Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Ophthalmic Epidemiol ; 28(2): 169-174, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32693661

RESUMEN

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.


Asunto(s)
Evisceración Orbitaria , Anciano , Estudios de Cohortes , Bases de Datos Factuales , Femenino , Francia/epidemiología , Humanos , Incidencia , Masculino , Estudios Retrospectivos
2.
J Fr Ophtalmol ; 43(2): 152-174, 2020 Feb.
Artículo en Francés | MEDLINE | ID: mdl-31831277

RESUMEN

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.


Asunto(s)
Evisceración Orbitaria , Neoplasias del Ojo/diagnóstico , Neoplasias del Ojo/epidemiología , Neoplasias del Ojo/cirugía , Neoplasias de los Párpados/diagnóstico , Neoplasias de los Párpados/epidemiología , Neoplasias de los Párpados/cirugía , Historia del Siglo XXI , Humanos , Esperanza de Vida/tendencias , Evisceración Orbitaria/historia , Evisceración Orbitaria/mortalidad , Evisceración Orbitaria/tendencias , Neoplasias Orbitales/diagnóstico , Neoplasias Orbitales/epidemiología , Neoplasias Orbitales/cirugía , Pronóstico , Procedimientos de Cirugía Plástica/métodos , Procedimientos de Cirugía Plástica/tendencias , Análisis de Supervivencia , Resultado del Tratamiento
3.
Oncogene ; 36(48): 6659-6667, 2017 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-28825724

RESUMEN

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.


Asunto(s)
Melanoma/metabolismo , Factor de Transcripción Asociado a Microftalmía/metabolismo , Neoplasias Cutáneas/metabolismo , Sumoilación , Animales , Senescencia Celular , Humanos , Melanoma/patología , Melanoma/terapia , Transducción de Señal , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/terapia
5.
Oncogene ; 35(21): 2735-45, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26364600

RESUMEN

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.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2/genética , Melanoma/genética , Melanoma/metabolismo , Animales , Línea Celular Tumoral , Regulación hacia Abajo , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Xenoinjertos , Humanos , Melanoma/patología , Ratones , Ratones Desnudos , Subunidad p52 de NF-kappa B/biosíntesis , Subunidad p52 de NF-kappa B/genética , Subunidad p52 de NF-kappa B/metabolismo , Activación Transcripcional
6.
Rev. chil. reumatol ; 32(1): 17-20, 2016. ilus
Artículo en Español | LILACS | ID: biblio-869807

RESUMEN

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.


Asunto(s)
Humanos , Femenino , Adulto Joven , Calcinosis/etiología , Calcinosis , Esclerodermia Sistémica/complicaciones , Esclerodermia Limitada/complicaciones
7.
Oncogene ; 31(19): 2461-70, 2012 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-21996743

RESUMEN

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.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Melanoma/secundario , Factor de Transcripción Asociado a Microftalmía/metabolismo , Neoplasias Cutáneas/patología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Hipoxia de la Célula , Silenciador del Gen , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Melanoma/metabolismo , Melanoma Experimental/metabolismo , Melanoma Experimental/secundario , Mesodermo/metabolismo , Mesodermo/patología , Ratones , Factor de Transcripción Asociado a Microftalmía/antagonistas & inhibidores , Factor de Transcripción Asociado a Microftalmía/genética , Invasividad Neoplásica , Metástasis de la Neoplasia , Neoplasias Cutáneas/metabolismo
8.
Cell Death Dis ; 2: e199, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21881601

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Metformina/farmacología , Metformina/toxicidad , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína 5 Relacionada con la Autofagia , Beclina-1 , Línea Celular Tumoral , Fase G1 , Humanos , Melanoma/tratamiento farmacológico , Melanoma/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Desnudos , Proteínas Asociadas a Microtúbulos/antagonistas & inhibidores , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Fase de Descanso del Ciclo Celular , Trasplante Heterólogo
9.
Oncogene ; 30(20): 2307-18, 2011 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-21278797

RESUMEN

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.


Asunto(s)
Melanoma Experimental/metabolismo , Melanoma/metabolismo , Factor de Transcripción Asociado a Microftalmía/metabolismo , Neoplasias Cutáneas/metabolismo , Animales , Diferenciación Celular/genética , División Celular , Línea Celular Tumoral , Transdiferenciación Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Silenciador del Gen , Humanos , Metástasis Linfática , Melanoma/patología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Factor de Transcripción Asociado a Microftalmía/genética , Trasplante de Neoplasias , Células Madre Neoplásicas/patología , Neoplasias Cutáneas/patología
10.
Oncogene ; 30(20): 2319-32, 2011 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-21258399

RESUMEN

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.


Asunto(s)
Reparación del ADN/genética , Replicación del ADN , Regulación Neoplásica de la Expresión Génica , Inestabilidad Genómica , Melanoma/genética , Factor de Transcripción Asociado a Microftalmía/metabolismo , Mitosis/genética , Neoplasias Cutáneas/genética , Sitios de Unión , Línea Celular Tumoral , Linaje de la Célula , Senescencia Celular , Técnicas de Inactivación de Genes , Humanos , Factor de Transcripción Asociado a Microftalmía/genética , Metástasis de la Neoplasia , Neoplasias Cutáneas/metabolismo
11.
Cell Death Differ ; 18(1): 109-21, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20596077

RESUMEN

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.


Asunto(s)
Antineoplásicos/uso terapéutico , Quimiocina CXCL1/metabolismo , Melanoma/tratamiento farmacológico , Factor de Transcripción Asociado a Microftalmía/metabolismo , Tiazolidinedionas/uso terapéutico , Animales , Apoptosis , Diferenciación Celular , Línea Celular Tumoral , Quimiocina CXCL1/genética , Quimiocina CXCL1/farmacología , Regulación hacia Abajo , Humanos , Melanoma/metabolismo , Ratones , Ratones Desnudos , Factor de Transcripción Asociado a Microftalmía/antagonistas & inhibidores , Factor de Transcripción Asociado a Microftalmía/fisiología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal , Trasplante Heterólogo
12.
Pigment Cell Melanoma Res ; 21(2): 139-46, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18426407

RESUMEN

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.


Asunto(s)
Apoptosis , Factores de Transcripción Forkhead/metabolismo , Melanoma/metabolismo , Apoptosis/genética , Western Blotting , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/genética , Células Cultivadas , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Humanos , Proteínas Inhibidoras de la Apoptosis , Factor I del Crecimiento Similar a la Insulina/metabolismo , Melanoma/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Reacción en Cadena de la Polimerasa , Transducción de Señal , Survivin
13.
Cytogenet Genome Res ; 106(2-4): 243-6, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15292598

RESUMEN

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.


Asunto(s)
Cromosomas/genética , Lagartos/genética , Animales
14.
Cell Death Differ ; 11(10): 1084-91, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15243584

RESUMEN

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.


Asunto(s)
Apoptosis/efectos de los fármacos , Melanocitos/efectos de los fármacos , Glicoproteínas de Membrana/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Proteínas Reguladoras de la Apoptosis , Células Cultivadas , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Melanocitos/citología , Melanocitos/enzimología , Melanocitos/metabolismo , Melanoma/patología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal/efectos de los fármacos , Factor de Células Madre/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF
15.
Cytogenet Genome Res ; 103(1-2): 128-34, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-15004475

RESUMEN

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.


Asunto(s)
Cromosomas , Lagartos/genética , Animales , Bandeo Cromosómico , Sitios Frágiles del Cromosoma , Cromosomas/ultraestructura , Femenino , Cariotipificación , Masculino , Región Organizadora del Nucléolo
16.
J Mol Endocrinol ; 29(3): 347-60, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12459036

RESUMEN

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.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Glucagón/farmacología , Conductos Pancreáticos/citología , Conductos Pancreáticos/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Precursores de Proteínas/farmacología , Animales , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Ciclinas/metabolismo , Citometría de Flujo , Regulación de la Expresión Génica/efectos de los fármacos , Péptido 1 Similar al Glucagón , Glucoquinasa/metabolismo , Glucosa/farmacología , Transportador de Glucosa de Tipo 2 , Humanos , Inmunohistoquímica , Insulina/análisis , Insulina/metabolismo , Secreción de Insulina , Proteínas de Transporte de Monosacáridos/metabolismo , Ratas , Proteínas Supresoras de Tumor/metabolismo
17.
Leukemia ; 16(4): 700-7, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11960352

RESUMEN

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.


Asunto(s)
Apoptosis/efectos de los fármacos , Linfoma de Burkitt/metabolismo , Caspasas/metabolismo , Desoxirribonucleasas/metabolismo , Leucemia de Células B/metabolismo , Leucemia de Células T/metabolismo , Proteínas/metabolismo , Antineoplásicos/farmacología , Proteínas Reguladoras de la Apoptosis , Factor Apoptótico 1 Activador de Proteasas , Western Blotting , Linfoma de Burkitt/patología , Supervivencia Celular/efectos de los fármacos , Grupo Citocromo c/metabolismo , Fragmentación del ADN , Cartilla de ADN/química , ADN de Neoplasias/análisis , Relación Dosis-Respuesta a Droga , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Citometría de Flujo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Leucemia de Células B/patología , Leucemia de Células T/patología , Glicoproteínas de Membrana/farmacología , Proteínas de Unión a Poli-ADP-Ribosa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estaurosporina/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
18.
Pigment Cell Res ; 14(5): 328-36, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11601654

RESUMEN

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.


Asunto(s)
Proteínas Morfogenéticas Óseas/farmacología , Regulación Enzimológica de la Expresión Génica , Melaninas/biosíntesis , Melanocitos/fisiología , Monofenol Monooxigenasa/genética , Cresta Neural/efectos de los fármacos , Factor de Crecimiento Transformador beta , 1-Metil-3-Isobutilxantina/farmacología , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Animales , Proteína Morfogenética Ósea 2 , Diferenciación Celular , Células Cultivadas , Colforsina/farmacología , Coturnix , AMP Cíclico/metabolismo , Melanocitos/efectos de los fármacos , Melanocitos/enzimología , Melanocitos/ultraestructura , Ratones , Monofenol Monooxigenasa/metabolismo , Cresta Neural/citología , Cresta Neural/enzimología , Cresta Neural/fisiología , Inhibidores de Fosfodiesterasa/farmacología , Pigmentación/fisiología , Transducción de Señal/fisiología
19.
Oncogene ; 20(16): 1964-72, 2001 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-11360180

RESUMEN

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


Asunto(s)
Isoenzimas/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteína Quinasa C/fisiología , Proteínas Proto-Oncogénicas pp60(c-src) , Proteínas Proto-Oncogénicas/fisiología , Linfocitos T/fisiología , Trombina/farmacología , Benzoquinonas , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Humanos , Isoenzimas/metabolismo , Células Jurkat , Lactamas Macrocíclicas , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/fisiología , Proteína Quinasa C/metabolismo , Proteína Quinasa C-delta , Proteína Quinasa C-epsilon , Proteínas Tirosina Quinasas/metabolismo , Proteínas Tirosina Quinasas/fisiología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-fyn , Quinonas/farmacología , Receptores de Trombina/fisiología , Rifabutina/análogos & derivados , Linfocitos T/efectos de los fármacos , Linfocitos T/enzimología , Proteínas Quinasas p38 Activadas por Mitógenos
20.
Eur Cytokine Netw ; 12(1): 126-34, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11282556

RESUMEN

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.


Asunto(s)
Apoptosis/fisiología , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas de Choque Térmico/metabolismo , Receptores del Factor de Necrosis Tumoral/fisiología , Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Ceramidas/farmacología , Activación Enzimática , Citometría de Flujo , Humanos , Células Jurkat , Estaurosporina/farmacología , Receptor fas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...