Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
J Cell Sci ; 134(14)2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34152366

RESUMEN

Oocyte-specific knockdown of pericentrin (PCNT) in transgenic (Tg) mice disrupts acentriolar microtubule-organizing center (aMTOC) formation, leading to spindle instability and error-prone meiotic division. Here, we show that PCNT-depleted oocytes lack phosphorylated Aurora A (pAURKA) at spindle poles, while overall levels are unaltered. To test aMTOC-associated AURKA function, metaphase II (MII) control (WT) and Tg oocytes were briefly exposed to a specific AURKA inhibitor (MLN8237). Similar defects were observed in Tg and MLN8237-treated WT oocytes, including altered spindle structure, increased chromosome misalignment and impaired microtubule regrowth. Yet, AURKA inhibition had a limited effect on Tg oocytes, revealing a critical role for aMTOC-associated AURKA in regulating spindle stability. Notably, spindle instability was associated with disrupted γ-tubulin and lack of the liquid-like meiotic spindle domain (LISD) in Tg oocytes. Analysis of this Tg model provides the first evidence that LISD assembly depends expressly on aMTOC-associated AURKA, and that Ran-mediated spindle formation ensues without the LISD. These data support that loss of aMTOC-associated AURKA and failure of LISD assembly contribute to error-prone meiotic division in PCNT-depleted oocytes, underscoring the essential role of aMTOCs for spindle stability.


Asunto(s)
Aurora Quinasa A , Centro Organizador de los Microtúbulos , Huso Acromático , Animales , Aurora Quinasa A/genética , Meiosis , Ratones , Oocitos , Huso Acromático/genética , Polos del Huso/genética
2.
Reproduction ; 159(3): 261-274, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31895686

RESUMEN

Acentriolar microtubule-organizing centers (aMTOCs) play a critical role in stable meiotic spindle assembly in oocytes, necessary for accurate chromosome segregation. Yet, there is a limited understanding of the essential regulatory components of these unique MTOCs. In somatic cells, CEP215 (Centrosomal Protein 215) serves as an important regulator of centrosome maturation and spindle organization. Here, we assessed whether it has a similar function in mouse oocytes. CEP215 was detected in oocyte lysates and specifically localized to aMTOCs throughout the progression of meiosis in a pericentrin-dependent manner. Super-resolution microscopy revealed CEP215 co-localization with pericentrin and a unique pore/ring-like structural organization of aMTOCs. Interestingly, inhibition of Aurora Kinase A in either MI or MII-stage oocytes resulted in a striking loss of the ring-like aMTOC organization and pronounced CEP215 clustering at spindle poles, as well as shorter spindles with highly focused poles. In vitro siRNA-mediated transcript knockdown effectively reduced CEP215 in approximately 85% of the oocytes. Maturation rates to MII were similar in the Cep215 siRNA and injected controls; however, a high percentage (~40%) of the Cep215-knockdown oocytes showed notable variations in spindle pole focusing. Surprisingly, pericentrin and γ-tubulin localization and fluorescence intensity at aMTOCs were unaltered in knockdown oocytes, contrasting with mitotic cells where CEP215 depletion reduced γ-tubulin at centrosomes. Our results demonstrate that CEP215 is a functional component of oocyte aMTOCs and participates in the regulation of meiotic spindle pole focusing. Moreover, these studies reveal a vital role for Aurora Kinase A activity in the maintenance of aMTOC organization in oocytes.


Asunto(s)
Aurora Quinasa A/metabolismo , Proteínas de Ciclo Celular/metabolismo , Meiosis , Centro Organizador de los Microtúbulos/metabolismo , Oocitos/fisiología , Animales , Antígenos/metabolismo , Femenino , Masculino , Ratones Endogámicos C57BL , Tubulina (Proteína)/metabolismo
3.
Development ; 142(10): 1806-17, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25926359

RESUMEN

A striking proportion of human cleavage-stage embryos exhibit chromosome instability (CIN). Notably, until now, no experimental model has been described to determine the origin and mechanisms of complex chromosomal rearrangements. Here, we examined mouse embryos deficient for the chromatin remodeling protein ATRX to determine the cellular mechanisms activated in response to CIN. We demonstrate that ATRX is required for silencing of major satellite transcripts in the maternal genome, where it confers epigenetic asymmetry to pericentric heterochromatin during the transition to the first mitosis. This stage is also characterized by a striking kinetochore size asymmetry established by differences in CENP-C protein between the parental genomes. Loss of ATRX results in increased centromeric mitotic recombination, a high frequency of sister chromatid exchanges and double strand DNA breaks, indicating the formation of mitotic recombination break points. ATRX-deficient embryos exhibit a twofold increase in transcripts for aurora kinase B, the centromeric cohesin ESCO2, DNMT1, the ubiquitin-ligase (DZIP3) and the histone methyl transferase (EHMT1). Thus, loss of ATRX activates a pathway that integrates epigenetic modifications and DNA repair in response to chromosome breaks. These results reveal the cellular response of the cleavage-stage embryo to CIN and uncover a mechanism by which centromeric fission induces the formation of large-scale chromosomal rearrangements. Our results have important implications to determine the epigenetic origins of CIN that lead to congenital birth defects and early pregnancy loss, as well as the mechanisms involved in the oocyte to embryo transition.


Asunto(s)
ADN Helicasas/metabolismo , Proteínas Nucleares/metabolismo , Acetiltransferasas/genética , Acetiltransferasas/metabolismo , Animales , Centrómero/genética , Ensamble y Desensamble de Cromatina/genética , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Helicasas/genética , Epigénesis Genética/genética , Epigénesis Genética/fisiología , Heterocromatina/genética , Heterocromatina/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Cinetocoros/metabolismo , Meiosis/genética , Meiosis/fisiología , Ratones , Proteínas Nucleares/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Proteína Nuclear Ligada al Cromosoma X
4.
Reproduction ; 150(4): 297-310, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26224097

RESUMEN

Endometriosis is associated with infertility and debilitating chronic pain. Abnormal epigenetic modifications in the human endometrium have recently been implicated in the pathogenesis of this condition. However, whether an altered epigenetic landscape contributes to pathological changes in the ovary is unknown. Using an established baboon endometriosis model, early-, and late-stage epigenetic changes in the ovary were investigated. Transcript profiling of key chromatin-modifying enzymes using pathway-focused PCR arrays on ovarian tissue from healthy control animals and at 3 and 15 months of endometriosis revealed dramatic changes in gene expression in a disease duration-dependent manner. Ingenuity Pathway Analysis indicated that transcripts for chromatin-remodeling enzymes associated with reproductive system disease and cancer development were abnormally regulated, most prominently the arginine methyltransferases CARM1, PRMT2, and PRMT8. Downregulation of CARM1 protein expression was also detected in the ovary, fully-grown oocytes and eutopic endometrium following 15 months of endometriosis. Sodium bisulfite sequencing revealed DNA hypermethylation within the PRMT8 promoter, suggesting that deregulated CpG methylation may play a role in transcriptional repression of this gene. These results demonstrate that endometriosis is associated with changes of epigenetic profiles in the primate ovary and suggest that arginine methyltransferases play a prominent role in mediating the ovarian response to endometriosis. Owing to the critical role of CARM1 in nuclear receptor-mediated transcription and maintenance of pluripotency in the cleavage stage embryo, our results suggest that epigenetic alterations in the ovary may have functional consequences for oocyte quality and the etiology of infertility associated with endometriosis.


Asunto(s)
Endometriosis/genética , Endometriosis/fisiopatología , Epigénesis Genética/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Ovario/enzimología , Papio , Proteína-Arginina N-Metiltransferasas/genética , Animales , Cromatina/metabolismo , Secuencia Conservada , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN , Endometrio/enzimología , Femenino , Heterocromatina/genética , Humanos , Infertilidad/genética , Isoenzimas/genética , Oocitos/enzimología , Embarazo , Activación Transcripcional
5.
Nat Cell Biol ; 8(12): 1448-54, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17115026

RESUMEN

Lymphoid specific helicase (Lsh) is a major epigenetic regulator that is essential for DNA methylation and transcriptional silencing of parasitic elements in the mammalian genome. However, whether Lsh is involved in the regulation of chromatin-mediated processes during meiosis is not known. Here, we show that Lsh is essential for the completion of meiosis and transcriptional repression of repetitive elements in the female gonad. Oocytes from Lsh knockout mice exhibit demethylation of transposable elements and tandem repeats at pericentric heterochromatin, as well as incomplete chromosome synapsis associated with persistent RAD51 foci and gammaH2AX phosphorylation. Failure to load crossover-associated foci results in the generation of non-exchange chromosomes. The severe oocyte loss observed and lack of ovarian follicle formation, together with the patterns of Lsh nuclear compartmentalization in the germ line, demonstrate that Lsh has a critical and previously unidentified role in epigenetic gene silencing and maintenance of genomic stability during female meiosis.


Asunto(s)
Emparejamiento Cromosómico , Cromosomas de los Mamíferos/metabolismo , ADN Helicasas/metabolismo , Silenciador del Gen , Meiosis , Oocitos/citología , Retroelementos/genética , Animales , ADN Helicasas/genética , Metilación de ADN , Femenino , Regulación de la Expresión Génica , Heterocromatina/metabolismo , Ratones , Ovario/citología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Recombinación Genética , Secuencias Repetitivas de Ácidos Nucleicos , Cromosoma X/metabolismo
6.
PLoS Genet ; 6(9): e1001137, 2010 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-20885787

RESUMEN

The α-thalassemia/mental retardation X-linked protein (ATRX) is a chromatin-remodeling factor known to regulate DNA methylation at repetitive sequences of the human genome. We have previously demonstrated that ATRX binds to pericentric heterochromatin domains in mouse oocytes at the metaphase II stage where it is involved in mediating chromosome alignment at the meiotic spindle. However, the role of ATRX in the functional differentiation of chromatin structure during meiosis is not known. To test ATRX function in the germ line, we developed an oocyte-specific transgenic RNAi knockdown mouse model. Our results demonstrate that ATRX is required for heterochromatin formation and maintenance of chromosome stability during meiosis. During prophase I arrest, ATRX is necessary to recruit the transcriptional regulator DAXX (death domain associated protein) to pericentric heterochromatin. At the metaphase II stage, transgenic ATRX-RNAi oocytes exhibit abnormal chromosome morphology associated with reduced phosphorylation of histone 3 at serine 10 as well as chromosome segregation defects leading to aneuploidy and severely reduced fertility. Notably, a large proportion of ATRX-depleted oocytes and 1-cell stage embryos exhibit chromosome fragments and centromeric DNA-containing micronuclei. Our results provide novel evidence indicating that ATRX is required for centromere stability and the epigenetic control of heterochromatin function during meiosis and the transition to the first mitosis.


Asunto(s)
Aneuploidia , Blastocisto/metabolismo , Centrómero/metabolismo , Inestabilidad Cromosómica/genética , ADN Helicasas/deficiencia , Proteínas Nucleares/deficiencia , Oocitos/metabolismo , Animales , Blastocisto/citología , Proteínas Portadoras/metabolismo , Segregación Cromosómica/genética , Cromosomas de los Mamíferos/metabolismo , Proteínas Co-Represoras , ADN Helicasas/genética , ADN Helicasas/metabolismo , Femenino , Fertilidad/genética , Técnicas de Silenciamiento del Gen , Heterocromatina/metabolismo , Histonas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones Transgénicos , Mitosis/genética , Modelos Animales , Chaperonas Moleculares , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oocitos/citología , Especificidad de Órganos/genética , Fosforilación , Interferencia de ARN , Proteína Nuclear Ligada al Cromosoma X
7.
Open Biol ; 13(11): 230133, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37935356

RESUMEN

In somatic cells, mitotic transcription of major satellite non-coding RNAs is tightly regulated and essential for heterochromatin formation and the maintenance of genome integrity. We recently demonstrated that major satellite transcripts are expressed, and chromatin-bound during mouse oocyte meiosis. Pericentric satellite RNAs are also expressed in human oocytes. However, the specific biological function(s) during oocyte meiosis remain to be established. Here, we use validated locked nucleic acid gapmers for major satellite RNA depletion followed by live cell imaging, and superresolution analysis to determine the role of pericentric non-coding RNAs during female meiosis. Depletion of satellite RNA induces mesoscale changes in pericentric heterochromatin structure leading to chromosome instability, kinetochore attachment errors and abnormal chromosome alignment. Chromosome misalignment is associated with spindle defects, microtubule instability and, unexpectedly, loss of acentriolar microtubule organizing centre (aMTOC) tethering to spindle poles. Pericentrin fragmentation and failure to assemble ring-like aMTOCs with loss of associated polo-like kinase 1 provide critical insight into the mechanisms leading to impaired spindle pole integrity. Inhibition of transcription or RNA splicing phenocopies the chromosome alignment errors and spindle defects, suggesting that pericentric transcription during oocyte meiosis is required to regulate heterochromatin structure, chromosome segregation and maintenance of spindle organization.


Asunto(s)
Heterocromatina , Huso Acromático , Ratones , Femenino , Humanos , Animales , Huso Acromático/genética , Polos del Huso , Meiosis/genética , Oocitos , Inestabilidad Cromosómica , Satélite de ARN , Segregación Cromosómica
8.
Commun Biol ; 6(1): 1258, 2023 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-38086992

RESUMEN

The mechanisms leading to changes in mesoscale chromatin organization during cellular aging are unknown. Here, we used transcriptional activator-like effectors, RNA-seq and superresolution analysis to determine the effects of genotoxic stress on oocyte chromatin structure. Major satellites are organized into tightly packed globular structures that coalesce into chromocenters and dynamically associate with the nucleolus. Acute irradiation significantly enhanced chromocenter mobility in transcriptionally inactive oocytes. In transcriptionally active oocytes, irradiation induced a striking unfolding of satellite chromatin fibers and enhanced the expression of transcripts required for protection from oxidative stress (Fermt1, Smg1), recovery from DNA damage (Tlk2, Rad54l) and regulation of heterochromatin assembly (Zfp296, Ski-oncogene). Non-irradiated, senescent oocytes exhibit not only high chromocenter mobility and satellite distension but also a high frequency of extra chromosomal satellite DNA. Notably, analysis of biological aging using an oocyte-specific RNA clock revealed cellular communication, posttranslational protein modifications, chromatin and histone dynamics as the top cellular processes that are dysregulated in both senescent and irradiated oocytes. Our results indicate that unfolding of heterochromatin fibers following acute genotoxic stress or cellular aging induced the formation of distended satellites and that abnormal chromatin structure together with increased chromocenter mobility leads to chromosome instability in senescent oocytes.


Asunto(s)
Heterocromatina , Oocitos , Animales , Heterocromatina/genética , Heterocromatina/metabolismo , Cromatina/genética , Cromatina/metabolismo , Histonas/metabolismo , Ensamble y Desensamble de Cromatina , Mamíferos/genética
9.
Chromosoma ; 120(3): 227-44, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21274552

RESUMEN

Pairing of the sex chromosomes during mammalian meiosis is characterized by the formation of a unique heterochromatin structure at the XY body. The mechanisms underlying the formation of this nuclear domain are reportedly highly conserved from marsupials to mammals. In this study, we demonstrate that in contrast to all eutherian species studied to date, partial synapsis of the heterologous sex chromosomes during pachytene stage in the horse is not associated with the formation of a typical macrochromatin domain at the XY body. While phosphorylated histone H2AX (γH2AX) and macroH2A1.2 are present as a diffuse signal over the entire macrochromatin domain in mouse pachytene spermatocytes, γH2AX, macroH2A1.2, and the cohesin subunit SMC3 are preferentially enriched at meiotic sex chromosome cores in equine spermatocytes. Moreover, although several histone modifications associated with this nuclear domain in the mouse such as H3K4me2 and ubH2A are conspicuously absent in the equine XY body, prominent RNA polymerase II foci persist at the sex chromosomes. Thus, the localization of key marker proteins and histone modifications associated with the XY body in the horse differs significantly from all other mammalian systems described. These results demonstrate that the epigenetic landscape and heterochromatinization of the equine XY body might be regulated by alternative mechanisms and that some features of XY body formation may be evolutionary divergent in the domestic horse. We propose equine spermatogenesis as a unique model system for the study of the regulatory networks leading to the epigenetic control of gene expression during XY body formation.


Asunto(s)
Cromatina/química , Emparejamiento Cromosómico , Caballos/genética , Cromosomas Sexuales/genética , Espermatogénesis/genética , Animales , Proteínas Cromosómicas no Histona/metabolismo , Epigenómica , Heterocromatina/genética , Histonas/genética , Ratones , Miosis/genética , Fase Paquiteno/genética
10.
Cells ; 11(20)2022 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-36291100

RESUMEN

The endocrine disrupting activity of bisphenol compounds is well documented, but less is known regarding their impact on cell division and early embryo formation. Here, we tested the effects of acute in vitro exposure to bisphenol A (BPA) and its common substitute, bisphenol F (BPF), during critical stages of mouse pre-implantation embryo development, including the first mitotic division, cell polarization, as well as morula and blastocyst formation. Timing of initial cleavage was determined by live-cell imaging, while subsequent divisions, cytoskeletal organization and lineage marker labeling were assessed by high-resolution fluorescence microscopy. Our analysis reveals that brief culture with BPA or BPF impeded cell division and disrupted embryo development at all stages tested. Surprisingly, BPF was more detrimental to the early embryo than BPA. Notably, poor embryo development was associated with cytoskeletal disruptions of the actomyosin network, apical domain formation during cell polarization, actin ring zippering for embryo sealing and altered cell lineage marker profiles. These results underscore that bisphenols can disrupt cytoskeletal integrity and remodeling that is vital for early embryo development and raise concerns regarding the use of BPF as a 'safe' BPA substitute.


Asunto(s)
Compuestos de Bencidrilo , Blastocisto , Citoesqueleto , Fenoles , Animales , Ratones , Actinas/metabolismo , Actomiosina/metabolismo , Blastocisto/efectos de los fármacos , Blastocisto/ultraestructura , Compuestos de Bencidrilo/toxicidad , Fenoles/toxicidad , Citoesqueleto/efectos de los fármacos , Citoesqueleto/ultraestructura
11.
Biol Reprod ; 84(6): 1235-41, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21349825

RESUMEN

Lymphoid-specific helicase (HELLS; also known as LSH) is a member of the SNF2 family of chromatin remodeling proteins. Because Hells-null mice die at birth, a phenotype in male meiosis cannot be studied in these animals. Allografting of testis tissue from Hells(-/-) to wild-type mice was employed to study postnatal germ cell differentiation. Testes harvested at Day 18.5 of gestation from Hells(-/-), Hells(+/-), and Hells(+/+) mice were grafted ectopically to immunodeficient mice. Bromodeoxyuridine incorporation at 1 wk postgrafting revealed fewer dividing germ cells in grafts from Hells(-/-) than from Hells(+/+) mice. Whereas spermatogenesis proceeded through meiosis with round spermatids in grafts from Hells heterozygote and wild-type donor testes, spermatogenesis arrested at stage IV, and midpachytene spermatocytes were the most advanced germ cell type in grafts from Hells(-/-) mice at 4, 6, and 8 wk after grafting. Analysis of meiotic configurations at 22 days posttransplantation revealed an increase in Hells(-/-) spermatocytes with abnormal chromosome synapsis. These results indicate that in the absence of HELLS, proliferation of spermatogonia is reduced and germ cell differentiation arrested at the midpachytene stage, implicating an essential role for HELLS during male meiosis. This study highlights the utility of testis tissue grafting to study spermatogenesis in animal models that cannot reach sexual maturity.


Asunto(s)
ADN Helicasas/metabolismo , Meiosis , Espermatocitos/citología , Espermatocitos/metabolismo , Animales , ADN Helicasas/genética , Regulación Enzimológica de la Expresión Génica , Masculino , Ratones , Espermatogénesis/fisiología
12.
Reproduction ; 142(2): 221-34, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21653732

RESUMEN

Functional differentiation of chromatin structure is essential for the control of gene expression, nuclear architecture, and chromosome stability. Compelling evidence indicates that alterations in chromatin remodeling proteins play an important role in the pathogenesis of human disease. Among these, α-thalassemia mental retardation X-linked protein (ATRX) has recently emerged as a critical factor involved in heterochromatin formation at mammalian centromeres and telomeres as well as facultative heterochromatin on the murine inactive X chromosome. Mutations in human ATRX result in an X-linked neurodevelopmental condition with various degrees of gonadal dysgenesis (ATRX syndrome). Patients with ATRX syndrome may exhibit skewed X chromosome inactivation (XCI) patterns, and ATRX-deficient mice exhibit abnormal imprinted XCI in the trophoblast cell line. Non-random or skewed XCI can potentially affect both the onset and severity of X-linked disease. Notably, failure to establish epigenetic modifications associated with the inactive X chromosome (Xi) results in several conditions that exhibit genomic and chromosome instability such as fragile X syndrome as well as cancer development. Insight into the molecular mechanisms of ATRX function and its interacting partners in different tissues will no doubt contribute to our understanding of the pathogenesis of ATRX syndrome as well as the epigenetic origins of aneuploidy. In turn, this knowledge will be essential for the identification of novel drug targets and diagnostic tools for cancer progression as well as the therapeutic management of global epigenetic changes commonly associated with malignant neoplastic transformation.


Asunto(s)
Cromatina/química , Cromatina/metabolismo , Inestabilidad Cromosómica , ADN Helicasas/fisiología , Enfermedades Genéticas Ligadas al Cromosoma X/metabolismo , Proteínas Nucleares/fisiología , Animales , Ensamble y Desensamble de Cromatina , Progresión de la Enfermedad , Epigénesis Genética , Femenino , Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/fisiopatología , Enfermedades Genéticas Ligadas al Cromosoma X/fisiopatología , Heterocromatina/química , Heterocromatina/metabolismo , Humanos , Masculino , Discapacidad Intelectual Ligada al Cromosoma X/metabolismo , Discapacidad Intelectual Ligada al Cromosoma X/fisiopatología , Inactivación del Cromosoma X , Proteína Nuclear Ligada al Cromosoma X , Talasemia alfa/metabolismo , Talasemia alfa/fisiopatología
13.
J Cell Biol ; 173(4): 497-507, 2006 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-16717126

RESUMEN

During meiosis, the arrangement of homologous chromosomes is tightly regulated by the synaptonemal complex (SC). Each SC consists of two axial/lateral elements (AEs/LEs), and numerous transverse filaments. SC protein 2 (SYCP2) and SYCP3 are integral components of AEs/LEs in mammals. We find that SYCP2 forms heterodimers with SYCP3 both in vitro and in vivo. An evolutionarily conserved coiled coil domain in SYCP2 is required for binding to SYCP3. We generated a mutant Sycp2 allele in mice that lacks the coiled coil domain. The fertility of homozygous Sycp2 mutant mice is sexually dimorphic; males are sterile because of a block in meiosis, whereas females are subfertile with sharply reduced litter size. Sycp2 mutant spermatocytes exhibit failure in the formation of AEs and chromosomal synapsis. Strikingly, the mutant SYCP2 protein localizes to axial chromosomal cores in both spermatocytes and fetal oocytes, but SYCP3 does not, demonstrating that SYCP2 is a primary determinant of AEs/LEs and, thus, is required for the incorporation of SYCP3 into SCs.


Asunto(s)
Proteínas Cromosómicas no Histona/genética , Emparejamiento Cromosómico/genética , Cromosomas/genética , Meiosis/genética , Espermatocitos/metabolismo , Complejo Sinaptonémico/metabolismo , Animales , Proteínas de Ciclo Celular , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/metabolismo , Secuencia Conservada/genética , Proteínas de Unión al ADN , Femenino , Infertilidad Femenina/genética , Infertilidad Masculina/genética , Masculino , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Mutación/genética , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oocitos/metabolismo , Oocitos/ultraestructura , Estructura Terciaria de Proteína/genética , Caracteres Sexuales , Diferenciación Sexual/genética , Espermatocitos/ultraestructura , Espermatogénesis/genética , Complejo Sinaptonémico/ultraestructura
14.
Epigenetics Chromatin ; 14(1): 58, 2021 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-34955095

RESUMEN

Directed differentiation of mouse embryonic stem cells (mESCs) or induced pluripotent stem cells (iPSCs) provides powerful models to dissect the molecular mechanisms leading to the formation of specific cell lineages. Treatment with histone deacetylase inhibitors can significantly enhance the efficiency of directed differentiation. However, the mechanisms are not well understood. Here, we use CUT&RUN in combination with ATAC-seq to determine changes in both histone modifications and genome-wide chromatin accessibility following valproic acid (VPA) exposure. VPA induced a significant increase in global histone H3 acetylation (H3K56ac), a core histone modification affecting nucleosome stability, as well as enrichment at loci associated with cytoskeletal organization and cellular morphogenesis. In addition, VPA altered the levels of linker histone H1 subtypes and the total histone H1/nucleosome ratio indicative of initial differentiation events. Notably, ATAC-seq analysis revealed changes in chromatin accessibility of genes involved in regulation of CDK serine/threonine kinase activity and DNA duplex unwinding. Importantly, changes in chromatin accessibility were evident at several key genomic loci, such as the pluripotency factor Lefty, cardiac muscle troponin Tnnt2, and the homeodomain factor Hopx, which play critical roles in cardiomyocyte differentiation. Massive parallel transcription factor (TF) footprinting also indicates an increased occupancy of TFs involved in differentiation toward mesoderm and endoderm lineages and a loss of footprints of POU5F1/SOX2 pluripotency factors following VPA treatment. Our results provide the first genome-wide analysis of the chromatin landscape following VPA-induced differentiation in mESCs and provide new mechanistic insight into the intricate molecular processes that govern departure from pluripotency and early lineage commitment.


Asunto(s)
Cromatina , Histonas , Acetilación , Animales , Diferenciación Celular , Células Madre Embrionarias/metabolismo , Histonas/metabolismo , Ratones , Ácido Valproico/toxicidad
15.
Dev Biol ; 331(2): 326-38, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19463809

RESUMEN

In spite of the impact of aneuploidy on human health little is known concerning the molecular mechanisms involved in the formation of structural or numerical chromosome abnormalities during meiosis. Here, we provide novel evidence indicating that lack of PARP-1 function during oogenesis predisposes the female gamete to genome instability. During prophase I of meiosis, a high proportion of Parp-1((-/-)) mouse oocytes exhibit a spectrum of meiotic defects including incomplete homologous chromosome synapsis or persistent histone H2AX phosphorylation in fully synapsed chromosomes at the late pachytene stage. Moreover, the X chromosome bivalent is also prone to exhibit persistent double strand DNA breaks (DSBs). In striking contrast, such defects were not detected in mutant pachytene spermatocytes. In fully-grown wild type oocytes at the germinal vesicle stage, PARP-1 protein associates with nuclear speckles and upon meiotic resumption, undergoes a striking re-localization towards spindle poles as well as pericentric heterochromatin domains at the metaphase II stage. Notably, a high proportion of in vivo matured Parp-1((-/-)) oocytes show lack of recruitment of the kinetochore-associated protein BUB3 to centromeric domains and fail to maintain metaphase II arrest. Defects in chromatin modifications in the form of persistent histone H2AX phosphorylation during prophase I of meiosis and deficient sister chromatid cohesion during metaphase II predispose mutant oocytes to premature anaphase II onset upon removal from the oviductal environment. Our results indicate that PARP-1 plays a critical role in the maintenance of chromosome stability at key stages of meiosis in the female germ line. Moreover, in the metaphase II stage oocyte PARP-1 is required for the regulation of centromere structure and function through a mechanism that involves the recruitment of BUB3 protein to centromeric domains.


Asunto(s)
Centrómero/fisiología , Emparejamiento Cromosómico , Histonas/metabolismo , Oocitos/fisiología , Poli(ADP-Ribosa) Polimerasas/fisiología , Animales , Cromatina/metabolismo , Aberraciones Cromosómicas , Epigénesis Genética , Femenino , Meiosis , Ratones , Ratones Noqueados , Fosforilación , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/genética
16.
Chromosoma ; 118(2): 209-22, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19005673

RESUMEN

Mammalian X chromosome inactivation (XCI) is an essential mechanism to compensate for dosage imbalances between male and female embryos. Although the molecular pathways are not fully understood, heterochromatinization of the Xi requires the coordinate recruitment of multiple epigenetic marks. Using fluorescence in situ hybridization analysis combined with immunocytochemistry, we demonstrate that the chromatin remodeling protein ATRX decorates the chromatids of a single, late replicating X chromosome in female somatic cells and co-localizes with the bona fide marker of the Xi, macroH2A1.2. Chromatin immunoprecipitation using somatic, embryonic stem (ES) cells and trophoblast stem (TS) cells as model for random and imprinted XCI, respectively, revealed that, in somatic and TS cells, ATRX exhibits a specific association with sequences located within the previously described H3K9me2-hotspot, a region 5' to the X inactive-specific transcript (Xist) locus. While no ATRX-Xi interaction was detectable in undifferentiated ES cells, an enrichment of ATRX was observed after 8 days of differentiation, indicating that ATRX associates with the Xi following the onset of random XCI, consistent with a potential role in maintenance of XCI. These results have important implications regarding a previously described escape from imprinted XCI in ATRX-deficient mice as well as cases of skewed XCI in patients with ATRX syndrome.


Asunto(s)
ADN Helicasas/genética , ADN Helicasas/metabolismo , Células Madre Embrionarias/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Trofoblastos/citología , Trofoblastos/metabolismo , Inactivación del Cromosoma X , Cromosoma X/genética , Cromosoma X/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Inmunoprecipitación de Cromatina , Cromosomas Humanos X/genética , Cromosomas Humanos X/metabolismo , ADN Helicasas/deficiencia , Células Madre Embrionarias/citología , Femenino , Impresión Genómica , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Masculino , Ratones , Ratones Noqueados , Modelos Genéticos , Proteínas Nucleares/deficiencia , Embarazo , ARN Largo no Codificante , ARN no Traducido/genética , ARN no Traducido/metabolismo , Síndrome , Proteína Nuclear Ligada al Cromosoma X
17.
J Cell Biol ; 219(11)2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-32870972

RESUMEN

The polycomb group protein CBX2 is an important epigenetic reader involved in cell proliferation and differentiation. While CBX2 overexpression occurs in a wide range of human tumors, targeted deletion results in homeotic transformation, proliferative defects, and premature senescence. However, its cellular function(s) and whether it plays a role in maintenance of genome stability remain to be determined. Here, we demonstrate that loss of CBX2 in mouse fibroblasts induces abnormal large-scale chromatin structure and chromosome instability. Integrative transcriptome analysis and ATAC-seq revealed a significant dysregulation of transcripts involved in DNA repair, chromocenter formation, and tumorigenesis in addition to changes in chromatin accessibility of genes involved in lateral sclerosis, basal transcription factors, and folate metabolism. Notably, Cbx2-/- cells exhibit prominent decondensation of satellite DNA sequences at metaphase and increased sister chromatid recombination events leading to rampant chromosome instability. The presence of extensive centromere and telomere defects suggests a prominent role for CBX2 in heterochromatin homeostasis and the regulation of nuclear architecture.


Asunto(s)
Transformación Celular Neoplásica/patología , Senescencia Celular , Aberraciones Cromosómicas , Fibroblastos/patología , Inestabilidad Genómica , Complejo Represivo Polycomb 1/fisiología , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Centrómero , Cromatina , Femenino , Fibroblastos/metabolismo , Masculino , Ratones , Ratones Noqueados , Transcriptoma
18.
Nat Commun ; 11(1): 4486, 2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32900989

RESUMEN

Centromeres are epigenetically determined nuclear domains strictly required for chromosome segregation and genome stability. However, the mechanisms regulating centromere and kinetochore chromatin modifications are not known. Here, we demonstrate that LSH is enriched at meiotic kinetochores and its targeted deletion induces centromere instability and abnormal chromosome segregation. Superresolution chromatin analysis resolves LSH at the inner centromere and kinetochores during oocyte meiosis. LSH knockout pachytene oocytes exhibit reduced HDAC2 and DNMT-1. Notably, mutant oocytes show a striking increase in histone H3 phosphorylation at threonine 3 (H3T3ph) and accumulation of major satellite transcripts in both prophase-I and metaphase-I chromosomes. Moreover, knockout oocytes exhibit centromere fusions, ectopic kinetochore formation and abnormal exchange of chromatin fibers between paired bivalents and asynapsed chromosomes. Our results indicate that loss of LSH affects the levels and chromosomal localization of H3T3ph and provide evidence that, by maintaining transcriptionally repressive heterochromatin, LSH may be essential to prevent deleterious meiotic recombination events at repetitive centromeric sequences.


Asunto(s)
ADN Helicasas/metabolismo , Meiosis/fisiología , Oocitos/citología , Oocitos/metabolismo , Animales , Centrómero/genética , Centrómero/metabolismo , ADN Helicasas/deficiencia , ADN Helicasas/genética , Femenino , Histonas/metabolismo , Cinetocoros/metabolismo , Masculino , Meiosis/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Transcripción Genética
19.
BMC Mol Biol ; 9: 29, 2008 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-18366812

RESUMEN

BACKGROUND: Establishment of chromosomal cytosine methylation and histone methylation patterns are critical epigenetic modifications required for heterochromatin formation in the mammalian genome. However, the nature of the primary signal(s) targeting DNA methylation at specific genomic regions is not clear. Notably, whether histone methylation and/or chromatin remodeling proteins play a role in the establishment of DNA methylation during gametogenesis is not known. The chromosomes of mouse neonatal spermatogonia display a unique pattern of 5-methyl cytosine staining whereby centromeric heterochromatin is hypo-methylated whereas chromatids are strongly methylated. Thus, in order to gain some insight into the relationship between global DNA and histone methylation in the germ line we have used neonatal spermatogonia as a model to determine whether these unique chromosomal DNA methylation patterns are also reflected by concomitant changes in histone methylation. RESULTS: Our results demonstrate that histone H3 tri-methylated at lysine 9 (H3K9me3), a hallmark of constitutive heterochromatin, as well as the chromatin remodeling protein ATRX remained associated with pericentric heterochromatin regions in spite of their extensive hypo-methylation. This suggests that in neonatal spermatogonia, chromosomal 5-methyl cytosine patterns are regulated independently of changes in histone methylation, potentially reflecting a crucial mechanism to maintain pericentric heterochromatin silencing. Furthermore, chromatin immunoprecipitation and fluorescence in situ hybridization, revealed that ATRX as well as H3K9me3 associate with Y chromosome-specific DNA sequences and decorate both arms of the Y chromosome, suggesting a possible role in heterochromatinization and the predominant transcriptional quiescence of this chromosome during spermatogenesis. CONCLUSION: These results are consistent with a role for histone modifications and chromatin remodeling proteins such as ATRX in maintaining transcriptional repression at constitutive heterochromatin domains in the absence of 5-methyl cytosine and provide evidence suggesting that the establishment and/or maintenance of repressive histone and chromatin modifications at pericentric heterochromatin following genome-wide epigenetic reprogramming in the germ line may precede the establishment of chromosomal 5-methyl cytosine patterns as a genomic silencing strategy in neonatal spermatogonia.


Asunto(s)
ADN Helicasas/metabolismo , Heterocromatina/metabolismo , Proteínas Nucleares/metabolismo , Espermatogonias/metabolismo , Cromosoma Y/metabolismo , Animales , Animales Recién Nacidos , Ensamble y Desensamble de Cromatina , Citosina/metabolismo , Metilación de ADN , Histonas/metabolismo , Masculino , Ratones , Espermatogonias/ultraestructura , Proteína Nuclear Ligada al Cromosoma X
20.
J Cell Biol ; 208(1): 53-69, 2015 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-25547156

RESUMEN

Postmeiotic gene expression is essential for development and maturation of sperm and eggs. We report that the dual bromodomain-containing protein BRWD1, which is essential for both male and female fertility, promotes haploid spermatid-specific transcription but has distinct roles in oocyte meiotic progression. Brwd1 deficiency caused down-regulation of ∼300 mostly spermatid-specific transcripts in testis, including nearly complete elimination of those encoding the protamines and transition proteins, but was not associated with global epigenetic changes in chromatin, which suggests that BRWD1 acts selectively. In females, Brwd1 ablation caused severe chromosome condensation and structural defects associated with abnormal telomere structure but only minor changes in gene expression at the germinal vesicle stage, including more than twofold overexpression of the histone methyltransferase MLL5 and LINE-1 elements transposons. Thus, loss of BRWD1 function interferes with the completion of oogenesis and spermatogenesis through sexually dimorphic mechanisms: it is essential in females for epigenetic control of meiotic chromosome stability and in males for haploid gene transcription during postmeiotic sperm differentiation.


Asunto(s)
Inestabilidad Cromosómica , Fertilidad , Histona Acetiltransferasas/metabolismo , Meiosis , Oogénesis , Espermátides/metabolismo , Espermatogénesis , Transcripción Genética , Animales , Células Cultivadas , Análisis por Conglomerados , Epigénesis Genética , Femenino , Perfilación de la Expresión Génica/métodos , Regulación del Desarrollo de la Expresión Génica , Genotipo , Haploidia , Histona Acetiltransferasas/genética , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Elementos de Nucleótido Esparcido Largo , Masculino , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Caracteres Sexuales , Factores Sexuales , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA