Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 271
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Endoscopy ; 44(2): 148-53, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22271025

RESUMEN

BACKGROUND AND STUDY AIMS: Confocal laser endomicroscopy (CLE) with intravenous infusion of fluorescein allows noninvasive, real-time in vivo visualization of gastrointestinal mucosa at ~ × 1000 magnification ("virtual biopsy"). Conventional biopsies obtained during these procedures serve as the reference and established diagnostic standard. The aim of the present study was to assess whether the standard histologic biopsies that are obtained during CLE retain fluorescein in the tissues and allow the visualization of mucosal structures without any additional staining. PATIENTS AND METHODS: CLE optical imaging of the mucosa was performed in 16 patients who were undergoing CLE colonoscopy. Standard conventional biopsies were also obtained from both normal colonic mucosa and colonic polyps. De-paraffinized mucosal sections were examined under a fluorescence microscope for the presence and distribution of fluorescein, and then underwent immunostaining for expression of vascular endothelial growth factor (VEGF). RESULTS: Standard mucosal biopsy sections from patients undergoing CLE displayed a strong fluorescence and showed well-delineated mucosal structures. In colonic adenomas, there was a 4.6-fold increased vascular permeability compared with normal mucosa (P<0.001), indicated by fluorescein leakage to the extravascular space. Immunostaining demonstrated an aberrantly increased expression of VEGF in the epithelium of colonic adenomas but not in the epithelium of normal mucosa or hyperplastic polyps. CONCLUSIONS: This study shows for the first time that standard colonic biopsies obtained during CLE retain fluorescein, show excellent delineation of mucosal structures without additional staining, allow the evaluation of mucosal microvasculature and vascular permeability, and are suitable for immunostaining.


Asunto(s)
Colon/patología , Pólipos del Colon/patología , Colonoscopía , Fluoresceína , Colorantes Fluorescentes , Mucosa Intestinal/patología , Adenoma/metabolismo , Adenoma/patología , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/metabolismo , Biopsia/métodos , Colon/metabolismo , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Pólipos del Colon/metabolismo , Estudios de Factibilidad , Femenino , Humanos , Mucosa Intestinal/metabolismo , Masculino , Microscopía Confocal , Microscopía Fluorescente , Persona de Mediana Edad , Estudios Prospectivos , Factor A de Crecimiento Endotelial Vascular/metabolismo
2.
Nat Med ; 5(12): 1418-23, 1999 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-10581086

RESUMEN

Angiogenesis, the formation of new capillary blood vessels, is essential not only for the growth and metastasis of solid tumors, but also for wound and ulcer healing, because without the restoration of blood flow, oxygen and nutrients cannot be delivered to the healing site. Nonsteroidal anti-inflammatory drugs (NSAIDs) such as aspirin, indomethacin and ibuprofen are the most widely used drugs for pain, arthritis, cardiovascular diseases and, more recently, the prevention of colon cancer and Alzheimer disease. However, NSAIDs produce gastroduodenal ulcers in about 25% of users (often with bleeding and/or perforations) and delay ulcer healing, presumably by blocking prostaglandin synthesis from cyclooxygenase (COX)-1 and COX-2 (ref. 10). The hypothesis that the gastrointestinal side effects of NSAIDs result from inhibition of COX-1, but not COX-2 (ref. 11), prompted the development of NSAIDs that selectively inhibit only COX-2 (such as celecoxib and rofecoxib). Our study demonstrates that both selective and nonselective NSAIDs inhibit angiogenesis through direct effects on endothelial cells. We also show that this action involves inhibition of mitogen-activated protein (MAP) kinase (ERK2) activity, interference with ERK nuclear translocation, is independent of protein kinase C and has prostaglandin-dependent and prostaglandin-independent components. Finally, we show that both COX-1 and COX-2 are important for the regulation of angiogenesis. These findings challenge the premise that selective COX-2 inhibitors will not affect the gastrointestinal tract and ulcer/wound healing.


Asunto(s)
Antiinflamatorios no Esteroideos/efectos adversos , Antiinflamatorios no Esteroideos/farmacología , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/prevención & control , Úlcera Péptica/inducido químicamente , Animales , Células Cultivadas , Ciclooxigenasa 1 , Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa/efectos adversos , Inhibidores de la Ciclooxigenasa/farmacología , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Flavonoides/farmacología , Humanos , Indometacina/farmacología , Isoenzimas/fisiología , Proteínas de la Membrana , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Neovascularización Patológica/enzimología , Nitrobencenos/farmacología , Úlcera Péptica/patología , Prostaglandina-Endoperóxido Sintasas/fisiología , Ratas , Sulfonamidas/farmacología , Cicatrización de Heridas/efectos de los fármacos
4.
J Clin Invest ; 81(4): 1081-9, 1988 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-3350966

RESUMEN

Isolated human gastric glands from surgical specimens were preincubated in an oxygenated medium with placebo or 16,16 dimethyl prostaglandin E2 (dmPGE2) and incubated at 37 degrees C in either medium alone, medium containing 4.43 mM indomethacin or medium containing 8% ethanol. We assessed the viability of gland cells with fast green exclusion, release of lactate dehydrogenase (LDH) into the medium, and ultrastructural damage by scanning and transmission electron microscopy. Both indomethacin and ethanol significantly reduced the viability of placebo-pretreated glands, increased LDH release into the medium, and produced prominent ultrastructural damage. DmPGE2 significantly reduced both indomethacin and ethanol-induced injury, increased the number of viable cells, reduced LDH release, and diminished the extent of ultrastructural damage. These studies indicate that PG protection of gastric mucosal cells has a direct cellular action that is not limited to replacement of depleted endogenous PGs. PG protection in our experiments did not depend on PG's previously described systemic actions, such as protection of the microvessels, preservation of the mucosal blood flow, or stimulation of bicarbonate and mucus secretion.


Asunto(s)
16,16-Dimetilprostaglandina E2/farmacología , Etanol/antagonistas & inhibidores , Mucosa Gástrica/efectos de los fármacos , Indometacina/antagonistas & inhibidores , Prostaglandinas E Sintéticas/farmacología , Supervivencia Celular/efectos de los fármacos , Mucosa Gástrica/ultraestructura , L-Lactato Deshidrogenasa/metabolismo , Microscopía Electrónica , Microscopía Electrónica de Rastreo
5.
Aliment Pharmacol Ther ; 25(12): 1443-9, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17539984

RESUMEN

BACKGROUND: Dysplasia and malignant transformation of colonocytes in ulcerative colitis are associated with overexpression of c-Myc and genes regulating cell survival. 5-Aminosalicylates such as mesalazine may reduce the development of colorectal cancer in ulcerative colitis, but the mechanisms of its chemopreventive action are not clear. AIMS: To examine whether mesalazine affects the expression of c-Myc in human colon cancer cell lines. METHODS: Human colon cancer cells were treated with vehicle or mesalazine (4 mm or 40 mm). We examined: (i) mRNA expression by gene array, (ii) protein expression by Western blotting and immunohistochemistry and (iii) apoptosis by Annexin V labelling. RESULTS: Mesalazine significantly reduced expression of c-Myc mRNA and protein. CONCLUSIONS: Mesalazine downregulates gene and protein expression of c-Myc. The apoptotic and growth inhibitory effects of mesalazine are dose-dependent. Expression of c-Myc is significantly reduced by mesalazine 40 mm.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Neoplasias del Colon/prevención & control , Regulación hacia Abajo , Mesalamina/farmacología , Anexina A5/farmacología , Antiinflamatorios no Esteroideos/uso terapéutico , Apoptosis/efectos de los fármacos , Western Blotting/métodos , Colitis Ulcerosa/tratamiento farmacológico , Neoplasias del Colon/metabolismo , Inhibidores Enzimáticos/farmacología , Expresión Génica/efectos de los fármacos , Humanos , Inmunohistoquímica/métodos , Mesalamina/uso terapéutico , Proteínas Proto-Oncogénicas c-myc/metabolismo , Resultado del Tratamiento
6.
J Physiol Pharmacol ; 68(6): 841-846, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29550796

RESUMEN

Regeneration of blood vessels (neovascularization) is critical for tissue injury healing. The contribution of bone marrow-derived endothelial progenitor cells (BMD-EPCs) to neovascularization during tissue injury healing is not fully elucidated and it is not clear whether BMD-EPCs can form new capillary blood vessels independently or jointly with fully differentiated endothelial cells (ECs). The aim of this study was to establish an in vitro model of vasculogenesis/angiogenesis by co-culture of BMD-EPCs and gastric endothelial cells (GECs) on Matrigel, examine direct interactions of these cells; and, identify the mechanisms involved. We isolated BMD-EPCs and GECs from bone marrow and stomach of rats, respectively. In these cells, we examined the expression of CD34, CD133, CD31, VEGF-R2, stromal derived factor 1 (SDF-1) and CXCR4, and, their ability to form capillary-like tubes when cultured separately or when co-cultured (1:5 ratio) on growth factor-reduced Matrigel. Fluorescence-labeled BMD-EPCs seeded alone on Matrigel formed capillary-like tubes reflecting in vitro vasculogenesis, and when co-cultured with GECs on Matrigel, formed 'hybrid' tubes containing BMD-EPCs nested between GECs thus reflecting in vitro angio-vasculogenesis. These 'hybrid' tubes were 1.5-fold wider (P < 0.001) and had more extensive (5.1-fold increase) loops (P < 0.01) at the junctions of BMD-EPCs and GECs versus tubes formed by GECs alone. GECs expressed SDF-1 that likely mediated homing of BMD-EPCs (which expressed the SDF-1 receptor, CXCR4) and their incorporation during neovascularization. BMD-EPCs can independently form capillary-like tubes on Matrigel, and when co-cultured with fully differentiated ECs on Matrigel, form capillary-like 'hybrid' tubes comprised of both cell types. Both BMD-EPCs and GECs express SDF-1 and CXCR4, which indicate direct paracrine interactions between these cells during neovascularization.


Asunto(s)
Quimiocina CXCL12/fisiología , Células Progenitoras Endoteliales/fisiología , Neovascularización Fisiológica , Receptores CXCR4/fisiología , Animales , Células de la Médula Ósea/citología , Capilares/fisiología , Células Cultivadas , Técnicas de Cocultivo , Ratas Endogámicas F344 , Estómago/citología
7.
J Physiol Pharmacol ; 68(4): 585-589, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29151075

RESUMEN

Regeneration of blood vessels (neovascularization) is critical for gastric ulcer (GU) healing. The contributions of bone marrow-derived endothelial progenitor cells (BMD-EPCs) to neovascularization during GU healing are not fully elucidated. Our specific aims were to determine whether in GU, BMD-EPCs are incorporated into blood vessels of GU granulation tissue jointly with ECs, thus forming hybrid vessels; or, form separate vessels consisting of only BMD-EPCs. GUs were induced in rats by serosal application of acetic acid. Vascular cast studies were performed at 7, 21 and 60 days after GU induction and tissue specimens were immunostained for CD34, CD133, VEGFR2, and SDF-1 at 14 days. Human relevance was determined using archival human GU specimens. In rat GU granulation tissue BMD-EPCs constituted 28 ± 3% of all cells lining newly formed blood vessels, and were nested between fully differentiated ECs. In rat GU granulation tissue, expression of stromal derived factor-1 (SDF-1) - the major chemoattractant for BMD-EPCs was strongly upregulated. In human GU specimens, BMD-EPCs were also present in granulation tissue constituting 34 ± 3% of all cells lining blood vessels and jointly formed hybrid vessels with differentiated ECs. Our study uncovered that BMD-EPCs incorporate into newly formed blood vessels in GU granulation tissue; and, together with ECs of pre-existing vessels, contribute to and support neovascularization through vasculogenesis. This study is the first demonstration that vasculogenesis occurs during GU healing in both humans and in rats.


Asunto(s)
Médula Ósea/fisiología , Células Progenitoras Endoteliales/fisiología , Neovascularización Fisiológica/fisiología , Úlcera Gástrica/fisiopatología , Animales , Antígenos CD/metabolismo , Médula Ósea/metabolismo , Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Células Progenitoras Endoteliales/metabolismo , Humanos , Ratas , Úlcera Gástrica/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
8.
J Physiol Pharmacol ; 67(6): 879-883, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28195068

RESUMEN

This study was aimed to determine the expression and localization of nerve growth factor (NGF) in the gastric mucosa. Transmural gastric specimens were obtained from euthanized rats. STUDIES: 1) expression of NGF and TrkA receptor by Western blotting; 2) histological evaluation of gastric wall architecture; 3) expression of NGF using immunostaining. Immunostaining showed strong and differential expression of NGF in neural elements of gastric myenteric and submucosal plexuses; in epithelial cells: mainly in chief and progenitor cells, in enterochromaffin-like (ECL) cells; and, in endothelial cells (ECs) lining blood vessels. We concluded that NGF expression in neural elements, epithelial cells and endothelial cells of blood vessels indicated a complex local interaction between neural, epithelial and endothelial cells that regulated gastric mucosal homeostasis and, likely, the protection against gastric injury and ulcer healing.


Asunto(s)
Células Endoteliales/metabolismo , Células Epiteliales/metabolismo , Mucosa Gástrica/metabolismo , Factor de Crecimiento Nervioso/metabolismo , Animales , Masculino , Ratas , Ratas Endogámicas F344 , Células Madre/metabolismo
9.
J Physiol Pharmacol ; 56(1): 3-13, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15795471

RESUMEN

Gastrointestinal ulcer healing is a complex process, involving cell migration, proliferation, angiogenesis and extracellular matrix deposition, all ultimately leading to reconstruction of tissue architecture within the ulcer scar. These processes are controlled by growth factors, cytokines and hormones. Transforming growth factor-beta (TGF-beta), one of the multifunctional peptide growth factors, has been reported to positively regulate gastrointestinal ulcer healing. Although TGF-beta inhibits cell proliferation in a variety of cells, it induces cell migration, angiogenesis, and enhances extracellular matrix production necessary for gastrointestinal ulcer healing. TGF-beta exerts its action by binding to its transmembrane serine/threonine kinase receptors, which in turn triggers activation of various intracellular signaling pathways. Smads are intermediate effector proteins that play key roles in biological activities of TGF-beta by transmitting the signals from the cell surface directly into the nucleus and initiating transcription. New insight into the mechanisms underlying TGF-beta-Smad modulation of gastrointestinal ulcer healing will likely enhance our understanding of the mechanisms controlling the healing processes of gastrointestinal ulcers.


Asunto(s)
Úlcera Péptica/metabolismo , Úlcera Péptica/fisiopatología , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/fisiología , Cicatrización de Heridas , Animales , Enfermedades Gastrointestinales/metabolismo , Enfermedades Gastrointestinales/fisiopatología , Humanos
10.
J Physiol Pharmacol ; 66(4): 617-21, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26348086

RESUMEN

A previous study has demonstrated that locally administered growth factors such as epidermal growth factor, basic fibroblast growth factor and hepatocyte growth factor can accelerate healing of experimental gastric ulcers in rats. That study indicates that locally administered growth factors can exert potent biological effects resulting in enhanced gastric ulcers healing. However, the fate of injected growth factors, their retention and localization to specific cellular compartments have not been examined. In our preliminary study, we demonstrated that local injection of nerve growth factor to the base of experimental gastric ulcers dramatically accelerates ulcer healing, increases angiogenesis - new blood vessel formation, and improves the quality of vascular and epithelial regeneration. Before embarking on larger, definitive and time sequence studies, we wished to determine whether locally injected nerve growth factor is retained in gastric ulcer's tissues and taken up by specific cells during gastric ulcer healing. Gastric ulcers were induced in anesthetized rats by local application of acetic acid using standard methods; and, 60 min later fluorescein isothiocyanate-labeled nerve growth factor was injected locally to the ulcer base. Rats were euthanized 2, 5 and 10 days later. Gastric specimens were obtained and processed for histology. Unstained paraffin sections were examined under a fluorescence microscope, and the incorporation of fluorescein isothiocyanate-labeled nerve growth factor into various gastric tissue cells was determined and quantified. In addition, we performed immunostaining for S100ß protein that is expressed in neural components. Five and ten days after ulcer induction labeled nerve growth factor (injected to the gastric ulcer base) was incorporated into endothelial cells of blood vessels, neuronal, glial and epithelial cells, myofibroblasts and muscle cells. This study demonstrates for the first time that during gastric ulcer healing locally administered exogenous nerve growth factor is retained in gastric tissue and is taken up by endothelial, neural, muscle and epithelial cells. This is likely the basis for the therapeutic action of locally administered nerve growth factor and its stimulation of angiogenesis, tissue regeneration and gastric ulcer healing.


Asunto(s)
Células Endoteliales/metabolismo , Células Epiteliales/metabolismo , Mucosa Gástrica/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Factor de Crecimiento Nervioso/farmacología , Neuroglía/metabolismo , Neuronas/metabolismo , Úlcera Gástrica/tratamiento farmacológico , Animales , Inyecciones , Masculino , Factor de Crecimiento Nervioso/administración & dosificación , Factor de Crecimiento Nervioso/metabolismo , Ratas , Ratas Sprague-Dawley , Regeneración/efectos de los fármacos , Subunidad beta de la Proteína de Unión al Calcio S100/metabolismo , Cicatrización de Heridas/efectos de los fármacos
11.
J Physiol Pharmacol ; 66(3): 415-20, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26084223

RESUMEN

UNLABELLED: This study was aimed to determine the expression and localization of nerve growth factor (NGF) and several neural peptides in porcine esophagus. Transmural esophageal specimens were obtained from euthanized pigs. STUDIES: 1) histologic evaluation, 2) expressions of NGF and its tropomyosin receptor kinase A (TrkA) receptor, calcitonin generelated peptide (CGRP), neuronal nitric oxide synthase (nNOS), and neuronal enolase using immunostaining and quantification of signal distribution and intensity. Immunostaining for NGF, CGRP, nNOS and neuronal specific enolase (NSE) showed their strong and differential expression and localization in the neuronal network. NGF was strongly expressed in the majority of neurons and nerves, distribution of TrkA was complementary; its signal was 1.5-fold weaker P < 0.001 than NGF). Quantitatively the signal intensity was: CGRP > nNOS > NGF > NES > TrkA. In addition to neural structures, nNOS, NGF and TrkA were expressed in keratinocyte progenitor cells of esophageal mucosa and in endothelial cells of blood vessels. We conclude that a strong expression of NGF in majority of esophageal neurons and nerves indicates important, but previously unrecognized regulatory roles in the esophagus; 2) This study showed expression of NGF and some of the neuropeptides in neural elements, keratinocyte progenitor cells and endothelial cells of blood vessels, which indicates local interactions between neural, epithelial and endothelial cells.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina/metabolismo , Esófago/metabolismo , Factor de Crecimiento Nervioso/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Receptor trkA/metabolismo , Animales , Células Endoteliales/metabolismo , Epitelio/metabolismo , Esófago/citología , Neuronas/metabolismo , Células Madre/metabolismo , Porcinos
12.
Front Biosci ; 4: D303-9, 1999 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-10077540

RESUMEN

Growth factors and their receptors play important roles in cell proliferation, migration, tissue injury repair and ulcer healing. In gastric mucosa, transforming growth factor alpha (TGF-alpha) and epidermal growth factor (EGF) by activating their common receptor, control cell proliferation. TGF-alpha predominantly plays this role under normal conditions and after acute injury, while EGF exerts its actions mainly during healing of chronic ulcers. During regeneration of injured gastric mucosa, these growth factors serve predominantly to restore the epithelial component. Other growth factors, basic fibroblast growth factor (bFGF) and vascular endothelial growth factor (VEGF) serve to promote restoration of the connective tissue and microvessels (angiogenesis) in injured mucosa. During healing of chronic ulcers, a new epithelial lineage secreting EGF and other growth peptides develops and the majority of cells lining the ulcer margin overexpress the EGF receptor. Activation of the EGF receptor induces dramatic increases in MAP (Erk -1 and -2) kinase activity and phosphorylation levels. Inhibition of this signaling pathway dramatically delays ulcer healing. Granulation connective tissue, which grows under the stimulation of bFGF and VEGF is the major source for regeneration of connective tissue lamina propria and microvessels within the ulcer scar. Other growth factors such as insulin - like growth factor, keratinocyte growth factor, hepatocyte growth factor and trefoil peptides have been implicated in gastrointestinal (gastric ulcers, colitis) regeneration following injury. This paper is intended to provide an overview of the role of growth factors in gastrointestinal mucosal regeneration.


Asunto(s)
Mucosa Gástrica/fisiología , Sustancias de Crecimiento/fisiología , Mucosa Intestinal/fisiología , Regeneración , Animales , División Celular , Colitis/fisiopatología , Factor de Crecimiento Epidérmico/fisiología , Mucosa Gástrica/irrigación sanguínea , Mucosa Gástrica/lesiones , Mucosa Gástrica/microbiología , Infecciones por Helicobacter/fisiopatología , Helicobacter pylori , Humanos , Mucosa Intestinal/irrigación sanguínea , Mucosa Intestinal/lesiones , Mucosa Intestinal/microbiología , Neovascularización Fisiológica , Transducción de Señal , Úlcera Gástrica/microbiología , Úlcera Gástrica/fisiopatología , Factor de Crecimiento Transformador alfa/fisiología , Cicatrización de Heridas
13.
Am J Med ; 79(2C): 19-23, 1985 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-2863980

RESUMEN

The abilities of antacid (Mylanta II), sucralfate, cimetidine, and ranitidine to protect the gastric mucosa against ethanol-induced necrosis were compared in a standardized, experimental rat model. Fasted rats received pretreatment with either saline, Mylanta II, 500 mg/kg of sucralfate, 50 mg/kg of cimetidine, or 50 mg/kg of ranitidine. This was followed one hour later by intragastric administration of 2 ml of 100 percent ethanol. Gastric mucosal injury was assessed four hours after administration of ethanol by quantitation of gross mucosal necrosis, assessment of mucosal histology, and determination of intragastric blood and protein concentrations. Pretreatment with Mylanta II or sucralfate significantly reduced ethanol-induced gastric mucosal necrosis. The protective effect of sucralfate was six to 10 times greater than that of Mylanta II. H2-receptor antagonists increased ethanol-induced gastric mucosal necrosis.


Asunto(s)
Aluminio/farmacología , Antiácidos/farmacología , Antiulcerosos , Mucosa Gástrica/efectos de los fármacos , Antagonistas de los Receptores H2 de la Histamina/farmacología , Hidróxido de Aluminio/farmacología , Animales , Cimetidina/farmacología , Combinación de Medicamentos/farmacología , Etanol/antagonistas & inhibidores , Mucosa Gástrica/patología , Hemorragia Gastrointestinal/inducido químicamente , Hemorragia Gastrointestinal/prevención & control , Hidróxido de Magnesio/farmacología , Masculino , Necrosis/inducido químicamente , Necrosis/prevención & control , Proteínas/metabolismo , Ranitidina/farmacología , Ratas , Ratas Endogámicas , Simeticona/farmacología , Sucralfato
14.
Am J Med ; 83(3B): 31-7, 1987 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-3661609

RESUMEN

In order to study whether sucralfate or cimetidine may protect human gastric mucosa against alcohol injury, 28 healthy volunteers were pretreated with either: (1) placebo 1 g; (2) cimetidine (Tagamet) 300 mg; or (3) sucralfate (Carafate) 1 g. One hour later, 100 ml of 40 percent ethanol was sprayed directly on the gastric mucosa of the greater curvature during an endoscopic examination. Gastric mucosal changes were assessed by endoscopic appearance (according to grading scale) and by histology. In placebo-pretreated subjects, alcohol produced prominent mucosal damage (endoscopic score, 3.9 +/- 0.3, histologic score, 4.0 +/- 1.1 at 30 minutes). Cimetidine alkalinized gastric pH but did not prevent alcohol-induced damage (endoscopic score, 4.0 +/- 0.6; histologic score, 3.8 +/- 1.1, at 30 minutes). Sucralfate reduced endoscopic and histologic features of alcohol injury (endoscopic score, 1.8 +/- 0.6; histologic score, 1.8 +/- 1.1, at 30 minutes) without affecting gastric luminal pH. Reduction of alcohol-induced injury of the human gastric mucosa by sucralfate but not cimetidine demonstrates that effective protection of the gastric mucosa can be achieved without neutralization or inhibition of gastric acid secretion and points out another clinical application for sucralfate.


Asunto(s)
Cimetidina/administración & dosificación , Etanol/toxicidad , Mucosa Gástrica/efectos de los fármacos , Sucralfato/administración & dosificación , Adolescente , Adulto , Cimetidina/uso terapéutico , Evaluación de Medicamentos , Ácido Gástrico/metabolismo , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patología , Gastritis/prevención & control , Gastroscopía , Humanos , Masculino , Placebos , Sucralfato/uso terapéutico
15.
Aliment Pharmacol Ther ; 5 Suppl 1: 79-90, 1991.
Artículo en Inglés | MEDLINE | ID: mdl-1888836

RESUMEN

It has long been assumed that the mucosa in areas of grossly 'healed' gastric or duodenal ulcers returns to normal, either spontaneously or after treatment. This assumption is based almost entirely upon visual, superficial examination by endoscopy. Few, if any, histological and ultrastructural studies examined the deeper mucosa in the areas of grossly healed ulcers. In several experimental studies, we analysed the development, evolution, and healing of acetic acid-induced gastric ulcers in rats and assessed the histological and ultrastructural features (structure and cellular composition) of the gastric mucosa in areas of grossly healed ulcers. The gastric mucosa of grossly 'healed' ulcers showed re-epithelialization of the mucosal surface at every study interval (2 weeks, 2, 3, and 4 months), but the subepithelial mucosa displayed prominent abnormalities. Two patterns of scarring were distinguished: (a) the mucosa in the area of healed ulcer was thinner (25-45% thinner than normal mucosa) with increased connective tissue and poor differentiation and/or degenerative changes in the glandular cells; and (b) the mucosa displayed a marked dilation of gastric glands with poor differentiation of the glandular cells and a reduction in the supportive microvascular network. It is theorized that these abnormalities could interfere with oxygenation, nutrient supply, and mucosal resistance and defence; therefore, they could be a basis for ulcer recurrence. These observations indicate that the quality of mucosal structural restoration rather than the speed of ulcer healing is the most important factor in determining risk of ulcer recurrence. The clinical relevance of these findings is supported by a preliminary study in which marked histological abnormalities were found in the subepithelial mucosa in patients with 'healed' duodenal ulcers.


Asunto(s)
Úlcera Gástrica/patología , Animales , Mucosa Gástrica/ultraestructura , Ratas
16.
Aliment Pharmacol Ther ; 15(10): 1673-9, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11564009

RESUMEN

BACKGROUND: Basic fibroblast growth factor has been shown to be mitogenic in colon cancer cell lines. In human malignant melanoma cells, antisense oligodeoxynucleotides targeted against basic fibroblast growth factor messenger RNA significantly inhibit cell growth. However, the efficacy of such an antisense oligodeoxynucleotide strategy has not been evaluated for colon cancer cells. AIM: To investigate whether basic fibroblast growth factor can stimulate the growth of HT-29 human colon cancer cells and whether antisense oligodeoxynucleotides can inhibit growth of these cells at baseline. METHODS: Western blotting analyses were used to confirm the presence of basic fibroblast growth factor protein in this cell line. Cell growth was assessed after 2, 4 and 6 days of treatment by cell counting using the trypan blue exclusion method. Phosphorothioate-modified oligodeoxynucleotides (10 microM) were used, complementary to codon 60 of the basic fibroblast growth factor messenger RNA. Cationic liposomes (DOTAP) were used to enhance the cellular uptake of the oligodeoxynucleotides. RESULTS: Western blotting demonstrated the presence of basic fibroblast growth factor protein in this cell line. Basic fibroblast growth factor (1-40 ng/mL) dose-dependently stimulated cell growth and peak values were obtained at a dose of 20 ng/mL. By contrast, antisense oligodeoxynucleotide treatment significantly inhibited cell growth compared with the sense oligodeoxynucleotide-treated cells (P=0.007). This inhibition was reversed by the addition of basic fibroblast growth factor, 20 ng/mL. CONCLUSION: Treatment targeted against basic fibroblast growth factor messenger RNA inhibits growth of HT-29 human colon cancer cells. This finding may provide a rationale for the therapeutic use of antisense oligodeoxynucleotides targeted at basic fibroblast growth factor for the treatment of colon cancer.


Asunto(s)
Neoplasias del Colon/tratamiento farmacológico , Factores de Crecimiento de Fibroblastos/genética , Oligonucleótidos Antisentido/uso terapéutico , Animales , Cationes , División Celular/efectos de los fármacos , Supervivencia Celular , Células HT29 , Humanos , Liposomas/metabolismo , Oligonucleótidos Antisentido/genética , Proyectos Piloto , ARN Mensajero/genética
17.
Aliment Pharmacol Ther ; 11(2): 373-80, 1997 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9146778

RESUMEN

INTRODUCTION: Relapse of erosive oesophagitis occurs in almost all patients if treatment is stopped after initial healing. AIM: To assess the potential of different therapeutic regimens of omeprazole to prevent relapse of erosive reflux oesophagitis after initial healing with omeprazole. PATIENTS AND METHODS: Patients whose active erosive reflux oesophagitis (grade > or = 2) had healed (grade 0 or 1) after 4-8 weeks of open-label omeprazole 40 mg daily (phase I) were eligible to join a multi-centre, 6-month double-blind, placebo-controlled maintenance study (phase II), which included endoscopy, symptom assessments, serum gastrin measurements, and gastric fundic biopsies. During phase I, endoscopy was performed at weeks 0, 4, and 8. At the end of phase I, 429 of 472 patients (91%) were healed, and there were significant reductions in heartburn, dysphagia and acid regurgitation. Of the 429 patients who healed, 406 joined phase II and were randomized to one of three groups: 20 mg omeprazole daily (n = 138), 20 mg omeprazole for 3 consecutive days each week (n = 137), or placebo (n = 131). During phase II, endoscopy was performed at months 1, 3, and 6 or at symptomatic relapse. RESULTS: The percentages of patients still in endoscopic remission at 6 months were 11% for placebo, 34% for omeprazole 3-days-a-week, and 70% for omeprazole daily. Both omeprazole regimens were superior to placebo in preventing recurrence of symptoms (P < 0.001); however, omeprazole 20 mg daily was superior to omeprazole 20 mg 3-days-a-week (P < 0.001). Compared to baseline, omeprazole therapy resulted in no significant differences among treatment groups in the distribution of gastric endocrine cells. CONCLUSIONS: These results show that after healing of erosive oesophagitis with 4-8 weeks of omeprazole, relapse of oesophagitis and recurrence of reflux symptoms can be prevented in 70% of patients with a maintenance regimen of 20 mg daily, but that intermittent dosing comprising 3 consecutive days each week significantly compromises efficacy.


Asunto(s)
Antiulcerosos/uso terapéutico , Esofagitis Péptica/prevención & control , Omeprazol/uso terapéutico , Antiulcerosos/administración & dosificación , Antiulcerosos/efectos adversos , Método Doble Ciego , Esofagitis Péptica/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Omeprazol/administración & dosificación , Omeprazol/efectos adversos , Recurrencia
18.
Microsc Res Tech ; 51(2): 156-68, 2000 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-11054866

RESUMEN

Recent studies suggest that an abnormal increase in intestinal tight junction (TJ) permeability may be an important etiologic factor in number of diseases including Crohn's disease, NSAID-associated enteritis, and various infectious diarrheal syndromes. The intracellular processes involved in regulation of intestinal epithelial TJ permeability, however, remain poorly understood. In this study, we used cultured Caco-2 intestinal epithelial cells to examine the intracellular processes involved in extracellular Ca(++) modulation of intestinal epithelial monolayer TJ barrier. Incubation of the filter-grown Caco-2 intestinal monolayers in Ca(++)-free solution (CFS), consisting of modified Krebs-buffer solution containing 0 mM Ca(++) and 1 mM EGTA, resulted in a rapid drop in Caco-2 epithelial resistance and increase in epithelial permeability to paracellular markers mannitol and inulin, indicating an increase in TJ permeability. The increase in Caco-2 TJ permeability was rapidly reversed by the re-introduction of Ca(++) (1.8 mM) into the incubation medium. The CFS-induced increase in Caco-2 TJ permeability was associated with separation of the cytoplasmic and transmembrane TJ proteins, ZO-1 and occludin, and formation of large intercellular openings between the adjoining cells. The CFS-induced modulation of TJ barrier was associated with activation of myosin light chain kinase (MLCK) activity and centripetal retraction of peri-junctional actin and myosin filaments. The inhibition of CFS-induced activation of Caco-2 MLCK with MLCK inhibitor (ML-7) prevented the CFS-induced retraction of actin and myosin filaments and the subsequent alteration of TJ barrier function and structure. Our results suggested that the CFS-induced alteration of TJ proteins and functional increase in TJ permeability was mediated by Caco-2 MLCK activation and the resultant contraction of the peri-junctionally located actin-myosin filaments. Consistent with the role of MLCK in this process, selected inhibitors of Mg(++)-myosin ATPase and metabolic energy, but not protein synthesis inhibitors, also prevented the CFS-induced retraction of actin and myosin filaments and the subsequent increase in TJ permeability. In conclusion, our results indicate that extracellular Ca(++) is crucial for the maintenance of intestinal epithelial TJ barrier function. The removal of extracellular Ca(++) from the incubation medium causes activation of Caco-2 MLCK, which in turn leads to an increase in intestinal monolayer TJ permeability.


Asunto(s)
Calcio/farmacología , Citoesqueleto/fisiología , Mucosa Intestinal/efectos de los fármacos , Uniones Estrechas/efectos de los fármacos , Actinas/antagonistas & inhibidores , Actinas/metabolismo , Azepinas/farmacología , ATPasa de Ca(2+) y Mg(2+)/antagonistas & inhibidores , Células CACO-2 , Permeabilidad de la Membrana Celular/efectos de los fármacos , Resistencia a Medicamentos , Inhibidores Enzimáticos/farmacología , Humanos , Mucosa Intestinal/fisiología , Proteínas de la Membrana/metabolismo , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Quinasa de Cadena Ligera de Miosina/metabolismo , Miosinas/antagonistas & inhibidores , Miosinas/metabolismo , Naftalenos/farmacología , Ocludina , Fosfoproteínas/metabolismo , Uniones Estrechas/fisiología , Proteína de la Zonula Occludens-1
19.
Surgery ; 119(4): 424-30, 1996 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-8644008

RESUMEN

BACKGROUND: Basic fibroblast growth factor (bFGF) enhances cell migration, proliferation, and tissue integrity. This especially pertinent to the smooth muscle cells in which it stimulates cell proliferation and promotes their growth. The aim of this study was to determine whether expression of bFGF and its receptors (FGFR-1 and -2) is altered in portal hypertensive esophageal mucosa, especially in the muscularis mucosal layer, which constitutes a physical barrier to variceal rupture. METHODS: Portal hypertension (PHT) was produced by staged portal vein ligation. In 30 PHT rats and 30 sham-operated controls 2 weeks after operation, specimens of lower esophagus were obtained for (1) quantitative histologic assessment including thickness of epithelium and muscularis mucosae; (2) immunostaining with specific antibodies against bFGF and its receptors 1 and 2 (intensity of bFGF, FGFR-1 and FGFR-2 immunostaining in esophageal structures was measured with a video image system); and (3) expression of bFGF and FGFR-1 and -2 mRNAs was assessed with reverse transcription-polymerase chain reaction. RESULTS: The esophageal muscularis mucosae and epithelium overlaying large submucosal veins in PHT rats significatly thinner than those in controls (muscularis mucosae, 28.3 +/- 1.4 versus 52.2 +/- 8.0 micrometer, respectively, p<0.05); epithelium, 39.0+/- 7.1 versus 49.3 +/- 1.9 micrometer, respectively, p<0.05). The immunostaining intensity of bFGF and FGFR-2 was significantly reduced in PHT rats (42.1 +/- 2.3 and 71.3 +/- 6.5 units, respectively) versus controls (49.5 +/- 5.6 and 78.6 +/- 5.7 units, respectively, p< 0.05). Expressions of bFGF and FGFR-2 mRNAs in PHT esophageal mucosa were significantly reduced versus controls by 30.8% and 30.3%, respectively (p < 0.01, p 0.05). CONCLUSIONS: (1) Esophageal mucosa of PHT rats has thinner muscularis mucosae and reduced bFGF and FGFR-2 mRNAs and proteins. (2) Because bFGF stimulates smooth muscle cell proliferation and their growth, our findings can explain thinning of esophageal muscularis mucosae in PHT rats, thus indicating a possible mechanism for rupture of varices in the esophagus.


Asunto(s)
Várices Esofágicas y Gástricas/complicaciones , Esófago/metabolismo , Factor 2 de Crecimiento de Fibroblastos/genética , Hipertensión Portal/metabolismo , ARN Mensajero/análisis , Receptores de Factores de Crecimiento de Fibroblastos/genética , Animales , Secuencia de Bases , Esófago/patología , Factor 2 de Crecimiento de Fibroblastos/análisis , Técnica del Anticuerpo Fluorescente , Hipertensión Portal/patología , Masculino , Datos de Secuencia Molecular , Membrana Mucosa/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Factores de Crecimiento de Fibroblastos/análisis
20.
Surgery ; 104(1): 79-85, 1988 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-3388181

RESUMEN

We assessed macroscopic, histologic, ultrastructural, and functional features of aspirin-induced gastric mucosal injury in portal hypertensive and sham-operated rats. Portal hypertension was produced by staged portal vein ligation. Four hours after intragastric acidified aspirin administration, intraluminal pH in portal hypertensive rats was 6.6 +/- 0.2 and 4.3 +/- 0.5 in sham-operated controls (p less than 0.01). Gross mucosal damage was significantly greater in portal hypertensive rats compared with controls (18 +/- 2 versus 7 +/- 1% of total mucosal area). Histologic deep necrosis involved 22 +/- 2% of mucosal section lengths in portal hypertensive rats compared with 7 +/- 1% in sham-operated rats (p less than 0.01). In portal hypertensive rats, histologic and ultrastructural evaluation demonstrated capillary endothelial abnormalities, arterialization of submucosal veins, and markedly greater severity of microvascular damage than in sham-operated controls. Neutralized aspirin (pH, 7.0) did not produce any significant damage detectable grossly, histologically, or by transmission electron microscopy in portal hypertensive rats. We conclude that acid-dependent aspirin-induced gastric mucosal damage is significantly increased in portal hypertension.


Asunto(s)
Aspirina , Mucosa Gástrica/ultraestructura , Gastritis/inducido químicamente , Hipertensión Portal/patología , Animales , Ácido Gástrico/metabolismo , Mucosa Gástrica/metabolismo , Gastritis/patología , Concentración de Iones de Hidrógeno , Masculino , Microscopía Electrónica , Microscopía Electrónica de Rastreo , Ratas , Ratas Endogámicas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA