Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell Biol Int ; 45(6): 1296-1305, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33739578

RESUMEN

The prognosis of advanced colorectal cancer (CRC) is currently still very poor, which suggests that the biological mechanisms of CRC oncogenesis are not fully understood. This study was conducted to explore the regulatory effect of SOX-17 on the expression of microRNA (miR)-302b-3p, and the involvement of SOX-17 in the invasion and apoptosis of CRC cells. The expression of SOX-17 and miR-302a,b,c,d-3p in colorectal cancer and normal colon epithelial cell lines was measured by real-time polymerase chain reaction and/or western blot. The regulatory effects of SOX-17 on miR-302b-3p gene in HT29 and LoVo cells were tested using the ChiP assay. The biological activities of SOX-17 and miR-302b-3p were evaluated by invasion and apoptosis assay. Results showed that transfection of SOX-17 small interfering RNA (siSOX-17) significantly increased, whereas transfection of SOX-17 overexpression vector (oeSOX-17) significantly decreased, miR-302b expression in HT29 and LoVo cells. Cotransfection of oeSOX-17 and miR-302b-3p inhibitor (INmiR-302b) significantly blocked the effects of SOX-17 in HT29 and LoVo cells. ChIP experiments showed that SOX-17 bonded to the miR-302b-3p promoter in HT29 and LoVo cells. Transfection of oeSOX-17 and miR-302b-3p mimics (MImiR-302b) significantly decreased, whereas transfection of siSOX-17 and INmiR-302b significantly increased, the invasion of HT29 and LoVo cells. In contrast, transfection of oeSOX-17 and MImiR-302b significantly increased, while transfection of siSOX-17 and INmiR-302b significantly decreased, apoptosis in HT29 and LoVo cells. Cotransfection of oeSOX-17 and INmiR-302b significantly blocked the effects of oeSOX-17 on cell invasion and apoptosis in HT29 and LoVo cells. These results suggested that SOX-17 can bind to the promoter of miR-302b-3p gene to regulate its expression, while both SOX-17 and miR-302b regulate the invasion and apoptosis in colorectal cancer cells.


Asunto(s)
Neoplasias Colorrectales/metabolismo , MicroARNs/fisiología , Factores de Transcripción SOXF/fisiología , Apoptosis , Línea Celular Tumoral , Movimiento Celular , Regulación Neoplásica de la Expresión Génica , Humanos
2.
Surg Today ; 51(3): 340-349, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32754843

RESUMEN

Organ liver transplantation and hepatocyte transplantation are not performed to their full potential because of donor shortage, which could be resolved by identifying new donor sources for the development of hepatocyte-like cells (HLCs). HLCs have been differentiated from some stem cell sources as alternative primary hepatocytes throughout the world; however, the currently available techniques cannot differentiate HLCs to the level of normal adult primary hepatocytes. The outstanding questions are as follows: which stem cells are the best cell sources? which protocol is the best way to differentiate them into HLCs? what is the definition of differentiated HLCs? how can we enforce the function of HLCs? what is the difference between HLCs and primary hepatocytes? what are the problems with HLC transplantation? This review summarizes the current status of HLCs, focusing on stem cell sources, the differentiation protocol for HLCs, the general characterization of HLCs, the generation of more functional HLCs, comparison with primary hepatocytes, and HLCs in cell-transplantation-based liver regeneration.


Asunto(s)
Diferenciación Celular , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Técnicas Citológicas/métodos , Hepatocitos/trasplante , Hepatopatías/terapia , Células Madre/fisiología , Proteínas Morfogenéticas Óseas/fisiología , Diferenciación Celular/genética , Células Cultivadas , Técnicas de Cocultivo , Factores de Crecimiento de Fibroblastos/fisiología , Factor Nuclear 4 del Hepatocito/fisiología , Proteínas de Homeodominio/fisiología , Humanos , Regeneración Hepática/fisiología , Factores de Transcripción SOXF/fisiología , Factores de Transcripción/fisiología
3.
Development ; 144(10): 1887-1895, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28512199

RESUMEN

SOX family proteins SOX2 and SOX18 have been reported as being essential in determining hair follicle type; however, the role they play during development remains unclear. Here, we demonstrate that Sox18 regulates the normal differentiation of the dermal papilla of all hair types. In guard (primary) hair dermal condensate (DC) cells, we identified transient Sox18 in addition to SOX2 expression at E14.5, which allowed fate tracing of primary DC cells until birth. Similarly, expression of Sox18 was detected in the DC cells of secondary hairs at E16.5 and in tertiary hair at E18.5. Dominant-negative Sox18 mutation (opposum) did not prevent DC formation in any hair type. However, it affected dermal papilla differentiation, restricting hair formation especially in secondary and tertiary hairs. This Sox18 mutation also prevented neonatal dermal cells or dermal papilla spheres from inducing hair in regeneration assays. Microarray expression studies identified WNT5A and TNC as potential downstream effectors of SOX18 that are important for epidermal WNT signalling. In conclusion, SOX18 acts as a mesenchymal molecular switch necessary for the formation and function of the dermal papilla in all hair types.


Asunto(s)
Diferenciación Celular/genética , Folículo Piloso/embriología , Cabello/embriología , Factores de Transcripción SOXF/fisiología , Animales , Dermis/embriología , Dermis/metabolismo , Embrión de Mamíferos , Células Epidérmicas , Epidermis/embriología , Femenino , Genes Dominantes , Genes de Cambio/fisiología , Cabello/metabolismo , Folículo Piloso/metabolismo , Masculino , Ratones , Ratones Transgénicos , Factores de Transcripción SOXF/genética
4.
Prostate ; 79(4): 370-378, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30488457

RESUMEN

BACKGROUND: PSMA expression in the prostate epithelium is controlled by a cis-element, PSMA enhancer (PSME). PSME contains multiple binding sites for Sox proteins, and in this study, we identified Sox7 protein as a negative regulator of PSMA expression through its interaction with PSME. METHODS: The statistical correlation between Sox7 and PSMA mRNA expression was evaluated using five prostate cancer studies from cBioportal. In vitro and in vivo interaction between Sox7 and PSME was evaluated by chromatin immunoprecipitation (ChIP), electrophoretic mobility shift assay (EMSA), and luciferase reporter assay. Synthetic oligonucleotides were generated to define the sites in PSME that interact with Sox7 protein. Sox7 mutants were generated to identify the region of this protein required to regulate PSMA expression. Sox7 was also stably expressed in LNCaP/C4-2 and 22Rv1 cells to validate the regulation of PSMA expression by Sox7 in vivo. RESULTS: Sox7 mRNA expression negatively correlated with PSMA/FOLH1 and PSMAL/FOLH1B mRNA expression in Broad/Cornell, TCGA and MSKCC studies, but not in two studies containing only metastatic prostate tumors. PC-3 cells mostly expressed the 48.5 KDa isoform 2 of Sox7, and the depletion of this isoform did not restore PSMA expression. Ectopic expression of canonical, wild-type Sox7 in C4-2 and 22Rv1 cells suppressed PSMA protein expression. ChIP assay revealed that canonical Sox7 protein preferentially interacts with PSME in vivo, and EMSA identified the SOX box sites #2 and #4 in PSME as required for its interaction. Sox7 was capable of directly binding to PSME and suppressed PSME-mediated transcription. The NLS regions of Sox7, but not its ß-catenin interacting motif, are essential for this suppressing activity. Furthermore, restoration of wild-type Sox7 expression but not Sox7-NLS mutant in Sox7-null prostate cancer cell lines suppressed PSMA expression. CONCLUSIONS: The inactivation of canonical Sox7 is responsible for the upregulated expression of PSMA in non-metastatic prostate cancer.


Asunto(s)
Antígenos de Superficie/genética , Elementos de Facilitación Genéticos/fisiología , Regulación Neoplásica de la Expresión Génica/fisiología , Glutamato Carboxipeptidasa II/genética , Próstata/metabolismo , Neoplasias de la Próstata/metabolismo , Factores de Transcripción SOXF/fisiología , Secuencia de Aminoácidos , Secuencia de Bases , Sitios de Unión , Línea Celular Tumoral , Humanos , Masculino , Neoplasias de la Próstata/química , ARN Mensajero/análisis , Factores de Transcripción SOXF/química , Vía de Señalización Wnt/fisiología
5.
Circulation ; 135(25): 2505-2523, 2017 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-28381471

RESUMEN

BACKGROUND: The mechanisms underlying the dedifferentiation and lineage conversion of adult human fibroblasts into functional endothelial cells have not yet been fully defined. Furthermore, it is not known whether fibroblast dedifferentiation recapitulates the generation of multipotent progenitors during embryonic development, which give rise to endothelial and hematopoietic cell lineages. Here we established the role of the developmental transcription factor SOX17 in regulating the bilineage conversion of fibroblasts by the generation of intermediate progenitors. METHODS: CD34+ progenitors were generated after the dedifferentiation of human adult dermal fibroblasts by overexpression of pluripotency transcription factors. Sorted CD34+ cells were transdifferentiated into induced endothelial cells and induced erythroblasts using lineage-specific growth factors. The therapeutic potential of the generated cells was assessed in an experimental model of myocardial infarction. RESULTS: Induced endothelial cells expressed specific endothelial cell surface markers and also exhibited the capacity for cell proliferation and neovascularization. Induced erythroblasts expressed erythroid surface markers and formed erythroid colonies. Endothelial lineage conversion was dependent on the upregulation of the developmental transcription factor SOX17, whereas suppression of SOX17 instead directed the cells toward an erythroid fate. Implantation of these human bipotential CD34+ progenitors into nonobese diabetic/severe combined immunodeficiency (NOD-SCID) mice resulted in the formation of microvessels derived from human fibroblasts perfused with mouse and human erythrocytes. Endothelial cells generated from human fibroblasts also showed upregulation of telomerase. Cell implantation markedly improved vascularity and cardiac function after myocardial infarction without any evidence of teratoma formation. CONCLUSIONS: Dedifferentiation of fibroblasts to intermediate CD34+ progenitors gives rise to endothelial cells and erythroblasts in a SOX17-dependent manner. These findings identify the intermediate CD34+ progenitor state as a critical bifurcation point, which can be tuned to generate functional blood vessels or erythrocytes and salvage ischemic tissue.


Asunto(s)
Antígenos CD34/fisiología , Desdiferenciación Celular/fisiología , Células Endoteliales/fisiología , Eritroblastos/fisiología , Fibroblastos/fisiología , Factores de Transcripción SOXF/fisiología , Células Madre/fisiología , Animales , Células Cultivadas , Humanos , Recién Nacido , Ratones , Ratones Endogámicos NOD , Ratones SCID
6.
Biol Reprod ; 99(3): 578-589, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29635272

RESUMEN

In mouse conceptus, two yolk-sac membranes, the parietal endoderm (PE) and visceral endoderm (VE), are involved in protecting and nourishing early-somite-stage embryos prior to the establishment of placental circulation. Both PE and VE membranes are tightly anchored to the marginal edge of the developing placental disk, in which the extraembryonic endoderm (marginal zone endoderm: ME) shows the typical flat epithelial morphology intermediate between those of PE and VE in vivo. However, the molecular characteristics and functions of the ME in mouse placentation remain unclear. Here, we show that SOX17, not SOX7, is continuously expressed in the ME cells, whereas both SOX17 and SOX7 are coexpressed in PE cells, by at least 10.5 days postconception. The Sox17-null conceptus, but not the Sox7-null one, showed the ectopic appearance of squamous VE-like epithelial cells in the presumptive ME region, together with reduced cell density and aberrant morphology of PE cells. Such aberrant ME formation in the Sox17-null extraembryonic endoderm was not rescued by the chimeric embryo replaced with the wild-type gut endoderm by the injection of wild-type ES cells into the Sox17-null blastocyst, suggesting the cell autonomous defects in the extraembryonic endoderm of Sox17-null concepti. These findings provide direct evidence of the crucial roles of SOX17 in proper formation and maintenance of the ME region, highlighting a novel entry point to understand the in vivo VE-to-PE transition in the marginal edge of developing placenta.


Asunto(s)
Desarrollo Embrionario/fisiología , Endodermo/fisiología , Proteínas HMGB/fisiología , Placentación/fisiología , Factores de Transcripción SOXF/fisiología , Saco Vitelino/fisiología , Animales , Proliferación Celular , Femenino , Expresión Génica , Genotipo , Proteínas HMGB/deficiencia , Proteínas HMGB/genética , Masculino , Ratones , Ratones Noqueados , Embarazo , Factores de Transcripción SOXF/deficiencia , Factores de Transcripción SOXF/genética
7.
Circ Res ; 119(7): 839-52, 2016 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-27528602

RESUMEN

RATIONALE: Vascular endothelial growth factor (VEGF) signaling is a key pathway for angiogenesis and requires highly coordinated regulation. Although the Notch pathway-mediated suppression of excessive VEGF activity via negative feedback is well known, the positive feedback control for augmenting VEGF signaling remains poorly understood. Transcription factor Sox17 is indispensable for angiogenesis, but its association with VEGF signaling is largely unknown. The contribution of other Sox members to angiogenesis also remains to be determined. OBJECTIVE: To reveal the genetic interaction of Sox7, another Sox member, with Sox17 in developmental angiogenesis and their functional relationship with VEGF signaling. METHODS AND RESULTS: Sox7 is expressed specifically in endothelial cells and its global and endothelial-specific deletion resulted in embryonic lethality with severely impaired angiogenesis in mice, substantially overlapping with Sox17 in both expression and function. Interestingly, compound heterozygosity for Sox7 and Sox17 phenocopied vascular defects of Sox7 or Sox17 homozygous knockout, indicating that the genetic cooperation of Sox7 and Sox17 is sensitive to their combined gene dosage. VEGF signaling upregulated both Sox7 and Sox17 expression in angiogenesis via mTOR pathway. Furthermore, Sox7 and Sox17 promoted VEGFR2 (VEGF receptor 2) expression in angiogenic vessels, suggesting a positive feedback loop between VEGF signaling and SoxF. CONCLUSIONS: Our findings demonstrate that SoxF transcription factors are indispensable players in developmental angiogenesis by acting as positive feedback regulators of VEGF signaling.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/metabolismo , Neovascularización Fisiológica/fisiología , Factores de Transcripción SOXF/fisiología , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Técnicas de Cultivo , Femenino , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Embarazo
8.
Dev Biol ; 414(2): 219-27, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27102016

RESUMEN

The importance of canonical Wnt signaling to murine uterine development is well established. Mouse models in which uterine-specific Wnt ligands, ß-catenin, or Lef1 are disrupted result in failure of postnatal endometrial gland development. Sox17 is a transcription factor characterized in numerous tissues as an antagonist of Wnt signaling. Thus, we hypothesized that conditional ablation of Sox17 would lead to hyperproliferation of endometrial glands in mice. Contrary to our prediction, disruption of Sox17 in epithelial and stromal compartments led to inhibition of endometrial adenogenesis and a loss of reproductive capacity. Epithelium-specific Sox17 disruption resulted in normal adenogenesis although reproductive capacity remained impaired. These findings suggest that non-epithelial, Sox17-positive cells are necessary for adenogenesis and that glands require Sox17 to properly function. To our knowledge, these findings are the first to implicate Sox17 in endometrial gland formation and reproductive success. The data presented herein underscore the importance of studying Sox17 in uterine homeostasis and function.


Asunto(s)
Endometrio/crecimiento & desarrollo , Células Epiteliales/fisiología , Glándulas Exocrinas/crecimiento & desarrollo , Proteínas HMGB/fisiología , Factores de Transcripción SOXF/fisiología , Animales , Endometrio/metabolismo , Endometrio/patología , Glándulas Exocrinas/metabolismo , Femenino , Proteínas HMGB/deficiencia , Proteínas HMGB/genética , Factor Nuclear 3-beta del Hepatocito/biosíntesis , Factor Nuclear 3-beta del Hepatocito/genética , Homeostasis , Hiperplasia , Infertilidad Femenina/genética , Infertilidad Femenina/patología , Proteínas Luminiscentes/análisis , Factor de Unión 1 al Potenciador Linfoide/biosíntesis , Factor de Unión 1 al Potenciador Linfoide/genética , Ratones , Ratones Noqueados , Embarazo , Factores de Transcripción SOXF/deficiencia , Factores de Transcripción SOXF/genética , Organismos Libres de Patógenos Específicos , Células del Estroma/fisiología , Proteínas Wnt/biosíntesis , Proteínas Wnt/genética , Vía de Señalización Wnt/fisiología
9.
J Hepatol ; 67(1): 72-83, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28237397

RESUMEN

BACKGROUND & AIMS: Cholangiocarcinoma (CCA) is a biliary malignancy linked to genetic and epigenetic abnormalities, such as hypermethylation of SOX17 promoter. Here, the role of SOX17 in cholangiocyte differentiation and cholangiocarcinogenesis was studied. METHODS: SOX17 expression/function was evaluated along the differentiation of human induced pluripotent stem cells (iPSC) into cholangiocytes, in the dedifferentiation process of normal human cholangiocytes (NHC) in culture and in cholangiocarcinogenesis. Lentiviruses for SOX17 overexpression or knockdown were used. Gene expression and DNA methylation profiling were performed. RESULTS: SOX17 expression is induced in the last stage of cholangiocyte differentiation from iPSC and regulates the acquisition of biliary markers. SOX17 becomes downregulated in NHC undergoing dedifferentiation; experimental SOX17 knockdown in differentiated NHC downregulated biliary markers and promoted baseline and Wnt-dependent proliferation. SOX17 expression is lower in human CCA than in healthy tissue, which correlates with worse survival after tumor resection. In CCA cells, SOX17 overexpression decreased their tumorigenic capacity in murine xenograft models, which was related to increased oxidative stress and apoptosis. In contrast, SOX17 overexpression in NHC did not affect their survival but inhibited their baseline proliferation. In CCA cells, SOX17 inhibited migration, anchorage-independent growth and Wnt/ß-catenin-dependent proliferation, and restored the expression of biliary markers and primary cilium length. In human CCA, SOX17 promoter was found hypermethylated and its expression inversely correlates with the methylation grade. In NHC, Wnt3a decreased SOX17 expression in a DNMT-dependent manner, whereas in CCA, DNMT1 inhibition or silencing upregulated SOX17. CONCLUSIONS: SOX17 regulates the differentiation and maintenance of the biliary phenotype and functions as a tumor suppressor for CCA, being a potential prognostic marker and a promising therapeutic target. LAY SUMMARY: Understanding the molecular mechanisms involved in the pathogenesis of CCA is key in finding new valuable diagnostic and prognostic biomarkers, as well as therapeutic targets. This study provides evidence that SOX17 regulates the differentiation and maintenance of the biliary phenotype, and its downregulation promotes their tumorigenic transformation. SOX17 acts as a tumor suppressor in CCA and its genetic, molecular and/or pharmacological restoration may represent a new promising therapeutic strategy. Moreover, SOX17 expression correlates with the outcome of patients after tumor resection, being a potential prognostic biomarker.


Asunto(s)
Neoplasias de los Conductos Biliares/etiología , Conductos Biliares/patología , Colangiocarcinoma/etiología , Factores de Transcripción SOXF/fisiología , Proteínas Supresoras de Tumor/fisiología , Animales , Diferenciación Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Factores de Transcripción SOXF/análisis , Factores de Transcripción SOXF/genética
10.
Circulation ; 131(11): 995-1005, 2015 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-25596186

RESUMEN

BACKGROUND: Intracranial aneurysm (IA) is a common vascular disorder that frequently leads to fatal vascular rupture. Although various acquired risk factors associated with IA have been identified, the hereditary basis of IA remains poorly understood. As a result, genetically modified animals accurately modeling IA and related pathogenesis have been lacking, and subsequent drug development has been delayed. METHODS AND RESULTS: The transcription factor Sox17 is robustly expressed in endothelial cells of normal intracerebral arteries. The combination of Sox17 deficiency and angiotensin II infusion in mice induces vascular abnormalities closely resembling the cardinal features of IA such as luminal dilation, wall thinning, tortuosity, and subarachnoid hemorrhages. This combination impairs junctional assembly, cell-matrix adhesion, regeneration capacity, and paracrine secretion in endothelial cells of intracerebral arteries, highlighting key endothelial dysfunctions that lead to IA pathogenesis. Moreover, human IA samples showed reduced Sox17 expression and impaired endothelial integrity, further strengthening the applicability of this animal model to clinical settings. CONCLUSIONS: Our findings demonstrate that Sox17 deficiency in mouse can induce IA under hypertensive conditions, suggesting Sox17 deficiency as a potential genetic factor for IA formation. The Sox17-deficient mouse model provides a novel platform to develop therapeutics for incurable IA.


Asunto(s)
Endotelio Vascular/patología , Proteínas HMGB/deficiencia , Aneurisma Intracraneal/genética , Factores de Transcripción SOXF/deficiencia , Factores de Transcripción SOXF/fisiología , Adulto , Anciano , Angiotensina II/toxicidad , Animales , Aorta/patología , Células Cultivadas , Arterias Cerebrales/química , Arterias Cerebrales/patología , Proteínas Inhibidoras de las Quinasas Dependientes de la Ciclina/biosíntesis , Proteínas Inhibidoras de las Quinasas Dependientes de la Ciclina/genética , Dilatación Patológica/genética , Dilatación Patológica/patología , Modelos Animales de Enfermedad , Endotelio Vascular/metabolismo , Femenino , Proteínas HMGB/genética , Proteínas HMGB/fisiología , Humanos , Hipertensión/complicaciones , Aneurisma Intracraneal/etiología , Aneurisma Intracraneal/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Miocitos del Músculo Liso/química , Comunicación Paracrina , Interferencia de ARN , Factores de Transcripción SOXF/análisis , Factores de Transcripción SOXF/genética , Organismos Libres de Patógenos Específicos , Hemorragia Subaracnoidea/etiología , Transcripción Genética , Regulación hacia Arriba , Venas/química
11.
Tumour Biol ; 37(9): 11753-11762, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27022736

RESUMEN

Interleukin-22 (IL-22) is an inflammatory cytokine mainly produced by activated Th17 and Th22 cells. The data presented here demonstrate that IL-22 induced the migration and invasion of papillary thyroid cancer (PTC) cells. MicroRNA expression analysis and functional studies indicated that IL-22-mediated migration and invasion is positively regulated by miR-595. Further mechanistic studies revealed that sex-determining region Y-box 17 (Sox17) is directly targeted by miR-595. We then demonstrated that IL-22 regulated migration and invasion of PTC cells via inhibiting Sox17 expression. Interestingly, in PTC cell lines and PTC tissues, expression of IL-22 and miR-595 was upregulated and Sox17 downregulated compared with normal thyroid, and their expression levels were closely correlated. Taken together, this present study suggests that IL-22 stimulation enhances the migration and invasion of PTC cells by regulating miR-595 and its target Sox17.


Asunto(s)
Carcinoma/patología , Interleucinas/farmacología , MicroARNs/fisiología , Factores de Transcripción SOXF/fisiología , Neoplasias de la Tiroides/patología , Carcinoma Papilar , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Humanos , Invasividad Neoplásica , Factores de Transcripción SOXF/genética , Cáncer Papilar Tiroideo , Interleucina-22
12.
Circ Res ; 115(2): 215-26, 2014 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-24755984

RESUMEN

RATIONALE: The Notch pathway stabilizes sprouting angiogenesis by favoring stalk cells over tip cells at the vascular front. Because tip and stalk cells have different properties in morphology and function, their transcriptional regulation remains to be distinguished. Transcription factor Sox17 is specifically expressed in endothelial cells, but its expression and role at the vascular front remain largely unknown. OBJECTIVE: To specify the role of Sox17 and its relationship with the Notch pathway in sprouting angiogenesis. METHODS AND RESULTS: Endothelial-specific Sox17 deletion reduces sprouting angiogenesis in mouse embryonic and postnatal vascular development, whereas Sox17 overexpression increases it. Sox17 promotes endothelial migration by destabilizing endothelial junctions and rearranging cytoskeletal structure and upregulates expression of several genes preferentially expressed in tip cells. Interestingly, Sox17 expression is suppressed in stalk cells in which Notch signaling is relatively high. Notch activation by overexpressing Notch intracellular domain reduces Sox17 expression both in primary endothelial cells and in retinal angiogenesis, whereas Notch inhibition by delta-like ligand 4 (Dll4) blockade increases it. The Notch pathway regulates Sox17 expression mainly at the post-transcriptional level. Furthermore, endothelial Sox17 ablation rescues vascular network from excessive tip cell formation and hyperbranching under Notch inhibition in developmental and tumor angiogenesis. CONCLUSIONS: Our findings demonstrate that the Notch pathway restricts sprouting angiogenesis by reducing the expression of proangiogenic regulator Sox17.


Asunto(s)
Células Endoteliales/metabolismo , Proteínas HMGB/fisiología , Neovascularización Patológica/fisiopatología , Neovascularización Fisiológica/fisiología , Receptores Notch/fisiología , Factores de Transcripción SOXF/fisiología , Transducción de Señal/fisiología , Animales , Carcinoma Pulmonar de Lewis/irrigación sanguínea , Diferenciación Celular , Movimiento Celular , Citoesqueleto/ultraestructura , Embrión de Mamíferos/irrigación sanguínea , Células Madre Embrionarias , Regulación de la Expresión Génica , Proteínas HMGB/biosíntesis , Proteínas HMGB/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Uniones Intercelulares/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Morfogénesis/genética , Estructura Terciaria de Proteína , ARN Interferente Pequeño/farmacología , Receptor Notch1/genética , Receptor Notch1/fisiología , Proteínas Recombinantes de Fusión , Vasos Retinianos/crecimiento & desarrollo , Factores de Transcripción SOXF/biosíntesis , Factores de Transcripción SOXF/genética , Organismos Libres de Patógenos Específicos , Transcripción Genética
13.
Blood ; 121(3): 417-8, 2013 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-23329691

RESUMEN

In this issue of Blood, Nakajima-Takagi et al use human embryonic stem (ES) and induced pluripotent stem (iPS) cells to identify Sox17 as a regulator of hemogenic endothelium, and use conditional expression of Sox17 to capture endothelial cells at the critical moment of hemogeneic transition.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Factores de Transcripción SOXF/genética , Factores de Transcripción SOXF/fisiología , Animales , Humanos
14.
Blood ; 121(3): 447-58, 2013 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-23169777

RESUMEN

To search for genes that promote hematopoietic development from human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs), we overexpressed several known hematopoietic regulator genes in hESC/iPSC-derived CD34(+)CD43(-) endothelial cells (ECs) enriched in hemogenic endothelium (HE). Among the genes tested, only Sox17, a gene encoding a transcription factor of the SOX family, promoted cell growth and supported expansion of CD34(+)CD43(+)CD45(-/low) cells expressing the HE marker VE-cadherin. SOX17 was expressed at high levels in CD34(+)CD43(-) ECs compared with low levels in CD34(+)CD43(+)CD45(-) pre-hematopoietic progenitor cells (pre-HPCs) and CD34(+)CD43(+)CD45(+) HPCs. Sox17-overexpressing cells formed semiadherent cell aggregates and generated few hematopoietic progenies. However, they retained hemogenic potential and gave rise to hematopoietic progenies on inactivation of Sox17. Global gene-expression analyses revealed that the CD34(+)CD43(+)CD45(-/low) cells expanded on overexpression of Sox17 are HE-like cells developmentally placed between ECs and pre-HPCs. Sox17 overexpression also reprogrammed both pre-HPCs and HPCs into HE-like cells. Genome-wide mapping of Sox17-binding sites revealed that Sox17 activates the transcription of key regulator genes for vasculogenesis, hematopoiesis, and erythrocyte differentiation directly. Depletion of SOX17 in CD34(+)CD43(-) ECs severely compromised their hemogenic activity. These findings suggest that SOX17 plays a key role in priming hemogenic potential in ECs, thereby regulating hematopoietic development from hESCs/iPSCs.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Factores de Transcripción SOXF/genética , Factores de Transcripción SOXF/fisiología , Animales , Diferenciación Celular/fisiología , División Celular/fisiología , Células Cultivadas , Técnicas de Cocultivo , Células Endoteliales/citología , Células Endoteliales/fisiología , Sangre Fetal/citología , Fibroblastos/citología , Hematopoyesis/genética , Humanos , Lentivirus/genética , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/fisiología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/fisiología , Transducción Genética/métodos
15.
J Neurosci ; 31(39): 13921-35, 2011 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-21957254

RESUMEN

The SRY-box (Sox) transcription factors regulate oligodendrocyte differentiation, but their signaling targets are largely unknown. We have identified a major signal transduction pathway regulated by Sox containing gene 17 (Sox17) in the oligodendrocyte lineage. Microarray analysis in oligodendrocyte progenitor cells (OPCs) after Sox17 attenuation revealed upregulated genes associated with cell cycle control and activation of the Wingless and integration site (Wnt)/ß-catenin pathway. Sox17 knockdown also increases the levels of cyclin D1, Axin2, and activated ß-catenin. In OPCs, the expression pattern of Sox17, cyclin D1, and secreted Frizzled-related protein-1 in the presence of platelet-derived growth factor (PDGF) was coordinately accelerated by addition of thyroid hormone, indicating differentiation-induced regulation of Sox17 targets. In developing white matter, decreased total ß-catenin, activated ß-catenin, and cyclin D1 levels coincided with the peak of Sox17 expression, and immunoprecipitates showed a developmentally regulated interaction among Sox17, T-cell transcription factor 4, and ß-catenin proteins. In OPCs, PDGF stimulated phosphorylation of glycogen synthase 3ß and the Wnt coreceptor LRP6, and enhanced ß-catenin-dependent gene expression. Sox17 overexpression inhibited PDGF-induced TOPFLASH and cyclin D1 promoter activity, and decreased endogenous cyclin D1, activated ß-catenin, as well as total ß-catenin levels. Recombinant Sox17 prevented Wnt3a from repressing myelin protein expression, and inhibition of Sox17-mediated proteasomal degradation of ß-catenin blocked myelin protein induction. These results indicate that Sox17 suppresses cyclin D1 expression and cell proliferation by directly antagonizing ß-catenin, whose activity in OPCs is stimulated not only by Wnt3a, but also by PDGF. Our identification of downstream targets of Sox17 thus defines signaling pathways and molecular mechanisms in OPCs that are regulated by Sox17 during cell cycle exit and the onset of differentiation in oligodendrocyte development.


Asunto(s)
Proteínas HMGB/fisiología , Oligodendroglía/fisiología , Factores de Transcripción SOXF/fisiología , Transducción de Señal/fisiología , Células Madre/fisiología , Proteínas Wnt/fisiología , beta Catenina/fisiología , Animales , Células Cultivadas , Técnicas de Sustitución del Gen , Proteínas HMGB/antagonistas & inhibidores , Proteínas HMGB/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de la Mielina/antagonistas & inhibidores , Proteínas de la Mielina/biosíntesis , Células 3T3 NIH , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Factores de Transcripción SOXF/antagonistas & inhibidores , Factores de Transcripción SOXF/genética , beta Catenina/antagonistas & inhibidores
16.
Dev Biol ; 350(2): 393-404, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21146513

RESUMEN

Cells of the primitive endoderm (PrE) and the pluripotent epiblast (EPI), the two lineages specified within the inner cell mass (ICM) of the mouse blastocyst stage embryo, are segregated into adjacent tissue layers by the end of the preimplantation period. The PrE layer which emerges as a polarized epithelium adjacent to the blastocoel, with a basement membrane separating it from the EPI, has two derivatives, the visceral and parietal endoderm. In this study we have investigated the localization of two transcriptional regulators of the SOX family, SOX17 and SOX7, within the PrE and its derivatives. We noted that SOX17 was first detected in a salt-and-pepper distribution within the ICM, subsequently becoming restricted to the nascent PrE epithelium. This dynamic distribution of SOX17 resembled the localization of GATA6 and GATA4, two other PrE lineage-specific transcription factors. By contrast, SOX7 was only detected in PrE cells positioned in contact with the blastocoel, raising the possibility that these cells are molecularly distinct. Our observations support a model of sequential GATA6 > SOX17 > GATA4 > SOX7 transcription factor activation within the PrE lineage, perhaps correlating with the consecutive periods of cell lineage 'naïvete', commitment and sorting. Furthermore our data suggest that co-expression of SOX17 and SOX7 within sorted PrE cells could account for the absence of a detectable phenotype of Sox17 mutant blastocysts. However, analysis of implantation-delayed blastocysts, revealed a role for SOX17 in the maintenance of PrE epithelial integrity, with the absence of SOX17 leading to premature delamination and migration of parietal endoderm.


Asunto(s)
Blastocisto/fisiología , Diferenciación Celular , Linaje de la Célula , Endodermo/fisiología , Proteínas HMGB/fisiología , Factores de Transcripción SOXF/fisiología , Factores de Transcripción/fisiología , Animales , Blastocisto/citología , Movimiento Celular , Polaridad Celular , Endodermo/citología , Femenino , Proteínas HMGB/análisis , Masculino , Ratones , Ratones Endogámicos ICR , Factores de Transcripción SOXF/análisis , Factores de Transcripción SOXF/genética
17.
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
18.
Blood ; 114(23): 4813-22, 2009 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-19801444

RESUMEN

The molecular mechanisms that regulate the balance between proliferation and differentiation of precursors at the onset of hematopoiesis specification are poorly understood. By using a global gene expression profiling approach during the course of embryonic stem cell differentiation, we identified Sox7 as a potential candidate gene involved in the regulation of blood lineage formation from the mesoderm germ layer. In the present study, we show that Sox7 is transiently expressed in mesodermal precursors as they undergo specification to the hematopoietic program. Sox7 knockdown in vitro significantly decreases the formation of both primitive erythroid and definitive hematopoietic progenitors as well as endothelial progenitors. In contrast, Sox7-sustained expression in the earliest committed hematopoietic precursors promotes the maintenance of their multipotent and self-renewing status. Removal of this differentiation block driven by Sox7-enforced expression leads to the efficient differentiation of hematopoietic progenitors to all erythroid and myeloid lineages. This study identifies Sox7 as a novel and important player in the molecular regulation of the first committed blood precursors. Furthermore, our data demonstrate that the mere sustained expression of Sox7 is sufficient to completely alter the balance between proliferation and differentiation at the onset of hematopoiesis.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Factores de Transcripción SOXF/fisiología , Animales , División Celular , Linaje de la Célula , Células Cultivadas/citología , Células Cultivadas/metabolismo , Células Eritroides/citología , Gástrula/citología , Gástrula/metabolismo , Técnicas de Silenciamiento del Gen , Vectores Genéticos/farmacología , Hemangioblastos/citología , Hemangioblastos/metabolismo , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Lentivirus/genética , Mesodermo/citología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Mieloides/citología , ARN Interferente Pequeño/genética , Proteínas Recombinantes de Fusión/fisiología , Factores de Transcripción SOXF/biosíntesis , Factores de Transcripción SOXF/deficiencia , Factores de Transcripción SOXF/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
20.
Biochem Biophys Res Commun ; 391(1): 357-63, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19913509

RESUMEN

In early-organogenesis-stage mouse embryos, the posteroventral foregut endoderm adjacent to the heart tube gives rise to liver, ventral pancreas and gallbladder. Hepatic and pancreatic primordia become specified in the posterior segment of the ventral foregut endoderm at early somite stages. The mechanisms for demarcating gallbladder and bile duct primordium, however, are poorly understood. Here, we demonstrate that the gallbladder and bile duct progenitors are specified in the paired lateral endoderm domains outside the heart field at almost the same timing as hepatic and pancreatic induction. In the anterior definitive endoderm, Sox17 reactivation occurs in a certain population within the most lateral domains posterolateral to the anterior intestinal portal (AIP) lip on both the left and right sides. During foregut formation, the paired Sox17-positive domains expand ventromedially to merge in the midline of the AIP lip and become localized between the liver and pancreatic primordia. In Sox17-null embryos, these lateral domains are missing, resulting in a complete loss of the gallbladder/bile-duct structure. Chimera analyses revealed that Sox17-null endoderm cells in the posteroventral foregut do not display any gallbladder/bile-duct molecular characters. Our findings show that Sox17 functions cell-autonomously to specify gallbladder/bile-duct in the mouse embryo.


Asunto(s)
Conductos Biliares/embriología , Vesícula Biliar/embriología , Proteínas HMGB/fisiología , Intestinos/embriología , Morfogénesis , Factores de Transcripción SOXF/fisiología , Animales , Conductos Biliares/anomalías , Conductos Biliares/metabolismo , Tipificación del Cuerpo , Embrión de Mamíferos/metabolismo , Endodermo/metabolismo , Femenino , Vesícula Biliar/anomalías , Vesícula Biliar/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas HMGB/genética , Mucosa Intestinal/metabolismo , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción SOXF/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA