Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Oncogene ; 36(13): 1849-1861, 2017 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-27748762

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Indoles/farmacología , Factores de Transcripción de Tipo Kruppel/antagonistas & inhibidores , Melanoma/metabolismo , Sulfonamidas/farmacología , Proteína con Dedos de Zinc GLI1/antagonistas & inhibidores , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Senescencia Celular/genética , Expresión Génica , Técnicas de Silenciamiento del Gen , Proteínas Hedgehog/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Melanoma/tratamiento farmacológico , Melanoma/genética , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Vemurafenib , Proteína con Dedos de Zinc GLI1/genética , Proteína con Dedos de Zinc GLI1/metabolismo , Proteína Gli2 con Dedos de Zinc
2.
Oncogene ; 30(20): 2304-6, 2011 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-21278792

RESUMEN

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.


Asunto(s)
Senescencia Celular/genética , Melanoma/genética , Factor de Transcripción Asociado a Microftalmía/genética , Neoplasias Cutáneas/genética , Ciclo Celular/genética , Diferenciación Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Melanocitos/metabolismo , Melanoma/mortalidad , Células Madre Neoplásicas/fisiología , Neoplasias Cutáneas/mortalidad
3.
Oncogene ; 27(7): 976-84, 2008 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-17700536

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Resistencia a Antineoplásicos , Poliploidía , Proteínas de Dominio T Box/metabolismo , Animales , Western Blotting , Proliferación Celular/efectos de los fármacos , Aberraciones Cromosómicas , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Citometría de Flujo , Inestabilidad Genómica , Humanos , Pulmón , Melanoma/genética , Melanoma/metabolismo , Melanoma/patología , Ratones , Mitosis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas de Dominio T Box/genética
4.
Nucleic Acids Res ; 33(16): 5235-42, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16166655

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteína Oncogénica p55(v-myc)/genética , Proteína Oncogénica p55(v-myc)/metabolismo , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico , Sitios de Unión , Embrión de Pollo , Dimerización , Secuencias Hélice-Asa-Hélice , Leucina Zippers , Datos de Secuencia Molecular , Mutación , Proteína Oncogénica p55(v-myc)/química , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-myc/química , Proteínas Proto-Oncogénicas c-myc/metabolismo , Eliminación de Secuencia
5.
Br J Cancer ; 89(6): 1072-8, 2003 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-12966428

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica , Sarcoma de Células Claras/metabolismo , Sarcoma/metabolismo , Factores de Transcripción/metabolismo , Western Blotting , Diferenciación Celular , Cloranfenicol O-Acetiltransferasa/metabolismo , Cartilla de ADN , Proteínas de Unión al ADN/genética , Humanos , Leucina Zippers , Melanocitos/metabolismo , Factor de Transcripción Asociado a Microftalmía , Proteínas de Neoplasias/genética , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Plásmidos , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas/genética , Isoformas de Proteínas , ARN Neoplásico/análisis , Sarcoma/genética , Sarcoma/patología , Sarcoma de Células Claras/genética , Sarcoma de Células Claras/patología , Factores de Transcripción/genética , Transcripción Genética , Células Tumorales Cultivadas
6.
EMBO J ; 20(17): 5022-31, 2001 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-11532965

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN , Regulación Enzimológica de la Expresión Génica/efectos de la radiación , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Monofenol Monooxigenasa/genética , Factores de Transcripción/metabolismo , Rayos Ultravioleta , Células 3T3 , Animales , Células COS , Chlorocebus aethiops , Cromatina/fisiología , Genes Reporteros , Glutatión Transferasa/genética , Secuencias Hélice-Asa-Hélice , Humanos , Melanocitos/citología , Melanocitos/metabolismo , Melanocitos/efectos de la radiación , Melanoma , Ratones , Regiones Promotoras Genéticas/efectos de la radiación , Proteínas Recombinantes de Fusión/análisis , Transfección , Células Tumorales Cultivadas , Factores Estimuladores hacia 5' , Proteínas Quinasas p38 Activadas por Mitógenos
7.
Mol Microbiol ; 38(2): 411-22, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11069666

RESUMEN

Saccharomyces cerevisiae strains lacking a functional Pho85 cyclin-dependent kinase (cdk) exhibit a complex phenotype, including deregulation of phosphatase genes controlled by the transcription factor Pho4, slow growth on rich media, failure to grow using galactose, lactate or glycerol as a carbon source and hyperaccumulation of glycogen. The ability of Pho85 to regulate the transcription factor Pho4 is mediated by its association the Pho80 cyclin. Some other regulatory functions of the Pho85 cdk have been shown to be mediated via its interaction with a recently identified family of Pho80-related cyclins (Pcls). Here, we show that the poorly characterized Pho80-like protein Pcl7 forms a functional kinase complex with the Pho85 cdk, and that the activity of this complex is inhibited in response to phosphate starvation. Additionally, we show that Pcl7 interacts with the phosphate-regulated cyclin-cdk inhibitor Pho81, and that the regulation of the Pcl7-Pho85 complex in response to changes in phosphate levels is dependent on Pho81. Thus, we demonstrate for the first time that the Pho81 regulator is not dedicated to regulating Pho80, but may act to co-ordinate the activity of both the Pho80-Pho85 and Pcl7-Pho85 cyclin-cdk complexes in response to phosphate levels. We also demonstrate that expression of Pcl7 is cell cycle regulated, with maximal activity occurring in mid to late S-phase, perhaps suggesting a role for Pcl7 in cell cycle progression. Finally, we describe the phenotype of pcl7Delta and pcl6Delta yeast strains that have defects in carbon source utilization.


Asunto(s)
Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , Inhibidores Enzimáticos/metabolismo , Proteínas Fúngicas/metabolismo , Proteínas Represoras , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae/metabolismo , Secuencia de Aminoácidos , Ciclo Celular , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/genética , Ciclinas/genética , Proteínas Fúngicas/genética , Datos de Secuencia Molecular , Fenotipo , Fosfatos/metabolismo , Saccharomyces cerevisiae/genética
8.
Forum (Genova) ; 10(3): 176-87, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11007928

RESUMEN

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.


Asunto(s)
Melanocitos , Melanoma/genética , Animales , Proteínas de Unión al ADN , Regulación Neoplásica de la Expresión Génica , Melanocitos/metabolismo , Melanocitos/patología , Melanoma/metabolismo , Melanoma/patología , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Ratones Mutantes , Mutación , Metástasis de la Neoplasia , Fenotipo , Transducción de Señal , Factores de Transcripción/genética , Transcripción Genética
9.
J Biol Chem ; 275(48): 37978-83, 2000 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-10973953

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Sordera/genética , Regulación de la Expresión Génica/fisiología , Proteínas del Grupo de Alta Movilidad/fisiología , Trastornos de la Pigmentación/genética , Regiones Promotoras Genéticas , Factores de Transcripción , Síndrome de Waardenburg/genética , Animales , Secuencia de Bases , Cartilla de ADN , Ratones , Factor de Transcripción Asociado a Microftalmía , Factores de Transcripción SOXE , Células Tumorales Cultivadas
11.
J Biol Chem ; 275(29): 21920-7, 2000 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-10770922

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/genética , Melanocitos/fisiología , Proteínas de Dominio T Box/genética , Factores de Transcripción , Animales , Secuencia de Bases , Células COS , Linaje de la Célula/genética , Regulación del Desarrollo de la Expresión Génica , Células HeLa , Humanos , Melanocitos/citología , Ratones , Factor de Transcripción Asociado a Microftalmía , Datos de Secuencia Molecular , Regiones Promotoras Genéticas
12.
Gene ; 259(1-2): 159-70, 2000 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-11163973

RESUMEN

Tyrosinase is the key enzyme required for the synthesis of melanin pigments. Sequence comparison and functional analysis of the 5' upstream regions of vertebrate tyrosinase genes have revealed the importance of conserved E-box motifs in regulating their specific expression in pigment cells, optic cup-derived retinal pigment epithelium (RPE) and neural crest-derived melanocytes. In ascidians (more basal protochordates), two pigment cells that resemble vertebrate RPE cells are formed and specifically express the orthologous tyrosinase gene (HrTyr) in the cerebral vesicle located at the anterior end of the neural tube. To define regulatory sequences required for pigment cell-lineage-specific expression of HrTyr during embryogenesis, a series of mutations of the 5' upstream region of HrTyr were fused to the lacZ reporter gene and were microinjected into fertilized eggs. We found that the -152bp upstream of the translational start site is essential for expression in pigment cell precursors of tailbud-stage embryos. Further, additional positive and unique restriction elements were identified in the region up to -1.8kb. Surprisingly, in the -152bp minimal promoter or in other regions with regulatory activities, there are no E-box motifs or sequences correlating with other conserved elements regulating vertebrate tyrosinase promoters. The possibility that Pax proteins regulate HrTyr expression is also discussed.


Asunto(s)
Melanocitos/enzimología , Monofenol Monooxigenasa/genética , Epitelio Pigmentado Ocular/enzimología , Urocordados/genética , Vertebrados/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Secuencia Conservada , ADN/química , ADN/genética , Precursores Enzimáticos/genética , Regulación Enzimológica de la Expresión Génica , Operón Lac/genética , Melanocitos/citología , Datos de Secuencia Molecular , Epitelio Pigmentado Ocular/citología , Regiones Promotoras Genéticas/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética , Alineación de Secuencia , Análisis de Secuencia de ADN , Eliminación de Secuencia , Homología de Secuencia de Ácido Nucleico , Urocordados/embriología , Urocordados/enzimología
13.
J Biol Chem ; 274(38): 26894-900, 1999 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-10480898

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Regulación de la Expresión Génica , Glicoproteínas de Membrana , Oxidorreductasas , Regiones Promotoras Genéticas , Proteínas/genética , Factores de Transcripción , Animales , Secuencia de Bases , Diferenciación Celular , Melanocitos/citología , Melanocitos/metabolismo , Melanoma/metabolismo , Ratones , Datos de Secuencia Molecular , Factor de Transcripción PAX3 , Factores de Transcripción Paired Box , Transfección , Células Tumorales Cultivadas
14.
Hum Mol Genet ; 8(8): 1431-41, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10400990

RESUMEN

Waardenburg syndrome type 2 (WS2) is an autosomal dominant disorder characterized by a combination of pigmentary and auditory abnormalities. Approximately 20% of WS2 cases are associated with mutations in the gene encoding microphthalmia-associated transcription factor (MITF). MITF plays a critical role in the development of both neural-crest-derived melanocytes and optic cup-derived retinal pigmented epithelium (RPE); the loss of a functional Mitf in mice results in complete absence of all pigment cells, which in turn induces microphthalmia and inner ear deafness. The black-eyed white Mitf mi-bw homozygous mouse normally has a pigmented RPE but lacks melanocytes essential for the pigmentation of the body and hearing. We show here that Mitf mi-bw is caused by an insertion into intron 3 of a 7.2 kb novel L1 element, L1bw, which belongs to an actively retrotransposing TF subfamily. The L1bw insertion reduces the amount of mRNAs for two Mitf isoforms, Mitf-A and Mitf-H, by affecting their overall expression levels and pre-mRNA splicing patterns, while it abolishes mRNA expression of another isoform, Mitf-M, which is specifically expressed in neural-crest-derived melanocytes. The consequence of the L1 insertion in the black-eyed white Mitf mi-bw mouse is that the developmental programme for RPE cells proceeds normally, most likely because of the presence of residual, full-length Mitf-A and Mitf-H proteins, whereas the lack of Mitf-M results in loss of the melanocyte population. The results suggest that melanocyte development depends critically on a single Mitf isoform, Mitf-M, and raise the possibility that specific mutations affecting MITF-M, the human equivalent of Mitf-M, may be responsible for a subset of WS2 conditions.


Asunto(s)
Proteínas de Unión al ADN/genética , Intrones/genética , Retroelementos , Factores de Transcripción , Alelos , Animales , Secuencia de Bases , Femenino , Regulación de la Expresión Génica , Variación Genética , Pérdida Auditiva Sensorineural/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Mutantes , Factor de Transcripción Asociado a Microftalmía , Datos de Secuencia Molecular , Mutagénesis Insercional , Fenotipo , Trastornos de la Pigmentación/genética , Isoformas de Proteínas/genética , Pigmentación de la Piel/genética , Síndrome de Waardenburg/genética
15.
Mol Cell Biol ; 18(12): 6930-8, 1998 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9819381

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Secuencias Hélice-Asa-Hélice/genética , Leucina Zippers/genética , Melanocitos/metabolismo , Factores de Transcripción/genética , Secuencia Conservada/genética , Proteínas de Unión al ADN/genética , Dimerización , Genes Reguladores/genética , Genes Reporteros/genética , Humanos , Factor de Transcripción Asociado a Microftalmía , Monofenol Monooxigenasa/genética , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-kit/genética , Factores Estimuladores hacia 5'
16.
Mol Cell Biol ; 18(10): 5818-27, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9742099

RESUMEN

Perhaps the best characterized example of an activator-induced chromatin transition is found in the activation of the Saccharomyces cerevisiae acid phosphatase gene PHO5 by the basic helix-loop-helix (bHLH) transcription factor Pho4. Transcription activation of the PHO5 promoter by Pho4 is accompanied by the remodeling of four positioned nucleosomes which is dependent on the Pho4 activation domain but independent of transcription initiation. Whether the requirements for transcription activation through the TATA sequence are different from those necessary for the chromatin transition remains a major outstanding question. In an attempt to understand better the ability of Pho4 to activate transcription and to remodel chromatin, we have initiated a detailed characterization of the Pho4 activation domain. Using both deletion and point mutational analysis, we have defined residues between positions 75 and 99 as being both essential and sufficient to mediate transcription activation. Significantly, there is a marked concordance between the ability of mutations in the Pho4 activation domain to induce chromatin opening and transcription activation. Interestingly, the requirements for transcription activation within the Pho4 activation domain differ significantly if fused to a heterologous bHLH-leucine zipper DNA-binding domain. The implications for transcription activation by Pho4 are discussed.


Asunto(s)
Cromatina , Proteínas de Unión al ADN/fisiología , Proteínas Fúngicas/fisiología , Secuencias Hélice-Asa-Hélice , Proteínas de Saccharomyces cerevisiae , Factores de Transcripción/fisiología , Activación Transcripcional , Secuencia de Aminoácidos , Aminoácidos , Sitios de Unión , Dicroismo Circular , ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Datos de Secuencia Molecular , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
17.
Mol Cell Biol ; 18(9): 5099-108, 1998 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9710594

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas Fetales , Melanocitos/metabolismo , Glicoproteínas de Membrana , Oxidorreductasas , Regiones Promotoras Genéticas , Biosíntesis de Proteínas , Proteínas/genética , Proteínas de Dominio T Box , Animales , Secuencia de Bases , Sitios de Unión , Secuencia de Consenso , Cartilla de ADN , Proteínas de Unión al ADN/biosíntesis , Regulación de la Expresión Génica , Ratones , Reacción en Cadena de la Polimerasa , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/metabolismo , Proteínas Represoras/metabolismo , Reticulocitos/metabolismo , Especificidad por Sustrato , Factores de Transcripción/metabolismo , Transcripción Genética , Transfección
19.
Oncogene ; 14(25): 3083-92, 1997 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-9223672

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Transactivadores/metabolismo , Transcripción Genética , Proteínas E1A de Adenovirus/metabolismo , Secuencia de Aminoácidos , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Sitios de Unión , Proteína de Unión a CREB , Proteínas de Unión al ADN/genética , Histona Acetiltransferasas , Melanocitos/metabolismo , Ratones , Factor de Transcripción Asociado a Microftalmía , Datos de Secuencia Molecular , Proteína MioD/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Coactivador 3 de Receptor Nuclear , Homología de Secuencia de Aminoácido , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
20.
J Immunol ; 159(12): 6176-83, 1997 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-9550420

RESUMEN

Activation of complement gene expression plays a major role in the response to antigenic challenge. The induction of complement synthesis occurs primarily in liver and in macrophages and is mediated, at least in part, by increased transcription of the complement genes. For example, transcription of the C4 complement gene, which plays a crucial role in the complement pathway, is induced in response to acute inflammation or tissue injury. Previous work has defined the elements present in the C4 complement gene promoter that are required for its expression. Particularly important is an E-box motif, E-C4, that is conserved between the mouse, human, and rat promoters and that directed up to 90% of transcription from the mouse C4 promoter. Here we have purified the E-C4-binding factor to homogeneity using a novel and rapid affinity purification procedure. Following N-terminal microsequencing and subsequent isolation of the corresponding cDNA, the factor binding the E-C4 element was identified as upstream stimulatory factor-1 (USF-1), a basic helix-loop-helix-leucine zipper transcription factor. We also show for the first time that in vivo USF-1 is a phosphoprotein, but that phosphorylation of USF-1 is severely reduced in cells in culture. Moreover, the phosphorylated form of USF-1 binds DNA preferentially, indicating that phosphorylation may enhance the ability of USF-1 to bind DNA. The implications of USF-1 phosphorylation for C4 complement gene expression and transcription regulation are discussed.


Asunto(s)
Proteínas Portadoras/genética , Complemento C4/genética , Proteínas de Unión al ADN , Regiones Promotoras Genéticas , Factores de Transcripción/genética , Células 3T3 , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Proteínas Portadoras/aislamiento & purificación , Proteínas Portadoras/metabolismo , Clonación Molecular , ADN Complementario/aislamiento & purificación , Humanos , Integrina alfaXbeta2 , Masculino , Ratones , Datos de Secuencia Molecular , Peso Molecular , Fosforilación , Ratas , Ratas Sprague-Dawley , Factores de Transcripción/aislamiento & purificación , Factores de Transcripción/metabolismo , Factores Estimuladores hacia 5'
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA