Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Exp Dermatol ; 25(8): 618-22, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27060463

RESUMO

CD147 has been implicated in melanoma invasion and metastasis mainly through increasing metalloproteinase synthesis and regulating VEGF/VEGFR signalling. In this study, the prognostic value of CD147 expression was investigated in a cohort of 196 cutaneous melanomas including 136 consecutive primary malignant melanomas, 30 lymph nodes, 16 in-transit and 14 visceral metastases. A series of 10 normal skin, 10 blue nevi and 10 dermal nevi was used as control. CD147 expression was assessed by immunohistochemistry, and the association of its expression with the clinicopathological characteristics of patients and survival was evaluated using univariate and multivariate statistical analyses. Univariate analysis showed that high CD147 expression was significantly associated with metastatic potential and with a reduced overall survival (P < 0.05 for both) in primary melanoma patients. CD147 expression level was correlated with histological factors which were associated with prognosis: Clark level, ulceration status and more particularly with Breslow index (r = 0.7, P < 10(-8) ). Multivariate analysis retained CD147 expression level and ulceration status as predicting factors for metastasis and overall survival (P < 0.05 for both). CD147 emerges as an important factor in the aggressive behaviour of melanoma and deserves further evaluation as an independent prognostic biomarker.


Assuntos
Basigina/metabolismo , Melanoma/metabolismo , Neoplasias Cutâneas/metabolismo , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/metabolismo , Estudos de Casos e Controles , Feminino , França/epidemiologia , Humanos , Masculino , Melanoma/diagnóstico , Melanoma/mortalidade , Melanoma/patologia , Pessoa de Meia-Idade , Prognóstico , Pele/patologia , Neoplasias Cutâneas/diagnóstico , Neoplasias Cutâneas/mortalidade , Neoplasias Cutâneas/patologia
2.
Biochim Biophys Acta ; 1840(8): 2581-8, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24608032

RESUMO

BACKGROUND: Elevated levels of EMMPRIN/CD147 in cancer tissues have been correlated with tumor progression but the regulation of its expression is not yet understood. Here, the regulation of EMMPRIN expression was investigated in testicular germ cell tumor (TGCTs) cell lines. METHODS: EMMPRIN expression in seminoma JKT-1 and embryonal carcinoma NT2/D1 cell lines was determined by Western blot, immunofluorescence and qRT-PCR. Membrane vesicles (MVs) secreted from these cells, treated or not with EMMPRIN siRNA, were isolated by differential centrifugations of their conditioned medium. MMP-2 was analyzed by zymography and qRT-PCR. RESULTS: The more aggressive embryonic carcinoma NT2/D1 cells expressed more EMMPRIN mRNA than the seminoma JKT-1 cells, but surprisingly contained less EMMPRIN protein, as determined by immunoblotting and immunostaining. The protein/mRNA discrepancy was not due to accelerated protein degradation in NT2/D1 cells, but by the secretion of EMMPRIN within MVs, as the vesicles released from NT2/D1 contained considerably more EMMPRIN than those released from JKT-1. EMMPRIN-containing MVs obtained from NT2/D1, but not from EMMPRIN-siRNA treated NT2/D1, increased MMP-2 production in fibroblasts to a greater extent than those from JKT-1 cells. CONCLUSION AND GENERAL SIGNIFICANCE: The data presented show that the more aggressive embryonic carcinoma cells synthesize more EMMPRIN than seminoma cells, but which they preferentially target to secreted MVs, unlike seminoma cells which retain EMMPRIN within the cell membrane. This cellular event points to a mechanism by which EMMPRIN expressed by malignant testicular cells can exert its MMP inducing effect on distant cells within the tumor microenvironment to promote tumor invasion. This article is part of a Special Issue entitled Matrix-mediated cell behaviour and properties.


Assuntos
Basigina/metabolismo , Comunicação Celular , Membrana Celular/metabolismo , Metaloproteinase 2 da Matriz/biossíntese , Neoplasias Embrionárias de Células Germinativas/enzimologia , Neoplasias Embrionárias de Células Germinativas/patologia , Vesículas Secretórias/metabolismo , Neoplasias Testiculares/enzimologia , Neoplasias Testiculares/patologia , Basigina/genética , Linhagem Celular Tumoral , Fibroblastos/enzimologia , Fibroblastos/patologia , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Microdomínios da Membrana/metabolismo , Neoplasias Embrionárias de Células Germinativas/genética , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células Estromais/patologia , Neoplasias Testiculares/genética
3.
Exp Dermatol ; 24(6): 443-8, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25807898

RESUMO

EMMPRIN is known to promote tumor invasion through extracellular matrix (ECM) degradation. Here we report that EMMPRIN can regulate melanoma cell adhesion to the ECM through an interaction with ß1 integrin involving kindlin-3. In this study, EMMPRIN knockdown in the human melanoma cell line M10 using siRNA decreased cell invasion and significantly increased cell adhesion and spreading. A morphological change from a round to a spread shape was observed associated with enhanced phalloidin-labelled actin staining. In situ proximity ligation assay and co-immunoprecipitation revealed that EMMPRIN silencing increased the interaction of ß1 integrin with kindlin-3, a focal adhesion protein. This was associated with an increase in ß1 integrin activation and a decrease in the phosphorylation of the downstream integrin kinase FAK. Moreover, the expression at both the transcript and protein level of kindlin-3 and of ß1 integrin was inversely regulated by EMMPRIN. EMMPRIN did not regulate either talin expression or its interaction with ß1 integrin. These results are consistent with our in vivo demonstration that EMMPRIN inhibition increased ß1 integrin activation and its interaction with kindlin-3. To conclude, these findings reveal a new role of EMMPRIN in tumor cell migration through ß1 integrin/kindlin-3-mediated adhesion pathway.


Assuntos
Basigina/fisiologia , Adesão Celular/fisiologia , Integrina beta1/fisiologia , Melanoma/patologia , Proteínas de Membrana/fisiologia , Proteínas de Neoplasias/fisiologia , Neoplasias Cutâneas/patologia , Animais , Basigina/efeitos dos fármacos , Basigina/genética , Linhagem Celular Tumoral , Forma Celular/fisiologia , Matriz Extracelular/fisiologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Xenoenxertos , Humanos , Técnicas In Vitro , Melanoma/fisiopatologia , Camundongos , Camundongos Nus , RNA Interferente Pequeno/farmacologia , Transdução de Sinais/fisiologia , Neoplasias Cutâneas/fisiopatologia
4.
Oncogene ; 43(21): 1620-1630, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38570692

RESUMO

The role of the focal adhesion protein kindlin-3 as a tumor suppressor and its interaction mechanisms with extracellular matrix constitute a major field of investigation to better decipher tumor progression. Besides the well-described role of kindlin-3 in integrin activation, evidence regarding modulatory functions between melanoma cells and tumor microenvironment are lacking and data are needed to understand mechanisms driven by kindlin-3 inactivation. Here, we show that kindlin-3 inactivation through knockdown or somatic mutations increases BRAFV600mut melanoma cells oncogenic properties via collagen-related signaling by decreasing cell adhesion and enhancing proliferation and migration in vitro, and by promoting tumor growth in mice. Mechanistic analysis reveals that kindlin-3 interacts with the collagen-activated tyrosine kinase receptor DDR1 (Discoidin domain receptor 1) modulating its expression and its interaction with ß1-integrin. Kindlin-3 knockdown or mutational inactivation disrupt DDR1/ß1-integrin complex in vitro and in vivo and its loss improves the anti-proliferative effect of DDR1 inhibition. In agreement, kindlin-3 downregulation is associated with DDR1 over-expression in situ and linked to worse melanoma prognosis. Our study reveals a unique mechanism of action of kindlin-3 in the regulation of tumorigenesis mediated by the collagen-activated tyrosine kinase receptor DDR1 thus paving the way for innovative therapeutic targeting approaches in melanoma.


Assuntos
Proliferação de Células , Receptor com Domínio Discoidina 1 , Melanoma , Proteínas de Membrana , Proteínas de Neoplasias , Humanos , Receptor com Domínio Discoidina 1/genética , Receptor com Domínio Discoidina 1/metabolismo , Animais , Melanoma/patologia , Melanoma/genética , Melanoma/metabolismo , Camundongos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proliferação de Células/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Linhagem Celular Tumoral , Integrina beta1/metabolismo , Integrina beta1/genética , Movimento Celular/genética , Adesão Celular/genética , Colágeno/metabolismo , Transdução de Sinais/genética , Regulação Neoplásica da Expressão Gênica
5.
J Biol Chem ; 287(48): 40339-49, 2012 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-23060448

RESUMO

BACKGROUND: The role of pleiotrophin and its receptors RPTPß/ζ and Syndecan-3 during tumor metastasis remains unknown. RESULTS: RPTPß/ζ knockdown initiates EMT, promotes pleiotrophin-mediated migration and attachment through Syndecan-3 and induces in vivo metastasis. CONCLUSION: RPTPß/ζ plays a suppressor-like role in prostate cancer metastasis. SIGNIFICANCE: Boosting RPTPß/ζ or attenuating Syndecan-3 signaling pathways may lead to more effective therapeutic strategies in treating prostate cancer metastasis. Pleiotrophin is a growth factor that induces carcinogenesis. Despite the fact that many published reports focused on the role of pleiotrophin and its receptors, receptor protein tyrosine phosphatase (RPTPß/ζ), and syndecan-3 during tumor development, no information is available regarding their function in tumor metastasis. To investigate the mechanism through which pleiotrophin regulates tumor metastasis, we used two different prostate carcinoma cell lines, DU145 and PC3, in which the expression of RPTPß/ζ or syndecan-3 was down-regulated by the RNAi technology. The loss of RPTPß/ζ expression initiated epithelial-to-mesenchymal transition (EMT) and increased the ability of the cells to migrate and invade. Importantly, the loss of RPTPß/ζ expression increased metastasis in nude mice in an experimental metastasis assay. We also demonstrate that RPTPß/ζ counterbalanced the pleiotrophin-mediated syndecan-3 pathway. While the inhibition of syndecan-3 expression inhibited the pleiotrophin-mediated cell migration and attachment through the Src and Fak pathway, the inhibition of RPTPß/ζ expression increased pleiotrophin-mediated migration and attachment through an interaction with Src and the subsequent activation of a signal transduction pathway involving Fak, Pten, and Erk1/2. Taken together, these results suggest that the loss of RPTPß/ζ may contribute to the metastasis of prostate cancer cells by inducing EMT and promoting pleiotrophin activity through the syndecan-3 pathway.


Assuntos
Metástase Neoplásica , Neoplasias da Próstata/enzimologia , Proteínas Tirosina Fosfatases Classe 5 Semelhantes a Receptores/deficiência , Animais , Linhagem Celular Tumoral , Movimento Celular , Humanos , Masculino , Camundongos , Camundongos Knockout , Camundongos Nus , Fosforilação , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Proteínas Tirosina Fosfatases Classe 5 Semelhantes a Receptores/genética , Transdução de Sinais , Sindecana-3/genética , Sindecana-3/metabolismo , Regulação para Cima
6.
J Biol Chem ; 287(52): 43685-93, 2012 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-23109338

RESUMO

Blockage of the metastasis process remains a significant clinical challenge, requiring innovative therapeutic approaches. For this purpose, molecules that inhibit matrix metalloproteinases activity or induce the expression of their natural inhibitor, the tissue inhibitor of metalloproteinases (TIMPs), are potentially interesting. In a previous study, we have shown that synthetic ligands binding to cell surface nucleolin/nucleophosmin and known as HB 19 for the lead compound and NucAnt 6L (N6L) for the most potent analog, inhibit both tumor growth and angiogenesis. Furthermore, they prevent metastasis in a RET transgenic mice model which develops melanoma. Here, we investigated the effect of N6L on the invasion capacity of MDA-MB-435 melanoma cells. Our results show that the multivalent pseudopeptide N6L inhibited Matrigel invasion of MDA-MB-435 cells in a modified Boyden chamber model. This was associated with an increase in TIMP-3 in the cell culture medium without a change in TIMP-3 mRNA expression suggesting its release from cell surface and/or extracellular matrix. This may be explained by our demonstrated N6L interaction with sulfated glycosaminoglycans and consequently the controlled bioavailability of glycosaminoglycan-bound TIMP-3. The implication of TIMP-3 in N6L-induced inhibition of cell invasion was evidenced by siRNA silencing experiments showing that the loss of TIMP-3 expression abrogated the effect of N6L. The inhibition of tumor cell invasion by N6L demonstrated in this study, in addition to its previously established inhibitory effect on tumor growth and angiogenesis, suggests that N6L represents a promising anticancer drug candidate warranting further investigation.


Assuntos
Antineoplásicos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Chumbo/farmacologia , Proteínas de Neoplasias/biossíntese , Neoplasias/metabolismo , Peptídeos/farmacologia , Inibidor Tecidual de Metaloproteinase-3/biossíntese , Animais , Antineoplásicos/química , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Chumbo/química , Camundongos , Camundongos Transgênicos , Invasividade Neoplásica , Proteínas de Neoplasias/genética , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/patologia , Peptídeos/química , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Neoplásico/biossíntese , Inibidor Tecidual de Metaloproteinase-3/genética
7.
Am J Pathol ; 179(3): 1278-86, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21777561

RESUMO

Dry eye is a common disease that develops as a result of alteration of tear fluid, leading to osmotic stress and a perturbed epithelial barrier. Matrix metalloproteinase-9 (MMP-9) may be important in dry eye disease, as its genetic knockout conferred resistance to the epithelial disruption. We show that extracellular matrix metalloproteinase inducer (EMMPRIN; also termed CD147), an inducer of MMP expression, participates in the pathogenesis of dry eye through MMP-mediated cleavage of occludin, an important component of tight junctions. EMMPRIN expression was increased on the ocular surface of dry eye patients and correlated with those of MMP-9. High osmolarity in cell culture, mimicking dry eye conditions, increased both EMMPRIN and MMP-9 and resulted in the disruption of epithelial junctions through the cleavage of occludin. Exogenously added recombinant EMMPRIN had similar effects that were abrogated in the presence of the MMP inhibitor marimastat. Membrane occludin immunostaining was markedly increased in the apical corneal epithelium of both EMMPRIN and MMP-9 knock-out mice. Furthermore, an inverse correlation between EMMPRIN and occludin membrane staining was consistently observed both in vitro and in vivo as a function of corneal epithelial cells differentiation. These data suggest a possible role of EMMPRIN in regulating the amount of occludin at the cell surface in homeostasis beyond pathological situations such as dry eye disease, and EMMPRIN may be essential for the formation and maintenance of organized epithelial structure.


Assuntos
Basigina/farmacologia , Síndromes do Olho Seco/etiologia , Inibidores de Metaloproteinases de Matriz , Proteínas de Membrana/efeitos dos fármacos , Animais , Basigina/metabolismo , Diferenciação Celular , Síndromes do Olho Seco/metabolismo , Epitélio Corneano/efeitos dos fármacos , Homeostase , Humanos , Camundongos , Camundongos Knockout , Ocludina , Concentração Osmolar , Proteínas Recombinantes/farmacologia
8.
BMC Cancer ; 12: 115, 2012 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-22443116

RESUMO

BACKGROUNDS: An elevated level of EMMPRIN in cancer tissues have been correlated with tumor invasion in numerous cancers including oral cavity and larynx. Although EMMPRIN's effect has been generally attributed to its MMP inducing activity, we have previously demonstrated in breast cancer model that EMMPRIN can also enhance invasion by upregulating uPA. In this study, the role of EMMPRIN in regulating uPA and invasion was investigated in oral squamous cell carcinoma (OSCC) progression. METHODS: Precancerous and invasive oral tumoral tissues were used as well as the corresponding cell lines, DOK and SCC-9 respectively. The paracrine regulation of uPA by EMMPRIN was investigated by treating culture cells with EMMPRIN-enriched membrane vesicles. UPA expression was analyzed by qPCR and immunostaining and the consequence on the invasion capacity was studied using modified Boyden chamber assay, in the presence or absence of EMMPRIN blocking antibody, the uPA inhibitor amiloride or the MMP inhibitor marimastat. RESULTS: OSCC tumors were shown to express more EMMPRIN and uPA compared to dysplastic lesions. The corresponding cell models, SCC-9 and DOK cells, displayed similar expression pattern. In both cell types EMMPRIN upregulated the expression of uPA as well as that of MMP-2 and MMP-9. EMMPRIN treatment led to a significant increase in cell invasion both in the invasive SCC-9 and in the less invasive dysplastic DOK cells, in an MMP and uPA dependent manner. CONCLUSIONS: Our results suggest that the upregulation of uPA contributes to EMMPRIN's effect in promoting oral tumor invasion.


Assuntos
Basigina/fisiologia , Carcinoma de Células Escamosas/metabolismo , Neoplasias Bucais/metabolismo , Proteínas de Neoplasias/fisiologia , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Western Blotting , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Progressão da Doença , Humanos , Imuno-Histoquímica , Neoplasias Bucais/patologia , Invasividade Neoplásica/patologia , Lesões Pré-Cancerosas/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Regulação para Cima
9.
Oncogene ; 41(15): 2254-2264, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35217792

RESUMO

More than 70% of human NRASmut melanomas are resistant to MEK inhibitors highlighting the crucial need for efficient therapeutic strategies for these tumors. CD147, a membrane receptor, is overexpressed in most cancers including melanoma and is associated with poor prognosis. We show here that CD147i, a specific inhibitor of CD147/VEGFR-2 interaction represents a potential therapeutic strategy for NRASmut melanoma cells. It significantly inhibited the malignant properties of NRASmut melanomas ex vivo and in vivo. Importantly, NRASmut patient's-derived xenografts, which were resistant to MEKi, became sensitive when combined with CD147i leading to decreased proliferation ex vivo and tumor regression in vivo. Mechanistic studies revealed that CD147i effects were mediated through STAT3 pathway. These data bring a proof of concept on the impact of the inhibition of CD147/VEGFR-2 interaction on melanoma progression and represents a new therapeutic opportunity for NRASmut melanoma when combined with MEKi.


Assuntos
Basigina , Melanoma , Receptor 2 de Fatores de Crescimento do Endotélio Vascular , Basigina/antagonistas & inibidores , Basigina/metabolismo , Linhagem Celular Tumoral , GTP Fosfo-Hidrolases/genética , GTP Fosfo-Hidrolases/metabolismo , Humanos , Melanoma/tratamento farmacológico , Melanoma/genética , Melanoma/metabolismo , Proteínas de Membrana/genética , Mutação , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas B-raf/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
10.
J Cell Physiol ; 226(1): 141-9, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20648565

RESUMO

Matrix metalloproteinases (MMPs) are thought to play an important role in skeletal muscle cell growth and differentiation. In view of the MMP inducing function of EMMPRIN/CD147, its role in myogenic cell differentiation was investigated. EMMPRIN level increased during differentiation of both rat primary myoblasts derived from satellite cells and mouse C2.7 myogenic cells and was associated with an alteration in its molecular forms. In parallel, expression of pro-MMP-9 gradually decreased and that of pro-MMP-2 and active MMP-2 increased. While small interfering RNA (siRNA) inhibition of EMMPRIN expression accelerated cell differentiation, exogenously added recombinant EMMPRIN inhibited differentiation by an MMP-mediated mechanism, as the MMP inhibitor marimastat abrogated EMMPRIN's effect. Our results further suggest that EMMPRIN regulates differentiation through an MMP activation of transforming growth factor beta (TGFß), a known inhibitor of myoblast's differentiation, as the increased activation and signaling of TGFß by EMMPRIN was attenuated in the presence of marimastat. EMMPRIN inhibition may thus represent a novel strategy in the treatment of muscular degenerative disorders.


Assuntos
Basigina/metabolismo , Diferenciação Celular/fisiologia , Inativação Gênica , Metaloproteinases da Matriz/metabolismo , Células Satélites de Músculo Esquelético/citologia , Animais , Basigina/genética , Linhagem Celular , Células Cultivadas , Indução Enzimática , Matriz Extracelular/enzimologia , Matriz Extracelular/fisiologia , Metaloproteinases da Matriz/biossíntese , Metaloproteinases da Matriz/genética , Camundongos , Desenvolvimento Muscular/fisiologia , Mioblastos/citologia , Mioblastos/fisiologia , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Wistar , Células Satélites de Músculo Esquelético/fisiologia , Fator de Crescimento Transformador beta/metabolismo
11.
Blood ; 114(27): 5547-56, 2009 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-19837976

RESUMO

Extracellular matrix metalloproteinase inducer (EMMPRIN/CD147) is thought to promote tumor angiogenesis mostly through its protease-inducing function and more recently by its ability to increase tumor cell expression of vascular endothelial growth factor (VEGF). In this study, we present evidence that EMMPRIN can promote angiogenesis by a direct effect on endothelial cells through a paracrine regulation of the VEGF/VEGF-receptor (VEGFR) system. Using human microvascular endothelial cell line-1 endothelial cells, we show that EMMPRIN selectively increased the soluble VEGF isoforms (121 and 165), but not the matrix-bound VEGF 189 form. In addition, EMMPRIN up-regulated the expression of VEGFR-2 without an effect on VEGFR-1. This increase in VEGFR-2 was responsible for the observed EMMPRIN stimulation of the migratory and tube formation capacity of endothelial cells. EMMPRIN's effects, which were matrix metalloproteinase and urokinase-type plasminogen activator independent, were mediated primarily through hypoxia-inducible factor-2alpha expression, also up-regulated by EMMPRIN. VEGFR-2 increase was also observed in vivo in a mouse model of xenograph tumors overexpressing EMMPRIN. These results suggest that in addition to increasing protease production, EMMPRIN may contribute to the formation of a reactive stroma also through the up-regulation of hypoxia-inducible factor-2alpha, VEGFR-2, and the soluble forms of VEGF in endothelial cells, thus directly regulating the angiogenic process.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Basigina/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Basigina/genética , Basigina/metabolismo , Células CHO , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Cricetinae , Cricetulus , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Humanos , Immunoblotting , Camundongos , Camundongos Nus , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Solubilidade , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
12.
Cancers (Basel) ; 13(19)2021 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-34638342

RESUMO

Malignant melanoma is one of the most aggressive skin cancers and is characterized by early lymph node metastasis and the capacity to develop resistance to therapies. Hence, understanding the regulation of lymphangiogenesis through mechanisms contributing to lymphatic vessel formation represents a treatment strategy for metastatic cancer. We have previously shown that CD147, a transmembrane glycoprotein overexpressed in melanoma, regulates the angiogenic process in endothelial cells. In this study, we show a correlation between high CD147 expression levels and the number of lymphatic vessels expressing LYVE-1, Podoplanin, and VEGFR-3 in human melanoma lymph nodes. CD147 upregulates in vitro lymphangiogenesis and its related mediators through the PROX-1 transcription factor. In vivo studies in a melanoma model confirmed that CD147 is involved in metastasis through a similar mechanism as in vitro. This study, demonstrating the paracrine role of CD147 in the lymphangiogenesis process, suggests that CD147 could be a promising target for the inhibition of melanoma-associated lymphangiogenesis.

13.
Prog Retin Eye Res ; 28(1): 19-33, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19056510

RESUMO

In the cornea, the epithelium and the underlying stroma are separated by the basement membrane and Bowman's layer. The disruption of these anatomical barriers during wound healing represents a key step which initiates tissue remodeling through the modification of the epithelial-stromal interactions (ESI). Diffusible cytokines are generally viewed as central modulators in the bidirectional communication between these epithelial and stromal compartments and their implication in all stages of the wound healing process has been an active area of research for many years. Our studies which aimed to explore mechanisms of matrix degradation in pathological corneal wound healing have shown that EMMPRIN, a glycoprotein expressed on corneal epithelial cell surface, can induce matrix metalloproteinase (MMP) production and myofibroblasts differentiation after direct interaction with corneal fibroblasts. EMMPRIN appears therefore as a potential mediator of ESI by direct cell-cell contact which represents a new mechanism for dysregulated MMPs' induction observed in corneal ulcerations. These direct epithelial-stromal interactions (direct-ESI) can occur when delayed epithelial healing prevents regeneration of the basement membrane and allows the two cell types to come into close proximity. We propose that prevention of these interactions through inhibition of EMMPRIN may represent a promising therapeutic strategy in the inhibition of MMP induction in ulceration.


Assuntos
Basigina/fisiologia , Comunicação Celular/fisiologia , Substância Própria/fisiologia , Epitélio Corneano/fisiologia , Metaloproteinases da Matriz/biossíntese , Cicatrização/fisiologia , Animais , Humanos
14.
Int J Biol Markers ; 25(2): 112-5, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20544684

RESUMO

When renal cell carcinoma (RCC) metastasizes to bone (a frequent site of systemic spread of this cancer) it becomes highly resistant to radiation therapy and chemotherapy. A better understanding of the biology of bone metastasis in RCC may permit to identify biomarkers for early detection of subclinical disease and better stratification of patients prior to treatment. We therefore investigated in this study, using a multiplex real-time RT-PCR assay, the expression of a panel of 16 biomarkers involved in angiogenesis and tumor invasion; the panel was applied to primary tumors and normal tissues obtained from clear-cell RCC patients with and without bone metastases. We identified a novel combination of biomarkers associated with the risk of bone metastasis. Among the transcripts of the genes studied, VEGFR-1, VEGFR-2, HIF-1alpha, uPA , and PA I-1 overexpression in tumor tissues was significantly associated with the presence of bone metastasis (p=0.02, p=0.02, p<0.0001, p=0.04, and p=0.03, respectively). No differences were found in the expression of these transcripts in the corresponding normal tissues. This preliminary study provides a promising tool that may help in the management of RCC patients with bone metastasis. Indeed, these predictive markers could be useful to identify subclinical disease, improve staging, and guide treatment decisions.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Ósseas/secundário , Carcinoma de Células Renais/patologia , Perfilação da Expressão Gênica , Neoplasias Renais/patologia , Idoso , Biomarcadores Tumorais/análise , Biomarcadores Tumorais/isolamento & purificação , Neoplasias Ósseas/diagnóstico , Neoplasias Ósseas/genética , Carcinoma de Células Renais/diagnóstico , Carcinoma de Células Renais/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Predisposição Genética para Doença , Humanos , Neoplasias Renais/diagnóstico , Neoplasias Renais/genética , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Fatores de Risco
15.
FASEB J ; 22(4): 1144-54, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17965264

RESUMO

Extracellular matrix metalloproteinase inducer (EMMPRIN) is a cell surface glycoprotein enriched on tumor cells and normal epithelia. It is mainly known for its ability to induce matrix metalloproteinase production in fibroblasts following epithelial-stromal interaction. We sought to examine whether EMMPRIN has a broader role promoting fibroblast-to-myofibroblast differentiation. Because alpha-smooth muscle actin (alphaSMA) is considered a marker of this differentiation process, we analyzed the effect of EMMPRIN on its expression in corneal and skin fibroblasts by Western blots, immunocytochemistry, and a functional assay of collagen lattice contraction. Increasing EMMPRIN expression by cDNA transfection or by treatment with exogenously added recombinant EMMPRIN resulted in an up-regulation of alphaSMA expression. EMMPRIN also increased the contractile properties of the treated fibroblasts as demonstrated by the immunohistochemical appearance of stress fibers and by the accelerated contraction of fibroblast-embedded collagen lattices. Blocking EMMPRIN expression by small interfering RNA inhibited alphaSMA and collagen gel contraction induced not only by EMMPRIN but also by transforming growth factor-beta, a major mediator of myofibroblast differentiation that also regulated EMMPRIN expression. These findings, combined with the fact that EMMPRIN and alphaSMA colocalized to the same cells in the stroma of pathological corneas, expand on the mechanism by which EMMPRIN remodels extracellular matrix during wound healing and cancer.


Assuntos
Actinas/metabolismo , Basigina/metabolismo , Colágeno/fisiologia , Fibroblastos/citologia , Músculo Liso/metabolismo , Basigina/análise , Diferenciação Celular , Linhagem Celular , Substância Própria/metabolismo , Humanos , Imuno-Histoquímica , Contração Muscular , Músculo Liso/citologia , Fator de Crescimento Transformador beta/metabolismo
16.
Eur Cell Mater ; 18: 84-95, 2009 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-19908197

RESUMO

Dentin Matrix Protein 1 (DMP1) plays a regulatory role in dentin mineralization and can also function as a signaling molecule. MMP-2 (matrix metalloproteinase-2) is a predominant protease in the dentin matrix that plays a prominent role in tooth formation and a potential role during the carious process. The possibility that MMP-2 can cleave DMP1 to release biologically active peptides was investigated in this study. DMP1, both in the recombinant form and in its native state within the dentin matrix, was shown to be a substrate for MMP-2. Proteolytic processing of DMP1 by MMP-2 produced two major peptides, one that contains the C-terminal region of the protein known to carry both the ASARM (aspartic acid and serine rich domain) domain involved in biomineralization and the DNA binding site of DMP1. In vitro experiments with recombinant N- and C-terminal polypeptides mimicking the MMP-2 cleavage products of DMP1 demonstrated an effect of the C-polypeptide on the differentiation of dental pulp stem/progenitor cells to a putative odontoblast phenotype. In vivo implantation of this peptide in a rat injured pulp model induced a rapid formation of a homogeneous dentin bridge covered by a palisade of orientated cells expressing dentin sialoprotein (DSP) and DMP1, attesting an efficient repair process. These data suggest that a peptide generated through the proteolytic processing of DMP1 by MMP-2 can regulate the differentiation of mesenchymal cells during dentinogenesis and thus sustain reparative dentin formation in pathological situations such as carious decay. In addition, these data open a new therapeutic possibility of using this peptide to regenerate dentin after an injury.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Polpa Dentária/citologia , Proteínas da Matriz Extracelular/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Fosfoproteínas/metabolismo , Células-Tronco/fisiologia , Adulto , Sequência de Aminoácidos , Animais , Dentinogênese/genética , Proteínas da Matriz Extracelular/fisiologia , Humanos , Camundongos , Dados de Sequência Molecular , Fragmentos de Peptídeos/fisiologia , Fosfoproteínas/fisiologia , Ratos , Proteínas Recombinantes/metabolismo , Células-Tronco/citologia
17.
Cancer Res ; 67(1): 9-15, 2007 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17210677

RESUMO

Extracellular matrix metalloproteinase inducer (EMMPRIN) is a membrane glycoprotein overexpressed in many cancer tissues and is known for its ability to stimulate MMP expression. In this work, we show that EMMPRIN is also a regulator of the urokinase-type plasminogen activation (uPA) system of serine proteases, thus participating to the increase of the overall proteolytic function of the cancer cells. Enhanced EMMPRIN expression in a tumorigenic breast epithelial cell line NS2T2A increased the levels of uPA, uPA receptor, and the uPA inhibitor plasminogen activator inhibitor-1 (PAI-1), as measured by quantitative reverse transcription-PCR, Western blot, and plasminogen-casein zymography. This response was down-regulated by either EMMPRIN small interfering RNA or a blocking antibody to EMMPRIN. EMMPRIN-containing purified membrane fraction from Chinese hamster ovary cells when added exogenously to NS2T2A cells induced a similar activation of the uPA/PAI-1 system. Additionally, overexpression of EMMPRIN in NS2T2A cells increased uPA levels in cocultured endothelial cells, showing a paracrine regulation loop involving a tumor-stroma interaction. EMMPRIN-expressing cells also exhibited enhanced invasive potential in vitro, and the use of amiloride (uPA inhibitor) and marimastat (MMP inhibitor) showed that the two proteolytic systems reduced alone and in combination the invasive potential mediated through EMMPRIN. These data show a novel regulatory pathway for uPA activity and suggest that EMMPRIN is involved in uPA dysregulation observed in cancer.


Assuntos
Basigina/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Ativador de Plasminogênio Tipo Uroquinase/biossíntese , Animais , Basigina/genética , Neoplasias da Mama/enzimologia , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Humanos , Metaloproteases/metabolismo , Camundongos , Camundongos Nus , Invasividade Neoplásica , Inibidor 1 de Ativador de Plasminogênio/biossíntese , Inibidor 1 de Ativador de Plasminogênio/genética , Receptores de Superfície Celular/biossíntese , Receptores de Superfície Celular/genética , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Transfecção , Regulação para Cima , Ativador de Plasminogênio Tipo Uroquinase/genética
18.
Cancers (Basel) ; 11(11)2019 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-31744072

RESUMO

Microenvironment plays a crucial role in tumor development and progression. Cancer cells modulate the tumor microenvironment, which also contribute to resistance to therapy. Identifying biomarkers involved in tumorigenesis and cancer progression represents a great challenge for cancer diagnosis and therapeutic strategy development. CD147 is a glycoprotein involved in the regulation of the tumor microenvironment and cancer progression by several mechanisms-in particular, by the control of glycolysis and also by its well-known ability to induce proteinases leading to matrix degradation, tumor cell invasion, metastasis and angiogenesis. Accumulating evidence has demonstrated the role of CD147 expression in tumor progression and prognosis, suggesting it as a relevant tumor biomarker for cancer diagnosis and prognosis, as well as validating its potential as a promising therapeutic target in cancers.

19.
Pigment Cell Melanoma Res ; 32(5): 697-707, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31271515

RESUMO

The discoidin domain receptor 1 (DDR1) is a member of the receptor tyrosine kinase family that signals in response to collagen and that has been implicated in cancer progression. In the present study, we investigated the expression and role of DDR1 in human melanoma progression. Immunohistochemical staining of human melanoma specimens (n = 52) shows high DDR1 expression in melanoma lesions that correlates with poor prognosis. DDR1 expression was associated with the clinical characteristics of Clark level and ulceration and with BRAF mutations. Downregulation of DDR1 by small interfering RNA (siRNA) in vitro inhibited melanoma cells malignant properties, migration, invasion, and survival in several human melanoma cell lines. A DDR tyrosine kinase inhibitor (DDR1-IN-1) significantly inhibited melanoma cell proliferation in vitro, and ex vivo and in tumor xenografts, underlining the promising potential of DDR1 inhibition in melanoma.


Assuntos
Proliferação de Células , Receptor com Domínio Discoidina 1/metabolismo , Melanoma/patologia , Pele/metabolismo , Animais , Apoptose , Estudos de Casos e Controles , Feminino , Humanos , Melanoma/genética , Melanoma/metabolismo , Camundongos , Camundongos Nus , Prognóstico , Taxa de Sobrevida , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Connect Tissue Res ; 49(3): 175-9, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18661337

RESUMO

Emmprin/CD147 is a cell membrane glycoprotein that belongs to the Ig superfamily and is involved in numerous physiological and pathological systems. Through its ability to interact with multiple partners within the cell surface and its potential to regulate the expression of several targets within the cell, emmprin may have different functions depending on the cell or tissue type. However, its role in tissue remodeling remains the most clearly demonstrated. Emmprin is able to induce, in the same cellular model, both the matrix metalloproteinases and the serine protease urokinase plasminogen activator, whose concerted action in the breakdown of the extracellular matrix (ECM) during various physiopathological situations has been reported. In addition, emmprin also promotes myofibroblasts' differentiation and tissue contraction through the induction of alpha smooth muscle actin, thus expanding on the mechanism by which emmprin remodels ECM.


Assuntos
Basigina/metabolismo , Diferenciação Celular , Matriz Extracelular/metabolismo , Metaloproteinases da Matriz/metabolismo , Neoplasias/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Animais , Basigina/química , Indução Enzimática , Humanos , Neoplasias/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA