Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Mol Cancer ; 21(1): 125, 2022 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-35681235

RESUMEN

BACKGROUND: The dynamic epigenome and proteins specialized in the interpretation of epigenetic marks critically contribute to leukemic pathogenesis but also offer alternative therapeutic avenues. Targeting newly discovered chromatin readers involved in leukemogenesis may thus provide new anticancer strategies. Accumulating evidence suggests that the PRC1 complex member CBX2 is overexpressed in solid tumors and promotes cancer cell survival. However, its role in leukemia is still unclear. METHODS: We exploited reverse genetic approaches to investigate the role of CBX2 in human leukemic cell lines and ex vivo samples. We also analyzed phenotypic effects following CBX2 silencing using cellular and molecular assays and related functional mechanisms by ATAC-seq and RNA-seq. We then performed bioinformatic analysis of ChIP-seq data to explore the influence of histone modifications in CBX2-mediated open chromatin sites. Lastly, we used molecular assays to determine the contribution of CBX2-regulated pathways to leukemic phenotype. RESULTS: We found CBX2 overexpressed in leukemia both in vitro and ex vivo samples compared to CD34+ cells. Decreased CBX2 RNA levels prompted a robust reduction in cell proliferation and induction of apoptosis. Similarly, sensitivity to CBX2 silencing was observed in primary acute myeloid leukemia samples. CBX2 suppression increased genome-wide chromatin accessibility followed by alteration of leukemic cell transcriptional programs, resulting in enrichment of cell death pathways and downregulation of survival genes. Intriguingly, CBX2 silencing induced epigenetic reprogramming at p38 MAPK-associated regulatory sites with consequent deregulation of gene expression. CONCLUSIONS: Our results identify CBX2 as a crucial player in leukemia progression and highlight a potential druggable CBX2-p38 MAPK network in AML.


Asunto(s)
Cromatina , Leucemia Mieloide Aguda , Complejo Represivo Polycomb 1 , Cromatina/genética , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 1/metabolismo , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
2.
Support Care Cancer ; 30(1): 585-591, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34347181

RESUMEN

Chemotherapy-induced nausea and vomiting (CINV) is one of the most frequent adverse events compromising quality of life (QoL) in patients undergoing autologous stem cell transplantation (ASCT). However, CINV prophylaxis is still lacking uniformity for high-dose melphalan (HDM), which is used to condition patients with multiple myeloma (MM). Netupitant/palonosetron (NEPA) is administered with dexamethasone (DEXA) for CINV prevention in several chemotherapy regimens. Our study aims to assess the efficacy of NEPA, without DEXA, in preventing CINV in 106 adult patients with MM receiving HDM and ASCT. All patients had antiemetic prophylaxis with multiple doses of NEPA 1 h before the start of conditioning and after 72 h and 120 h. A complete response (CR) was observed in 99 (93%) patients at 120 h (overall phase). The percentage of patients with complete control was 93%. The CR rate during the acute phase was 94% (n = 100). During the delayed phase, the CR rate was 95% (n = 101). Grade 1 nausea and vomiting were experienced by 82% and 12% of the patients, respectively. Grade 2 nausea was reported in 18% and vomiting in 10% of patients. Our results showed, for the first time, that NEPA, without DEXA, was a well-tolerated and effective antiemetic option for MM patients receiving HDM followed by ASCT.


Asunto(s)
Antieméticos , Trasplante de Células Madre Hematopoyéticas , Mieloma Múltiple , Antieméticos/uso terapéutico , Dexametasona/uso terapéutico , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Melfalán/efectos adversos , Mieloma Múltiple/tratamiento farmacológico , Náusea/inducido químicamente , Náusea/tratamiento farmacológico , Náusea/prevención & control , Palonosetrón/uso terapéutico , Piridinas , Calidad de Vida , Quinuclidinas/uso terapéutico , Trasplante Autólogo , Vómitos/inducido químicamente , Vómitos/tratamiento farmacológico , Vómitos/prevención & control
3.
ACS Med Chem Lett ; 11(11): 2268-2276, 2020 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-33214839

RESUMEN

Histone deacetylase inhibitors (HDACi) have emerged as promising therapeutics for the treatment of neurodegeneration, cancer, and rare disorders. Herein, we report the development of a series of spiroindoline-based HDAC6 isoform-selective inhibitors based on the X-ray crystal studies of the hit 6a. We identified compound 6j as the most potent and selective hHDAC6 inhibitor of the series. Biological investigation of compounds 6b, 6h, and 6j demonstrated their antiproliferative activity against several cancer cell lines. Western blotting studies indicated that they were able to increase tubulin acetylation, without significant variation in histone acetylation state, and induced PARP cleavage indicating their apoptotic potential at the molecular level. 6j induced HDAC6-dependent pSTAT3 inhibition.

4.
Cancers (Basel) ; 12(7)2020 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-32674429

RESUMEN

The ubiquitin proteasome system (UPS) is the main cellular degradation machinery designed for controlling turnover of critical proteins involved in cancer pathogenesis, including hematological malignancies. UPS plays a functional role in regulating turnover of key proteins involved in cell cycle arrest, apoptosis and terminal differentiation. When deregulated, it leads to several disorders, including cancer. Several studies indicate that, in some subtypes of human hematological neoplasms such as multiple myeloma and Burkitt's lymphoma, abnormalities in the UPS made it an attractive therapeutic target due to pro-cancer activity. In this review, we discuss the aberrant role of UPS evaluating its impact in hematological malignancies. Finally, we also review the most promising therapeutic approaches to target UPS as powerful strategies to improve treatment of blood cancers.

5.
Cancers (Basel) ; 11(6)2019 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-31146471

RESUMEN

Glioblastoma (GBM), a high-grade glioma (WHO grade IV), is the most aggressive form of brain cancer. Available treatment options for GBM involve a combination of surgery, radiation and chemotherapy but result in a poor survival outcome. GBM is a high-vascularized tumor and antiangiogenic drugs are widely used in GBM therapy as adjuvants to control abnormal vasculature. Vasculogenic mimicry occurs in GBM as an alternative vascularization mechanism, providing a means whereby GBM can escape anti-angiogenic therapies. Here, using an in vitro tube formation assay on Matrigel®, we evaluated the ability of different histone deacetylase inhibitors (HDACis) to interfere with vasculogenic mimicry. We found that vorinostat (SAHA) and MC1568 inhibit tube formation by rat glioma C6 cells. Moreover, at sublethal doses for GBM cells, SAHA, trichostatin A (TSA), entinostat (MS275), and MC1568 significantly decrease tube formation by U87MG and by patient-derived human GBM cancer stem cells (CSCs). The reduced migration and invasion of HDACis-treated U87 cells, at least in part, may account for the inhibition of tube formation. In conclusion, our results indicate that HDACis are promising candidates for blocking vascular mimicry in GBM.

6.
Cell Death Dis ; 10(5): 338, 2019 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-31000698

RESUMEN

Leukemia is characterized by genetic and epigenetic mutations resulting in selection of cancer cells, which are unable to differentiate. Although genetic alterations are difficult to target, the epigenome is intrinsically dynamic and readily offers new therapeutic strategies. Thus, identifying cancer-specific context-dependent targets and unraveling their biological function may open up new therapeutic perspectives. Here we identify bromodomain-containing protein 9 (BRD9) as a critical target required in acute myeloid leukemia (AML). We show that BRD9 is overexpressed in AML cells including ex vivo primary blasts compared with CD34+ cells. By targeting BRD9 expression in AML, we observed an alteration in proliferation and survival, ultimately resulting in the induction of apoptosis. Intriguingly, genome-wide profiling revealed that BRD9 binds enhancer regions in a cell type-specific manner, regulating cell type-related processes. We unveil a novel BRD9-sustained STAT5 pathway activation via regulation of SOCS3 expression levels. Our findings identify a previously undescribed BRD9-STAT5 axis as critical for leukemia maintenance, suggesting BRD9 as a potential therapeutic target.


Asunto(s)
Cromatina/metabolismo , Leucemia Mieloide Aguda/patología , Factor de Transcripción STAT5/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Antígenos CD34/metabolismo , Proliferación Celular , Supervivencia Celular , Células HeLa , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Factor de Transcripción STAT5/antagonistas & inhibidores , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Transcriptoma , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor/antagonistas & inhibidores
7.
Oncogene ; 37(19): 2559-2572, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29467492

RESUMEN

Polycomb group (PcG) proteins regulate transcription, playing a key role in stemness and differentiation. Deregulation of PcG members is known to be involved in cancer pathogenesis. Emerging evidence suggests that CBX2, a member of the PcG protein family, is overexpressed in several human tumors, correlating with lower overall survival. Unraveling the mechanisms regulating CBX2 expression may thus provide a promising new target for anticancer strategies. Here we show that the HDAC inhibitor SAHA regulates CBX2 stability via a SUMO-triggered ubiquitin-mediated pathway in leukemia. We identify CBX4 and RNF4 as the E3 SUMO and E3 ubiquitin ligase, respectively, and describe the specific molecular mechanism regulating CBX2 protein stability. Finally, we show that CBX2-depleted leukemic cells display impaired proliferation, underscoring its critical role in regulating leukemia cell tumorogenicity. Our results show that SAHA affects CBX2 stability, revealing a potential SAHA-mediated anti-leukemic activity though SUMO2/3 pathway.


Asunto(s)
Inhibidores de Histona Desacetilasas/farmacología , Leucemia Mieloide Aguda/metabolismo , Complejo Represivo Polycomb 1/química , Vorinostat/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HL-60 , Humanos , Células K562 , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Ligasas/metabolismo , Proteínas Nucleares/metabolismo , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 1/metabolismo , Proteínas del Grupo Polycomb/metabolismo , Estabilidad Proteica/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/metabolismo
8.
Arch Toxicol ; 88(9): 1651-68, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25085708

RESUMEN

Alteration of the chromatin orchestra seems to play a critical role in cancer. In recent years, in-depth studies of epigenetic machinery and its deregulation have led to the development and use of a wide range of modulatory molecules directed not only at chromatin enzymes (histone acetyltransferases, histone deacetylases, histone methyltransferases, histone demethylases and DNA methyltransferases) but also toward the emerging class of chromatin-associated proteins, so-called "histone readers." Chromatin modifiers are attractive therapeutic targets for the development of new cancer therapies. Many are currently approved by the US Food and Drug Administration and used to treat different malignancies. Specifically, inhibitors of DNA methyltransferases, such as azacitidine and decitabine, have been approved for the treatment of myelodysplastic syndrome, while inhibitors of histone deacetylases, including vorinostat and romidepsin, have been approved for cutaneous T-cell lymphoma. The bromodomain and extra-terminal inhibitors JQ1, IBET762 and IBET151 have performed extremely well in preclinical settings, suggesting that they may be promising molecules for the treatment of some type of tumors. This review focuses on epidrugs and their possible application, with particular emphasis on their mechanism of action as well as their present status in clinical and preclinical trials.


Asunto(s)
Antineoplásicos/uso terapéutico , Drogas en Investigación/uso terapéutico , Epigénesis Genética/efectos de los fármacos , Modelos Biológicos , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Investigación Biomédica/tendencias , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Diseño de Fármacos , Descubrimiento de Drogas/tendencias , Drogas en Investigación/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo
9.
J Biol Chem ; 287(36): 30170-80, 2012 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-22787154

RESUMEN

The Y-box binding protein 1 (YB-1) belongs to the cold-shock domain protein superfamily, one of the most evolutionarily conserved nucleic acid-binding proteins currently known. YB-1 performs a wide variety of cellular functions, including transcriptional and translational regulation, DNA repair, drug resistance, and stress responses to extracellular signals. Inasmuch as the level of YB-1 drastically increases in tumor cells, this protein is considered to be one of the most indicative markers of malignant tumors. Here, we present evidence that ΔNp63α, the predominant p63 protein isoform in squamous epithelia and YB-1, can physically interact. Into the nucleus, ΔNp63α and YB-1 cooperate in PI3KCA gene promoter activation. Moreover, ΔNp63α promotes YB-1 nuclear accumulation thereby reducing the amount of YB-1 bound to its target transcripts such as that encoding the SNAIL1 protein. Accordingly, ΔNp63α enforced expression was associated with a reduction of the level of SNAIL1, a potent inducer of epithelial to mesenchymal transition. Furthermore, ΔNp63α depletion causes morphological change and enhanced formation of actin stress fibers in squamous cancer cells. Mechanistic studies indicate that ΔNp63α affects cell movement and can reverse the increase of cell motility induced by YB-1 overexpression. These data thus suggest that ΔNp63α provides inhibitory signals for cell motility. Deficiency of ΔNp63α gene expression promotes cell mobilization, at least partially, through a YB-1-dependent mechanism.


Asunto(s)
Movimiento Celular , Núcleo Celular/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteína 1 de Unión a la Caja Y/metabolismo , Transporte Activo de Núcleo Celular/genética , Línea Celular Tumoral , Núcleo Celular/genética , Supervivencia Celular/genética , Humanos , Isoformas de Proteínas , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética , Proteína 1 de Unión a la Caja Y/genética
10.
J Cell Physiol ; 226(8): 2189-97, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21520071

RESUMEN

The homeodomain transcription factors play crucial roles in many developmental processes ranging from organization of the body plan to differentiation of individual tissues. The homeodomain protein Distal-less-3 (DLX3) has an essential role in epidermal stratification and development of ectodermal appendages, placenta and bones. A four-nucleotide deletion in the human DLX3 gene is etiologic for the human hereditary tricho-dento-osseous (TDO) ectodermal dysplasia, a dominant syndrome characterized by abnormalities in hair, nails, teeth, and bones. We have previously demonstrated that DLX3 gene expression induces degradation of ΔNp63α, a specific product of the TP63 gene, a master regulator of multi-layered epithelia. Here we show that the DLX3(TDO) mutant protein is unable to promote ΔNp63α protein degradation and impairs the expression of cell cycle regulatory proteins and skin differentiation markers. However, we found that in cell expressing equal amounts of mutant and wild-type DLX3, ΔNp63α protein level is efficiently regulated implying that genetic heterozygosity at the DLX3 locus protects TDO patients from developing severe p63-associated skin defects.


Asunto(s)
Genes Dominantes , Proteínas de Homeodominio/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Anomalías Múltiples/genética , Biomarcadores/análisis , Proteínas de Ciclo Celular/genética , Diferenciación Celular/genética , Línea Celular , Células Cultivadas , Anomalías Craneofaciales , Hipoplasia del Esmalte Dental/genética , Hipoplasia del Esmalte Dental/metabolismo , Regulación hacia Abajo , Displasia Ectodérmica/genética , Femenino , Enfermedades del Cabello/genética , Enfermedades del Cabello/metabolismo , Proteínas de Homeodominio/genética , Humanos , Eliminación de Secuencia/genética , Piel/metabolismo , Factores de Transcripción/genética
11.
J Proteome Res ; 9(4): 2042-8, 2010 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-20085233

RESUMEN

p63, a transcription factor related to the p53 tumor suppressor, plays a key role in epidermal differentiation and limb development. The gene has two distinct promoters that allow the formation of proteins that either contain (TA) or lack (DeltaN) a transactivation domain. DeltaNp63alpha is the most widely expressed isoform, at all stages of development and in adult tissues. It supports the regenerative capacity of basal keratinocytes and its upregulation is a hallmark of human squamous carcinomas. To get insight into the complex biology of DeltaNp63alpha, we set out to identify DeltaNp63alpha interacting proteins by co-immunoprecipitation in mammalian cells and mass spectrometry analysis. A total of 49 potential DeltaNp63alpha binding proteins, including several heterogeneous ribonucleoproteins (hnRNPs), were identified. Integration of the proteomic data with a Human Coexpression Network highlighted 5 putative p63 protein interactors whose expression is significantly comodulated with p63: hnRNPA/B, hnRNPK, hnRNPQ, FUS/TLS and Keratin 5. hnRNPA/B was already described as a p63 partner, but the others were novel. Interaction of DeltaNp63alpha with hnRNPQ, hnRNPK and FUS/TLS was confirmed by reciprocal co-immunoprecipitations in human keratinocytes. The finding that DeltaNp63alpha exists in complexes with several RNA-binding proteins lays the premises for the analysis of the role of DeltaNp63alpha in mRNA metabolism and transport.


Asunto(s)
Mapeo de Interacción de Proteínas/métodos , Espectrometría de Masas en Tándem/métodos , Transactivadores/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Línea Celular Tumoral , Análisis por Conglomerados , Bases de Datos de Proteínas , Ribonucleoproteínas Nucleares Heterogéneas/química , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Humanos , Isoformas de Proteínas , Proteínas/química , Proteínas/metabolismo , Proteína FUS de Unión a ARN/química , Proteína FUS de Unión a ARN/metabolismo , Transactivadores/química , Factores de Transcripción , Proteínas Supresoras de Tumor/química
12.
Cell Cycle ; 8(21): 3545-51, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19829080

RESUMEN

The tumor suppressor p14(ARF) inhibits cell growth in response to oncogenic stress in a p53-dependent and independent manner. However, new physiologic roles for ARF activation have been proposed. We have previously demonstrated that ARF interacts with p63, influencing its transcriptional activity. p63 is a member of the p53 family involved in skin and limb development, as well as in the homeostasis of mature epidermis. Here, we show that, in human keratinocytes, as well as in tumor-derived cell lines, ARF targets DeltaNp63alpha, the most abundantly expressed p63 isoform, to proteasomal degradation by stimulating its sumoylation. Interestingly, we have observed an increase of ARF expression in differentiating keratinocytes, that is concomitant to the already described upregulation of SUMO2/3. Remarkably, we found that DeltaNp63alpha is preferentially sumoylated by SUMO2, instead of SUMO1, and p14(ARF) increases the efficiency of this process.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Queratinocitos/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Transactivadores/metabolismo , Proteína p14ARF Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Línea Celular Tumoral , Células Cultivadas , Cisteína Endopeptidasas/efectos de los fármacos , Inhibidores de Cisteína Proteinasa/farmacología , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Células HeLa , Humanos , Queratinocitos/efectos de los fármacos , Leupeptinas/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/efectos de los fármacos , Factores de Transcripción , Proteína p14ARF Supresora de Tumor/antagonistas & inhibidores
13.
Cell Cycle ; 8(8): 1185-95, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19282665

RESUMEN

The epidermis is a stratified epithelium which develops depending on the transcription factor p63, a member of the p53 family of transcription factors. p63 is strongly expressed in the innermost basal layer where highly proliferative epithelial cells reside. p63 functions as a molecular switch that initiates epithelial stratification or cell fate determination while regulating proliferation and differentiation of developmentally mature keratinocytes. p63 acts upstream of Dlx3 homeobox gene in a transcriptional regulatory pathway relevant to ectodermal dysplasia. Here we show that Dlx3 triggers p63 protein degradation by a proteasome-dependent pathway. Mutant DeltaNp63alpha in which Threonine397 and Serine383 were replaced with Alanine as well as C-terminal truncated versions of DeltaNp63alpha are resistant to Dlx3-mediated degradation. Transient expression of Dlx3 is associated with Raf1 phosphorylation. Dlx3 is unable to promote p63 degradation in Raf1 depleted MEF cells or upon pharmacological knockdown of Raf1. Our data support a previously unrecognized role for Dlx3 in posttranslational regulation of DeltaNp63alpha protein level, a mechanism that may contribute to reduce the abundance of DeltaNp63alpha during differentiation of stratified epithelia.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Procesamiento Proteico-Postraduccional , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Secuencia de Aminoácidos , Aminoácidos/metabolismo , Animales , Línea Celular , Regulación hacia Abajo , Semivida , Humanos , Queratinocitos/citología , Queratinocitos/metabolismo , Ratones , Datos de Secuencia Molecular , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Transducción de Señal , Relación Estructura-Actividad , Transactivadores/química , Proteínas Supresoras de Tumor/química , Quinasas raf/metabolismo
14.
Cell Cycle ; 5(1): 78-87, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16319531

RESUMEN

p63 mutations have been associated with several human hereditary disorders characterized by ectodermal dysplasia such as EEC (ectrodactyly, ectodermal dysplasia, clefting) syndrome, ADULT (acro, dermato, ungual, lacrimal, tooth) syndrome and AEC (ankyloblepharon, ectodermal dysplasia, clefting) syndrome (also called Hay-Wells syndrome). The location and functional effects of the mutations that underlie these syndromes reveal a striking genotype-phenotype correlation. Unlike EEC and ADULT that result from missense mutations in the DNA-binding domain of p63, AEC is solely caused by missense mutations in the SAM domain of p63. In this paper we report a study on the TAp63alpha isoform, the first to be expressed during development of the embryonic epithelia, and on its naturally occurring Q540L mutant derived from an AEC patient. To assess the effects of the Q540L mutation, we generated stable cell lines expressing TAp63alpha wt, DeltaNp63alpha or the TAp63alpha-Q540L mutant protein and used them to systematically compare the cell growth regulatory activity of the mutant and wt p63 proteins and to generate, by microarray analysis, a comprehensive profile of differential gene expression. We found that the Q540L substitution impairs the transcriptional activity of TAp63alpha and causes misregulation of genes involved in the control of cell growth and epidermal differentiation.


Asunto(s)
Displasia Ectodérmica/metabolismo , Displasia Ectodérmica/patología , Glutamina/genética , Mutación/genética , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Transcripción Genética/genética , Línea Celular Tumoral , Proliferación Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Proteínas de Unión al ADN , Displasia Ectodérmica/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Genes Supresores de Tumor , Glutamina/metabolismo , Humanos , Regiones Promotoras Genéticas/genética , Isoformas de Proteínas , Proteínas Proto-Oncogénicas c-mdm2/genética , Factores de Transcripción , Proteínas Supresoras de Tumor
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA