Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-36262892

RESUMEN

Introduction: Studies have demonstrated that primary care clinicians can achieve the same excellent outcomes in treatment of hepatitis C (HCV) infection as specialist physicians but there is a dearth of literature on experiences and outcomes of treatment of HCV infection in residency clinics. We sought to describe the perspectives of internal medicine resident physicians in one community-based residency program toward treating HCV infection before and after launching treatment of HCV infection within the residency clinic. Further, this study examined outcomes of patients treated by the resident physicians. Methods: Treatment of HCV infection was initiated in 2019. Residents were invited to complete a baseline survey. Residents who treated at least one patient with HCV infection were invited to complete a subsequent survey. Comparative analysis was performed using Fisher's Exact test. Sustained virologic response at least 12 weeks (SVR-12) after completion of treatment in patients initiated on therapy in the residency clinic was assessed. Results: Residents (n = 12) who treated patients for HCV infection reported significantly greater knowledge in evaluating and treating patients with HCV infection and preparedness to provide this care after residency than residents (n = 34) who completed the baseline survey (p < 0.001). Twenty-six patients were initiated on direct-acting antiviral (DAA) therapy. All 21 patients who were tested achieved SVR-12. Conclusions: Training resident physicians to evaluate and treat HCV infection can improve outcomes for underserved patients in residency clinics while preparing a pool of physicians to provide this care after residency.

3.
Mol Cancer Res ; 14(7): 647-59, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27084312

RESUMEN

UNLABELLED: The mechanism underlying the progression of normal esophageal mucosa to esophageal adenocarcinoma remains elusive. WNT5A is a noncanonical WNT, which mainly functions via the receptor tyrosine kinase-like orphan receptor 2 (ROR2), and has an unclear role in carcinogenesis. In this study, we aimed to determine the role of WNT5A/ROR2 signaling in esophageal adenocarcinoma. Analysis of WNT5A and ROR2 expression patterns in healthy controls, Barrett and esophageal adenocarcinoma patients' esophageal clinical specimens as well as in various esophageal cell lines demonstrated a ROR2 overexpression in esophageal adenocarcinoma tissues compared with Barrett and healthy mucosa, whereas WNT5A expression was found significantly downregulated toward esophageal adenocarcinoma formation. Treatment of esophageal adenocarcinoma OE33 cells with human recombinant WNT5A (rhWNT5A) significantly suppressed proliferation, survival, and migration in a dose-dependent fashion. rhWNT5A was found to inhibit TOPflash activity in ROR2 wild-type cells, whereas increased TOPflash activity in ROR2-knockdown OE33 cells. In addition, ROR2 knockdown alone abolished cell proliferation and weakened the migration properties of OE33 cells. These findings support an early dysregulation of the noncanonical WNT5A/ROR2 pathway in the pathogenesis of esophageal adenocarcinoma, with the loss of WNT5A expression together with the ROR2 overexpression to be consistent with tumor promotion. IMPLICATIONS: The dysregulation of WNT5A/ROR2 noncanonical WNT signaling in Barrett-associated esophageal adenocarcinoma introduces possible prognostic markers and novel targets for tailored therapy of this malignancy. Mol Cancer Res; 14(7); 647-59. ©2016 AACR.


Asunto(s)
Adenocarcinoma/metabolismo , Esófago de Barrett/metabolismo , Neoplasias Esofágicas/metabolismo , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/metabolismo , Proteína Wnt-5a/metabolismo , Adenocarcinoma/patología , Adulto , Anciano , Esófago de Barrett/patología , Proliferación Celular/fisiología , Progresión de la Enfermedad , Neoplasias Esofágicas/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
4.
Neoplasia ; 17(7): 598-611, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26297437

RESUMEN

INTRODUCTION: Wnt/ß-catenin signaling activation has been reported only during the late steps of Barrett's esophagus (BE) neoplastic progression, but not in BE metaplasia, based on the absence of nuclear ß-catenin. However, ß-catenin transcriptional activity has been recorded in absence of robust nuclear accumulation. Thus, we aimed to investigate the Wnt/ß-catenin signaling in nondysplastic BE. METHODS: Esophageal tissues from healthy and BE patients without dysplasia were analyzed for Wnt target gene expression by quantitative reverse transcription polymerase chain reaction (qRT-PCR) and immunohistochemistry. Esophageal squamous (EPC1-& EPC2-hTERT), BE metaplastic (CP-A), and adenocarcinoma (OE33) cell lines were characterized for Wnt activation by qRT-PCR, Western blot, and luciferase assay. Wnt activity regulation was examined by using recombinant Wnt3a and Dickkopf-1 (Dkk1) as well as Dkk1 short interfering RNA. RESULTS: Wnt target genes (AXIN2, c-MYC, Cyclin D1, Dkk1) and Wnt3a were significantly upregulated in nondysplastic BE compared with squamous mucosa. Elevated levels of dephosphorylated ß-catenin were detected in nondysplastic BE. Nuclear active ß-catenin and TOPflash activity were increased in CP-A and OE33 cells compared with squamous cells. Wnt3a-mediated ß-catenin signaling activation was abolished by Dkk1 in CP-A cells. TOPFlash activity was elevated following Dkk1 silencing in CP-A but not in OE33 cells. Dysplastic and esophageal adenocarcinoma tissues demonstrated further Dkk1 and AXIN2 overexpression. CONCLUSIONS: Despite the absence of robust nuclear accumulation, ß-catenin is transcriptionally active in nondysplastic BE. Dkk1 overexpression regulates ß-catenin signaling in BE metaplastic but not in adenocarcinoma cells, suggesting that early perturbation of Dkk1-mediated signaling suppression may contribute to BE malignant transformation.


Asunto(s)
Adenocarcinoma/patología , Esófago de Barrett/patología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Vía de Señalización Wnt/fisiología , beta Catenina/metabolismo , Proteína Axina/biosíntesis , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Ciclina D1/biosíntesis , Activación Enzimática , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Interferencia de ARN , ARN Interferente Pequeño , Proteína Wnt3A/biosíntesis , Proteína Wnt3A/genética , Proteína Wnt3A/metabolismo , beta Catenina/genética
5.
Microvasc Res ; 97: 167-80, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25446010

RESUMEN

Polyphenolic compounds (anthocyanins, flavonoid glycosides) in berries prevent the initiation, promotion, and progression of carcinogenesis in rat's digestive tract and esophagus, in part, via anti-inflammatory pathways. Angiogenesis has been implicated in the pathogenesis of chronic inflammation and tumorigenesis. In this study, we investigated the anti-inflammatory and anti-angiogenic effects of black raspberry extract (BRE) on two organ specific primary human intestinal microvascular endothelial cells, (HIMEC) and human esophageal microvascular endothelial cells (HEMEC), isolated from surgically resected human intestinal and donor discarded esophagus, respectively. HEMEC and HIMEC were stimulated with TNF-α/IL-1ß with or without BRE. The anti-inflammatory effects of BRE were assessed based upon COX-2, ICAM-1 and VCAM-1 gene and protein expression, PGE2 production, NFκB p65 subunit nuclear translocation as well as endothelial cell-leukocyte adhesion. The anti-angiogenic effects of BRE were assessed on cell migration, proliferation and tube formation following VEGF stimulation as well as on activation of Akt, MAPK and JNK signaling pathways. BRE inhibited TNF-α/IL-1ß-induced NFκB p65 nuclear translocation, PGE2 production, up-regulation of COX-2, ICAM-1 and VCAM-1 gene and protein expression and leukocyte binding in HEMEC but not in HIMEC. BRE attenuated VEGF-induced cell migration, proliferation and tube formation in both HEMEC and HIMEC. The anti-angiogenic effect of BRE is mediated by inhibition of Akt, MAPK and JNK phosphorylations. BRE exerted differential anti-inflammatory effects between HEMEC and HIMEC following TNF-α/IL-1ß activation whereas demonstrated similar anti-angiogenic effects following VEGF stimulation in both cell lines. These findings may provide more insight into the anti-tumorigenic capacities of BRE in human disease and cancer.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antiinflamatorios/farmacología , Células Endoteliales/efectos de los fármacos , Esófago/irrigación sanguínea , Intestinos/irrigación sanguínea , Microvasos/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Extractos Vegetales/farmacología , Rubus , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Endoteliales/metabolismo , Frutas , Humanos , Mediadores de Inflamación/metabolismo , Microvasos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Fitoterapia , Plantas Medicinales , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
6.
Am J Physiol Cell Physiol ; 307(9): C859-77, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25163519

RESUMEN

Endothelial-mesenchymal transition (EndoMT) has been recognized as a key determinant of tumor microenvironment in cancer progression and metastasis. Endothelial cells undergoing EndoMT lose their endothelial markers, acquire the mesenchymal phenotype, and become more invasive with increased migratory abilities. Early stages of esophageal adenocarcinoma (EAC) are characterized by strong microvasculature whose impact in tumor progression remains undefined. Our aim was to determine the role of EndoMT in EAC by investigating the impact of tumor cells on normal primary human esophageal microvascular endothelial cells (HEMEC). HEMEC were either cocultured with OE33 adenocarcinoma cells or treated with IL-1ß and transforming growth factor-ß2 (TGF-ß2) for indicated periods and analyzed for EndoMT-associated changes by real-time PCR, Western blotting, immunofluorescence staining, and functional assays. Additionally, human EAC tissues were investigated for detection of EndoMT-like cells. Our results demonstrate an increased expression of mesenchymal markers [fibroblast-specific protein 1 (FSP1), collagen1α2, vimentin, α-smooth muscle actin (α-SMA), and Snail], decreased expression of endothelial markers [CD31, von Willebrand factor VIII (vWF), and VE-cadherin], and elevated migration ability in HEMEC following coculture with OE33 cells. The EndoMT-related changes were inhibited by IL-1ß and TGF-ß2 gene silencing in OE33 cells. Recombinant IL-1ß and TGF-ß2 induced EndoMT in HEMEC. Although the level of VEGF expression was elevated in EndoMT cells, the angiogenic property of these cells was diminished. In vivo, by immunostaining EndoMT-like cells were detected at the invasive front of EAC. Our findings underscore a significant role for EndoMT in EAC and provide new insights into the mechanisms and significance of EndoMT in the context of tumor progression.


Asunto(s)
Adenocarcinoma/patología , Células Endoteliales/citología , Neoplasias Esofágicas/patología , Esófago/citología , Interleucina-1beta/fisiología , Mesodermo/citología , Factor de Crecimiento Transformador beta2/fisiología , Microambiente Tumoral , Adenocarcinoma/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Células Endoteliales/metabolismo , Neoplasias Esofágicas/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Interleucina-1beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta2/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo
7.
Am J Physiol Gastrointest Liver Physiol ; 306(7): G557-74, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24481601

RESUMEN

Squamous esophageal epithelium adapts to acid reflux-mediated injury by proliferation and differentiation via signal transduction pathways. Induction of the Wnt antagonist Dickkopf-1 (Dkk1) is involved in tissue repair during inflammation and cellular injury. In this study, we aimed to identify the biological role of Dkk1 in human reflux esophagitis with respect to cell growth and regulation of Wnt signaling. Esophageal biopsies from reflux-esophagitis patients (n = 15) and healthy individuals (n = 10) were characterized in terms of Dkk1 expression. The role of Dkk1 in response to acid-mediated epithelial injury was analyzed by cellular assays in vitro utilizing squamous esophageal epithelial cell lines (EPC1-hTERT, EPC2-hTERT, and HEEC). Dkk1 was significantly overexpressed in human reflux-esophagitis tissue compared with healthy esophageal mucosa at transcriptional and translational levels. After acute and chronic acid (pH 4) exposure, esophageal squamous epithelial cell lines expressed and secreted high levels of Dkk1 in response to stress-associated DNA injury. High extracellular levels of human recombinant Dkk1 inhibited epithelial cell growth and induced cellular senescence in vitro, as demonstrated by reduced cell proliferation, G0/G1 cell cycle arrest, elevated senescence-associated ß-galactosidase activity, and upregulation of p16. Acid pulsing induced Dkk1-mediated senescence, which was directly linked to the ability of Dkk1 to antagonize the canonical Wnt/ß-catenin signaling. In healthy esophageal mucosa, Dkk1 expression was associated with low expression of transcriptionally active ß-catenin, while in reflux-esophagitis tissue, Dkk1 overexpression correlated with increased senescence-associated ß-galactosidase activity and p16 upregulation. The data indicate that, in human reflux esophagitis, Dkk1 functions as a secreted growth inhibitor by suppressing Wnt/ß-catenin signaling and promoting cellular senescence. These findings suggest a significant role for Dkk1 and cellular senescence in esophageal tissue homeostasis during reflux esophagitis.


Asunto(s)
Senescencia Celular , Células Epiteliales/metabolismo , Esofagitis Péptica/metabolismo , Esófago/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Vía de Señalización Wnt , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Puntos de Control del Ciclo Celular , Línea Celular , Proliferación Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Células Epiteliales/patología , Esofagitis Péptica/genética , Esofagitis Péptica/patología , Esófago/patología , Femenino , Humanos , Concentración de Iones de Hidrógeno , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Persona de Mediana Edad , Membrana Mucosa/metabolismo , Membrana Mucosa/patología , Interferencia de ARN , Factores de Tiempo , Transfección , Regulación hacia Arriba , Adulto Joven , beta Catenina/metabolismo , beta-Galactosidasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA