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










Base de datos
Intervalo de año de publicación
1.
Behav Pharmacol ; 33(6): 377-394, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35947066

RESUMEN

Drugs targeting mu opioid receptors are the mainstay of clinical practice for treating moderate-to-severe pain. While they can offer excellent analgesia, their use can be limited by adverse effects, including constipation, respiratory depression, tolerance, and abuse liability. Multifunctional ligands acting at mu opioid and nociceptin/orphanin FQ peptide receptors might provide antinociception with substantially improved adverse-effect profiles. This study explored one of these ligands, OREX-1038 (BU10038), in several assays in rodents and nonhuman primates. Binding and functional studies confirmed OREX-1038 to be a low-efficacy agonist at mu opioid and nociceptin/orphanin FQ peptide receptors and an antagonist at delta and kappa opioid receptors with selectivity for opioid receptors over other proteins. OREX-1038 had long-acting antinociceptive effects in postsurgical and complete Freund's adjuvant (CFA)-induced thermal hyperalgesia assays in rats and a warm water tail-withdrawal assay in monkeys. OREX-1038 was active for at least 24 h in each antinociception assay, and its effects in monkeys did not diminish over 22 days of daily administration. This activity was coupled with limited effects on physiological signs (arterial pressure, heart rate, and body temperature) and no evidence of withdrawal after administration of naltrexone or discontinuation of treatment in monkeys receiving OREX-1038 daily. Over a range of doses, OREX-1038 was only transiently self-administered, which diminished rapidly to nonsignificant levels; overall, both OREX-1038 and buprenorphine maintained less responding than remifentanil. These results support the concept of dual mu and nociceptin/orphanin FQ peptide receptor partial agonists having improved pharmacological profiles compared with opioids currently used to treat pain.


Asunto(s)
Analgésicos Opioides , Dolor , Analgésicos Opioides/efectos adversos , Animales , Isoquinolinas , Naltrexona/análogos & derivados , Dolor/tratamiento farmacológico , Fenilpropionatos , Ratas , Receptores Opioides/agonistas , Receptores Opioides mu/agonistas
2.
Int J Obes (Lond) ; 43(10): 2085-2094, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30664661

RESUMEN

BACKGROUND/OBJECTIVES: Prolonged-release (PR) naltrexone 32 mg/bupropion 360 mg (NB) is approved for chronic weight management as an adjunct to reduced-calorie diet and increased physical activity. Central nervous system-active medications have the potential to affect mood; therefore, post hoc analysis of clinical trial data was conducted to evaluate psychiatric adverse events (PAEs) and effects on mood of NB therapy versus placebo. SUBJECTS/METHODS: Data were pooled from 5 prospective, double-blind, randomized, placebo-controlled clinical trials (duration range, 24-56 weeks) of NB in subjects with overweight or obesity. PAEs were collected via AE preferred terms, organized into major subtopics (e.g., anxiety, depression, sleep disorders), and divided into category terms (e.g., anxiety, potential anxiety symptoms). Additionally, the Inventory of Depressive Symptomatology Self Report (IDS-SR; score range 0-84) and the Columbia Classification Algorithm of Suicide Assessment (C-CASA) evaluated treatment-emergent depressive/anxiety symptoms and suicidal behavior/ideation, respectively. RESULTS: Baseline characteristics and comorbidities were comparable for placebo (n = 1515) and NB (n = 2545). Most common PAEs in the NB group (using category grouping; NB vs placebo) were sleep disorders (12.7 vs 7.9%, P < 0.001), anxiety (5.4 vs 3.3%, P = 0.029), and depression (1.8 vs 2.7%, P = 0.014); PAEs were more frequent during dose escalation and generally mild or moderate. Mean (SD) changes in IDS-SR total score from baseline to endpoint were small in both groups: 0.13 (5.83) for NB and -0.45 (5.65) for placebo. Retrospective AE categorization via C-CASA confirmed no completed suicides, suicide attempts, or preparatory acts toward imminent suicidal behavior. CONCLUSIONS: This large pooled analysis of 5 clinical trials provides additional safety information about the NB PAE profile. Anxiety and sleep disorder-related PAEs were more frequent with NB versus placebo but were mostly mild to moderate and generally occurred early. Depression-related PAEs were less common with NB than placebo, and NB was not associated with suicidal ideation or behavior in this patient population.


Asunto(s)
Fármacos Antiobesidad/efectos adversos , Bupropión/efectos adversos , Trastornos del Humor/inducido químicamente , Naltrexona/efectos adversos , Obesidad/tratamiento farmacológico , Sobrepeso/tratamiento farmacológico , Fármacos Antiobesidad/uso terapéutico , Bupropión/uso terapéutico , Método Doble Ciego , Combinación de Medicamentos , Humanos , Naltrexona/uso terapéutico , Estudios Prospectivos , Ensayos Clínicos Controlados Aleatorios como Asunto
3.
BMC Gastroenterol ; 18(1): 3, 2018 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-29304731

RESUMEN

BACKGROUND: Pathogenesis in non-alcoholic steatohepatitis (NASH) involves abnormal cholesterol metabolism and hepatic accumulation of toxic free cholesterol. Apical sodium-dependent bile acid transporter (ASBT) inhibition in the terminal ileum may facilitate removal of free cholesterol from the liver by reducing recirculation of bile acids (BAs) to the liver, thereby stimulating new BA synthesis from cholesterol. The aim of this phase 1 study in adult healthy volunteers (HVs) and patients with type 2 diabetes mellitus (T2DM) was to assess the safety, tolerability, pharmacokinetics and pharmacodynamics of ASBT inhibition with volixibat (SHP626; formerly LUM002). METHODS: Participants were randomised 3:1 to receive once-daily oral volixibat (0.5 mg, 1 mg, 5 mg or 10 mg) or placebo for 28 days in two cohorts (HV and T2DM). Assessments included safety, faecal BA and serum 7α-hydroxy-4-cholesten-3-one (C4; BA synthesis biomarker). RESULTS: Sixty-one individuals were randomised (HVs: placebo, n = 12; volixibat, n = 38; T2DM: placebo, n = 3; volixibat, n = 8). No deaths or treatment-related serious adverse events were reported. Mild or moderate gastrointestinal adverse events were those most frequently reported with volixibat. With volixibat, mean total faecal BA excretion on day 28 was ~1.6-3.2 times higher in HVs (643.73-1239.3 µmol/24 h) and ~8 times higher in T2DM (1786.0 µmol/24 h) than with placebo (HVs: 386.93 µmol/24 h; T2DM: 220.00 µmol/24 h). With volixibat, mean C4 concentrations increased by ~1.3-5.3-fold from baseline to day 28 in HVs and by twofold in T2DM. CONCLUSIONS: Volixibat was generally well tolerated. Increased faecal BA excretion and serum C4 levels support the mechanistic rationale for exploring ASBT inhibition in NASH. The study was registered with the Dutch clinical trial authority (Centrale Commissie Mensgebonden Onderzoek; trial registration number NL44732.056.13; registered 24 May 2013).


Asunto(s)
Benzotiepinas/administración & dosificación , Benzotiepinas/efectos adversos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Glicósidos/administración & dosificación , Glicósidos/efectos adversos , Glicoproteínas de Membrana/antagonistas & inhibidores , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Transportadores de Anión Orgánico Sodio-Dependiente/antagonistas & inhibidores , Simportadores/antagonistas & inhibidores , Adolescente , Adulto , Anciano , Benzotiepinas/farmacocinética , Ácidos y Sales Biliares/análisis , Ácidos y Sales Biliares/metabolismo , Glucemia/metabolismo , Colestenonas/sangre , Diabetes Mellitus Tipo 2/metabolismo , Método Doble Ciego , Heces/química , Femenino , Glicósidos/farmacocinética , Homeostasis , Humanos , Metabolismo de los Lípidos , Masculino , Persona de Mediana Edad , Adulto Joven
4.
Stem Cell Reports ; 4(6): 1075-88, 2015 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-26004631

RESUMEN

To gain insight into the cellular and molecular cues that promote neurovascular co-patterning at the earliest stages of human embryogenesis, we developed a human embryonic stem cell model to mimic the developing epiblast. Contact of ectoderm-derived neural cells with mesoderm-derived vasculature is initiated via the neural crest (NC), not the neural tube (NT). Neurovascular co-patterning then ensues with specification of NC toward an autonomic fate requiring vascular endothelial cell (EC)-secreted nitric oxide (NO) and direct contact with vascular smooth muscle cells (VSMCs) via T-cadherin-mediated homotypic interactions. Once a neurovascular template has been established, NT-derived central neurons then align themselves with the vasculature. Our findings reveal that, in early human development, the autonomic nervous system forms in response to distinct molecular cues from VSMCs and ECs, providing a model for how other developing lineages might coordinate their co-patterning.


Asunto(s)
Vasos Sanguíneos/fisiología , Células Madre Embrionarias Humanas/citología , Neuronas/metabolismo , Animales , Cadherinas/antagonistas & inhibidores , Cadherinas/genética , Cadherinas/metabolismo , Técnicas de Cultivo de Célula , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Técnicas de Cocultivo , Ectodermo/citología , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Humanos , Inmunohistoquímica , Masculino , Mesodermo/citología , Ratones , Ratones Noqueados , Modelos Biológicos , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Cresta Neural/citología , Cresta Neural/metabolismo , Neuronas/citología , Óxido Nítrico/metabolismo , Donantes de Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Periferinas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Tubulina (Proteína)/metabolismo
5.
Curr Protoc Stem Cell Biol ; 29: 1C.16.1-13, 2014 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-24838913

RESUMEN

The umbilical cord is tissue that is normally discarded after the delivery of the infant, but it has been shown to be a rich source of stem cells from the cord blood, Wharton's jelly, and umbilical endothelial cells. Patient-specific human induced pluripotent stem cells (hiPSCs) reprogrammed from patient specific human umbilical vein endothelial cells in the neonatal intensive care unit (NICU) population (specifically, premature neonates) have not been shown in the literature. This unit describes a protocol for the generation and expansion of hiPSCs originating from umbilical cords collected from patients in the NICU.


Asunto(s)
Diferenciación Celular , Técnicas de Reprogramación Celular/métodos , Células Endoteliales de la Vena Umbilical Humana , Células Madre Pluripotentes Inducidas , Cordón Umbilical , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Recién Nacido , Cordón Umbilical/citología , Cordón Umbilical/metabolismo
6.
Dev Cell ; 22(1): 146-57, 2012 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-22264731

RESUMEN

Endothelial cells (ECs) form cell-cell adhesive junctional structures maintaining vascular integrity. This barrier is dynamically regulated by vascular endothelial growth factor (VEGF) receptor signaling. We created an inducible knockin mouse model to study the contribution of the integrin-associated focal adhesion tyrosine kinase (FAK) signaling on vascular function. Here we show that genetic or pharmacological FAK inhibition in ECs prevents VEGF-stimulated permeability downstream of VEGF receptor or Src tyrosine kinase activation in vivo. VEGF promotes tension-independent FAK activation, rapid FAK localization to cell-cell junctions, binding of the FAK FERM domain to the vascular endothelial cadherin (VE-cadherin) cytoplasmic tail, and direct FAK phosphorylation of ß-catenin at tyrosine-142 (Y142) facilitating VE-cadherin-ß-catenin dissociation and EC junctional breakdown. Kinase inhibited FAK is in a closed conformation that prevents VE-cadherin association and limits VEGF-stimulated ß-catenin Y142 phosphorylation. Our studies establish a role for FAK as an essential signaling switch within ECs regulating adherens junction dynamics.


Asunto(s)
Permeabilidad Capilar/fisiología , Movimiento Celular/fisiología , Endotelio Vascular/metabolismo , Quinasa 1 de Adhesión Focal/fisiología , Neovascularización Fisiológica , Factor A de Crecimiento Endotelial Vascular/metabolismo , Uniones Adherentes/metabolismo , Animales , Antígenos CD/metabolismo , Cadherinas/metabolismo , Adhesión Celular , Comunicación Celular , Células Cultivadas , Endotelio Vascular/citología , Femenino , Adhesiones Focales/fisiología , Corazón/fisiología , Integrasas/metabolismo , Pulmón/citología , Pulmón/metabolismo , Masculino , Ratones , Fosforilación , Transducción de Señal , Tirosina/metabolismo , beta Catenina/metabolismo , Familia-src Quinasas/metabolismo
7.
Mol Cancer Ther ; 10(6): 972-82, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21518727

RESUMEN

Although nanoparticle-based drug delivery formulations can improve the effectiveness and safety of certain anticancer drugs, many drugs, due to their chemical composition, are unsuitable for nanoparticle loading. Here, we describe a targeted nanogel drug delivery platform that can (i) encapsulate a wide range of drug chemotypes, including biological, small molecule, and cytotoxic agents; (ii) display targeting ligands and polymeric coatings on the surface; (iii) enhance drug retention within the nanogel core after photo-cross-linking; and (iv) retain therapeutic activity after lyophilization allowing for long-term storage. For therapeutic studies, we used integrin αvß3-targeted lipid-coated nanogels with cross-linked human serum albumin in the core for carrying therapeutic cargoes. These particles exhibited potent activity in tumor cell viability assays with drugs of distinct chemotype, including paclitaxel, docetaxel, bortezomib, 17-AAG, sorafenib, sunitinib, bosutinib, and dasatinib. Treatment of orthotopic breast and pancreas tumors in mice with taxane-loaded nanogels produced a 15-fold improvement in antitumor activity relative to Abraxane by blocking both primary tumor growth and spontaneous metastasis. With a modifiable surface and core, the lipid-coated nanogel represents a platform technology that can be easily adapted for specific drug delivery applications to treat a wide range of malignant diseases.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Sistemas de Liberación de Medicamentos/métodos , Neoplasias Pancreáticas/tratamiento farmacológico , Polietilenglicoles/administración & dosificación , Polietilenglicoles/química , Polietileneimina/administración & dosificación , Polietileneimina/química , Paclitaxel Unido a Albúmina , Albúminas/administración & dosificación , Animales , Antineoplásicos/química , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Liofilización/métodos , Humanos , Integrina alfaVbeta3/metabolismo , Lípidos/química , Ratones , Ratones Desnudos , Nanogeles , Paclitaxel/administración & dosificación , Neoplasias Pancreáticas/metabolismo , Polietilenglicoles/síntesis química , Polietileneimina/síntesis química , Polímeros/química , Taxoides/administración & dosificación
8.
Nat Med ; 16(8): 909-14, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20676106

RESUMEN

Although it is well established that tumors initiate an angiogenic switch, the molecular basis of this process remains incompletely understood. Here we show that the miRNA miR-132 acts as an angiogenic switch by targeting p120RasGAP in the endothelium and thereby inducing neovascularization. We identified miR-132 as a highly upregulated miRNA in a human embryonic stem cell model of vasculogenesis and found that miR-132 was highly expressed in the endothelium of human tumors and hemangiomas but was undetectable in normal endothelium. Ectopic expression of miR-132 in endothelial cells in vitro increased their proliferation and tube-forming capacity, whereas intraocular injection of an antagomir targeting miR-132, anti-miR-132, reduced postnatal retinal vascular development in mice. Among the top-ranking predicted targets of miR-132 was p120RasGAP, which we found to be expressed in normal but not tumor endothelium. Endothelial expression of miR-132 suppressed p120RasGAP expression and increased Ras activity, whereas a miRNA-resistant version of p120RasGAP reversed the vascular response induced by miR-132. Notably, administration of anti-miR-132 inhibited angiogenesis in wild-type mice but not in mice with an inducible deletion of Rasa1 (encoding p120RasGAP). Finally, vessel-targeted nanoparticle delivery of anti-miR-132 restored p120RasGAP expression in the tumor endothelium, suppressed angiogenesis and decreased tumor burden in an orthotopic xenograft mouse model of human breast carcinoma. We conclude that miR-132 acts as an angiogenic switch by suppressing endothelial p120RasGAP expression, leading to Ras activation and the induction of neovascularization, whereas the application of anti-miR-132 inhibits neovascularization by maintaining vessels in the resting state.


Asunto(s)
Endotelio Vascular/patología , MicroARNs/fisiología , Neovascularización Patológica/genética , Proteína Activadora de GTPasa p120/genética , Animales , Anticuerpos Monoclonales/farmacología , Proliferación Celular , Células Cultivadas , Evaluación Preclínica de Medicamentos , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Endotelio Vascular/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , MicroARNs/metabolismo , Neovascularización Patológica/metabolismo , Interferencia de ARN/fisiología , ARN Interferente Pequeño/farmacología , Arteria Retiniana/efectos de los fármacos , Arteria Retiniana/metabolismo , Arteria Retiniana/patología , Regulación hacia Arriba/genética , Regulación hacia Arriba/fisiología , Proteína Activadora de GTPasa p120/metabolismo
9.
Proc Natl Acad Sci U S A ; 107(9): 4299-304, 2010 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-20154271

RESUMEN

Kinases are known to regulate fundamental processes in cancer including tumor proliferation, metastasis, neovascularization, and chemoresistance. Accordingly, kinase inhibitors have been a major focus of drug development, and several kinase inhibitors are now approved for various cancer indications. Typically, kinase inhibitors are selected via high-throughput screening using catalytic kinase domains at low ATP concentration, and this process often yields ATP mimetics that lack specificity and/or function poorly in cells where ATP levels are high. Molecules targeting the allosteric site in the inactive kinase conformation (type II inhibitors) provide an alternative for developing selective inhibitors that are physiologically active. By applying a rational design approach using a constrained amino-triazole scaffold predicted to stabilize kinases in the inactive state, we generated a series of selective type II inhibitors of PDGFRbeta and B-RAF, important targets for pericyte recruitment and endothelial cell survival, respectively. These molecules were designed in silico and screened for antivascular activity in both cell-based models and a Tg(fli1-EGFP) zebrafish embryogenesis model. Dual inhibition of PDGFRbeta and B-RAF cellular signaling demonstrated synergistic antiangiogenic activity in both zebrafish and murine models of angiogenesis, and a combination of previously characterized PDGFRbeta and RAF inhibitors validated the synergy. Our lead compound was selected as an orally active molecule with favorable pharmacokinetic properties which demonstrated target inhibition in vivo leading to suppression of murine orthotopic tumors in both the kidney and pancreas.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Carcinoma de Células Renales/patología , División Celular/efectos de los fármacos , Neoplasias Renales/patología , Neovascularización Patológica , Neoplasias Pancreáticas/patología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Administración Oral , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/metabolismo , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Pez Cebra
10.
J Pharmacol Exp Ther ; 328(3): 758-65, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19056934

RESUMEN

Phosphatidylinositol 3-kinases (PI3Ks) are key elements in the signaling cascades that lie downstream of many cellular receptors. In particular, PI3K delta and gamma isoforms contribute to inflammatory cell recruitment and subsequent activation. For this reason, in a series of preclinical studies, we tested the potential of a recently developed small-molecule inhibitor of these two isoforms, TG100-115 [3-[2,4-diamino-6-(3-hydroxyphenyl)pteridin-7-yl]phenol], as a form of anti-inflammatory therapy for respiratory diseases such as asthma and chronic obstructive pulmonary disease (COPD). To determine pharmacokinetic profiles, aerosolized formulations of the drug were delivered to mice by a nose-only inhalation route, yielding high pulmonary TG100-115 levels with minimal systemic exposure. Safety assessments were favorable, with no clinical or histological changes noted after 21 days of daily dosing. In a murine asthma model, aerosolized TG100-115 markedly reduced the pulmonary eosinophilia and the concomitant interleukin-13 and mucin accumulation characteristic of this disease. As a functional benefit, interventional dosing schedules of this inhibitor also reduced airway hyper-responsiveness. To model the pulmonary neutrophilia characteristic of COPD, mice were exposed to either intranasal lipopolysaccharide or inhaled smoke. Aerosolized TG100-115 again inhibited these inflammatory patterns, most notably in the smoke model, where interventional therapy overcame the steroid-resistant nature of the pulmonary inflammation. In conclusion, aerosolized TG100-115 displays pharmacokinetic, safety, and biological activity profiles favorable for further development as a therapy for both asthma and COPD. Furthermore, these studies support the hypothesis that PI3K delta and gamma are suitable molecular targets for these diseases.


Asunto(s)
Antiinflamatorios/uso terapéutico , Asma/tratamiento farmacológico , Fenoles/uso terapéutico , Pteridinas/uso terapéutico , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Administración Intranasal , Aerosoles , Animales , Antiinflamatorios/administración & dosificación , Hiperreactividad Bronquial/tratamiento farmacológico , Fosfatidilinositol 3-Quinasa Clase Ib , Modelos Animales de Enfermedad , Isoenzimas/metabolismo , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/toxicidad , Ratones , Neutrófilos/efectos de los fármacos , Neutrófilos/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo
11.
Cancer Cell ; 14(6): 429-30, 2008 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-19061833

RESUMEN

Infantile hemangiomas represent the most common tumor of endothelial cell (EC) origin, yet the mechanisms regulating hemangioma EC behavior are poorly understood. A new study by Jinnin et al. demonstrates that enhanced VEGFR2 signaling in hemangioma ECs is caused by suppression of NFAT (nuclear factor of activated T cells)-dependent VEGFR1 expression.


Asunto(s)
Hemangioma/metabolismo , Factores de Transcripción NFATC/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Movimiento Celular , Proliferación Celular , Humanos , Integrinas/metabolismo , Modelos Biológicos , Transducción de Señal , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
12.
Nature ; 456(7223): 809-13, 2008 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-18997771

RESUMEN

Angiogenesis does not only depend on endothelial cell invasion and proliferation: it also requires pericyte coverage of vascular sprouts for vessel stabilization. These processes are coordinated by vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF) through their cognate receptors on endothelial cells and vascular smooth muscle cells (VSMCs), respectively. PDGF induces neovascularization by priming VSMCs/pericytes to release pro-angiogenic mediators. Although VEGF directly stimulates endothelial cell proliferation and migration, its role in pericyte biology is less clear. Here we define a role for VEGF as an inhibitor of neovascularization on the basis of its capacity to disrupt VSMC function. Specifically, under conditions of PDGF-mediated angiogenesis, VEGF ablates pericyte coverage of nascent vascular sprouts, leading to vessel destabilization. At the molecular level, VEGF-mediated activation of VEGF-R2 suppresses PDGF-Rbeta signalling in VSMCs through the assembly of a previously undescribed receptor complex consisting of PDGF-Rbeta and VEGF-R2. Inhibition of VEGF-R2 not only prevents assembly of this receptor complex but also restores angiogenesis in tissues exposed to both VEGF and PDGF. Finally, genetic deletion of tumour cell VEGF disrupts PDGF-Rbeta/VEGF-R2 complex formation and increases tumour vessel maturation. These findings underscore the importance of VSMCs/pericytes in neovascularization and reveal a dichotomous role for VEGF and VEGF-R2 signalling as both a promoter of endothelial cell function and a negative regulator of VSMCs and vessel maturation.


Asunto(s)
Vasos Sanguíneos/metabolismo , Neovascularización Fisiológica/fisiología , Pericitos/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Inhibidores de la Angiogénesis/farmacología , Animales , Línea Celular , Células Cultivadas , Fibrosarcoma/irrigación sanguínea , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Neovascularización Fisiológica/efectos de los fármacos , Pericitos/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/farmacología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Transducción de Señal
14.
J Cell Biol ; 180(1): 101-12, 2008 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-18195103

RESUMEN

Prohibitin 1 (PHB1) is a highly conserved protein that is mainly localized to the inner mitochondrial membrane and has been implicated in regulating mitochondrial function in yeast. Because mitochondria are emerging as an important regulator of vascular homeostasis, we examined PHB1 function in endothelial cells. PHB1 is highly expressed in the vascular system and knockdown of PHB1 in endothelial cells increases mitochondrial production of reactive oxygen species via inhibition of complex I, which results in cellular senescence. As a direct consequence, both Akt and Rac1 are hyperactivated, leading to cytoskeletal rearrangements and decreased endothelial cell motility, e.g., migration and tube formation. This is also reflected in an in vivo angiogenesis assay, where silencing of PHB1 blocks the formation of functional blood vessels. Collectively, our results provide evidence that PHB1 is important for mitochondrial function and prevents reactive oxygen species-induced senescence and thereby maintains the angiogenic capacity of endothelial cells.


Asunto(s)
Senescencia Celular , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Mitocondrias/fisiología , Neovascularización Fisiológica , Proteínas Represoras/fisiología , Animales , Aorta/citología , Aorta/metabolismo , Células Cultivadas , Citoesqueleto/metabolismo , Complejo I de Transporte de Electrón/genética , Complejo I de Transporte de Electrón/metabolismo , Células Endoteliales/citología , Endotelio Vascular/citología , Humanos , Ratones , Ratones Endogámicos , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Neovascularización Fisiológica/genética , Neuropéptidos/genética , Neuropéptidos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Prohibitinas , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal , Proteínas de Unión al GTP rac/genética , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1
15.
Blood ; 111(5): 2674-80, 2008 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-18180379

RESUMEN

Semaphorin 3A (Sema3A), a known inhibitor of axonal sprouting, also alters vascular patterning. Here we show that Sema3A selectively interferes with VEGF- but not bFGF-induced angiogenesis in vivo. Consistent with this, Sema3A disrupted VEGF- but not bFGF-mediated endothelial cell signaling to FAK and Src, key mediators of integrin and growth factor signaling; however, signaling to ERK by either growth factor was unperturbed. Since VEGF is also a vascular permeability (VP) factor, we examined the role of Sema3A on VEGF-mediated VP in mice. Surprisingly, Sema3A not only stimulated VEGF-mediated VP but also potently induced VP in the absence of VEGF. Sema3A-mediated VP was inhibited either in adult mice expressing a conditional deletion of endothelial neuropilin-1 (Nrp-1) or in wild-type mice systemically treated with a function-blocking Nrp-1 antibody. While both Sema3A- and VEGF-induced VP was Nrp-1 dependent, they use distinct downstream effectors since VEGF- but not Sema3A-induced VP required Src kinase signaling. These findings define a novel role for Sema3A both as a selective inhibitor of VEGF-mediated angiogenesis and a potent inducer of VP.


Asunto(s)
Permeabilidad Capilar/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Semaforina-3A/farmacología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/farmacología , Animales , Células Cultivadas , Pollos , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Inhibidores Enzimáticos/farmacología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Humanos , Ratones , Ratones Endogámicos BALB C , Neuropilina-1/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo
16.
J Biol Chem ; 282(42): 30745-53, 2007 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-17702744

RESUMEN

Myoferlin and dysferlin are members of the ferlin family of membrane proteins. Recent studies have shown that mutation or genetic disruption of myoferlin or dysferlin promotes muscular dystrophy-related phenotypes in mice, which are the result of impaired plasma membrane integrity. However, no biological functions have been ascribed to myoferlin in non-muscle tissues. Herein, using a proteomic analysis of endothelial cell (EC) caveolae/lipid raft microdomains we identified myoferlin in these domains and show that myoferlin is highly expressed in ECs and vascular tissues. The loss of myoferlin results in lack of proliferation, migration, and nitric oxide (NO) release in response to vascular endothelial growth factor (VEGF). Western blotting and surface biotinylation experiments show that loss of myoferlin reduces the expression level and autophosphorylation of VEGF receptor-2 (VEGFR-2) in native ECs. In a reconstituted cell system, transfection of myoferlin increases VEGFR-2 membrane expression and autophosphorylation in response to VEGF. In vivo, VEGFR-2 levels and VEGF-induced permeability are impaired in myoferlin-deficient mice. Mechanistically, myoferlin forms a complex with dynamin-2 and VEGFR-2, which prevents CBL-dependent VEGFR-2 polyubiquitination and proteasomal degradation. These data are the first to report novel biological activities for myoferlin and reveal the role of membrane integrity to VEGF signaling.


Asunto(s)
Caveolas/metabolismo , Células Endoteliales/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Musculares/metabolismo , Procesamiento Proteico-Postraduccional , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Proteínas de Unión al Calcio , Permeabilidad Capilar/efectos de los fármacos , Permeabilidad Capilar/genética , Caveolas/patología , Línea Celular , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Dinamina II/genética , Dinamina II/metabolismo , Disferlina , Células Endoteliales/patología , Eliminación de Gen , Humanos , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Proteínas Musculares/genética , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/patología , Óxido Nítrico/genética , Óxido Nítrico/metabolismo , Especificidad de Órganos/efectos de los fármacos , Especificidad de Órganos/genética , Fenotipo , Fosforilación/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Procesamiento Proteico-Postraduccional/genética , Proteómica , Proteínas Proto-Oncogénicas c-cbl/genética , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Ubiquitina/genética , Ubiquitina/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
17.
Cancer Res ; 67(6): 2766-72, 2007 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-17363598

RESUMEN

Genotoxic stress induced by anticancer drugs can lead to apoptosis of both angiogenic endothelial cells (ECs) and proliferating tumor cells. However, growth factors such as basic fibroblast growth factor (bFGF) and vascular endothelial cell growth factor (VEGF) present within the tumor microenvironment can promote chemoresistance by suppressing apoptotic mechanisms in these cells. Here, we have identified apoptosis signal-regulating kinase 1 (ASK1), a proapoptotic member of the MAP3K family, as a target of bFGF-mediated survival signaling in ECs. Evidence is provided that ASK1 is required for EC apoptosis in response to the genotoxic chemotherapeutic agent doxorubicin, and that bFGF, but not VEGF, neutralizes the death-promoting activity of ASK1. Specifically, bFGF stimulation promotes the formation of a Raf-1/ASK1 complex at the mitochondria, inhibits ASK1 kinase activity, and protects ECs from genotoxic stress. Mutation of the Raf-1 activation domain (SS338/9AA) not only prevents Raf-1/ASK1 complex formation but abolishes bFGF-mediated EC protection from genotoxic stress. In line with these observations, bFGF, but not VEGF, neutralizes the antiangiogenic effects of doxorubicin in vivo. These findings reveal a new pathway of EC survival signaling and define a molecular mechanism for chemoresistance induced by bFGF.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Factor 2 de Crecimiento de Fibroblastos/farmacología , MAP Quinasa Quinasa Quinasa 5/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-raf/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Células Endoteliales/citología , Humanos , MAP Quinasa Quinasa Quinasa 5/metabolismo , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica/efectos de los fármacos , Proteínas Proto-Oncogénicas c-raf/genética , Fracciones Subcelulares/enzimología , Fracciones Subcelulares/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología
18.
Proc Natl Acad Sci U S A ; 103(29): 10997-1002, 2006 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-16835300

RESUMEN

Nogo isoforms (Nogo-A and -B) have been implicated in regulating neural and cardiovascular functions, such as cell spreading and chemotaxis. Unlike the loop domain (Nogo-66) found in all Nogo isoforms that can interact with a neural-specific Nogo-66 receptor, the receptor for the amino terminus of Nogo-B that mediates vascular function is unknown. Here, we identify a previously uncharacterized Nogo-B receptor specific for the amino terminus of Nogo-B and show that Nogo-B receptor localizes with the ligand Nogo-B during VEGF and wound healing angiogenesis in vivo, mediates chemotaxis in a heterologous expression system and chemotaxis, and 3D tube formation in native endothelial cells. Thus, identification of this receptor may lead to the discovery of agonists or antagonists of this pathway to regulate vascular remodeling and angiogenesis.


Asunto(s)
Forma de la Célula , Quimiotaxis , Células Endoteliales/citología , Células Endoteliales/metabolismo , Proteínas de la Mielina/metabolismo , Receptores de Superficie Celular/metabolismo , Secuencia de Aminoácidos , Animales , Células Cultivadas , Chlorocebus aethiops , Cricetinae , Humanos , Ratones , Datos de Secuencia Molecular , Proteínas de la Mielina/genética , Proteínas de la Mielina/farmacología , Proteínas Nogo , Unión Proteica , Isoformas de Proteínas/metabolismo , Receptores de Superficie Celular/química
19.
J Biol Chem ; 279(29): 30349-57, 2004 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-15136572

RESUMEN

The heterogeneous localization of endothelial nitricoxide synthase (eNOS) on the Golgi complex versus the plasma membrane has made it difficult to dissect the regulation of each pool of enzyme. Here, we generated fusion proteins that specifically target the plasma membrane or cytoplasmic aspects of the Golgi complex and have assessed eNOS activation. Plasma membrane-targeted eNOS constructs were constitutively active, phosphorylated, and responsive to transmembrane calcium fluxes, yet were insensitive to further activation by Akt-mediated phosphorylation. In contrast, cis-Golgi complex-targeted eNOS behaved similarly to wild-type eNOS and was less sensitive to calcium-dependent activation and highly responsive to Akt-dependent phosphorylation compared with plasma membrane versions. In plasma membrane- and Golgi complex-targeted constructs, Ser1179 is critical for NO production. This study provides clear evidence for functional roles of plasma membrane- and Golgi complex-localized eNOS and supports the concept that proteins thought to be regulated and to function exclusively in the plasma membrane of cells can indeed signal and be regulated in internal Golgi membranes.


Asunto(s)
Calcio/metabolismo , Membrana Celular/metabolismo , Citoplasma/enzimología , Aparato de Golgi/enzimología , Óxido Nítrico Sintasa/biosíntesis , Óxido Nítrico/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Western Blotting , Células COS , Calcio/química , Calmodulina/química , Cisteína/química , Endotelio Vascular/metabolismo , Aparato de Golgi/metabolismo , Humanos , Microscopía Fluorescente , Ácidos Mirísticos/metabolismo , Óxido Nítrico Sintasa/química , Óxido Nítrico Sintasa de Tipo III , Ácidos Palmíticos/metabolismo , Fosforilación , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-akt , Serina/química , Transfección , Venas Umbilicales/metabolismo
20.
Nat Med ; 10(4): 382-8, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15034570

RESUMEN

Although Nogo-A has been identified in the central nervous system as an inhibitor of axonal regeneration, the peripheral roles of Nogo isoforms remain virtually unknown. Here, using a proteomic analysis to identify proteins enriched in caveolae and/or lipid rafts (CEM/LR), we show that Nogo-B is highly expressed in cultured endothelial and smooth muscle cells, as well as in intact blood vessels. The N terminus of Nogo-B promotes the migration of endothelial cells but inhibits the migration of vascular smooth muscle (VSM) cells, processes necessary for vascular remodeling. Vascular injury in Nogo-A/B-deficient mice promotes exaggerated neointimal proliferation, and adenoviral-mediated gene transfer of Nogo-B rescues the abnormal vascular expansion in those knockout mice. Our discovery that Nogo-B is a regulator of vascular homeostasis and remodeling broadens the functional scope of this family of proteins.


Asunto(s)
Endotelio Vascular/fisiología , Músculo Liso Vascular/fisiología , Proteínas de la Mielina/fisiología , Animales , Línea Celular , Movimiento Celular , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Humanos , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Proteínas de la Mielina/metabolismo , Proteínas Nogo , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA