Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Invest New Drugs ; 33(1): 75-85, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25404487

RESUMEN

Angiogenesis plays a pivotal role in tumorigenesis and also contributes to the pathogenesis of hematologic malignancies. A number of plant compounds have shown efficacy in preclinical and clinical studies and some of them possess an anti-angiogenic activity. Our present findings report anti-angiogenic activities of ethoxyfagaronine (etxfag), a synthetic derivative of fagaronine. Once determined the non-cytotoxic concentration of etxfag, we showed that the drug inhibits VEGF-induced angiogenesis in a Matrigel™ plug assay and suppresses ex vivo sprouting from VEGF-treated aortic rings. Each feature leading to neovascularization was then investigated and results demonstrate that etxfag prevents VEGF-induced migration and tube formation in human umbilical vein endothelial cells (HUVEC). Moreover, etxfag also suppresses VEGF-induced VEGFR-2 phosphorylation and inhibits FAK phosphorylation at Y-861 as well as focal adhesion complex turnover. Beside these effects, etxfag modifies MT1-MMP localization at the endothelial cell membrane. Finally, immunoprecipitation assay revealed that etxfag decreases VEGF binding to VEGFR-2. As we previously reported that etxfag is able to prevent leukemic cell invasiveness and adhesion to fibronectin, all together our data collectively support the anti-angiogenic activities of etxfag which could represent an additional approach to current anti-cancer therapies.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Benzofenantridinas/farmacología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Aorta/efectos de los fármacos , Aorta/fisiología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Ratones Endogámicos C57BL , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
2.
Biochem Biophys Res Commun ; 432(4): 549-52, 2013 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-23454380

RESUMEN

The NC1 domains from the different α(IV) collagen chains were found to exert anti-tumorigenic and/or anti-angiogenic activities. A limitation to the therapeutic use of these matrikines is the large amount of purified recombinant proteins, in the milligram range in mice that should be administered daily throughout the experimental procedures. In the current study, we developed a new therapeutic approach based on tumstatin (NC1α3(IV)) overexpression in vivo in a mouse melanoma model. Gene electrotransfer of naked plasmid DNA (pDNA) is particularly attractive because of its simplicity, its lack of immune responsiveness and its safety. The pDNA electrotransfer in muscle mediates a substantial gene expression that lasts several months. A pVAX1© vector containing the tumstatin cDNA was injected into the legs of C57BL/6 mice and submitted to electrotranfer. Sera were collected at different times and tumstatin was quantified by ELISA. Tumstatin secretion reached a plateau at day 21 with an expression level of 12 µg/mL. For testing the effects of tumstatin expression on tumor growth in vivo, B16F1 melanoma cells were subcutaneously injected in mice 7 days after empty pVAX1© (Mock) or pVAX1©-tumstatin electrotransfer. Tumstatin expression triggered a large decrease in tumor growth and an increase in mouse survival. This new therapeutic approach seems promising to inhibit tumor progression in vivo.


Asunto(s)
Autoantígenos/genética , Colágeno Tipo IV/genética , Electroquimioterapia , Melanoma Experimental/terapia , Músculo Esquelético , Animales , Técnicas de Transferencia de Gen , Ratones , Ratones Endogámicos C57BL , Plásmidos/administración & dosificación , Plásmidos/genética
3.
Int J Cancer ; 126(5): 1055-66, 2010 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-19551865

RESUMEN

We previously demonstrated that the CNYYSNS peptide derived from tumstatin inhibited in vivo tumor progression. The YSNS motif formed a beta-turn crucial for biological activity. More recently, a YSNSG cyclopeptide with a constrained beta-turn on the YSNS residues was designed. Intraperitoneal administration of the YSNSG cyclopeptide inhibited in vivo melanoma progression more efficiently than the native linear peptide. In the present article, we showed that the YSNSG cyclopeptide also triggered an inhibition of in vivo tumor neovascularization and we further analyzed its in vitroantiangiogenic effect. The YSNSG cyclopeptide did not alter endothelial cell proliferation but inhibited cell migration by 83% in an in vitro wound healing model. The inhibition was mediated by a decrease in active MT1-MMP at the migration front as well as a decrease in u-PA and u-PAR expression. The cyclopeptide also altered beta1-integrin distribution in endothelial cell lamellipodia, induced a strong decrease in the phosphorylated focal adhesion kinase (p125(FAK)), disorganized F-actin stress fibers and decreased the number of lamellipodia, resulting in a non migratory phenotype. Our results confirm the YSNSG cyclopeptide as a potent antitumor agent, through both the inhibition of invasive properties of tumor cells and the antiangiogenic activity.


Asunto(s)
Antineoplásicos/farmacología , Movimiento Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Neovascularización Patológica/tratamiento farmacológico , Péptidos Cíclicos/farmacología , Animales , Autoantígenos/química , Western Blotting , Proliferación Celular/efectos de los fármacos , Colágeno Tipo IV/química , Regulación hacia Abajo , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Metaloproteinasa 14 de la Matriz/efectos de los fármacos , Metaloproteinasa 14 de la Matriz/metabolismo , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Receptores del Activador de Plasminógeno Tipo Uroquinasa/efectos de los fármacos , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/efectos de los fármacos , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Mol Cancer Ther ; 6(2): 506-14, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17308049

RESUMEN

Type XIX collagen is a minor collagen that localizes to basement membrane zones, together with types IV, XV, and XVIII collagens. Because several NC1 COOH-terminal domains of other chains from basement membrane collagens were reported to exhibit antitumor activity, we decided to study the effects of the NC1(XIX) collagen domain on tumor progression using an experimental in vivo model of mouse melanoma. We observed a 70% reduction in tumor volume in NC1(XIX)-treated mice compared with the corresponding controls. Histologic examination of the tumors showed a strong decrease in tumor vascularization in treated mice. In vitro, NC1(XIX) inhibited the migrating capacity of tumor cells and their capacity to invade Matrigel. It also inhibited the capacity of human microvascular endothelial cells to form pseudotubes in Matrigel. This effect was accompanied by a strong inhibition of membrane type-1 matrix metalloproteinase (matrix metalloproteinase-14) and vascular endothelial growth factor expression. Collectively, our data indicate that the NC1 domain of type XIX collagen exerts antitumor activity. This effect is mediated by a strong inhibition of the invasive capacities of tumor cells and antiangiogenic effects. NC1(XIX) should now be considered as a new member of the basement membrane collagen-derived matrikine family with antitumor and antiangiogenic activity.


Asunto(s)
Colágeno/uso terapéutico , Melanoma/prevención & control , Neovascularización Patológica/tratamiento farmacológico , Animales , Apoptosis , Membrana Basal/metabolismo , Western Blotting , Adhesión Celular , Proliferación Celular , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Matriz Extracelular/metabolismo , Humanos , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Metaloproteinasa 14 de la Matriz/metabolismo , Melanoma/irrigación sanguínea , Melanoma/patología , Ratones , Ratones Endogámicos C57BL , Fragmentos de Péptidos/metabolismo , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/prevención & control , Factor A de Crecimiento Endotelial Vascular/metabolismo
5.
Chem Biol ; 13(12): 1307-15, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17185226

RESUMEN

We previously demonstrated that the NC1[alpha3(IV)185-191] CNYYSNS peptide inhibited in vivo tumor progression. The YSNS motif formed a beta turn crucial for biological activity. The aim of the present study was to design a YSNSG cyclopeptide with a constrained beta turn on the YSNS residues more stable than CNYYSNS. By nuclear magnetic resonance and molecular modeling, we demonstrated that the YSNSG cyclopeptide actually adopted the expected beta-turn conformation. It promoted melanoma cell adhesion and prevented their adhesion to the native peptide. It inhibited in vitro cell proliferation and migration through Matrigel by downregulating proteolytic cascades. Moreover, intraperitoneal administration of the YSNSG cyclopeptide inhibited melanoma progression far more efficiently than the native peptide. The increased solubility and stability at low pH of the YSNSG cyclopeptide suggest this peptide as a potent antitumor therapeutic agent.


Asunto(s)
Antineoplásicos/farmacología , Autoantígenos/química , Colágeno Tipo IV/química , Neoplasias Pulmonares/terapia , Péptidos Cíclicos/química , Animales , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Dicroismo Circular , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Espectroscopía de Resonancia Magnética , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Péptidos Cíclicos/farmacología , Conformación Proteica
6.
PLoS One ; 9(7): e103839, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25075518

RESUMEN

Tissue inhibitor of metalloproteinases-1 (TIMP-1) regulates the extracellular matrix turnover by inhibiting the proteolytic activity of matrix metalloproteinases (MMPs). TIMP-1 also displays MMP-independent activities that influence the behavior of various cell types including neuronal plasticity, but the underlying molecular mechanisms remain mostly unknown. The trans-membrane receptor low-density lipoprotein receptor-related protein-1 (LRP-1) consists of a large extracellular chain with distinct ligand-binding domains that interact with numerous ligands including TIMP-2 and TIMP-3 and a short transmembrane chain with intracellular motifs that allow endocytosis and confer signaling properties to LRP-1. We addressed TIMP-1 interaction with recombinant ligand-binding domains of LRP-1 expressed by CHO cells for endocytosis study, or linked onto sensor chips for surface plasmon resonance analysis. Primary cortical neurons bound and internalized endogenous TIMP-1 through a mechanism mediated by LRP-1. This resulted in inhibition of neurite outgrowth and increased growth cone volume. Using a mutated inactive TIMP-1 variant we showed that TIMP-1 effect on neurone morphology was independent of its MMP inhibitory activity. We conclude that TIMP-1 is a new ligand of LRP-1 and we highlight a new example of its MMP-independent, cytokine-like functions.


Asunto(s)
Receptores de LDL/fisiología , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Proteínas Supresoras de Tumor/fisiología , Animales , Células CHO , Cricetinae , Cricetulus , Citocinas/metabolismo , Endocitosis , Conos de Crecimiento/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Ratones , Neuritas/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas
7.
FEBS Lett ; 587(10): 1524-8, 2013 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-23583449

RESUMEN

TIMP-1, a well-known MMP inhibitor, displays other biological activities such as cell survival, proliferation and differentiation in hematopoietic cells. In this report, we investigated the role of the Src-related kinase Lyn in TIMP-1 induced UT-7 erythroleukemic cell survival. We showed that (i) tyrosine 507 of Lyn was dephosphorylated and Lyn kinase activity enhanced by TIMP-1, (ii) Lyn silencing suppressed TIMP-1 anti-apoptotic activity and (iii) Lyn was activated upstream the JAK2/PI 3-kinase/Akt pathway. Our data suggest a novel role for Lyn in erythroid cell survival.


Asunto(s)
Células Eritroides , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Familia-src Quinasas/fisiología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Activación Enzimática/efectos de los fármacos , Células Eritroides/citología , Células Eritroides/metabolismo , Células Eritroides/patología , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Humanos , Janus Quinasa 2/metabolismo , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/metabolismo , Leucemia Eritroblástica Aguda/patología , Modelos Biológicos , Proteína Oncogénica v-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , ARN Interferente Pequeño/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/fisiología , Inhibidor Tisular de Metaloproteinasa-1/fisiología , Células Tumorales Cultivadas , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
8.
Biol Aujourdhui ; 206(2): 111-23, 2012.
Artículo en Francés | MEDLINE | ID: mdl-22748049

RESUMEN

Tumor microenvironment is a complex system composed of a largely altered extracellular matrix (ECM) with different cell types that determine the angiogenic response. Upon the influence of hypoxia, tumor cells secrete cytokines that activate stromal cells to produce proteases and angiogenic factors. The proteases degrade the stromal ECM and participate in the release of various ECM fragments, named matrikines or matricryptins, capable to control tumor invasion and metastasis dissemination. We will focus on the matrikines derived from the NC1 domains of the different constitutive chains of basement membrane-associated collagens and mainly collagen IV. The putative targets of the matrikine action are the proliferation and invasive properties of tumor or inflammatory cells, and the angiogenic and lymphangiogenic responses. For example, canstatin, tumstatin and tetrastatin, respectively derived from the NC1 domains of α2, α3 and α4 chains of collagen IV, inhibit in vivo tumor growth in various experimental cancer models. Their anti-cancer activity comprises an anti-proliferative effect on tumor cells and on endothelial cells by induction of cell apoptosis or cell cycle blockade and the induction of a loss of their migratory phenotype. Matrikines constitute a new family of potent anticancer agents that could be used under various therapeutic strategies: i) induction of their overexpression by cancer cells or by the host cells, ii) use of recombinant proteins or synthetic peptides or structural analogues designed from the structure of the active sequences. These matrikines could be used in combination with conventional chemotherapy or radiotherapy to limit tumor progression.


Asunto(s)
Antineoplásicos/uso terapéutico , Citocinas/fisiología , Citocinas/uso terapéutico , Matriz Extracelular/metabolismo , Neoplasias/tratamiento farmacológico , Animales , Membrana Basal/efectos de los fármacos , Membrana Basal/metabolismo , Membrana Basal/fisiología , Ensayos Clínicos como Asunto , Colágeno/química , Colágeno/metabolismo , Colágeno/fisiología , Citocinas/química , Citocinas/metabolismo , Humanos , Modelos Biológicos , Neoplasias/metabolismo
9.
PLoS One ; 7(4): e29587, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22539938

RESUMEN

BACKGROUND: NC1 domains from α1, α2, α3 and α6(IV) collagen chains were shown to exert anti-tumor or anti-angiogenic activities, whereas the NC1 domain of the α4(IV) chain did not show such activities so far. METHODOLOGY/PRINCIPAL FINDINGS: We demonstrate in the present paper that the NC1 α4(IV) domain exerts a potent anti-tumor activity both in vitro and in an experimental human melanoma model in vivo. The overexpression of NC1 α4(IV) in human UACC-903 melanoma cells strongly inhibited their in vitro proliferative (-38%) and invasive (-52%) properties. MT1-MMP activation was largely decreased and its cellular distribution was modified, resulting in a loss of expression at the migration front associated with a loss of migratory phenotype. In an in vivo xenograft model in athymic nude mice, the subcutaneous injection of NC1 α4(IV)-overexpressing melanoma cells induced significantly smaller tumors (-80% tumor volume) than the Mock cells, due to a strong inhibition of tumor growth. Exogenously added recombinant human NC1 α4(IV) reproduced the inhibitory effects of NC1 α4(IV) overexpression in UACC-903 cells but not in dermal fibroblasts. An anti-αvß3 integrin blocking antibody inhibited cell adhesion on recombinant human NC1 α4(IV) substratum. The involvement of αvß3 integrin in mediating NC1 α4(IV) effect was confirmed by surface plasmon resonance (SPR) binding assays showing that recombinant human NC1 α4(IV) binds to αvß3 integrin (K(D) = 148 ± 9.54 nM). CONCLUSION/SIGNIFICANCE: Collectively, our results demonstrate that the NC1 α4(IV) domain, named tetrastatin, is a new endogenous anti-tumor matrikine.


Asunto(s)
Antineoplásicos/uso terapéutico , Colágeno Tipo IV/química , Melanoma Experimental/tratamiento farmacológico , Animales , Anticuerpos/inmunología , Antineoplásicos/farmacología , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Humanos , Integrina alfaVbeta3/inmunología , Integrina alfaVbeta3/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Desnudos , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Resonancia por Plasmón de Superficie , Trasplante Heterólogo
10.
Eur J Med Genet ; 51(6): 598-607, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18822396

RESUMEN

Interstitial 2q36 deletion is a rare event. We report on a patient with a de novo del(2)(q36.2q36.3) interstitial deletion of the long arm of chromosome 2 diagnosed by classical banding. The phenotype comprised facial dysmorphism, enlarged kidneys with multiple renal cysts, abnormal minora labia, asymmetric lower limbs with dysplastic patella, and severe mental retardation. By physical mapping, using array-comparative genomic hybridisation (CGH) confirmed by Fluorescent In Situ Hybridisation (FISH), the breakpoints of the deletion were mapped and the size of the deletions was measured: 5.61+/-0.19Mb. A skin biopsy was analysed using electronic microscopy showing an alteration of the structure and organisation of the dermal and peri-neuronal basement membrane. The relation between the phenotype and the deletion of both COL4A4 and COL4A3 genes, located in 2q36.3 loci, as well as the disruption of TRIP12 were discussed.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 2 , Quistes/genética , Discapacidad Intelectual/genética , Enfermedades Renales/genética , Preescolar , Humanos , Hibridación Fluorescente in Situ , Cariotipificación , Hibridación de Ácido Nucleico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA