Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 42(12): 113586, 2023 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-38113139

RESUMEN

Melanoma is the deadliest form of skin cancer due to its propensity to metastasize. It arises from melanocytes, which are attached to keratinocytes within the basal epidermis. Here, we hypothesize that, in addition to melanocyte-intrinsic modifications, dysregulation of keratinocyte functions could initiate early-stage melanoma cell invasion. We identified the lysolipid sphingosine 1-phosphate (S1P) as a tumor paracrine signal from melanoma cells that modifies the keratinocyte transcriptome and reduces their adhesive properties, leading to tumor invasion. Mechanistically, tumor cell-derived S1P reduced E-cadherin expression in keratinocytes via S1P receptor dependent Snail and Slug activation. All of these effects were blocked by S1P2/3 antagonists. Importantly, we showed that epidermal E-cadherin expression was inversely correlated with the expression of the S1P-producing enzyme in neighboring tumors and the Breslow thickness in patients with early-stage melanoma. These findings support the notion that E-cadherin loss in the epidermis initiates the metastatic cascade in melanoma.


Asunto(s)
Melanoma , Humanos , Melanoma/patología , Esfingolípidos/metabolismo , Comunicación Paracrina , Queratinocitos/metabolismo , Cadherinas/metabolismo , Esfingosina/metabolismo , Lisofosfolípidos/metabolismo
2.
J Invest Dermatol ; 142(9): 2488-2498.e8, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35150661

RESUMEN

Obesity is a recognized factor for increased risk and poor prognosis of many cancers, including melanoma. In this study, using genetically engineered mouse models of melanoma (NrasQ61K transgenic expression, associated or not with Cdkn2a heterozygous deletion), we show that obesity increases melanoma initiation and progression by supporting tumor growth and metastasis, thereby reducing survival. This effect is associated with a decrease in p16INK4A expression in tumors. Mechanistically, adipocytes downregulate p16INK4A in melanoma cells through ß-catenin-dependent regulation, which increases cell motility. Furthermore, ß-catenin is directly transferred from adipocytes to melanoma cells in extracellular vesicles, thus increasing its level and activity, which represses CDKN2A transcription. Adipocytes from individuals with obesity have a stronger effect than those from lean individuals, mainly owing to an increase in the number of vesicles secreted, thus increasing the amount of ß-catenin delivered to melanoma cells and, consequently, amplifying their effect. In conclusion, in this study, we reveal that adipocyte extracellular vesicles control p16INK4A expression in melanoma, which promotes tumor progression. This work expands our understanding of the cooperation between adipocytes and tumors, particularly in obesity.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina , Vesículas Extracelulares , Melanoma , Obesidad , Adipocitos/metabolismo , Animales , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Vesículas Extracelulares/metabolismo , Melanoma/genética , Melanoma/metabolismo , Ratones , Obesidad/genética , Obesidad/metabolismo , beta Catenina/metabolismo
3.
FASEB Bioadv ; 3(6): 407-419, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34124596

RESUMEN

As the largest human energy reservoir, adipocytes drive an intense dialog with other cells/organs throughout the body to regulate the size of adipose tissue and to communicate with other metabolic tissues and the brain to regulate energy supply. Adipokines have long been described as mediators of this crosstalk, participating in obesity-associated complications. Recently, adipocyte-derived extracellular vesicles (Ad-EVs) have emerged as new key actors in this communication due to their powerful capacity to convey complex messages between cells. Ad-EVs convey specific subpopulations of RNA, proteins, and lipids from their parental cells, and can transfer these cargoes into various recipient cells, modulating their metabolism and cell cycle. In healthy individuals, Ad-EVs actively participate in adipose tissue remodeling to compensate energy supply variations by exchanging information between adipocytes or stroma-vascular cells, including immune cells. Besides this, recent evidence points out that Ad-EV secretion and composition from dysfunctional adipocytes are strongly impacted within adipose tissue where they modulate local intercellular communication, contributing to inflammation, fibrosis, abnormal angiogenesis, and at distance with other cells/tissues intrinsically linked to fat (muscle, hepatocytes and even cancer cells). Additionally, some data even suggests that Ad-EVs might have a systemic action. In this review, we will describe the particular properties of Ad-EVs and their involvement in health and diseases, with a particular focus on metabolic and cardiovascular diseases as well as cancer.

4.
Cancers (Basel) ; 12(11)2020 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-33121001

RESUMEN

Metabolic reprogramming contributes to the pathogenesis and heterogeneity of melanoma. It is driven both by oncogenic events and the constraints imposed by a nutrient- and oxygen-scarce microenvironment. Among the most prominent metabolic reprogramming features is an increased rate of lipid synthesis. Lipids serve as a source of energy and form the structural foundation of all membranes, but have also emerged as mediators that not only impact classical oncogenic signaling pathways, but also contribute to melanoma progression. Various alterations in fatty acid metabolism have been reported and can contribute to melanoma cell aggressiveness. Elevated expression of the key lipogenic fatty acid synthase is associated with tumor cell invasion and poor prognosis. Fatty acid uptake from the surrounding microenvironment, fatty acid ß-oxidation and storage also appear to play an essential role in tumor cell migration. The aim of this review is (i) to focus on the major alterations affecting lipid storage organelles and lipid metabolism. A particular attention has been paid to glycerophospholipids, sphingolipids, sterols and eicosanoids, (ii) to discuss how these metabolic dysregulations contribute to the phenotype plasticity of melanoma cells and/or melanoma aggressiveness, and (iii) to highlight therapeutic approaches targeting lipid metabolism that could be applicable for melanoma treatment.

5.
EMBO J ; 39(3): e102525, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-31919869

RESUMEN

Extracellular vesicles are emerging key actors in adipocyte communication. Notably, small extracellular vesicles shed by adipocytes stimulate fatty acid oxidation and migration in melanoma cells and these effects are enhanced in obesity. However, the vesicular actors and cellular processes involved remain largely unknown. Here, we elucidate the mechanisms linking adipocyte extracellular vesicles to metabolic remodeling and cell migration. We show that adipocyte vesicles stimulate melanoma fatty acid oxidation by providing both enzymes and substrates. In obesity, the heightened effect of extracellular vesicles depends on increased transport of fatty acids, not fatty acid oxidation-related enzymes. These fatty acids, stored within lipid droplets in cancer cells, drive fatty acid oxidation upon being released by lipophagy. This increase in mitochondrial activity redistributes mitochondria to membrane protrusions of migrating cells, which is necessary to increase cell migration in the presence of adipocyte vesicles. Our results provide key insights into the role of extracellular vesicles in the metabolic cooperation that takes place between adipocytes and tumors with particular relevance to obesity.


Asunto(s)
Adipocitos/citología , Vesículas Extracelulares/metabolismo , Ácidos Grasos/metabolismo , Melanoma/metabolismo , Obesidad/complicaciones , Células 3T3 , Adipocitos/metabolismo , Animales , Autofagia , Línea Celular Tumoral , Movimiento Celular , Humanos , Metabolismo de los Lípidos , Masculino , Ratones , Mitocondrias/metabolismo , Dinámicas Mitocondriales , Obesidad/metabolismo , Oxidación-Reducción
6.
Breast Cancer Res ; 21(1): 7, 2019 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-30654824

RESUMEN

INTRODUCTION: Clinical studies suggest that obesity, in addition to promoting breast cancer aggressiveness, is associated with a decrease in chemotherapy efficacy, although the mechanisms involved remain elusive. As chemotherapy is one of the main treatments for aggressive or metastatic breast cancer, we investigated whether adipocytes can mediate resistance to doxorubicin (DOX), one of the main drugs used to treat breast cancer, and the mechanisms associated. METHODS: We used a coculture system to grow breast cancer cells with in vitro differentiated adipocytes as well as primary mammary adipocytes isolated from lean and obese patients. Drug cellular accumulation, distribution, and efflux were studied by immunofluorescence, flow cytometry, and analysis of extracellular vesicles. Results were validated by immunohistochemistry in a series of lean and obese patients with cancer. RESULTS: Adipocytes differentiated in vitro promote DOX resistance (with cross-resistance to paclitaxel and 5-fluorouracil) in a large panel of human and murine breast cancer cell lines independently of their subtype. Subcellular distribution of DOX was altered in cocultivated cells with decreased nuclear accumulation of the drug associated with a localized accumulation in cytoplasmic vesicles, which then are expelled into the extracellular medium. The transport-associated major vault protein (MVP), whose expression was upregulated by adipocytes, mediated both processes. Coculture with human mammary adipocytes also induced chemoresistance in breast cancer cells (as well as the related MVP-induced DOX efflux) and their effect was amplified by obesity. Finally, in a series of human breast tumors, we observed a gradient of MVP expression, which was higher at the invasive front, where tumor cells are at close proximity to adipocytes, than in the tumor center, highlighting the clinical relevance of our results. High expression of MVP in these tumor cells is of particular interest since they are more likely to disseminate to give rise to chemoresistant metastases. CONCLUSIONS: Collectively, our study shows that adipocytes induce an MVP-related multidrug-resistant phenotype in breast cancer cells, which could contribute to obesity-related chemoresistance.


Asunto(s)
Adipocitos/metabolismo , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Obesidad/complicaciones , Partículas Ribonucleoproteicas en Bóveda/metabolismo , Células 3T3 , Tejido Adiposo/citología , Adulto , Anciano , Animales , Antineoplásicos/uso terapéutico , Mama/citología , Mama/patología , Mama/cirugía , Neoplasias de la Mama/patología , Neoplasias de la Mama/cirugía , Línea Celular Tumoral , Técnicas de Cocultivo , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Femenino , Fluorouracilo/farmacología , Fluorouracilo/uso terapéutico , Humanos , Mastectomía , Ratones , Persona de Mediana Edad , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Cultivo Primario de Células , ARN Interferente Pequeño/metabolismo , Partículas Ribonucleoproteicas en Bóveda/genética
7.
Mol Cancer Res ; 17(3): 821-835, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30606769

RESUMEN

Prostate gland is surrounded by periprostatic adipose tissue (PPAT), which is increasingly believed to play a paracrine role in prostate cancer progression. Our previous work demonstrates that adipocytes promote homing of prostate cancer cells to PPAT and that this effect is upregulated by obesity. Here, we show that once tumor cells have invaded PPAT (mimicked by an in vitro model of coculture), they establish a bidirectional crosstalk with adipocytes, which promotes tumor cell invasion. Indeed, tumor cells induce adipocyte lipolysis and the free fatty acids (FFA) released are taken up and stored by tumor cells. Incubation with exogenous lipids also stimulates tumor cell invasion, underlining the importance of lipid transfer in prostate cancer aggressiveness. Transferred FFAs (after coculture or exogenous lipid treatment) stimulate the expression of one isoform of the pro-oxidant enzyme NADPH oxidase, NOX5. NOX5 increases intracellular reactive oxygen species (ROS) that, in turn, activate a HIF1/MMP14 pathway, which is responsible for the increased tumor cell invasion. In obesity, tumor-surrounding adipocytes are more prone to activate the depicted signaling pathway and to induce tumor invasion. Finally, the expression of NOX5 and MMP14 is upregulated at the invasive front of human tumors where cancer cells are in close proximity to adipocytes and this process is amplified in obese patients, underlining the clinical relevance of our results. IMPLICATIONS: Our work emphasizes the key role of adjacent PPAT in prostate cancer dissemination and proposes new molecular targets for the treatment of obese patients exhibiting aggressive diseases.


Asunto(s)
Tejido Adiposo/fisiopatología , Obesidad/complicaciones , Neoplasias de la Próstata/etiología , Animales , Técnicas de Cultivo de Célula , Humanos , Masculino , Ratones , Estrés Oxidativo , Neoplasias de la Próstata/patología , Transfección
8.
J Lipid Res ; 59(10): 1793-1804, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29678957

RESUMEN

Cancer cells must adapt their metabolism in order to meet the energy requirements for cell proliferation, survival in nutrient-deprived environments, and dissemination. In particular, FA metabolism is emerging as a critical process for tumors. FA metabolism can be modulated through intrinsic changes in gene expression or signaling between tumor cells and also in response to signals from the surrounding microenvironment. Among these signals, extracellular vesicles (EVs) could play an important role in FA metabolism remodeling. In this review, we will present the role of EVs in tumor progression and especially in metabolic reprogramming. Particular attention will be granted to adipocytes. These cells, which are specialized in storing and releasing FAs, are able to shift tumor metabolism toward the use of FAs and, subsequently, increase tumor aggressiveness. Recent work demonstrates the involvement of EVs in this metabolic symbiosis.


Asunto(s)
Vesículas Extracelulares/metabolismo , Neoplasias/patología , Adipocitos/metabolismo , Adipocitos/patología , Animales , Carcinogénesis , Progresión de la Enfermedad , Humanos , Neoplasias/metabolismo
9.
Pigment Cell Melanoma Res ; 30(3): 294-306, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28222242

RESUMEN

Over the last decade, it has become increasingly clear that adipose tissue, and particularly adipocytes, contributes to tumor progression. Obesity, an ever-increasing worldwide phenomenon, exacerbates this effect. The influence of obesity on melanoma remains poorly studied, although recent data do underline an association between the two diseases in both humans and murine models. Herein, we review the impact of obesity on melanoma incidence and progression and discuss the underlying mechanisms known to be involved. Adipose tissue favors the proliferation and aggressiveness of melanoma cells through a direct dialog, mediated by soluble factors and by exosomes, and through remodeling of the tumor microenvironment. This knowledge could, in the future, help to design new personalized therapeutic options for obese melanoma patients.


Asunto(s)
Adiposidad , Melanoma/complicaciones , Obesidad/complicaciones , Tejido Adiposo/patología , Animales , Progresión de la Enfermedad , Humanos , Melanoma/patología , Modelos Biológicos , Obesidad/patología
10.
JCI Insight ; 2(4): e87489, 2017 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-28239646

RESUMEN

In breast cancer, a key feature of peritumoral adipocytes is their loss of lipid content observed both in vitro and in human tumors. The free fatty acids (FFAs), released by adipocytes after lipolysis induced by tumor secretions, are transferred and stored in tumor cells as triglycerides in lipid droplets. In tumor cell lines, we demonstrate that FFAs can be released over time from lipid droplets through an adipose triglyceride lipase-dependent (ATGL-dependent) lipolytic pathway. In vivo, ATGL is expressed in human tumors where its expression correlates with tumor aggressiveness and is upregulated by contact with adipocytes. The released FFAs are then used for fatty acid ß-oxidation (FAO), an active process in cancer but not normal breast epithelial cells, and regulated by coculture with adipocytes. However, in cocultivated cells, FAO is uncoupled from ATP production, leading to AMPK/acetyl-CoA carboxylase activation, a circle that maintains this state of metabolic remodeling. The increased invasive capacities of tumor cells induced by coculture are completely abrogated by inhibition of the coupled ATGL-dependent lipolysis/FAO pathways. These results show a complex metabolic symbiosis between tumor-surrounding adipocytes and cancer cells that stimulate their invasiveness, highlighting ATGL as a potential therapeutic target to impede breast cancer progression.


Asunto(s)
Adipocitos/metabolismo , Neoplasias de la Mama/metabolismo , Carcinoma Ductal de Mama/metabolismo , Carcinoma Lobular/metabolismo , Ácidos Grasos no Esterificados/metabolismo , Lipasa/metabolismo , Lipólisis , Acetil-CoA Carboxilasa/metabolismo , Adenosina Trifosfato/metabolismo , Adenilato Quinasa/metabolismo , Anciano , Animales , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/patología , Carcinoma Lobular/patología , Línea Celular Tumoral , Proliferación Celular , Técnicas de Cocultivo , Femenino , Humanos , Ratones , Persona de Mediana Edad , Invasividad Neoplásica , Oxidación-Reducción , Triglicéridos/metabolismo
11.
Cancer Res ; 76(14): 4051-7, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27216185

RESUMEN

Malignant progression results from a dynamic cross-talk between stromal and cancer cells. Recent evidence suggests that this cross-talk is mediated to a significant extent by exosomes, nanovesicles secreted by most cell types and which allow the transfer of proteins, lipids, and nucleic acids between cells. Adipocytes are a major component of several tumor microenvironments, including that of invasive melanoma, where cells have migrated to the adipocyte-rich hypodermic layer of the skin. We show that adipocytes secrete exosomes in abundance, which are then taken up by tumor cells, leading to increased migration and invasion. Using mass spectrometry, we analyzed the proteome of adipocyte exosomes. Interestingly, these vesicles carry proteins implicated in fatty acid oxidation (FAO), a feature highly specific to adipocyte exosomes. We further show that, in the presence of adipocyte exosomes, FAO is increased in melanoma cells. Inhibition of this metabolic pathway completely abrogates the exosome-mediated increase in migration. Moreover, in obese mice and humans, both the number of exosomes secreted by adipocytes as well as their effect on FAO-dependent cell migration are amplified. These observations might in part explain why obese melanoma patients have a poorer prognosis than their nonobese counterparts. Cancer Res; 76(14); 4051-7. ©2016 AACR.


Asunto(s)
Adipocitos/fisiología , Exosomas/fisiología , Ácidos Grasos/metabolismo , Melanoma/patología , Obesidad/complicaciones , Células 3T3 , Animales , Movimiento Celular , Humanos , Masculino , Melanoma/etiología , Melanoma/metabolismo , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción
12.
Nat Commun ; 7: 10230, 2016 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-26756352

RESUMEN

Obesity favours the occurrence of locally disseminated prostate cancer in the periprostatic adipose tissue (PPAT) surrounding the prostate gland. Here we show that adipocytes from PPAT support the directed migration of prostate cancer cells and that this event is strongly promoted by obesity. This process is dependent on the secretion of the chemokine CCL7 by adipocytes, which diffuses from PPAT to the peripheral zone of the prostate, stimulating the migration of CCR3 expressing tumour cells. In obesity, higher secretion of CCL7 by adipocytes facilitates extraprostatic extension. The observed increase in migration associated with obesity is totally abrogated when the CCR3/CCL7 axis is inhibited. In human prostate cancer tumours, expression of the CCR3 receptor is associated with the occurrence of aggressive disease with extended local dissemination and a higher risk of biochemical recurrence, highlighting the potential benefit of CCR3 antagonists in the treatment of prostate cancer.


Asunto(s)
Adenocarcinoma/metabolismo , Adipocitos/metabolismo , Movimiento Celular , Quimiocina CCL7/metabolismo , Recurrencia Local de Neoplasia/metabolismo , Obesidad/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores CCR3/metabolismo , Adenocarcinoma/patología , Tejido Adiposo/citología , Adulto , Anciano , Animales , Línea Celular Tumoral , Progresión de la Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Inmunohistoquímica , Masculino , Espectrometría de Masas , Ratones , Persona de Mediana Edad , Pronóstico , Próstata , Neoplasias de la Próstata/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Pigment Cell Melanoma Res ; 28(4): 464-75, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25950383

RESUMEN

Exosomes are important mediators in cell-to-cell communication and, recently, their role in melanoma progression has been brought to light. Here, we characterized exosomes secreted by seven melanoma cell lines with varying degrees of aggressivity. Extensive proteomic analysis of their exosomes confirmed the presence of characteristic exosomal markers as well as melanoma-specific antigens and oncogenic proteins. Importantly, the protein composition differed among exosomes from different lines. Exosomes from aggressive cells contained specific proteins involved in cell motility, angiogenesis, and immune response, while these proteins were less abundant or absent in exosomes from less aggressive cells. Interestingly, when exposed to exosomes from metastatic lines, less aggressive cells increased their migratory capacities, likely due to transfer of pro-migratory exosomal proteins to recipient cells. Hence, this study shows that the specific protein composition of melanoma exosomes depends on the cells' aggressivity and suggests that exosomes influence the behavior of other tumor cells and their microenvironment.


Asunto(s)
Exosomas/metabolismo , Melanoma/metabolismo , Proteoma/metabolismo , Línea Celular Tumoral , Movimiento Celular , Humanos , Espectrometría de Masas , Metástasis de la Neoplasia , Proteínas de Neoplasias/metabolismo , Proteómica
14.
Med Sci (Paris) ; 30(4): 398-404, 2014 Apr.
Artículo en Francés | MEDLINE | ID: mdl-24801034

RESUMEN

Adipose tissue is found in close proximity whith many invasive cancers. In breast cancer, early local tumour invasion results in close interactions of cancer cells with fully differentiated adipocytes. Aside from their energy-storing function, mature adipocytes are also active endocrine cells prone to influence tumour behaviour through heterotypic signaling processes. After a short description of anatomical depots specificities of adipose tissue, we describe the phenotypic changes induced by tumor secretion in tumour-surrounding adipocytes. These cells (that we named CAA for cancer-associated adipocytes) by their ability to secrete pro-inflammatory cytokines, extra-cellular matrix proteins and proteases involved in its remodeling, as well as to release free fatty acid, stimulate tumor proliferation, invasiveness and drug resistance. These results support the concept that adipocytes participate in a deleterious crosstalk with cancer cells to support tumour progression, that might be amplified in obesity conditions and explain the poor prognosis of cancers observed in this subset of patients.


Asunto(s)
Tejido Adiposo/patología , Neoplasias/patología , Adipocitos/fisiología , Tejido Adiposo/fisiopatología , Humanos , Factores de Riesgo , Microambiente Tumoral
15.
Mol Cell Proteomics ; 12(3): 736-48, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23275444

RESUMEN

Hypoxia-inducible factors (HIFs) are heterodimeric transcription factors that play a key role in cellular adaptation to hypoxia. HIF proteins are composed of an α subunit regulated by oxygen pressure (essentially HIF1α or HIF2α) and a constitutively expressed ß subunit. These proteins are often overexpressed in cancer cells, and HIF overexpression frequently correlates with poor prognosis, making HIF proteins promising therapeutic targets. HIF proteins are involved in melanoma initiation and progression; however, the specific function of HIF2 in melanoma has not yet been studied comprehensively. Identifying protein complexes is a valuable way to uncover protein function, and affinity purification coupled with mass spectrometry and label-free quantification is a reliable method for this approach. We therefore applied quantitative interaction proteomics to identify exhaustively the nuclear complexes containing HIF2α in a human melanoma cell line, 501mel. We report, for the first time, a high-throughput analysis of the interactome of an HIF subunit. Seventy proteins were identified that interact with HIF2α, including some well-known HIF partners and some new interactors. The new HIF2α partners microphthalmia-associated transcription factor, SOX10, and AP2α, which are master actors of melanoma development, were confirmed via co-immunoprecipitation experiments. Their ability to bind to HIF1α was also tested: microphthalmia-associated transcription factor and SOX10 were confirmed as HIF1α partners, but the transcription factor AP2α was not. AP2α expression correlates with low invasive capacities. Interestingly, we demonstrated that when HIF2α was overexpressed, only cells expressing large amounts of AP2α exhibited decreased invasive capacities in hypoxia relative to normoxia. The simultaneous presence of both transcription factors therefore reduces cells' invasive properties. Knowledge of the HIF2α interactome is thus a useful resource for investigating the general mechanisms of HIF function and regulation, and here we reveal unexpected, distinct roles for the HIF1 and HIF2 isoforms in melanoma progression.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Núcleo Celular/metabolismo , Proteoma/metabolismo , Proteómica/métodos , Secuencia de Aminoácidos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Western Blotting , Hipoxia de la Célula , Línea Celular Tumoral , Movimiento Celular , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inmunoprecipitación , Espectrometría de Masas/métodos , Melanoma/genética , Melanoma/metabolismo , Melanoma/patología , Factor de Transcripción Asociado a Microftalmía/genética , Factor de Transcripción Asociado a Microftalmía/metabolismo , Modelos Biológicos , Datos de Secuencia Molecular , Unión Proteica , Mapeo de Interacción de Proteínas/métodos , Proteoma/genética , Interferencia de ARN , Factores de Transcripción SOXE/genética , Factores de Transcripción SOXE/metabolismo , Homología de Secuencia de Aminoácido , Factor de Transcripción AP-2/genética , Factor de Transcripción AP-2/metabolismo
16.
Mol Cell Biol ; 32(7): 1237-47, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22290434

RESUMEN

MITF-M and PAX3 are proteins central to the establishment and transformation of the melanocyte lineage. They control various cellular mechanisms, including migration and proliferation. BRN2 is a POU domain transcription factor expressed in melanoma cell lines and is involved in proliferation and invasion, at least in part by regulating the expression of MITF-M and PAX3. The T361 and S362 residues of BRN2, both in the POU domain, are conserved throughout the POU protein family and are targets for phosphorylation, but their roles in vivo remain unknown. To examine the role of this phosphorylation, we generated mutant BRN2 in which these two residues were replaced with alanines (BRN2TS→BRN2AA). When expressed in melanocytes in vitro or in the melanocyte lineage in transgenic mice, BRN2TS induced proliferation and repressed migration, whereas BRN2AA repressed both proliferation and migration. BRN2TS and BRN2AA bound and repressed the MITF-M promoter, whereas PAX3 transcription was induced by BRN2TS but repressed by BRN2AA. Expression of the BRN2AA transgene in a Mitf heterozygous background and in a Pax3 mutant background enhanced the coat color phenotype. Our findings show that melanocyte migration and proliferation are controlled both through the regulation of PAX3 by nonphosphorylated BRN2 and through the regulation of MITF-M by the overall BRN2 level.


Asunto(s)
Proliferación Celular , Melanocitos/citología , Proteínas del Tejido Nervioso/metabolismo , Factores del Dominio POU/metabolismo , Factores de Transcripción Paired Box/genética , Animales , Línea Celular Tumoral , Movimiento Celular , Humanos , Melanocitos/metabolismo , Melanoma/genética , Melanoma/metabolismo , Ratones , Ratones Transgénicos , Factor de Transcripción Asociado a Microftalmía/genética , Mutación , Proteínas del Tejido Nervioso/genética , Factor de Transcripción PAX3 , Factores del Dominio POU/genética , Fenotipo , Fosforilación , Regiones Promotoras Genéticas , Transcripción Genética
17.
Pigment Cell Melanoma Res ; 23(3): 404-18, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20337985

RESUMEN

POU3F2 is a POU-Homeodomain transcription factor expressed in neurons and melanoma cells. In melanoma lesions, cells expressing high levels of POU3F2 show enhanced invasive and metastatic capacity that can in part be explained by repression of Micropthalmia-associated Transcription Factor (MITF) expression via POU3F2 binding to its promoter. To identify other POU3F2 target genes that may be involved in modulating the properties of melanoma cells, we performed ChIP-chip experiments in 501Mel melanoma cells. 2108 binding loci located in the regulatory regions of 1700 potential target genes were identified. Bioinformatic and experimental assays showed the presence of known POU3F2-binding motifs, but also many AT-rich sequences with only partial similarity to the known motifs at the occupied loci. Functional analysis indicates that POU3F2 regulates the stem cell factor (Kit ligand, Kitl) promoter via a cluster of four closely spaced binding sites located in the proximal promoter. Our results suggest that POU3F2 may regulate the properties of melanoma cells via autocrine KIT ligand signalling.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Genoma Humano/genética , Estudio de Asociación del Genoma Completo , Proteínas de Homeodominio/genética , Melanoma/genética , Factores del Dominio POU/genética , Regiones Promotoras Genéticas , Factor de Células Madre/genética , Secuencias de Aminoácidos , Secuencia de Bases , Sitios de Unión , Antígenos CD36/genética , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Genes Relacionados con las Neoplasias/genética , Sitios Genéticos/genética , Proteínas de Homeodominio/química , Humanos , Melanoma/patología , Factor de Transcripción Asociado a Microftalmía/genética , Datos de Secuencia Molecular , Factores del Dominio POU/química , Unión Proteica , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Activación Transcripcional/genética , Proteínas Wnt/genética
18.
Protein Expr Purif ; 64(1): 39-46, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18996486

RESUMEN

This report describes the first purification procedure of the human full-length N Oct-3 protein in amounts suitable for structural studies and proteomic investigations. N Oct-3 is a transcription factor member of the POU protein family. It possesses a large N-terminal transactivation domain and a DNA-binding domain (DBD) which is composed of two subdomains, POUs and POUh, which are joined by a linker peptide. N Oct-3 is a master gene for central nervous system development but also for melanoma progression. Previous structural studies have all been performed using N Oct-3 DBD only. In this study, the full-length N Oct-3 protein was bacterially expressed and purified to homogeneity. The purified protein gave a single band at approximately 53 kDa on SDS-PAGE, while cDNA sequence analysis revealed a calculated molecular mass of 47 kDa confirmed by mass spectroscopy. Size-exclusion chromatography experiments indicated that in solution, full-length N Oct-3 was a monomer. Circular dichroïsm and intrinsic tryptophan fluorescence showed that full-length N Oct-3 was folded, with a significant alpha-helix content probably located in its DBD. Comparison with the purified N Oct-3 DBD demonstrated that, at least in vitro, the affinity of the protein for its DNA targets was similar. This suggests that the transactivation domain of N Oct-3 was not involved in N Oct-3 DNA interaction.


Asunto(s)
Proteínas de Unión al ADN/aislamiento & purificación , Proteínas de Unión al ADN/metabolismo , Proteínas de Homeodominio/aislamiento & purificación , Proteínas de Homeodominio/metabolismo , Melanoma/genética , Factores del Dominio POU/aislamiento & purificación , Factores del Dominio POU/metabolismo , Secuencia de Aminoácidos , Fenómenos Biológicos , ADN Complementario/química , ADN Complementario/genética , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Proteínas de Homeodominio/química , Proteínas de Homeodominio/genética , Humanos , Hidrólisis , Datos de Secuencia Molecular , Peso Molecular , Factores del Dominio POU/química , Factores del Dominio POU/genética , Fragmentos de Péptidos/química , Plásmidos , Pliegue de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Tripsina/farmacología
19.
J Mol Biol ; 370(4): 687-700, 2007 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-17543985

RESUMEN

N Oct-3, a transcription factor member of the POU protein family, is implicated in normal central nervous system development but also in melanoma growth. Its DNA-binding domain (DBD) comprises two subdomains, POUs and POUh, joined by a linker peptide. We have previously shown that N Oct-3 can interact with the already described PORE and MORE DNA motifs, but also with a new structural element we have termed NORE. Having observed that both the PORE and NORE DNA-association modes depend on a strong anchoring of the POUh subdomain rigid arm into the DNA-target minor groove, in contrast to the MORE mode, we have formulated the hypothesis that phosphorylation of the conserved Ser101 residue located in the N Oct-3 POUh arm could lead to differential results in DNA binding according to the type of target. Here we demonstrate that, in vitro, Ser101 is phosphorylated by protein kinase A (PKA), either purified or contained in melanoma (624 mel) nuclear extract, and that this phosphorylation indeed significantly reduced N Oct-3 DBD binding to PORE and NORE motifs, most likely by hampering the POUh rigid arm insertion in the DNA minor groove. Conversely, no effect was observed on the binding of N Oct-3 DBD to MORE sequences. Finally, once bound to its DNA targets, N Oct-3 DBD is less susceptible to PKA activity. We conclude that transcription of genes exhibiting a MORE motif in their promoter should be less affected by N Oct-3 phosphorylation than that of genes switched on by PORE or NORE sequences.


Asunto(s)
ADN/química , ADN/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/química , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Dominio Catalítico , Extractos Celulares , Núcleo Celular/enzimología , Simulación por Computador , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Mutación/genética , Conformación de Ácido Nucleico , Factor 3 de Transcripción de Unión a Octámeros/genética , Factores del Dominio POU/química , Factores del Dominio POU/clasificación , Factores del Dominio POU/metabolismo , Fosforilación , Fosfoserina/metabolismo , Unión Proteica , Estructura Terciaria de Proteína
20.
Nucleic Acids Res ; 33(5): 1513-23, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15767276

RESUMEN

N-Oct-3 is a neuronal transcription factor widely expressed in the developing mammalian central nervous system, and necessary to maintain neural cell differentiation. The key role of N-Oct-3 in the transcriptional regulation of a multiplicity of genes is primarily due to the structural plasticity of its so-called 'POU' (acronym of Pit, Oct, Unc) DNA-binding domain. We have recently reported about the unusual dual neuro-specific transcriptional regulation displayed by N-Oct-3 [Blaud,M., Vossen,C., Joseph,G., Alazard,R., Erard,M. and Nieto,L. (2004) J. Mol. Biol., 339, 1049-1058]. To elucidate the underlying molecular mechanisms, we have now made use of molecular modeling, DNA footprinting and electrophoretic mobility shift assay techniques. This combined approach has allowed us to uncover a novel mode of homodimerization adopted by the N-Oct-3 POU domain bound to the neuronal aromatic amino acids de-carboxylase and corticotropin-releasing hormone gene promoters and to demonstrate that this pattern is induced by a structural motif that we have termed 'NORE' (N-Oct-3 responsive element), comprising the 14 bp sequence element TNNRTAAATAATRN. In addition, we have been able to explain how the same structural motif can also induce the formation of a heterodimer in association with hepatocyte nuclear factor 3beta(/Forkhead box a2). Finally, we discuss the possible role of the NORE motif in relation to neuroendocrine lung tumor formation, and in particular the development of small cell lung cancer.


Asunto(s)
Proteínas de Unión al ADN/química , Neuronas/metabolismo , Elementos de Respuesta , Factores de Transcripción/química , Secuencias de Aminoácidos , Animales , Descarboxilasas de Aminoácido-L-Aromático/genética , Secuencia de Bases , Sitios de Unión , Carcinoma de Células Pequeñas/genética , Hormona Liberadora de Corticotropina/genética , Huella de ADN , Proteínas de Unión al ADN/metabolismo , Dimerización , Factor Nuclear 3-beta del Hepatocito , Humanos , Neoplasias Pulmonares/genética , Modelos Moleculares , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Factor 3 de Transcripción de Unión a Octámeros , Regiones Promotoras Genéticas , Estructura Terciaria de Proteína , Ratas , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...