Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Int J Cancer ; 141(6): 1190-1200, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28577299

RESUMO

In this manuscript, we show the involvement of the uPA/uPAR system in the regulation of aerobic glycolysis of melanoma cells. uPAR over-expression in human melanoma cells controls an invasive and glycolytic phenotype in normoxic conditions. uPAR down-regulation by siRNA or its uncoupling from integrins, and hence from integrin-linked tyrosine kinase receptors (IL-TKRs), by an antagonist peptide induced a striking inhibition of the PI3K/AKT/mTOR/HIF1α pathway, resulting into impairment of glucose uptake, decrease of several glycolytic enzymes and of PKM2, a checkpoint that controls metabolism of cancer cells. Further, binding of uPA to uPAR regulates expression of molecules that govern cell invasion, including extracellular matrix metallo-proteinases inducer (EMPPRIN) and enolase, a glycolytyc enzyme that also serves as a plasminogen receptor, thus providing a common denominator between tumor metabolism and phenotypic invasive features. Such effects depend on the α5ß1-integrin-mediated uPAR connection with EGFR in melanoma cells with engagement of the PI3K-mTOR-HIFα pathway. HIF-1α trans-activates genes whose products mediate tumor invasion and glycolysis, thus providing the common denominator between melanoma metabolism and its invasive features. These findings unveil a unrecognized interaction between the invasion-related uPAR and IL-TKRs in the control of glycolysis and disclose a new pharmacological target (i.e., uPAR/IL-TKRs axis) for the therapy of melanoma.


Assuntos
Melanoma/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Animais , Linhagem Celular Tumoral , Regulação para Baixo , Feminino , Glicólise , Células HEK293 , Xenoenxertos , Humanos , Melanoma/patologia , Camundongos , Camundongos Nus , Camundongos SCID , Invasividade Neoplásica , Fenótipo
2.
J Cell Mol Med ; 19(1): 113-23, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25313007

RESUMO

Gangliosides and the urokinase plasminogen activator receptor (uPAR) tipically partition in specialized membrane microdomains called lipid-rafts. uPAR becomes functionally important in fostering angiogenesis in endothelial progenitor cells (EPCs) upon recruitment in caveolar-lipid rafts. Moreover, cell membrane enrichment with exogenous GM1 ganglioside is pro-angiogenic and opposite to the activity of GM3 ganglioside. On these basis, we first checked the interaction of uPAR with membrane models enriched with GM1 or GM3, relying on the adoption of solid-supported mobile bilayer lipid membranes with raft-like composition formed onto solid hydrophilic surfaces, and evaluated by surface plasmon resonance (SPR) the extent of uPAR recruitment. We estimated the apparent dissociation constants of uPAR-GM1/GM3 complexes. These preliminary observations, indicating that uPAR binds preferentially to GM1-enriched biomimetic membranes, were validated by identifying a pro-angiogenic activity of GM1-enriched EPCs, based on GM1-dependent uPAR recruitment in caveolar rafts. We have observed that addition of GM1 to EPCs culture medium promotes matrigel invasion and capillary morphogenesis, as opposed to the anti-angiogenesis activity of GM3. Moreover, GM1 also stimulates MAPKinases signalling pathways, typically associated with an angiogenesis program. Caveolar-raft isolation and Western blotting of uPAR showed that GM1 promotes caveolar-raft partitioning of uPAR, as opposed to control and GM3-challenged EPCs. By confocal microscopy, we have shown that in EPCs uPAR is present on the surface in at least three compartments, respectively, associated to GM1, GM3 and caveolar rafts. Following GM1 exogenous addition, the GM3 compartment is depleted of uPAR which is recruited within caveolar rafts thereby triggering angiogenesis.


Assuntos
Cavéolas/metabolismo , Células Progenitoras Endoteliais/metabolismo , Gangliosídeo G(M1)/farmacologia , Gangliosídeo G(M3)/farmacologia , Microdomínios da Membrana/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Cavéolas/efeitos dos fármacos , Caveolina 1/metabolismo , Ensaio de Unidades Formadoras de Colônias , Células Progenitoras Endoteliais/efeitos dos fármacos , Humanos , Recém-Nascido , Cinética , Microdomínios da Membrana/efeitos dos fármacos , Fenótipo , Transdução de Sinais
3.
PLoS One ; 11(4): e0154422, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27123929

RESUMO

This study tested the hypothesis that subsets of human T helper cells can orchestrate leukocyte adhesion to synovial fibroblasts (SFbs), thus regulating the retention of leukocytes in the joints of juvenile idiopathic arthritis (JIA) patients. Several cell types, such as monocytes/macrophages, granulocytes, T and B lymphocytes, SFbs and osteoclasts participate in joint tissue damage JIA. Among T cells, an enrichment of classic and non-classic Th1 subsets, has been found in JIA synovial fluid (SF), compared to peripheral blood (PB). Moreover, it has been shown that IL-12 in the SF of inflamed joints mediates the shift of Th17 lymphocytes towards the non-classic Th1 subset. Culture supernatants of Th17, classic and non-classic Th1 clones, have been tested for their ability to stimulate proliferation, and to induce expression of adhesion molecules on SFbs, obtained from healthy donors. Culture supernatants of both classic and non-classic Th1, but not of Th17, clones, were able to induce CD106 (VCAM-1) up-regulation on SFbs. This effect, mediated by tumor necrosis factor (TNF)-α, was crucial for the adhesion of circulating leukocytes on SFbs. Finally, we found that SFbs derived from SF of JIA patients expressed higher levels of CD106 than those from healthy donors, resembling the phenotype of SFbs activated in vitro with Th1-clones supernatants. On the basis of these findings, we conclude that classic and non-classic Th1 cells induce CD106 expression on SFbs through TNF-α, an effect that could play a role in leukocytes retention in inflamed joints.


Assuntos
Artrite Juvenil/imunologia , Meios de Cultivo Condicionados/farmacologia , Cápsula Articular/imunologia , Células Th1/imunologia , Molécula 1 de Adesão de Célula Vascular/imunologia , Adolescente , Adulto , Artrite Juvenil/genética , Artrite Juvenil/patologia , Estudos de Casos e Controles , Adesão Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Criança , Pré-Escolar , Técnicas de Cocultura , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/imunologia , Fibroblastos/patologia , Expressão Gênica , Humanos , Cápsula Articular/patologia , Masculino , Cultura Primária de Células , Líquido Sinovial/citologia , Células Th1/citologia , Células Th1/metabolismo , Células Th17/citologia , Células Th17/imunologia , Células Th17/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Molécula 1 de Adesão de Célula Vascular/genética
4.
J Mol Med (Berl) ; 93(7): 783-94, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25694039

RESUMO

UNLABELLED: The capacity of cancer cells to undergo epithelial-to-mesenchymal transition (EMT) is now considered a hallmark of tumor progression, and it is known that interactions between cancer cells and mesenchymal stem cells (MSCs) of tumor microenvironment may promote this program. Herein, we demonstrate that MSC-conditioned medium (MSC-CM) is a potent inducer of EMT in melanoma cells. The EMT profile acquired by MSC-CM-exposed melanoma cells is characterized by an enhanced level of mesenchymal markers, including TGFß/TGFß-receptors system upregulation, by increased invasiveness and uPAR expression, and in vivo tumor growth. Silencing TGFß in MSC is found to abrogate ability of MSC to promote EMT characteristics in melanoma cells, together with uPAR expression, and this finding is strengthened using an antagonist peptide of TGFßRIII, the so-called P17. Finally, we demonstrate that the uPAR antisense oligonucleotide (uPAR aODN) may inhibit EMT of melanoma cells either stimulated by exogenous TGFß or MSC-CM. Thus, uPAR upregulation in melanoma cells exposed to MSC-medium drives TGFß-mediated EMT. On the whole, TGFß/uPAR dangerous liaison in cancer cell/MSC interactions may disclose a new strategy to abrogate melanoma progression. KEY MESSAGE: Mesenchymal stem cell (MSC)-conditioned medium induces EMT-like profile in melanoma. MSC-derived TGFß promotes uPAR and TGFß/TGFß-receptor upregulation in melanoma. TGFß gene silencing in MSCs downregulates uPAR expression and EMT in melanoma. uPAR downregulation prevents MSC-induced EMT-like profile in melanoma cells. Inhibition of the dangerous TGFß/uPAR relationship might abrogate melanoma progression.


Assuntos
Transição Epitelial-Mesenquimal/fisiologia , Melanoma/patologia , Células-Tronco Mesenquimais/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/antagonistas & inibidores , Fator de Crescimento Transformador beta/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Meios de Cultivo Condicionados/farmacologia , Humanos , Camundongos , Transplante de Neoplasias , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Transplante Heterólogo , Microambiente Tumoral
5.
Oncotarget ; 5(6): 1538-53, 2014 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-24681666

RESUMO

The receptor for the urokinase plasminogen activator (uPAR) is up-regulated in malignant tumors. Historically the function of uPAR in cancer cell invasion is strictly related to its property to promote uPA-dependent proteolysis of extracellular matrix and to open a path to malignant cells. These features are typical of mesenchymal motility. Here we show that the full-length form of uPAR is required when prostate and melanoma cancer cells convert their migration style from the "path generating" mesenchymal to the "path finding" amoeboid one, thus conferring a plasticity to tumor cell invasiveness across three-dimensional matrices. Indeed, in response to a protease inhibitors-rich milieu, prostate and melanoma cells activated an amoeboid invasion program connoted by retraction of cell protrusions, RhoA-mediated rounding of the cell body, formation of a cortical ring of actin and a reduction of Rac-1 activation. While the mesenchymal movement was reduced upon silencing of uPAR expression, the amoeboid one was almost completely abolished, in parallel with a deregulation of small Rho-GTPases activity. In melanoma and prostate cancer cells we have shown uPAR colocalization with ß1/ß3 integrins and actin cytoskeleton, as well integrins-actin co-localization under both mesenchymal and amoeboid conditions. Such co-localizations were lost upon treatment of cells with a peptide that inhibits uPAR-integrin interactions. Similarly to uPAR silencing, the peptide reduced mesenchymal invasion and almost abolished the amoeboid one. These results indicate that full-length uPAR bridges the mesenchymal and amoeboid style of movement by an inward-oriented activity based on its property to promote integrin-actin interactions and the following cytoskeleton assembly.


Assuntos
Movimento Celular , Extensões da Superfície Celular/patologia , Melanoma/patologia , Mesoderma/patologia , Neoplasias da Próstata/patologia , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Citoesqueleto de Actina/metabolismo , Western Blotting , Proliferação de Células , Extensões da Superfície Celular/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Imunofluorescência , Humanos , Integrina beta1/genética , Integrina beta1/metabolismo , Masculino , Melanoma/genética , Melanoma/metabolismo , Mesoderma/metabolismo , Invasividade Neoplásica , Fenótipo , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Ativador de Plasminogênio Tipo Uroquinase/antagonistas & inibidores , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas
6.
Oncotarget ; 5(11): 3711-27, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25003596

RESUMO

The receptor for the urokinase-type plasminogen activator (uPAR) accounts for many features of cancer progression, and is therefore considered a target for anti-tumoral therapy. Only full length uPAR mediates tumor progression. Matrix-metallo-proteinase-12 (MMP12)-dependent uPAR cleavage results into the loss of invasion properties and angiogenesis. MMP12 can be employed in the field of "targeted therapies" as a biological drug to be delivered directly in patient's tumor mass. Endothelial Progenitor Cells (EPCs) are selectively recruited within the tumor and could be used as cellular vehicles for delivering anti-cancer molecules. The aim of our study is to inhibit cancer progression by engeneering ECFCs, a subset of EPC, with a lentivirus encoding the anti-tumor uPAR-degrading enzyme MMP12. Ex vivo manipulated ECFCs lost the capacity to perform capillary morphogenesis and acquired the anti-tumor and anti-angiogenetic activity. In vivo MMP12-engineered ECFCs cleaved uPAR within the tumor mass and strongly inhibited tumor growth, tumor angiogenesis and development of lung metastasis. The possibility to exploit tumor homing and activity of autologous MMP12-engineered ECFCs represents a novel way to combat melanoma by a "personalized therapy", without rejection risk. The i.v. injection of radiolabelled MMP12-ECFCs can thus provide a new theranostic approach to control melanoma progression and metastasis.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Células Progenitoras Endoteliais/fisiologia , Metaloproteinase 12 da Matriz/administração & dosagem , Melanoma/terapia , Receptores de Ativador de Plasminogênio Tipo Uroquinase/administração & dosagem , Animais , Linhagem Celular Tumoral , Proliferação de Células , Células Progenitoras Endoteliais/citologia , Células Progenitoras Endoteliais/enzimologia , Humanos , Masculino , Metaloproteinase 12 da Matriz/biossíntese , Metaloproteinase 12 da Matriz/genética , Melanoma/genética , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Receptores de Ativador de Plasminogênio Tipo Uroquinase/biossíntese , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Transfecção , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa