Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Cancer Ther ; 16(12): 2668-2676, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28978721

RESUMEN

An increasing number of cancers are known to harbor mutations, translocations, or amplifications in the fibroblast growth factor receptor (FGFR) family of kinases. The FGFR inhibitors evaluated in clinical trials to date have shown promise at treating these cancers. Here, we describe PRN1371, an irreversible covalent inhibitor of FGFR1-4 targeting a cysteine within the kinase active site. PRN1371 demonstrated strong FGFR potency and excellent kinome-wide selectivity in a number of biochemical and cellular assays, including in various cancer cell lines exhibiting FGFR alterations. Furthermore, PRN1371 maintained FGFR inhibition in vivo, not only when circulating drug levels were high but also after the drug had been cleared from circulation, indicating the possibility of sustained FGFR inhibition in the clinic without the need for continuous drug exposure. Durable tumor regression was also obtained in multiple tumor xenografts and patient-derived tumor xenograft models and was sustained even using an intermittent dosing strategy that provided drug holidays. PRN1371 is currently under clinical investigation for treatment of patients with solid tumors. Mol Cancer Ther; 16(12); 2668-76. ©2017 AACR.


Asunto(s)
Piridonas/uso terapéutico , Pirimidinas/uso terapéutico , Receptores de Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Proliferación Celular , Humanos , Ratones , Piridonas/farmacología , Pirimidinas/farmacología , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Med Chem ; 60(15): 6516-6527, 2017 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-28665128

RESUMEN

Aberrant signaling of the FGF/FGFR pathway occurs frequently in cancers and is an oncogenic driver in many solid tumors. Clinical validation of FGFR as a therapeutic target has been demonstrated in bladder, liver, lung, breast, and gastric cancers. Our goal was to develop an irreversible covalent inhibitor of FGFR1-4 for use in oncology indications. An irreversible covalent binding mechanism imparts many desirable pharmacological benefits including high potency, selectivity, and prolonged target inhibition. Herein we report the structure-based design, medicinal chemistry optimization, and unique ADME assays of our irreversible covalent drug discovery program which culminated in the discovery of compound 34 (PRN1371), a highly selective and potent FGFR1-4 inhibitor.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias/tratamiento farmacológico , Piridonas/farmacología , Pirimidinas/farmacología , Receptores de Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/síntesis química , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Perros , Diseño de Fármacos , Estabilidad de Medicamentos , Femenino , Humanos , Absorción Intestinal , Macaca fascicularis , Masculino , Piridonas/administración & dosificación , Piridonas/síntesis química , Piridonas/farmacocinética , Pirimidinas/administración & dosificación , Pirimidinas/síntesis química , Pirimidinas/farmacocinética , Ratas Sprague-Dawley , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Solubilidad , Relación Estructura-Actividad
3.
Nat Chem Biol ; 11(7): 525-31, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26006010

RESUMEN

Drugs with prolonged on-target residence times often show superior efficacy, yet general strategies for optimizing drug-target residence time are lacking. Here we made progress toward this elusive goal by targeting a noncatalytic cysteine in Bruton's tyrosine kinase (BTK) with reversible covalent inhibitors. Using an inverted orientation of the cysteine-reactive cyanoacrylamide electrophile, we identified potent and selective BTK inhibitors that demonstrated biochemical residence times spanning from minutes to 7 d. An inverted cyanoacrylamide with prolonged residence time in vivo remained bound to BTK for more than 18 h after clearance from the circulation. The inverted cyanoacrylamide strategy was further used to discover fibroblast growth factor receptor (FGFR) kinase inhibitors with residence times of several days, demonstrating the generalizability of the approach. Targeting of noncatalytic cysteines with inverted cyanoacrylamides may serve as a broadly applicable platform that facilitates 'residence time by design', the ability to modulate and improve the duration of target engagement in vivo.


Asunto(s)
Acrilamidas/farmacocinética , Linfocitos B/efectos de los fármacos , Cianoacrilatos/farmacocinética , Inhibidores de Proteínas Quinasas/farmacocinética , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Acrilamidas/síntesis química , Agammaglobulinemia Tirosina Quinasa , Animales , Linfocitos B/enzimología , Linfocitos B/patología , Línea Celular Tumoral , Cristalografía por Rayos X , Cianoacrilatos/síntesis química , Dasatinib , Femenino , Expresión Génica , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Estructura Terciaria de Proteína , Proteínas Tirosina Quinasas/química , Proteínas Tirosina Quinasas/genética , Pirimidinas/farmacocinética , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Células Sf9 , Spodoptera , Relación Estructura-Actividad , Especificidad por Sustrato , Tiazoles/farmacocinética , Factores de Tiempo
4.
PLoS One ; 6(3): e17692, 2011 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-21408192

RESUMEN

BACKGROUND: Many proteins that are dysregulated or mutated in cancer cells rely on the molecular chaperone HSP90 for their proper folding and activity, which has led to considerable interest in HSP90 as a cancer drug target. The diverse array of HSP90 client proteins encompasses oncogenic drivers, cell cycle components, and a variety of regulatory factors, so inhibition of HSP90 perturbs multiple cellular processes, including mitogenic signaling and cell cycle control. Although many reports have investigated HSP90 inhibition in the context of the cell cycle, no large-scale studies have examined potential correlations between cell genotype and the cell cycle phenotypes of HSP90 inhibition. METHODOLOGY/PRINCIPAL FINDINGS: To address this question, we developed a novel high-content, high-throughput cell cycle assay and profiled the effects of two distinct small molecule HSP90 inhibitors (XL888 and 17-AAG [17-allylamino-17-demethoxygeldanamycin]) in a large, genetically diverse panel of cancer cell lines. The cell cycle phenotypes of both inhibitors were strikingly similar and fell into three classes: accumulation in M-phase, G2-phase, or G1-phase. Accumulation in M-phase was the most prominent phenotype and notably, was also correlated with TP53 mutant status. We additionally observed unexpected complexity in the response of the cell cycle-associated client PLK1 to HSP90 inhibition, and we suggest that inhibitor-induced PLK1 depletion may contribute to the striking metaphase arrest phenotype seen in many of the M-arrested cell lines. CONCLUSIONS/SIGNIFICANCE: Our analysis of the cell cycle phenotypes induced by HSP90 inhibition in 25 cancer cell lines revealed that the phenotypic response was highly dependent on cellular genotype as well as on the concentration of HSP90 inhibitor and the time of treatment. M-phase arrest correlated with the presence of TP53 mutations, while G2 or G1 arrest was more commonly seen in cells bearing wt TP53. We draw upon previous literature to suggest an integrated model that accounts for these varying observations.


Asunto(s)
Compuestos de Azabiciclo/farmacología , Ciclo Celular , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Ensayos Analíticos de Alto Rendimiento/métodos , Ácidos Ftálicos/farmacología , Benzoquinonas/farmacología , Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Citometría de Flujo , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Lactamas Macrocíclicas/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Factores de Tiempo , Quinasa Tipo Polo 1
5.
Microcirculation ; 17(2): 128-36, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20163539

RESUMEN

OBJECTIVE: To test the hypothesis that rapamycin inhibits induced microvascular hyperpermeability directly in vivo. METHODS: Male golden Syrian hamsters (80-120 g) were treated with either rapamycin (at 0.1, 0.5, 2, and 10 mg/kg i.p.) or vehicle at 24 hours and at 1 hour prior to preparation of the cheek pouch. Caveolin-1 scaffolding (1 mg/kg; positive inhibitory control) was injected i.p. 24 hours prior to the experiment. 10(-8) M vascular endothelial growth factor (VEGF) or 10(-7) M platelet-activating factor (PAF) were topically applied to the cheek pouch. Microvascular permeability and arteriolar diameter were assessed using integrated optical intensity (IOI) and vascular wall imaging, respectively. RESULTS: Rapamycin at 0.1 and 0.5 mg/kg significantly reduced VEGF-stimulated mean IOI from 63.0 +/- 4.2 to 9.7 +/- 5.0 (85% reduction, P < 0.001) and 3.6 +/- 2.7 (95% reduction, P < 0.001), respectively. Rapamycin at 2 mg/kg also lowered VEGF-stimulated hyperpermeability (40% reduction, P < 0.05). However, 10 mg/kg rapamycin increased VEGF-induced microvascular hyperpermeability. Rapamycin at 0.5 mg/kg attenuated VEGF-induced vasodilation and PAF-induced hyperpermeability, but did not inhibit PAF-induced vasoconstriction. CONCLUSIONS: At therapeutically relevant concentrations, rapamycin inhibits VEGF- and PAF-induced microvascular permeability. This inhibition is (i) a direct effect on the endothelial barrier, and (ii) independent of arteriolar vasodilation. Rapamycin at 10 mg/kg stimulates effectors that increase microvascular permeability.


Asunto(s)
Permeabilidad Capilar/efectos de los fármacos , Microcirculación/efectos de los fármacos , Sirolimus/farmacología , Factor A de Crecimiento Endotelial Vascular/farmacología , Animales , Arteriolas/efectos de los fármacos , Arteriolas/fisiología , Velocidad del Flujo Sanguíneo/efectos de los fármacos , Mejilla , Cricetinae , Masculino , Mesocricetus , Microcirculación/fisiología , Mucosa Bucal/irrigación sanguínea , Mucosa Bucal/efectos de los fármacos , Factor de Activación Plaquetaria/antagonistas & inhibidores , Factor de Activación Plaquetaria/farmacología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Vasodilatación/efectos de los fármacos
6.
Clin Cancer Res ; 14(8): 2465-75, 2008 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-18413839

RESUMEN

PURPOSE: Mutations associated with resistance to kinase inhibition are an important mechanism of intrinsic or acquired loss of clinical efficacy for kinase-targeted therapeutics. We report the prospective discovery of ErbB2 mutations that confer resistance to the small-molecule inhibitor lapatinib. EXPERIMENTAL DESIGN: We did in vitro screening using a randomly mutagenized ErbB2 expression library in Ba/F3 cells, which were dependent on ErbB2 activity for survival and growth. RESULTS: Lapatinib resistance screens identified mutations at 16 different ErbB2 amino acid residues, with 12 mutated amino acids mapping to the kinase domain. Mutations conferring the greatest lapatinib resistance cluster in the NH2-terminal kinase lobe and hinge region. Structural computer modeling studies suggest that lapatinib resistance is caused by multiple mechanisms; including direct steric interference and restriction of conformational flexibility (the inactive state required for lapatinib binding is energetically unfavorable). ErbB2 T798I imparts the strongest lapatinib resistance effect and is analogous to the epidermal growth factor receptor T790M, ABL T315I, and cKIT T670I gatekeeper mutations that are associated with clinical drug resistance. ErbB2 mutants associated with lapatinib resistance transformed NIH-3T3 cells, including L755S and T733I mutations known to occur in human breast and gastric carcinomas, supporting a direct mechanism for lapatinib resistance in ErbB2-driven human cancers. The epidermal growth factor receptor/ErbB2/vascular endothelial growth factor receptor inhibitor EXEL-7647 was found to inhibit almost all lapatinib resistance-associated mutations. Furthermore, no ErbB2 mutations were found to be associated with EXEL-7647 resistance and lapatinib sensitivity. CONCLUSIONS: Taken together, these data suggest potential target-based mechanisms of resistance to lapatinib and suggest that EXEL-7647 may be able to circumvent these effects.


Asunto(s)
Antineoplásicos/farmacología , Transformación Celular Neoplásica , Mutación , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas/farmacología , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-2/genética , Supervivencia Celular , Resistencia a Antineoplásicos , Humanos , Lapatinib , Fosforilación , Conformación Proteica , Receptor ErbB-2/química
7.
Microcirculation ; 12(1): 129-140, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15825252

RESUMEN

This review addresses a rapidly growing area of vascular biology, i.e. genomic variations in vascular genes that underlie different human phenotypes. Two of the most important molecular in vascular biology, endothelial nitric oxide synthase (eNOS) and vascular endothelial cell growth factor (VEGF)are discussed. Variations in the eNOS gene have been correlated with a number of human diseases including hypertension, coronary vasospasm, smoking dependent risk of coronary disease, myocardial infarction and placental disruption. Similarly, variations in the VEGF gene have been associated with increased risk of various cancers, DiGeorge syndrome, psoriasis, diabetic renal disease and amyotropic lateral sclerosis. Understanding the molecular basis of these genetic variations and how they contribute to the pathophysiology provides new and important insights into human disease.


Asunto(s)
Óxido Nítrico Sintasa/genética , Factor A de Crecimiento Endotelial Vascular/genética , Enfermedades Cardiovasculares/genética , Variación Genética , Humanos , Óxido Nítrico Sintasa de Tipo III
8.
Vitam Horm ; 70: 105-35, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15727803

RESUMEN

Stanniocalcin was originally described as a hormone with calcitonin-like actions in fish. During the last decade, mammalian forms of stanniocalcin have been identified, and this discovery has led to important advances in our understanding of this enigmatic polypeptide hormone. This review briefly covers some early studies on stanniocalcin in fish and then provides a more in-depth look at some of the more intriguing, new aspects of its functions in mammals. The roles of stanniocalcin in renal function, metabolism, angiogenesis, pregnancy and lactation, bone formation, and neural protection are discussed, along with new information relating to its receptor-mediated sequestration and accumulation in target cell organelles.


Asunto(s)
Glicoproteínas/fisiología , Secuencia de Aminoácidos , Animales , Femenino , Peces , Glicoproteínas/química , Glicoproteínas/genética , Glicoproteínas/historia , Historia del Siglo XIX , Historia del Siglo XX , Humanos , Mamíferos , Ratones , Ratones Transgénicos , Mitocondrias , Datos de Secuencia Molecular , Embarazo
10.
Endothelium ; 11(5-6): 285-91, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15763948

RESUMEN

Vascular endothelial cells sense and respond to pressure by molecular mechanism(s) which, to date, remain poorly understood. The present study investigated basic fibroblast growth factor (bFGF) signaling as a putative mechanotransduction pathway involved in the proliferative responses of human umbilical vein endothelia cells (HUVECs) to 60/20 mm Hg cyclic pressure at 1 Hz for 24 h. Under these conditions, the enhanced proliferative response of these HUVECs was not associated with an increased synthesis/release of bFGF, but involved rapid (within 30 min from the onset of exposure to pressure) tyrosine phosphorylation of the bFGF receptor, FGFR-2. Furthermore, monoclonal antibodies to either bFGF or FGFR-2 attenuated the increased proliferation of HUVECs exposed to 60/20 mm Hg cyclic pressure. HUVECs proliferation under 60/20 mm Hg at 1 Hz cyclic pressure is, therefore, dependent upon bFGF and involves FGFR-2 activation.


Asunto(s)
División Celular/fisiología , Células Endoteliales/fisiología , Factor 2 de Crecimiento de Fibroblastos/fisiología , Receptores de Factores de Crecimiento de Fibroblastos/fisiología , Transducción de Señal/fisiología , Anticuerpos Monoclonales/inmunología , Células Endoteliales/citología , Endotelio Vascular/fisiología , Factor 2 de Crecimiento de Fibroblastos/inmunología , Humanos , Fosforilación , Receptores de Factores de Crecimiento de Fibroblastos/inmunología , Tirosina/metabolismo
11.
Br J Pharmacol ; 140(4): 595-610, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14504135

RESUMEN

Hepatocyte growth factor (HGF) and vascular endothelial cell growth factor (VEGF) are two potent endothelial mitogens with demonstrated angiogenic activities in animal models of therapeutic angiogenesis. Several recent studies suggest that these growth factors may act synergistically, although the mechanism of this interaction is not understood. Changes in the gene expression profile of human umbilical vein endothelial cells treated with HGF, VEGF or the combination of the two were analyzed with high-density oligonucleotide arrays, representing approximately 22000 genes. Notably, the genes significantly up- and downregulated by VEGF versus HGF exhibited very little overlap, indicating distinct signal transduction pathways. The combination of HGF and VEGF markedly increased the number of significantly up- and downregulated genes. At 4 h, the combination of the two growth factors induced a number of chemokine and cytokines and their receptors (IL-8, IL-6, IL-11, CCR6, CXCR1,CXC1 and IL17RC), numerous genes involved in growth factor signal transduction (egr-1, fosB, grb10, grb14,MAP2K3,MAP3K8, MAPKAP2,MPK3, DUSP4 and DUSP6), as well as a number of other growth factors (PDGFA, BMP2, Hb-EGF, FGF16, heuregulin beta 1, c-kit ligand, angiopoietin 2 and angiopoietin 4 and VEGFC). In addition, the VEGF receptors neuropilin-1 and flt-1 were also upregulated. At 24 h, a clear 'cell cycle' signature is noted, with the upregulated expression of various cell cycle control proteins and gene involved in the regulation of mitosis and mitotic spindle assembly. The receptor for HGF, c-met, is also upregulated. These data are consistent with the hypothesis that the combination of HGF and VEGF results in the cooperative upregulation of a number of different molecular pathways leading to a more robust proliferative response, that is, growth factor(s), receptors, molecules involved in growth factor signal transduction, as well as, at later time points, upregulation of the necessary cellular proteins required for cells to escape cell cycle arrest and enter the cell cycle.


Asunto(s)
Células Endoteliales/metabolismo , Perfilación de la Expresión Génica/métodos , Factor de Crecimiento de Hepatocito/metabolismo , Sondas Moleculares/química , Factores de Crecimiento Endotelial Vascular/metabolismo , Factor de Crecimiento de Hepatocito/química , Factor de Crecimiento de Hepatocito/genética , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Factores de Crecimiento Endotelial Vascular/química , Factores de Crecimiento Endotelial Vascular/genética
12.
J Biol Chem ; 278(48): 47654-9, 2003 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-14500721

RESUMEN

Stanniocalcin 1 (STC1) is a secreted glycoprotein originally described as a hormone involved in calcium and phosphate homeostasis in bony fishes. We recently identified the mammalian homolog of this molecule to be highly up-regulated in an in vitro model of angiogenesis, as well as focally and intensely expressed at sites of pathological angiogenesis (e.g. tumor vasculature). In the present study, we report that STC1 is a selective modulator of hepatocyte growth factor (HGF)-induced endothelial migration and morphogenesis, but not proliferation. STC1 did not inhibit proliferative or migratory responses to vascular endothelial growth factor or basic fibroblast growth factor. The mechanism of STC1 inhibitory effects on HGF-induced endothelial migration seem to occur secondary to receptor activation because STC1 did not inhibit HGF-induced c-met receptor phosphorylation, but did block HGF-induced focal adhesion kinase activation. In the mouse femoral artery ligation model of angiogenesis, STC1 expression closely paralleled that of the endothelial marker CD31, and the peak level of STC1 expression occurred after an increase in HGF expression. We propose that STC1 may play a selective modulatory role in angiogenesis, possibly serving as a "stop signal" or stabilizing factor contributing to the maturation of newly formed blood vessels. HGF is a mesenchyme-derived pleiotropic factor with mitogenic, motogenic, and morphogenic activities on a number of different cell types. HGF effects are mediated through a specific tyrosine kinase, c-met, and aberrant HGF and c-met expression are frequently observed in a variety of tumors. Recent studies have shown HGF to be a potent growth factor implicated in wound healing, tissue regeneration, and angiogenesis.


Asunto(s)
Endotelio/metabolismo , Glicoproteínas/fisiología , Factor de Crecimiento de Hepatocito/metabolismo , Neovascularización Fisiológica , Animales , Anticuerpos Monoclonales/química , División Celular , Movimiento Celular , Células Cultivadas , Colágeno/farmacología , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Endotelio Vascular/citología , Activación Enzimática , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Vectores Genéticos , Humanos , Laminina/farmacología , Masculino , Mesodermo/metabolismo , Ratones , Ratones Endogámicos C57BL , Fosforilación , Proteínas Tirosina Quinasas/metabolismo , Proteoglicanos/farmacología , Proteínas Proto-Oncogénicas c-met/metabolismo , ARN Mensajero/metabolismo , Factores de Tiempo , Venas Umbilicales/citología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Cicatrización de Heridas
13.
J Biol Chem ; 278(35): 33232-8, 2003 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-12807873

RESUMEN

We have previously utilized a combination of high throughput sequencing and genome-wide microarray profiling analyses to identify novel cell-surface proteins expressed in human umbilical vein endothelial cells. One gene identified by this approach encodes a type I transmembrane receptor that shares sequence homology with the intracellular domain of members of the interleukin-17 (IL-17) receptor family. Real-time quantitative PCR and Northern analyses revealed that this gene is highly expressed in human umbilical vein endothelial cells and in several highly vascularized tissues such as kidney, colon, skeletal muscle, heart, and small intestine. In addition, we also found that it is also highly expressed in the ductal epithelial cells of human salivary glands, seminal vesicles, and the collecting tubules of the kidney by in situ hybridization. This putative receptor, which we have termed human SEF (hSEF), is also expressed in a variety of breast cancer tissues. In co-immunoprecipitation assays, this receptor is capable of forming homomeric complexes and can interact with fibroblast growth factor (FGF) receptor 1. Overexpression of this receptor inhibits FGF induction of an FGF-responsive reporter gene in human 293T cells. This appears to occur as a result of specific inhibition of p42/p44 ERK in the absence of upstream MEK inhibition. This inhibitory effect is dependent upon a functional intracellular domain since deletion mutants missing the IL-17 receptor-like domain lack this inhibitory effect. These findings are consistent with the recent discovery of the zebrafish homologue, Sef (similar expression to fgf genes), which specifically antagonizes FGF signaling when ectopically expressed in zebrafish or Xenopus laevis embryos. Based on sequence and functional similarities, this novel IL-17 receptor homologue represents a potential human SEF and is likely to play critical roles in endothelial or epithelial functions such as proliferation, migration, and angiogenesis.


Asunto(s)
Endotelio Vascular/citología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Receptores de Interleucina/química , Receptores de Interleucina/metabolismo , Proteínas Recombinantes/química , Transducción de Señal , Venas Umbilicales/citología , Secuencia de Aminoácidos , Animales , Northern Blotting , Western Blotting , División Celular , Línea Celular , Movimiento Celular , Células Cultivadas , Clonación Molecular , Genes Reporteros , Humanos , Hibridación in Situ , Luciferasas/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Datos de Secuencia Molecular , Neovascularización Fisiológica , Plásmidos/metabolismo , Pruebas de Precipitina , Unión Proteica , Estructura Terciaria de Proteína , ARN Mensajero/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Receptores de Interleucina/genética , Receptores de Interleucina-17 , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Distribución Tisular , Transfección , Células Tumorales Cultivadas
14.
Microcirculation ; 10(1): 63-81, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12610664

RESUMEN

The process of endothelial differentiation into a network of tube-like structures with patent lumens requires an integrated program of gene expression. To identify genes upregulated in endothelial cells during the process of tube formation, RNA was prepared from several different time points (0, 4, 8, 24, 40, and 48 hours) and from three different experimental models of human endothelial tube formation: in collagen gels and fibrin gels driven by the combination of PMA (80), bFGF (40 ng/ml) and bFGF (40 ng/ml) or in collagen gels driven by the combination of HGF (40 ng/ml) and VEGF (40 ng/ml). Gene expression was evaluated using Affymetrix Gene Chip oligonucleotide arrays. Over 1000 common genes were upregulated greater than twofold over baseline at one or more time points in the three different models. In the present study, we discuss the identified genes that could be assigned to major functional classes: apoptosis, cytoskeleton, proteases, matrix, and matrix turnover, pumps and transporters, membrane lipid turnover, and junctional molecules or adhesion proteins.


Asunto(s)
Endotelio Vascular/citología , Regulación del Desarrollo de la Expresión Génica , Neovascularización Fisiológica/fisiología , Animales , Apoptosis/genética , Moléculas de Adhesión Celular/biosíntesis , Moléculas de Adhesión Celular/genética , Diferenciación Celular/genética , Células Cultivadas/metabolismo , Colágeno , Proteínas del Citoesqueleto/biosíntesis , Proteínas del Citoesqueleto/genética , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Fibrina , Geles , Perfilación de la Expresión Génica , Sustancias de Crecimiento/farmacología , Humanos , Lípidos de la Membrana/biosíntesis , Neovascularización Fisiológica/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Acetato de Tetradecanoilforbol/farmacología
15.
Arterioscler Thromb Vasc Biol ; 22(11): 1797-803, 2002 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-12426207

RESUMEN

OBJECTIVE: This study evaluated the relative roles of the vascular endothelial growth factor (VEGF) receptors KDR and Flt-1 in the mediation of altered gene expression elicited by VEGF. METHODS AND RESULTS: We used mutants of VEGF selective for the KDR and Flt-1 receptors to differentiate gene expression patterns mediated by wild-type VEGF (VEGFwt) in human umbilical vein endothelial cells. RNA was extracted from cells treated for 24 hours with 1 nmol/L of each ligand, and gene expression was monitored by using oligonucleotide arrays (Affymetrix U95A). We report that activation of KDR was sufficient to upregulate all the genes induced by VEGFwt. In contrast, there were no genes selectively upregulated by the Flt-selective mutant. However, high concentrations of the Flt-selective mutant could augment the expression of some genes induced by submaximal concentrations of VEGFwt but not the KDR-selective mutant. CONCLUSIONS: The binding of VEGF to its receptor, KDR, is necessary and sufficient to induce the gene expression profile induced by this growth factor. Furthermore, in human umbilical vein endothelial cells, the Flt-1 receptor appears to act as a decoy receptor, tempering the response to lower concentrations of VEGF.


Asunto(s)
Factores de Crecimiento Endotelial/fisiología , Endotelio Vascular/química , Endotelio Vascular/metabolismo , Regulación de la Expresión Génica/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Linfocinas/fisiología , Venas Umbilicales/química , Venas Umbilicales/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/fisiología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/fisiología , Células Cultivadas , Factores de Crecimiento Endotelial/genética , Factores de Crecimiento Endotelial/farmacología , Endotelio Vascular/citología , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/farmacología , Linfocinas/genética , Linfocinas/farmacología , Mutación/genética , Mutación/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa/métodos , Receptores CXCR4/biosíntesis , Factores de Tiempo , Venas Umbilicales/citología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología , Factor A de Crecimiento Endotelial Vascular , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Factores de Crecimiento Endotelial Vascular
16.
Physiol Genomics ; 11(3): 245-51, 2002 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-12388793

RESUMEN

Mechanical forces modulate endothelial cell functions through several mechanisms including regulation of gene transcription. In the present study, gene transcription by human umbilical vein endothelial cells (HUVEC) either maintained under control pressure (that is, standard cell culture conditions equivalent to 0.15 mmHg sustained hydrostatic pressure) or exposed to 60/20 mmHg sinusoidal pressures at 1 Hz were compared using Affymetrix GeneChip microarrays to identify cellular/molecular mechanisms associated with endothelial cell responses to cyclic pressure. Cyclic pressure selectively affected transcription of 14 genes that included a set of mechanosensitive proteins involved in hemostasis (tissue plasminogen activator), cell adhesion (integrin-alpha2), and cell signaling (Rho B, cytosolic phospholipase A2), as well as a unique subset of cyclic pressure-sensitive genes such as vascular endothelial growth factor (VEGF)-C and transforming growth factor (TGF)-beta2. The present study also provided first evidence that VEGF-C, the most highly induced gene under 60/20 mmHg, mediated HUVEC proliferation in response to this cyclic pressure. Cyclic pressure is, therefore, a mechanical force that modulates endothelial cell functions (such as proliferation) by activating a specific transcriptional program.


Asunto(s)
Factores de Crecimiento Endotelial/fisiología , Endotelio Vascular/metabolismo , Transcripción Genética , División Celular , Células Cultivadas , Factores de Crecimiento Endotelial/genética , Endotelio Vascular/efectos de los fármacos , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Periodicidad , Presión , ARN Mensajero/biosíntesis , Estrés Mecánico , Factor C de Crecimiento Endotelial Vascular
17.
Physiol Genomics ; 11(3): 263-72, 2002 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-12399448

RESUMEN

DNA microarrays were used to measure the time course of gene expression during skeletal muscle damage and regeneration in mice following femoral artery ligation (FAL). We found 1,289 known sequences were differentially expressed between the FAL and control groups. Gene expression peaked on day 3, and the functional cluster "inflammation" contained the greatest number of genes. Muscle function was depressed for 3 days postligation, but returned to normal by day 7. Decreased muscle function was accompanied by reduced expression of genes involved in mitochondrial energy production, muscle contraction, and calcium handling. The induction of MyoD on day 1 denoted the beginning of muscle regeneration and was followed by the reemergence of the embryonic forms of muscle contractile proteins, which peaked at day 7. Transcriptional analysis indicated that the ischemic skeletal muscle may transition through a functional adaptation stage with recovery of contractile force prior to full regeneration. Several members of the insulin-like growth factor axis were coordinately induced in a time frame consistent with their playing a role in the regenerative process.


Asunto(s)
Isquemia/genética , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiología , Regeneración , Enfermedad Aguda , Animales , Citocinas/biosíntesis , Citocinas/genética , Arteria Femoral/cirugía , Perfilación de la Expresión Génica , Isquemia/metabolismo , Isquemia/patología , Cinética , Ligadura , Extremidad Inferior , Masculino , Ratones , Ratones Endogámicos C57BL , Contracción Muscular , Músculo Esquelético/patología , Miosinas/biosíntesis , Miosinas/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/biosíntesis , Receptores de Citocinas/biosíntesis , Receptores de Citocinas/genética , Somatomedinas/biosíntesis , Somatomedinas/genética , Transcripción Genética
18.
Endocrinology ; 143(9): 3681-90, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12193584

RESUMEN

Fish stanniocalcin (STC) inhibits uptake of calcium and stimulates phosphate reabsorption. To determine the role of the highly homologous mammalian protein, STC-1, we created and characterized transgenic mice that express STC-1 under control of a muscle-specific promoter. STC-1 transgenic mice were smaller than wild-type littermates and had normal growth plate cartilage morphology but increased cartilage matrix synthesis. In STC-1 mice, the rate of bone formation, but not bone mineralization, was decreased. Increased cortical bone thickness and changes in trabeculae number, density, and thickness in STC-1 mice indicated a concomitant suppression of osteoclast activity, which was supported by microcomputed tomography analyses and histochemistry. Skeletal muscles were disproportionately small and showed altered function and response to injury in STC-1 mice. Electron microscopy indicated that muscle mitochondria were dramatically enlarged in STC-1 mice. These changes in STC-1 mice could not be explained by deficits in blood vessel formation, as vascularity in organs and skeletal tissues was increased as was induction of vascularity in response to femoral artery ligation. Our results indicate that STC-1 can affect calcium homeostasis, bone and muscle mass and structure, and angiogenesis through effects on osteoblasts, osteoclasts, myoblasts/myocytes, and endothelial cells.


Asunto(s)
Huesos/anatomía & histología , Huesos/fisiología , Glicoproteínas/fisiología , Hormonas/fisiología , Músculo Esquelético/anatomía & histología , Músculo Esquelético/fisiología , Animales , Composición Corporal , Constitución Corporal , Densidad Ósea , Desarrollo Óseo , Matriz Ósea/metabolismo , Calcificación Fisiológica , Calcio/sangre , Cartílago/metabolismo , Femenino , Expresión Génica , Glicoproteínas/genética , Crecimiento/genética , Placa de Crecimiento/anatomía & histología , Hormonas/genética , Masculino , Ratones , Ratones Transgénicos , Microscopía Electrónica , Neovascularización Fisiológica , Osteoclastos/fisiología , Cráneo/diagnóstico por imagen , Tomografía Computarizada por Rayos X
19.
Physiol Genomics ; 10(1): 13-20, 2002 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-12118101

RESUMEN

The objective of this study was to use gene expression data from well-defined cell culture models, in combination with expression data from diagnostic samples of human diseased tissues, to identify potential therapeutic targets and markers of disease. Using Affymetrix oligonucleotide array technology, we identified a common profile of genes upregulated during endothelial morphogenesis into tubelike structures in three in vitro models of angiogenesis. Rigorous data selection criteria were used to identify a list of over 1,000 genes whose expression was increased more than twofold over baseline at either 4, 8, 24, 40 or 50 h. To further refine and prioritize this list, we used standard bioinformatic algorithms to identify potential transmembrane and secreted proteins. We then overlapped this gene set with genes upregulated in colon tumors vs. normal colon, resulting in a subset of 128 genes in common with our endothelial list. We removed from this list those genes expressed in 6 different colon tumor lines, resulting in a list of 24 putative, vascular-specific angiogenesis-associated genes. Three genes, gp34, stanniocalcin-1 (STC-1), and GA733-1, were expressed at levels 10-fold or more in colon tumors compared with normal mucosa. We validated the vascular-specific expression of one of these genes, STC-1, by in situ hybridization. The ability to combine in vitro and in vivo data sets should permit one to identify putative angiogenesis target genes in various tumors, chronic inflammation, and other disorders where therapeutic manipulation of angiogenesis is a desirable treatment modality.


Asunto(s)
Bases de Datos Genéticas , Perfilación de la Expresión Génica/métodos , Neovascularización Patológica/genética , Neovascularización Fisiológica/genética , Animales , Línea Celular , Endotelio Vascular/química , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiología , Marcación de Gen/métodos , Humanos , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Ratas , Ratas Sprague-Dawley , Células Tumorales Cultivadas , Venas Umbilicales
20.
Ann Biomed Eng ; 30(3): 297-304, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12051615

RESUMEN

The present study investigated the proliferative and apoptotic responses of human umbilical vein endothelial cells (HUVECs) to well-defined, sinusoidal pressures (60/20, 100/60, and 140/100 mm Hg/mm Hg) at 1 Hz for up to 24 h under Media 199 containing either 1% FBS and 0.04% bovine brain extract (BBE) (low serum/growth factor conditions) or 10% FBS and 0.4% BBE (normal serum/growth factor conditions). Controls were HUVEC maintained under 0.2 mm Hg sustained pressure, but otherwise, similar experimental conditions. Under low serum/growth factor conditions, exposure of HUVEC to 60/20 mm Hg/mm Hg cyclic pressure at 1 Hz for time periods up to 24 h resulted in increases in total cell population density, apoptosis, and DNA synthesis. Under normal serum/growth factor conditions, exposure of HUVEC to either 60/20 or 100/60 mm Hg/mm Hg cyclic pressures resulted in increased DNA synthesis but did not significantly affect cell density or the apoptotic index. A reduced rate of cell death was observed in HUVEC under low serum/growth factor conditions after exposure to 140/100 mm Hg/mm Hg. Under normal serum/growth factor conditions. HUVEC exposed to 140/100 mm Hg/mm Hg cyclic pressure exhibited reduced DNA synthesis. Endothelial cells. therefore, sense and respond to physiologic levels of cyclic pressure by modifying cell proliferation and apoptosis in a mean-pressure-selective manner.


Asunto(s)
Apoptosis/fisiología , Endotelio Vascular/citología , Endotelio Vascular/fisiología , Mecanotransducción Celular/fisiología , Recuento de Células , Células Cultivadas , ADN/biosíntesis , Humanos , Periodicidad , Presión , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Venas Umbilicales/citología , Venas Umbilicales/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA