Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
EMBO Mol Med ; 12(7): e11099, 2020 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-32558295

RESUMO

A subset of lung adenocarcinomas is driven by the EML4-ALK translocation. Even though ALK inhibitors in the clinic lead to excellent initial responses, acquired resistance to these inhibitors due to on-target mutations or parallel pathway alterations is a major clinical challenge. Exploring these mechanisms of resistance, we found that EML4-ALK cells parental or resistant to crizotinib, ceritinib or alectinib are remarkably sensitive to inhibition of CDK7/12 with THZ1 and CDK9 with alvocidib or dinaciclib. These compounds robustly induce apoptosis through transcriptional inhibition and downregulation of anti-apoptotic genes. Importantly, alvocidib reduced tumour progression in xenograft mouse models. In summary, our study takes advantage of the transcriptional addiction hypothesis to propose a new treatment strategy for a subset of patients with acquired resistance to first-, second- and third-generation ALK inhibitors.


Assuntos
Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Proteínas de Fusão Oncogênica/genética , Inibidores de Proteínas Quinases/farmacologia , Transcrição Gênica/efeitos dos fármacos , Animais , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Feminino , Humanos , Camundongos , Proteínas de Fusão Oncogênica/antagonistas & inibidores , Inibidores de Proteínas Quinases/uso terapêutico
2.
Nat Commun ; 11(1): 822, 2020 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-32054838

RESUMO

High-grade serous ovarian carcinoma is characterised by TP53 mutation and extensive chromosome instability (CIN). Because our understanding of CIN mechanisms is based largely on analysing established cell lines, we developed a workflow for generating ex vivo cultures from patient biopsies to provide models that support interrogation of CIN mechanisms in cells not extensively cultured in vitro. Here, we describe a "living biobank" of ovarian cancer models with extensive replicative capacity, derived from both ascites and solid biopsies. Fifteen models are characterised by p53 profiling, exome sequencing and transcriptomics, and karyotyped using single-cell whole-genome sequencing. Time-lapse microscopy reveals catastrophic and highly heterogeneous mitoses, suggesting that analysis of established cell lines probably underestimates mitotic dysfunction in advanced human cancers. Drug profiling reveals cisplatin sensitivities consistent with patient responses, demonstrating that this workflow has potential to generate personalized avatars with advantages over current pre-clinical models and the potential to guide clinical decision making.


Assuntos
Bancos de Espécimes Biológicos , Mitose/genética , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Instabilidade Cromossômica , Resistencia a Medicamentos Antineoplásicos , Feminino , Expressão Gênica , Perfilação da Expressão Gênica , Técnicas Histológicas/métodos , Humanos , Imageamento Tridimensional , Hibridização in Situ Fluorescente , Técnicas In Vitro , Cariotipagem , Modelos Biológicos , Mutação , Neoplasias Ovarianas/tratamento farmacológico , Paclitaxel/farmacologia , Análise de Célula Única , Imagem com Lapso de Tempo , Proteína Supressora de Tumor p53/genética , Sequenciamento do Exoma
3.
Elife ; 72018 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-29911973

RESUMO

TALE factors are broadly expressed embryonically and known to function in complexes with transcription factors (TFs) like Hox proteins at gastrula/segmentation stages, but it is unclear if such generally expressed factors act by the same mechanism throughout embryogenesis. We identify a TALE-dependent gene regulatory network (GRN) required for anterior development and detect TALE occupancy associated with this GRN throughout embryogenesis. At blastula stages, we uncover a novel functional mode for TALE factors, where they occupy genomic DECA motifs with nearby NF-Y sites. We demonstrate that TALE and NF-Y form complexes and regulate chromatin state at genes of this GRN. At segmentation stages, GRN-associated TALE occupancy expands to include HEXA motifs near PBX:HOX sites. Hence, TALE factors control a key GRN, but utilize distinct DNA motifs and protein partners at different stages - a strategy that may also explain their oncogenic potential and may be employed by other broadly expressed TFs.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Genes Essenciais/genética , Proteínas de Homeodomínio/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/genética , Animais , Blástula/embriologia , Blástula/metabolismo , Fator de Ligação a CCAAT/genética , Fator de Ligação a CCAAT/metabolismo , Técnicas de Silenciamento de Genes , Redes Reguladoras de Genes , Proteínas de Homeodomínio/metabolismo , Ligação Proteica , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
4.
Sci Rep ; 7(1): 6725, 2017 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-28751734

RESUMO

Nephrotic syndrome (NS) occurs when the glomerular filtration barrier becomes excessively permeable leading to massive proteinuria. In childhood NS, immune system dysregulation has been implicated and increasing evidence points to the central role of podocytes in the pathogenesis. Children with NS are typically treated with an empiric course of glucocorticoid (Gc) therapy; a class of steroids that are activating ligands for the glucocorticoid receptor (GR) transcription factor. Although Gc-therapy has been the cornerstone of NS management for decades, the mechanism of action, and target cell, remain poorly understood. We tested the hypothesis that Gc acts directly on the podocyte to produce clinically useful effects without involvement of the immune system. In human podocytes, we demonstrated that the basic GR-signalling mechanism is intact and that Gc induced an increase in podocyte barrier function. Defining the GR-cistrome identified Gc regulation of motility genes. These findings were functionally validated with live-cell imaging. We demonstrated that treatment with Gc reduced the activity of the pro-migratory small GTPase regulator Rac1. Furthermore, Rac1 inhibition had a direct, protective effect on podocyte barrier function. Our studies reveal a new mechanism for Gc action directly on the podocyte, with translational relevance to designing new selective synthetic Gc molecules.


Assuntos
Glucocorticoides/farmacologia , Podócitos/efeitos dos fármacos , Prednisolona/farmacologia , Substâncias Protetoras/farmacologia , Puromicina Aminonucleosídeo/antagonistas & inibidores , Receptores de Glucocorticoides/genética , Proteínas rac1 de Ligação ao GTP/genética , Antimetabólitos Antineoplásicos/toxicidade , Transporte Biológico/efeitos dos fármacos , Linhagem Celular Transformada , Membrana Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Impedância Elétrica , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Análise em Microsséries , Podócitos/citologia , Podócitos/metabolismo , Puromicina Aminonucleosídeo/toxicidade , Receptores de Glucocorticoides/antagonistas & inibidores , Receptores de Glucocorticoides/metabolismo , Transdução de Sinais , Transcriptoma , Proteínas rac1 de Ligação ao GTP/antagonistas & inibidores , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo
5.
PLoS Genet ; 13(6): e1006828, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28604778

RESUMO

Cleft palate is a common congenital disorder that affects up to 1 in 2500 live births and results in considerable morbidity to affected individuals and their families. The aetiology of cleft palate is complex with both genetic and environmental factors implicated. Mutations in the transcription factor p63 are one of the major individual causes of cleft palate; however, the gene regulatory networks in which p63 functions remain only partially characterized. Our findings demonstrate that p63 functions as an essential regulatory molecule in the spatio-temporal control of palatal epithelial cell fate to ensure appropriate fusion of the palatal shelves. Initially, p63 induces periderm formation and controls its subsequent maintenance to prevent premature adhesion between adhesion-competent, intra-oral epithelia. Subsequently, TGFß3-induced down-regulation of p63 in the medial edge epithelia of the palatal shelves is a pre-requisite for palatal fusion by facilitating periderm migration from, and reducing the proliferative potential of, the midline epithelial seam thereby preventing cleft palate.


Assuntos
Fissura Palatina/genética , Redes Reguladoras de Genes/genética , Fosfoproteínas/genética , Transativadores/genética , Fator de Crescimento Transformador beta3/genética , Animais , Movimento Celular/genética , Proliferação de Células/genética , Fissura Palatina/fisiopatologia , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , Mutação , Fosfoproteínas/biossíntese , Transdução de Sinais/genética , Transativadores/biossíntese
6.
Dev Cell ; 32(3): 265-77, 2015 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-25640223

RESUMO

Hox transcription factors (TFs) are essential for vertebrate development, but how these evolutionary conserved proteins function in vivo remains unclear. Because Hox proteins have notoriously low binding specificity, they are believed to bind with cofactors, mainly homeodomain TFs Pbx and Meis, to select their specific targets. We mapped binding of Meis, Pbx, and Hoxa2 in the branchial arches, a series of segments in the developing vertebrate head. Meis occupancy is largely similar in Hox-positive and -negative arches. Hoxa2, which specifies second arch (IIBA) identity, recognizes a subset of Meis prebound sites that contain Hox motifs. Importantly, at these sites Meis binding is strongly increased. This enhanced Meis binding coincides with active enhancers, which are linked to genes highly expressed in the IIBA and regulated by Hoxa2. These findings show that Hoxa2 operates as a tissue-specific cofactor, enhancing Meis binding to specific sites that provide the IIBA with its anatomical identity.


Assuntos
Região Branquial/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Homeodomínio/metabolismo , Animais , Linhagem Celular , Camundongos , Proteína Meis1 , Proteínas de Neoplasias/metabolismo , Fatores de Transcrição/metabolismo
7.
Nucleic Acids Res ; 37(22): 7368-80, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19789270

RESUMO

ETS-domain transcription factors play important roles in controlling gene expression in a variety of different contexts; however, these proteins bind to very similar sites and it is unclear how in vivo specificity is achieved. In silico analysis is unlikely to reveal specific targets for individual family members and direct experimental approaches are therefore required. Here, we take advantage of an inducible dominant-negative expression system to identify a group of novel target genes for the ETS-domain transcription factor Elk-1. Elk-1 is thought to mainly function through cooperation with a second transcription factor SRF, but the targets we identify are largely SRF-independent. Furthermore, we demonstrate that there is a high degree of overlapping, cell type-specific, target gene binding by Elk-1 and other ETS-domain transcription factors. Our results are therefore consistent with the notion that there is a high degree of functional redundancy in target gene regulation by ETS-domain transcription factors in addition to the specific target gene regulation that can be dictated through heterotypic interactions exemplified by the Elk-1-SRF complex.


Assuntos
Regiões Promotoras Genéticas , Proteínas Elk-1 do Domínio ets/metabolismo , Sítios de Ligação , Linhagem Celular , Regulação da Expressão Gênica , Humanos , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-ets/química , Proteínas Proto-Oncogênicas c-ets/metabolismo , Fator de Resposta Sérica/metabolismo , Proteínas Elk-1 do Domínio ets/química
8.
Blood ; 114(9): 1820-30, 2009 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-19571317

RESUMO

The discovery of JAK2V617F as an acquired mutation in the majority of patients with myeloproliferative disorders (MPDs) and the key role of the JAK2-STAT5 signaling cascade in normal hematopoiesis has focused attention on the downstream transcriptional targets of STAT5. Despite evidence of its vital role in normal erythropoiesis and its ability to recapitulate many of the features of myeloid malignancies, including the MPDs, few functionally validated targets of STAT5 have been described. Here we used a combination of comparative genomics and chromatin immunoprecipitation assays to identify ID1 as a novel target of the JAK2-STAT5 signaling axis in erythroid cells. STAT5 binds and transactivates a downstream enhancer of ID1, and ID1 expression levels correlate with the JAK2V617F mutation in both retrovirally transfected fetal liver cells and polycythemia vera patients. Knockdown and overexpression studies in a well-characterized erythroid differentiation assay from primary murine fetal liver cells demonstrated a survival-promoting action of ID1. This hitherto unrecognized function implicates ID1 in the expansion of erythroblasts during terminal differentiation and suggests that ID1 plays an important role in the pathogenesis of polycythemia vera. Furthermore, our findings contribute to an increasing body of evidence implicating ID proteins in a wider range of cellular functions than initially appreciated.


Assuntos
Células Eritroides/citologia , Proteína 1 Inibidora de Diferenciação/metabolismo , Janus Quinase 2/metabolismo , Fator de Transcrição STAT5/metabolismo , Animais , Separação Celular , Citometria de Fluxo , Humanos , Janus Quinase 2/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Retroviridae/genética , Transdução de Sinais
9.
Blood ; 113(22): 5456-65, 2009 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-19346495

RESUMO

The basic helix-loop-helix transcription factor Scl/Tal1 controls the development and subsequent differentiation of hematopoietic stem cells (HSCs). However, because few Scl target genes have been validated to date, the underlying mechanisms have remained largely unknown. In this study, we have used ChIP-Seq technology (coupling chromatin immunoprecipitation with deep sequencing) to generate a genome-wide catalog of Scl-binding events in a stem/progenitor cell line, followed by validation using primary fetal liver cells and comprehensive transgenic mouse assays. Transgenic analysis provided in vivo validation of multiple new direct Scl target genes and allowed us to reconstruct an in vivo validated network consisting of 17 factors and their respective regulatory elements. By coupling ChIP-Seq in model cell lines with in vivo transgenic validation and sophisticated bioinformatic analysis, we have identified a widely applicable strategy for the reconstruction of stem cell regulatory networks in which biologic material is otherwise limiting. Moreover, in addition to revealing multiple previously unrecognized links to known HSC regulators, as well as novel links to genes not previously implicated in HSC function, comprehensive transgenic analysis of regulatory elements provided substantial new insights into the transcriptional control of several important hematopoietic regulators, including Cbfa2t3h/Eto2, Cebpe, Nfe2, Zfpm1/Fog1, Erg, Mafk, Gfi1b, and Myb.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento , Hematopoese/genética , Proteínas Proto-Oncogênicas/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Sítios de Ligação/genética , Células Cultivadas , Embrião de Mamíferos/irrigação sanguínea , Perfilação da Expressão Gênica , Genoma , Humanos , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Ligação Proteica , Proteínas Proto-Oncogênicas/metabolismo , Proteína 1 de Leucemia Linfocítica Aguda de Células T , Transcrição Gênica
10.
Blood ; 113(23): 5783-92, 2009 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-19171877

RESUMO

The Lmo2 gene encodes a transcriptional cofactor critical for the development of hematopoietic stem cells. Ectopic LMO2 expression causes leukemia in T-cell acute lymphoblastic leukemia (T-ALL) patients and severe combined immunodeficiency patients undergoing retroviral gene therapy. Tightly controlled Lmo2 expression is therefore essential, yet no comprehensive analysis of Lmo2 regulation has been published so far. By comparative genomics, we identified 17 highly conserved noncoding elements, 9 of which revealed specific acetylation marks in chromatin-immunoprecipitation and microarray (ChIP-chip) assays performed across 250 kb of the Lmo2 locus in 11 cell types covering different stages of hematopoietic differentiation. All candidate regulatory regions were tested in transgenic mice. An extended LMO2 proximal promoter fragment displayed strong endothelial activity, while the distal promoter showed weak forebrain activity. Eight of the 15 distal candidate elements functioned as enhancers, which together recapitulated the full expression pattern of Lmo2, directing expression to endothelium, hematopoietic cells, tail, and forebrain. Interestingly, distinct combinations of specific distal regulatory elements were required to extend endothelial activity of the LMO2 promoter to yolk sac or fetal liver hematopoietic cells. Finally, Sfpi1/Pu.1, Fli1, Gata2, Tal1/Scl, and Lmo2 were shown to bind to and transactivate Lmo2 hematopoietic enhancers, thus identifying key upstream regulators and positioning Lmo2 within hematopoietic regulatory networks.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição GATA/metabolismo , Leucemia/metabolismo , Metaloproteínas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Telomerase/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Linhagem Celular , Imunoprecipitação da Cromatina , Proteínas de Ligação a DNA/genética , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Fatores de Transcrição GATA/genética , Regulação da Expressão Gênica no Desenvolvimento , Genoma/genética , Proteínas com Domínio LIM , Leucemia/genética , Metaloproteínas/genética , Camundongos , Ligação Proteica , Proteínas Proto-Oncogênicas/genética , Telomerase/genética , Análise Serial de Tecidos , Transativadores/genética
11.
Genome Res ; 18(9): 1422-32, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18687876

RESUMO

Altered cis-regulation is thought to underpin much of metazoan evolution, yet the underlying mechanisms remain largely obscure. The stem cell leukemia TAL1 (also known as SCL) transcription factor is essential for the normal development of blood stem cells and we have previously shown that the Tal1 +19 enhancer directs expression to hematopoietic stem cells, hematopoietic progenitors, and to endothelium. Here we demonstrate that an adjacent region 1 kb upstream (+18 element) is in an open chromatin configuration and carries active histone marks but does not function as an enhancer in transgenic mice. Instead, it boosts activity of the +19 enhancer both in stable transfection assays and during differentiation of embryonic stem (ES) cells carrying single-copy reporter constructs targeted to the Hprt locus. The +18 element contains a mammalian interspersed repeat (MIR) which is essential for the +18 function and which was transposed to the Tal1 locus approximately 160 million years ago at the time of the mammalian/marsupial branchpoint. Our data demonstrate a previously unrecognized mechanism whereby enhancer activity is modulated by a transposon exerting a "booster" function which would go undetected by conventional transgenic approaches.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Elementos Facilitadores Genéticos/genética , Evolução Molecular , Sequências Repetitivas Dispersas/genética , Proteínas Proto-Oncogênicas/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular , Linhagem da Célula , Embrião de Mamíferos/metabolismo , Células-Tronco Embrionárias/metabolismo , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Filogenia , Proteínas Proto-Oncogênicas/metabolismo , Proteína 1 de Leucemia Linfocítica Aguda de Células T , Transfecção
12.
Proc Natl Acad Sci U S A ; 104(3): 840-5, 2007 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-17213321

RESUMO

Hematopoietic stem cell (HSC) development is regulated by several signaling pathways and a number of key transcription factors, which include Scl/Tal1, Runx1, and members of the Smad family. However, it remains unclear how these various determinants interact. Using a genome-wide computational screen based on the well characterized Scl +19 HSC enhancer, we have identified a related Smad6 enhancer that also targets expression to blood and endothelial cells in transgenic mice. Smad6, Bmp4, and Runx1 transcripts are concentrated along the ventral aspect of the E10.5 dorsal aorta in the aorta-gonad-mesonephros region from which HSCs originate. Moreover, Smad6, an inhibitor of Bmp4 signaling, binds and inhibits Runx1 activity, whereas Smad1, a positive mediator of Bmp4 signaling, transactivates the Runx1 promoter. Taken together, our results integrate three key determinants of HSC development; the Scl transcriptional network, Runx1 activity, and the Bmp4/Smad signaling pathway.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Proteínas Proto-Oncogênicas/genética , Transdução de Sinais , Animais , Sequência de Bases , Proteína Morfogenética Óssea 4 , Proteínas Morfogenéticas Ósseas/genética , Biologia Computacional , Sequência Conservada , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Células Endoteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Humanos , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Família Multigênica , Células NIH 3T3 , Ligação Proteica , Elementos Reguladores de Transcrição/genética , Alinhamento de Sequência , Proteína Smad6/química , Proteína Smad6/genética , Proteína Smad6/metabolismo
13.
Blood ; 109(5): 1908-16, 2007 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17053063

RESUMO

Transcription factors are key regulators of hematopoietic stem cells (HSCs), yet the molecular mechanisms that control their expression are largely unknown. Previously, we demonstrated that expression of Scl/Tal1, a transcription factor required for the specification of HSCs, is controlled by Ets and GATA factors. Here we characterize the molecular mechanisms controlling expression of Lyl1, a paralog of Scl also required for HSC function. Two closely spaced promoters directed expression to hematopoietic progenitor, megakaryocytic, and endothelial cells in transgenic mice. Conserved binding sites required for promoter activity were bound in vivo by GATA-2 and the Ets factors Fli1, Elf1, Erg, and PU.1. However, despite coregulation of Scl and Lyl1 by the same Ets and GATA factors, Scl expression was initiated prior to Lyl1 in embryonic stem (ES) cell differentiation assays. Moreover, ectopic expression of Scl but not Lyl1 rescued hematopoietic differentiation in Scl-/- ES cells, thus providing a molecular explanation for the vastly different phenotypes of Scl-/- and Lyl1-/- mouse embryos. Furthermore, coregulation of Scl and Lyl1 later during development may explain the mild phenotype of Scl-/- adult HSCs.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Fator de Transcrição GATA2/metabolismo , Hematopoese , Proteínas de Neoplasias/metabolismo , Proteína Proto-Oncogênica c-ets-1/metabolismo , Proteínas Proto-Oncogênicas/deficiência , Sequência de Aminoácidos , Animais , Sequência de Bases , Fatores de Transcrição Hélice-Alça-Hélice Básicos/química , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Linhagem Celular , Sequência Conservada , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Células Endoteliais/metabolismo , Expressão Gênica , Humanos , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Fenótipo , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Alinhamento de Sequência , Proteína 1 de Leucemia Linfocítica Aguda de Células T , Fatores de Tempo
14.
Genome Res ; 16(10): 1310-9, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16963707

RESUMO

The identification of cis-regulatory elements is central to understanding gene transcription. Hypersensitivity of cis-regulatory elements to digestion with DNaseI remains the gold-standard approach to locating such elements. Traditional methods used to identify DNaseI hypersensitive sites are cumbersome and can only be applied to short stretches of DNA at defined locations. Here we report the development of a novel genomic array-based approach to DNaseI hypersensitive site mapping (ADHM) that permits precise, large-scale identification of such sites from as few as 5 million cells. Using ADHM we identified all previously recognized hematopoietic regulatory elements across 200 kb of the mouse T-cell acute lymphocytic leukemia-1 (Tal1) locus, and, in addition, identified two novel elements within the locus, which show transcriptional regulatory activity. We further validated the ADHM protocol by mapping the DNaseI hypersensitive sites across 250 kb of the human TAL1 locus in CD34+ primary stem/progenitor cells and K562 cells and by mapping the previously known DNaseI hypersensitive sites across 240 kb of the human alpha-globin locus in K562 cells. ADHM provides a powerful approach to identifying DNaseI hypersensitive sites across large genomic regions.


Assuntos
Desoxirribonuclease I/metabolismo , Genômica/métodos , Análise em Microsséries/métodos , Elementos Reguladores de Transcrição/genética , Mapeamento por Restrição/métodos , Algoritmos , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Estudos de Avaliação como Assunto , Humanos , Camundongos , Proteínas Proto-Oncogênicas/genética , Proteína 1 de Leucemia Linfocítica Aguda de Células T , Ubiquitina-Proteína Ligases/genética
15.
Blood ; 107(12): 4737-45, 2006 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-16484587

RESUMO

Angiogenesis is critical to the growth and regeneration of tissue but is also a key component of tumor growth and chronic inflammatory disorders. Endoglin plays a key role in angiogenesis by modulating cellular responses to transforming growth factor-beta (TGF-beta) signaling and is upregulated in proliferating endothelial cells. To gain insights into the transcriptional hierarchies that govern endoglin expression, we used a combination of comparative genomic, biochemical, and transgenic approaches. Both the promoter and a region 8 kb upstream of exon 1 were active in transfection assays in endothelial cells. In transgenic mice, the promoter directed low-level expression to a subset of endothelial cells. By contrast, inclusion of the -8 enhancer resulted in robust endothelial activity with additional staining in developing ear mesenchyme. Subsequent molecular analysis demonstrated that both the -8 enhancer and the promoter depend on conserved Ets sites, which were bound in endothelial cells in vivo by Fli-1, Erg, and Elf-1. This study therefore establishes the transcriptional framework within which endoglin functions during angiogenesis.


Assuntos
Endotélio Vascular/metabolismo , Elementos Facilitadores Genéticos/fisiologia , Efrina-A2/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Regiões Promotoras Genéticas/fisiologia , Proteína Proto-Oncogênica c-fli-1/metabolismo , Animais , Cães , Endoglina , Efrina-A2/genética , Éxons/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Camundongos Transgênicos , Células NIH 3T3 , Neovascularização Fisiológica/fisiologia , Gambás , Proteína Proto-Oncogênica c-fli-1/genética , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Regulação para Cima/fisiologia
16.
Blood ; 106(8): 2680-7, 2005 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-15994290

RESUMO

Transcriptional control has been identified as a key mechanism regulating the formation and subsequent behavior of hematopoietic stem cells. We have used a comparative genomics approach to identify transcriptional regulatory elements of the LMO2 gene, a transcriptional cofactor originally identified through its involvement in T-cell leukemia and subsequently shown to be critical for normal hematopoietic and endothelial development. Of the 2 previously characterized LMO2 promoters, the second (proximal) promoter was highly conserved in vertebrates ranging from mammals to fish. Real-time reverse transcriptase-polymerase chain reaction (RT-PCR) expression analysis identified this promoter as the predominant source of transcription in hematopoietic tissue. Transient and stable transfections indicated that the proximal promoter was active in hematopoietic progenitor and endothelial cell lines and this activity was shown to depend on 3 conserved Ets sites that were bound in vivo by E74-like factor 1 (Elf1), Friend leukemia integration 1 (Fli1), and erythroblastosis virus oncogene homolog E twenty-six-1 (Ets1). Finally, transgenic analysis demonstrated that the LMO2 proximal promoter is sufficient for expression in endothelial cells in vivo. No hematopoietic expression was observed, indicating that additional enhancers are required to mediate transcription from the proximal promoter in hematopoietic cells. Together, these results suggest that the conserved proximal promoter is central to LMO2 transcription in hematopoietic and endothelial cells, where it is regulated by Ets factors.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Células Endoteliais/metabolismo , Efrina-A2/metabolismo , Regulação da Expressão Gênica/genética , Metaloproteínas/genética , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas/metabolismo , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Sequência de Bases , Sítios de Ligação , Linhagem Celular , Sequência Conservada/genética , Cães , Efrina-A2/genética , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Hematopoéticas/metabolismo , Humanos , Proteínas com Domínio LIM , Camundongos , Dados de Sequência Molecular , Proteína Proto-Oncogênica c-ets-1 , Proteína Proto-Oncogênica c-fli-1 , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-ets , Ratos , Elementos de Resposta/genética , Alinhamento de Sequência , Transativadores/genética , Fatores de Transcrição/genética , Sítio de Iniciação de Transcrição , Transcrição Gênica/genética
17.
Hum Mol Genet ; 14(5): 595-601, 2005 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-15649946

RESUMO

The development of blood has long served as a model for mammalian cell type specification and differentiation, and yet the underlying transcriptional networks remain ill defined. Characterization of such networks will require genome-wide identification of cis-regulatory sequences and an understanding of how regulatory information is encoded in the primary DNA sequence. Despite progress in lower organisms, genome-wide computational identification of mammalian cis-regulatory sequences has been hindered by increased genomic complexity and cumbersome transgenic assays. Starting with a well-characterized blood stem cell enhancer from the SCL gene, we have developed computational tools for the identification of functionally related gene regulatory sequences. Two candidate enhancers discovered in this way were located in intron 1 of the Fli-1 and PRH/Hex genes, both transcription factors previously implicated in controlling blood and endothelial development. Subsequent transgenic and biochemical analysis demonstrated that the two computationally identified enhancers are functionally related to the SCL stem cell enhancer. The approach developed here may therefore be useful for identifying additional enhancers involved in the control of early blood and endothelial development, and may be adapted to decipher transcriptional regulatory codes controlling a broad range of mammalian developmental programmes.


Assuntos
Diferenciação Celular/genética , Endotélio/fisiologia , Elementos Facilitadores Genéticos , Células-Tronco Hematopoéticas/fisiologia , Antígenos CD34/genética , Antígenos CD34/metabolismo , Sequência de Bases , Diferenciação Celular/fisiologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Endotélio/citologia , Células-Tronco Hematopoéticas/citologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Dados de Sequência Molecular , Proteína Proto-Oncogênica c-fli-1 , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Software , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
18.
Genome Res ; 14(2): 313-8, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14718377

RESUMO

Comparative analysis of genomic sequences is becoming a standard technique for studying gene regulation. However, only a limited number of tools are currently available for the analysis of multiple genomic sequences. An extensive data set for the testing and training of such tools is provided by the SCL gene locus. Here we have expanded the data set to eight vertebrate species by sequencing the dog SCL locus and by annotating the dog and rat SCL loci. To provide a resource for the bioinformatics community, all SCL sequences and functional annotations, comprising a collation of the extensive experimental evidence pertaining to SCL regulation, have been made available via a Web server. A Web interface to new tools specifically designed for the display and analysis of multiple sequence alignments was also implemented. The unique SCL data set and new sequence comparison tools allowed us to perform a rigorous examination of the true benefits of multiple sequence comparisons. We demonstrate that multiple sequence alignments are, overall, superior to pairwise alignments for identification of mammalian regulatory regions. In the search for individual transcription factor binding sites, multiple alignments markedly increase the signal-to-noise ratio compared to pairwise alignments.


Assuntos
Bases de Dados Genéticas/tendências , Marcadores Genéticos/genética , Genoma Humano , Genoma , Internet/tendências , Leucemia/genética , Liases/genética , Sistemas On-Line/tendências , Alinhamento de Sequência/métodos , Animais , Cães , Genes Neoplásicos/genética , Genômica/métodos , Humanos , Leucemia/enzimologia , Camundongos , Dados de Sequência Molecular , Ratos , Projetos de Pesquisa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA