Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Genes Dev ; 34(15-16): 1039-1050, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32561546

RESUMEN

The FoxA transcription factors are critical for liver development through their pioneering activity, which initiates a highly complex regulatory network thought to become progressively resistant to the loss of any individual hepatic transcription factor via mutual redundancy. To investigate the dispensability of FoxA factors for maintaining this regulatory network, we ablated all FoxA genes in the adult mouse liver. Remarkably, loss of FoxA caused rapid and massive reduction in the expression of critical liver genes. Activity of these genes was reduced back to the low levels of the fetal prehepatic endoderm stage, leading to necrosis and lethality within days. Mechanistically, we found FoxA proteins to be required for maintaining enhancer activity, chromatin accessibility, nucleosome positioning, and binding of HNF4α. Thus, the FoxA factors act continuously, guarding hepatic enhancer activity throughout adult life.


Asunto(s)
Factores de Transcripción Forkhead/fisiología , Redes Reguladoras de Genes , Hígado/metabolismo , Animales , Sitios de Unión , Cromatina/metabolismo , Elementos de Facilitación Genéticos , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Factor Nuclear 3-alfa del Hepatocito/genética , Factor Nuclear 3-beta del Hepatocito/genética , Factor Nuclear 3-gamma del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/metabolismo , Hígado/patología , Fallo Hepático/etiología , Fallo Hepático/patología , Masculino , Ratones , Nucleosomas
2.
Development ; 149(2)2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-35051273

RESUMEN

Although it is well known that DNA methylation serves to repress gene expression, precisely how it functions during the process of development remains unclear. Here, we propose that the overall pattern of DNA methylation established in the early embryo serves as a sophisticated mechanism for maintaining a genome-wide network of gene regulatory elements in an inaccessible chromatin structure throughout the body. As development progresses, programmed demethylation in each cell type then provides the specificity for maintaining select elements in an open structure. This allows these regulatory elements to interact with a large range of transcription factors and thereby regulate the gene expression profiles that define cell identity.


Asunto(s)
Metilación de ADN , Regulación del Desarrollo de la Expresión Génica , Animales , Ensamble y Desensamble de Cromatina , Humanos
3.
Genes Dev ; 29(9): 923-33, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25934504

RESUMEN

DNA methylation patterns are set up in a relatively fixed programmed manner during normal embryonic development and are then stably maintained. Using genome-wide analysis, we discovered a postnatal pathway involving gender-specific demethylation that occurs exclusively in the male liver. This demodification is programmed to take place at tissue-specific enhancer sequences, and our data show that the methylation state at these loci is associated with and appears to play a role in the transcriptional regulation of nearby genes. This process is mediated by the secretion of testosterone at the time of sexual maturity, but the resulting methylation profile is stable and therefore can serve as an epigenetic memory even in the absence of this inducer. These findings add a new dimension to our understanding of the role of DNA methylation in vivo and provide the foundations for deciphering how environment can impact on the epigenetic regulation of genes in general.


Asunto(s)
Metilación de ADN , Epigénesis Genética/genética , Hígado/metabolismo , Andrógenos/farmacología , Animales , Castración , Metilación de ADN/efectos de los fármacos , Elementos de Facilitación Genéticos/genética , Epigénesis Genética/efectos de los fármacos , Femenino , Regulación del Desarrollo de la Expresión Génica , Estudio de Asociación del Genoma Completo , Histonas/genética , Histonas/metabolismo , Humanos , Hígado/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Caracteres Sexuales , Testosterona/metabolismo , Testosterona/farmacología
4.
Proc Natl Acad Sci U S A ; 113(18): 5018-23, 2016 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-27091986

RESUMEN

There is ample evidence that somatic cell differentiation during development is accompanied by extensive DNA demethylation of specific sites that vary between cell types. Although the mechanism of this process has not yet been elucidated, it is likely to involve the conversion of 5mC to 5hmC by Tet enzymes. We show that a Tet2/Tet3 conditional knockout at early stages of B-cell development largely prevents lineage-specific programmed demethylation events. This lack of demethylation affects the expression of nearby B-cell lineage genes by impairing enhancer activity, thus causing defects in B-cell differentiation and function. Thus, tissue-specific DNA demethylation appears to be necessary for proper somatic cell development in vivo.


Asunto(s)
Linfocitos B/citología , Linfocitos B/fisiología , Metilación de ADN/genética , Proteínas de Unión al ADN/genética , Epigénesis Genética/genética , Animales , Diferenciación Celular/genética , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Especificidad de Órganos/genética
5.
PLoS Genet ; 8(2): e1002477, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22383887

RESUMEN

Fundamental aspects of embryonic and post-natal development, including maintenance of the mammalian female germline, are largely unknown. Here we employ a retrospective, phylogenetic-based method for reconstructing cell lineage trees utilizing somatic mutations accumulated in microsatellites, to study female germline dynamics in mice. Reconstructed cell lineage trees can be used to estimate lineage relationships between different cell types, as well as cell depth (number of cell divisions since the zygote). We show that, in the reconstructed mouse cell lineage trees, oocytes form clusters that are separate from hematopoietic and mesenchymal stem cells, both in young and old mice, indicating that these populations belong to distinct lineages. Furthermore, while cumulus cells sampled from different ovarian follicles are distinctly clustered on the reconstructed trees, oocytes from the left and right ovaries are not, suggesting a mixing of their progenitor pools. We also observed an increase in oocyte depth with mouse age, which can be explained either by depth-guided selection of oocytes for ovulation or by post-natal renewal. Overall, our study sheds light on substantial novel aspects of female germline preservation and development.


Asunto(s)
Envejecimiento , Linaje de la Célula/genética , Células Germinativas , Envejecimiento/genética , Animales , Femenino , Células Germinativas/citología , Células Germinativas/metabolismo , Mutación de Línea Germinal , Células Madre Mesenquimatosas/citología , Ratones , Oogénesis/genética , Especificidad de Órganos , Ovario/citología , Ovario/fisiología , Ovulación
6.
PLoS Comput Biol ; 9(11): e1003297, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24244121

RESUMEN

Organism cells proliferate and die to build, maintain, renew and repair it. The cellular history of an organism up to any point in time can be captured by a cell lineage tree in which vertices represent all organism cells, past and present, and directed edges represent progeny relations among them. The root represents the fertilized egg, and the leaves represent extant and dead cells. Somatic mutations accumulated during cell division endow each organism cell with a genomic signature that is unique with a very high probability. Distances between such genomic signatures can be used to reconstruct an organism's cell lineage tree. Cell populations possess unique features that are absent or rare in organism populations (e.g., the presence of stem cells and a small number of generations since the zygote) and do not undergo sexual reproduction, hence the reconstruction of cell lineage trees calls for careful examination and adaptation of the standard tools of population genetics. Our lab developed a method for reconstructing cell lineage trees by examining only mutations in highly variable microsatellite loci (MS, also called short tandem repeats, STR). In this study we use experimental data on somatic mutations in MS of individual cells in human and mice in order to validate and quantify the utility of known lineage tree reconstruction algorithms in this context. We employed extensive measurements of somatic mutations in individual cells which were isolated from healthy and diseased tissues of mice and humans. The validation was done by analyzing the ability to infer known and clear biological scenarios. In general, we found that if the biological scenario is simple, almost all algorithms tested can infer it. Another somewhat surprising conclusion is that the best algorithm among those tested is Neighbor Joining where the distance measure used is normalized absolute distance. We include our full dataset in Tables S1, S2, S3, S4, S5 to enable further analysis of this data by others.


Asunto(s)
Algoritmos , Linaje de la Célula/genética , Repeticiones de Microsatélite/genética , Mutación/genética , Filogenia , Animales , Células de la Médula Ósea , Células Cultivadas , Análisis por Conglomerados , Biología Computacional/métodos , Simulación por Computador , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Modelos Genéticos
7.
PLoS Genet ; 7(7): e1002192, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21829376

RESUMEN

Stem cell dynamics in vivo are often being studied by lineage tracing methods. Our laboratory has previously developed a retrospective method for reconstructing cell lineage trees from somatic mutations accumulated in microsatellites. This method was applied here to explore different aspects of stem cell dynamics in the mouse colon without the use of stem cell markers. We first demonstrated the reliability of our method for the study of stem cells by confirming previously established facts, and then we addressed open questions. Our findings confirmed that colon crypts are monoclonal and that, throughout adulthood, the process of monoclonal conversion plays a major role in the maintenance of crypts. The absence of immortal strand mechanism in crypts stem cells was validated by the age-dependent accumulation of microsatellite mutations. In addition, we confirmed the positive correlation between physical and lineage proximity of crypts, by showing that the colon is separated into small domains that share a common ancestor. We gained new data demonstrating that colon epithelium is clustered separately from hematopoietic and other cell types, indicating that the colon is constituted of few progenitors and ruling out significant renewal of colonic epithelium from hematopoietic cells during adulthood. Overall, our study demonstrates the reliability of cell lineage reconstruction for the study of stem cell dynamics, and it further addresses open questions in colon stem cells. In addition, this method can be applied to study stem cell dynamics in other systems.


Asunto(s)
Linaje de la Célula , Colon/citología , Células Madre/citología , Animales , Linfocitos B/metabolismo , Linaje de la Célula/genética , Colon/metabolismo , Epitelio/metabolismo , Estudio de Asociación del Genoma Completo , Células Madre Hematopoyéticas , Células Secretoras de Insulina/metabolismo , Mucosa Intestinal/metabolismo , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Páncreas/citología , Páncreas/metabolismo , Células Madre/metabolismo
8.
FASEB J ; 26(11): 4495-505, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22859367

RESUMEN

Completion of the first meiotic division, manifested by extrusion of the first polar body (PBI), depends on proteasomal degradation of cyclin B1 and securin and the subsequent respective CDK1 inactivation and chromosome segregation. We aimed at identifying the polyubiquitin signal that mediates proteasomal action and at a better characterization of the role of CDK1 inactivation at this stage of meiosis. Microinjections of mutated ubiquitin proteins into mouse oocytes revealed that interference with lysine-11 polyubiquitin chains abrogated chromosome segregation and reduced PBI extrusion by 63% as compared to WT ubiquitin-injected controls. Inactivation of CDK1 in oocytes arrested at first metaphase by a proteasome inhibitor fully rescued PBI extrusion. However, removal of CDK1 inhibition failed to allow progression to the second metaphase, rather, inducing PBI reengulfment in 62% of the oocytes. Inhibition of either PLK1 or MEK1/2 during the first anaphase changed spindle dimensions. The PLK1 inhibitor also blocked PBI emission and prevented RhoA translocation. Our results identified lysine-11 rather than the canonic lysine-48 ubiquitin chains as the degradation signal in oocytes resuming meiosis, further disclosing that CDK1 inactivation is necessary and sufficient for PBI emission. This information significantly contributes to our understanding of faulty chromosome segregation that may lead to aneuploidy.


Asunto(s)
Proteína Quinasa CDC2/metabolismo , Oocitos/citología , Oocitos/metabolismo , Cuerpos Polares/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Animales , Proteína Quinasa CDC2/genética , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Segregación Cromosómica , Citocinesis , Femenino , Regulación Enzimológica de la Expresión Génica , Sistema de Señalización de MAP Quinasas , Meiosis/fisiología , Ratones , Cuerpos Polares/citología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Securina , Transducción de Señal , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo , Quinasa Tipo Polo 1
9.
Nat Commun ; 14(1): 6708, 2023 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-37872177

RESUMEN

Telomeres, the ends of eukaryotic chromosomes, protect genome integrity and enable cell proliferation. Maintaining optimal telomere length in the germline and throughout life limits the risk of cancer and enables healthy aging. Telomeres in the house mouse, Mus musculus, are about five times longer than human telomeres, limiting the use of this common laboratory animal for studying the contribution of telomere biology to aging and cancer. We identified a key amino acid variation in the helicase RTEL1, naturally occurring in the short-telomere mouse species M. spretus. Introducing this variation into M. musculus is sufficient to reduce the telomere length set point in the germline and generate mice with human-length telomeres. While these mice are fertile and appear healthy, the regenerative capacity of their colonic epithelium is compromised. The engineered Telomouse reported here demonstrates a dominant role of RTEL1 in telomere length regulation and provides a unique model for aging and cancer.


Asunto(s)
Genoma , Neoplasias , Humanos , Ratones , Animales , Modelos Animales de Enfermedad , Telómero/genética , Proliferación Celular , Neoplasias/genética , ADN Helicasas/genética
10.
Cells ; 11(8)2022 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-35455969

RESUMEN

Vascular endothelial growth factor A (VEGF-A) is a secreted protein that stimulates angiogenesis in response to hypoxia. Under hypoxic conditions, a non-canonical long isoform called L-VEGF is concomitantly expressed with VEGF-A. Once translated, L-VEGF is proteolytically cleaved to generate N-VEGF and VEGF-A. Interestingly, while VEGF-A is secreted and affects the surrounding cells, N-VEGF is mobilized to the nucleus. This suggests that N-VEGF participates in transcriptional response to hypoxia. In this study, we performed a series of complementary experiments to examine the functional role of N-VEGF. Strikingly, we found that the mere expression of N-VEGF followed by its hypoxia-independent mobilization to the nucleus was sufficient to induce key genes associated with angiogenesis, such as Hif1α,VEGF-A isoforms, as well as genes associated with cell survival under hypoxia. Complementarily, when N-VEGF was genetically depleted, key hypoxia-induced genes were downregulated and cells were significantly susceptible to hypoxia-mediated apoptosis. This is the first report of N-VEGF serving as an autoregulatory arm of VEGF-A. Further experiments will be needed to determine the role of N-VEGF in cancer and embryogenesis.


Asunto(s)
Neovascularización Patológica , Factor A de Crecimiento Endotelial Vascular , Apoptosis , Hipoxia de la Célula , Humanos , Hipoxia , Neovascularización Patológica/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
11.
Dev Cell ; 56(5): 602-612.e4, 2021 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-33636105

RESUMEN

Tissue-specific DNA methylation patterns are created by transcription factors that recruit methylation and demethylation enzymes to cis-regulatory elements. To date, it is not known whether transcription factors are needed to continuously maintain methylation profiles in development and mature tissues or whether they only establish these marks during organ development. We queried the role of the pioneer factor FoxA in generating hypomethylated DNA at liver enhancers. We discovered a set of FoxA-binding sites that undergo regional, FoxA-dependent demethylation during organ development. Conditional ablation of FoxA genes in the adult liver demonstrated that continued FoxA presence was not required to maintain the hypomethylated state, even when massive cell proliferation was induced. This study provides strong evidence for the stable, epigenetic nature of tissue-specific DNA methylation patterns directed by lineage-determining transcription factors during organ development.


Asunto(s)
Diferenciación Celular , Metilación de ADN , Elementos de Facilitación Genéticos , Epigénesis Genética , Factor Nuclear 3-alfa del Hepatocito/fisiología , Factor Nuclear 3-beta del Hepatocito/fisiología , Hígado/metabolismo , Animales , Sitios de Unión , Cromatina/química , Cromatina/genética , Cromatina/metabolismo , Desmetilación , Regulación del Desarrollo de la Expresión Génica , Hígado/embriología , Masculino , Ratones , Ratones Noqueados
12.
Nat Cancer ; 2(10): 1055-1070, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-35121883

RESUMEN

Stochastic transition of cancer cells between drug-sensitive and drug-tolerant persister phenotypes has been proposed to play a key role in non-genetic resistance to therapy. Yet, we show here that cancer cells actually possess a highly stable inherited chance to persist (CTP) during therapy. This CTP is non-stochastic, determined pre-treatment and has a unimodal distribution ranging from 0 to almost 100%. Notably, CTP is drug specific. We found that differential serine/threonine phosphorylation of the insulin receptor substrate 1 (IRS1) protein determines the CTP of lung and of head and neck cancer cells under epidermal growth factor receptor inhibition, both in vitro and in vivo. Indeed, the first-in-class IRS1 inhibitor NT219 was highly synergistic with anti-epidermal growth factor receptor therapy across multiple in vitro and in vivo models. Elucidation of drug-specific mechanisms that determine the degree and stability of cellular CTP may establish a framework for the elimination of cancer persisters, using new rationally designed drug combinations.


Asunto(s)
Receptores ErbB , Neoplasias , Receptores ErbB/genética , Proteínas Sustrato del Receptor de Insulina/genética , Fosforilación , Probabilidad
13.
Dev Biol ; 313(1): 1-12, 2008 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-18005958

RESUMEN

Gap junctions, predominantly comprising connexin43 (Cx43), mediate cell-to-cell communication within the ovarian follicle. However, the partaking of Cx43 in the formation of the gap junction channels, between the oocyte and the somatic cells, is controversial. We addressed this dispute by crossing females that carry a Cx43 coding region, flanked by loxP recognition sites, with males expressing the Cre recombinase under the control of Zp3 promoter. Oocytes of the resultant Zp3Cre;Gja1(lox/lox) mice did not express Cx43 and were referred to as Cx43(del/del). Unexpectedly, a decrease in Cx43 was observed in cumulus/granulosa cells of some follicles as well. Nevertheless, no histological abnormalities were detected in the ovaries of the Zp3Cre;Gja1(lox/lox) mice. Furthermore, these mice ovulated normally and developed fully functional corpora lutea. Additionally, the ovarian Cx43(del/del) oocytes were meiotically arrested and transferred Lucifer yellow to the surrounding cumulus cells. However, mating Zp3Cre;Gja1(lox/lox) females with wild-type males resulted in a reduced rate of parturition and a substantial decrease in litter size. Further examination revealed that although preimplantation development of Zp3Cre;Gja1(lox/+) embryos was normal, the blactocysts exhibited impaired implantation. Our data suggest that total ablation of Cx43 in the oocyte, combined with its decrease in the surrounding somatic cells, allows normal oogenesis and folliculogenesis, ovulation and early embryonic development but severely impairs the implantation capacity of the resulting blactocysts.


Asunto(s)
Conexina 43/metabolismo , Infertilidad , Oocitos/metabolismo , Animales , Implantación del Embrión , Embrión de Mamíferos/metabolismo , Femenino , Uniones Comunicantes/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
14.
PLoS Comput Biol ; 4(4): e1000058, 2008 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-18404205

RESUMEN

The depth of a cell of a multicellular organism is the number of cell divisions it underwent since the zygote, and knowing this basic cell property would help address fundamental problems in several areas of biology. At present, the depths of the vast majority of human and mouse cell types are unknown. Here, we show a method for estimating the depth of a cell by analyzing somatic mutations in its microsatellites, and provide to our knowledge for the first time reliable depth estimates for several cells types in mice. According to our estimates, the average depth of oocytes is 29, consistent with previous estimates. The average depth of B cells ranges from 34 to 79, linearly related to the mouse age, suggesting a rate of one cell division per day. In contrast, various types of adult stem cells underwent on average fewer cell divisions, supporting the notion that adult stem cells are relatively quiescent. Our method for depth estimation opens a window for revealing tissue turnover rates in animals, including humans, which has important implications for our knowledge of the body under physiological and pathological conditions.


Asunto(s)
Linfocitos B/citología , Linfocitos B/fisiología , Senescencia Celular/genética , Análisis Mutacional de ADN/métodos , Células Híbridas/fisiología , Repeticiones de Microsatélite/genética , Análisis de Secuencia de ADN/métodos , Animales , Secuencia de Bases , Células Cultivadas , Ratones , Datos de Secuencia Molecular
15.
Nat Commun ; 9(1): 2040, 2018 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-29795194

RESUMEN

Development in mammals is accompanied by specific de novo and demethylation events that are thought to stabilize differentiated cell phenotypes. We demonstrate that a large percentage of the tissue-specific methylation pattern is generated postnatally. Demethylation in the liver is observed in thousands of enhancer-like sequences associated with genes that undergo activation during the first few weeks of life. Using. conditional gene ablation strategy we show that the removal of these methyl groups is stable and necessary for assuring proper hepatocyte gene expression and function through its effect on chromatin accessibility. These postnatal changes in methylation come about through exposure to hormone signaling. These results define the molecular rules of 5-methyl-cytosine regulation as an epigenetic mechanism underlying cellular responses to. changing environment.


Asunto(s)
Desmetilación del ADN , Epigénesis Genética/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Hígado/crecimiento & desarrollo , Transducción de Señal/fisiología , 5-Metilcitosina/metabolismo , Animales , Animales Recién Nacidos , Células Cultivadas , Proteínas de Unión al ADN/genética , Dioxigenasas , Femenino , Hepatocitos/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Hígado/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Cultivo Primario de Células , Proteínas Proto-Oncogénicas/genética , Análisis de Secuencia de ARN
16.
PLoS One ; 6(10): e25605, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22022423

RESUMEN

Myofiber cultures give rise to myogenic as well as to non-myogenic cells. Whether these myofiber-associated non-myogenic cells develop from resident stem cells that possess mesenchymal plasticity or from other stem cells such as mesenchymal stem cells (MSCs) remain unsolved. To address this question, we applied a method for reconstructing cell lineage trees from somatic mutations to MSCs and myogenic and non-myogenic cells from individual myofibers that were cultured at clonal density.Our analyses show that (i) in addition to myogenic progenitors, myofibers also harbor non-myogenic progenitors of a distinct, yet close, lineage; (ii) myofiber-associated non-myogenic and myogenic cells share the same muscle-bound primordial stem cells of a lineage distinct from bone marrow MSCs; (iii) these muscle-bound primordial stem-cells first part to individual muscles and then differentiate into myogenic and non-myogenic stem cells.


Asunto(s)
Linaje de la Célula , Desarrollo de Músculos , Fibras Musculares Esqueléticas/citología , Células Madre/citología , Adipogénesis/genética , Envejecimiento/fisiología , Animales , Adhesión Celular , Diferenciación Celular , Forma de la Célula , Células Clonales , Análisis por Conglomerados , Regulación de la Expresión Génica , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Desarrollo de Músculos/genética , Fibras Musculares Esqueléticas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Células Madre/metabolismo
17.
Mol Endocrinol ; 24(2): 402-11, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20009084

RESUMEN

Mammalian reproduction depends on the release of a mature oocyte from the ovarian follicle. Maturation of the oocyte and rupture of the follicle wall constitute part of the responses to the preovulatory surge of LH, which also include cumulus expansion and granulosa cell luteinization. It was previously shown that the epidermal growth factor receptor (EGFR) mediates the ovulatory response to LH in the ovarian follicle. We hypothesized that it is a sustained activity of the EGFR that generates oocyte maturation and cumulus expansion. We demonstrated that, whereas a transient exposure of rat isolated, intact, preovulatory follicles to either LH or forskolin was sufficient to induce oocyte maturation and cumulus expansion, these LH-induced responses were only generated upon a prolonged activity of the EGFR. In addition, the continuous activity of the EGFR is essential for the chronic phosphorylation of the ERK1/2 downstream signaling molecules, which were shown to be essential for oocyte maturation and cumulus expansion. Interestingly, EGFR-sustained activity was also necessary to maintain the up-regulation of Ptgs2, a gene essential for cumulus expansion. The unusual prolonged duration of ERK1/2 activity may possibly be attributed to the late induction of the ERK-specific phosphatase 3, demonstrated herein. These new data shed light on the unique characteristics of EGFR-ERK1/2 activity in the ovarian follicle and emphasize the fact that the ovulatory process involves a nonclassical activation of this pathway.


Asunto(s)
Células del Cúmulo/fisiología , Receptores ErbB/metabolismo , Hormona Luteinizante/fisiología , Oocitos/fisiología , Oogénesis/fisiología , Adenilil Ciclasas , Animales , Colforsina/farmacología , Células del Cúmulo/efectos de los fármacos , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Activadores de Enzimas/farmacología , Receptores ErbB/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Isoenzimas/genética , Isoenzimas/metabolismo , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/genética , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/metabolismo , Oocitos/efectos de los fármacos , Oogénesis/efectos de los fármacos , Folículo Ovárico/fisiología , Fosforilación/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Factores de Tiempo
18.
Endocrinology ; 151(2): 755-65, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19996184

RESUMEN

Completion of the first meiosis in oocytes is achieved by the extrusion of the first polar body (PBI), a particular example of cell division. In mitosis, the small GTPase RhoA, which is activated by epithelial cell transforming protein 2 (ECT2), orchestrates contractile ring constriction, thus enabling cytokinesis. However, the involvement of this pathway in mammalian oocytes has not been established. To characterize the role of ECT2 in PBI emission in mouse oocytes, the small interfering RNA approach was employed. We found that ECT2 depletion significantly reduces PBI emission, induces first metaphase arrest, and generates oocytes containing two properly formed spindles of the second metaphase. Moreover, we describe, for the first time, that before PBI emission, RhoA forms a ring that is preceded by a dome-like accumulation at the oocyte cortex, next to the spindle. This unique mode of RhoA translocation failed to occur in the absence of ECT2. We further found that the Rho-dependent kinase, a main RhoA effector, is essential for PBI emission. In addition, we demonstrate herein that ECT2 is subjected to phosphorylation/dephosphorylation throughout meiosis in oocytes and further reveal that PBI emission is temporally associated with ECT2 dephosphorylation. Our data provide the first demonstration that an active cyclin-dependent kinase 1, the catalytic subunit of the maturation-promoting factor, phosphorylates ECT2 during the first meiotic metaphase and that cyclin-dependent kinase 1 inactivation at anaphase allows ECT2 dephosphorylation. In conclusion, our study demonstrates the indispensable role of the maturation-promoting factor/ECT2/RhoA pathway in PBI extrusion in mouse oocytes.


Asunto(s)
Metafase/fisiología , Oocitos/citología , Proteínas Proto-Oncogénicas/deficiencia , Huso Acromático/fisiología , Animales , Proteína Quinasa CDC2/metabolismo , Técnicas de Cultivo de Célula , ADN Complementario/genética , Femenino , Gonadotropinas Equinas/farmacología , Ratones , Ratones Endogámicos C57BL , Oocitos/fisiología , Folículo Ovárico/efectos de los fármacos , Folículo Ovárico/fisiología , Fosforilación , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/fisiología , ARN/genética , ARN/aislamiento & purificación , ARN Interferente Pequeño/genética , Proteína de Unión al GTP rhoA/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA