Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
2.
EMBO Mol Med ; 15(12): e17719, 2023 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-37966164

RESUMEN

Metastatic uveal melanomas are highly resistant to all existing treatments. To address this critical issue, we performed a kinome-wide CRISPR-Cas9 knockout screen, which revealed the LKB1-SIK2 module in restraining uveal melanoma tumorigenesis. Functionally, LKB1 loss enhances proliferation and survival through SIK2 inhibition and upregulation of the sodium/calcium (Na+ /Ca2+ ) exchanger SLC8A1. This signaling cascade promotes increased levels of intracellular calcium and mitochondrial reactive oxygen species, two hallmarks of cancer. We further demonstrate that combination of an SLC8A1 inhibitor and a mitochondria-targeted antioxidant promotes enhanced cell death efficacy in LKB1- and SIK2-negative uveal melanoma cells compared to control cells. Our study also identified an LKB1-loss gene signature for the survival prognostic of patients with uveal melanoma that may be also predictive of response to the therapy combination. Our data thus identify not only metabolic vulnerabilities but also new prognostic markers, thereby providing a therapeutic strategy for particular subtypes of metastatic uveal melanoma.


Asunto(s)
Melanoma , Neoplasias de la Úvea , Humanos , Calcio , Proliferación Celular , Melanoma/tratamiento farmacológico , Especies Reactivas de Oxígeno , Neoplasias de la Úvea/genética , Neoplasias de la Úvea/patología
3.
Mol Cancer ; 20(1): 12, 2021 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-33413419

RESUMEN

Resistances to immunotherapies remains a major hurdle towards a cure for melanoma in numerous patients. An increase in the mesenchymal phenotype and a loss of differentiation have been clearly associated with resistance to targeted therapies. Similar phenotypes have been more recently also linked to resistance to immune checkpoint therapies. We demonstrated here that the loss of MIcrophthalmia associated Transcription Factor (MITF), a pivotal player in melanocyte differentiation, favors the escape of melanoma cells from the immune system. We identified Integrin beta-like protein 1 (ITGBL1), a secreted protein, upregulated in anti-PD1 resistant patients and in MITFlow melanoma cells, as the key immunomodulator. ITGBL1 inhibited immune cell cytotoxicity against melanoma cells by inhibiting NK cells cytotoxicity and counteracting beneficial effects of anti-PD1 treatment, both in vitro and in vivo. Mechanistically, MITF inhibited RUNX2, an activator of ITGBL1 transcription. Interestingly, VitaminD3, an inhibitor of RUNX2, improved melanoma cells to death by immune cells. In conclusion, our data suggest that inhibition of ITGBL1 might improve melanoma response to immunotherapies.


Asunto(s)
Carcinogénesis/patología , Citotoxicidad Inmunológica , Factores Inmunológicos/metabolismo , Integrina beta1/metabolismo , Células Asesinas Naturales/inmunología , Melanoma/inmunología , Animales , Línea Celular Tumoral , Proliferación Celular , Melanoma/patología , Ratones Endogámicos C57BL , Factor de Transcripción Asociado a Microftalmía/metabolismo
4.
Cell Death Differ ; 28(6): 1990-2000, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33462406

RESUMEN

Intratumor heterogeneity has been recognized in numerous cancers as a major source of metastatic dissemination. In uveal melanomas, the existence and identity of specific subpopulations, their biological function and their contribution to metastasis remain unknown. Here, in multiscale analyses using single-cell RNA sequencing of six different primary uveal melanomas, we uncover an intratumoral heterogeneity at the genomic and transcriptomic level. We identify distinct transcriptional cell states and diverse tumor-associated populations in a subset of the samples. We also decipher a gene regulatory network underlying an invasive and poor prognosis state driven in part by the transcription factor HES6. HES6 heterogenous expression has been validated by RNAscope assays within primary human uveal melanomas, which further unveils the existence of these cells conveying a dismal prognosis in tumors diagnosed with a favorable outcome using bulk analyses. Depletion of HES6 impairs proliferation, migration and metastatic dissemination in vitro and in vivo using the chick chorioallantoic membrane assay, demonstrating the essential role of HES6 in uveal melanomas. Thus, single-cell analysis offers an unprecedented view of primary uveal melanoma heterogeneity, identifies bona fide biomarkers for metastatic cells in the primary tumor, and reveals targetable modules driving growth and metastasis formation. Significantly, our findings demonstrate that HES6 is a valid target to stop uveal melanoma progression.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Melanoma/genética , Proteínas Represoras/metabolismo , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Neoplasias de la Úvea/genética , Línea Celular Tumoral , Humanos , Metástasis de la Neoplasia , Pronóstico
6.
J Invest Dermatol ; 140(11): 2253-2259.e4, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32240722

RESUMEN

Integration of chromatin immunoprecipitation-sequencing and microarray data enabled us to identify previously unreported MITF-target genes, among which the amino acid transporter SLC7A5 is also included. We reported that small interfering RNA-mediated SLC7A5 knockdown decreased pigmentation in B16F10 cells but neither affected morphology nor dendricity. Treatment with the SLC7A5 inhibitors 2-amino-2-norbornanecarboxylic acid (BCH) or JPH203 also decreased melanin synthesis in B16F10 cells. Our findings indicated that BCH was as potent as reference depigmenting agent, kojic acid, but acted through a different pathway not affecting tyrosinase activity. BCH also decreased pigmentation in human MNT1 melanoma cells or normal human melanocytes. Finally, we tested BCH on a more physiological model, using reconstructed human epidermis and confirmed a strong inhibition of pigmentation, revealing the clinical potential of SLC7A5 inhibition and positioning BCH as a depigmenting agent suitable for cosmetic or dermatological intervention in hyperpigmentation diseases.


Asunto(s)
Transportador de Aminoácidos Neutros Grandes 1/fisiología , Melaninas/biosíntesis , Animales , Ácidos Carboxílicos/farmacología , Línea Celular Tumoral , Humanos , Transportador de Aminoácidos Neutros Grandes 1/genética , Melaninas/análisis , Ratones , Factor de Transcripción Asociado a Microftalmía/genética , Factor de Transcripción Asociado a Microftalmía/fisiología , Norbornanos/farmacología , Pigmentación/efectos de los fármacos , Pironas/farmacología , ARN Interferente Pequeño/genética
7.
Oncogene ; 38(8): 1282-1295, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30254208

RESUMEN

Phenotypic plasticity and subsequent generation of intratumoral heterogeneity underly key traits in malignant melanoma such as drug resistance and metastasis. Melanoma plasticity promotes a switch between proliferative and invasive phenotypes characterized by different transcriptional programs of which MITF is a critical regulator. Here, we show that the acid ceramidase ASAH1, which controls sphingolipid metabolism, acted as a rheostat of the phenotypic switch in melanoma cells. Low ASAH1 expression was associated with an invasive behavior mediated by activation of the integrin alphavbeta5-FAK signaling cascade. In line with that, human melanoma biopsies revealed heterogeneous staining of ASAH1 and low ASAH1 expression at the melanoma invasive front. We also identified ASAH1 as a new target of MITF, thereby involving MITF in the regulation of sphingolipid metabolism. Together, our findings provide new cues to the mechanisms underlying the phenotypic plasticity of melanoma cells and identify new anti-metastatic targets.


Asunto(s)
Ceramidasa Ácida/genética , Proliferación Celular/genética , Melanoma/genética , Factor de Transcripción Asociado a Microftalmía/genética , Línea Celular Tumoral , Femenino , Quinasa 1 de Adhesión Focal/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Melanoma/patología , Invasividad Neoplásica/genética , Proteínas Proto-Oncogénicas B-raf , Receptores de Vitronectina/genética , Transducción de Señal
8.
Genes Dev ; 32(5-6): 448-461, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29567766

RESUMEN

In BRAFV600E melanoma cells, a global metabolomic analysis discloses a decrease in nicotinamide adenine dinucleotide (NAD+) levels upon PLX4032 treatment that is conveyed by a STAT5 inhibition and a transcriptional regulation of the nicotinamide phosphoribosyltransferase (NAMPT) gene. NAMPT inhibition decreases melanoma cell proliferation both in vitro and in vivo, while forced NAMPT expression renders melanoma cells resistant to PLX4032. NAMPT expression induces transcriptomic and epigenetic reshufflings that steer melanoma cells toward an invasive phenotype associated with resistance to targeted therapies and immunotherapies. Therefore, NAMPT, the key enzyme in the NAD+ salvage pathway, appears as a rational target in targeted therapy-resistant melanoma cells and a key player in phenotypic plasticity of melanoma cells.


Asunto(s)
Citocinas/metabolismo , Resistencia a Antineoplásicos/genética , Melanoma/enzimología , Melanoma/genética , Invasividad Neoplásica/genética , Nicotinamida Fosforribosiltransferasa/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Citocinas/genética , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Indoles/farmacología , Melanoma/fisiopatología , Metaboloma , Ratones , Ratones Desnudos , NAD/metabolismo , Nicotinamida Fosforribosiltransferasa/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Factor de Transcripción STAT5/antagonistas & inhibidores , Factor de Transcripción STAT5/genética , Sulfonamidas/farmacología , Activación Transcripcional/efectos de los fármacos , Vemurafenib
9.
J Natl Cancer Inst ; 109(8)2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28376192

RESUMEN

Background: MITF encodes an oncogenic lineage-specific transcription factor in which a germline mutation ( MITFE318K ) was identified in human patients predisposed to both nevus formation and, among other tumor types, melanoma. The molecular mechanisms underlying the oncogenic activity of MITF E318K remained uncharacterized. Methods: Here, we compared the SUMOylation status of endogenous MITF by proximity ligation assay in melanocytes isolated from wild-type (n = 3) or E318K (n = 4) MITF donors. We also used a newly generated Mitf E318K knock-in (KI) mouse model to assess the role of Mitf E318K (n = 7 to 13 mice per group) in tumor development in vivo and performed transcriptomic analysis of the tumors to identify the molecular mechanisms. Finally, using immortalized or normal melanocytes (wild-type or E318K MITF, n = 2 per group), we assessed the role of MITF E318K on the induction of senescence mediated by BRAF V600E . All statistical tests were two-sided. Results: We demonstrated a decrease in endogenous MITF SUMOylation in melanocytes from MITF E318K patients (mean of cells with hypoSUMOylated MITF, MITF E318K vs MITF WT , 94% vs 44%, difference = 50%, 95% CI = 21.8% to 67.2%, P = .004). The Mitf E318K mice were slightly hypopigmented (mean melanin content Mitf WT vs Mitf E318K/+ , 0.54 arbitrary units [AU] vs 0.36 AU, difference = -0.18, 95% CI = -0.36 to -0.007, P = .04). We provided genetic evidence that Mitf E318K enhances BRaf V600E -induced nevus formation in vivo (mean nevus number for Mitf E318K , BRaf V600E vs Mitf WT , BRaf V600E , 68 vs 44, difference = 24, 95% CI = 9.1 to 38.9, P = .006). Importantly, although Mitf E318K was not sufficient to cooperate with BRaf V600E alone in promoting metastatic melanoma, it accelerated tumor formation on a BRaf V600E , Pten-deficient background (median survival, Mitf E318K/+ = 42 days, 95% CI = 31 to 46 vs Mitf WT = 51 days, 95% CI = 50 to 55, P < .001). Transcriptome analysis suggested a decrease in senescence in tumors from Mitf E318K mice. We confirmed this hypothesis by in vitro experiments, demonstrating that Mitf E318K impaired the ability of human melanocytes to undergo BRAF V600E -induced senescence. Conclusions: We characterized the functions of melanoma-associated MITF E318K mutations. Our results demonstrate that MITF E318K reduces the program of senescence to potentially favor melanoma progression in vivo.


Asunto(s)
Melanoma/genética , Factor de Transcripción Asociado a Microftalmía/genética , Factor de Transcripción Asociado a Microftalmía/metabolismo , Nevo/genética , Proteínas Proto-Oncogénicas B-raf/genética , Neoplasias Cutáneas/genética , Adulto , Anciano , Animales , Línea Celular Tumoral , Senescencia Celular/genética , Modelos Animales de Enfermedad , Mutación de Línea Germinal , Humanos , Melanocitos , Ratones , Persona de Mediana Edad , Fosfohidrolasa PTEN/genética , Cultivo Primario de Células , Sumoilación , Transcriptoma
11.
Oncotarget ; 5(8): 2085-95, 2014 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-24742694

RESUMEN

SIRT1 operates as both a tumor suppressor and oncogenic factor depending on the cell context. Whether SIRT1 plays a role in melanoma biology remained poorly elucidated. Here, we demonstrate that SIRT1 is a critical regulator of melanoma cell proliferation. SIRT1 suppression by genetic or pharmacological approaches induces cell cycle arrest and a senescence-like phenotype. Gain and loss of function experiments show that M-MITF regulates SIRT1 expression, thereby revealing a melanocyte-specific control of SIRT1. SIRT1 over-expression relieves the senescence-like phenotype and the proliferation arrest caused by MITF suppression, demonstrating that SIRT1 is an effector of MITF-induced proliferation in melanoma cells. Interestingly, SIRT1 level and activity are enhanced in the PLX4032-resistant BRAF(V600E)-mutated melanoma cells compared with their sensitive counterpart. SIRT1 inhibition decreases melanoma cell growth and rescues the sensibility to PLX4032 of PLX4032-resistant BRAF(V600E)-mutated melanoma cells. In conclusion, we provide the first evidence that inhibition of SIRT1 warrants consideration as an anti-melanoma therapeutic option.


Asunto(s)
Proliferación Celular/fisiología , Senescencia Celular/fisiología , Melanoma/metabolismo , Sirtuina 1/metabolismo , Western Blotting , Técnica del Anticuerpo Fluorescente , Humanos , Microscopía Confocal , Fenotipo , ARN Interferente Pequeño/genética , Transfección
12.
Oncotarget ; 4(12): 2212-24, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24344100

RESUMEN

Here, we showed that the secretome of senescent melanoma cells drives basal melanoma cells towards a mesenchymal phenotype, with characteristic of stems illustrated by increased level of the prototype genes FN1, SNAIL, OCT4 and NANOG. This molecular reprogramming leads to an increase in the low-MITF and slow-growing cell population endowed with melanoma-initiating cell features. The secretome of senescent melanoma cells induces a panel of 52 genes, involved in cell movement and cell/cell interaction, among which AXL and ALDH1A3 have been implicated in melanoma development. We found that the secretome of senescent melanoma cells activates the STAT3 pathway and STAT3 inhibition prevents secretome effects, including the acquisition of tumorigenic properties. Collectively, the findings provide insights into how the secretome of melanoma cells entering senescence upon chemotherapy treatments increases the tumorigenicity of naïve melanoma cells by inducing, through STAT3 activation, a melanoma-initiating cell phenotype that could favor chemotherapy resistance and relapse.


Asunto(s)
Melanoma/patología , Células Madre Neoplásicas/patología , Factor de Transcripción STAT3/antagonistas & inhibidores , Animales , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Senescencia Celular/fisiología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/genética , Melanoma/metabolismo , Ratones , Ratones Desnudos , Fenotipo , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Distribución Aleatoria , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
13.
J Biol Chem ; 287(35): 29887-98, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22767597

RESUMEN

Metastatic melanoma is a deadly skin cancer and is resistant to almost all existing treatment. Vemurafenib, which targets the BRAFV600E mutation, is one of the drugs that improves patient outcome, but the patients next develop secondary resistance and a return to cancer. Thus, new therapeutic strategies are needed to treat melanomas and to increase the duration of v-Raf murine sarcoma viral oncogene homolog B1 (BRAF) inhibitor response. The ERK pathway controls cell proliferation, and Aurora B plays a pivotal role in cell division. Here, we confirm that Aurora B is highly expressed in metastatic melanoma cells and that Aurora B inhibition triggers both senescence-like phenotypes and cell death in melanoma cells. Furthermore, we show that the BRAF/ERK axis controls Aurora B expression at the transcriptional level, likely through the transcription factor FOXM1. Our results provide insight into the mechanism of Aurora B regulation and the first molecular basis of Aurora B regulation in melanoma cells. The inhibition of Aurora B expression that we observed in vemurafenib-sensitive melanoma cells was rescued in cells resistant to this drug. Consistently, these latter cells remain sensitive to the effect of the Aurora B inhibitor. Noteworthy, wild-type BRAF melanoma cells are also sensitive to Aurora B inhibition. Collectively, our findings, showing that Aurora B is a potential target in melanoma cells, particularly in those vemurafenib-resistant, may open new avenues to improve the treatment of metastatic melanoma.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Sistema de Señalización de MAP Quinasas , Melanoma/enzimología , Proteínas Serina-Treonina Quinasas/biosíntesis , Neoplasias Cutáneas/enzimología , Sustitución de Aminoácidos , Animales , Aurora Quinasa B , Aurora Quinasas , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , División Celular/efectos de los fármacos , División Celular/genética , Línea Celular Tumoral , Senescencia Celular/efectos de los fármacos , Senescencia Celular/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Proteína Forkhead Box M1 , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Indoles/farmacología , Melanoma/genética , Melanoma/patología , Melanoma/terapia , Ratones , Mutación Missense , Metástasis de la Neoplasia , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/terapia , Sulfonamidas/farmacología , Vemurafenib
14.
Nature ; 480(7375): 94-8, 2011 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-22012259

RESUMEN

So far, no common environmental and/or phenotypic factor has been associated with melanoma and renal cell carcinoma (RCC). The known risk factors for melanoma include sun exposure, pigmentation and nevus phenotypes; risk factors associated with RCC include smoking, obesity and hypertension. A recent study of coexisting melanoma and RCC in the same patients supports a genetic predisposition underlying the association between these two cancers. The microphthalmia-associated transcription factor (MITF) has been proposed to act as a melanoma oncogene; it also stimulates the transcription of hypoxia inducible factor (HIF1A), the pathway of which is targeted by kidney cancer susceptibility genes. We therefore proposed that MITF might have a role in conferring a genetic predisposition to co-occurring melanoma and RCC. Here we identify a germline missense substitution in MITF (Mi-E318K) that occurred at a significantly higher frequency in genetically enriched patients affected with melanoma, RCC or both cancers, when compared with controls. Overall, Mi-E318K carriers had a higher than fivefold increased risk of developing melanoma, RCC or both cancers. Codon 318 is located in a small-ubiquitin-like modifier (SUMO) consensus site (ΨKXE) and Mi-E318K severely impaired SUMOylation of MITF. Mi-E318K enhanced MITF protein binding to the HIF1A promoter and increased its transcriptional activity compared to wild-type MITF. Further, we observed a global increase in Mi-E318K-occupied loci. In an RCC cell line, gene expression profiling identified a Mi-E318K signature related to cell growth, proliferation and inflammation. Lastly, the mutant protein enhanced melanocytic and renal cell clonogenicity, migration and invasion, consistent with a gain-of-function role in tumorigenesis. Our data provide insights into the link between SUMOylation, transcription and cancer.


Asunto(s)
Carcinoma de Células Renales/genética , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal , Melanoma/genética , Factor de Transcripción Asociado a Microftalmía/genética , Movimiento Celular/genética , Frecuencia de los Genes , Humanos , Invasividad Neoplásica/genética , Sumoilación
15.
Genes Dev ; 25(12): 1245-61, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21646373

RESUMEN

Melanoma cells can enter the process of senescence, but whether they express a secretory phenotype, as reported for other cells, is undetermined. This is of paramount importance, because this secretome can alter the tumor microenvironment and the response to chemotherapeutic drugs. More generally, the molecular events involved in formation of the senescent-associated secretome have yet to be determined. We reveal here that melanoma cells experiencing senescence in response to diverse stimuli, including anti-melanoma drugs, produce an inflammatory secretory profile, where the chemokine ligand-2 (CCL2) acts as a critical effector. Thus, we reveal how senescence induction might be involved in therapeutic failure in melanoma. We further provide a molecular relationship between senescence induction and secretome formation by revealing that the poly(ADP-ribose) polymerase-1 (PARP-1)/nuclear factor-κB (NF-κB) signaling cascade, activated during senescence, drives the formation of a secretome endowed with protumoral and prometastatic properties. Our findings also point to the existence of the PARP-1 and NF-κB-associated secretome, termed the PNAS, in nonmelanoma cells. Most importantly, inhibition of PARP-1 or NF-κB prevents the proinvasive properties of the secretome. Collectively, identification of the PARP-1/NF-κB axis in secretome formation opens new avenues for therapeutic intervention against cancers.


Asunto(s)
FN-kappa B/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Línea Celular Tumoral , Senescencia Celular , Quimiocina CCL2/metabolismo , Daño del ADN , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/fisiopatología , Invasividad Neoplásica/patología , Poli(ADP-Ribosa) Polimerasa-1 , Transducción de Señal
16.
Cancer Res ; 70(9): 3813-22, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20388797

RESUMEN

Apoptosis and senescence are cellular failsafe programs that counteract excessive mitogenic signaling observed in cancer cells. Melanoma is known for its notorious resistance to apoptotic processes; therefore, senescence, which remains poorly understood in melanomas, can be viewed as a therapeutic alternative. Microphthalmia-associated transcription factor (MITF), in which its M transcript is specifically expressed in melanocyte cells, plays a critical role in melanoma proliferation, and its specific inhibition is associated with G(0)-G(1) growth arrest. Interestingly, decreased MITF expression has been described in senescent melanocytes, and we have observed an inhibition of MITF expression in melanoma cells exposed to chemotherapeutic drugs that induce their senescence. All these observations thereby question the role of MITF in controlling senescence in melanoma cells. Here, we report that long-term depletion of MITF in melanoma cells triggers a senescence program characterized by typical morphologic and biochemical changes associated with a sustained growth arrest. Further, we show that MITF-silenced cells engage a DNA damage response (DDR) signaling pathway, leading to p53 upregulation, which is critically required for senescence entry. This study uncovers the existence of a lineage-restricted DDR/p53 signaling pathway that is inhibited by MITF to prevent senescence and favor melanoma cell proliferation.


Asunto(s)
Daño del ADN , Melanoma/genética , Factor de Transcripción Asociado a Microftalmía/deficiencia , Animales , Línea Celular Tumoral , Linaje de la Célula/fisiología , Senescencia Celular/fisiología , Humanos , Melanoma/metabolismo , Melanoma/patología , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Factor de Transcripción Asociado a Microftalmía/genética , Factor de Transcripción Asociado a Microftalmía/metabolismo , Mitosis/genética , Transducción de Señal , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
17.
J Biol Chem ; 284(28): 18699-706, 2009 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-19389708

RESUMEN

Melanins are synthesized in melanocytes within specialized organelles called melanosomes. Numerous studies have shown that the pH of melanosome plays a key role in the regulation of melanin synthesis. However, until now, acute regulation of melanosome pH by a physiological stimulus has never been demonstrated. In the present study, we show that the activation of the cAMP pathway by alphaMSH or forskolin leads to an alkalinization of melanosomes and a concomitant regulation of vacuolar ATPases and ion transporters of the solute carrier family. The solute carrier family members include SLC45A2, which is mutated in oculocutaneous albinism type IV, SLC24A4 and SLC24A5, proteins implicated in the control of eye, hair, and skin pigmentation, and the P protein, encoded by the oculocutaneous albinism type II locus. Interestingly, H89, a pharmacological inhibitor of protein kinase A (PKA), prevents the cAMP-induced pigmentation and induces acidification of melanosomes. The drastic depigmenting effect of H89 is not due to an inhibition of tyrosinase expression. Indeed, H89 blocks the induction of melanogenesis induced by LY294002, a potent inhibitor of the PI 3-kinase pathway, without any effect on tyrosinase expression. Furthermore, PKA is not involved in the inhibition of pigmentation promoted by H89 because LY294002 induces pigmentation independently of PKA. Also, other PKA inhibitors do not affect pigmentation. Taken together, our results strengthen the support for a key role of melanosome pH in the regulation of melanin synthesis and, for the first time, demonstrate that melanosome pH is regulated by cAMP and alphaMSH. Notably, these are both mediators of the response to solar UV radiation, the main physiological stimulus of skin pigmentation.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/química , AMP Cíclico/metabolismo , Melanosomas/metabolismo , alfa-MSH/metabolismo , Animales , Cromonas/farmacología , Inhibidores Enzimáticos/farmacología , Regulación Enzimológica de la Expresión Génica , Humanos , Concentración de Iones de Hidrógeno , Isoquinolinas/farmacología , Melaninas/química , Melanoma Experimental , Ratones , Modelos Biológicos , Monofenol Monooxigenasa/antagonistas & inhibidores , Monofenol Monooxigenasa/metabolismo , Morfolinas/farmacología , Pigmentación de la Piel , Sulfonamidas/farmacología
18.
J Biol Chem ; 283(18): 12635-42, 2008 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-18281284

RESUMEN

Melanosomes are lysosome-related organelles specialized in melanin synthesis and transport. In this study, we show that microphthalmia-associated transcription factor (MITF) silencing induces melanosome gathering around the nucleus and causes the relocalization of Rab27A, Slac2a-Mlph, and Myo5a that control the transport of melanosomes on the actin network. In an attempt to elucidate the mechanism by which MITF controls melanosome distribution, we identify RAB27A as a new MITF target gene. Indeed, MITF silencing leads to a dramatic decrease in Rab27A expression and blocks the stimulation of Rab27A expression evoked by cAMP. Further, forced expression of MITF increases Rab27A expression, indicating that MITF is required and sufficient for Rab27A expression in melanoma cells. MITF binds to two E-boxes in the proximal region of the Rab27A promoter and stimulates its transcriptional activity. Finally, re-expression of Rab27A, in MITF-depleted cells, restores the transport of melanosomes to the cell periphery. These results show that RAB27A is a new direct transcriptional target of MITF and link MITF to melanosome transport, another key parameter of melanocyte differentiation and skin pigmentation. Interestingly, Rab27A is involved in other fundamental physiological functions, such as the transport of lytic granules and insulin secretion. Thus our results, deciphering the mechanism of Rab27A transcriptional regulation, have an interest that goes beyond the skin pigmentation field.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Melanosomas/metabolismo , Factor de Transcripción Asociado a Microftalmía/metabolismo , Proteínas de Unión al GTP rab/genética , Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Secuencia de Bases , Transporte Biológico , Línea Celular Tumoral , Elementos E-Box/genética , Silenciador del Gen , Humanos , Ratones , Factor de Transcripción Asociado a Microftalmía/deficiencia , Datos de Secuencia Molecular , Miosina Tipo V/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica , Transcripción Genética , Proteínas de Unión al GTP rab/metabolismo , Proteínas rab27 de Unión a GTP
19.
J Invest Dermatol ; 128(6): 1499-505, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18079751

RESUMEN

IGF1 plays a key role in the development and growth of multiple tumors and in the prevention of apoptosis. In melanoma cells, IGF1 has been shown to mediate resistance to anoikis-induced apoptosis. However, the effect of IGF1 on other proapoptotic stimuli has never been reported. Further, the molecular mechanisms by which IGF1 mediates its prosurvival properties in melanoma cells remain unknown. Here, we demonstrate that IGF1 impairs the onset of tumor necrosis factor-related apoptosis-inducing ligand and staurosporine-induced apoptosis in melanoma cells expressing either wild-type or oncogenic B-Raf. Further, we show that IGF1 inhibits mitochondrial damage that occurs during apoptosis, thereby indicating that IGF1 acts at the level of mitochondria to mediate its antiapoptotic stimuli. Accordingly, IGF1 increases the mRNA levels and protein expression of antiapoptotic members of the BCL2 family--BCL2 and BCL-X(L)--and that of the inhibitor of apoptosis protein, survivin. Further, their specific silencing by small interfering RNA prevents the protective effect of IGF1. These findings therefore delineate the molecular mechanisms by which IGF1 mediates its prosurvival properties and provide a basis for clinical strategies designed to neutralize IGF1 or its target genes.


Asunto(s)
Apoptosis , Regulación Neoplásica de la Expresión Génica , Factor I del Crecimiento Similar a la Insulina/fisiología , Melanoma/metabolismo , Melanoma/patología , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína bcl-X/metabolismo , Línea Celular Tumoral , Inhibidores Enzimáticos/farmacología , Humanos , Proteínas Inhibidoras de la Apoptosis , Factor I del Crecimiento Similar a la Insulina/metabolismo , Modelos Biológicos , Fosforilación , Proteínas Proto-Oncogénicas B-raf/metabolismo , Estaurosporina/farmacología , Survivin , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo
20.
J Biol Chem ; 282(19): 14140-7, 2007 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-17371876

RESUMEN

The MET proto-oncogene encodes for the hepatocyte growth factor (HGF) receptor, a plasma membrane tyrosine kinase that is involved in melanocyte growth and melanoma development. In mouse melanoma cells, Met expression is increased by alphaMSH via the activation of the cAMP pathway. However, the mechanism by which cAMP regulates MET and the biological consequences of this increase were not known. In the present report, we show that alphaMSH regulates MET expression in both human melanocytes and mouse melanoma cells through a transcriptional mechanism that requires MITF. Furthermore, the adenovirus driven expression of MITF is sufficient to increase MET in melanoma cells. Functional analysis of the MET promoter allows us to identify an E-box motif conserved in both human and mouse promoter that mediates the effect of MITF. Interestingly, up-regulation of MET expression by cAMP leads to an exacerbated HGF signaling and allows HGF to protect melanocytes and melanoma cells from apoptosis. Thus, physiological stimuli or pathological events that would induce MITF expression may lead to increased MET expression thereby favoring melanoma survival. These observations strengthen the roles of MITF and MET in melanoma development.


Asunto(s)
Apoptosis , Factor de Crecimiento de Hepatocito/metabolismo , Melanocitos/efectos de los fármacos , Melanoma Experimental/tratamiento farmacológico , Factor de Transcripción Asociado a Microftalmía/farmacología , Proteínas Proto-Oncogénicas c-met/metabolismo , alfa-MSH/farmacología , Adenoviridae/genética , Animales , Secuencia de Bases , Western Blotting , Núcleo Celular/metabolismo , Células Cultivadas , Cloranfenicol O-Acetiltransferasa/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Prepucio/citología , Prepucio/metabolismo , Regulación de la Expresión Génica , Humanos , Recién Nacido , Luciferasas/metabolismo , Masculino , Melanocitos/metabolismo , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Datos de Secuencia Molecular , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-met/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Ácido Nucleico , Transfección , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA