Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38798672

RESUMEN

Synovial sarcoma (SyS) is an aggressive soft-tissue malignancy characterized by a pathognomonic chromosomal translocation leading to the formation of the SS18::SSX fusion oncoprotein. SS18::SSX associates with mammalian BAF complexes suggesting deregulation of chromatin architecture as the oncogenic driver in this tumour type. To examine the epigenomic state of SyS we performed comprehensive multi-omics analysis on 52 primary pre-treatment human SyS tumours. Our analysis revealed a continuum of epigenomic states across the cohort at fusion target genes independent of rare somatic genetic lesions. We identify cell-of-origin signatures defined by enhancer states and reveal unexpected relationships between H2AK119Ub1 and active marks. The number of bivalent promoters, dually marked by the repressive H3K27me3 and activating H3K4me3 marks, has strong prognostic value and outperforms tumor grade in predicting patient outcome. Finally, we identify SyS defining epigenomic features including H3K4me3 expansion associated with striking promoter DNA hypomethylation in which SyS displays the lowest mean methylation level of any sarcoma subtype. We explore these distinctive features as potential vulnerabilities in SyS and identify H3K4me3 inhibition as a promising therapeutic strategy.

2.
iScience ; 26(6): 106795, 2023 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-37213235

RESUMEN

Runt-related transcription factor 1 (RUNX1) is oncogenic in diverse types of leukemia and epithelial cancers where its expression is associated with poor prognosis. Current models suggest that RUNX1 cooperates with other oncogenic factors (e.g., NOTCH1, TAL1) to drive the expression of proto-oncogenes in T cell acute lymphoblastic leukemia (T-ALL) but the molecular mechanisms controlled by RUNX1 and its cooperation with other factors remain unclear. Integrative chromatin and transcriptional analysis following inhibition of RUNX1 and NOTCH1 revealed a surprisingly widespread role of RUNX1 in the establishment of global H3K27ac levels and that RUNX1 is required by NOTCH1 for cooperative transcription activation of key NOTCH1 target genes including MYC, DTX1, HES4, IL7R, and NOTCH3. Super-enhancers were preferentially sensitive to RUNX1 knockdown and RUNX1-dependent super-enhancers were disrupted following the treatment of a pan-BET inhibitor, I-BET151.

3.
Leukemia ; 36(3): 809-820, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34588613

RESUMEN

TET2 loss-of-function mutations are recurrent events in a wide range of hematological malignancies and a physiologic occurrence in blood cells of healthy older adults. It is currently unknown what determines if a person harboring a somatic TET2 mutation will progress to myelodysplastic syndrome or acute myeloid leukemia. Here we develop a zebrafish tet2 mutant through which we show that tet2 loss leads to restricted hematopoietic differentiation combined with a modest upregulation of p53, which is also characteristic of many inherited bone marrow failure syndromes. Uniquely in the context of emergency hematopoiesis by external stimuli, such as infection or cytokine stimulation, lack of tet2 leads hematopoietic stem cells to undergo excessive proliferation, resulting in an accumulation of immature cells, which are poised to become leukemogenic following additional genetic/epigenetic perturbations. This same phenomenon observed in zebrafish extends to human hematopoietic stem cells, identifying TET2 as a critical relay switch in the context of stress hematopoiesis.


Asunto(s)
Dioxigenasas/genética , Neoplasias Hematológicas/genética , Hematopoyesis , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Animales , Proliferación Celular , Modelos Animales de Enfermedad , Eliminación de Gen , Silenciador del Gen , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Leucemia Mieloide Aguda/genética , Mutación con Pérdida de Función , Síndromes Mielodisplásicos/genética
5.
Blood ; 136(24): 2764-2773, 2020 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-33301029

RESUMEN

Hematopoietic clones with leukemogenic mutations arise in healthy people as they age, but progression to acute myeloid leukemia (AML) is rare. Recent evidence suggests that the microenvironment may play an important role in modulating human AML population dynamics. To investigate this concept further, we examined the combined and separate effects of an oncogene (c-MYC) and exposure to interleukin-3 (IL-3), granulocyte-macrophage colony-stimulating factor (GM-CSF), and stem cell factor (SCF) on the experimental genesis of a human AML in xenografted immunodeficient mice. Initial experiments showed that normal human CD34+ blood cells transduced with a lentiviral MYC vector and then transplanted into immunodeficient mice produced a hierarchically organized, rapidly fatal, and serially transplantable blast population, phenotypically and transcriptionally similar to human AML cells, but only in mice producing IL-3, GM-CSF, and SCF transgenically or in regular mice in which the cells were exposed to IL-3 or GM-CSF delivered using a cotransduction strategy. In their absence, the MYC+ human cells produced a normal repertoire of lymphoid and myeloid progeny in transplanted mice for many months, but, on transfer to secondary mice producing the human cytokines, the MYC+ cells rapidly generated AML. Indistinguishable diseases were also obtained efficiently from both primitive (CD34+CD38-) and late granulocyte-macrophage progenitor (GMP) cells. These findings underscore the critical role that these cytokines can play in activating a malignant state in normally differentiating human hematopoietic cells in which MYC expression has been deregulated. They also introduce a robust experimental model of human leukemogenesis to further elucidate key mechanisms involved and test strategies to suppress them.


Asunto(s)
Regulación Leucémica de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Interleucina-3/metabolismo , Leucemia Mieloide Aguda/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Animales , Línea Celular Tumoral , Xenoinjertos , Humanos , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Trasplante de Neoplasias
6.
Epigenomics ; 12(12): 1053-1070, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32677466

RESUMEN

Aim: To provide a comprehensive understanding of gene regulatory networks in the developing human brain and a foundation for interpreting pathogenic deregulation. Materials & methods: We generated reference epigenomes and transcriptomes of dissected brain regions and primary neural progenitor cells (NPCs) derived from cortical and ganglionic eminence tissues of four normal human fetuses. Results: Integration of these data across developmental stages revealed a directional increase in active regulatory states, transcription factor activities and gene transcription with developmental stage. Consistent with differences in their biology, NPCs derived from cortical and ganglionic eminence regions contained common, region specific, and gestational week specific regulatory states. Conclusion: We provide a high-resolution regulatory network for NPCs from different brain regions as a comprehensive reference for future studies.


Asunto(s)
Encéfalo/embriología , Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Epigenoma , Femenino , Feto , Humanos , Células-Madre Neurales , Embarazo , Transcriptoma , Gemelos
7.
Cancer Res ; 80(17): 3480-3491, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32641414

RESUMEN

The somatic missense point mutation c.402C>G (p.C134W) in the FOXL2 transcription factor is pathognomonic for adult-type granulosa cell tumors (AGCT) and a diagnostic marker for this tumor type. However, the molecular consequences of this mutation and its contribution to the mechanisms of AGCT pathogenesis remain unclear. To explore these mechanisms, we engineered V5-FOXL2WT- and V5-FOXL2C134W-inducible isogenic cell lines and performed chromatin immunoprecipitation sequencing and transcriptome profiling. FOXL2C134W associated with the majority of the FOXL2 wild-type DNA elements as well as a large collection of unique elements genome wide. This model enabled confirmation of altered DNA-binding specificity for FOXL2C134W and identification of unique targets of FOXL2C134W including SLC35F2, whose expression increased sensitivity to YM155. Our results suggest FOXL2C134W drives AGCT by altering the binding affinity of FOXL2-containing complexes to engage an oncogenic transcriptional program. SIGNIFICANCE: A mechanistic understanding of FOXL2C134W-induced regulatory state alterations drives discovery of a rationally designed therapeutic strategy.


Asunto(s)
ADN/metabolismo , Proteína Forkhead Box L2/genética , Proteína Forkhead Box L2/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Tumor de Células de la Granulosa/genética , Línea Celular Tumoral , Femenino , Tumor de Células de la Granulosa/metabolismo , Humanos , Mutación Missense , Mutación Puntual , Unión Proteica
8.
Epigenetics Chromatin ; 12(1): 63, 2019 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-31601272

RESUMEN

BACKGROUND: MeCP2-a chromatin-binding protein associated with Rett syndrome-has two main isoforms, MeCP2-E1 and MeCP2-E2, differing in a few N-terminal amino acid residues. Previous studies have shown brain region-specific expression of these isoforms which, in addition to their different cellular localization and differential expression during brain development, suggest that they may also have non-overlapping molecular mechanisms. However, differential functions of MeCP2-E1 and E2 remain largely unexplored. RESULTS: Here, we show that the N-terminal domains (NTD) of MeCP2-E1 and E2 modulate the ability of the methyl-binding domain (MBD) to interact with DNA as well as influencing the turn-over rates, binding dynamics, response to neuronal depolarization, and circadian oscillations of the two isoforms. Our proteomics data indicate that both isoforms exhibit unique interacting protein partners. Moreover, genome-wide analysis using ChIP-seq provide evidence for a shared as well as a specific regulation of different sets of genes. CONCLUSIONS: Our study supports the idea that Rett syndrome might arise from simultaneous impairment of cellular processes involving non-overlapping functions of MECP2 isoforms. For instance, MeCP2-E1 mutations might impact stimuli-dependent chromatin regulation, while MeCP2-E2 mutations could result in aberrant ribosomal expression. Overall, our findings provide insight into the functional complexity of MeCP2 by dissecting differential aspects of its two isoforms.


Asunto(s)
ADN/metabolismo , Proteína 2 de Unión a Metil-CpG/metabolismo , Animales , Línea Celular Tumoral , Cromatina/metabolismo , Ritmo Circadiano/genética , Humanos , Proteína 2 de Unión a Metil-CpG/química , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Ratones Noqueados , Neuronas/metabolismo , Unión Proteica , Dominios Proteicos , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratas , Ratas Sprague-Dawley , Síndrome de Rett/genética , Síndrome de Rett/patología
10.
Cell Stem Cell ; 23(5): 714-726.e7, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30269902

RESUMEN

Acute leukemias are aggressive malignancies of developmentally arrested hematopoietic progenitors. We sought here to explore the possibility that changes in hematopoietic stem/progenitor cells during development might alter the biology of leukemias arising from this tissue compartment. Using a mouse model of acute T cell leukemia, we found that leukemias generated from fetal liver (FL) and adult bone marrow (BM) differed dramatically in their leukemia stem cell activity with FL leukemias showing markedly reduced serial transplantability as compared to BM leukemias. We present evidence that this difference is due to NOTCH1-driven autocrine IGF1 signaling, which is active in FL cells but restrained in BM cells by EZH2-dependent H3K27 trimethylation. Further, we confirmed this mechanism is operative in human disease and show that enforced IGF1 signaling effectively limits leukemia stem cell activity. These findings demonstrate that resurrecting dormant fetal programs in adult cells may represent an alternate therapeutic approach in human cancer.


Asunto(s)
Células de la Médula Ósea/metabolismo , Epigénesis Genética/genética , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Células Madre Neoplásicas/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Transducción de Señal/genética , Animales , Células de la Médula Ósea/patología , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Células Madre Neoplásicas/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología
11.
J Endocrinol ; 235(2): 153-165, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28808080

RESUMEN

The thyroid gland, necessary for normal human growth and development, functions as an essential regulator of metabolism by the production and secretion of appropriate levels of thyroid hormone. However, assessment of abnormal thyroid function may be challenging suggesting a more fundamental understanding of normal function is needed. One way to characterize normal gland function is to study the epigenome and resulting transcriptome within its constituent cells. This study generates the first published reference epigenomes for human thyroid from four individuals using ChIP-seq and RNA-seq. We profiled six histone modifications (H3K4me1, H3K4me3, H3K27ac, H3K36me3, H3K9me3, H3K27me3), identified chromatin states using a hidden Markov model, produced a novel quantitative metric for model selection and established epigenomic maps of 19 chromatin states. We found that epigenetic features characterizing promoters and transcription elongation tend to be more consistent than regions characterizing enhancers or Polycomb-repressed regions and that epigenetically active genes consistent across all epigenomes tend to have higher expression than those not marked as epigenetically active in all epigenomes. We also identified a set of 18 genes epigenetically active and consistently expressed in the thyroid that are likely highly relevant to thyroid function. Altogether, these epigenomes represent a powerful resource to develop a deeper understanding of the underlying molecular biology of thyroid function and provide contextual information of thyroid and human epigenomic data for comparison and integration into future studies.


Asunto(s)
Epigénesis Genética/fisiología , Epigenómica/métodos , Regulación de la Expresión Génica/fisiología , Glándula Tiroides/fisiología , Cromatina , Histonas/genética , Histonas/metabolismo , Humanos , Regiones Promotoras Genéticas , Transcriptoma
12.
Nature ; 549(7671): 227-232, 2017 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-28854171

RESUMEN

Human glioblastomas harbour a subpopulation of glioblastoma stem cells that drive tumorigenesis. However, the origin of intratumoural functional heterogeneity between glioblastoma cells remains poorly understood. Here we study the clonal evolution of barcoded glioblastoma cells in an unbiased way following serial xenotransplantation to define their individual fate behaviours. Independent of an evolving mutational signature, we show that the growth of glioblastoma clones in vivo is consistent with a remarkably neutral process involving a conserved proliferative hierarchy rooted in glioblastoma stem cells. In this model, slow-cycling stem-like cells give rise to a more rapidly cycling progenitor population with extensive self-maintenance capacity, which in turn generates non-proliferative cells. We also identify rare 'outlier' clones that deviate from these dynamics, and further show that chemotherapy facilitates the expansion of pre-existing drug-resistant glioblastoma stem cells. Finally, we show that functionally distinct glioblastoma stem cells can be separately targeted using epigenetic compounds, suggesting new avenues for glioblastoma-targeted therapy.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Rastreo Celular , Glioblastoma/patología , Células Madre Neoplásicas/patología , Animales , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Proliferación Celular , Células Clonales/efectos de los fármacos , Células Clonales/patología , Epigénesis Genética , Femenino , Glioblastoma/tratamiento farmacológico , Xenoinjertos , Humanos , Ratones , Invasividad Neoplásica , Trasplante de Neoplasias , Células Madre Neoplásicas/efectos de los fármacos , Fenotipo , Procesos Estocásticos
13.
Exp Hematol ; 53: 48-58, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28602946

RESUMEN

We report here the first use of whole-genome sequencing (WGS) to examine the initial clonal dynamics in an unusual patient with chronic myeloid leukemia (CML), who presented in chronic phase (CP) with doubly marked BCR-ABL1+/JAK2V617F-mutant cells and, over a 9-year period, progressed into an accelerated phase (AP) and then terminal blast phase (BP). WGS revealed that the diagnostic cells also contained mutations in ASXL1, SEC23B, MAD1L1, and RREB1 as well as 12,000 additional uncommon DNA variants. WGS of endothelial cells generated from circulating precursors revealed many of these were shared with the CML clone. Surprisingly, WGS of induced pluripotent stem cells (iPSCs) derived from the AP cells revealed only six additional coding somatic mutations, despite retention by the hematopoietic progeny of the parental AP cell levels of BCR-ABL1 expression and sensitivity to imatinib and pimozide. Limited analysis of BP cells revealed independent subclonal progression to homozygosity of the MAD1L1 and RREB1 variants. MAD1L1 and SEC23B mutations were also identified in 2 of 101 cases of myeloproliferative neoplasms, but not in 42 healthy subjects. These findings challenge historic concepts of clonal evolution in CML.


Asunto(s)
Janus Quinasa 2/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Mutación , Proteínas de Ciclo Celular/genética , Proteínas de Unión al ADN/genética , Estudio de Asociación del Genoma Completo , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Masculino , Persona de Mediana Edad , Proteínas Nucleares/genética , Factores de Transcripción/genética
14.
Cell Rep ; 17(8): 2060-2074, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27851968

RESUMEN

The normal adult human mammary gland is a continuous bilayered epithelial system. Bipotent and myoepithelial progenitors are prominent and unique components of the outer (basal) layer. The inner (luminal) layer includes both luminal-restricted progenitors and a phenotypically separable fraction that lacks progenitor activity. We now report an epigenomic comparison of these three subsets with one another, with their associated stromal cells, and with three immortalized, non-tumorigenic human mammary cell lines. Each genome-wide analysis contains profiles for six histone marks, methylated DNA, and RNA transcripts. Analysis of these datasets shows that each cell type has unique features, primarily within genomic regulatory regions, and that the cell lines group together. Analyses of the promoter and enhancer profiles place the luminal progenitors in between the basal cells and the non-progenitor luminal subset. Integrative analysis reveals networks of subset-specific transcription factors.


Asunto(s)
Mama/metabolismo , Elementos de Facilitación Genéticos/genética , Epigénesis Genética , Redes Reguladoras de Genes , Factores de Transcripción/metabolismo , Adulto , Separación Celular , Cromatina/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Humanos , Fenotipo , Regiones Promotoras Genéticas , Reproducibilidad de los Resultados
15.
Cell Rep ; 17(8): 2112-2124, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27851972

RESUMEN

Nucleosome position, density, and post-translational modification are widely accepted components of mechanisms regulating DNA transcription but still incompletely understood. We present a modified native ChIP-seq method combined with an analytical framework that allows MNase accessibility to be integrated with histone modification profiles. Application of this methodology to the primitive (CD34+) subset of normal human cord blood cells enabled genomic regions enriched in one versus two nucleosomes marked by histone 3 lysine 4 trimethylation (H3K4me3) and/or histone 3 lysine 27 trimethylation (H3K27me3) to be associated with their transcriptional and DNA methylation states. From this analysis, we defined four classes of promoter-specific profiles and demonstrated that a majority of bivalent marked promoters are heterogeneously marked at a single-cell level in this primitive cell type. Interestingly, extension of this approach to human embryonic stem cells revealed an altered relationship between chromatin modification state and nucleosome content at promoters, suggesting developmental stage-specific organization of histone methylation states.


Asunto(s)
Inmunoprecipitación de Cromatina , Nucleosomas/metabolismo , Análisis de Secuencia de ARN , Antígenos CD34/metabolismo , Islas de CpG/genética , ADN/metabolismo , Metilación de ADN/genética , Sangre Fetal/citología , Sangre Fetal/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Histonas/metabolismo , Células Madre Embrionarias Humanas/metabolismo , Humanos , Nucleasa Microcócica/metabolismo , Regiones Promotoras Genéticas , Procesamiento Proteico-Postraduccional , ARN/genética , ARN/metabolismo
16.
Cancer Cell ; 29(3): 394-406, 2016 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-26977886

RESUMEN

Malignant rhabdoid tumors (MRTs) are rare lethal tumors of childhood that most commonly occur in the kidney and brain. MRTs are driven by SMARCB1 loss, but the molecular consequences of SMARCB1 loss in extra-cranial tumors have not been comprehensively described and genomic resources for analyses of extra-cranial MRT are limited. To provide such data, we used whole-genome sequencing, whole-genome bisulfite sequencing, whole transcriptome (RNA-seq) and microRNA sequencing (miRNA-seq), and histone modification profiling to characterize extra-cranial MRTs. Our analyses revealed gene expression and methylation subgroups and focused on dysregulated pathways, including those involved in neural crest development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Tumor Rabdoide/genética , Línea Celular Tumoral , Proteínas Cromosómicas no Histona/genética , Metilación de ADN/genética , Proteínas de Unión al ADN/genética , Expresión Génica/genética , Histonas/genética , Humanos , MicroARNs/genética , Proteína SMARCB1 , Factores de Transcripción/genética , Transcriptoma/genética
17.
Nature ; 528(7581): 267-71, 2015 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-26633636

RESUMEN

Most human breast cancers have diversified genomically and biologically by the time they become clinically evident. Early events involved in their genesis and the cellular context in which these events occur have thus been difficult to characterize. Here we present the first formal evidence of the shared and independent ability of basal cells and luminal progenitors, isolated from normal human mammary tissue and transduced with a single oncogene (KRAS(G12D)), to produce serially transplantable, polyclonal, invasive ductal carcinomas within 8 weeks of being introduced either subrenally or subcutaneously into immunodeficient mice. DNA barcoding of the initial cells revealed a dramatic change in the numbers and sizes of clones generated from them within 2 weeks, and the first appearance of many 'new' clones in tumours passaged into secondary recipients. Both primary and secondary tumours were phenotypically heterogeneous and primary tumours were categorized transcriptionally as 'normal-like'. This system challenges previous concepts that carcinogenesis in normal human epithelia is necessarily a slow process requiring the acquisition of multiple driver mutations. It also presents the first description of initial events that accompany the genesis and evolution of malignant human mammary cell populations, thereby contributing new understanding of the rapidity with which heterogeneity in their properties can develop.


Asunto(s)
Neoplasias de la Mama/fisiopatología , Carcinoma Ductal de Mama/fisiopatología , Transformación Celular Neoplásica , Glándulas Mamarias Humanas/fisiopatología , Animales , Neoplasias de la Mama/genética , Carcinoma Ductal de Mama/genética , Linaje de la Célula/genética , Células Cultivadas , Código de Barras del ADN Taxonómico , Femenino , Perfilación de la Expresión Génica , Xenoinjertos , Humanos , Lentivirus/genética , Glándulas Mamarias Humanas/citología , Ratones , Ratones Endogámicos , Ratones SCID , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas p21(ras) , Factores de Tiempo , Transducción Genética , Proteínas ras/genética
18.
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
19.
Nat Commun ; 6: 6351, 2015 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-25690954

RESUMEN

While significant effort has been dedicated to the characterization of epigenetic changes associated with prenatal differentiation, relatively little is known about the epigenetic changes that accompany post-natal differentiation where fully functional differentiated cell types with limited lifespans arise. Here we sought to address this gap by generating epigenomic and transcriptional profiles from primary human breast cell types isolated from disease-free human subjects. From these data we define a comprehensive human breast transcriptional network, including a set of myoepithelial- and luminal epithelial-specific intronic retention events. Intersection of epigenetic states with RNA expression from distinct breast epithelium lineages demonstrates that mCpG provides a stable record of exonic and intronic usage, whereas H3K36me3 is dynamic. We find a striking asymmetry in epigenomic reprogramming between luminal and myoepithelial cell types, with the genomes of luminal cells harbouring more than twice the number of hypomethylated enhancer elements compared with myoepithelial cells.


Asunto(s)
Mama/metabolismo , Epigénesis Genética , Regulación de la Expresión Génica , Mama/citología , Ciclo Celular , Diferenciación Celular , Separación Celular , Cromatina/química , Inmunoprecipitación de Cromatina , Islas de CpG , Epigenómica , Células Epiteliales/citología , Exones , Femenino , Citometría de Flujo , Genoma Humano , Histonas/química , Humanos , Intrones , Cariotipificación , MicroARNs/metabolismo , Análisis de Secuencia de ARN , Transcripción Genética
20.
Nat Commun ; 5: 5871, 2014 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-25532760

RESUMEN

Genomic and phenotypic analyses indicate extensive intra- as well as intertumoral heterogeneity in primary human malignant cell populations despite their clonal origin. Cellular DNA barcoding offers a powerful and unbiased alternative to track the number and size of multiple subclones within a single human tumour xenograft and their response to continued in vivo passaging. Using this approach we find clone-initiating cell frequencies that vary from ~1/10 to ~1/10,000 cells transplanted for two human breast cancer cell lines and breast cancer xenografts derived from three different patients. For the cell lines, these frequencies are negatively affected in transplants of more than 20,000 cells. Serial transplants reveal five clonal growth patterns (unchanging, expanding, diminishing, fluctuating or of delayed onset), whose predominance is highly variable both between and within original samples. This study thus demonstrates the high growth potential and diverse growth properties of xenografted human breast cancer cells.


Asunto(s)
Neoplasias de la Mama/genética , Proliferación Celular , Animales , Neoplasias de la Mama/fisiopatología , Línea Celular Tumoral , Células Clonales , Código de Barras del ADN Taxonómico , Femenino , Humanos , Cinética , Ratones , Trasplante de Neoplasias , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...