Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
EMBO J ; 39(3): e102525, 2020 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-31919869

RESUMO

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.


Assuntos
Adipócitos/citologia , Vesículas Extracelulares/metabolismo , Ácidos Graxos/metabolismo , Melanoma/metabolismo , Obesidade/complicações , Células 3T3 , Adipócitos/metabolismo , Animais , Autofagia , Linhagem Celular Tumoral , Movimento Celular , Humanos , Metabolismo dos Lipídeos , Masculino , Camundongos , Mitocôndrias/metabolismo , Dinâmica Mitocondrial , Obesidade/metabolismo , Oxirredução
2.
Breast Cancer Res ; 21(1): 7, 2019 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-30654824

RESUMO

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.


Assuntos
Adipócitos/metabolismo , Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Obesidade/complicações , Partículas de Ribonucleoproteínas em Forma de Abóbada/metabolismo , Células 3T3 , Tecido Adiposo/citologia , Adulto , Idoso , Animais , Antineoplásicos/uso terapêutico , Mama/citologia , Mama/patologia , Mama/cirurgia , Neoplasias da Mama/patologia , Neoplasias da Mama/cirurgia , Linhagem Celular Tumoral , Técnicas de Cocultura , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Feminino , Fluoruracila/farmacologia , Fluoruracila/uso terapêutico , Humanos , Mastectomia , Camundongos , Pessoa de Meia-Idade , Paclitaxel/farmacologia , Paclitaxel/uso terapêutico , Cultura Primária de Células , RNA Interferente Pequeno/metabolismo , Partículas de Ribonucleoproteínas em Forma de Abóbada/genética
3.
J Lipid Res ; 59(10): 1793-1804, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29678957

RESUMO

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.


Assuntos
Vesículas Extracelulares/metabolismo , Neoplasias/patologia , Adipócitos/metabolismo , Adipócitos/patologia , Animais , Carcinogênese , Progressão da Doença , Humanos , Neoplasias/metabolismo
4.
Mol Cell Proteomics ; 12(3): 736-48, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23275444

RESUMO

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.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Núcleo Celular/metabolismo , Proteoma/metabolismo , Proteômica/métodos , Sequência de Aminoácidos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Western Blotting , Hipóxia Celular , Linhagem Celular Tumoral , Movimento Celular , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Imunoprecipitação , Espectrometria de Massas/métodos , Melanoma/genética , Melanoma/metabolismo , Melanoma/patologia , Fator de Transcrição Associado à Microftalmia/genética , Fator de Transcrição Associado à Microftalmia/metabolismo , Modelos Biológicos , Dados de Sequência Molecular , Ligação Proteica , Mapeamento de Interação de Proteínas/métodos , Proteoma/genética , Interferência de RNA , Fatores de Transcrição SOXE/genética , Fatores de Transcrição SOXE/metabolismo , Homologia de Sequência de Aminoácidos , Fator de Transcrição AP-2/genética , Fator de Transcrição AP-2/metabolismo
5.
Med Sci (Paris) ; 30(4): 398-404, 2014 Apr.
Artigo em Francês | MEDLINE | ID: mdl-24801034

RESUMO

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.


Assuntos
Tecido Adiposo/patologia , Neoplasias/patologia , Adipócitos/fisiologia , Tecido Adiposo/fisiopatologia , Humanos , Fatores de Risco , Microambiente Tumoral
6.
Cell Rep ; 42(12): 113586, 2023 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-38113139

RESUMO

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.


Assuntos
Melanoma , Humanos , Melanoma/patologia , Esfingolipídeos/metabolismo , Comunicação Parácrina , Queratinócitos/metabolismo , Caderinas/metabolismo , Esfingosina/metabolismo , Lisofosfolipídeos/metabolismo
7.
J Invest Dermatol ; 142(9): 2488-2498.e8, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35150661

RESUMO

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.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina , Vesículas Extracelulares , Melanoma , Obesidade , Adipócitos/metabolismo , Animais , Inibidor p16 de Quinase Dependente de Ciclina/genética , Vesículas Extracelulares/metabolismo , Melanoma/genética , Melanoma/metabolismo , Camundongos , Obesidade/genética , Obesidade/metabolismo , beta Catenina/metabolismo
8.
FASEB Bioadv ; 3(6): 407-419, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34124596

RESUMO

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.

9.
Cancers (Basel) ; 12(11)2020 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-33121001

RESUMO

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.

10.
Protein Expr Purif ; 64(1): 39-46, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18996486

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/isolamento & purificação , Proteínas de Ligação a DNA/metabolismo , Proteínas de Homeodomínio/isolamento & purificação , Proteínas de Homeodomínio/metabolismo , Melanoma/genética , Fatores do Domínio POU/isolamento & purificação , Fatores do Domínio POU/metabolismo , Sequência de Aminoácidos , Fenômenos Biológicos , DNA Complementar/química , DNA Complementar/genética , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Proteínas de Homeodomínio/química , Proteínas de Homeodomínio/genética , Humanos , Hidrólise , Dados de Sequência Molecular , Peso Molecular , Fatores do Domínio POU/química , Fatores do Domínio POU/genética , Fragmentos de Peptídeos/química , Plasmídeos , Dobramento de Proteína , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Tripsina/farmacologia
11.
Mol Cancer Res ; 17(3): 821-835, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30606769

RESUMO

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.


Assuntos
Tecido Adiposo/fisiopatologia , Obesidade/complicações , Neoplasias da Próstata/etiologia , Animais , Técnicas de Cultura de Células , Humanos , Masculino , Camundongos , Estresse Oxidativo , Neoplasias da Próstata/patologia , Transfecção
12.
J Mol Biol ; 370(4): 687-700, 2007 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-17543985

RESUMO

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.


Assuntos
DNA/química , DNA/metabolismo , Fator 3 de Transcrição de Octâmero/química , Fator 3 de Transcrição de Octâmero/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Domínio Catalítico , Extratos Celulares , Núcleo Celular/enzimologia , Simulação por Computador , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Mutação/genética , Conformação de Ácido Nucleico , Fator 3 de Transcrição de Octâmero/genética , Fatores do Domínio POU/química , Fatores do Domínio POU/classificação , Fatores do Domínio POU/metabolismo , Fosforilação , Fosfosserina/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína
13.
Nucleic Acids Res ; 33(5): 1513-23, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15767276

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/química , Neurônios/metabolismo , Elementos de Resposta , Fatores de Transcrição/química , Motivos de Aminoácidos , Animais , Descarboxilases de Aminoácido-L-Aromático/genética , Sequência de Bases , Sítios de Ligação , Carcinoma de Células Pequenas/genética , Hormônio Liberador da Corticotropina/genética , Pegada de DNA , Proteínas de Ligação a DNA/metabolismo , Dimerização , Fator 3-beta Nuclear de Hepatócito , Humanos , Neoplasias Pulmonares/genética , Modelos Moleculares , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Fator 3 de Transcrição de Octâmero , Regiões Promotoras Genéticas , Estrutura Terciária de Proteína , Ratos , Fatores de Transcrição/metabolismo
14.
Pigment Cell Melanoma Res ; 30(3): 294-306, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28222242

RESUMO

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.


Assuntos
Adiposidade , Melanoma/complicações , Obesidade/complicações , Tecido Adiposo/patologia , Animais , Progressão da Doença , Humanos , Melanoma/patologia , Modelos Biológicos , Obesidade/patologia
15.
JCI Insight ; 2(4): e87489, 2017 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-28239646

RESUMO

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.


Assuntos
Adipócitos/metabolismo , Neoplasias da Mama/metabolismo , Carcinoma Ductal de Mama/metabolismo , Carcinoma Lobular/metabolismo , Ácidos Graxos não Esterificados/metabolismo , Lipase/metabolismo , Lipólise , Acetil-CoA Carboxilase/metabolismo , Trifosfato de Adenosina/metabolismo , Adenilato Quinase/metabolismo , Idoso , Animais , Neoplasias da Mama/patologia , Carcinoma Ductal de Mama/patologia , Carcinoma Lobular/patologia , Linhagem Celular Tumoral , Proliferação de Células , Técnicas de Cocultura , Feminino , Humanos , Camundongos , Pessoa de Meia-Idade , Invasividade Neoplásica , Oxirredução , Triglicerídeos/metabolismo
16.
Cancer Res ; 76(14): 4051-7, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27216185

RESUMO

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.


Assuntos
Adipócitos/fisiologia , Exossomos/fisiologia , Ácidos Graxos/metabolismo , Melanoma/patologia , Obesidade/complicações , Células 3T3 , Animais , Movimento Celular , Humanos , Masculino , Melanoma/etiologia , Melanoma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Oxirredução
17.
Nat Commun ; 7: 10230, 2016 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-26756352

RESUMO

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.


Assuntos
Adenocarcinoma/metabolismo , Adipócitos/metabolismo , Movimento Celular , Quimiocina CCL7/metabolismo , Recidiva Local de Neoplasia/metabolismo , Obesidade/metabolismo , Neoplasias da Próstata/metabolismo , Receptores CCR3/metabolismo , Adenocarcinoma/patologia , Tecido Adiposo/citologia , Adulto , Idoso , Animais , Linhagem Celular Tumoral , Progressão da Doença , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Masculino , Espectrometria de Massas , Camundongos , Pessoa de Meia-Idade , Prognóstico , Próstata , Neoplasias da Próstata/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
J Mol Biol ; 339(5): 1049-58, 2004 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-15178247

RESUMO

N Oct-3, a neurospecific POU protein, homodimerizes in a non-cooperative fashion on the neuronal aromatic l-amino acid decarboxylase gene promoter and generates heterodimers with HNF-3beta. Several other neuronal gene promoters, the corticotropin releasing hormone and the aldolase C gene promoters also contain overlapping binding sites for N Oct-3 and HNF-3beta. We have demonstrated that N Oct-3 presents a non-cooperative homodimerization on these two additional targets and can also give rise to heterodimers with HNF-3beta. Surprisingly, despite the high degree of conservation of the respective POU subunits, the ubiquitous POU protein Oct-1 can only form monomers even in the presence of either N Oct-3 or HNF-3beta on these DNA targets. Our data indicate that this difference is correlated with the specific ability of a portion of the N Oct-3 linker to fold as an alpha-helix, a property shared by class III POU proteins. These results suggest that this novel binding pattern permits the heterodimerization of N Oct-3 and HNF-3beta on the neuronal promoters, which could be a key issue in the development of the nervous system and possibly tumors of neural origin.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Neurônios/fisiologia , Regiões Promotoras Genéticas , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Dicroísmo Circular , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Dimerização , Fator 3-beta Nuclear de Hepatócito , Fator C1 de Célula Hospedeira , Humanos , Dados de Sequência Molecular , Proteínas Nucleares/metabolismo , Fator 1 de Transcrição de Octâmero , Fator 3 de Transcrição de Octâmero , Ligação Proteica , Estrutura Secundária de Proteína , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Alinhamento de Sequência , Fatores de Transcrição/química , Fatores de Transcrição/genética
19.
Pigment Cell Melanoma Res ; 28(4): 464-75, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25950383

RESUMO

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.


Assuntos
Exossomos/metabolismo , Melanoma/metabolismo , Proteoma/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Humanos , Espectrometria de Massas , Metástase Neoplásica , Proteínas de Neoplasias/metabolismo , Proteômica
20.
Mol Cell Biol ; 32(7): 1237-47, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22290434

RESUMO

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.


Assuntos
Proliferação de Células , Melanócitos/citologia , Proteínas do Tecido Nervoso/metabolismo , Fatores do Domínio POU/metabolismo , Fatores de Transcrição Box Pareados/genética , Animais , Linhagem Celular Tumoral , Movimento Celular , Humanos , Melanócitos/metabolismo , Melanoma/genética , Melanoma/metabolismo , Camundongos , Camundongos Transgênicos , Fator de Transcrição Associado à Microftalmia/genética , Mutação , Proteínas do Tecido Nervoso/genética , Fator de Transcrição PAX3 , Fatores do Domínio POU/genética , Fenótipo , Fosforilação , Regiões Promotoras Genéticas , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa