Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Stem Cell Reports ; 17(6): 1476-1492, 2022 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-35523177

RESUMEN

Advances in the study of neurological conditions have been possible because of pluripotent stem cell technologies and organoids. Studies have described the generation of neural ectoderm-derived retinal and brain structures from pluripotent stem cells. However, the field is still troubled by technical challenges, including high culture costs and variability. Here, we describe a simple and economical protocol that reproducibly gives rise to the neural retina and cortical brain regions from confluent cultures of stem cells. The spontaneously generated cortical organoids are transcriptionally comparable with organoids generated by other methods. Furthermore, these organoids showed spontaneous functional network activity and proteomic analysis confirmed organoids maturity. The generation of retinal and brain organoids in close proximity enabled their mutual isolation. Suspension culture of this complex organoid system demonstrated the formation of nerve-like structures connecting retinal and brain organoids, which might facilitate the investigation of neurological diseases of the eye and brain.


Asunto(s)
Células Madre Pluripotentes Inducidas , Células Madre Pluripotentes , Encéfalo , Diferenciación Celular , Organoides , Proteómica , Retina
2.
Cell Rep ; 36(12): 109722, 2021 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-34551299

RESUMEN

DNA replication timing and three-dimensional (3D) genome organization are associated with distinct epigenome patterns across large domains. However, whether alterations in the epigenome, in particular cancer-related DNA hypomethylation, affects higher-order levels of genome architecture is still unclear. Here, using Repli-Seq, single-cell Repli-Seq, and Hi-C, we show that genome-wide methylation loss is associated with both concordant loss of replication timing precision and deregulation of 3D genome organization. Notably, we find distinct disruption in 3D genome compartmentalization, striking gains in cell-to-cell replication timing heterogeneity and loss of allelic replication timing in cancer hypomethylation models, potentially through the gene deregulation of DNA replication and genome organization pathways. Finally, we identify ectopic H3K4me3-H3K9me3 domains from across large hypomethylated domains, where late replication is maintained, which we purport serves to protect against catastrophic genome reorganization and aberrant gene transcription. Our results highlight a potential role for the methylome in the maintenance of 3D genome regulation.


Asunto(s)
Metilación de ADN , Momento de Replicación del ADN/fisiología , Genoma Humano , Línea Celular Tumoral , Cromatina/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1/genética , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Bases de Datos Genéticas , Expresión Génica , Histonas/metabolismo , Humanos , Análisis de Secuencia de ADN/métodos
3.
Nat Commun ; 11(1): 54, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31911579

RESUMEN

The architectural protein CTCF is a mediator of chromatin conformation, but how CTCF binding to DNA is orchestrated to maintain long-range gene expression is poorly understood. Here we perform RNAi knockdown to reduce CTCF levels and reveal a shared subset of CTCF-bound sites are robustly resistant to protein depletion. The 'persistent' CTCF sites are enriched at domain boundaries and chromatin loops constitutive to all cell types. CRISPR-Cas9 deletion of 2 persistent CTCF sites at the boundary between a long-range epigenetically active (LREA) and silenced (LRES) region, within the Kallikrein (KLK) locus, results in concordant activation of all 8 KLK genes within the LRES region. CTCF genome-wide depletion results in alteration in Topologically Associating Domain (TAD) structure, including the merging of TADs, whereas TAD boundaries are not altered where persistent sites are maintained. We propose that the subset of essential CTCF sites are involved in cell-type constitutive, higher order chromatin architecture.


Asunto(s)
Factor de Unión a CCCTC/metabolismo , Cromatina/metabolismo , Epigénesis Genética , Sitios de Unión , Factor de Unión a CCCTC/genética , Cromatina/química , Cromatina/genética , ADN/genética , ADN/metabolismo , Humanos , Regiones Promotoras Genéticas , Unión Proteica , Dominios Proteicos
4.
Nat Commun ; 11(1): 320, 2020 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-31949157

RESUMEN

Endocrine therapy resistance frequently develops in estrogen receptor positive (ER+) breast cancer, but the underlying molecular mechanisms are largely unknown. Here, we show that 3-dimensional (3D) chromatin interactions both within and between topologically associating domains (TADs) frequently change in ER+ endocrine-resistant breast cancer cells and that the differential interactions are enriched for resistance-associated genetic variants at CTCF-bound anchors. Ectopic chromatin interactions are preferentially enriched at active enhancers and promoters and ER binding sites, and are associated with altered expression of ER-regulated genes, consistent with dynamic remodelling of ER pathways accompanying the development of endocrine resistance. We observe that loss of 3D chromatin interactions often occurs coincidently with hypermethylation and loss of ER binding. Alterations in active A and inactive B chromosomal compartments are also associated with decreased ER binding and atypical interactions and gene expression. Together, our results suggest that 3D epigenome remodelling is a key mechanism underlying endocrine resistance in ER+ breast cancer.


Asunto(s)
Sitios de Unión , Neoplasias de la Mama/genética , Cromatina/metabolismo , Epigénesis Genética , Receptores de Estrógenos/química , Receptores de Estrógenos/metabolismo , Antineoplásicos Hormonales/farmacología , Neoplasias de la Mama/metabolismo , Factor de Unión a CCCTC/química , Factor de Unión a CCCTC/metabolismo , Cromatina/química , Cromatina/genética , Metilación de ADN , Epigénesis Genética/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Células MCF-7 , Proteínas de Neoplasias/genética , Regiones Promotoras Genéticas/efectos de los fármacos , Dominios y Motivos de Interacción de Proteínas , Secuenciación Completa del Genoma
5.
iScience ; 21: 773-788, 2019 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-31727574

RESUMEN

Acetylation of the histone variant H2A.Z (H2A.Zac) occurs at active regulatory regions associated with gene expression. Although the Tip60 complex is proposed to acetylate H2A.Z, functional studies suggest additional enzymes are involved. Here, we show that p300 acetylates H2A.Z at multiple lysines. In contrast, we found that although Tip60 does not efficiently acetylate H2A.Z in vitro, genetic inhibition of Tip60 reduces H2A.Zac in cells. Importantly, we found that interaction between the p300-bromodomain and H4 acetylation (H4ac) enhances p300-driven H2A.Zac. Indeed, H2A.Zac and H4ac show high genomic overlap, especially at active promoters. We also reveal unique chromatin features and transcriptional states at enhancers correlating with co-occurrence or exclusivity of H4ac and H2A.Zac. We propose that differential H4 and H2A.Z acetylation signatures can also define the enhancer state. In conclusion, we show both Tip60 and p300 contribute to H2A.Zac and reveal molecular mechanisms of writer/reader crosstalk between H2A.Z and H4 acetylation through p300.

6.
Stem Cell Reports ; 13(4): 669-683, 2019 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-31522977

RESUMEN

Here we elucidate the effect of Alzheimer disease (AD)-predisposing genetic backgrounds, APOE4, PSEN1ΔE9, and APPswe, on functionality of human microglia-like cells (iMGLs). We present a physiologically relevant high-yield protocol for producing iMGLs from induced pluripotent stem cells. Differentiation is directed with small molecules through primitive erythromyeloid progenitors to re-create microglial ontogeny from yolk sac. The iMGLs express microglial signature genes and respond to ADP with intracellular Ca2+ release distinguishing them from macrophages. Using 16 iPSC lines from healthy donors, AD patients and isogenic controls, we reveal that the APOE4 genotype has a profound impact on several aspects of microglial functionality, whereas PSEN1ΔE9 and APPswe mutations trigger minor alterations. The APOE4 genotype impairs phagocytosis, migration, and metabolic activity of iMGLs but exacerbates their cytokine secretion. This indicates that APOE4 iMGLs are fundamentally unable to mount normal microglial functionality in AD.


Asunto(s)
Precursor de Proteína beta-Amiloide/genética , Apolipoproteína E4/genética , Diferenciación Celular/genética , Células Madre Pluripotentes Inducidas/metabolismo , Microglía/metabolismo , Fenotipo , Presenilina-1/genética , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Apolipoproteína E4/metabolismo , Calcio/metabolismo , Células Cultivadas , Quimiocinas/metabolismo , Citocinas/metabolismo , Hematopoyesis , Humanos , Células Madre Pluripotentes Inducidas/citología , Mediadores de Inflamación/metabolismo , Microglía/citología , Mutación , Fagocitosis , Presenilina-1/metabolismo , Proteolisis
7.
Cancer Cell ; 35(2): 297-314.e8, 2019 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-30753827

RESUMEN

Promoter CpG islands are typically unmethylated in normal cells, but in cancer a proportion are subject to hypermethylation. Using methylome sequencing we identified CpG islands that display partial methylation encroachment across the 5' or 3' CpG island borders. CpG island methylation encroachment is widespread in prostate and breast cancer and commonly associates with gene suppression. We show that the pattern of H3K4me1 at CpG island borders in normal cells predicts the different modes of cancer CpG island hypermethylation. Notably, genetic manipulation of Kmt2d results in concordant alterations in H3K4me1 levels and CpG island border DNA methylation encroachment. Our findings suggest a role for H3K4me1 in the demarcation of CpG island methylation borders in normal cells, which become eroded in cancer.


Asunto(s)
Islas de CpG , Metilación de ADN , ADN de Neoplasias/metabolismo , Histonas/metabolismo , Neoplasias/metabolismo , 5-Metilcitosina/análogos & derivados , 5-Metilcitosina/metabolismo , Animales , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/genética , Humanos , Masculino , Metilación , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/genética , Regiones Promotoras Genéticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA