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

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Endoscopy ; 44(2): 148-53, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22271025

RESUMO

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.


Assuntos
Colo/patologia , Pólipos do Colo/patologia , Colonoscopia , Fluoresceína , Corantes Fluorescentes , Mucosa Intestinal/patologia , Adenoma/metabolismo , Adenoma/patologia , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/metabolismo , Biópsia/métodos , Colo/metabolismo , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Pólipos do Colo/metabolismo , Estudos de Viabilidade , Feminino , Humanos , Mucosa Intestinal/metabolismo , Masculino , Microscopia Confocal , Microscopia de Fluorescência , Pessoa de Meia-Idade , Estudos Prospectivos , Fator A de Crescimento do Endotélio Vascular/metabolismo
2.
Nat Med ; 5(12): 1418-23, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10581086

RESUMO

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.


Assuntos
Anti-Inflamatórios não Esteroides/efeitos adversos , Anti-Inflamatórios não Esteroides/farmacologia , Neoplasias/irrigação sanguínea , Neoplasias/tratamento farmacológico , Neovascularização Patológica/prevenção & controle , Úlcera Péptica/induzido quimicamente , Animais , Células Cultivadas , Ciclo-Oxigenase 1 , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase/efeitos adversos , Inibidores de Ciclo-Oxigenase/farmacologia , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Flavonoides/farmacologia , Humanos , Indometacina/farmacologia , Isoenzimas/fisiologia , Proteínas de Membrana , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Neovascularização Patológica/enzimologia , Nitrobenzenos/farmacologia , Úlcera Péptica/patologia , Prostaglandina-Endoperóxido Sintases/fisiologia , Ratos , Sulfonamidas/farmacologia , Cicatrização/efeitos dos fármacos
4.
J Clin Invest ; 81(4): 1081-9, 1988 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-3350966

RESUMO

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.


Assuntos
16,16-Dimetilprostaglandina E2/farmacologia , Etanol/antagonistas & inibidores , Mucosa Gástrica/efeitos dos fármacos , Indometacina/antagonistas & inibidores , Prostaglandinas E Sintéticas/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Mucosa Gástrica/ultraestrutura , L-Lactato Desidrogenase/metabolismo , Microscopia Eletrônica , Microscopia Eletrônica de Varredura
5.
Aliment Pharmacol Ther ; 25(12): 1443-9, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17539984

RESUMO

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.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Neoplasias do Colo/prevenção & controle , Regulação para Baixo , Mesalamina/farmacologia , Anexina A5/farmacologia , Anti-Inflamatórios não Esteroides/uso terapêutico , Apoptose/efeitos dos fármacos , Western Blotting/métodos , Colite Ulcerativa/tratamento farmacológico , Neoplasias do Colo/metabolismo , Inibidores Enzimáticos/farmacologia , Expressão Gênica/efeitos dos fármacos , Humanos , Imuno-Histoquímica/métodos , Mesalamina/uso terapêutico , Proteínas Proto-Oncogênicas c-myc/metabolismo , Resultado do Tratamento
6.
J Physiol Pharmacol ; 68(4): 585-589, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29151075

RESUMO

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.


Assuntos
Medula Óssea/fisiologia , Células Progenitoras Endoteliais/fisiologia , Neovascularização Fisiológica/fisiologia , Úlcera Gástrica/fisiopatologia , Animais , Antígenos CD/metabolismo , Medula Óssea/metabolismo , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Células Progenitoras Endoteliais/metabolismo , Humanos , Ratos , Úlcera Gástrica/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
7.
J Physiol Pharmacol ; 68(6): 841-846, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29550796

RESUMO

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.


Assuntos
Quimiocina CXCL12/fisiologia , Células Progenitoras Endoteliais/fisiologia , Neovascularização Fisiológica , Receptores CXCR4/fisiologia , Animais , Células da Medula Óssea/citologia , Capilares/fisiologia , Células Cultivadas , Técnicas de Cocultura , Ratos Endogâmicos F344 , Estômago/citologia
8.
J Physiol Pharmacol ; 67(6): 879-883, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28195068

RESUMO

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.


Assuntos
Células Endoteliais/metabolismo , Células Epiteliais/metabolismo , Mucosa Gástrica/metabolismo , Fator de Crescimento Neural/metabolismo , Animais , Masculino , Ratos , Ratos Endogâmicos F344 , Células-Tronco/metabolismo
9.
J Physiol Pharmacol ; 56(1): 3-13, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15795471

RESUMO

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.


Assuntos
Úlcera Péptica/metabolismo , Úlcera Péptica/fisiopatologia , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Cicatrização , Animais , Gastroenteropatias/metabolismo , Gastroenteropatias/fisiopatologia , Humanos
10.
J Physiol Pharmacol ; 66(4): 617-21, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26348086

RESUMO

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.


Assuntos
Células Endoteliais/metabolismo , Células Epiteliais/metabolismo , Mucosa Gástrica/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Fator de Crescimento Neural/farmacologia , Neuroglia/metabolismo , Neurônios/metabolismo , Úlcera Gástrica/tratamento farmacológico , Animais , Injeções , Masculino , Fator de Crescimento Neural/administração & dosagem , Fator de Crescimento Neural/metabolismo , Ratos , Ratos Sprague-Dawley , Regeneração/efeitos dos fármacos , Subunidade beta da Proteína Ligante de Cálcio S100/metabolismo , Cicatrização/efeitos dos fármacos
11.
J Physiol Pharmacol ; 66(3): 415-20, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26084223

RESUMO

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.


Assuntos
Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Esôfago/metabolismo , Fator de Crescimento Neural/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Fosfopiruvato Hidratase/metabolismo , Receptor trkA/metabolismo , Animais , Células Endoteliais/metabolismo , Epitélio/metabolismo , Esôfago/citologia , Neurônios/metabolismo , Células-Tronco/metabolismo , Suínos
12.
Front Biosci ; 4: D303-9, 1999 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-10077540

RESUMO

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.


Assuntos
Mucosa Gástrica/fisiologia , Substâncias de Crescimento/fisiologia , Mucosa Intestinal/fisiologia , Regeneração , Animais , Divisão Celular , Colite/fisiopatologia , Fator de Crescimento Epidérmico/fisiologia , Mucosa Gástrica/irrigação sanguínea , Mucosa Gástrica/lesões , Mucosa Gástrica/microbiologia , Infecções por Helicobacter/fisiopatologia , Helicobacter pylori , Humanos , Mucosa Intestinal/irrigação sanguínea , Mucosa Intestinal/lesões , Mucosa Intestinal/microbiologia , Neovascularização Fisiológica , Transdução de Sinais , Úlcera Gástrica/microbiologia , Úlcera Gástrica/fisiopatologia , Fator de Crescimento Transformador alfa/fisiologia , Cicatrização
13.
Am J Med ; 79(2C): 19-23, 1985 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-2863980

RESUMO

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.


Assuntos
Alumínio/farmacologia , Antiácidos/farmacologia , Antiulcerosos , Mucosa Gástrica/efeitos dos fármacos , Antagonistas dos Receptores H2 da Histamina/farmacologia , Hidróxido de Alumínio/farmacologia , Animais , Cimetidina/farmacologia , Combinação de Medicamentos/farmacologia , Etanol/antagonistas & inibidores , Mucosa Gástrica/patologia , Hemorragia Gastrointestinal/induzido quimicamente , Hemorragia Gastrointestinal/prevenção & controle , Hidróxido de Magnésio/farmacologia , Masculino , Necrose/induzido quimicamente , Necrose/prevenção & controle , Proteínas/metabolismo , Ranitidina/farmacologia , Ratos , Ratos Endogâmicos , Simeticone/farmacologia , Sucralfato
14.
Am J Med ; 83(3B): 31-7, 1987 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-3661609

RESUMO

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.


Assuntos
Cimetidina/administração & dosagem , Etanol/toxicidade , Mucosa Gástrica/efeitos dos fármacos , Sucralfato/administração & dosagem , Adolescente , Adulto , Cimetidina/uso terapêutico , Avaliação de Medicamentos , Ácido Gástrico/metabolismo , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patologia , Gastrite/prevenção & controle , Gastroscopia , Humanos , Masculino , Placebos , Sucralfato/uso terapêutico
15.
Aliment Pharmacol Ther ; 5 Suppl 1: 79-90, 1991.
Artigo em Inglês | MEDLINE | ID: mdl-1888836

RESUMO

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.


Assuntos
Úlcera Gástrica/patologia , Animais , Mucosa Gástrica/ultraestrutura , Ratos
16.
Aliment Pharmacol Ther ; 15(10): 1673-9, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11564009

RESUMO

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.


Assuntos
Neoplasias do Colo/tratamento farmacológico , Fatores de Crescimento de Fibroblastos/genética , Oligonucleotídeos Antissenso/uso terapêutico , Animais , Cátions , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular , Células HT29 , Humanos , Lipossomos/metabolismo , Oligonucleotídeos Antissenso/genética , Projetos Piloto , RNA Mensageiro/genética
17.
Aliment Pharmacol Ther ; 11(2): 373-80, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9146778

RESUMO

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.


Assuntos
Antiulcerosos/uso terapêutico , Esofagite Péptica/prevenção & controle , Omeprazol/uso terapêutico , Antiulcerosos/administração & dosagem , Antiulcerosos/efeitos adversos , Método Duplo-Cego , Esofagite Péptica/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Omeprazol/administração & dosagem , Omeprazol/efeitos adversos , Recidiva
18.
Microsc Res Tech ; 51(2): 156-68, 2000 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11054866

RESUMO

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.


Assuntos
Cálcio/farmacologia , Citoesqueleto/fisiologia , Mucosa Intestinal/efeitos dos fármacos , Junções Íntimas/efeitos dos fármacos , Actinas/antagonistas & inibidores , Actinas/metabolismo , Azepinas/farmacologia , ATPase de Ca(2+) e Mg(2+)/antagonistas & inibidores , Células CACO-2 , Permeabilidade da Membrana Celular/efeitos dos fármacos , Resistência a Medicamentos , Inibidores Enzimáticos/farmacologia , Humanos , Mucosa Intestinal/fisiologia , Proteínas de Membrana/metabolismo , Quinase de Cadeia Leve de Miosina/antagonistas & inibidores , Quinase de Cadeia Leve de Miosina/metabolismo , Miosinas/antagonistas & inibidores , Miosinas/metabolismo , Naftalenos/farmacologia , Ocludina , Fosfoproteínas/metabolismo , Junções Íntimas/fisiologia , Proteína da Zônula de Oclusão-1
19.
Surgery ; 104(1): 79-85, 1988 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-3388181

RESUMO

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.


Assuntos
Aspirina , Mucosa Gástrica/ultraestrutura , Gastrite/induzido quimicamente , Hipertensão Portal/patologia , Animais , Ácido Gástrico/metabolismo , Mucosa Gástrica/metabolismo , Gastrite/patologia , Concentração de Íons de Hidrogênio , Masculino , Microscopia Eletrônica , Microscopia Eletrônica de Varredura , Ratos , Ratos Endogâmicos
20.
Surgery ; 112(2): 341-6, 1992 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-1641773

RESUMO

BACKGROUND: Epidermal growth factor (EGF) protects gastric mucosa against a variety of injurious agents, but the mechanism is unclear. Because the abnormal microvasculature of portal hypertensive (PHT) gastric mucosa is a major target of ischemia/reperfusion (I/R) injury, we used this model to assess EGF's protective role at the microvascular level. METHODS: Rats with PHT (staged portal vein ligation) received either EGF, 20 micrograms/kg, or saline solution intravenously, with or without indomethacin pretreatment (20 mg/kg subcutaneously). I/R was produced by withdrawing blood to systemic pressures of 30 mm Hg for 20 minutes and reinfusing it. Stomachs were excised 20 minutes later and evaluated for gross and histologic necrosis, microvascular permeability, mucosal ultrastructure and vimentin, and cyclooxygenase immunofluorescence. RESULTS: In saline-treated rats, gross and histologic damage involved 46% +/- 3% of glandular mucosa and 23% +/- 3% of mucosal sections, respectively. Microvascular permeability was increased 43-fold over that of normal control rats. Vimentin immunofluorescence intensity was reduced to 36% +/- 4% that of normal control rats. EGF pretreatment reduced histologic necrosis to 2% +/- 1% (p less than 0.01). Microvascular permeability and vimentin intensity were almost normalized. Indomethacin partially reversed the mucosal protection induced by EGF. CONCLUSIONS: EGF significantly reduces I/R injury to PHT gastric mucosa. Microvascular endothelia of PHT gastric mucosa are the major target of I/R injury and the site of EGF's protective action. Prostaglandins in part mediate EGF's protective action.


Assuntos
Capilares/fisiopatologia , Fator de Crescimento Epidérmico/farmacologia , Mucosa Gástrica/efeitos dos fármacos , Hipertensão Portal/fisiopatologia , Isquemia/fisiopatologia , Animais , Endotélio Vascular/fisiologia , Imunofluorescência , Mucosa Gástrica/irrigação sanguínea , Mucosa Gástrica/fisiopatologia , Prostaglandinas/fisiologia , Ratos , Ratos Endogâmicos , Vimentina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA