Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 54(4): 675-82, 2014 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-24768537

RESUMEN

A complex network of regulatory pathways links transcription to cell growth and proliferation. Here we show that cellular quiescence alters chromatin structure by promoting trimethylation of histone H4 at lysine 20 (H4K20me3). In contrast to pericentric or telomeric regions, recruitment of the H4K20 methyltransferase Suv4-20h2 to rRNA genes and IAP elements requires neither trimethylation of H3K9 nor interaction with HP1 proteins but depends on long noncoding RNAs (lncRNAs) that interact with Suv4-20h2. Growth factor deprivation and terminal differentiation lead to upregulation of these lncRNAs, increase in H4K20me3, and chromatin compaction. The results uncover a lncRNA-mediated mechanism that guides Suv4-20h2 to specific genomic loci to establish a more compact chromatin structure in growth-arrested cells.


Asunto(s)
Cromatina/metabolismo , Silenciador del Gen , Genes de ARNr , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Lisina/metabolismo , ARN Largo no Codificante/metabolismo , Animales , Proliferación Celular , Cromatina/genética , Regulación de la Expresión Génica , Genes de Partícula A Intracisternal , Sitios Genéticos , N-Metiltransferasa de Histona-Lisina/genética , Histonas/química , Histonas/genética , Metilación , Ratones , Células 3T3 NIH , ARN Largo no Codificante/genética , Telómero/genética
2.
Nature ; 520(7548): 549-52, 2015 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-25707806

RESUMEN

Haematopoietic stem cells (HSCs) are responsible for the lifelong production of blood cells. The accumulation of DNA damage in HSCs is a hallmark of ageing and is probably a major contributing factor in age-related tissue degeneration and malignant transformation. A number of accelerated ageing syndromes are associated with defective DNA repair and genomic instability, including the most common inherited bone marrow failure syndrome, Fanconi anaemia. However, the physiological source of DNA damage in HSCs from both normal and diseased individuals remains unclear. Here we show in mice that DNA damage is a direct consequence of inducing HSCs to exit their homeostatic quiescent state in response to conditions that model physiological stress, such as infection or chronic blood loss. Repeated activation of HSCs out of their dormant state provoked the attrition of normal HSCs and, in the case of mice with a non-functional Fanconi anaemia DNA repair pathway, led to a complete collapse of the haematopoietic system, which phenocopied the highly penetrant bone marrow failure seen in Fanconi anaemia patients. Our findings establish a novel link between physiological stress and DNA damage in normal HSCs and provide a mechanistic explanation for the universal accumulation of DNA damage in HSCs during ageing and the accelerated failure of the haematopoietic system in Fanconi anaemia patients.


Asunto(s)
Ciclo Celular , Daño del ADN , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Animales , Médula Ósea/patología , Muerte Celular , Proliferación Celular , Anemia de Fanconi/metabolismo , Ratones , Especies Reactivas de Oxígeno/metabolismo , Estrés Fisiológico
3.
Hepatology ; 69(5): 2091-2106, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30615206

RESUMEN

Intrahepatic cholangiocarcinoma (iCCA) is the second most common primary liver cancer. It is defined by cholangiocytic differentiation and has poor prognosis. Recently, epigenetic processes have been shown to play an important role in cholangiocarcinogenesis. We performed an integrative analysis on 52 iCCAs using both genetic and epigenetic data with a specific focus on DNA methylation components. We found recurrent isocitrate dehydrogenase 1 (IDH1) and IDH2 (28%) gene mutations, recurrent arm-length copy number alterations (CNAs), and focal alterations such as deletion of 3p21 or amplification of 12q15, which affect BRCA1 Associated Protein 1, polybromo 1, and mouse double minute 2 homolog. DNA methylome analysis revealed excessive hypermethylation of iCCA, affecting primarily the bivalent genomic regions marked with both active and repressive histone modifications. Integrative clustering of genetic and epigenetic data identified four iCCA subgroups with prognostic relevance further designated as IDH, high (H), medium (M), and low (L) alteration groups. The IDH group consisted of all samples with IDH1 or IDH2 mutations and showed, together with the H group, a highly disrupted genome, characterized by frequent deletions of chromosome arms 3p and 6q. Both groups showed excessive hypermethylation with distinct patterns. The M group showed intermediate characteristics regarding both genetic and epigenetic marks, whereas the L group exhibited few methylation changes and mutations and a lack of CNAs. Methylation-based latent component analysis of cell-type composition identified differences among these four groups. Prognosis of the H and M groups was significantly worse than that of the L group. Conclusion: Using an integrative genomic and epigenomic analysis approach, we identified four major iCCA subgroups with widespread genomic and epigenomic differences and prognostic implications. Furthermore, our data suggest differences in the cell-of-origin of the iCCA subtypes.


Asunto(s)
Neoplasias de los Conductos Biliares/clasificación , Neoplasias de los Conductos Biliares/genética , Colangiocarcinoma/clasificación , Colangiocarcinoma/genética , Metilación de ADN , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de los Conductos Biliares/diagnóstico , Colangiocarcinoma/diagnóstico , Femenino , Genes p53 , Humanos , Isocitrato Deshidrogenasa/genética , Masculino , Persona de Mediana Edad , Mutación , Pronóstico
4.
Semin Cancer Biol ; 51: 12-21, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29366906

RESUMEN

Analogous to life on earth, tumor cells evolve through space and time and adapt to different micro-environmental conditions. As a result, tumors are composed of millions of genetically diversified cells at the time of diagnosis. Profiling these variants contributes to understanding tumors' clonal origins and might help to better understand response to therapy. However, even genetically homogenous cell populations show remarkable diversity in their response to different environmental stimuli, suggesting that genetic heterogeneity does not explain the full spectrum of tumor plasticity. Understanding epigenetic diversity across cancer cells provides important additional information about the functional state of subclones and therefore allows better understanding of tumor evolution and resistance to current therapies.


Asunto(s)
Evolución Clonal , Epigénesis Genética , Heterogeneidad Genética , Neoplasias/genética , Animales , Humanos , Neoplasias/patología
5.
Cancer Discov ; 14(1): 142-157, 2024 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-37934007

RESUMEN

Suboptimal functional persistence limits the efficacy of adoptive T-cell therapies. CD28-based chimeric antigen receptors (CAR) impart potent effector function to T cells but with a limited lifespan. We show here that the genetic disruption of SUV39H1, which encodes a histone-3, lysine-9 methyl-transferase, enhances the early expansion, long-term persistence, and overall antitumor efficacy of human CAR T cells in leukemia and prostate cancer models. Persisting SUV39H1-edited CAR T cells demonstrate improved expansion and tumor rejection upon multiple rechallenges. Transcriptional and genome accessibility profiling of repeatedly challenged CAR T cells shows improved expression and accessibility of memory transcription factors in SUV39H1-edited CAR T cells. SUV39H1 editing also reduces expression of inhibitory receptors and limits exhaustion in CAR T cells that have undergone multiple rechallenges. Our findings thus demonstrate the potential of epigenetic programming of CAR T cells to balance their function and persistence for improved adoptive cell therapies. SIGNIFICANCE: T cells engineered with CD28-based CARs possess robust effector function and antigen sensitivity but are hampered by limited persistence, which may result in tumor relapse. We report an epigenetic strategy involving disruption of the SUV39H1-mediated histone-silencing program that promotes the functional persistence of CD28-based CAR T cells. See related article by López-Cobo et al., p. 120. This article is featured in Selected Articles from This Issue, p. 5.


Asunto(s)
Leucemia , Receptores Quiméricos de Antígenos , Masculino , Humanos , Linfocitos T , Receptores de Antígenos de Linfocitos T , Histonas/metabolismo , Antígenos CD28/genética , Antígenos CD28/metabolismo , Inmunoterapia Adoptiva , Leucemia/metabolismo , Metilación , Ensayos Antitumor por Modelo de Xenoinjerto , Metiltransferasas/genética , Metiltransferasas/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
6.
Philos Trans R Soc Lond B Biol Sci ; 375(1795): 20190345, 2020 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-32075565

RESUMEN

Next-generation sequencing approaches have fundamentally changed the types of questions that can be asked about gene function and regulation. With the goal of approaching truly genome-wide quantifications of all the interaction partners and downstream effects of particular genes, these quantitative assays have allowed for an unprecedented level of detail in exploring biological interactions. However, many challenges remain in our ability to accurately describe and quantify the interactions that take place in those hard to reach and extremely repetitive regions of our genome comprised mostly of transposable elements (TEs). Tools dedicated to TE-derived sequences have lagged behind, making the inclusion of these sequences in genome-wide analyses difficult. Recent improvements, both computational and experimental, allow for the better inclusion of TE sequences in genomic assays and a renewed appreciation for the importance of TE biology. This review will discuss the recent improvements that have been made in the computational analysis of TE-derived sequences as well as the areas where such analysis still proves difficult. This article is part of a discussion meeting issue 'Crossroads between transposons and gene regulation'.


Asunto(s)
Elementos Transponibles de ADN/genética , Estudio de Asociación del Genoma Completo , Genómica , Secuenciación de Nucleótidos de Alto Rendimiento , Evolución Molecular
7.
Leukemia ; 33(4): 945-956, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30470836

RESUMEN

DNA methyltransferase inhibitors (DNMTi) approved for older AML patients are clinically tested in combination with histone deacetylase inhibitors (HDACi). The mechanism of action of these drugs is still under debate. In colon cancer cells, 5-aza-2'-deoxycytidine (DAC) can downregulate oncogenes and metabolic genes by reversing gene body DNA methylation, thus implicating gene body methylation as a novel drug target. We asked whether DAC-induced gene body demethylation in AML cells is also associated with gene repression, and whether the latter is enhanced by HDACi.Transcriptome analyses revealed that a combined treatment with DAC and the HDACi panobinostat or valproic acid affected significantly more transcripts than the sum of the genes regulated by either treatment alone, demonstrating a quantitative synergistic effect on genome-wide expression in U937 cells. This effect was particularly striking for downregulated genes. Integrative methylome and transcriptome analyses showed that a massive downregulation of genes, including oncogenes (e.g., MYC) and epigenetic modifiers (e.g., KDM2B, SUV39H1) often overexpressed in cancer, was associated predominantly with gene body DNA demethylation and changes in acH3K9/27. These findings have implications for the mechanism of action of combined epigenetic treatments, and for a better understanding of responses in trials where this approach is clinically tested.


Asunto(s)
Metilación de ADN , Decitabina/farmacología , Sinergismo Farmacológico , Inhibidores Enzimáticos/farmacología , Regulación Neoplásica de la Expresión Génica , Inhibidores de Histona Desacetilasas/farmacología , Leucemia Mieloide Aguda/genética , Biomarcadores de Tumor/genética , Metilasas de Modificación del ADN/antagonistas & inhibidores , Desmetilación , Regulación hacia Abajo , Epigénesis Genética , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/patología , Panobinostat/farmacología , Ácido Valproico/farmacología
8.
Cell Cycle ; 17(7): 811-822, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29633898

RESUMEN

Inhibitors of DNA methyltransferases (DNMTis) or histone deacetylases (HDACis) are epigenetic drugs which are investigated since decades. Several have been approved and are applied in the treatment of hematopoietic and lymphatic malignancies, although their mode of action has not been fully understood. Two recent findings improved mechanistic insights: i) activation of human endogenous retroviral elements (HERVs) with concomitant synthesis of double-stranded RNAs (dsRNAs), and ii) massive activation of promoters from long terminal repeats (LTRs) which originated from past HERV invasions. These dsRNAs activate an antiviral response pathway followed by apoptosis. LTR promoter activation leads to synthesis of non-annotated transcripts potentially encoding novel or cryptic proteins. Here, we discuss the current knowledge of the molecular effects exerted by epigenetic drugs with a focus on DNMTis and HDACis. We highlight the role in LTR activation and provide novel data from both in vitro and in vivo epigenetic drug treatment.


Asunto(s)
Antineoplásicos/uso terapéutico , ADN (Citosina-5-)-Metiltransferasas/genética , Retrovirus Endógenos/efectos de los fármacos , Inhibidores Enzimáticos/uso terapéutico , Epigénesis Genética , Neoplasias Hematológicas/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/uso terapéutico , Histona Desacetilasas/genética , Apoptosis/efectos de los fármacos , Apoptosis/genética , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Retrovirus Endógenos/genética , Retrovirus Endógenos/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Hematológicas/enzimología , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/patología , Histona Desacetilasas/metabolismo , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/genética , Humanos , Regiones Promotoras Genéticas , ARN Bicatenario , Secuencias Repetidas Terminales , Activación Viral/efectos de los fármacos
10.
Nat Commun ; 8(1): 2126, 2017 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-29259247

RESUMEN

Juvenile myelomonocytic leukemia (JMML) is an aggressive myeloproliferative disorder of early childhood characterized by mutations activating RAS signaling. Established clinical and genetic markers fail to fully recapitulate the clinical and biological heterogeneity of this disease. Here we report DNA methylome analysis and mutation profiling of 167 JMML samples. We identify three JMML subgroups with unique molecular and clinical characteristics. The high methylation group (HM) is characterized by somatic PTPN11 mutations and poor clinical outcome. The low methylation group is enriched for somatic NRAS and CBL mutations, as well as for Noonan patients, and has a good prognosis. The intermediate methylation group (IM) shows enrichment for monosomy 7 and somatic KRAS mutations. Hypermethylation is associated with repressed chromatin, genes regulated by RAS signaling, frequent co-occurrence of RAS pathway mutations and upregulation of DNMT1 and DNMT3B, suggesting a link between activation of the DNA methylation machinery and mutational patterns in JMML.


Asunto(s)
Metilación de ADN , Leucemia Mielomonocítica Juvenil/genética , Síndrome de Noonan/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Transducción de Señal/genética , Antineoplásicos/uso terapéutico , Biopsia , Niño , Preescolar , Cromatina/genética , Cromatina/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Análisis Mutacional de ADN , Epigenómica , Femenino , Regulación Leucémica de la Expresión Génica , Trasplante de Células Madre Hematopoyéticas , Humanos , Lactante , Leucemia Mielomonocítica Juvenil/mortalidad , Leucemia Mielomonocítica Juvenil/patología , Leucemia Mielomonocítica Juvenil/terapia , Masculino , Mutación , Síndrome de Noonan/patología , Pronóstico , Estudios Prospectivos , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteínas Proto-Oncogénicas c-cbl , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Regulación hacia Arriba , ADN Metiltransferasa 3B
11.
Nat Genet ; 49(7): 1052-1060, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28604729

RESUMEN

Several mechanisms of action have been proposed for DNA methyltransferase and histone deacetylase inhibitors (DNMTi and HDACi), primarily based on candidate-gene approaches. However, less is known about their genome-wide transcriptional and epigenomic consequences. By mapping global transcription start site (TSS) and chromatin dynamics, we observed the cryptic transcription of thousands of treatment-induced non-annotated TSSs (TINATs) following DNMTi and HDACi treatment. The resulting transcripts frequently splice into protein-coding exons and encode truncated or chimeric ORFs translated into products with predicted abnormal or immunogenic functions. TINAT transcription after DNMTi treatment coincided with DNA hypomethylation and gain of classical promoter histone marks, while HDACi specifically induced a subset of TINATs in association with H2AK9ac, H3K14ac, and H3K23ac. Despite this mechanistic difference, both inhibitors convergently induced transcription from identical sites, as we found TINATs to be encoded in solitary long terminal repeats of the ERV9/LTR12 family, which are epigenetically repressed in virtually all normal cells.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , Proteínas Quinasas Asociadas a Muerte Celular/genética , Código de Histonas , Inhibidores de Histona Desacetilasas/farmacología , Secuencias Repetidas Terminales/genética , Sitio de Iniciación de la Transcripción/efectos de los fármacos , Empalme Alternativo/genética , Animales , Bencimidazoles/farmacología , Línea Celular Tumoral , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/fisiología , Metilación de ADN , Proteínas Quinasas Asociadas a Muerte Celular/antagonistas & inhibidores , Represión Epigenética , Exones/genética , Femenino , Perfilación de la Expresión Génica , Silenciador del Gen , Humanos , Ácidos Hidroxámicos/farmacología , Intrones/genética , Ratones , Ratones Desnudos , Interferencia de ARN , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Vorinostat
12.
Nat Genet ; 48(3): 253-64, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26780610

RESUMEN

Charting differences between tumors and normal tissue is a mainstay of cancer research. However, clonal tumor expansion from complex normal tissue architectures potentially obscures cancer-specific events, including divergent epigenetic patterns. Using whole-genome bisulfite sequencing of normal B cell subsets, we observed broad epigenetic programming of selective transcription factor binding sites coincident with the degree of B cell maturation. By comparing normal B cells to malignant B cells from 268 patients with chronic lymphocytic leukemia (CLL), we showed that tumors derive largely from a continuum of maturation states reflected in normal developmental stages. Epigenetic maturation in CLL was associated with an indolent gene expression pattern and increasingly favorable clinical outcomes. We further uncovered that most previously reported tumor-specific methylation events are normally present in non-malignant B cells. Instead, we identified a potential pathogenic role for transcription factor dysregulation in CLL, where excess programming by EGR and NFAT with reduced EBF and AP-1 programming imbalances the normal B cell epigenetic program.


Asunto(s)
Linfocitos B/metabolismo , Metilación de ADN/genética , Epigénesis Genética , Leucemia Linfocítica Crónica de Células B/genética , Adulto , Anciano , Anciano de 80 o más Años , Linfocitos B/inmunología , Sitios de Unión , Islas de CpG/genética , Femenino , Regulación Leucémica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Leucemia Linfocítica Crónica de Células B/inmunología , Leucemia Linfocítica Crónica de Células B/patología , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Fenotipo , Regiones Promotoras Genéticas
13.
Clin Epigenetics ; 7: 94, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26357527

RESUMEN

Scientific data has been transformed into music in order to raise awareness in the non-scientific community. While the general public is nowadays familiar with the genetic code, there is still a lack of knowledge regarding epigenetic regulation. By making use of the binary nature of the methylome, we here describe a method that transforms methylation patterns into music. The resulting musical pieces show decent complexity and allow the audible recognition between music and underlying methylation state. This approach might therefore facilitate the recognition of complex methylation patterns and increase awareness for epigenetic regulation in the general public.

14.
Cell Rep ; 8(3): 798-806, 2014 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-25066126

RESUMEN

Despite much evidence on epigenetic abnormalities in cancer, it is currently unclear to what extent epigenetic alterations can be associated with tumors' clonal genetic origins. Here, we show that the prostate intratumor heterogeneity in DNA methylation and copy-number patterns can be explained by a unified evolutionary process. By assaying multiple topographically distinct tumor sites, premalignant lesions, and lymph node metastases within five cases of prostate cancer, we demonstrate that both DNA methylation and copy-number heterogeneity consistently reflect the life history of the tumors. Furthermore, we show cases of genetic or epigenetic convergent evolution and highlight the diversity in the evolutionary origins and aberration spectrum between tumor and metastatic subclones. Importantly, DNA methylation can complement genetic data by serving as a proxy for activity at regulatory domains, as we show through identification of high epigenetic heterogeneity at androgen-receptor-bound enhancers. Epigenome variation thereby expands on the current genome-centric view on tumor heterogeneity.


Asunto(s)
Adenocarcinoma/genética , Evolución Clonal , Metilación de ADN , Heterogeneidad Genética , Neoplasias de la Próstata/genética , Adenocarcinoma/patología , Variaciones en el Número de Copia de ADN , Epigénesis Genética , Humanos , Metástasis Linfática , Masculino , Persona de Mediana Edad , Neoplasias de la Próstata/patología
15.
Cancer Discov ; 4(3): 348-61, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24356097

RESUMEN

Although clonal selection by genetic driver aberrations in cancer is well documented, the ability of epigenetic alterations to promote tumor evolution is undefined. We used 450k arrays and next-generation sequencing to evaluate intratumor heterogeneity and evolution of DNA methylation and genetic aberrations in chronic lymphocytic leukemia (CLL). CLL cases exhibit vast interpatient differences in intratumor methylation heterogeneity, with genetically clonal cases maintaining low methylation heterogeneity and up to 10% of total CpGs in a monoallelically methylated state. Increasing methylation heterogeneity correlates with advanced genetic subclonal complexity. Selection of novel DNA methylation patterns is observed only in cases that undergo genetic evolution, and independent genetic evolution is uncommon and is restricted to low-risk alterations. These results reveal that although evolution of DNA methylation occurs in high-risk, clinically progressive cases, positive selection of novel methylation patterns entails coevolution of genetic alteration(s) in CLL.


Asunto(s)
Metilación de ADN , Inestabilidad Genómica , Leucemia Linfocítica Crónica de Células B/genética , Anciano , Cromosomas Humanos , Islas de CpG , Progresión de la Enfermedad , Epigénesis Genética , Femenino , Regulación Leucémica de la Expresión Génica , Heterogeneidad Genética , Genoma Humano , Humanos , Leucemia Linfocítica Crónica de Células B/diagnóstico , Leucemia Linfocítica Crónica de Células B/patología , Masculino , Persona de Mediana Edad
16.
Cell Stem Cell ; 15(4): 507-522, 2014 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-25158935

RESUMEN

In this study, we present integrated quantitative proteome, transcriptome, and methylome analyses of hematopoietic stem cells (HSCs) and four multipotent progenitor (MPP) populations. From the characterization of more than 6,000 proteins, 27,000 transcripts, and 15,000 differentially methylated regions (DMRs), we identified coordinated changes associated with early differentiation steps. DMRs show continuous gain or loss of methylation during differentiation, and the overall change in DNA methylation correlates inversely with gene expression at key loci. Our data reveal the differential expression landscape of 493 transcription factors and 682 lncRNAs and highlight specific expression clusters operating in HSCs. We also found an unexpectedly dynamic pattern of transcript isoform regulation, suggesting a critical regulatory role during HSC differentiation, and a cell cycle/DNA repair signature associated with multipotency in MPP2 cells. This study provides a comprehensive genome-wide resource for the functional exploration of molecular, cellular, and epigenetic regulation at the top of the hematopoietic hierarchy.


Asunto(s)
Metilación de ADN/genética , Redes Reguladoras de Genes , Células Madre Hematopoyéticas/metabolismo , Proteoma/metabolismo , Transcriptoma/genética , Adulto , Diferenciación Celular/genética , Linaje de la Célula/genética , Análisis por Conglomerados , Epigénesis Genética , Perfilación de la Expresión Génica , Genoma Humano/genética , Impresión Genómica , Células Madre Hematopoyéticas/citología , Humanos , Datos de Secuencia Molecular , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN Largo no Codificante/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
17.
Cell Cycle ; 13(22): 3476-87, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25483069

RESUMEN

Epigenetic alterations during cellular differentiation are a key molecular mechanism which both instructs and reinforces the process of lineage commitment. Within the haematopoietic system, progressive changes in the DNA methylome of haematopoietic stem cells (HSCs) are essential for the effective production of mature blood cells. Inhibition or loss of function of the cellular DNA methylation machinery has been shown to lead to a severe perturbation in blood production and is also an important driver of malignant transformation. HSCs constitute a very rare cell population in the bone marrow, capable of life-long self-renewal and multi-lineage differentiation. The low abundance of HSCs has been a major technological barrier to the global analysis of the CpG methylation status within both HSCs and their immediate progeny, the multipotent progenitors (MPPs). Within this Extra View article, we review the current understanding of how the DNA methylome regulates normal and malignant hematopoiesis. We also discuss the current methodologies that are available for interrogating the DNA methylation status of HSCs and MPPs and describe a new data set that was generated using tagmentation-based whole genome bisulfite sequencing (TWGBS) in order to comprehensively map methylated cytosines using the limited amount of genomic DNA that can be harvested from rare cell populations. Extended analysis of this data set clearly demonstrates the added value of genome-wide sequencing of methylated cytosines and identifies novel important cis-acting regulatory regions that are dynamically remodeled during the first steps of haematopoietic differentiation.


Asunto(s)
Diferenciación Celular/genética , Metilación de ADN/genética , Células Madre Hematopoyéticas , Secuencias Reguladoras de Ácidos Nucleicos/genética , Epigénesis Genética , Genoma , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA