Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
Chem Commun (Camb) ; 50(93): 14504-7, 2014 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-25308125

RESUMEN

Pressure-driven orbital reordering in the quantum magnet [CuF2(H2O)2(pyz)], (pyz = pyrazine), dramatically affects its magnetic exchange interactions. The crystal chemistry of this system is enriched with a new phase above 3 GPa, surprisingly concomitant with other polymorphs. Moreover, we discovered an unprecedented compound with a different stoichiometry, [(CuF2(H2O)2)2(pyz)], featuring magnetic bi-layers.

2.
Aliment Pharmacol Ther ; 39(5): 507-17, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24428642

RESUMEN

BACKGROUND: Early detection and treatment of colorectal adenomatous polyps (AP) and colorectal cancer (CRC) is associated with decreased mortality for CRC. However, accurate, non-invasive and compliant tests to screen for AP and early stages of CRC are not yet available. A blood-based screening test is highly attractive due to limited invasiveness and high acceptance rate among patients. AIM: To demonstrate whether gene expression signatures in the peripheral blood mononuclear cells (PBMC) were able to detect the presence of AP and early stages CRC. METHODS: A total of 85 PBMC samples derived from colonoscopy-verified subjects without lesion (controls) (n = 41), with AP (n = 21) or with CRC (n = 23) were used as training sets. A 42-gene panel for CRC and AP discrimination, including genes identified by Digital Gene Expression-tag profiling of PBMC, and genes previously characterised and reported in the literature, was validated on the training set by qPCR. Logistic regression analysis followed by bootstrap validation determined CRC- and AP-specific classifiers, which discriminate patients with CRC and AP from controls. RESULTS: The CRC and AP classifiers were able to detect CRC with a sensitivity of 78% and AP with a sensitivity of 46% respectively. Both classifiers had a specificity of 92% with very low false-positive detection when applied on subjects with inflammatory bowel disease (n = 23) or tumours other than CRC (n = 14). CONCLUSION: This pilot study demonstrates the potential of developing a minimally invasive, accurate test to screen patients at average risk for colorectal cancer, based on gene expression analysis of peripheral blood mononuclear cells obtained from a simple blood sample.


Asunto(s)
Pólipos Adenomatosos/diagnóstico , Biomarcadores de Tumor/genética , Neoplasias Colorrectales/diagnóstico , Leucocitos Mononucleares/metabolismo , Transcriptoma , Pólipos Adenomatosos/genética , Adulto , Anciano , Anciano de 80 o más Años , Colonoscopía , Neoplasias Colorrectales/genética , Detección Precoz del Cáncer , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Sensibilidad y Especificidad , Adulto Joven
3.
Oncogene ; 33(44): 5163-72, 2014 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-24213569

RESUMEN

TAT-RasGAP(317-326), a peptide corresponding to the 317-326 sequence of p120 RasGAP coupled with a cell-permeable TAT-derived peptide, sensitizes the death response of various tumor cells to several anticancer treatments. We now report that this peptide is also able to increase cell adherence, prevent cell migration and inhibit matrix invasion. This is accompanied by a marked modification of the actin cytoskeleton and focal adhesion redistribution. Interestingly, integrins and the small Rho GTP-binding protein, which are well-characterized proteins modulating actin fibers, adhesion and migration, do not appear to be required for the pro-adhesive properties of TAT-RasGAP(317-326). In contrast, deleted in liver cancer-1, a tumor suppressor protein, the expression of which is often deregulated in cancer cells, was found to be required for TAT-RasGAP(317-326) to promote cell adherence and inhibit migration. These results show that TAT-RasGAP(317-326), besides its ability to favor tumor cell death, hampers cell migration and invasion.


Asunto(s)
Adhesión Celular/efectos de los fármacos , Proteínas Activadoras de GTPasa/metabolismo , Proteínas Activadoras de GTPasa/farmacología , Fragmentos de Péptidos/farmacología , Proteínas Supresoras de Tumor/metabolismo , Actinas/metabolismo , Animales , Línea Celular Tumoral/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Fibroblastos , Proteínas Activadoras de GTPasa/genética , Humanos , Receptores de Hialuranos/metabolismo , Integrinas/metabolismo , Ratones , Procesamiento Proteico-Postraduccional , Proteínas Supresoras de Tumor/genética , Quinasas Asociadas a rho/metabolismo
4.
Oncogene ; 31(1): 48-59, 2012 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-21666716

RESUMEN

Cyclooxyganase-2 (COX-2), a rate-limiting enzyme in the prostaglandin synthesis pathway, is overexpressed in many cancers and contributes to cancer progression through tumor cell-autonomous and paracrine effects. Regular use of non-steroidal anti-inflammatory drugs or selective COX-2 inhibitors (COXIBs) reduces the risk of cancer development and progression, in particular of the colon. The COXIB celecoxib is approved for adjunct therapy in patients with Familial adenomatous polyposis at high risk for colorectal cancer (CRC) formation. Long-term use of COXIBs, however, is associated with potentially severe cardiovascular complications, which hampers their broader use as preventive anticancer agents. In an effort to better understand the tumor-suppressive mechanisms of COXIBs, we identified MAGUK with Inverted domain structure-1 (MAGI1), a scaffolding protein implicated in the stabilization of adherens junctions, as a gene upregulated by COXIB in CRC cells and acting as tumor suppressor. Overexpression of MAGI1 in CRC cell lines SW480 and HCT116 induced an epithelial-like morphology; stabilized E-cadherin and ß-catenin localization at cell-cell junctions; enhanced actin stress fiber and focal adhesion formation; increased cell adhesion to matrix proteins and suppressed Wnt signaling, anchorage-independent growth, migration and invasion in vitro. Conversely, MAGI1 silencing decreased E-cadherin and ß-catenin localization at cell-cell junctions; disrupted actin stress fiber and focal adhesion formation; and enhanced Wnt signaling, anchorage-independent growth, migration and invasion in vitro. MAGI1 overexpression suppressed SW480 and HCT116 subcutaneous primary tumor growth, attenuated primary tumor growth and spontaneous lung metastasis in an orthotopic model of CRC, and decreased the number and size of metastatic nodules in an experimental model of lung metastasis. Collectively, these results identify MAG1 as a COXIB-induced inhibitor of the Wnt/ß-catenin signaling pathway, with tumor-suppressive and anti-metastatic activity in experimental colon cancer.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/fisiología , Neoplasias Colorrectales/tratamiento farmacológico , Inhibidores de la Ciclooxigenasa 2/farmacología , Proteínas Supresoras de Tumor/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Adhesión Celular , Moléculas de Adhesión Celular , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Neoplasias Colorrectales/patología , Femenino , Guanilato-Quinasas , Humanos , Neoplasias Pulmonares/secundario , Ratones , Invasividad Neoplásica , Vía de Señalización Wnt , beta Catenina/fisiología
5.
Oncogene ; 31(12): 1521-32, 2012 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-21841820

RESUMEN

Milk fat globule-EGF factor 8 (MFG-E8) is a glycoprotein highly expressed in breast cancer that contributes to tumor progression through largely undefined mechanisms. By analyzing publicly available gene expression profiles of breast carcinomas, we found that MFG-E8 is highly expressed in primary and metastatic breast carcinomas, associated with absent estrogen receptor expression. Immunohistochemistry analysis of breast cancer biopsies revealed that MFG-E8 is expressed on the cell membrane as well as in the cytoplasm and nucleus. We also show that increased expression of MFG-E8 in mammary carcinoma cells increases their tumorigenicity in immunodeficient mice, and conversely, its downregulation reduces their in vivo growth. Moreover, expression of MFG-E8 in immortalized mammary epithelial cells promotes their growth and branching in three-dimensional collagen matrices and induces the expression of cyclins D1/D3 and N-cadherin. A mutant protein unable to bind integrins can in part exert these effects, indicating that MFG-E8 function is only partially dependent on integrin activation. We conclude that MFG-E8-dependent signaling stimulates cell proliferation and the acquisition of mesenchymal properties and contributes to mammary carcinoma development.


Asunto(s)
Antígenos de Superficie/fisiología , Ciclina D1/metabolismo , Ciclina D3/metabolismo , Células Epiteliales/metabolismo , Neoplasias Mamarias Experimentales/metabolismo , Animales , Mama/metabolismo , Cadherinas/metabolismo , Línea Celular , Proliferación Celular , Transición Epitelial-Mesenquimal , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones SCID , Proteínas de la Leche , Trasplante de Neoplasias , Transducción de Señal/fisiología
6.
Nuklearmedizin ; 50(6): 225-33, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21989840

RESUMEN

AIM: To visualize neovasculature and/or tumour integrin αvß3 we selected the binding moiety Arg-Gly-Asp-D-Tyr-Lys (RGDyK) coupled to NODAGA for labeling with 68Ga. METHODS: NODAGA-RGDyK (ABX) was labeled with the 68Ga eluate from the 68Ge generator IGG100 using the processor unit PharmTracer. Biodistribution was measured in female Hsd mice sacrificed 10, 30, 60 and 90 min after i.v. injection of 68Ga-NODAGA-RGDyK for OLINDA dosimetry extrapolated to humans. Tumour targeting was studied in SCID mice bearing A431 and other tumour transplants using microPET and biodistribution measurements. RESULTS: Effective half-life of 68Ga-NODAGA-RGDyK was ~25 min for total body and most organs except liver and spleen that showed stable activity retention. With a bladder voiding interval of 0.5 h the calculated effective dose (ED) was 0.012 and 0.016 mSv/MBq for males and females, respectively. Rapid uptake within 10 min was observed in A431 tumours with dynamic PET followed by a slow release. Biodistribution measurements showed a 68Ga-NODAGA-RGDyK uptake in A431 tumours of 3.4±0.4 and 2.7±0.3%ID/g at 1 and 2 h, respectively. Similar uptakes were observed in a mouse and human breast and ovarian cancer xenografts. Co-injection of excess (5 mg/kg) unlabeled NODAGA-RGDyK with the radiotracer reduced tumour uptake at one hour to 0.23±0.01%ID/g, but similarly decreased uptake in normal organs as well. When unlabeled peptide was injected 15 min after 68Ga-NODAGA-RGDyK, uptake diminished particularly in tumour and adrenals, suggestive of a different binding mode compared with other normal tissues. CONCLUSION: NODAGA-RGDyK was reliably labeled with 68Ga and revealed a predicted ED of 0.014 mSv/MBq. Tumour uptake was rapid and significant and was chased with unlabeled RGDyK in a similar manner as adrenal uptake.


Asunto(s)
Carcinoma de Células Escamosas/diagnóstico por imagen , Carcinoma de Células Escamosas/metabolismo , Integrina alfaVbeta3/metabolismo , Oligopéptidos/farmacocinética , Tomografía de Emisión de Positrones/métodos , Animales , Carga Corporal (Radioterapia) , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Femenino , Radioisótopos de Galio , Compuestos Heterocíclicos/química , Compuestos Heterocíclicos/farmacocinética , Compuestos Heterocíclicos con 1 Anillo , Humanos , Integrina alfaVbeta3/química , Tasa de Depuración Metabólica , Ratones , Ratones Endogámicos ICR , Especificidad de Órganos , Radiofármacos/síntesis química , Radiofármacos/farmacocinética , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Recuento Corporal Total
7.
J Proteome Res ; 8(10): 4779-88, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19711906

RESUMEN

Supernatants from cell cultures (also called conditioned media, CMs) are commonly analyzed to study the pool of secreted proteins (secretome). To reduce the exogenous protein background, serum-free media are often used to obtain CMs. Serum deprivation, however, can severely affect cell viability and phenotype, including protein secretion. We present a strategy to analyze the proteins secreted by cells in fetal bovine serum-containing CMs, which combines the advantage of metabolic labeling and protein concentration linearization techniques. Incubation of CMs with a hexapeptide ligand library was used to reduce the dynamic range of the samples and led to the identification of 3 times more proteins than in untreated CM samples. Labeling with a deuterated amino acid was used to distinguish between cellular proteins and homologous bovine proteins contained in the medium. Application of the strategy to two breast cancer cell lines led to the identification of proteins secreted in different amounts and which could correlate with their varying degree of aggressiveness. Selected reaction monitoring (SRM)-based quantitation of three proteins of interest in the crude samples yielded data in good agreement with the results from concentration-equalized samples.


Asunto(s)
Proteínas Sanguíneas , Medios de Cultivo Condicionados/metabolismo , Proteínas , Proteómica/métodos , Suero/metabolismo , Animales , Proteínas Sanguíneas/química , Proteínas Sanguíneas/aislamiento & purificación , Bovinos , Línea Celular Tumoral , Medios de Cultivo Condicionados/química , Humanos , Marcaje Isotópico , Espectrometría de Masas , Fragmentos de Péptidos/análisis , Fragmentos de Péptidos/química , Mapeo Peptídico , Proteínas/análisis , Proteínas/química , Proteínas/metabolismo , Proteoma/análisis , Proteoma/química , Proteoma/metabolismo , Reproducibilidad de los Resultados
8.
Oncogene ; 26(39): 5722-32, 2007 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-17369858

RESUMEN

Tumor necrosis factor (TNF) is a pro-inflammatory cytokine exerting pleiotropic effects on endothelial cells. Depending on the vascular context it can induce endothelial cell activation and survival or death. The microenvironmental cues determining whether endothelial cells will survive or die, however, have remained elusive. Here we report that integrin ligation acts permissive for TNF-induced protein kinase B (PKB/Akt) but not nuclear factor (NF)-kappaB activation. Concomitant activation of PKB/Akt and NF-kappaB is essential for the survival of endothelial cells exposed to TNF. Active PKB/Akt strengthens integrin-dependent endothelial cell adhesion, whereas disruption of actin stress fibers abolishes the protective effect of PKB/Akt. Integrin-mediated adhesion also represses TNF-induced JNK activation, but JNK activity is not required for cell death. The alphaVbeta3/alphaVbeta5 integrin inhibitor EMD121974 sensitizes endothelial cells to TNF-dependent cytotoxicity and active PKB/Akt attenuates this effect. Interferon gamma synergistically enhanced TNF-induced endothelial cell death in all conditions tested. Taken together, these observations reveal a novel permissive role for integrins in TNF-induced PKB/Akt activation and prevention of TNF-induced death distinct of NF-kappaB, and implicate the actin cytoskeleton in PKB/Akt-mediated cell survival. The sensitizing effect of EMD121974 on TNF cytotoxicity may open new perspectives to the therapeutic use of TNF as anticancer agent.


Asunto(s)
Apoptosis/fisiología , Adhesión Celular , Endotelio Vascular/citología , Integrina alfaVbeta3/metabolismo , Integrinas/metabolismo , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Vitronectina/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Actinas/metabolismo , Western Blotting , Células Cultivadas , Citoesqueleto/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Endotelio Vascular/metabolismo , Citometría de Flujo , Humanos , Etiquetado Corte-Fin in Situ , Integrina alfaVbeta3/antagonistas & inhibidores , Integrinas/antagonistas & inhibidores , MAP Quinasa Quinasa 4/metabolismo , FN-kappa B/genética , Fosforilación , Receptores de Vitronectina/antagonistas & inhibidores , Transducción de Señal , Esferoides Celulares
9.
J Thromb Haemost ; 5(1): 166-73, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17059425

RESUMEN

BACKGROUND: Recent evidence indicates that zoledronate, a nitrogen-containing bisphosphonate used to treat conditions of increased bone resorption, may have anti-angiogenic activity. The endothelial cells signaling events modulated by zoledronate remain largely elusive. OBJECTIVES: The aim of this work was to identify signaling events suppressed by zoledronate in endothelial cells and responsible for some of its biological effects. METHODS: Human umbilical vein endothelial cells (HUVEC) were exposed to zoledronate, isoprenoid analogs (i.e. farnesol and geranylgeraniol) and various inhibitors of signaling, and the effect on adhesion, survival, migration, actin cytoskeleton and signaling events characterized. RESULTS: Zoledronate reduced Ras prenylation, Ras and RhoA translocation to the membrane, and sustained ERK1/2 phosphorylation and tumor necrosis factor (TNF) induced JNK phosphorylation. Isoprenoid analogs attenuated zoledronate effects on HUVEC adhesion, actin stress fibers and focal adhesions, migration and survival. Isoprenoid analogs also restored Ras prenylation, RhoA translocation to the membrane, sustained FAK and ERK1/2 phosphorylation and prevented suppression of protein kinase B (PKB) and JNK phosphorylation in HUVEC exposed to TNF in the presence of zoledronate. Pharmacological inhibition of Rock, a RhoA target mediating actin fiber formation, phosphatidylinositol 3-kinase, an activator of PKB, MEK1/2, an activator of ERK1/2, and JNK, recapitulated individual zoledronate effects, consistent with the involvement of these molecules and pathways and their inhibition in the zoledronate effects. CONCLUSIONS: This work has demonstrated that zoledronate inhibits HUVEC adhesion, survival, migration and actin stress fiber formation by interfering with protein prenylation and has identified ERK1/2, JNK, Rock, FAK and PKB as kinases affected by zoledronate in a prenylation-dependent manner.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Difosfonatos/farmacología , Células Endoteliales/efectos de los fármacos , Imidazoles/farmacología , Prenilación de Proteína/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Conservadores de la Densidad Ósea/farmacología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Diterpenos/farmacología , Células Endoteliales/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Farnesol/farmacología , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , MAP Quinasa Quinasa 4/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor de Necrosis Tumoral alfa/toxicidad , Venas Umbilicales , Ácido Zoledrónico , Quinasas Asociadas a rho , Proteína de Unión al GTP rhoA/metabolismo
10.
Bull Cancer ; 93(8): E90-100, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16935777

RESUMEN

Recombinant human TNF (rhTNF) has a selective effect on endothelial cells in tumour angiogenic vessels. Its clinical use has been limited because of its property to induce vascular collapsus. TNF administration through isolated limb perfusion (ILP) for regionally advanced melanomas and soft tissue sarcomas of the limbs was shown to be safe and efficient. When combined to the alkylating agent melphalan, a single ILP produces a very high objective response rate. ILP with TNF and melphalan provided the proof of concept that a vasculotoxic strategy combined to chemotherapy may produce a strong anti-tumour effect. The registered indication of TNF-based ILP is a regional therapy for regionally spread tumours. In soft tissue sarcomas, it is a limb sparing neoadjuvant treatment and, in melanoma in-transit metastases, a curative treatment. Despite its demonstrated regional efficiency TNF-based ILP is unlikely to have any impact on survival. High TNF dosages induce endothelial cells apoptosis, leading to vascular destruction. However, lower TNF dosage produces a very strong effect that is to increase the drug penetration into the tumour, presumably by decreasing the intratumoural hypertension resulting in better tumour uptake. TNF-ILP allowed the identification of the role of alphaVbeta3 integrin deactivation as an important mechanism of antiangiogenesis. Several recent studies have shown that TNF targeting is possible, paving the way to a new opportunity to administer TNF systemically for improving cancer drug penetration. TNF was the first agent registered for the treatment of cancer that improves drug penetration in tumours and selectively destroys angiogenic vessels.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Factor de Necrosis Tumoral alfa/uso terapéutico , Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Vasos Sanguíneos/efectos de los fármacos , Quimioterapia del Cáncer por Perfusión Regional/métodos , Humanos , Interferón gamma/administración & dosificación , Melanoma/tratamiento farmacológico , Melanoma/mortalidad , Melfalán/administración & dosificación , Neoplasias/irrigación sanguínea , Proyectos Piloto , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/uso terapéutico , Sarcoma/irrigación sanguínea , Sarcoma/tratamiento farmacológico , Factor de Necrosis Tumoral alfa/administración & dosificación
12.
Cell Mol Life Sci ; 60(6): 1135-57, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12861381

RESUMEN

New blood vessel formation, a process referred to as angiogenesis, is essential for embryonic development and for many physiological and pathological processes during postnatal life, including cancer progression. Endothelial cell adhesion molecules of the integrin family have emerged as critical mediators and regulators of angiogenesis and vascular homeostasis. Integrins provide the physical interaction with the extracellular matrix necessary for cell adhesion, migration and positioning, and induction of signaling events essential for cell survival, proliferation and differentiation. Antagonists of integrin alpha V beta 3 suppress angiogenesis in many experimental models and are currently tested in clinical trials for their therapeutic efficacy against angiogenesis-dependent diseases, including cancer. Furthermore, interfering with signaling pathways downstream of integrins results in suppression of angiogenesis and may have relevant therapeutic implications. In this article we review the role of integrins in endothelial cell function and angiogenesis. In the light of recent advances in the field, we will discuss their relevance as a therapeutic target to suppress tumor angiogenesis.


Asunto(s)
Vasos Sanguíneos/fisiología , Integrinas/fisiología , Proteínas Serina-Treonina Quinasas , Animales , Vasos Sanguíneos/citología , Vasos Sanguíneos/crecimiento & desarrollo , Moléculas de Adhesión Celular/fisiología , Supervivencia Celular , Ciclooxigenasa 2 , Endotelio Vascular/citología , Endotelio Vascular/fisiología , Homeostasis , Humanos , Isoenzimas/metabolismo , Sistema Linfático/citología , Sistema Linfático/crecimiento & desarrollo , Sistema Linfático/fisiología , Sistema de Señalización de MAP Quinasas , Microdominios de Membrana/metabolismo , Proteínas de la Membrana , Modelos Biológicos , FN-kappa B/metabolismo , Neovascularización Patológica , Neovascularización Fisiológica , Prostaglandina-Endoperóxido Sintasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Proteínas de Unión al GTP rho/metabolismo
13.
Endothelium ; 9(3): 151-60, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12380640

RESUMEN

Tumor angiogenesis is an essential step in tumor progression and metastasis formation. Suppression of tumor angiogenesis results in the inhibition of tumor growth. Recent evidence indicates that vascular integrins, in particular alpha V beta 3, are important regulators of angiogenesis, including tumor angiogenesis. Integrin alpha V beta 3 antagonists, such as blocking antibodies or peptides, suppress tumor angiogenesis and tumor progression in many preclinical tumor models. The potential therapeutic efficacy of extracellular integrin antagonists in human cancer is currently being tested in clinical trials. Selective disruption of the tumor vasculature by high doses of tumor necrosis factor (TNF) and interferon gamma (IFN-gamma), and the antiangiogenic activity of nonsteroidal anti-inflammatory drugs are associated with the suppression of integrin alpha V beta 3 function and signaling in endothelial cells. Furthermore, expression of isolated integrin cytoplasmic domains disrupts integrin-dependent adhesion, resulting in endothelial cell detachment and apoptosis. These results confirm the critical role of vascular integrins in promoting endothelial cell survival and angiogenesis and suggest that intracellular targeting of integrin function and signaling may be an alternative strategy to extracellular integrin antagonists for the therapeutic inhibition of tumor angiogenesis.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Endotelio Vascular/efectos de los fármacos , Integrinas/antagonistas & inhibidores , Neoplasias/irrigación sanguínea , Inhibidores de la Angiogénesis/farmacología , Moléculas de Adhesión Celular/fisiología , Ciclooxigenasa 2 , Sistemas de Liberación de Medicamentos , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Humanos , Integrina alfaVbeta3/antagonistas & inhibidores , Interferón gamma/farmacología , Isoenzimas/farmacología , Proteínas de la Membrana , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neovascularización Patológica , Prostaglandina-Endoperóxido Sintasas/farmacología , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Proteínas de Unión al GTP rho/metabolismo
14.
Infection ; 30(3): 168-70, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12120945

RESUMEN

A 71-year-old female patient was hospitalized with membranous laryngopharyngitis typical of classical diphtheria. A toxigenic strain of Corynebacterium ulcerans was isolated from the throat. The patient was treated for 6 days with amoxicillin-clavulanic acid and recovered without complications. This second reported case of diphtheric laryngopharyngitis caused by C. ulcerans in Switzerland is a reminder that C. ulcerans should be included as a possible agent in patients with classical diphtheria symptoms.


Asunto(s)
Infecciones por Corynebacterium/diagnóstico , Corynebacterium/aislamiento & purificación , Difteria/diagnóstico , Laringitis/microbiología , Anciano , Antibacterianos , Corynebacterium/clasificación , Infecciones por Corynebacterium/tratamiento farmacológico , Diagnóstico Diferencial , Quimioterapia Combinada/administración & dosificación , Femenino , Estudios de Seguimiento , Humanos , Laringitis/diagnóstico , Laringitis/tratamiento farmacológico , Suiza , Resultado del Tratamiento
16.
J Immunol ; 167(12): 7150-6, 2001 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-11739538

RESUMEN

Many tumor-associated Ags represent tissue differentiation Ags that are poorly immunogenic. Their weak immunogenicity may be due to immune tolerance to self-Ags. Prostatic acid phosphatase (PAP) is just such an Ag that is expressed by both normal and malignant prostate tissue. We have previously demonstrated that PAP can be immunogenic in a rodent model. However, generation of prostate-specific autoimmunity was seen only when a xenogeneic homolog of PAP was used as the immunogen. To explore the potential role of xenoantigen immunization in cancer patients, we performed a phase I clinical trial using dendritic cells pulsed with recombinant mouse PAP as a tumor vaccine. Twenty-one patients with metastatic prostate cancer received two monthly vaccinations of xenoantigen-loaded dendritic cells with minimal treatment-associated side effects. All patients developed T cell immunity to mouse PAP following immunization. Eleven of the 21 patients also developed T cell proliferative responses to the homologous self-Ag. These responses were associated with Ag-specific IFN-gamma and/or TNF-alpha secretion, but not IL-4, consistent with induction of Th1 immunity. Finally, 6 of 21 patients had clinical stabilization of their previously progressing prostate cancer. All six of these patients developed T cell immunity to human PAP following vaccination. These results demonstrate that xenoantigen immunization can break tolerance to a self-Ag in humans, resulting in a clinically significant antitumor effect.


Asunto(s)
Antígenos Heterófilos/inmunología , Vacunas contra el Cáncer/uso terapéutico , Células Dendríticas/trasplante , Inmunoterapia Adoptiva , Neoplasias de la Próstata/terapia , Fosfatasa Ácida , Animales , Citocinas/biosíntesis , Ensayo de Inmunoadsorción Enzimática , Humanos , Cinética , Masculino , Ratones , Persona de Mediana Edad , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/inmunología , Proteínas Tirosina Fosfatasas/inmunología , Células TH1/inmunología , Resultado del Tratamiento
17.
J Immunol ; 167(11): 6431-40, 2001 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-11714809

RESUMEN

Several lines of evidences have suggested that T cell activation could be impaired in the tumor environment, a condition referred to as tumor-induced immunosuppression. We have previously shown that tenascin-C, an extracellular matrix protein highly expressed in the tumor stroma, inhibits T lymphocyte activation in vitro, raising the possibility that this molecule might contribute to tumor-induced immunosuppression in vivo. However, the region of the protein mediating this effect has remained elusive. Here we report the identification of the minimal region of tenascin-C that can inhibit T cell activation. Recombinant fragments corresponding to defined regions of the molecule were tested for their ability to inhibit in vitro activation of human peripheral blood T cells induced by anti-CD3 mAbs in combination with fibronectin or IL-2. A recombinant protein encompassing the alternatively spliced fibronectin type III domains of tenascin-C (TnFnIII A-D) vigorously inhibited both early and late lymphocyte activation events including activation-induced TCR/CD8 down-modulation, cytokine production, and DNA synthesis. In agreement with this, full length recombinant tenascin-C containing the alternatively spliced region suppressed T cell activation, whereas tenascin-C lacking this region did not. Using a series of smaller fragments and deletion mutants issued from this region, we have identified the TnFnIII A1A2 domain as the minimal region suppressing T cell activation. Single TnFnIII A1 or A2 domains were no longer inhibitory, while maximal inhibition required the presence of the TnFnIII A3 domain. Altogether, these data demonstrate that the TnFnIII A1A2 domain mediate the ability of tenascin-C to inhibit in vitro T cell activation and provide insights into the immunosuppressive activity of tenascin-C in vivo.


Asunto(s)
Empalme Alternativo/inmunología , Citocinas/antagonistas & inhibidores , Fibronectinas/fisiología , Inmunosupresores/farmacología , Activación de Linfocitos/inmunología , Fragmentos de Péptidos/fisiología , Linfocitos T/inmunología , Tenascina/fisiología , Citocinas/biosíntesis , Citotoxicidad Inmunológica/genética , Citotoxicidad Inmunológica/inmunología , Regulación hacia Abajo/genética , Regulación hacia Abajo/inmunología , Fibronectinas/genética , Humanos , Activación de Linfocitos/genética , Fragmentos de Péptidos/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Estructura Terciaria de Proteína/genética , Complejo Receptor-CD3 del Antígeno de Linfocito T/antagonistas & inhibidores , Complejo Receptor-CD3 del Antígeno de Linfocito T/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Secuencias Repetitivas de Aminoácido/genética , Secuencias Repetitivas de Aminoácido/inmunología , Linfocitos T/metabolismo , Tenascina/genética , Células Tumorales Cultivadas
18.
Nat Med ; 7(9): 1041-7, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11533708

RESUMEN

Cyclooxygenase-2 (COX-2), a key enzyme in arachidonic acid metabolism, is overexpressed in many cancers. Inhibition of COX-2 by nonsteroidal anti-inflammatory drugs (NSAIDs) reduces the risk of cancer development in humans and suppresses tumor growth in animal models. The anti-cancer effect of NSAIDs seems to involve suppression of tumor angiogenesis, but the underlying mechanism is not completely understood. Integrin alpha V beta 3 is an adhesion receptor critically involved in mediating tumor angiogenesis. Here we show that inhibition of endothelial-cell COX-2 by NSAIDs suppresses alpha V beta 3-dependent activation of the small GTPases Cdc42 and Rac, resulting in inhibition of endothelial-cell spreading and migration in vitro and suppression of fibroblast growth factor-2-induced angiogenesis in vivo. These results establish a novel functional link between COX-2, integrin alpha V beta 3 and Cdc42-/Rac-dependent endothelial-cell migration. Moreover, they provide a rationale to the understanding of the anti-angiogenic activity of NSAIDs.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Endotelio Vascular/citología , Receptores de Vitronectina/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Animales , División Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa/farmacología , Dinoprostona/farmacología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Humanos , Isoenzimas/antagonistas & inhibidores , Proteínas de la Membrana , Ratones , Ratones Desnudos , Neovascularización Fisiológica/efectos de los fármacos , Nitrobencenos/farmacología , Prostaglandina-Endoperóxido Sintasas , Receptores de Vitronectina/antagonistas & inhibidores , Sulfonamidas/farmacología , Tromboxano A2/farmacología , Proteína de Unión al GTP cdc42/efectos de los fármacos , Proteínas de Unión al GTP rac/efectos de los fármacos
19.
Curr Biol ; 11(13): 1028-38, 2001 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-11470407

RESUMEN

BACKGROUND: Thy-1 is an abundant neuronal glycoprotein in mammals. Despite such prevalence, Thy-1 function remains largely obscure in the absence of a defined ligand. Astrocytes, ubiquitous cells of the brain, express a putative Thy-1 ligand that prevents neurite outgrowth. In this paper, a ligand molecule for Thy-1 was identified, and the consequences of Thy-1 binding for astrocyte function were investigated. RESULTS: Thy-1 has been implicated in cell adhesion and, indeed, all known Thy-1 sequences were found to contain an integrin binding, RGD-like sequence. Thy-1 interaction with beta3 integrin on astrocytes was demonstrated in an adhesion assay using a thymoma line (EL-4) expressing high levels of Thy-1. EL-4 cells bound to astrocytes five times more readily than EL-4(-f), control cells lacking Thy-1. Binding was blocked by either anti-Thy-1 or anti-beta3 antibodies, by RGD-related peptides, or by soluble Thy-1-Fc chimeras. However, neither RGE/RLE peptides nor Thy-1(RLE)-Fc fusion protein inhibited the interaction. Immobilized Thy-1-Fc, but not Thy-1(RLE)-Fc fusion protein supported the attachment and spreading of astrocytes in a Mn(2+)-dependent manner. Binding to Thy-1-Fc was inhibited by RGD peptides. Moreover, vitronectin, fibrinogen, denatured collagen (dcollagen), and a kistrin-derived peptide, but not fibronectin, also mediated Mn(2+)-dependent adhesion, suggesting the involvement of beta3 integrin. The addition of Thy-1 to matrix-bound astrocytes induced recruitment of paxillin, vinculin, and focal adhesion kinase (FAK) to focal contacts and increased tyrosine phosphorylation of proteins such as p130(Cas) and FAK. Furthermore, astrocyte binding to immobilized Thy-1-Fc alone was sufficient to promote focal adhesion formation and phosphorylation on tyrosine. CONCLUSIONS: Thy-1 binds to beta3 integrin and triggers tyrosine phosphorylation of focal adhesion proteins in astrocytes, thereby promoting focal adhesion formation, cell attachment, and spreading.


Asunto(s)
Antígenos CD/metabolismo , Astrocitos/metabolismo , Adhesiones Focales/metabolismo , Glicoproteínas de Membrana Plaquetaria/metabolismo , Antígenos Thy-1/metabolismo , Antígenos Thy-1/fisiología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/inmunología , Antígenos CD/inmunología , Encéfalo/metabolismo , Adhesión Celular , Células Cultivadas , Proteínas del Citoesqueleto/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Integrina beta3 , Ratones , Datos de Secuencia Molecular , Neuronas/metabolismo , Oligopéptidos/farmacología , Fosfotirosina/metabolismo , Glicoproteínas de Membrana Plaquetaria/inmunología , Ratas , Homología de Secuencia de Aminoácido , Transducción de Señal , Antígenos Thy-1/química , Células Tumorales Cultivadas
20.
Drug Metab Dispos ; 29(5): 748-53, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11302943

RESUMEN

Cytochrome P450 inhibition studies are performed in the pharmaceutical industry in the discovery stage to screen candidates that may have the potential for clinical drug-drug interactions. A 96-well microtiter plate assay using recombinant cytochrome P450 (Supersomes) has been used to increase the overall throughput. The IC(50) values for the inhibition of CYP3A4 by 52 new chemical entities (NCEs) were determined using the Supersomes assay with resorufin benzyl ether as a substrate, and the data were compared with those obtained in human liver microsomes (HLM) using midazolam as a substrate. Among the 52 compounds tested, 25 showed IC(50) values within a 5-fold difference in the two assays. For all compounds that showed a >5-fold difference, the IC(50) values in the Supersomes assay were lower than those obtained in HLM, except for one compound. Further studies suggested that this discrepancy was not related to difference in protein concentrations between the two assays. In addition, the IC(50) values for 16 compounds with a wide range of inhibition potency were determined in HLM using testosterone and dextromethorphan as substrates. The results showed an 80 to 93% match within a 5-fold difference between the three probe substrates. However, for certain compounds including ketoconazole, there were substrate-dependent differences in the inhibition. The results suggest that the difference between the Supersomes and HLM could be partially attributed to differences in the substrate used, and to metabolism by other cytochrome P450s present in the HLM but not in the Supersomes. Furthermore, multiple CYP3A4 substrates should be used to improve the reliability of estimating potential drug-drug interaction of NCEs.


Asunto(s)
Inhibidores Enzimáticos del Citocromo P-450 , Microsomas Hepáticos/enzimología , Oxigenasas de Función Mixta/antagonistas & inhibidores , Citocromo P-450 CYP3A , Sistema Enzimático del Citocromo P-450/metabolismo , Humanos , Oxigenasas de Función Mixta/metabolismo , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA