Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
2.
Oncogene ; 38(7): 1050-1066, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30194450

RESUMEN

Vascular endothelial growth factor-A (VEGF-A) is highly subjected to alternative pre-mRNA splicing that generates several splice variants. The VEGFxxx and VEGFxxxb families encode splice variants of VEGF-A that differ only at the level of six amino acids in their C-terminal part. The expression level of VEGFxxx splice variants and their function as pro-angiogenic factors during tumor neo-angiogenesis have been well-described. The role of VEGFxxxb isoforms is less well known, but they have been shown to inhibit VEGFxxx-mediated angiogenesis, while being partial or weak activators of VEGFR receptors in endothelial cells. On the opposite, their role on tumor cells expressing VEGFRs at their surface remains largely unknown. In this study, we find elevated levels of VEGF165b, the main VEGFxxxb isoform, in 36% of non-small cell lung carcinoma (NSCLC), mainly lung adenocarcinoma (46%), and show that a high VEGF165b/VEGF165 ratio correlates with the presence of lymph node metastases. At the molecular level, we demonstrate that VEGF165b stimulates proliferation and invasiveness of two lung tumor cell lines through a VEGFR/ß1 integrin loop. We further provide evidence that the isoform-specific knockdown of VEGF165b reduces tumor growth, demonstrating a tumor-promoting autocrine role for VEGF165b in lung cancer cells. Importantly, we show that bevacizumab, an anti-angiogenic compound used for the treatment of lung adenocarcinoma patients, increases the expression of VEGF165b and activates the invasive VEGFR/ß1 integrin loop. Overall, these data highlight an unexpected role of the VEGF165b splice variant in the progression of lung tumors and their response to anti-angiogenic therapies.


Asunto(s)
Empalme Alternativo , Comunicación Autocrina/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Integrina beta1/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas de Neoplasias/metabolismo , Neovascularización Patológica/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Inhibidores de la Angiogénesis/farmacología , Bevacizumab/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Femenino , Humanos , Integrina beta1/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Proteínas de Neoplasias/genética , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/genética , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/genética
3.
Oncotarget ; 6(16): 14669-86, 2015 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-26036640

RESUMEN

We previously identified 1-(2,4-dimethoxyphenyl)-3-(1-methylindolyl) propenone (IPP51), a new chalcone derivative that is capable of inducing prometaphase arrest and subsequent apoptosis of bladder cancer cells. Here, we demonstrate that IPP51 selectively inhibits proliferation of tumor-derived cells versus normal non-tumor cells. IPP51 interfered with spindle formation and mitotic chromosome alignment. Accumulation of cyclin B1 and mitotic checkpoint proteins Bub1 and BubR1 on chromosomes in IPP51 treated cells indicated the activation of spindle-assembly checkpoint, which is consistent with the mitotic arrest. The antimitotic actions of other chalcones are often associated with microtubule disruption. Indeed, IPP51 inhibited tubulin polymerization in an in vitro assay with purified tubulin. In cells, IPP51 induced an increase in soluble tubulin. Furthermore, IPP51 inhibited in vitro capillary-like tube formation by endothelial cells, indicating that it has anti-angiogenic activity. Molecular docking showed that the indol group of IPP51 can be accommodated in the colchicine binding site of tubulin. This characteristic was confirmed by an in vitro competition assay demonstrating that IPP51 can compete for colchicine binding to soluble tubulin. Finally, in a human bladder xenograft mouse model, IPP51 inhibited tumor growth without signs of toxicity. Altogether, these findings suggest that IPP51 is an attractive new microtubule-targeting agent with potential chemotherapeutic value.


Asunto(s)
Microtúbulos/genética , Neoplasias de la Vejiga Urinaria/genética , Animales , Proliferación Celular , Humanos , Ratones , Microtúbulos/metabolismo , Simulación del Acoplamiento Molecular , Estructura Molecular , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/patología , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Oncotarget ; 6(13): 11421-33, 2015 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-25823656

RESUMEN

Adrenal carcinoma (ACC) is a rare neoplasm with a poor outcome. Aberrant expression of ß-catenin has been found in approximatively 30% of ACC. We herein studied its effects on the growth of the human ACC cell line H295R. The cells were infected with short hairpin RNA (shRNA)-mediated silencing ß-catenin. Two shRNAs used induced down-regulation of ß-catenin protein levels. The expression of these shRNAs decreased cell growth and increased H295R cells in S and G2/M phases. This cytostatic effect is due to a decrease of phosphorylated MAPK and to an up-regulation expression of the cyclin-dependent kinase inhibitors p57(KIP2), p21(CIP) and p27(KIP1). In addition, the knockdown of ß-catenin decreased phosphorylated Akt and increased apoptosis. Finally, loss of ß-catenin was sufficient to induce the reversal of the epithelial-to-mesenchymal transition. We then transplanted these genetically modified H295R cells in Scid mice. Tumor growth suppression was achieved by the two shRNAs showing in vitro efficacy. Proliferation was not reduced in silenced tumors. In contrast, p57, p27 and p21 proteins were found expressed at high levels in silenced tumors along with an increase in apoptotic cells. These findings indicate that ß-catenin loss in H295R cells inhibits tumor growth by inducing transcriptional and functional changes.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/metabolismo , Carcinoma Corticosuprarrenal/metabolismo , Proliferación Celular , Transición Epitelial-Mesenquimal , beta Catenina/metabolismo , Neoplasias de la Corteza Suprarrenal/genética , Neoplasias de la Corteza Suprarrenal/patología , Neoplasias de la Corteza Suprarrenal/terapia , Carcinoma Corticosuprarrenal/genética , Carcinoma Corticosuprarrenal/patología , Carcinoma Corticosuprarrenal/terapia , Animales , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación hacia Abajo , Puntos de Control de la Fase G2 del Ciclo Celular , Regulación Neoplásica de la Expresión Génica , Genotipo , Humanos , Ratones SCID , Fenotipo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Tratamiento con ARN de Interferencia , Puntos de Control de la Fase S del Ciclo Celular , Transducción de Señal , Factores de Tiempo , Transfección , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto , beta Catenina/genética
5.
Oncotarget ; 6(7): 5382-411, 2015 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-25742784

RESUMEN

The efficacy of anti-angiogenic therapies on cancer patients is limited by the emergence of drug resistance, urging the search for second-generation drugs. In this study, we screened an academic chemical library (DCM, University of Grenoble-Alpes) and identified a leader molecule, COB223, that inhibits endothelial cell migration and proliferation. It inhibits also Lewis lung carcinoma (LLC/2) cell proliferation whereas it does not affect fibroblast proliferation. The anti-angiogenic activity of COB223 was confirmed using several in vitro and in vivo assays. In a mouse LLC/2 tumor model, ip administration of doses as low as 4 mg/kg COB223 efficiently reduced the tumor growth rate. We observed that COB223 inhibits endothelial cell ERK1/2 phosphorylation induced by VEGF, FGF-2 or serum and that it acts downstream of PKC and upstream of Ras. This molecule represents a novel anti-angiogenic and anti-tumorigenic agent with an original mechanism of action that deserves further development as an anti-cancer drug.


Asunto(s)
Antineoplásicos/farmacología , Carbamatos/farmacología , Carcinoma Pulmonar de Lewis/prevención & control , Transformación Celular Neoplásica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neovascularización Patológica/prevención & control , Sulfonamidas/farmacología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Proteínas ras/metabolismo , Animales , Antineoplásicos/síntesis química , Apoptosis/efectos de los fármacos , Western Blotting , Carbamatos/síntesis química , Carcinoma Pulmonar de Lewis/irrigación sanguínea , Carcinoma Pulmonar de Lewis/patología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Ensayos Analíticos de Alto Rendimiento , Humanos , Técnicas para Inmunoenzimas , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neovascularización Patológica/patología , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Piel/citología , Piel/efectos de los fármacos , Piel/metabolismo , Sulfonamidas/síntesis química , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Eur J Pharm Biopharm ; 88(3): 683-94, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25204521

RESUMEN

In a previous study, we reported on the formulation of Artemisinin-loaded surface-decorated nanoparticles (nanospheres and nanoreservoirs) by co-nanoprecipitation of PEG derivatives (PEG1500 and PEG4000-stearate, polysorbate 80) and biosynthesized γ-CD fatty esters. In the present study, the co-nanoprecipitation was extended to the use of a PEGylated phospholipid, namely DMPE-PEG2000. As our goal was to prepare long-circulating nanocarriers for further systemic delivery of Artemisinin (ART), here, we have investigated, on the one hand, the in vitro behavior of these surface-modified γ-CD-C10 particles toward the immune system (complement activation and macrophage uptake assays) and, on the other hand, their biodistribution features in mice. These experiments showed that the in vitro plasma protein adsorption and phagocytosis by macrophage cells triggered by γ-CD-C10 nanoparticles were significantly reduced when their surface was decorated with amphiphilic PEGylated molecules, in particular PEG1500-stearate, DMPE-mPEG2000 or polysorbate 80. The prolonged blood circulation time assessed by fluorescence imaging was demonstrated for unloaded γ-CD-C10-based nanospheres and nanoreservoir particles containing DMPE-PEG2000 and polysorbate80, respectively. These nanoparticles also proved to be non-hemolytic at the concentration range used in vivo. Within the limits of the conducted experiments, the co-nanoprecipitation technique may be considered as an alternative for surface modification of amphiphilic CD-based drug delivery systems and may be applied to the systemic delivery of ART.


Asunto(s)
Antiinfecciosos/administración & dosificación , Artemisininas/administración & dosificación , Ciclodextrinas/química , Portadores de Fármacos/química , Sistema Inmunológico/efectos de los fármacos , Nanopartículas/química , Animales , Antiinfecciosos/sangre , Antiinfecciosos/farmacocinética , Artemisininas/sangre , Artemisininas/farmacocinética , Línea Celular , Activación de Complemento/efectos de los fármacos , Activación de Complemento/inmunología , Estabilidad de Medicamentos , Eritrocitos/efectos de los fármacos , Hemólisis/efectos de los fármacos , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones Desnudos , Tamaño de la Partícula , Fagocitosis/efectos de los fármacos , Fagocitosis/inmunología , Ovinos , Propiedades de Superficie , Distribución Tisular
7.
J Cell Biol ; 202(3): 545-61, 2013 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-23918940

RESUMEN

The endothelial CCM complex regulates blood vessel stability and permeability. Loss-of-function mutations in CCM genes are responsible for human cerebral cavernous malformations (CCMs), which are characterized by clusters of hemorrhagic dilated capillaries composed of endothelium lacking mural cells and altered sub-endothelial extracellular matrix (ECM). Association of the CCM1/2 complex with ICAP-1, an inhibitor of ß1 integrin, prompted us to investigate whether the CCM complex interferes with integrin signaling. We demonstrate that CCM1/2 loss resulted in ICAP-1 destabilization, which increased ß1 integrin activation and led to increased RhoA-dependent contractility. The resulting abnormal distribution of forces led to aberrant ECM remodeling around lesions of CCM1- and CCM2-deficient mice. ICAP-1-deficient vessels displayed similar defects. We demonstrate that a positive feedback loop between the aberrant ECM and internal cellular tension led to decreased endothelial barrier function. Our data support that up-regulation of ß1 integrin activation participates in the progression of CCM lesions by destabilizing intercellular junctions through increased cell contractility and aberrant ECM remodeling.


Asunto(s)
Fibronectinas/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Integrina beta1/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Adhesión Celular , Células Cultivadas , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Proteína KRIT1 , Ratones , Ratones Endogámicos , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/deficiencia , Modelos Biológicos , Proteínas Proto-Oncogénicas/deficiencia
8.
J Biomed Opt ; 18(10): 101311, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23900442

RESUMEN

A new contrast agent, LipImage™ 815, has been designed and compared to previously described indocyanine green (ICG)-loaded lipid nanoparticles (ICG-lipidots®). Both contrast agents display similar size (50-nm diameter), zeta potential, high IC50 in cellular studies, near-infrared absorption and emission wavelengths in the "imaging window," long-term shelf colloidal and optical stabilities with high brightness (>106 L mol-1 cm-1) in ready-to-use storage conditions in aqueous buffer (4°C in dark), therefore being promising fluorescence contrast agents for in vivo imaging. However, while ICG-lipidots® display a relatively short plasma lifetime, LipImage™ 815 circulates in blood for longer times, allowing the efficient uptake of fluorescence signal in human prostate cancer cells implanted in mice. Prolonged tumor labeling is observed for more than 21 days.


Asunto(s)
Colorantes Fluorescentes/química , Lípidos/química , Nanopartículas/química , Imagen Óptica/métodos , Espectroscopía Infrarroja Corta/métodos , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Femenino , Colorantes Fluorescentes/farmacocinética , Colorantes Fluorescentes/toxicidad , Humanos , Lípidos/farmacocinética , Masculino , Ratones , Ratones Desnudos , Tamaño de la Partícula , Distribución Tisular
9.
Stem Cell Res Ther ; 4(2): 41, 2013 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-23628074

RESUMEN

INTRODUCTION: Understanding the multiple biological functions played by human mesenchymal stem cells (hMSCs) as well as their development as therapeutics in regenerative medicine or in cancer treatment are major fields of research. Indeed, it has been established that hMSCs play a central role in the pathogenesis and progression of tumours, but their impact on tumour growth remains controversial. METHODS: In this study, we investigated the influence of hMSCs on the growth of pre-established tumours. We engrafted nude mice with luciferase-positive mouse adenocarcinoma cells (TSA-Luc+) to obtain subcutaneous or lung tumours. When tumour presence was confirmed by non-invasive bioluminescence imaging, hMSCs were injected into the periphery of the SC tumours or delivered by systemic intravenous injection in mice bearing either SC tumours or lung metastasis. RESULTS: Regardless of the tumour model and mode of hMSC injection, hMSC administration was always associated with decreased tumour growth due to an inhibition of tumour cell proliferation, likely resulting from deep modifications of the tumour angiogenesis. Indeed, we established that although hMSCs can induce the formation of new blood vessels in a non-tumoural cellulose sponge model in mice, they do not modify the overall amount of haemoglobin delivered into the SC tumours or lung metastasis. We observed that these tumour vessels were reduced in number but were longer. CONCLUSIONS: Our results suggest that hMSCs injection decreased solid tumour growth in mice and modified tumour vasculature, which confirms hMSCs could be interesting to use for the treatment of pre-established tumours.


Asunto(s)
Neoplasias Pulmonares/patología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Neovascularización Fisiológica , Actinas/metabolismo , Animales , Células de la Médula Ósea/citología , Línea Celular Tumoral , Humanos , Inyecciones Intravenosas , Inyecciones Subcutáneas , Antígeno Ki-67/metabolismo , Neoplasias Pulmonares/metabolismo , Imagen por Resonancia Magnética , Ratones , Ratones Desnudos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo
10.
Mol Imaging Biol ; 15(3): 239-44, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23054555

RESUMEN

PURPOSE: There is a real need to adapt simple and reproducible imaging methodologies to evaluate noninvasively pro- and antiangiogenic activities of new treatments in a physiological context in mice. PROCEDURE: The angiogenic response to fibroblast growth factor 2 (FGF-2) in a model of subcutaneously implanted cellulose sponges was measured in parallel after an intravenous injection of a fluorescent αvß3 integrin-targeting molecule (Angiolone(TM)) and an fluorescence diffuse optical tomography optical imaging system and by measuring the hemoglobin content in the sponges. RESULTS: Optical measurements of angiogenesis correlated perfectly with the values obtained using hemoglobin quantification. This assay can be used to follow the activity of a pro- or antiangiogenic treatment like demonstrated after FGF-2 or angiostatin, respectively. CONCLUSION: The perfectly controlled quality of cellulose sponges combined to this noninvasive optical method allow rapid, accurate, and reproducible measurements of angiogenic activities in vivo at the preclinical level.


Asunto(s)
Imagen Molecular/métodos , Neovascularización Patológica/patología , Neovascularización Fisiológica/efectos de los fármacos , Oligopéptidos/farmacología , Tejido Subcutáneo/irrigación sanguínea , Tapones Quirúrgicos de Gaza , Inhibidores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/uso terapéutico , Angiostatinas/farmacología , Angiostatinas/uso terapéutico , Animales , Femenino , Factor 2 de Crecimiento de Fibroblastos/farmacología , Fluorescencia , Ratones , Ratones Desnudos , Neovascularización Patológica/tratamiento farmacológico , Tejido Subcutáneo/efectos de los fármacos
11.
J Biomed Opt ; 17(10): 106014, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23052561

RESUMEN

Over the last few years, near-infrared (NIR) fluorescence imaging has witnessed rapid growth and is already used in clinical trials for various procedures. However, most clinically compatible imaging systems are optimized for large, open-surgery procedures. Such systems cannot be employed during head and neck oncologic surgeries because the system is not able to image inside deep cavities or allow the surgeon access to certain tumors due to the large footprint of the system. We describe a miniaturized, low-cost, NIR fluorescence system optimized for clinical use during oral oncologic surgeries. The system, termed FluoSTIC, employs a miniature, high-quality, consumer-grade lipstick camera for collecting fluorescence light and a novel custom circular optical fiber array for illumination that combines both white light and NIR excitation. FluoSTIC maintains fluorescence imaging quality similar to that of current large-size imaging systems and is 22 mm in diameter and 200 mm in height and weighs less than 200 g.


Asunto(s)
Miniaturización/instrumentación , Espectrometría de Fluorescencia/instrumentación , Cirugía Asistida por Computador/instrumentación , Animales , Femenino , Tecnología de Fibra Óptica/instrumentación , Humanos , Neoplasias Hepáticas/patología , Ratones , Ratones Desnudos , Espectrometría de Fluorescencia/métodos , Espectroscopía Infrarroja Corta , Cirugía Asistida por Computador/métodos
12.
Biomaterials ; 32(7): 1978-85, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21168204

RESUMEN

Polyethyleneimine (PEI) is a cationic polymer that is effective in gene delivery in vivo. Plasmid DNA incorporating the Sleeping-Beauty (SB) transposon has been shown to induce long-term transgene expression in mouse lungs after PEI-mediated delivery. In the current report, we followed the reporter gene expression mediated by PEI/SB delivery in lungs of mice using the non-invasive bioluminescent imaging (BLI) technology. After delivery, the reporter gene signal showed a rapid decay in the first two weeks to a nearly undetectable level, but then the signal augmented gradually in the following weeks and finally reached a stable level that maintained until the natural death of animals. The stabilization of transgene expression is associated with the multiplication of a small number of PEI/SB-labeled alveolar cells, which proliferated both under normal conditions and in response to acute local injury for epithelia repair, and may play a role in long-term homeostatic maintenance in alveoli. The data presented here suggests that systemic delivery of PEI/SB induces stable transfection specifically in a small population of alveolar progenitor cells. The technique provides a promising platform for future research in distal lung biology and tissue regenerative therapy.


Asunto(s)
Elementos Transponibles de ADN/genética , Diagnóstico por Imagen/métodos , Genes Reporteros/genética , Pulmón/metabolismo , Polietileneimina/química , Animales , Femenino , Técnicas de Transferencia de Gen , Ratones , Ratones SCID
13.
Exp Cell Res ; 315(19): 3406-18, 2009 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19769963

RESUMEN

In embryogenesis, coronary blood vessels are formed by vasculogenesis from epicardium-derived progenitors. Subsequently, growing or regenerating myocardium increases its vasculature by angiogenesis, forming new vessels from the pre-existing ones. Recently, cell therapies for myocardium ischemia that used different protocols have given promising results, using either extra-cardiac blood vessel cell progenitors or stimulating the cardiac angiogenesis. We have questioned whether cardiomyocytes could sustain both vasculogenesis and angiogenesis. We used a 3D culture model of tissue-like spheroids in co-cultures of cardiomyocytes supplemented either with endothelial cells or with bone marrow-derived mesenchymal stroma cells. Murine foetal cardiomyocytes introduced into non-adherent U-wells formed 3D contractile structures. They were coupled by gap junctions. Cardiomyocytes segregated inside the 3D structure into clumps separated by connective tissue septa, rich in fibronectin. Three vascular endothelial growth factor isoforms were produced (VEGF 120, 164 and 188). When co-cultured with human umbilical cord endothelial cells, vascular structures were produced in fibronectin-rich external layer and in radial septa, followed by angiogenic sprouting into the cardiomyocyte microtissue. Presence of vascular structures led to the maintenance of long-term survival and contractile capacity of cardiac microtissues. Conversely, bone marrow mesenchymal cells formed isolated cell aggregates, which progressively expressed the endothelial markers von Willebrand's antigen and CD31. They proceeded to typical vasculogenesis forming new blood vessels organised in radial pattern. Our results indicate that the in vitro 3D model of cardiomyocyte spheroids provides the two basic elements for formation of new blood vessels: fibronectin and VEGF. Within the myocardial environment, endothelial and mesenchymal cells can proceed to formation of new blood vessels either through angiogenesis or vasculogenesis, respectively.


Asunto(s)
Vasos Coronarios/fisiología , Células Endoteliales/citología , Células Madre Mesenquimatosas/citología , Miocitos Cardíacos/citología , Neovascularización Fisiológica , Animales , Vasos Sanguíneos/crecimiento & desarrollo , Técnicas de Cocultivo , Fibronectinas/biosíntesis , Ratones , Factor A de Crecimiento Endotelial Vascular/biosíntesis
14.
Antimicrob Agents Chemother ; 53(11): 4694-701, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19738024

RESUMEN

Chagas' disease induced by Trypanosoma cruzi infection is an important cause of mortality and morbidity affecting the cardiovascular system for which presently available therapies are largely inadequate. We previously reported that transforming growth factor beta (TGF-beta) is implicated in several regulatory aspects of T. cruzi invasion and growth and in host tissue fibrosis. This prompted us to evaluate the therapeutic action of an inhibitor of TGF-beta signaling (SB-431542) administered during the acute phase of experimental Chagas' disease. Male Swiss mice were infected intraperitoneally with 10(4) trypomastigotes of T. cruzi (Y strain) and evaluated clinically for the following 30 days. SB-431542 treatment significantly reduced mortality and decreased parasitemia. Electrocardiography showed that SB-431542 treatment was effective in protecting the cardiac conduction system. By 14 day postinfection, enzymatic biomarkers of tissue damage indicated that muscle injury was decreased by SB-431542 treatment, with significantly lower blood levels of aspartate aminotransferase and creatine kinase. In conclusion, inhibition of TGF-beta signaling in vivo appears to potently decrease T. cruzi infection and to prevent heart damage in a preclinical mouse model. This suggests that this class of molecules may represent a new therapeutic agent for acute and chronic Chagas' disease that warrants further clinical exploration.


Asunto(s)
Benzamidas/uso terapéutico , Cardiomiopatía Chagásica/prevención & control , Enfermedad de Chagas/tratamiento farmacológico , Dioxoles/uso terapéutico , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Animales , Bradicardia/prevención & control , Masculino , Ratones , Miocardio/patología , Parasitemia/tratamiento farmacológico , Proteínas Serina-Treonina Quinasas/fisiología , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/fisiología
15.
Cytokine Growth Factor Rev ; 19(5-6): 405-13, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18938102

RESUMEN

This paper summarizes recent data from the literature suggesting that transforming growth factor-beta (TGF-beta) participates at least in four different processes influencing development of myocardiopathy in Chagas disease, a major parasitic illness caused by Trypanosoma cruzi infection: (a) invasion of cardiac fibroblasts and myocytes; (b) intracellular parasite cycle; (c) regulation of inflammation and immune response; (d) fibrosis and heart remodeling during acute and chronic disease. All these effects point to an important role of TGF-beta in Chagas disease myocardiopathy and suggest that monitoring the circulating levels of this cytokine could be of help in clinical prognosis and management of patients. Moreover, TGF-beta-interfering therapies appear as interesting adjuvant interventions during acute and chronic phases of T. cruzi infection.


Asunto(s)
Cardiomiopatía Chagásica/etiología , Enfermedad de Chagas/inmunología , Factor de Crecimiento Transformador beta/fisiología , Trypanosoma cruzi/fisiología , Animales , Cardiomiopatía Chagásica/inmunología , Interacciones Huésped-Parásitos/fisiología , Humanos
16.
Circ Res ; 102(8): 914-22, 2008 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-18309101

RESUMEN

Angiogenesis is a complex process, requiring a finely tuned balance between numerous stimulatory and inhibitory signals. ALK1 (activin receptor like-kinase 1) is an endothelial-specific type 1 receptor of the transforming growth factor-beta receptor family. Heterozygotes with mutations in the ALK1 gene develop hereditary hemorrhagic telangiectasia type 2 (HHT2). Recently, we reported that bone morphogenetic protein (BMP)9 and BMP10 are specific ligands for ALK1 that potently inhibit microvascular endothelial cell migration and growth. These data lead us to suggest that these factors may play a role in the control of vascular quiescence. To test this hypothesis, we checked their presence in human serum. We found that human serum induced Smad1/5 phosphorylation. To identify the active factor, we tested neutralizing antibodies against BMP members and found that only the anti-BMP9 inhibited serum-induced Smad1/5 phosphorylation. The concentration of circulating BMP9 was found to vary between 2 and 12 ng/mL in sera and plasma from healthy humans, a value well above its EC(50) (50 pg/mL). These data indicated that BMP9 is circulating at a biologically active concentration. We then tested the effects of BMP9 in 2 in vivo angiogenic assays. We found that BMP9 strongly inhibited sprouting angiogenesis in the mouse sponge angiogenesis assay and that BMP9 could inhibit blood circulation in the chicken chorioallantoic membrane assay. Taken together, our results demonstrate that BMP9, circulating under a biologically active form, is a potent antiangiogenic factor that is likely to play a physiological role in the control of adult blood vessel quiescence.


Asunto(s)
Receptores de Activinas Tipo II/fisiología , Proteínas Morfogenéticas Óseas/fisiología , Neovascularización Fisiológica , Células 3T3 , Receptores de Activinas Tipo II/genética , Adulto , Proteínas Angiogénicas , Animales , Proteínas Morfogenéticas Óseas/sangre , Estudios de Casos y Controles , Embrión de Pollo , Femenino , Factor 2 de Diferenciación de Crecimiento , Factores de Diferenciación de Crecimiento , Humanos , Masculino , Ratones , Persona de Mediana Edad , Proteínas Smad/metabolismo , Telangiectasia Hemorrágica Hereditaria/sangre , Telangiectasia Hemorrágica Hereditaria/genética , Transfección
17.
J Endocrinol ; 196(3): 473-82, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18310443

RESUMEN

Endocrine gland-derived vascular endothelial growth factor (EG-VEGF) and its homolog Bombina variegata (Bv8), also termed prokineticin-1 and -2 (PK1 and PK2) respectively, are newly identified peptides with specific mitogenic activity on endocrine gland-derived endothelial cells. In the present study, we analyzed the sites of expression of EG-VEGF/PK1, Bv8/PK2, and their receptors (PKR1 and PKR2) in the adrenal cortex and checked for new biological functions of these factors on the endocrine cell compartment. RT-PCR and immunostaining analyses revealed that glomerulosa and fasciculata cells express both factors and both receptors. EG-VEGF/PK1 had no effect on the steroidogenic activity of both bovine glomerulosa and fasciculata cells but appeared to be mitogenic for both cell types. Binding of EG-VEGF/PK1 to fasciculata cells stimulated the phosphorylation of ERK1/2. Pretreatment with pertussis toxin suppressed this effect, indicating that it was Gi mediated. EG-VEGF/PK1 also increased the phosphorylation of Akt in endocrine cells of the adrenal cortex. EG-VEGF/PK1 and Bv8/PK2 thus represent new regulatory peptides acting as autocrine mitogens for endocrine cells.


Asunto(s)
Corteza Suprarrenal/metabolismo , Hormonas Gastrointestinales/metabolismo , Sustancias de Crecimiento/metabolismo , Neuropéptidos/metabolismo , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/metabolismo , Corteza Suprarrenal/citología , Hormona Adrenocorticotrópica/farmacología , Angiotensina II/farmacología , Animales , Bovinos , División Celular/efectos de los fármacos , División Celular/fisiología , Células Cultivadas , Hormonas Gastrointestinales/genética , Hormonas Gastrointestinales/farmacología , Sustancias de Crecimiento/genética , Sustancias de Crecimiento/farmacología , Inmunohistoquímica , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neuropéptidos/genética , Neuropéptidos/farmacología , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropéptido/genética , Receptores de Neuropéptido/metabolismo , Proteínas Recombinantes/farmacología , Esteroides/metabolismo , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/genética , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/farmacología
18.
Antimicrob Agents Chemother ; 51(8): 2905-10, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17526757

RESUMEN

The antiinflammatory cytokine transforming growth factor beta (TGF-beta) plays an important role in Chagas disease, a parasitic infection caused by the protozoan Trypanosoma cruzi. In the present study, we show that SB-431542, an inhibitor of the TGF-beta type I receptor (ALK5), inhibits T. cruzi-induced activation of the TGF-beta pathway in epithelial cells and in cardiomyocytes. Further, we demonstrate that addition of SB-431542 greatly reduces cardiomyocyte invasion by T. cruzi. Finally, SB-431542 treatment significantly reduces the number of parasites per infected cell and trypomastigote differentiation and release. Taken together, these data further confirm the major role of the TGF-beta signaling pathway in both T. cruzi infection and T. cruzi cell cycle completion. Our present data demonstrate that small inhibitors of the TGF-beta signaling pathway might be potential pharmacological tools for the treatment of Chagas disease.


Asunto(s)
Benzamidas/farmacología , Dioxoles/farmacología , Miocitos Cardíacos/parasitología , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/efectos de los fármacos , Trypanosoma cruzi/patogenicidad , Animales , Apoptosis , Ciclo Celular/efectos de los fármacos , Células Cultivadas , Enfermedad de Chagas , Chlorocebus aethiops , Células Epiteliales/parasitología , Ratones , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Trypanosoma cruzi/citología , Trypanosoma cruzi/efectos de los fármacos , Trypanosoma cruzi/crecimiento & desarrollo , Células Vero
19.
Am J Pathol ; 167(4): 993-1003, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16192635

RESUMEN

The cytokine transforming growth factor-beta (TGF-beta) plays various functions in the control of Trypanosoma cruzi infectivity and in the progression of Chagas' disease. When we immunostained T. cruzi-infected cardiomyocytes (after either in vivo or in vitro infections) for TGF-beta, we observed stronger immunoreactivity in parasites than in host cells. TGF-beta immunoreactivity evolved during parasite cycle progression, with intense staining in amastigotes versus very faint staining in trypomastigotes. TGF-beta was present on the surface of amastigotes, in the flagellar pocket, and in intraparasitic vesicles as revealed by electron microscopy. However, no ortholog TGF-beta gene could be identified in the genome of T. cruzi by in silico analysis or by extensive polymerase chain reaction and reverse transcriptase-polymerase chain reaction studies. Immunoreactive TGF-beta was most probably taken up by the parasite from the host cell cytoplasm because such an internalization process of biotinylated TGF-beta could be observed in axenic amastigotes in vitro. These observations represent the first example of a novel mechanism by which a primitive unicellular protozoan can use host cell TGF-beta to control its own intracellular life cycle.


Asunto(s)
Estadios del Ciclo de Vida , Miocitos Cardíacos/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Trypanosoma cruzi/crecimiento & desarrollo , Trypanosoma cruzi/parasitología , Actinas/metabolismo , Animales , Células Cultivadas , Enfermedad de Chagas/metabolismo , Embrión de Mamíferos , Embrión no Mamífero , Fluoresceína-5-Isotiocianato , Técnica del Anticuerpo Fluorescente Indirecta , Colorantes Fluorescentes , Inmunohistoquímica , Indoles , Ratones , Microscopía Confocal , Miocitos Cardíacos/parasitología , Faloidina/metabolismo , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Rodaminas , Factores de Tiempo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/inmunología , Trypanosoma cruzi/ultraestructura
20.
Am J Ophthalmol ; 139(4): 589-96, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15808152

RESUMEN

PURPOSE: Vascular endothelial growth factor (VEGF) and angiopoietins are key regulators of angiogenesis. The purpose of this study was to measure mRNA levels of these factors and of their receptors in surgically excised subfoveal membranes from patients with age-related macular degeneration (AMD) and to evaluate their relevance as prognostic markers of postsurgical recurrence of choroidal neovascularization (CNV). DESIGN: Prospective observational case series. METHODS: setting: Institutional. study population: In a prospective series of 24 patients (aged 51 to 91 years) with classic CNV of AMD diagnosed less than 6 months previously, 24 subfoveal membranes (one eye per patient) were surgically removed and collected. Thirteen patients underwent treatment for recurrence of CNV within 6 months of surgery. main outcome measures: Four 8-mu sections were prepared from each membrane for immunohistochemical determination of vascular density (CD31 immunostaining). The remaining tissue was used for preparation of total RNA. The levels of VEGF-A, VEGF-R1, VEGF-R2, neuropilin-1, angiopoietin-1, angiopoietin-2, Tie-2, and hypoxanthine phosphoribosyltransferase mRNAs were determined by real-time reverse-transcriptase polymerase chain reaction. RESULTS: Vascular endothelial growth factor, angiopoietin-1, and angiopoietin-2 appeared to be expressed to variable levels in most samples, whereas Tie-2, VEGF-R1, and VEGF-R2 were undetectable. Low levels of VEGF expression correlated with postsurgical recurrence of CNV (P = .07). Angiopoietin-1 and angiopoietin-2 levels did not predict recurrence (P > .1). CONCLUSION: The results indicate that at the time of surgical excision, subfoveal membranes express angiopoietin-1, VEGF, and, to a lesser degree, angiopoietin-2. Because CNV appears to recur less often in membranes expressing high levels of VEGF, we hypothesize that VEGF acts as a stabilizer of neovessels at this stage of the disease.


Asunto(s)
Angiopoyetinas/genética , Fóvea Central/metabolismo , Expresión Génica , Degeneración Macular/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Anciano , Anciano de 80 o más Años , Angiopoyetinas/metabolismo , Biomarcadores/metabolismo , Neovascularización Coroidal/etiología , Neovascularización Coroidal/metabolismo , Neovascularización Coroidal/cirugía , Humanos , Hipoxantina Fosforribosiltransferasa/genética , Degeneración Macular/complicaciones , Degeneración Macular/cirugía , Membranas/metabolismo , Persona de Mediana Edad , Neuropilina-1/genética , Complicaciones Posoperatorias , Estudios Prospectivos , ARN Mensajero/metabolismo , Receptor TIE-2/genética , Recurrencia , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Vitrectomía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA