Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Semin Cell Dev Biol ; 135: 93-101, 2023 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35249811

RESUMEN

Variants of the histone H2A occupy distinct locations in the genome. There is relatively little known about the mechanisms responsible for deposition of specific H2A variants. Notable exceptions are chromatin remodelers that control the dynamics of H2A.Z at promoters. Here we review the steps that identified the role of a specific class of chromatin remodelers, including LSH and DDM1 that deposit the variants macroH2A in mammals and H2A.W in plants, respectively. The function of these remodelers in heterochromatin is discussed together with their multiple roles in genome stability.


Asunto(s)
Heterocromatina , Histonas , Animales , Histonas/genética , Histonas/metabolismo , Heterocromatina/genética , Cromatina , Regiones Promotoras Genéticas , Mamíferos/genética , Biología , Nucleosomas
2.
Nucleic Acids Res ; 49(14): 8024-8036, 2021 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-34223906

RESUMEN

LSH, a homologue of the ISWI/SNF2 family of chromatin remodelers, is required in vivo for deposition of the histone variants macroH2A1 and macroH2A2 at specific genomic locations. However, it remains unknown whether LSH is directly involved in this process or promotes other factors. Here we show that recombinant LSH interacts in vitro with macroH2A1-H2B and macroH2A2-H2B dimers, but not with H2A.Z-H2B dimers. Moreover, LSH catalyzes the transfer of macroH2A into mono-nucleosomes reconstituted with canonical core histones in an ATP dependent manner. LSH requires the ATP binding site and the replacement process is unidirectional leading to heterotypic and homotypic nucleosomes. Both variants macroH2A1 and macroH2A2 are equally well incorporated into the nucleosome. The histone exchange reaction is specific for histone variant macroH2A, since LSH is not capable to incorporate H2A.Z. These findings define a previously unknown role for LSH in chromatin remodeling and identify a novel molecular mechanism for deposition of the histone variant macroH2A.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , Cromatina/genética , ADN Helicasas/genética , Histonas/genética , Adenosina Trifosfato/genética , Sitios de Unión/genética , Humanos , Nucleosomas/genética
3.
Proc Natl Acad Sci U S A ; 117(33): 20100-20108, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32727902

RESUMEN

Mutation of HELLS (Helicase, Lymphoid-Specific)/Lsh in human DNA causes a severe immunodeficiency syndrome, but the nature of the defect remains unknown. We assessed here the role of Lsh in hematopoiesis using conditional Lsh knockout mice with expression of Mx1 or Vav Cre-recombinase. Bone marrow transplantation studies revealed that Lsh depletion in hematopoietic stem cells severely reduced B cell numbers and impaired B cell development in a hematopoietic cell-autonomous manner. Lsh-deficient mice without bone marrow transplantation exhibited lower Ig levels in vivo compared to controls despite normal peripheral B cell numbers. Purified B lymphocytes proliferated normally but produced less immunoglobulins in response to in vitro stimulation, indicating a reduced capacity to undergo class switch recombination (CSR). Analysis of germline transcripts, examination of double-stranded breaks using biotin-labeling DNA break assay, and End-seq analysis indicated that the initiation of the recombination process was unscathed. In contrast, digestion-circularization PCR analysis and high-throughput sequencing analyses of CSR junctions and a chromosomal break repair assay indicated an impaired ability of the canonical end-joining pathway in Lsh-deficient B cells. Our data suggest a hematopoietic cell-intrinsic role of Lsh in B cell development and in CSR providing a potential target for immunodeficiency therapy.


Asunto(s)
Linfocitos B/fisiología , ADN Helicasas/metabolismo , Inmunoglobulinas/metabolismo , Animales , Línea Celular , ADN Helicasas/genética , Silenciador del Gen , Humanos , Inmunoglobulinas/genética , Ratones , Ratones Noqueados , Mutación
4.
Genome Res ; 24(10): 1613-23, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25170028

RESUMEN

Cytosine methylation is critical in mammalian development and plays a role in diverse biologic processes such as genomic imprinting, X chromosome inactivation, and silencing of repeat elements. Several factors regulate DNA methylation in early embryogenesis, but their precise role in the establishment of DNA methylation at a given site remains unclear. We have generated a comprehensive methylation map in fibroblasts derived from the murine DNA methylation mutant Hells(-/-) (helicase, lymphoid specific, also known as LSH). It has been previously shown that HELLS can influence de novo methylation of retroviral sequences and endogenous genes. Here, we describe that HELLS controls cytosine methylation in a nuclear compartment that is in part defined by lamin B1 attachment regions. Despite widespread loss of cytosine methylation at regulatory sequences, including promoter regions of protein-coding genes and noncoding RNA genes, overall relative transcript abundance levels in the absence of HELLS are similar to those in wild-type cells. A subset of promoter regions shows increases of the histone modification H3K27me3, suggesting redundancy of epigenetic silencing mechanisms. Furthermore, HELLS modulates CG methylation at all classes of repeat elements and is critical for repression of a subset of repeat elements. Overall, we provide a detailed analysis of gene expression changes in relation to DNA methylation alterations, which contributes to our understanding of the biological role of cytosine methylation.


Asunto(s)
Citosina/metabolismo , ADN Helicasas/genética , Metilación de ADN , ADN/metabolismo , Regulación del Desarrollo de la Expresión Génica , Animales , Núcleo Celular/genética , Células Madre Embrionarias , Epigénesis Genética , Fibroblastos/citología , Fibroblastos/metabolismo , Técnicas de Inactivación de Genes , Histonas/metabolismo , Lamina Tipo B/metabolismo , Ratones , Datos de Secuencia Molecular , Mutación , Secuencias Reguladoras de Ácidos Nucleicos , Secuencias Repetitivas de Ácidos Nucleicos
5.
Nucleic Acids Res ; 43(3): 1444-55, 2015 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-25578963

RESUMEN

Lsh, a chromatin remodeling protein of the SNF2 family, is critical for normal heterochromatin structure. In particular, DNA methylation at repeat elements, a hallmark of heterochromatin, is greatly reduced in Lsh(-/-) (KO) cells. Here, we examined the presumed nucleosome remodeling activity of Lsh on chromatin in the context of DNA methylation. We found that dynamic CG methylation was dependent on Lsh in embryonic stem cells. Moreover, we demonstrate that ATP function is critical for de novo methylation at repeat sequences. The ATP binding site of Lsh is in part required to promote stable association of the DNA methyltransferase 3b with the repeat locus. By performing nucleosome occupancy assays, we found distinct nucleosome occupancy in KO ES cells compared to WT ES cells after differentiation. Nucleosome density was restored to wild-type level by re-expressing wild-type Lsh but not the ATP mutant in KO ES cells. Our results suggest that ATP-dependent nucleosome remodeling is the primary molecular function of Lsh, which may promote de novo methylation in differentiating ES cells.


Asunto(s)
Adenosina Trifosfato/metabolismo , Ensamble y Desensamble de Cromatina , ADN Helicasas/metabolismo , Metilación de ADN , Nucleosomas/metabolismo , Secuencias Repetitivas de Ácidos Nucleicos , Sitios de Unión , Células Cultivadas , Humanos
6.
Proc Natl Acad Sci U S A ; 111(16): 5890-5, 2014 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-24711395

RESUMEN

DNA methylation patterns are established in early embryogenesis and are critical for cellular differentiation. To investigate the role of CG methylation in potential enhancer formation, we assessed H3K4me1 modification in murine embryonic fibroblasts (MEFs) derived from the DNA methylation mutant Lsh(-/-) mice. We report here de novo formation of putative enhancer elements at CG hypomethylated sites that can be dynamically altered. We found a subset of differentially enriched H3K4me1 regions clustered at neuronal lineage genes and overlapping with known cis-regulatory elements present in brain tissue. Reprogramming of Lsh(-/-) MEFs into induced pluripotent stem (iPS) cells leads to increased neuronal lineage gene expression of premarked genes and enhanced differentiation potential of Lsh(-/-) iPS cells toward the neuronal lineage pathway compared with WT iPS cells in vitro and in vivo. The state of CG hypomethylation and H3K4me1 enrichment is partially maintained in Lsh(-/-) iPS cells. The acquisition of H3K27ac and activity of subcloned fragments in an enhancer reporter assay indicate functional activity of several of de novo H3K4me1-marked sequences. Our results suggest a functional link of H3K4me1 enrichment at CG hypomethylated sites, enhancer formation, and cellular plasticity.


Asunto(s)
Islas de CpG/genética , ADN Helicasas/deficiencia , Metilación de ADN/genética , Embrión de Mamíferos/citología , Fibroblastos/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Animales , Biomarcadores/metabolismo , Linaje de la Célula , ADN Helicasas/metabolismo , Elementos de Facilitación Genéticos/genética , Epigénesis Genética , Fibroblastos/citología , Células HeLa , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Ratones Noqueados , Neuronas/citología , Unión Proteica , Transducción de Señal , Factores de Transcripción/metabolismo
7.
EMBO J ; 31(4): 972-85, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22157815

RESUMEN

The activating E2F-transcription factors are best known for their dependence on the Retinoblastoma protein and their role in cellular proliferation. E2F3 is uniquely amplified in specific human tumours where its expression is inversely correlated with the survival of patients. Here, E2F3B interaction partners were identified by mass spectrometric analysis. We show that the SNF2-like helicase HELLS interacts with E2F3A in vivo and cooperates with its oncogenic functions. Depletion of HELLS severely perturbs the induction of E2F-target genes, hinders cell-cycle re-entry and growth. Using chromatin immmunoprecipitation coupled to sequencing, we identified genome-wide targets of HELLS and E2F3A/B. HELLS binds promoters of active genes, including the trithorax-related MLL1, and co-regulates E2F3-dependent genes. Strikingly, just as E2F3, HELLS is overexpressed in human tumours including prostate cancer, indicating that either factor may contribute to the malignant progression of tumours. Our work reveals that HELLS is important for E2F3 in tumour cell proliferation.


Asunto(s)
Transformación Celular Neoplásica , ADN Helicasas/fisiología , Factor de Transcripción E2F3/fisiología , Transcripción Genética/fisiología , Ciclo Celular , Inmunoprecipitación de Cromatina , ADN Helicasas/metabolismo , Humanos , Masculino , Neoplasias de la Próstata/patología , Unión Proteica
8.
Proc Natl Acad Sci U S A ; 108(14): 5626-31, 2011 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-21427231

RESUMEN

DNA methylation is critical for normal development and plays important roles in genome organization and transcriptional regulation. Although DNA methyltransferases have been identified, the factors that establish and contribute to genome-wide methylation patterns remain elusive. Here, we report a high-resolution cytosine methylation map of the murine genome modulated by Lsh, a chromatin remodeling family member that has previously been shown to regulate CpG methylation at repetitive sequences. We provide evidence that Lsh also controls genome-wide cytosine methylation at nonrepeat sequences and relate those changes to alterations in H4K4me3 modification and gene expression. Deletion of Lsh alters the allocation of cytosine methylation in chromosomal regions of 50 kb to 2 Mb and, in addition, leads to changes in the methylation profile at the 5' end of genes. Furthermore, we demonstrate that loss of Lsh promotes--as well as prevents--cytosine methylation. Our data indicate that Lsh is an epigenetic modulator that is critical for normal distribution of cytosine methylation throughout the murine genome.


Asunto(s)
Citosina/metabolismo , ADN Helicasas/metabolismo , Metilación de ADN , Epigenómica , Animales , Southern Blotting , Línea Celular , Inmunoprecipitación de Cromatina , Cromatografía Líquida de Alta Presión , Perfilación de la Expresión Génica , Genómica , Ratones , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Estadísticas no Paramétricas
9.
Biochim Biophys Acta ; 1819(7): 757-62, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22381140

RESUMEN

Methylation regulates DNA by altering chromatin and limiting accessibility of transcription factors and RNA polymerase. In this way, DNA methylation controls gene expression and plays a role in ES cell regulation, tissue differentiation and the development of the organism. In abnormal circumstances methylation can also induce diseases and promote cancer progression. Chromatin remodeling proteins such as the SNF2 family member Lsh regulates genome-wide cytosine methylation patterns during mammalian development. Lsh promotes methylation by targeting and repressing repeat sequences that are imbedded in heterochromatin. Lsh also regulates cytosine methylation at unique loci. Alterations in histone modifications (such as H3K4me3, histone acetylation, H3K27me3 and H2Aub) can be associated with DNA methylation changes making Lsh-mediated cytosine methylation part of a larger epigenetic network defining gene expression and cellular differentiation during development. This article is part of a Special Issue entitled: Chromatin in time and space.


Asunto(s)
Diferenciación Celular/genética , Metilación de ADN , Animales , Citosina/metabolismo , ADN Helicasas/fisiología , Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Genoma Humano , Humanos , Secuencias Repetitivas de Ácidos Nucleicos , Transcripción Genética
10.
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
11.
Nucleic Acids Res ; 39(22): 9508-20, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21880597

RESUMEN

Inactivation of tumor suppressor genes plays an important role in tumorigenesis, and epigenetic modifications such as DNA methylation are frequently associated with transcriptional repression. Here, we show that gene silencing at selected genes with signs of DNA hypermethylation in breast cancer cells involves Pol II stalling. We studied several repressed genes with DNA hypermethylation within a region 1-kb upstream of the transcriptional start site that were upregulated after treatment with DNA demethylating agents, such as Azacytidine and several natural products. All those selected genes had stalled Pol II at their transcriptional start site and showed enhanced ser2 phosphorylated Pol II and elevated transcripts after drug treatment indicating successful elongation. In addition, a decrease of the epigenetic regulator LSH in a breast cancer cell line by siRNA treatment reduced DNA methylation and overcame Pol II stalling, whereas overexpression of LSH in a normal breast epithelial cell line increased DNA methylation and resulted in repression. Decrease of LSH was associated with reduced DNMT3b binding to promoter sequences, and depletion of DNMT3b by siRNA could release Pol II suggesting that DNMT3b is functionally involved. The release of paused Pol II was accompanied by a dynamic switch from repressive to active chromatin marks. Thus release of Pol II stalling can act as a mechanism for gene reactivation at specific target genes after DNA demethylating treatment in cancer cells.


Asunto(s)
Azacitidina/farmacología , Neoplasias de la Mama/genética , Metilación de ADN/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , ARN Polimerasa II/metabolismo , Neoplasias de la Mama/enzimología , Línea Celular Tumoral , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Femenino , Histonas/metabolismo , Humanos , Proteínas del Grupo Polycomb , Proteínas Represoras/metabolismo , Sitio de Iniciación de la Transcripción , Transcripción Genética/efectos de los fármacos
12.
Nat Commun ; 14(1): 5695, 2023 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-37709749

RESUMEN

HELLS/LSH (Helicase, Lymphoid Specific) is a SNF2-like chromatin remodelling protein involved in DNA methylation. Its loss-of-function in humans causes humoral immunodeficiency, called ICF4 syndrome (Immunodeficiency, Centromeric Instability, Facial anomalies). Here we show by our newly generated B-cell-specific Hells conditional knockout mouse model that HELLS plays a pivotal role in T-dependent B-cell responses. HELLS deficiency induces accelerated decay of germinal center (GC) B cells and impairs the generation of high affinity memory B cells and circulating antibodies. Mutant GC B cells undergo dramatic DNA hypomethylation and massive de-repression of evolutionary recent retrotransposons, which surprisingly does not directly affect their survival. Instead, they prematurely upregulate either memory B cell markers or the transcription factor ATF4, which is driving an mTORC1-dependent metabolic program typical of plasma cells. Treatment of wild type mice with a DNMT1-specific inhibitor phenocopies the accelerated kinetics, thus pointing towards DNA-methylation maintenance by HELLS being a crucial mechanism to fine-tune the GC transcriptional program and enable long-lasting humoral immunity.


Asunto(s)
Linfocitos B , Metilación de ADN , Animales , Humanos , Ratones , ADN , ADN Helicasas , Centro Germinal , Células Plasmáticas
13.
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
14.
Nat Commun ; 12(1): 3520, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-34112784

RESUMEN

The Immunodeficiency Centromeric Instability Facial Anomalies (ICF) 4 syndrome is caused by mutations in LSH/HELLS, a chromatin remodeler promoting incorporation of histone variant macroH2A. Here, we demonstrate that LSH depletion results in degradation of nascent DNA at stalled replication forks and the generation of genomic instability. The protection of stalled forks is mediated by macroH2A, whose knockdown mimics LSH depletion and whose overexpression rescues nascent DNA degradation. LSH or macroH2A deficiency leads to an impairment of RAD51 loading, a factor that prevents MRE11 and EXO1 mediated nascent DNA degradation. The defect in RAD51 loading is linked to a disbalance of BRCA1 and 53BP1 accumulation at stalled forks. This is associated with perturbed histone modifications, including abnormal H4K20 methylation that is critical for BRCA1 enrichment and 53BP1 exclusion. Altogether, our results illuminate the mechanism underlying a human syndrome and reveal a critical role of LSH mediated chromatin remodeling in genomic stability.


Asunto(s)
ADN Helicasas/metabolismo , Replicación del ADN , Inestabilidad Genómica , Histonas/metabolismo , Recombinasa Rad51/metabolismo , Animales , Proteína BRCA1/metabolismo , Línea Celular Tumoral , Supervivencia Celular/genética , Ensamble y Desensamble de Cromatina/genética , Secuenciación de Inmunoprecipitación de Cromatina , ADN Helicasas/deficiencia , ADN Helicasas/genética , Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/metabolismo , Replicación del ADN/genética , Epigénesis Genética , Exodesoxirribonucleasas/genética , Exodesoxirribonucleasas/metabolismo , Inestabilidad Genómica/genética , Histonas/deficiencia , Histonas/genética , Humanos , Proteína Homóloga de MRE11/genética , Proteína Homóloga de MRE11/metabolismo , Metilación , Ratones , ARN Interferente Pequeño , Recombinasa Rad51/genética , Regulación hacia Arriba
15.
Stem Cells ; 27(11): 2691-702, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19650037

RESUMEN

Transcriptional control of stem cell genes is a critical step in differentiation of embryonic stem cells and in reprogramming of somatic cells into stem cells. Here we report that Lsh, a regulator of repressive chromatin at retrotransposons, also plays an important role in silencing of stem cell-specific genes such as Oct4. We found that CpG methylation is gained during in vitro differentiation of several stem cell-specific genes (in 11 of 12 promoter regions) and thus appears to be a common epigenetic mark. Lsh depletion prevents complete silencing of stem cell gene expression and moreover promotes the maintenance of stem cell characteristics in culture. Lsh is required for establishment of DNA methylation patterns at stem cell genes during differentiation, in part by regulating access of Dnmt3b to its genomic targets. Our results indicate that Lsh is involved in the control of stem cell genes and suggest that Lsh is an important epigenetic modulator during early stem cell differentiation.


Asunto(s)
ADN Helicasas/metabolismo , Metilación de ADN , Silenciador del Gen , Animales , Southern Blotting , Western Blotting , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Línea Celular , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Proteínas Cromosómicas no Histona , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Helicasas/genética , Factor de Crecimiento Epidérmico/genética , Factor 3 de Diferenciación de Crecimiento/genética , Proteínas de Homeodominio/genética , Glicoproteínas de Membrana/genética , Ratones , Proteína Homeótica Nanog , Proteínas de Neoplasias/genética , Factor 3 de Transcripción de Unión a Octámeros/genética , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas/genética , Unión Proteica , Proteínas Represoras/genética , ADN Metiltransferasa 3B
16.
Mol Reprod Dev ; 77(2): 105-13, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19921744

RESUMEN

Development from separate parental germ cells through fertilization and proceeding to a fully functioning adult animal occurs through an intricate program of transcriptional and chromatin changes. Epigenetic alterations such as DNA methylation are an important part of this process. This review looks at the role of DNA methylation in early embryonic development, as well as how this epigenetic mark affects stem cell differentiation and tissue-specific gene expression in somatic cells.


Asunto(s)
Diferenciación Celular/fisiología , Metilación de ADN/fisiología , Desarrollo Embrionario/fisiología , Células Madre Embrionarias/fisiología , Animales , Ensamble y Desensamble de Cromatina/fisiología , Células Madre Embrionarias/citología , Humanos , Especificidad de Órganos/fisiología , Transcripción Genética/fisiología
17.
Nat Commun ; 11(1): 5647, 2020 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-33159050

RESUMEN

The human Immunodeficiency Centromeric Instability Facial Anomalies (ICF) 4 syndrome is a severe disease with increased mortality caused by mutation in the LSH gene. Although LSH belongs to a family of chromatin remodeling proteins, it remains unknown how LSH mediates its function on chromatin in vivo. Here, we use chemical-induced proximity to rapidly recruit LSH to an engineered locus and find that LSH specifically induces macroH2A1.2 and macroH2A2 deposition in an ATP-dependent manner. Tethering of LSH induces transcriptional repression and silencing is dependent on macroH2A deposition. Loss of LSH decreases macroH2A enrichment at repeat sequences and results in transcriptional reactivation. Likewise, reduction of macroH2A by siRNA interference mimicks transcriptional reactivation. ChIP-seq analysis confirmed that LSH is a major regulator of genome-wide macroH2A distribution. Tethering of ICF4 mutations fails to induce macroH2A deposition and ICF4 patient cells display reduced macroH2A deposition and transcriptional reactivation supporting a pathogenic role for altered marcoH2A deposition. We propose that LSH is a major chromatin modulator of the histone variant macroH2A and that its ability to insert marcoH2A into chromatin and transcriptionally silence is disturbed in the ICF4 syndrome.


Asunto(s)
ADN Helicasas/metabolismo , Histonas/metabolismo , Enfermedades de Inmunodeficiencia Primaria/metabolismo , Animales , Cromatina/genética , Cromatina/metabolismo , ADN Helicasas/genética , Regulación hacia Abajo , Femenino , Histonas/genética , Humanos , Masculino , Ratones , Enfermedades de Inmunodeficiencia Primaria/enzimología , Enfermedades de Inmunodeficiencia Primaria/genética , Transcripción Genética
18.
Sci Signal ; 13(645)2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32817374

RESUMEN

Synthetic lethality between poly(ADP-ribose) polymerase (PARP) inhibition and BRCA deficiency is exploited to treat breast and ovarian tumors. However, resistance to PARP inhibitors (PARPis) is common. To identify potential resistance mechanisms, we performed a genome-wide RNAi screen in BRCA2-deficient mouse embryonic stem cells and validation in KB2P1.21 mouse mammary tumor cells. We found that resistance to multiple PARPi emerged with reduced expression of TET2 (ten-eleven translocation), which promotes DNA demethylation by oxidizing 5-methylcytosine (5mC) to 5-hydroxymethycytosine (5hmC) and other products. TET2 knockdown in BRCA2-deficient cells protected stalled replication forks (RFs). Increasing 5hmC abundance induced the degradation of stalled RFs in KB2P1.21 and human cancer cells by recruiting the base excision repair-associated apurinic/apyrimidinic endonuclease APE1, independent of the BRCA2 status. TET2 loss did not affect the recruitment of the repair protein RAD51 to sites of double-strand breaks (DSBs) or the abundance of proteins associated with RF integrity. The loss of TET2, of its product 5hmC, and of APE1 recruitment to stalled RFs promoted resistance to the chemotherapeutic cisplatin. Our findings reveal a previously unknown role for the epigenetic mark 5hmC in maintaining the integrity of stalled RFs and a potential resistance mechanism to PARPi and cisplatin.


Asunto(s)
Neoplasias de la Mama/genética , Replicación del ADN/genética , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Desoxicitidina/análogos & derivados , Inestabilidad Genómica/genética , Neoplasias Ováricas/genética , 5-Metilcitosina/metabolismo , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Línea Celular , Línea Celular Tumoral , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Desoxicitidina/metabolismo , Resistencia a Antineoplásicos/genética , Femenino , Humanos , Ratones , Ratones Desnudos , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Ftalazinas/farmacología , Piperazinas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología
19.
Epigenetics ; 14(3): 277-293, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30861354

RESUMEN

Dynamic regulation of chromatin accessibility is a key feature of cellular differentiation during embryogenesis, but the precise factors that control access to chromatin remain largely unknown. Lsh/HELLS is critical for normal development and mutations of Lsh in human cause the ICF (Immune deficiency, Centromeric instability, Facial anomalies) syndrome, a severe immune disorder with multiple organ deficiencies. We report here that Lsh, previously known to regulate DNA methylation level, has a genome wide chromatin remodeling function. Using micrococcal nuclease (MNase)-seq analysis, we demonstrate that Lsh protects MNase accessibility at transcriptional regulatory regions characterized by DNase I hypersensitivity and certain histone 3 (H3) tail modifications associated with enhancers. Using an auxin-inducible degron system, allowing proteolytical degradation of Lsh, we show that Lsh mediated changes in nucleosome occupancy are independent of DNA methylation level and are characterized by reduced H3 occupancy. While Lsh mediated nucleosome occupancy prevents binding sites for transcription factors in wild type cells, depletion of Lsh leads to an increase in binding of ectopically expressed tissue specific transcription factors to their respective binding sites. Our data suggests that Lsh mediated chromatin remodeling can modulate nucleosome positioning at a subset of putative enhancers contributing to the preservation of cellular identity through regulation of accessibility.


Asunto(s)
Ensamble y Desensamble de Cromatina/fisiología , ADN Helicasas/metabolismo , Elementos de Facilitación Genéticos , Factores de Transcripción/metabolismo , Animales , Sitios de Unión , ADN Helicasas/genética , Metilación de ADN , Código de Histonas , Ratones Noqueados , Nucleasa Microcócica/metabolismo , Nucleosomas/genética , Nucleosomas/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos , Factores de Transcripción/genética
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA