Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Nutrients ; 14(5)2022 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-35268073

RESUMEN

BACKGROUND: Triple-negative breast cancer (TNBC) cells secretome induces a pro-inflammatory microenvironment within the adipose tissue, which hosts both mature adipocytes and adipose-derived mesenchymal stem/stromal cells (ADMSC). The subsequent acquisition of a cancer-associated adipocyte (CAA)-like phenotype is, however, unknown in ADMSC. While epidemiological studies suggest that consuming a polyphenol-rich diet reduces the incidence of some obesity-related cancers, the chemopreventive impact of green tea-derived epigallocatechin-3-gallate (EGCG) against the cues that trigger the CAA phenotype remain undocumented in ADMSC. METHODS: Human ADMSC were exposed to human TNBC-derived MDA-MB-231 conditioned media (TNBC cells secretome) supplemented or not with EGCG. Differential gene expression was assessed through RNA-Seq analysis and confirmed by RT-qPCR. Protein expression levels and the activation status of signal transduction pathways mediators were determined by Western blotting. ADMSC chemotaxis was assessed by a real-time cell migration assay. RESULTS: The TNBC cells secretome induced in ADMSC the expression of the CAA cytokines CCL2, CCL5, IL-1ß, and IL-6, and of immunomodulators COX2, HIF-1α, VEGFα, and PD-L1. The epithelial-to-mesenchymal biomarker Snail was found to control the CAA phenotype. EGCG inhibited the induction of CAA genes and the activation status of Smad2 and NF-κB. The induced chemotactic response was also inhibited by EGCG. CONCLUSION: The induction of an inflammatory and CAA-like phenotype in ADMSC can be triggered by the TNBC cells secretome, while still efficiently prevented by diet-derived polyphenols.


Asunto(s)
Células Madre Mesenquimatosas , Neoplasias de la Mama Triple Negativas , Adipocitos , Catequina/análogos & derivados , Humanos , Células Madre Mesenquimatosas/metabolismo , Fenotipo , Secretoma , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/prevención & control , Microambiente Tumoral
2.
Clin Nutr ; 40(6): 3842-3851, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34130031

RESUMEN

BACKGROUND AND AIMS: Anastomotic leak (AL) is a major complication in colorectal surgery. Recent evidence suggests that the gut microbiota may affect healing and may cause or prevent AL. Butyrate is a beneficial short-chain fatty acid (SCFA) that is produced as a result of bacterial fermentation of dietary oligosaccharides and has been described as beneficial in the maintenance of colonic health. To assess the impact of oligosaccharides on colonic anastomotic healing in mice, we propose to modulate the microbiota with oligosaccharides to increase butyrate production via enhancement of butyrate-producing bacteria and, consequently, improve anastomotic healing in mice. METHODS: Animal experiments were conducted in mice that were subjected to diets supplemented with inulin, galacto-oligosaccharides (GOS) or cellulose, as a control, for two weeks before undergoing a surgical colonic anastomosis. Macroscopic and histological assessment of the anastomosis was performed. Extent of epithelial proliferation was assessed by Ki-67 immunohistochemistry. Gelatin zymography was used to evaluate the extent of matrix metalloproteinase (MMP) hydrolytic activity. RESULTS: Inulin and GOS diets were associated with increased butyrate production and better anastomotic healing. Histological analysis revealed an enhanced mucosal continuity, and this was associated with an increased re-epithelialization of the wound as determined by increased epithelial proliferation. Collagen concentration in peri-anastomotic tissue was higher with inulin and GOS diets and MMP activity, a marker of collagen degradation, was lower with both oligosaccharides. Inulin and GOS diets were further associated with lower bacterial translocation. CONCLUSIONS: Dietary supplementation with inulin and GOS may improve anastomotic healing and reinforce the gut barrier in mice.


Asunto(s)
Fuga Anastomótica/prevención & control , Enfermedades del Colon/cirugía , Ácidos Grasos Volátiles/administración & dosificación , Inulina/administración & dosificación , Animales , Suplementos Dietéticos , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos C57BL , Periodo Perioperatorio , Complicaciones Posoperatorias/prevención & control , Resultado del Tratamiento , Cicatrización de Heridas
3.
Molecules ; 26(6)2021 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-33801973

RESUMEN

Obese subjects have an increased risk of developing triple-negative breast cancer (TNBC), in part associated with the chronic low-grade inflammation state. On the other hand, epidemiological data indicates that increased consumption of polyphenol-rich fruits and vegetables plays a key role in reducing incidence of some cancer types. Here, we tested whether green tea-derived epigallocatechin-3-gallate (EGCG) could alter adipose-derived mesenchymal stem cell differentiation into adipocytes, and how this impacts the secretome profile and paracrine regulation of the TNBC invasive phenotype. Here, cell differentiation was performed and conditioned media (CM) from preadipocytes and mature adipocytes harvested. Human TNBC-derived MDA-MB-231 real-time cell migration was performed using the exCELLigence system. Differential gene arrays and RT-qPCR were used to assess gene expression levels. Western blotting was used to assess protein expression and phosphorylation status levels. In vitro vasculogenic mimicry (VM) was assessed with Matrigel. EGCG was found to inhibit the induction of key adipogenic biomarkers, including lipoprotein lipase, adiponectin, leptin, fatty acid synthase, and fatty acid binding protein 4. Increased TNBC-derived MDA-MB-231 cell chemotaxis and vasculogenic mimicry were observed in response to mature adipocytes secretome, and this was correlated with increased STAT3 phosphorylation status. This invasive phenotype was prevented by EGCG, the JAK/STAT inhibitors Tofacitinib and AG490, as well as upon STAT3 gene silencing. In conclusion, dietary catechin-mediated interventions could, in part through the inhibition of adipogenesis and modulation of adipocytes secretome profile, prevent the onset of an obesogenic environment that favors TNBC development.


Asunto(s)
Catequina/análogos & derivados , Células Madre Mesenquimatosas/efectos de los fármacos , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Adipogénesis/efectos de los fármacos , Adiponectina/metabolismo , Animales , Catequina/metabolismo , Catequina/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Medios de Cultivo Condicionados , Femenino , Humanos , Leptina/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Comunicación Paracrina/efectos de los fármacos , Factor de Transcripción STAT3/metabolismo , Té/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo
4.
J Nutr Biochem ; 87: 108518, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33017609

RESUMEN

The galloyl moiety is a specific structural feature which dictates, in part, the chemopreventive properties of diet-derived catechins. In ovarian cancer cells, galloylated catechins were recently demonstrated to target the transforming growth factor (TGF)-ß-mediated control of the epithelial-mesenchymal transition process. The specific impact of the galloyl moiety on such signaling, however, remains poorly understood. Here, we questioned whether the sole galloyl moiety interacted with TGF-ß-receptors to alter signal transduction and chemotactic migratory response in an ES-2 serous carcinoma-derived ovarian cancer cell model. In line with the LogP and LogS values of the tested molecules, we found that TGF-ß-induced Smad-3 phosphorylation and cell migration were optimally inhibited, provided that the lateral aliphatic chain of the galloyl moiety reached 8-10 carbons. Functional inhibition of the TGF-ß receptor (TGF-ßR1) kinase activity was supported by surface plasmon resonance assays showing direct physical interaction between TGF-ßR1 and the galloyl moiety. In silico molecular docking analysis predicted a model where galloylated catechins may bind TGF-ßR1 within its adenosine triphosphate binding cleft in a site analogous to that of Galunisertib, a selective adenosine triphosphate-mimetic competitive inhibitor of TGF-ßR1. In conclusion, our data suggest that the galloyl moiety of the diet-derived catechins provides specificity of action to galloylated catechins by positioning them within the kinase domain of the TGF-ßR1 in order to antagonize TGF-ß-mediated signaling that is required for ovarian cancer cell invasion and metastasis.


Asunto(s)
Catequina/farmacología , Ácido Gálico/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Receptor Tipo I de Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Té/química , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/aislamiento & purificación , Antineoplásicos Fitogénicos/farmacología , Catequina/química , Catequina/aislamiento & purificación , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Femenino , Ácido Gálico/análogos & derivados , Ácido Gálico/aislamiento & purificación , Humanos , Neoplasias Ováricas/metabolismo , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/aislamiento & purificación , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo
5.
Artículo en Inglés | MEDLINE | ID: mdl-31827554

RESUMEN

Emerging drugs aim at targeting the genomic integrity and replication machinery in ovarian cancer. While the antiproliferative activity of Xanthium strumarium L. extract (XFC), a traditional herbal medicine, is believed to alter the mitotic apparatus of Chinese hamster ovary epithelial cells, its capacity to target and overcome the chemoresistance phenotype in ovarian cancer is unknown. Among the cancer cell lines tested, we found that the best proliferation inhibitory effect for XFC was against ovarian cancer cells and ranged from 30 to 35 µg/mL. XFC efficiently targeted both the cytotoxic drug chemoresistance phenotype of SKOV-3 cells and of the chemosensitive ES-2 cells. Early apoptosis and late apoptosis were effectively induced by XFC extract in ES-2 cells, whereas late apoptosis and necrosis events were triggered in SKOV-3 cells. Cell cycling regulation was trapped by XFC extract in the G2/M phase in both the ES-2 and SKOV-3 cell models. This effect was, in part, attributable to increased dose-dependent tubulin polymerization, which was increased in SKOV-3 cells. Whereas XFC extract triggered poly (ADP-Ribose) polymerase (PARP) cleavage in both ES-2 and SKOV-3 cells, it only lowered Nrf2 in ES-2 cells and phosphorylated Akt levels in SKOV-3 cells. Interestingly, cell cycling regulators Cdk4, Cyclin D3, and p27 were all decreased in SKOV-3 cells. XFC extracts were effective in inhibiting in vitro migration in both ovarian cancer cell models. Our data support the potential anticancer targeting of chemoresistant human ovarian cancer cells phenotype by XFC extract.

6.
Biophys Chem ; 234: 34-41, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29407769

RESUMEN

Membrane type-1 matrix metalloproteinase (MT1-MMP) is a transmembrane MMP which triggers intracellular signaling and regulates extracellular matrix proteolysis, two functions that are critical for tumor-associated angiogenesis and inflammation. While green tea catechins, particularly epigallocatechin gallate (EGCG), are considered very effective in preventing MT1-MMP-mediated functions, lack of structure-function studies and evidence regarding their direct interaction with MT1-MMP-mediated biological activities remain. Here, we assessed the impact in both cellular and biophysical assays of four ungallated catechins along with their gallated counterparts on MT1-MMP-mediated functions and molecular binding partners. Concanavalin-A (ConA) was used to trigger MT1-MMP-mediated proMMP-2 activation, expression of MT1-MMP and of endoplasmic reticulum stress biomarker GRP78 in U87 glioblastoma cells. We found that ConA-mediated MT1-MMP induction was inhibited by EGCG and catechin gallate (CG), that GRP78 induction was inhibited by EGCG, CG, and gallocatechin gallate (GCG), whereas proMMP-2 activation was inhibited by EGCG and GCG. Surface plasmon resonance was used to assess direct interaction between catechins and MT1-MMP interactors. We found that gallated catechins interacted better than their ungallated analogs with MT1-MMP as well as with MT1-MMP binding partners MMP-2, TIMP-2, MTCBP-1 and LRP1-clusterIV. Overall, current structure-function evidence supports a role for the galloyl moiety in both direct and indirect interactions of green tea catechins with MT1-MMP-mediated oncogenic processes.


Asunto(s)
Catequina/análogos & derivados , Metaloproteinasa 14 de la Matriz/metabolismo , Té/química , Carcinogénesis/efectos de los fármacos , Catequina/metabolismo , Catequina/farmacología , Línea Celular Tumoral , Concanavalina A/farmacología , Chaperón BiP del Retículo Endoplásmico , Precursores Enzimáticos/antagonistas & inhibidores , Gelatinasas/antagonistas & inhibidores , Glioblastoma/patología , Proteínas de Choque Térmico/antagonistas & inhibidores , Humanos , Metaloproteinasa 14 de la Matriz/farmacología , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Unión Proteica , Relación Estructura-Actividad
7.
Leuk Res ; 31(9): 1277-84, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17081606

RESUMEN

Matrix metalloproteinase (MMP)-9 expression is linked with myeloid cell differentiation, as well as inflammation and angiogenesis processes related to cancer progression. MMP-9 secretion and macrophage-like HL-60 myeloid leukemia cells differentiation were triggered by the tumor-promoting agent PMA. The chemopreventive effects of green tea catechins epigallocatechin-gallate, catechin-gallate, and epicatechin-gallate, but not those catechins that lack a 3'-galloyl group, inhibited in a time- and dose-dependent manner MMP-9 secretion. The gene and protein expression of MMP-9 and of the mRNA stabilizing factor HuR were also inhibited, while that of the 67 kDa laminin receptor remained unaffected. Specific catechins may help optimize current chemotherapeutic treatment protocols for leukemia.


Asunto(s)
Anticarcinógenos/farmacología , Catequina/análogos & derivados , Diferenciación Celular/efectos de los fármacos , Macrófagos/efectos de los fármacos , Inhibidores de la Metaloproteinasa de la Matriz , Proteínas de Unión al ARN/antagonistas & inhibidores , Té/química , Antígenos de Superficie/metabolismo , Carcinógenos/farmacología , Catequina/farmacología , Adhesión Celular/efectos de los fármacos , Proteínas ELAV , Proteína 1 Similar a ELAV , Células HL-60/efectos de los fármacos , Células HL-60/metabolismo , Humanos , Immunoblotting , Metaloproteinasa 9 de la Matriz/metabolismo , Proteínas de Unión al ARN/metabolismo , Acetato de Tetradecanoilforbol/farmacología
8.
J Neurooncol ; 80(2): 111-21, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16715350

RESUMEN

The microvasculature of brain tumors has been proposed as the primary target for ionizing radiation (IR)-induced apoptosis. However, the contribution of low dose IR-induced non-apoptotic cell death pathways has not been investigated. This study aimed to characterize the effect of IR on human brain microvascular endothelial cells (HBMEC) and to assess the combined effect of epigallocatechin-3-gallate (EGCg), a green tea-derived anti-angiogenic molecule. HBMEC were treated with EGCg, irradiated with a sublethal (< or =10 Gy) single dose. Cell survival was assessed 48 h later by nuclear cell counting and Trypan blue exclusion methods. Cell cycle distribution and DNA fragmentation were evaluated by flow cytometry (FC), cell death was assessed by fluorimetric caspase-3 activity, FC and immunoblotting for pro-apoptotic proteins. While low IR doses alone reduced cell survival by 30%, IR treatment was found more effective in EGCg pretreated-cells reaching 70% cell death. Analysis of cell cycle revealed that IR-induced cell accumulation in G2-phase. Expression of cyclin-dependent kinase inhibitors p21(CIP/Waf1) and p27(Kip) were increased by EGCg and IR. Although random DNA fragmentation increased by approximately 40% following combined EGCg/IR treatments, the synergistic reduction of cell survival was not related to increased pro-apoptotic caspase-3, caspase-9 and cytochrome C proteins. Cell necrosis increased 5-fold following combined EGCg/IR treatments while no changes in early or late apoptosis were observed. Our results suggest that the synergistic effects of combined EGCg/IR treatments may be related to necrosis, a non-apoptotic cell death pathway. Strategies sensitizing brain tumor-derived EC to IR may enhance the efficacy of radiotherapy and EGCg may represent such a potential agent.


Asunto(s)
Encéfalo/citología , Camellia/química , Catequina/análogos & derivados , Células Endoteliales/efectos de los fármacos , Células Endoteliales/efectos de la radiación , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Encéfalo/efectos de los fármacos , Caspasa 3/metabolismo , Catequina/farmacología , Ciclo Celular/efectos de los fármacos , Ciclo Celular/efectos de la radiación , Muerte Celular/efectos de los fármacos , Muerte Celular/efectos de la radiación , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Fragmentación del ADN/efectos de los fármacos , Fragmentación del ADN/efectos de la radiación , Citometría de Flujo , Humanos , Immunoblotting , Necrosis , Fotones
9.
Brain Res ; 1071(1): 1-9, 2006 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-16412397

RESUMEN

INTRODUCTION: Glioblastoma multiforme's (GBM) aggressiveness is potentiated in radioresistant tumor cells. The combination of radiotherapy and chemotherapy has been envisioned as a therapeutic approach for GBM. The goal of this study is to determine if epigallocatechin-3-gallate (EGCg), a green tea-derived anti-cancer molecule, can modulate GBMs' response to ionizing radiation (IR) and whether this involves mediators of intracellular signaling and inhibitors of apoptosis proteins. MATERIAL AND METHODS: U-87 human GBM cells were cultured and transfected with cDNAs encoding for Survivin, RhoA or Caveolin-1. Mock and transfected cells were irradiated at sublethal single doses. Cell proliferation was analyzed by nuclear cell counting. Apoptosis was detected using a fluorometric caspase-3 assay. Analysis of protein expression was accomplished by Western immunoblotting. RESULTS: IR (10 Gy) reduced control U-87 cell proliferation by 40% through a caspase-independent mechanism. The overexpression of Survivin induced a cytoprotective effect against IR, while the overexpression of RhoA conferred a cytosensitizing effect upon IR. Control U-87 cells pretreated with EGCg exhibited a dose-dependent decrease in their proliferation rate. The growth inhibitory effect of EGCg was not antagonized by overexpressed Survivin. However, Survivin -transfected cells pretreated with EGCg became sensitive to IR, and their RhoA expression was downregulated. A potential therapeutic effect of EGCg targeting the prosurvival intracellular pathways of cancer cells is suggested to act synergistically with IR. CONCLUSION: The radioresistance of GBM is possibly mediated by a mechanism dependent on Survivin in conjunction with RhoA. The combination of natural anti-cancerous molecules such as EGCg with radiotherapy could improve the efficacy of IR treatments.


Asunto(s)
Anticarcinógenos/uso terapéutico , Catequina/análogos & derivados , Glioblastoma/tratamiento farmacológico , Glioblastoma/radioterapia , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Western Blotting/métodos , Caspasa 3 , Caspasas/metabolismo , Catequina/uso terapéutico , Caveolina 1/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de la radiación , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Técnica del Anticuerpo Fluorescente/métodos , Expresión Génica/efectos de los fármacos , Expresión Génica/efectos de la radiación , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Humanos , Proteínas Inhibidoras de la Apoptosis , Fenotipo , Probabilidad , Survivin , Transfección/métodos
10.
Cancer Biol Ther ; 2(6): 642-9, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14688468

RESUMEN

Radiation therapy is a widely-used option for the treatment of a variety of solid tumors. Although effective, ionizing radiation (IR) may give rise to various side effects, including secondary tumors. In agreement with this, recent reports have demonstrated increased invasive potential in different tumor-derived cell lines following radiation treatment. Many of the molecular effects of IR specifically on the endothelial cells involved in tumor neo-vascularization remain unknown. In this study, we found that low sublethal single doses of IR applied to human umbilical vein endothelial cells stimulated cell migration and in vitro tubulogenesis. This correlated with an increase in membrane type-1 matrix metalloproteinase (MT1-MMP) protein expression, a crucial enzyme that promotes endothelial cell migration and tube formation, and of caveolin-1, a protein that regulates tube formation. Cell adhesion was also promoted by IR, reflected in increased gene expression levels of cell surface beta(3) integrin. Pretreatment of the cells with epigallocatechin-3-gallate (EGCg), a green tea catechin that possesses anti-angiogenic properties, prevented most of the IR-induced cellular and molecular events. These observations suggest that current protocols involving radiation therapy for the treatment of cancer can paradoxically promote angiogenesis, but can be improved by combination with anti-angiogenic molecules such as EGCg to target those tumor-derived endothelial cells that escaped IR-induced apoptosis.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Catequina/análogos & derivados , Catequina/farmacología , Células Endoteliales/efectos de los fármacos , Morfogénesis/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Té/química , Western Blotting , Caspasas/análisis , Caspasas/metabolismo , Caveolina 1 , Caveolinas/efectos de los fármacos , Caveolinas/efectos de la radiación , Adhesión Celular/efectos de los fármacos , Adhesión Celular/efectos de la radiación , División Celular/efectos de los fármacos , División Celular/efectos de la radiación , Línea Celular , Movimiento Celular/efectos de los fármacos , Movimiento Celular/efectos de la radiación , Colágeno/metabolismo , Relación Dosis-Respuesta en la Radiación , Combinación de Medicamentos , Células Endoteliales/efectos de la radiación , Endotelio Vascular/citología , Flavonoides/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Integrina beta3/efectos de los fármacos , Integrina beta3/efectos de la radiación , Laminina/metabolismo , Metaloproteinasas de la Matriz Asociadas a la Membrana , Metaloendopeptidasas/efectos de los fármacos , Metaloendopeptidasas/efectos de la radiación , Modelos Biológicos , Morfogénesis/efectos de la radiación , Neovascularización Fisiológica/efectos de la radiación , Fenoles/farmacología , Polifenoles , Proteoglicanos/metabolismo , Radiación Ionizante , Factores de Tiempo , Transglutaminasas/efectos de los fármacos , Transglutaminasas/efectos de la radiación , Venas Umbilicales/citología , Regulación hacia Arriba/efectos de la radiación
11.
J Cell Biochem ; 90(4): 745-55, 2003 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-14587030

RESUMEN

Epigallocatechin-3-gallate (EGCG), the major green tea polyphenol, can reach the brain following oral intake and could thus act as an anti-tumoral agent targeting several key steps of brain cancer cells invasive activity. Because integrin-mediated extracellular matrix recognition is crucial during the cell adhesion processes involved in carcinogenesis, we have investigated the effects of EGCG on different cellular integrins of the pediatric brain tumor-derived medulloblastoma cell line DAOY. Using flow cytometry, we report the levels of expression of several cell surface integrins in DAOY. These include high expression of alpha2, alpha3, and beta1 integrins, as well as alphav and beta3 integrins. Moreover, we provide evidence that EGCG can antagonize DAOY cell migration specifically on collagen by increasing cell adhesive ability through specific gene and protein upregulation of the beta1 integrin subunit. Our results suggest that this naturally occurring green tea polyphenol may thus be used as a nutraceutical therapeutic agent in targeting the invasive character of medulloblastomas.


Asunto(s)
Catequina/análogos & derivados , Catequina/farmacología , Movimiento Celular/efectos de los fármacos , Meduloblastoma/patología , Invasividad Neoplásica/prevención & control , Té/química , Apoptosis/efectos de los fármacos , Caspasa 3 , Caspasas/metabolismo , Catequina/uso terapéutico , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Colágeno/metabolismo , Citometría de Flujo , Humanos , Integrina beta1/genética , Integrina beta1/metabolismo , Meduloblastoma/tratamiento farmacológico , Regulación hacia Arriba/efectos de los fármacos
12.
Biochim Biophys Acta ; 1542(1-3): 209-20, 2002 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-11853893

RESUMEN

We have recently shown that green tea polyphenols, and especially (-)-epigallocatechin 3-gallate (EGCg), acted as potent inhibitors of matrix metalloproteinase activities as well as of proMMP-2 activation (M. Demeule, M. Brossard, M. Page, D. Gingras, R. Beliveau, Biochim. Biophys. Acta 1478 (2000)). In the present work, we sought to examine the involvement of MT1-MMP in the EGCg-induced inhibition of proMMP-2 activation. The incubation of U-87 glioblastoma cells in the presence of concanavalin A or cytochalasin D, two potent activators of MT1-MMP, resulted in proMMP-2 activation that was correlated with the cell surface proteolytic processing of MT1-MMP to its inactive 43 kDa form. Addition of EGCg strongly inhibited the MT1-MMP-dependent proMMP-2 activation. The inhibitory effect of EGCg on MT1-MMP was also demonstrated by the down-regulation of MT1-MMP transcript levels and by the inhibition of MT1-MMP-driven cell migration of transfected COS-7 cells. These observations suggest that this catechin may act at both the MT1-MMP gene and protein expression levels. In addition, treatment of cells with non-cytotoxic doses of EGCg significantly reduced the amount of secreted proMMP-2, and led to a concomitant increase in intracellular levels of that protein. This effect was similar to that observed using well-characterized secretion inhibitors such as brefeldin A and manumycin, suggesting that EGCg could also potentially act on intracellular secretory pathways. Taken together, these results indicate that EGCg targets multiple MMP-mediated cellular events in cancer cells and provides a new mechanism for the anticancer properties of that molecule.


Asunto(s)
Camellia sinensis , Catequina/farmacología , Flavonoides , Inhibidores de la Metaloproteinasa de la Matriz , Metaloendopeptidasas/antagonistas & inhibidores , Fenoles/farmacología , Polímeros/farmacología , Animales , Antineoplásicos/farmacología , Células COS , Catequina/análogos & derivados , Catequina/aislamiento & purificación , Movimiento Celular/efectos de los fármacos , Medios de Cultivo Condicionados , Inhibidores Enzimáticos/farmacología , Precursores Enzimáticos/antagonistas & inhibidores , Gelatina/metabolismo , Gelatinasas/antagonistas & inhibidores , Glioblastoma , Humanos , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasas de la Matriz Asociadas a la Membrana , Metaloendopeptidasas/genética , Fenoles/aislamiento & purificación , Polímeros/aislamiento & purificación , Transcripción Genética/efectos de los fármacos , Transfección , Células Tumorales Cultivadas/efectos de los fármacos
13.
Curr Med Chem Anticancer Agents ; 2(4): 441-63, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12678730

RESUMEN

The concept of cancer prevention by use of naturally occuring substances that could be included in the diet is under investigation as a practical approach towards reducing cancer incidence, and therefore the mortality and morbidity associated with this disease. Tea, which is the most popularly consumed beverage aside from water, has been particularly associated with decreased risk of various proliferative diseases such as cancer and atherosclerosis in humans. Various studies have provided evidence that polyphenols are the strongest biologically active agents in green tea. Green tea polyphenols (GTPs) mainly consist of catechins (3-flavanols), of which (-)-epigallocatechin gallate is the most abundant and the most extensively studied. Recent observations have raised the possibility that green tea catechins, in addition to their antioxidative properties, also affect the molecular mechanisms involved in angiogenesis, extracellular matrix degradation, regulation of cell death and multidrug resistance. This article will review the effects and the biological activities of green tea catechins in relation to these mechanisms, each of which plays a crucial role in the development of cancer in humans. The extraction of polyphenols from green tea, as well as their bioavailability, are also discussed since these two important parameters affect blood and tissue levels of the GTPs and consequently their biological activities. In addition, general perspectives on the application of dietary GTPs as novel antiangiogenic and antitumor compounds are also presented.


Asunto(s)
Inhibidores de la Angiogénesis/química , Antineoplásicos/química , Catequina/farmacología , Flavonoides , Té/química , Inhibidores de la Angiogénesis/farmacocinética , Inhibidores de la Angiogénesis/farmacología , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Catequina/química , Catequina/farmacocinética , Humanos , Fenoles/química , Fenoles/farmacocinética , Fenoles/farmacología , Polímeros/química , Polímeros/farmacocinética , Polímeros/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA