Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Commun Biol ; 6(1): 771, 2023 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-37488353

RESUMEN

The Zfp296 gene encodes a zinc finger-type protein. Its expression is high in mouse embryonic stem cells (ESCs) but rapidly decreases following differentiation. Zfp296-knockout (KO) ESCs grew as flat colonies, which were reverted to rounded colonies by exogenous expression of Zfp296. KO ESCs could not form teratomas when transplanted into mice but could efficiently contribute to germline-competent chimeric mice following blastocyst injection. Transcriptome analysis revealed that Zfp296 deficiency up- and down-regulates a distinct group of genes, among which Dppa3, Otx2, and Pou3f1 were markedly downregulated. Chromatin immunoprecipitation sequencing demonstrated that ZFP296 binding is predominantly seen in the vicinity of the transcription start sites (TSSs) of a number of genes, and ZFP296 was suggested to negatively regulate transcription. Consistently, chromatin accessibility assay clearly showed that ZFP296 binding reduces the accessibility of the TSS regions of target genes. Zfp296-KO ESCs showed increased histone H3K9 di- and trimethylation. Co-immunoprecipitation analyses revealed interaction of ZFP296 with G9a and GLP. These results show that ZFP296 plays essential roles in maintaining the global epigenetic state of ESCs through multiple mechanisms including activation of Dppa3, attenuation of chromatin accessibility, and repression of H3K9 methylation, but that Zfp296-KO ESCs retain a unique state of pluripotency while lacking the teratoma-forming ability.


Asunto(s)
Cromatina , Teratoma , Animales , Ratones , Células Madre Embrionarias , Histonas , Células Madre Embrionarias de Ratones , Bioensayo , Proteínas Cromosómicas no Histona , Factor 6 de Transcripción de Unión a Octámeros
2.
Biochem Biophys Res Commun ; 556: 134-141, 2021 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-33839409

RESUMEN

Oxidative stress is a deteriorating factor for pancreatic ß-cells under chronic hyperglycemia in diabetes. However, the molecular mechanism underlying the increase in oxidative stress in ß-cells under diabetic conditions remains unclear. We demonstrated previously that the selective alleviation of glucotoxicity ameliorated the downregulation of several ß-cell factors, including Cox6a2. Cox6a2 encodes a subunit of the respiratory chain complex IV in mitochondria. In this study, we analyzed the role of Cox6a2 in pancreatic ß-cell function and its pathophysiological significance in diabetes mellitus. Cox6a2-knockdown experiments in MIN6-CB4 cells indicated an increased production of reactive oxygen species as detected by CellROX Deep Red reagent using flow cytometry. In systemic Cox6a2-knockout mice, impaired glucose tolerance was observed under a high-fat high-sucrose diet. However, insulin resistance was reduced when compared with control littermates. This indicates a relative insufficiency of ß-cell function. To examine the transcriptional regulation of Cox6a2, ATAC-seq with islet DNA was performed and an open-chromatin area within the Cox6a2 enhancer region was detected. Reporter gene analysis using this area revealed that MafA directly regulates Cox6a2 expression. These findings suggest that the decreased expression of Cox6a2 increases the levels of reactive oxygen species and that Mafa is associated with decreased Cox6a2 expression under glucotoxic conditions.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Proteínas Musculares/deficiencia , Especies Reactivas de Oxígeno/metabolismo , Animales , Línea Celular , Diabetes Mellitus Experimental/metabolismo , Complejo IV de Transporte de Electrones/biosíntesis , Complejo IV de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/metabolismo , Regulación de la Expresión Génica , Glucosa/metabolismo , Intolerancia a la Glucosa/genética , Células HEK293 , Humanos , Insulina/metabolismo , Resistencia a la Insulina/genética , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Proteínas Musculares/biosíntesis , Proteínas Musculares/genética , Estrés Oxidativo , Transcripción Genética
3.
Sci Rep ; 11(1): 477, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33436850

RESUMEN

A pancreatic ß-cell line MIN6 was previously established in our lab from an insulinoma developed in an IT6 transgenic mouse expressing the SV40 T antigen in ß-cells. This cell line has been widely used for in vitro analysis of ß-cell function, but tends to lose the mature ß-cell features, including glucose-stimulated insulin secretion (GSIS), in long-term culture. The aim of this study was to develop a stable ß-cell line that retains the characteristics of mature ß-cells. Considering that mice derived from a cross between C3H and C57BL/6 strains are known to exhibit higher insulin secretory capacity than C57BL/6 mice, an IT6 male mouse of this hybrid background was used to isolate insulinomas, which were independently cultured. After 7 months of continuous culturing, we obtained the MIN6-CB4 ß-cell line, which stably maintains its GSIS. It has been noted that ß-cell lines express the glucagon (Gcg) gene at certain levels. MIN6-CB4 cells were utilized to assess the effects of differential Gcg expression on ß-cell function. Our data show the functional importance of Gcg expression and resulting basal activation of the GLP-1 receptor in ß-cells. MIN6-CB4 cells can serve as an invaluable tool for studying the regulatory mechanisms of insulin secretion, such as the GLP-1/cAMP signaling, in ß-cells.


Asunto(s)
Receptor del Péptido 1 Similar al Glucagón/metabolismo , Glucagón/fisiología , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Insulinoma/metabolismo , Neoplasias Pancreáticas/metabolismo , Animales , Femenino , Células Secretoras de Insulina/citología , Insulinoma/patología , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias Pancreáticas/patología
4.
Development ; 147(2)2020 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-31852686

RESUMEN

Lymphangiogenesis plays important roles in normal fetal development and postnatal growth. However, its molecular regulation remains unclear. Here, we have examined the function of forkhead box protein O1 (FOXO1) transcription factor, a known angiogenic factor, in developmental dermal lymphangiogenesis using endothelial cell-specific FOXO1-deficient mice. FOXO1-deficient mice showed disconnected and dilated lymphatic vessels accompanied with increased proliferation and decreased apoptosis in the lymphatic capillaries. Comprehensive DNA microarray analysis of the causes of in vivo phenotypes in FOXO1-deficient mice revealed that the gene encoding C-X-C chemokine receptor 4 (CXCR4) was the most drastically downregulated in FOXO1-deficient primary lymphatic endothelial cells (LECs). CXCR4 was expressed in developing dermal lymphatic capillaries in wild-type mice but not in FOXO1-deficient dermal lymphatic capillaries. Furthermore, FOXO1 suppression impaired migration toward the exogenous CXCR4 ligand, C-X-C chemokine ligand 12 (CXCL12), and coordinated proliferation in LECs. These results suggest that FOXO1 serves an essential role in normal developmental lymphangiogenesis by promoting LEC migration toward CXCL12 and by regulating their proliferative activity. This study provides valuable insights into the molecular mechanisms underlying developmental lymphangiogenesis.


Asunto(s)
Dermis/metabolismo , Proteína Forkhead Box O1/metabolismo , Regulación del Desarrollo de la Expresión Génica , Linfangiogénesis/genética , Receptores CXCR4/genética , Cola (estructura animal)/metabolismo , Regulación hacia Arriba/genética , Animales , Animales Recién Nacidos , Antígenos CD/metabolismo , Apoptosis , Secuencia de Bases , Cadherinas/metabolismo , Muerte Celular , Proliferación Celular , Quimiocina CXCL12/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Elementos de Facilitación Genéticos/genética , Eliminación de Gen , Integrasas/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Regiones Promotoras Genéticas/genética , Unión Proteica , Receptores CXCR4/metabolismo
6.
J Cell Biol ; 217(4): 1287-1301, 2018 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-29507125

RESUMEN

In mammalian pancreatic ß cells, the IRE1α-XBP1 pathway is constitutively and highly activated under physiological conditions. To elucidate the precise role of this pathway, we constructed ß cell-specific Ire1α conditional knockout (CKO) mice and established insulinoma cell lines in which Ire1α was deleted using the Cre-loxP system. Ire1α CKO mice showed the typical diabetic phenotype including impaired glycemic control and defects in insulin biosynthesis postnatally at 4-20 weeks. Ire1α deletion in pancreatic ß cells in mice and insulinoma cells resulted in decreased insulin secretion, decreased insulin and proinsulin contents in cells, and decreased oxidative folding of proinsulin along with decreased expression of five protein disulfide isomerases (PDIs): PDI, PDIR, P5, ERp44, and ERp46. Reconstitution of the IRE1α-XBP1 pathway restored the proinsulin and insulin contents, insulin secretion, and expression of the five PDIs, indicating that IRE1α functions as a key regulator of the induction of catalysts for the oxidative folding of proinsulin in pancreatic ß cells.


Asunto(s)
Endorribonucleasas/metabolismo , Células Secretoras de Insulina/enzimología , Insulina/metabolismo , Proinsulina/metabolismo , Pliegue de Proteína , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína 1 de Unión a la X-Box/metabolismo , Factor de Transcripción Activador 6/metabolismo , Animales , Sitios de Unión , Glucemia/metabolismo , Línea Celular Tumoral , Diabetes Mellitus/sangre , Diabetes Mellitus/enzimología , Diabetes Mellitus/genética , Endorribonucleasas/deficiencia , Endorribonucleasas/genética , Insulina/genética , Insulinoma/enzimología , Insulinoma/genética , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Noqueados , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Oxidación-Reducción , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/genética , Fosforilación , Proinsulina/química , Proinsulina/genética , Regiones Promotoras Genéticas , Proteína Disulfuro Isomerasas/genética , Proteína Disulfuro Isomerasas/metabolismo , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal , Tiorredoxinas/genética , Tiorredoxinas/metabolismo , Proteína 1 de Unión a la X-Box/genética , eIF-2 Quinasa/metabolismo
7.
EMBO Rep ; 19(4)2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29437694

RESUMEN

The piRNA pathway is a piRNA-guided retrotransposon silencing system which includes processing of retrotransposon transcripts by PIWI-piRNAs in secondary piRNA biogenesis. Although several proteins participate in the piRNA pathway, the ones crucial for the cleavage of target RNAs by PIWI-piRNAs have not been identified. Here, we show that GTSF1, an essential factor for retrotransposon silencing in male germ cells in mice, associates with both MILI and MIWI2, mouse PIWI proteins that function in prospermatogonia. GTSF1 deficiency leads to a severe defect in the production of secondary piRNAs, which are generated from target RNAs of PIWI-piRNAs. Furthermore, in Gtsf1 mutants, a known target RNA of PIWI-piRNAs is left unsliced at the cleavage site, and the generation of secondary piRNAs from this transcript is defective. Our findings indicate that GTSF1 is a crucial factor for the slicing of target RNAs by PIWI-piRNAs and thus affects secondary piRNA biogenesis in prospermatogonia.


Asunto(s)
Regulación de la Expresión Génica , Proteínas/metabolismo , ARN Interferente Pequeño/genética , Transcripción Genética , Células Madre Germinales Adultas/metabolismo , Animales , Núcleo Celular/metabolismo , Amplificación de Genes , Silenciador del Gen , Genes de Partícula A Intracisternal , Péptidos y Proteínas de Señalización Intracelular , Elementos de Nucleótido Esparcido Largo , Masculino , Ratones , Ratones Noqueados , Modelos Biológicos , Complejos Multiproteicos/metabolismo , Unión Proteica , Transporte de Proteínas , Proteínas/genética , Interferencia de ARN , Proteínas Recombinantes de Fusión , Retroelementos , Testículo/metabolismo
8.
Angiogenesis ; 21(2): 203-214, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29185141

RESUMEN

Forkhead box protein O1 (FoxO1) is a transcription factor and a critical regulator of angiogenesis. Various environmental stimuli, including growth factors, nutrients, shear stress, oxidative stress and hypoxia, affect FoxO1 subcellular localization and strongly influence its transcriptional activity; however, FoxO1-localization patterns in endothelial cells (ECs) during development have not been clarified in vivo. Here, we reported that FoxO1 expression was observed in three layers of angiogenic vessels in developing mouse retinas and that among these layers, the front layer showed high levels of FoxO1 expression in the nuclei of most tip ECs. Because tip ECs migrate toward the avascular hypoxic area, we focused on hypoxia as a major stimulus regulating FoxO1 subcellular localization in tip cells. In cultured ECs, FoxO1 accumulated into the nucleus under hypoxic conditions, with hypoxia also inducing expression of tip-cell-specific genes, including endothelial-specific molecule 1 (ESM1), which was suppressed by FoxO1 knockdown. Additionally, in murine models, EC-specific FoxO1 deletion resulted in reduced ESM1 expression and suppressed tip-cell migration during angiogenesis. These findings indicated roles for FoxO1 in tip-cell migration and that its transcriptional activity is regulated by hypoxia.


Asunto(s)
Células Endoteliales/metabolismo , Proteína Forkhead Box O1/metabolismo , Regulación de la Expresión Génica , Hipoxia/metabolismo , Retina/crecimiento & desarrollo , Neovascularización Retiniana/metabolismo , Animales , Células Endoteliales/patología , Proteína Forkhead Box O1/genética , Técnicas de Silenciamiento del Gen , Humanos , Hipoxia/genética , Hipoxia/patología , Ratones , Ratones Transgénicos , Retina/patología , Neovascularización Retiniana/genética , Neovascularización Retiniana/patología
9.
Sci Rep ; 7(1): 12462, 2017 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-28963472

RESUMEN

The Cys2/His2-type zinc finger protein Zfp296 has been implicated in stem cell pluripotency and tumor pathogenesis. However, its mechanisms remain elusive. Here, we demonstrated that a Zfp296 deficiency in mice impairs germ-cell development and embryonic growth. Zfp296 was intracellularly localized to heterochromatin in embryos. A GST-Zfp296 pull-down experiment using ES cell nuclear extract followed by LC-MS/MS showed that Zfp296 interacts with component proteins of heterochromatin (such as HP1, Dnmt1, Dnmt3b, and ATRX) and the NuRD complex. We focused on H3K9 methylation as a hallmark of heterochromatin, and found that Zfp296 overexpression in cultured cells reduces the Suv39h1-mediated H3K9 methylation. Consistent with this finding, in Zfp296 -/- mouse embryos, we observed a global increase in H3K9 methylation in a developmental stage-dependent manner, and showed, by ChIP-qPCR, that the H3K9me3 levels at major satellite repeats were elevated in Zfp296 -/- embryos. Our results demonstrate that Zfp296 is a component of heterochromatin that affects embryonic development by negatively regulating H3K9 methylation.


Asunto(s)
Proteínas de Unión al ADN/genética , Desarrollo Embrionario/genética , Heterocromatina/metabolismo , Histonas/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Procesamiento Proteico-Postraduccional , Animales , Diferenciación Celular , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1/genética , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Proteínas de Unión al ADN/deficiencia , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica , Heterocromatina/química , Histonas/genética , Masculino , Metilación , Metiltransferasas/genética , Metiltransferasas/metabolismo , Ratones , Ratones Noqueados , Células Madre Embrionarias de Ratones/citología , Ovario/anomalías , Ovario/crecimiento & desarrollo , Ovario/metabolismo , Cultivo Primario de Células , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Testículo/anomalías , Testículo/crecimiento & desarrollo , Testículo/metabolismo , Proteína Nuclear Ligada al Cromosoma X/genética , Proteína Nuclear Ligada al Cromosoma X/metabolismo , ADN Metiltransferasa 3B
10.
PLoS One ; 11(8): e0161190, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27526291

RESUMEN

A promising approach to new diabetes therapies is to generate ß cells from other differentiated pancreatic cells in vivo. Because the acinar cells represent the most abundant cell type in the pancreas, an attractive possibility is to reprogram acinar cells into ß cells. The transcription factor Pdx1 (Pancreas/duodenum homeobox protein 1) is essential for pancreatic development and cell lineage determination. Our objective is to examine whether exogenous expression of Pdx1 in acinar cells of adult mice might induce reprogramming of acinar cells into ß cells. We established a transgenic mouse line in which Pdx1 and EGFP (enhanced green fluorescent protein) could be inducibly expressed in the acinar cells. After induction of Pdx1, we followed the acinar cells for their expression of exocrine and endocrine markers using cell-lineage tracing with EGFP. The acinar cell-specific expression of Pdx1 in adult mice reprogrammed the acinar cells as endocrine precursor cells, which migrated into the pancreatic islets and differentiated into insulin-, somatostatin-, or PP (pancreatic polypeptide)-producing endocrine cells, but not into glucagon-producing cells. When the mice undergoing such pancreatic reprogramming were treated with streptozotocin (STZ), the newly generated insulin-producing cells were able to ameliorate STZ-induced diabetes. This paradigm of in vivo reprogramming indicates that acinar cells hold promise as a source for new islet cells in regenerative therapies for diabetes.


Asunto(s)
Células Acinares/citología , Transdiferenciación Celular/genética , Páncreas/citología , Transactivadores/genética , Células Acinares/metabolismo , Animales , Glucemia/metabolismo , Reprogramación Celular , Glándulas Exocrinas/citología , Expresión Génica , Proteínas Fluorescentes Verdes/genética , Insulina/metabolismo , Ratones , Ratones Transgénicos
11.
PLoS One ; 11(3): e0151927, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26986842

RESUMEN

Elucidating the regulation of glucose-stimulated insulin secretion (GSIS) in pancreatic ß cells is important for understanding and treating diabetes. The pancreatic ß cell line, MIN6, retains GSIS but gradually loses it in long-term culture. The MIN6 subclone, MIN6c4, exhibits well-regulated GSIS even after prolonged culture. We previously used DNA microarray analysis to compare gene expression in the parental MIN6 cells and MIN6c4 cells and identified several differentially regulated genes that may be involved in maintaining GSIS. Here we investigated the potential roles of six of these genes in GSIS: Tmem59l (Transmembrane protein 59 like), Scgn (Secretagogin), Gucy2c (Guanylate cyclase 2c), Slc29a4 (Solute carrier family 29, member 4), Cdhr1 (Cadherin-related family member 1), and Celsr2 (Cadherin EGF LAG seven-pass G-type receptor 2). These genes were knocked down in MIN6c4 cells using lentivirus vectors expressing gene-specific short hairpin RNAs (shRNAs), and the effects of the knockdown on insulin expression and secretion were analyzed. Suppression of Tmem59l, Scgn, and Gucy2c expression resulted in significantly decreased glucose- and/or KCl-stimulated insulin secretion from MIN6c4 cells, while the suppression of Slc29a4 expression resulted in increased insulin secretion. Tmem59l overexpression rescued the phenotype of the Tmem59l knockdown MIN6c4 cells, and immunostaining analysis indicated that the TMEM59L protein colocalized with insulin and GM130, a Golgi complex marker, in MIN6 cells. Collectively, our findings suggested that the proteins encoded by Tmem59l, Scgn, Gucy2c, and Slc29a4 play important roles in regulating GSIS. Detailed studies of these proteins and their functions are expected to provide new insights into the molecular mechanisms involved in insulin secretion.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Animales , Western Blotting , Cadherinas/fisiología , Línea Celular , Femenino , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Genes Reguladores/fisiología , Glucosa/fisiología , Insulina/fisiología , Secreción de Insulina , Células Secretoras de Insulina/fisiología , Proteínas de la Membrana/fisiología , Proteínas de Transporte de Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Receptores de Enterotoxina , Receptores Acoplados a la Guanilato-Ciclasa/fisiología , Receptores de Péptidos/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Secretagoginas/fisiología
12.
PLoS One ; 11(3): e0150390, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26930067

RESUMEN

The unknown protein family 0224 (UPF0224) includes three members that are expressed in germ-line cells in mice: Gtsf1, Gtsf1l, and BC048502 (Gtsf2). These genes produce proteins with two repeats of the CHHC Zn-finger domain, a predicted RNA-binding motif, in the N terminus. We previously reported that Gtsf1 is essential for spermatogenesis and retrotransposon suppression. In this study, we investigated the expression patterns and functions of Gtsf1l and Gtsf2. Interestingly, Gtsf1l and Gtsf2 were found to be sequentially but not simultaneously expressed in gonocytes and spermatids. Pull-down experiments showed that both GTSF1L and GTSF2 can interact with PIWI-protein complexes. Nevertheless, knocking out Gtsf1, Gtsf2, or both did not cause defects in spermatogenesis or retrotransposon suppression in mice.


Asunto(s)
Proteínas Portadoras/genética , Regulación del Desarrollo de la Expresión Génica , Células Germinativas/metabolismo , Proteínas/genética , Espermátides/metabolismo , Espermatogénesis/genética , Secuencia de Aminoácidos , Animales , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Proteínas Portadoras/metabolismo , Línea Celular , Células Cultivadas , Femenino , Técnica del Anticuerpo Fluorescente , Células Germinativas/crecimiento & desarrollo , Immunoblotting , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Unión Proteica , Proteínas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Espermátides/crecimiento & desarrollo , Testículo/citología , Testículo/metabolismo
13.
PLoS One ; 8(8): e71645, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23940776

RESUMEN

Tcl1 is highly expressed in embryonic stem (ES) cells, but its expression rapidly decreases following differentiation. To assess Tcl1's roles in ES cells, we generated Tcl1-deficient and -overexpressing mouse ES cell lines. We found that Tcl1 was neither essential nor sufficient for maintaining the undifferentiated state. Tcl1 is reported to activate Akt and to enhance cell proliferation. We found that Tcl1 expression levels correlated positively with the proliferation rate and negatively with the apoptosis of ES cells, but did not affect Akt phosphorylation. On the other hand, the phosphorylation level of ß-catenin decreased in response to Tcl1 overexpression. We measured the ß-catenin activity using the TOPflash reporter assay, and found that wild-type ES cells had low activity, which Tcl1 overexpression enhanced 1.8-fold. When the canonical Wnt signaling is activated by ß-catenin stabilization, it reportedly helps maintain ES cells in the undifferentiated state. We then performed DNA microarray analyses between the Tcl1-deficient and -expressing ES cells. The results revealed that Tcl1 expression downregulated a distinct group of genes, including Ndp52, whose expression is very high in blastocysts but reduced in the primitive ectoderm. Based on these results, we discuss the possible roles of Tcl1 in ES cells.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Proteínas Proto-Oncogénicas/fisiología , Animales , Apoptosis/genética , Diferenciación Celular/genética , Línea Celular , Proliferación Celular , Perfilación de la Expresión Génica , Técnicas de Transferencia de Gen , Ratones , Ratones Noqueados , Análisis por Micromatrices , Vía de Señalización Wnt/genética
14.
PLoS One ; 8(4): e61211, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23560115

RESUMEN

Elucidating the regulation of glucose-stimulated insulin secretion (GSIS) in pancreatic islet ß cells is important for understanding and treating diabetes. MIN6 cells, a transformed ß-cell line derived from a mouse insulinoma, retain GSIS and are a popular in vitro model for insulin secretion. However, in long-term culture, MIN6 cells' GSIS capacity is lost. We previously isolated a subclone, MIN6 clone 4, from the parental MIN6 cells, that shows well-regulated insulin secretion in response to glucose, glybenclamide, and KCl, even after prolonged culture. To investigate the molecular mechanisms responsible for preserving GSIS in this subclone, we compared four groups of MIN6 cells: Pr-LP (parental MIN6, low passage number), Pr-HP (parental MIN6, high passage number), C4-LP (MIN6 clone 4, low passage number), and C4-HP (MIN6 clone 4, high passage number). Based on their capacity for GSIS, we designated the Pr-LP, C4-LP, and C4-HP cells as "responder cells." In a DNA microarray analysis, we identified a group of genes with high expression in responder cells ("responder genes"), but extremely low expression in the Pr-HP cells. Another group of genes ("non-responder genes") was expressed at high levels in the Pr-HP cells, but at extremely low levels in the responder cells. Some of the responder genes were involved in secretory machinery or glucose metabolism, including Chrebp, Scgn, and Syt7. Among the non-responder genes were Car2, Maf, and Gcg, which are not normally expressed in islet ß cells. Interestingly, we found a disproportionate number of known imprinted genes among the responder genes. Our findings suggest that the global expression profiling of GSIS-competent and GSIS-incompetent MIN6 cells will help delineate the gene regulatory networks for insulin secretion.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Anhidrasa Carbónica II/genética , Anhidrasa Carbónica II/metabolismo , Línea Celular , Proliferación Celular , Células Clonales , Redes Reguladoras de Genes , Glucosa/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/citología , Ratones , Análisis por Micromatrices , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Especificidad de Órganos , Proglucagón/genética , Proglucagón/metabolismo , Proteínas Proto-Oncogénicas c-maf/genética , Proteínas Proto-Oncogénicas c-maf/metabolismo , Secretagoginas , Sinaptotagminas/genética , Sinaptotagminas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
15.
Biochem Biophys Res Commun ; 430(2): 604-9, 2013 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-23219817

RESUMEN

Obtaining a homogenous population of central nervous system neurons has been a significant challenge in neuroscience research; however, a recent study established a retinoic acid-treated embryoid bodies-based differentiation protocol that permits the effective generation of highly homogeneous glutamatergic cortical pyramidal neurons from embryonic stem cells. We were able to reproduce this protocol regarding the purity of glutamatergic neurons, but these neurons were not sufficiently healthy for long-term observation under the same conditions that were originally described. Here, we achieved a substantial improvement in cell survival by applying a simple technique: We changed the medium for glutamatergic neurons from the original complete medium to commercially available SBM (the Nerve-Cell Culture Medium manufactured by Sumitomo Bakelite Co. Ltd.) and finally succeeded in maintaining healthy neurons for at least 3 weeks without decreasing their purity. Because SBM contains glial conditioned medium, we postulated that brain-derived neurotrophic factor or basic fibroblast growth factor is the key components responsible for pro-survival effect of SBM on neurons, and examined their effects by adding them to CM. As a result, neither of them had pro-survival effect on pure glutamatergic neuronal population.


Asunto(s)
Técnicas de Cultivo de Célula , Células Madre Embrionarias/citología , Ácido Glutámico/metabolismo , Neurogénesis , Neuronas/citología , Animales , Apoptosis , Factor Neurotrófico Derivado del Encéfalo/farmacología , Caspasa 3/metabolismo , Supervivencia Celular , Células Madre Embrionarias/efectos de los fármacos , Factores de Crecimiento de Fibroblastos/farmacología , Ratones , Tubulina (Proteína)/metabolismo
16.
PLoS One ; 7(10): e47536, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23077629

RESUMEN

Tubular complexes (TCs) are aggregates of duct-like monolayered cells in the developing and regenerating pancreas. Recent studies showed that TCs have regenerative potential, including islet neogenesis. We previously delivered adenovirus vector (AdV) into exocrine cells of the pancreas by intra-common bile ductal (ICBD) injection, and found that AdV expressing Pdx1, a pancreas-specific transcription factor, causes TC formation and islet neogenesis. We also established RTF-Pdx1-EGFP mice, which ubiquitously express Pdx1 when tetracycline is removed from the drinking water. However, exogenous Pdx1 expression in adult RTF-Pdx1-EGFP mice did not cause any pathological changes in the pancreas during three weeks of observation after tetracycline withdrawal. To examine whether the host immune response induced by AdV was involved in TC formation, we delivered AdVs expressing pancreas-related transcription factors or an irrelevant protein into the pancreas of RTF-Pdx1-EGFP mice. Histological analyses showed that both AdV injection and Pdx1 expression are required for TC formation. We also analyzed the effects of these ICBD-injected AdVs. AdV expressing Isl1, a proendocrine transcription factor, effectively induced TC formation through acinar-to-ductal metaplasia, and exogenous Pdx1 expression facilitated this process. Considering the regenerative potential of TCs, a strategy that efficiently induces TC formation may lead to novel therapies for diabetes.


Asunto(s)
Proteínas de Homeodominio , Proteínas con Homeodominio LIM , Metaplasia , Páncreas , Transactivadores , Factores de Transcripción , Adenoviridae , Animales , Diabetes Mellitus/genética , Diabetes Mellitus/terapia , Regulación Neoplásica de la Expresión Génica , Vectores Genéticos , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Metaplasia/genética , Metaplasia/metabolismo , Ratones , Páncreas/metabolismo , Páncreas/patología , Regeneración/genética , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
17.
Genes Cells ; 17(9): 758-67, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22845550

RESUMEN

Several reports have suggested that Foxo1, a key regulator in differentiation, growth and metabolism, is involved in pancreatic ß-cell function. However, detailed analyses have been hampered by a lack of Foxo1-deficient ß cells. To elucidate Foxo1's function in ß cells, we produced a ß-cell line with inducible Foxo1 deletion. We generated a conditional knockout mouse line, in which Cre recombinase deletes the Foxo1 gene. We then established a ß-cell line from an insulinoma induced in this knockout mouse by the ß-cell-specific expression of simian virus 40 T antigen. In this cell line, designated MIN6-Foxo1flox/flox, adenovirus-mediated Cre expression ablates the Foxo1 gene, generating MIN6-Foxo1-KO cells. Using these knockout and floxed cell lines, we found that Foxo1 ablation enhanced the glucose-stimulated insulin secretion (GSIS) at high glucose concentrations and enhanced ß-cell proliferation. We also conducted DNA microarray analyses of MIN6-Foxo1-KO cells infected with either an adenovirus vector expressing a constitutively active FOXO1 or a control vector and identified several Foxo1-regulated genes, including some known to be related to ß-cell function. These cells should be useful for further studies on Foxo1's roles in ß-cells and may lead to novel strategies for treating the impaired insulin secretion in type 2 diabetes mellitus.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Alelos , Animales , Antígenos Transformadores de Poliomavirus/genética , Antígenos Transformadores de Poliomavirus/metabolismo , Apoptosis , Línea Celular , Proliferación Celular , Quimera/genética , Quimera/metabolismo , Femenino , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Glucosa/farmacología , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Integrasas/genética , Integrasas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Virus 40 de los Simios/genética , Virus 40 de los Simios/metabolismo
18.
Diabetes ; 59(11): 2854-61, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20798333

RESUMEN

OBJECTIVE: Retinoid X receptors (RXRs) are members of the nuclear hormone receptor superfamily and are thought to be key regulators in differentiation, cellular growth, and gene expression. Although several experiments using pancreatic ß-cell lines have shown that the ligands of nuclear hormone receptors modulate insulin secretion, it is not clear whether RXRs have any role in insulin secretion. RESEARCH DESIGN AND METHODS: To elucidate the function of RXRs in pancreatic ß-cells, we generated a double-transgenic mouse in which a dominant-negative form of RXRß was inducibly expressed in pancreatic ß-cells using the Tet-On system. We also established a pancreatic ß-cell line from an insulinoma caused by the ß-cell-specific expression of simian virus 40 T antigen in the above transgenic mouse. RESULTS: In the transgenic mouse, expression of the dominant-negative RXR enhanced the insulin secretion with high glucose stimulation. In the pancreatic ß-cell line, the suppression of RXRs also enhanced glucose-stimulated insulin secretion at a high glucose concentration, while 9-cis-retinoic acid, an RXR agonist, repressed it. High-density oligonucleotide microarray analysis showed that expression of the dominant-negative RXR affected the expression levels of a number of genes, some of which have been implicated in the function and/or differentiation of ß-cells. CONCLUSIONS: These results suggest that endogenous RXR negatively regulates the glucose-stimulated insulin secretion. Given these findings, we propose that the modulation of endogenous RXR in ß-cells may be a new therapeutic approach for improving impaired insulin secretion in type 2 diabetes.


Asunto(s)
Glucosa/farmacología , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Receptores X Retinoide/fisiología , Animales , Secuencia Conservada , Cruzamientos Genéticos , Cartilla de ADN , ADN Complementario/genética , Femenino , Humanos , Insulina/genética , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Masculino , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas , Receptor alfa X Retinoide/genética , Receptor alfa X Retinoide/fisiología , Receptor beta X Retinoide/genética , Receptor beta X Retinoide/fisiología , Receptores X Retinoide/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Genes Cells ; 15(8): 813-28, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20590823

RESUMEN

In a search for genes specifically expressed in mouse embryonic stem cells, we identified one we called Ces5. We found that it corresponded to the Ooep gene, which was recently reported to be expressed specifically in oocytes. Mouse Ces5/Ooep, also called Moep19 or Floped, encoded a 164-amino acid protein, which was detected in the cytoplasm of developing and mature oocytes and in embryos throughout the preimplantation period. To examine its function, we carried out targeted disruption of this gene. The Ces5/Ooep-null mice were grossly normal, but the females were infertile. Although the ovaries and ovulation appeared normal, the embryos from Ces5/Ooep-null females mated with wild-type males showed developmental arrest at the two- or four-cell stage. In addition, their first cleavage was considerably delayed and often asymmetrical. Thus, Ces5/Ooep is a maternal-effect gene. By electron microscopy, we found that the eggs from Ces5/Ooep-null females lacked oocyte cytoplasmic lattices (CPLs), which have long been predicted to function as a storage form for components that are maternally contributed to the early embryo. Further analysis showed that CES5/OOEP was directly associated with the CPLs. These results indicate that CES5/OOEP is an essential component of the CPLs and is required for embryonic development at the maternal-zygotic stage transition.


Asunto(s)
Citoplasma/metabolismo , Embrión de Mamíferos/embriología , Oocitos/citología , Oocitos/metabolismo , Proteínas de Unión al ARN/metabolismo , Cigoto/metabolismo , Secuencia de Aminoácidos , Animales , Embrión de Mamíferos/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Noqueados , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Transcripción Genética/genética , Cigoto/citología
20.
Dev Biol ; 335(1): 216-27, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19735653

RESUMEN

We recently reported that the Gtsf1/Cue110 gene, a member of the evolutionarily conserved UPF0224 family, is expressed predominantly in male germ cells, and that the GTSF1/CUE110 protein is localized to the cytoplasm of these cells in the adult testis. Here, to analyze the roles of the Gtsf1/Cue110 gene in spermatogenesis, we produced Gtsf1/Cue110-null mice by gene targeting. The Gtsf1/Cue110-null mice grew normally and appeared healthy; however, the males were sterile due to massive apoptotic death of their germ cells after postnatal day 14. In contrast, the null females were fertile. Detailed analyses revealed that the Gtsf1/Cue110-null male meiocytes ceased meiotic progression before the zygotene stage. Thus, the Gtsf1/Cue110 gene is essential for spermatogenesis beyond the early meiotic phase. Furthermore, the loss of the Gtsf1/Cue110 gene caused increased transcription of the long interspersed nucleotide element (Line-1) and the intracisternal A-particle (IAP) retrotransposons, accompanied by demethylation of their promoter regions. These observations indicate that Gtsf1/Cue110 is required for spermatogenesis and involved in retrotransposon suppression in male germ cells.


Asunto(s)
Proteínas , Retroelementos , Espermatogénesis/fisiología , Testículo , Dedos de Zinc , Animales , Metilación de ADN , Femenino , Fertilidad/fisiología , Regulación de la Expresión Génica , Marcación de Gen , Péptidos y Proteínas de Señalización Intracelular , Masculino , Meiosis/fisiología , Ratones , Ratones Noqueados , Proteínas/genética , Proteínas/metabolismo , Testículo/citología , Testículo/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...