Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 146
Filtrar
2.
Oncogene ; 25(42): 5777-86, 2006 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-16652147

RESUMO

RUNX1 (AML1) is a gene that is frequently disrupted by chromosomal translocations in acute leukemia. Like its Drosophila homolog Runt, RUNX1 both activates and represses transcription. Both Runt and RUNX1 are required for gene silencing during development and a central domain of RUNX1, termed repression domain 2 (RD2), was defined as being required for transcriptional repression and for the silencing of CD4 during T-cell maturation in thymic organ cultures. Although transcriptional co-repressors are known to contact other repression domains in RUNX1, the factors that bind to RD2 had not been defined. Therefore, we tested whether RD2 contacts histone-modifying enzymes that may mediate both repression and gene silencing. We found that RD2 contacts SUV39H1, a histone methyltransferase, via two motifs and that endogenous Suv39h1 associates with a Runx1-regulated repression element in murine erythroleukemia cells. In addition, one of these SUV39H1-binding motifs is also sufficient for binding to histone deacetylases 1 and 3, and both of these domains are required for full RUNX1-mediated transcriptional repression. The association between RUNX1, histone deacetylases and SUV39H1 provides a molecular mechanism for repression and possibly gene silencing mediated by RUNX1.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Histona Desacetilases/metabolismo , Metiltransferases/metabolismo , Proteínas Repressoras/metabolismo , Animais , Células COS , Chlorocebus aethiops , Humanos , Células Jurkat , Transfecção
3.
Bioinformatics ; 17(12): 1228-9, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11751233

RESUMO

UNLABELLED: Through sequencing projects and, more recently, array-based expression analysis experiments, a wealth of genetic data has become accessible via online resources. In contrast, few of the (molecular-) cytogenetic aberration data collected in the last decades are available in a format suitable for data mining procedures. www.progenetix.net is a new online repository for previously published chromosomal aberration data, allowing the addition of band-specific information about chromosomal imbalances to oncologic data analysis efforts. AVAILABILITY: http://www.progenetix.net CONTACT: mbaudis@stanford.edu


Assuntos
Aberrações Cromossômicas , Bases de Dados de Ácidos Nucleicos , Internet , Citogenética
4.
J Cell Sci ; 114(Pt 16): 2921-7, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11686296

RESUMO

Sbf1 (SET binding factor 1) is a pseudo-phosphatase related to the myotubularin family of dual specificity phosphatases, some of which have been implicated in cellular growth and differentiation by virtue of their mutation in human genetic disorders. Sbf1 contains germline-encoded alterations of its myotubularin homology domain that render it non-functional as a phosphatase. We report here the complete structure of Sbfl and further characterization of its growth regulatory properties. In addition to its similarity to myotubularin, the predicted full-length Sbf1 protein contains pleckstrin (PH) and GEF homology domains that are conserved in several proteins implicated in signaling and growth control. Forced expression of wild-type Sbfl in NIH 3T3 cells inhibited their proliferation and altered their morphology. These effects required intact PH, GEF and myotubularin homology domains, implying that growth inhibition may be an intrinsic property of wild-type Sbf1. Conversely, deletion of its conserved N-terminal 44 amino acids alone was sufficient to convert Sbf1 from an inhibitor of cellular growth to a transforming protein in NIH 3T3 cells. Oncogenic forms of Sbf1 partially localized to the nucleus, in contrast to the exclusively cytoplasmic subcellular localization of endogenous Sbf1 in all cell lines and mammalian tissues tested. These data show that the N-terminal GEF homology domain serves to inhibit the transforming effects of Sbf1, possibly sequestering the protein to the cytoplasm, and suggest that this region may be a modulatory domain that relays growth control signals.


Assuntos
Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Transformação Celular Neoplásica/patologia , Fatores de Troca do Nucleotídeo Guanina/química , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Tirosina Fosfatases/química , Células 3T3 , Sequência de Aminoácidos , Animais , Proteínas de Transporte/genética , Divisão Celular , Núcleo Celular/metabolismo , Tamanho Celular , Transformação Celular Neoplásica/metabolismo , Mapeamento Cromossômico , Sequência Conservada , Citoplasma/metabolismo , Humanos , Camundongos , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Transporte Proteico , Proteínas Tirosina Fosfatases não Receptoras , Deleção de Sequência/genética , Transdução de Sinais
6.
Oncogene ; 20(40): 5695-707, 2001 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-11607819

RESUMO

The MLL (Mixed Lineage Leukemia) gene is a common target for chromosomal translocations associated with human acute leukemias. These translocations result in a gain of MLL function by generating novel chimeric proteins containing the amino-terminus of MLL fused in-frame with one of 30 distinct partner proteins. Structure/function studies using an in vitro myeloid progenitor immortalization assay have revealed that at least four nuclear partner proteins contribute transcriptional effector properties to MLL to produce a range of chimeric transcription factors with leukemogenic potential. Mouse models suggest that expression of an MLL fusion protein is necessary but not sufficient for leukemogenesis. Interestingly, whilst all MLL fusion proteins tested so far phenocopy each other with respect to in vitro immortalization, the latency period required for the onset of acute leukemia in vivo is variable and partner protein dependent. We discuss potential mechanisms that may account for the ability of distinct MLL fusion proteins to promote short or long latency leukemogenesis.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Leucemia/etiologia , Leucemia/metabolismo , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Proto-Oncogenes , Fatores de Transcrição , Animais , Proteínas de Ligação a DNA/química , Células-Tronco Hematopoéticas/metabolismo , Histona-Lisina N-Metiltransferase , Humanos , Leucemia/genética , Camundongos , Modelos Genéticos , Proteína de Leucina Linfoide-Mieloide , Proteínas de Fusão Oncogênica/química , Ligação Proteica , Estrutura Terciária de Proteína , Relação Estrutura-Atividade
7.
Development ; 128(18): 3543-57, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11566859

RESUMO

Pbx1 and a subset of homeodomain proteins collaboratively bind DNA as higher-order molecular complexes with unknown consequences for mammalian development. Pbx1 contributions were investigated through characterization of Pbx1-deficient mice. Pbx1 mutants died at embryonic day 15/16 with severe hypoplasia or aplasia of multiple organs and widespread patterning defects of the axial and appendicular skeleton. An obligatory role for Pbx1 in limb axis patterning was apparent from malformations of proximal skeletal elements, but distal structures were unaffected. In addition to multiple rib and vertebral malformations, neural crest cell-derived skeletal structures of the second branchial arch were morphologically transformed into elements reminiscent of first arch-derived cartilages. Although the skeletal malformations did not phenocopy single or compound Hox gene defects, they were restricted to domains specified by Hox proteins bearing Pbx dimerization motifs and unaccompanied by alterations in Hox gene expression. In affected domains of limbs and ribs, chondrocyte proliferation was markedly diminished and there was a notable increase of hypertrophic chondrocytes, accompanied by premature ossification of bone. The pattern of expression of genes known to regulate chondrocyte differentiation was not perturbed in Pbx1-deficient cartilage at early days of embryonic skeletogenesis, however precocious expression of Col1a1, a marker of bone formation, was found. These studies demonstrate a role for Pbx1 in multiple developmental programs and reveal a novel function in co-ordinating the extent and/or timing of proliferation with terminal differentiation. This impacts on the rate of endochondral ossification and bone formation and suggests a mechanistic basis for most of the observed skeletal malformations.


Assuntos
Padronização Corporal , Osso e Ossos/embriologia , Cartilagem/embriologia , Condrócitos/citologia , Proteínas de Ligação a DNA/metabolismo , Proteínas de Homeodomínio/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fatores Etários , Animais , Osso e Ossos/anormalidades , Região Branquial/embriologia , Cartilagem/anormalidades , Diferenciação Celular , Divisão Celular , Cruzamentos Genéticos , Proteínas de Ligação a DNA/genética , Genes Homeobox , Proteínas de Homeodomínio/genética , Homozigoto , Camundongos , Camundongos Knockout , Morfogênese , Osteogênese , Fenótipo , Fator de Transcrição 1 de Leucemia de Células Pré-B , Proteínas Proto-Oncogênicas/genética
8.
Blood ; 98(3): 618-26, 2001 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-11468159

RESUMO

Pbx1 is the product of a proto-oncogene originally discovered at the site of chromosomal translocations in acute leukemias. It binds DNA as a complex with a broad subset of homeodomain proteins, but its contributions to hematopoiesis have not been established. This paper reports that Pbx1 is expressed in hematopoietic progenitors during murine embryonic development and that its absence results in severe anemia and embryonic lethality at embryonic day 15 (E15) or E16. Definitive myeloerythroid lineages are present in Pbx1(-/-) fetal livers, but the total numbers of colony-forming cells are substantially reduced. Fetal liver hypoplasia reflects quantitative as well as qualitative defects in the most primitive multilineage progenitors and their lineage-restricted progeny. Hematopoietic stem cells from Pbx1(-/-) embryos have reduced colony-forming activity and are unable to establish multilineage hematopoiesis in competitive reconstitution experiments. Common myeloid progenitors (CMPs), the earliest known myeloerythroid-restricted progenitors, are markedly depleted in Pbx1(-/-) embryos at E14 and display clonogenic defects in erythroid colony formation. Comparative cell-cycle indexes suggest that these defects result largely from insufficient proliferation. Megakaryocyte- and erythrocyte-committed progenitors are also reduced in number and show decreased erythroid colony-forming potential. Taken together, these data indicate that Pbx1 is essential for the function of hematopoietic progenitors with erythropoietic potential and that its loss creates a proliferative constriction at the level of the CMP. Thus, Pbx1 is required for the maintenance, but not the initiation, of definitive hematopoiesis and contributes to the mitotic amplifications of progenitor subsets through which mature erythrocytes are generated. (Blood. 2001;98:618-626)


Assuntos
Proteínas de Ligação a DNA/farmacologia , Hematopoese/efeitos dos fármacos , Proteínas de Homeodomínio/farmacologia , Fígado/embriologia , Proteínas Proto-Oncogênicas/farmacologia , Anemia/embriologia , Anemia/etiologia , Anemia/mortalidade , Animais , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/metabolismo , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/efeitos dos fármacos , Células Precursoras Eritroides/metabolismo , Feto/metabolismo , Feto/fisiologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Imuno-Histoquímica , Fígado/química , Fígado/fisiologia , Camundongos , Camundongos Knockout , Fator de Transcrição 1 de Leucemia de Células Pré-B , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Transcrição/farmacologia
9.
Anim Reprod Sci ; 65(1-2): 135-47, 2001 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-11182515

RESUMO

The Northern Hairy-nosed Wombat (Lasiorhinus krefftii) is a highly endangered marsupial species and every possible option for sustaining the species needs to be explored. One important approach may be the development of assisted reproductive technologies in the non-endangered Common Wombat (Vombatus ursinus) and Southern Hairy-nosed Wombat (Lasiorhinus latifrons) for application in breeding the Northern Hairy-nosed Wombat. In this study, it was examined whether cryopreserved Wombat ovarian tissue would develop following xenografting to immunologically deficient rats. Ovarian tissue was collected from Common Wombats (n = 3) and cryopreserved as small cortical pieces. After thawing the cortical pieces were grafted underneath the kidney capsule of Nude rats (n = 16). The grafts were recovered at 2, 4, and 10 weeks after transplantation and their gross and histological appearance investigated. Two weeks after grafting (n = 2), the tissue was revascularized and healthy primordial follicles were present. At week 4 (n = 2), some follicular development was present. At week 10, six rats received human chorionic gonadotrophin (hCG) to trigger follicle and oocyte maturation while another six rats were not given any treatment. The administration of hCG did not induce preovulatory follicles and oocyte maturation although type 5 follicles were present in ovarian tissue collected 10 weeks posttransplantation in both treated and untreated groups. This study demonstrates for the first time that Wombat ovarian tissue can survive and function when grafted into immunocompromized rats and that Wombat ovarian follicles can be recruited to growth and development in an ovarian xenograft. This model system has the potential to produce mature oocytes from endangered species for use in assisted reproductive technologies such as in vitro fertilization (IVF), intracytoplasmic sperm injection (ICSI), and mature oocytes from non-endangered species for nuclear transfer which may be necessary for the preservation of critically endangered species.


Assuntos
Marsupiais/fisiologia , Folículo Ovariano/crescimento & desenvolvimento , Ovário/transplante , Transplante Heterólogo/veterinária , Animais , Conservação dos Recursos Naturais/métodos , Criopreservação/veterinária , Feminino , Histocitoquímica/veterinária , Ovário/crescimento & desenvolvimento , Ovário/fisiologia , Ratos , Ratos Nus , Transplante Heterólogo/métodos
10.
Mech Dev ; 100(1): 131-5, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11118899

RESUMO

Mammalian Pbx genes (Pbx1-3) encode a family of TALE homeodomain proteins that function as transcriptional regulators in numerous cell types (Curr. Opin. Genet. Dev. 8 (1998) 423). The present study highlights distinctive features of Pbx1b expression during mouse embryonic development as a framework to understand its biological functions. Immunohistochemical analyses demonstrate extensive expression of Pbx1b throughout post-implantation development, with highest levels observed during early to mid-gestation. Its initial distribution is predominantly associated with condensing mesoderm, however, Pbx1b displays dynamic expression patterns in derivatives of all principal germ layers. In particular, Pbx1b localizes to sites of mesenchymal-epithelial interactions during periods of active morphogenesis in tissues such as the lung, kidney, tooth buds and vibrissae follicles. Furthermore, BrdU labeling studies reveal that Pbx1b expression domains partially overlap with regions of cellular proliferation. Taken together, these data suggest that Pbx1b contributes to multiple cellular processes during embryogenesis, which may include roles in cell-autonomous regulation as well as in the mediation of tissue interactions.


Assuntos
Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas/genética , Animais , Bromodesoxiuridina/metabolismo , Divisão Celular , Epitélio/embriologia , Imuno-Histoquímica , Mesoderma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fator de Transcrição 1 de Leucemia de Células Pré-B , Fatores de Tempo
11.
Blood ; 96(12): 3887-93, 2000 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11090074

RESUMO

The t(11;19)(q23;p13.1) chromosomal translocation in acute myeloid leukemias fuses the gene encoding transcriptional elongation factor ELL to the MLL gene with consequent expression of an MLL-ELL chimeric protein. To identify potential mechanisms of leukemogenesis by MLL-ELL, its transcriptional and oncogenic properties were investigated. Fusion with MLL preserves the transcriptional elongation activity of ELL but relocalizes it from a diffuse nuclear distribution to the nuclear bodies characteristic of MLL. Using a serial replating assay, it was demonstrated that the MLL-ELL chimeric protein is capable of immortalizing clonogenic myeloid progenitors in vitro after its retroviral transduction into primary murine hematopoietic cells. However, a structure-function analysis indicates that the elongation domain is not essential for myeloid transformation because mutants lacking elongation activity retain a potent ability to immortalize myeloid progenitors. Rather, the highly conserved carboxyl terminal R4 domain is both a necessary and a sufficient contribution from ELL for the immortalizing activity associated with MLL-ELL. The R4 domain demonstrates potent transcriptional activation properties and is required for transactivation of a HoxA7 promoter by MLL-ELL in a transient transcriptional assay. These data indicate that neoplastic transformation by the MLL-ELL fusion protein is likely to result from aberrant transcriptional activation of MLL target genes. Thus, in spite of the extensive diversity of MLL fusion partners, these data, in conjunction with previous studies of MLL-ENL, suggest that conversion of MLL to a constitutive transcriptional activator may be a general model for its oncogenic conversion in myeloid leukemias. (Blood. 2000;96:3887-3893)


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/farmacologia , Leucemia Mieloide/etiologia , Células Progenitoras Mieloides/efeitos dos fármacos , Proteínas de Neoplasias , Fatores de Alongamento de Peptídeos/farmacologia , Proto-Oncogenes , Fatores de Transcrição/farmacologia , Doença Aguda , Sequência de Aminoácidos , Animais , Células da Medula Óssea , Núcleo Celular/química , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Proteínas de Ligação a DNA/química , Embrião de Mamíferos/citologia , Fibroblastos/citologia , Histona-Lisina N-Metiltransferase , Interleucina-3/farmacologia , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patologia , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Células Progenitoras Mieloides/química , Células Progenitoras Mieloides/citologia , Proteína de Leucina Linfoide-Mieloide , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/farmacologia , Fatores de Alongamento de Peptídeos/genética , Estrutura Terciária de Proteína , RNA Polimerase II/metabolismo , Relação Estrutura-Atividade , Fatores de Transcrição/química , Fatores de Transcrição/genética , Transcrição Gênica/efeitos dos fármacos , Fatores de Elongação da Transcrição , Transfecção , Translocação Genética
12.
Blood ; 96(5): 1906-13, 2000 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-10961893

RESUMO

PBX1 is a proto-oncogene that plays important roles in pattern formation during development. It was discovered as a fusion with the E2A gene after chromosomal translocations in a subset of acute leukemias. The resulting E2a-Pbx1 chimeric proteins display potent oncogenic properties that appear to require dimerization with Hox DNA binding partners. To define molecular pathways that may be impacted by E2a-Pbx1, a genetic screen consisting of neonatal retroviral infection was used to identify genes that accelerate development of T-cell tumors in E2A-PBX1 transgenic mice. Retroviral insertions in the Notch1 gene were observed in 88% of tumors arising with a shortened latency. Among these, approximately half created a Notch(IC) allele, encoding the intracellular, signaling portion of Notch1, suggesting a synergistic interaction between the Notch and E2a-Pbx1 pathways in oncogenesis. The remaining proviral insertions involving Notch1 occurred in a more 3' exon, resulting in truncating mutations that deleted the carboxy-terminal region of Notch1 containing negative regulatory sequences (Notch1(DeltaC)). In contrast to Notch(IC), forced expression of Notch1(DeltaC) in transgenic mice did not perturb thymocyte growth or differentiation. However, mice transgenic for both the E2A-PBX1 and Notch1(DeltaC) genes displayed a substantially shortened latency for tumor development compared with E2A-PBX1 single transgenic mice. These studies reveal a novel mechanism for oncogenic activation of Notch1 and demonstrate a collaborative relationship between 2 cellular oncogenes that also contribute to cell fate determination during embryonic development. (Blood. 2000;96:1906-1913)


Assuntos
Proteínas de Homeodomínio/genética , Linfoma de Células T/genética , Proteínas de Membrana/genética , Proteínas de Fusão Oncogênica/genética , Receptores de Superfície Celular , Neoplasias do Timo/genética , Fatores de Transcrição , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Sequência de Bases , Feminino , Deleção de Genes , Regulação Neoplásica da Expressão Gênica , Linfoma de Células T/etiologia , Linfoma de Células T/patologia , Masculino , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Vírus da Leucemia Murina de Moloney/genética , Mutação , Receptor Notch1 , Infecções por Retroviridae/complicações , Infecções por Retroviridae/virologia , Análise de Sobrevida , Neoplasias do Timo/etiologia , Neoplasias do Timo/patologia
13.
Mol Cell Biol ; 20(13): 4900-9, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10848615

RESUMO

Mammalian SET domain-containing proteins define a distinctive class of chromatin-associated factors that are targets for growth control signals and oncogenic activation. SUV39H1, a mammalian ortholog of Drosophila Su(var)3-9, contains both SET and chromo domains, signature motifs for proteins that contribute to epigenetic control of gene expression through effects on the regional organization of chromatin structure. In this report we demonstrate that SUV39H1 represses transcription in a transient transcriptional assay when tethered to DNA through the GAL4 DNA binding domain. Under these conditions, SUV39H1 displays features of a long-range repressor capable of acting over several kilobases to silence basal promoters. A possible role in chromatin-mediated gene silencing is supported by the localization of exogenously expressed SUV39H1 to nuclear bodies with morphologic features suggestive of heterochromatin in interphase cells. In addition, we show that SUV39H1 is phosphorylated specifically at the G(1)/S cell cycle transition and when forcibly expressed suppresses cell growth. Growth suppression as well as the ability of SUV39H1 to form nuclear bodies and silence transcription are antagonized by the oncogenic antiphosphatase Sbf1 that when hyperexpressed interacts with the SET domain and stabilizes the phosphorylated form of SUV39H1. These studies suggest a phosphorylation-dependent mechanism for regulating the chromatin organizing activity of a mammalian su(var) protein and implicate the SET domain as a gatekeeper motif that integrates upstream signaling pathways to epigenetic regulation and growth control.


Assuntos
Proteínas de Transporte/metabolismo , Divisão Celular/fisiologia , Proteínas de Drosophila , Peptídeos e Proteínas de Sinalização Intracelular , Metiltransferases , Proteínas Repressoras/metabolismo , Células 3T3 , Motivos de Aminoácidos , Animais , Proteínas de Transporte/genética , DNA/metabolismo , Drosophila/genética , Fase G1 , Regulação da Expressão Gênica , Inativação Gênica , Humanos , Camundongos , Mutação , Organelas/metabolismo , Fosforilação , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Repressoras/genética , Fase S , Transcrição Gênica
14.
Oncogene ; 19(5): 608-16, 2000 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-10698505

RESUMO

Specific Hox genes are implicated in leukemic transformation, and their selective genetic collaboration with TALE homeobox genes, Pbx and Meis, accentuates their oncogenic potential. The molecular mechanisms underlying these coordinate functions, however, have not been characterized. In this study, we demonstrate that HoxA9 requires its Pbx interaction motif as well as its amino terminus to enhance the clonogenic potential of myeloid progenitors in vitro. We further show that HoxA9 forms functional trimeric DNA binding complexes with Pbx and Meis-like proteins on a modified enhancer. DNA binding complexes containing HoxA9 and TALE homeoproteins display cooperative transcriptional activity and are present in leukemic cells. Trimeric complex formation on its own, however, is not sufficient for HoxA9-mediated immortalization. Rather, structure-function analyses demonstrate that domains of HoxA9 which are necessary for cellular transformation are coincident with those required for trimer-mediated transcriptional activation. Furthermore, the amino terminus of HoxA9 provides essential transcriptional effector properties and its requirement for myeloid transformation can be functionally replaced by the VP16 activation domain. These data suggest that biochemical interactions between HoxA9 and TALE homeoproteins mediate cellular transformation in hematopoietic cells, and that their transcriptional activity in higher order DNA binding complexes provides a molecular basis for their collaborative roles in leukemogenesis.


Assuntos
Células-Tronco Hematopoéticas/metabolismo , Proteínas de Homeodomínio/metabolismo , Proteínas de Homeodomínio/fisiologia , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Repressoras , Motivos de Aminoácidos , Animais , Biopolímeros/genética , Biopolímeros/metabolismo , Divisão Celular/genética , Linhagem Celular , Linhagem Celular Transformada , Transformação Celular Neoplásica/patologia , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Hematopoéticas/patologia , Células-Tronco Hematopoéticas/fisiologia , Proteínas de Homeodomínio/química , Leucemia Mieloide/genética , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patologia , Camundongos , Camundongos Endogâmicos C57BL , Proteína Meis1 , Proteínas de Neoplasias/química , Proteínas de Neoplasias/fisiologia , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/fisiologia , Células Tumorais Cultivadas
17.
Mol Cell Biol ; 19(7): 5134-42, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10373562

RESUMO

Pbx/exd proteins modulate the DNA binding affinities and specificities of Hox proteins and contribute to the execution of Hox-dependent developmental programs in arthropods and vertebrates. Pbx proteins also stably heterodimerize and bind DNA with Meis and Pknox1-Prep1, additional members of the TALE (three-amino-acid loop extension) superclass of homeodomain proteins that function on common genetic pathways with a subset of Hox proteins. In this study, we demonstrated that Pbx and Meis bind DNA as heterotrimeric complexes with Hoxb1 on a genetically defined Hoxb2 enhancer, r4, that mediates the cross-regulatory transcriptional effects of Hoxb1 in vivo. The DNA binding specificity of the heterotrimeric complex for r4 is mediated by a Pbx-Hox site in conjunction with a distal Meis site, which we showed to be required for ternary complex formation and Meis-enhanced transcription. Formation of heterotrimeric complexes in which all three homeodomains bind their cognate DNA sites is topologically facilitated by the ability of Pbx and Meis to interact through their amino termini and bind DNA without stringent half-site orientation and spacing requirements. Furthermore, Meis site mutation in the Hoxb2 enhancer phenocopies Pbx-Hox site mutation to abrogate enhancer-directed expression of a reporter transgene in the murine embryonic hindbrain, demonstrating that DNA binding by all three proteins is required for trimer function in vivo. Our data provide in vitro and in vivo evidence for the combinatorial regulation of Hox and TALE protein functions that are mediated, in part, by their interdependent DNA binding activities as ternary complexes. As a consequence, Hoxb1 employs Pbx and Meis-related proteins, as a pair of essential cofactors in a higher-order molecular complex, to mediate its transcriptional effects on an endogenous Hox response element.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Elementos Facilitadores Genéticos , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Transcrição/genética , Animais , Sítios de Ligação , Células COS , Sequência Consenso , DNA/metabolismo , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Dimerização , Proteínas de Homeodomínio/química , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Transgênicos , Proteína Meis1 , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Fator de Transcrição 1 de Leucemia de Células Pré-B , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/genética , Rombencéfalo/metabolismo , Soluções , Transcrição Gênica
18.
Mol Cell Biol ; 19(6): 4443-51, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10330184

RESUMO

The hepatic leukemia factor (HLF) gene codes for a basic region-leucine zipper (bZIP) protein that is disrupted by chromosomal translocations in a subset of pediatric acute lymphoblastic leukemias. HLF undergoes fusions with the E2A gene, resulting in chimeric E2a-Hlf proteins containing the E2a transactivation domains and the Hlf bZIP DNA binding and dimerization motifs. To investigate the in vivo role of this chimeric bZIP protein in oncogenic transformation, its expression was directed to the lymphoid compartments of transgenic mice. Within the thymus, E2a-Hlf induced profound hypoplasia, premature involution, and progressive accumulation of a T-lineage precursor population arrested at an early stage of maturation. In the spleen, mature T cells were present but in reduced numbers, and they lacked expression of the transgene, suggesting further that E2a-Hlf expression was incompatible with T-cell differentiation. In contrast, mature splenic B cells expressed E2a-Hlf but at lower levels and without apparent adverse or beneficial effects on their survival. Approximately 60% of E2A-HLF mice developed lymphoid malignancies with a mean latency of 10 months. Tumors were monoclonal, consistent with a requirement for secondary genetic events, and displayed phenotypes of either mid-thymocytes or, rarely, B-cell progenitors. We conclude that E2a-Hlf disrupts the differentiation of T-lymphoid progenitors in vivo, leading to profound postnatal thymic depletion and rendering B- and T-cell progenitors susceptible to malignant transformation.


Assuntos
Transformação Celular Neoplásica , Proteínas de Ligação a DNA/fisiologia , Linfócitos/fisiologia , Proteínas de Fusão Oncogênica/fisiologia , Fatores Etários , Animais , Linfócitos B/metabolismo , Fatores de Transcrição de Zíper de Leucina Básica , Proteínas de Ligação a DNA/análise , Citometria de Fluxo , Imuno-Histoquímica , Linfoma/patologia , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Modelos Biológicos , Modelos Genéticos , Neoplasias Experimentais , Proteínas de Fusão Oncogênica/análise , Células-Tronco/fisiologia , Sobrevida , Linfócitos T/metabolismo , Timo/metabolismo , Fatores de Tempo
19.
Cell ; 96(4): 587-97, 1999 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-10052460

RESUMO

Hox homeodomain proteins are developmental regulators that determine body plan in a variety of organisms. A majority of the vertebrate Hox proteins bind DNA as heterodimers with the Pbx1 homeodomain protein. We report here the 2.35 A structure of a ternary complex containing a human HoxB1-Pbx1 heterodimer bound to DNA. Heterodimer contacts are mediated by the hexapeptide of HoxB1, which binds in a pocket in the Pbx1 protein formed in part by a three-amino acid insertion in the Pbx1 homeodomain. The Pbx1 DNA-binding domain is larger than the canonical homeodomain, containing an additional alpha helix that appears to contribute to binding of the HoxB1 hexapeptide and to stable binding of Pbx1 to DNA. The structure suggests a model for modulation of Hox DNA binding activity by Pbx1 and related proteins.


Assuntos
Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/química , Proteínas de Homeodomínio/genética , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/genética , DNA/química , Dimerização , Humanos , Dados de Sequência Molecular , Fragmentos de Peptídeos/genética , Fator de Transcrição 1 de Leucemia de Células Pré-B , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos
20.
Mol Cell Biol ; 19(1): 764-76, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9858599

RESUMO

Genes encoding the Phe-Gly (FG) repeat-containing nucleoporins NUP98 and CAN/NUP214 are at the breakpoints of several chromosomal translocations associated with human acute myeloid leukemia (AML), but their role in oncogenesis is unclear. Here we demonstrate that the NUP98-HOXA9 fusion gene encodes two nuclear oncoproteins with either 19 or 37 NUP98 FG repeats fused to the DNA binding and PBX heterodimerization domains of the transcription factor HOXA9. Both NUP98-HOXA9 chimeras transformed NIH 3T3 fibroblasts, and this transformation required the HOXA9 domains for DNA binding and PBX interaction. Surprisingly, the FG repeats acted as very potent transactivators of gene transcription. This NUP98-derived activity is essential for transformation and can be replaced by the bona fide transactivation domain of VP16. Interestingly, FG repeat-containing segments derived from the nucleoporins NUP153 and CAN/NUP214 functioned similarly to those from NUP98. We further demonstrate that transactivation by FG repeat-rich segments of NUP98 correlates with their ability to interact functionally and physically with the transcriptional coactivators CREB binding protein (CBP) and p300. This finding shows, for the first time, that a translocation-generated fusion protein appears to recruit CBP/p300 as an important step of its oncogenic mechanism. Together, our results suggest that NUP98-HOXA9 chimeras are aberrant transcription factors that deregulate HOX-responsive genes through the transcriptional activation properties of nucleoporin-specific FG repeats that recruit CBP/p300. Indeed, FG repeat-mediated transactivation may be a shared pathogenic function of nucleoporins implicated human AML.


Assuntos
Glicina/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Membrana/genética , Complexo de Proteínas Formadoras de Poros Nucleares , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fenilalanina/metabolismo , Transativadores/metabolismo , Transcrição Gênica , Células 3T3 , Animais , Fusão Gênica Artificial , Proteína de Ligação a CREB , Transformação Celular Neoplásica , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Dimerização , Humanos , Proteínas de Membrana/metabolismo , Camundongos , Fator de Transcrição 1 de Leucemia de Células Pré-B , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Ativação Transcricional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...