Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 66
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Oncogene ; 36(48): 6659-6667, 2017 11 30.
Article de Anglais | MEDLINE | ID: mdl-28825724

RÉSUMÉ

Cutaneous melanoma is a deadly skin cancer that originates from melanocytes. The development of cutaneous melanoma involves a complex interaction between environmental factors, mainly ultraviolet radiation from sunlight, and genetic alterations. Melanoma can also occur from a pre-existing nevus, a benign lesion formed from melanocytes harboring oncogenic mutations that trigger proliferative arrest and senescence entry. Senescence is a potent barrier against tumor progression. As such, the acquisition of mutations that suppress senescence and promote cell division is mandatory for cancer development. This topic appears central to melanoma development because, in humans, several somatic and germline mutations are related to the control of cellular senescence and proliferative activity. Consequently, primary melanoma can be viewed as a paradigm of senescence evasion. In support of this notion, a sumoylation-defective germline mutation in microphthalmia-associated transcription factor (MITF), a master regulator of melanocyte homeostasis, is associated with the development of melanoma. Interestingly, this MITF variant has also been recently reported to negatively impact the program of senescence. This article reviews the genetic alterations that have been shown to be involved in melanoma and that alter the process of senescence to favor melanoma development. Then, the transcription factor MITF and its sumoylation-defective mutant are described. How sumoylation misregulation can change MITF activity and impact the process of senescence is discussed. Finally, the contribution of such information to the development of anti-malignant melanoma strategies is evaluated.


Sujet(s)
Mélanome/métabolisme , Facteur de transcription associé à la microphtalmie/métabolisme , Tumeurs cutanées/métabolisme , Sumoylation , Animaux , Vieillissement de la cellule , Humains , Mélanome/anatomopathologie , Mélanome/thérapie , Transduction du signal , Tumeurs cutanées/anatomopathologie , Tumeurs cutanées/thérapie
3.
Oncogene ; 35(21): 2735-45, 2016 05.
Article de Anglais | MEDLINE | ID: mdl-26364600

RÉSUMÉ

Enhancer of Zeste homologue 2 (EZH2) belongs to the polycomb repressive complex 2 and catalyzes the methylation of histone H3 lysine 27. These pivotal epigenetic marks are altered in many cancers, including melanoma, as a result of EZH2 overexpression. Here, we show that the non-canonical-NF-kB pathway accounts for most of the NF-kB activity in melanoma cells, in contrast to non-cancer cells. We identify the non-canonical-NF-kB pathway as a key regulator of EZH2 expression in melanoma. We show a striking correlation between NF-kB2 and EZH2 expression in human melanoma metastases. We demonstrate that inhibition of the non-canonical NF-kB pathway by targeting NF-kB2/p52 or the upstream kinase NIK restores the senescence program in melanoma cells through the decrease of EZH2. On the contrary, the overexpression of NF-kB2/p52 in normal human melanocytes prevents stress- and oncogene-induced senescence. Finally, we show in mouse models that the inhibition of the non-canonical NF-kB pathway restores senescence and induces a dramatic reduction in tumor growth compared with controls, thus providing potential drug targets for the re-induction of senescence in melanoma and other cancers where EZH2 is overexpressed.


Sujet(s)
Protéine-2 homologue de l'activateur de Zeste/génétique , Mélanome/génétique , Mélanome/métabolisme , Animaux , Lignée cellulaire tumorale , Régulation négative , Protéine-2 homologue de l'activateur de Zeste/métabolisme , Hétérogreffes , Humains , Mélanome/anatomopathologie , Souris , Souris nude , Sous-unité p52 de NF-kappa B/biosynthèse , Sous-unité p52 de NF-kappa B/génétique , Sous-unité p52 de NF-kappa B/métabolisme , Activation de la transcription
5.
Oncogene ; 31(19): 2461-70, 2012 May 10.
Article de Anglais | MEDLINE | ID: mdl-21996743

RÉSUMÉ

Melanomas are very aggressive neoplasms with notorious resistance to therapeutics. It was recently proposed that the remarkable phenotypic plasticity of melanoma cells allows for the rapid development of both resistance to chemotherapeutic drugs and invasive properties. Indeed, the capacity of melanoma cells to form distant metastases is the main cause of mortality in melanoma patients. Therefore, the identification of the mechanism controlling melanoma phenotype is of paramount importance. In the present report, we show that deletion of microphthalmia-associated transcription factor (MITF), the master gene in melanocyte differentiation, is sufficient to increase the metastatic potential of mouse and human melanoma cells. MITF silencing also increases fibronectin and Snail, two mesenchymal markers that might explain the increased invasiveness in vitro and in vivo. Furthermore, ablation of this population by Forskolin-induced differentiation or MITF-forced expression significantly decreases tumour and metastasis formation, suggesting that eradication of low-MITF cells might improve melanoma treatment. Moreover, we demonstrate that a hypoxic microenvironment decreases MITF expression through an indirect, hypoxia-inducible factor 1 (HIF1)α-dependant transcriptional mechanism, and increases the tumourigenic and metastatic properties of melanoma cells. We identified Bhlhb2, a new factor in melanoma biology, as the mediator of hypoxia/HIF1α inhibitory effect on MITF expression. Our results reveal a hypoxia-HIF1α-BHLHB2-MITF cascade controlling the phenotypic plasticity in melanoma cells and favouring metastasis development. Targeting this pathway might be helpful in the design of new anti-melanoma therapies.


Sujet(s)
Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Mélanome/secondaire , Facteur de transcription associé à la microphtalmie/métabolisme , Tumeurs cutanées/anatomopathologie , Animaux , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Hypoxie cellulaire , Extinction de l'expression des gènes , Humains , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Mélanome/métabolisme , Mélanome expérimental/métabolisme , Mélanome expérimental/secondaire , Mésoderme/métabolisme , Mésoderme/anatomopathologie , Souris , Facteur de transcription associé à la microphtalmie/antagonistes et inhibiteurs , Facteur de transcription associé à la microphtalmie/génétique , Invasion tumorale , Métastase tumorale , Tumeurs cutanées/métabolisme
6.
Cell Death Dis ; 2: e199, 2011 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-21881601

RÉSUMÉ

Metformin is the most widely used antidiabetic drug because of its proven efficacy and limited secondary effects. Interestingly, recent studies have reported that metformin can block the growth of different tumor types. Here, we show that metformin exerts antiproliferative effects on melanoma cells, whereas normal human melanocytes are resistant to these metformin-induced effects. To better understand the basis of this antiproliferative effect of metformin in melanoma, we characterized the sequence of events underlying metformin action. We showed that 24 h metformin treatment induced a cell cycle arrest in G0/G1 phases, while after 72 h, melanoma cells underwent autophagy as demonstrated by electron microscopy, immunochemistry, and by quantification of the autolysosome-associated LC3 and Beclin1 proteins. In addition, 96 h post metformin treatment we observed robust apoptosis of melanoma cells. Interestingly, inhibition of autophagy by knocking down LC3 or ATG5 decreased the extent of apoptosis, and suppressed the antiproliferative effect of metformin on melanoma cells, suggesting that apoptosis is a consequence of autophagy. The relevance of these observations were confirmed in vivo, as we showed that metformin treatment impaired the melanoma tumor growth in mice, and induced autophagy and apoptosis markers. Taken together, our data suggest that metformin has an important impact on melanoma growth, and may therefore be beneficial in patients with melanoma.


Sujet(s)
Antinéoplasiques/pharmacologie , Antinéoplasiques/toxicité , Apoptose/effets des médicaments et des substances chimiques , Autophagie/effets des médicaments et des substances chimiques , Metformine/pharmacologie , Metformine/toxicité , Animaux , Protéines régulatrices de l'apoptose/métabolisme , Protéine-5 associée à l'autophagie , Bécline-1 , Lignée cellulaire tumorale , Phase G1 , Humains , Mélanome/traitement médicamenteux , Mélanome/métabolisme , Protéines membranaires/métabolisme , Souris , Souris nude , Protéines associées aux microtubules/antagonistes et inhibiteurs , Protéines associées aux microtubules/génétique , Protéines associées aux microtubules/métabolisme , Interférence par ARN , Petit ARN interférent/métabolisme , Phase G0 , Transplantation hétérologue
7.
Oncogene ; 30(49): 4887-900, 2011 Dec 08.
Article de Anglais | MEDLINE | ID: mdl-21685937

RÉSUMÉ

Aberrant expression of Secreted Protein Acidic and Rich in Cysteine (SPARC)/osteonectin has been associated with an invasive tumor cell phenotype and poor outcome in human melanomas. Although it is known that SPARC controls melanoma tumorigenesis, the precise role of SPARC in melanoma cell survival is still unclear. Here, we show that SPARC has a cell-autonomous survival activity, which requires Akt-dependent regulation of p53. Suppression of SPARC by RNA interference in several human melanoma cells and xenografted A375 tumors triggers apoptotic cell death through the mitochondrial intrinsic pathway and activation of caspase-3. Cell death induced by depletion of SPARC is dependent on p53 and induction of Bax, and results in the generation of ROS. Stabilization of p53 in SPARC-depleted cells is associated with a decrease in Akt-mediated activating phosphorylation of MDM2. Inhibition of Akt signaling pathway is important for the observed changes as overexpression of constitutively active Akt protects cells against apoptosis induced by SPARC depletion. Conversely, increased expression of SPARC stimulates Akt and MDM2 phosphorylation, thus facilitating p53 degradation. Finally, we show that overexpression of SPARC renders cells more resistant to the p53-mediated cytotoxic effects of the DNA-damaging drug actinomycin-D. Our study indicates that SPARC functions through activation of Akt and MDM2 to limit p53 levels and that acquired expression of SPARC during melanoma development would confer survival advantages through suppression of p53-dependent apoptotic pathways.


Sujet(s)
Mélanome/anatomopathologie , Ostéonectine/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-mdm2/métabolisme , Protéine p53 suppresseur de tumeur/métabolisme , Animaux , Antinéoplasiques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Dactinomycine/pharmacologie , Techniques de knock-down de gènes , Humains , Mélanome/génétique , Mélanome/métabolisme , Souris , Ostéonectine/déficit , Ostéonectine/génétique , Stress oxydatif/effets des médicaments et des substances chimiques , Phosphorylation/effets des médicaments et des substances chimiques , Liaison aux protéines/effets des médicaments et des substances chimiques , Interférence par ARN , Espèces réactives de l'oxygène/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Facteurs temps , Régulation positive/effets des médicaments et des substances chimiques , Protéine Bax/métabolisme
8.
Oncogene ; 30(20): 2307-18, 2011 May 19.
Article de Anglais | MEDLINE | ID: mdl-21278797

RÉSUMÉ

In melanoma, as well as in other solid tumors, the cells within a given tumor exhibit strong morphological, functional and molecular heterogeneity that might reflect the existence of different cancer cell populations, among which are melanoma-initiating cells (MICs) with 'stemness' properties and their differentiated, fast-growing progeny. The existence of a slow-growing population might explain the resistance of melanoma to classical chemotherapies that target fast growing cells. Therefore, elucidating the biologic properties of MICs and, more importantly, the molecular mechanisms that drive the transition between MICs and their proliferating progeny needs to be addressed to develop an efficient melanoma therapy. Using B16 mouse melanoma cells and syngeneic mice, we show that the inhibition of microphthalmia-associated transcription factor (Mitf), the master regulator of melanocyte differentiation, increases the tumorigenic potential of melanoma cells and upregulates the stem cell markers Oct4 and Nanog. Notably, p27, the CDK inhibitor, is increased in Mitf-depleted cells and is required for exacerbation of the tumorigenic properties of melanoma cells. Further, a slow-growing population with low-Mitf level and high tumorigenic potential exists spontaneously in melanoma. Ablation of this population dramatically decreases tumor formation. Importantly, these data were confirmed using human melanoma cell lines and freshly isolated human melanoma cell from lymph node and skin melanoma metastasis. Taken together our data, identified Mitf and p27 as the key molecular switches that control the transition between MICs and their differentiated progeny. Eradication of low-Mitf cells might be an appealing strategy to cure melanoma.


Sujet(s)
Mélanome expérimental/métabolisme , Mélanome/métabolisme , Facteur de transcription associé à la microphtalmie/métabolisme , Tumeurs cutanées/métabolisme , Animaux , Différenciation cellulaire/génétique , Division cellulaire , Lignée cellulaire tumorale , Transdifférenciation cellulaire , Inhibiteur p27 de kinase cycline-dépendante/métabolisme , Extinction de l'expression des gènes , Humains , Métastase lymphatique , Mélanome/anatomopathologie , Mélanome expérimental/anatomopathologie , Souris , Souris de lignée C57BL , Facteur de transcription associé à la microphtalmie/génétique , Transplantation tumorale , Cellules souches tumorales/anatomopathologie , Tumeurs cutanées/anatomopathologie
9.
Oncogene ; 30(20): 2319-32, 2011 May 19.
Article de Anglais | MEDLINE | ID: mdl-21258399

RÉSUMÉ

Malignant melanoma is an aggressive cancer known for its notorious resistance to most current therapies. The basic helix-loop-helix microphthalmia transcription factor (MITF) is the master regulator determining the identity and properties of the melanocyte lineage, and is regarded as a lineage-specific 'oncogene' that has a critical role in the pathogenesis of melanoma. MITF promotes melanoma cell proliferation, whereas sustained supression of MITF expression leads to senescence. By combining chromatin immunoprecipitation coupled to high throughput sequencing (ChIP-seq) and RNA sequencing analyses, we show that MITF directly regulates a set of genes required for DNA replication, repair and mitosis. Our results reveal how loss of MITF regulates mitotic fidelity, and through defective replication and repair induces DNA damage, ultimately ending in cellular senescence. These findings reveal a lineage-specific control of DNA replication and mitosis by MITF, providing new avenues for therapeutic intervention in melanoma. The identification of MITF-binding sites and gene-regulatory networks establish a framework for understanding oncogenic basic helix-loop-helix factors such as N-myc or TFE3 in other cancers.


Sujet(s)
Réparation de l'ADN/génétique , Réplication de l'ADN , Régulation de l'expression des gènes tumoraux , Instabilité du génome , Mélanome/génétique , Facteur de transcription associé à la microphtalmie/métabolisme , Mitose/génétique , Tumeurs cutanées/génétique , Sites de fixation , Lignée cellulaire tumorale , Lignage cellulaire , Vieillissement de la cellule , Techniques de knock-out de gènes , Humains , Facteur de transcription associé à la microphtalmie/génétique , Métastase tumorale , Tumeurs cutanées/métabolisme
10.
Cell Death Differ ; 18(1): 109-21, 2011 Jan.
Article de Anglais | MEDLINE | ID: mdl-20596077

RÉSUMÉ

We have previously demonstrated that the thiazolidinedione ciglitazone inhibited, independently of PPARγ activation, melanoma cell growth. Further investigations now show that ciglitazone effects are mediated through the regulation of secreted factors. Q-PCR screening of several genes involved in melanoma biology reveals that ciglitazone inhibits expression of the CXCL1 chemokine gene. CXCL1 is overexpressed in melanoma and contributes to tumorigenicity. We show that ciglitazone induces a diminution of CXCL1 level in different human melanoma cell lines. This effect is mediated by the downregulation of microphthalmia-associated transcription factor, MITF, the master gene in melanocyte differentiation and involved in melanoma development. Further, recombinant CXCL1 protein is sufficient to abrogate thiazolidinedione effects such as apoptosis induction, whereas extinction of the CXCL1 pathway mimics phenotypic changes observed in response to ciglitazone. Finally, inhibition of human melanoma tumor development in nude mice treated with ciglitazone is associated with a strong decrease in MITF and CXCL1 levels. Our results show that anti-melanoma effects of thiazolidinediones involve an inhibition of the MITF/CXCL1 axis and highlight the key role of this specific pathway in melanoma malignancy.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Chimiokine CXCL1/métabolisme , Mélanome/traitement médicamenteux , Facteur de transcription associé à la microphtalmie/métabolisme , Thiazolidinediones/usage thérapeutique , Animaux , Apoptose , Différenciation cellulaire , Lignée cellulaire tumorale , Chimiokine CXCL1/génétique , Chimiokine CXCL1/pharmacologie , Régulation négative , Humains , Mélanome/métabolisme , Souris , Souris nude , Facteur de transcription associé à la microphtalmie/antagonistes et inhibiteurs , Facteur de transcription associé à la microphtalmie/physiologie , Interférence par ARN , Petit ARN interférent/métabolisme , Protéines recombinantes/génétique , Protéines recombinantes/métabolisme , Protéines recombinantes/pharmacologie , Transduction du signal , Transplantation hétérologue
11.
Pigment Cell Melanoma Res ; 21(2): 139-46, 2008 Apr.
Article de Anglais | MEDLINE | ID: mdl-18426407

RÉSUMÉ

Melanoma is a highly aggressive tumour characterized by a strong resistance to apoptotic stimuli that give rise to a selective advantage for tumour progression and metastasis formation. Therefore, it is of paramount importance to better understand the mechanisms involved in this resistance to apoptosis. In this report, we focused our attention on FKHRL1, a member of the forkhead family of transcription factors, which controls expression of genes involved in cell cycle progression and apoptosis. In melanoma cells, we show that IGF1, which exerts pro-survival properties, induces the phosphorylation and nuclear exclusion of FKHRL1 in a PI3K/AKT-dependent pathway. Moreover, we observe that over-expression of a non-phosphorylable mutant of FKHRL1 (FKHRL1-TM), constitutively localized to the nucleus, promotes apoptotic cell death of melanoma cells. Finally, we find that FKHRL1-TM decreases the expression of survivin, a member of the inhibitor of apoptosis protein and that survivin re-expression partially rescues the deleterious effects of FKHRL1. Taken together, these findings reveal, in melanoma cells, that endogenous FKHRL1 is a downstream target of the PI3K/AKT pathway and suggest that the phosphorylation of this transcription factor may be involved in the pro-survival effects of growth factors such as IGF1. On the other hand, forced nuclear localization of FKHRL1 decreases melanoma cell growth and may serve as a therapeutic strategy against melanoma.


Sujet(s)
Apoptose , Facteurs de transcription Forkhead/métabolisme , Mélanome/métabolisme , Apoptose/génétique , Technique de Western , Lignée cellulaire , Lignée cellulaire tumorale , Survie cellulaire/génétique , Cellules cultivées , Protéine O3 à motif en tête de fourche , Facteurs de transcription Forkhead/génétique , Humains , Protéines IAP , Facteur de croissance IGF-I/métabolisme , Mélanome/génétique , Protéines associées aux microtubules/métabolisme , Protéines tumorales/métabolisme , Réaction de polymérisation en chaîne , Transduction du signal , Survivine
12.
Cell Death Differ ; 11(10): 1084-91, 2004 Oct.
Article de Anglais | MEDLINE | ID: mdl-15243584

RÉSUMÉ

Melanocytes are cells of the epidermis that synthesize melanin, which is responsible for skin pigmentation. Transformation of melanocytes leads to melanoma, a highly aggressive neoplasm, which displays resistance to apoptosis. In this report, we demonstrate that TNF-related apoptosis-inducing ligand (TRAIL), which was thought to kill only transformed cells, promotes very efficiently apoptosis of primary human melanocytes, leading to activation of caspases 8, 9 and 3, and the cleavage of vital proteins. Further, we show that stem cell factor (SCF), a physiologic melanocyte growth factor that activates both the phosphatidyl-inositol-3 kinase (PI3K) and the extracellular regulated kinase (ERK) pathways, strongly protects melanocytes from TRAIL and staurosporine killing. Interestingly, inhibition of PI3K or its downstream target AKT completely blocks the antiapoptotic effect of SCF, while inhibition of ERK has only a moderate effect. Our data indicate that protection evoked by SCF/PI3K/AKT cascade is not mediated by an increase in the intracellular level of FLIP. Further, only a sustained PI3K activity can protect melanocytes from apoptosis, thereby indicating that the PI3K/AKT pathway plays a pivotal role in melanocyte survival. The results gathered in this report bring new information on the molecular mechanisms involved in primary melanocyte apoptosis and survival that would help to better understand the process by which melanomas acquire their resistance to apoptosis.


Sujet(s)
Apoptose/effets des médicaments et des substances chimiques , Mélanocytes/effets des médicaments et des substances chimiques , Glycoprotéines membranaires/pharmacologie , Phosphatidylinositol 3-kinases/métabolisme , Facteur de nécrose tumorale alpha/pharmacologie , Protéines régulatrices de l'apoptose , Cellules cultivées , Activation enzymatique , Antienzymes/pharmacologie , Extracellular Signal-Regulated MAP Kinases/antagonistes et inhibiteurs , Extracellular Signal-Regulated MAP Kinases/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Humains , Kératinocytes/effets des médicaments et des substances chimiques , Kératinocytes/métabolisme , Mélanocytes/cytologie , Mélanocytes/enzymologie , Mélanocytes/métabolisme , Mélanome/anatomopathologie , Inhibiteurs des phosphoinositide-3 kinases , Protein-Serine-Threonine Kinases/métabolisme , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes c-akt , Transduction du signal/effets des médicaments et des substances chimiques , Facteur de croissance des cellules souches/pharmacologie , Ligand TRAIL
13.
Pigment Cell Res ; 14(5): 328-36, 2001 Oct.
Article de Anglais | MEDLINE | ID: mdl-11601654

RÉSUMÉ

Cells of the vertebrate neural crest (crest cells) differentiate in vitro to melanocytes and sympathoadrenal (SA) progenitor cells. We have shown previously, using primary J. quail neural crest cultures, the combinatorial effect of bone morphogenetic protein-2 (BMP-2) and cAMP signaling on SA cell development. Herein, we report that in primary J. quail neural crest cultures, BMP-2 and cAMP signaling similarly exert a combinatorial effect on melanocyte development. We demonstrate that BMP-2 treatment of neural crest cells increases melanogenesis by promoting the synthesis of melanin. This increased melanin synthesis by BMP-2 is effected by the selective increase in the transcription of the tyrosinase gene, encoding the rate-limiting enzyme of the melanin biosynthetic pathway. By contrast, BMP-2 exerts no effect on the expression of the tyrosine-related proteins 1 and 2 (Tyrpl and Dct), also involved in the melanin biosynthetic process, or on the expression of microphalmia (Mitf) gene, supporting the fact that BMP-2 does not affect melanocyte differentiation. Employing transient transfection analysis of tyrosinase-reporter constructs in B16 melanoma cells, we demonstrate that the BMP-2 response-element is localized between 900 and 1,100 bp upstream from the tyrosinase transcriptional start site. These studies support a role for BMP-2 in melanogenesis by selectively targeting the expression of the tyrosinase gene involved in melanin biosynthesis.


Sujet(s)
Protéines morphogénétiques osseuses/pharmacologie , Régulation de l'expression des gènes codant pour des enzymes , Mélanines/biosynthèse , Mélanocytes/physiologie , Monophenol monooxygenase/génétique , Crête neurale/effets des médicaments et des substances chimiques , Facteur de croissance transformant bêta , Xanthine(isobutyl-3 methyl-1)/pharmacologie , 8-Bromo AMP cyclique/pharmacologie , Animaux , Protéine morphogénétique osseuse de type 2 , Différenciation cellulaire , Cellules cultivées , Colforsine/pharmacologie , Coturnix , AMP cyclique/métabolisme , Mélanocytes/effets des médicaments et des substances chimiques , Mélanocytes/enzymologie , Mélanocytes/ultrastructure , Souris , Monophenol monooxygenase/métabolisme , Crête neurale/cytologie , Crête neurale/enzymologie , Crête neurale/physiologie , Inhibiteurs de la phosphodiestérase/pharmacologie , Pigmentation/physiologie , Transduction du signal/physiologie
14.
J Cell Biol ; 152(4): 843-50, 2001 Feb 19.
Article de Anglais | MEDLINE | ID: mdl-11266474

RÉSUMÉ

Normal pigmentation depends on the uniform distribution of melanin-containing vesicles, the melanosomes, in the epidermis. Griscelli syndrome (GS) is a rare autosomal recessive disease, characterized by an immune deficiency and a partial albinism that has been ascribed to an abnormal melanosome distribution. GS maps to 15q21 and was first associated with mutations in the myosin-V gene. However, it was demonstrated recently that GS can also be caused by a mutation in the Rab27a gene. These observations prompted us to investigate the role of Rab27a in melanosome transport. Using immunofluorescence and immunoelectron microscopy studies, we show that in normal melanocytes Rab27a colocalizes with melanosomes. In melanocytes isolated from a patient with GS, we show an abnormal melanosome distribution and a lack of Rab27a expression. Finally, reexpression of Rab27a in GS melanocytes restored melanosome transport to dendrite tips, leading to a phenotypic reversion of the diseased cells. These results identify Rab27a as a key component of vesicle transport machinery in melanocytes.


Sujet(s)
Hypopigmentation/métabolisme , Déficits immunitaires/métabolisme , Mélanocytes/métabolisme , Mélanosomes/métabolisme , Myosine de type V , Protéines G rab/métabolisme , Transport biologique , Protéines de liaison à la calmoduline/isolement et purification , Compartimentation cellulaire , Technique d'immunofluorescence , Humains , Mélanocytes/ultrastructure , Mélanosomes/ultrastructure , Microscopie immunoélectronique , Moteurs moléculaires , Protéines de tissu nerveux/isolement et purification , Syndrome , Protéines G rab/génétique , Protéines G rab/isolement et purification , Protéines G rab/ultrastructure , Protéines rab27 liant le GTP
15.
J Biol Chem ; 275(48): 37978-83, 2000 Dec 01.
Article de Anglais | MEDLINE | ID: mdl-10973953

RÉSUMÉ

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


Sujet(s)
Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/physiologie , Surdité/génétique , Régulation de l'expression des gènes/physiologie , Protéines HMG/physiologie , Troubles de la pigmentation/génétique , Régions promotrices (génétique) , Facteurs de transcription , Syndrome de Waardenburg/génétique , Animaux , Séquence nucléotidique , Amorces ADN , Souris , Facteur de transcription associé à la microphtalmie , Facteurs de transcription SOX-E , Cellules cancéreuses en culture
16.
J Invest Dermatol ; 115(3): 381-7, 2000 Sep.
Article de Anglais | MEDLINE | ID: mdl-10951272

RÉSUMÉ

Compelling evidence suggest a role for melanocortins in the regulation of melanogenesis by ultraviolet radiation. Within the epidermis, melanocytes and keratinocytes produce alpha-melanocyte-stimulating hormone and adrenocorticotropic hormone. The persistence and the strength of the biologic signal delivered by these peptides depend on their local concentration, which is controlled by the rate of peptide production and by the rate of its degradation. In this study, we investigated the mechanism of melanocortin degradation by melanocytes and the effect of ultraviolet on this process. We have focused our attention on a neutral endopeptidase, neprilysin, which has been implicated in the ending of numerous peptidergic signals. We have shown that this enzyme is expressed at the surface of human melanocytes. Interestingly, its activity and its expression are dramatically downregulated by ultraviolet B treatment. Moreover, in the presence of phosphoramidon, a stable inhibitor of neprilysin, we observed an increased efficiency of alpha-melanocyte-stimulating hormone and adrenocorticotropic hormone to stimulate both tyrosinase activity and microphthalmia expression. Taken together, these data indicate that neprilysin expressed by melanocytes has a physiologic role in the regulation of melanogenesis by proopiomelanocortin peptide. Further, its downregulation by ultraviolet B irradiation shed light on a new and appealing mechanism of ultraviolet B induced melanogenesis via the control of melanocortins degradation.


Sujet(s)
Mélanines/biosynthèse , Mélanines/effets des radiations , Néprilysine/effets des radiations , Rayons ultraviolets , Hormone mélanotrope alpha/physiologie , Cellules cultivées , Régulation négative/effets des radiations , Humains , Néprilysine/physiologie , Pigmentation de la peau/effets des radiations
17.
J Biol Chem ; 275(40): 30757-60, 2000 Oct 06.
Article de Anglais | MEDLINE | ID: mdl-10938265

RÉSUMÉ

The absence of melanocytes from the cochlea and epidermis is responsible of deafness and hypopigmentation, two symptoms shared by the four Waardenburg syndrome (WS) subtypes. Microphthalmia-associated transcription factor (MITF) controls melanocyte survival and differentiation. Mutations, which impair MITF function or expression, result in an abnormal melanocyte development leading to the WS2. WS1 and WS3 are caused by mutation in the gene encoding the transcription factor Pax3, which regulates MITF expression. Recently, mutations in SOX10, a gene encoding a SRY-related transcription factor, have been reported in patients with WS4. However, the molecular basis of the defective melanocyte development in these patients remained to be elucidated. In the present report, we demonstrate that Sox10 is a strong activator of the MITF promoter, and we identify a Sox10 binding site between -264 and -266 of the MITF promoter. Finally, we show that three SOX10 mutations found in WS4 abolish the transcriptional activity of the resulting Sox10 proteins toward the MITF promoter. Taken together, our observations bring new and meaningful information concerning the molecular process that leads to a defective melanocyte development in WS4 patients with SOX10 mutations.


Sujet(s)
Protéines de liaison à l'ADN/génétique , Régulation de l'expression des gènes , Protéines HMG/génétique , Syndrome de Waardenburg/génétique , Cellules 3T3 , Animaux , Technique de Western , Codon , Codon non-sens , Protéine de liaison à l'élément de réponse à l'AMP cyclique/métabolisme , Protéines de liaison à l'ADN/biosynthèse , Protéines de liaison à l'ADN/métabolisme , Endothéline-3/génétique , Épistasie , Mutation avec décalage du cadre de lecture , Délétion de gène , Gènes rapporteurs , Protéines HMG/biosynthèse , Humains , Luciferases/métabolisme , Facteur de transcription LEF-1 , Mélanocytes/métabolisme , Souris , Facteur de transcription associé à la microphtalmie , Mutation , Facteur de transcription PAX3 , Facteurs de transcription PAX , Plasmides/métabolisme , Régions promotrices (génétique) , Liaison aux protéines , Facteurs de transcription SOX-E , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Transcription génétique , Activation de la transcription , Transfection , Cellules cancéreuses en culture
18.
EMBO J ; 19(12): 2900-10, 2000 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-10856235

RÉSUMÉ

In melanocytes and melanoma cells, cAMP activates extracellular signal-regulated kinases (ERKs) and MEK-1 by an unknown mechanism. We demonstrate that B-Raf is activated by cAMP in melanocytes. A dominant-negative mutant of B-Raf, but not of Raf-1, blocked the cAMP-induced activation of ERK, indicating that B-Raf is the MEK-1 upstream regulator mediating this cAMP effect. Studies using Clostridium sordelii lethal toxin and Clostridium difficile toxin B have suggested that Rap-1 or Ras might transduce cAMP action. We show that Ras, but not Rap-1, is activated cell-specifically and mediates the cAMP-dependent activation of ERKs, while Rap-1 is not involved in this process in melanocytes. Our results suggest a novel, cell-specific mechanism involving Ras small GTPase and B-Raf kinase as mediators of ERK activation by cAMP. Also, in melanocytes, Ras or ERK activation by cAMP is not mediated through protein kinase A activation. Neither the Ras exchange factor, Son of sevenless (SOS), nor the cAMP-responsive Rap-1 exchange factor, Epac, participate in the cAMP-dependent activation of Ras. These findings suggest the existence of a melanocyte-specific Ras exchange factor directly regulated by cAMP.


Sujet(s)
AMP cyclique/métabolisme , Système de signalisation des MAP kinases , Mélanocytes/métabolisme , Mitogen-Activated Protein Kinases/métabolisme , Protéines G ras/métabolisme , Animaux , Cellules cultivées , Cyclic AMP-Dependent Protein Kinases/métabolisme , Activation enzymatique , Facteurs d'échange de nucléotides guanyliques/génétique , Facteurs d'échange de nucléotides guanyliques/métabolisme , MAP Kinase Kinase Kinases/métabolisme , Mélanome expérimental , Souris , Modèles biologiques , Cellules PC12 , Protéines proto-oncogènes c-raf/métabolisme , Rats , Protéines Son of sevenless/génétique , Protéines Son of sevenless/métabolisme , Protéines G ras/génétique
19.
Pigment Cell Res ; 13(2): 60-9, 2000 Apr.
Article de Anglais | MEDLINE | ID: mdl-10841026

RÉSUMÉ

Compelling evidence has been gathered indicating that pro-opiomelanocortin peptides, alpha-melanocyte stimulating hormone (alpha-MSH) and adrenocorticotropic hormone (ACTH), through the cyclic AMP pathway, play a pivotal role in melanocyte differentiation and in the regulation of melanogenesis. Recently, the molecular events linking cAMP to melanogenesis up-regulation have been elucidated. This cascade involves the activation of protein kinase A and CREB transcription factor, leading to the up-regulation of the expression of Microphthalmia associated transcription factor (MITF). MITF has been found mutated in patients with Waardenburg syndrome 2A, and plays a crucial role in melanocyte development. MITF binds and activates melanogenic gene promoters, thereby increasing their expression which results in an increased melanin synthesis. Beyond this simplified scheme, It appears that melanogenic gene expression is controlled by a complex network of regulation involving other transcription factors such as Brn2, TBX2, PAX3 and SOX10. Further studies are required to better understand the respective roles of these factors in the regulation of melanin synthesis. In addition, other intracellular signaling pathways, like the phosphatidyl inositol 3-kinase pathway, as well as the molecular cascade of events governed by the small GTP-binding protein Rho, seem to be involved in the regulation of melanogenesis and melanocyte dendricity. Finally, it should be mentioned that cAMP activates a melanocyte-specific pathway leading to MAP kinase activation. MAP kinase, ERK2, phosphorylates MITF, thereby targeting the transcription factor to proteasomes for degradation. Thus, in addition to the complex transcriptional regulation, melanogenesis is also subjected to a post-translational regulation that controls MITF or tyrosinase function. Taken together, these complex molecular processes would finally allow a fine tuning of melanocyte differentiation leading to melanin synthesis.


Sujet(s)
AMP cyclique/métabolisme , Systèmes de seconds messagers/physiologie , Pigmentation de la peau/physiologie , Animaux , Humains
20.
Mol Endocrinol ; 14(3): 449-56, 2000 Mar.
Article de Anglais | MEDLINE | ID: mdl-10707962

RÉSUMÉ

Microphthalmia gene encodes a basic helix-loop-helix-leucine zipper (bHLH-Zip) transcription factor involved in the development of the melanocyte lineage and plays a key role in the transcriptional regulation of the melanogenic enzymes, tyrosinase and TyrpI. Recently, we have shown that Microphthalmia mediates the melanogenic effects elicited by alphaMSH that up-regulates the expression of tyrosinase through the activation of the cAMP pathway. Therefore, Microphthalmia appears as a principal gene in melanocyte development and functioning. Among the transcription factors of the bHLH-Zip family, TFE3 and TFEB show a remarkably elevated homology with Microphthalmia. These observations prompted us to investigate the role of TFE3 and TFEB in the regulation of tyrosinase and TyrpI gene transcription. We show in this report that overexpression of TFE3 stimulates the tyrosinase and TyrpI promoter activities, while TFEB acts only on the TyrpI promoter. TFE3 and TFEB elicit their effects mainly through the binding to Mbox (AGTCATGTGCT) and Ebox motifs (CATGTG) of tyrosinase and TyrpI promoters. In B16 melanoma cells, the high basal expression of TFE3 is down-regulated by forskolin and by alphaMSH. Interestingly, endogenous TFE3 cannot bind as homodimers to the Mbox, and we did not detect TFE3/Mi heterodimers. According to these data, TFE3 is clearly endowed with the capacity to regulate tyrosinase and TyrpI gene expression. However, TFE3 binding to the melanogenic gene promoters is hindered, thereby preventing its potential melanogenic action. In specific physiological or pathological conditions, the recovery of its binding function would make TFE3 an important element in melanogenesis regulation.


Sujet(s)
Protéines de liaison à l'ADN/physiologie , Glycoprotéines membranaires , Monophenol monooxygenase/métabolisme , Oxidoreductases , Protéines/métabolisme , Facteurs de transcription/physiologie , Activation de la transcription , Cellules 3T3 , Animaux , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines , Colforsine/pharmacologie , AMP cyclique/physiologie , Protéines de liaison à l'ADN/composition chimique , Protéines de liaison à l'ADN/génétique , Dimérisation , Régulation de l'expression des gènes , Cellules HeLa , Motifs à hélice-boucle-hélice/génétique , Motifs à hélice-boucle-hélice/physiologie , Humains , Kératinocytes/effets des médicaments et des substances chimiques , Kératinocytes/métabolisme , Glissières à leucine/génétique , Glissières à leucine/physiologie , Mélanines/métabolisme , Mélanocytes/effets des médicaments et des substances chimiques , Mélanocytes/métabolisme , Mélanome expérimental/anatomopathologie , Souris , Facteur de transcription associé à la microphtalmie , Régions promotrices (génétique) , Protéines de fusion recombinantes/physiologie , Systèmes de seconds messagers/effets des médicaments et des substances chimiques , Délétion de séquence , Similitude de séquences d'acides nucléiques , Facteurs de transcription/composition chimique , Cellules cancéreuses en culture/effets des médicaments et des substances chimiques , Cellules cancéreuses en culture/métabolisme , Hormone mélanotrope alpha/physiologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE