Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Immunol ; 199(7): 2215-2224, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28827284

RESUMO

Orosomucoid like 3 (ORMDL3), a gene localized to chromosome 17q21, has been linked in epidemiologic studies to childhood asthma and rhinovirus (RV) infections. As the single nucleotide polymorphisms linking ORMDL3 to asthma are associated with increased expression of ORMDL3, we have used hORMDL3zp3-Cre mice (which have universal increased expression of human ORMDL3) to determine whether infection of these transgenic mice with RV influences levels of airway inflammation or RV viral load. RV infection of hORMDL3zp3-Cre mice resulted in reduced RV viral load assessed by quantitative real-time PCR (lung and airway epithelium), as well as reduced airway inflammation (total bronchoalveolar lavage cells, neutrophils, macrophages, and lymphocytes) compared with RV-infected wild-type mice. Levels of the antiviral pathways including IFNs (IFN-α, IFN-ß, IFN-λ) and RNAse L were significantly increased in the lungs of RV-infected hORMDL3zp3-Cre mice. Levels of the antiviral mouse oligoadenylate synthetase (mOas)1g pathway and RNAse L were upregulated in the lungs of unchallenged hORMDL3zp3-Cre mice. In addition, levels of mOas2, but not mOas1 (mOas1a, mOas1b, mOas1g), or mOas3 pathways were significantly more upregulated by IFNs (IFN-α, IFN-ß, IFN-λ) in epithelial cells from hORMDL3zp3-Cre mice compared with RV-infected wild-type mouse epithelial cells. RNAse L-deficient mice infected with RV had increased RV viral load. Overall, these studies suggest that increased levels of ORMDL3 contribute to antiviral defense to RV infection in mice through pathways that may include IFNs (IFN-α, IFN-ß, IFN-λ), OAS, and RNAse L.


Assuntos
Pulmão/virologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Infecções por Picornaviridae/imunologia , Infecções por Picornaviridae/virologia , Rhinovirus/isolamento & purificação , 2',5'-Oligoadenilato Sintetase/genética , 2',5'-Oligoadenilato Sintetase/metabolismo , Animais , Asma/imunologia , Asma/virologia , Endorribonucleases/deficiência , Endorribonucleases/genética , Endorribonucleases/metabolismo , Células Epiteliais/virologia , Inflamação/imunologia , Inflamação/virologia , Interferon beta/biossíntese , Interferon beta/genética , Interferon beta/imunologia , Interferons/biossíntese , Interferons/genética , Interferons/imunologia , Pulmão/imunologia , Camundongos , Camundongos Transgênicos , Infecções por Picornaviridae/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Carga Viral
2.
J Immunol ; 198(8): 3017-3022, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28275141

RESUMO

In this study, we used cre-lox techniques to generate mice selectively deficient in ORMDL3 in airway epithelium (Ormdl3Δ2-3/Δ2-3/CC10) to simulate an inhaled therapy that effectively inhibited ORMDL3 expression in the airway. In contrast to the anticipated reduction in airway hyperresponsiveness (AHR), OVA allergen-challenged Ormdl3Δ2-3/Δ2-3/CC10 mice had a significant increase in AHR compared with wild-type mice. Levels of airway inflammation, mucus, fibrosis, and airway smooth muscle were no different in Ormdl3Δ2-3/Δ2-3/CC10 and wild-type mice. However, levels of sphingosine-1-phosphate (S1P) were significantly increased in Ormdl3Δ2-3/Δ2-3/CC10 mice as well as in airway epithelial cells in which ORMDL3 was inhibited with small interfering RNA. Incubation of S1P with airway smooth muscle cells significantly increased contractility. Overall, Ormdl3Δ2-3/Δ2-3/CC10 mice exhibit increased allergen-induced AHR independent of inflammation and associated with increased S1P generation. These studies raise concerns for inhaled therapies that selectively and effectively inhibit ORMDL3 in airway epithelium in asthma.


Assuntos
Asma/metabolismo , Proteínas de Membrana/antagonistas & inibidores , Hipersensibilidade Respiratória/metabolismo , Animais , Asma/imunologia , Modelos Animais de Doenças , Lisofosfolipídeos/imunologia , Lisofosfolipídeos/metabolismo , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase , Hipersensibilidade Respiratória/imunologia , Esfingosina/análogos & derivados , Esfingosina/imunologia , Esfingosina/metabolismo
3.
Proc Natl Acad Sci U S A ; 113(46): 13132-13137, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27799535

RESUMO

Gasdermin B (GSDMB) on chromosome 17q21 demonstrates a strong genetic linkage to asthma, but its function in asthma is unknown. Here we identified that GSDMB is highly expressed in lung bronchial epithelium in human asthma. Overexpression of GSDMB in primary human bronchial epithelium increased expression of genes important to both airway remodeling [TGF-ß1, 5-lipoxygenase (5-LO)] and airway-hyperresponsiveness (AHR) (5-LO). Interestingly, hGSDMBZp3-Cre mice expressing increased levels of the human GSDMB transgene showed a significant spontaneous increase in AHR and a significant spontaneous increase in airway remodeling, with increased smooth muscle mass and increased fibrosis in the absence of airway inflammation. In addition, hGSDMBZp3-Cre mice showed increases in the same remodeling and AHR mediators (TGF-ß1, 5-LO) observed in vitro in GSDMB-overexpressing epithelial cells. GSDMB induces TGF-ß1 expression via induction of 5-LO, because knockdown of 5-LO in epithelial cells overexpressing GSDMB inhibited TGF-ß1 expression. These studies demonstrate that GSDMB, a gene highly linked to asthma but whose function in asthma is previously unknown, regulates AHR and airway remodeling without airway inflammation through a previously unrecognized pathway in which GSDMB induces 5-LO to induce TGF-ß1 in bronchial epithelium.


Assuntos
Remodelação das Vias Aéreas/genética , Asma/genética , Hiper-Reatividade Brônquica/genética , Proteínas de Neoplasias/genética , Remodelação das Vias Aéreas/imunologia , Animais , Antígenos de Dermatophagoides/imunologia , Araquidonato 5-Lipoxigenase/genética , Araquidonato 5-Lipoxigenase/metabolismo , Asma/imunologia , Asma/metabolismo , Hiper-Reatividade Brônquica/imunologia , Hiper-Reatividade Brônquica/metabolismo , Células Cultivadas , Colágeno/metabolismo , Citocinas/genética , Citocinas/imunologia , Citocinas/metabolismo , Células Epiteliais/metabolismo , Humanos , Pulmão/citologia , Pulmão/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Camundongos Transgênicos , Fenótipo , RNA Mensageiro/metabolismo , Mucosa Respiratória/metabolismo
4.
J Allergy Clin Immunol ; 141(1): 329-338.e12, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28366795

RESUMO

BACKGROUND: Group 2 innate lymphoid cells (ILC2s) expand in the lungs of mice during type 2 inflammation induced by the fungal allergen Alternaria alternata. The increase in ILC2 numbers in the lung has been largely attributed to local proliferation and whether ILC2s migrate from the circulation to the lung after Alternaria exposure is unknown. OBJECTIVE: We examined whether human (lung, lymph node, and blood) and mouse lung ILC2s express ß1 and ß2 integrin adhesion molecules and whether these integrins are required for trafficking of ILC2s into the lungs of mice. METHODS: Human and mouse ILC2s were assessed for surface expression of ß1 and ß2 integrin adhesion molecules by using flow cytometry. The role of ß1 and ß2 integrins in ILC2 trafficking to the lungs was assessed by in vivo blocking of these integrins before airway exposure to Alternaria in mice. RESULTS: Both human and mouse lung ILC2s express high levels of ß1 and ß2 integrin adhesion receptors. Intranasal administration of Alternaria challenge reduced ILC2 numbers in the bone marrow and concurrently increased blood and lung ILC2 numbers. In vivo blocking of ß2 integrins (CD18) significantly reduced ILC2 numbers in the lungs but did not alter ILC2 proliferation, apoptosis, and function. In contrast, in vivo blocking of ß1 integrins or α4 integrins did not affect lung ILC2 numbers. CONCLUSION: ILC2 numbers increase in the mouse lung not only through local proliferation but also through trafficking from the circulation into the lung using ß2 rather than ß1 or α4 integrins.


Assuntos
Alternaria/imunologia , Antígenos CD18/metabolismo , Imunidade Inata , Integrina beta1/metabolismo , Pulmão/imunologia , Pulmão/metabolismo , Subpopulações de Linfócitos/imunologia , Subpopulações de Linfócitos/metabolismo , Animais , Apoptose/imunologia , Biomarcadores , Medula Óssea/imunologia , Medula Óssea/metabolismo , Antígenos CD18/genética , Citocinas/metabolismo , Citometria de Fluxo , Expressão Gênica , Humanos , Integrina beta1/genética , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Selectina L/genética , Selectina L/metabolismo , Pulmão/patologia , Contagem de Linfócitos , Camundongos , Células Th2/imunologia , Células Th2/metabolismo
5.
J Immunol ; 195(8): 3546-56, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26355153

RESUMO

Chronic asthma is associated with airway remodeling and decline in lung function. In this article, we show that follistatin-like 1 (Fstl1), a mediator not previously associated with asthma, is highly expressed by macrophages in the lungs of humans with severe asthma. Chronic allergen-challenged Lys-Cre(tg) /Fstl1(Δ/Δ) mice in whom Fstl1 is inactivated in macrophages/myeloid cells had significantly reduced airway remodeling and reduced levels of oncostatin M (OSM), a cytokine previously not known to be regulated by Fstl1. The importance of the Fstl1 induction of OSM to airway remodeling was demonstrated in murine studies in which administration of Fstl1 induced airway remodeling and increased OSM, whereas administration of an anti-OSM Ab blocked the effect of Fstl1 on inducing airway remodeling, eosinophilic airway inflammation, and airway hyperresponsiveness, all cardinal features of asthma. Overall, these studies demonstrate that the Fstl1/OSM pathway may be a novel pathway to inhibit airway remodeling in severe human asthma.


Assuntos
Remodelação das Vias Aéreas/imunologia , Asma/imunologia , Proteínas Relacionadas à Folistatina/imunologia , Oncostatina M/imunologia , Transdução de Sinais/imunologia , Remodelação das Vias Aéreas/efeitos dos fármacos , Remodelação das Vias Aéreas/genética , Animais , Anticorpos/imunologia , Anticorpos/farmacologia , Asma/genética , Asma/patologia , Feminino , Proteínas Relacionadas à Folistatina/genética , Humanos , Macrófagos/imunologia , Macrófagos/patologia , Masculino , Camundongos , Oncostatina M/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
6.
J Immunol ; 192(8): 3475-87, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24623133

RESUMO

Orosomucoid-like (ORMDL)3 has been strongly linked with asthma in genetic association studies. Because allergen challenge induces lung ORMDL3 expression in wild-type mice, we have generated human ORMDL3 zona pellucida 3 Cre (hORMDL3(zp3-Cre)) mice that overexpress human ORMDL3 universally to investigate the role of ORMDL3 in regulating airway inflammation and remodeling. These hORMDL3(zp3-Cre) mice have significantly increased levels of airway remodeling, including increased airway smooth muscle, subepithelial fibrosis, and mucus. hORMDL3(zp3-Cre) mice had spontaneously increased airway responsiveness to methacholine compared to wild-type mice. This increased airway remodeling was associated with selective activation of the unfolded protein response pathway transcription factor ATF6 (but not Ire1 or PERK). The ATF6 target gene SERCA2b, implicated in airway remodeling in asthma, was strongly induced in the lungs of hORMDL3(zp3-Cre) mice. Additionally, increased levels of expression of genes associated with airway remodeling (TGF-ß1, ADAM8) were detected in airway epithelium of these mice. Increased levels of airway remodeling preceded increased levels of airway inflammation in hORMDL3(zp3-Cre) mice. hORMDL3(zp3-Cre) mice had increased levels of IgE, with no change in levels of IgG, IgM, and IgA. These studies provide evidence that ORMDL3 plays an important role in vivo in airway remodeling potentially through ATF6 target genes such as SERCA2b and/or through ATF6-independent genes (TGF-ß1, ADAM8).


Assuntos
Remodelação das Vias Aéreas/genética , Remodelação das Vias Aéreas/imunologia , Asma/genética , Asma/imunologia , Proteínas de Membrana/genética , Fator 6 Ativador da Transcrição/metabolismo , Alérgenos/imunologia , Animais , Especificidade de Anticorpos/imunologia , Asma/patologia , Hiper-Reatividade Brônquica/induzido quimicamente , Quimiocinas CC/metabolismo , Quimiocinas CXC/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Eosinófilos/imunologia , Eosinófilos/metabolismo , Expressão Gênica , Ordem dos Genes , Marcação de Genes , Humanos , Imunoglobulina E/sangue , Imunoglobulina E/imunologia , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Cloreto de Metacolina/administração & dosagem , Camundongos , Camundongos Transgênicos , Ovalbumina/imunologia , Células Th2/imunologia , Células Th2/metabolismo , Transgenes , Resposta a Proteínas não Dobradas , eIF-2 Quinase/metabolismo
7.
J Pediatr Gastroenterol Nutr ; 59(1): 10-6, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24590208

RESUMO

OBJECTIVES: Eosinophilic esophagitis (EoE) is a food-triggered disease associated with esophageal fibrosis and stricture formation in a subset of patients. In the present study we used a murine model of egg (ovalbumin [OVA])-induced EoE to determine whether inhibiting transforming growth factor-ß1 (TGF-ß1) signaling through the Smad3 pathway would inhibit features of esophageal remodeling including fibrosis, angiogenesis, and basal zone hyperplasia. METHODS: Wild-type (WT) and Smad3-deficient (KO [knockout]) mice were sensitized intraperitoneally and then challenged chronically with intraesophageal OVA for 1 month. Levels of esophageal eosinophils, esophageal TGF-ß1+ and vascular endothelial growth factor (VEGF)+ cells, and features of esophageal remodeling (fibrosis, angiogenesis, basal zone hyperplasia) were quantitated by immunohistochemistry and image analysis. RESULTS: OVA challenge induced a similar increase in the levels of esophageal major basic protein (MBP)+ eosinophils and esophageal TGF-ß1+ cells in WT and Smad3 KO mice. Smad3 KO mice challenged with OVA had significantly less esophageal fibrosis and esophageal angiogenesis compared with OVA-challenged WT mice. The reduced esophageal angiogenesis in Smad3 KO mice was associated with reduced numbers of VEGF+ cells in the esophagus. There was a trend toward OVA-challenged Smad3 KO to have reduced basal zone hyperplasia, but this was not statistically significant. CONCLUSIONS: In a mouse model of egg-induced EoE, Smad3-deficient mice have significantly less esophageal remodeling, especially fibrosis and angiogenesis that is associated with reduced expression of VEGF. Targeting the TGF-ß1/Smad3 pathway may be a novel strategy to reduce esophageal fibrosis and its associated complications such as esophageal strictures in EoE.


Assuntos
Esofagite Eosinofílica/metabolismo , Esofagite Eosinofílica/patologia , Esôfago/metabolismo , Esôfago/patologia , Proteína Smad3/deficiência , Animais , Modelos Animais de Doenças , Proteína Básica Maior de Eosinófilos/metabolismo , Esofagite Eosinofílica/induzido quimicamente , Eosinófilos/química , Esôfago/irrigação sanguínea , Feminino , Fibrose , Hiperplasia , Camundongos , Camundongos Knockout , Neovascularização Patológica/patologia , Ovalbumina , RNA Mensageiro/metabolismo , Transdução de Sinais , Proteína Smad3/genética , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
10.
Nat Commun ; 14(1): 5814, 2023 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-37726288

RESUMO

Epithelial plasticity has been suggested in lungs of mice following genetic depletion of stem cells but is of unknown physiological relevance. Viral infection and chronic lung disease share similar pathological features of stem cell loss in alveoli, basal cell (BC) hyperplasia in small airways, and innate immune activation, that contribute to epithelial remodeling and loss of lung function. We show that a subset of distal airway secretory cells, intralobar serous (IS) cells, are activated to assume BC fates following influenza virus infection. Injury-induced hyperplastic BC (hBC) differ from pre-existing BC by high expression of IL-22Ra1 and undergo IL-22-dependent expansion for colonization of injured alveoli. Resolution of virus-elicited inflammation results in BC to IS re-differentiation in repopulated alveoli, and increased local expression of protective antimicrobial factors, but fails to restore normal alveolar epithelium responsible for gas exchange.


Assuntos
Células Epiteliais , Alvéolos Pulmonares , Animais , Camundongos , Diferenciação Celular , Hiperplasia , Imunidade Inata
13.
Biomolecules ; 12(9)2022 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-36139112

RESUMO

The alveolar epithelium serves as a barrier between the body and the external environment. To maintain efficient gas exchange, the alveolar epithelium has evolved to withstand and rapidly respond to an assortment of inhaled, injury-inducing stimuli. However, alveolar damage can lead to loss of alveolar fluid barrier function and exuberant, non-resolving inflammation that manifests clinically as acute respiratory distress syndrome (ARDS). This review discusses recent discoveries related to mechanisms of alveolar homeostasis, injury, repair, and regeneration, with a contemporary emphasis on virus-induced lung injury. In addition, we address new insights into how the alveolar epithelium coordinates injury-induced lung inflammation and review maladaptive lung responses to alveolar damage that drive ARDS and pathologic lung remodeling.


Assuntos
Lesão Pulmonar , Síndrome do Desconforto Respiratório , Epitélio , Humanos , Pulmão/patologia , Lesão Pulmonar/patologia , Mucosa Respiratória
15.
Front Immunol ; 9: 2817, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30564235

RESUMO

Using an in vivo model of tolerance to TLR7-induced skin inflammation, we found a critical role for macrophage-derived MMP10 in mediating immune hypo-responsiveness. Cutaneous exposure to Imiquimod (IMQ), a TLR7 agonist, induced acute expression of pro-inflammatory factors (IL1ß, IL6, CXCL1) and neutrophil influx equally in both wildtype and Mmp10-/- mice. However, whereas subsequent exposure (11 and 12 days later) to IMQ led to marked abrogation of pro-inflammatory factor expression in wildtype mice, Mmp10-/- mice responded similarly as they did to the first application. In addition, the second exposure led to increased expression of negative regulators of TLR signaling (TNFAIP3, IRAK3) and immunosuppressive cytokines (IL10, TGFß1) in wildtype mice but not in Mmp10-/- mice. In vitro studies demonstrated that prior exposure of IMQ to bone marrow-derived macrophages (BMDM) made wildtype cells refractory to subsequent stimulation but did not for Mmp10-/- macrophages. These findings expand the critical roles MMP10 plays in controlling macrophage activation to indicate that the development of immune tolerance to TLR7 ligand is dependent on this macrophage-derived proteinase.


Assuntos
Tolerância Imunológica/imunologia , Macrófagos/imunologia , Metaloproteinase 10 da Matriz/imunologia , Glicoproteínas de Membrana/imunologia , Receptor 7 Toll-Like/imunologia , Animais , Citocinas/imunologia , Feminino , Imiquimode/farmacologia , Tolerância Imunológica/efeitos dos fármacos , Ativação de Macrófagos/efeitos dos fármacos , Ativação de Macrófagos/imunologia , Macrófagos/efeitos dos fármacos , Masculino , Glicoproteínas de Membrana/agonistas , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Receptor 7 Toll-Like/agonistas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA