Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Epigenetics ; 19(1): 2374979, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38970823

RESUMEN

TET1/2/3 dioxygenases iteratively demethylate 5-methylcytosine, beginning with the formation of 5-hydroxymethylcytosine (5hmC). The post-mitotic brain maintains higher levels of 5hmC than most peripheral tissues, and TET1 ablation studies have underscored the critical role of TET1 in brain physiology. However, deletion of Tet1 precludes the disentangling of the catalytic and non-catalytic functions of TET1. Here, we dissect these functions of TET1 by comparing adult cortex of Tet1 wildtype (Tet1 WT), a novel Tet1 catalytically dead mutant (Tet1 HxD), and Tet1 knockout (Tet1 KO) mice. Using DNA methylation array, we uncover that Tet1 HxD and KO mutations perturb the methylation status of distinct subsets of CpG sites. Gene ontology (GO) analysis on specific differential 5hmC regions indicates that TET1's catalytic activity is linked to neuronal-specific functions. RNA-Seq further shows that Tet1 mutations predominantly impact the genes that are associated with alternative splicing. Lastly, we performed High-performance Liquid Chromatography Mass-Spectrometry lipidomics on WT and mutant cortices and uncover accumulation of lysophospholipids lysophosphatidylethanolamine and lysophosphatidylcholine in Tet1 HxD cortex. In summary, we show that Tet1 HxD does not completely phenocopy Tet1 KO, providing evidence that TET1 modulates distinct cortical functions through its catalytic and non-catalytic roles.


Asunto(s)
5-Metilcitosina , Corteza Cerebral , Metilación de ADN , Proteínas Proto-Oncogénicas , Animales , Ratones , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , 5-Metilcitosina/metabolismo , 5-Metilcitosina/análogos & derivados , Corteza Cerebral/metabolismo , Ratones Noqueados , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Islas de CpG , Mutación
2.
Hepatology ; 78(2): 486-502, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-36037289

RESUMEN

BACKGROUND AND AIMS: Assessing mammalian gene function in vivo has traditionally relied on manipulation of the mouse genome in embryonic stem cells or perizygotic embryos. These approaches are time-consuming and require extensive breeding when simultaneous mutations in multiple genes is desired. The aim of this study is to introduce a rapid in vivo multiplexed editing (RIME) method and provide proof of concept of this system. APPROACH AND RESULTS: RIME, a system wherein CRISPR/caspase 9 technology, paired with adeno-associated viruses (AAVs), permits the inactivation of one or more genes in the adult mouse liver. The method is quick, requiring as little as 1 month from conceptualization to knockout, and highly efficient, enabling editing in >95% of target cells. To highlight its use, we used this system to inactivate, alone or in combination, genes with functions spanning metabolism, mitosis, mitochondrial maintenance, and cell proliferation. CONCLUSIONS: RIME enables the rapid, efficient, and inexpensive analysis of multiple genes in the mouse liver in vivo .


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Ratones , Animales , Edición Génica/métodos , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Hígado , Mamíferos
3.
Mol Cell ; 82(19): 3613-3631.e7, 2022 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-36108632

RESUMEN

Allele-specific expression of imprinted gene clusters is governed by gametic DNA methylation at master regulators called imprinting control regions (ICRs). Non-gametic or secondary differentially methylated regions (DMRs) at promoters and exonic regions reinforce monoallelic expression but do not control an entire cluster. Here, we unveil an unconventional secondary DMR that is indispensable for tissue-specific imprinting of two previously unlinked genes, Grb10 and Ddc. Using polymorphic mice, we mapped an intronic secondary DMR at Grb10 with paternal-specific CTCF binding (CBR2.3) that forms contacts with Ddc. Deletion of paternal CBR2.3 removed a critical insulator, resulting in substantial shifting of chromatin looping and ectopic enhancer-promoter contacts. Destabilized gene architecture precipitated abnormal Grb10-Ddc expression with developmental consequences in the heart and muscle. Thus, we redefine the Grb10-Ddc imprinting domain by uncovering an unconventional intronic secondary DMR that functions as an insulator to instruct the tissue-specific, monoallelic expression of multiple genes-a feature previously ICR exclusive.


Asunto(s)
Impresión Genómica , ARN Largo no Codificante , Alelos , Animales , Cromatina/genética , Metilación de ADN , Proteína Adaptadora GRB10/genética , Corazón , Ratones
4.
Mol Cell ; 81(4): 859-869.e8, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33352108

RESUMEN

Active DNA demethylation via ten-eleven translocation (TET) family enzymes is essential for epigenetic reprogramming in cell state transitions. TET enzymes catalyze up to three successive oxidations of 5-methylcytosine (5mC), generating 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC), or 5-carboxycytosine (5caC). Although these bases are known to contribute to distinct demethylation pathways, the lack of tools to uncouple these sequential oxidative events has constrained our mechanistic understanding of the role of TETs in chromatin reprogramming. Here, we describe the first application of biochemically engineered TET mutants that unlink 5mC oxidation steps, examining their effects on somatic cell reprogramming. We show that only TET enzymes proficient for oxidation to 5fC/5caC can rescue the reprogramming potential of Tet2-deficient mouse embryonic fibroblasts. This effect correlated with rapid DNA demethylation at reprogramming enhancers and increased chromatin accessibility later in reprogramming. These experiments demonstrate that DNA demethylation through 5fC/5caC has roles distinct from 5hmC in somatic reprogramming to pluripotency.


Asunto(s)
5-Metilcitosina/metabolismo , Reprogramación Celular , Proteínas de Unión al ADN/metabolismo , Embrión de Mamíferos/metabolismo , Elementos de Facilitación Genéticos , Epigénesis Genética , Fibroblastos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Proteínas de Unión al ADN/genética , Dioxigenasas , Embrión de Mamíferos/citología , Fibroblastos/citología , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Mutación , Células 3T3 NIH , Proteínas Proto-Oncogénicas/genética
5.
Physiol Rep ; 7(19): e14244, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31609547

RESUMEN

Epigenetic dysregulation of long noncoding RNA H19 was recently found to be associated with calcific aortic valve disease (CAVD) in humans by repressing NOTCH1 transcription. This finding offers a possible epigenetic explanation for the abundance of cases of CAVD that are not explained by any clear genetic mutation. In this study, we examined the effect of age and sex on epigenetic dysregulation of H19 and subsequent aortic stenosis. Cohorts of littermate, wild-type C57BL/6 mice were studied at developmental ages analogous to human middle age through advanced age. Cardiac and aortic valve function were assessed with M-mode echocardiography and pulsed wave Doppler ultrasound, respectively. Bisulfite sequencing was used to determine methylation-based epigenetic regulation of H19, and RT-PCR was used to determine changes in gene expression profiles. Male mice were found to have higher peak systolic velocities than females, with several of the oldest mice showing signs of early aortic stenosis. The imprinting control region of H19 was not hypomethylated with age, and H19 expression was lower in the aortic valves of older mice than in the youngest group. These results suggest that age-related upregulation of H19 is not observed in murine aortic valves and that other factors may initiate H19-related CAVD in humans.


Asunto(s)
Estenosis de la Válvula Aórtica/genética , Válvula Aórtica/patología , Calcinosis/genética , Metilación de ADN/genética , ARN Largo no Codificante/genética , Envejecimiento , Animales , Válvula Aórtica/metabolismo , Estenosis de la Válvula Aórtica/metabolismo , Calcinosis/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Largo no Codificante/metabolismo , Factores Sexuales , Regulación hacia Arriba
6.
Sci Rep ; 9(1): 2535, 2019 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-30796277

RESUMEN

Imatinib is an oral chemotherapeutic used primarily to treat chronic myeloid leukemia (CML) and gastrointestinal stromal tumors (GIST). The potential effects of cancer treatments on a patient's future fertility  are a major concern affecting the quality of life for cancer survivors. The effects of imatinib on future fertility are unknown. It is teratogenic. Therefore, patients are advised to stop treatment before pregnancy. Unfortunately, CML and GIST have high rates of recurrence in the absence of the drug, therefore halting imatinib during pregnancy endangers the mother. Possible long-term (post-treatment) effects of imatinib on reproduction have not been studied. We have used a mouse model to examine the effects of imatinib on the placenta and implantation after long-term imatinib exposure. We found significant changes in epigenetic markers of key imprinted genes in the placenta. There was a significant decrease in the labyrinth zone and vasculature of the placenta, which could impact fetal growth later in pregnancy. These effects on placental growth occurred even when imatinib was stopped prior to pregnancy. These results indicate potential long-term effects of imatinib on pregnancy and implantation. A prolonged wash-out period prior to pregnancy or extra monitoring for possible placental insufficiency may be advisable.


Asunto(s)
Implantación del Embrión/efectos de los fármacos , Embrión de Mamíferos/efectos de los fármacos , Mesilato de Imatinib/efectos adversos , Placentación/efectos de los fármacos , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/uso terapéutico , Femenino , Tumores del Estroma Gastrointestinal/tratamiento farmacológico , Mesilato de Imatinib/uso terapéutico , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Ratones , Modelos Animales , Embarazo
7.
Proc Natl Acad Sci U S A ; 115(44): E10370-E10378, 2018 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-30322909

RESUMEN

The success of mammalian development following fertilization depends on a series of transient increases in egg cytoplasmic Ca2+, referred to as Ca2+ oscillations. Maintenance of these oscillations requires Ca2+ influx across the plasma membrane, which is mediated in part by T-type, CaV3.2 channels. Here we show using genetic mouse models that TRPM7 channels are required to support this Ca2+ influx. Eggs lacking both TRPM7 and CaV3.2 stop oscillating prematurely, indicating that together they are responsible for the majority of Ca2+ influx immediately following fertilization. Fertilized eggs lacking both channels also frequently display delayed resumption of Ca2+ oscillations, which appears to require sperm-egg fusion. TRPM7 and CaV3.2 channels almost completely account for Ca2+ influx observed following store depletion, a process previously attributed to canonical store-operated Ca2+ entry mediated by STIM/ORAI interactions. TRPM7 serves as a membrane sensor of extracellular Mg2+ and Ca2+ concentrations and mediates the effects of these ions on Ca2+ oscillation frequency. When bred to wild-type males, female mice carrying eggs lacking TRPM7 and CaV3.2 are subfertile, and their offspring have increased variance in postnatal weight. These in vivo findings confirm previous observations linking in vitro experimental alterations in Ca2+ oscillatory patterns with developmental potential and offspring growth. The identification of TRPM7 and CaV3.2 as key mediators of Ca2+ influx following fertilization provides a mechanistic basis for the rational design of culture media that optimize developmental potential in research animals, domestic animals, and humans.


Asunto(s)
Canales de Calcio Tipo T/metabolismo , Señalización del Calcio/fisiología , Calcio/metabolismo , Fertilización/fisiología , Canales Catiónicos TRPM/metabolismo , Cigoto/metabolismo , Animales , Membrana Celular/metabolismo , Citoplasma/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Oocitos/metabolismo , Espermatozoides/metabolismo , Molécula de Interacción Estromal 1/metabolismo
8.
Horm Behav ; 102: 93-104, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29763587

RESUMEN

Fetal exposure to endocrine disrupting chemicals (EDCs) has been associated with adverse neurobehavioral outcomes across the lifespan and can persist across multiple generations of offspring. However, the underlying mechanisms driving these changes are not well understood. We investigated the molecular perturbations associated with EDC-induced behavioral changes in first (F1) and second (F2) filial generations, using the model EDC bisphenol A (BPA). C57BL/6J dams were exposed to BPA from preconception until lactation through the diet at doses (10 µg/kg bw/d-lower dose or 10 mg/kg bw/d-upper dose) representative of human exposure levels. As adults, F1 male offspring exhibited increased depressive-like behavior, measured by the forced swim test, while females were unaffected. These behavioral changes were limited to the F1 generation and were not associated with altered maternal care. Transcriptome analysis by RNA-sequencing in F1 control and upper dose BPA-exposed adult male hippocampus revealed neurotransmitter systems as major pathways disrupted by developmental BPA exposure. High performance liquid chromatography demonstrated a male-specific reduction in hippocampal serotonin. Administration of the selective serotonin reuptake inhibitor fluoxetine (20 mg/kg bw) rescued the depressive-like phenotype in males exposed to lower, but not upper, dose BPA, suggesting distinct mechanisms of action for each exposure dose. Finally, high resolution mass spectrometry revealed reduced circulating levels of the neuroactive steroid dehydroepiandrosterone in BPA-exposed males, suggesting another potential mechanism underlying the depressive-like phenotype. Thus, behavioral changes associated with early life BPA exposure may be mediated by sex-specific disruptions in the serotonergic system and/or sex steroid biogenesis in male offspring.


Asunto(s)
Conducta Animal/efectos de los fármacos , Compuestos de Bencidrilo/farmacología , Depresión/inducido químicamente , Disruptores Endocrinos/farmacología , Hipotálamo/efectos de los fármacos , Conducta Materna/efectos de los fármacos , Fenoles/farmacología , Efectos Tardíos de la Exposición Prenatal/psicología , Animales , Fármacos del Sistema Nervioso Central/metabolismo , Depresión/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Hipotálamo/metabolismo , Hipotálamo/fisiología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Neurotransmisores/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Esteroides/metabolismo
10.
Biol Reprod ; 90(2): 22, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24337315

RESUMEN

Assisted reproductive technologies (ART) have enabled millions of couples with compromised fertility to conceive children. Nevertheless, there is a growing concern regarding the safety of these procedures due to an increased incidence of imprinting disorders, premature birth, and low birth weight in ART-conceived offspring. An integral aspect of ART is the oxygen concentration used during in vitro development of mammalian embryos, which is typically either atmospheric (~20%) or reduced (5%). Both oxygen tension levels have been widely used, but 5% oxygen improves preimplantation development in several mammalian species, including that of humans. To determine whether a high oxygen tension increases the frequency of epigenetic abnormalities in mouse embryos subjected to ART, we measured DNA methylation and expression of several imprinted genes in both embryonic and placental tissues from concepti generated by in vitro fertilization (IVF) and exposed to 5% or 20% oxygen during culture. We found that placentae from IVF embryos exhibit an increased frequency of abnormal methylation and expression profiles of several imprinted genes, compared to embryonic tissues. Moreover, IVF-derived placentae exhibit a variety of epigenetic profiles at the assayed imprinted genes, suggesting that these epigenetic defects arise by a stochastic process. Although culturing embryos in both of the oxygen concentrations resulted in a significant increase of epigenetic defects in placental tissues compared to naturally conceived controls, we did not detect significant differences between embryos cultured in 5% and those cultured in 20% oxygen. Thus, further optimization of ART should be considered to minimize the occurrence of epigenetic errors in the placenta.


Asunto(s)
Aberraciones Cromosómicas/estadística & datos numéricos , Técnicas de Cultivo de Embriones , Impresión Genómica , Enfermedades Placentarias/genética , Placenta/metabolismo , Técnicas Reproductivas Asistidas/efectos adversos , Animales , Blastocisto/citología , Aberraciones Cromosómicas/embriología , Técnicas de Cultivo de Embriones/estadística & datos numéricos , Embrión de Mamíferos , Epigénesis Genética , Femenino , Incidencia , Masculino , Ratones , Ratones Endogámicos C57BL , Placenta/patología , Enfermedades Placentarias/patología , Embarazo , Técnicas Reproductivas Asistidas/estadística & datos numéricos , Procesos Estocásticos
11.
Genetics ; 192(3): 1095-107, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22887817

RESUMEN

During the development of female mammals, one of the two X chromosomes is inactivated, serving as a dosage-compensation mechanism to equalize the expression of X-linked genes in females and males. While the choice of which X chromosome to inactivate is normally random, X chromosome inactivation can be skewed in F1 hybrid mice, as determined by alleles at the X chromosome controlling element (Xce), a locus defined genetically by Cattanach over 40 years ago. Four Xce alleles have been defined in inbred mice in order of the tendency of the X chromosome to remain active: Xce(a) < Xce(b) < Xce(c) < Xce(d). While the identity of the Xce locus remains unknown, previous efforts to map sequences responsible for the Xce effect in hybrid mice have localized the Xce to candidate regions that overlap the X chromosome inactivation center (Xic), which includes the Xist and Tsix genes. Here, we have intercrossed 129S1/SvImJ, which carries the Xce(a) allele, and Mus musculus castaneus EiJ, which carries the Xce(c) allele, to generate recombinant lines with single or double recombinant breakpoints near or within the Xce candidate region. In female progeny of 129S1/SvImJ females mated to recombinant males, we have measured the X chromosome inactivation ratio using allele-specific expression assays of genes on the X chromosome. We have identified regions, both proximal and distal to Xist/Tsix, that contribute to the choice of which X chromosome to inactivate, indicating that multiple elements on the X chromosome contribute to the Xce.


Asunto(s)
Inactivación del Cromosoma X , Cromosoma X , Alelos , Animales , Cruzamiento , Femenino , Genes Ligados a X , Masculino , Ratones , Sitios de Carácter Cuantitativo
12.
Dev Biol ; 355(2): 349-57, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21600199

RESUMEN

Expression of coregulated imprinted genes, H19 and Igf2, is monoallelic and parent-of-origin-dependent. Like most imprinted genes, H19 and Igf2 are regulated by a differentially methylated imprinting control region (ICR). CTCF binding sites and DNA methylation at the ICR have previously been identified as key cis-acting elements required for proper H19/Igf2 imprinting. Here, we use mouse models to elucidate further the mechanism of ICR-mediated gene regulation. We specifically address the question of whether sequences outside of CTCF sites at the ICR are required for paternal H19 repression. To this end, we generated two types of mutant ICRs in the mouse: (i) deletion of intervening sequence between CTCF sites (H19(ICR∆IVS)), which changes size and CpG content at the ICR; and (ii) CpG depletion outside of CTCF sites (H19(ICR-8nrCG)), which only changes CpG content at the ICR. Individually, both mutant alleles (H19(ICR∆IVS) and H19(ICR-8nrCG)) show loss of imprinted repression of paternal H19. Interestingly, this loss of repression does not coincide with a detectable change in methylation at the H19 ICR or promoter. Thus, neither intact CTCF sites nor hypermethylation at the ICR is sufficient for maintaining the fully repressed state of the paternal H19 allele. Our findings demonstrate, for the first time in vivo, that sequence outside of CTCF sites at the ICR is required in cis for ICR-mediated imprinted repression at the H19/Igf2 locus. In addition, these results strongly implicate a novel role of ICR size and CpG density in paternal H19 repression.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Impresión Genómica/fisiología , ARN no Traducido/metabolismo , Elementos Reguladores de la Transcripción/fisiología , Proteínas Represoras/metabolismo , Animales , Southern Blotting , Factor de Unión a CCCTC , Cruzamientos Genéticos , Metilación de ADN/genética , Cartilla de ADN/genética , Electroporación , Vectores Genéticos/genética , Impresión Genómica/genética , Patrón de Herencia/genética , Ratones , Ratones Endogámicos C57BL , Mutagénesis Sitio-Dirigida , ARN Largo no Codificante , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Eliminación de Secuencia/genética
13.
Mol Cell Biol ; 30(16): 3916-28, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20547750

RESUMEN

Imprinted genes are expressed in a monoallelic, parent-of-origin-specific manner. Clusters of imprinted genes are regulated by imprinting control regions (ICRs) characterized by DNA methylation of one allele. This methylation is critical for imprinting; a reduction in the DNA methyltransferase DNMT1 causes a widespread loss of imprinting. To better understand the role of DNA methylation in the regulation of imprinting, we characterized the effects of Dnmt1 mutations on the expression of a panel of imprinted genes in the embryo and placenta. We found striking differences among imprinted domains. The Igf2 and Peg3 domains showed imprinting perturbations with both null and partial loss-of-function mutations, and both domains had pairs of coordinately regulated genes with opposite responses to loss of DNMT1 function, suggesting these domains employ similar regulatory mechanisms. Genes in the Kcnq1 domain were less sensitive to the absence of DNMT1. Cdkn1c exhibited imprinting perturbations only in null mutants, while Kcnq1 and Ascl2 were largely unaffected by a loss of DNMT1 function. These results emphasize the critical role for DNA methylation in imprinting and reveal the different ways it controls gene expression.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/deficiencia , Metilación de ADN , Impresión Genómica , Alelos , Animales , Secuencia de Bases , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Cruzamientos Genéticos , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/química , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Cartilla de ADN/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Factor II del Crecimiento Similar a la Insulina/genética , Canal de Potasio KCNQ1/genética , Factores de Transcripción de Tipo Kruppel/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Familia de Multigenes , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Placenta/metabolismo , Embarazo , Estructura Terciaria de Proteína
14.
Genetics ; 164(4): 1481-94, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12930754

RESUMEN

The mammalian epigenetic phenomena of X inactivation and genomic imprinting are incompletely understood. X inactivation equalizes X-linked expression between males and females by silencing genes on one X chromosome during female embryogenesis. Genomic imprinting functionally distinguishes the parental genomes, resulting in parent-specific monoallelic expression of particular genes. N-ethyl-N-nitrosourea (ENU) mutagenesis was used in the mouse to screen for mutations in novel factors involved in X inactivation. Previously, we reported mutant pedigrees identified through this screen that segregate aberrant X-inactivation phenotypes and we mapped the mutation in one pedigree to chromosome 15. We now have mapped two additional mutations to the distal chromosome 5 and the proximal chromosome 10 in a second pedigree and show that each of the mutations is sufficient to induce the mutant phenotype. We further show that the roles of these factors are specific to embryonic X inactivation as neither genomic imprinting of multiple genes nor imprinted X inactivation is perturbed. Finally, we used mice bearing selected X-linked alleles that regulate X chromosome choice to demonstrate that the phenotypes of all three mutations are consistent with models in which the mutations have affected molecules involved specifically in the choice or the initiation of X inactivation.


Asunto(s)
Alquilantes/farmacología , Epigénesis Genética , Etilnitrosourea/farmacología , Ratones/genética , Mutagénesis , Mutación , Alelos , Animales , Mapeo Cromosómico , Cromosomas de los Mamíferos , Compensación de Dosificación (Genética) , Relación Dosis-Respuesta a Droga , Femenino , Fertilidad , Expresión Génica , Silenciador del Gen , Impresión Genómica , Masculino , Ratones/embriología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos , Modelos Genéticos , Linaje , Cromosoma X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA