Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Nucleic Acids Res ; 52(D1): D1138-D1142, 2024 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-37933860

RESUMO

BloodSpot is a specialised database integrating gene expression data from acute myeloid leukaemia (AML) patients related to blood cell development and maturation. The database and interface has helped numerous researchers and clinicians to quickly get an overview of gene expression patterns in healthy and malignant haematopoiesis. Here, we present an update to our framework that includes protein expression data of sorted single cells. With this update we also introduce datasets broadly spanning age groups, which many users have requested, with particular interest for researchers studying paediatric leukaemias. The backend of the database has been rewritten and migrated to a cloud-based environment to accommodate the growth, and provide a better user-experience for our many international users. Users can now enjoy faster transfer speeds and a more responsive interface. In conclusion, the continuing popularity of the database and emergence of new data modalities has prompted us to rewrite and futureproof the back-end, including paediatric centric views, as well as single cell protein data, allowing us to keep the database updated and relevant for the years to come. The database is freely available at www.bloodspot.eu.


Assuntos
Hematopoese , Leucemia Mieloide Aguda , Criança , Humanos , Células Sanguíneas , Diferenciação Celular , Bases de Dados Genéticas , Hematopoese/genética , Leucemia Mieloide Aguda/genética , Proteínas/genética
2.
Genome Res ; 33(3): 332-345, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36927987

RESUMO

SWI/SNF and NuRD are protein complexes that antagonistically regulate DNA accessibility. However, repression of their activities often leads to unanticipated changes in target gene expression (paradoxical), highlighting our incomplete understanding of their activities. Here we show that SWI/SNF and NuRD are in a tug-of-war to regulate PRC2 occupancy at lowly expressed and bivalent genes in mouse embryonic stem cells (mESCs). In contrast, at promoters of average or highly expressed genes, SWI/SNF and NuRD antagonistically modulate RNA polymerase II (Pol II) release kinetics, arguably owing to accompanying alterations in H3.3 and H2A.Z levels at promoter-flanking nucleosomes, leading to paradoxical changes in gene expression. Owing to this mechanism, the relative activities of the two remodelers potentiate gene promoters toward Pol II-dependent open or PRC2-dependent closed chromatin states. Our results highlight RNA Pol II occupancy as the key parameter in determining the direction of gene expression changes in response to SWI/SNF and NuRD inactivation at gene promoters in mESCs.


Assuntos
RNA Polimerase II , Fatores de Transcrição , Animais , Camundongos , RNA Polimerase II/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Histonas/metabolismo , Nucleossomos/genética , Expressão Gênica
3.
Cell Rep ; 39(6): 110793, 2022 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-35545054

RESUMO

Ribosomopathies constitute a range of disorders associated with defective protein synthesis mainly affecting hematopoietic stem cells (HSCs) and erythroid development. Here, we demonstrate that deletion of poly-pyrimidine-tract-binding protein 1 (PTBP1) in the hematopoietic compartment leads to the development of a ribosomopathy-like condition. Specifically, loss of PTBP1 is associated with decreases in HSC self-renewal, erythroid differentiation, and protein synthesis. Consistent with its function as a splicing regulator, PTBP1 deficiency results in splicing defects in hundreds of genes, and we demonstrate that the up-regulation of a specific isoform of CDC42 partly mimics the protein-synthesis defect associated with loss of PTBP1. Furthermore, PTBP1 deficiency is associated with a marked defect in ribosome biogenesis and a selective reduction in the translation of mRNAs encoding ribosomal proteins. Collectively, this work identifies PTBP1 as a key integrator of ribosomal functions and highlights the broad functional repertoire of RNA-binding proteins.


Assuntos
Células-Tronco Hematopoéticas , Ribossomos , Eritrócitos/metabolismo , Eritropoese , Células-Tronco Hematopoéticas/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas/genética , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Proteína de Ligação a Regiões Ricas em Polipirimidinas/genética , Proteína de Ligação a Regiões Ricas em Polipirimidinas/metabolismo , Proteínas Ribossômicas/metabolismo , Ribossomos/metabolismo
4.
Sci Adv ; 8(11): eabf8627, 2022 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-35302840

RESUMO

Activation of interferon genes constitutes an important anticancer pathway able to restrict proliferation of cancer cells. Here, we demonstrate that the H3K9me3 histone methyltransferase (HMT) suppressor of variegation 3-9 homolog 1 (SUV39H1) is required for the proliferation of acute myeloid leukemia (AML) and find that its loss leads to activation of the interferon pathway. Mechanistically, we show that this occurs via destabilization of a complex composed of SUV39H1 and the two H3K9me2 HMTs, G9A and GLP. Indeed, loss of H3K9me2 correlated with the activation of key interferon pathway genes, and interference with the activities of G9A/GLP largely phenocopied loss of SUV39H1. Last, we demonstrate that inhibition of G9A/GLP synergized with DNA demethylating agents and that SUV39H1 constitutes a potential biomarker for the response to hypomethylation treatment. Collectively, we uncovered a clinically relevant role for H3K9me2 in safeguarding cancer cells against activation of the interferon pathway.

5.
Leukemia ; 35(7): 2030-2042, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33299144

RESUMO

Most AML patients exhibit mutational activation of the PI3K/AKT signaling pathway, which promotes downstream effects including growth, survival, DNA repair, and resistance to chemotherapy. Herein we demonstrate that the inv(16)/KITD816Y AML mouse model exhibits constitutive activation of PI3K/AKT signaling, which was enhanced by chemotherapy-induced DNA damage through DNA-PK-dependent AKT phosphorylation. Strikingly, inhibitors of either PI3K or DNA-PK markedly reduced chemotherapy-induced AKT phosphorylation and signaling leading to increased DNA damage and apoptosis of inv(16)/KITD816Y AML cells in response to chemotherapy. Consistently, combinations of chemotherapy and PI3K or DNA-PK inhibitors synergistically inhibited growth and survival of clonogenic AML cells without substantially inhibiting normal clonogenic bone marrow cells. Moreover, treatment of inv(16)/KITD816Y AML mice with combinations of chemotherapy and PI3K or DNA-PK inhibitors significantly prolonged survival compared to untreated/single-treated mice. Mechanistically, our findings implicate that constitutive activation of PI3K/AKT signaling driven by mutant KIT, and potentially other mutational activators such as FLT3 and RAS, cooperates with chemotherapy-induced DNA-PK-dependent activation of AKT to promote survival, DNA repair, and chemotherapy resistance in AML. Hence, our study provides a rationale to select AML patients exhibiting constitutive PI3K/AKT activation for simultaneous treatment with chemotherapy and inhibitors of DNA-PK and PI3K to improve chemotherapy response and clinical outcome.


Assuntos
Antineoplásicos/farmacologia , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/metabolismo , Mutação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase/farmacologia , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
6.
Cell Res ; 29(3): 221-232, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30617251

RESUMO

Several developmental stages of spermatogenesis are transcriptionally quiescent which presents major challenges associated with the regulation of gene expression. Here we identify that the zygotene to pachytene transition is not only associated with the resumption of transcription but also a wave of programmed mRNA degradation that is essential for meiotic progression. We explored whether terminal uridydyl transferase 4- (TUT4-) or TUT7-mediated 3' mRNA uridylation contributes to this wave of mRNA degradation during pachynema. Indeed, both TUT4 and TUT7 are expressed throughout most of spermatogenesis, however, loss of either TUT4 or TUT7 does not have any major impact upon spermatogenesis. Combined TUT4 and TUT7 (TUT4/7) deficiency results in embryonic growth defects, while conditional gene targeting revealed an essential role for TUT4/7 in pachytene progression. Loss of TUT4/7 results in the reduction of miRNA, piRNA and mRNA 3' uridylation. Although this reduction does not greatly alter miRNA or piRNA expression, TUT4/7-mediated uridylation is required for the clearance of many zygotene-expressed transcripts in pachytene cells. We find that TUT4/7-regulated transcripts in pachytene spermatocytes are characterized by having long 3' UTRs with length-adjusted enrichment for AU-rich elements. We also observed these features in TUT4/7-regulated maternal transcripts whose dosage was recently shown to be essential for sculpting a functional maternal transcriptome and meiosis. Therefore, mRNA 3' uridylation is a critical determinant of both male and female germline transcriptomes. In conclusion, we have identified a novel requirement for 3' uridylation-programmed zygotene mRNA clearance in pachytene spermatocytes that is essential for male meiotic progression.


Assuntos
Prófase Meiótica I/genética , Estágio Paquíteno/genética , Processamento Pós-Transcricional do RNA/fisiologia , Espermatogênese/genética , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estabilidade de RNA/genética , RNA Mensageiro/genética , UDPglucose-Hexose-1-Fosfato Uridiltransferase/metabolismo
7.
Nat Commun ; 10(1): 172, 2019 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-30635567

RESUMO

Cancer sequencing studies have implicated regulators of pre-mRNA splicing as important disease determinants in acute myeloid leukemia (AML), but the underlying mechanisms have remained elusive. We hypothesized that "non-mutated" splicing regulators may also play a role in AML biology and therefore conducted an in vivo shRNA screen in a mouse model of CEBPA mutant AML. This has led to the identification of the splicing regulator RBM25 as a novel tumor suppressor. In multiple human leukemic cell lines, knockdown of RBM25 promotes proliferation and decreases apoptosis. Mechanistically, we show that RBM25 controls the splicing of key genes, including those encoding the apoptotic regulator BCL-X and the MYC inhibitor BIN1. This mechanism is also operative in human AML patients where low RBM25 levels are associated with high MYC activity and poor outcome. Thus, we demonstrate that RBM25 acts as a regulator of MYC activity and sensitizes cells to increased MYC levels.


Assuntos
Regulação Leucêmica da Expressão Gênica , Leucemia Experimental/metabolismo , Leucemia Mieloide Aguda/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas com Motivo de Reconhecimento de RNA/metabolismo , Fatores de Processamento de RNA/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Leucemia Mieloide Aguda/mortalidade , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares , Splicing de RNA , Proteínas Supressoras de Tumor/metabolismo
8.
Cell Rep ; 24(3): 766-780, 2018 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-30021172

RESUMO

Hematopoietic stem cells (HSCs) are considered a heterogeneous cell population. To further resolve the HSC compartment, we characterized a retinoic acid (RA) reporter mouse line. Sub-fractionation of the HSC compartment in RA-CFP reporter mice demonstrated that RA-CFP-dim HSCs were largely non-proliferative and displayed superior engraftment potential in comparison with RA-CFP-bright HSCs. Gene expression analysis demonstrated higher expression of RA-target genes in RA-CFP-dim HSCs, in contrast to the RA-CFP reporter expression, but both RA-CFP-dim and RA-CFP-bright HSCs responded efficiently to RA in vitro. Single-cell RNA sequencing (RNA-seq) of >1,200 HSCs showed that differences in cell cycle activity constituted the main driver of transcriptional heterogeneity in HSCs. Moreover, further analysis of the single-cell RNA-seq data revealed that stochastic low-level expression of distinct lineage-affiliated transcriptional programs is a common feature of HSCs. Collectively, this work demonstrates the utility of the RA-CFP reporter line as a tool for the isolation of superior HSCs.


Assuntos
Compartimento Celular , Ciclo Celular/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Transcrição Gênica , Animais , Ciclo Celular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Genes Reporter , Genoma , Hematopoese/efeitos dos fármacos , Hematopoese/genética , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/efeitos dos fármacos , Proteínas Luminescentes/metabolismo , Camundongos , Transdução de Sinais/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Transcriptoma/genética , Tretinoína/farmacologia
9.
Cell Rep ; 23(9): 2744-2757, 2018 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-29847803

RESUMO

Transcription factors PU.1 and CEBPA are required for the proper coordination of enhancer activity during granulocytic-monocytic (GM) lineage differentiation to form myeloid cells. However, precisely how these factors control the chronology of enhancer establishment during differentiation is not known. Through integrated analyses of enhancer dynamics, transcription factor binding, and proximal gene expression during successive stages of murine GM-lineage differentiation, we unravel the distinct kinetics by which PU.1 and CEBPA coordinate GM enhancer activity. We find no evidence of a pioneering function of PU.1 during late GM-lineage differentiation. Instead, we delineate a set of enhancers that gain accessibility in a CEBPA-dependent manner, suggesting a pioneering function of CEBPA. Analyses of Cebpa null bone marrow demonstrate that CEBPA controls PU.1 levels and, unexpectedly, that the loss of CEBPA results in an early differentiation block. Taken together, our data provide insights into how PU.1 and CEBPA functionally interact to drive GM-lineage differentiation.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/deficiência , Diferenciação Celular/genética , Elementos Facilitadores Genéticos/genética , Células Mieloides/citologia , Proteínas Proto-Oncogênicas/metabolismo , Transativadores/metabolismo , Animais , Sequência de Bases , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Linhagem Celular , Linhagem da Célula , Cromatina/metabolismo , Feminino , Regulação da Expressão Gênica , Granulócitos/citologia , Granulócitos/metabolismo , Camundongos , Monócitos/citologia , Monócitos/metabolismo , Células Mieloides/metabolismo , Ligação Proteica
11.
Cell ; 163(7): 1663-77, 2015 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-26627738

RESUMO

Within the bone marrow, stem cells differentiate and give rise to diverse blood cell types and functions. Currently, hematopoietic progenitors are defined using surface markers combined with functional assays that are not directly linked with in vivo differentiation potential or gene regulatory mechanisms. Here, we comprehensively map myeloid progenitor subpopulations by transcriptional sorting of single cells from the bone marrow. We describe multiple progenitor subgroups, showing unexpected transcriptional priming toward seven differentiation fates but no progenitors with a mixed state. Transcriptional differentiation is correlated with combinations of known and previously undefined transcription factors, suggesting that the process is tightly regulated. Histone maps and knockout assays are consistent with early transcriptional priming, while traditional transplantation experiments suggest that in vivo priming may still allow for plasticity given strong perturbations. These data establish a reference model and general framework for studying hematopoiesis at single-cell resolution.


Assuntos
Hematopoese , Células Progenitoras Mieloides/citologia , Células Progenitoras Mieloides/metabolismo , Análise de Célula Única , Transcriptoma , Animais , Transplante de Medula Óssea , Proteínas Estimuladoras de Ligação a CCAAT/genética , Técnicas de Inativação de Genes , Sequenciamento de Nucleotídeos em Larga Escala , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sequência de RNA , Fatores de Transcrição/metabolismo
12.
Genes Dev ; 29(18): 1915-29, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26385962

RESUMO

The balance between self-renewal and differentiation is crucial for the maintenance of hematopoietic stem cells (HSCs). Whereas numerous gene regulatory factors have been shown to control HSC self-renewal or drive their differentiation, we have relatively few insights into transcription factors that serve to restrict HSC differentiation. In the present work, we identify ETS (E-twenty-six)-related gene (ERG) as a critical factor protecting HSCs from differentiation. Specifically, loss of Erg accelerates HSC differentiation by >20-fold, thus leading to rapid depletion of immunophenotypic and functional HSCs. Molecularly, we could demonstrate that ERG, in addition to promoting the expression of HSC self-renewal genes, also represses a group of MYC targets, thereby explaining why Erg loss closely mimics Myc overexpression. Consistently, the BET domain inhibitor CPI-203, known to repress Myc expression, confers a partial phenotypic rescue. In summary, ERG plays a critical role in coordinating the balance between self-renewal and differentiation of HSCs.


Assuntos
Diferenciação Celular/genética , Células-Tronco Hematopoéticas/citologia , Proteínas Oncogênicas/metabolismo , Fatores de Transcrição/metabolismo , Animais , Células da Medula Óssea/fisiologia , Adesão Celular/genética , Movimento Celular/genética , Transformação Celular Neoplásica/genética , Células Cultivadas , Deleção de Genes , Camundongos , Proteínas Oncogênicas/genética , Fatores de Transcrição/genética , Regulador Transcricional ERG
13.
BMC Bioinformatics ; 15: 81, 2014 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-24655717

RESUMO

BACKGROUND: RNA-seq data is currently underutilized, in part because it is difficult to predict the functional impact of alternate transcription events. Recent software improvements in full-length transcript deconvolution prompted us to develop spliceR, an R package for classification of alternative splicing and prediction of coding potential. RESULTS: spliceR uses the full-length transcript output from RNA-seq assemblers to detect single or multiple exon skipping, alternative donor and acceptor sites, intron retention, alternative first or last exon usage, and mutually exclusive exon events. For each of these events spliceR also annotates the genomic coordinates of the differentially spliced elements, facilitating downstream sequence analysis. For each transcript isoform fraction values are calculated to identify transcript switching between conditions. Lastly, spliceR predicts the coding potential, as well as the potential nonsense mediated decay (NMD) sensitivity of each transcript. CONCLUSIONS: spliceR is an easy-to-use tool that extends the usability of RNA-seq and assembly technologies by allowing greater depth of annotation of RNA-seq data. spliceR is implemented as an R package and is freely available from the Bioconductor repository ( http://www.bioconductor.org/packages/2.13/bioc/html/spliceR.html).


Assuntos
Processamento Alternativo/genética , RNA Mensageiro/genética , Análise de Sequência de RNA/métodos , Software , Sequência de Bases , Éxons/genética , Células HCT116 , Humanos , Íntrons/genética , Isoformas de Proteínas/genética
14.
J Exp Med ; 211(1): 5-13, 2014 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-24367003

RESUMO

MLL-fusion proteins are potent inducers of oncogenic transformation, and their expression is considered to be the main oncogenic driving force in ∼10% of human acute myeloid leukemia (AML) patients. These oncogenic fusion proteins are responsible for the initiation of a downstream transcriptional program leading to the expression of factors such as MEIS1 and HOXA9, which in turn can replace MLL-fusion proteins in overexpression experiments. To what extent MLL fusion proteins act on their own during tumor initiation, or if they collaborate with other transcriptional regulators, is unclear. Here, we have compared gene expression profiles from human MLL-rearranged AML to normal progenitors and identified the myeloid tumor suppressor C/EBPα as a putative collaborator in MLL-rearranged AML. Interestingly, we find that deletion of Cebpa rendered murine hematopoietic progenitors completely resistant to MLL-ENL-induced leukemic transformation, whereas C/EBPα was dispensable in already established AMLs. Furthermore, we show that Cebpa-deficient granulocytic-monocytic progenitors were equally resistant to transformation and that C/EBPα collaborates with MLL-ENL in the induction of a transcriptional program, which is also apparent in human AML. Thus, our studies demonstrate a key role of C/EBPα in MLL fusion-driven transformation and find that it sharply demarcates tumor initiation and maintenance.


Assuntos
Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Transformação Celular Neoplásica/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Leucemia Mieloide Aguda/metabolismo , Proteína de Leucina Linfoide-Mieloide/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Animais , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Biologia Computacional , Primers do DNA/genética , Citometria de Fluxo , Deleção de Genes , Perfilação da Expressão Gênica , Histona-Lisina N-Metiltransferase , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Proteína Meis1 , Proteínas de Neoplasias/metabolismo , Reação em Cadeia da Polimerase
15.
Genome Res ; 23(4): 592-603, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23403033

RESUMO

Dynamic shifts in transcription factor binding are central to the regulation of biological processes by allowing rapid changes in gene transcription. However, very few genome-wide studies have examined how transcription factor occupancy is coordinated temporally in vivo in higher animals. Here, we quantified the genome-wide binding patterns of two key hepatocyte transcription factors, CEBPA and CEBPB (also known as C/EBPalpha and C/EBPbeta), at multiple time points during the highly dynamic process of liver regeneration elicited by partial hepatectomy in mouse. Combining these profiles with RNA polymerase II binding data, we find three temporal classes of transcription factor binding to be associated with distinct sets of regulated genes involved in the acute phase response, metabolic/homeostatic functions, or cell cycle progression. Moreover, we demonstrate a previously unrecognized early phase of homeostatic gene expression prior to S-phase entry. By analyzing the three classes of CEBP bound regions, we uncovered mutually exclusive sets of sequence motifs, suggesting temporal codes of CEBP recruitment by differential cobinding with other factors. These findings were validated by sequential ChIP experiments involving a panel of central transcription factors and/or by comparison to external ChIP-seq data. Our quantitative investigation not only provides in vivo evidence for the involvement of many new factors in liver regeneration but also points to similarities in the circuitries regulating self-renewal of differentiated cells. Taken together, our work emphasizes the power of global temporal analyses of transcription factor occupancy to elucidate mechanisms regulating dynamic biological processes in complex higher organisms.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Genes cdc , Homeostase/genética , Regeneração Hepática/genética , Sequências Reguladoras de Ácido Nucleico , Animais , Sítios de Ligação , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Imunoprecipitação da Cromatina , Análise por Conglomerados , Proteínas de Ligação a DNA/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Regulação da Expressão Gênica , Masculino , Camundongos , Anotação de Sequência Molecular , Motivos de Nucleotídeos , Transcrição Gênica
16.
Nat Neurosci ; 15(3): 381-8, S1, 2012 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-22246437

RESUMO

Postsynaptic density protein 95 (PSD-95) is essential for synaptic maturation and plasticity. Although its synaptic regulation has been widely studied, the control of PSD-95 cellular expression is not understood. We found that Psd-95 was controlled post-transcriptionally during neural development. Psd-95 was transcribed early in mouse embryonic brain, but most of its product transcripts were degraded. The polypyrimidine tract binding proteins PTBP1 and PTBP2 repressed Psd-95 (also known as Dlg4) exon 18 splicing, leading to premature translation termination and nonsense-mediated mRNA decay. The loss of first PTBP1 and then of PTBP2 during embryonic development allowed splicing of exon 18 and expression of PSD-95 late in neuronal maturation. Re-expression of PTBP1 or PTBP2 in differentiated neurons inhibited PSD-95 expression and impaired the development of glutamatergic synapses. Thus, expression of PSD-95 during early neural development is controlled at the RNA level by two PTB proteins whose sequential downregulation is necessary for synapse maturation.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Guanilato Quinases/metabolismo , Proteínas de Membrana/metabolismo , Neurogênese/fisiologia , Neurônios/fisiologia , Proteína de Ligação a Regiões Ricas em Polipirimidinas/metabolismo , Fatores Etários , Animais , Proteínas de Transporte/genética , Diferenciação Celular/genética , Células Cultivadas , Córtex Cerebral/citologia , Dendritos/genética , Proteína 4 Homóloga a Disks-Large , Estimulação Elétrica , Ensaio de Desvio de Mobilidade Eletroforética , Embrião de Mamíferos , Potenciais Pós-Sinápticos Excitadores/genética , Potenciais Pós-Sinápticos Excitadores/fisiologia , Éxons/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Guanilato Quinases/genética , Hipocampo/citologia , Proteínas de Homeodomínio/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , Células-Tronco Neurais/fisiologia , Neuroblastoma/patologia , Neurogênese/genética , Neurônios/citologia , Técnicas de Patch-Clamp , Proteína de Ligação a Regiões Ricas em Polipirimidinas/genética , Isoformas de RNA/genética , Isoformas de RNA/metabolismo , Splicing de RNA/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA , Fatores de Transcrição/genética , Transfecção
17.
Biofactors ; 35(3): 227-31, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19322763

RESUMO

The role of the transcription factor CCATT/enhancer binding protein alpha (C/EBPalpha) as a lineage instructive determinant in myelopoiesis is widely accepted. Furthermore, early mutational events ultimately leading to acute myeloid leukemia (AML) often involve abrogation of C/EBPalpha expression and/or function. The main focus of this review is the progression from a preclinical state to AML, and which preleukemic cell population(s) might-in general and in particular in patients with CEBPA mutations-be a target for the secondary genetic and epigenetic events leading to this progression.


Assuntos
Proteína alfa Estimuladora de Ligação a CCAAT/fisiologia , Hematopoese/fisiologia , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/metabolismo , Animais , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Hematopoese/genética , Humanos , Modelos Biológicos
18.
Genes Dev ; 22(10): 1381-96, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18483223

RESUMO

Nonsense-mediated mRNA decay (NMD) is a post-transcriptional surveillance process that eliminates mRNAs containing premature termination codons (PTCs). NMD has been hypothesized to impact on several aspects of cellular function; however, its importance in the context of a mammalian organism has not been addressed in detail. Here we use mouse genetics to demonstrate that hematopoietic-specific deletion of Upf2, a core NMD factor, led to the rapid, complete, and lasting cell-autonomous extinction of all hematopoietic stem and progenitor populations. In contrast, more differentiated cells were only mildly affected in Upf2-null mice, suggesting that NMD is mainly essential for proliferating cells. Furthermore, we show that UPF2 loss resulted in the accumulation of nonproductive rearrangement by-products from the Tcrb locus and that this, as opposed to the general loss of NMD, was particularly detrimental to developing T-cells. At the molecular level, gene expression analysis showed that Upf2 deletion led to a profound skewing toward up-regulated mRNAs, highly enriched in transcripts derived from processed pseudogenes, and that NMD impacts on regulated alternative splicing events. Collectively, our data demonstrate a unique requirement of NMD for organismal survival.


Assuntos
Códon sem Sentido/fisiologia , Rearranjo Gênico/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Células Progenitoras Linfoides/metabolismo , Estabilidade de RNA/genética , Deleção de Sequência/fisiologia , Animais , Sequência de Bases , Proteínas de Transporte/genética , Células Cultivadas , Códon sem Sentido/genética , Perfilação da Expressão Gênica , Rearranjo Gênico/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Biológicos , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas de Ligação a RNA
19.
Biochim Biophys Acta ; 1766(1): 88-103, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16616425

RESUMO

The CCATT/enhancer binding protein alpha, C/EBPalpha, is a key transcription factor involved in late differentiation events of several cell types. Besides acting as a classical transcription factor, C/EBPalpha is also a well-characterized inhibitor of mitotic growth in most cell lines tested. In line with its anti-mitotic properties, C/EBPalpha has been shown to interact with, and alter the activities of, several cell cycle related proteins and a number of models as to the mechanistics of C/EBPalpha-mediated growth repression have been proposed. More recently, several reports have indicated that C/EBPalpha acts as a tumour suppressor in the hematopoietic system and that mutation within C/EBPalpha is sufficient to induce tumourigenesis. Here, we will review these data and probe the possibility that C/EBPalpha also act as a tumour suppressor in other C/EBPalpha-expressing tissues.


Assuntos
Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Genes Supressores de Tumor/fisiologia , Neoplasias/metabolismo , Animais , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Humanos , Neoplasias/genética , Neoplasias/patologia , Distribuição Tecidual
20.
Mol Cell Biol ; 26(3): 1028-37, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16428455

RESUMO

The C/EBPalpha transcription factor regulates growth and differentiation of several tissues during embryonic development. Several hypotheses as to how C/EBPalpha inhibits cellular growth in vivo have been derived, mainly from studies of tissue culture cells. In fetal liver it has been proposed that a short, centrally located, 15-amino-acid proline-histidine-rich region (PHR) of C/EBPalpha is responsible for the growth-inhibitory function of the protein through its ability to interact with CDK2 and CDK4, thereby inhibiting their activities. Homozygous Cebpa(DeltaPHR/DeltaPHR) (DeltaPHR) mice, carrying a modified cebpa allele lacking amino acids 180 to 194, were born at the Mendelian ratio, reached adulthood, and displayed no apparent adverse phenotypes. When fetal livers from the DeltaPHR mice were analyzed for their expression of cell cycle markers, bromodeoxyuridine incorporation, cyclin-dependent kinase 2 kinase activity, and global gene expression, we failed to detect any cell cycle or developmental differences between the DeltaPHR mice and their control littermates. These in vivo data demonstrate that any C/EBPalpha-mediated growth repression via the PHR as well as the basic region is dispensable for proper embryonic development of, and cell cycle control in, the liver. Surprisingly, control experiments performed in C/EBPalpha null fetal livers yielded similar results.


Assuntos
Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Histidina/química , Fígado/embriologia , Prolina/química , Adipócitos/citologia , Sequência de Aminoácidos , Animais , Proteína alfa Estimuladora de Ligação a CCAAT/química , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Diferenciação Celular , Quinase 2 Dependente de Ciclina/metabolismo , Quinase 4 Dependente de Ciclina/metabolismo , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário/genética , Histidina/genética , Humanos , Fígado/metabolismo , Camundongos , Camundongos Mutantes , Dados de Sequência Molecular , Prolina/genética , Estrutura Terciária de Proteína , Ratos , Deleção de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA