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










Base de dados
Intervalo de ano de publicação
1.
Blood ; 132(21): e24-e34, 2018 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-30249787

RESUMO

The transforming growth factor beta (TGF-ß) signaling pathway controls hematopoietic stem cell (HSC) behavior in the marrow niche; however, TGF-ß signaling becomes chronic in early-stage myelodysplastic syndrome (MDS). Although TGF-ß signaling normally induces negative feedback, in early-stage MDS, high levels of microRNA-21 (miR-21) contribute to chronic TGF-ß signaling. We found that a TGF-ß signal-correlated gene signature is sufficient to identify an MDS patient population with abnormal RNA splicing (eg, CSF3R) independent of splicing factor mutations and coincident with low HNRNPK activity. Levels of SKI messenger RNA (mRNA) encoding a TGF-ß antagonist are sufficient to identify these patients. However, MDS patients with high SKI mRNA and chronic TGF-ß signaling lack SKI protein because of miR-21 activity. To determine the impact of SKI loss, we examined murine Ski -/- HSC function. First, competitive HSC transplants revealed a profound defect in stem cell fitness (competitive disadvantage) but not specification, homing, or multilineage production. Aged recipients of Ski -/- HSCs exhibited mild phenotypes similar to phenotypes in those with macrocytic anemia. Second, blastocyst complementation revealed a dramatic block in Ski -/- hematopoiesis in the absence of transplantation. Similar to SKI-high MDS patient samples, Ski -/- HSCs strikingly upregulated TGF-ß signaling and deregulated expression of spliceosome genes (including Hnrnpk). Moreover, novel single-cell splicing analyses demonstrated that Ski -/- HSCs and high levels of SKI expression in MDS patient samples share abnormal alternative splicing of common genes (including those that encode splicing factors). We conclude that miR-21-mediated loss of SKI activates TGF-ß signaling and alternative splicing to impair the competitive advantage of normal HSCs (fitness), which could contribute to selection of early-stage MDS-genic clones.


Assuntos
Proteínas de Ligação a DNA/genética , Deleção de Genes , Células-Tronco Hematopoéticas/patologia , Síndromes Mielodisplásicas/genética , Proteínas Proto-Oncogênicas/genética , Splicing de RNA , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Síndromes Mielodisplásicas/metabolismo , Síndromes Mielodisplásicas/patologia , Proteínas Proto-Oncogênicas/metabolismo , RNA Mensageiro/genética
2.
J Clin Invest ; 124(1): 222-36, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24334453

RESUMO

Acute myelogenous leukemia (AML) subtypes that result from oncogenic activation of homeobox (HOX) transcription factors are associated with poor prognosis. The HOXA9 transcription activator and growth factor independent 1 (GFI1) transcriptional repressor compete for occupancy at DNA-binding sites for the regulation of common target genes. We exploited this HOXA9 versus GFI1 antagonism to identify the genes encoding microRNA-21 and microRNA-196b as transcriptional targets of HOX-based leukemia oncoproteins. Therapeutic inhibition of microRNA-21 and microRNA-196b inhibited in vitro leukemic colony forming activity and depleted in vivo leukemia-initiating cell activity of HOX-based leukemias, which led to leukemia-free survival in a murine AML model and delayed disease onset in xenograft models. These data establish microRNA as functional effectors of endogenous HOXA9 and HOX-based leukemia oncoproteins, provide a concise in vivo platform to test RNA therapeutics, and suggest therapeutic value for microRNA antagonists in AML.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Leucemia Mieloide Aguda/metabolismo , MicroRNAs/genética , Células-Tronco Neoplásicas/fisiologia , Animais , Sequência de Bases , Sítios de Ligação , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Terapia Combinada , Citarabina/administração & dosagem , Proteínas de Ligação a DNA/metabolismo , Doxorrubicina/administração & dosagem , Regulação Leucêmica da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Humanos , Quimioterapia de Indução , Leucemia Mieloide Aguda/patologia , Leucemia Mieloide Aguda/terapia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , MicroRNAs/metabolismo , Proteína Meis1 , Proteínas de Neoplasias/metabolismo , Oligonucleotídeos Fosforotioatos/genética , Fator de Transcrição 1 de Leucemia de Células Pré-B , Ligação Proteica , Proteínas Proto-Oncogênicas/metabolismo , Sequências Reguladoras de Ácido Nucleico , Fatores de Transcrição/metabolismo , Transcriptoma , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Methods Mol Biol ; 928: 185-95, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22956143

RESUMO

MicroRNAs (miRNAs) are highly conserved small RNAs which regulate gene expression primarily through base pairing to the 3' untranslated region of target messenger RNA (mRNA), leading to mRNA degradation or translation inhibition depending on the complementarity between the miRNA and target mRNA. Single miRNA regulates multiple target mRNA. miRNAs have been shown to regulate gene expression in the hematopoietic stem cells, as well as at key decision points for various lineages. However, aberrant expression of miRNAs has been documented in cancer and disease models. Rigorous dissection of miRNA pathways and biology requires facile loss of function modeling. This chapter describes detailed protocol for knockdown miRNA-21 which is involved in myelopoiesis using antagomiRs in primary murine bone marrow stem/progenitor cells.


Assuntos
Células da Medula Óssea/metabolismo , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Oligorribonucleotídeos/genética , Oligorribonucleotídeos/farmacologia , Animais , Células da Medula Óssea/efeitos dos fármacos , Camundongos
4.
Blood ; 118(26): 6871-80, 2011 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-22042697

RESUMO

Protracted inhibition of osteoblast (OB) differentiation characterizes multiple myeloma (MM) bone disease and persists even when patients are in long-term remission. However, the underlying pathophysiology for this prolonged OB suppression is unknown. Therefore, we developed a mouse MM model in which the bone marrow stromal cells (BMSCs) remained unresponsive to OB differentiation signals after removal of MM cells. We found that BMSCs from both MM-bearing mice and MM patients had increased levels of the transcriptional repressor Gfi1 compared with controls and that Gfi1 was a novel transcriptional repressor of the critical OB transcription factor Runx2. Trichostatin-A blocked the effects of Gfi1, suggesting that it induces epigenetic changes in the Runx2 promoter. MM-BMSC cell-cell contact was not required for MM cells to increase Gfi1 and repress Runx2 levels in MC-4 before OBs or naive primary BMSCs, and Gfi1 induction was blocked by anti-TNF-α and anti-IL-7 antibodies. Importantly, BMSCs isolated from Gfi1(-/-) mice were significantly resistant to MM-induced OB suppression. Strikingly, siRNA knockdown of Gfi1 in BMSCs from MM patients significantly restored expression of Runx2 and OB differentiation markers. Thus, Gfi1 may have an important role in prolonged MM-induced OB suppression and provide a new therapeutic target for MM bone disease.


Assuntos
Neoplasias Ósseas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Mieloma Múltiplo/metabolismo , Osteoblastos/metabolismo , Células Estromais/metabolismo , Fatores de Transcrição/metabolismo , Células 3T3 , Animais , Western Blotting , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Neoplasias Ósseas/genética , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Proteínas de Ligação a DNA/genética , Feminino , Expressão Gênica , Humanos , Interleucina-7/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Osteoblastos/patologia , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Estromais/patologia , Fatores de Transcrição/genética , Fator de Necrose Tumoral alfa/metabolismo
5.
Proc Natl Acad Sci U S A ; 107(43): 18505-10, 2010 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-20937872

RESUMO

T-cell homeostasis is essential for normal functioning of the immune system. IL-7 receptor (IL-7R) and T-cell receptor (TCR) signaling are pivotal for T-cell homeostatic regulation. The detailed mechanisms regulating T-cell homeostasis and how IL-7R and TCR signaling are coordinated are largely unknown. Here we demonstrate that T cell-specific deletion of cell-division cycle 42 (Cdc42) GTPase causes a profound loss of mature T cells. Deletion of Cdc42 leads to a markedly increased expression of growth factor independence-1 (Gfi-1) and represses expression of IL-7Rα. In the absence of Cdc42, aberrant ERK1/2 MAP kinase activity results in enhanced, TCR-mediated T-cell proliferation. In vivo reconstitution of effector-binding-defective Cdc42 mutants and the effector p21 protein-activated kinase 1 (PAK1) into Cdc42-deficient T cells showed that PAK1 is both necessary and sufficient for Cdc42-regulated T-cell homeostasis. Thus, T-cell homeostasis is maintained through a concerted regulation of Gfi-1-IL-7R-controlled cytokine responsiveness and ERK-mediated TCR signaling strength by the Cdc42-PAK1 signaling axis.


Assuntos
Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Interleucina-7/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo , Animais , Sequência de Bases , Diferenciação Celular , Proliferação de Células , Primers do DNA/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Expressão Gênica , Homeostase , Ativação Linfocitária , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Linfócitos T/citologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteína cdc42 de Ligação ao GTP/deficiência , Proteína cdc42 de Ligação ao GTP/genética , Quinases Ativadas por p21/genética , Quinases Ativadas por p21/metabolismo
6.
Exp Hematol ; 38(8): 629-640.e1, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20399246

RESUMO

OBJECTIVE: The transcription factor PU.1 (encoded by Sfpi1) promotes myeloid differentiation, but it is unclear what downstream genes are involved. Micro RNAs (miRNAs) are a class of small RNAs that regulate many cellular pathways, including proliferation, survival, and differentiation. The objective of this study was to identify miRNAs downstream of PU.1 that regulate hematopoietic development. MATERIALS AND METHODS: miRNAs that change expression in a PU.1-inducible cell line were identified with microarrays. The promoter for an miRNA cluster upregulated by PU.1 induction was analyzed for PU.1 binding by electrophoretic mobility shift and chromatin immunoprecipitation assays. Retroviral transduction of hematopoietic progenitors was performed to evaluate the effect of miRNA expression on hematopoietic development in vitro and in vivo. RESULTS: We identified an miRNA cluster whose pri-transcript is regulated by PU.1. The pri-miRNA encodes three mature miRNAs: miR-23a, miR-27a, and miR-24-2. Each miRNA is more abundant in myeloid cells compared to lymphoid cells. When hematopoietic progenitors expressing the 23a cluster miRNAs were cultured in B-cell-promoting conditions, we observed a dramatic decrease in B lymphopoiesis and an increase in myelopoiesis compared to control cultures. In vivo, hematopoietic progenitors expressing the miR-23a cluster generate reduced numbers of B cells compared to control cells. CONCLUSIONS: The miR-23a cluster is a downstream target of PU.1 involved in antagonizing lymphoid cell fate acquisition. Although miRNAs have been identified downstream of PU.1 in mediating development of monocytes and granulocytes, the 23a cluster is the first downstream miRNA target implicated in regulating development of myeloid vs lymphoid cells.


Assuntos
Linfócitos B/metabolismo , Diferenciação Celular/fisiologia , Regulação da Expressão Gênica/fisiologia , Linfopoese/fisiologia , MicroRNAs/biossíntese , Proteínas Proto-Oncogênicas/metabolismo , Transativadores/metabolismo , Animais , Linfócitos B/citologia , Linhagem Celular , Células-Tronco Hematopoéticas , Camundongos , MicroRNAs/genética , Células Mieloides/citologia , Células Mieloides/metabolismo , Proteínas Proto-Oncogênicas/genética , Transativadores/genética
7.
PLoS One ; 4(9): e6995, 2009 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-19746166

RESUMO

Chromatin insulators separate active transcriptional domains and block the spread of heterochromatin in the genome. Studies on the chicken hypersensitive site-4 (cHS4) element, a prototypic insulator, have identified CTCF and USF-1/2 motifs in the proximal 250 bp of cHS4, termed the "core", which provide enhancer blocking activity and reduce position effects. However, the core alone does not insulate viral vectors effectively. The full-length cHS4 has excellent insulating properties, but its large size severely compromises vector titers. We performed a structure-function analysis of cHS4 flanking lentivirus-vectors and analyzed transgene expression in the clonal progeny of hematopoietic stem cells and epigenetic changes in cHS4 and the transgene promoter. We found that the core only reduced the clonal variegation in expression. Unique insulator activity resided in the distal 400 bp cHS4 sequences, which when combined with the core, restored full insulator activity and open chromatin marks over the transgene promoter and the insulator. These data consolidate the known insulating activity of the canonical 5' core with a novel 3' 400 bp element with properties similar to the core. Together, they have excellent insulating properties and viral titers. Our data have important implications in understanding the molecular basis of insulator function and design of gene therapy vectors.


Assuntos
Elementos Isolantes , Regiões 3' não Traduzidas , Motivos de Aminoácidos , Animais , Linhagem Celular , Galinhas , Epigênese Genética , Vetores Genéticos , Células-Tronco Hematopoéticas/citologia , Lentivirus/genética , Camundongos , Regiões Promotoras Genéticas , Relação Estrutura-Atividade , Transgenes
8.
Blood ; 114(14): 2909-16, 2009 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-19671922

RESUMO

Cdc42 is a member of the Rho GTPase family that has been implicated in several cell functions including proliferation and migration, but its physiologic role needs to be dissected in each cell type. We achieved B-cell and hematopoietic stem cell deletion of Cdc42 by conditional gene targeting in mice. Deletion of Cdc42 from proB/preB-cell stage significantly blocked B-cell development at T1 and later stages, resulting in reduced mature B-cell populations and reduced antigen-specific immunoglobulin M (IgM), IgG1, and IgG3 production. The Cdc42(-/-) B cells, themselves, were abnormal with impaired proliferation and survival. The mutant B cells were further characterized by a B-cell receptor (BCR) signaling defect with increased Erk and decreased Akt activation, as well as a defect in BCR-mediated B-cell-activating factor (BAFF) receptor up-regulation and subsequent BAFF receptor signaling in mature resting B cells. Surprisingly, Cdc42 was dispensable for stromal cell-derived factor-1alpha (SDF-1alpha)- or B-lymphocyte chemoattractant (BLC)-induced B-cell migration. Finally, loss of Cdc42 from hematopoietic stem cells did not alter common lymphoid progenitor production but severely reduced proB/preB- and immature B-cell populations, indicating that Cdc42 is also involved in B-cell precursor differentiation. These results reveal multifaceted roles of Cdc42 in B-cell development and activation.


Assuntos
Linfócitos B/citologia , Linfócitos B/metabolismo , Ativação Linfocitária , Proteína cdc42 de Ligação ao GTP/fisiologia , Animais , Receptor do Fator Ativador de Células B/metabolismo , Western Blotting , Diferenciação Celular , Movimento Celular , Proliferação de Células , Quimiocina CXCL12/metabolismo , Ensaio de Imunoadsorção Enzimática , Perfilação da Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Imunoglobulina M/deficiência , Integrases/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Células Precursoras de Linfócitos B/metabolismo , Receptores de Antígenos de Linfócitos B/metabolismo , Transdução de Sinais
9.
Blood ; 113(22): 5466-75, 2009 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-19346496

RESUMO

In patients with severe congenital neutropenia (SCN) and mice with growth factor independent-1 (Gfi1) loss of function, arrested myeloid progenitors accumulate, whereas terminal granulopoiesis is blocked. One might assume that Gfi-null progenitors accumulate because they lack the ability to differentiate. Instead, our data indicate that Gfi1 loss of function deregulates 2 separable transcriptional programs, one of which controls the accumulation and lineage specification of myeloid progenitors, but not terminal granulopoiesis. We demonstrate that Gfi1 directly represses HoxA9, Pbx1, and Meis1 during normal myelopoiesis. Gfi1-/- progenitors exhibit elevated levels of HoxA9, Pbx1 and Meis1, exaggerated HoxA9-Pbx1-Meis1 activity, and progenitor transformation in collaboration with oncogenic K-Ras. Limiting HoxA9 alleles corrects, in a dose-dependent manner, in vivo and in vitro phenotypes observed with loss of Gfi1 in myeloid progenitor cells but did not rescue Gfi1-/- blocked granulopoiesis. Thus, Gfi1 integrates 2 events during normal myeloid differentiation; the suppression of a HoxA9-Pbx1-Meis1 progenitor program and the induction of a granulopoietic transcription program.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Células Precursoras de Granulócitos/fisiologia , Granulócitos/fisiologia , Fatores de Transcrição/fisiologia , Animais , Diferenciação Celular/genética , Células Cultivadas , Proteínas de Ligação a DNA/genética , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Predisposição Genética para Doença , Células Precursoras de Granulócitos/metabolismo , Granulócitos/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Leucemia/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Meis1 , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Fator de Transcrição 1 de Leucemia de Células Pré-B , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica/fisiologia
10.
Blood ; 113(19): 4720-8, 2009 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-19278956

RESUMO

The zinc finger protein growth factor independent-1 (Gfi1) is a transcriptional repressor that is critically required for normal granulocytic differentiation. GFI1 loss-of-function mutations are found in some patients with severe congenital neutropenia (SCN). The SCN-associated GFI1-mutant proteins act as dominant negatives to block granulopoiesis through selective deregulation of a subset of GFI1 target genes. Here we show that Gfi1 is a master regulator of microRNAs, and that deregulated expression of these microRNAs recapitulates a Gfi1 loss-of-function block to granulocyte colony-stimulating factor (G-CSF)-stimulated granulopoiesis. Specifically, bone marrow cells from a GFI1-mutant SCN patient and Gfi1(-/-) mice display deregulated expression of miR-21 and miR-196B expression. Flow cytometric analysis and colony assays reveal that the overexpression or depletion of either miR induces changes in myeloid development. However, coexpression of miR-21 and miR-196b (as seen in Gfi1(-/-) mice and a GFI1N382S SCN patient) completely blocks G-CSF-induced granulopoiesis. Thus, our results not only identify microRNAs whose regulation is required during myelopoiesis, but also provide an example of synergy in microRNA biologic activity and illustrate potential mechanisms underlying SCN disease pathogenesis.


Assuntos
Medula Óssea/fisiologia , Proteínas de Ligação a DNA/fisiologia , MicroRNAs/genética , Mielopoese/fisiologia , Fatores de Transcrição/fisiologia , Animais , Transplante de Medula Óssea , Imunoprecipitação da Cromatina , Ensaio de Unidades Formadoras de Colônias , Ensaio de Desvio de Mobilidade Eletroforética , Feminino , Citometria de Fluxo , Perfilação da Expressão Gênica , Fator Estimulador de Colônias de Granulócitos/farmacologia , Granulócitos/citologia , Granulócitos/fisiologia , Células-Tronco Hematopoéticas , Immunoblotting , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/fisiologia , Análise de Sequência com Séries de Oligonucleotídeos , RNA Interferente Pequeno/farmacologia
11.
Blood ; 113(14): 3314-22, 2009 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-19188669

RESUMO

Chromosomal translocations involving the Mixed Lineage Leukemia (MLL) gene produce chimeric proteins that cause abnormal expression of a subset of HOX genes and leukemia development. Here, we show that MLL normally regulates expression of mir-196b, a hematopoietic microRNA located within the HoxA cluster, in a pattern similar to that of the surrounding 5' Hox genes, Hoxa9 and Hoxa10, during embryonic stem (ES) cell differentiation. Within the hematopoietic lineage, mir-196b is most abundant in short-term hematopoietic stem cells and is down-regulated in more differentiated hematopoietic cells. Leukemogenic MLL fusion proteins cause overexpression of mir-196b, while treatment of MLL-AF9 transformed bone marrow cells with mir-196-specific antagomir abrogates their replating potential in methylcellulose. This demonstrates that mir-196b function is necessary for MLL fusion-mediated immortalization. Furthermore, overexpression of mir-196b was found specifically in patients with MLL associated leukemias as determined from analysis of 55 primary leukemia samples. Overexpression of mir-196b in bone marrow progenitor cells leads to increased proliferative capacity and survival, as well as a partial block in differentiation. Our results suggest a mechanism whereby increased expression of mir-196b by MLL fusion proteins significantly contributes to leukemia development.


Assuntos
Transformação Celular Neoplásica/genética , Regulação da Expressão Gênica , MicroRNAs/genética , Proteína de Leucina Linfoide-Mieloide/fisiologia , Animais , Sequência de Bases , Diferenciação Celular/genética , Proliferação de Células , Transformação Celular Neoplásica/patologia , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/patologia , Células-Tronco Embrionárias/fisiologia , Regulação da Expressão Gênica/fisiologia , Histona-Lisina N-Metiltransferase , Leucemia/etiologia , Leucemia/genética , Leucemia/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/fisiologia , Dados de Sequência Molecular , Proteínas Recombinantes de Fusão/fisiologia , Homologia de Sequência do Ácido Nucleico , Regulação para Cima/fisiologia
12.
J Biol Chem ; 283(46): 32056-65, 2008 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-18805794

RESUMO

Growth factor independent-1 (Gfi1) is a zinc finger protein with a SNAG-transcriptional repressor domain. Ajuba is a LIM domain protein that shuttles between the cytoplasm and the nucleus. Ajuba functions as a co-repressor for synthetic Gfi1 SNAG-repressor domain-containing constructs, but a role for Ajuba co-repression of the cognate DNA bound Gfi1 protein has not been defined. Co-immunoprecipitation of synthetic and endogenous proteins and co-elution with gel filtration suggest that an endogenous Ajuba.Gfi1.HDAC multiprotein complex is possible. Active histone deacetylase activity co-immunoprecipitates with Ajuba or Gfi1, and both proteins depend upon histone deacetylases for full transcriptional repression activity. Ajuba LIM domains directly bind to Gfi1, but the association is not SNAG domain-dependent. ChIP analysis and reciprocal knockdown experiments suggest that Ajuba selectively functions as a co-repressor for Gfi1 autoregulation. The data suggest that Ajuba is utilized as a corepressor selectively on Gfi1 target genes.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo , Histona Desacetilases/metabolismo , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição/metabolismo , Linhagem Celular , Sobrevivência Celular , Proteínas de Ligação a DNA/genética , Proteínas de Homeodomínio/genética , Humanos , Proteínas com Domínio LIM , Ligação Proteica , Fatores de Transcrição/genética
13.
Immunity ; 28(3): 370-80, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18328744

RESUMO

Severe congenital neutropenia (SCN) is characterized by a deficiency of mature neutrophils, leading to recurrent bacterial and fungal infections. Although mutations in Elastase-2, neutrophil (ELA2) predominate in human SCN, mutation of Ela2 in mice does not recapitulate SCN. The growth factor independent-1 (GFI1) transcription factor regulates ELA2. Mutations in GFI1 are associated with human SCN, and genetic deletion of Gfi1 results in murine neutropenia. We examined whether human SCN-associated GFI1N382S mutant proteins are causal in SCN and found that GFI1 functions as a rate-limiting granulopoietic molecular switch. The N382S mutation inhibited GFI1 DNA binding and resulted in a dominant-negative block to murine granulopoiesis. Moreover, Gfi1N382S selectively derepressed the monopoietic cytokine CSF1 and its receptor. Gfi1N382S-expressing Csf1-/- cells formed neutrophils. These results reveal a common transcriptional program that underlies both human and murine myelopoiesis, and that is central to the pathogenesis of SCN associated with mutations in GFI1. This shared transcriptional pathway may provide new avenues for understanding SCN caused by mutations in other genes and for clinical intervention into human neutropenias.


Assuntos
Proteínas de Ligação a DNA/genética , Granulócitos/citologia , Hematopoese/genética , Fator Estimulador de Colônias de Macrófagos/metabolismo , Neutropenia/genética , Fatores de Transcrição/genética , Animais , Diferenciação Celular/genética , Linhagem da Célula , Ensaio de Desvio de Mobilidade Eletroforética , Citometria de Fluxo , Células-Tronco Hematopoéticas/citologia , Humanos , Immunoblotting , Imunoprecipitação , Camundongos , Mutação , Neutropenia/congênito , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica
14.
Biochemistry ; 46(26): 7765-80, 2007 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-17555331

RESUMO

The cellular mechanisms that modulate the redox state of p53 tumor suppressor remain unclear, although its DNA binding function is known to be strongly inhibited by oxidative and nitrosative stresses. We show that human p53 is subjected to a new and reversible posttranslational modification, namely, S-glutathionylation in stressed states, including DNA damage. First, a rapid and direct incorporation of biotinylated GSH or GSSG into the purified recombinant p53 protein was observed. The modified p53 had a significantly weakened ability to bind its consensus DNA sequence. Reciprocal immunoprecipitations and a GST overlay assay showed that p53 in tumor cells was marginally glutathionylated; however, the level of modification increased greatly after oxidant and DNA-damaging treatments. GSH modification coexisted with the serine phophorylations in activated p53, and the thiol-conjugated protein was present in nuclei. When tumor cells treated with camptothecin or cisplatin were subsequently exposed to glutathione-enhancing agents, p53 underwent dethiolation accompanied by detectable increases in the level of p21waf1 expression, relative to the DNA-damaging drugs alone. Mass spectrometry of GSH-modified p53 protein identified cysteines 124, 141, and 182, all present in the proximal DNA-binding domain, as the sites of glutathionylation. Biotinylated maleimide also reacted rapidly with Cys141, implying that this is the most reactive cysteine on the p53 surface. The glutathionylatable cysteines were found to exist in a negatively charged microenvironment in cellular p53. Molecular modeling studies located Cys124 and -141 at the dimer interface of p53 and showed glutathionylation of either residue would inhibit p53-DNA association and also interfere with protein dimerization. These results show for the first time that shielding of reactive cysteines contributes to a negative regulation for human p53 and imply that such an inactivation of the transcription factor may represent an acute defensive response with significant consequences for oncogenesis.


Assuntos
Cisteína/química , Glutationa/química , Estresse Oxidativo/fisiologia , Proteína Supressora de Tumor p53/antagonistas & inibidores , Acetilcisteína/farmacologia , Sequência de Aminoácidos , Sítios de Ligação , Butionina Sulfoximina/farmacologia , Camptotecina/farmacologia , Linhagem Celular Tumoral , Reagentes de Ligações Cruzadas/química , Dano ao DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Diamida/farmacologia , Eletroforese em Gel de Poliacrilamida , Escherichia coli/metabolismo , Glutaral/química , Glutationa/análogos & derivados , Glutationa/farmacologia , Dissulfeto de Glutationa/química , Humanos , Peróxido de Hidrogênio/farmacologia , Modelos Moleculares , Estresse Oxidativo/efeitos dos fármacos , Fosforilação , Proteínas Recombinantes/metabolismo , Espectrometria de Massas em Tandem , terc-Butil Hidroperóxido/farmacologia
15.
Arch Biochem Biophys ; 444(2): 174-84, 2005 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-16297848

RESUMO

S-Glutathionylation is emerging as a novel regulatory and adoptive mechanism by which glutathione (GSH or GSSG) conjugation can modify functionally important reactive cysteines in redox-sensitive proteins. The dynamics of generation and reversal of this modification in cells is poorly understood. This study describes the ability and applicability of GSH- and GSSG-affinity matrices to quantitatively bind proteins which harbor reactive cysteines and undergo glutathionylation. We showed that purified proteins, known to be modified by S-thiolation, bind to these matrices, are selectively eluted by dithiothreitol and rapidly incorporate biotin-labeled GSH or GSSG in vitro. Chromatography of extracts from tumor cells that had been treated with oxidants (diamide, H(2)O(2), tert-butyl hydroperoxide) on GSH-Sepharose showed the specific binding of many proteins, whose levels increased transiently (2- to 6-fold) soon after treatments. However, when these cells were post-incubated in drug/oxidant-free media, protein binding decreased gradually to control levels over 3-12h, thereby demonstrating the central role of cysteine redox status in the binding. Immunoblotting of eluates from GSH-Sepharose showed the presence of known (actin, ubiquitin-activating enzyme E1, NF-kappaB, and proteasome) and putative (p53, glutathione-S-transferase P1) targets for glutathionation. After oxidant withdrawal, many of these proteins displayed unique kinetics in their loss of binding to GSH-matrix, reflecting their differential abilities to recover from cysteine redox changes in cellular milieu. Further, we correlated the kinetics of S-thiolation susceptibility of the proteasome and ubiquitin-E1 proteins with altered levels of protein ubiquitination in H(2)O(2)-treated cells. Our study reveals the hitherto underutilized ability of glutathione matrices for analyzing the kinetics of cysteine redox in cellular proteins and allows easy identification of S-thiolatable proteins.


Assuntos
Biomimética/métodos , Cisteína/metabolismo , Glutationa/metabolismo , Meduloblastoma/metabolismo , Proteínas de Neoplasias/metabolismo , Mapeamento de Interação de Proteínas/métodos , Compostos de Sulfidrila/metabolismo , Sítios de Ligação , Linhagem Celular Tumoral , Cisteína/química , Glutationa/química , Humanos , Cinética , Meduloblastoma/química , Proteínas de Neoplasias/química , Oxirredução , Ligação Proteica , Compostos de Sulfidrila/química
16.
Biochem Biophys Res Commun ; 337(4): 1176-84, 2005 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-16226712

RESUMO

Recent evidence suggests that human O(6)-methylguanine-DNA methyltransferase (MGMT), a DNA repair protein that protects the genome against mutagens and accords tumor resistance to many anticancer alkylating agents, may have other roles besides repair. Therefore, we isolated MGMT-interacting proteins from extracts of HT29 human colon cancer cells using affinity chromatography on MGMT-Sepharose. Specific proteins bound to this column were identified by electrospray ionization tandem mass spectrometry and/or Western blotting. These procedures identified >60 MGMT-interacting proteins with diverse functions including those involved in DNA replication and repair (MCM2, PCNA, ORC1, DNA polymerase delta, MSH-2, and DNA-dependent protein kinase), cell cycle progression (CDK1, cyclin B, CDK2, CDC7, CDC10, 14-3-3 protein, and p21(waf1/cip1)), RNA processing and translation (poly(A)-binding protein, nucleolin, heterogeneous nuclear ribonucleoproteins, A2/B1, and elongation factor-1alpha), several histones (H4, H3.4, and H2A.1), and topoisomerase I. The heat shock proteins, HSP-90alpha and beta, also bound strongly with MGMT. The DNA repair activity of MGMT was greatly enhanced in the presence of interacting proteins or histones. These data, for the first time, suggest that human MGMT is likely to have additional functions, possibly, in sensing and integrating the DNA damage/repair-related signals with replication, cell cycle progression, and genomic stability.


Assuntos
O(6)-Metilguanina-DNA Metiltransferase/química , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Proteômica , Sequência de Aminoácidos , Western Blotting , Ciclo Celular , Linhagem Celular Tumoral , Cromatografia de Afinidade , DNA/genética , Reparo do DNA/genética , Replicação do DNA/genética , Histonas/metabolismo , Humanos , Espectrometria de Massas , Dados de Sequência Molecular , Ligação Proteica
17.
J Agric Food Chem ; 52(23): 7033-8, 2004 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-15537314

RESUMO

Fatty acids are the largest component of lipids and have become a useful tool in the determination of live feeds to a variety of cultured species. Bioencapsulation is a technique which allows high-level incorporation of desired components (i.e., fatty acids, vitamins, antibiotics, etc.) in live feeds, which in turn can be supplemented to the consumer organisms. The procedure described in the present study serves as a platform of technology for enriching the Streptocephalus dichotomus. Uptake of two enrichment diets (ALGAMAC2000 and DHA-SELCO) by adult S. dichotomus was investigated. The fatty acid profile supports the hypothesis that the enrichment diet increases the level of essential fatty acids, such as linolic, linolenic, eicosapentenoic, and docosahexaenoic acids. The average content (percent of total fatty acids detected) of the enriched organism by different highly unsaturated fatty acid (HUFA) products were as follows: ALGAMAC2000 showed 14-22% saturated fatty acid (SFA), 17-18% monounsaturated fatty acid (MUFA), 28-41% polyunsaturated fatty acid (PUFA), 23-34% n-3, and 4.9-7.5% n-6, whereas DHA-SELCO showed about 20-23% SFA, 20-26% MUFA, 38% PUFA, 28-31% n-3, and 7.5-10% n-6. Our present investigation proves that both HUFA-rich diets appear to be an appropriate enrichment diet, and further provides an additional rationale for using fairy shrimp as a maturation diet for any cultivable freshwater organism.


Assuntos
Anostraca/química , Dieta , Emulsões/química , Ácidos Graxos Insaturados/análise , Ácidos Graxos/análise , Animais , Anostraca/crescimento & desenvolvimento , Emulsões/administração & dosagem , Ácidos Graxos Insaturados/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...