Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 2 de 2
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 10(7): e0134057, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26207385

RESUMO

Globally, asthma is a chronic inflammatory respiratory disease affecting over 300 million people. Some asthma patients remain poorly controlled by conventional therapies and experience more life-threatening exacerbations. Vitamin D, as an adjunct therapy, may improve disease control in severe asthma patients since vitamin D enhances glucocorticoid responsiveness and mitigates airway smooth muscle (ASM) hyperplasia. We sought to characterize differences in transcriptome responsiveness to vitamin D between fatal asthma- and non-asthma-derived ASM by using RNA-Seq to measure ASM transcript expression in five donors with fatal asthma and ten non-asthma-derived donors at baseline and with vitamin D treatment. Based on a Benjamini-Hochberg corrected p-value <0.05, 838 genes were differentially expressed in fatal asthma vs. non-asthma-derived ASM at baseline, and vitamin D treatment compared to baseline conditions induced differential expression of 711 and 867 genes in fatal asthma- and non-asthma-derived ASM, respectively. Functional gene categories that were highly represented in all groups included extracellular matrix, and responses to steroid hormone stimuli and wounding. Genes differentially expressed by vitamin D also included cytokine and chemokine activity categories. Follow-up qPCR and individual analyte ELISA experiments were conducted for four cytokines (i.e. CCL2, CCL13, CXCL12, IL8) to measure TNFα-induced changes by asthma status and vitamin D treatment. Vitamin D inhibited TNFα-induced IL8 protein secretion levels to a comparable degree in fatal asthma- and non-asthma-derived ASM even though IL8 had significantly higher baseline levels in fatal asthma-derived ASM. Our findings identify vitamin D-specific gene targets and provide transcriptomic data to explore differences in the ASM of fatal asthma- and non-asthma-derived donors.


Assuntos
Asma/metabolismo , Músculo Liso/efeitos dos fármacos , Sistema Respiratório/efeitos dos fármacos , Transcriptoma/efeitos dos fármacos , Vitamina D/farmacologia , Adolescente , Adulto , Asma/tratamento farmacológico , Asma/genética , Quimiocinas/genética , Quimiocinas/metabolismo , Criança , Citocinas/genética , Citocinas/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Pessoa de Meia-Idade , Músculo Liso/metabolismo , Sistema Respiratório/metabolismo , Vitamina D/uso terapêutico , Adulto Jovem
2.
PLoS One ; 9(6): e99625, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24926665

RESUMO

Asthma is a chronic inflammatory respiratory disease that affects over 300 million people worldwide. Glucocorticoids are a mainstay therapy for asthma because they exert anti-inflammatory effects in multiple lung tissues, including the airway smooth muscle (ASM). However, the mechanism by which glucocorticoids suppress inflammation in ASM remains poorly understood. Using RNA-Seq, a high-throughput sequencing method, we characterized transcriptomic changes in four primary human ASM cell lines that were treated with dexamethasone--a potent synthetic glucocorticoid (1 µM for 18 hours). Based on a Benjamini-Hochberg corrected p-value <0.05, we identified 316 differentially expressed genes, including both well known (DUSP1, KLF15, PER1, TSC22D3) and less investigated (C7, CCDC69, CRISPLD2) glucocorticoid-responsive genes. CRISPLD2, which encodes a secreted protein previously implicated in lung development and endotoxin regulation, was found to have SNPs that were moderately associated with inhaled corticosteroid resistance and bronchodilator response among asthma patients in two previously conducted genome-wide association studies. Quantitative RT-PCR and Western blotting showed that dexamethasone treatment significantly increased CRISPLD2 mRNA and protein expression in ASM cells. CRISPLD2 expression was also induced by the inflammatory cytokine IL1ß, and small interfering RNA-mediated knockdown of CRISPLD2 further increased IL1ß-induced expression of IL6 and IL8. Our findings offer a comprehensive view of the effect of a glucocorticoid on the ASM transcriptome and identify CRISPLD2 as an asthma pharmacogenetics candidate gene that regulates anti-inflammatory effects of glucocorticoids in the ASM.


Assuntos
Anti-Inflamatórios/farmacologia , Asma/tratamento farmacológico , Moléculas de Adesão Celular/genética , Dexametasona/farmacologia , Fatores Reguladores de Interferon/genética , Pulmão/citologia , Administração por Inalação , Anti-Inflamatórios/administração & dosagem , Asma/genética , Asma/metabolismo , Moléculas de Adesão Celular/metabolismo , Linhagem Celular , Dexametasona/administração & dosagem , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Fatores Reguladores de Interferon/metabolismo , Pulmão/patologia , Músculo Liso/efeitos dos fármacos , Polimorfismo de Nucleotídeo Único , Análise de Sequência de RNA/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA