Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Genes Dev ; 31(1): 18-33, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28096186

RESUMEN

The intratumor microenvironment generates phenotypically distinct but interconvertible malignant cell subpopulations that fuel metastatic spread and therapeutic resistance. Whether different microenvironmental cues impose invasive or therapy-resistant phenotypes via a common mechanism is unknown. In melanoma, low expression of the lineage survival oncogene microphthalmia-associated transcription factor (MITF) correlates with invasion, senescence, and drug resistance. However, how MITF is suppressed in vivo and how MITF-low cells in tumors escape senescence are poorly understood. Here we show that microenvironmental cues, including inflammation-mediated resistance to adoptive T-cell immunotherapy, transcriptionally repress MITF via ATF4 in response to inhibition of translation initiation factor eIF2B. ATF4, a key transcription mediator of the integrated stress response, also activates AXL and suppresses senescence to impose the MITF-low/AXL-high drug-resistant phenotype observed in human tumors. However, unexpectedly, without translation reprogramming an ATF4-high/MITF-low state is insufficient to drive invasion. Importantly, translation reprogramming dramatically enhances tumorigenesis and is linked to a previously unexplained gene expression program associated with anti-PD-1 immunotherapy resistance. Since we show that inhibition of eIF2B also drives neural crest migration and yeast invasiveness, our results suggest that translation reprogramming, an evolutionarily conserved starvation response, has been hijacked by microenvironmental stress signals in melanoma to drive phenotypic plasticity and invasion and determine therapeutic outcome.


Asunto(s)
Plasticidad de la Célula/genética , Reprogramación Celular/genética , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/genética , Melanoma/genética , Factor de Transcripción Asociado a Microftalmía/genética , Biosíntesis de Proteínas/genética , Animales , Microambiente Celular , Evolución Molecular , Retroalimentación Fisiológica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glutamina/farmacología , Humanos , Inmunoterapia , Melanoma/tratamiento farmacológico , Melanoma/metabolismo , Invasividad Neoplásica/genética , Cresta Neural/citología , Fenotipo , Factores de Transcripción/metabolismo , Pez Cebra/embriología
2.
Proc Natl Acad Sci U S A ; 110(29): 11893-8, 2013 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-23818617

RESUMEN

The mechanisms by which arterial fate is established and maintained are not clearly understood. Although a number of signaling pathways and transcriptional regulators have been implicated in arterio-venous differentiation, none are essential for arterial formation, and the manner in which widely expressed factors may achieve arterial-specific gene regulation is unclear. Using both mouse and zebrafish models, we demonstrate here that arterial specification is regulated combinatorially by Notch signaling and SoxF transcription factors, via direct transcriptional gene activation. Through the identification and characterization of two arterial endothelial cell-specific gene enhancers for the Notch ligand Delta-like ligand 4 (Dll4), we show that arterial Dll4 expression requires the direct binding of both the RBPJ/Notch intracellular domain and SOXF transcription factors. Specific combinatorial, but not individual, loss of SOXF and RBPJ DNA binding ablates all Dll4 enhancer-transgene expression despite the presence of multiple functional ETS binding sites, as does knockdown of sox7;sox18 in combination with loss of Notch signaling. Furthermore, triple knockdown of sox7, sox18 and rbpj also results in ablation of endogenous dll4 expression. Fascinatingly, this combinatorial ablation leads to a loss of arterial markers and the absence of a detectable dorsal aorta, demonstrating the essential roles of SoxF and Notch, together, in the acquisition of arterial identity.


Asunto(s)
Arterias/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Receptores Notch/metabolismo , Factores de Transcripción SOXF/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Arterias/metabolismo , Proteínas de Unión al Calcio , Inmunoprecipitación de Cromatina , Clonación Molecular , Ensayo de Cambio de Movilidad Electroforética , Técnicas de Silenciamiento del Gen , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Transgénicos , Pez Cebra
3.
Exp Cell Res ; 318(10): 1146-59, 2012 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-22484375

RESUMEN

Melanoma, the most aggressive form of skin cancer, is notoriously resistant to all current modalities of cancer therapy, including to the drug methotrexate. Melanosomal sequestration and cellular exportation of methotrexate have been proposed to be important melanoma-specific mechanisms that contribute to the resistance of melanoma to methotrexate. In addition, other mechanisms of resistance that are present in most epithelial cancer cells are also operative in melanoma. This report elucidates how melanoma orchestrates these mechanisms to become extremely resistant to methotrexate, where both E2F1 and checkpoint kinase 1 (Chk1), two molecules with dual roles in survival/apoptosis, play prominent roles. The results indicated that MTX induced the depletion of dihydrofolate in melanoma cells, which stimulated the transcriptional activity of E2F1. The elevate expression of dihydrofolate reductase and thymidylate synthase, two E2F1-target genes involved in folate metabolism and required for G(1) progression, favored dTTP accumulation, which promoted DNA single strand breaks and the subsequent activation of Chk1. Under these conditions, melanoma cells are protected from apoptosis by arresting their cell cycle in S phase. Excess of dTTP could also inhibit E2F1-mediated apoptosis in melanoma cells.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Supervivencia Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Melanoma/metabolismo , Metotrexato/farmacología , Neoplasias Cutáneas/metabolismo , Secuencia de Aminoácidos , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Factor de Transcripción E2F1/química , Factor de Transcripción E2F1/metabolismo , Activación Enzimática/efectos de los fármacos , Ácido Fólico/metabolismo , Humanos , Melanoma/patología , Ratones , Datos de Secuencia Molecular , Proteínas Quinasas/metabolismo , Procesamiento Proteico-Postraduccional , Puntos de Control de la Fase S del Ciclo Celular , Neoplasias Cutáneas/patología , Tetrahidrofolato Deshidrogenasa/genética , Tetrahidrofolato Deshidrogenasa/metabolismo , Timidilato Sintasa/genética , Timidilato Sintasa/metabolismo , Nucleótidos de Timina/metabolismo , Transcripción Genética
4.
Cell Death Dis ; 14(11): 761, 2023 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-37996408

RESUMEN

Pancreatic cancer cells with mutant KRAS require strong basal autophagy for viability and growth. Here, we observed that some processes that allow the maintenance of basal autophagy in pancreatic cancer cells are controlled by protein methylation. Thus, by maintaining the methylation status of proteins such as PP2A and MRAS, these cells can sustain their autophagic activity. Protein methylation disruption by a hypomethylating treatment (HMT), which depletes cellular S-adenosylmethionine levels while inducing S-adenosylhomocysteine accumulation, resulted in autophagy inhibition and endoplasmic reticulum stress-induced apoptosis in pancreatic cancer cells. We observed that by reducing the membrane localization of MRAS, hypomethylation conditions produced an imbalance in KRAS signaling, resulting in the partial inactivation of ERK and hyperactivation of the PI3K/AKT-mTORC1 pathway. Interestingly, HMT impeded CRAF activation by disrupting the ternary SHOC2 complex (SHOC2/MRAS/PP1), which functions as a CRAF-S259 holophosphatase. The demethylation events that resulted in PP2A inactivation also favored autophagy inhibition by preventing ULK1 activation while restoring the cytoplasmic retention of the MiT/TFE transcription factors. Since autophagy provides pancreatic cancer cells with metabolic plasticity to cope with various metabolic stress conditions, while at the same time promoting their pathogenesis and resistance to KRAS pathway inhibitors, this hypomethylating treatment could represent a therapeutic opportunity for pancreatic adenocarcinomas.


Asunto(s)
Neoplasias Pancreáticas , Proteínas Proto-Oncogénicas p21(ras) , Humanos , Proteínas Proto-Oncogénicas p21(ras)/genética , Fosfatidilinositol 3-Quinasas , Metilación , Línea Celular Tumoral , Neoplasias Pancreáticas/genética , Autofagia/genética , Péptidos y Proteínas de Señalización Intracelular
5.
Int J Biochem Cell Biol ; 139: 106059, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34400318

RESUMEN

In recent years considerable progress has been made in identifying the impact of mRNA translation in tumour progression. Cancer cells hijack the pre-existing translation machinery to thrive under the adverse conditions originating from intrinsic oncogenic programs, that increase their energetic demand, and from the hostile microenvironment. A key translation program frequently dysregulated in cancer is the Integrated Stress Response, that reprograms translation by attenuating global protein synthesis to decrease metabolic demand while increasing translation of specific mRNAs that support survival, migration, immune escape. In this review we provide an overview of the Integrated Stress Response, emphasise its dual role during tumorigenesis and cancer progression, and highlight the therapeutic strategies available to target it.


Asunto(s)
Factor de Transcripción Activador 4 , Factor 2 Eucariótico de Iniciación , Fosforilación
6.
J Exp Clin Cancer Res ; 40(1): 117, 2021 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-33789714

RESUMEN

BACKGROUND: The application of immune-based therapies has revolutionized cancer treatment. Yet how the immune system responds to phenotypically heterogeneous populations within tumors is poorly understood. In melanoma, one of the major determinants of phenotypic identity is the lineage survival oncogene MITF that integrates diverse microenvironmental cues to coordinate melanoma survival, senescence bypass, differentiation, proliferation, invasion, metabolism and DNA damage repair. Whether MITF also controls the immune response is unknown. METHODS: By using several mouse melanoma models, we examine the potential role of MITF to modulate the anti-melanoma immune response. ChIP-seq data analysis, ChIP-qPCR, CRISPR-Cas9 genome editing, and luciferase reporter assays were utilized to identify ADAM10 as a direct MITF target gene. Western blotting, confocal microscopy, flow cytometry, and natural killer (NK) cytotoxicity assays were used to determine the underlying mechanisms by which MITF-driven phenotypic plasticity modulates melanoma NK cell-mediated killing. RESULTS: Here we show that MITF regulates expression of ADAM10, a key sheddase that cleaves the MICA/B family of ligands for NK cells. By controlling melanoma recognition by NK-cells MITF thereby controls the melanoma response to the innate immune system. Consequently, while melanoma MITFLow cells can be effectively suppressed by NK-mediated killing, MITF-expressing cells escape NK cell surveillance. CONCLUSION: Our results reveal how modulation of MITF activity can impact the anti-melanoma immune response with implications for the application of anti-melanoma immunotherapies.


Asunto(s)
Inmunidad Innata/inmunología , Melanoma/inmunología , Factor de Transcripción Asociado a Microftalmía/metabolismo , Animales , Femenino , Humanos , Ratones , Ratones Desnudos , Transfección
7.
Nat Commun ; 12(1): 3707, 2021 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-34140478

RESUMEN

While the major drivers of melanoma initiation, including activation of NRAS/BRAF and loss of PTEN or CDKN2A, have been identified, the role of key transcription factors that impose altered transcriptional states in response to deregulated signaling is not well understood. The POU domain transcription factor BRN2 is a key regulator of melanoma invasion, yet its role in melanoma initiation remains unknown. Here, in a BrafV600E PtenF/+ context, we show that BRN2 haplo-insufficiency promotes melanoma initiation and metastasis. However, metastatic colonization is less efficient in the absence of Brn2. Mechanistically, BRN2 directly induces PTEN expression and in consequence represses PI3K signaling. Moreover, MITF, a BRN2 target, represses PTEN transcription. Collectively, our results suggest that on a PTEN heterozygous background somatic deletion of one BRN2 allele and temporal regulation of the other allele elicits melanoma initiation and progression.


Asunto(s)
Carcinogénesis/metabolismo , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica/genética , Genes Supresores de Tumor , Proteínas de Homeodominio/metabolismo , Melanoma/metabolismo , Factores del Dominio POU/metabolismo , Neoplasias Cutáneas/metabolismo , Animales , Carcinogénesis/genética , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Estudios de Cohortes , Variaciones en el Número de Copia de ADN , Progresión de la Enfermedad , Técnicas de Silenciamiento del Gen , Haploinsuficiencia , Proteínas de Homeodominio/genética , Humanos , Inmunohistoquímica , Melanoma/genética , Melanoma/mortalidad , Melanoma/secundario , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Análisis por Micromatrices , Factor de Transcripción Asociado a Microftalmía/metabolismo , Mutación , Factores del Dominio POU/genética , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas B-raf/genética , ARN Interferente Pequeño , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/mortalidad , Neoplasias Cutáneas/secundario , Melanoma Cutáneo Maligno
8.
J Cell Biochem ; 110(6): 1399-409, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20564235

RESUMEN

In our search to improve the stability and cellular absorption of tea polyphenols, we synthesized 3-O-(3,4,5-trimethoxybenzoyl)-(-)-epicatechin (TMECG), which showed high antiproliferative activity against melanoma. TMECG downregulates dihydrofolate reductase (DHFR) expression in melanoma cells and we detail the sequential mechanisms that result from this even. TMECG is specifically activated in melanoma cells to form a stable quinone methide (TMECG-QM). TMECG-QM has a dual action on these cells. First, it acts as a potent antifolate compound, disrupting folate metabolism and increasing intracellular oxidized folate coenzymes, such as dihydrofolate, which is a non-competitive inhibitor of dihydropterine reductase, an enzyme essential for tetrahydrobiopterin (H(4)B) recycling. Such inhibition results in H(4)B deficiency, endothelial nitric oxide synthase (eNOS) uncoupling and superoxide production. Second, TMECG-QM acts as an efficient superoxide scavenger and promotes intra-cellular H(2)O(2) accumulation. Here, we present evidence that TMECG markedly reduces melanoma H(4)B and NO bioavailability and that TMECG action is abolished by the eNOS inhibitor N(omega)-nitro-L-arginine methyl ester or the H(2)O(2) scavenger catalase, which strongly suggests H(2)O(2)-dependent DHFR downregulation. In addition, the data presented here indicate that the simultaneous targeting of important pathways for melanoma survival, such as the folate cycle, H(4)B recycling, and the eNOS reaction, could represent an attractive strategy for fighting this malignant skin pathology.


Asunto(s)
Catequina/análogos & derivados , Regulación hacia Abajo/efectos de los fármacos , Tetrahidrofolato Deshidrogenasa/metabolismo , Apoptosis/efectos de los fármacos , Biopterinas/análogos & derivados , Biopterinas/metabolismo , Western Blotting , Catequina/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Dihidropteridina Reductasa/antagonistas & inhibidores , Dihidropteridina Reductasa/genética , Dihidropteridina Reductasa/metabolismo , Ácido Fólico/análogos & derivados , Ácido Fólico/metabolismo , Ácido Fólico/farmacología , Humanos , Peróxido de Hidrógeno/farmacología , Melanoma/genética , Melanoma/metabolismo , Melanoma/patología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Oxidantes/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tetrahidrofolato Deshidrogenasa/genética
9.
Cancers (Basel) ; 13(1)2020 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-33396270

RESUMEN

Hypoxia-inducible factor (HIF)-1α is constitutively expressed in melanoma cells under normoxic conditions and its elevated expression correlates with the aggressiveness of melanoma tumors. Here, we used acriflavine, a potent inhibitor of HIF-1α dimerization, as a tool to investigate whether HIF-1α-regulated pathways contribute to the growth of melanoma cells under normoxia. We observed that acriflavine differentially modulated HIF-1α-regulated targets in melanoma under normoxic conditions, although acriflavine treatment resulted in over-expression of vascular endothelial growth factor (VEGF), its action clearly downregulated the expression of pyruvate dehydrogenase kinase 1 (PDK1), a well-known target of HIF-1α. Consequently, downregulation of PDK1 by acrifavine resulted in reduced glucose availability and suppression of the Warburg effect in melanoma cells. In addition, by inhibiting the AKT and RSK2 phosphorylation, acriflavine also avoided protective pathways necessary for survival under conditions of oxidative stress. Interestingly, we show that acriflavine targets activating transcription factor 4 (ATF4) for proteasomal degradation while suppressing the expression of microphthalmia-associated transcription factor (MITF), a master regulator of melanocyte development and a melanoma oncogene. Since acriflavine treatment results in the consistent death of melanoma cells, our results suggest that inhibition of HIF-1α function in melanoma could open new avenues for the treatment of this deadly disease regardless of the hypoxic condition of the tumor.

10.
Sci Rep ; 10(1): 13275, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32764667

RESUMEN

The therapeutic effect of irradiation is thought to come from DNA damage that affects rapidly proliferating cancer cells; however, resistant cells rapidly initiate mechanisms to repair such damage. While DNA repair mechanisms responsible for cancer cell survival following DNA damage are understood, less is known about the epigenetic mechanisms resulting in resistance to radiotherapy. Although changes in DNA methylation are related to mechanisms of long-term resistance, it is more likely that the methylation state of a series of proteins could be responsible for the first-line of defense of cancer cells against irradiation. In this study, we observed that irradiation of breast cancer cells was accompanied by an overproduction in S-adenosylmethionine, which increases the activity of cellular methylases. We found that by activating PRMT1, irradiation triggers a BRCA1-dependent program that results in efficient DNA repair and inhibition of apoptosis. Depletion of PRMT1 in irradiated cells resulted in a switch of BRCA1 functions from repair and survival in the nucleus to activation of cell death signals in the cytoplasm. We conclude that by modulating the cellular localization of BRCA1, PRMT1 is an important regulator of the oncogenic functions of BRCA1, contributing to the epigenetic defense of breast cancer cells against ionizing radiation.


Asunto(s)
Proteína BRCA1/metabolismo , Neoplasias de la Mama/patología , Proteína-Arginina N-Metiltransferasas/metabolismo , Tolerancia a Radiación , Proteínas Represoras/metabolismo , S-Adenosilmetionina/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Supervivencia Celular , Citoplasma/metabolismo , Reparación del ADN , Epigénesis Genética , Femenino , Humanos , Células MCF-7 , Ratones , Radiación Ionizante , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Int J Mol Sci ; 10(12): 5398-5410, 2009 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-20054477

RESUMEN

Dihydrofolate reductase (DHFR) is the subject of intensive investigation since it appears to be the primary target enzyme for antifolate drugs. Fluorescence quenching experiments show that the ester bond-containing tea polyphenols (-)-epigallocatechin gallate (EGCG) and (-)-epicatechin gallate (ECG) are potent inhibitors of DHFR with dissociation constants (K(D))of 0.9 and 1.8 microM, respectively, while polyphenols lacking the ester bound gallate moiety [e.g., (-)-epigallocatechin (EGC) and (-)-epicatechin (EC)] did not bind to this enzyme. To avoid stability and bioavailability problems associated with tea catechins we synthesized a methylated derivative of ECG (3-O-(3,4,5-trimethoxybenzoyl)-(-)-epicatechin; TMECG), which effectively binds to DHFR (K(D) = 2.1 microM). In alkaline solution, TMECG generates a stable quinone methide product that strongly binds to the enzyme with a K(D) of 8.2 nM. Quercetin glucuronides also bind to DHFR but its effective binding was highly dependent of the sugar residue, with quercetin-3-xyloside being the stronger inhibitor of the enzyme with a K(D) of 0.6 microM. The finding that natural polyphenols are good inhibitors of human DHFR could explain the epidemiological data on their prophylactic effects for certain forms of cancer and open a possibility for the use of natural and synthetic polyphenols in cancer chemotherapy.


Asunto(s)
Antagonistas del Ácido Fólico/química , Polifenoles/química , Tetrahidrofolato Deshidrogenasa/química , Secuencia de Aminoácidos , Sitios de Unión , Catequina/análogos & derivados , Catequina/síntesis química , Catequina/química , Antagonistas del Ácido Fólico/farmacología , Glucurónidos/química , Glucurónidos/farmacología , Humanos , Indolquinonas/química , Indolquinonas/farmacología , Simulación del Acoplamiento Molecular , Datos de Secuencia Molecular , Polifenoles/farmacología , Unión Proteica , Quercetina/química , Tetrahidrofolato Deshidrogenasa/metabolismo
12.
Int J Cancer ; 123(10): 2446-55, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18729182

RESUMEN

The higher expression of methionine cycle genes in melanoma cells than in normal melanocytes may be related with increased protein synthesis and transmethylation reactions and the subsequent need for high levels of methionine. 3-O-(3,4,5-trimethoxybenzoyl)-(-)-epicatechin (TMECG), a trimethoxy derivative of epicatechin-3-gallate (ECG), effectively suppressed proliferation of melanoma cells in cultures by inducing apoptosis. TMECG modulates the expression of genes involved in methionine metabolism, cellular methylation and glutathione synthesis in melanoma cells. TMECG treatment of melanoma cells resulted in the downregulation of antiapoptotic Bcl-2, the upregulation of proapoptotic Bax and the activation of caspase-3; however, it did not induce the expression of the apoptosis protease-activating factor-1 (Apaf-1). Having elucidated the effects of TMECG on the melanoma methionine cycle, we designed therapeuthical strategies to increase its effectiveness. Combinations of TMECG with S-adenosylmethionine or compounds that modulate the intracellular concentration of adenosine strongly increase the antiproliferative effects of TMECG. The ability of TMECG to target multiple aspects related with melanoma survival, with a high degree of potency, points to its clinical value in melanoma therapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Catequina/análogos & derivados , Melanoma/tratamiento farmacológico , Metionina/metabolismo , Adenosina/metabolismo , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Secuencia de Bases , Western Blotting , Caspasa 3/metabolismo , Catequina/farmacología , Catequina/uso terapéutico , Línea Celular Tumoral , Cartilla de ADN , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glutatión/biosíntesis , Humanos , Metilación , Ratones , Microscopía Confocal , Microscopía Electrónica , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
J Med Chem ; 51(7): 2018-26, 2008 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-18324763

RESUMEN

Despite presenting bioavailability problems, tea catechins have emerged as promising chemopreventive agents because of their observed efficacy in various animal models. To improve the stability and cellular absorption of tea polyphenols, we developed a new catechin-derived compound, 3- O-(3,4,5-trimethoxybenzoyl)-(-)-epicatechin (TMECG), which has shown significant antiproliferative activity against several cancer cell lines, especially melanoma. The presence of methoxy groups in its ester-bound gallyl moiety drastically decreased its antioxidant and prooxidant properties without affecting its cell-antiproliferative effects, and the data indicated that the 3-gallyl moiety was essential for its biological activity. As regards its action mechanism, we demonstrated that TMECG binds efficiently to human dihydrofolate reductase and down-regulates folate cycle gene expression in melanoma cells. Disruption of the folate cycle by TMECG is a plausible explanation for its observed biological effects and suggests that, like other antifolate compounds, TMECG could be of clinical value in cancer therapy.


Asunto(s)
Antioxidantes/síntesis química , Antioxidantes/farmacología , Catequina/análogos & derivados , Antagonistas del Ácido Fólico/síntesis química , Antagonistas del Ácido Fólico/farmacología , Antioxidantes/química , Catequina/síntesis química , Catequina/química , Catequina/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Ácido Fólico/efectos de los fármacos , Ácido Fólico/metabolismo , Antagonistas del Ácido Fólico/química , Perfilación de la Expresión Génica , Humanos , Metilenotetrahidrofolato Reductasa (NADPH2)/efectos de los fármacos , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Modelos Moleculares , Estructura Molecular , NADP/química , NADP/efectos de los fármacos , ARN Mensajero/efectos de los fármacos , ARN Mensajero/genética , Estereoisomerismo , Relación Estructura-Actividad , Té/química , Tetrahidrofolato Deshidrogenasa/efectos de los fármacos , Tetrahidrofolato Deshidrogenasa/genética , Timidilato Sintasa/efectos de los fármacos , Timidilato Sintasa/genética , Factores de Tiempo
15.
Neurochem Int ; 50(3): 531-9, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17178175

RESUMEN

The laminin-alpha2 chain, referred to as merosin, forms part of the laminin-2 heterotrimer (alpha2beta1gamma1), which is principally expressed in the basement membrane of muscle. Nearly half of patients suffering from congenital muscular dystrophy (CMD) have abnormalities in the laminin-alpha2 chain (LAMA2) gene, and the merosin-deficient Lama2dy mouse shows CMD. The expression of merosin in thymus, the abnormalities in the gland of Lama2dy mice, and the presence of acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) in thymus prompted us to study the possible effects of the deficiency of merosin on thymus BuChE. We found that, while AChE activity decreased by approximately 50% in merosin-deficient thymus, the deficiency had little effect on BuChE activity. About 65% of thymus BuChE activity was extracted with a saline buffer and 30% with 1% Triton X-100. Sedimentation analyses and phenyl-agarose chromatography showed that thymus contained amphiphilic BuChE monomers (G(1)(A),44%) and dimers (G(2)(A),33%), and hydrophilic tetramers (G(4)(H),23%). Binding assays with various plant lectins revealed differences between the oligoglycans linked to BuChE tetramers and lighter components. The deficiency of merosin had no effect on the biosynthesis of thymus BuChE as judged by the lack of major changes between control and Lama2dy mice thymuses in the distribution of BuChE molecules and the level of lectin binding. The detoxifying action of BuChE, its role as a backup to AChE, and the relevance of the cholinergic dialogue between T cells and stromal cells for T lymphocyte proliferation, maturation and survival support a physiological function for BuChE in thymus.


Asunto(s)
Butirilcolinesterasa/metabolismo , Laminina/fisiología , Timo/enzimología , Animales , Butirilcolinesterasa/aislamiento & purificación , Laminina/genética , Ratones
16.
Int J Biochem Cell Biol ; 38(7): 1092-101, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16412684

RESUMEN

We have previously shown that mouse muscle and liver contain catalytically active and inactive ecto-5'-nucleotidase (eNT) variants and that eNT activity in these tissues increases in laminin alpha2 (merosin)-deficient Lama2dy mice. These results prompted us to study whether: (1) the increase of eNT activity depends on the change in the content of merosin between healthy and dystrophic organs; (2) the active and inactive eNT variants arise from the same or distinct mRNAs; (3) the enhancement of the activity is caused by an increase in the eNT mRNA content. Compared to healthy organs, the activity in dystrophic organs increased four-fold in muscle, 1.7-fold in liver, 1.4-fold in heart and not at all in kidney and lung. The level of immunolabelled eNT protein per unit of activity suggested a similar ratio of inactive to active eNT in healthy liver, kidney, heart and muscle, which increased greatly in lung. The size of the eNT subunit in liver, kidney, heart and muscle (72 kDa) decreased to 66 kDa in lung. The identification of a single RT-PCR product suggested that active and inactive eNT arise from the same mRNA and are generated by a differential post-translational processing. Compared to the content in muscle, the amount of eNT mRNA was 12-fold higher in liver and kidney, eight-fold in heart and five-fold in lung. The relative content of eNT mRNA was unaffected by the deficiency of merosin.


Asunto(s)
5'-Nucleotidasa/metabolismo , Activación Enzimática/genética , Laminina/deficiencia , Hígado/metabolismo , Músculo Esquelético/metabolismo , Miocardio/metabolismo , ARN Mensajero/metabolismo , 5'-Nucleotidasa/análisis , Animales , Laminina/química , Laminina/genética , Hígado/química , Ratones , Ratones Mutantes/genética , Músculo Esquelético/química , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/metabolismo , Miocardio/química , Procesamiento Proteico-Postraduccional , ARN Mensajero/análisis
17.
Toxins (Basel) ; 8(7)2016 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-27399772

RESUMEN

Melanoma, the most threatening form of skin cancer, has a very poor prognosis and is characterized by its very invasive and chemoresistant properties. Despite the recent promising news from the field of immunotherapy, there is an urgent need for new therapeutic approaches that are free of resistance mechanisms and side effects. Anti-neoplasic properties have been highlighted for different disintegrins from snake venom including Lebein; however, the exact effect of Lebein on melanoma has not yet been defined. In this study, we showed that Lebein blocks melanoma cell proliferation and induces a more differentiated phenotype with inhibition of extracellular signal-regulated kinase (ERK) phosphorylation and microphthalmia-associated transcription factor (MITF) overexpression. Melanoma cells became detached but were less invasive with upregulation of E-cadherin after Lebein exposure. Lebein induced a caspase-independent apoptotic program with apoptosis inducing factor (AIF), BCL-2-associated X protein (BAX) and Bim overexpression together with downregulation of B-cell lymphoma-2 (BCL-2). It generated a distinct response in reactive oxygen species (ROS) generation and p53 levels depending on the p53 cell line status (wild type or mutant). Therefore, we propose Lebein as a new candidate for development of potential therapies for melanoma.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Melanoma/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , Venenos de Víboras/farmacología , Antígenos CD , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Adhesión Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Melanoma/genética , Melanoma/metabolismo , Melanoma/patología , Factor de Transcripción Asociado a Microftalmía/genética , Factor de Transcripción Asociado a Microftalmía/metabolismo , Fenotipo , Fosforilación , Transducción de Señal/efectos de los fármacos , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Factores de Tiempo
18.
Pigment Cell Melanoma Res ; 28(6): 736-40, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26301891

RESUMEN

The multifunctional Ig-like carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) is neo-expressed in the majority of malignant melanoma lesions. CEACAM1 acts as a driver of tumor cell invasion, and its expression correlates with poor patient prognosis. Despite its importance in melanoma progression, how CEACAM1 expression is regulated is largely unknown. Here, we show that CEACAM1 expression in melanoma cell lines and melanoma tissue strongly correlates with that of the microphthalmia-associated transcription factor (MITF), a key regulator of melanoma proliferation and invasiveness. MITF is revealed as a direct and positive regulator for CEACAM1 expression via binding to an M-box motif located in the CEACAM1 promoter. Taken together, our study provides novel insights into the regulation of CEACAM1 expression and suggests an MITF-CEACAM1 axis as a potential determinant of melanoma progression.


Asunto(s)
Antígenos CD/metabolismo , Moléculas de Adhesión Celular/metabolismo , Melanoma/metabolismo , Factor de Transcripción Asociado a Microftalmía/metabolismo , Secuencia de Bases , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/genética , Datos de Secuencia Molecular , Motivos de Nucleótidos/genética , Unión Proteica , Neoplasias Cutáneas , Melanoma Cutáneo Maligno
19.
Neoplasia ; 15(7): 826-39, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23814494

RESUMEN

Human melanoma is a significant clinical problem. As most melanoma patients relapse with lethal drug-resistant disease, understanding and preventing mechanism(s) of resistance is one of the highest priorities to improve melanoma therapy. Melanosomal sequestration and the cellular exportation of cytotoxic drugs have been proposed to be important melanoma-specific mechanisms that contribute to multidrug resistance in melanoma. Concretely, we found that treatment of melanoma with methotrexate (MTX) altered melanogenesis and accelerated the exportation of melanosomes; however, the cellular and molecular processes by which MTX is trapped into melanosomes and exported out of cells have not been elucidated. In this study, we identified myosin Va (MyoVa) as a possible mediator of these cellular processes. The results demonstrated that melanoma treatment with MTX leads to Akt2-dependent MyoVa phosphorylation, which enhances its ability to interact with melanosomes and accelerates their exportation. To understand the mechanism(s) by which MTX activates Akt2, we examined the effects of this drug on the activity of protein phosphatase 2A, an Akt inhibitor activated by the methylation of its catalytic subunit. Taken together, this study identified a novel trafficking pathway in melanoma that promotes tumor resistance through Akt2/MyoVa activation. Because of these findings, we explored several MTX combination therapies to increase the susceptibility of melanoma to this drug. By avoiding MTX exportation, we observed that the E2F1 apoptotic pathway is functional in melanoma, and its induction activates p73 and apoptosis protease-activating factor 1 following a p53-autonomous proapoptotic signaling event.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Melanoma/metabolismo , Metotrexato/farmacología , Cadenas Pesadas de Miosina/metabolismo , Miosina Tipo V/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular Tumoral , Modelos Animales de Enfermedad , Factor de Transcripción E2F1/metabolismo , Activación Enzimática/efectos de los fármacos , Femenino , Silenciador del Gen , Humanos , Melanoma/genética , Metilación/efectos de los fármacos , Ratones , Cadenas Pesadas de Miosina/genética , Miosina Tipo V/genética , Fosfoproteínas Fosfatasas/metabolismo , Fosforilación/efectos de los fármacos , Proteína Fosfatasa 2C , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Estaurosporina/análogos & derivados , Estaurosporina/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Cancer Cell ; 24(1): 105-19, 2013 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-23792190

RESUMEN

Therapeutic resistance in melanoma and other cancers arises via irreversible genetic, and dynamic phenotypic, heterogeneity. Here, we use directed phenotype switching in melanoma to sensitize melanoma cells to lineage-specific therapy. We show that methotrexate (MTX) induces microphthalmia-associated transcription factor (MITF) expression to inhibit invasiveness and promote differentiation-associated expression of the melanocyte-specific Tyrosinase gene. Consequently, MTX sensitizes melanomas to a tyrosinase-processed antifolate prodrug 3-O-(3,4,5-trimethoxybenzoyl)-(-)-epicatechin (TMECG), that inhibits the essential enzyme DHFR with high affinity. The combination of MTX and TMECG leads to depletion of thymidine pools, double-strand DNA breaks, and highly efficient E2F1-mediated apoptosis in culture and in vivo. Importantly, this drug combination delivers an effective and tissue-restricted antimelanoma therapy in vitro and in vivo irrespective of BRAF, MEK, or p53 status.


Asunto(s)
Melanoma/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Catequina/análogos & derivados , Catequina/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Daño del ADN , Factor de Transcripción E2F1/fisiología , Humanos , Melanoma/patología , Metotrexato/farmacología , Factor de Transcripción Asociado a Microftalmía/genética , Fenotipo , Nucleótidos de Timina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA