Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Stem Cell Res Ther ; 10(1): 157, 2019 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-31151411

RESUMEN

BACKGROUND: Small nucleolar RNA host gene 3 (Snhg3) is a long non-coding RNA (lncRNA) that was shown to participate in the tumorigenesis of certain cancers. However, little is known about its role in embryonic stem cells (ESCs). METHODS: Here, we investigated the role of Snhg3 in mouse ESCs (mESCs) through both loss-of-function (knockdown) and gain-of-function (overexpression) approaches. Alkaline phosphatase staining, secondary colony formation, propidium iodide staining, western blotting, and quantitative reverse transcription polymerase chain reaction (qRT-PCR) were used to access self-renewal capacity, whereas immunofluorescence, qRT-PCR, and embryoid body formation were performed to examine pluripotency. In addition, the effect of Snhg3 on mouse embryonic development was determined based on the morphological changes, blastocyst rate, and altered pluripotency marker (Nanog, Oct4) expression. Moreover, the relationship between Snhg3 and key pluripotency factors was evaluated by chromatin immunoprecipitation qPCR, qRT-PCR, subcellular fractionation, and RNA immunoprecipitation. Finally, RNA pull-down and mass spectrometry were applied to explore the potential interacting proteins of Snhg3 in mESCs. RESULTS: We demonstrated that Snhg3 is essential for self-renewal and pluripotency maintenance in mESCs. In addition, Snhg3 knockdown disrupted mouse early embryo development. Mechanistically, Snhg3 formed a positive feedback network with Nanog and Oct4, and 126 Snhg3-interacting proteins were identified in mESCs. CONCLUSIONS: Snhg3 is essential for mESC self-renewal and pluripotency, as well as mouse early embryo development.


Asunto(s)
Autorrenovación de las Células/fisiología , Células Madre Embrionarias de Ratones/metabolismo , Células Madre Pluripotentes/metabolismo , ARN Largo no Codificante/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Ratones
2.
Reprod Biomed Online ; 37(5): 613-623, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30224242

RESUMEN

RESEARCH QUESTION: What is the expression pattern of long non-coding RNAs (lncRNA) in ovarian granulosa cells of women with polycystic ovary syndrome (PCOS) with or without hyperandrogenism? DESIGN: Microarray screening of lncRNA was conducted in ovarian granulosa cells collected from women with PCOS with hyperandrogenism (PCOS-T) or without hyperandrogenism (PCOS-N) and control participants, with four samples in each group. This was followed by hierarchy clustering, gene ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses. Several candidate lncRNA were randomly selected for quantitative polymerase chain reaction validation in another 54 patients. To predict the regulatory effect of lncRNA on hyperandrogenism, a co-expression network was plotted using differentially hexpressed lncRNA with statistical significance (≥ twofold; P < 0.05) in PCOS-T compared with PCOS-N. RESULTS: A total of 3000 and 1030 differentially expressed lncRNA (≥ twofold change) were detected in PCOS-T compared with control and PCOS-N, respectively. A total of 1361 differentially expressed lncRNA were detected in PCOS-N compared with controls. Corticotropin releasing hormone binding protein is consistently the up-regulated lncRNA with the highest fold-change in PCOS-T compared with either control or PCOS-N. Gene ontology and pathway analysis showed that dysregulated lncRNA in PCOS-T have a regulatory role in mitochondrial function by interacting with transcription factors such as YY1 and SIX5. CONCLUSIONS: The expression patterns of lncRNA in women with PCOS were ascertained by microarray. Many lncRNA were differentially expressed in PCOS-T compared with PCOS-N, suggesting that they may play a key role in steroid genesis and metabolism.


Asunto(s)
Células de la Granulosa/metabolismo , Hiperandrogenismo/genética , Síndrome del Ovario Poliquístico/genética , ARN Largo no Codificante/metabolismo , Adulto , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Hiperandrogenismo/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Mitocondrias/fisiología , Ovario/metabolismo , Síndrome del Ovario Poliquístico/metabolismo , ARN Largo no Codificante/fisiología
3.
Mol Med Rep ; 15(4): 1441-1454, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28259916

RESUMEN

A subset of basal cell carcinomas (BCCs) are directly derived from hair follicles (HFs). In some respects, HFs can be defined as 'ordered' skin appendage growths, while BCCs can be regarded as 'disordered' skin appendage growths. The aim of the present study was to examine HFs and BCCs to define the expression of common and unique signaling pathways in each skin appendage. Human nodular BCCs, along with HFs and non­follicular skin epithelium from normal individuals, were examined using microarrays, qPCR, and immunohistochemistry. Subsequently, BCC cells and root sheath keratinocyte cells from HFs were cultured and treated with Notch signaling peptide Jagged1 (JAG1). Gene expression, protein levels, and cell apoptosis susceptibility were assessed using qPCR, immunoblotting, and flow cytometry, respectively. Specific molecular mechanisms were found to be involved in the process of cell self­renewal in the HFs and BCCs, including Notch and Hedgehog signaling pathways. However, several key Notch signaling factors showed significant differential expression in BCCs compared with HFs. Stimulating Notch signaling with JAG1 induced apoptosis of BCC cells by increasing Fas ligand expression and downstream caspase-8 activation. The present study showed that Notch signaling pathway activity is suppressed in BCCs, and is highly expressed in HFs. Elements of the Notch pathway could, therefore, represent targets for the treatment of BCCs and potentially in hair follicle engineering.


Asunto(s)
Apoptosis , Carcinoma Basocelular/metabolismo , Carcinoma Basocelular/patología , Receptores Notch/metabolismo , Transducción de Señal , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Carcinoma Basocelular/genética , Análisis por Conglomerados , Proteína Ligando Fas/metabolismo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Ontología de Genes , Redes Reguladoras de Genes/efectos de los fármacos , Folículo Piloso/metabolismo , Folículo Piloso/patología , Humanos , Proteína Jagged-1/farmacología , Análisis de Secuencia por Matrices de Oligonucleótidos , Reproducibilidad de los Resultados , Transducción de Señal/genética , Neoplasias Cutáneas/genética
4.
Oncotarget ; 8(66): 110176-110186, 2017 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-29299139

RESUMEN

Endometriosis is a condition which involves the presence of uterine stroma and glands outside of the uterine cavity and represents one of the most prevalent disorders of the female reproductive tract. The key symptom of endometriosis is pain, including dysmenorrhea, deep dyspareunia, and chronic pelvic pain. As such, endometriosis has significant economic consequences within the healthcare system and can influence the daily quality of life in affected patients. However, the pathophysiology of this disease and the mechanisms in which this condition generates pain are very unclear. This study, involving 30 women with endometriosis and 28 controls without endometriosis, aimed to investigate relative levels of estrogen receptor alpha (ERα) splice variants in the endometrium of women with and without endometriosis and investigate potential links to the severity of pain. Wild type (wt)-ERα was dominantly expressed in human endometrium while the expression of ERα-del.4, ERα-del.7, and ERα-del.3,4 was significantly reduced in endometriosis patients compared with healthy patients (p < 0.05). Furthermore, the relative ratios of wtERα:ERα-del.4, and wtERα:ERα-del.3,4 were associated with the severity of pain in endometriosis patients (p < 0.05). Consequently, analyzing differences in the relative levels of four types of ERα splice variant in the endometrium of patients with endometriosis may help in the development of endometriosis-targeted treatment and the development of appropriate therapies.

5.
J Clin Endocrinol Metab ; 101(11): 4237-4243, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27583471

RESUMEN

CONTEXT: Although thyroid dysfunction in early pregnancy may have adverse effects on pregnancy outcome and offspring, few prospective studies have evaluated these effects. OBJECTIVE: Our aim was to evaluate the correlations between different thyroid hormone levels in early pregnancy and the incidence of gestational diabetes mellitus (GDM). SETTING AND PARTICIPANTS: The study comprised 27 513 mothers who provided early pregnancy serum samples for analyses of thyroid function. GDM was diagnosed using a 2 hours, 75-g oral glucose tolerance test, and the mothers were grouped and compared according to the results. MAIN OUTCOME MEASURES: We focused on GDM during the index pregnancy. RESULTS: The incidence of GDM in pregnant women tended to increase with age (5.83%, 10.18%, 14.95%, and 22.40%; P < .0001). The incidence of GDM increased with increasing prepregnancy body mass index (P < .0001). Pregnant women with a family history of diabetes had a much higher incidence of GDM than those without a family history of diabetes (21.09% vs 12.92%; P < .0001). The level of free T4 (FT4) in early pregnancy in GDM women was lower than that in non GDM women (P < .0001). With increasing early pregnancy FT4, the rate of incident GDM was decreasing (P < .0001). CONCLUSIONS: Low thyroid hormone levels in early pregnancy are a risk factor for GDM incidence.


Asunto(s)
Dextrotiroxina/sangre , Diabetes Gestacional/sangre , Diabetes Gestacional/epidemiología , Adulto , China/epidemiología , Femenino , Humanos , Embarazo , Primer Trimestre del Embarazo , Riesgo , Adulto Joven
6.
J Invest Dermatol ; 136(8): 1617-1626, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27094591

RESUMEN

Alopecia areata (AA) is believed to be a cell-mediated autoimmune hair loss disease. Both CD4 and cytotoxic CD8 T cells (CTLs) are important for the onset and progression of AA. Hair follicle (HF) keratinocyte and/or melanocyte antigen epitopes are suspected potential targets of autoreactive CTLs, but the specific epitopes have not yet been identified. We investigated the potential for a panel of known epitopes, expressed by HF keratinocytes and melanocytes, to induce activation of CTL populations in peripheral blood mononuclear cells. Specific synthetic epitopes derived from HF antigens trichohyalin and tyrosinase-related protein-2 induced significantly higher frequencies of response in AA CTLs compared with healthy controls (IFN-gamma secretion). Apoptosis assays revealed conditioned media from AA peripheral blood mononuclear cells stimulated with trichohyalin peptides elevated the expression of apoptosis markers in primary HF keratinocytes. A cytokine array revealed higher expression of IL-13 and chemokine ligand 5 (CCL5, RANTES) from AA peripheral blood mononuclear cells stimulated with trichohyalin peptides compared with controls. The data indicate that AA affected subjects present with an increased frequency of CTLs responsive to epitopes originating from keratinocytes and melanocytes; the activated CTLs secreted soluble factors that induced apoptosis in HF keratinocytes. Potentially, CTL response to self-antigen epitopes, particularly trichohyalin epitopes, could be a prognostic marker for human AA.


Asunto(s)
Alopecia Areata/sangre , Alopecia Areata/inmunología , Autoantígenos/inmunología , Epítopos/inmunología , Adulto , Anciano , Algoritmos , Apoptosis , Medios de Cultivo Condicionados/química , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Antígeno HLA-A2/metabolismo , Haplotipos , Humanos , Interferón gamma/metabolismo , Proteínas de Filamentos Intermediarios/metabolismo , Queratinocitos/inmunología , Queratinocitos/metabolismo , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Masculino , Melanocitos/inmunología , Melanocitos/metabolismo , Persona de Mediana Edad , Pronóstico , Adulto Joven
7.
Chin Med J (Engl) ; 129(7): 785-91, 2016 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-26996472

RESUMEN

BACKGROUND: Maternal thyroid dysfunction is common during pregnancy, and physiological changes during pregnancy can lead to the overdiagnosis of hyperthyroidism and misdiagnosis of hypothyroidism with nongestation-specific reference intervals. Our aim was to compare sequential with nonsequential methods for the evaluation of thyroid function in pregnant women. METHODS: We tested pregnant women who underwent their trimester prenatal screening at our hospital from February 2011 to September 2012 for serum thyroid stimulating hormone (TSH) and free thyroxine (FT4) using the Abbott and Roche kits. There were 447 and 200 patients enrolled in the nonsequential and sequential groups, respectively. The central 95% range between the 2.5th and the 97.5th percentiles was used as the reference interval for the thyroid function parameter. RESULTS: The nonsequential group exhibited a significantly larger degree of dispersion in the TSH reference interval during the 2nd and 3rd trimesters as measured using both the Abbott and Roche kits (all P < 0.05). The TSH reference intervals were significantly larger in the nonsequential group than in the sequential group during the 3rd trimester as measured with both the Abbott (4.95 vs. 3.77 mU/L, P < 0.001) and Roche kits (6.62 vs. 5.01 mU/L, P = 0.004). The nonsequential group had a significantly larger FT4 reference interval as measured with the Abbott kit during all trimesters (12.64 vs. 5.82 pmol/L; 7.96 vs. 4.77 pmol/L; 8.10 vs. 4.77 pmol/L, respectively, all P < 0.05), whereas a significantly larger FT4 reference interval was only observed during the 2nd trimester with the Roche kit (7.76 vs. 5.52 pmol/L, P = 0.002). CONCLUSIONS: It was more reasonable to establish reference intervals for the evaluation of maternal thyroid function using the sequential method during each trimester of pregnancy. Moreover, the exclusion of pregnancy-related complications should be considered in the inclusion criteria for thyroid function tests.


Asunto(s)
Embarazo/fisiología , Glándula Tiroides/fisiología , Femenino , Humanos , Valores de Referencia , Tirotropina/sangre , Tiroxina/sangre
8.
Cell Stem Cell ; 17(3): 273-86, 2015 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-26340527

RESUMEN

In mammals, DNA methylation is essential for protecting repetitive sequences from aberrant transcription and recombination. In some developmental contexts (e.g., preimplantation embryos) DNA is hypomethylated but repetitive elements are not dysregulated, suggesting that alternative protection mechanisms exist. Here we explore the processes involved by investigating the role of the chromatin factors Daxx and Atrx. Using genome-wide binding and transcriptome analysis, we found that Daxx and Atrx have distinct chromatin-binding profiles and are co-enriched at tandem repetitive elements in wild-type mouse ESCs. Global DNA hypomethylation further promoted recruitment of the Daxx/Atrx complex to tandem repeat sequences, including retrotransposons and telomeres. Knockdown of Daxx/Atrx in cells with hypomethylated genomes exacerbated aberrant transcriptional de-repression of repeat elements and telomere dysfunction. Mechanistically, Daxx/Atrx-mediated repression seems to involve Suv39h recruitment and H3K9 trimethylation. Our data therefore suggest that Daxx and Atrx safeguard the genome by silencing repetitive elements when DNA methylation levels are low.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Portadoras/metabolismo , ADN Helicasas/metabolismo , Metilación de ADN , Histonas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lisina/metabolismo , Proteínas Nucleares/metabolismo , Secuencias Repetidas en Tándem/genética , Animales , Sitios de Unión , Cromatina/metabolismo , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/metabolismo , Proteínas Co-Represoras , Desarrollo Embrionario , Genoma , Inestabilidad Genómica , Humanos , Metiltransferasas/metabolismo , Ratones , Ratones Noqueados , Chaperonas Moleculares , Células Madre Embrionarias de Ratones/metabolismo , Unión Proteica , Proteínas Represoras/metabolismo , Telómero/metabolismo , Transcripción Genética , Proteína Nuclear Ligada al Cromosoma X
9.
Mol Cell Endocrinol ; 412: 290-301, 2015 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-26001835

RESUMEN

Activins are homo- or heterodimers of inhibin ß subunits that play important roles in the reproductive system. Our previous work has shown that activins A (ßAßA), B (ßBßB) and AB (ßAßB) induce aromatase/estradiol, but suppress StAR/progesterone production in human granulosa-lutein cells. However, the underlying molecular determinants of these effects have not been examined. In this continuing study, we used immortalized human granulosa cells (SVOG) to investigate the effects of activins in regulating StAR/progesterone and the potential mechanisms of action. In SVOG cells, activins A, B and AB produced comparable down-regulation of StAR expression and progesterone production. In addition, all three activin isoforms induced equivalent phosphorylation of both SMAD2 and SMAD3. Importantly, the activin-induced down-regulation of StAR, increase in SMAD2/3 phosphorylation, and decrease in progesterone were abolished by the TGF-ß type I receptor inhibitor SB431542. Interestingly, the small interfering RNA-mediated knockdown of ALK4 but not ALK5 reversed the activin-induced suppression of StAR. Furthermore, the knockdown of SMAD4 or SMAD2 but not SMAD3 abolished the inhibitory effects of all three activin isoforms on StAR expression. These results provide evidence that activins A, B and AB down-regulate StAR expression and decrease progesterone production in human granulosa cells, likely via an ALK4-mediated SMAD2/SMAD4-dependent pathway. Our findings provide important insights into the molecular mechanisms underlying the regulatory effects of activins on human granulosa cell steroidogenesis.


Asunto(s)
Activinas/fisiología , Células de la Granulosa/metabolismo , Fosfoproteínas/metabolismo , Progesterona/biosíntesis , Receptores de Activinas Tipo I/fisiología , Células Cultivadas , Regulación hacia Abajo , Femenino , Regulación de la Expresión Génica , Humanos , Fosfoproteínas/genética , Proteínas Serina-Treonina Quinasas/fisiología , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Transducción de Señal , Proteína Smad2/metabolismo , Proteína smad3/metabolismo
10.
Sci Rep ; 5: 10086, 2015 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-25961186

RESUMEN

UNLABELLED: While the intrauterine environment is essential for the health of offspring, the impact of high maternal serum estradiol (E2) on lipid metabolism in offspring and the mechanisms are unknown. We found that ovarian stimulation (OS) could result in high E2 levels in women throughout pregnancy. Strikingly, their newborns showed elevated total cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C) levels that were positively related with E2 in newborns. In vitro, E2 dose-dependently stimulated TC and LDL-C secretion, and increased expression of the cholesterol synthesis rate-limiting enzyme 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) in HepG2 cells and mouse fetal hepatocytes. In vivo, high maternal E2 was detected and fetal livers also showed significantly higher HMGCR expression in an OS mouse model. Notably, an estrogen response element (ERE) was identified in the HMGCR promoter, indicating that high maternal serum E2 could up-regulate HMGCR expression in fetal hepatocytes via an ERE that in turn induces elevated levels of TC and LDL-C in offspring. CONCLUSION: OS can induce a high maternal E2 environment, which up-regulates HMGCR expression in fetal hepatocytes via an ERE in the promoter, and induces elevated levels of TC and LDL-C in newborns that may be related to increased risk of metabolic disease in adulthood.


Asunto(s)
Dislipidemias/sangre , Estradiol/sangre , Enfermedades del Recién Nacido/sangre , Complicaciones del Embarazo/sangre , Animales , LDL-Colesterol/sangre , Modelos Animales de Enfermedad , Dislipidemias/etiología , Femenino , Células Hep G2 , Humanos , Hidroximetilglutaril-CoA Reductasas/metabolismo , Recién Nacido , Enfermedades del Recién Nacido/etiología , Masculino , Ratones , Ratones Endogámicos ICR , Embarazo
11.
Stem Cells ; 33(6): 1782-93, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25802002

RESUMEN

Actl6a (actin-like protein 6A, also known as Baf53a or Arp4) is a subunit shared by multiple complexes including esBAF, INO80, and Tip60-p400, whose main components (Brg1, Ino80, and p400, respectively) are crucial for the maintenance of embryonic stem cells (ESCs). However, whether and how Actl6a functions in ESCs has not been investigated. ESCs originate from the epiblast (EPI) that is derived from the inner cell mass (ICM) in blastocysts, which also give rise to primitive endoderm (PrE). The molecular mechanisms for EPI/PrE specification remain unclear. In this study, we provide the first evidence that Actl6a can protect mouse ESCs (mESCs) from differentiating into PrE. While RNAi knockdown of Actl6a, which appeared highly expressed in mESCs and downregulated during differentiation, induced mESCs to differentiate towards the PrE lineage, ectopic expression of Actl6a was able to repress PrE differentiation. Our work also revealed that Actl6a could interact with Nanog and Sox2 and promote Nanog binding to pluripotency genes such as Oct4 and Sox2. Interestingly, cells depleted of p400, but not of Brg1 or Ino80, displayed similar PrE differentiation patterns. Mutant Actl6a with impaired ability to bind Tip60 and p400 failed to block PrE differentiation induced by Actl6a dysfunction. Finally, we showed that Actl6a could target to the promoters of key PrE regulators (e.g., Sall4 and Fgf4), repressing their expression and inhibiting PrE differentiation. Our findings uncover a novel function of Actl6a in mESCs, where it acts as a gatekeeper to prevent mESCs from entering into the PrE lineage through a Yin/Yang regulating pattern.


Asunto(s)
Actinas/metabolismo , Blastocisto/citología , Diferenciación Celular/fisiología , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/metabolismo , Endodermo/citología , Estratos Germinativos/citología , Células Madre Embrionarias de Ratones/citología , Animales , Linaje de la Célula/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Ratones , Factor 3 de Transcripción de Unión a Octámeros/metabolismo
12.
J Biol Chem ; 288(29): 20776-20784, 2013 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-23729667

RESUMEN

As a member of the Tet (Ten-eleven translocation) family proteins that can convert 5-methylcytosine (5mC) to 5-hydroxylmethylcytosine (5hmC), Tet1 has been implicated in regulating global DNA demethylation and gene expression. Tet1 is highly expressed in embryonic stem (ES) cells and appears primarily to repress developmental genes for maintaining pluripotency. To understand how Tet1 may regulate gene expression, we conducted large scale immunoprecipitation followed by mass spectrometry of endogenous Tet1 in mouse ES cells. We found that Tet1 could interact with multiple chromatin regulators, including Sin3A and NuRD complexes. In addition, we showed that Tet1 could also interact with the O-GlcNAc transferase (Ogt) and be O-GlcNAcylated. Depletion of Ogt led to reduced Tet1 and 5hmC levels on Tet1-target genes, whereas ectopic expression of wild-type but not enzymatically inactive Ogt increased Tet1 levels. Mutation of the putative O-GlcNAcylation site on Tet1 led to decreased O-GlcNAcylation and level of the Tet1 protein. Our results suggest that O-GlcNAcylation can positively regulate Tet1 protein concentration and indicate that Tet1-mediated 5hmC modification and target repression is controlled by Ogt.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/enzimología , Regulación del Desarrollo de la Expresión Génica , N-Acetilglucosaminiltransferasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Animales , Línea Celular , Cromatina/metabolismo , Proteínas de Unión al ADN/genética , Células Madre Embrionarias/citología , Genes del Desarrollo , Glicosilación , Ratones , N-Acetilglucosaminiltransferasas/antagonistas & inhibidores , Células Madre Pluripotentes/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas/genética
13.
J Biol Chem ; 288(7): 5039-48, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23239880

RESUMEN

Embryonic stem (ES) cells exhibit general characteristics of open chromatin, a state that may be necessary for ES cells to efficiently self-renew while remaining poised for differentiation. Histone H3K4 and H3K9 trimethylation associate as a general rule, with open and silenced chromatin, respectively, for ES cell pluripotency maintenance. However, how histone modifications are regulated to maintain open chromatin in ES cells remains largely unknown. Here, we demonstrate that trithorax protein Ash2l, homologue of the Drosophila Ash2 (absent, small, homeotic-2) protein, is a key regulator of open chromatin in ES cells. Consistent with Ash2l being a core subunit of mixed lineage leukemia methyltransferase complex, RNAi knockdown of Ash2l was sufficient to reduce H3K4 methylation levels and drive ES cells to a silenced chromatin state with high H3K9 trimethylation. Genome-wide ChIP-seq analysis indicated that Ash2l is recruited to target loci through two distinct modes and enriched at a family of genes implicated in open chromatin regulation, including chromatin remodeler Cdh7, transcription factor c-Myc, and H3K9 demethylase Kdm4c. Our results underscore the importance of Ash2l in open chromatin regulation and provide insight into how the open chromatin landscape is maintained in ES cells.


Asunto(s)
Cromatina/química , Proteínas de Unión al ADN/fisiología , Células Madre Embrionarias/citología , Regulación del Desarrollo de la Expresión Génica , Proteínas Nucleares/fisiología , Factores de Transcripción/fisiología , Animales , Diferenciación Celular , Cromatina/metabolismo , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/metabolismo , Genoma , Histonas/metabolismo , Metilación , Ratones , Microscopía Fluorescente/métodos , Modelos Biológicos , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas Nucleares/metabolismo , Células Madre Pluripotentes/citología , Interferencia de ARN , Factores de Transcripción/metabolismo
14.
Breast Cancer Res Treat ; 134(2): 531-41, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22576469

RESUMEN

While breast cancer mortality rate has seen a steady decline in the last few decades, advances in better treatment and diagnostic tools remain important as we come into the age of personalized therapy. In this report, we describe our studies of SGK3's role in breast cancer. SGK3 (also known as CISK) is a member of the AGC family of kinases. Our previous work indicates that SGK3 functions downstream of the PI 3-kinase cascade and shares molecular and biochemical similarities with Akt. Here, we show that SGK3 expression is linked to estrogen receptor (ER) both in breast caner cell lines and in primary tumor samples. Our analysis also indicated a positive correlation between SGK3 expression and tumor prognosis. Importantly, our immunochemistry analysis of human tumor samples established a clinical link between SGK3 expression and ER+ tumors. These findings implicate SGK3 as an additional component to a complex and heterogeneous disease, and point to the potential benefits of incorporating SGK3 into the process of breast cancer diagnosis and treatment.


Asunto(s)
Neoplasias de la Mama/metabolismo , Carcinoma Ductal de Mama/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Estrógenos/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/mortalidad , Carcinoma Ductal de Mama/patología , Línea Celular Tumoral , Supervivencia sin Enfermedad , Femenino , Expresión Génica , Humanos , Modelos Logísticos , Persona de Mediana Edad , Análisis Multivariante , Pronóstico , Modelos de Riesgos Proporcionales , Proteínas Serina-Treonina Quinasas/genética
15.
PLoS One ; 6(8): e22866, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21829661

RESUMEN

BACKGROUND: We have demonstrated that growth differentiation factor 9 (GDF9) enhances activin A-induced inhibin ß(B)-subunit mRNA levels in human granulosa-lutein (hGL) cells by regulating receptors and key intracellular components of the activin signaling pathway. However, we could not exclude its effects on follistatin (FST) and follistatin-like 3 (FSTL3), well recognized extracellular inhibitors of activin A. METHODOLOGY: hGL cells from women undergoing in vitro fertilization (IVF) treatment were cultured with and without siRNA transfection of FST, FSTL3 or GDF9 and then treated with GDF9, activin A, FST, FSTL3 or combinations. FST, FSTL3 and inhibin ß(B)-subunit mRNA, and FST, FSTL3 and inhibin B protein levels were assessed with real-time RT-PCR and ELISA, respectively. Data were log transformed before ANOVA followed by Tukey's test. PRINCIPAL FINDINGS: GDF9 suppressed basal FST and FSTL3 mRNA and protein levels in a time- and dose-dependent manner and inhibited activin A-induced FST and FSTL3 mRNA and protein expression, effects attenuated by BMPR2 extracellular domain (BMPR2 ECD), a GDF9 antagonist. After GDF9 siRNA transfection, basal and activin A-induced FST and FSTL3 mRNA and protein levels increased, but changes were reversed by adding GDF9. Reduced endogenous FST or FSTL3 expression with corresponding siRNA transfection augmented activin A-induced inhibin ß(B)-subunit mRNA levels as well as inhibin B levels (P values all <0.05). Furthermore, the enhancing effects of GDF9 in activin A-induced inhibin ß(B)-subunit mRNA and inhibin B production were attenuated by adding FST. CONCLUSION: GDF9 decreases basal and activin A-induced FST and FSTL3 expression, and this explains, in part, its enhancing effects on activin A-induced inhibin ß(B)-subunit mRNA expression and inhibin B production in hGL cells.


Asunto(s)
Proteínas Relacionadas con la Folistatina/biosíntesis , Folistatina/antagonistas & inhibidores , Folistatina/biosíntesis , Células de la Granulosa/metabolismo , Factor 9 de Diferenciación de Crecimiento/fisiología , Luteína/metabolismo , Análisis de Varianza , Secuencia de Bases , Cartilla de ADN , Femenino , Folistatina/genética , Proteínas Relacionadas con la Folistatina/genética , Técnicas de Silenciamiento del Gen , Factor 9 de Diferenciación de Crecimiento/genética , Humanos , Reacción en Cadena de la Polimerasa
16.
J Clin Endocrinol Metab ; 95(10): E172-80, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20660033

RESUMEN

BACKGROUND: We have reported that growth differentiation factor 9 (GDF9) can enhance activin A (ß(A)ß(A))-induced inhibin B (αß(B)) secretion in human granulosa-lutein (hGL) cells, but its effects on steroidogenic acute regulatory protein (StAR), ovarian steroidogenic enzymes, and progesterone production are unknown. We undertook this study to further evaluate GDF9 in this regard. METHODS: hGL cells from women undergoing in vitro fertilization treatment were cultured with and without small interfering RNA (siRNA) transfection targeted at inhibin α-subunit or GDF9 before treatment with GDF9, activin A, FSH, or combinations. We compared StAR, P450 side-chain cleavage enzyme, and 3ß-hydroxysteroid dehydrogenase expression in hGL cells and progesterone levels in culture media after these treatments. mRNA, protein, and hormone levels were assessed with real-time RT-PCR, immunoblotting, and ELISA, respectively. Data were analyzed by ANOVA followed by Tukey's test. RESULTS: Activin A alone reduced basal and FSH-induced progesterone production by decreasing the expression of StAR protein, which regulates the rate-limiting step in steroidogenesis but not P450 side-chain cleavage enzyme and 3ß-hydroxysteroid dehydrogenase. GDF9 attenuated these activin A effects on StAR and progesterone. After transfection of α-subunit siRNA, activin A level increased (P < 0.001), whereas basal and activin A-induced inhibin B levels (with and without GDF9) decreased. Furthermore, the effects of GDF9 in reversing activin A suppression of progesterone production were attenuated (P < 0.001). Transfection of GDF9 siRNA decreased GDF9 as expected and led to lower StAR expression and progesterone secretion than those observed with activin A treatment alone. CONCLUSION: GDF9 attenuates the suppressive effects of activin A on StAR expression and progesterone production by increasing the expression of inhibin B, which acts as an activin A competitor.


Asunto(s)
Activinas/antagonistas & inhibidores , Factor 9 de Diferenciación de Crecimiento/farmacología , Células Lúteas/efectos de los fármacos , Células Lúteas/metabolismo , Fosfoproteínas/genética , Progesterona/metabolismo , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Antagonismo de Drogas , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Factor 9 de Diferenciación de Crecimiento/antagonistas & inhibidores , Factor 9 de Diferenciación de Crecimiento/genética , Humanos , Inhibinas/antagonistas & inhibidores , Inhibinas/genética , Fosfoproteínas/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Transfección
17.
J Clin Endocrinol Metab ; 94(12): 5108-16, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19846738

RESUMEN

BACKGROUND: We recently reported on the effects of exogenous growth differentiation factor 9 (GDF9) in enhancing activin A-induced inhibin beta(B)-subunit mRNA and inhibin B levels in human granulosa-lutein (hGL) cells by modulating key components of the activin signaling pathway. We undertook the following study to characterize the role of endogenous GDF9 in this regard. METHODS: We compared inhibin subunit (alpha, beta(A), and beta(B)) mRNA and inhibin B levels and activation of activin receptors (ACVRs) and Smad signaling pathway in hGL cells obtained from women undergoing in vitro fertilization and cultured with and without activin A treatment after GDF9-targeting small interfering RNA transfection. GDF9, inhibin subunits, ACVR2B/1B and Smad2/3/4/7 mRNA and/or protein levels, Smad phosphorylation, and inhibin B were assessed with RT-PCR, immunoblotting, and ELISA. Data were analyzed by ANOVA followed by Tukey's test. RESULTS: GDF9 was detected as mRNA and protein in hGL cells and protein in follicular fluid from all 11 patients tested. Reduced endogenous GDF9 expression after targeting small interfering RNA transfection was associated with decreased ACVR2B/1B and Smad2/3/4 but increased inhibitory Smad7 mRNA and protein levels and, consequently, reduced activin A-induced beta(B)-subunit mRNA and inhibin B levels. CONCLUSIONS: We report here for the first time autocrine roles for endogenous GDF9 in hGL cells in enhancing activin A-induced beta(B)-subunit mRNA and inhibin B levels via key components of the activin signaling pathway. However, the relative contributions of GDF9 in granulosa cells vs. oocyte as autocrine/paracrine regulators of beta(B)-subunit and inhibin B production in normal and abnormal human ovarian functions remain to be determined.


Asunto(s)
Activinas/farmacología , Células de la Granulosa/metabolismo , Factor 9 de Diferenciación de Crecimiento/genética , Factor 9 de Diferenciación de Crecimiento/fisiología , Inhibinas/biosíntesis , Células Lúteas/metabolismo , Receptores de Activinas/efectos de los fármacos , Adulto , Western Blotting , Células Cultivadas , Femenino , Células de la Granulosa/efectos de los fármacos , Humanos , Células Lúteas/efectos de los fármacos , Ciclo Menstrual/fisiología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Transducción de Señal/fisiología , Proteínas Smad/metabolismo , Transfección
18.
Endocrinology ; 150(8): 3540-6, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19423755

RESUMEN

Activin A or growth differentiation factor 9 (GDF9) alone can increase beta(B)-mRNA level in human granulosa-lutein cells from women undergoing in vitro fertilization, but their potential interactions and related cell signaling pathways involved are unknown. We therefore compared inhibin subunit and inhibin levels and activation of activin receptors (ACVRs) and Smad signaling pathway in these human granulosa-lutein cells with and without GDF9 and/or activin A treatment. Inhibin subunit (alpha, beta(A), beta(B)), ACVR, and Smad2/3/4/7 mRNA levels, inhibin A and B production, and Smad phosphorylation were assessed by real-time RT-PCR, ELISA, and immunoblotting, respectively. Data were analyzed by ANOVA followed by Tukey's test. Activin A (1-50 ng/ml) or GDF9 (1-200 ng/ml) alone had only little stimulatory effects on alpha- and beta(A)-mRNA levels. In contrast, GDF9 could stimulate beta(B)-subunit levels but to a lesser degree than the dose- and time-dependent effects of activin A. Compared with untreated cells, GDF9 pretreatment for 24 h significantly enhanced activin A-induced beta(B)-mRNA levels, inhibin B secretion, and Smad2/3 phosphorylation (effects attenuated by bone morphogenetic protein receptor 2 extracellular domain, a GDF9 antagonist); and induced ACVR2B/1B and Smad2/3 but reduced Smad7 (an inhibitory Smad) mRNA levels. We report here for the first time that GDF9 enhances cell response to activin A by modulating key components of the activin signaling pathway in regulating inhibin subunits and hence inhibin B production in human granulosa-lutein cells.


Asunto(s)
Activinas/farmacología , Células de la Granulosa/efectos de los fármacos , Células de la Granulosa/metabolismo , Factor 9 de Diferenciación de Crecimiento/farmacología , Inhibinas/metabolismo , Receptores de Activinas/genética , Western Blotting , Células Cultivadas , Femenino , Humanos , Inhibinas/genética , Fosforilación/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína smad3/genética , Proteína smad3/metabolismo , Proteína smad7/genética , Proteína smad7/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...