Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Nature ; 546(7660): 676-680, 2017 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-28658220

RESUMO

Cutaneous melanoma is a type of cancer with an inherent potential for lymph node colonization, which is generally preceded by neolymphangiogenesis. However, sentinel lymph node removal does not necessarily extend the overall survival of patients with melanoma. Moreover, lymphatic vessels collapse and become dysfunctional as melanomas progress. Therefore, it is unclear whether (and how) lymphangiogenesis contributes to visceral metastasis. Soluble and vesicle-associated proteins secreted by tumours and/or their stroma have been proposed to condition pre-metastatic sites in patients with melanoma. Still, the identities and prognostic value of lymphangiogenic mediators remain unclear. Moreover, our understanding of lymphangiogenesis (in melanomas and other tumour types) is limited by the paucity of mouse models for live imaging of distal pre-metastatic niches. Injectable lymphatic tracers have been developed, but their limited diffusion precludes whole-body imaging at visceral sites. Vascular endothelial growth factor receptor 3 (VEGFR3) is an attractive 'lymphoreporter' because its expression is strongly downregulated in normal adult lymphatic endothelial cells, but is activated in pathological situations such as inflammation and cancer. Here, we exploit this inducibility of VEGFR3 to engineer mouse melanoma models for whole-body imaging of metastasis generated by human cells, clinical biopsies or endogenously deregulated oncogenic pathways. This strategy revealed early induction of distal pre-metastatic niches uncoupled from lymphangiogenesis at primary lesions. Analyses of the melanoma secretome and validation in clinical specimens showed that the heparin-binding factor midkine is a systemic inducer of neo-lymphangiogenesis that defines patient prognosis. This role of midkine was linked to a paracrine activation of the mTOR pathway in lymphatic endothelial cells. These data support the use of VEGFR3 reporter mice as a 'MetAlert' discovery platform for drivers and inhibitors of metastasis.


Assuntos
Citocinas/metabolismo , Vasos Linfáticos/metabolismo , Metástase Neoplásica/diagnóstico por imagem , Metástase Neoplásica/patologia , Imagem Corporal Total/métodos , Animais , Modelos Animais de Doenças , Progressão da Doença , Células Endoteliais/metabolismo , Feminino , Genes Reporter , Humanos , Linfangiogênese , Vasos Linfáticos/patologia , Masculino , Melanoma/diagnóstico por imagem , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Midkina , Comunicação Parácrina , Prognóstico , Recidiva , Reprodutibilidade dos Testes , Serina-Treonina Quinases TOR/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/análise , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Angiogenesis ; 19(3): 433-45, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26993803

RESUMO

The lymphatic system is essential in many physiological and pathological processes. Still, much remains to be known about the molecular mechanisms that control its development and function and how to modulate them therapeutically. The study of these mechanisms will benefit from better controlled genetic mouse models targeting specifically lymphatic endothelial cells. Among the genes expressed predominantly in lymphatic endothelium, Vegfr3 was the first one identified and is still considered to be one of the best lymphatic markers and a key regulator of the lymphatic system. Here, we report the generation of a Vegfr3-CreER (T2) knockin mouse by gene targeting in embryonic stem cells. This mouse expresses the tamoxifen-inducible CreER(T2) recombinase under the endogenous transcriptional control of the Vegfr3 gene without altering its physiological expression or regulation. The Vegfr3-CreER (T2) allele drives efficient recombination of floxed sequences upon tamoxifen administration specifically in Vegfr3-expressing cells, both in vitro, in primary lymphatic endothelial cells, and in vivo, at different stages of mouse embryonic development and postnatal life. Thus, our Vegfr3-CreER (T2) mouse constitutes a new powerful genetic tool for lineage tracing analysis and for conditional gene manipulation in the lymphatic endothelium that will contribute to improve our current understanding of this system.


Assuntos
Sistema Linfático/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/genética , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Introdução de Genes/métodos , Integrases/genética , Sistema Linfático/citologia , Sistema Linfático/crescimento & desenvolvimento , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Gravidez , Tamoxifeno/farmacologia
3.
Nat Rev Cancer ; 7(6): 415-28, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17508028

RESUMO

The molecular mechanisms that underlie tumour progression are still poorly understood, but recently our knowledge of particular aspects of some of these processes has increased. Specifically, the identification of Snail, ZEB and some basic helix-loop-helix (bHLH) factors as inducers of epithelial-mesenchymal transition (EMT) and potent repressors of E-cadherin expression has opened new avenues of research with potential clinical implications.


Assuntos
Caderinas/fisiologia , Epitélio/fisiologia , Sequências Hélice-Volta-Hélice/fisiologia , Proteínas de Homeodomínio/fisiologia , Invasividade Neoplásica , Neoplasias/patologia , Fatores de Transcrição/fisiologia , Adesão Celular , Movimento Celular , Progressão da Doença , Humanos , Modelos Biológicos , Fenótipo , Processamento de Proteína Pós-Traducional , Proteínas Repressoras/fisiologia , Transdução de Sinais , Fatores de Transcrição da Família Snail , Homeobox 1 de Ligação a E-box em Dedo de Zinco
4.
Int J Cancer ; 136(4): E62-73, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25178837

RESUMO

Cell plasticity is emerging as a key regulator of tumor progression and metastasis. During carcinoma dissemination epithelial cells undergo epithelial to mesenchymal transition (EMT) processes characterized by the acquisition of migratory/invasive properties, while the reverse, mesenchymal to epithelial transition (MET) process, is also essential for metastasis outgrowth. Different transcription factors, called EMT-TFs, including Snail, bHLH and Zeb families are drivers of the EMT branch of epithelial plasticity, and can be post-transcriptionally downregulated by several miRNAs, as the miR-200 family. The specific or redundant role of different EMT-TFs and their functional interrelations are not fully understood. To study the interplay between different EMT-TFs, comprehensive gain and loss-of-function studies of Snail1, Snail2 and/or Zeb1 factors were performed in the prototypical MDCK cell model system. We here describe that Snail1 and Zeb1 are mutually required for EMT induction while continuous Snail1 and Snail2 expression, but not Zeb1, is needed for maintenance of the mesenchymal phenotype in MDCK cells. In this model system, EMT is coordinated by Snail1 and Zeb1 through transcriptional and epigenetic downregulation of the miR-200 family. Interestingly, Snail1 is involved in epigenetic CpG DNA methylation of the miR-200 loci, essential to maintain the mesenchymal phenotype. The present results thus define a novel functional interplay between Snail and Zeb EMT-TFs in miR-200 family regulation providing a molecular link to their previous involvement in the generation of EMT process in vivo.


Assuntos
Transição Epitelial-Mesenquimal , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/fisiologia , MicroRNAs/genética , Fatores de Transcrição/fisiologia , Animais , Metilação de DNA , Cães , Epigênese Genética , Células HEK293 , Humanos , Células Madin Darby de Rim Canino , MicroRNAs/metabolismo , Fenótipo , Regiões Promotoras Genéticas , Fatores de Transcrição da Família Snail , Homeobox 1 de Ligação a E-box em Dedo de Zinco
5.
Proc Natl Acad Sci U S A ; 109(16): 6223-8, 2012 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-22474390

RESUMO

Lymphatic vessel growth or lymphangiogenesis occurs during embryonic development and wound healing and plays an important role in tumor metastasis and inflammatory diseases. However, the possibility of noninvasive detection and quantification of lymphangiogenesis has been lacking. Here, we present the Vegfr3(EGFPLuc) mouse model, where an EGFP-luciferase fusion protein, expressed under the endogenous transcriptional control of the Vegfr3 gene, allows the monitoring of physiological and pathological lymphangiogenesis in vivo. We show tracking of lymphatic vessel development during embryogenesis as well as lymphangiogenesis induced by specific growth factors, during wound healing and in contact hypersensitivity (CHS)--induced inflammation where we also monitor down-regulation of lymphangiogenesis by the glucocorticoid dexamethasone. Importantly, the Vegfr3-reporter allowed us to tracking tumor-induced lymphangiogenesis at the tumor periphery and in lymph nodes in association with the metastatic process. This is the first reporter mouse model for luminescence imaging of lymphangiogenesis. It should provide an important tool for studying the involvement of lymphangiogenesis in pathological processes.


Assuntos
Diagnóstico por Imagem/métodos , Inflamação/metabolismo , Vasos Linfáticos/metabolismo , Cicatrização , Animais , Linhagem Celular Tumoral , Dexametasona/farmacologia , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Feminino , Glucocorticoides/farmacologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Inflamação/genética , Luciferases/genética , Luciferases/metabolismo , Medições Luminescentes/métodos , Linfangiogênese/efeitos dos fármacos , Metástase Linfática , Vasos Linfáticos/embriologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Fatores de Tempo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo
6.
Blood ; 119(19): 4565-76, 2012 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-22446484

RESUMO

Membrane-anchored ephrinB2 and its receptor EphB4 are involved in the formation of blood and lymphatic vessels in normal and pathologic conditions. Eph/ephrin activation requires cell-cell interactions and leads to bidirectional signaling pathways in both ligand- and receptor-expressing cells. To investigate the functional consequences of blocking ephrinB2 activity, 2 highly specific human single-chain Fv (scFv) Ab fragments against ephrinB2 were generated and characterized. Both Ab fragments suppressed endothelial cell migration and tube formation in vitro in response to VEGF and provoked abnormal cell motility and actin cytoskeleton alterations in isolated endothelial cells. As only one of them (B11) competed for binding of ephrinB2 to EphB4, these data suggest an EphB-receptor-independent blocking mechanism. Anti-ephrinB2 therapy reduced VEGF-induced neovascularization in a mouse Matrigel plug assay. Moreover, systemic administration of ephrinB2-blocking Abs caused a drastic reduction in the number of blood and lymphatic vessels in xenografted mice and a concomitant reduction in tumor growth. Our results show for the first time that specific Ab-based ephrinB2 targeting may represent an effective therapeutic strategy to be used as an alternative or in combination with existing antiangiogenic drugs for treating patients with cancer and other angiogenesis-related diseases.


Assuntos
Anticorpos/farmacologia , Efrina-B2/antagonistas & inibidores , Linfangiogênese/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Neovascularização Patológica/prevenção & controle , Sequência de Aminoácidos , Animais , Anticorpos/química , Anticorpos/metabolismo , Anticorpos/uso terapêutico , Especificidade de Anticorpos , Regulação para Baixo/efeitos dos fármacos , Efrina-B2/imunologia , Efrina-B2/metabolismo , Feminino , Células HEK293 , Células Endoteliais da Veia Umbilical Humana , Humanos , Imunoterapia/métodos , Linfangiogênese/fisiologia , Camundongos , Camundongos Nus , Camundongos SCID , Terapia de Alvo Molecular , Neoplasias/irrigação sanguínea , Neoplasias/patologia , Neovascularização Patológica/tratamento farmacológico , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Genesis ; 49(1): 36-45, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21254335

RESUMO

Cre/loxP-dependent expression of fluorescent proteins represents a powerful biological tool for cell lineage, fate-mapping, and genetic analysis. Live tissue imaging has significantly improved with the development of far-red fluorescent proteins, with optimized spectral characteristics for in vivo applications. Here, we report the generation of the first transgenic mouse line expressing the far-red fluorescent protein Katushka, driven by the hybrid CAG promoter upon Cre-mediated recombination. After germ line or tissue-specific Cre-driven reporter activation, Katushka expression is strong and ubiquitous, without toxic effects, allowing fluorescence detection in fresh and fixed samples from all tissues examined. Moreover, fluorescence can be detected by in vivo noninvasive whole-body imaging when Katuhska is expressed exclusively in a specific cell population deep within the animal body such as pancreatic beta cells. Thus, this reporter model enables early, widespread, and sensitive in vivo detection of Cre activity and should provide a versatile tool for a wide spectrum of fluorescence and live-imaging applications.


Assuntos
Integrases/genética , Proteínas Luminescentes/genética , Animais , Citometria de Fluxo/métodos , Genes Reporter , Camundongos , Camundongos Transgênicos , Microscopia Confocal/métodos , Regiões Promotoras Genéticas , Recombinação Genética , Proteína Vermelha Fluorescente
9.
Adv Drug Deliv Rev ; 175: 113833, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34147531

RESUMO

Imaging of the lymphatic vasculature has gained great attention in various fields, not only because lymphatic vessels act as a key draining system in the body, but also for their implication in autoimmune diseases, organ transplant, inflammation and cancer. Thus, neolymphangiogenesis, or the generation of new lymphatics, is typically an early event in the development of multiple tumor types, particularly in aggressive ones such as malignant melanoma. Still, the understanding of how lymphatic endothelial cells get activated at distal (pre)metastatic niches and their impact on therapy is still unclear. Addressing these questions is of particular interest in the case of immune modulators, because endothelial cells may favor or halt inflammatory processes depending on the cellular context. Therefore, there is great interest in visualizing the lymphatic vasculature in vivo. Here, we review imaging tools and mouse models used to analyze the lymphatic vasculature during tumor progression. We also discuss therapeutic approaches based on nanomedicines to target the lymphatic system and the potential use of extracellular vesicles to track and target sentinel lymph nodes. Finally, we summarize main pre-clinical models developed to visualize the lymphatic vasculature in vivo, discussing their applications with a particular focus in metastatic melanoma.


Assuntos
Vesículas Extracelulares/metabolismo , Sistema Linfático/diagnóstico por imagem , Sistemas de Liberação de Fármacos por Nanopartículas , Animais , Vesículas Extracelulares/patologia , Humanos , Linfonodos/diagnóstico por imagem , Linfonodos/efeitos dos fármacos , Linfonodos/patologia , Sistema Linfático/efeitos dos fármacos , Sistema Linfático/patologia , Vasos Linfáticos/diagnóstico por imagem , Vasos Linfáticos/efeitos dos fármacos , Vasos Linfáticos/patologia , Linfonodo Sentinela/diagnóstico por imagem , Linfonodo Sentinela/patologia
10.
EMBO Mol Med ; 13(12): e12924, 2021 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-34762341

RESUMO

Long-range communication between tumor cells and the lymphatic vasculature defines competency for metastasis in different cancer types, particularly in melanoma. Nevertheless, the discovery of selective blockers of lymphovascular niches has been compromised by the paucity of experimental systems for whole-body analyses of tumor progression. Here, we exploit immunocompetent and immunodeficient mouse models for live imaging of Vegfr3-driven neolymphangiogenesis, as a versatile platform for drug screening in vivo. Spatiotemporal analyses of autochthonous melanomas and patient-derived xenografts identified double-stranded RNA mimics (dsRNA nanoplexes) as potent inhibitors of neolymphangiogenesis, metastasis, and post-surgical disease relapse. Mechanistically, dsRNA nanoplexes were found to exert a rapid dual action in tumor cells and in their associated lymphatic vasculature, involving the transcriptional repression of the lymphatic drivers Midkine and Vegfr3, respectively. This suppressive function was mediated by a cell-autonomous type I interferon signaling and was not shared by FDA-approved antimelanoma treatments. These results reveal an alternative strategy for targeting the tumor cell-lymphatic crosstalk and underscore the power of Vegfr3-lymphoreporters for pharmacological testing in otherwise aggressive cancers.


Assuntos
Melanoma , RNA de Cadeia Dupla , Animais , Humanos , Melanoma/tratamento farmacológico , Melanoma/patologia , Camundongos , Camundongos Nus , Transdução de Sinais
11.
Pharm Res ; 27(12): 2544-55, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20857179

RESUMO

PURPOSE: To design hyaluronic acid (HA) and chitosan-g-poly(ethylene glycol) (CS-g-PEG) nanoparticles intended for a broad range of gene delivery applications. METHODS: Nanoparticles formulated at different HA/CS-g-PEG mass ratios were developed to associate either pDNA or siRNA. The physico-chemical characteristics, morphology, association efficiency and nuclease protection ability of the nanocarriers were compared for these two molecules. Their biological performance, including transfection effciency, nanoparticle cellular uptake and citotoxicity, was assesed. RESULTS: The resulting nanoparticles showed an adequate size (between 130 and 180 nm), and their surface charge could be modulated according to the nanoparticle composition (from +30 mV to -20 mV). All prototypes exhibited a greater association efficiency and nuclease protection for pDNA than for siRNA. However, cell culture experiments evidenced that HA/CS-g-PEG nanoparticles were effective carriers for the delivery of both, siRNA and pDNA, eliciting a biological response with minimal cytotoxicity. Moreover, experiments performed in the HEK-EGFP-Snail1 cell line showed the potential of the HA/CS-g-PEG nanoparticles to silence the expression of the Snail1 transcription factor, an important mediator in tumor progression. CONCLUSIONS: HA/CS-g-PEG nanoparticles can be easily modulated for the delivery of different types of gene molecules, offering great potential for gene therapy applications, as evidenced by their biological performance.


Assuntos
Quitosana/química , DNA/administração & dosagem , Terapia Genética , Ácido Hialurônico/química , Nanopartículas , Plasmídeos , Polietilenoglicóis/química , RNA Interferente Pequeno/administração & dosagem , Sequência de Bases , Linhagem Celular , Técnicas de Silenciamento de Genes , Humanos
12.
Nat Med ; 26(12): 1865-1877, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33077955

RESUMO

An open question in aggressive cancers such as melanoma is how malignant cells can shift the immune system to pro-tumorigenic functions. Here we identify midkine (MDK) as a melanoma-secreted driver of an inflamed, but immune evasive, microenvironment that defines poor patient prognosis and resistance to immune checkpoint blockade. Mechanistically, MDK was found to control the transcriptome of melanoma cells, allowing for coordinated activation of nuclear factor-κB and downregulation of interferon-associated pathways. The resulting MDK-modulated secretome educated macrophages towards tolerant phenotypes that promoted CD8+ T cell dysfunction. In contrast, genetic targeting of MDK sensitized melanoma cells to anti-PD-1/anti-PD-L1 treatment. Emphasizing the translational relevance of these findings, the expression profile of MDK-depleted tumors was enriched in key indicators of a good response to immune checkpoint blockers in independent patient cohorts. Together, these data reveal that MDK acts as an internal modulator of autocrine and paracrine signals that maintain immune suppression in aggressive melanomas.


Assuntos
Carcinogênese/efeitos dos fármacos , Melanoma Experimental/terapia , Midkina/genética , Microambiente Tumoral/genética , Animais , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/genética , Linfócitos T CD8-Positivos/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/genética , Terapia Genética , Humanos , Melanoma Experimental/genética , Melanoma Experimental/patologia , Camundongos , Midkina/farmacologia , NF-kappa B/genética , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Transcriptoma/genética
13.
Cancer Cell ; 35(1): 46-63.e10, 2019 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-30581152

RESUMO

Modulators of mRNA stability are not well understood in melanoma, an aggressive tumor with complex changes in the transcriptome. Here we report the ability of p62/SQSTM1 to extend mRNA half-life of a spectrum of pro-metastatic factors. These include FERMT2 and other transcripts with no previous links to melanoma. Transcriptomic, proteomic, and interactomic analyses, combined with validation in clinical biopsies and mouse models, identified a selected set of RNA-binding proteins (RBPs) recruited by p62, with IGF2BP1 as a key partner. This p62-RBP interaction distinguishes melanoma from other tumors where p62 controls autophagy or oxidative stress. The relevance of these data is emphasized by follow-up analyses of patient prognosis revealing p62 and FERMT2 as adverse determinants of disease-free survival.


Assuntos
Melanoma/metabolismo , Proteínas de Membrana/genética , Proteínas de Neoplasias/genética , RNA Mensageiro/química , Proteínas de Ligação a RNA/metabolismo , Proteína Sequestossoma-1/metabolismo , Animais , Linhagem Celular Tumoral , Progressão da Doença , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica , Humanos , Melanoma/genética , Proteínas de Membrana/química , Camundongos , Proteínas de Neoplasias/química , Transplante de Neoplasias , Mapas de Interação de Proteínas , Proteômica/métodos , Estabilidade de RNA , Análise Serial de Tecidos
14.
Mol Biol Cell ; 14(7): 2844-60, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12857869

RESUMO

Beta-catenin is a multifunctional protein involved in cell-cell adhesion and Wnt signal transduction. Beta-catenin signaling has been proposed to act as inducer of cell proliferation in different tumors. However, in some developmental contexts and cell systems beta-catenin also acts as a positive modulator of apoptosis. To get additional insights into the role of beta-catenin in the regulation of the cell cycle and apoptosis, we have analyzed the levels and subcellular localization of endogenous beta-catenin and its relation with adenomatous polyposis coli (APC) during the cell cycle in S-phase-synchronized epithelial cells. Beta-catenin levels increase in S phase, reaching maximum accumulation at late G2/M and then abruptly decreasing as the cells enter into a new G1 phase. In parallel, an increased cytoplasmic and nuclear localization of beta-catenin and APC is observed during S and G2 phases. In addition, strong colocalization of APC with centrosomes, but not beta-catenin, is detected in M phase. Interestingly, overexpression of a stable form of beta-catenin, or inhibition of endogenous beta-catenin degradation, in epidermal keratinocyte cells induces a G2 cell cycle arrest and leads to apoptosis. These results support a role for beta-catenin in the control of cell cycle and apoptosis at G2/M in normal and transformed epidermal keratinocytes.


Assuntos
Proteína da Polipose Adenomatosa do Colo/metabolismo , Apoptose/fisiologia , Proteínas do Citoesqueleto/metabolismo , Fase G2/fisiologia , Mitose/fisiologia , Transativadores/metabolismo , Animais , Adesão Celular/fisiologia , Núcleo Celular/metabolismo , Células Cultivadas , Centrossomo/metabolismo , Cães , Citometria de Fluxo , Humanos , Camundongos , Microscopia Confocal , beta Catenina
15.
Nat Commun ; 8(1): 2249, 2017 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-29269732

RESUMO

Melanomas are well-known for their altered mRNA expression profiles. Yet, the specific contribution of mRNA binding proteins (mRBPs) to melanoma development remains unclear. Here we identify a cluster of melanoma-enriched genes under the control of CUGBP Elav-like family member 1 (CELF1). CELF1 was discovered with a distinct prognostic value in melanoma after mining the genomic landscape of the 692 known mRBPs across different cancer types. Genome-wide transcriptomic, proteomic, and RNA-immunoprecipitation studies, together with loss-of-function analyses in cell lines, and histopathological evaluation in clinical biopsies, revealed an intricate repertoire of CELF1-RNA interactors with minimal overlap with other malignancies. This systems approach uncovered the oncogene DEK as an unexpected target and downstream effector of CELF1. Importantly, CELF1 and DEK were found to represent early-induced melanoma genes and adverse indicators of overall patient survival. These results underscore novel roles of CELF1 in melanoma, illustrating tumor type-restricted functions of RBPs in cancer.


Assuntos
Proteínas CELF1/fisiologia , Melanoma/genética , Oncogenes , Biologia de Sistemas , Regiões 3' não Traduzidas , Biópsia , Proteínas CELF1/genética , Proteínas CELF1/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Proteínas Cromossômicas não Histona/metabolismo , Humanos , Imunoprecipitação , Melanoma/patologia , Proteínas Oncogênicas/metabolismo , Proteínas de Ligação a Poli-ADP-Ribose/metabolismo , Prognóstico , Proteômica , RNA Neoplásico/genética , Análise de Sobrevida , Transcriptoma
16.
Cancer Res ; 64(18): 6732-9, 2004 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-15374991

RESUMO

Breast cancer is still one of the most important tumors among women in industrialized countries. Improvement in both understanding the molecular events associated with the disease and the development of new additional treatments is still an important goal to be achieved. Choline kinase (ChoK) is increased in human mammary tumors with high incidence, and this activation is associated with clinical variable indicators of greater malignancy. Here, we have investigated the role of ChoK in the development of breast cancer and found that ChoK is both necessary and sufficient for growth factor-induced proliferation in primary human mammary epithelial cells and an absolute requirement for the specific mitogenic response to heregulin in breast tumor-derived cells. These results demonstrate that ChoK plays an essential role in both normal human mammary epithelial cell proliferation and breast tumor progression. Furthermore, inhibition of ChoK shows a strong in vivo antitumor activity against human breast cancer xenografts. Thus, ChoK constitutes a novel bona fide molecular target for the treatment of breast cancer patients.


Assuntos
Neoplasias da Mama/enzimologia , Colina Quinase/antagonistas & inibidores , Colina Quinase/metabolismo , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Colina Quinase/genética , Progressão da Doença , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/enzimologia , Feminino , Humanos , Glândulas Mamárias Humanas/citologia , Glândulas Mamárias Humanas/efeitos dos fármacos , Glândulas Mamárias Humanas/enzimologia , Camundongos , Camundongos Nus , Neuregulina-1/farmacologia , Fosforilcolina/metabolismo , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
17.
FEBS J ; 283(1): 25-38, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26443003

RESUMO

A common feature of solid tumors is their ability to incite the formation of new blood and lymph vessels trough the processes of angiogenesis and lymphangiogenesis, respectively, to support tumor growth and favor metastatic dissemination. As a result of the lack of feedback regulatory control mechanisms or due to the exacerbated presence of pro-angiogenic signals within the tumor microenvironment, the tumor endothelium receives continuous signals to sprout and develop, generating vessels that are structurally and functionally abnormal. An emerging mechanism playing a central role in shaping the tumor vasculature is the endothelial-vesicular network that regulates trafficking/export and degradation of key signaling proteins and membrane receptors, including the vascular endothelial growth-factor receptor-2/3 and members of the Notch pathway. Here we will discuss recent evidence highlighting how vesicular trafficking mechanisms in endothelial cells contribute to pathological angiogenesis/lymphangiogenesis and can provide novel and exploitable targets in antiangiogenic therapies.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Neoplasias/irrigação sanguínea , Neoplasias/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Animais , Células Endoteliais/patologia , Humanos , Linfangiogênese/efeitos dos fármacos , Neoplasias/metabolismo , Neovascularização Patológica/metabolismo , Transporte Proteico/efeitos dos fármacos
18.
Autophagy ; 12(10): 1776-1790, 2016 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-27464255

RESUMO

Melanoma is a paradigm of aggressive tumors with a complex and heterogeneous genetic background. Still, melanoma cells frequently retain developmental traits that trace back to lineage specification programs. In particular, lysosome-associated vesicular trafficking is emerging as a melanoma-enriched lineage dependency. However, the contribution of other lysosomal functions such as autophagy to melanoma progression is unclear, particularly in the context of metastasis and resistance to targeted therapy. Here we mined a broad spectrum of cancers for a meta-analysis of mRNA expression, copy number variation and prognostic value of 13 core autophagy genes. This strategy identified heterozygous loss of ATG5 at chromosome band 6q21 as a distinctive feature of advanced melanomas. Importantly, partial ATG5 loss predicted poor overall patient survival in a manner not shared by other autophagy factors and not recapitulated in other tumor types. This prognostic relevance of ATG5 copy number was not evident for other 6q21 neighboring genes. Melanocyte-specific mouse models confirmed that heterozygous (but not homozygous) deletion of Atg5 enhanced melanoma metastasis and compromised the response to targeted therapy (exemplified by dabrafenib, a BRAF inhibitor in clinical use). Collectively, our results support ATG5 as a therapeutically relevant dose-dependent rheostat of melanoma progression. Moreover, these data have important translational implications in drug design, as partial blockade of autophagy genes may worsen (instead of counteracting) the malignant behavior of metastatic melanomas.


Assuntos
Proteína 5 Relacionada à Autofagia/genética , Perda de Heterozigosidade/genética , Melanoma/genética , Melanoma/patologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Animais , Autofagia/efeitos dos fármacos , Autofagia/genética , Mapeamento Cromossômico , Variações do Número de Cópias de DNA/genética , Metilação de DNA/genética , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Heterozigoto , Camundongos , Metástase Neoplásica , Nevo/genética , Nevo/patologia , Pigmentação/genética , Prognóstico , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas B-raf/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Risco , Análise de Sobrevida
19.
Oncotarget ; 7(48): 78971-78984, 2016 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-27806339

RESUMO

Vascular Endotelial Growth Factors C and D (VEGF-C and VEGF-D) are crucial regulators of lymphangiogenesis, a main event in the metastatic spread of breast cancer tumors. Although inhibition of lymphangiogenic gene expression might be a useful therapeutic strategy to restrict the progression of cancer, the factors involved in the transcriptional repression of these genes are still unknown. We have previously shown that Nuclear Receptor Corepressor 1 (NCoR) and the thyroid hormone receptor ß1 (TRß) inhibit tumor invasion. Here we show that these molecules repress VEGF-C and VEGF-D gene transcription in breast cancer cells, reducing lymphatic vessel density and sentinel lymph node invasion in tumor xenografts. The clinical significance of these results is stressed by the finding that NCoR and TRß transcripts correlate negatively with those of the lymphangiogenic genes and the lymphatic vessel marker LYVE-1 in human breast tumors. Our results point to the use of NCoR and TRß as potential biomarkers for diagnosis or prognosis in breast cancer and suggest that further studies of these molecules as potential targets for anti-lymphangiogenic therapy are warranted.


Assuntos
Neoplasias da Mama/genética , Metástase Linfática/patologia , Correpressor 1 de Receptor Nuclear/metabolismo , Receptores beta dos Hormônios Tireóideos/genética , Animais , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Células MCF-7 , Camundongos , Invasividade Neoplásica , Transplante de Neoplasias , Correpressor 1 de Receptor Nuclear/genética , Prognóstico , Transcrição Gênica , Fator C de Crescimento do Endotélio Vascular/genética , Fator D de Crescimento do Endotélio Vascular/genética , Proteínas de Transporte Vesicular/genética
20.
Cancer Cell ; 30(5): 694-707, 2016 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-27908735

RESUMO

RNA binding proteins (RBPs) modulate cancer progression through poorly understood mechanisms. Here we show that the RBP UNR/CSDE1 is overexpressed in melanoma tumors and promotes invasion and metastasis. iCLIP sequencing, RNA sequencing, and ribosome profiling combined with in silico studies unveiled sets of pro-metastatic factors coordinately regulated by UNR as part of RNA regulons. In addition to RNA steady-state levels, UNR was found to control many of its targets at the level of translation elongation/termination. Key pro-oncogenic targets of UNR included VIM and RAC1, as validated by loss- and gain-of-function studies. Our results identify UNR as an oncogenic modulator of melanoma progression, unravel the underlying molecular mechanisms, and identify potential targets for this therapeutically challenging malignancy.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Melanoma/patologia , Proteínas de Ligação a RNA/metabolismo , Regulação para Cima , Vimentina/genética , Proteínas rac1 de Ligação ao GTP/genética , Animais , Proteínas de Ligação a DNA/genética , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Humanos , Melanoma/genética , Melanoma/metabolismo , Camundongos , Invasividade Neoplásica , Metástase Neoplásica , Transplante de Neoplasias , Processamento Pós-Transcricional do RNA , Proteínas de Ligação a RNA/genética , Ribossomos/genética , Análise de Sequência de RNA/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA