Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 49
Filtrer
1.
Ophthalmic Epidemiol ; 28(2): 169-174, 2021 04.
Article de Anglais | MEDLINE | ID: mdl-32693661

RÉSUMÉ

PURPOSE: Orbital exenteration is a radical and disfiguring surgery mainly performed for treating orbital malignancies. Recently, several studies found favorable results in terms of overall survival with eye-sparing surgeries combined with targeted therapies and/or radiotherapy. The aim of this study was to assess the incidence of orbital exenteration and its evolution in France between 2006 and 2017. METHODS: A national observational cohort study was conducted in France between January 2006 and December 2017. Data were collected from the national PMSI (Programme de Médicalisation des Systèmes d'Information) database provided by the CNAM (Caisse Nationale de l'Assurance Maladie). All patients undergoing orbital exenteration over the study period in France were included. RESULTS: One thousand and fifty-seven patients were included. The mean annual number of orbital exenterations was 88.1 (63-117), corresponding to a mean incidence of 0.1/100,000 inhabitants/year. A male predominance was noted (n = 626, 59.2%). Exenteration was mainly performed between 75 and 79 years. The underlying etiology was available for 821 patients (77.7%): malignancies were the most common (n = 755; 92.0%) followed by infectious diseases (n = 16; 1.9%). Over the study period, no statistical difference in the mean incidence of orbital exenteration was found (p = .132). CONCLUSION: The mean annual incidence of orbital exenteration was 0.1/100,000 inhabitants in France and was not significantly modified during the study period.


Sujet(s)
Éviscération de l'orbite , Sujet âgé , Études de cohortes , Bases de données factuelles , Femelle , France/épidémiologie , Humains , Incidence , Mâle , Études rétrospectives
2.
J Fr Ophtalmol ; 43(2): 152-174, 2020 Feb.
Article de Français | MEDLINE | ID: mdl-31831277

RÉSUMÉ

INTRODUCTION: Orbital exenteration is a radical anatomically and psychologically disfiguring procedure. It is mostly performed for management of orbital cancers or cancers with orbital involvement. The lack of benefit in terms of overall survival and the development of new molecular therapies (targeted therapies, immunotherapy) in recent years leads us to question its use. The goal of our review is to answer to the following question: is orbital exenteration a viable procedure in 2019? MATERIALS AND METHODS: A literature review was performed using the PUBMED and MEDLINE databases. The following terms were used then crossed with each other: "orbital exenteration", "exenterated socket", "overall survival", "life expectancy", "orbital reconstruction", "socket reconstruction". Oncology articles from the past 15 years were included and separated into those in the oculoplastic literature and those in the ENT literature. RESULTS: Nineteen articles were included in this review. Eyelid tumours represent the main etiology of orbital exenteration. Basal cell carcinoma is the most frequently incriminated tumor, while sebaceous carcinoma and conjunctival squamous cell carcinoma are the most frequently encountered in Asian series. Non-conservative orbital exenteration is the most prevalent surgery performed. Orbital reconstruction depends on the surgeon's speciality: healing by secondary intention and split thickness skin grafts are mostly performed by oculoplastic surgeons, whereas regional or free flaps are mostly performed by ENT surgeons. Cerebrospinal fluid leakage is the most common intraoperative complication, encountered in 0 to 13 % of cases. The most common postoperative complications are ethmoid fistula and infection of the operative site, encountered in 0 to 50 % and 0 to 43 % of cases respectively. Orbital exenteration allows surgical resection of R0 tumors in 42.5 % to 97 % of cases. Overall survival following orbital exenteration is 83 % (50.5-97) and 65 % (37-92) at 1 and 5 years respectively. Identified risk factors for poor overall survival are: age, tumor histology (worse prognosis with choroidal melanoma, better prognosis with basal cell carcinoma), non-R0 surgical resection, locally advanced tumors (size>20mm, BCVA<20/400 and the presence of metastases at diagnosis). Recent studies have demonstrated favorable outcomes when managing locally advanced basal cell carcinoma, lacrimal gland cancer and conjunctival melanoma with targeted therapies or immunotherapies without performing orbital exenteration. CONCLUSION: Orbital exenteration remains a major part of our therapeutic arsenal. Although orbital exenteration has failed to demonstrate any overall survival benefit, it allows satisfactory local control of the disease with an increasingly less invasive procedure. The development of targeted therapies and immunotherapies may change our therapeutic decisions in the future.


Sujet(s)
Éviscération de l'orbite , Tumeurs de l'oeil/diagnostic , Tumeurs de l'oeil/épidémiologie , Tumeurs de l'oeil/chirurgie , Tumeurs de la paupière/diagnostic , Tumeurs de la paupière/épidémiologie , Tumeurs de la paupière/chirurgie , Histoire du 21ème siècle , Humains , Espérance de vie/tendances , Éviscération de l'orbite/histoire , Éviscération de l'orbite/mortalité , Éviscération de l'orbite/tendances , Tumeurs de l'orbite/diagnostic , Tumeurs de l'orbite/épidémiologie , Tumeurs de l'orbite/chirurgie , Pronostic , /méthodes , /tendances , Analyse de survie , Résultat thérapeutique
3.
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
5.
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
6.
Rev. chil. reumatol ; 32(1): 17-20, 2016. ilus
Article de Espagnol | LILACS | ID: biblio-869807

RÉSUMÉ

La calcinosis es una manifestación frecuente en la esclerodermia sistémica. Sin embargo, la calcinosis pseudotumoral es de presentación excepcional. Las radiografías o tomografía axial computada son de utilidad para el diagnóstico y el seguimiento. Aunque no existe un tratamiento estandarizado efectivo, la remoción quirúrgica de las lesiones sintomáticas y algunos fármacos son útiles en algunos enfermos. Se presenta el caso de una paciente de 23 años con esclerodermia sistémica variedad limitada que desarrolló múltiples lesiones pseudotumorales.


Soft tissue calcinosis is a common cutaneous manifestation in systemic sclerosis. However, pseudotumoral calcinosis is rare. Radiographs or computed tomography are useful tools to diagnose and assess this condition. Although no treatment was uniformly effective, surgical excision of symptomatic lesions and medical treatment provided benefit for some patients. We report here a case of a 23 year old woman, suffering from limited cutaneous systemic sclerosis, who developed many pseudo tumoural calcinosis lesions.


Sujet(s)
Humains , Femelle , Jeune adulte , Calcinose/étiologie , Calcinose , Sclérodermie systémique/complications , Sclérodermie limitée/complications
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
Cytogenet Genome Res ; 106(2-4): 243-6, 2004.
Article de Anglais | MEDLINE | ID: mdl-15292598

RÉSUMÉ

Although B chromosomes have been reported in many species of plants and animals, few studies have revealed the presence of these extra chromosomes in lizards. B chromosomes of lizards show different morphologies and sizes, from microchromosomes to macrochromosomes, or elements of intermediate size between smaller and larger A chromosomes, and number variability at intra- and inter-individual levels. In most cases, they are late-replicating and show either heterochromatic or no distinctive patterns after C-banding. The great majority of the publications about supernumerary chromosomes in this group have been based on conventional staining analyses, and there is no study designed to address questions related to their composition and structure or origin and evolution.


Sujet(s)
Chromosomes/génétique , Lézards/génétique , Animaux
14.
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
15.
Cytogenet Genome Res ; 103(1-2): 128-34, 2003.
Article de Anglais | MEDLINE | ID: mdl-15004475

RÉSUMÉ

The karyotypes of three species of sphaerodactyl gekkonid lizards are described after conventional and differential staining. Karyotypes of Gonatodeshumeralis and G. hasemani are formed by a gradual series of 32 acrocentric chromosomes, similar to those already published for other species of the genus. G. humeralis shows multiple Ag-NORs with intra-individual variability, and positive C-bands located at centromeric and telomeric regions of several chromosome pairs. Coleodactylus amazonicus, the first non-Gonatodes sphaerodactyl studied so far karyologically, exhibits 36 acrocentric/subtelocentric chromosomes and a single pair of Ag- NORs. Fragile sites were detected on two medium-sized chromosome pairs in the karyotype of G. humeralis, most of them obtained in BrdU-treated culture preparations. These sites may represent a putative fission/fusion spot involved in the differentiation of G. humeralis-like 2n = 32 and C. amazonicus-like 2n = 36 karyotypes. Our results, especially on the location of Ag-NORs and the description of fragile sites, are relevant in improving our knowledge about the events of chromosome evolution in this extremely variable and poorly known group of lizards.


Sujet(s)
Chromosomes , Lézards/génétique , Animaux , Zébrage chromosomique , Sites fragiles de chromosome , Chromosomes/ultrastructure , Femelle , Caryotypage , Mâle , Organisateur nucléolaire
16.
J Mol Endocrinol ; 29(3): 347-60, 2002 Dec.
Article de Anglais | MEDLINE | ID: mdl-12459036

RÉSUMÉ

The intestinal hormone glucagon-like peptide-1 (GLP-1) has been shown to promote an increase in pancreatic beta-cell mass via proliferation of islet cells and differentiation of non-insulin-secreting cells. In this study, we have characterized some of the events that lead to the differentiation of pancreatic ductal cells in response to treatment with human GLP-1. Rat pancreatic ductal (ARIP) cells were cultured in the presence of GLP-1 and analyzed for cell counting, cell cycle distribution, expression of cyclin-dependent-kinase (Cdk) inhibitors, transcription of beta-cell-specific genes, loss of ductal-like phenotype and acquisition of beta-cell-like gene expression profile. Exposure of ARIP cells to 10 nM GLP-1 induced a significant reduction in the cell replication rate and a significant decrease in the percentage of cells in S phase of the cell cycle. This was associated with an increase in the number of cells in G0-G1 phase and a reduction of cells in G2-M phase. Western blot analysis for the Cdk inhibitors, kinase inhibitor protein 1 (p27(Kip1)) and Cdk-interacting protein 1 (p21(Cip1)), demonstrated a significant increase in p27(Kip1) and p21(Cip1) levels within the first 24 h from the beginning of GLP-1 treatment. As cells slowed down their proliferation rate, GLP-1 also induced a time-dependent expression of various beta-cell-specific mRNAs. The glucose transporter GLUT-2 was the first of those factors to be expressed (24 h treatment), followed by insulin (44 h) and finally by the enzyme glucokinase (56 h). In addition, immunocytochemistry analysis showed that GLP-1 induced a time-dependent down-regulation of the ductal marker cytokeratin-20 (CK-20) and a time-dependent induction of insulin expression. Finally, GLP-1 promoted a glucose-dependent secretion of insulin, as demonstrated by HPLC and RIA analyses of the cell culture medium. The present study has demonstrated that GLP-1 induces a cell cycle re-distribution with a decrease in cell proliferation rate prior to promoting the differentiation of cells towards an endocrine-like phenotype.


Sujet(s)
Cycle cellulaire/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Glucagon/pharmacologie , Conduits pancréatiques/cytologie , Conduits pancréatiques/effets des médicaments et des substances chimiques , Fragments peptidiques/pharmacologie , Précurseurs de protéines/pharmacologie , Animaux , Protéines du cycle cellulaire/métabolisme , Cellules cultivées , Inhibiteur p21 de kinase cycline-dépendante , Inhibiteur p27 de kinase cycline-dépendante , Cyclines/métabolisme , Cytométrie en flux , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Glucagon-like peptide 1 , Glucokinase/métabolisme , Glucose/pharmacologie , Transporteur de glucose de type 2 , Humains , Immunohistochimie , Insuline/analyse , Insuline/métabolisme , Sécrétion d'insuline , Transporteurs de monosaccharides/métabolisme , Rats , Protéines suppresseurs de tumeurs/métabolisme
17.
Leukemia ; 16(4): 700-7, 2002 Apr.
Article de Anglais | MEDLINE | ID: mdl-11960352

RÉSUMÉ

The execution phase of apoptosis occurs through the activation and function of caspases which cleave key substrates that orchestrate the death process. Here, we have compared the sensitivity of various T and B cell lines to death receptor or staurosporine-induced apoptosis. First, we found a lack of correlation between death receptor expression and sensitivity to Fas or Trail. By contrast, a correlation between caspase activation, DNA fragmentation and cell death in T cell lines was evidenced. Among T cells, CEM underwent apoptosis in response to CH11 but were resistant to Trail in agreement with the absence of Trail receptors (DR4 and DR5) on their surface. The B cell line SKW 6.4 was sensitive to CH11 and staurosporine but resistant to Trail. As B cell lines expressed significant levels of DR4 and DR5, resistance to Trail in SKW 6.4 is likely due to the expression of the decoy receptor DcR1. Burkitt's lymphoma such as RPMI 8866 and Raji did not exhibit DNA fragmentation in response to CH11, Trail or staurosporine but showed long-term caspase-dependent loss of viability upon effector treatment. The B cell lines used in this study express very weak or undetectable levels of DFF40 and relatively high levels of DFF45. Interestingly, cytosolic extracts from RPMI 88.66 but not other B lymphoma exhibit altered levels of cytochrome c-dependent caspase activation. Taken together, our results show that: (1) death receptor expression does not correlate with sensitivity to apoptosis; (2) the very low ratio of DFF40 vs. DFF45 is unlikely to explain by itself the lack of DNA fragmentation observed in certain B cell lines; and (3) a defective cytochrome c-dependent caspase activation might account at least in part for the insensitivity of certain Burkitt's lymphoma (RPMI 88.66) to apoptosis. Thus it seems that resistance of Burkitt's lymphoma to apoptosis is not governed by a general mechanism, but is rather multifactorial and differs from one cell line to another.


Sujet(s)
Apoptose/effets des médicaments et des substances chimiques , Lymphome de Burkitt/métabolisme , Caspases/métabolisme , Désoxyribonucléases/métabolisme , Leucémie B/métabolisme , Leucémie à cellules T/métabolisme , Protéines/métabolisme , Antinéoplasiques/pharmacologie , Protéines régulatrices de l'apoptose , Facteur-1 activateur des protéases apoptotiques , Technique de Western , Lymphome de Burkitt/anatomopathologie , Survie cellulaire/effets des médicaments et des substances chimiques , Cytochromes de type c/métabolisme , Fragmentation de l'ADN , Amorces ADN/composition chimique , ADN tumoral/analyse , Relation dose-effet des médicaments , Activation enzymatique , Antienzymes/pharmacologie , Cytométrie en flux , Humains , Protéines et peptides de signalisation intracellulaire , Leucémie B/anatomopathologie , Leucémie à cellules T/anatomopathologie , Glycoprotéines membranaires/pharmacologie , Protéines liant le poly-adp-ribose , RT-PCR , Staurosporine/pharmacologie , Ligand TRAIL , Cellules cancéreuses en culture/effets des médicaments et des substances chimiques , Cellules cancéreuses en culture/métabolisme , Facteur de nécrose tumorale alpha/pharmacologie
18.
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
19.
Oncogene ; 20(16): 1964-72, 2001 Apr 12.
Article de Anglais | MEDLINE | ID: mdl-11360180

RÉSUMÉ

Activation of the mitogen-activated protein kinase (MAPK) cascade is a well documented mechanism for the G-protein-coupled receptors. Here, we have analysed the requirements for ERKs and p38 MAPK activation by thrombin in Jurkat T cells. We show that thrombin-mediated ERKs activation requires both PTK and PKC activities, whereas p38 MAPK activation is dependent only on PTKs. Thrombin-induced ERK and p38 MAPK activation was more pronounced in p56Lck deficient cells indicating that this PTK exerts a negative control on MAPK activity. Accordingly, overexpression of p50 Csk a kinase that inactivates p56Lck induced constitutive activation of ERKs. Requirement for a Src kinase was evidenced by expression of a constitutively active form of p59Fyn in Jurkat cells. Besides its effect on tyrosine phosphorylation events, thrombin also triggered a rapid and robust redistribution of PKCepsilon and delta from the cytosol to the membrane. Expression of constitutively active and dominant negative PKCepsilon demonstrates the pivotal role of this PKC isoform in ERKs activation by thrombin. These data are consistent with a model where thrombin induces ERK activation via both PKC-dependent and independent pathways, whereas p38 MAPK activation requires only PTKs. The PKC-independent pathway requires Src kinases other than p56Lck more likely p59Fyn, while the PKC-dependent mechanism depends on PKCepsilon


Sujet(s)
Isoenzymes/physiologie , Système de signalisation des MAP kinases/physiologie , Mitogen-Activated Protein Kinases/métabolisme , Protéine kinase C/physiologie , Protéines proto-oncogènes pp60(c-src) , Protéines proto-oncogènes/physiologie , Lymphocytes T/physiologie , Thrombine/pharmacologie , Benzoquinones , Activation enzymatique , Antienzymes/pharmacologie , Humains , Isoenzymes/métabolisme , Cellules Jurkat , Lactames macrocycliques , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Mitogen-Activated Protein Kinase 1/métabolisme , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/antagonistes et inhibiteurs , Mitogen-Activated Protein Kinases/physiologie , Protéine kinase C/métabolisme , Protein kinase C-delta , Protein kinase C-epsilon , Protein-tyrosine kinases/métabolisme , Protein-tyrosine kinases/physiologie , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes c-fyn , Quinones/pharmacologie , Récepteurs à la thrombine/physiologie , Rifabutine/analogues et dérivés , Lymphocytes T/effets des médicaments et des substances chimiques , Lymphocytes T/enzymologie , p38 Mitogen-Activated Protein Kinases
20.
Eur Cytokine Netw ; 12(1): 126-34, 2001 Mar.
Article de Anglais | MEDLINE | ID: mdl-11282556

RÉSUMÉ

Ligation of Fas induces an apoptotic program in Jurkat cells (Jd). We describe a Jurkat T cell variant (Jr) which shows total resistance to Fas-mediated apoptosis but which exhibits sensitivity to non-death-receptor pro-apoptotic stimuli such as staurosporine. Resistance to Fas-induced apoptosis in Jr cells is correlated with high expression of Hsps. A prior heat-shock increases Hsp27 and 70 expression and protects Jd and Jr cells from Fas- and staurosporine-induced apoptosis. Staurosporine, but not the anti-Fas antibody CH11, abrogates constitutive Hsp70 expression at 37 degrees C and staurosporine also inhibit Hsp27 expression in Jd and Jr cells at 42 degrees C. These data suggest that constitutive expression of Hsp27 inhibits Fas-mediated apoptosis, but only induced expression of Hsp70 can protect T cells from staurosporine-induced apoptosis. Thus, Hsp27 could play a role in the regulation of death receptor-mediated apoptosis, while Hsp70 could regulate mitochondrial-dependent cell death.


Sujet(s)
Apoptose/physiologie , Protéines du choc thermique HSP70/métabolisme , Protéines du choc thermique/métabolisme , Récepteurs aux facteurs de nécrose tumorale/physiologie , Apoptose/effets des médicaments et des substances chimiques , Caspases/métabolisme , Céramides/pharmacologie , Activation enzymatique , Cytométrie en flux , Humains , Cellules Jurkat , Staurosporine/pharmacologie , Antigènes CD95/physiologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE