Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Cell Biol ; 105(5): 2409-15, 1987 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-3680388

RESUMEN

Thrombospondin induces the migration of human melanoma and carcinoma cells. Using a modified Boyden chamber assay, tumor cells migrated to a gradient of soluble thrombospondin (chemotaxis). Checkerboard analysis indicated that directional migration was induced 27-fold greater than stimulation of random motility. Tumor cells also migrated in a dose-dependent manner to a gradient of substratum-bound thrombospondin (haptotaxis). A series of human melanoma and carcinoma cells were compared for their relative motility stimulation by thrombospondin haptotaxis vs. chemotaxis. Some cell lines exhibited a stronger haptotactic response compared to their chemotactic response while other lines exhibited little or no migration response to thrombospondin. Human A2058 melanoma cells which exhibit a strong haptotactic and chemotactic response to thrombospondin were used to study the structural domains of thrombospondin required for the response. Monoclonal antibody C6.7, which binds to the COOH-terminal region of thrombospondin, inhibited haptotaxis in a dose-dependent optimal manner. C6.7 had no significant effect on thrombospondin chemotaxis. In contrast, monoclonal antibody A2.5, heparin, and fucoidan, which bind to the NH2-terminal heparin-binding domain of thrombospondin, inhibited thrombospondin chemotaxis but not haptotaxis. Monoclonal antibody A6.1 directed against the internal core region of thrombospondin had no significant effect on haptotaxis or chemotaxis. Synthetic peptides GRGDS (50 micrograms/ml), but not GRGES, blocked tumor cell haptotaxis on fibronectin, but had minimal effect on thrombospondin or laminin haptotaxis. The 140-kD fragment of thrombospondin lacking the heparin-binding amino-terminal region retained the property to fully mediate haptotaxis but not chemotaxis. When the COOH region of the 140-kD fragment, containing the C6.7-binding site, was cleaved off, the resulting 120-kD fragment (which retains the RGDA sequence) failed to induce haptotaxis. Separate structural domains of thrombospondin are therefore required for tumor cell haptotaxis vs. chemotaxis. This may have implications during hematogenous cancer metastases formation.


Asunto(s)
Plaquetas/fisiología , Quimiotaxis , Glicoproteínas/fisiología , Células Tumorales Cultivadas/fisiología , Neoplasias de la Mama , Carcinoma , Línea Celular , Movimiento Celular/efectos de los fármacos , Femenino , Humanos , Laminina/farmacología , Trombospondinas , Células Tumorales Cultivadas/efectos de los fármacos
2.
J Cell Biol ; 111(2): 765-72, 1990 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-1696271

RESUMEN

Components of the extracellular matrix have been shown to modulate the interaction of endothelial cells with their microenvironment. Here we report that thrombospondin (TSP), an extracellular matrix component, induces adhesion and spreading of murine lung capillary (LE-II) and bovine aortic (BAEC) endothelial cells. This TSP-induced spreading was inhibited by heparin and fucoidan, known to bind the amino-terminal globular domain of the molecule. In addition, endothelial cells were induced to migrate by a gradient of soluble TSP (chemotaxis). The chemotactic response was inhibited by heparin and fucoidan, as well as by the mAb A2.5, which also binds to the amino-terminal domain. These data are in agreement with our previous observation that the TSP aminoterminal heparin binding region is responsible for the induction of tumor cell spreading and chemotactic motility. The inhibition of chemotaxis and spreading by antibodies against the beta 3 but not the beta 1 chain of the integrin receptor points to a role for the integrins in the interaction of endothelial cells with TSP. We also found that TSP modulates endothelial cell growth. When added to quiescent LE-II cells, it inhibited the mitogenic effects of serum and the angiogenic factor bFGF, in a dose-dependent manner. The inhibition of DNA synthesis detected in the mitogenic assay resulted in a true inhibition of BAEC and LE-II cell growth, as assessed by proliferation assay. This work indicates that TSP affects endothelial cell adhesion, spreading, motility and growth. TSP, therefore, has the potential to modulate the angiogenic process.


Asunto(s)
Plaquetas/fisiología , Endotelio Vascular/fisiología , Glicoproteínas de Membrana/farmacología , Neovascularización Patológica , Animales , Bovinos , Adhesión Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Quimiotaxis/efectos de los fármacos , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Factores de Crecimiento de Fibroblastos/farmacología , Cinética , Trombospondinas
3.
J Natl Cancer Inst ; 81(10): 781-8, 1989 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-2523976

RESUMEN

The effects of estradiol and two synthetic progestins (ORG2058 and R5020) on the expression of the high-affinity, metastasis-associated laminin receptor in two human breast carcinoma cell lines were examined. The T47D cell line contains estrogen and progesterone receptors, but the MDA-MB 231 cell line lacks both receptors. Treatment of T47D cells with 10(-9) M estradiol alone results in a three-fold increase (P less than or equal to .05) in the steady-state level of laminin receptor mRNA determined by RNA blot analysis as well as in cell-surface, laminin receptor expression that is evaluated by immunofluorescence. No effects of estradiol on the receptor-negative MDA-MB 231 cells were observed. Untreated and steroid-treated MDA-MB 231 cells had higher levels of laminin receptor mRNA than did untreated or estradiol-treated T47D cells. A more dramatic increase (five-fold; P less than or equal to .005) of mRNA and cell-surface expression in T47D cells was observed after treatment with estradiol plus 10(-8) M progestin or with progestin alone. Estradiol treatment also increased chemotaxis and haptotaxis of T47D cells but not of MDA-MB 231 cells to laminin; it had no effect on the attachment of these latter cells to laminin. Interestingly, treatment with estradiol plus progestin or progestin alone significantly increased the attachment of T47D cells to laminin but did not have an effect on either haptotaxis or chemotaxis to laminin. These results suggest that the various cell-laminin interactions are mediated by different mechanisms. The augmentation of laminin receptor mRNA by estrogen and progesterone treatment in hormone receptor-positive cells, but not in cells that lack these receptors, may relate functionally to the difference in the clinical aggressiveness between classes of breast cancers.


Asunto(s)
Neoplasias de la Mama/metabolismo , Estradiol/farmacología , Laminina/metabolismo , Congéneres de la Progesterona/farmacología , Receptores Inmunológicos/biosíntesis , Movimiento Celular/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Humanos , Laminina/farmacología , Pregnenodionas/farmacología , Promegestona/farmacología , ARN Mensajero/metabolismo , Receptores Inmunológicos/efectos de los fármacos , Receptores de Laminina , Células Tumorales Cultivadas/efectos de los fármacos
4.
J Natl Cancer Inst ; 85(3): 235-40, 1993 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-8423629

RESUMEN

BACKGROUND: Stable anchorage of circulating cancer cells to the vasculature is a critical step in the formation of hematogenous metastases. Although the basement membrane glycoprotein laminin clearly plays a crucial role in this event, the exact interactive pathways among cancer cells, laminin, and the vessel wall have not been elucidated. In a previous study, we identified synthetic peptide G, which contains the laminin-binding domain of the 67-kd laminin receptor and which inhibits tumor cell adhesion to endothelial cells. PURPOSE: To assess the role of the interaction between laminin and the 67-kd laminin receptor in hematogenous metastasis formation, we studied the effect of peptide G on melanoma cell behavior in vivo and in vitro. METHODS: The effect of peptide G and control peptides was studied in vivo on lung retention and colonizing potential of murine (B16BL6) and human (A2058) melanoma cells injected intravenously in C57BL/6 and nude mice, respectively. In addition, their effect on cell adhesion and chemotaxis to laminin and on binding of iodine 125-labeled laminin to cells was studied in vitro. RESULTS: In vivo, pretreatment of cells with peptide G resulted in a two- to 10-fold significant increase in the number of experimental lung metastases. A significant relative increase in lung retention of peptide G-treated tumor cells was observed 48 hours after injection, although after 4 hours a partial reduction was observed. In vitro, peptide G significantly increased laminin binding and cancer cell adhesion to laminin and subendothelial matrix, whereas chemotaxis to laminin was significantly inhibited. CONCLUSIONS: Peptide G differentially affected the biological response of cancer cells to laminin. In vitro, it increased laminin binding and cell adhesion to laminin and subendothelial matrix, whereas it inhibited cell chemotaxis to laminin. In vivo, the overall effect of peptide G was an augmentation of lung metastasis. IMPLICATIONS: Our findings suggest that direct adhesion of tumor cells to the subendothelial matrix is a main pathway for hematogenous metastases and that tumor cell-matrix interaction may be more relevant than tumor cell-endothelial cell attachment in this process.


Asunto(s)
Laminina/metabolismo , Melanoma/patología , Células Neoplásicas Circulantes , Péptidos/farmacología , Precursores de Proteínas , Receptores de Laminina , Secuencia de Aminoácidos , Animales , Adhesión Celular/efectos de los fármacos , Humanos , Melanoma Experimental/patología , Ratones , Datos de Secuencia Molecular
5.
J Natl Cancer Inst ; 87(4): 293-8, 1995 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-7535861

RESUMEN

BACKGROUND: The importance of matrix metalloproteinases in angiogenesis, tumor growth, and metastasis is well known. However, little is known about the role of matrix metalloproteinases in the formation of hemangiomas and about the possible therapeutic use of matrix metalloproteinase inhibitors in aggressive vascular tumors. PURPOSE: To study the role of matrix metalloproteinase in vascular tumors, we tested the antineoplastic activity of a synthetic inhibitor of matrix metalloproteinases, batimastat, on an experimental model of hemangioma, formed by murine endothelioma cells transformed by polyoma middle-T oncogene (eEnd.1). METHODS: The effect of batimastat was studied in vivo on the formation of hemorrhaging, cavernous hemangiomas by eEnd.1 endothelioma cells injected subcutaneously in nude mice and on the angiogenic response induced by an endothelioma cell supernatant embedded in a pellet of reconstituted basement membrane (Matrigel). The effect of batimastat was investigated in vitro on endothelial cell proliferation, motility, and invasion of a layer of Matrigel. RESULTS: Daily treatment with batimastat (30, 3, and 0.3 mg/kg at the site of eEnd.1 cell injection) inhibited tumor growth, with increased doubling time. The carboxamide derivative of batimastat, BB-374, a poor inhibitor of matrix metalloproteinase activity, was less active in reducing hemangioma growth. Histologic analysis of treated tumors indicated a reduction in the size of blood-filled spaces and in hemorrhage. Batimastat also inhibited the angiogenic response induced by cultured eEnd.1 endothelioma cell supernatant embedded in a pellet of Matrigel. Batimastat significantly inhibited endothelial cell invasion in vitro through a layer of Matrigel, but it showed no direct cytotoxic activity. CONCLUSIONS: Batimastat reduces in vivo growth of experimental hemangiomas, most probably by blocking endothelial cell recruitment by the transformed cells or by interfering with cell organization in vascular structures. IMPLICATIONS: These results confirm the importance of matrix metalloproteinase in endothelial cell recruitment that occurs in angiogenesis and in the formation of vascular tumors and suggest a therapeutic potential for synthetic matrix metalloproteinase inhibitors.


Asunto(s)
Hemangioendotelioma/tratamiento farmacológico , Metaloendopeptidasas/antagonistas & inhibidores , Neovascularización Patológica/tratamiento farmacológico , Fenilalanina/análogos & derivados , Tiofenos/farmacología , Animales , Relación Dosis-Respuesta a Droga , Femenino , Hemangioendotelioma/patología , Ratones , Invasividad Neoplásica , Fenilalanina/farmacología , Factores de Tiempo
6.
Cancer Res ; 51(20): 5672-8, 1991 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-1833053

RESUMEN

Stable attachment of cancer cells to the endothelium is a key step in the formation of metastasis. In this study, we have investigated the possibility that interaction between laminin and its Mr 67,000 high-affinity receptor (67 LR) could play a major role in this process. Scatchard analysis of laminin-binding studies showed that bovine aortic endothelial cells exhibit 46,000 high-affinity receptors that mediate, at least in part, the attachment of highly invasive melanoma cells. This endothelial cell-melanoma cell interaction was significantly inhibited by soluble laminin and by anti-laminin antibodies. Peptide G, an eicosapeptide derived from the complementary DNA sequence of the 67 LR precursor (IPCNNKGAHSVGLMWWMLAR) that specifically binds to laminin and presumably contains the active ligand-binding site of the receptor, specifically prevented attachment of the melanoma cells to both the bovine aortic endothelial cell monolayer and human umbilical vein endothelium. Thus, peptide G may selectively interfere with the metastatic cascade by inhibiting tumor cell attachment to endothelium via the laminin-67 LR pathway and is a potential new antimetastatic agent.


Asunto(s)
Endotelio Vascular/fisiopatología , Laminina/fisiología , Melanoma/fisiopatología , Receptores Inmunológicos/fisiología , Secuencia de Aminoácidos , Adhesión Celular/fisiología , Humanos , Laminina/genética , Laminina/metabolismo , Datos de Secuencia Molecular , Metástasis de la Neoplasia , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Receptores de Laminina , Células Tumorales Cultivadas
7.
Cancer Res ; 53(16): 3812-6, 1993 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-8393382

RESUMEN

Polyoma virus middle T-transformed murine endothelioma cell lines provide a useful model for studying vascular lesions such as hemangiomas, hemangiosarcomas, and Kaposi's sarcoma and tumor-associated angiogenesis. In vivo they produce fast-growing, hemorrhaging, cavernous blood-filled hemangiomas, mainly formed by recruited host endothelial cells, suggesting an angiogenesis-like process underlying the lesion. The molecular mechanism(s) responsible for the recruitment of host endothelial cells by endothelioma cells has not yet been identified. We found that five different cultured endothelioma cell lines produced a soluble factor, named endothelioma-derived motility factor (EDMF) that stimulates chemotaxis (motility induced by a gradient of soluble attractant), haptotaxis (motility in response to substrate-bound attractant), and chemoinvasion (migration through a layer of reconstituted basement membrane, Matrigel) of normal human, bovine, and murine endothelial cells. The inhibitory effect of actinomycin D and of enzymatic treatment on its activity proved that EDMF is a protein. EDMF binds to heparin, since its activity was inhibited by heparin, and it was retained on a heparin-Sepharose column. Its molecular weight, as assessed by Sephacryl S-200 gel filtration, ranges from 40,000-65,000. Although in many aspects EDMF is similar to vascular permeability factor-vascular endothelial growth factor, this was not detected in endothelioma cell supernatants, as assessed by enzyme-linked immunosorbent assay, thus indicating that EDMF might be related to, but is not identical with, vascular permeability factor. Our findings support the notion that recruitment of host endothelial cells by endothelioma cells in vivo might be mediated by a still unidentified, soluble factor that stimulates and directs endothelial cell migration.


Asunto(s)
Factores Quimiotácticos/metabolismo , Hemangioendotelioma/metabolismo , Invasividad Neoplásica , Proteínas de Neoplasias/metabolismo , Animales , Línea Celular Transformada , Movimiento Celular , Factores Quimiotácticos/química , Factores Quimiotácticos/fisiología , Hemangioendotelioma/patología , Ratones , Peso Molecular , Proteínas de Neoplasias/química , Proteínas de Neoplasias/fisiología , Poliomavirus
8.
Cancer Res ; 47(21): 5691-8, 1987 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-2959357

RESUMEN

Polyclonal antisera were made against biochemically purified laminin receptor protein as well as against synthetic peptides deduced from a complementary DNA clone corresponding to the COOH-terminal end of the laminin receptor (U.M. Wewer et al., Proc. Natl. Acad. Sci. USA, 83: 7137-7141, 1986). These antisera were used to study the potential role of laminin receptor in laminin-mediated attachment and haptotactic migration of human A2058 melanoma cells. The anti-laminin receptor antisera reacted with the surface of suspended, nonpermeabilized melanoma and carcinoma cells. The anti-laminin receptor antisera blocked the surface interaction of A2058 cells with endogenous laminin, resulting in the inhibition of laminin-mediated cell attachment. The A2058 melanoma cells migrated toward a gradient of solid phase laminin or fibronectin (haptotaxis). Anti-laminin antiserum abolished haptotaxis on laminin but not on fibronectin. Synthetic peptide GRGDS corresponding to the fibronectin cell-binding domain inhibited haptotaxis on fibronectin but not on laminin. Both types of anti-laminin receptor antisera inhibited haptotaxis on laminin but not on fibronectin. Using immunohistochemistry, invading human carcinoma cells in vivo exhibited a marked cytoplasmic immunoreactivity for the receptor antigen. Together these findings indicate a specific role for the laminin receptor in laminin-mediated migration and that the ligand binding of the laminin receptor is encompassed in the COOH-terminal end of the protein.


Asunto(s)
Neoplasias/patología , Receptores Inmunológicos/fisiología , Adhesión Celular , Movimiento Celular , Ensayo de Inmunoadsorción Enzimática , Fibronectinas/metabolismo , Humanos , Sueros Inmunes/inmunología , Laminina/metabolismo , Melanoma/patología , Invasividad Neoplásica , Receptores Inmunológicos/análisis , Receptores Inmunológicos/inmunología , Receptores de Laminina
9.
Thromb Res ; 140 Suppl 1: S182, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27161705

RESUMEN

INTRODUCTION: Platelet thrombospondin-1 (TSP-1) is a major endogenous regulator of growth factor activity in physiological and pathological processes, including tumor onset, progression and angiogenesis. We previously demonstrated that TSP-1 binds to FGF-2, sequestering the growth factor and inhibiting its angiogenic activity. We also identified a non-peptidic antiangiogenic compound (SM27) that retains the structural and functional properties of the FGF2-binding sequence of TSP-1. AIM: To identify new small molecule inhibitors of FGF2 that recapitulate the structure and functional properties of the FGF-2-binding site of TSP-1, by investigating the chemical space around SM27. MATERIALS AND METHODS: A similarity-based screening of small molecule libraries has been used to identify candidates, followed by docking calculations, and evaluation of the activity of the resulting compounds in biochemical and biophysical assays, to assess interaction with FGF2, and in experimental models of angiogenesis, to assess biological activity. RESULTS: The used integrated approach allowed selecting 7 bi-naphthalenic compounds that bound FGF2 inhibiting FGF2 binding to both heparan sulfate proteoglycans and FGFR1. The compounds inhibited FGF2-induced endothelial cell proliferation, vessel sprouting from aortic rings and angiogenesis in the chorioallantoic membrane assay, with improved potency over SM27. CONCLUSIONS: We have identified new compounds that are valuable as FGF inhibitors for potential therapeutic purposes. Moreover, these compounds are useful chemical tools to identify the minimal stereochemical requirements for FGF2 binding and activity to improve the design of new agents for antineoplastic therapy. ACKNOWLEDGEMENT: Supported by AIRC (Associazione Italiana per la Ricerca sul Cancro).

10.
Oncogene ; 20(50): 7293-300, 2001 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-11704858

RESUMEN

Tumor neovascularization is controlled by a balance between positive and negative effectors, whose production can be regulated by oncogenes and tumor suppressor genes. The aim of this study was to investigate whether the angiogenic potential of tumors could also be controlled by p73, a gene homologous to the tumor suppressor p53, whose involvement in tumor angiogenesis is known. We have studied the production of proangiogenic (VEGF, FGF-2, PIGF and PDGF) and antiangiogenic (TSP-1) factors in two p73 overexpressing clones obtained from the human ovarian carcinoma cells A2780. TSP-1 was downregulated in both clones compared to mock transfected cells, both at mRNA and protein level. Conversely, both clones showed an increased production of VEGF mRNA and protein. For both TSP-1 and VEGF, regulation of expression was partially due to modulation of the promoter activity, and was dependent on p53 status. Production of the other angiogenic factors FGF-2, PIGF and PDGF-B was also increased in p73 overexpressing clones. The two clones were more angiogenic than parental cells, as shown in vitro by their increased chemotactic activity for endothelial cells, and in vivo by the generation of more vascularized tumors. These findings suggest a potential role of p73 in tumor angiogenesis.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Factores de Crecimiento Endotelial/biosíntesis , Linfocinas/biosíntesis , Neovascularización Patológica/genética , Proteínas Nucleares/fisiología , Trombospondina 1/biosíntesis , Animales , Northern Blotting , Carcinoma/patología , Quimiotaxis/efectos de los fármacos , Medios de Cultivo Condicionados/química , Medios de Cultivo Condicionados/farmacología , Medio de Cultivo Libre de Suero , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Factores de Crecimiento Endotelial/genética , Endotelio Vascular/citología , Ensayo de Inmunoadsorción Enzimática , Femenino , Factor 2 de Crecimiento de Fibroblastos/biosíntesis , Factor 2 de Crecimiento de Fibroblastos/genética , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Genes p53 , Humanos , Linfocinas/genética , Proteínas de la Membrana , Ratones , Ratones Desnudos , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Neoplasias Experimentales/irrigación sanguínea , Neovascularización Patológica/metabolismo , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Neoplasias Ováricas/patología , Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Factor de Crecimiento Derivado de Plaquetas/genética , Regiones Promotoras Genéticas , Biosíntesis de Proteínas , Proteínas/genética , ARN Mensajero/biosíntesis , ARN Neoplásico/biosíntesis , Proteínas Recombinantes de Fusión/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trombospondina 1/genética , Transfección , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/metabolismo , Proteína Tumoral p73 , Proteína p53 Supresora de Tumor/fisiología , Proteínas Supresoras de Tumor , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
11.
Biochim Biophys Acta ; 1500(2): 227-40, 2000 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-10657592

RESUMEN

It has been shown that deletion of the chemokine receptor, CXCR4, causes disordered angiogenesis in mouse models. In the present studies, we examined the distribution and trafficking of CXCR4 in human endothelial cells, tested their responses to the CXCR4 ligand, SDF-1, and asked whether endothelial cell CXCR4 can serve as a cell surface receptor for the binding of viruses. The results show that CXCR4 is present on endothelial cells from coronary arteries, iliac arteries and umbilical veins (HUVEC), but expression was heterogeneous, with some cells expressing CXCR4 on their surface, while others did not. Addition of SDF-1 caused a rapid decrease in CXCR4 surface expression. It also caused CXCR4-mediated activation of MAPK, release of PGI(2), endothelial migration, and the formation of capillary-like structures by endothelial cells in culture. Co-culture of HUVEC with lymphoid cells that were chronically infected with a CD4-independent/CXCR4-tropic variant of HIV-2 resulted in the formation of multinucleated syncytia. Formation of the syncytia was inhibited by each of several different CXCR4 antibodies. Thus, our findings indicate: (1) that CXCR4 is widely expressed on human endothelial cells; (2) the CXCR4 ligand, SDF-1, can evoke a wide variety of responses from human endothelial cells; and (3) CXCR4 on endothelial cells can serve as a receptor for isolates of HIV that can utilize chemokine receptors in the absence of CD4.


Asunto(s)
Endotelio Vascular/metabolismo , VIH-2/fisiología , Receptores CXCR4/fisiología , Fármacos Anti-VIH/farmacología , Señalización del Calcio/efectos de los fármacos , Capilares/citología , Fusión Celular/efectos de los fármacos , Quimiocina CXCL12 , Quimiocinas CXC/farmacología , Quimiotaxis/efectos de los fármacos , Colágeno , Vasos Coronarios/citología , Efecto Citopatogénico Viral/efectos de los fármacos , Regulación hacia Abajo , Combinación de Medicamentos , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/virología , Epoprostenol/metabolismo , Citometría de Flujo , Expresión Génica , Humanos , Arteria Ilíaca/citología , Técnicas para Inmunoenzimas , Laminina , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Microscopía Fluorescente , Morfogénesis/efectos de los fármacos , Proteoglicanos , Receptor Cross-Talk , Receptor PAR-1 , Receptores CXCR4/genética , Receptores de Trombina/fisiología , Acetato de Tetradecanoilforbol/farmacología , Venas Umbilicales/citología
12.
Leukemia ; 17(1): 52-9, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12529660

RESUMEN

The mechanism by which aplidine, a marine natural product in early clinical development as an anticancer agent, induces cell growth inhibition and apoptosis has been investigated in the human leukemia cell line MOLT-4. This cell line is characterized not only by the ability to secrete VEGF, but also for the presence on its surface of the VEGF receptor-1 (VEGFR-1). Previous studies from our laboratory concerned with evaluating early changes in gene expression induced by aplidine in MOLT-4 cells have shown that the drug decreases the expression of VEGFR-1 (Marchini et al. Proc Am Assoc Cancer Res 2000; 41: 833). Here, we report the ability of aplidine to block the VEGF/VEGFR-1 loop. We found that aplidine blocked VEGF secretion that was temporally followed by a decrease in both VEGF and VEGFR-1 production. Aplidine did not directly affect either VEGF transcription or stabilization of its mRNA. Transfection of MOLT-4 cells with an antisense VEGF cDNA construct, resulted in inhibition of colony formations. One clone, transfected with sense VEGF cDNA, secreting 8-10 times more VEGF than parental cells, was less sensitive to aplidine-induced cytotoxicity and apoptosis than control cells. Moreover, addition of VEGF in the medium decreased the activity of aplidine in MOLT-4 cells. These data demonstrate that aplidine inhibits the growth and induces apoptosis in MOLT-4 cells through the inhibition of VEGF secretion which blocks the VEGF/VEGFR-1 autocrine loop necessary for the growth of these cells.


Asunto(s)
Antineoplásicos/farmacología , Depsipéptidos , Factores de Crecimiento Endotelial/antagonistas & inhibidores , Leucemia de Células T/tratamiento farmacológico , Linfocinas/antagonistas & inhibidores , Péptidos Cíclicos/farmacología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Comunicación Autocrina , División Celular/efectos de los fármacos , Cartilla de ADN/química , Dactinomicina/farmacología , Ensayo de Cambio de Movilidad Electroforética , Factores de Crecimiento Endotelial/genética , Factores de Crecimiento Endotelial/metabolismo , Semivida , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Leucemia de Células T/genética , Leucemia de Células T/metabolismo , Luciferasas/metabolismo , Linfocinas/genética , Linfocinas/metabolismo , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas/efectos de los fármacos , Inhibidores de la Síntesis de la Proteína/farmacología , ARN Mensajero/metabolismo , Transducción de Señal , Transfección , Células Tumorales Cultivadas/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Factores de Crecimiento Endotelial Vascular
13.
Clin Cancer Res ; 2(10): 1725-30, 1996 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9816123

RESUMEN

The effect of paclitaxel on the adhesive and motility properties of human ovarian carcinoma cell lines was investigated. Paclitaxel significantly inhibited the motility of OVCAR 5, SK-OV-3, and HOC-1OTC ovarian carcinoma cell lines (IC50 = 2.1 x 10(-8), 2 x 10(-9), and 1.9 x 10(-8) m, respectively) but did not affect the adhesion of these cells to the subendothelial matrix. The association between inhibition of motility and cytotoxic activity was investigated using an A2780 subclone (1A9) and three paclitaxel-resistant variants (designated 1A9/PTX22, 1A9/PTX10, and 1A9/PTX18). Although paclitaxel did not significantly affect the adhesion to subendothelial matrix of the sublines, it completely inhibited their migration. Inhibition of migration was similar in 1A9 cells and the resistant sublines, with an IC50 of 1 x 10(-8) for 1A9 cells and 5.4 x 10(-9), 1.1 x 10(-8), and 5.2 x 10(-9) m for 1A9/PTX22, 1A9/PTX10, and 1A9/PTX18, respectively. Paclitaxel inhibited motility induced by soluble attractant (chemotaxis) and immobilized attractant (haptotaxis). Inhibition of cell motility occurred in the absence of an antiproliferative effect, because higher concentrations of paclitaxel were required to inhibit tumor cell proliferation (IC50 = 1.9 x 10(-7) and 4.6 x 10(-6), 1 x 10(-5), and 3.1 x 10(-6) m for 1A9 and 1A9/PTX22, 1A9/PTX10, and 1A9/PTX18, respectively). These data show that paclitaxel is a potent inhibitor of ovarian carcinoma cell motility and that this activity is independent of its cytotoxic activity.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Movimiento Celular/efectos de los fármacos , Neoplasias Ováricas/tratamiento farmacológico , Paclitaxel/farmacología , Adhesión Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Femenino , Humanos , Neoplasias Ováricas/patología , Células Tumorales Cultivadas
14.
Clin Cancer Res ; 2(11): 1843-9, 1996 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9816139

RESUMEN

Endothelial cell migration is a critical event during angiogenesis, and inhibitors of cell motility can affect the angiogenic process. Paclitaxel (Taxol(R)), a microtubule-stabilizing antineoplastic cytotoxic drug, inhibits motility and invasiveness of several cell types. The aim of this study was to investigate the effect of paclitaxel on endothelial cell functions and on angiogenesis. In vivo, paclitaxel (20-28 mg/kg i.v.) significantly inhibited the angiogenic response induced by tumor cell supernatant embedded in a pellet of reconstituted basement membrane (Matrigel) injected s.c. into C57BL/6N mice. In vitro, paclitaxel inhibited endothelial cell proliferation, motility, invasiveness, and cord formation on Matrigel in a dose-dependent manner. The antiangiogenic activity of paclitaxel was not linked to its cytotoxicity, since inhibition of endothelial cell chemotaxis and invasiveness occurred at drug concentrations which did not affect endothelial cell proliferation. Another cytotoxic drug, cisplatin, that inhibited endothelial cell proliferation in vitro, did not affect angiogenesis in vivo. These data indicate that paclitaxel has a strong antiangiogenic activity, a property that might contribute to its antineoplastic activity in vivo.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Microtúbulos/efectos de los fármacos , Neovascularización Patológica/prevención & control , Paclitaxel/farmacología , Animales , Antineoplásicos/farmacología , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Cisplatino/farmacología , Colágeno/metabolismo , Combinación de Medicamentos , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiología , Laminina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales/irrigación sanguínea , Neoplasias Experimentales/tratamiento farmacológico , Proteoglicanos/metabolismo
15.
Clin Cancer Res ; 4(4): 985-92, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9563894

RESUMEN

Batimastat (also known as BB-94) is a synthetic matrix metalloproteinase inhibitor that has shown antineoplastic and antiangiogenic activity in various tumor models. In this study, two human ovarian carcinoma (HOC) xenografts (HOC22 and HOC8) were used to investigate the effect of batimastat on the antineoplastic activity of cisplatin. Both xenografts produced ascites and solid lesions in the peritoneal cavity of nude mice. HOC cells were inoculated i.p. in nude mice, and treatment was started at different stages of the disease. Batimastat was administered alone or concurrently with or subsequent to cisplatin therapy. In all of the protocols, the response of HOC xenografts was confirmed by cytological analysis of ascites and histological examination of the organs in the peritoneal cavity. Treatment of nude mice bearing early-stage (3 days after tumor implantation) HOC22 or HOC8 with cisplatin or batimastat alone delayed tumor growth and increased the survival time of the mice, although all animals eventually died. In contrast, treatment with batimastat (60 mg/kg i.p. every other day, for a total of eight injections) concomitantly with cisplatin (4 mg/kg i.v., every 7 days for a total of three injections) completely prevented growth and spread of both xenografts, and all animals were alive and healthy on day 200. The potentiation of cisplatin's activity by batimastat was dose dependent and was observed in the treatment of both advanced (7 days after tumor inoculation) and late-stage (20 days after inoculation) tumor. The administration of batimastat following cisplatin therapy also led to significant improvement in the survival of mice compared to treatment with cisplatin alone. These results suggest a potentiation of the antineoplastic activity of cisplatin by batimastat and support the use of the two agents in combination in the treatment of ovarian cancer patients.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma/tratamiento farmacológico , Cisplatino/uso terapéutico , Metaloendopeptidasas/antagonistas & inhibidores , Neoplasias Ováricas/tratamiento farmacológico , Fenilalanina/análogos & derivados , Inhibidores de Proteasas/farmacología , Tiofenos/farmacología , Animales , Antineoplásicos/administración & dosificación , Carcinoma/mortalidad , Cisplatino/administración & dosificación , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Quimioterapia Combinada , Femenino , Humanos , Ratones , Ratones Desnudos , Neoplasias Ováricas/mortalidad , Fenilalanina/administración & dosificación , Fenilalanina/farmacología , Inhibidores de Proteasas/administración & dosificación , Análisis de Supervivencia , Tiofenos/administración & dosificación , Trasplante Heterólogo
16.
Clin Cancer Res ; 5(11): 3603-7, 1999 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-10589777

RESUMEN

Matrix metalloproteinases (MMPs) have been implicated in tumor cell invasion, metastasis, and angiogenesis. BAY 12-9566, a novel, non-peptidic biphenyl MMP inhibitor, has shown preclinical activity on a broad range of tumor models and is currently in clinical development. The purpose of this study was to investigate the antiangiogenic activity of BAY 12-9566. In vitro, BAY 12-9566 prevented matrix invasion by endothelial cells in a concentration-dependent manner (IC50 = 8.4x10(-7) M), without affecting cell proliferation. In vivo, oral daily administration of BAY 12-9566 (50-200 mg/kg) inhibited angiogenesis induced by basic fibroblast growth factor in the Matrigel plug assay, reducing the hemoglobin content of the pellets. Histological analysis showed a reduction in the amount of functional vessels within the Matrigel. We conclude that the MMP inhibitor BAY 12-9566 inhibits angiogenesis, a property that further supports its clinical development as an antimetastatic agent.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antineoplásicos/farmacología , Endotelio Vascular/fisiología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Inhibidores de la Metaloproteinasa de la Matriz , Neovascularización Patológica/prevención & control , Neovascularización Fisiológica/efectos de los fármacos , Compuestos Orgánicos , Animales , Compuestos de Bifenilo , División Celular/efectos de los fármacos , Células Cultivadas , Colágeno , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Humanos , Laminina , Ratones , Ratones Endogámicos C57BL , Fenilbutiratos , Proteoglicanos , Venas Umbilicales
17.
Curr Opin Pharmacol ; 1(4): 378-84, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11710736

RESUMEN

The great interest in the potential antineoplastic effect of targeting tumor-associated blood vessels has generated an expanding armamentarium of therapeutic tools that include antiangiogenic compounds, aimed at preventing the formation of vessels, and antivascular compounds, targeted to the existing tumor vasculature. Following promising preclinical studies, antiangiogenic drugs have rapidly gained access to clinical trials.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Vasos Sanguíneos/efectos de los fármacos , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Animales , Terapia Combinada , Humanos , Flujo Sanguíneo Regional/efectos de los fármacos
18.
J Leukoc Biol ; 39(1): 113-9, 1986 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-3455708

RESUMEN

Three malignant tumors (3LL carcinoma, mFS6 sarcoma, and B16 F1 melanoma) were transplanted in mice with congenital (beige) or acquired (anti-asialo GM1-treated) defects of natural killer cell (NK) activity. The macrophage content of the neoplastic tissues was not influenced by host NK activity levels. These data suggest that NK cell-mediated resistance does not play an appreciable role in the regulation of the levels of tumor-associated macrophages in established malignancy.


Asunto(s)
Carcinoma/inmunología , Inmunidad Innata , Células Asesinas Naturales/inmunología , Macrófagos/inmunología , Melanoma/inmunología , Sarcoma Experimental/inmunología , Animales , Línea Celular , Heterocigoto , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Mutantes , Especificidad de la Especie
19.
Crit Rev Oncol Hematol ; 37(1): 53-60, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11164719

RESUMEN

Inhibitors of matrix metalloproteinases (MMPs), enzymes involved in the processes of tumor growth, angiogenesis, invasion and metastasis, represent a promising new potential approach to cancer therapy. Several synthetic inhibitors of MMPs have been developed, many of which are currently in clinical trials. This review will describe some inhibitors of MMPs, presenting results of preclinical studies and, where available, their current clinical status as well. Issues concerning the use of MMP inhibitors, the design of clinical trials and the assessment of clinical response will also be addressed.


Asunto(s)
Antineoplásicos/uso terapéutico , Metaloendopeptidasas/antagonistas & inhibidores , Inhibidores Enzimáticos/uso terapéutico , Humanos , Invasividad Neoplásica/prevención & control , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Neovascularización Patológica/tratamiento farmacológico
20.
Eur J Cancer ; 40(6): 881-9, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15120043

RESUMEN

The development of a functional vasculature within a tumour is a requisite for its growth and progression. This fact has led to the design of therapies directed toward the tumour vasculature, aiming either to prevent the formation of new vessels (anti-angiogenic) or to damage existing vessels (vascular targeting). The development of agents with different mechanisms of action requires powerful preclinical models for the analysis and optimization of these therapies. This review concerns 'classical' assays of angiogenesis in vitro and in vivo, recent approaches to target identification (analysis of gene and protein expression), and the study of morphological and functional changes in the vasculature in vivo (imaging techniques). It mainly describes assays designed for anti-angiogenic compounds, indicating, where possible, their application to the study of vascular-targeting agents.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Neoplasias/irrigación sanguínea , Animales , Apoptosis , División Celular , Evaluación de Medicamentos/métodos , Ensayos de Selección de Medicamentos Antitumorales/métodos , Humanos , Modelos Biológicos , Neoplasias/patología , Neovascularización Patológica/tratamiento farmacológico , Pez Cebra
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA