Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 36(13): 1849-1861, 2017 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-27748762

RESUMO

BRAF inhibitor (BRAFi) therapy for melanoma patients harboring the V600E mutation is initially highly effective, but almost all patients relapse within a few months. Understanding the molecular mechanisms underpinning BRAFi-based therapy is therefore an important issue. Here we identified a previously unsuspected mechanism of BRAFi resistance driven by elevated Hedgehog (Hh) pathway activation that is observed in a cohort of melanoma patients after vemurafenib treatment. Specifically, we demonstrate that melanoma cell lines, with acquired in vitro-induced vemurafenib resistance, show increased levels of glioma-associated oncogene homolog 1 and 2 (GLI1/GLI2) compared with naïve cells. We also observed these findings in clinical melanoma specimens. Moreover, the increased expression of the transcription factors GLI1/GLI2 was independent of canonical Hh signaling and was instead correlated with the noncanonical Hh pathway, involving TGFß/SMAD (transforming growth factor-ß/Sma- and Mad-related family) signaling. Knockdown of GLI1 and GLI2 restored sensitivity to vemurafenib-resistant cells, an effect associated with both growth arrest and senescence. Treatment of vemurafenib-resistant cells with the GLI1/GLI2 inhibitor Gant61 led to decreased invasion of the melanoma cells in a three-dimensional skin reconstruct model and was associated with a decrease in metalloproteinase (MMP2/MMP9) expression and microphthalmia transcription factor upregulation. Gant61 monotherapy did not alter the drug sensitivity of naïve cells, but could reverse the resistance of melanoma cells chronically treated with vemurafenib. We further noted that alternating dosing schedules of Gant61 and vemurafenib prevented the onset of BRAFi resistance, suggesting that this could be a potential therapeutic strategy for the prevention of therapeutic escape. Our results suggest that targeting the Hh pathway in BRAFi-resistant melanoma may represent a viable therapeutic strategy to restore vemurafenib sensitivity, reducing or even inhibiting the acquired chemoresistance in melanoma patients.


Assuntos
Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos , Indóis/farmacologia , Fatores de Transcrição Kruppel-Like/antagonistas & inibidores , Melanoma/metabolismo , Sulfonamidas/farmacologia , Proteína GLI1 em Dedos de Zinco/antagonistas & inibidores , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Senescência Celular/genética , Expressão Gênica , Técnicas de Silenciamento de Genes , Proteínas Hedgehog/metabolismo , Humanos , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Melanoma/tratamento farmacológico , Melanoma/genética , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Vemurafenib , Proteína GLI1 em Dedos de Zinco/genética , Proteína GLI1 em Dedos de Zinco/metabolismo , Proteína Gli2 com Dedos de Zinco
2.
Oncogene ; 30(20): 2304-6, 2011 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-21278792

RESUMO

The Mitf gene has a key role in melanocytes and melanoma by regulating cell cycle progression, survival and differentiation. Two papers in this issue of Oncogene (Cheli et al., 2011; Strub et al., 2011) reveal that low-Mitf cells can initiate tumors with high efficiency, and that Mitf blocks senescence by regulating genes implicated in S-phase progression and mitosis.


Assuntos
Senescência Celular/genética , Melanoma/genética , Fator de Transcrição Associado à Microftalmia/genética , Neoplasias Cutâneas/genética , Ciclo Celular/genética , Diferenciação Celular/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Melanócitos/metabolismo , Melanoma/mortalidade , Células-Tronco Neoplásicas/fisiologia , Neoplasias Cutâneas/mortalidade
3.
Oncogene ; 27(7): 976-84, 2008 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-17700536

RESUMO

T-box factors play critical roles in embryonic development and have been implicated in cell cycle regulation and cancer. For example, Tbx2 can suppress senescence through a mechanism involving the repression of the cyclin-dependent kinase inhibitors, p19(ARF) and p21(WAF1/CIP1/SDII), and the Tbx2 gene is deregulated in melanoma, breast and pancreatic cancers. In this study, several transformed human lung fibroblast cell lines were shown to downregulate Tbx2. To further investigate the role of Tbx2 in oncogenesis we therefore stably reexpressed Tbx2 in one such cell line. Compared to their parental cells, the resulting Tbx2-expressing cells are larger, with binucleate and lobular nuclei containing double the number of chromosomes. Moreover, these cells had an increase in frequency of several features of genomic instability such as chromosome missegregation, chromosomal rearrangements and polyploidy. While grossly abnormal, these cells still divide and give rise to cells that are resistant to the chemotherapeutic drug cisplatin. Furthermore, this is shown to be neither species nor cell type dependent, as ectopically expressing Tbx2 in a murine melanoma cell line also induce mitotic defects and polyploidy. These results have important implications for our understanding of the role of Tbx2 in tumorigenesis because polyploidy frequently precedes aneuploidy, which is associated with high malignancy and poor prognosis.


Assuntos
Antineoplásicos/farmacologia , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos , Poliploidia , Proteínas com Domínio T/metabolismo , Animais , Western Blotting , Proliferação de Células/efeitos dos fármacos , Aberrações Cromossômicas , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Citometria de Fluxo , Instabilidade Genômica , Humanos , Pulmão , Melanoma/genética , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Mitose , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas com Domínio T/genética
4.
Nucleic Acids Res ; 33(16): 5235-42, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16166655

RESUMO

Interaction with Max via the helix-loop-helix/leucine zipper (HLH-LZ) domain is essential for Myc to function as a transcription factor. Myc is commonly upregulated in tumours, however, its activity can also be potentiated by virally derived mutations. vMyc, derived from the virus, MC29 gag-Myc, differs from its cellular counterpart by five amino acids. The N-terminal mutation stabilizes the protein, however, the significance of the other mutations is not known. We now show that vMyc can sustain longer deletions in the LZ domain than cMyc before complete loss in transforming activity, implicating the viral mutations in contributing to Myc:Max complex formation. We confirmed this both in vitro and in vivo, with loss of Max binding correlating with a loss in the biological activity of Myc. A specific viral mutation, isoleucine383>leucine (I383>L) in helix 2 of the HLH domain, extends the LZ domain from four to five heptad repeats. Significantly, introduction of I383>L into a Myc mutant that is defective for Max binding substantially restored its ability to complex with Max in vitro and in vivo. We therefore propose that this virally derived mutation is functional by significantly contributing to establishing a more hydrophobic interface between the LZs of Myc and Max.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteína Oncogênica p55(v-myc)/genética , Proteína Oncogênica p55(v-myc)/metabolismo , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Animais , Fatores de Transcrição de Zíper de Leucina Básica , Sítios de Ligação , Embrião de Galinha , Dimerização , Sequências Hélice-Alça-Hélice , Zíper de Leucina , Dados de Sequência Molecular , Mutação , Proteína Oncogênica p55(v-myc)/química , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-myc/química , Proteínas Proto-Oncogênicas c-myc/metabolismo , Deleção de Sequência
5.
Br J Cancer ; 89(6): 1072-8, 2003 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-12966428

RESUMO

Clear cell sarcoma (CCS) is associated with the EWS/ATF1 oncogene that is created by chromosomal fusion of the Ewings Sarcoma oncogene (EWS) and the cellular transcription factor ATF1. The melanocytic character of CCS suggests that the microphthalmia-associated transcription factor (Mitf), a major inducer of melanocytic differentiation, may be miss-expressed in CCS. Accordingly, we show that the mRNA and protein of the melanocyte-specific isoform of Mitf (Mitf-M) are present in several cultured CCS cell lines (Su-ccs-1, DTC1, Kao, MST-1, MST-2 and MST-3). The above cell lines thus provide a valuable experimental resource for examining the role of Mitf-M in both CCS and melanocyte differentiation. Melanocyte-specific expression of Mitf-M is achieved via an ATF-dependent melanocyte-specific cAMP-response element in the Mitf-M promoter, and expression of Mitf-M in CCS cells suggests that EWS/ATF1 (a potent and promiscuous activator of cAMP-inducible promoters) may activate the Mitf-M promoter. Surprisingly, however, the Mitf-M promoter is not activated by EWS/ATF1 in transient assays employing CCS cells, melanocytes or nonmelanocytic cells. Thus, our results indicate that Mitf-M promoter activation may require an appropriate chromosomal context in CCS cells or alternatively that the Mitf-M promoter is not directly activated by EWS/ATF1.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Regulação Neoplásica da Expressão Gênica , Sarcoma de Células Claras/metabolismo , Sarcoma/metabolismo , Fatores de Transcrição/metabolismo , Western Blotting , Diferenciação Celular , Cloranfenicol O-Acetiltransferase/metabolismo , Primers do DNA , Proteínas de Ligação a DNA/genética , Humanos , Zíper de Leucina , Melanócitos/metabolismo , Fator de Transcrição Associado à Microftalmia , Proteínas de Neoplasias/genética , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Plasmídeos , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas/genética , Isoformas de Proteínas , RNA Neoplásico/análise , Sarcoma/genética , Sarcoma/patologia , Sarcoma de Células Claras/genética , Sarcoma de Células Claras/patologia , Fatores de Transcrição/genética , Transcrição Gênica , Células Tumorais Cultivadas
6.
EMBO J ; 20(17): 5022-31, 2001 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-11532965

RESUMO

The stress-activated signalling cascade leading to phosphorylation of the p38 family of kinases plays a crucial role during development and in the cellular response to a wide variety of stress-inducing agents. Although alterations in gene expression characteristic of the stress response require the regulation of key transcription factors by the p38 family, few downstream targets for this signalling pathway have been identified. By examining the ability of pigment cells to respond to UV irradiation as part of the UV-induced tanning response, we show that while the microphthalmia-associated transcription factor Mitf regulates basal Tyrosinase expression, it is the ubiquitous basic helix-loop-helix-leucine zipper transcription factor Usf-1 that is required for the UV activation of the Tyrosinase promoter. Consistent with this we demonstrate that Usf-1 is phosphorylated and activated by the stress-responsive p38 kinase. The results suggest that activation of Usf-1 by p38 at a wide variety of viral and cellular promoters will provide a link between stimuli as diverse as UV irradiation, glucose, viral infection and pro-inflammatory cytokines, and the changes in gene expression associated with the stress response.


Assuntos
Proteínas de Ligação a DNA , Regulação Enzimológica da Expressão Gênica/efeitos da radiação , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Monofenol Mono-Oxigenase/genética , Fatores de Transcrição/metabolismo , Raios Ultravioleta , Células 3T3 , Animais , Células COS , Chlorocebus aethiops , Cromatina/fisiologia , Genes Reporter , Glutationa Transferase/genética , Sequências Hélice-Alça-Hélice , Humanos , Melanócitos/citologia , Melanócitos/metabolismo , Melanócitos/efeitos da radiação , Melanoma , Camundongos , Regiões Promotoras Genéticas/efeitos da radiação , Proteínas Recombinantes de Fusão/análise , Transfecção , Células Tumorais Cultivadas , Fatores Estimuladores Upstream , Proteínas Quinases p38 Ativadas por Mitógeno
7.
Forum (Genova) ; 10(3): 176-87, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11007928

RESUMO

Malignant melanoma is a notoriously aggressive disease that can affect relatively young individuals and whose incidence is rising at an alarming rate. Unlike many cancers, metastatic melanoma is poorly responsive to current therapies and mutations affecting p53, the retinoblastoma gene product or Ras which occur frequently in many other cancer types, appear to be rare or at least relatively late events in the progression of the disease. Recent advances in our understanding of the disease at the molecular level have indicated that in addition to the loss of cell cycle checkpoints which may be common to all cancers, malignant melanoma shares many characteristics in common with developmental precursors to melanocytes, the mature pigment producing cells of the skin and hair follicles which are responsible for skin and hair colour. This review therefore focuses on the signalling pathways that play a crucial role in the development of the melanocyte lineage which are subject to deregulation in malignant melanoma namely signalling by receptor tyrosine kinases, the Wnt signalling pathway, as well as loss of the p16INK4a cyclin-dependent kinase inhibitor. Intriguingly all three pathways impact on the expression or function of the microphthalmia-associated transcription factor which plays an essential role in melanocyte development.


Assuntos
Melanócitos , Melanoma/genética , Animais , Proteínas de Ligação a DNA , Regulação Neoplásica da Expressão Gênica , Melanócitos/metabolismo , Melanócitos/patologia , Melanoma/metabolismo , Melanoma/patologia , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Mutantes , Mutação , Metástase Neoplásica , Fenótipo , Transdução de Sinais , Fatores de Transcrição/genética , Transcrição Gênica
8.
J Biol Chem ; 275(48): 37978-83, 2000 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-10973953

RESUMO

The transcription factor Sox10 is genetically linked with Waardenburg syndrome 4 (WS4) in humans and the Dominant megacolon (Dom) mouse model for this disease. The pigmentary defects observed in the Dom mouse and WS4 are reminiscent of those associated with mutations in the microphthalmia (Mitf) gene, which encodes a transcription factor essential for the development of the melanocyte lineage. We demonstrate here that wild type Sox10 directly binds and activates transcription of the MITF promoter, whereas a mutant form of the Sox10 protein genetically linked with WS4 acts as a dominant-negative repressor of MITF expression and can reduce endogenous MITF protein levels. The ability of Sox10 to activate transcription of the MITF promoter implicates Sox10 in the regulation of melanocyte development and provides a molecular basis for the hypopigmentation and deafness associated with WS4.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Surdez/genética , Regulação da Expressão Gênica/fisiologia , Proteínas de Grupo de Alta Mobilidade/fisiologia , Transtornos da Pigmentação/genética , Regiões Promotoras Genéticas , Fatores de Transcrição , Síndrome de Waardenburg/genética , Animais , Sequência de Bases , Primers do DNA , Camundongos , Fator de Transcrição Associado à Microftalmia , Fatores de Transcrição SOXE , Células Tumorais Cultivadas
10.
J Biol Chem ; 275(29): 21920-7, 2000 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-10770922

RESUMO

Commitment to the melanocyte lineage is characterized by the onset of microphthalmia-associated transcription factor (Mitf) expression. Mitf plays a fundamental role in melanocyte development, with mice lacking Mitf being entirely devoid of pigment cells. In the absence of functional Mitf protein, melanoblasts expressing Mitf mRNA disappear around 2 days after their first appearance either by apoptosis or by losing their identity and adopting an alternative cell fate. The role of Mitf must therefore be to regulate genes required for melanoblast survival, proliferation, or the maintenance of melanoblast identity. Yet to date, Mitf has been shown to regulate genes such as Tyrosinase, Tyrp-1, and Dct, which are required for pigmentation, a differentiation-specific process. Because expression of these genes cannot account for the complete absence of pigment cells in Mitf-negative mice, Mitf must regulate the expression of other as yet uncharacterized genes. Here we provide several lines of evidence to suggest that Mitf may regulate the expression of the Tbx2 transcription factor, a member of the T-box family of proteins implicated in the maintenance of cell identity. First, isolation and sequencing of the entire murine Tbx2 gene revealed that the Tbx2 promoter contains a full consensus Mitf recognition element; second, Mitf could bind the promoter in vitro and activate Tbx2 expression in vivo in an E box-dependent fashion; and third, Tbx2 is expressed in melanoma cell lines expressing Mitf, but not in a line in which Mitf expression was not detectable. Taken together, with the fact that Tbx2 is expressed in Mitf-positive melanoblasts and melanocytes, but not in Mitf-negative melanoblast precursor cells, the evidence suggests that the Tbx2 gene may represent one of the first known targets for Mitf that is not a gene involved directly in the manufacture of pigment.


Assuntos
Proteínas de Ligação a DNA/genética , Melanócitos/fisiologia , Proteínas com Domínio T/genética , Fatores de Transcrição , Animais , Sequência de Bases , Células COS , Linhagem da Célula/genética , Regulação da Expressão Gênica no Desenvolvimento , Células HeLa , Humanos , Melanócitos/citologia , Camundongos , Fator de Transcrição Associado à Microftalmia , Dados de Sequência Molecular , Regiões Promotoras Genéticas
11.
J Biol Chem ; 274(38): 26894-900, 1999 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-10480898

RESUMO

Previous work has established that the melanocyte-specific tyrosinase-related protein-1 (TRP-1) promoter is regulated positively by the microphthalmia-associated transcription factor Mitf, acting through the conserved M box and negatively by the T-box factor Tbx2, which can bind two "melanocyte-specific elements" termed the MSEu and MSEi. Both the MSEu and MSEi, which share a 6-base pair GTGTGA consensus, are also recognized by a previously unidentified melanocyte-specific factor, MSF. Here we show using a combination of DNA binding assays, proteolytic clipping, and anti-Pax3 antibodies that MSF is indistinguishable from Pax3, a paired homeodomain transcription factor implicated genetically in melanocyte development and the regulation of the Mitf promoter. Consistent with Pax3 being able to bind the TRP-1 promoter, Pax3 is expressed in melanocytes and melanomas, and TRP-1 promoter activity is up-regulated by Pax3. The results identify a novel role for Pax3 in the expression of TRP-1, and the potential role of Pax3 in the melanocyte lineage is discussed.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Regulação da Expressão Gênica , Glicoproteínas de Membrana , Oxirredutases , Regiões Promotoras Genéticas , Proteínas/genética , Fatores de Transcrição , Animais , Sequência de Bases , Diferenciação Celular , Melanócitos/citologia , Melanócitos/metabolismo , Melanoma/metabolismo , Camundongos , Dados de Sequência Molecular , Fator de Transcrição PAX3 , Fatores de Transcrição Box Pareados , Transfecção , Células Tumorais Cultivadas
12.
Mol Cell Biol ; 18(12): 6930-8, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9819381

RESUMO

The development of melanocytes, which are pigment-producing cells responsible for skin, hair, and eye color, is absolutely dependent on the action of the microphthalmia basic helix-loop-helix-leucine zipper (bHLH-LZ) transcription factor (Mi); mice lacking a functional Mi protein are entirely devoid of pigment cells. Mi has been shown to activate transcription of the tyrosinase, TRP-1, TRP-2, and QNR-71 genes through specific E-box elements, most notably the highly conserved M box. We investigated the mechanism which enables Mi to be recruited specifically to a restricted subset of E boxes in target promoters while being prevented from binding E-box elements in other promoters. We show both in vitro and in vivo that the presence of a T residue flanking a CATGTG E box is an essential determinant of the ability of Mi to bind DNA, and we successfully predict that the CATGTG E box from the P gene would not bind Mi. In contrast, no specific requirement for the sequences flanking a CACGTG E box was observed, and no binding to an atypical E box in the c-Kit promoter was detected. The relevance of these observations to the control of melanocyte-specific gene expression was highlighted by the fact that the E-box elements located in the tyrosinase, TRP-1, TRP-2, and QNR-71 promoters without exception possess a 5' flanking T residue which is entirely conserved between species as diverse as man and turtle. The ability of Mi to discriminate between different E-box motifs provides a mechanism to restrict the repertoire of genes which are likely to be regulated by Mi and provides insight into the ability of bHLH-LZ transcription factors to achieve the specificity required for the precise coordination of transcription during development.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Sequências Hélice-Alça-Hélice/genética , Zíper de Leucina/genética , Melanócitos/metabolismo , Fatores de Transcrição/genética , Sequência Conservada/genética , Proteínas de Ligação a DNA/genética , Dimerização , Genes Reguladores/genética , Genes Reporter/genética , Humanos , Fator de Transcrição Associado à Microftalmia , Monofenol Mono-Oxigenase/genética , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas c-kit/genética , Fatores Estimuladores Upstream
13.
Mol Cell Biol ; 18(9): 5099-108, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9710594

RESUMO

Previous work has demonstrated that two key melanocyte-specific elements termed the MSEu and MSEi play critical roles in the expression of the melanocyte-specific tyrosinase-related protein 1 (TRP-1) promoter. Both the MSEu and MSEi, located at position -237 and at the initiator, respectively, bind a melanocyte-specific factor termed MSF but are also recognized by a previously uncharacterized repressor, since mutations affecting either of these elements result in strong up-regulation of TRP-1 promoter activity in melanoma cells. Here we demonstrate that repression mediated by the MSEu and MSEi also operates in melanocytes. We also report that both the MSEu and MSEi are recognized by the brachyury-related transcription factor Tbx2, a member of the recently described T-box family, and that Tbx2 is expressed in melanocyte and melanoblast cell lines but not in melanoblast precursor cells. Although Tbx2 and MSF each recognize the TRP-1 MSEu and MSEi motifs, it is binding by Tbx-2, not binding by MSF, that correlates with repression. Several lines of evidence tend to point to the brachyury-related transcription factor Tbx2 as being the repressor of TRP-1 expression: both the MSEu and MSEi bind Tbx2, and mutations in either element that result in derepression of the TRP-1 promoter diminish binding by Tbx2; the TRP-1 promoter, but not the tyrosinase, microphthalmia, or glyceraldehyde-3-phosphate dehydrogenase (G3PDH) promoter, is repressed by Tbx2 in cotransfection assays; a high-affinity consensus brachyury/Tbx2-binding site is able to constitutively repress expression of the heterologous IE110 promoter; and a low-affinity brachyury/Tbx2 binding site is able to mediate Tbx2-dependent repression of the G3PDH promoter. Although we cannot rule out the presence of an additional, as yet unidentified factor playing a role in the negative regulation of TRP-1 in vivo, the evidence presented here suggests that Tbx2 most likely is the previously unidentified repressor of TRP-1 expression and as such is likely to represent the first example of transcriptional repression by a T-box family member.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas Fetais , Melanócitos/metabolismo , Glicoproteínas de Membrana , Oxirredutases , Regiões Promotoras Genéticas , Biossíntese de Proteínas , Proteínas/genética , Proteínas com Domínio T , Animais , Sequência de Bases , Sítios de Ligação , Sequência Consenso , Primers do DNA , Proteínas de Ligação a DNA/biossíntese , Regulação da Expressão Gênica , Camundongos , Reação em Cadeia da Polimerase , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/metabolismo , Proteínas Repressoras/metabolismo , Reticulócitos/metabolismo , Especificidade por Substrato , Fatores de Transcrição/metabolismo , Transcrição Gênica , Transfecção
15.
Oncogene ; 14(25): 3083-92, 1997 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-9223672

RESUMO

The Microphthalmia basic-Helix-Loop-Helix-Leucine Zipper (bHLH-LZ) transcription factor (Mi) plays a crucial role in the genesis of melanocytes; mice deficient for a functional (Microphthalmia) gene product lack all pigment cells. We show here that the Mi activation domain resides N-terminal to the DNA-binding domain and that as little as 18 amino acids are sufficient to mediate transcription activation. The minimal activation region of Mi is highly conserved in the related transcription factor TFE3 and is predicted to adopt an amphipathic alpha-helical conformation. This region of Mi is also highly conserved with a region of E1A known to be essential for binding the CBP/p300 transcription cofactor. Consistent with these observations, the Mi activation domain can interact in vitro with CBP specifically through a region of CBP required for complex formation with E1A, P/CAF and c-Fos, and anti p300 antibodies can co-immunoprecipitate Mi from both melanocyte and melanoma cell lines. In addition, co-transfection of a vector expressing CBP2 (aas 1621-1891) fused to the VP16 activation domain potentiated the ability of Mi to activate transcription, confirming the significance of the CBP-Mi interaction observed in vitro. These data suggest that transcription activation by Mi is achieved at least in part by recruitment of CBP. The parallels between transcription regulation by Microphthalmia in melanocytes and MyoD in muscle cells are discussed.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Transativadores/metabolismo , Transcrição Gênica , Proteínas E1A de Adenovirus/metabolismo , Sequência de Aminoácidos , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Sítios de Ligação , Proteína de Ligação a CREB , Proteínas de Ligação a DNA/genética , Histona Acetiltransferases , Melanócitos/metabolismo , Camundongos , Fator de Transcrição Associado à Microftalmia , Dados de Sequência Molecular , Proteína MyoD/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Coativador 3 de Receptor Nuclear , Homologia de Sequência de Aminoácidos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
16.
Nucleic Acids Res ; 24(16): 3216-21, 1996 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-8774903

RESUMO

Chimaeric alleles were constructed to assay the biological functions of an N-terminal deletion and C-terminal mutations which were found in a naturally occurring mutant of feline vMyc, T17. The mutant alleles were assayed for their ability to transform chick embryo fibroblasts in vitro by a number of criteria, namely the ability to induce morphological transformation, an accelerated growth rate and growth in soft agar. Feline cMyc could transform the avian cells, whilst T17 vMyc could not, and the N-terminal deletion was responsible for conferring the primary transformation defect on the mutant protein. The C-terminal mutations which consist of a point mutation adjacent to the nuclear localisation signal and a point mutation/amino acid insertion within the basic region (BR) could, however, dissociate the Myc-induced parameters of transformation. This effect was a specific function of the BR mutation alone, and the mutation could be transferred into avian cMyc with comparable biological consequences. The BR mutation did not disrupt the sequence specific DNA binding activity of the protein in vivo, despite exerting a biological effect. These data suggest a novel phenotype where the mutation may affect a subset of Myc-regulated genes through altered DNA binding specificity or protein-protein interactions.


Assuntos
Transformação Celular Neoplásica/genética , Genes myc , Mutação , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Gatos , Embrião de Galinha , Proteínas de Ligação a DNA , Dados de Sequência Molecular , Proteína Oncogênica p55(v-myc)/genética , Fenótipo , Ligação Proteica , Estrutura Secundária de Proteína , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Recombinantes de Fusão , Deleção de Sequência
17.
Oncogene ; 11(10): 2157-64, 1995 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-7478537

RESUMO

Previous work has shown that melanoma cell lines express a distinct octamer binding protein. Given the role of octamer-binding proteins in cell differentiation and development, the role this factor is a key issue in understanding melanocyte differentiation and transformation. Using a proteolytic clipping assay, we show that the melanoma-specific octamer factor is Brn-2/N-Oct3, a POU domain protein previously known to be expressed in adult brain and in the developing nervous system. N-Oct3 mRNA was detected in a range of human melanoma cell lines and was around 10-fold elevated compared to normal human melanocytes while mRNA for Brn-2 was also detected in a mouse melanoblast cell line. Expression of Brn-2/N-Oct3, in melanoma cells in cotransfection assays activated the expression of the MHC class II DR alpha promoter but repressed the activity of the melanocyte-specific tyrosinase promoter. Repression correlated with Brn-2/N-Oct3 binding in a mutually exclusive fashion with basic-helix-loop-helix-leucine-zipper (bHLH-LZ) transcription factor USF in vitro and with Brn-2 expression preventing activation of the tyrosinase promoter by the bHLH-LZ factor Microphthalmia in vivo. The potential role of Brn-2/N-Oct3 in melanocyte differentiation and gene expression is discussed.


Assuntos
Melanócitos/fisiologia , Melanoma/genética , Melanoma/metabolismo , Fatores de Transcrição/biossíntese , Antígenos de Neoplasias , Sequência de Bases , Sítios de Ligação , DNA de Neoplasias/genética , DNA de Neoplasias/metabolismo , Regulação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Antígenos HLA-DR/genética , Sequências Hélice-Alça-Hélice/fisiologia , Proteínas de Homeodomínio , Humanos , Melanócitos/metabolismo , Antígenos Específicos de Melanoma , Dados de Sequência Molecular , Monofenol Mono-Oxigenase/genética , Monofenol Mono-Oxigenase/metabolismo , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Fatores do Domínio POU , Regiões Promotoras Genéticas/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição/genética , Transfecção , Células Tumorais Cultivadas
18.
Oncogene ; 10(1): 123-34, 1995 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-7824265

RESUMO

Little is known of the molecular mechanisms underlying the differentiation of the melanocyte from the melanoblast or the progression from the melanocyte to a malignant melanoma. Since the adenovirus E1A products have proved a useful tool for understanding control of differentiation in other systems, we explored the possibility of using E1A as a probe for factors controlling melanocyte-specific gene expression and differentiation. The results obtained show that the adenovirus E1A 13S, but not the 12S, product can transform the highly pigmented and TPA-dependent melanocyte cell line melan-a. Transformation is characterised by a morphological change, loss of TPA-dependence, the ability to grow in soft agar and strikingly, loss of pigmentation which correlates with loss of expression of the melanocyte-specific TRP-1 and tyrosinase genes. Cotransfection assays demonstrated that repression of TRP-1 by E1A correlated with E1A binding to p105Rb and p300, with the target in the TRP-1 promoter being the M-box, and 11 bp basic-Helix-loop-Helix (bHLH) factor-binding motif conserved between melanocyte-specific promoters. Consistent with the M-box acting as a target for E1a-mediated transcription repression, we also show that the basic-helix-loop-helix-leucine zipper (bHLH-LZ) protein (Mi) encoded by the microphthalmia gene (mi), which is required for pigment cell differentiation, is a positive acting transcription factor which can interact with the retinoblastoma product in vitro and activate the TRP-1 promoter. Moreover, expression of the mi gene was reduced around 50-fold in the non-pigmented E1a-transformed melan-a cells compared to the nontransformed melan-a cell line, with ectopic expression of Mi able to prevent repression of the tyrosinase and TRP-1 promoters in the presence of E1A. Mi therefore appears to play a crucial role in melanocyte-specific gene expression. The parallels between repression of myogenesis and muscle cell bHLH factors, and Mi and melanocyte differentiation are discussed.


Assuntos
Proteínas E1A de Adenovirus/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Melanócitos/metabolismo , Glicoproteínas de Membrana , Oxirredutases , Proteína do Retinoblastoma/metabolismo , Transativadores , Animais , Sequência de Bases , Diferenciação Celular , Linhagem Celular , Primers do DNA , Proteínas de Ligação a DNA/genética , Regulação para Baixo , Proteína p300 Associada a E1A , Melanócitos/enzimologia , Camundongos , Fator de Transcrição Associado à Microftalmia , Dados de Sequência Molecular , Monofenol Mono-Oxigenase/genética , Proteínas Nucleares/metabolismo , Proteínas/genética , Proteína do Retinoblastoma/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica
19.
Mol Cell Biol ; 14(5): 3494-503, 1994 May.
Artigo em Inglês | MEDLINE | ID: mdl-8164694

RESUMO

For a gene to be transcribed in a tissue-specific fashion, expression must be achieved in the appropriate cell type and also be prevented in other tissues. As an approach to understanding the regulation of tissue-specific gene expression, we have analyzed the requirements for melanocyte-specific expression of the tyrosinase-related protein 1 (TRP-1) promoter. Positive regulation of TRP-1 expression is mediated by both an octamer-binding motif and an 11-bp element, termed the M box, which is conserved between the TRP-1 and other melanocyte-specific promoters. We show here that, consistent with its ability to activate transcription in a non-tissue-specific fashion, the M box binds the basic-helix-loop-helix factor USF in vitro. With the use of a combination of site-directed mutagenesis and chimeric promoter constructs, additional elements involved in regulating TRP-1 expression were identified. These include the TATA region, which appears to contribute to the melanocyte specificity of the TRP-1 promoter. Mutational analysis also identified two repressor elements, one at the start site, the other located at -240, which function both in melanoma and nonmelanoma cells. In addition, a melanocyte-specific factor, MSF, binds to sites which overlap both repressor elements, with substitution mutations demonstrating that binding by MSF is not required for repression. Although a functional role for MSF has not been unequivocally determined, the location of its binding sites leads us to speculate that it may act as a melanocyte-specific antirepressor during transcription of the endogenous TRP-1 gene.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Expressão Gênica , Melanócitos/metabolismo , Glicoproteínas de Membrana , Oxirredutases , Regiões Promotoras Genéticas , Transcrição Gênica , Animais , Sequência de Bases , Linhagem Celular , Cloranfenicol O-Acetiltransferase/biossíntese , Cloranfenicol O-Acetiltransferase/metabolismo , Proteínas de Ligação a DNA/biossíntese , Humanos , Melanoma Experimental , Camundongos , Dados de Sequência Molecular , Mutagênese Insercional , Mutagênese Sítio-Dirigida , Sondas de Oligonucleotídeos , Biossíntese de Proteínas , Proteínas/genética , Transfecção , Células Tumorais Cultivadas
20.
EMBO J ; 12(13): 5075-82, 1993 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-8262050

RESUMO

The Myc oncoprotein has been implicated in control of cell growth, division and differentiation. Although Myc contains a bHLH-LZ motif, it fails to bind DNA alone but can do so by forming heterodimers with an unrelated bHLH-LZ protein, Max. Max homodimers and Myc-Max heterodimers share the ability to bind CACGTG or CATGTG elements. Current models, based on experimentally induced overexpression of Myc and Max in mammalian cells, propose that Max-Max homodimers repress while Myc-Max heterodimers activate transcription through CACGTG binding sites. The interpretation of the results using mammalian cells is complicated by the presence of numerous unrelated CACGTG binding transcription activators and the existence of two alternative Max dimerization partners, Mad and Mxi-1. Thus, the mechanism whereby overexpression of Max leads to transcriptional repression remains to be established. Using a yeast system we show that Max homodimers have the potential to activate transcription through CACGTG motifs. Activation by Max requires DNA binding and amino acids outside the bHLH-LZ domain but is reduced compared with activation by Myc-Max heterodimers. Moreover, transcriptional activation by Myc-Max heterodimers, but not Max-Max homodimers, is strongly inhibited in vivo by specific sequences flanking the core CACGTG binding motif, presumably reflecting reduced DNA binding affinity. These results suggest a mechanism for directing the Myc-Max complex to a specific subset of CACGTG-containing target genes.


Assuntos
Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Proteínas Proto-Oncogênicas c-myc/genética , Fatores de Transcrição/genética , Transcrição Gênica , Sequência de Aminoácidos , Sequência de Bases , Fatores de Transcrição de Zíper de Leucina Básica , Sequências Hélice-Alça-Hélice , Zíper de Leucina , Dados de Sequência Molecular , Proteínas Recombinantes , Sequências Reguladoras de Ácido Nucleico , Saccharomyces cerevisiae , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA