Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros











Intervalo de año de publicación
1.
Eur J Neurosci ; 24(12): 3333-42, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17229082

RESUMEN

The G protein-coupled pheromone receptor neurons (V1R and V2R) of the vomeronasal organ (VNO) are continually replaced throughout the lifetime of the mouse. Moreover, active signalling of V2Rs via the transient receptor potential 2(TRPC2) channel is necessary for regeneration of receptors, as the TRPC2 null mutant mouse showed a 75% reduction of V2Rs by the age of two months. Here we describe V2R mediated signalling in a neuronal line established from vomeronasal stem cells taken from postnatal female mice. Cells were immunoreactive for Galpha(o) and V2R, whereas V1R and Galpha(i) immunoreactivity could not be detected. Biological ligands (dilute urine and its protein fractions) were found to increase proliferation and survival of these neurons. Dilute mouse urine but not artificial urine also induced ERK, Akt and CREB signalling in a dose dependent way. The volatile fraction of male mouse urine alone was without effect while the fraction containing peptides (> 5 kDa) also stimulated ERK and Akt phosphorylation. The ERK, Akt and CREB phosphorylation response was sensitive to pertussis toxin, confirming the involvement of V2R linked Galpha(o). Dilute mouse urine or its high molecular weight protein fraction increased survival and proliferation of these neurons. Hence, urinary pheromones, which signal important social information via mature neurons, also promote survival and proliferation of their regenerating precursors. These data show that regenerating V2Rs respond to urine and the urinary peptides by activation of the Ras-ERK and PI3-Akt pathways, which appear to be important for vomeronasal neural survival and proliferation.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regeneración Nerviosa/efectos de los fármacos , Neuronas/efectos de los fármacos , Feromonas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Feromonas/metabolismo , Órgano Vomeronasal/citología , Análisis de Varianza , Animales , Animales Recién Nacidos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/genética , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Feromonas/orina , Fosforilación/efectos de los fármacos , Proteínas/farmacología , Proteínas Proto-Oncogénicas c-akt/genética , ARN Mensajero/metabolismo , Receptores de Feromonas/clasificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Transducción de Señal/efectos de los fármacos
2.
Circulation ; 110(10): 1219-25, 2004 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-15337705

RESUMEN

BACKGROUND: Ginseng is a commonly used nutraceutical. Intriguingly, existing literature reports both wound-healing and antitumor effects of ginseng extract through opposing activities on the vascular system. To elucidate this perplexity, we merged a chemical fingerprinting approach with a deconstructional study of the effects of pure molecules from ginseng extract on angiogenesis. METHODS AND RESULTS: A mass spectrometric compositional analysis of American, Chinese and Korean, and Sanqi ginseng revealed distinct "sterol ginsenoside" fingerprints, especially in the ratio between a triol, Rg1, and a diol, Rb1, the 2 most prevalent constituents. Using a Matrigel implant model and reconstituting the extracts using distinct ratios of the 2 ginsenosides, we demonstrate that the dominance of Rg1 leads to angiogenesis, whereas Rb1 exerts an opposing effect. Rg1 also promoted functional neovascularization into a polymer scaffold in vivo and the proliferation of, chemoinvasion of, and tubulogenesis by endothelial cells in vitro, an effect mediated through the expression of nitric oxide synthase and the phosphatidylinositol-3 kinase-->Akt pathway. In contrast, Rb1 inhibited the earliest step in angiogenesis, the chemoinvasion of endothelial cells. CONCLUSIONS: The present study explains, for the first time, the ambiguity about the effects of ginseng in vascular pathophysiology based on the existence of opposing active principles in the extract. We also unraveled a speciogeographic variation impinging on the compositional fingerprint that may modulate the final phenotype. This emphasizes the need for regulations standardizing herbal therapy, currently under the Dietary Supplement and Health Education Act. Furthermore, we propose that Rg1 could be a prototype for a novel group of nonpeptide molecules that can induce therapeutic angiogenesis, such as in wound healing.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/farmacología , Células Endoteliales/efectos de los fármacos , Ginsenósidos/análisis , Panax/química , Fitoterapia , Américas , Inductores de la Angiogénesis/química , Inhibidores de la Angiogénesis/química , Animales , Células Cultivadas/efectos de los fármacos , China , Implantes de Medicamentos , Células Endoteliales/citología , Endotelio Vascular/citología , Inhibidores Enzimáticos/farmacología , Ginsenósidos/antagonistas & inhibidores , Ginsenósidos/farmacología , Humanos , Corea (Geográfico) , Masculino , Ratones , Ratones Endogámicos C57BL , Estructura Molecular , NG-Nitroarginina Metil Éster/farmacología , Neovascularización Patológica/inducido químicamente , Panax/clasificación , Fosfatidilinositol 3-Quinasas/fisiología , Fitoterapia/normas , Transducción de Señal/efectos de los fármacos , Especificidad de la Especie , Espectrometría de Masa por Ionización de Electrospray , Tapones Quirúrgicos de Gaza/efectos adversos , Venas Umbilicales
3.
Mol Pharmacol ; 65(4): 979-85, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15044628

RESUMEN

Investigation of rat recombinant P2X(6) receptors has been limited because of the difficulty in obtaining functional expression in heterologous systems. In this study, we demonstrate glycosylation-dependent regulation of recombinant P2X(6) receptor function and associated conferral of a novel phenotype that is sensitive to the P2X(1) and P2X(3) receptor agonist, alphabeta-methylene ATP. In cells functionally expressing P2X(6) receptors, ATP and alphabeta-methylene ATP evoked slowly desensitizing inward currents (EC(50) values, 0.5 and 0.6 microM, respectively) with slow kinetics of current decay on agonist washout. 2',3'-O-(2,4,6-trinitrophenyl ATP) and iso-pyridoxalphosphate-6-azophenyl-2'-5'-disulfonate were effective antagonists (IC(50) values, 0.8 and 22 microM, respectively); however, suramin was relatively ineffective. Reverse transcription-polymerase chain reaction analysis confirmed the absence of other P2X receptor subunits. Western analysis of membrane fractions from functional and nonfunctional clones confirmed the presence of P2X(6) at the cell membrane but revealed a difference in apparent molecular mass of immunoreactive products ( approximately 70 and approximately 60 kDa, respectively). N-glycosidase F treatment of both functional and nonfunctional receptor cell membranes increased the electrophoretic mobilities of immunoreactive products, with both proteins migrating at approximately 55 kDa, demonstrating an increased level of glycosylation of the P2X(6) receptor in functional compared with nonfunctional cells. This study demonstrates that nonfunctional rat recombinant P2X(6) receptors 1) are expressed on the membrane surface of human embryonic kidney cells and 2) are glycosylated. Expression of the novel functional receptor phenotype is associated with further glycosylation, resulting in an apparently larger molecular mass. These results suggest that P2X(6) receptor subunits contribute to alphabeta-methylene ATP sensitivity.


Asunto(s)
Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/farmacología , Glicosilación , Receptores Purinérgicos P2/metabolismo , Animales , Membrana Celular/metabolismo , Células Cultivadas , Humanos , Fenotipo , Subunidades de Proteína/metabolismo , Ratas , Receptores Purinérgicos P2/efectos de los fármacos
4.
Angiogenesis ; 7(4): 285-94, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15886872

RESUMEN

OBJECTIVE: There is limited knowledge about potential therapeutic targets in Hepatocyte growth factor/scatter factor (HGF)-induced pathophysiological angiogenesis. Recent candidates have included phosphatidylinositol-3-kinase, which is an upstream activator for endothelial nitric oxide (NO) synthase (NOS III). The current study is the first to evaluate the possible involvement of NOS-NO cascade in HGF-induced angiogenesis. METHODS AND RESULTS: NOS III inhibitors blocked the HGF-induced functional neovascularization in vivo, as quantified using vessel counts, 133Xe-clearance, and immunohistology. This was reversed by L-arginine. Western blot analysis of HGF-treated cells also revealed a temporal increase in HGF-induced phosphorylation. In a deconstructional approach, HGF induced the proliferation and chemokinesis of human endothelial cells. These phenotypic effects were inhibited by NOS inhibitors, L-NAME and L-NIO, and the NO scavenger, carboxy PTIO, but unaltered by 1400W, a NOS II inhibitor. This inhibition was reversed by spermine NONOate, a NO donor, which independently exerted a biphasic effect on endothelial cell proliferation. The modulation of NO did not alter HGF-induced chemoinvasion of endothelial cells, while spermine-NONOate destabilized HGF-induced tubulogenesis, suggesting that a single assay is not sufficient for predicting the final phenotypic outcome on angiogenesis. CONCLUSIONS: The study is the first to demonstrate that the NOS III nitric oxide is a key signal cascade in HGF-induced angiogenesis, and represents a promising target for the clinical management of pathological conditions characterized by overt HGF signaling.


Asunto(s)
Factor de Crecimiento de Hepatocito/fisiología , Neovascularización Fisiológica/fisiología , Óxido Nítrico/fisiología , Animales , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Guanilato Ciclasa/antagonistas & inhibidores , Humanos , Isoenzimas/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos BALB C , NG-Nitroarginina Metil Éster/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/metabolismo , Fosforilación
5.
Cancer Res ; 63(23): 8351-9, 2003 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-14678996

RESUMEN

Epidemiological studies have indicated a reduced risk of malignancies with the use of nonsteroidal anti-inflammatory drugs (NSAIDs), although the exact mechanisms are debated. NSAIDs inhibit angiogenesis, which is a key step for tumor growth. Hepatocyte growth factor/scatter factor (HGF/SF), a potent and independent angiogenic factor, has been implicated in tumorigenesis, but limited knowledge exists on the potential targets for inhibiting HGF/SF-induced pathological angiogenesis. The current study was designed to elucidate the possible role of cyclooxygenase (COX) downstream of HGF/SF during angiogenesis and to evaluate the potential for harnessing NSAIDs as a therapeutic strategy. Known NSAIDs were classified as COX-1 or COX-2 selective based on their activity in a platelet aggregation experiment. The inhibitors were administered into a polyether polyurethane scaffold implant in mice at the selected doses, and the total neovascularization after the administration of HGF/SF was quantified using a (133)Xenon clearance technique, vessel counts, and immunohistochemistry. Angiogenesis was also quantized into chemoinvasion, migration, proliferation, and tube formation events in vitro, and the effects of the NSAIDs were evaluated on HGF/SF-induced activity of human umbilical vein endothelial cells (HUVECs). HGF/SF accelerated the angiogenic process in the murine implant, and this activity was inhibited by COX-2-selective meloxicam and NS398. The COX-1 inhibitors ketoprofen and SC560 failed to inhibit the HGF/SF-induced angiogenic events in vitro and in vivo. A COX-2 blockade inhibited the HGF/SF-induced chemoinvasion and migration of human umbilical vein endothelial cells, without affecting the proliferative or tubulogenic responses. Western blots revealed the induction COX-2 expression after HGF/SF treatment, and the pharmacological inhibition of COX-2 executed a temporal inhibition of phosphorylation of the mitogen-activated protein kinases. The current study, for the first time, implicates COX-2 as a downstream signal during HGF/SF-induced angiogenesis, temporally impinging on the mitogen-activated protein kinase signaling. However, the mediation is restricted to only the early events of the angiogenic process, emphasizing the chemopreventive role for NSAIDs. Few therapeutic options currently exist for HGF/SF-induced pathological angiogenesis, and the vast knowledge on COX-2 inhibitors can be harnessed to design a newer therapeutic approach.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antiinflamatorios no Esteroideos/farmacología , Inhibidores de la Ciclooxigenasa/farmacología , Factor de Crecimiento de Hepatocito/antagonistas & inhibidores , Isoenzimas/antagonistas & inhibidores , Neovascularización Fisiológica/efectos de los fármacos , Animales , División Celular/efectos de los fármacos , Ciclooxigenasa 1 , Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa 2 , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/enzimología , Factor de Crecimiento de Hepatocito/farmacología , Factor de Crecimiento de Hepatocito/fisiología , Humanos , Isoenzimas/biosíntesis , Lipooxigenasa/metabolismo , Inhibidores de la Lipooxigenasa/farmacología , Masculino , Proteínas de la Membrana , Ratones , Ratones Endogámicos BALB C , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Fosforilación , Prostaglandina-Endoperóxido Sintasas/biosíntesis
6.
J Neurochem ; 87(4): 1043-51, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14622133

RESUMEN

The drug transporter, P-glycoprotein (P-gp) on brain microvessel endothelium, influences movement of lipophilic substances in and out of the brain. Pathways regulating P-gp expression, both basal and hydrogen peroxide (H2O2)-induced, are here examined in primary cultured rat brain endothelial cells. Activation of extracellular-signal regulated kinases (ERK1/2), protein kinase C (PKC), the p46 isoform of stress-activated protein kinase (SAPK) and its downstream transcription factor, c-Jun, occurred in a time- and concentration-dependent manner following exposure of cells to H2O2 with concomitant increases in P-gp expression. Blockade of ERK activation with U0126, of PKC with Gö6976 and of SAPK with SP600125 decreased basal P-gp but did not abolish the H2O2-induced increase. Blockade of Akt with PI3-kinase inhibitor, LY294002, lowered basal P-gp and prevented the H2O2-induced increase. Inhibition of nuclear factor-kappaB (NF-kappaB), either by blocking dissociation from its inhibitory factor, IkappaB, with MG132 or its nuclear translocation with SN50 enhanced basal P-gp, obscuring the H2O2-induced increase. H2O2 itself produced no detectable activation of IkappaB, but inhibited that induced by 5 ng/mL tumour necrosis factor-alpha (TNF-alpha). P-gp expression may involve positive inputs from ERK1/2, SAPK, Akt and PKC and inhibitory influences of NF-kappaB. By depressing NF-kappaB signalling, H2O2 may still augment P-gp expression when ERK1/2, PKC or SAPK are inhibited.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/biosíntesis , Barrera Hematoencefálica/metabolismo , Endotelio Vascular/metabolismo , Peróxido de Hidrógeno/farmacología , Proteínas Serina-Treonina Quinasas , Transducción de Señal/fisiología , Animales , Barrera Hematoencefálica/citología , Encéfalo/irrigación sanguínea , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Expresión Génica/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Oxidantes/farmacología , Péptidos/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas c-jun/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos
7.
Circulation ; 107(23): 2955-61, 2003 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-12782568

RESUMEN

BACKGROUND: Hepatocyte growth factor/scatter factor (HGF/SF) can sufficiently and independently induce pathophysiological angiogenesis. However, the treatment strategies have mostly been unsuccessful. The present study is the first to evaluate the possible targeting of downstream signals for the inhibition of HGF/SF-induced angiogenesis. METHODS AND RESULTS: In a multichannel scratch assay with human endothelial cells (ECs), HGF/SF induced a strong and prolonged activation of MAPK and cell proliferation that was inhibited by PD98059 and LY294002/wortmannin, selective inhibitors of MAPK and PI3K signaling modules, respectively. Western blotting demonstrated a temporal relation between the activation of the two pathways. Chemical inhibition of the PI3K and MAPK signals inhibited HGF/SF-induced chemoinvasion of ECs in vitro and blocked the HGF/SF-induced neovascularization into a polymer scaffold in vivo, as quantified by vessel counts and the clearance of radioactive 133Xe. CONCLUSIONS: These data indicate that MEK and PI3K inhibitors represent a promising approach to the clinical management of pathological conditions characterized by overt HGF/SF-induced angiogenesis.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Factor de Crecimiento de Hepatocito/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Neovascularización Fisiológica/efectos de los fármacos , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Serina-Treonina Quinasas , Androstadienos/farmacología , Animales , Bioensayo , División Celular/efectos de los fármacos , Células Cultivadas , Cromonas/farmacología , Colágeno , Combinación de Medicamentos , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Flavonoides/farmacología , Factor de Crecimiento de Hepatocito/farmacología , Humanos , Laminina , Masculino , Ratones , Ratones Endogámicos BALB C , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Morfolinas/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Proteoglicanos , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal/efectos de los fármacos , Wortmanina
8.
Br J Pharmacol ; 139(2): 219-31, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12770927

RESUMEN

1 Thymidine phosphorylase (TP) is elevated in the plasma of cancer patients, and has been implicated in pathophysiological angiogenesis. However, the downstream signals underlying this implication remain obscure. The purpose of the present study was to examine the effects of TP on the neovascularisation response in vitro and in vivo. 2 Both TP and its catalytic product, 2-deoxy-D-ribose-1-phosphate, and downstream 2-deoxy-D-ribose (2-DDR) promoted endothelial tubulogenesis in vitro, and the regeneration of a wounded monolayer of endothelial cells without exerting any mitogenic effect. In vivo, both TP and 2-DDR promoted the development of functional vasculature into an avascular sponge. A TP inhibitor, 6-amino-5-chlorouracil, was able to partially reverse the effects of TP, but had no effect on the 2-DDR-induced angiogenesis. 3 Enhanced monolayer regeneration was observed with TP-cDNA-transfected bladder carcinoma cells. The transfection of TP-cDNA, however, did not confer any proliferative advantage. The regeneration of TP overexpressing cells was associated with a time-dependent expression of the enzyme haeme-oxygenase (HO-1). 4 The present study demonstrates that both TP and its ribose-sugar metabolites induce angiogenesis by mediating a cohesive interplay between carcinoma and endothelial cells. The induction of HO-1 in TP-transfected cells suggests that it could be a possible downstream signal for the angiogenic effects of TP. Furthermore, reducing sugars have been shown to induce oxidative stress, and ribose could be a possible cause for the upregulation of HO-1, which has been implicated in the release of angiogenic factors. Therefore, we postulate that 2-DDR could be mediating the angiogenic effects of TP possibly through an oxidative stress mechanism and additionally getting integrated in the endothelial metabolic machinery.


Asunto(s)
Neovascularización Patológica/metabolismo , Timidina Fosforilasa/metabolismo , Inductores de la Angiogénesis/farmacología , Animales , Células Cultivadas , Desoxirribosa/química , Desoxirribosa/farmacología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/enzimología , Endotelio Vascular/patología , Hemo Oxigenasa (Desciclizante)/biosíntesis , Hemo-Oxigenasa 1 , Humanos , Masculino , Proteínas de la Membrana , Ratones , Ratones Endogámicos BALB C , Neovascularización Patológica/patología , Ribosamonofosfatos/farmacología , Estereoisomerismo , Relación Estructura-Actividad , Timidina Fosforilasa/antagonistas & inhibidores , Timidina Fosforilasa/farmacología , Transfección , Células Tumorales Cultivadas
9.
Arterioscler Thromb Vasc Biol ; 23(1): 69-75, 2003 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-12524227

RESUMEN

OBJECTIVE: Hepatocyte growth factor/scatter factor (HGF/SF) promotes vascular endothelial growth factor (VEGF) expression and induces angiogenesis in multiple pathological conditions. The present study was designed to delineate the HGF/SF and VEGF signaling cascades during angiogenesis by using PTK787, a selective VEGF receptor antagonist. METHODS AND RESULTS: PTK787 produced a concentration-dependent (10(-8) to 10(-6) mol/L) inhibition of VEGF-induced angiogenesis, without altering the basal or HGF/SF-induced response in vitro. In contrast, the nonspecific kinase inhibitor genistein blocked the HGF/SF-induced effect. Both VEGF and HGF/SF induced a rapid phosphorylation of extracellular receptor kinases-1 and -2 (ERKs) and Akt. PTK787 inhibited the VEGF-induced activation of Akt and ERKs, without affecting the HGF/SF-induced phosphorylation. Treatment with VEGF and HGF/SF increased total neovascularization in a murine scaffold granuloma model, but no additive or synergistic interactions were observed. PTK787 (50 mg/kg) blocked the VEGF-induced response without altering the basal or HGF/SF-induced neovascularization. CONCLUSIONS: We demonstrate that HGF/SF can induce angiogenesis independently of VEGF, possibly through the direct activation of the Akt and ERKs. These results demonstrate the necessity of a multitargeted approach for the rational design of newer therapies to inhibit pathophysiological angiogenesis.


Asunto(s)
Factores de Crecimiento Endotelial/fisiología , Factor de Crecimiento de Hepatocito/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Linfocinas/fisiología , Neovascularización Patológica/fisiopatología , Proteínas Proto-Oncogénicas , Piridinas , Animales , División Celular/efectos de los fármacos , División Celular/fisiología , Células Cultivadas , Factores de Crecimiento Endotelial/antagonistas & inhibidores , Factores de Crecimiento Endotelial/biosíntesis , Factores de Crecimiento Endotelial/genética , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/enzimología , Endotelio Vascular/lesiones , Endotelio Vascular/patología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/genética , Linfocinas/antagonistas & inhibidores , Linfocinas/biosíntesis , Linfocinas/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones , Ratones Endogámicos BALB C , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/fisiología , Fosforilación/efectos de los fármacos , Ftalazinas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptores de Factores de Crecimiento Endotelial Vascular/fisiología , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Transfección , Células Tumorales Cultivadas , Venas Umbilicales/efectos de los fármacos , Venas Umbilicales/enzimología , Venas Umbilicales/patología , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA