Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 153(5): 1134-48, 2013 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-23664764

RESUMEN

Epigenetic mechanisms have been proposed to play crucial roles in mammalian development, but their precise functions are only partially understood. To investigate epigenetic regulation of embryonic development, we differentiated human embryonic stem cells into mesendoderm, neural progenitor cells, trophoblast-like cells, and mesenchymal stem cells and systematically characterized DNA methylation, chromatin modifications, and the transcriptome in each lineage. We found that promoters that are active in early developmental stages tend to be CG rich and mainly engage H3K27me3 upon silencing in nonexpressing lineages. By contrast, promoters for genes expressed preferentially at later stages are often CG poor and primarily employ DNA methylation upon repression. Interestingly, the early developmental regulatory genes are often located in large genomic domains that are generally devoid of DNA methylation in most lineages, which we termed DNA methylation valleys (DMVs). Our results suggest that distinct epigenetic mechanisms regulate early and late stages of ES cell differentiation.


Asunto(s)
Metilación de ADN , Células Madre Embrionarias/metabolismo , Epigenómica , Regulación del Desarrollo de la Expresión Génica , Animales , Diferenciación Celular , Cromatina/metabolismo , Islas de CpG , Células Madre Embrionarias/citología , Histonas/metabolismo , Humanos , Metilación , Neoplasias/genética , Regiones Promotoras Genéticas , Pez Cebra/embriología
2.
Proc Natl Acad Sci U S A ; 120(36): e2304851120, 2023 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-37639608

RESUMEN

Memory formation and forgetting unnecessary memory must be balanced for adaptive animal behavior. While cyclic AMP (cAMP) signaling via dopamine neurons induces memory formation, here we report that cyclic guanine monophosphate (cGMP) signaling via dopamine neurons launches forgetting of unconsolidated memory in Drosophila. Genetic screening and proteomic analyses showed that neural activation induces the complex formation of a histone H3K9 demethylase, Kdm4B, and a GMP synthetase, Bur, which is necessary and sufficient for forgetting unconsolidated memory. Kdm4B/Bur is activated by phosphorylation through NO-dependent cGMP signaling via dopamine neurons, inducing gene expression, including kek2 encoding a presynaptic protein. Accordingly, Kdm4B/Bur activation induced presynaptic changes. Our data demonstrate a link between cGMP signaling and synapses via gene expression in forgetting, suggesting that the opposing functions of memory are orchestrated by distinct signaling via dopamine neurons, which affects synaptic integrity and thus balances animal behavior.


Asunto(s)
Neuronas Dopaminérgicas , Proteómica , Animales , Sistemas de Mensajero Secundario , Transducción de Señal , Memoria , Drosophila , Guanina , Histona Demetilasas
3.
Mol Cell ; 64(6): 1062-1073, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27916660

RESUMEN

The methylcytosine oxidase TET proteins play important roles in DNA demethylation and development. However, it remains elusive how exactly they target substrates and execute oxidation. Interestingly, we found that, in mice, the full-length TET1 isoform (TET1e) is restricted to early embryos, embryonic stem cells (ESCs), and primordial germ cells (PGCs). By contrast, a short isoform (TET1s) is preferentially expressed in somatic cells, which lacks the N terminus including the CXXC domain, a DNA-binding module that often recognizes CpG islands (CGIs) where TET1 predominantly occupies. Unexpectedly, TET1s can still bind CGIs despite the fact that its global chromatin binding is significantly reduced. Interestingly, global chromatin binding, but not targeted binding at CGIs, is correlated with TET1-mediated demethylation. Finally, mice with exclusive expression of Tet1s failed to erase imprints in PGCs and displayed developmental defects in progeny. These data show that isoform switch of TET1 regulates epigenetic memory erasure and mouse development.


Asunto(s)
Proteínas de Unión al ADN/genética , Impresión Genómica , Células Madre Embrionarias de Ratones/metabolismo , Óvulo/metabolismo , Proteínas Proto-Oncogénicas/genética , Espermatozoides/metabolismo , Animales , Sitios de Unión , Cromatina/química , Cromatina/metabolismo , Islas de CpG , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Células Madre Embrionarias de Ratones/citología , Óvulo/citología , Regiones Promotoras Genéticas , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/metabolismo , Espermatozoides/citología
4.
Int J Mol Sci ; 24(22)2023 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-38003607

RESUMEN

Retrotransposons are invasive genetic elements that constitute substantial portions of mammalian genomes. They have the potential to influence nearby gene expression through their cis-regulatory sequences, reverse transcription machinery, and the ability to mold higher-order chromatin structures. Due to their multifaceted functions, it is crucial for host fitness to maintain strict regulation of these parasitic sequences to ensure proper growth and development. This review explores how subsets of retrotransposons have undergone evolutionary exaptation to enhance the complexity of mammalian genomes. It also highlights the significance of regulating these elements, drawing on recent studies conducted in human and murine systems.


Asunto(s)
Genoma , Retroelementos , Animales , Ratones , Humanos , Retroelementos/genética , Evolución Molecular , Mamíferos/genética
5.
J Neurosci ; 41(45): 9286-9307, 2021 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-34593604

RESUMEN

Adult-onset neurodegenerative diseases are often accompanied by evidence of a chronic inflammation that includes activation of microglial cells and altered levels of brain cytokines. Aspects of this response are likely secondary reactions to neurodegeneration, but for many illnesses the inflammation may itself be an early and even causative disease event. In such cases, the inflammation is referred to as "sterile" as it occurs in the absence of an actual bacterial or viral pathogen. A potent trigger of sterile inflammation in CNS microglia has been shown to be the presence of DNA in the cytoplasm (cytoDNA) induced either by direct DNA damage or by inhibited DNA repair. We have shown that cytoDNA comes from the cell nucleus as a result of insufficient DNA damage repair. Using wild-type and Atm-/- mouse microglia, we extend these observations here by showing that its genomic origins are not random, but rather are heavily biased toward transcriptionally inactive, intergenic regions, in particular repetitive elements and AT-rich sequences. Once released from the genome, in both males and females, we show that cytoDNA is actively exported to the cytoplasm by a CRM1-dependent mechanism. In the cytoplasm, it is degraded either by a cytosolic exonuclease, Trex1, or an autophagy pathway that ends with degradation in the lysosome. Blocking the accumulation of cytoDNA prevents the emergence of the sterile inflammation reaction. These findings offer new insights into the emergence of sterile inflammation and offer novel approaches that may be of use in combatting a wide range of neurodegenerative conditions.SIGNIFICANCE STATEMENT Sterile inflammation describes a state where the defenses of the immune system are activated in the absence of a true pathogen. A potent trigger of this unorthodox response is the presence of DNA in the cytoplasm, which immune cells interpret as an invading virus or pathogen. We show that when DNA damage increases, fragments of the cell's own genome are actively exported to the cytoplasm where they are normally degraded. If this degradation is incomplete an immune reaction is triggered. Both age and stress increase DNA damage, and as age-related neurodegenerative diseases are frequently accompanied by a chronic low-level inflammation, strategies that reduce the induction of cytoplasmic DNA or speed its clearance become attractive therapeutic targets.


Asunto(s)
Citoplasma/inmunología , Daño del ADN/inmunología , ADN/inmunología , Inflamación/inmunología , Secuencias Repetitivas de Ácidos Nucleicos/inmunología , Animales , Citoplasma/metabolismo , ADN/metabolismo , Reparación del ADN , Femenino , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/inmunología , Microglía/metabolismo
6.
Geophys Res Lett ; 49(8): e2021GL097287, 2022 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-35866061

RESUMEN

Aircraft measurement campaigns have revealed that super coarse dust (diameter >10 µm) surprisingly accounts for approximately a quarter of aerosols by mass in the atmosphere. However, most global aerosol models either underestimate or do not include super coarse dust abundance. To address this problem, we use brittle fragmentation theory to develop a parameterization for the emitted dust size distribution that includes emission of super coarse dust. We implement this parameterization in the Community Earth System Model (CESM) and find that it brings the model in good agreement with aircraft measurements of super coarse dust close to dust source regions. However, the CESM still underestimates super coarse dust in dust outflow regions. Thus, we conclude that the model underestimation of super coarse atmospheric dust is in part due to the underestimation of super coarse dust emission and likely in part due to errors in deposition processes.

7.
Nature ; 523(7559): 212-6, 2015 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-26030523

RESUMEN

Understanding the diversity of human tissues is fundamental to disease and requires linking genetic information, which is identical in most of an individual's cells, with epigenetic mechanisms that could have tissue-specific roles. Surveys of DNA methylation in human tissues have established a complex landscape including both tissue-specific and invariant methylation patterns. Here we report high coverage methylomes that catalogue cytosine methylation in all contexts for the major human organ systems, integrated with matched transcriptomes and genomic sequence. By combining these diverse data types with each individuals' phased genome, we identified widespread tissue-specific differential CG methylation (mCG), partially methylated domains, allele-specific methylation and transcription, and the unexpected presence of non-CG methylation (mCH) in almost all human tissues. mCH correlated with tissue-specific functions, and using this mark, we made novel predictions of genes that escape X-chromosome inactivation in specific tissues. Overall, DNA methylation in several genomic contexts varies substantially among human tissues.


Asunto(s)
Metilación de ADN , Epigénesis Genética , Factores de Edad , Alelos , Mapeo Cromosómico , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Variación Genética , Humanos , Masculino , Especificidad de Órganos
8.
Nature ; 518(7539): 350-354, 2015 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-25693566

RESUMEN

Allelic differences between the two homologous chromosomes can affect the propensity of inheritance in humans; however, the extent of such differences in the human genome has yet to be fully explored. Here we delineate allelic chromatin modifications and transcriptomes among a broad set of human tissues, enabled by a chromosome-spanning haplotype reconstruction strategy. The resulting large collection of haplotype-resolved epigenomic maps reveals extensive allelic biases in both chromatin state and transcription, which show considerable variation across tissues and between individuals, and allow us to investigate cis-regulatory relationships between genes and their control sequences. Analyses of histone modification maps also uncover intriguing characteristics of cis-regulatory elements and tissue-restricted activities of repetitive elements. The rich data sets described here will enhance our understanding of the mechanisms by which cis-regulatory elements control gene expression programs.


Asunto(s)
Alelos , Epigénesis Genética/genética , Epigenómica , Haplotipos/genética , Acetilación , Cromatina/genética , Cromatina/metabolismo , Cromosomas Humanos/genética , Conjuntos de Datos como Asunto , Elementos de Facilitación Genéticos/genética , Variación Genética/genética , Histonas/metabolismo , Humanos , Motivos de Nucleótidos , Especificidad de Órganos/genética , Transcripción Genética/genética
9.
Nature ; 518(7539): 317-30, 2015 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-25693563

RESUMEN

The reference human genome sequence set the stage for studies of genetic variation and its association with human disease, but epigenomic studies lack a similar reference. To address this need, the NIH Roadmap Epigenomics Consortium generated the largest collection so far of human epigenomes for primary cells and tissues. Here we describe the integrative analysis of 111 reference human epigenomes generated as part of the programme, profiled for histone modification patterns, DNA accessibility, DNA methylation and RNA expression. We establish global maps of regulatory elements, define regulatory modules of coordinated activity, and their likely activators and repressors. We show that disease- and trait-associated genetic variants are enriched in tissue-specific epigenomic marks, revealing biologically relevant cell types for diverse human traits, and providing a resource for interpreting the molecular basis of human disease. Our results demonstrate the central role of epigenomic information for understanding gene regulation, cellular differentiation and human disease.


Asunto(s)
Epigénesis Genética/genética , Epigenómica , Genoma Humano/genética , Secuencia de Bases , Linaje de la Célula/genética , Células Cultivadas , Cromatina/química , Cromatina/genética , Cromatina/metabolismo , Cromosomas Humanos/química , Cromosomas Humanos/genética , Cromosomas Humanos/metabolismo , ADN/química , ADN/genética , ADN/metabolismo , Metilación de ADN , Conjuntos de Datos como Asunto , Elementos de Facilitación Genéticos/genética , Variación Genética/genética , Estudio de Asociación del Genoma Completo , Histonas/metabolismo , Humanos , Especificidad de Órganos/genética , ARN/genética , Valores de Referencia
11.
Proc Natl Acad Sci U S A ; 111(18): 6690-5, 2014 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-24757056

RESUMEN

During mammalian development, DNA methylation patterns need to be reset in primordial germ cells (PGCs) and preimplantation embryos. However, many LTR retrotransposons and imprinted genes are impervious to such global epigenetic reprogramming via hitherto undefined mechanisms. Here, we report that a subset of such genomic regions are resistant to widespread erasure of DNA methylation in mouse embryonic stem cells (mESCs) lacking the de novo DNA methyltransferases (Dnmts) Dnmt3a and Dnmt3b. Intriguingly, these loci are enriched for H3K9me3 in mESCs, implicating this mark in DNA methylation homeostasis. Indeed, deletion of the H3K9 methyltransferase SET domain bifurcated 1 (Setdb1) results in reduced H3K9me3 and DNA methylation levels at specific loci, concomitant with increased 5-hydroxymethylation (5hmC) and ten-eleven translocation 1 binding. Taken together, these data reveal that Setdb1 promotes the persistence of DNA methylation in mESCs, likely reflecting one mechanism by which DNA methylation is maintained at LTR retrotransposons and imprinted genes during developmental stages when DNA methylation is reprogrammed.


Asunto(s)
Metilación de ADN , Impresión Genómica , N-Metiltransferasa de Histona-Lisina/metabolismo , Retroelementos/genética , Animales , Células Cultivadas , ADN (Citosina-5-)-Metiltransferasas/deficiencia , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/genética , ADN Metiltransferasa 3A , Células Madre Embrionarias/metabolismo , Retrovirus Endógenos/genética , Retrovirus Endógenos/metabolismo , Epigénesis Genética , Femenino , N-Metiltransferasa de Histona-Lisina/deficiencia , N-Metiltransferasa de Histona-Lisina/genética , Histonas/química , Histonas/genética , Histonas/metabolismo , Masculino , Ratones , Ratones Noqueados , ADN Metiltransferasa 3B
12.
Trends Biochem Sci ; 37(4): 127-33, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22178137

RESUMEN

Retrotransposons, such as endogenous retroviruses (ERVs), have colonized the genomes of all metazoans. As retrotransposition can be deleterious, numerous pathways have evolved to repress the expression of these parasitic elements. For example, methylation of the fifth carbon of the cytosine base in DNA (5-methylcytosine, 5mC) is required for transcriptional silencing of ERVs in differentiated cells. However, this epigenetic mark is generally dispensable for ERV silencing during early stages of mouse embryogenesis and in mouse embryonic stem cells (mESCs). In this Opinion, we evaluate recent findings on the exceptional role of covalent modifications of histones in ERV silencing in these cell types. In addition, we discuss the potential role of TET proteins, which catalyze the oxidation of 5mC to 5-hydroxymethylcytosine (5hmC), in perturbing transcriptional silencing, and propose that histone modification-based pathways may be used to silence ERVs during those developmental stages when DNA methylation-mediated silencing is compromised.


Asunto(s)
Retrovirus Endógenos/genética , Histonas/metabolismo , 5-Metilcitosina/metabolismo , Animales , Metilación de ADN/genética , Embrión de Mamíferos/metabolismo , N-Metiltransferasa de Histona-Lisina , Ratones , Proteína Metiltransferasas/genética , Proteína Metiltransferasas/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
13.
Nature ; 464(7290): 927-31, 2010 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-20164836

RESUMEN

Endogenous retroviruses (ERVs), retrovirus-like elements with long terminal repeats, are widely dispersed in the euchromatic compartment in mammalian cells, comprising approximately 10% of the mouse genome. These parasitic elements are responsible for >10% of spontaneous mutations. Whereas DNA methylation has an important role in proviral silencing in somatic and germ-lineage cells, an additional DNA-methylation-independent pathway also functions in embryonal carcinoma and embryonic stem (ES) cells to inhibit transcription of the exogenous gammaretrovirus murine leukaemia virus (MLV). Notably, a recent genome-wide study revealed that ERVs are also marked by histone H3 lysine 9 trimethylation (H3K9me3) and H4K20me3 in ES cells but not in mouse embryonic fibroblasts. However, the role that these marks have in proviral silencing remains unexplored. Here we show that the H3K9 methyltransferase ESET (also called SETDB1 or KMT1E) and the Krüppel-associated box (KRAB)-associated protein 1 (KAP1, also called TRIM28) are required for H3K9me3 and silencing of endogenous and introduced retroviruses specifically in mouse ES cells. Furthermore, whereas ESET enzymatic activity is crucial for HP1 binding and efficient proviral silencing, the H4K20 methyltransferases Suv420h1 and Suv420h2 are dispensable for silencing. Notably, in DNA methyltransferase triple knockout (Dnmt1(-/-)Dnmt3a(-/-)Dnmt3b(-/-)) mouse ES cells, ESET and KAP1 binding and ESET-mediated H3K9me3 are maintained and ERVs are minimally derepressed. We propose that a DNA-methylation-independent pathway involving KAP1 and ESET/ESET-mediated H3K9me3 is required for proviral silencing during the period early in embryogenesis when DNA methylation is dynamically reprogrammed.


Asunto(s)
Células Madre Embrionarias/enzimología , Células Madre Embrionarias/virología , Retrovirus Endógenos/genética , Silenciador del Gen , N-Metiltransferasa de Histona-Lisina/metabolismo , Proteína Metiltransferasas/metabolismo , Provirus/genética , Animales , Línea Celular , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/deficiencia , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/genética , ADN Metiltransferasa 3A , Células Madre Embrionarias/metabolismo , Fibroblastos , Eliminación de Gen , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/deficiencia , N-Metiltransferasa de Histona-Lisina/genética , Ratones , Proteínas Nucleares/metabolismo , Proteína Metiltransferasas/deficiencia , Proteína Metiltransferasas/genética , Proteínas Represoras/metabolismo , Proteína 28 que Contiene Motivos Tripartito , ADN Metiltransferasa 3B
14.
Hum Mol Genet ; 21(11): 2572-87, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22378147

RESUMEN

Biallelic mutations of the DNA annealing helicase SMARCAL1 (SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin, subfamily a-like 1) cause Schimke immuno-osseous dysplasia (SIOD, MIM 242900), an incompletely penetrant autosomal recessive disorder. Using human, Drosophila and mouse models, we show that the proteins encoded by SMARCAL1 orthologs localize to transcriptionally active chromatin and modulate gene expression. We also show that, as found in SIOD patients, deficiency of the SMARCAL1 orthologs alone is insufficient to cause disease in fruit flies and mice, although such deficiency causes modest diffuse alterations in gene expression. Rather, disease manifests when SMARCAL1 deficiency interacts with genetic and environmental factors that further alter gene expression. We conclude that the SMARCAL1 annealing helicase buffers fluctuations in gene expression and that alterations in gene expression contribute to the penetrance of SIOD.


Asunto(s)
Alelos , Arteriosclerosis/genética , ADN Helicasas/genética , Expresión Génica , Síndromes de Inmunodeficiencia/genética , Mutación , Síndrome Nefrótico/genética , Osteocondrodisplasias/genética , Embolia Pulmonar/genética , Animales , Arteriosclerosis/metabolismo , Cromatina/metabolismo , ADN Helicasas/metabolismo , Modelos Animales de Enfermedad , Drosophila/enzimología , Embrión no Mamífero/metabolismo , Ambiente , Humanos , Síndromes de Inmunodeficiencia/metabolismo , Ratones , Síndrome Nefrótico/metabolismo , Osteocondrodisplasias/metabolismo , Penetrancia , Enfermedades de Inmunodeficiencia Primaria , Embolia Pulmonar/metabolismo
15.
Proc Natl Acad Sci U S A ; 108(14): 5718-23, 2011 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-21427230

RESUMEN

Methylation on lysine 9 of histone H3 (H3K9me) and DNA methylation play important roles in the transcriptional silencing of specific genes and repetitive elements. Both marks are detected on class I and II endogenous retroviruses (ERVs) in murine embryonic stem cells (mESCs). Recently, we reported that the H3K9-specific lysine methyltransferase (KMTase) Eset/Setdb1/KMT1E is required for H3K9me3 and the maintenance of silencing of ERVs in mESCs. In contrast, G9a/Ehmt2/KMT1C is dispensable, despite the fact that this KMTase is required for H3K9 dimethylation (H3K9me2) and efficient DNA methylation of these retroelements. Transcription of the exogenous retrovirus (XRV) Moloney murine leukemia virus is rapidly extinguished after integration in mESCs, concomitant with de novo DNA methylation. However, the role that H3K9 KMTases play in this process has not been addressed. Here, we demonstrate that G9a, but not Suv39h1 or Suv39h2, is required for silencing of newly integrated Moloney murine leukemia virus-based vectors in mESCs. The silencing defect in G9a(-/-) cells is accompanied by a reduction of H3K9me2 at the proviral LTR, indicating that XRVs are direct targets of G9a. Furthermore, de novo DNA methylation of newly integrated proviruses is impaired in the G9a(-/-) line, phenocopying proviral DNA methylation and silencing defects observed in Dnmt3a-deficient mESCs. Once established, however, maintenance of silencing of XRVs, like ERVs, is dependent exclusively on the KMTase Eset. Taken together, these observations reveal that in mESCs, the H3K9 KMTase G9a is required for the establishment, but not for the maintenance, of silencing of newly integrated proviruses.


Asunto(s)
Metilación de ADN/genética , Células Madre Embrionarias/virología , N-Metiltransferasa de Histona-Lisina/metabolismo , Virus de la Leucemia Murina de Moloney/genética , Animales , Western Blotting , Inmunoprecipitación de Cromatina , ADN (Citosina-5-)-Metiltransferasas/genética , Retrovirus Endógenos/genética , Citometría de Flujo , Silenciador del Gen , Vectores Genéticos/genética , N-Metiltransferasa de Histona-Lisina/genética , Ratones , Ratones Noqueados , Provirus/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Nat Commun ; 15(1): 15, 2024 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-38167730

RESUMEN

SETDB1 is an essential histone methyltransferase that deposits histone H3 lysine 9 trimethylation (H3K9me3) to transcriptionally repress genes and repetitive elements. The function of differential H3K9me3 enrichment between cell-types remains unclear. Here, we demonstrate mutual exclusivity of H3K9me3 and CTCF across mouse tissues from different developmental timepoints. We analyze SETDB1 depleted cells and discover that H3K9me3 prevents aberrant CTCF binding independently of DNA methylation and H3K9me2. Such sites are enriched with SINE B2 retrotransposons. Moreover, analysis of higher-order genome architecture reveals that large chromatin structures including topologically associated domains and subnuclear compartments, remain intact in SETDB1 depleted cells. However, chromatin loops and local 3D interactions are disrupted, leading to transcriptional changes by modifying pre-existing chromatin landscapes. Specific genes with altered expression show differential interactions with dysregulated cis-regulatory elements. Collectively, we find that cell-type specific targets of SETDB1 maintain cellular identities by modulating CTCF binding, which shape nuclear architecture and transcriptomic networks.


Asunto(s)
Cromatina , Histonas , Animales , Ratones , Histonas/metabolismo , Metilación de ADN , Retroelementos , Secuencias Reguladoras de Ácidos Nucleicos , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo
17.
Nat Cell Biol ; 25(7): 1047-1060, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37400500

RESUMEN

During pregnancy the maternal-fetal interface plays vital roles in fetal development. Its disruption is frequently found in pregnancy complications. Recent studies show increased incidences of adverse pregnancy outcomes in patients with COVID-19; however, the mechanism remains unclear. Here we analysed the molecular impacts of SARS-CoV-2 infection on the maternal-fetal interface. Generating bulk and single-nucleus transcriptomic and epigenomic profiles from patients with COVID-19 and control samples, we discovered aberrant immune activation and angiogenesis patterns in distinct cells from patients. Surprisingly, retrotransposons were also dysregulated in specific cell types. Notably, reduced enhancer activities of LTR8B elements were functionally linked to the downregulation of pregnancy-specific glycoprotein genes in syncytiotrophoblasts. Our findings revealed that SARS-CoV-2 infection induced substantial changes to the epigenome and transcriptome at the maternal-fetal interface, which may be associated with pregnancy complications.


Asunto(s)
COVID-19 , Complicaciones Infecciosas del Embarazo , Embarazo , Femenino , Humanos , COVID-19/genética , Transcriptoma , SARS-CoV-2 , Epigenómica , Complicaciones Infecciosas del Embarazo/genética , Análisis de la Célula Individual
18.
EMBO J ; 27(20): 2691-701, 2008 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-18818693

RESUMEN

Histone H3K9 methylation is required for DNA methylation and silencing of repetitive elements in plants and filamentous fungi. In mammalian cells however, deletion of the H3K9 histone methyltransferases (HMTases) Suv39h1 and Suv39h2 does not affect DNA methylation of the endogenous retrovirus murine leukaemia virus, indicating that H3K9 methylation is dispensable for DNA methylation of retrotransposons, or that a different HMTase is involved. We demonstrate that embryonic stem (ES) cells lacking the H3K9 HMTase G9a show a significant reduction in DNA methylation of retrotransposons, major satellite repeats and densely methylated CpG-rich promoters. Surprisingly, demethylated retrotransposons remain transcriptionally silent in G9a(-/-) cells, and show only a modest decrease in H3K9me2 and no decrease in H3K9me3 or HP1alpha binding, indicating that H3K9 methylation per se is not the relevant trigger for DNA methylation. Indeed, introduction of catalytically inactive G9a transgenes partially 'rescues' the DNA methylation defect observed in G9a(-/-) cells. Taken together, these observations reveal that H3K9me3 and HP1alpha recruitment to retrotransposons occurs independent of DNA methylation in ES cells and that G9a promotes DNA methylation independent of its HMTase activity.


Asunto(s)
Metilación de ADN , Células Madre Embrionarias/citología , Regulación Enzimológica de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/metabolismo , Metiltransferasas/metabolismo , Animales , Catálisis , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/metabolismo , Islas de CpG , Histonas/química , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Modelos Genéticos
19.
Hong Kong Med J ; 18(4): 318-24, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22865176

RESUMEN

OBJECTIVE. Medical professionalism has been widely discussed in western scholarly literature. However, since Hong Kong has a mixed Chinese-western culture, it remains uncertain whether Hong Kong health care professionals, medical students, and patients see medical professionalism in exactly the same way as westerners. The objective of the present study was to explore perceptions of medical professionalism in Hong Kong. DESIGN. Individual semi-structured interviews. SETTING. Medical faculty preceptors, residents, interns, nurses, and students from the Li Ka Shing Faculty of Medicine of the University of Hong Kong. Subjects were recruited at an out-patient clinic of Queen Mary Hospital. PARTICIPANTS. We interviewed 39 subjects, including six medical faculty preceptors, six hospital residents, four medical interns, eight nurses, eight out-patients, and seven medical students. The interviews were transcribed and coded. Grounded theory was employed for framing and analysing the interviews. RESULTS. A total of 30 primary themes were identified and grouped under three secondary themes, ie 'Expectations of a professional doctor', 'Work values', and 'Patient care'. In general, the primary themes were consistent with recognised professional attributes in western bioethics, such as knowledge and skills, holistic care, and communication skills. A closer analysis suggested that traditional Chinese thought also played an important role in shaping the medical professionalism of Hong Kong. Challenges to be faced by Hong Kong doctors due to recent social changes were also identified. CONCLUSIONS. Medical professionalism in Hong Kong is shaped by both western medical ethics and traditional Chinese thought. The values treasured by Hong Kong health care professionals as well as technological advance, and the city's proximity to Mainland China makes Hong Kong health care unique. It is important to maintain the present work attitudes and at the same time adapt to new social changes.


Asunto(s)
Competencia Clínica , Investigación Cualitativa , Comunicación , Hong Kong , Humanos , Entrevistas como Asunto , Atención al Paciente , Percepción , Relaciones Médico-Paciente
20.
Genes Genomics ; 43(3): 227-236, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33638813

RESUMEN

Chronic inflammatory diseases (CIDs) have complex pathologies that result from aberrant and persistent immune responses. However, the precise triggers and mechanisms remain elusive. An important aspect of CID research focuses on epigenetics modifications, which regulate gene expression and provide a dynamic transcriptional response to inflammation. In recent years, mounting evidence has demonstrated an association between epigenomic and transcriptomic dysregulation and the phenotypes of CIDs. In particular, epigenetic changes at cis-regulatory elements have provided new insights for immune cell-specific alterations that contribute to disease etiology. Furthermore, the advancements in single-cell genomics provide novel solutions to cell type heterogeneity, which has long posed challenges for CID diagnosis and treatment. In this review, we discuss the current state of epigenomics research of CID and the insights derived from single-cell transcriptomic and epigenomic studies.


Asunto(s)
Epigénesis Genética , Inflamación/genética , Animales , Enfermedad Crónica , Perfilación de la Expresión Génica , Humanos , Inflamación/diagnóstico , Ratones , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA