Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell Tissue Res ; 375(3): 743-754, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30377784

RESUMEN

We previously demonstrated that female Runx3 knockout (Runx3-/-) mice were anovulatory and their uteri were atrophic and that Runx3 mRNA was expressed in granulosa cells. To clarify how Runx3 regulates folliculogenesis and ovulation, we examine the effects of Runx3 knockout on the gene expression of growth factors associated with folliculogenesis and enzymes associated with steroidogenesis. In Runx3-/- mouse ovaries, the numbers of primary and antral follicles were lower than those in wild-type (wt) mice at 3 weeks of age, indicating that the loss of Runx3 affects folliculogenesis. The expression of genes encoding activin and inhibin subunits (Inha, Inhba and Inhbb) was also decreased in ovaries from the Runx3-/- mice compared with that in wt mice. Moreover, the expression of the genes Cyp11a1 and Cyp19a1 encoding steroidogenic enzymes was also decreased. In cultured granulosa cells from 3-week-old mouse ovaries, Cyp19a1 mRNA levels were lower in Runx3-/- mice than those in wt mice. Follicle-stimulating hormone (FSH) treatment increased Cyp19a1 mRNA levels in both wt and Runx3-/- granulosa cells in culture but the mRNA level in Runx3-/- granulosa cells was lower than that in wt ones, indicating that granulosa cells could not fully function in the absence of Runx3. At 3 weeks of age, gonadotropin α subunit, FSHß subunit and luteinizing hormone (LH) ß subunit mRNA levels were decreased in Runx3-/- mice. These findings suggest that Runx3 plays a key role in female reproduction by regulating folliculogenesis and steroidogenesis in granulosa cells.


Asunto(s)
Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Células de la Granulosa/metabolismo , Organogénesis , Esteroides/biosíntesis , Animales , Subunidad alfa 3 del Factor de Unión al Sitio Principal/deficiencia , Estradiol/biosíntesis , Femenino , Hormona Folículo Estimulante/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Células de la Granulosa/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Ratones Endogámicos BALB C , Organogénesis/efectos de los fármacos , Progesterona/biosíntesis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Gonadotropina/genética , Receptores de Gonadotropina/metabolismo
2.
Br J Cancer ; 118(7): 972-984, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29527007

RESUMEN

BACKGROUND: Diffuse-type gastric cancer (DGC) exhibits rapid disease progression and poor patient prognosis. We have previously established an E-cadherin/p53 double conditional knockout (DCKO) mouse line as the first genetically engineered one, which morphologically and molecularly recapitulates human DGC. In this study, we explored low-molecular-weight drugs selectively eliminating mouse and human DGC cells. METHODS: We derived mouse gastric cancer (GC) cell lines from DGC of the DCKO mice demonstrating enhanced tumourigenic activity in immunodeficient mice and acquired tolerance to cytotoxic anti-cancer agents. RESULTS: We performed a synthetic lethal screening of 1535 annotated chemical compounds, and identified 27 candidates selectively killing the GC cell lines. The most potent drug mestranol, an oestrogen derivative, and other oestrogen receptor modulators specifically attenuated cell viability of the GC cell lines by inducing apoptosis preceded by DNA damage. Moreover, mestranol could significantly suppress tumour growth of the GC cells subcutaneously transplanted into nude mice, consistent with longer survival time in the female DCKO mice than in the male. Expectedly, human E-cadherin-mutant and -low gastric cancer cells showed higher susceptibility to oestrogen drugs in contrast to E-cadherin-intact ones in vitro and in vivo. CONCLUSIONS: These findings may lead to the development of novel therapeutic strategies targeting DGC.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Animales , Antineoplásicos/clasificación , Antineoplásicos/uso terapéutico , Proteínas Cdh1/genética , Línea Celular Tumoral , Modelos Animales de Enfermedad , Ensayos de Selección de Medicamentos Antitumorales , Masculino , Ratones , Ratones Noqueados , Ratones Desnudos , Neoplasias Gástricas/patología , Proteína p53 Supresora de Tumor/genética
3.
J Reprod Dev ; 62(5): 479-486, 2016 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-27301496

RESUMEN

We previously demonstrated that the Runx3 transcription factor is expressed in the hypothalami, pituitaries, and ovaries of mice, and that Runx3 knockout (Runx3-/-) mice are anovulatory and their uteri are atrophic. Runx3 mRNA expression was detected in the granulosa cells of ovarian follicles, and in the anteroventral periventricular nucleus (AVPV) and arcuate nucleus (ARC). In the present study, we examined the effects of Runx3 knockout on the gene expression of enzymes associated with steroidogenesis. We found decreased Cyp11a1 mRNA expression in Runx3-/- mouse ovaries compared with that in wild-type (wt) mouse ovaries at the age of 8 weeks. In situ hybridization analysis showed that the percentages of Cyp11a1 mRNA-expressing theca cells in follicles of Runx3-/- mice were decreased compared with those of wt mice. In accord with the alterations in Runx3-/- mouse ovaries, Kiss1 mRNA levels in ARC were increased, whereas mRNA levels of kisspeptin in AVPV were decreased, and gonadotropin-releasing hormone in the preoptic area and follicle-stimulating hormone ß subunit gene were increased in Runx3-/- mice. Following an ovarian transplantation experiment between Runx3-/- mice and wt mice, corpora lutea were observed when ovaries from Runx3-/- mice were transplanted into wt mice, but not when those from wt mice were transplanted into Runx3-/- mice, suggesting that Runx3 in the hypothalamo-pituitary system may drive gonadotropin release to induce ovulation in the ovary. These findings indicate that Runx3 plays a crucial role in the hypothalamo-pituitary-gonadal axis.


Asunto(s)
Subunidad alfa 3 del Factor de Unión al Sitio Principal/fisiología , Ovario/fisiología , Ovulación/fisiología , Animales , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/metabolismo , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Femenino , Gonadotropinas/metabolismo , Células de la Granulosa/citología , Sistema Hipotálamo-Hipofisario , Hipotálamo/metabolismo , Hipotálamo Anterior/fisiología , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Transgénicos , Folículo Ovárico/fisiología , ARN Mensajero/metabolismo , Esteroides/química , Factores de Transcripción/metabolismo
4.
Dev Growth Differ ; 55(9): 786-91, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24299059

RESUMEN

The importance of epithelial-mesenchymal interaction on the development of gastro-intestinal (GI) organs has been repeatedly reported, but its molecular mechanism has not been fully understood though several factors including hepatocyte growth factor and endothelin-3 have been shown to mediate it. Activins have been demonstrated to play important roles in the regulation of organogenesis in vertebrates, but their roles in the regulation of growth and differentiation of GI organs remain to be solved. In the present study, we examined expression of activins in developing rat GI tract, and found that inhibin bA encoding activin A was specifically expressed by GI mesenchymes, while inhibin bB encoding activin B was expressed by both epithelial and mesenchymal components. We then examined the effect of activin A on the growth of fetal rat GI epithelial cells in primary culture. We found that activin A inhibited the growth of forestomach and glandular stomach epithelial cells while it stimulated the growth of colonic epithelial cells. These results suggest that activin A secreted from GI mesenchymes region-specifically regulates the growth of attaching epithelial cells. We thus conclude that activin A mediates epithelial-mesenchymal interaction in the developing GI tract.


Asunto(s)
Proliferación Celular/fisiología , Células Epiteliales/fisiología , Transición Epitelial-Mesenquimal/fisiología , Tracto Gastrointestinal/citología , Subunidades beta de Inhibinas/metabolismo , Animales , Células Cultivadas , Cartilla de ADN/genética , Células Epiteliales/metabolismo , Tracto Gastrointestinal/crecimiento & desarrollo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
Stem Cells ; 30(10): 2088-99, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22899304

RESUMEN

The transcription factor RUNX3 functions as a tumor suppressor in the gastrointestinal epithelium, where its loss is an early event in carcinogenesis. While RUNX3 acts concurrently as a mediator of TGF-ß signaling and an antagonist of Wnt, the cellular changes that follow its loss and their contribution to tumorigenicity are not fully understood. Here, we report that the loss of Runx3 in gastric epithelial cells results in spontaneous epithelial-mesenchymal transition (EMT). This produces a tumorigenic stem cell-like subpopulation, which remarkably expresses the gastric stem cell marker Lgr5. This phenomenon is due to the compounding effects of the dysregulation of the TGF-ß and Wnt pathways. Specifically, Runx3(-/-) p53(-/-) gastric epithelial cells were unexpectedly sensitized for TGF-ß-induced EMT, during which the resultant induction of Lgr5 was enhanced by an aberrantly activated Wnt pathway. These data demonstrate a protective role for RUNX3 in safeguarding gastric epithelial cells against aberrant growth factor signaling and the resultant cellular plasticity and stemness.


Asunto(s)
Transformación Celular Neoplásica/genética , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal/genética , Neoplasias Gastrointestinales/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Animales , Diferenciación Celular , Línea Celular Tumoral , Transformación Celular Neoplásica/efectos de los fármacos , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Células Epiteliales/patología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Neoplasias Gastrointestinales/genética , Neoplasias Gastrointestinales/patología , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Trasplante de Neoplasias , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transfección , Factor de Crecimiento Transformador beta/farmacología , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , beta Catenina/genética , beta Catenina/metabolismo
6.
Nat Genet ; 33(3): 375-81, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12598897

RESUMEN

The vacuolating cytotoxin VacA produced by Helicobacter pylori causes massive cellular vacuolation in vitro and gastric tissue damage in vivo, leading to gastric ulcers, when administered intragastrically. Here we report that mice deficient in protein tyrosine phosphatase receptor type Z (Ptprz, also called PTP-zeta or RPTP-beta, encoded by Ptprz) do not show mucosal damage by VacA, although VacA is incorporated into the gastric epithelial cells to the same extent as in wild-type mice. Primary cultures of gastric epithelial cells from Ptprz+/+ and Ptprz-/- mice also showed similar incorporation of VacA, cellular vacuolation and reduction in cellular proliferation, but only Ptprz+/+ cells showed marked detachment from a reconstituted basement membrane 24 h after treatment with VacA. VacA bound to Ptprz, and the levels of tyrosine phosphorylation of the G protein-coupled receptor kinase-interactor 1 (Git1), a Ptprz substrate, were higher after treatment with VacA, indicating that VacA behaves as a ligand for Ptprz. Furthermore, pleiotrophin (PTN), an endogenous ligand of Ptprz, also induced gastritis specifically in Ptprz+/+ mice when administered orally. Taken together, these data indicate that erroneous Ptprz signaling induces gastric ulcers.


Asunto(s)
Proteínas Bacterianas/fisiología , Proteínas de Ciclo Celular , Infecciones por Helicobacter/etiología , Fosfoproteínas , Proteínas Tirosina Fosfatasas/deficiencia , Úlcera Gástrica/etiología , Animales , Proteínas Bacterianas/toxicidad , Femenino , Proteínas Activadoras de GTPasa/metabolismo , Gastritis/etiología , Gastritis/patología , Infecciones por Helicobacter/patología , Helicobacter pylori/patogenicidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Fosfatasas/fisiología , Transducción de Señal , Úlcera Gástrica/patología , Virulencia
7.
Gut ; 61(3): 344-53, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21865403

RESUMEN

BACKGROUND: Gastric cancer is the second most frequent cause of death from cancer in the world, diffuse-type gastric cancer (DGC) exhibiting a poor prognosis. Germline mutations of CDH1, encoding E-cadherin, have been reported in hereditary DGC, and genetic and/or epigenetic alterations of CDH1 are frequently detected in sporadic DGC. Genetic alterations of TP53 are also frequently found in DGC. To examine the synergistic effect of the loss of E-cadherin and p53 on gastric carcinogenesis, a mouse line was established in which E-cadherin and p53 are specifically inactivated in the stomach parietal cell lineage. METHODS: Atp4b-Cre mice were crossed with Cdh1(loxP/loxP) and Trp53(loxP/loxP) mice, and the gastric phenotype of Atp4b-Cre(+);Cdh1(loxP/loxP);Trp53(loxP/loxP) double conditional knockout (DCKO) mice was examined. RESULTS: Non-polarised E-cadherin-negative parietal cells and proton pump-negative atypical foci were observed in DCKO mice. Intramucosal cancers and invasive cancers composed of poorly differentiated carcinoma cells and signet ring cells, histologically very similar to those in humans, were found from 6 to 9 months, respectively. Fatal DGC developed at 100% penetrance within a year, frequently metastasised to lymph nodes, and had tumourigenic activity in immunodeficient mice. Gene expression profiles of DGC in DCKO mice also resembled those of human DGC, and mesenchymal markers and epithelial-mesenchymal transition-related genes were highly expressed in mouse DGC as in human DGC. CONCLUSION: This mouse line is the first genetically engineered mouse model of DGC and is very useful for clarifying the mechanism underlying gastric carcinogenesis, and provides a new approach to the treatment and prevention of DGC.


Asunto(s)
Cadherinas/fisiología , Transformación Celular Neoplásica/metabolismo , Neoplasias Gástricas/metabolismo , Proteína p53 Supresora de Tumor/fisiología , Animales , Cadherinas/deficiencia , Polaridad Celular/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , ADN de Neoplasias/genética , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica/métodos , Predisposición Genética a la Enfermedad , Tolerancia Inmunológica , Metástasis Linfática , Ratones , Ratones Noqueados , Ratones Desnudos , Invasividad Neoplásica , Trasplante de Neoplasias , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Células Parietales Gástricas/metabolismo , Células Parietales Gástricas/patología , Bombas de Protones/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Proteína p53 Supresora de Tumor/deficiencia
8.
Gastroenterology ; 140(5): 1536-46.e8, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21277301

RESUMEN

BACKGROUND & AIMS: RUNX3 is a tumor suppressor originally identified in gastric cancer. The mutation R122C in RUNX3 promotes gastric carcinogenesis by unclear mechanisms. We investigated how Runx3-deficiency contributes to distinct changes in the gastric epithelium that precede neoplasia. METHODS: Runx3-deficient (Runx3(-/-)) and wild-type BALB/c adult mice were subjected to histological analyses. Gastric cancer formation after administration of N-methyl-N-nitrosourea was evaluated. Runx3(+/+) and Runx3(-/-) gastric epithelial cell lines were used to investigate the molecular basis underlying Runx3 function. RESULTS: The gastric epithelia in Runx3(-)/(-) adult mice was hyperplastic, with loss of chief cells and development of mucin 6- and trefoil factor-2-expressing metaplasia. The gastric epithelium of Runx3(-)/(-) mice had an intestinal phenotype that expressed Cdx2. After addition of N-methyl-N-nitrosourea, Runx3- mice, unlike wild-type mice, consistently developed adenocarcinomas, indicating that Runx3-deficiency leads to premalignant changes in the gastric epithelia. RUNX3, but not the RUNX3 mutant R122C, repressed Cdx2 expression by attenuation of oncogenic beta(symbol)-catenin and Tcfs. CONCLUSIONS: Runx3-deficiency leads to a precancerous state in the gastric epithelia of mice, characterized by loss of chief cells but not parietal cells; inflammation did not appear to be involved.


Asunto(s)
Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , ADN de Neoplasias/genética , Mucosa Gástrica/metabolismo , Mutación , Lesiones Precancerosas , Neoplasias Gástricas/genética , Animales , Línea Celular Tumoral , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Mucosa Gástrica/patología , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología
9.
J Reprod Dev ; 58(5): 592-8, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22785323

RESUMEN

Runx3 is a transcription factor that belongs to the Runx family. We studied the localization of Runx3 mRNA in the mouse uterus, and its function in the mouse endometrium using Runx3 knockout (Runx3(-/-)) mice. Runx3 mRNA was detected in the endometrial luminal epithelial cells, glandular epithelial cells and stromal cells below the epithelial cell layer on the luminal side. The uteri of Runx3(-/-) mice were smaller than those of wt mice. The endometrial layer and uterine glands of Runx3(-/-) mice were less developed than those of wild-type mice, and the endometrial stromal layer was thinner. Transforming growth factor ß1 and ß3 (TGFß1 and ß3) mRNA levels in endometrial stromal cells of Runx3(-/-) mice were low compared with those of wild-type mice. Estradiol-17ß (E2) increased Tgfb2 mRNA levels in endometrial stromal cells of Runx3(-/-) mice, but not in those of wild-type mice. E2 increased epidermal growth factor (EGF) mRNA levels in endometrial stromal cells of wild-type mice, but did not increase those of Runx3(-/-) mice. The diminished Tgfb1 and Tgfb3 mRNA expressions may lead to the reduced proliferation of endometrial stromal cells. Alterations of E2-associated expressions of Tgfb2 and Egf mRNA in endometrial stromal cells of Runx3(-/-) mice may be associated with suppression of E2-dependent endometrial epithelial cell proliferation in Runx3(-/-) mice. Thus, Runx3 is likely to be a regulatory factor responsible for endometrial growth.


Asunto(s)
Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Endometrio/crecimiento & desarrollo , Endometrio/metabolismo , Regulación del Desarrollo de la Expresión Génica , Animales , Proliferación Celular , Células Cultivadas , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Cruzamientos Genéticos , Endometrio/citología , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Epidérmico/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Estradiol/metabolismo , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Tamaño de los Órganos , Organogénesis , Ovario/citología , Ovario/crecimiento & desarrollo , Ovario/metabolismo , Células del Estroma/citología , Células del Estroma/metabolismo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta3/genética , Factor de Crecimiento Transformador beta3/metabolismo , Útero/citología , Útero/crecimiento & desarrollo , Útero/metabolismo
10.
Cancer Sci ; 102(5): 942-50, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21276134

RESUMEN

Alterations in the E-cadherin gene are associated with sporadic and hereditary diffuse-type gastric cancer. To determine how the loss of function of E-cadherin affects gastric epithelial cell phenotypes, we generated transgenic mice using the Cre-loxP system in which the E-cadherin gene is specifically knocked out in the parietal cell lineage. In the transgenic mice, expression of E-cadherin was lost or reduced in proton pump-expressing parietal cells, which became round in shape and were pushed out of the glands to accumulate in the stromal area. Additionally, gastric mucosa exhibited hyperplasia from 3 months in the mice, some cells of which later became positive for trefoil factor 2, a marker of spasmolytic polypeptide-expressing metaplasia. From 6 months, E-cadherin-negative/proton pump-negative cells appeared from the parietal cell lineage, which increased in number to form cell clusters. Moreover, signet ring-like cells, which are morphologically similar to signet ring carcinoma cells, were found in the cell clusters from 12 months. However, no invasive gastric adenocarcinomas were found in the E-cadherin-deficient mice, even at 24 months or later. These data indicate that the loss of E-cadherin induces possible pre-cancerous lesions in the gastric mucosa but may not be sufficient for its malignant conversion.


Asunto(s)
Cadherinas/metabolismo , Células Epiteliales/metabolismo , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patología , Lesiones Precancerosas/genética , Neoplasias Gástricas/genética , Animales , Cadherinas/genética , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Ratones , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Factor Trefoil-2
11.
Cancer Sci ; 102(7): 1313-21, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21457403

RESUMEN

CD133 is a universal marker of tissue stem/progenitor cells as well as cancer stem cells, but its physiological significance remains to be elucidated. Here we examined the relationship between expression of CD133 and features of gastric epithelial cells, and found that CD133-positive (CD133[+]) tumor cell lines formed well-differentiated tumors while CD133-negative (CD133[-]) lines formed poorly differentiated ones when subcutaneously injected into nude mice. We also found that CD133(+) and CD133(-) cell populations co-existed in some cell lines. FACS analysis showed that CD133(+) cells were mother cells because CD133(+) cells formed both CD133(+) and CD133(-) cells, but CD133(-) cells did not form CD133(+) cells. In these cell lines, CD133(+) cells formed well-differentiated tumors while CD133(-) cells formed poorly differentiated ones. In human gastric cancers, CD133 was exclusively expressed on the luminal surface membrane of gland-forming cells, and it was never found on poorly differentiated diffuse-type cells. Considering that poorly differentiated tumors often develop from well-differentiated tumors during tumor progression, these results suggest that loss of expression of CD133 might be related to gastric tumor progression. Microarray analysis showed that CD133(+) cells specifically expressed Sox17, a tumor suppressor in gastric carcinogenesis. Forced expression of SOX17 induced expression of CD133 in CD133(-) cells, and reduction of SOX17 caused by siRNA in CD133(+) cells induced a reduction in the level of CD133. These results indicate that Sox17 might be a key transcription factor controlling CD133 expression, and that it might also play a role in the control of gastric tumor progression.


Asunto(s)
Antígenos CD/fisiología , Glicoproteínas/fisiología , Proteínas HMGB/fisiología , Células Madre Neoplásicas/química , Péptidos/fisiología , Factores de Transcripción SOXF/fisiología , Neoplasias Gástricas/patología , Antígeno AC133 , Animales , Antígenos CD/análisis , Femenino , Mucosa Gástrica/patología , Perfilación de la Expresión Génica , Glicoproteínas/análisis , Proteínas HMGB/análisis , Humanos , Ratones , Péptidos/análisis , Factores de Transcripción SOXF/análisis , Neoplasias Gástricas/química
12.
Gastroenterology ; 138(1): 255-65.e1-3, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19706291

RESUMEN

BACKGROUND & AIMS: The transcription factor RUNX3 is a gastric tumor suppressor. Tumorigenic Runx3(-/-) gastric epithelial cells attach weakly to each other, compared with nontumorigenic Runx3(+/+) cells. We aimed to identify RUNX3 target genes that promote cell-cell contact to improve our understanding of RUNX3's role in suppressing gastric carcinogenesis. METHODS: We compared gene expression profiles of Runx3(+/+) and Runx3(-/-) cells and observed down-regulation of genes associated with cell-cell adhesion in Runx3(-/-) cells. Reporter, mobility shift, and chromatin immunoprecipitation assays were used to examine the regulation of these genes by RUNX3. Tumorigenesis assays and immunohistological analyses of human gastric tumors were performed to confirm the role of the candidate genes in gastric tumor development. RESULTS: Mobility shift and chromatin immunoprecipitation assays revealed that the promoter activity of the gene that encodes the tight junction protein claudin-1 was up-regulated via the binding of RUNX3 to the RUNX consensus sites. The tumorigenicity of gastric epithelial cells from Runx3(-/-) mice was significantly reduced by restoration of claudin-1 expression, whereas knockdown of claudin-1 increased the tumorigenicity of human gastric cancer cells. Concomitant expression of RUNX3 and claudin-1 was observed in human normal gastric epithelium and cancers. CONCLUSIONS: The tight junction protein claudin-1 has gastric tumor suppressive activity and is a direct transcriptional target of RUNX3. Claudin-1 is down-regulated during the epithelial-mesenchymal transition; RUNX3 might therefore act as a tumor suppressor to antagonize the epithelial-mesenchymal transition.


Asunto(s)
Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Proteínas de la Membrana/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Animales , Comunicación Celular/fisiología , Línea Celular , Línea Celular Tumoral , Claudina-1 , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Regulación hacia Abajo/fisiología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Genes Reporteros , Humanos , Proteínas de la Membrana/genética , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Estómago/patología , Uniones Estrechas/metabolismo , Trasplante Heterólogo
13.
Int J Cancer ; 127(5): 1106-14, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20020497

RESUMEN

To clarify the role of micro (mi) RNAs in gastric carcinogenesis, we studied the expression and function of miRNAs in gastric carcinoma (GC) cells. Initially, we performed microarray analysis using total RNA from 3 human GC cell lines and noncancerous gastric tissue. Among the downregulated miRNAs in GC cells, miR-212 expression was decreased in all 8 GC cell lines examined and a significant decrease of miR-212 expression in human primary GC tissues was also observed in 6 of 11 cases. Transfection of the precursor miR-212 molecule induced decreased growth of 3 GC cell lines. Using 3 different databases, methyl-CpG-binding protein MeCP2 was postulated to be a target of miR-212. As seen on reporter assaying, miR-212 repressed the construct with the MECP2 3'-UTR. Ectopic expression of miR-212 repressed expression of the MeCP2 protein but not the MECP2 mRNA level. These data suggest that downregulation of miR-212 may be related to gastric carcinogenesis through its target genes, such as MECP2.


Asunto(s)
Neoplasias Intestinales/genética , Proteína 2 de Unión a Metil-CpG/antagonistas & inhibidores , MicroARNs/genética , Neoplasias Gástricas/genética , Apoptosis , Western Blotting , Proliferación Celular , Metilación de ADN , Regulación hacia Abajo , Femenino , Humanos , Técnicas para Inmunoenzimas , Neoplasias Intestinales/metabolismo , Neoplasias Intestinales/patología , Luciferasas/metabolismo , Masculino , Proteína 2 de Unión a Metil-CpG/genética , Proteína 2 de Unión a Metil-CpG/metabolismo , MicroARNs/metabolismo , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Biosíntesis de Proteínas , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Células Tumorales Cultivadas
14.
Mol Cell Biol ; 26(12): 4474-88, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16738314

RESUMEN

Genes involved in the transforming growth factor beta (TGF-beta) signaling pathway are frequently altered in several types of cancers, and a gastric tumor suppressor RUNX3 appears to be an integral component of this pathway. We reported previously that apoptosis is notably reduced in Runx3-/- gastric epithelial cells. In the present study, we show that a proapoptotic gene Bim was transcriptionally activated by RUNX3 in the gastric cancer cell lines SNU16 and SNU719 treated with TGF-beta. The human Bim promoter contains RUNX sites, which are required for its activation. Furthermore, a dominant negative form of RUNX3 comprised of amino acids 1 to 187 increased tumorigenicity of SNU16 by inhibiting Bim expression. In Runx3-/- mouse gastric epithelium, Bim was down-regulated, and apoptosis was reduced to the same extent as that in Bim-/- gastric epithelium. We confirmed comparable expression of TGF-beta1 and TGF-beta receptors between wild-type and Runx3-/- gastric epithelia and reduction of Bim in TGF-beta1-/- stomach. These results demonstrate that RUNX3 is responsible for transcriptional up-regulation of Bim in TGF-beta-induced apoptosis.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Mucosa Gástrica/citología , Mucosa Gástrica/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transporte Activo de Núcleo Celular , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteínas Reguladoras de la Apoptosis/genética , Secuencia de Bases , Proteína 11 Similar a Bcl2 , Línea Celular Tumoral , Subunidad alfa 3 del Factor de Unión al Sitio Principal/deficiencia , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , ADN/genética , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Mucosa Gástrica/efectos de los fármacos , Humanos , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Mutagénesis Sitio-Dirigida , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética , Proteínas Smad/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Factor de Crecimiento Transformador beta/farmacología , Regulación hacia Arriba
15.
Cancer Res ; 67(9): 4311-9, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17483344

RESUMEN

Matrix metalloproteinase-2 (MMP-2) is a stroma-derived MMP belonging to the type IV collagenase family. It is believed to mediate tumor cell behavior by degrading deposits of type IV collagen, a major component of the basement membrane. The membrane type 1-MMP (MT1-MMP) is a highly potent activator of MMP-2 and is expressed in many tumor and stromal cells. However, the roles played by stromal MMP-2 in tumor progression in vivo remain poorly understood. We established a colon epithelial cell line from an Mt1-mmp(-/-) mouse strain and transfected these cells with an inducible expression system for MT1-MMP (MT1rev cells). Following s.c. implantation into Mmp-2(+/+) mice and induction of MT1-MMP expression, MT1rev cells grew rapidly, whereas they grew very slowly in Mmp-2(-/-) mice, even in the presence of MT1-MMP. This MT1-MMP-dependent tumor growth of MT1rev cells was enhanced in Mmp-2(-/-) mice as long as MMP-2 was supplied via transfection or coimplantation of MMP-2-positive fibroblasts. MT1rev cells cultured in vitro in a three-dimensional collagen gel matrix also required the MT1-MMP/MMP-2 axis for rapid proliferation. MT1rev cells deposit type IV collagen primarily at the cell-collagen interface, and these deposits seem scarce at sites of invasion and proliferation. These data suggest that cooperation between stroma-derived MMP-2 and tumor-derived MT1-MMP may play a role in tumor invasion and proliferation via remodeling of the tumor-associated basement membrane. To our knowledge, this is the first study demonstrating that MT1-MMP-dependent tumor growth in vivo requires stromal-derived MMP-2. It also suggests that MMP-2 represents a potential target for tumor therapeutics.


Asunto(s)
Metaloproteinasa 14 de la Matriz/fisiología , Metaloproteinasa 2 de la Matriz/fisiología , Neoplasias Experimentales/enzimología , Neoplasias Experimentales/patología , Animales , Procesos de Crecimiento Celular , Colágeno Tipo I/metabolismo , Colágeno Tipo IV/biosíntesis , Colágeno Tipo IV/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Metaloproteinasa 14 de la Matriz/biosíntesis , Metaloproteinasa 14 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 2 de la Matriz/genética , Ratones , Neoplasias Experimentales/genética , Células del Estroma/enzimología , Células del Estroma/patología , Transducción Genética , Transfección
16.
PLoS One ; 14(8): e0221880, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31465502

RESUMEN

The potential of genomic selection (GS) is currently being evaluated for fruit breeding. GS models are usually constructed based on information from both the genotype and phenotype of population. However, information from phenotyped but non-genotyped relatives can also be used to construct GS models, and this additional information can improve their accuracy. In the present study, we evaluated the utility of single-step genomic best linear unbiased prediction (ssGBLUP) in citrus breeding, which is a genomic prediction method that combines the kinship information from genotyped and non-genotyped relatives into a single relationship matrix for a mixed model to apply GS. Fruit weight, sugar content, and acid content of 1,935 citrus individuals, of which 483 had genotype data of 2,354 genome-wide single nucleotide polymorphisms, were evaluated from 2009-2012. The prediction accuracy of ssGBLUP for genotyped individuals was similar to or higher than that of usual genomic best linear unbiased prediction method using only genotyped individuals, especially for sugar content. Therefore, ssGBLUP could yield higher accuracy in genotyped individuals by adding information from non-genotyped relatives. The prediction accuracy of ssGBLUP for non-genotyped individuals was also slightly higher than that of conventional best linear unbiased prediction method using pedigree information. This indicates that ssGBLUP can enhance prediction accuracy of breeding values for non-genotyped individuals using genomic information of genotyped relatives. These results demonstrate the potential of ssGBLUP for fruit breeding, including citrus.


Asunto(s)
Citrus/genética , Genoma de Planta , Genómica , Genotipo , Fenotipo , Algoritmos , Genómica/métodos , Modelos Genéticos , Sitios de Carácter Cuantitativo , Carácter Cuantitativo Heredable , Reproducibilidad de los Resultados
17.
J Exp Clin Cancer Res ; 38(1): 127, 2019 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-30866995

RESUMEN

BACKGROUND: Mechanistic target of rapamycin (mTOR) pathway is essential for the growth of gastric cancer (GC), but mTOR inhibitor everolimus was not effective for the treatment of GCs. The Cancer Genome Atlas (TCGA) researchers reported that most diffuse-type GCs were genomically stable (GS). Pathological analysis suggested that some diffuse-type GCs developed from intestinal-type GCs. METHODS: We established patient-derived xenograft (PDX) lines from diffuse-type GCs, and searched for drugs that suppressed their growth. Diffuse-type GCs were classified into subtypes by their gene expression profiles. RESULTS: mTOR inhibitor temsirolimus strongly suppressed the growth of PDX-derived diffuse-type GC-initiating cells, which was regulated via Wnt-mTOR axis. These cells were microsatellite unstable (MSI) or chromosomally unstable (CIN), inconsistent with TCGA report. Diffuse-type GCs in TCGA cohort could be classified into two clusters, and GS subtype was major in cluster I while CIN and MSI subtypes were predominant in cluster II where PDX-derived diffuse-type GC cells were included. We estimated that about 9 and 55% of the diffuse-type GCs in cluster II were responders to mTOR inhibitors and checkpoint inhibitors, respectively, by identifying PIK3CA mutations and MSI condition in TCGA cohort. These ratios were far greater than those of diffuse-type GCs in cluster I or intestinal-type GCs. Further analysis suggested that diffuse-type GCs in cluster II developed from intestinal-type GCs while those in cluster I from normal gastric epithelial cells. CONCLUSION: mTOR inhibitors and checkpoint inhibitors might be useful for the treatment of a subset of diffuse-type GCs which may develop from intestinal-type GCs.


Asunto(s)
Neoplasias Gástricas/tratamiento farmacológico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Humanos , Ratones , Inestabilidad de Microsatélites , Neoplasias Gástricas/patología
18.
Int J Cancer ; 122(5): 1040-6, 2008 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-17957782

RESUMEN

Invasion and metastases of cancer cells are the main causes of treatment failure in cancer. IQ motif-containing GTPase activating protein 1 (IQGAP1), plays pivotal roles in intercellular adhesion, migration, invasion and metastases in various cancer cells. However, the role of another family member, IQGAP2, in carcinogenesis remains unknown. Here, we investigated IQGAP2 functions in gastric cancers. We found that IQGAP2 protein expression was lost in 5 of the 9 gastric cancer cell lines. Through analysis by the methylation-specific PCR, aberrant IQGAP2 methylation was detected in 3 gastric cancer cell lines. IQGAP2 mRNA was found to be activated after 5-aza-2'-deoxycytidine treatment of the methylation-positive cells. Moreover, IQGAP2 methylation was detected in 28 of the 59 (47%) primary gastric cancer tissues, but not in 12 normal gastric mucosa samples. Immunohistochemical staining revealed that 7 of the 8 (88%) gastric cancer tissues without methylation signals displayed IQGAP2 expression, whereas among 10 with methylation signals none expressed IQGAP2 (p = 0.0002), indicating that IQGAP2 methylation is highly associated with loss of the IQGAP2 expression in the primary gastric cancer tissues as well as gastric cancer cell lines. Furthermore, IQGAP2 methylation was also associated with tumor invasion and a poor prognosis. IQGAP2 knockdown with small interfering RNA increased the invasive capacity of a gastric cancer cell line. These results suggest that silencing of IQGAP2 by promoter methylation may contribute to gastric cancer development.


Asunto(s)
Metilación de ADN , Invasividad Neoplásica/genética , Regiones Promotoras Genéticas/genética , Neoplasias Gástricas/genética , Proteínas Activadoras de ras GTPasa/genética , Línea Celular Tumoral , Femenino , Técnica del Anticuerpo Fluorescente , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Gástricas/mortalidad , Neoplasias Gástricas/patología , Transfección , Proteínas Activadoras de ras GTPasa/metabolismo
19.
Cancer Sci ; 99(4): 671-6, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18377419

RESUMEN

We have reported that a lack of RUNX3 function is causally associated with gastric carcinogenesis. We have also presented evidence that loss of Runx3 may be related to the genesis of intestinal metaplasia because expression of RUNX3 is reduced in some intestinal metaplasias, and some Runx3(-/-)p53(-/-) gastric epithelial cells differentiate into intestinal type cells in vivo. Recently several reports have indicated that blood cells play important roles in the gastric carcinogenesis. In the present study, we therefore examined whether Runx3(-/-)p53(-/-) gastric epithelial cells differentiate autonomously into intestinal type cells, or whether the presence of other cells is necessary for the differentiation in vitro. When Runx3(-/-)p53(-/-) gastric epithelial cells were cultured with collagen gels, they did not exhibit any morphogenesis and differentiated poorly. When cultured with fetal mouse gastric mesenchymes, the cells formed glandular structures and differentiated into surface mucous cells, but differentiation of intestinal type cells was never observed. When cultured with Matrigel, the cells formed glandular structures, and some cells differentiated into intestinal type cells in vitro. Reverse transcription-polymerase chain reaction analysis showed that the cells expressed stomach-specific genes, and their levels decreased gradually during the culture. The cells expressed some intestine-specific genes weakly at the start of culture, and their levels were increased with time in culture. We therefore conclude that Runx3(-/-)p53(-/-) gastric epithelial cells differentiate into intestinal type cells in combination with Matrigel in the absence of other cell types. Extracellular matrix, not blood cells, may play a role in the genesis of intestinal metaplasia.


Asunto(s)
Diferenciación Celular , Subunidad alfa 3 del Factor de Unión al Sitio Principal/fisiología , Mucosa Gástrica/citología , Mucosa Intestinal/citología , Neoplasias Gástricas/etiología , Proteína p53 Supresora de Tumor/fisiología , Animales , Diferenciación Celular/genética , Línea Celular , Colágeno/química , Colágeno/metabolismo , Colágeno/farmacología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/análisis , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Combinación de Medicamentos , Matriz Extracelular/genética , Matriz Extracelular/ultraestructura , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/fisiología , Expresión Génica , Mucosa Intestinal/fisiología , Laminina/química , Laminina/metabolismo , Laminina/farmacología , Ratones , Técnicas de Cultivo de Órganos , Proteoglicanos/química , Proteoglicanos/metabolismo , Proteoglicanos/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Proteína p53 Supresora de Tumor/análisis , Proteína p53 Supresora de Tumor/genética
20.
Mol Reprod Dev ; 75(11): 1653-61, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18361415

RESUMEN

Runx3 is a transcription factor that belongs to the Runx family. We studied the function of Runx3 in the mouse ovary and uterus using the Runx3 knockout (Runx3(-/-)) mouse. Ovaries were collected from 8-week-old wild type (wt) and Runx3(-/-) mice. Histological studies showed that follicles were present at various developmental stages in the Runx3(-/-) and wt mouse ovaries. The numbers of primary, preantral and antral follicles in the Runx3(-/-) mice were significantly less than those in the wt mice while the number of primordial follicles in the Runx3(-/-) mice was not significantly different from that in the wt mice. Corpora lutea were not detected in the Runx3(-/-) mouse ovary. Gonadotropin treatment in immature female mice induced ovulation in Runx3(-/-) mice as well as in wt mice, indicating that ovaries of Runx3(-/-) mice respond to gonadotropin treatment as those in wt mouse ovaries. This suggests that failure of ovulation is due to dysfunction of regulatory mechanism of gonadotropin secretion. In addition, the uteri of Runx3(-/-) mice were atrophic, showed thin epithelial layers compared with those of the wt mice, and did not respond to estrogen in terms of DNA replication in endometrial epithelial cells. These results suggest that Runx3 takes part in the regulation of reproductive functions.


Asunto(s)
Proliferación Celular , Subunidad alfa 3 del Factor de Unión al Sitio Principal/deficiencia , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Endometrio/citología , Células Epiteliales/citología , Estrógenos/fisiología , Ovulación/genética , Animales , Atrofia/genética , Células Cultivadas , Subunidad alfa 3 del Factor de Unión al Sitio Principal/fisiología , Endometrio/patología , Células Epiteliales/patología , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Folículo Ovárico/patología , Folículo Ovárico/fisiología , Útero/metabolismo , Útero/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA