Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Dev Dyn ; 247(3): 542-554, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28631378

RESUMEN

BACKGROUND: Although normally linked to bone and cartilage development, the Runt-related transcription factor, RUNX2, was reported in the mouse heart during development of the valves. We examined RUNX2 expression and function in the developing avian heart as it related to the epithelial-mesenchymal transition (EMT) in the atrioventricular canal. EMT can be separated into an activation stage involving hypertrophy and cell separation and an invasion stage where cells invade the extracellular matrix. The localization and activity of RUNX2 was explored in relation to these steps in the heart. As RUNX2 was also reported in cancer tissues, we examined its expression in the progression of esophageal cancer in staged tissues. RESULTS: A specific isoform, RUNX2-I, is present and required for EMT by endothelia of the atrioventricular canal. Knockdown of RUNX2-I inhibits the cell-cell separation that is characteristic of initial activation of EMT. Loss of RUNX2-I altered expression of EMT markers to a greater extent during activation than during subsequent cell invasion. Transforming growth factor beta 2 (TGFß2) mediates activation during cardiac endothelial EMT. Consistent with a role in activation, RUNX2-I is regulated by TGFß2 and its activity is independent of similarly expressed Snai2 in regulation of EMT. Examination of RUNX2 expression in esophageal cancer showed its upregulation concomitant with the development of dysplasia and continued expression in adenocarcinoma. CONCLUSIONS: These data introduce the RUNX2-I isoform as a critical early transcription factor mediating EMT in the developing heart after induction by TGFß2. Its expression in tumor tissue suggests a similar role for RUNX2 in the EMT of metastasis. Developmental Dynamics 247:542-554, 2018. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Subunidad alfa 1 del Factor de Unión al Sitio Principal/fisiología , Transición Epitelial-Mesenquimal , Animales , Embrión de Pollo , Pollos , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Neoplasias/metabolismo , Isoformas de Proteínas , Activación Transcripcional , Factor de Crecimiento Transformador beta
2.
Am J Physiol Gastrointest Liver Physiol ; 305(8): G585-92, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-23989006

RESUMEN

Enterohepatic helicobacter species (EHS) infect the intestinal tract and biliary tree, triggering intestinal and hepatic disorders. Helicobacter hepaticus, the prototypic murine EHS, is also associated with inflammation. Necrotizing enterocolitis (NEC) is a devastating disease of premature infants. The cause of NEC is not fully understood, but anomalies of bacterial colonization (dysbiosis) are thought to play an important role in disease onset. To evaluate the effect of H. hepaticus infection on the development of NEC, premature formula-fed rats were kept either in H. hepaticus-free conditions or colonized with H. hepaticus; both groups were exposed to asphyxia and cold stress. The incidence of NEC, expression of Toll-like receptors (TLRs), production of cytokines and mucins, and presence of autophagy regulators were evaluated at the site of injury. H. hepaticus infection increased the incidence of NEC from 39 to 71% and significantly increased levels of TLR4 receptor, expression of proinflammatory cytokines CXCL1, IL-1ß, IL-12, and IL-23, and altered activation of autophagy. H. hepaticus induces inflammation and increases the incidence and severity of experimental NEC; this is consistent with observations in neonates of blooms of proinflammatory microbes just before the onset of NEC. Future studies using rodent NEC models should include testing for H. hepaticus infection. Further studies in neonates of early identification and/or diminution of proinflammatory microbes may be beneficial in decreasing the incidence of NEC.


Asunto(s)
Enterocolitis Necrotizante/microbiología , Infecciones por Helicobacter/microbiología , Helicobacter hepaticus/fisiología , Animales , Autofagia , Citocinas/genética , Citocinas/metabolismo , Enterocolitis Necrotizante/patología , Femenino , Regulación de la Expresión Génica/fisiología , Infecciones por Helicobacter/patología , Íleon/metabolismo , Íleon/microbiología , Íleon/patología , Inflamación/metabolismo , Inflamación/patología , Embarazo , ARN Bacteriano/aislamiento & purificación , Ratas , Ratas Sprague-Dawley
3.
Am J Physiol Gastrointest Liver Physiol ; 302(8): G864-72, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22301112

RESUMEN

Beclin-1 has a central role in the regulation of autophagy. Barrett's esophagus (BE) is associated with a significantly increased risk for the development of esophageal adenocarcinoma (EAC). In the current study, we evaluated the role of Beclin-1 and autophagy in the EAC. Biopsies obtained from patients with BE and EAC, tissues from a rat model of BE and EAC, and esophageal cell lines were evaluated for the expression of Beclin-1 by immunohistochemistry, immunoblotting, or RT-PCR. Since reflux of bile acids is important in EAC, we also evaluated the effect of exposure to deoxycholic acid (DCA) on autophagy and Beclin-1 expression. Beclin-1 expression was high in squamous epithelium and nondysplastic BE, whereas its expression was low in dysplastic BE and EAC. The same pattern of expression was observed in rat tissues and in esophageal cell lines. Normal esophageal epithelium and HET-1A cells (derived from normal squamous epithelium) show high levels of Beclin-1, but lower levels of Beclin-1 were found in BE and EAC cell lines (CP-A, CP-C, and OE33). Acute exposure to DCA led to increased Beclin-1 expression and increased autophagy as evaluated by electron microscopy and counting percentage of GFP-LC3-positive BE cells with punctate pattern. In contrast, chronic exposure to DCA did not result in the alteration of Beclin-1 levels or autophagy. In summary, these data suggest that autophagy is initially activated in response to bile acids, but chronic exposure to bile acids leads to decreased Beclin-1 expression and autophagy resistance.


Asunto(s)
Adenocarcinoma/patología , Proteínas Reguladoras de la Apoptosis/biosíntesis , Proteínas Reguladoras de la Apoptosis/fisiología , Ácido Desoxicólico/fisiología , Neoplasias Esofágicas/patología , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/fisiología , Aminoácidos/deficiencia , Animales , Autofagia , Esófago de Barrett/patología , Beclina-1 , Ácidos y Sales Biliares/metabolismo , Western Blotting , Línea Celular Tumoral , Proliferación Celular , Ácido Desoxicólico/farmacología , Progresión de la Enfermedad , Humanos , Inmunohistoquímica , Análisis por Micromatrices , Microscopía Confocal , Microscopía Electrónica de Transmisión , ARN/biosíntesis , ARN/genética , ARN Interferente Pequeño/farmacología , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa
4.
Am J Physiol Gastrointest Liver Physiol ; 300(2): G292-302, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21127259

RESUMEN

Barrett's esophagus (BE) is a premalignant condition, where normal squamous epithelium is replaced by intestinal epithelium. BE is associated with an increased risk of developing esophageal adenocarcinoma (EAC). However, the BE cell of origin is not clear. We hypothesize that BE tissue originates from esophageal squamous cells, which can differentiate to columnar cells as a result of repeated exposure to gastric acid and bile acids, two components of refluxate implicated in BE pathology. To test this hypothesis, we repeatedly exposed squamous esophageal HET1A cells to 0.2 mM bile acid (BA) cocktail at pH 5.5 and developed an HET1AR-resistant cell line. These cells are able to survive and proliferate after repeated 2-h treatments with BA at pH 5.5. HET1AR cells are resistant to acidification and express markers of columnar differentiation, villin, CDX2, and cytokeratin 8/18. HET1AR cells have increased amounts of reactive oxygen species, concomitant with a decreased level and activity of manganese superoxide dismutase compared with parental cells. Furthermore, HET1AR cells express proteins and activate signaling pathways associated with inflammation, cell survival, and tumorigenesis that are thought to contribute to BE and EAC development. These include STAT3, NF-κB, epidermal growth factor receptor (EGFR), cyclooxygenase-2, interleukin-6, phosphorylated mammalian target of rapamycin (p-mTOR), and Mcl-1. The expression of prosurvival and inflammatory proteins and resistance to cell death could be partially modified by inhibition of STAT3 signaling. In summary, our study shows that long-term exposure of squamous cells to BA at acidic pH causes the cells to display the same characteristics and markers as BE.


Asunto(s)
Esófago de Barrett/etiología , Ácidos y Sales Biliares/farmacología , Esófago/efectos de los fármacos , Ácidos/farmacología , Adenocarcinoma/metabolismo , Biomarcadores/metabolismo , Línea Celular Transformada , Regulación hacia Abajo , Resistencia a Medicamentos , Epitelio/metabolismo , Neoplasias Esofágicas/metabolismo , Esófago/citología , Esófago/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Interleucina-6/metabolismo , Metaplasia/metabolismo , FN-kappa B/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Superóxido Dismutasa/metabolismo , Regulación hacia Arriba
5.
Gut ; 59(12): 1606-16, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20876775

RESUMEN

OBJECTIVE: Barrett's oesophagus is a premalignant disease associated with oesophageal adenocarcinoma. The major goal of this study was to determine the mechanism responsible for bile acid-induced alteration in intracellular pH (pH(i)) and its effect on DNA damage in cells derived from normal oesophagus (HET1A) or Barrett's oesophagus (CP-A). DESIGN: Cells were exposed to bile acid cocktail (BA) and/or acid in the presence/absence of inhibitors of nitric oxide synthase (NOS) or sodium-hydrogen exchanger (NHE). Nitric oxide (NO), pH(i) and DNA damage were measured by fluorescent imaging and comet assay. Expression of NHE1 and NOS in cultured cells and biopsies from Barrett's oesophagus or normal squamous epithelium was determined by RT-PCR, immunoblotting or immunohistochemistry. RESULTS: A dose dependent decrease in pH(i) was observed in CP-A cells exposed to BA. This effect of BA is the consequence of NOS activation and increased NO production, which leads to NHE inhibition. Exposure of oesophageal cells to acid in combination with BA synergistically decreased pH(i). The decrease was more pronounced in CP-A cells and resulted in >2-fold increase in DNA damage compared to acid treatment alone. Examination of biopsies and cell lines revealed robust expression of NHE1 in Barrett's oesophagus but an absence of NHE1 in normal epithelium. CONCLUSIONS: The results of this study identify a new mechanism of bile acid-induced DNA damage. We found that bile acids present in the refluxate activate immediately all three isoforms of NOS, which leads to an increased NO production and NHE inhibition. This consequently results in increased intracellular acidification and DNA damage, which may lead to mutations and cancer progression. Therefore, we propose that in addition to gastric reflux, bile reflux should be controlled in patients with Barrett's oesophagus.


Asunto(s)
Esófago de Barrett/genética , Ácidos y Sales Biliares/farmacología , Daño del ADN , Esófago/efectos de los fármacos , Intercambiadores de Sodio-Hidrógeno/genética , Esófago de Barrett/metabolismo , Esófago de Barrett/patología , Proteínas de Transporte de Catión/biosíntesis , Proteínas de Transporte de Catión/genética , Línea Celular , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Activación Enzimática/efectos de los fármacos , Esófago/metabolismo , Humanos , Ácido Clorhídrico/farmacología , Concentración de Iones de Hidrógeno/efectos de los fármacos , Óxido Nítrico/fisiología , Óxido Nítrico Sintasa/metabolismo , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Intercambiador 1 de Sodio-Hidrógeno , Intercambiadores de Sodio-Hidrógeno/biosíntesis , Intercambiadores de Sodio-Hidrógeno/fisiología
6.
Am J Physiol Gastrointest Liver Physiol ; 299(3): G614-22, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20539009

RESUMEN

Necrotizing enterocolitis (NEC) is a devastating intestinal disease of premature infants. Epidermal growth factor (EGF) is one of the most promising candidates in NEC prophylaxis. Autophagy regulates cell homeostasis, but uncontrolled activation of autophagy may lead to cellular injury. The aim was to evaluate the effects of EGF on intestinal autophagy in epithelial cells and in the rat NEC model and measure autophagy in NEC patients. Intestinal epithelial cells (IEC-6) and the rat NEC model were used to study the effect of EGF on intestinal autophagy. Protein levels of Beclin 1 and LC3II were measured in the intestinal epithelium in both in vivo and in vitro models. Ultrastructural changes in intestinal epithelium were studied by electron microscopy. Expression of Beclin 1, LC3II, and p62 protein was evaluated in biopsies from NEC patients. Autophagy was induced in IEC-6 cells and inhibited by adding EGF into the culture. In the rat NEC model, EGF treatment of NEC reduced expression of Beclin 1 and LC3II in ileal epithelium. Morphologically, typical signs of autophagy were observed in the epithelium of the NEC group, but not in the EGF group. A strong signal for Beclin 1 and LC3II was detected in the intestine from patients with NEC. Autophagy is activated in the intestinal epithelium of NEC patients and in the ileum of NEC rats. Supplementation of EGF blocks intestinal autophagy in both in vivo and in vitro conditions. Results from this study indicate that EGF-mediated protection against NEC injury is associated with regulation of intestinal autophagy.


Asunto(s)
Autofagia , Enterocolitis Necrotizante/tratamiento farmacológico , Factor de Crecimiento Epidérmico/farmacología , Factor de Crecimiento Epidérmico/uso terapéutico , Mucosa Intestinal/patología , Administración Oral , Animales , Animales Recién Nacidos , Proteínas Reguladoras de la Apoptosis/metabolismo , Beclina-1 , Línea Celular , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/patología , Factor de Crecimiento Epidérmico/administración & dosificación , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Humanos , Incidencia , Mucosa Intestinal/efectos de los fármacos , Proteínas Asociadas a Microtúbulos/metabolismo , Ratas , Ratas Sprague-Dawley
7.
Am J Physiol Gastrointest Liver Physiol ; 297(5): G940-9, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20501441

RESUMEN

Neonatal necrotizing enterocolitis (NEC) is a major cause of morbidity and mortality in premature infants. Oral administration of probiotics has been suggested as a promising strategy for prevention of NEC. However, little is known about the mechanism(s) of probiotic-mediated protection against NEC. The aim of this study was to evaluate the effects of Bifidobacterium bifidum treatment on development of NEC, cytokine regulation, and intestinal integrity in a rat model of NEC. Premature rats were divided into three groups: dam fed (DF), hand fed with formula (NEC), or hand fed with formula supplemented with 5 x 10(6) CFU B. bifidum per day (B. bifidum). All groups were exposed to asphyxia and cold stress to develop NEC. Intestinal injury, mucin and trefoil factor 3 (Tff3) production, cytokine levels, and composition of tight junction (TJ) and adherens junction (AJ) proteins were evaluated in the terminal ileum. B. bifidum decreased the incidence of NEC from 57 to 17%. Increased levels of IL-6, mucin-3, and Tff3 in the ileum of NEC rats was normalized in B. bifidum treated rats. Reduced mucin-2 production in the NEC rats was not affected by B. bifidum. Administration of B. bifidum normalized the expression and localization of TJ and AJ proteins in the ileum compared with animals with NEC. In conclusion, administration of B. bifidum protects against NEC in the neonatal rat model. This protective effect is associated with reduction of inflammatory reaction in the ileum, regulation of main components of mucus layer, and improvement of intestinal integrity.


Asunto(s)
Bifidobacterium , Enterocolitis Necrotizante/microbiología , Enterocolitis Necrotizante/terapia , Intestinos/microbiología , Intestinos/patología , Probióticos/uso terapéutico , Uniones Adherentes/metabolismo , Animales , Animales Recién Nacidos , Asfixia/complicaciones , Cadherinas/metabolismo , Cateninas/metabolismo , Claudina-3 , Frío , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/metabolismo , Enterocolitis Necrotizante/patología , Enterocitos/metabolismo , Expresión Génica/genética , Íleon/metabolismo , Íleon/patología , Incidencia , Interleucinas/genética , Mucosa Intestinal/metabolismo , Proteínas de la Membrana/metabolismo , Mucina 2/genética , Mucina 2/metabolismo , Mucina 3/genética , Neuropéptidos/genética , Neuropéptidos/metabolismo , Ocludina , Ratas , Ratas Sprague-Dawley , Estrés Fisiológico , Uniones Estrechas/metabolismo , Factor Trefoil-3 , Factor de Necrosis Tumoral alfa/genética
8.
Am J Gastroenterol ; 104(2): 302-9, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19174784

RESUMEN

OBJECTIVES: Barrett's esophagus (BE) is a metaplastic lesion characterized by replacement of the normal squamous epithelium by columnar intestinal epithelium containing goblet cells. It is speculated that this process is an adaptation to protect cells from components of refluxate, such as gastric acid and bile acids. In contrast to the normal squamous epithelium, enterocytes of the distal ileum are adapted to transport bile acids from the intestinal lumen. Several bile acid transporters are utilized for effective removal of bile acids, including the apical sodium-dependent bile acid transporter (ASBT), the ileal bile acid-binding protein (IBABP), and the multidrug-resistant protein 3 (MRP3). We hypothesized that one of the possible functions of newly arising metaplastic epithelium, in the esophagus, is to transport bile acids. Our major goal was to evaluate the expression of bile acid transporters in normal squamous epithelium, BE with different grades of dysplasia, and esophageal adenocarcinoma (EAC). METHODS: A total of 101 patients were included in this study. Immunohistochemistry (IHC) and reverse transcriptase (RT)-PCR were used to detect the expression of these transporters at the mRNA and protein levels. RESULTS: Our immunohistochemical studies showed that all three bile acid transporters are expressed in BE glands, but not in squamous epithelium. ASBT was found in the apical border in BE biopsies. The highest frequency of ASBT expression was in patients with nondysplastic BE (9 of 15, 60%), and a progressive loss of ASBT was observed through the stages of dysplasia. ASBT was not detected in EAC (0 of 15). IBABP staining was observed in the cytoplasm of BE epithelial surface cells. Expression of IBABP was found in 100% of nondysplastic BE (14 of 14), in 93% of low-grade dysplasia (LGD, 15 of 16), in 73% of high-grade dysplasia (HGD, 10 of 14), and in 33% of EAC (5 of 15). MRP3 was expressed in the basolateral membrane in 93% of nondysplastic BE (13 of 14), in 60% of LGD (10 of 16), and in 86% of HGD (11 of 13). Only weak MRP3 staining was detected in EAC biopsies (5 of 15, 33%). In addition, RT-PCR studies showed increased expression of mRNA coding for ASBT (6.1x), IBABP (9.1x), and MRP3 (2.4x) in BE (N=13) compared with normal squamous epithelium (N=15). Significantly increased mRNA levels of IBABP (10.1x) and MRP3 (2.5x) were also detected in EAC (N=21) compared with normal squamous epithelium. CONCLUSIONS: We found that bile acid transporters expression is increased in BE tissue at the mRNA and protein levels and that expression of bile acid transporter proteins decreased with progression to cancer.


Asunto(s)
Adenocarcinoma/metabolismo , Esófago de Barrett/metabolismo , Proteínas Portadoras/metabolismo , Neoplasias Esofágicas/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Transportadores de Anión Orgánico Sodio-Dependiente/metabolismo , Simportadores/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/patología , Adulto , Anciano , Anciano de 80 o más Años , Esófago de Barrett/genética , Esófago de Barrett/patología , Proteínas Portadoras/genética , Estudios de Casos y Controles , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Femenino , Humanos , Masculino , Glicoproteínas de Membrana/genética , Metaplasia , Persona de Mediana Edad , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Transportadores de Anión Orgánico Sodio-Dependiente/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Simportadores/genética
9.
Pediatr Res ; 66(2): 140-4, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19390485

RESUMEN

Necrotizing enterocolitis (NEC) is a devastating disease of premature babies. Previously, we have shown that EGF reduces NEC and that overproduction of hepatic TNF-alpha is associated with intestinal damage. Leakage of TNF-alpha may be a consequence of epithelial hepatic cellular junction dysfunction. The aim of this study was to investigate changes in the composition of hepatic tight junctions (TJs) and adherens junctions (AJs). Using an established rat model of NEC, animals were divided into the following groups: dam fed (DF), formula fed (NEC), or fed with formula supplemented with EGF (EGF). Serum EGF and histologic localization of major TJ and AJ proteins were evaluated. Distribution patterns of hepatic TJ and AJ proteins were significantly altered in the NEC group compared with those in DF or EGF groups. Cytoplasmic accumulation of occludin, claudin-2, and ZO-1 with reduction of claudin-3 signal was detected in the liver of NEC rats. Localization of beta-catenin was associated with the hepatocyte membrane in EGF and DF groups, but diffused in the NEC group. These data show that hepatic cellular junctions are significantly altered during NEC pathogenesis. EGF-mediated reduction of experimental NEC is associated with protection of hepatic integrity and structure.


Asunto(s)
Enterocolitis Necrotizante , Factor de Crecimiento Epidérmico , Hepatocitos , Uniones Intercelulares , Alimentación Animal , Animales , Cadherinas/metabolismo , Claudina-1 , Claudina-3 , Claudinas , Dieta , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/tratamiento farmacológico , Enterocolitis Necrotizante/patología , Factor de Crecimiento Epidérmico/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Factor de Crecimiento Epidérmico/uso terapéutico , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Humanos , Uniones Intercelulares/química , Uniones Intercelulares/efectos de los fármacos , Uniones Intercelulares/ultraestructura , Proteínas de la Membrana/metabolismo , Ocludina , Fosfoproteínas/metabolismo , Ratas , Ratas Sprague-Dawley , Factor de Necrosis Tumoral alfa/metabolismo , Proteína de la Zonula Occludens-1 , Proteína de la Zonula Occludens-2 , alfa Catenina/metabolismo , beta Catenina/metabolismo
10.
Pathol Oncol Res ; 25(1): 349-359, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29127628

RESUMEN

The major aim of this study was to evaluate the performance of anti-BRAF V600E (VE1) antibody in colorectal tumors with and without KRAS mutation. KRAS and BRAF are two major oncogenic drivers of colorectal cancer (CRC) that have been frequently described as mutually exclusive, thus the BRAF V600E mutation is not expected to be present in the cases with KRAS mutation. In addition, a review of 25 studies comparing immunohistochemistry (IHC) using the anti-BRAF V600E (VE1) antibody with BRAF V600E molecular testing in 4041 patient samples was included. One-hundred and twenty cases with/without KRAS or BRAF mutations were acquired. The tissue were immunostained with anti-BRAF V600E (VE1) antibody with OptiView DAB IHC detection kit. The KRAS mutated cases with equivocal immunostaining were further evaluated by Sanger sequencing for BRAF V600E mutation. Thirty cases with BRAF V600E mutation showed unequivocal, diffuse, uniform, positive cytoplasmic staining and 30 cases with wild-type KRAS and BRAF showed negative staining with anti-BRAF V600E (VE1) antibody. Out of 60 cases with KRAS mutation, 56 cases (93.3%) were negative for BRAF V600E mutation by IHC. Four cases showed weak, equivocal, heterogeneous, cytoplasmic staining along with nuclear staining in 25-90% of tumor cells. These cases were confirmed to be negative for BRAF V600E mutation by Sanger sequencing. Overall, IHC with anti-BRAF V600E (VE1) antibody using recommended protocol with OptiView detection is optimal for detection of BRAF V600E mutation in CRC. Our data are consistent with previous reports indicating that KRAS and BRAF V600E mutation are mutually exclusive.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Neoplasias Colorrectales/diagnóstico , Inmunohistoquímica/métodos , Mutación , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/inmunología , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/inmunología , Biomarcadores de Tumor/metabolismo , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/metabolismo , Humanos , Ratones , Pronóstico , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética
11.
Methods Protoc ; 2(4)2019 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-31731599

RESUMEN

Archived Hematoxylin and Eosin (H&E) stained pathology slides are routinely stored to index formalin-fixed paraffin-embedded (FFPE) sample tissue blocks. FFPE blocks are clinically annotated human tumor specimens that can be valuable in studies decades after the tissue is collected. If stored properly, they have the potential to yield a valuable number of serial sectioned slides for diagnostic or research purposes. However, some retrospective studies are limited in scope because the tissue samples have been depleted or not enough material is available in stored blocks for serial sections. The goal of these studies was to determine if archived H&E-stained slides can be directly reutilized by optimizing methods to de-stain and then re-stain the H&E stained slides to allow the detection of several biomarkers of interest using a conjugated antibody with chromogen multiplex immunohistochemistry procedure. This simple but innovative procedure, combined with image analysis techniques, demonstrates the ability to perform precise detection of relevant markers correlated to disease progression in initially identified tumor regions in tissue. This may add clinical value in retaining H&E slides for further use.

12.
Cancer Lett ; 260(1-2): 1-10, 2008 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-18164807

RESUMEN

A field of defective tissue may represent a pre-malignant stage in progression to many cancers. However, field defects are often overlooked in studies of cancer progression through assuming tissue at some distance from the cancer is normal. We indicate, however, the generality of field defects in gastrointestinal cancers, including cancers of the oropharynx, esophagus, stomach, bile duct, pancreas, small intestine and colon/rectum. Common features of these field defects are reduced apoptosis competence, aberrant proliferation and genomic instability. These features are often associated with high bile acid exposure and may explain the association of dietary-related factors with cancer progression.


Asunto(s)
Transformación Celular Neoplásica/patología , Neoplasias Gastrointestinales/patología , Lesiones Precancerosas/patología , Adenocarcinoma/patología , Animales , Apoptosis , Ácidos y Sales Biliares/metabolismo , Neoplasias de los Conductos Biliares/patología , Carcinoma de Células Escamosas/patología , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Colangiocarcinoma/patología , Progresión de la Enfermedad , Neoplasias Esofágicas/patología , Neoplasias Gastrointestinales/genética , Neoplasias Gastrointestinales/metabolismo , Regulación Neoplásica de la Expresión Génica , Inestabilidad Genómica , Neoplasias de Cabeza y Cuello/patología , Humanos , Neoplasias Intestinales/patología , Neoplasias Pancreáticas/patología , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo , Factores de Riesgo , Neoplasias Gástricas/patología
13.
Clin Cancer Res ; 13(18 Pt 1): 5305-13, 2007 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-17875759

RESUMEN

OBJECTIVES: The molecular factors contributing to the development of Barrett's esophagus (BE) are unclear. Our previous studies showed that BE tissues secrete interleukin-6 (IL-6) and express proteins associated with IL-6 signaling, including IL-6 receptor, activated signal transducer and activators of transcription 3 (STAT3), and antiapoptotic proteins Bcl-x(L) and Mcl-1. Here, we test the hypothesis that bile acids and gastric acids, two components of refluxate associated with gastresophageal reflux disease, activate the IL-6/STAT3 pathway. MATERIALS AND METHODS: Immunohistochemistry was used to assess levels of phosphorylated STAT3 in esophageal tissue samples from BE patients with different grades of dysplasia. Seg-1 esophageal adenocarcinoma cells were evaluated for STAT3 activation and IL-6 and Bcl-x(L) expression by molecular biology techniques, including Western blot, reverse transcription-PCR, and ELISA after exposure to control media (pH 7.4), media supplemented with a 0.1 mmol/L bile acid cocktail with media at pH 4 or media at pH 4 with bile acid cocktail. RESULTS: Immunohistochemical analysis showed that activated, phosphorylated STAT3 is expressed in nuclei of dysplastic BE and cancer tissues. Treatment of Seg-1 cells with media containing bile acid cocktail and acidified to pH 4 resulted in increased activation of STAT3, IL-6 secretion, and increased expression of Bcl-x(L). Inhibition of the STAT3 pathway using STAT3 small interfering RNA or Janus-activated kinase inhibitor resulted in increased apoptosis. CONCLUSIONS: The IL-6/STAT3 antiapoptotic pathway is induced by short exposure to bile acid cocktail and low pH. This alteration, if persistent in vivo, may underlie the development of dysplastic BE and tumor progression.


Asunto(s)
Esófago de Barrett/metabolismo , Ácidos y Sales Biliares/metabolismo , Esófago/metabolismo , Interleucina-6/metabolismo , Factor de Transcripción STAT3/metabolismo , Adenocarcinoma/química , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Adulto , Anciano , Anciano de 80 o más Años , Apoptosis , Esófago de Barrett/patología , Ácidos y Sales Biliares/farmacología , Neoplasias Esofágicas/química , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Esófago/efectos de los fármacos , Esófago/patología , Femenino , Ácido Gástrico/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Interleucina-6/genética , Masculino , Persona de Mediana Edad , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Factor de Transcripción STAT3/análisis , Factor de Transcripción STAT3/genética , Células Tumorales Cultivadas , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
14.
Inflamm Bowel Dis ; 12(4): 278-93, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16633050

RESUMEN

BACKGROUND: A high-fat diet is a risk factor for the development of inflammatory bowel disease (IBD) in humans. Deoxycholate (DOC) is increased in the colonic contents in response to a high-fat diet. Thus, an elevated level of DOC in the colonic lumen may play a role in the natural course of development of IBD. METHODS: Wild-type B6.129 mice were fed an AIN-93G diet, either supplemented with 0.2% DOC or unsupplemented and sacrificed at 1 week, 1 month, 3 months, 4 months, and 8 months. Colon samples were assessed by histopathological, immunohistochemical, and cDNA microarray analyses. RESULTS: Mice fed the DOC-supplemented diet developed focal areas of colonic inflammation associated with increases in angiogenesis, nitrosative stress, DNA/RNA damage, and proliferation. Genes that play a central role in inflammation and angiogenesis and other related processes such as epithelial barrier function, oxidative stress, apoptosis, cell proliferation/cell cycle/DNA repair, membrane transport, and the ubiquitin-proteasome pathway showed altered expression in the DOC-fed mice compared with the control mice. Changes in expression of individual genes (increases or reductions) correlated over time. These changes were greatest 1 month after the start of DOC feeding. CONCLUSIONS: The results suggest that exposure of the colonic mucosa to DOC may be a key etiologic factor in IBD. The DOC-fed mouse model may reflect the natural course of development of colitis/IBD in humans, and thus may be useful for determining new preventive strategies and lifestyle changes in affected individuals.


Asunto(s)
Colitis/etiología , Grasas de la Dieta/toxicidad , Animales , Apoptosis/genética , Proliferación Celular , Colitis/patología , ADN/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Estudios de Seguimiento , Expresión Génica , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Ratones , Estrés Oxidativo , Antígeno Nuclear de Célula en Proliferación/genética
15.
Eur J Gastroenterol Hepatol ; 18(10): 1047-51, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16957509

RESUMEN

The mechanisms that regulate ionic balance, fluid absorption and secretion in the gastrointestinal tract are complex. Disturbance of this homeostatic state by bile acids, bacterial enterotoxins and neoplasm-derived secretagogues, for example, can lead to diarrhea. Determining the causes of chronic diarrhea in individual patients may, therefore, represent a diagnostic challenge. The gastrointestinal tract has finely tuned mechanisms to maintain ionic balance, fluid absorption and secretion. To achieve this homeostasis, various ionic transport proteins/complexes are targeted to the apical, basal or basolateral membranes of epithelial cells. Na, K and Cl ion transport proteins are regulated by endogenous activators (e.g. cAMP, PGE2, Ca), dietary-related factors (e.g. bile acids) and through post-translational modifications (e.g. phosphorylation). In certain pathophysiologic states, this homeostatic ionic/fluid exchange becomes dysfunctional as a result of failure of compensatory pro-absorptive/anti-secretory mechanisms. The excessive secretion of Na and Cl ions followed by the release of a large amount of H2O into the colonic lumen results in diarrhea, which may be acute or chronic, and can be life-threatening if not ameliorated. Diverse infectious organisms (e.g. bacteria, protozoa, viruses) utilize different mechanisms to cause intestinal disease and diarrhea. These pathophysiologic mechanisms include alterations in ion transport, disruption of tight junctions and elicitation of inflammatory responses. Studies of patients infected with certain pathogens, those with ulcerative colitis and those harboring extra-colonic or colonic neoplasms have elucidated some of these pathophysiologic mechanisms of diarrhea. Determining the etiology of chronic diarrhea in specific cases may be a diagnostic challenge for both gastroenterologists and primary care physicians.


Asunto(s)
Diarrea/etiología , Enfermedades Intestinales/complicaciones , Enfermedades Intestinales/diagnóstico , Adulto , Enfermedades Transmisibles/complicaciones , Enfermedades Transmisibles/diagnóstico , Humanos , Enfermedades Inflamatorias del Intestino/complicaciones , Enfermedades Inflamatorias del Intestino/diagnóstico , Neoplasias Intestinales/complicaciones , Neoplasias Intestinales/diagnóstico
16.
Cancer Epidemiol Biomarkers Prev ; 14(9): 2066-75, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16172211

RESUMEN

There is an increasing demand for the development of intermediate biomarkers to assess colon cancer risk. We previously determined that a live cell bioassay, which assesses apoptosis resistance in the nonneoplastic colonic mucosa, detects approximately 50% of patients with colon cancer. A hypothesis-driven biomarker that reflects apoptosis resistance in routine formalin-fixed, paraffin-embedded tissue would be easier to use. Cytochrome c oxidase is a critical enzyme that controls mitochondrial respiration and is central to apoptosis. We did an immunohistochemical study of cytochrome c oxidase subunit I expression in 46 colonic mucosal samples from 16 patients who had undergone a colonic resection. These included five patients without evidence of colonic neoplasia (three normal and two diverticulitis), three patients with tubulovillous adenomas, and eight patients with colonic adenocarcinomas. Analysis of aberrancies in expression of cytochrome c oxidase subunit I showed that, compared with nonneoplasia, the patients with neoplasia had a higher mean incidence of crypts having decreased expression (1.7 versus 22.8, P = 0.03) and a higher mean incidence having crypt-restricted loss (0.6 versus 3.2, P = 0.06). The percentage with segmented loss was low and was similar in the two groups. Combining these results, the mean % normal (i.e., with none of the three types of abnormality) was 96.7 in nonneoplasia versus only 73.2 in patients with neoplasia (P = 0.02). It should be noted that a defect in cytochrome c oxidase subunit I immunostaining was not detected in all biopsy samples from each patient for whom some abnormality was found, indicating a "patchiness" in the cytochrome c oxidase subunit I field defect. As a result of this "patchiness," the increased variability in the incidence of crypt-restricted loss of cytochrome c oxidase subunit I expression was a statistically significant feature of the neoplasia group. Crypt-restricted loss of cytochrome c oxidase subunit I has not been previously reported in colonic mucosa and is presumably the result of a crypt-restricted stem cell mutation. Decreased cytochrome c oxidase subunit I expression also significantly correlated with apoptosis resistance, a factor known to contribute to carcinogenesis. The results suggest, however, that aberrant cytochrome c oxidase subunit I expression may be a better biomarker than loss of apoptosis competence for increased colon cancer risk.


Asunto(s)
Adenocarcinoma/genética , Biomarcadores de Tumor/análisis , Neoplasias del Colon/genética , Complejo IV de Transporte de Electrones/genética , Perfilación de la Expresión Génica , Apoptosis/genética , Transformación Celular Neoplásica , Complejo IV de Transporte de Electrones/biosíntesis , Humanos , Inmunohistoquímica , Mucosa Intestinal/patología , Medición de Riesgo
17.
Pathology ; 46(6): 509-17, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25014730

RESUMEN

The most common of all activating BRAF mutations (T1799A) leads to a substitution of valine (V) to glutamic acid (E) at the position 600 of the amino acid sequence. The major goal of this study was to compare detection of the BRAF V600E mutation by DNA sequencing with immunohistochemistry (IHC) using the anti-BRAF V600E (VE1) antibody. Archival formalin fixed, paraffin embedded tissues from 352 patients with colon adenocarcinoma (n = 279) and papillary thyroid carcinoma (n = 73) were evaluated for the BRAF V600E mutation by sequencing and IHC. The discordant cases were re-evaluated by repeat IHC, SNaPshot and next-generation sequencing (NGS). Furthermore, the effect of pre-analytical variables on the utility of this antibody was evaluated in two xenograft mouse models.After resolving 15 initially discordant cases, 212 cases were negative for the BRAF V600E mutation by IHC. Of these, 210 cases (99.1%) were also negative by sequencing and two cases (0.9%) remained discordant. Of the 140 cases that were IHC positive for BRAF V600E, 138 cases were confirmed by sequencing (98.6%) and two cases remained discordant (1.4%). Overall, the negative predictive value was 99.1%, positive predictive value 98.6%, sensitivity 98.6%, specificity 99.1% and overall percentage agreement 98.9% (348/352 cases). Tissue fixation studies indicated that tissues should be fixed for 12-24 h within 2 h of tissue collection with 10% neutral buffered formalin.


Asunto(s)
Adenocarcinoma/patología , Anticuerpos Monoclonales/inmunología , Biomarcadores de Tumor/metabolismo , Carcinoma/patología , Neoplasias Colorrectales/patología , Proteínas Proto-Oncogénicas B-raf/genética , Neoplasias de la Tiroides/patología , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Animales , Secuencia de Bases , Biomarcadores de Tumor/genética , Carcinoma/genética , Carcinoma/metabolismo , Carcinoma Papilar , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Modelos Animales de Enfermedad , Femenino , Xenoinjertos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Inmunohistoquímica , Masculino , Ratones , Mutación , Proteínas Proto-Oncogénicas B-raf/inmunología , Proteínas Proto-Oncogénicas B-raf/metabolismo , Sensibilidad y Especificidad , Análisis de Secuencia de ADN , Cáncer Papilar Tiroideo , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/metabolismo
18.
World J Gastroenterol ; 19(15): 2307-12, 2013 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-23613623

RESUMEN

Barrett's esophagus (BE) is a metaplastic lesion of the distal esophagus arising as a consequence of chronic gastroesophageal reflux disease. Multiple studies show that BE is associated with increased risk of esophageal adenocarcinoma (EAC). Epidemiological studies and animal models demonstrate that chronic inflammation triggered by repeated exposure to refluxate predisposes to the development of BE and EAC. The chronic inflammation is associated with cytokine alterations. Interleukin 6 (IL-6) is a cytokine that stimulates cell proliferation and apoptosis resistance is frequently increased in different cancers. Importantly, IL-6 and transcriptional factor signal transducer and activator of transcription 3 (STAT3) that is activated by IL-6 are also increased in BE and EAC. This review critically appraises the role of IL-6/STAT3 pathway in progression of BE to EAC from the published evidence currently available.


Asunto(s)
Esófago de Barrett/metabolismo , Regulación de la Expresión Génica , Interleucina-6/fisiología , Animales , Apoptosis , Ácidos y Sales Biliares/metabolismo , Neoplasias del Colon/metabolismo , Neoplasias Esofágicas/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Inflamación , Masculino , Ratones , Factor de Transcripción STAT3/metabolismo , Factores Sexuales
19.
Ann N Y Acad Sci ; 1300: 187-199, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24117642

RESUMEN

This paper presents commentaries on animal models used for Barrett's esophagus (BE) and esophageal adenocarcinoma (EAC) research; acid- and bile-induced chromosomal instability and clonal selection during the progression of BE to EAC; how the components of gastric refluxate, especially acid and bile salts, promote carcinogenesis in metaplastic BE; genome-wide changes in DNA methylation and transcription involved in BE carcinogenesis; the potential role of miRNA in the development of BE and EAC; the effect of inflammatory cytokines linked to obesity on the activation of cell-death pathways and cell survival in BE and esophageal cancer; and the role of autophagy in esophageal cancer development.


Asunto(s)
Adenocarcinoma/patología , Esófago de Barrett/patología , Transformación Celular Neoplásica/patología , Neoplasias Esofágicas/patología , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Animales , Esófago de Barrett/genética , Esófago de Barrett/metabolismo , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Metilación de ADN , Modelos Animales de Enfermedad , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Humanos
20.
Ann N Y Acad Sci ; 1300: 296-314, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24117650

RESUMEN

The following paper on the molecular biology of Barrett's esophagus (BE) includes commentaries on signaling pathways central to the development of BE including Hh, NF-κB, and IL-6/STAT3; surgical approaches for esophagectomy and classification of lesions by appropriate therapy; the debate over the merits of minimally invasive esophagectomy versus open surgery; outcomes for patients with pharyngolaryngoesophagectomy; the applications of neoadjuvant chemotherapy and chemoradiotherapy; animal models examining the surgical models of BE and esophageal adenocarcinoma; the roles of various morphogens and Cdx2 in BE; and the use of in vitro BE models for chemoprevention studies.


Asunto(s)
Adenocarcinoma/patología , Esófago de Barrett/patología , Transformación Celular Neoplásica/patología , Neoplasias Esofágicas/patología , Esófago/patología , Adenocarcinoma/genética , Adenocarcinoma/cirugía , Esófago de Barrett/genética , Esófago de Barrett/cirugía , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/cirugía , Esofagectomía , Esófago/metabolismo , Esófago/cirugía , Humanos , Interleucina-6/genética , FN-kappa B/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA