de Doenças , Inflamação/complicações , Interferon gama/efeitos dos fármacos , Antagonistas de Leucotrienos/farmacologia , Leucotrienos , Camundongos , Camundongos Endogâmicos BALB C , Recidiva , Hipersensibilidade Respiratória/etiologia , Sulfetos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...