Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS One ; 10(2): e0114831, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25723478

RESUMEN

Eosinophilic Esophagitis (EoE) is a chronic allergic disorder, whose pathobiology is incompletely understood. Histamine-producing cells including mast cells and basophils have been implicated in EoE. However, very little is currently known about the role of histamine and histamine receptor (HR) expression and signaling in the esophageal epithelium. Herein, we characterized HR (H1R, H2R, H3R, and H4R) expression in human esophageal biopsies and investigate the role of histamine signaling in inducible cytokine expression in human esophageal epithelial cells in vitro. HR expression was quantified in esophageal biopsies from non-EoE control (N = 23), inactive EoE (<15 eos/hpf, N = 26) and active EoE (>15 eos/hpf, N = 22) subjects using qRT-PCR and immunofluorescent localization. HR expression and histamine-mediated cytokine secretion were evaluated in human primary and telomerase-immortalized esophageal epithelial cells. H1R, H2R, and H4R expression were increased in active EoE biopsies compared to inactive EoE and controls. H2R was the most abundantly expressed receptor, and H3R expression was negligible in all 3 cohorts. Infiltrating eosinophils expressed H1R, H2R, and H4R, which contributed to the observed increase in HR in active subjects. H1R and H2R, but not H3R or H4R, were constitutively expressed by primary and immortalized cells, and epithelial histamine stimulation induced GM-CSF, TNFα, and IL-8, but not TSLP or eotaxin-3 secretion. Epithelial priming with the TLR3 ligand poly (I:C) induced H1R and H2R expression, and enhanced histamine-induced GM-CSF, TNFα, and IL-8 secretion. These effects were primarily suppressed by H1R antagonists, but unaffected by H2R antagonism. Histamine directly activates esophageal epithelial cytokine secretion in vitro in an H1R dependent fashion. However, H1R, H2R and H4R are induced in active inflammation in EoE in vivo. While systemic antihistamine (anti-H1R) therapy may not induce clinical remission in EoE, our study suggests that further study of histamine receptor signaling in EoE is warranted and that targeting of additional histamine receptors may lead to novel treatment strategies for this important disease.


Asunto(s)
Esofagitis Eosinofílica/genética , Expresión Génica , Receptores Histamínicos/genética , Adolescente , Biopsia , Recuento de Células , Línea Celular , Niño , Preescolar , Esofagitis Eosinofílica/etiología , Esofagitis Eosinofílica/metabolismo , Esofagitis Eosinofílica/patología , Eosinófilos/patología , Células Epiteliales/metabolismo , Femenino , Estudios de Asociación Genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Histamina/metabolismo , Humanos , Lactante , Interleucina-8/metabolismo , Masculino , Membrana Mucosa/metabolismo , Membrana Mucosa/patología , Receptores Histamínicos/metabolismo , Receptores Histamínicos H1/genética , Receptores Histamínicos H1/metabolismo , Receptores Histamínicos H2/genética , Receptores Histamínicos H2/metabolismo , Receptores Histamínicos H3/genética , Receptores Histamínicos H3/metabolismo , Receptor Toll-Like 3/agonistas , Receptor Toll-Like 3/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
2.
Exp Cell Res ; 319(6): 850-9, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23237990

RESUMEN

BACKGROUND: Esophageal fibrosis is a complication of eosinophilic esophagitis (EoE) which has been attributed to both subepithelial fibrosis and to epithelial to mesenchymal transition (EMT), a process by which epithelial cells acquire mesenchymal features. Common to both causes of EoE-fibrosis is the notion that granulocyte-derived TGF-ß, induces myofibroblast differentiation of the target cell. To date, the role of esophageal epithelial cells as effector cells in esophageal fibrosis has never been explored. Herein, we investigated consequences of cross-talk between esophageal epithelial cells and fibroblasts, and identified profibrotic cytokines which influence the development of EMT in vitro. METHODS AND RESULTS: Stimulation of primary fetal esophageal fibroblasts (FEF3) with conditioned media (CEM) from esophageal epithelial cells (EPC2-hTERT), primed FEF3 cells to secrete IL-1ß and TNFα, but not TGFß. To determine whether these cytokines signaled in a paracrine fashion to esophageal epithelial cells, FEF3 cells were stimulated with CEM, followed by transfer of this fibroblast conditioned media (FCM) to EPC2-hTERT cells. Epithelial FCM stimulation increased expression of mesenchymal markers and reduced E-cadherin expression, features of EMT which were TNFα and IL-1ß-dependent. Using organotypic culture models, primary EoE epithelial cells exhibited features of EMT compared to non-EoE cells, corresponding to patterns of EMT in native biopsies. CONCLUSIONS: Esophageal epithelial cell and fibroblast cross-talk contributes to esophageal fibrosis. Our results suggest that features of EMT can develop independent of TGF-ß and granulocytes, which may have important implications in treatment of EoE.


Asunto(s)
Comunicación Celular , Esofagitis Eosinofílica/patología , Células Epiteliales/citología , Transición Epitelial-Mesenquimal , Esófago/citología , Adolescente , Biopsia , Cadherinas/metabolismo , Diferenciación Celular , Células Cultivadas , Niño , Medios de Cultivo Condicionados/metabolismo , Medios de Cultivo Condicionados/farmacología , Esofagitis Eosinofílica/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Esófago/efectos de los fármacos , Esófago/metabolismo , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Granulocitos/metabolismo , Humanos , Interleucina-1beta/metabolismo , Interleucina-1beta/farmacología , Masculino , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
3.
Artículo en Inglés | MEDLINE | ID: mdl-21474651

RESUMEN

The mechanisms by which gastroesophageal reflux disease esophagitis develops are controversial. Although many support the notion that caustic injury leads to reflux esophagitis, others have proposed that reflux esophagitis is caused by esophageal epithelial cytokine-mediated inflammation. We previously demonstrated that Toll-like receptor 3 (TLR3) is highly expressed and functional in the nontransformed human esophageal epithelial cell line EPC2-hTERT. In addition to activation by viral double-stranded RNA, TLR3 can be activated by endogenous mRNA released by necrotic cells. In the present study, we investigated the role of esophageal epithelial TLR3 to sense danger signals released by necrotic esophageal epithelial cells in vitro. Following induction of freeze-thaw necrosis, necrotic EPC2-hTERT cell supernatants (NCS) were used to stimulate EPC2-hTERT monolayers, leading to NF-κB-dependent induction of IL-8 mRNA expression. Responses to self-derived NCS were not observed in transformed gastrointestinal epithelial cell lines, including TE-1 and Caco-2 cells, suggesting that the ability to sense endogenous danger signals is unique to nontransformed esophageal epithelial cells. To determine the immunostimulatory role of epithelial RNA, EPC2-hTERT cells were stimulated with self-derived mRNA, which significantly induced IL-8 mRNA expression. Finally, suppression of TLR3 signaling in a DN-TLR3 cell line, hTERT-ΔTIR-TLR3, led to reduced NCS-induced IL-8 induction by both NCS and mRNA stimulation. Our results demonstrate that human esophageal epithelial cells can sense endogenous danger signals, in part through TLR3 signaling. This supports the concept that epithelial injury plays an inciting role in the pathogenesis of reflux-induced esophagitis, providing important insights into the mechanisms by which epithelial injury leads to inflammation.


Asunto(s)
Esófago/metabolismo , Esófago/patología , Receptor Toll-Like 3/metabolismo , Células CACO-2 , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Interleucina-8/biosíntesis , FN-kappa B/metabolismo , Necrosis/metabolismo , Necrosis/patología , Transducción de Señal
4.
Am J Physiol Gastrointest Liver Physiol ; 297(6): G1172-80, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19779021

RESUMEN

Despite its position at the front line against ingested pathogens, very little is presently known about the role of the esophageal epithelium in host innate immune defense. As a key player in the innate immune response, Toll-like receptor (TLR) signaling has not been well characterized in human esophageal epithelial cells. In the present study, we investigated the inflammatory response and signaling pathways activated by TLR stimulation of human esophageal cells in vitro. Using quantitative RT-PCR, we profiled the expression pattern of human TLRs 1-10 in primary esophageal keratinocytes (EPC2), immortalized nontransformed esophageal keratinocytes (EPC2-hTERT), and normal human esophageal mucosal biopsies and found that TLRs 1, 2, 3, and 5 were expressed both in vivo and in vitro. Using the cytokine IL-8 as a physiological read out of the inflammatory response, we found that TLR3 is the most functional of the expressed TLRs in both primary and immortalized esophageal epithelial cell lines in response to its synthetic ligand polyinosinic polycytidylic acid [poly(I:C)]. Through reporter gene studies, we show that poly(I:C)-induced NF-kappaB activation is critical for the transactivation of the IL-8 promoter in vitro and that nuclear translocation of NF-kappaB occurs at an early time point following poly(I:C) stimulation of esophageal epithelial cells. Importantly, we also show that poly(I:C) stimulation induces the NF-kappaB-dependent esophageal epithelial expression of TLR2, leading to enhanced epithelial responsiveness of EPC2-hTERT cells to TLR2 ligand stimulation, suggesting an important regulatory role for TLR3-mediated NF-kappaB signaling in the innate immune response of esophageal epithelial cells. Our findings demonstrate for the first time that TLR3 is highly functional in the human esophageal epithelium and that TLR3-mediated NF-kappaB signaling may play an important regulatory role in esophageal epithelial homeostasis.


Asunto(s)
Esófago/inmunología , Inmunidad Innata , Queratinocitos/inmunología , Transducción de Señal , Receptor Toll-Like 3/metabolismo , Factor de Transcripción ReIA/metabolismo , Transporte Activo de Núcleo Celular , Biopsia , Células Cultivadas , Esófago/efectos de los fármacos , Humanos , Inmunidad Innata/efectos de los fármacos , Mediadores de Inflamación/metabolismo , Interleucina-8/metabolismo , Queratinocitos/efectos de los fármacos , Membrana Mucosa/inmunología , Poli I-C/farmacología , Interferencia de ARN , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Telomerasa/genética , Factores de Tiempo , Receptor Toll-Like 1/metabolismo , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 3/genética , Receptor Toll-Like 5/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...