Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Gynecol Obstet Fertil Senol ; 46(4): 395-402, 2018 Apr.
Artículo en Francés | MEDLINE | ID: mdl-29602693

RESUMEN

OBJECTIVES: The aim of this article is to question the feeling of IVF patients towards embryonic cryopreservation, in order to understand their potential reluctance to freeze embryos and their difficulties to consider the fate of their frozen embryos once their parental project completed. METHODS: Twenty-seven semi-directive interviews with homologous IVF patients were conducted. These persons were followed in two fertility centres in Marseille. RESULTS: If all the patients interviewed have accepted embryonic cryopreservation or have accepted on principle, a majority have an ambivalent attitude towards this technique. If some share the "pragmatic" vision of professionals (embryologists, technicians and gynaecologists), they are numerous to worry about a possible deterioration of embryonic quality, or again about a disrupted order of generation. Finally, it appears that patients do not anticipate the possible fate of their frozen embryos if they are uninscribed from their parental project. CONCLUSIONS: Patients are mainly ambivalent towards embryonic cryopreservation. They prioritize different rationality depending on the situations and issues they are dealing with.


Asunto(s)
Criopreservación , Embrión de Mamíferos , Fertilización In Vitro/psicología , Adulto , Toma de Decisiones , Transferencia de Embrión , Femenino , Francia , Humanos , Masculino , Persona de Mediana Edad
2.
Br J Pharmacol ; 173(12): 2016-29, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27060978

RESUMEN

BACKGROUND AND PURPOSE: We evaluated the extent to which individual versus combination treatments that specifically target airway epithelial damage [trefoil factor-2 (TFF2)], airway fibrosis [serelaxin (RLX)] or airway inflammation [dexamethasone (DEX)] reversed the pathogenesis of chronic allergic airways disease (AAD). EXPERIMENTAL APPROACH: Following induction of ovalbumin (OVA)-induced chronic AAD in 6­8 week female Balb/c mice, animals were i.p. administered naphthalene (NA) on day 64 to induce epithelial damage, then received daily intranasal administration of RLX (0.8 mg·mL(−1)), TFF2 (0.5 mg·mL(−1)), DEX (0.5 mg·mL(−1)), RLX + TFF2 or RLX + TFF2 + DEX from days 67­74. On day 75, lung function was assessed by invasive plethysmography, before lung tissue was isolated for analyses of various measures. The control group was treated with saline + corn oil (vehicle for NA). KEY RESULTS: OVA + NA-injured mice demonstrated significantly increased airway inflammation, airway remodelling (AWR) (epithelial damage/thickness; subepithelial myofibroblast differentiation, extracellular matrix accumulation and fibronectin deposition; total lung collagen concentration), and significantly reduced airway dynamic compliance (cDyn). RLX + TFF2 markedly reversed several measures of OVA + NA-induced AWR and normalized the reduction in cDyn. The combined effects of RLX + TFF2 + DEX significantly reversed peribronchial inflammation score, airway epithelial damage, subepithelial extracellular matrix accumulation/fibronectin deposition and total lung collagen concentration (by 50­90%) and also normalized the reduction of cDyn. CONCLUSIONS AND IMPLICATIONS: Combining an epithelial repair factor and anti-fibrotic provides an effective means of treating the AWR and dysfunction associated with AAD/asthma and may act as an effective adjunct therapy to anti-inflammatory corticosteroids


Asunto(s)
Asma/tratamiento farmacológico , Dexametasona/uso terapéutico , Hipersensibilidad/tratamiento farmacológico , Animales , Asma/complicaciones , Quimioterapia Combinada , Femenino , Fibrosis/prevención & control , Hipersensibilidad/complicaciones , Ratones , Ratones Endogámicos BALB C , Factor Trefoil-2/efectos de los fármacos
3.
Am J Physiol Gastrointest Liver Physiol ; 310(1): G13-25, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26514775

RESUMEN

We tested whether the T helper (Th) type 2 (Th2) cell agonist and allergenic ligand IL-33 was associated with eosinophilic esophagitis (EoE) development in a pediatric cohort and whether IL-33 protein could induce disease symptoms in mice. Biopsies from EoE patients or controls were used to measure IL-33 mRNA and protein expression. Increased expression of IL-33 mRNA was found in the esophageal mucosa in EoE. IL-33 protein was detected in cells negative for CD45, mast cells, and epithelial cell markers near blood vessels. Circulating levels of IL-33 were not increased. The time course for IL-33 gene expression was quantified in an established Aspergillus fumigatus allergen mouse model of EoE. Because IL-33 induction was transient in this model and chronicity of IL-33 expression has been demonstrated in humans, naive mice were treated with recombinant IL-33 for 1 wk and esophageal pathology was evaluated. IL-33 application produced changes consistent with phenotypically early EoE, including transmural eosinophilia, mucosal hyperproliferation, and upregulation of eosinophilic genes and chemokines. Th2 cytokines, including IL-13, along with innate lymphoid cell group 2, Th1/17, and M2 macrophage marker genes, were increased after IL-33 application. IL-33-induced eosinophilia was ablated in IL-13 null mice. In addition, IL-33 induced a profound inhibition of the regulatory T cell gene signature. We conclude that IL-33 gene expression is associated with pediatric EoE development and that application of recombinant protein in mice phenocopies the early clinical phase of the human disease in an IL-13-dependent manner. IL-33 inhibition of esophageal regulatory T cell function may induce loss of antigenic tolerance, thereby providing a mechanistic rationale for EoE development.


Asunto(s)
Esofagitis Eosinofílica/inducido químicamente , Esofagitis Eosinofílica/metabolismo , Esófago/metabolismo , Mediadores de Inflamación/metabolismo , Interleucina-33/metabolismo , Inmunidad Adaptativa , Adolescente , Animales , Aspergillus fumigatus/patogenicidad , Biopsia , Estudios de Casos y Controles , Proliferación Celular , Quimiocina CCL26 , Quimiocinas CC/metabolismo , Niño , Preescolar , Modelos Animales de Enfermedad , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Esofagitis Eosinofílica/genética , Esofagitis Eosinofílica/inmunología , Esofagitis Eosinofílica/microbiología , Esofagitis Eosinofílica/patología , Esófago/inmunología , Esófago/microbiología , Esófago/patología , Humanos , Tolerancia Inmunológica , Inmunidad Innata , Interleucina-13/deficiencia , Interleucina-13/genética , Interleucina-33/genética , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones Endogámicos BALB C , Ratones Noqueados , Fenotipo , ARN Mensajero/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Factores de Tiempo , Regulación hacia Arriba
4.
Oncogene ; 34(22): 2856-66, 2015 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-25088199

RESUMEN

Runt domain transcription factor 3 (RUNX3) is widely regarded as a tumour-suppressor gene inactivated by DNA hypermethylation of its canonical CpG (cytidine-phosphate-guanidine) island (CGI) promoter in gastric cancer (GC). Absence of RUNX3 expression from normal gastric epithelial cells (GECs), the progenitors to GC, coupled with frequent RUNX3 overexpression in GC progression, challenge this longstanding paradigm. However, epigenetic models to better describe RUNX3 deregulation in GC have not emerged. Here, we identify lineage-specific DNA methylation at an alternate, non-CGI promoter (P1) as a new mechanism of RUNX3 epigenetic control. In normal GECs, P1 was hypermethylated and repressed, whereas in immune lineages P1 was hypomethylated and widely expressed. In human GC development, we detected aberrant P1 hypomethylation signatures associated with the early inflammatory, preneoplastic and tumour stages. Aberrant P1 hypomethylation was fully recapitulated in mouse models of gastric inflammation and tumorigenesis. Cell sorting showed that P1 hypomethylation reflects altered cell-type composition of the gastric epithelium/tumour microenvironment caused by immune cell recruitment, not methylation loss. Finally, via long-term culture of gastric tumour epithelium, we revealed that de novo methylation of the RUNX3 canonical CGI promoter is a bystander effect of oncogenic immortalization and not likely causal in GC pathogenesis as previously argued. We propose a new model of RUNX3 epigenetic control in cancer, based on immune-specific, non-CGI promoter hypomethylation. This novel epigenetic signature may have utility in early detection of GC and possibly other epithelial cancers with premalignant immune involvement.


Asunto(s)
Linaje de la Célula/genética , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Metilación de ADN , Lesiones Precancerosas/genética , Lesiones Precancerosas/inmunología , Neoplasias Gástricas/genética , Neoplasias Gástricas/inmunología , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/inmunología , Células Cultivadas , Islas de CpG , Mucosa Gástrica/inmunología , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Especificidad de Órganos/genética , Especificidad de Órganos/inmunología , Regiones Promotoras Genéticas , Neoplasias Gástricas/patología
5.
Peptides ; 31(2): 284-90, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19944725

RESUMEN

We have previously demonstrated that pregnant ovine endometrium expresses the gastrin-releasing peptide (GRP) gene at a high level following conceptus implantation. Here we report the isolation, characterization and biological activity of ovine GRP 1-46, the primary product of this gene in the pregnant endometrium. Full thickness 125-140-day pregnant sheep uterus (term is 145 day) was homogenized in 80% acetonitrile/2% trifluoroacetic acid (1:7 ACN/TFA), concentrated on reverse-phase C18 cartridges and chromatographed successively on gel filtration (Sephadex G-50) and reverse-phase HPLC (C18 muBondapak). Purification was monitored by RIA. Purified GRP peptide was analysed by mass spectrometry giving a major mass ion at 4963 which corresponds exactly to GRP 1-46. Other mass ions from pro-GRP did not contain a biologically active N-terminus or antigenic determinant. Proteolytic cleavage of pro-GRP to give rise to GRP(1-46) would require preferential cleavage at the Glu-Glu bond by a Glu-C2-like enzyme, rather than the trypsin-like and C-terminal amidation enzymes (PAM) that produce GRP(18-27) and GRP(1-27) in other tissues. GRP 1-46 was synthesized and receptor binding and biological activity tested on a range of rodent and human cell lines that express GRP-related receptors GRPR, NMBR and BRS3. GRP 1-46 bound GRPR and NMBR with low affinity, and mobilized inositol phosphate in cell lines expressing the GRPR and NMBR, but not BRS-3. This study describes a new processed product of the GRP gene, GRP 1-46, which is highly expressed in the pregnant sheep endometrium and which acts as a weak agonist at the GRPR and NMBR.


Asunto(s)
Endometrio/química , Péptido Liberador de Gastrina/aislamiento & purificación , Péptido Liberador de Gastrina/farmacología , Secuencia de Aminoácidos , Animales , Línea Celular , Línea Celular Tumoral , Femenino , Péptido Liberador de Gastrina/análisis , Péptido Liberador de Gastrina/metabolismo , Humanos , Indoles/farmacología , Fosfatos de Inositol/metabolismo , Ratones , Datos de Secuencia Molecular , Peso Molecular , Péptidos/genética , Embarazo , Unión Proteica/fisiología , Precursores de Proteínas/genética , Piridinas/farmacología , Ratas , Receptores de Bombesina/antagonistas & inhibidores , Receptores de Bombesina/genética , Receptores de Bombesina/metabolismo , Ovinos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Transfección , Fosfolipasas de Tipo C/metabolismo
6.
Biochim Biophys Acta ; 1793(11): 1623-33, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19665497

RESUMEN

Cytokine signalling pathways that depend on gp130 are dysregulated in several epithelial cancers including gastric cancer. It has been established that blockade of SHP2 activation of MAPK signalling results in hyperactivation of STAT3 resulting in increased cell proliferation, angiogenesis, inflammation and inhibition of both immunocyte and epithelial cell apoptosis. Additionally, key genes regulated downstream of gp130 via MAPK activation such as the stomach-specific tumor suppressor gene tff1 are suppressed, contributing to the oncogenic outcome. The main cytokine driver of gp130 signalling in the stomach is IL-11, with IL-6 having little activity in the antral stomach in which most pathology initiates. IL-11 is up-regulated in both mouse and human gastric cancer and in pre-neoplastic mucosa. A characteristic gene signature specifically associated with IL-11 drive has been observed, although the prognostic value of the signature has not yet been assessed. Infection of human or mouse stomach with Helicobacter pylori, especially that expressing the CagA cytotoxin, produces constitutive MAPK activation, but also activated STAT3 and increases IL-11 expression. The possibility of designing and utilising small molecule inhibitors of either IL-11 or STAT3 activation may be worthwhile in developing new cancer therapeutics.


Asunto(s)
Receptor gp130 de Citocinas/metabolismo , Interleucina-11/metabolismo , Interleucina-6/metabolismo , Sistema de Señalización de MAP Quinasas , Neoplasias Gástricas/metabolismo , Animales , Antígenos Bacterianos/metabolismo , Apoptosis , Proteínas Bacterianas/metabolismo , Proliferación Celular , Activación Enzimática , Mucosa Gástrica/metabolismo , Regulación Neoplásica de la Expresión Génica , Infecciones por Helicobacter/tratamiento farmacológico , Infecciones por Helicobacter/metabolismo , Helicobacter pylori , Humanos , Interleucina-11/antagonistas & inhibidores , Ratones , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Neovascularización Patológica/microbiología , Péptidos/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/metabolismo , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/microbiología , Factor Trefoil-1 , Proteínas Supresoras de Tumor/metabolismo
7.
J Pathol ; 213(2): 140-51, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17724739

RESUMEN

H. pylori infection accounts for most cases of gastric cancer, but the initiating events remain unclear. The principal H. pylori pathogenicity-associated CagA protein disrupts intracellular SHP-2 signalling pathways including those used by the IL-6 family cytokines, IL-6 and IL-11. Imbalanced IL-6 family cytokine signalling in the gp130(757FF) mouse model of gastric cancer arising from hyperactivation of oncogenic STAT3 after altered SHP-2 : ERK1/2 signalling produces dysplastic antral tumours preceded by gastritis and metaplasia. In a cohort of patient gastric biopsies with known H. pylori and CagA status, we investigated whether (i) STAT3 and ERK1/2 activation is altered in H. pylori-dependent gastritis; (ii) these profiles are more pronounced in CagA+ H. pylori infection; and (iii) the expression of pro-inflammatory cytokines that activate STAT3 and ERK 1/2 pathways is associated with progression to gastric cancer. IL-6, IL-11, and activated STAT3 and ERK1/2 were quantified in antral biopsies from gastritic stomach, metaplastic tissue, and resected gastric cancer tissues. We observed significantly increased STAT3 and ERK1/2 activation (p = 0.001) in H. pylori-dependent gastritis, which was further enhanced in the presence of CagA+ H. pylori strains. Of known gastric ligands that drive STAT3 activation, IL-6 expression was increased after H. pylori infection and both IL-6 and IL-11 were strongly up-regulated in the gastric cancer biopsies. This suggests a mechanism by which IL-11 drives STAT3 activation and proliferation during gastric cancer progression. We addressed this using an in vitro approach, demonstrating that recombinant human IL-11 activates STAT3 and concomitantly increases proliferation of MKN28 gastric epithelial cells. In summary, we show increased STAT3 and ERK1/2 activation in H. pylori-dependent gastritis that is likely driven in an IL-6-dependent fashion. IL-11 expression is associated with adenocarcinoma development, but not gastritic lesions, and we identify a novel mechanism for IL-11 as a potent inducer of proliferation in the human gastric cancer setting.


Asunto(s)
Interleucina-6/metabolismo , Neoplasias Gástricas/inmunología , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/microbiología , Adenocarcinoma/patología , Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Biopsia , Proliferación Celular , Progresión de la Enfermedad , Activación Enzimática , Mucosa Gástrica/metabolismo , Mucosa Gástrica/microbiología , Gastritis/metabolismo , Gastritis/microbiología , Regulación Neoplásica de la Expresión Génica , Infecciones por Helicobacter/complicaciones , Helicobacter pylori , Humanos , Interleucina-11/metabolismo , Interleucina-8/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas de Neoplasias/metabolismo , Inhibidores de la Bomba de Protones , Antro Pilórico/microbiología , Antro Pilórico/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Neoplasias Gástricas/genética , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/patología , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/metabolismo
8.
Artículo en Inglés | MEDLINE | ID: mdl-16935852

RESUMEN

Gastric trefoil peptides mediate mucosal repair by stimulating cell migration, inhibiting apoptosis and inflammation, and likely augmenting the barrier function of mucus. One of these, tff1, is a gastric-specific tumor suppressor gene, which when repressed is associated with gastric cancer progression. IL-6 family cytokines play an important role in maintaining gastric homeostasis by regulating tff1 and other mediators of mucosal proliferation, inflammation, angiogenesis, and apoptosis. In this review the signaling cascades downstream of the common IL-6 cytokine family coreceptor gp130 that contribute to control of this homeostasis are described, as are the pathological outcomes of imbalancing these pathways.


Asunto(s)
Mucosa Gástrica/citología , Mucosa Gástrica/fisiología , Interleucina-6/fisiología , Animales , Estrógenos/fisiología , Homeostasis , Humanos , Transducción de Señal , Factor Trefoil-1 , Proteínas Supresoras de Tumor/fisiología
9.
Cell Mol Life Sci ; 62(24): 2921-31, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16374580

RESUMEN

The expression of trefoil peptides and their biological consequences are regulated in a multifactorial fashion, and much more work is required in order to fully understand the underlying molecular mechanisms. Central to this will be the identification and functional analysis of trefoil peptide receptors and the full complement of binding proteins. This review summarizes the often fragmentary information available on the environmental, chemical and local regulatory molecules that control trefoil gene expression. Special attention is paid to the nature of the signaling cascades that are activated and the binding proteins that modulate gene transcription. Epigenetic regulation of trefoil gene expression, particularly the role of (de)methylation is described, and the signaling pathways downstream of trefoil peptide activation of target cells are enumerated, as are their physiological and pathological outcomes.


Asunto(s)
Regulación de la Expresión Génica/genética , Péptidos/genética , Péptidos/metabolismo , Transducción de Señal/fisiología , Animales , Humanos
10.
J Pathol ; 207(1): 43-52, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15983982

RESUMEN

H+/K+-ATPase beta-subunit-deficient mice (129/Sv background) display numerous pathologies in the stomach. Expression of the mutation in BALB/cCrSlc mice results in the development of an aberrant 'mucus-rich' cell population. 'Mucus-rich' cells have been described in stomachs of mice with autoimmune gastritis, a disease mediated by CD4+ T cells. Other pathological features of autoimmune gastritis are similar to those in H+/K+ beta-deficient mice and include a mononuclear cell infiltrate in the gastric mucosa, non-functional or absent parietal cells, depletion of zymogenic cells, hypergastrinaemia, and gastric unit hypertrophy caused by immature cell hyperplasia. The present study investigates further the aberrant gastric 'mucus-rich' cell lineage and analyses the mRNA expression of mucus cell products TFF1 and TFF2. 'Mucus-rich' cells stained for both acidic and neutral mucins, and with a TFF2-specific antibody. Stomachs from both models expressed decreased TFF1 mRNA and reciprocally increased TFF2 mRNA. The involvement of gastrin in regulating trefoil mRNA expression was also investigated using gastrin-deficient mice. In contrast to previous findings, gastrin did not positively regulate TFF1 mRNA expression, but there was possible augmentation of TFF2. Additionally, a clear role for inflammation was established involving both polymorphonuclear and mononuclear cells in these models, and a link was found between mucosal hypertrophy and increased interleukin-11 (IL-11) expression.


Asunto(s)
Mucosa Gástrica/patología , Gastritis/metabolismo , Mucinas/metabolismo , Proteínas Musculares/metabolismo , Péptidos/metabolismo , Animales , Enfermedades Autoinmunes/metabolismo , Citocinas/biosíntesis , Citocinas/genética , Citocinas/fisiología , Modelos Animales de Enfermedad , Mucosa Gástrica/metabolismo , Gastrinas/sangre , Regulación de la Expresión Génica , ATPasa Intercambiadora de Hidrógeno-Potásio/deficiencia , ATPasa Intercambiadora de Hidrógeno-Potásio/genética , Hiperplasia/metabolismo , Hipertrofia/metabolismo , Interleucina-11/biosíntesis , Interleucina-11/genética , Interleucina-11/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Mutantes , Mucinas/genética , Mucinas/fisiología , Proteínas Musculares/genética , Proteínas Musculares/fisiología , Péptidos/genética , Péptidos/fisiología , ARN Mensajero/genética , Especificidad de la Especie , Factor Trefoil-1 , Factor Trefoil-2
11.
J Endocrinol ; 175(2): 447-57, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12429042

RESUMEN

High concentrations of a peptide related to gastrin-releasing peptide (GRP) are produced in the utero-placental unit of the human and sheep and secreted into the general circulation. This suggests an endocrine role in addition to its role as a neurotransmitter/neuromodulator. The GRP is larger than the previously described form GRP(1-27) but it is not known whether the larger form is the product of a related GRP-like gene or differences in post-translational processing. We have therefore cloned the gene for the sheep homologue of the GRP gene and determined its distribution. Only a single GRP gene was found in the sheep. This had a similar organisation to the human GRP gene with three exons and two introns. The larger form of GRP in the pregnant endometrium therefore appears to be the result of an alteration in processing of the GRP prohormone. The expression of GRP mRNA in the pregnant uterus was extraordinarily high comprising one-third of all mRNA synthesised by the pregnant endometrium. As the endometrial GRP mRNA arises solely from the glandular epithelium, the localised synthesis of GRP mRNA would be far higher. GRP mRNA was expressed in a wide variety of fetal tissues (fundus, colon, jejunum, ileum, duodenum, kidney, adrenal, lung, heart and pancreas) with a corresponding presence of GRP immunoreactivity. The expression of GRP in the fetal lung was biphasic with peaks at mid-term and near parturition but none in the adult supporting the concept of a specific developmental role of GRP in the lung.


Asunto(s)
Feto/fisiología , Péptido Liberador de Gastrina/genética , Genes/genética , Preñez/genética , Ovinos/genética , Útero/fisiología , Animales , Secuencia de Bases , Northern Blotting , ADN Circular/genética , Endometrio/fisiología , Femenino , Expresión Génica/genética , Datos de Secuencia Molecular , Embarazo , ARN Mensajero/análisis , ARN Mensajero/genética , Secuencias Repetitivas de Ácidos Nucleicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Histopathology ; 40(3): 261-8, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11895492

RESUMEN

AIMS: Maintenance of the cellular integrity of the biliary epithelium may involve the production of mucins and mucin-associated peptides. In the luminal gastrointestinal tract, mucins and the mucin-associated trefoil peptides (TFF) are integral to cytoprotection and cellular repair of the mucosa. METHODS AND RESULTS: Samples of normal and diseased human liver tissue were examined using histological and immunohistochemical techniques, for the expression of TFF and mucins. Bile ducts were classified as small, medium or large depending upon the number of biliary epithelial cells. TFF expression was demonstrated in biliary epithelial cells of both normal and diseased liver tissue. TFF expression was greatest in the large bile ducts. In normal liver tissue, expression of at least one TFF was demonstrated in 2-7% of small bile ducts, 5-31% of medium bile ducts and 31-85% of large bile ducts. Seventy-seven percent of large bile ducts secreted mucins and all three TFF concurrently, compared with 3% of medium bile ducts and no small bile ducts. Biliary disease resulted in an increased expression of TFF1 and TFF3 in the medium bile ducts. CONCLUSIONS: The biliary epithelial cells in normal and diseased human liver tissue express TFF, particularly in the larger bile ducts. TFF expression may be up-regulated or induced in biliary diseases as a response to injury, as is seen in epithelial damage elsewhere in the gastrointestinal tract.


Asunto(s)
Conductos Biliares/patología , Enfermedades de las Vías Biliares/patología , Sustancias de Crecimiento/biosíntesis , Mucinas , Proteínas Musculares , Neuropéptidos , Adulto , Anciano , Conductos Biliares/química , Enfermedades de las Vías Biliares/metabolismo , Femenino , Humanos , Inmunohistoquímica , Hígado/química , Hígado/patología , Hepatopatías/metabolismo , Hepatopatías/patología , Masculino , Persona de Mediana Edad , Péptidos , Factor Trefoil-2 , Factor Trefoil-3
13.
Regul Pept ; 101(1-3): 35-41, 2001 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-11495677

RESUMEN

The trefoil factor TFF3 is a peptide predominantly produced by mucus-secreting cells in the small and large intestines. It has been implicated in intestinal protection and repair. The mechanisms that govern TFF3 secretion are poorly understood. The aim of this study was, therefore, to evaluate the influence of neurotransmitters, hormonal peptides and mediators of inflammation on the release of TFF3. For this purpose, an isolated vascularly perfused rat colon preparation was used. After a bolus administration of 1 ml isotonic saline into the lumen, TFF3 secretion was induced by a 30-min intra-arterial infusion of the compounds to be tested. TFF3 was evaluated in the luminal effluent using a newly developed radioimmunoassay. TFF3 was barely detected in crude luminal samples. In contrast, dithiothreitol (DTT) treatment of the effluent revealed TFF3 immunoreactivity, which amounted to about 0.3 pmol min(-1) cm(-1) in the basal state. Gel chromatography of DTT-treated luminal samples revealed a single peak that co-eluted with the monomeric form of TFF3. TFF3 was not detected in the portal effluent. Bethanechol (10(-6)-10(-4) M), vasoactive intestinal peptide (VIP, 10(-8)-10(-7) M) or bombesin (10(-8)-10(-7) M) induced a dose-dependent release of TFF3. In contrast, substance P evoked a modest release of TFF3, whereas calcitonin gene-related peptide (CGRP), somatostatin, neurotensin or peptide YY (PYY) did not modify TFF3 secretion. The degranulator compound bromolasalocid, 16,16-dimethyl PGE2 (dmPGE2) or interleukin-1-beta (IL-1-beta) also evoked a marked release of TFF3. In conclusion, TFF3 in the colonic effluent is present in a complex. This association presumably involves a disulfide bond. Additionally, the present results suggest a role for enteric nervous system and resident immune cells in mediation of colonic TFF3 secretion.


Asunto(s)
Colon/metabolismo , Lasalocido/análogos & derivados , Mucinas , Proteínas Musculares , Neuropéptidos/farmacología , Proteínas/metabolismo , 16,16-Dimetilprostaglandina E2/administración & dosificación , 16,16-Dimetilprostaglandina E2/farmacología , Animales , Betanecol/administración & dosificación , Betanecol/farmacología , Bombesina/administración & dosificación , Bombesina/farmacología , Colon/irrigación sanguínea , Colon/efectos de los fármacos , Infusiones Intraarteriales , Interleucina-1/administración & dosificación , Interleucina-1/farmacología , Lasalocido/administración & dosificación , Lasalocido/farmacología , Masculino , Neuropéptidos/administración & dosificación , Neurotransmisores/administración & dosificación , Neurotransmisores/farmacología , Péptidos , Perfusión , Proteínas/inmunología , Radioinmunoensayo , Ratas , Ratas Wistar , Factor Trefoil-3 , Péptido Intestinal Vasoactivo/administración & dosificación , Péptido Intestinal Vasoactivo/farmacología
14.
J Gastroenterol Hepatol ; 16(5): 506-12, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11350545

RESUMEN

BACKGROUND: The trefoil peptide (TFF1) is a member of a family of mucin-associated regulatory peptides that are widely distributed in gastrointestinal tissues and have been implicated in the maintenance of the gastric mucosa. The role of TFF1 in gastric mucosal repair was examined by analysis of the spatio-temporal expression of TFF1 following gastric ulceration in the rat. METHODS: Gastric ulcers were induced in rats by application of glacial acetic acid to the serosa of the fundus. At various time points post injury (0-28 days), macroscopic and microscopic examination of the gastric mucosa was performed. In addition, the spatio-temporal expression of TFF1 protein and proliferating cell nuclear antigen were identified by immunohistochemistry, TFF1 message by in situ hybridization, and acidic/neutral secreting mucins by Alcian blue-periodic acid-Schiff staining. RESULTS: In normal rat gastric tissue, TFF1 peptide and mRNA were expressed in mucosal cells of the superficial epithelium. Trefoil peptide and mRNA were significantly induced between 4 and 28 days post ulceration, with expression extending beyond the superficial epithelium and being localized to acidic mucin-producing cells deep within the repairing mucosa. CONCLUSIONS: Spatio-temporal expression of TFF1 mRNA and peptide following macroscopic repair implicates TFF1 as a potential mediator of late stage-repair processes. Whether this is through direct stimulation of cellular differentiation or the enhancement of mucosal protective properties through an interaction with gastric mucins remains to be elucidated.


Asunto(s)
Mucosa Gástrica/metabolismo , Proteínas/metabolismo , Úlcera Gástrica/metabolismo , Animales , Femenino , Inmunohistoquímica , Hibridación in Situ , Masculino , Mucinas/análisis , Antígeno Nuclear de Célula en Proliferación/análisis , ARN Mensajero/análisis , Ratas , Ratas Long-Evans , Factores de Tiempo , Factor Trefoil-1 , Proteínas Supresoras de Tumor
15.
J Biol Chem ; 276(26): 24088-96, 2001 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-11301329

RESUMEN

To further examine the function of the trefoil factor family (TFF), the expression of which is up-regulated at sites of injury, we have produced transgenic mice that chronically express rat TFF3 within the jejunum (using a rat fatty acid-binding protein promoter). The expression of rat TFF3 was limited to the villi of the jejunum and had no effect on base-line morphology. Rat TFF3 expression did result, however, in a reduced sensitivity to indomethacin (85 mg/kg subcutaneously), which only caused a 29% reduction in villus height in transgenics versus 51% reduction in controls (p < 0.01). Indomethacin increased initial intestinal epithelial cell proliferation and migration, but the presence of rat TFF3 caused no additional change in proliferation (bromodeoxyuridine), cell migration ([(3)H]thymidine and bromodeoxyuridine), apoptosis (terminal deoxyuridine nucleotidyl nick end labeling), or E-cadherin immunostaining. In vitro studies following changes in resistance of intestinal strips in Ussing chambers (voltage-clamp technique) showed increased base-line resistance in the rat TFF3-expressing region (326 +/- 60 versus 195 +/- 48 ohm.cm(2) in controls, p < 0.05) and reduced the fall in resistance following HCl exposure by about 40% (p < 0.01). Overexpression of TFF3 stabilizes the mucosa against noxious agents, supporting its role in mucosal protection/repair. It may therefore provide a novel approach to the prevention and/or treatment of intestinal ulceration.


Asunto(s)
Yeyuno/metabolismo , Mucinas , Proteínas Musculares , Proteínas de Neoplasias , Proteínas del Tejido Nervioso , Neuropéptidos , Proteínas/fisiología , Animales , Apoptosis/efectos de los fármacos , Fusión Artificial Génica , Proteínas Portadoras/genética , División Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proteína de Unión a los Ácidos Grasos 7 , Proteínas de Unión a Ácidos Grasos , Sustancias de Crecimiento/fisiología , Indometacina/farmacología , Enfermedades Intestinales/metabolismo , Enfermedades Intestinales/patología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Yeyuno/efectos de los fármacos , Yeyuno/patología , Ratones , Ratones Transgénicos , Técnicas de Placa-Clamp , Péptidos/fisiología , Proteínas/genética , Proteínas/metabolismo , Ratas , Factor Trefoil-2 , Factor Trefoil-3 , Cicatrización de Heridas
16.
Lab Invest ; 81(3): 397-408, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11310832

RESUMEN

The trefoil peptides spasmolytic polypeptide (SP), intestinal trefoil factor (ITF), and pS2 show lineage-specific expression in the normal gut and are strongly induced after mucosal injury. We assessed the relationship between this induction and the development of the regenerative epithelial lineage over time in the rat stomach and verified these observations in the metaplastic and dysplastic human stomach. Antral or colonic ulcers were induced in Wistar rats by application of serosal acetic acid and tissues harvested 2 hours to 125 days later. Human endoscopic biopsies or gastric resection specimens were also assessed. Tissues were examined by radioimmunoassay, immunoblotting, or immunohistochemistry for ITF, SP, and transforming growth factor alpha (rat) or ITF and pS2 (human) expression. ITF and SP mRNA in antral ulcer margins was localized by in situ hybridization. ITF and SP peptide expression rose steadily in ulcer margins after 4 days, with the rise in ITF being more pronounced. By 40 days, several hundred-fold elevations in ITF levels were present, with a field effect in uninvolved mucosa. Hyperproliferative, elongated glands of undifferentiated cells expressing abundant trefoil peptides and acid sulfomucins were present after day 12 and persisted after ulcer healing. ITF mRNA was aberrantly expressed in basal and mid-regions of these regenerative glands. In contrast, transforming growth factor alpha peptide expression rose promptly after injury then fell to baseline levels with healing. Seven months after injury, gastric atrophy, intestinal metaplasia, and severe dysplasia with conserved ITF expression were seen. ITF was also induced in human intestinal metaplasia and conserved in all gastric cancers, whereas expression of the gastric peptide pS2 was progressively reduced in the progression from metaplasia to dysplasia. Persistent, selective overexpression of ITF, possibly acting in an autocrine fashion, is a feature of regeneration after antral ulceration, and may provide insight into the nature of metaplastic phenotypes arising from chronic gastric injury. The loss of pS2 expression in metaplasia and cancer supports a role for this protein in gastric tumor suppression.


Asunto(s)
Adenocarcinoma/patología , Mucosa Gástrica/patología , Sustancias de Crecimiento/genética , Mucinas , Proteínas Musculares , Neuropéptidos , Péptidos/genética , Proteínas/genética , Neoplasias Gástricas/patología , Adenocarcinoma/metabolismo , Animales , Anticuerpos , Biopsia , Diferenciación Celular/fisiología , Mucosa Gástrica/metabolismo , Mucosa Gástrica/fisiología , Gastritis/metabolismo , Gastritis/patología , Expresión Génica/fisiología , Sustancias de Crecimiento/metabolismo , Humanos , Masculino , Metaplasia/metabolismo , Metaplasia/patología , Péptidos/metabolismo , Proteínas/análisis , Proteínas/inmunología , ARN Mensajero/análisis , Conejos , Ratas , Ratas Wistar , Regeneración , Neoplasias Gástricas/metabolismo , Úlcera Gástrica/metabolismo , Úlcera Gástrica/patología , Sustancia P/genética , Factor Trefoil-1 , Factor Trefoil-2 , Factor Trefoil-3 , Proteínas Supresoras de Tumor , Regulación hacia Arriba/fisiología
18.
Int J Dev Biol ; 45(8): 887-93, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11804032

RESUMEN

The signals that guide the morphogenesis and differentiation of rat fetal gastric mucosa remain largely unknown. We have investigated the role of capsulin in pit/gland formation and epithelial cell differentiation in cultured stomach tissue. Embryonic day 16.5 (E 16.5) stomach tissue cultured for three days in the presence of 1 microM hydrocortisone underwent dramatic transformation, from undifferentiated, stratified cells to differentiated epithelia composed of polarised columnar cells with mucous cells and pit/glands. In the presence of capsulin antisense oligonucleotides directed against capsulin mRNA, tissues do not undergo further development. Significantly, both mucous granules and pit/gland formation were inhibited compared to capsulin sense/scrambled oligonucleotide treated controls. However, in tissues treated with specific anti-rat HGF-antiserum to neutralise secreted HGF, pit/gland formation was inhibited, but the number of mucous granules remained unchanged compared to controls treated with non-specific antiserum (mouse monoclonal cytokeratin 8 antiserum). This data suggests that capsulin may have a role in the morphogenesis of pit/glands and mucin granule formation in the developing rat gastric mucosa. We discuss the possibility that this role of capsulin may be partly mediated through the actions of HGF.


Asunto(s)
Mucosa Gástrica/embriología , Factores de Transcripción/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Diferenciación Celular , Células Cultivadas , Femenino , Mucosa Gástrica/citología , Morfogénesis , Oligonucleótidos Antisentido , Fenotipo , Embarazo , Ratas , Ratas Wistar , Factores de Transcripción/genética
19.
J Gastroenterol Hepatol ; 16(11): 1222-7, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11903739

RESUMEN

BACKGROUND: Omeprazole is an inhibitor of the H+K+ ATPase of the gastric parietal cell, which is used clinically to suppress gastric acid secretion. It has also been found to inhibit gastric mucin production; however, its effects on the synthesis and secretion of the trefoil peptides, which are also expressed by mucus cells, and which play a key role in cytoprotection and epithelial repair, are unknown. METHODS: Rats (n=8) were given either omeprazole (30 mg/kg per day; p.o.) or inert carrier for 1 week, and the effects on synthesis and peptide expression of the gastric trefoil peptides, TFF1/pS2 and TFF2/SP, were compared. RESULTS: As expected, omeprazole treatment abolished H+ ion production with a mean gastric juice pH of 7.2 compared with 2.4 for controls. The omeprazole group had elevated total protein levels of 35-fold and TFF1/pS2 peptide levels elevated fourfold, respectively, but not TFF2/SP peptide in gastric juice, suggesting that the increased pH reduced the viscosity of adherent mucus, thereby increasing gastric juice concentrations by dissolution of adherent TFF1/pS2 and increased secretion. Concomitant with increased TFF1/pS2 secretion was a fall in predominantly antral mucosal trefoil peptide concentrations. In contrast to trefoil secretory rates, the steady-state synthesis of both TFF1/pS2 and TFF2/SP was unchanged after omeprazole treatment, implying both a large cellular pool of processed peptide and rapid secretion. CONCLUSION: The increase in the concentration of TFF1/pS2 in gastric secretions during chronic omeprazole-induced achlorhydria may be important in preventing tissue injury and promoting repair in response to an increased luminal bacterial population.


Asunto(s)
Aclorhidria/metabolismo , Mucosa Gástrica/metabolismo , Sustancias de Crecimiento/biosíntesis , Mucinas , Proteínas Musculares , Neuropéptidos , Aclorhidria/inducido químicamente , Animales , Masculino , Omeprazol , Péptidos , Ratas , Ratas Wistar , Factor Trefoil-2 , Factor Trefoil-3
20.
J Endocrinol ; 166(3): 649-58, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10974658

RESUMEN

In the ovine endometrium, dramatic increases in gastrin-releasing peptide (GRP) mRNA and immunoreactivity are observed during the luteal regression phase of the oestrous cycle (24-fold) and during pregnancy (at least 150-fold). This study sought to determine whether oestrogen and/or progesterone were responsible for the temporal regulation of GRP observed in the uterus. Ovariectomized sheep were divided into four groups (n=4), as follows: 1, untreated; 2, given subcutaneous and intravaginal progesterone implants; 3, given subcutaneous oestrogen implants; and 4, treated with both oestrogen and progesterone. After 10 days, the animals were sacrificed and plasma, pituitary and endometrium were obtained. A fifth group of sheep with intact ovaries was included. Analysis of endometrial GRP-immunoreactivity (GRP-ir) revealed a twofold drop for groups treated with oestrogen, progesterone or both hormones. A dramatic reduction in endometrial GRP mRNA was o! bserved in the group treated with both hormones. GRP-ir was measured in whole pituitaries and found to vary greatly (1.7-53.7 pmol/g tissue) within all groups of ovariectomized animals. There were no significant differences between any of the five groups. A significant reduction in circulating GRP-ir was observed after 10 days of treatment with either oestrogen or progesterone. These studies demonstrate that, in sheep, the synthesis, storage and secretion of GRP are differentially affected by oestrogen and progesterone. Regulation appears to be tissue specific since GRP content in the pituitary is unchanged by oestrogen or progesterone whereas GRP expression in the endometrium is inhibited. Changes in GRP mRNA expression did not correlate with changes in endometrial expression of mRNA for oestrogen receptor alpha, oestrogen receptor beta and the progesterone receptor. This study is the first reported demonstration that expression of the GRP gene can be influenced by the presence of ovarian steroids, with the conclusion that oestrogen and/or progesterone act as negative regulators of endometrial GRP expression.


Asunto(s)
Endometrio/metabolismo , Estrógenos/farmacología , Péptido Liberador de Gastrina/metabolismo , Progesterona/farmacología , Actinas/genética , Animales , Northern Blotting , Implantes de Medicamentos , Endometrio/efectos de los fármacos , Femenino , Péptido Liberador de Gastrina/biosíntesis , Ovariectomía , Hipófisis/metabolismo , ARN Mensajero/análisis , Receptores de Estrógenos/genética , Receptores de Progesterona/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ovinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...