Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Curr Oncol Rep ; 21(3): 22, 2019 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-30806847

RESUMEN

PURPOSE OF REVIEW: The purpose of this review is to discuss the current understanding of the Tie2-angiopoietin system and its role in tumor growth and metastasis. This review also focuses on preclinical and clinical data published to date that have evaluated Tie2-angiopoietin inhibition. RECENT FINDINGS: Tie2 inhibition has shown significant promise in preclinical models, notable for decreased tumor burden and fewer sites of metastatic disease across various malignancies. However, data from human clinical trials have shown more mixed results. Trebananib, rebastanib, and MEDI3617 are the three Tie2-angiopoietin inhibitors that have been most widely evaluated in phase I and II trials. Further investigation into these therapies is ongoing. The Tie2-angiopoietin pathway continues to show promise in preclinical and some clinical trials, including studies on recurrent or metastatic breast and renal cell carcinomas. Further evaluation of these therapies, however, is warranted to better understand their optimal clinical utility.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Angiopoyetina 1/antagonistas & inhibidores , Angiopoyetina 2/antagonistas & inhibidores , Ensayos Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Animales , Humanos , Neoplasias/irrigación sanguínea , Neoplasias/metabolismo , Neoplasias/patología , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología
2.
Stem Cells ; 35(7): 1849-1859, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28376568

RESUMEN

Microvesicles (MVs) derived from human mesenchymal stem cells (MSC MVs) were demonstrated to ameliorate inflammation in lungs. We have found their content of mRNA for keratinocyte growth factor was partly involved in their therapeutic effects. As MSC MVs also contained a substantial quantity of angiopoietin-1 (Ang-1) mRNA, which plays an essential role in vascular stabilization and resolving inflammation, we hypothesized that Ang-1 mRNA might similarly account for a part of their therapeutic effects. We downregulated Ang-1 mRNA expression in MVs, using a lentivirus vector carrying Ang-1 short hairpin RNA to transfect MSCs. A mouse model of lipopolysaccharide induced acute lung injury (ALI) was used in vivo. We also studied in vitro interactions between Ang-1 mRNA deficient MVs on macrophages and human lung microvascular endothelial cells. Compared with negative control, Ang-1 mRNA deficient MVs increased the influx of neutrophils and macrophage inflammatory protein-2 levels in bronchoalveolar lavage fluid by 136% and 105%, respectively, suggesting a deteriorative lung inflammation and a failure to restore pulmonary capillary permeability assessed by Evan's blue dye and bronchoalveolar lavage albumin level. In vitro, the addition of Ang-1 mRNA deficient MVs failed to maintain the integrity of endotoxin-stimulated microvascular endothelial cells and abrogated the decrease in tumor necrosis factor-α level and the increase in interleukin-10 level mediated by negative control in RAW 264.7 cells. In summary, the therapeutic effects of MVs in ALI, and their immunomodulatory properties on macrophages were partly mediated through their content of Ang-1 mRNA. Stem Cells 2017;35:1849-1859.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Angiopoyetina 1/genética , Micropartículas Derivadas de Células/metabolismo , Pulmón/metabolismo , Células Madre Mesenquimatosas/metabolismo , ARN Mensajero/genética , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/patología , Angiopoyetina 1/antagonistas & inhibidores , Angiopoyetina 1/metabolismo , Animales , Líquido del Lavado Bronquioalveolar/química , Permeabilidad Capilar/genética , Micropartículas Derivadas de Células/química , Micropartículas Derivadas de Células/trasplante , Quimiocina CXCL2/genética , Quimiocina CXCL2/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/citología , Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Humanos , Interleucina-10/genética , Interleucina-10/metabolismo , Lipopolisacáridos , Pulmón/patología , Masculino , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila/genética , Neutrófilos/metabolismo , Neutrófilos/patología , Cultivo Primario de Células , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
3.
Lancet ; 388(10043): 518-29, 2016 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-26853587

RESUMEN

Angiogenesis, the formation of new blood vessels from pre-existing vessels, has been validated as a target in several tumour types through randomised trials, incorporating vascular endothelial growth factor (VEGF) pathway inhibitors into the therapeutic armoury. Although some tumours such as renal cell carcinoma, ovarian and cervical cancers, and pancreatic neuroendocrine tumours are sensitive to these drugs, others such as prostate cancer, pancreatic adenocarcinoma, and melanoma are resistant. Even when drugs have yielded significant results, improvements in progression-free survival, and, in some cases, overall survival, are modest. Thus, a crucial issue in development of these drugs is the search for predictive biomarkers-tests that predict which patients will, and will not, benefit before initiation of therapy. Development of biomarkers is important because of the need to balance efficacy, toxicity, and cost. Novel combinations of these drugs with other antiangiogenics or other classes of drugs are being developed, and the appreciation that these drugs have immunomodulatory and other modes of action will lead to combination regimens that capitalise on these newly understood mechanisms.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Biomarcadores de Tumor/metabolismo , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Inhibidores de la Angiogénesis/farmacología , Angiopoyetina 1/antagonistas & inhibidores , Biomarcadores de Tumor/sangre , Supervivencia sin Enfermedad , Resistencia a Antineoplásicos , Femenino , Humanos , Terapia Molecular Dirigida/métodos , Neoplasias/irrigación sanguínea , Neoplasias/metabolismo , Neovascularización Patológica/prevención & control , Niacinamida/análogos & derivados , Niacinamida/uso terapéutico , Compuestos de Fenilurea/uso terapéutico , Ftalazinas/uso terapéutico , Piperazinas/uso terapéutico , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Quinazolinas/uso terapéutico , Quinolinas/uso terapéutico , Receptor TIE-2/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Sorafenib , Factor A de Crecimiento Endotelial Vascular/metabolismo
4.
Reprod Fertil Dev ; 28(6): 690-9, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25485810

RESUMEN

Ovarian hyperstimulation syndrome (OHSS) is a complication of ovarian stimulation with gonadotrophins following human chorionic gonadotrophin (hCG) administration. The relationship between hCG and OHSS is partly mediated via the production of angiogenic factors, such as vascular endothelial growth factor A (VEGFA) and angiopoietins (ANGPTs). Here, we investigated the effect of ANGPT1 inhibition on ovarian angiogenesis in follicular fluid (FF) from women at risk of OHSS, using the chorioallantoic membrane (CAM) of quail embryos as an experimental model. We also analysed cytoskeletal changes and endothelial junction protein expression induced by this FF in the presence or absence of an ANGPT1-neutralising antibody in endothelial cell cultures. The presence of this antibody restored the number of vascular branch points and integrin αvß3 levels in the CAMs to control values. ANGPT1 inhibition in FF from OHSS patients also restored the levels of claudin-5, vascular endothelial cadherin and phosphorylated ß-catenin and partially reversed actin redistribution in endothelial cells. Our findings suggest that ANGPT1 increases pathophysiological angiogenesis in patients at risk of OHSS by acting on tight and adherens junction proteins. Elucidating the mechanisms by which ANGPT1 regulates vascular development and cell-cell junctions in OHSS will contribute to identifying new therapeutic targets for the treatment of human diseases with aberrant vascular leakage.


Asunto(s)
Uniones Adherentes/metabolismo , Angiopoyetina 1/metabolismo , Endotelio Vascular/metabolismo , Neovascularización Patológica/etiología , Folículo Ovárico/metabolismo , Síndrome de Hiperestimulación Ovárica/fisiopatología , Uniones Estrechas/metabolismo , Uniones Adherentes/efectos de los fármacos , Uniones Adherentes/patología , Adulto , Angiopoyetina 1/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/farmacología , Argentina/epidemiología , Bioensayo , Biomarcadores/metabolismo , Línea Celular , Células Cultivadas , Membrana Corioalantoides/irrigación sanguínea , Membrana Corioalantoides/efectos de los fármacos , Membrana Corioalantoides/metabolismo , Coturnix , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Endotelio Vascular/fisiopatología , Femenino , Líquido Folicular/citología , Líquido Folicular/metabolismo , Humanos , Folículo Ovárico/irrigación sanguínea , Folículo Ovárico/efectos de los fármacos , Folículo Ovárico/patología , Síndrome de Hiperestimulación Ovárica/epidemiología , Síndrome de Hiperestimulación Ovárica/metabolismo , Síndrome de Hiperestimulación Ovárica/patología , Riesgo , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/patología
5.
Invest New Drugs ; 33(3): 691-9, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25895965

RESUMEN

BACKGROUND: Trebananib is an anti-angiogenic peptibody under investigation in patients with advanced cancer. This study evaluated the pharmacokinetic (PK) drug-drug interaction of paclitaxel and trebananib. PATIENTS AND METHODS: Patients with advanced solid tumors received weekly 80 mg/m(2) intravenous (IV) paclitaxel (3 weeks on/1 week off) with weekly 15 mg/kg IV trebananib starting at Week 2. Blood samples for PK analysis were collected at Week 1 (paclitaxel alone), Week 6 (paclitaxel and trebananib), and Week 8 (trebananib alone). An absence of interaction was to be concluded if the 90 % confidence intervals (CI) for the differences in paclitaxel exposure fell within the 0.80-1.25 interval. RESULTS: The primary study was conducted between 7/2012 and 10/2013. Thirty-five patients were enrolled and 34 received both treatments. Most patients were white (91 %) and female (59 %); mean age was 61 years. The most common tumor types were ovarian (32 %) and bladder (27 %), 71 % of patients had stage IV disease, and all had Eastern Cooperative Oncology Group (ECOG) scores of 0 or 1. PK parameter analysis was done on patients with evaluable PK data at both assessments (with and without concomitant therapy; n = 28). The geometric least squares mean (GLSM) ratio (90 % CI) of paclitaxel AUCinf with and without trebananib was 1.17 (1.10, 1.25). The GLSM ratio (90 % CI) of trebananib AUCtau,ss with and without paclitaxel was 0.92 (0.87, 0.97). The most common adverse events were fatigue, local edema, peripheral edema, and nausea. CONCLUSIONS: This study showed no evidence of clinically meaningful PK interaction between paclitaxel and trebananib.


Asunto(s)
Angiopoyetina 1/antagonistas & inhibidores , Angiopoyetina 2/antagonistas & inhibidores , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Paclitaxel/farmacocinética , Proteínas Recombinantes de Fusión/farmacocinética , Adulto , Anciano , Anciano de 80 o más Años , Angiopoyetina 1/metabolismo , Angiopoyetina 2/metabolismo , Anticuerpos Antineoplásicos/sangre , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/sangre , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Demografía , Interacciones Farmacológicas , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Paclitaxel/efectos adversos , Paclitaxel/sangre , Paclitaxel/uso terapéutico , Proteínas Recombinantes de Fusión/efectos adversos , Proteínas Recombinantes de Fusión/uso terapéutico , Resultado del Tratamiento
6.
Gynecol Oncol ; 138(3): 513-8, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26171911

RESUMEN

OBJECTIVES: Ang1 & 2 (angiopoietin-1; -2) interact with Tie2 receptors on endothelial cells to mediate vascular remodeling in an angiogenesis signaling pathway distinct from the VEGF axis. Trebananib is a peptide Fc fusion protein that binds Ang1 and 2 and prevents interaction with Tie2. The efficacy of trebananib in recurrent/persistent endometrial cancer (EC) was studied. METHODS: The primary objective was to determine the frequency of patients with objective tumor responses (ORR) and event-free survival for ≥6months (6-month EFS) and determine toxicity of trebananib at a dose and schedule of 15mg/kg, IV QW. Recurrent/persistent EC, measurable disease, and ≤2 prior chemotherapy lines were required. RESULTS: Thirty-two patients were eligible and treated. The most common histologies were G1/2 endometrioid (31%), G3 endometrioid (28%) and serous (31.3%). 78% of patients had 1 prior regimen. Patients received 1-9+ cycles of trebananib; 24 patients (75%) received ≤2cycles. One patient had a partial response (3.1%); 8 patients had stable disease (25%) and 5 patients (15.6%) had 6 month EFS. Median progression-free survival and overall-survival were 1.97 months (90% CI 1.77-2.1) and 6.6 months (90% CI 4.01-14.75), respectively. Most common adverse events (AEs) were fatigue, anemia, and GI issues. Grade 3 and 4 AEs were: GI 31 and 0%; vascular 22 and 0%; metabolism/nutrition 19 and 3%; and general (including edema) 16 and 0%. CONCLUSIONS: Trebananib has insufficient single agent activity in recurrent EC to warrant further investigation at this dose/schedule.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Neoplasias Endometriales/tratamiento farmacológico , Proteínas Recombinantes de Fusión/uso terapéutico , Adulto , Anciano , Inhibidores de la Angiogénesis/efectos adversos , Angiopoyetina 1/antagonistas & inhibidores , Angiopoyetina 1/metabolismo , Angiopoyetina 2/antagonistas & inhibidores , Angiopoyetina 2/metabolismo , Neoplasias Endometriales/irrigación sanguínea , Neoplasias Endometriales/metabolismo , Femenino , Humanos , Persona de Mediana Edad , Proteínas Recombinantes de Fusión/efectos adversos
7.
Lancet Oncol ; 15(8): 799-808, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24950985

RESUMEN

BACKGROUND: Angiogenesis is a valid target in the treatment of epithelial ovarian cancer. Trebananib inhibits the binding of angiopoietins 1 and 2 to the Tie2 receptor, and thereby inhibits angiogenesis. We aimed to assess whether the addition of trebananib to single-agent weekly paclitaxel in patients with recurrent epithelial ovarian cancer improved progression-free survival. METHODS: For this randomised, double-blind phase 3 study undertaken between Nov 10, 2010, and Nov 19, 2012, we enrolled women with recurrent epithelial ovarian cancer from 32 countries. Patient eligibility criteria included having been treated with three or fewer previous regimens, and a platinum-free interval of less than 12 months. We enrolled patients with a computerised interactive voice response system, and patients were randomly assigned using a permuted block method (block size of four) in a 1:1 ratio to receive weekly intravenous paclitaxel (80 mg/m(2)) plus either weekly masked intravenous placebo or trebananib (15 mg/kg). Patients were stratified on the basis of platinum-free interval (≥0 and ≤6 months vs >6 and ≤12 months), presence or absence of measurable disease, and region (North America, western Europe and Australia, or rest of world). The sponsor, investigators, site staff, and patients were masked to the treatment assignment. The primary endpoint was progression-free survival assessed in the intention-to-treat population. The trial is registered with ClinicalTrials.gov, NCT01204749, and is no longer accruing patients. FINDINGS: 919 patients were enrolled, of whom 461 were randomly assigned to the trebananib group and 458 to the placebo group. Median progression-free survival was significantly longer in the trebananib group than in the placebo group (7·2 months [5·8-7·4] vs 5·4 months [95% CI 4·3-5·5], respectively, hazard ratio 0·66, 95% CI 0·57-0·77, p<0·0001). Incidence of grade 3 or higher adverse events was similar between treatment groups (244 [54%] of 452 patients in the placebo group vs 258 [56%] of 461 patients in the trebananib group). Trebananib was associated with more adverse event-related treatment discontinuations than was placebo (77 [17%] patients vs 27 [6%], respectively) and higher incidences of oedema (294 [64%] patients had any-grade oedema in the trebananib group vs 127 [28%] patients in the placebo group). Grade 3 or higher adverse events included ascites (34 [8%] in the placebo group vs 52 [11%] in the trebananib group), neutropenia (40 [9%] vs 26 [6%]), and abdominal pain (21 [5%] vs 22 [5%]). We recorded serious adverse events in 125 (28%) patients in the placebo group and 159 (34%) patients in the trebananib group. There was a difference of 2% or less in class-specific adverse events associated with anti-VEGF therapy (hypertension, proteinuria, wound-healing complications, thrombotic events, gastrointestinal perforations), except bleeding, which was more common in the placebo group than in the trebananib group (75 [17%] vs 46 [10%]). INTERPRETATION: Inhibition of angiopoietins 1 and 2 with trebananib provided a clinically meaningful prolongation in progression-free survival. This non-VEGF anti-angiogenesis option for women with recurrent epithelial ovarian cancer should be investigated in other settings and in combination with additional agents. Although oedema was increased, typical anti-VEGF associated adverse events were not prominent. FUNDING: Amgen.


Asunto(s)
Inhibidores de la Angiogénesis/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Neoplasias Glandulares y Epiteliales/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Proteínas Recombinantes de Fusión/administración & dosificación , Anciano , Inhibidores de la Angiogénesis/efectos adversos , Angiopoyetina 1/antagonistas & inhibidores , Angiopoyetina 2/antagonistas & inhibidores , Carcinoma Epitelial de Ovario , Supervivencia sin Enfermedad , Método Doble Ciego , Edema/inducido químicamente , Femenino , Humanos , Persona de Mediana Edad , Paclitaxel/administración & dosificación , Proteínas Recombinantes de Fusión/efectos adversos , Privación de Tratamiento
8.
Pharmacol Res ; 80: 43-51, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24407281

RESUMEN

The growth factor angiopoietin-1 (Ang-1) plays an essential role in angiogenesis and vascular homeostasis. Nevertheless, the role of Ang-1 in regulating vascular tone and blood flow is largely unexplored. Endothelial nitric oxide synthase (eNOS) and the junctional protein VE-cadherin are part of the complex signalling cascade initiated by Ang-1 in endothelial cells. In this study, we aimed to investigate the mechanisms underlying acute effects of Ang-1 on microvascular reactivity, permeability and blood flow, and hypothesise that eNOS and VE-cadherin underpin Ang-1 mediated vascular effects that are independent of angiogenesis and proliferation. Myography of isolated microarterioles from male C3H/HeN mice (7-10 weeks) was employed to measure vascular reactivity in vitro. Microcirculatory function in vivo was evaluated by intravital microscopy and Doppler fluximetry in dorsal window chambers. Ang-1 and its stable variant MAT.Ang-1 induced a concentration-dependent vasodilation of arterioles in vitro, which was blocked with nitric oxide (NO) synthesis inhibitor l-NAME. In vivo, MAT.Ang-1 restored to control levels l-NAME induced peripheral vasoconstriction, decreased blood flow and microvascular hyperpermeability. Tissue protein expression of VE-cadherin was reduced by NOS inhibition and restored to control levels by MAT.Ang-1, whilst VE-cadherin phosphorylation was increased by l-NAME and subsequently reduced by MAT.Ang-1 administration. Moreover, MAT.Ang-1 alone did not modulate systemic levels of angiogenetic factors. Our novel findings report that Ang-1 induces arteriolar vasodilation via release of NO, suggesting that Ang-1 is an important regulator of microvascular tone. As MAT.Ang-1 ameliorates detrimental effects on the microcirculation induced by inhibition of NO synthesis and stabilizes the endothelial barrier function through VE-cadherin, we propose that this Ang-1 variant may serve as a novel therapeutic agent to protect the microcirculation against endothelial dysfunction.


Asunto(s)
Angiopoyetina 1/fisiología , Antígenos CD/fisiología , Cadherinas/fisiología , Permeabilidad Capilar/fisiología , Microcirculación/fisiología , Óxido Nítrico Sintasa de Tipo III/fisiología , Angiopoyetina 1/antagonistas & inhibidores , Angiopoyetina 1/farmacología , Animales , Antígenos CD/biosíntesis , Antígenos CD/efectos de los fármacos , Arteriolas/efectos de los fármacos , Arteriolas/fisiología , Cadherinas/biosíntesis , Cadherinas/efectos de los fármacos , Permeabilidad Capilar/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Masculino , Ratones , Microcirculación/efectos de los fármacos , Músculo Estriado/irrigación sanguínea , Músculo Estriado/efectos de los fármacos , Músculo Estriado/fisiología , NG-Nitroarginina Metil Éster/antagonistas & inhibidores , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación , Vasoconstricción/efectos de los fármacos , Vasoconstricción/fisiología , Vasodilatación/efectos de los fármacos , Vasodilatación/fisiología
9.
Glia ; 60(12): 1954-63, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22927341

RESUMEN

Vascular endothelial growth factors (VEGFs) and angiopoietins (ANGs) are involved in pathophysiological responses in damaged nerve tissues. Astrocytes produce VEGFs and ANGs upon brain ischemia and traumatic injury. To clarify the extracellular signals regulating VEGF and ANG production, effects of endothelins (ETs), a family of endothelium-derived peptides, were examined in cultured rat astrocytes. ET-1 (100 nM) and Ala(1,3,11,15)-ET-1 (100 nM), an ET(B) receptor agonist, increased VEGF-A mRNA levels in cultured astrocytes, while ANG-1 mRNA was decreased by ETs. ET-1 did not affect astrocytic VEGF-B, placental growth factor (PLGF), and ANG-2 mRNA levels. The effects of ET-1 on VEGF-A and ANG-1 mRNAs were inhibited by BQ788, an ET(B) antagonist. Release of VEGF-A proteins from cultured astrocytes was increased by ET-1. In contrast, ET-1 reduced release of astrocytic ANG-1. Exogenous ET-1 (100 nM) and VEGF(165) (100 ng/mL), an isopeptide of VEGF-A, stimulated bromodeoxyuridine (BrdU) incorporation into cultured astrocytes. Treatment with ET-1 and VEGF(165) increased the numbers of cyclin D1-positive astrocytes. Exogenous ANG-1 (250 ng/mL) did not stimulate the BrdU incorporation. Increases in BrdU incorporation by ET-1 and VEGF(165) were not affected by ANG-1. In 60-70% confluent cultures, SU4312 (10 µM), a VEGF receptor tyrosine kinase inhibitor, partially reduced the effects of ET-1 on BrdU incorporation and cyclin D1 expression. ET-induced BrdU incorporation and cyclin D1 expression were reduced by a neutralizing antibody against VEGF-A. Our findings suggest that ET-1 is a factor regulating astrocytic VEGF-A and ANG-1, and that increased VEGF-A production potentiates ET-induced astrocytic proliferation by an autocrine mechanism.


Asunto(s)
Angiopoyetina 1/biosíntesis , Astrocitos/metabolismo , Proliferación Celular , Endotelina-1/fisiología , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Angiopoyetina 1/antagonistas & inhibidores , Angiopoyetina 1/genética , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Comunicación Autocrina/efectos de los fármacos , Comunicación Autocrina/fisiología , Bromodesoxiuridina/antagonistas & inhibidores , Bromodesoxiuridina/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Endotelina-1/antagonistas & inhibidores , Endotelina-1/metabolismo , Oligopéptidos/farmacología , Piperidinas/farmacología , ARN Mensajero/biosíntesis , Ratas , Ratas Wistar , Regulación hacia Arriba/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/genética
10.
Pharm Res ; 29(4): 1057-65, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22189693

RESUMEN

PURPOSE: To investigate the mechanisms of clearance of AMG 386, an investigational recombinant peptide-Fc fusion protein (peptibody) that blocks tumor angiogenesis by neutralizing the interaction between angiopoietin-1 and -2 and the Tie2 receptor. METHODS: The role of the neonatal Fc receptor (FcRn) in AMG 386 clearance was assessed in wild-type and FcRn-knockout mice; the roles of the spleen and kidneys were assessed in splenectomized and 5/6th nephrectomized rats, respectively, compared with sham-operated rats. Animals were administered AMG 386 as a single intravenous dose of 3 or 10 mg/kg. Blood samples for pharmacokinetic analysis were collected periodically throughout a 504-hour postdose period. RESULTS: Compared with wild-type mice, AMG 386 clearance in FcRn-knockout mice was 18-fold faster at the 3-mg/kg dose (FcRn knockout, 13.2 mL/h/kg; wild-type, 0.728 mL/h/kg) and 14-fold faster at the 10-mg/kg dose (FcRn knockout, 10.7 mL/h/kg; wild-type, 0.777 mL/h/kg). Clearance in nephrectomized rats was slower than in sham-operated rats at both the 3-mg/kg dose (nephrectomized, 1.23 mL/h/kg; sham-operated, 1.75 mL/h/kg) and the 10-mg/kg dose (nephrectomized, 1.14 mL/h/kg; sham-operated, 1.65 mL/h/kg). Splenectomy had no apparent effect on the pharmacokinetics of AMG 386. CONCLUSIONS: The FcRn is integral to maintaining circulating levels of AMG 386 in mice. Renal clearance contributed approximately 30% to total AMG 386 clearance in rats.


Asunto(s)
Angiopoyetina 1/antagonistas & inhibidores , Angiopoyetina 2/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/farmacocinética , Angiopoyetina 1/metabolismo , Angiopoyetina 2/metabolismo , Animales , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales/farmacología , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Riñón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor TIE-2/antagonistas & inhibidores , Receptor TIE-2/metabolismo , Receptores Fc/genética , Receptores Fc/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Bazo/metabolismo
11.
Bioorg Med Chem Lett ; 22(7): 2388-92, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22406116

RESUMEN

The angiopoietin-Tie2 binding and related signal transduction pathways are crucial for vascular angiogenesis, blood vessel integrity and maturation. In this study, we preformed a virtual screening of small molecules targeting to Tie2. The binding site was selected at the extracellular ligand binding region of Tie2, rather than its conventional endocellular ATP binding region. It was found that loperamide, a widely-used antidiarrhea drug, was among the top hits. The binding between loperamide and Tie2 was confirmed by surface plasmon resonance (SPR) assay. Loperamide competitively inhibited the binding of both angiopoietin1 and angiopoietin2. These results indicate that loperamide is an antagonist of angiopoietin1 and angiopoietin2.


Asunto(s)
Angiopoyetina 1/química , Angiopoyetina 2/química , Antidiarreicos/química , Loperamida/química , Receptor TIE-2/química , Angiopoyetina 1/antagonistas & inhibidores , Angiopoyetina 2/antagonistas & inhibidores , Antidiarreicos/farmacología , Unión Competitiva , Células Endoteliales/citología , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Ensayos Analíticos de Alto Rendimiento , Humanos , Ligandos , Loperamida/farmacología , Modelos Moleculares , Unión Proteica , Receptor TIE-2/antagonistas & inhibidores , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/química , Transducción de Señal , Resonancia por Plasmón de Superficie , Termodinámica , Interfaz Usuario-Computador
12.
Sci Rep ; 11(1): 1114, 2021 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-33441910

RESUMEN

Neovascularization of the erectile tissue emerges as a beneficial curative approach to treat erectile dysfunction (ED). Here we for the first time report the unexpected role of vasohibin-1 (VASH1), mainly known as an anti-angiogenic factor, in restoring erectile function in diabetic mice. A diabetic patient has lower cavernous VASH1 expression than in the potent man. VASH1 was mainly expressed in endothelial cells. There were significant decreases in cavernous endothelial cell and pericyte contents in VASH1 knockout mice compared with those in wild-type mice, which resulted in impairments in erectile function. Intracavernous injection of VASH1 protein successfully restored erectile function in the diabetic mice (~ 90% of control values). VASH1 protein reinstated endothelial cells, pericytes, and endothelial cell-cell junction proteins and induced phosphorylation of eNOS (Ser1177) in the diabetic mice. The induction of angiogenic factors, such as angiopoietin-1 and vascular endothelial growth factor, is responsible for cavernous angiogenesis and the restoration of erectile function mediated by VASH1. Altogether, these findings suggest that VASH1 is proangiogenic in diabetic penis and is a new potential target for diabetic ED.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/uso terapéutico , Diabetes Mellitus Experimental/complicaciones , Disfunción Eréctil/tratamiento farmacológico , Disfunción Eréctil/metabolismo , Erección Peniana , Pene/metabolismo , Angiopoyetina 1/antagonistas & inhibidores , Angiopoyetina 1/metabolismo , Animales , Proteínas de Ciclo Celular/administración & dosificación , Diabetes Mellitus Experimental/metabolismo , Regulación hacia Abajo , Células Endoteliales/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica , Óxido Nítrico Sintasa de Tipo III/metabolismo , Pene/irrigación sanguínea , Pericitos/fisiología , Fosforilación , Proteínas de Uniones Estrechas/metabolismo , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo
13.
Am J Pathol ; 175(5): 2159-70, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19815705

RESUMEN

Angiopoietin-1 (Ang1) and angiopoietin-2 (Ang2) have complex actions in angiogenesis and vascular remodeling due to their effects on Tie2 receptor signaling. Ang2 blocks Ang1-mediated activation of Tie2 in endothelial cells under certain conditions but is a Tie2 receptor agonist in others. We examined the effects of selective inhibitors of Ang1 (mL4-3) or Ang2 (L1-7[N]), alone or in combination, on the vasculature of human Colo205 tumors in mice. The Ang2 inhibitor decreased the overall abundance of tumor blood vessels by reducing tumor growth and keeping vascular density constant. After inhibition of Ang2, tumor vessels had many features of normal blood vessels (normalization), as evidenced by junctional accumulation of vascular endothelial-cadherin, junctional adhesion molecule-A, and platelet/endothelial cell adhesion molecule-1 in endothelial cells, increased pericyte coverage, reduced endothelial sprouting, and remodeling into smaller, more uniform vessels. The Ang1 inhibitor by itself had little noticeable effect on the tumor vasculature. However, when administered with the Ang2 inhibitor, the Ang1 inhibitor prevented tumor vessel normalization, but not the reduction in tumor vascularity produced by the Ang2 inhibitor. These findings are consistent with a model whereby inhibition of Ang2 leads to normalization of tumor blood vessels by permitting the unopposed action of Ang1, but decreases tumor vascularity primarily by blocking Ang2 actions.


Asunto(s)
Angiopoyetina 1/antagonistas & inhibidores , Angiopoyetina 2/antagonistas & inhibidores , Vasos Sanguíneos/anatomía & histología , Vasos Sanguíneos/metabolismo , Neoplasias/irrigación sanguínea , Neoplasias/patología , Neovascularización Patológica/metabolismo , Angiopoyetina 1/metabolismo , Angiopoyetina 2/metabolismo , Animales , Vasos Sanguíneos/patología , Células Endoteliales/citología , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias/metabolismo , Pericitos/citología , Pericitos/metabolismo , Fenotipo , Transducción de Señal/fisiología
14.
Acta Virol ; 53(1): 7-13, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19301945

RESUMEN

Hemorrhagic fever with renal syndrome (HFRS) caused by Hantaan virus (HTNV) is characterized by vascular hemorrhage and acute renal failure. Angiopoietin-1 (Ang-1) is a glycoprotein that maintains vessel integrity and reduces endothelial permeability. We found that in HTNV-infected human umbilical vein endothelial cells (HUVECs) the levels of Ang-1 mRNA and protein were reduced on days 2 and 3 post-infection (p.i.), when endothelial permeability was increased. The HTNV-stimulated permeability was reduced by treatment of HUVECs with Ang-1. The plasma Ang-1 level was lower in HFRS patients than in healthy persons. Paired plasma samples of HFRS patients revealed markedly lower Ang-1 levels during the acute phase of HFRS as compared to the convalescent phase. These findings suggested that HTNV reduced the Ang-1 expression in endothelial cells that might play an important role in the increase of vascular permeability in HFRS.


Asunto(s)
Angiopoyetina 1/antagonistas & inhibidores , Angiopoyetina 1/biosíntesis , Permeabilidad Capilar , Células Endoteliales/virología , Virus Hantaan/fisiología , Angiopoyetina 1/sangre , Células Cultivadas , Perfilación de la Expresión Génica , Humanos , Plasma/química , Venas Umbilicales/citología
15.
Invest Ophthalmol Vis Sci ; 60(13): 4097-4108, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31574535

RESUMEN

Purpose: Investigate a significant, dose-related increase in IOP, leading to glaucomatous damage to the neuroretina and optic nerve following intravitreal (ITV) administration of a bispecific F(ab')2 [anti-VEGF/Angiopoietins [ANGPT]F(ab')2] molecule in adult monkeys. Methods: ITV ocular tolerability and investigation of anti-VEGF/ANGPT F(ab')2 (blocking both ANGPT1 and ANGPT2) was done in monkeys; mechanistic studies were done in neonatal mice. Results: Following the second ITV dose of anti-VEGF/ANGPT F(ab')2, all 1.5- and 4-mg/eye treated monkeys developed elevated IOP, which eventually was associated with optic disc cupping and thinning of the neuroretinal rim. Histopathologic examination showed nonreversible axonal degeneration in the optic nerves of animals administered 1.5 mg/eye and higher that was considered secondary to high IOP. Anti-ANGPT Fab also caused elevated IOP in monkeys, but anti-VEGF Fab did not contribute to the IOP increase. In addition, an anti-ANGPT2-selective antibody did not change IOP. In mice simultaneous blockade of ANGPT1 and ANGPT2 impaired the expansion and formation of Schlemm's canal (SC) vessels, similar to genetic ablation of Angpt1/Angpt2 and their receptor TIE2. As previously reported, blocking ANGPT2 alone did not affect SC formation in mice. Conclusions: Dual inhibition of ANGPT1/ANGPT2, but not ANGPT2 alone, leads to increased IOP and glaucomatous damage in monkeys. This confirms a role for TIE2/ANGPT signaling in the control of IOP in adults, a finding initially identified in transgenic mice. Dual pharmacologic inhibition of ANGPT1/ANGPT2 may affect aqueous drainage and homeostasis in adult monkeys and may be useful in developing novel models of glaucoma.


Asunto(s)
Angiopoyetina 1/antagonistas & inhibidores , Angiopoyetina 2/antagonistas & inhibidores , Humor Acuoso/metabolismo , Glaucoma/fisiopatología , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Angiopoyetina 1/fisiología , Angiopoyetina 2/fisiología , Animales , Anticuerpos/farmacología , Presión Intraocular , Primates , Factor A de Crecimiento Endotelial Vascular/fisiología
16.
Expert Opin Investig Drugs ; 28(10): 861-869, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31513439

RESUMEN

Introduction: The Tie-2/Angiopoietin pathway is a therapeutic target for the treatment of neovascular age-related macular degeneration (nAMD) and diabetic macular edema (DME). Activation of Tie-2 receptor via Ang-1 maintains vascular stability to limit exudation. Ang-2, a competitive antagonist to Ang-1, and VE-PTP, an endothelial-specific phosphatase, interfere with the Tie-2-Ang-1 axis, resulting in vascular leakage. Areas covered: Faricimab, a bispecific antibody that inhibits VEGF-A and Ang-2, is in phase 3 trials for nAMD and DME. Nesvacumab is an Ang-2 inhibitor; when coformulated with aflibercept, it failed to show benefit over aflibercept monotherapy in achieving visual gains in phase 2 studies of nAMD and DME. ARP-1536 is an intravitreally administered VE-PTP inhibitor undergoing preclinical studies. AKB-9778 is a subcutaneously administered VE-PTP inhibitor that, when combined with monthly ranibizumab, reduced DME more effectively than ranibizumab monotherapy in a phase 2 study. AKB-9778 monotherapy did not reduce diabetic retinopathy severity score compared to placebo. AXT107, currently in the preclinical phase, promotes conversion of Ang-2 into a Tie-2 agonist and blocks signaling through VEGFR2 and other receptor tyrosine-kinases. Expert opinion: Tie-2/Angiopoietin pathway modulators show promise to reduce treatment burden and improve visual outcomes in nAMD and DME, with potential to treat cases refractory to current treatment modalities.


Asunto(s)
Inhibidores de la Angiogénesis/administración & dosificación , Retinopatía Diabética/tratamiento farmacológico , Degeneración Macular/tratamiento farmacológico , Inhibidores de la Angiogénesis/farmacología , Angiopoyetina 1/antagonistas & inhibidores , Angiopoyetina 2/antagonistas & inhibidores , Animales , Retinopatía Diabética/fisiopatología , Humanos , Degeneración Macular/fisiopatología , Edema Macular/tratamiento farmacológico , Edema Macular/fisiopatología , Receptor TIE-2/efectos de los fármacos , Receptor TIE-2/metabolismo , Transducción de Señal/efectos de los fármacos
17.
Neuroscience ; 156(1): 155-64, 2008 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-18691637

RESUMEN

Neovascularization may contribute to functional recovery after neural injury. Combination treatment of stroke with a nitric oxide donor, (Z)-1-[N-(2-aminoethyl)-N-(2-ammonioethyl) amino] diazen-1-ium-1, 2-diolate (DETA-NONOate) and bone marrow stromal cells promotes functional recovery. However, the mechanisms underlying functional improvement have not been elucidated. In this study, we tested the hypothesis that combination treatment upregulates angiopoietin-1 and its receptor Tie2 in the ischemic brain and bone marrow stromal cells, thereby enhancing cerebral neovascularization after stroke. Adult wild type male C57BL/6 mice were i.v. administered PBS, bone marrow stromal cells 5x10(5), DETA-NONOate 0.4 mg/kg or combination DETA-NONOate with bone marrow stromal cells (n=12/group) after middle cerebral artery occlusion. Combination treatment significantly upregulated angiopoietin-1/Tie2 and tight junction protein (occludin) expression, and increased the number, diameter and perimeter of blood vessels in the ischemic brain compared with vehicle control (mean+ or -S.E., P<0.05). In vitro, DETA-NONOate significantly increased angiopoietin-1/Tie2 protein (n=6/group) and Tie2 mRNA (n=3/group) expression in bone marrow stromal cells. DETA-NONOate also significantly increased angiopoietin-1 protein (n=6/group) and mRNA (n=3/group) expression in mouse brain endothelial cells (P<0.05). Angiopoietin-1 mRNA (n=3/group) was significantly increased in mouse brain endothelial cells treated with DETA-NONOate in combination with bone marrow stromal cell-conditioned medium compared with cells treated with bone marrow stromal cell-conditioned medium or DETA-NONOate alone. Mouse brain endothelial cell capillary tube-like formation assays (n=6/group) showed that angiopoietin-1 peptide, the supernatant of bone marrow stromal cells and DETA-NONOate significantly increased capillary tube formation compared with vehicle control. Combination treatment significantly increased capillary tube formation compared with DETA-NONOate treatment alone. Inhibition of angiopoietin-1 significantly attenuated combination treatment-induced tube formation. Our data indicated that combination treatment of stroke with DETA-NONOate and bone marrow stromal cells promotes neovascularization, which is at least partially mediated by upregulation of the angiopoietin-1/Tie2 axis.


Asunto(s)
Angiopoyetina 1/metabolismo , Trasplante de Médula Ósea/métodos , Neovascularización Fisiológica/efectos de los fármacos , Compuestos Nitrosos/farmacología , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/cirugía , Angiopoyetina 1/agonistas , Angiopoyetina 1/antagonistas & inhibidores , Animales , Capilares/efectos de los fármacos , Capilares/metabolismo , Células Cultivadas , Arterias Cerebrales/efectos de los fármacos , Arterias Cerebrales/metabolismo , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica/fisiología , Donantes de Óxido Nítrico/farmacología , Donantes de Óxido Nítrico/uso terapéutico , Compuestos Nitrosos/uso terapéutico , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Receptor TIE-2/genética , Regeneración/efectos de los fármacos , Regeneración/fisiología , Células del Estroma/trasplante , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
18.
Mol Reprod Dev ; 75(4): 623-31, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17874466

RESUMEN

An adequate vascular supply is important to provide endocrine and paracrine signals during follicular development. We evaluated the direct in vivo effects of both the GnRH-agonist Leuprolide acetate (LA) and the GnRH-antagonist Antide (Ant) on the expression of VEGF-A and ANPT-1 and their receptors in ovarian follicles from prepubertal eCG-treated rats. We also examined whether the changes observed in apoptosis by GnRH-I analogs have an effect on the caspase cascade. LA significantly decreased the levels of VEGF-A, its receptor Flk-1, and ANPT-1 when compared to controls, while the co-injection of Ant interfered with this effect. No changes were observed in the levels of Tie-2 after treatment with these analogs. When we measured the follicular content of caspase-3 protein, we observed that LA significantly increased the level of the active form. The co-injection of Ant interfered with this effect and Ant alone significantly decreased caspase-3 cleavage. IHC analyses corroborated these data. Notably, while LA increased caspase-3 activity levels, Ant decreased them when compared to controls. In follicles obtained from LA-treated rats, cleavage of PARP (a substrate of caspase-3) from the intact 113-kDa protein showed a significant enhancement in an 85-kDa fragment. The co-injection of Ant interfered with this effect. Ant alone significantly decreased PARP cleavage as compared to controls. We conclude that the decrease in VEGF-A, its receptor Flk-1/KDR, and ANPT-1 produced by the administration of GnRH-I agonist is one of the mechanisms involved in ovarian cell apoptosis. This suggests an intraovarian role of an endogenous GnRH-like peptide in gonadotropin-induced follicular development.


Asunto(s)
Apoptosis/efectos de los fármacos , Hormona Liberadora de Gonadotropina/análogos & derivados , Hormona Liberadora de Gonadotropina/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Ovario/efectos de los fármacos , Angiopoyetina 1/antagonistas & inhibidores , Angiopoyetina 1/biosíntesis , Animales , Apoptosis/fisiología , Caspasa 3/efectos de los fármacos , Caspasa 3/metabolismo , Activación Enzimática/efectos de los fármacos , Femenino , Inyecciones Subcutáneas , Leuprolida/administración & dosificación , Neovascularización Fisiológica/fisiología , Oligopéptidos/administración & dosificación , Folículo Ovárico/citología , Folículo Ovárico/efectos de los fármacos , Folículo Ovárico/metabolismo , Ovario/irrigación sanguínea , Ovario/citología , Ratas , Ratas Sprague-Dawley , Receptor TIE-2/biosíntesis , Receptor TIE-2/efectos de los fármacos , Relación Estructura-Actividad , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/biosíntesis , Receptor 2 de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos
19.
Respiration ; 75(3): 328-38, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18073453

RESUMEN

BACKGROUND: Angiopoietins, newly discovered vascular-specific growth factors, and vascular endothelial growth factors (VEGF) play distinct and complementary roles in angiogenesis and vascular maturation. However, the exact roles of angiogenic factors in the adult pulmonary vasculature remain unclear. OBJECTIVE: To elucidate possible roles of angiopoietins and VEGF in the development of hypoxic pulmonary hypertension (PH), changes in the expression of angiogenic factors were examined. METHODS: The cellular distribution and expression of angiopoietins and their receptor Tie2 and VEGF were investigated by RT-PCR, immunoblot, and immunohistochemical methods in rat lung under normal and hypoxic conditions. RESULTS: During the development of PH with vascular remodeling characterized by a decrease in vessel density of intrapulmonary arteries, protein expression of angiopoietin-1 (Ang-1), Tie2, and VEGF significantly decreased in the pulmonary arteries, and Tie2 receptor was inactivated in the lung. The expression of angiopoietin-3 (Ang-3), an endogenous antagonist of Ang-1, significantly increased in the intima under hypoxic conditions. CONCLUSIONS: Since both Ang-1/Tie2 and VEGF promote angiogenesis and vascular survival, and play protective roles in the adaptation of microvascular changes during the onset of PH, the downregulation of both Ang-1/Tie2 and VEGF and upregulation of Ang-3 appear to be associated with vascular rarefaction and the development of hypoxic PH.


Asunto(s)
Angiopoyetina 1/antagonistas & inhibidores , Angiopoyetina 1/metabolismo , Regulación hacia Abajo/fisiología , Hipertensión Pulmonar/fisiopatología , Arteria Pulmonar/metabolismo , Receptor TIE-2/metabolismo , Animales , Western Blotting , Caspasa 3/metabolismo , Enfermedad Crónica , Cartilla de ADN , Hipoxia/metabolismo , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Pulmón/irrigación sanguínea , Pulmón/metabolismo , Masculino , Neovascularización Patológica/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología
20.
Cancer Res ; 66(12): 6167-74, 2006 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-16778190

RESUMEN

Angiopoietin-1 (Ang1) mediates angiogenesis by enhancing endothelial cell survival and migration. It is also known that Ang1 activates Tie2, an endothelial-specific tyrosine kinase receptor, but the molecular mechanism of this process is not clear. In this study, we investigated whether reactive oxygen species (ROS) production plays a role in Ang1-mediated angiogenesis. We found that human umbilical vein endothelial cells treated with Ang1 produce ROS transiently, which was suppressed by NADPH oxidase inhibitor, diphenylene-iodonium chloride, and rotenone. The Ang1-induced ROS was identified as hydrogen peroxide (H2O2) using adenovirus-catalase infection. Removal of H2O2 by adenovirus-catalase significantly suppressed Ang1-induced in vitro endothelial cell migration, in vivo tubule formation and angiogenesis, and activation of p44/42 mitogen-activated protein kinase (MAPK), involved in cell migration, and delayed the deactivation of Akt phosphorylation involved in cell survival. Supporting to in vitro data, Ang1-induced vascular remodeling in catalase (-/-) mice was more prominent than in catalase (+/+) mice: Ang1-induced increases of the diameter of terminal arterioles and the postcapillary venules in catalase (-/-) mice were significant compared with catalase (+/+) mice. These results show that Ang1-induced H2O2 plays an important role in Ang1-mediated angiogenesis by modulating p44/42 MAPK activity.


Asunto(s)
Angiopoyetina 1/farmacología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Peróxido de Hidrógeno/metabolismo , Angiopoyetina 1/antagonistas & inhibidores , Angiopoyetina 1/metabolismo , Animales , Catalasa/genética , Catalasa/metabolismo , Células Cultivadas , Embrión de Pollo , Membrana Corioalantoides/irrigación sanguínea , Membrana Corioalantoides/efectos de los fármacos , Células Endoteliales/citología , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , NG-Nitroarginina Metil Éster/farmacología , Neovascularización Patológica/inducido químicamente , Neovascularización Patológica/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA