Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochem J ; 474(1): 21-45, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-28008089

RESUMO

Cell-cell communication is critical across an assortment of physiological and pathological processes. Extracellular vesicles (EVs) represent an integral facet of intercellular communication largely through the transfer of functional cargo such as proteins, messenger RNAs (mRNAs), microRNA (miRNAs), DNAs and lipids. EVs, especially exosomes and shed microvesicles, represent an important delivery medium in the tumour micro-environment through the reciprocal dissemination of signals between cancer and resident stromal cells to facilitate tumorigenesis and metastasis. An important step of the metastatic cascade is the reprogramming of cancer cells from an epithelial to mesenchymal phenotype (epithelial-mesenchymal transition, EMT), which is associated with increased aggressiveness, invasiveness and metastatic potential. There is now increasing evidence demonstrating that EVs released by cells undergoing EMT are reprogrammed (protein and RNA content) during this process. This review summarises current knowledge of EV-mediated functional transfer of proteins and RNA species (mRNA, miRNA, long non-coding RNA) between cells in cancer biology and the EMT process. An in-depth understanding of EVs associated with EMT, with emphasis on molecular composition (proteins and RNA species), will provide fundamental insights into cancer biology.


Assuntos
Micropartículas Derivadas de Células/metabolismo , DNA de Neoplasias/metabolismo , Transição Epitelial-Mesenquimal , Lipídeos , Neoplasias/metabolismo , RNA Neoplásico/metabolismo , Animais , Micropartículas Derivadas de Células/genética , Micropartículas Derivadas de Células/patologia , DNA de Neoplasias/genética , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Neoplasias/genética , Neoplasias/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Neoplásico/genética
2.
Expert Rev Proteomics ; 14(1): 69-95, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27838931

RESUMO

INTRODUCTION: Extracellular vesicles (EVs) are critical mediators of intercellular communication, capable of regulating the transcriptional landscape of target cells through horizontal transmission of biological information, such as proteins, lipids, and RNA species. This capability highlights their potential as novel targets for disease intervention. Areas covered: This review focuses on the emerging importance of discovery proteomics (high-throughput, unbiased quantitative protein identification) and targeted proteomics (hypothesis-driven quantitative protein subset analysis) mass spectrometry (MS)-based strategies in EV biology, especially exosomes and shed microvesicles. Expert commentary: Recent advances in MS hardware, workflows, and informatics provide comprehensive, quantitative protein profiling of EVs and EV-treated target cells. This information is seminal to understanding the role of EV subtypes in cellular crosstalk, especially when integrated with other 'omics disciplines, such as RNA analysis (e.g., mRNA, ncRNA). Moreover, high-throughput MS-based proteomics promises to provide new avenues in identifying novel markers for detection, monitoring, and therapeutic intervention of disease.


Assuntos
Micropartículas Derivadas de Células/genética , Exossomos/genética , Vesículas Extracelulares/genética , Proteômica , Biomarcadores , Comunicação Celular/genética , Humanos , Espectrometria de Massas
3.
Sci Rep ; 6: 28321, 2016 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-27324842

RESUMO

Epithelial-mesenchymal transition (EMT) enhances the migration and invasion of cancer cells, and is regulated by various molecular mechanisms including extracellular matrix metalloproteinase (MMP) activity. Previously, we reported transformation of epithelial Madin-Darby canine kidney (MDCK) cells with oncogenic H-Ras (21D1 cells) induces EMT, and significantly elevates MMP1 expression. To explore the biological significance, in this study we characterized 21D1 cells with knocked-down MMP1 expression (21D1(-MMP1)). MMP1 silencing diminished 21D1 cell migration, invasion and anchorage-independent growth in vitro. Additionally, 21D1(-MMP1) cells displayed reduced tumour volume when grown as in vivo subcutaneous xenografts in mice. Depletion of MMP1 lowered the ability of the cellular secretome (extracellular culture medium) to influence recipient cell behaviour. For example, supplementation with 21D1 secretome elevated cell migration of recipient fibroblasts, and enhanced endothelial cell angiogenesis (vessel length and branching). By contrast, 21D1(-MMP1) secretome was less potent in both functional assays. We reveal laminin subunit alpha-5 (LAMA5) as a novel biological substrate of MMP1, that generates internal and C-terminal proteolytic fragments in 21D1 secretome. Furthermore, antibody-based inhibition of integrin αvß3 on endothelial cells nullified the angiogenic capability of 21D1 secretome. Therefore, we report this as a new VEGF-independent mechanism that oncogenic cells may employ to promote tumour angiogenesis.


Assuntos
Células Endoteliais da Veia Umbilical Humana/fisiologia , Laminina/fisiologia , Metaloproteinase 1 da Matriz/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Transformação Celular Neoplásica , Cães , Transição Epitelial-Mesenquimal , Técnicas de Silenciamento de Genes , Humanos , Células Madin Darby de Rim Canino , Masculino , Camundongos Endogâmicos NOD , Camundongos SCID , Transplante de Neoplasias , Neovascularização Patológica/enzimologia , Fragmentos de Peptídeos/fisiologia , Proteólise , Proteoma/metabolismo
4.
Oncotarget ; 7(15): 19709-22, 2016 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-26919098

RESUMO

The metastatic cascade describes the escape of primary tumour cells to distant secondary sites. Cells at the leading tumour edge are thought to undergo epithelial-mesenchymal transition (EMT), to enhance their motility and invasion for spreading. Whether EMT cells directly promote tumour angiogenesis, and the role of exosomes (30-150 nm extracellular vesicles) remains largely unknown. We examined the functional effects of exosomes from MDCK cells, MDCK cells stably expressing YBX1 (MDCKYBX1, intermediate EMT), and Ras-transformed MDCK cells (21D1 cells, complete EMT). 2F-2B cell motility and tube formation (length and branching) was significantly increased following supplementation with MDCKYBX1 or 21D1 exosomes, but not MDCK exosomes. Next, Matrigel™ plugs containing exosome-supplemented 2F-2B cells were subcutaneously injected into mice. Systemic perfusion was only observed for plugs supplemented with MDCKYBX1 or 21D1 exosomes. Comparative proteomics revealed that 21D1 exosomes contained VEGF-associated proteins, while MDCKYBX1 exosomes were enriched with activated Rac1 and PAK2. To validate, 2F-2B cells and HUVECs were pre-treated with PAK inhibitors prior to exosome supplementation. PAK inhibition nullified the effects of MDCKYBX1 exosomes by reducing the tube length and branching to baseline levels. By contrast, the effects of 21D1 exosomes were not significantly decreased. Our results demonstrate for the first time that oncogenic cells undergoing EMT can communicate with endothelial cells via exosomes, and establish exosomal Rac1/PAK2 as angiogenic promoters that may function from early stages of the metastatic cascade.


Assuntos
Células Endoteliais/metabolismo , Células Epiteliais/metabolismo , Exossomos/metabolismo , Quinases Ativadas por p21/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Linhagem Celular , Movimento Celular , Transformação Celular Neoplásica/metabolismo , Cães , Transição Epitelial-Mesenquimal , Humanos , Células Madin Darby de Rim Canino , Neovascularização Patológica/metabolismo , Proteômica/métodos
5.
Oncotarget ; 7(13): 16070-89, 2016 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-26893367

RESUMO

Podoplanin (PDPN) is a transmembrane glycoprotein that plays crucial roles in embryonic development, the immune response, and malignant progression. Here, we report that cells ectopically or endogenously expressing PDPN release extracellular vesicles (EVs) that contain PDPN mRNA and protein. PDPN incorporates into membrane shed microvesicles (MVs) and endosomal-derived exosomes (EXOs), where it was found to colocalize with the canonical EV marker CD63 by immunoelectron microscopy. We have previously found that expression of PDPN in MDCK cells induces an epithelial-mesenchymal transition (EMT). Proteomic profiling of MDCK-PDPN cells compared to control cells shows that PDPN-induced EMT is associated with upregulation of oncogenic proteins and diminished expression of tumor suppressors. Proteomic analysis of exosomes reveals that MDCK-PDPN EXOs were enriched in protein cargos involved in cell adhesion, cytoskeletal remodeling, signal transduction and, importantly, intracellular trafficking and EV biogenesis. Indeed, expression of PDPN in MDCK cells stimulated both EXO and MV production, while knockdown of endogenous PDPN in human HN5 squamous carcinoma cells reduced EXO production and inhibited tumorigenesis. EXOs released from MDCK-PDPN and control cells both stimulated in vitro angiogenesis, but only EXOs containing PDPN were shown to promote lymphatic vessel formation. This effect was mediated by PDPN on the surface of EXOs, as demonstrated by a neutralizing specific monoclonal antibody. These results contribute to our understanding of PDPN-induced EMT in association to tumor progression, and suggest an important role for PDPN in EV biogenesis and/or release and for PDPN-EXOs in modulating lymphangiogenesis.


Assuntos
Transição Epitelial-Mesenquimal/fisiologia , Exossomos/metabolismo , Vesículas Extracelulares/metabolismo , Linfangiogênese/fisiologia , Glicoproteínas de Membrana/metabolismo , Animais , Linhagem Celular Tumoral , Transformação Celular Neoplásica/metabolismo , Cães , Xenoenxertos , Humanos , Células Madin Darby de Rim Canino , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia
6.
Oncotarget ; 6(15): 13718-30, 2015 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-25980435

RESUMO

Epithelial-mesenchymal transition (EMT) describes a morphogenetic program which confers mesenchymal cell properties, such as reduced cell-cell contact and increased cell migration and invasion, to epithelial cells. Here we investigate the role of the pleiotropic transcription/splicing factor and RNA-binding protein nuclease-sensitive element-binding protein 1 (YBX1/YB-1) in increasing the oncogenic potential of epithelial MDCK cells. Characterization of MDCK cells expressing YBX1 (MDCKYBX1 cells) revealed a partial EMT phenotype, including cytosolic relocalization of E-cadherin, increased cell scattering, and anchorage-independent growth. Subcutaneous injection of parental MDCK cells into NOD/SCID mice did not form tumours. Critically, MDCKYBX1 cells established viable tumour xenografts, and immuno-histochemical staining indicated murine vascularization by CD31+ endothelial cells. We analysed the total secretome (containing soluble and extracellular vesicles) of MDCKYBX1 cells to investigate regulation of the tumour microenvironment. YBX1 expression elevated release of secreted factors known to enhance angiogenesis (TGF-ß, CSF-1, NGF, VGF, ADAM9 and ADAM17), compared to MDCK cells. Importantly, treatment with MDCKYBX1 cell-derived secretome increased recipient 2F-2B endothelial cell motility. This defines YBX1 as an oncogenic enhancer that can regulate tumour angiogenesis via release of secreted modulators into the extracellular microenvironment.


Assuntos
Indutores da Angiogênese/metabolismo , Proteína 1 de Ligação a Y-Box/genética , Proteína 1 de Ligação a Y-Box/metabolismo , Animais , Movimento Celular , Cães , Transição Epitelial-Mesenquimal , Células Madin Darby de Rim Canino , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Neovascularização Patológica , Microambiente Tumoral/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Semin Cell Dev Biol ; 40: 72-81, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25724562

RESUMO

Exosomes, a subset of extracellular vesicles (EVs), function as a mode of intercellular communication and molecular transfer. Exosomes facilitate the direct extracellular transfer of proteins, lipids, and miRNA/mRNA/DNAs between cells in vitro and in vivo. The immunological activities of exosomes affect immunoregulation mechanisms including modulating antigen presentation, immune activation, immune suppression, immune surveillance, and intercellular communication. Besides immune cells, cancer cells secrete immunologically active exosomes that influence both physiological and pathological processes. The observation that exosomes isolated from immune cells such as dendritic cells (DCs) modulate the immune response has enforced the way these membranous vesicles are being considered as potential immunotherapeutic reagents. Indeed, tumour- and immune cell-derived exosomes have been shown to carry tumour antigens and promote immunity, leading to eradication of established tumours by CD8(+) T cells and CD4(+) T cells, as well as directly suppressing tumour growth and resistance to malignant tumour development. Further understanding of these areas of exosome biology, and especially of molecular mechanisms involved in immune cell targeting, interaction and manipulation, is likely to provide significant insights into immunorecognition and therapeutic intervention. Here, we review the emerging roles of exosomes in immune regulation and the therapeutic potential in cancer.


Assuntos
Exossomos/imunologia , Neoplasias/metabolismo , Animais , Apresentação de Antígeno , Exossomos/metabolismo , Humanos , Monitorização Imunológica , Proteínas Opsonizantes/metabolismo , Linfócitos T/imunologia , Microambiente Tumoral
8.
Semin Cell Dev Biol ; 40: 60-71, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25721809

RESUMO

Epithelial-mesenchymal transition (EMT) is a highly conserved process defined by the loss of epithelial characteristics, and acquisition of the mesenchymal phenotype. In addition to its central role in development, EMT has been implicated as a cellular process during tumourigenesis which facilitates tumour cell invasion and metastasis. The EMT process has been largely defined by signal transduction networks and transcriptional factors that activate mesenchymal-associated gene expression. Knowledge of secretome components that influence EMT including secreted proteins/peptides and membrane-derived extracellular vesicles (EVs) (i.e., exosomes) has emerged. Here we review EV cargo associated with inducing the hallmarks of EMT and cancer progression, modulators of cell transformation, invasion/migration, angiogenesis, and components involved in establishing the metastatic niche.


Assuntos
Transição Epitelial-Mesenquimal , Exossomos/metabolismo , Neoplasias/patologia , Animais , Transformação Celular Neoplásica , Humanos , Neoplasias/metabolismo
9.
Mol Cell Proteomics ; 12(8): 2148-59, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23645497

RESUMO

Epithelial-mesenchymal transition (EMT) is a highly conserved morphogenic process defined by the loss of epithelial characteristics and the acquisition of a mesenchymal phenotype. EMT is associated with increased aggressiveness, invasiveness, and metastatic potential in carcinoma cells. To assess the contribution of extracellular vesicles following EMT, we conducted a proteomic analysis of exosomes released from Madin-Darby canine kidney (MDCK) cells, and MDCK cells transformed with oncogenic H-Ras (21D1 cells). Exosomes are 40-100 nm membranous vesicles originating from the inward budding of late endosomes and multivesicular bodies and are released from cells on fusion of multivesicular bodies with the plasma membrane. Exosomes from MDCK cells (MDCK-Exos) and 21D1 cells (21D1-Exos) were purified from cell culture media using density gradient centrifugation (OptiPrep™), and protein content identified by GeLC-MS/MS proteomic profiling. Both MDCK- and 21D1-Exos populations were morphologically similar by cryo-electron microscopy and contained stereotypical exosome marker proteins such as TSG101, Alix, and CD63. In this study we show that the expression levels of typical EMT hallmark proteins seen in whole cells correlate with those observed in MDCK- and 21D1-Exos, i.e. reduction of characteristic inhibitor of angiogenesis, thrombospondin-1, and epithelial markers E-cadherin, and EpCAM, with a concomitant up-regulation of mesenchymal makers such as vimentin. Further, we reveal that 21D1-Exos are enriched with several proteases (e.g. MMP-1, -14, -19, ADAM-10, and ADAMTS1), and integrins (e.g. ITGB1, ITGA3, and ITGA6) that have been recently implicated in regulating the tumor microenvironment to promote metastatic progression. A salient finding of this study was the unique presence of key transcriptional regulators (e.g. the master transcriptional regulator YBX1) and core splicing complex components (e.g. SF3B1, SF3B3, and SFRS1) in mesenchymal 21D1-Exos. Taken together, our findings reveal that exosomes from Ras-transformed MDCK cells are reprogrammed with factors which may be capable of inducing EMT in recipient cells.


Assuntos
Transição Epitelial-Mesenquimal , Exossomos/metabolismo , Proteínas ras/metabolismo , Animais , Anexinas/metabolismo , Transformação Celular Neoplásica/metabolismo , Cães , Genes ras , Integrinas/metabolismo , Células Madin Darby de Rim Canino , Peptídeo Hidrolases/metabolismo , Proteoma , Tetraspaninas/metabolismo
10.
J Proteomics ; 78: 545-57, 2013 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-23099347

RESUMO

Human cancer mortality is predominantly determined by the spread of tumour cells from a primary to secondary anatomical location. The metastatic cascade follows tumour cell dissemination, passage through the blood and/or lymphatic system, and colonisation at a distant site. Increased cell motility of cancer cells at the leading tumour edge has been attributed to the epithelial-mesenchymal transition (EMT) which facilitates their release and invasiveness. Mechanisms behind cancer cell dissemination and homing of metastatic cells to a secondary site remain largely unknown. However, certain cancers have a propensity to metastasise to particular organs, and one possible explanation is the seed and soil hypothesis, which is predicated on circulating tumour cells settling in favourable conditions. Extracellular vesicles mediate communication in the tumour microenvironment, stimulate cell migration and invasion, and prepare the pre-metastatic niche. In addition, the tumour stroma can initiate EMT in cancer cells at the invasive front which, in turn, secrete a distinct suite of molecules into the tumour microenvironment. As a first step towards understanding precise regulatory mechanisms, it is important to identify secreted modulators residing within the metastatic niche. Determining the signals and stimuli required to initiate and propagate cancer cell spreading will potentially unearth candidates to limit metastasis.


Assuntos
Transição Epitelial-Mesenquimal , Neoplasias/metabolismo , Microambiente Tumoral , Animais , Humanos , Invasividade Neoplásica , Neoplasias/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...