Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Genome Res ; 34(3): 484-497, 2024 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-38580401

RESUMEN

Transcriptional regulation controls cellular functions through interactions between transcription factors (TFs) and their chromosomal targets. However, understanding the fate conversion potential of multiple TFs in an inducible manner remains limited. Here, we introduce iTF-seq as a method for identifying individual TFs that can alter cell fate toward specific lineages at a single-cell level. iTF-seq enables time course monitoring of transcriptome changes, and with biotinylated individual TFs, it provides a multi-omics approach to understanding the mechanisms behind TF-mediated cell fate changes. Our iTF-seq study in mouse embryonic stem cells identified multiple TFs that trigger rapid transcriptome changes indicative of differentiation within a day of induction. Moreover, cells expressing these potent TFs often show a slower cell cycle and increased cell death. Further analysis using bioChIP-seq revealed that GCM1 and OTX2 act as pioneer factors and activators by increasing gene accessibility and activating the expression of lineage specification genes during cell fate conversion. iTF-seq has utility in both mapping cell fate conversion and understanding cell fate conversion mechanisms.


Asunto(s)
Diferenciación Celular , Factores de Transcripción , Animales , Ratones , Diferenciación Celular/genética , Linaje de la Célula/genética , Perfilación de la Expresión Génica/métodos , Células Madre Embrionarias de Ratones/metabolismo , Células Madre Embrionarias de Ratones/citología , Multiómica , ARN Citoplasmático Pequeño/genética , ARN Citoplasmático Pequeño/metabolismo , RNA-Seq/métodos , Análisis de Secuencia de ARN/métodos , Análisis de Expresión Génica de una Sola Célula , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Transcriptoma
2.
Blood ; 142(7): 658-674, 2023 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-37267513

RESUMEN

Myeloid cell heterogeneity is known, but whether it is cell-intrinsic or environmentally-directed remains unclear. Here, an inducible/reversible system pausing myeloid differentiation allowed the definition of clone-specific functions that clustered monocytes into subsets with distinctive molecular features. These subsets were orthogonal to the classical/nonclassical categorization and had inherent, restricted characteristics that did not shift under homeostasis, after irradiation, or with infectious stress. Rather, their functional fate was constrained by chromatin accessibility established at or before the granulocyte-monocyte or monocyte-dendritic progenitor level. Subsets of primary monocytes had differential ability to control distinct infectious agents in vivo. Therefore, monocytes are a heterogeneous population of functionally restricted subtypes defined by the epigenome of their progenitors that are differentially selected by physiologic challenges with limited plasticity to transition from one subset to another.


Asunto(s)
Granulocitos , Monocitos , Células Progenitoras Mieloides , Epigenoma , Epigénesis Genética , Diferenciación Celular/genética
3.
Nucleic Acids Res ; 51(8): 3806-3819, 2023 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-36951126

RESUMEN

The placenta is an essential organ that supports the growth and development of the fetus during pregnancy. However, cell type-specific enhancers and transcription factors (TFs), and the mechanisms underlying the maintenance and differentiation of trophoblast stem cell (TSC) populations in the human placenta remain elusive. Here, using human TSCs as a model system, we identify 31,362 enhancers that are enriched with the motifs of previously reported TSC-pivotal TFs, including TEAD4, GATA2/3 and TFAP2C. Subsequently, we identify 580 super-enhancers (SEs) and 549 SE-associated genes. These genes are robustly expressed in the human placenta and include numerous TFs, implying that SE-associated TFs (SE-TFs) may play crucial roles in placental development. Additionally, we identify the global binding sites of five TSC-pivotal SE-TFs (FOS, GATA2, MAFK, TEAD4 and TFAP2C), revealing that they preferentially co-occupy enhancers, regulate each other and form a trophoblast-active gene regulatory network. Loss-of-function studies unveil that the five TFs promote self-renewal of TSCs by activating proliferation-associated genes while repressing developmental genes. We further reveal that the five TFs exert conserved and unique functions on placental development between humans and mice. Our study provides important insights into the roles of human TSC-pivotal TFs in regulating placenta-specific gene expression programs.


Asunto(s)
Factores de Transcripción , Trofoblastos , Humanos , Femenino , Embarazo , Ratones , Animales , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Placenta/metabolismo , Células Madre/metabolismo , Diferenciación Celular/genética , Expresión Génica , Factores de Transcripción de Dominio TEA
4.
Proc Natl Acad Sci U S A ; 119(20): e2115709119, 2022 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-35537047

RESUMEN

The placenta is a transient but important multifunctional organ crucial for healthy pregnancy for both mother and fetus. Nevertheless, limited access to human placenta samples and the paucity of a proper in vitro model system have hampered our understanding of the mechanisms underlying early human placental development and placenta-associated pregnancy complications. To overcome these constraints, we established a simple procedure with a short-term treatment of bone morphogenetic protein 4 (BMP4) in trophoblast stem cell culture medium (TSCM) to convert human primed pluripotent stem cells (PSCs) to trophoblast stem-like cells (TSLCs). These TSLCs show not only morphology and global gene expression profiles comparable to bona fide human trophoblast stem cells (TSCs) but also long-term self-renewal capacity with bipotency that allows the cells to differentiate into functional extravillous trophoblasts (EVT) and syncytiotrophoblasts (ST). These indicate that TSLCs are equivalent to genuine human TSCs. Our data suggest a straightforward approach to make human TSCs directly from preexisting primed PSCs and provide a valuable opportunity to study human placenta development and pathology from patients with placenta-related diseases.


Asunto(s)
Placentación , Células Madre Pluripotentes , Trofoblastos , Biomarcadores , Proteína Morfogenética Ósea 4 , Diferenciación Celular , Femenino , Humanos , Modelos Biológicos , Placenta , Embarazo , Trofoblastos/metabolismo
5.
Biol Reprod ; 110(3): 431-449, 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38134961

RESUMEN

Long non-coding RNAs are cellular transcripts that have ˃200 nucleotides in length and do not code for proteins. Due to their low expression levels, long non-coding RNAs were previously considered as mere transcriptional noise. However, current evidence indicates that they regulate a myriad of biological processes such as cell proliferation, invasion, and apoptosis. Hence, their expression patterns are crucial indicators of the physiological or pathological states of cells, tissues, and organs. The utilization of long non-coding RNAs as biomarkers and therapeutic targets for the clinical management of several diseases have been suggested. Gradually, long non-coding RNAs are gaining a substantial attention in the field of feto-maternal medicine. After embryo implantation, the interactions between the trophoblast cells from the embryo and the uterus of the mother facilitate placenta development and pregnancy progression. These processes are tightly regulated, and their impairments result in pregnancy pathologies such as miscarriage and preeclampsia. Accumulating evidence implicates long non-coding RNAs in these processes. Herein, we have summarized the roles of several long non-coding RNAs in human placenta development, have proposed some mechanisms by which they participate in physiological and pathological placentation, have revealed some knowledge deficits, and have recommended ideal experimental approaches that will facilitate the clarification of the mechanistic actions of each long non-coding RNA at the feto-maternal interface during healthy and pathological pregnancies.


Asunto(s)
Placentación , ARN Largo no Codificante , Embarazo , Femenino , Humanos , Placentación/genética , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Placenta/metabolismo , Trofoblastos/metabolismo , Implantación del Embrión
6.
Mol Biol Rep ; 51(1): 301, 2024 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-38353828

RESUMEN

Long non-coding RNAs (LncRNAs) are being unveiled as crucial regulators of several biological processes and pathways. Among the lncRNAs is metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), which is also known as nuclear enriched abundant transcript 2 (NEAT2). MALAT1 is highly conserved in mammals, and controls cellular processes such as proliferation, migration, invasion, angiogenesis, and apoptosis in both physiological and pathological conditions. Roles of MALAT1 in the female reproductive system are gradually getting explored. Within the ovarian micro-environment, the physiological expression of MALAT1 potentially modulates folliculogenesis while its upregulation promotes the metastasis of epithelial ovarian cancers. Interestingly, women with polycystic ovary syndrome have been shown to exhibit aberrant ovarian expression of MALAT1 and this is believed to contribute to the development of the disease. At the feto-maternal interface, MALAT1 potentially promotes trophoblast development. While its placental downregulation is linked to the pathogenesis of preeclampsia, its placental upregulation is associated with placenta increta and placenta percreta. Hence, abnormal expression of MALAT1 is a candidate molecular biomarker and therapeutic target for the treatment of these obstetric and gynecologic anomalies. To enhance a quick uncovering and detailed characterization of the mechanistic actions of MALAT1 in the female reproductive system, we have highlighted some knowledge deficits and have recommended ideal experimental models to be employed in prospective investigations.


Asunto(s)
Neoplasias Ováricas , ARN Largo no Codificante , Embarazo , Animales , Femenino , Humanos , ARN Largo no Codificante/genética , Estudios Prospectivos , Placenta , Mamíferos , Microambiente Tumoral
7.
Cell Biochem Funct ; 42(1): e3907, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38269505

RESUMEN

As the first long noncoding RNA to be discovered, H19 has gained substantial attention as a key regulator of several biological processes and its roles in female reproductive biology are gradually getting revealed. Herein, we have summarized the current evidence regarding H19 expression pattern and involvement in the developmental and pathological processes associated with the ovary and the placenta. The findings indicate that within the ovaries, H19 is expressed in the antral and cystic atretic follicles as well as in the corpora lutea but absent in the primordial, primary, and secondary follicles. Its normal expression promotes the maturation of antral follicles and prevents their premature selection for the ovulatory journey while its aberrant induction promotes polycystic ovary syndrome development and ovarian cancer metastasis. In the placenta, H19 is highly expressed in the cytotrophoblasts and extravillous trophoblasts but weakly expressed in the syncytiotrophoblast layer and potentially controls trophoblast cell fate decisions during placenta development. Abnormal expression of H19 is observed in the placental villi of pregnancies affected by pre-eclampsia and fetal growth restriction. Therefore, dysregulated H19 is a candidate biomarker and therapeutic target for the mitigation of ovarian and placenta-associated diseases.


Asunto(s)
Ovario , ARN Largo no Codificante , Embarazo , Humanos , Femenino , ARN Largo no Codificante/genética , Placenta , Placentación , Biología
8.
Glia ; 71(12): 2866-2883, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37584502

RESUMEN

The zebrafish retina possesses tremendous regenerative potential. Müller glia underlie retinal regeneration through their ability to reprogram and generate multipotent neuronal progenitors that re-differentiate into lost neurons. Many factors required for Müller glia reprogramming and proliferation have been identified; however, we know little about the epigenetic and transcriptional regulation of these genes during regeneration. Here, we determined whether transcriptional regulation by members of the Bromodomain (Brd) family is required for Müller glia-dependent retinal regeneration. Our data demonstrate that three brd genes were expressed in Müller glia upon injury. brd2a and brd2b were expressed in all Müller glia and brd4 was expressed only in reprogramming Müller glia. Utilizing (+)-JQ1, a pharmacological inhibitor of Brd function, we demonstrate that transcriptional regulation by Brds plays a critical role in Müller glia reprogramming and regeneration. (+)-JQ1 treatment prevented cell cycle re-entry of Müller glia and the generation of neurogenic progenitors. Modulating the (+)-JQ1 exposure window, we identified the first 48 h post-injury as the time-period during which Müller glia reprogramming occurs. (+)-JQ1 treatments after 48 h post-injury had no effect on the re-differentiation of UV cones, indicating that Brd function is required only for Müller glia reprogramming and not subsequent specification/differentiation events. Brd inhibition also prevented the expression of reprogramming genes like ascl1a and lepb in Müller glia, but not effector genes like mmp9, nor did it affect microglial recruitment after injury. These results demonstrate that transcriptional regulation by Brds plays a critical role during Müller glia-dependent retinal regeneration in zebrafish.

9.
Genes Dev ; 28(20): 2219-32, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25319825

RESUMEN

Despite their origin from the inner cell mass, embryonic stem (ES) cells undergo differentiation to the trophectoderm (TE) lineage by repression of the ES cell master regulator Oct4 or activation of the TE master regulator Caudal-type homeobox 2 (Cdx2). In contrast to the in-depth studies of ES cell self-renewal and pluripotency, few TE-specific regulators have been identified, thereby limiting our understanding of mechanisms underlying the first cell fate decision. Here we show that up-regulation and nuclear entry of AT-rich interactive domain 3a (Arid3a) drives TE-like transcriptional programs in ES cells, maintains trophoblast stem (TS) cell self-renewal, and promotes further trophoblastic differentiation both upstream and independent of Cdx2. Accordingly, Arid3a(-/-) mouse post-implantation placental development is severely impaired, resulting in early embryonic death. We provide evidence that Arid3a directly activates TE-specific and trophoblast lineage-specific genes while directly repressing pluripotency genes via differential regulation of epigenetic acetylation or deacetylation. Our results identify Arid3a as a critical regulator of TE and placental development through execution of the commitment and differentiation phases of the first cell fate decision.


Asunto(s)
Diferenciación Celular/genética , Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica , Factores de Transcripción/metabolismo , Transporte Activo de Núcleo Celular , Animales , Linaje de la Célula/genética , Proteínas de Unión al ADN/genética , Femenino , Células HEK293 , Humanos , Ratones , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Placentación , Embarazo , Factores de Transcripción/genética
10.
Nucleic Acids Res ; 46(9): 4382-4391, 2018 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-29529258

RESUMEN

CpG islands (CGIs) have long been implicated in the regulation of vertebrate gene expression. However, the involvement of CGIs in chromosomal architectures and associated gene expression regulations has not yet been thoroughly explored. By combining large-scale integrative data analyses and experimental validations, we show that CGIs clearly reconcile two competing models explaining nuclear gene localizations. We first identify CGI-containing (CGI+) and CGI-less (CGI-) genes are non-randomly clustered within the genome, which reflects CGI-dependent spatial gene segregation in the nucleus and corresponding gene regulatory modes. Regardless of their transcriptional activities, CGI+ genes are mainly located at the nuclear center and encounter frequent long-range chromosomal interactions. Meanwhile, nuclear peripheral CGI- genes forming heterochromatin are activated and internalized into the nuclear center by local enhancer-promoter interactions. Our findings demonstrate the crucial implications of CGIs on chromosomal architectures and gene positioning, linking the critical importance of CGIs in determining distinct mechanisms of global gene regulation in three-dimensional space in the nucleus.


Asunto(s)
Cromosomas de los Mamíferos/química , Islas de CpG , Regulación de la Expresión Génica , Animales , Línea Celular , Núcleo Celular/genética , Cromatina/química , Ratones , Células 3T3 NIH , Transcripción Genética
11.
Nucleic Acids Res ; 45(17): 10103-10114, 2017 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-28973471

RESUMEN

Direct reprogramming can be achieved by forced expression of master transcription factors. Yet how such factors mediate repression of initial cell-type-specific genes while activating target cell-type-specific genes is unclear. Through embryonic stem (ES) to trophoblast stem (TS)-like cell reprogramming by introducing individual TS cell-specific 'CAG' factors (Cdx2, Arid3a and Gata3), we interrogate their chromosomal target occupancies, modulation of global transcription and chromatin accessibility at the initial stage of reprogramming. From the studies, we uncover a sequential, two-step mechanism of cellular reprogramming in which repression of pre-existing ES cell-associated gene expression program is followed by activation of TS cell-specific genes by CAG factors. Therefore, we reveal that CAG factors function as both decommission and pioneer factors during ES to TS-like cell fate conversion.


Asunto(s)
Factor de Transcripción CDX2/fisiología , Técnicas de Reprogramación Celular , Proteínas de Unión al ADN/fisiología , Células Madre Embrionarias/citología , Factor de Transcripción GATA3/fisiología , Regulación del Desarrollo de la Expresión Génica , Factores de Transcripción/fisiología , Trofoblastos/citología , Animales , Factor de Transcripción CDX2/genética , Células Cultivadas , Cromatina/genética , Cromatina/metabolismo , Medios de Cultivo Condicionados , Proteínas de Unión al ADN/genética , Elementos de Facilitación Genéticos , Fibroblastos , Factor de Transcripción GATA3/genética , Ontología de Genes , Código de Histonas , Ratones , Factores de Transcripción/genética , Transcripción Genética
12.
Nucleic Acids Res ; 45(12): 7151-7166, 2017 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-28453857

RESUMEN

Histone H2B lysine 120 mono-ubiquitination (H2Bub1) catalyzed by Rnf20 has been implicated in normal differentiation of embryonic stem (ES) and adult stem cells. However, it remains unknown how Rnf20 is recruited to its specific target chromosomal loci for the establishment of H2Bub1. Here, we reveal that Fbxl19, a CxxC domain-containing protein, promotes H2Bub1 at the promoters of CpG island-containing genes by interacting with Rnf20. We show that up-regulation of Fbxl19 increases the level of global H2Bub1 in mouse ES cells, while down-regulation of Fbxl19 reduces the level of H2Bub1. Our genome-wide target mapping unveils the preferential occupancy of Fbxl19 on CpG island-containing promoters, and we further discover that chromosomal binding of Fbxl19 is required for H2Bub1 of its targets. Moreover, we reveal that Fbxl19 is critical for proper differentiation of ES cells in collaboration with Rnf20. Altogether, our results demonstrate that Fbxl19 recruitment to CpG islands is required for Rnf20-mediated H2B mono-ubiquitination.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas F-Box/metabolismo , Histonas/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Procesamiento Proteico-Postraduccional , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Islas de CpG , Proteínas de Unión al ADN/genética , Proteínas F-Box/genética , Células HEK293 , Histonas/genética , Humanos , Lisina/metabolismo , Ratones , Células Madre Embrionarias de Ratones/citología , Regiones Promotoras Genéticas , Unión Proteica , Transducción de Señal , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
13.
Proc Natl Acad Sci U S A ; 113(5): E577-86, 2016 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-26787899

RESUMEN

High expression of the forkhead box P1 (FOXP1) transcription factor distinguishes the aggressive activated B cell (ABC) diffuse large B-cell lymphoma (DLBCL) subtype from the better prognosis germinal center B-cell (GCB)-DLBCL subtype and is highly correlated with poor outcomes. A genetic or functional role for FOXP1 in lymphomagenesis, however, remains unknown. Here, we report that sustained FOXP1 expression is vital for ABC-DLBCL cell-line survival. Genome-wide analyses revealed direct and indirect FOXP1 transcriptional enforcement of ABC-DLBCL hallmarks, including the classical NF-κB and MYD88 (myeloid differentiation primary response gene 88) pathways. FOXP1 promoted gene expression underlying transition of the GCB cell to the plasmablast--the transient B-cell stage targeted in ABC-DLBCL transformation--by antagonizing pathways distinctive of GCB-DLBCL, including that of the GCB "master regulator," BCL6 (B-cell lymphoma 6). Cell-line derived FOXP1 target genes that were highly correlated with FOXP1 expression in primary DLBCL accurately segregated the corresponding clinical subtypes of a large cohort of primary DLBCL isolates and identified conserved pathways associated with ABC-DLBCL pathology.


Asunto(s)
Linfocitos B/inmunología , Factores de Transcripción Forkhead/fisiología , Linfoma de Células B Grandes Difuso/inmunología , Proteínas Represoras/fisiología , Diferenciación Celular , Línea Celular Tumoral , Humanos , Activación de Linfocitos , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/patología , Transcripción Genética
14.
EMBO Rep ; 17(4): 519-29, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26917425

RESUMEN

Yap1 is a transcriptional co-activator of the Hippo pathway. The importance of Yap1 in early cell fate decision during embryogenesis has been well established, though its role in embryonic stem (ES) cells remains elusive. Here, we report that Yap1 plays crucial roles in normal differentiation rather than self-renewal of ES cells. Yap1-depleted ES cells maintain undifferentiated state with a typical colony morphology as well as robust alkaline phosphatase activity. These cells also retain comparable levels of the core pluripotent factors, such as Pou5f1 and Sox2, to the levels in wild-type ES cells without significant alteration of lineage-specific marker genes. Conversely, overexpression of Yap1 in ES cells promotes nuclear translocation of Yap1, resulting in disruption of self-renewal and triggering differentiation by up-regulating lineage-specific genes. Moreover, Yap1-deficient ES cells show impaired induction of lineage markers during differentiation. Collectively, our data demonstrate that Yap1 is a required factor for proper differentiation of mouse ES cells, while remaining dispensable for self-renewal.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Diferenciación Celular , Células Madre Embrionarias/fisiología , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Fosfatasa Alcalina/metabolismo , Animales , Proteínas de Ciclo Celular , Línea Celular , Proliferación Celular , Vía de Señalización Hippo , Ratones , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Fosfoproteínas/deficiencia , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Regulación hacia Arriba , Proteínas Señalizadoras YAP
15.
Nature ; 489(7414): 75-82, 2012 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-22955617

RESUMEN

DNase I hypersensitive sites (DHSs) are markers of regulatory DNA and have underpinned the discovery of all classes of cis-regulatory elements including enhancers, promoters, insulators, silencers and locus control regions. Here we present the first extensive map of human DHSs identified through genome-wide profiling in 125 diverse cell and tissue types. We identify ∼2.9 million DHSs that encompass virtually all known experimentally validated cis-regulatory sequences and expose a vast trove of novel elements, most with highly cell-selective regulation. Annotating these elements using ENCODE data reveals novel relationships between chromatin accessibility, transcription, DNA methylation and regulatory factor occupancy patterns. We connect ∼580,000 distal DHSs with their target promoters, revealing systematic pairing of different classes of distal DHSs and specific promoter types. Patterning of chromatin accessibility at many regulatory regions is organized with dozens to hundreds of co-activated elements, and the transcellular DNase I sensitivity pattern at a given region can predict cell-type-specific functional behaviours. The DHS landscape shows signatures of recent functional evolutionary constraint. However, the DHS compartment in pluripotent and immortalized cells exhibits higher mutation rates than that in highly differentiated cells, exposing an unexpected link between chromatin accessibility, proliferative potential and patterns of human variation.


Asunto(s)
Cromatina/genética , Cromatina/metabolismo , ADN/genética , Enciclopedias como Asunto , Genoma Humano/genética , Anotación de Secuencia Molecular , Secuencias Reguladoras de Ácidos Nucleicos/genética , Huella de ADN , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Desoxirribonucleasa I/metabolismo , Evolución Molecular , Genómica , Humanos , Tasa de Mutación , Regiones Promotoras Genéticas/genética , Factores de Transcripción/metabolismo , Sitio de Iniciación de la Transcripción , Transcripción Genética
17.
Proc Natl Acad Sci U S A ; 112(26): E3355-64, 2015 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-26080425

RESUMEN

TRAIL (TNF-related apoptosis-inducing ligand) is a promising anticancer agent that can be potentially used as an alternative or complementary therapy because of its specific antitumor activity. However, TRAIL can also stimulate the proliferation of cancer cells through the activation of NF-κB, but the exact mechanism is still poorly understood. In this study, we show that chronic exposure to subtoxic concentrations of TRAIL results in acquired resistance. This resistance is associated with the increase in miR-21, miR-30c, and miR-100 expression, which target tumor-suppressor genes fundamental in the response to TRAIL. Importantly, down-regulation of caspase-8 by miR-21 blocks receptor interacting protein-1 cleavage and induces the activation of NF-κB, which regulates these miRNAs. Thus, TRAIL activates a positive feedback loop that sustains the acquired resistance and causes an aggressive phenotype. Finally, we prove that combinatory treatment of NF-κB inhibitors and TRAIL is able to revert resistance and reduce tumor growth, with important consequences for the clinical practice.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Neoplasias Pulmonares/patología , MicroARNs/fisiología , FN-kappa B/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , MicroARNs/metabolismo , Transducción de Señal , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Transcripción Genética
19.
PLoS Genet ; 10(11): e1004798, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25411781

RESUMEN

Associating genetic variation with quantitative measures of gene regulation offers a way to bridge the gap between genotype and complex phenotypes. In order to identify quantitative trait loci (QTLs) that influence the binding of a transcription factor in humans, we measured binding of the multifunctional transcription and chromatin factor CTCF in 51 HapMap cell lines. We identified thousands of QTLs in which genotype differences were associated with differences in CTCF binding strength, hundreds of them confirmed by directly observable allele-specific binding bias. The majority of QTLs were either within 1 kb of the CTCF binding motif, or in linkage disequilibrium with a variant within 1 kb of the motif. On the X chromosome we observed three classes of binding sites: a minority class bound only to the active copy of the X chromosome, the majority class bound to both the active and inactive X, and a small set of female-specific CTCF sites associated with two non-coding RNA genes. In sum, our data reveal extensive genetic effects on CTCF binding, both direct and indirect, and identify a diversity of patterns of CTCF binding on the X chromosome.


Asunto(s)
Cromosomas Humanos X/genética , Sitios de Carácter Cuantitativo , Proteínas Represoras/genética , Alelos , Factor de Unión a CCCTC , Femenino , Humanos , Desequilibrio de Ligamiento , Unión Proteica , ARN no Traducido/genética , ARN no Traducido/metabolismo , Proteínas Represoras/metabolismo
20.
Proc Natl Acad Sci U S A ; 111(11): E998-1006, 2014 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-24591644

RESUMEN

The plasmacytoid dendritic cell (pDC) is vital to the coordinated action of innate and adaptive immunity. pDC development has not been unequivocally traced, nor has its transcriptional regulatory network been fully clarified. Here we confirm an essential requirement for the BCL11A transcription factor in fetal pDC development, and demonstrate this lineage-specific requirement in the adult organism. Furthermore, we identify BCL11A gene targets and provide a molecular mechanism for its action in pDC commitment. Embryonic germ-line deletion of Bcl11a revealed an absolute cellular, molecular, and functional absence of pDCs in fetal mice. In adults, deletion of Bcl11a in hematopoietic stem cells resulted in perturbed yet continued generation of progenitors, loss of downstream pDC and B-cell lineages, and persisting myeloid, conventional dendritic, and T-cell lineages. Challenge with virus resulted in a marked reduction of antiviral response in conditionally deleted adults. Genome-wide analyses of BCL11A DNA binding and expression revealed that BCL11A regulates transcription of E2-2 and other pDC differentiation modulators, including ID2 and MTG16. Our results identify BCL11A as an essential, lineage-specific factor that regulates pDC development, supporting a model wherein differentiation into pDCs represents a primed "default" pathway for common dendritic cell progenitors.


Asunto(s)
Proteínas Portadoras/inmunología , Diferenciación Celular/inmunología , Células Dendríticas/inmunología , Regulación de la Expresión Génica/inmunología , Redes Reguladoras de Genes/inmunología , Proteínas Nucleares/inmunología , Animales , Linfocitos B/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN , Células Dendríticas/citología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Perfilación de la Expresión Génica , Ratones , Ratones Noqueados , Ratones Mutantes , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Represoras , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA