Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Clin Endocrinol Metab ; 100(4): 1699-708, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25664604

RESUMEN

BACKGROUND: A novel oral octreotide formulation was tested for efficacy and safety in a phase III, multicenter, open-label, dose-titration, baseline-controlled study in patients with acromegaly. METHODS: We enrolled 155 complete or partially controlled patients (IGF-1 <1.3 × upper limit of normal [ULN], and 2-h integrated GH <2.5 ng/mL) receiving injectable somatostatin receptor ligand (SRL) for ≥ 3 months. Subjects were switched to 40 mg/d oral octreotide capsules (OOCs), and the dose escalated to 60 and then up to 80 mg/d to control IGF-1. Subsequent fixed doses were maintained for a 7-month core treatment, followed by a voluntary 6-month extension. RESULTS: Of 151 evaluable subjects initiating OOCs, 65% maintained response and achieved the primary endpoint of IGF-1 <1.3 × ULN and mean integrated GH <2.5 ng/mL at the end of the core treatment period and 62% at the end of treatment (up to 13 mo). The effect was durable, and 85 % of subjects initially controlled on OOCs maintained this response up to 13 months. When controlled on OOCs, GH levels were reduced compared to baseline, and acromegaly-related symptoms improved. Of 102 subjects completing the core treatment, 86% elected to enroll in the 6-month extension. Twenty-six subjects who were considered treatment failures (IGF-1 ≥ 1.3 × ULN) terminated early, and 23 withdrew for adverse events, consistent with those known for octreotide or disease related. CONCLUSIONS: OOC, an oral therapeutic peptide, achieves efficacy in controlling IGF-1 and GH after switching from injectable SRLs for up to 13 months, with a safety profile consistent with approved SRLs. OOC appears to be effective and safe as an acromegaly monotherapy.


Asunto(s)
Acromegalia/tratamiento farmacológico , Antineoplásicos Hormonales/administración & dosificación , Antineoplásicos Hormonales/efectos adversos , Octreótido/administración & dosificación , Octreótido/efectos adversos , Acromegalia/metabolismo , Adenoma/tratamiento farmacológico , Adenoma/metabolismo , Administración Oral , Adulto , Anciano , Antineoplásicos Hormonales/farmacocinética , Relación Dosis-Respuesta a Droga , Femenino , Adenoma Hipofisario Secretor de Hormona del Crecimiento/tratamiento farmacológico , Adenoma Hipofisario Secretor de Hormona del Crecimiento/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Persona de Mediana Edad , Octreótido/farmacocinética , Resultado del Tratamiento
2.
Pathology ; 46(5): 416-23, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24977735

RESUMEN

Neuropilin-1 (NRP1) is a receptor for vascular endothelial growth factor (VEGF). A soluble isoform of Nrp1 (sNrp1) has not been described in the mouse. Our goal was to examine the expression of mouse sNrp1 during liver development and regeneration.sNrp1 was cloned from mouse liver. The expression of sNrp1 and VEGF was examined in mouse liver during post-natal development and regeneration using northern blot, western blot, in situ hybridisation, and immunohistochemical analyses. HGF/NRP1 binding was examined in vitro.A novel 588-amino acid sNrp1 isoform was found to contain the ligand binding regions of Nrp1. The adult liver expressed more sNrp1 than full-length Nrp1. In vivo, hepatocytes constitutively expressed VEGF and sNrp1 in the quiescent state. sNrp1 was highly up-regulated at P20, a time point coinciding with a plateau in liver and body weights. Following hepatectomy, endogenous levels of sNrp1 decreased during the rapid growth phase, and VEGF levels were highest just prior to and during the angiogenic phase. sNrp1 levels again rose 5-10 days post-hepatectomy, presumably to control regeneration. HGF protein bound NRP1 and binding was competed with sNRP1.We cloned a novel mouse sNrp1 isoform from liver and provide evidence that this endogenous angiogenesis inhibitor may regulate VEGF or HGF bioavailability during normal physiological growth and development as well as during liver regeneration.


Asunto(s)
Regeneración Hepática/fisiología , Hígado/crecimiento & desarrollo , Hígado/metabolismo , Neovascularización Fisiológica/fisiología , Neuropilina-1/biosíntesis , Animales , Secuencia de Bases , Northern Blotting , Western Blotting , Humanos , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Datos de Secuencia Molecular , Homología de Secuencia de Ácido Nucleico , Factor A de Crecimiento Endotelial Vascular/biosíntesis
3.
Pharm Res ; 31(8): 2010-21, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24558008

RESUMEN

PURPOSE: Medium chain fatty acid salts promote absorption by increasing paracellular permeability of the intestinal epithelium. Novel oily suspension (OS) formulation disperses a powder containing sodium caprylate and macromolecules such as octreotide or fluorescent dextran (FD). Formulation safety, macromolecule absorption and pharmacokinetic (PK)/pharmacodynamic (PD) were evaluated. METHODS: Octreotide/OS toxicity was evaluated in monkeys following 9 months of daily oral enteric-coated capsule administration. The OS permeation effect was also assessed in rats, using FD/OS and octreotide/OS preparations. Octreotide/OS effects on circulating growth hormone (GH) levels were also measured. RESULTS: Safety assessment of octreotide/OS in monkeys after 9 months showed minor drug-related findings, comparable to the injectable octreotide. Octreotide exposure levels were similar across the treatment periods. In rats, OS facilitated FD permeation up to 70 kDa in a reversible, spatial and dose-dependent manner, independent of the intestinal dosing site. Following OS administration, the staining pattern of the tight-junction protein, ZO-1, changed transiently, and a paracellular penetration marker, LC-biotin, permeated between adjacent epithelial cells. Enteral octreotide/OS absorption was dose-dependent and suppressed rat GH levels. CONCLUSIONS: Oral octreotide/OS dosing was shown to be safe in monkeys. OS enhances intestinal absorption of active octreotide, likely by transient alteration of the tight junction protein complex.


Asunto(s)
Absorción Intestinal/fisiología , Sustancias Macromoleculares/química , Sustancias Macromoleculares/metabolismo , Octreótido/química , Octreótido/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Química Farmacéutica/métodos , Femenino , Absorción Intestinal/efectos de los fármacos , Macaca fascicularis , Sustancias Macromoleculares/farmacología , Masculino , Octreótido/farmacología , Ratas , Ratas Sprague-Dawley , Comprimidos Recubiertos
4.
MAbs ; 2(2): 199-208, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20190562

RESUMEN

CT-322 is a new anti-angiogenic therapeutic agent based on an engineered variant of the tenth type III domain of human fibronectin, i.e., an Adnectin™, designed to inhibit vascular endothelial growth factor receptor (VEGFR)-2. This PE Gylated Adnectin was developed using an mRNA display technology. CT-322 bound human VEGFR-2 with high affinity (K(D), 11 nM), but did not bind VEGFR-1 or VEGFR-3 at concentrations up to 100 nM, as determined by surface plasmon resonance studies. Western blot analysis showed that CT-322 blocked VEGF-induced phosphorylation of VEGFR-2 and mitogen-activated protein kinase in human umbilical vascular endothelial cells. CT-322 significantly inhibited the growth of human tumor xenograft models of colon carcinoma and glioblastoma at doses of 15-60 mg/kg administered 3 times/week. Anti-tumor effects of CT-322 were comparable to those of sorafenib or sunitinib, which inhibit multiple kinases, in a colon carcinoma xenograft model, although CT-322 caused less overt adverse effects than the kinase inhibitors. CT-322 also enhanced the anti-tumor activity of the chemotherapeutic agent temsirolimus in the colon carcinoma model. The high affinity and specificity of CT-322 binding to VEGFR-2 and its anti-tumor activities establish CT-322 as a promising anti-angiogenic therapeutic agent. Our results further suggest that Adnectins are an important new class of targeted biologics that can be developed as potential treatments for a wide variety of diseases.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antineoplásicos/farmacología , Carcinoma/tratamiento farmacológico , Neoplasias del Colon/tratamiento farmacológico , Endotelio Vascular/efectos de los fármacos , Fibronectinas/farmacología , Glioblastoma/tratamiento farmacológico , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Carcinoma/patología , Línea Celular Tumoral , Neoplasias del Colon/patología , Técnicas Químicas Combinatorias , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Fibronectinas/genética , Fibronectinas/metabolismo , Glioblastoma/patología , Humanos , Ratones , Unión Proteica/efectos de los fármacos , Ingeniería de Proteínas , Resonancia por Plasmón de Superficie , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
5.
BMC Cancer ; 8: 352, 2008 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-19038046

RESUMEN

BACKGROUND: Pancreatic cancer continues to have a 5-year survival of less than 5%. Therefore, more effective therapies are necessary to improve prognosis in this disease. Angiogenesis is required for tumor growth, and subsequently, mediators of angiogenesis are attractive targets for therapy. Vascular endothelial growth factor (VEGF) is a well-characterized mediator of tumor angiogenesis that functions primarily by binding and activating VEGF receptor 2 (VEGFR2). In this study, we evaluate the use of CT-322, a novel biologic (Adnectin). This small protein is based on a human fibronectin domain and has beneficial properties in that it is fully human, stable, and is produced in bacteria. CT-322 binds to and inhibits activation of VEGFR2. METHODS: The efficacy of CT-322 was evaluated in vivo using two orthotopic pancreatic tumor models. The first model was a human tumor xenograft where MiaPaCa-2 cells were injected into the tail of the pancreas of nude mice. The second model was a syngeneic tumor using Pan02 cells injected into pancreas of C57BL/6J mice. In both models, therapy was initiated once primary tumors were established. Mice bearing MiaPaCa-2 tumors were treated with vehicle or CT-322 alone. Gemcitabine alone or in combination with CT-322 was added to the treatment regimen of mice bearing Pan02 tumors. Therapy was given twice a week for six weeks, after which the animals were sacrificed and evaluated (grossly and histologically) for primary and metastatic tumor burden. Primary tumors were also evaluated by immunohistochemistry for the level of apoptosis (TUNEL), microvessel density (MECA-32), and VEGF-activated blood vessels (Gv39M). RESULTS: Treatment with CT-322 was effective at preventing pancreatic tumor growth and metastasis in orthotopic xenograft and syngeneic models of pancreatic cancer. Additionally, CT-322 treatment increased apoptosis, reduced microvessel density and reduced the number of VEGF-activated blood vessels in tumors. Finally, CT-322, in combination with gemcitabine was safe and effective at controlling the growth of syngeneic pancreatic tumors in immunocompetent mice. CONCLUSION: We conclude that CT-322 is an effective anti-VEGFR2 agent and that further investigation of CT-322 for the treatment of pancreatic cancer is warranted.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Antineoplásicos/uso terapéutico , Neovascularización Patológica/tratamiento farmacológico , Neoplasias Pancreáticas/tratamiento farmacológico , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Inhibidores de la Angiogénesis/farmacología , Animales , Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Línea Celular Tumoral , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Desoxicitidina/uso terapéutico , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Necrosis , Metástasis de la Neoplasia/tratamiento farmacológico , Neoplasias Pancreáticas/irrigación sanguínea , Neoplasias Pancreáticas/patología , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
6.
Angiogenesis ; 8(3): 217-27, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16328161

RESUMEN

Neuropilin 1 (NRP1) is a co-receptor for vascular endothelial growth factor (VEGF(165)), an inducer of vascular permeability and angiogenesis. Numerous physiological factors enhance VEGF expression and function but only a few have been shown to be negative regulators. Previously, we have shown that the naturally occurring soluble form of NRP1 (sNRP1) inhibits binding of VEGF(165) to endothelial cells in vitro and impairs tumor growth in vivo. To investigate the role of sNRP1 in the regulation of vascular development and function, sNRP1 expression was targeted to the skin, where it is not normally expressed, using a keratin 14 (K14) promoter expression construct. K14-sNRP1 transgenic mice displayed normal skin architecture with a subtle abnormal vascular phenotype. While the overall number of skin blood vessels remained unchanged, the lumen size of smooth muscle-associated dermal vessels was reduced. K14-sNRP1 mice had reduced vascular permeability in response to VEGF(165), but also to VEGF(121) and platelet activating factor, suggesting that the lack of permeability was not solely due to the sequestration of VEGF. sNRP1 also reversed the increase in inflammation and edema induced by transgenic VEGF overexpression in cutaneous delayed-type hypersensitivity reactions. In summary, sNRP1 appears to primarily regulate vessel permeability while its effect on physiological angiogenesis is less evident in this model.


Asunto(s)
Permeabilidad Capilar/fisiología , Neovascularización Fisiológica/fisiología , Neuropilina-1/metabolismo , Piel/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Northern Blotting , Western Blotting , Vectores Genéticos/genética , Inmunohistoquímica , Queratina-14 , Queratinas/genética , Ratones , Ratones Transgénicos , Piel/irrigación sanguínea
7.
Cancer Cell ; 7(3): 251-61, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15766663

RESUMEN

Angiogenesis inhibitors, such as TNP-470 and the nontoxic HPMA copolymer-TNP-470 (caplostatin), are emerging as a class of anticancer drugs. We report that TNP-470 and caplostatin inhibit vascular hyperpermeability of tumor blood vessels as well as that induced in mouse skin by different mediators. Treatment with TNP-470 or angiostatin for 3 days was sufficient to reduce permeability of tumor blood vessels, delayed-type hypersensitivity, and pulmonary edema induced by IL-2. TNP-470 also inhibited VPF/VEGF-induced phosphorylation of VEGFR-2, calcium influx, and RhoA activation in endothelial cells. These results identify an activity of TNP-470, that of inhibiting vessel hyperpermeability. This activity likely contributes to TNP-470's antiangiogenic effect and suggests that caplostatin can be used in the treatment of cancer and inflammation.


Asunto(s)
Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/farmacología , Capilares/efectos de los fármacos , Permeabilidad Capilar/efectos de los fármacos , Sesquiterpenos/química , Sesquiterpenos/farmacología , Angiostatinas/farmacología , Animales , Calcio/metabolismo , Capilares/metabolismo , Capilares/ultraestructura , Permeabilidad Capilar/fisiología , Movimiento Celular/efectos de los fármacos , Ciclohexanos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/ultraestructura , Femenino , Hipersensibilidad Tardía , Interleucina-2/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neoplasias/irrigación sanguínea , Neoplasias/metabolismo , Neoplasias/patología , O-(Cloroacetilcarbamoil) Fumagilol , Edema Pulmonar/inducido químicamente , Piel/irrigación sanguínea , Piel/efectos de los fármacos , Piel/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
8.
FASEB J ; 18(15): 1952-4, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15479766

RESUMEN

Vascular endothelial growth factor (VEGF) is a potent regulator of vascular function through its control of multiple endothelial cell functions. In addition to its key role in vascularization, VEGF has recently been shown to have neurotrophic activity during hypoxic stress. In the central and peripheral motor nervous system, VEGF treatment increased neuronal vascularization and perfusion, as well as having direct trophic effects on neurons and Schwann cells. However, the role of VEGF in the sensory nervous system remains unclear. To characterize the differential effects of VEGF on endothelial cells and neurons in sensory ganglia, we used explanted mouse dorsal root ganglia (DRG), a culture system containing neurons and endothelial cells in close apposition. We show that VEGF is expressed by neurons and satellite cells, but not by endothelial cells or pericytes. On the other hand, the tyrosine kinase VEGF receptor VEGFR-2 was robustly expressed by endothelial cells throughout the extensive DRG capillary network, but not found at either the transcript or protein level in sensory neurons or other nonendothelial cells of the DRG. Both soluble receptor sequestration of VEGF and small molecule kinase inhibition of VEGFR-2 signaling rapidly disrupted the connectivity, branching, and structural integrity of the capillary network of embryonic DRG; this effect was no longer evident postnatally. However, VEGF inhibition showed no detectable effect on neuronal health at any stage analyzed. These data suggest that endogenous VEGF is a strict requirement for vascular, but not neuronal, maintenance in developing sensory ganglia.


Asunto(s)
Ganglios Espinales/irrigación sanguínea , Ganglios Espinales/embriología , Neuronas/metabolismo , Factor A de Crecimiento Endotelial Vascular/fisiología , Animales , Axones/fisiología , Capilares/embriología , Endotelio Vascular/embriología , Endotelio Vascular/metabolismo , Ganglios Espinales/citología , Ratones , Células Satélites Perineuronales/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
9.
Development ; 131(9): 2161-71, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15073147

RESUMEN

To directly examine the role of vascular endothelial growth factor (VEGFA) in cartilage development, we conditionally knocked out Vegfa in chondrocytes, using the Col2a1 promoter to drive expression of Cre recombinase. Our study of Vegfa conditional knockout (CKO) mice provides new in-vivo evidence for two important functions of VEGFA in bone formation. First, VEGFA plays a significant role in both early and late stages of cartilage vascularization, since Vegfa CKO mice showed delayed invasion of blood vessels into primary ossification centers and delayed removal of terminal hypertrophic chondrocytes. Second, VEGFA is crucial for chondrocyte survival, since massive cell death was seen in joint and epiphyseal regions of Vegfa CKO endochondral bones. Chondrocytes in these regions were found to upregulate expression of Vegfa in wild-type mice at the time when massive cell death occurred in the Vegfa CKO mice. The expression of the VEGFA receptors Npr1 and Npr2 in epiphyseal chondrocytes and lack of blood vessel reduction in the vicinity of the cartilaginous elements in the Vegfa CKO mice raise the possibility that the observed cell death is the result of a direct involvement of VEGFA in chondrocyte survival. Interestingly, the extensive cell death seen in Vegfa CKO null bones had a striking similarity to the cell death phenotype observed when hypoxia-inducible factor 1 alpha (Hif1a) expression was abolished in developing cartilage. This similarity of cell death phenotypes and the deficient VEGFA production in Hif1a null epiphyseal chondrocytes demonstrate that HIF1 alpha and VEGFA are components of a key pathway to support chondrocyte survival during embryonic bone development.


Asunto(s)
Desarrollo Óseo/fisiología , Cartílago/crecimiento & desarrollo , Supervivencia Celular , Condrocitos/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Apoptosis/fisiología , Huesos/citología , Huesos/metabolismo , Calcificación Fisiológica , Cartílago/irrigación sanguínea , Cartílago/citología , Diferenciación Celular , Condrocitos/citología , Embrión de Mamíferos/anatomía & histología , Embrión de Mamíferos/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia , Ratones , Ratones Noqueados , Neovascularización Fisiológica , Embarazo , Factores de Transcripción/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética
10.
Dev Biol ; 255(1): 77-98, 2003 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-12618135

RESUMEN

Peripheral nerves and blood vessels have similar patterns in quail forelimb development. Usually, nerves extend adjacent to existing blood vessels, but in a few cases, vessels follow nerves. Nerves have been proposed to follow vascular smooth muscle, endothelium, or their basal laminae. Focusing on the major axial blood vessels and nerves, we found that when nerves grow into forelimbs at E3.5-E5, vascular smooth muscle was not detectable by smooth muscle actin immunoreactivity. Additionally, transmission electron microscopy at E5.5 confirmed that early blood vessels lacked smooth muscle and showed that the endothelial cell layer lacks a basal lamina, and we did not observe physical contact between peripheral nerves and these endothelial cells. To test more generally whether lack of nerves affected blood vessel patterns, forelimb-level neural tube ablations were performed at E2 to produce aneural limbs; these had completely normal vascular patterns up to at least E10. To test more generally whether vascular perturbation affected nerve patterns, VEGF(165), VEGF(121), Ang-1, and soluble Flt-1/Fc proteins singly and in combination were focally introduced via beads implanted into E4.5 forelimbs. These produced significant alterations to the vascular patterns, which included the formation of neo-vessels and the creation of ectopic avascular spaces at E6, but in both under- and overvascularized forelimbs, the peripheral nerve pattern was normal. The spatial distribution of semaphorin3A protein immunoreactivity was consistent with a negative regulation of neural and/or vascular patterning. Semaphorin3A bead implantations into E4.5 forelimbs caused failure of nerves and blood vessels to form and to deviate away from the bead. Conversely, semaphorin3A antibody bead implantation was associated with a local increase in capillary formation. Furthermore, neural tube electroporation at E2 with a construct for the soluble form of neuropilin-1 caused vascular malformations and hemorrhage as well as altered nerve trajectories and peripheral nerve defasciculation at E5-E6. These results suggest that neurovascular congruency does not arise from interdependence between peripheral nerves and blood vessels, but supports the hypothesis that it arises by a shared patterning mechanism that utilizes semaphorin3A.


Asunto(s)
Vasos Sanguíneos/embriología , Tipificación del Cuerpo , Embrión no Mamífero/irrigación sanguínea , Miembro Anterior/irrigación sanguínea , Miembro Anterior/embriología , Neuropilina-1/metabolismo , Semaforina-3A/metabolismo , Angiotensina II/farmacología , Animales , Vasos Sanguíneos/anomalías , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/ultraestructura , Células Cultivadas , Cricetinae , Técnicas de Cultivo , Embrión no Mamífero/ultraestructura , Factores de Crecimiento Endotelial/farmacología , Proteínas de la Matriz Extracelular/farmacología , Miembro Anterior/efectos de los fármacos , Miembro Anterior/inervación , Miembro Anterior/ultraestructura , Péptidos y Proteínas de Señalización Intercelular/farmacología , Linfocinas/farmacología , Fragmentos de Péptidos/farmacología , Nervios Periféricos/irrigación sanguínea , Nervios Periféricos/embriología , Nervios Periféricos/ultraestructura , Codorniz , Semaforina-3A/farmacología , Factor A de Crecimiento Endotelial Vascular , Receptor 1 de Factores de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
11.
J Biol Chem ; 277(27): 24818-25, 2002 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-11986311

RESUMEN

Neuroplin-1 (NRP1), a receptor for vascular endothelial growth factor (VEGF) family members, has three distinct extracellular domains, a1a2, b1b2, and c. To determine the VEGF(165) and placenta growth factor 2 (PlGF-2)-binding sites of NRP1, recombinant NRP1 domains were expressed in mammalian cells as Myc-tagged, soluble proteins, and used in co-precipitation experiments with 125I-VEGF165 and 125I-PlGF-2. Anti-Myc antibodies immunoprecipitated 125I-VEGF165 and 125I-PlGF-2 in the presence of the b1b2 but not of the a1a2 and c domains. Neither b1 nor b2 alone was capable of binding 125I-VEGF165. In competition experiments, VEGF165 competed PlGF-2 binding to the NRP1 b1b2 domain, suggesting that the binding sites of VEGF165 and PlGF-2 overlap. The presence of the a1a2 domain greatly enhanced VEGF165, but not PlGF-2 binding to b1b2. Heparin enhanced the binding of both 125I-VEGF165 and 125I-PlGF-2 to the b1b2 domain by 20- and 4-fold, respectively. A heparin chain of at least 20-24 monosaccharides was necessary for binding. In addition, the b1b2 domain of NRP1 could bind heparin directly, requiring heparin oligomers of at least 8 monosaccharide units. It was concluded that an intact b1b2 domain serves as the VEGF165-, PlGF-2-, and heparin-binding sites in NRP1, and that heparin is a critical component for regulating VEGF165 and PlGF-2 interactions with NRP1 by physically interacting with both receptor and ligands.


Asunto(s)
Factores de Crecimiento Endotelial/metabolismo , Heparina/metabolismo , Linfocinas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Gestacionales/metabolismo , Inductores de la Angiogénesis/química , Inductores de la Angiogénesis/metabolismo , Secuencia de Bases , Sitios de Unión , Neoplasias de la Mama , Células Cultivadas , Clonación Molecular , Cartilla de ADN , Factores de Crecimiento Endotelial/química , Endotelio Vascular/metabolismo , Heparina/química , Humanos , Cinética , Linfocinas/química , Proteínas del Tejido Nervioso/química , Neuropilina-1 , Factor de Crecimiento Placentario , Proteínas Gestacionales/química , Proteínas Recombinantes/metabolismo , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
12.
Adv Exp Med Biol ; 515: 33-48, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12613541

RESUMEN

Neuropilin-1 (NRP1) and NRP2 are related transmembrane receptors that function as mediators of neuronal guidance and angiogenesis. NRPs bind members of the class 3 semaphorin family, regulators of neuronal guidance, and of the vascular endothelial growth factor (VEGF) family of angiogenesis factors. There is substantial evidence that NRPs serve as mediators of developmental and tumor angiogenesis. NRPs are expressed in endothelial cells (EC) and bind VEGF165. NRP1 is a co-receptor for VEGF receptor-2 (VEGFR2) that enhances the binding of VEGF165 to VEGFR2 and VEGF165-mediated chemotaxis. NRP1 expression is regulated in EC by tumor necrosis factor-alpha, the transcription factors dHAND and Ets-1, and vascular injury. During avian blood vessel development NRP1 is expressed only in arteries whereas NRP2 is expressed in veins. Transgenic mouse models demonstrate that NRP1 plays a critical role in embryonic vascular development. Overexpression of NRP1 results in the formation of excess capillaries and hemorrhaging. NRP1 knockouts have defects in yolk sac, embryo and neuronal vascularization, and in development of large vessels in the heart. Tumor cells express NRPs and bind VEGF165. NRP1 upregulation is positively correlated with the progression of various tumors. Overexpression of NRPI in rat tumor cells results in enlarged tumors and substantially enhanced tumor angiogenesis. On the other hand, soluble NRP1 (sNRP1) is an antagonist of tumor angiogenesis. Semaphorin 3A binds to EC and tumor cells. It also inhibits EC motility and capillary sprouting in vitro. VEGF165 and Sema3A are competitive inhibitors for NRP1 mediated functions in EC and neurons. These results suggest that NRP1 is a novel regulator of the vascular system.


Asunto(s)
Vasos Sanguíneos/fisiología , Neoplasias/fisiopatología , Neuropilinas/fisiología , Animales , Vasos Sanguíneos/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Humanos , Neoplasias/metabolismo , Neovascularización Patológica/patología , Neuropilinas/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA