Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 141
Filtrar
2.
Shock ; 56(6): 1049-1056, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33756505

RESUMEN

BACKGROUND: Vascular barrier breakdown in sepsis represents a key component of the maladaptive host response to infection and the release of endothelial Angiopoietin-2 (Angpt-2) is a mechanistic driver of endothelial hyperpermeability. Angpt-2 is associated with morbidity and mortality but a targeted therapeutic approach is not available. We screened for U.S. Food and Drug Administration (FDA) approved drugs that might have off-target effects decreasing Angpt-2 and therefore, ameliorating capillary leakage. METHODS: Endothelial cells were isolated from human umbilical veins (HUVECs) and used for in vitro studies at baseline and after stimulation (FDA-library screening, RT-PCR, ELISA, immunocytochemistry, MTT assay). On the functional level, we assessed real-time transendothelial electrical resistance (TER) using an electric cell-substrate impedance sensing device. RESULTS: We found that the anti-fungal Bifonazole (BIFO) reduces spontaneous Angpt-2 release in a time- and dose-dependent manner after 8, 12, and 24 h (24 h: veh: 15.6 ±â€Š0.7 vs. BIFO: 8.6 ±â€Š0.8 ng/mL, P < 0.0001). Furthermore, we observed a reduction in its intra-cellular content by 33% (P < 0.001). Stimulation with tumor necrosis factor α induced a strong release of Angpt-2 that could analogously be blocked by additional treatment with BIFO (veh: 1.58 ±â€Š0.2 vs. BIFO: 1.02 ±â€Š0.1, P < 0.0001). Quantification of endothelial permeability by TER revealed that BIFO was sufficient to reduce Thrombin-induced barrier breakdown (veh: 0.82 ±â€Š0.1 vs. BIFO: 1.01 ±â€Š0.02, P < 0.05). CONCLUSION: The antifungal BIFO reduces both release and biosynthesis of the endothelial-destabilizing factor Angpt-2 in vitro thereby improving vascular barrier function. Additional studies are needed to further investigate the underlying mechanism and to translate these findings to in vivo models.


Asunto(s)
Angiopoyetina 2/fisiología , Antifúngicos/farmacología , Permeabilidad Capilar/efectos de los fármacos , Imidazoles/farmacología , Inflamación/fisiopatología , Células Cultivadas , Células Endoteliales , Humanos
3.
Cell Commun Signal ; 18(1): 46, 2020 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-32183816

RESUMEN

BACKGROUND: Hepatocellular carcinoma (HCC) is the most common primary liver cancer and is a highly vascularized solid tumor. Angiopoietin-2 (ANGPT2) has been described as an attractive target for antiangiogenic therapy. Exosomes are small extracellular vesicles secreted by most cell types and contribute to cell-to-cell communication by delivering functional cargo to recipient cells. The expression of ANGPT2 in tumor-derived exosomes remains unknown. METHODS: We detected the ANGPT2 expression in HCC-derived exosomes by immunoblotting, enzyme-linked immunosorbent assay and immunogold labeling, then observed exosomal ANGPT2 internalization and recycling by confocal laser scanning microscopy, co-immunoprecipitation and immunoblotting. We used two HCC cell lines (Hep3B and MHCC97H) to overexpress ANGPT2 by lentivirus infection or knockdown ANGPT2 by the CRISPR/Cas system, then isolated exosomes to coculture with human umbilical vein endothelial cells (HUVECs) and observed the angiogenesis by Matrigel microtubule formation assay, transwell migration assay, wound healing assay, cell counting kit-8 assay, immunoblotting and in vivo tumorigenesis assay. RESULTS: We found that HCC-derived exosomes carried ANGPT2 and delivered it into HUVECs by exosome endocytosis, this delivery led to a notable increase in angiogenesis by a Tie2-independent pathway. Concomitantly, we observed that HCC cell-secreted exosomal ANGPT2 was recycled by recipient HUVECs and might be reused. In addition, the CRISPR-Cas systems to knock down ANGPT2 significantly inhibited the angiogenesis induced by HCC cell-secreted exosomal ANGPT2, and obviously suppressed the epithelial-mesenchymal transition activation in HCC. CONCLUSIONS: Taken together, these results reveal a novel pathway of tumor angiogenesis induced by HCC cell-secreted exosomal ANGPT2 that is different from the classic ANGPT2/Tie2 pathway. This way may be a potential therapeutic target for antiangiogenic therapy. Video Abstract.


Asunto(s)
Angiopoyetina 2/fisiología , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Exosomas/metabolismo , Regulación Neoplásica de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana , Humanos , Neovascularización Patológica
4.
Arch. Soc. Esp. Oftalmol ; 95(2): 75-83, feb. 2020. tab
Artículo en Español | IBECS | ID: ibc-195330

RESUMEN

La degeneración macular asociada a la edad (DMAE), y en especial su forma neovascular, supone la principal causa de baja visión en países desarrollados. Aunque en años recientes la introducción de los fármacos conocidos como anti-VEGF ha supuesto una revolución en el manejo de esta enfermedad, su precio, la necesidad de inyecciones intravítreas repetidas y la pérdida de efectividad a largo plazo en una importante proporción de pacientes son problemas aún por resolver. En la actualidad existen en desarrollo una serie de nuevas estrategias que tratan de ofrecer soluciones a las limitaciones que presentan los fármacos intravítreos actuales. Entre ellos destacan nuevos anti-VEGF como brolucizumab o abicipar, fármacos contra el receptor de angiopoyetina-2 como faricimab, sistemas de liberación sostenida de macromoléculas o inhibidores de la tirosina cinasa. Respecto a la DMAE seca, la neuroprotección, la modulación de la vía del complemento y, en última instancia, la terapia con células madre son las líneas de investigación más prometedoras en la actualidad


Age-related macular degeneration and especially neovascular age-related macular degeneration is the leading cause of low vision in developed countries. Even though the introduction of anti-VEGF drugs in recent years completely changed the management of this condition, its cost, the need for repeated intravitreal injections, and loss of efficacy in the long term are still issues to deal with. Currently, a new generation of novel therapies under development is attempting to address some of these limitations. Some of the most prominent among them are new anti-VEGFs such as brolucizumab or abicipar, drugs against angiopoietin-2 receptor such as faricimab, sustained-release systems, or tyrosine kinase inhibitors. As regards dry age-related macular degeneration, neuroprotection, the complement pathway, and stem cell therapy are the most promising targets currently under investigation


Asunto(s)
Humanos , Degeneración Macular/terapia , Inhibidores de la Angiogénesis/uso terapéutico , Angiopoyetina 2/fisiología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Preparaciones de Acción Retardada/uso terapéutico , Terapia Genética/métodos , Degeneración Macular/tratamiento farmacológico , Degeneración Macular/prevención & control , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Ranibizumab/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Factores de Riesgo
5.
Arch Soc Esp Oftalmol (Engl Ed) ; 95(2): 75-83, 2020 Feb.
Artículo en Inglés, Español | MEDLINE | ID: mdl-31787390

RESUMEN

Age-related macular degeneration and especially neovascular age-related macular degeneration is the leading cause of low vision in developed countries. Even though the introduction of anti-VEGF drugs in recent years completely changed the management of this condition, its cost, the need for repeated intravitreal injections, and loss of efficacy in the long term are still issues to deal with. Currently, a new generation of novel therapies under development is attempting to address some of these limitations. Some of the most prominent among them are new anti-VEGFs such as brolucizumab or abicipar, drugs against angiopoietin-2 receptor such as faricimab, sustained-release systems, or tyrosine kinase inhibitors. As regards dry age-related macular degeneration, neuroprotection, the complement pathway, and stem cell therapy are the most promising targets currently under investigation.


Asunto(s)
Degeneración Macular/terapia , Inhibidores de la Angiogénesis/uso terapéutico , Angiopoyetina 2/fisiología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Preparaciones de Acción Retardada/uso terapéutico , Terapia Genética/métodos , Humanos , Degeneración Macular/tratamiento farmacológico , Degeneración Macular/prevención & control , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Ranibizumab/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Factores de Riesgo
6.
Life Sci ; 239: 117080, 2019 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-31756341

RESUMEN

The angiopoietin signal transduction system is a complex of vascular-specific kinase pathways that plays a crucial role in angiogenesis and maintenance of vascular homeostasis. Angiopoietin1 (Ang1) and 2 (Ang2), the ligand proteins of the pathway, belong to a family of glycoproteins that signal primarily through the transmembrane Tyrosine-kinase-2 receptor. Despite a considerable sequence homology, Ang1 and Ang2 manifest antagonistic effects in pathophysiological conditions. While Ang1 promotes the activation of survival pathways and the stabilization of the normal mature vessels, Ang2 can either favor vessel destabilization and leakage or promote abnormal EC proliferation in a context-dependent manner. Altered Ang1/Ang2 balance has been reported in various pathological conditions in association with inflammation and deregulated angiogenesis. In particular, increased Ang2 levels have been documented in human cancer and cardiovascular disease (CVD), including ischemic myocardial injury, heart failure and other cardiovascular complications secondary to diabetes, chronic renal damage and hypertension. Despite the obvious phenotypic differences, CVD and cancer share some common Ang2-dependent etiopathological mechanisms such as inflammation, epithelial (or endothelial) to mesenchymal transition, and adverse vascular network remodeling. Interestingly, both cancer and CVD are negatively affected by thyroid hormone dyshomeostasis. This review provides an overview of the complex Ang2-dependent signaling involved in CVD and cancer, as well as a survey of the related clinical literature. Moreover, on the basis of recent molecular acquisitions in an experimental model of post ischemic cardiac disease, the putative novel role of the thyroid hormone in the regulation of Ang1/Ang2 balance is also briefly discussed.


Asunto(s)
Angiopoyetina 2/metabolismo , Angiopoyetina 2/fisiología , Neovascularización Patológica/metabolismo , Inductores de la Angiogénesis/metabolismo , Enfermedades Cardiovasculares/metabolismo , Endotelio Vascular/metabolismo , Humanos , Inflamación/metabolismo , Glicoproteínas de Membrana/metabolismo , Neoplasias/metabolismo , Neovascularización Fisiológica , Transducción de Señal/fisiología , Remodelación Vascular
7.
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
8.
Arterioscler Thromb Vasc Biol ; 39(9): 1843-1858, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31315435

RESUMEN

Objective Weibel-Palade bodies (WPBs) are endothelial cell (EC)-specific organelles formed by vWF (von Willebrand factor) polymerization and that contain the proangiogenic factor Ang-2 (angiopoietin-2). WPB exocytosis has been shown to be implicated for vascular repair and inflammatory responses. Here, we investigate the role of WPBs during angiogenesis and vessel stabilization. Approach and Results WPB density in ECs decreased at the angiogenic front of retinal vascular network during development and neovascularization compared with stable vessels. In vitro, VEGF (vascular endothelial growth factor) induced a VEGFR-2 (vascular endothelial growth factor receptor-2)-dependent exocytosis of WPBs that contain Ang-2 and consequently the secretion of vWF and Ang-2. Blocking VEGF-dependant WPB exocytosis and Ang-2 secretion promoted pericyte migration toward ECs. Pericyte migration was inhibited by adding recombinant Ang-2 or by silencing Ang-1 (angiopoietin-1) or Tie2 (angiopoietin-1 receptor) in pericytes. Consistently, in vivo anti-VEGF treatment induced accumulation of WPBs in retinal vessels because of the inhibition of WPB exocytosis and promoted the increase of pericyte coverage of retinal vessels during angiogenesis. In tumor angiogenesis, depletion of WPBs in vWF knockout tumor-bearing mice promoted an increase of tumor angiogenesis and a decrease of pericyte coverage of tumor vessels. By another approach, normalized tumor vessels had higher WPB density. Conclusions We demonstrate that WPB exocytosis and Ang-2 secretion are regulated during angiogenesis to limit pericyte coverage of remodeling vessels by disrupting Ang-1/Tie2 autocrine signaling in pericytes.


Asunto(s)
Neovascularización Patológica/fisiopatología , Neovascularización Fisiológica/fisiología , Pericitos/fisiología , Cuerpos de Weibel-Palade/fisiología , Angiopoyetina 2/fisiología , Animales , Células Cultivadas , Células Endoteliales/fisiología , Exocitosis , Humanos , Ratones , Ratones Endogámicos C57BL , Neoplasias/irrigación sanguínea , Retina/fisiología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/fisiología
9.
Hepatology ; 69(3): 1087-1104, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30259536

RESUMEN

Angiogenesis contributes to the development of nonalcoholic steatohepatitis (NASH) and promotes inflammation, fibrosis, and progression to hepatocellular carcinoma (HCC). Angiopoietin-2 (Ang-2) is a key regulator of angiogenesis. We aimed to investigate the role of Ang-2 and its potential as a therapeutic target in NASH using human samples, in vivo mouse models, and in vitro assays. Serum Ang-2 levels were determined in 104 obese patients undergoing bariatric surgery and concomitant liver biopsy. The effect of the Ang-2/Tie2 receptor inhibiting peptibody L1-10 was evaluated in the methionine-choline deficient (MCD) and streptozotocin-western diet nonalcoholic fatty liver disease mouse models, and in vitro on endothelial cells and bone marrow-derived macrophages. The hepatic vasculature was visualized with µCT scans and scanning electron microscopy of vascular casts. Serum Ang-2 levels were increased in patients with histological NASH compared with patients with simple steatosis and correlated with hepatic CD34 immunoreactivity as a marker of hepatic angiogenesis. Serum and hepatic Ang-2 levels were similarly increased in mice with steatohepatitis. Both preventive and therapeutic L1-10 treatment reduced hepatocyte ballooning and fibrosis in MCD diet-fed mice and was associated with reduced hepatic angiogenesis and normalization of the vascular micro-architecture. Liver-isolated endothelial cells and monocytes from MCD-fed L1-10-treated mice showed reduced expression of leukocyte adhesion and inflammatory markers, respectively, compared with cells from untreated MCD diet-fed mice. In the streptozotocin-western diet model, therapeutic Ang-2 inhibition was able to reverse NASH and attenuate HCC progression. In vitro, L1-10 treatment mitigated increased cytokine production in lipopolysaccharide-stimulated endothelial cells but not in macrophages. Conclusion: Our findings provide evidence for Ang-2 inhibition as a therapeutic strategy to target pathological angiogenesis in NASH.


Asunto(s)
Angiopoyetina 2/fisiología , Hígado/irrigación sanguínea , Neovascularización Patológica , Enfermedad del Hígado Graso no Alcohólico/etiología , Adulto , Angiopoyetina 2/antagonistas & inhibidores , Angiopoyetina 2/sangre , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Neovascularización Patológica/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/sangre , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Estudios Prospectivos
10.
Cardiol Rev ; 27(1): 8-13, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30520778

RESUMEN

Continuous-flow left ventricular assist devices (CF-LVADs) are increasingly used for the management of advanced heart failure refractory to optimal medical therapy. Despite the encouraging outcomes with CF-LVADs, gastrointestinal bleeding (GIB) continues to be a rather concerning complication resulting in increased rates of readmission and increased morbidity. The exact pathophysiology of CF-LVAD-associated GIB remains poorly understood, and this lack of knowledge limits our ability to control this morbid complication. What is clear, however, is that the majority of GIB episodes in LVAD patients are due to fragile GI arteriovenous malformations or angiodysplasias, in the setting of CF-LVAD-associated acquired von Willebrand syndrome. We will, herein, appraise the proposed interactions between different pathophysiological processes thought to be causing angiodysplasias and GIB in patients on CF-LVAD support.


Asunto(s)
Hemorragia Gastrointestinal/etiología , Corazón Auxiliar/efectos adversos , Angiodisplasia/complicaciones , Angiopoyetina 2/fisiología , Hemorragia Gastrointestinal/diagnóstico , Hemorragia Gastrointestinal/epidemiología , Hemorragia Gastrointestinal/terapia , Humanos , Hipoxia/complicaciones , Incidencia , Trombina/metabolismo
12.
Kidney Blood Press Res ; 43(2): 545-554, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29642068

RESUMEN

BACKGROUND/AIMS: Diabetic nephropathy is the leading cause of end-stage renal disease and accounts for 30∼40% of patients requiring maintenance dialysis, thereby increasing the burden on health insurance programs. Diabetic nephropathy is also the strongest predictor of cardiovascular morbidity and mortality. The aim of this study was to examine whether angiopoietin-2 (Angpt2), a modulator of endothelial function, affects the clinical outcomes of diabetic patients. METHODS: This study enrolled 236 patients with diabetes mellitus with estimated glomerular filtration rate (eGFR) < 60ml/min/1.73m2 from January 2006 to December 2011, who were followed until June 2017. Clinical outcomes included renal outcomes (commencing dialysis and rapid decline in renal function (eGFR decline > 3 ml/min per 1.73 m2/year)), major adverse cardiovascular events (MACEs), and all-cause mortality. RESULTS: Over a mean follow-up period of 3.9±2.7 years, 135 (57.2%) patients commenced dialysis, 106 (44.9%) had rapid decline in renal function, and 50 (21.2%) had MACEs or died from all-causes. Log-formed Angpt2 was significantly associated with increased risks of commencing dialysis (HR: 3.91, 95% CI: 1.56-9.76), rapid renal function decline (OR: 6.81, 95% CI: 1.06-43.88), and MACEs or all-cause mortality (HR: 6.34, 95% CI: 1.18-33.97) in the adjusted analysis. Patients in the highest quartile had hazard ratios of 2.90 and 3.11 for commencing dialysis and rapid renal function decline, respectively, compared to those in the lowest quartile after adjustments. Similar significant dose-response results were found in composite outcomes of either MACEs or all-cause mortality. CONCLUSION: Angpt2 is an independent predictor of adverse clinical outcomes in diabetic patients. Further studies are needed to identify the pathogenic role of Angpt2 in renal deterioration and cardiovascular complications of diabetes mellitus.


Asunto(s)
Angiopoyetina 2/sangre , Enfermedades Cardiovasculares/etiología , Nefropatías Diabéticas , Insuficiencia Renal Crónica/etiología , Anciano , Angiopoyetina 2/fisiología , Nefropatías Diabéticas/complicaciones , Nefropatías Diabéticas/mortalidad , Nefropatías Diabéticas/fisiopatología , Femenino , Estudios de Seguimiento , Tasa de Filtración Glomerular , Humanos , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Pronóstico
13.
J Am Soc Nephrol ; 29(4): 1097-1107, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29237738

RESUMEN

Urinary concentrating ability is central to mammalian water balance and depends on a medullary osmotic gradient generated by a countercurrent multiplication mechanism. Medullary hyperosmolarity is protected from washout by countercurrent exchange and efficient removal of interstitial fluid resorbed from the loop of Henle and collecting ducts. In most tissues, lymphatic vessels drain excess interstitial fluid back to the venous circulation. However, the renal medulla is devoid of classic lymphatics. Studies have suggested that the fenestrated ascending vasa recta (AVRs) drain the interstitial fluid in this location, but this function has not been conclusively shown. We report that late gestational deletion of the angiopoietin receptor endothelial tyrosine kinase 2 (Tie2) or both angiopoietin-1 and angiopoietin-2 prevents AVR formation in mice. The absence of AVR associated with rapid accumulation of fluid and cysts in the medullary interstitium, loss of medullary vascular bundles, and decreased urine concentrating ability. In transgenic reporter mice with normal angiopoietin-Tie2 signaling, medullary AVR exhibited an unusual hybrid endothelial phenotype, expressing lymphatic markers (prospero homeobox protein 1 and vascular endothelial growth factor receptor 3) as well as blood endothelial markers (CD34, endomucin, platelet endothelial cell adhesion molecule 1, and plasmalemmal vesicle-associated protein). Taken together, our data redefine the AVRs as Tie2 signaling-dependent specialized hybrid vessels and provide genetic evidence of the critical role of AVR in the countercurrent exchange mechanism and the structural integrity of the renal medulla.


Asunto(s)
Angiopoyetina 1/fisiología , Angiopoyetina 2/fisiología , Líquido Extracelular/metabolismo , Capacidad de Concentración Renal/fisiología , Médula Renal/irrigación sanguínea , Receptor TIE-2/fisiología , Angiopoyetina 1/deficiencia , Angiopoyetina 1/genética , Angiopoyetina 2/deficiencia , Angiopoyetina 2/genética , Animales , Tipificación del Cuerpo , Linaje de la Célula , Endotelio Vascular , Genes Reporteros , Edad Gestacional , Proteínas de Homeodominio/análisis , Enfermedades Renales Quísticas/genética , Médula Renal/embriología , Médula Renal/fisiología , Ratones , Ratones Noqueados , Ratones Transgénicos , Miofibroblastos/patología , Ósmosis , Receptor TIE-2/deficiencia , Receptor TIE-2/genética , Circulación Renal , Transducción de Señal , Proteínas Supresoras de Tumor/análisis , Receptor 3 de Factores de Crecimiento Endotelial Vascular/análisis
14.
PLoS One ; 12(3): e0173906, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28333979

RESUMEN

BACKGROUND: Fluid overload is not only the characteristic but also an important complication in chronic kidney disease (CKD) patients. Angiopoietin-2 (Angpt2) disturbs endothelium and vessel permeability, which may induce fluid overload. The aim of this study is to examine the interaction between fluid status and Angpt2 in adverse renal outcomes of CKD. METHODS: This cohort study enrolled 290 patients with CKD stages 3-5 from January 2011 to December 2011 and followed up until December 2015. Fluid status was presented as overhydration (OH) value measured by body composition monitor, while OH>1.1L was defined as fluid overload. Renal outcomes were defined as commencing dialysis and rapid renal function decline (the slope of estimated glomerular filtration rate < -5 ml/min/1.73 m2/y). RESULTS: During a mean follow-up of 38.6±18.3 months, 125 (43.1%) patients progressed to commencing dialysis and 99(34.7%) patients presented rapid renal function decline. All patients were stratified by OH of 1.1L and the median of circulating Angpt2. These patients with both OH>1.1L and high circulating Angpt2 were more likely to reach commencing dialysis compared to other groups. The risks for commencing dialysis and rapid renal function decline were significantly higher in patients with OH>1.1L and high circulating Angpt2 level compared to those with OH≦1.1L and low circulating Angpt2 (2.14, 1.21-3.78, P = 0.009; 4.96, 1.45-16.97, P = 0.01). There was a significant interaction between OH level and circulating Angpt2 in entering dialysis (P-interaction = 0.02). CONCLUSIONS: Fluid overload and Angpt2 might have a synergistic effect on adverse renal outcomes in CKD patients.


Asunto(s)
Angiopoyetina 2/sangre , Insuficiencia Renal Crónica/fisiopatología , Equilibrio Hidroelectrolítico/fisiología , Anciano , Angiopoyetina 2/fisiología , Creatinina/sangre , Progresión de la Enfermedad , Líquido Extracelular , Femenino , Humanos , Estimación de Kaplan-Meier , Riñón/fisiopatología , Masculino , Persona de Mediana Edad
15.
Curr Diab Rep ; 16(12): 126, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27778249

RESUMEN

Tie2 is a tyrosine kinase receptor located predominantly on vascular endothelial cells that plays a central role in vascular stability. Angiopoietin-1 (Angpt1), produced by perivascular cells, binds, clusters, and activates Tie2, leading to Tie2 autophosphorylation and downstream signaling. Activated Tie2 increases endothelial cell survival, adhesion, and cell junction integrity, thereby stabilizing the vasculature. Angiopoietin-2 (Angpt2) and vascular endothelial-protein tyrosine phosphatase (VE-PTP) are negative regulators increased by hypoxia; they inactivate Tie2, destabilizing the vasculature and increasing responsiveness to vascular endothelial growth factor (VEGF) and other inflammatory cytokines that stimulate vascular leakage and neovascularization. AKB-9778 is a small-molecule antagonist of VE-PTP which increases phosphorylation of Tie2 even in the presence of high Angpt2 levels. In preclinical studies, AKB-9778 reduced VEGF-induced leakage and ocular neovascularization (NV) and showed additive benefit when combined with VEGF suppression. In two clinical trials in diabetic macular edema (DME) patients, subcutaneous injections of AKB-9778 were safe and provided added benefit to VEGF suppression. Preliminary data suggest that AKB-9778 monotherapy improves diabetic retinopathy. These data suggest that Tie2 activation may be a valuable strategy to treat or prevent diabetic retinopathy.


Asunto(s)
Compuestos de Anilina/uso terapéutico , Retinopatía Diabética/tratamiento farmacológico , Edema Macular/tratamiento farmacológico , Receptor TIE-2/antagonistas & inhibidores , Ácidos Sulfónicos/uso terapéutico , Angiopoyetina 1/fisiología , Angiopoyetina 2/fisiología , Humanos , Receptor TIE-2/fisiología , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/fisiología , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo
16.
Medicine (Baltimore) ; 95(33): e4524, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27537575

RESUMEN

This study is aimed to investigate whether serum angiostatic factors (thrombospondin-1 [TSP-1] and endostatin) or angiogenic factors (angiopoietin-2 [Ang-2]) are related to coronary collateral vessel development in patients with chronic total occlusion (CTO).A total of 149 patients were enrolled in the study, and 39 patients with coronary artery disease but without significant stenosis were included in control group. In 110 patients with CTO lesion, 79 with Rentrop grades 2 to 3 collaterals were grouped as good collateral, while 31 with Rentrop grades 0 to 1 collaterals were grouped as poor collateral. Serum TSP-1, endostatin, and Ang-2 levels were studied.Serum endostatin level was significantly higher in poor collateral group compared with control group and good collateral group, respectively (96.2 ±â€Š30.4 vs 77.8 ±â€Š16.5 ng/mL, P = 0.007; 96.2 ±â€Š30.4 vs 81.2 ±â€Š30.4 ng/mL, P = 0.018). In multivariate analysis, decreased serum endostatin level was independently related to good coronary collateral development. Serum TSP-1 level was lower in patients with CTO compared with control group. However, no difference in TSP-1 level was detected between poor and good collateral group. The serum Ang-2 level did not show a significant difference among 3 groups.Circulatory endostatin may be a useful biomarker for coronary collateral development and potential target for therapeutic angiogenesis in patients with CTO.


Asunto(s)
Angiopoyetina 2/sangre , Circulación Colateral/fisiología , Oclusión Coronaria/sangre , Endostatinas/sangre , Trombospondina 1/sangre , Angiopoyetina 2/fisiología , Biomarcadores/sangre , Estudios de Casos y Controles , Enfermedad Crónica , Enfermedad de la Arteria Coronaria/sangre , Enfermedad de la Arteria Coronaria/fisiopatología , Oclusión Coronaria/fisiopatología , Endostatinas/fisiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Trombospondina 1/fisiología
17.
Cytokine ; 80: 1-6, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26908294

RESUMEN

To assess the role of angiopoietin (Ang)-1 and Ang-2 and to investigate the clinical significance of serum levels of them in systemic juvenile idiopathic arthritis (s-JIA)-associated macrophage activation syndrome (MAS), we determined these levels in 51 patients with s-JIA, 11 patients with polyarticular JIA (poly-JIA), 12 patients with virus associated hemophagocytic syndrome (VAHS), 12 patients with Kawasaki disease (KD), and 15 age-matched healthy controls (HC). The results were compared with clinical features of MAS. During the MAS phase, serum Ang-1 levels were significantly decreased compared with those during the active and inactive phases. Serum Ang-2/1 ratio were significantly elevated during the MAS phase, compared with those during the active and inactive phases. There was a rapid increase in the Ang-2/1 ratio at the onset of MAS. Serum Ang-1 and the Ang-2/1 ratio significantly correlated with measures of disease activity, including AST and LDH. Ang-2/1 dysregulation was also observed in patients with VAHS, whereas not observed in most cases of KD. The homeostasis of vascular endothelial function by Ang-1 and Ang-2 is disrupted in MAS. Serum Ang-1 levels and the Ang-2/1 ratio might represent promising indicators of disease activity for MAS.


Asunto(s)
Angiopoyetina 1/sangre , Angiopoyetina 2/sangre , Artritis Juvenil/inmunología , Endotelio Vascular/fisiología , Homeostasis , Síndrome de Activación Macrofágica/fisiopatología , Adolescente , Angiopoyetina 1/fisiología , Angiopoyetina 2/fisiología , Artritis Juvenil/complicaciones , Niño , Preescolar , Citocinas/sangre , Femenino , Humanos , Lactante , Linfohistiocitosis Hemofagocítica/fisiopatología , Linfohistiocitosis Hemofagocítica/virología , Síndrome de Activación Macrofágica/etiología , Masculino , Síndrome Mucocutáneo Linfonodular/fisiopatología
18.
Dev Ophthalmol ; 55: 28-37, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26502333

RESUMEN

Systematic study of the mechanisms underlying pathological ocular neovascularization has yielded a wealth of knowledge about pro- and anti-angiogenic factors that modulate diseases such as neovascular age-related macular degeneration. The evidence implicating vascular endothelial growth factor (VEGF) in particular has led to the development of a number of approved anti-VEGF therapies. Additional proangiogenic targets that have emerged as potential mediators of ocular neovascularization include hypoxia-inducible factor-1, angiopoietin-2, platelet-derived growth factor-B and components of the alternative complement pathway. As for VEGF, knowledge of these factors has led to a product pipeline of many more novel agents that are in various stages of clinical development in the setting of ocular neovascularization. These agents are represented by a range of drug classes and, in addition to novel small- and large-molecule VEGF inhibitors, include gene therapies, small interfering RNA agents and tyrosine kinase inhibitors. In addition, combination therapy is beginning to emerge as a strategy to improve the efficacy of individual therapies. Thus, a variety of agents, whether administered alone or as adjunctive therapy with agents targeting VEGF, offer the promise of expanding the range of treatments for ocular neovascular diseases.


Asunto(s)
Angiopoyetina 2/fisiología , Vía Alternativa del Complemento/fisiología , Ojo/irrigación sanguínea , Factor 1 Inducible por Hipoxia/fisiología , Neovascularización Patológica/fisiopatología , Proteínas Proto-Oncogénicas c-sis/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología , Humanos
20.
Crit Care Med ; 43(7): e230-40, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25855898

RESUMEN

OBJECTIVE: The recent withdrawal of a targeted sepsis therapy has diminished pharmaceutical enthusiasm for developing novel drugs for the treatment of sepsis. Angiopoietin-2 is an endothelial-derived protein that potentiates vascular inflammation and leakage and may be involved in sepsis pathogenesis. We screened approved compounds for putative inhibitors of angiopoietin-2 production and investigated underlying molecular mechanisms. DESIGN: Laboratory and animal research plus prospective placebo-controlled randomized controlled trial (NCT00529139) and retrospective analysis (NCT00676897). SETTING: Research laboratories of Hannover Medical School and Harvard Medical School. PATIENTS: Septic patients/C57Bl/6 mice and human endothelial cells. INTERVENTIONS: Food and Drug Administration-approved library screening. MEASUREMENTS AND MAIN RESULTS: In a cell-based screen of more than 650 Food and Drug Administration-approved compounds, we identified multiple members of the 3-hydroxy-3-methyl-glutaryl-CoA reductase inhibitor drug class (referred to as statins) that suppressed angiopoietin-2. Simvastatin inhibited 3-hydroxy-3-methyl-glutaryl-CoA reductase, which in turn activated PI3K-kinase. Downstream of this signaling, PI3K-dependent phosphorylation of the transcription factor Foxo1 at key amino acids inhibited its ability to shuttle to the nucleus and bind cis-elements in the angiopoietin-2 promoter. In septic mice, transient inhibition of angiopoietin-2 expression by liposomal siRNA in vivo improved absolute survival by 50%. Simvastatin had a similar effect, but the combination of angiopoietin-2 siRNA and simvastatin showed no additive benefit. To verify the link between statins and angiopoietin-2 in humans, we performed a pilot matched case-control study and a small randomized placebo-controlled trial demonstrating beneficial effects on angiopoietin-2. CONCLUSIONS: 3-hydroxy-3-methyl-glutaryl-CoA reductase inhibitors may operate through a novel Foxo1-angiopoietin-2 mechanism to suppress de novo production of angiopoietin-2 and thereby ameliorate manifestations of sepsis. Given angiopoietin-2's dual role as a biomarker and candidate disease mediator, early serum angiopoietin-2 measurement may serve as a stratification tool for future trials of drugs targeting vascular leakage.


Asunto(s)
Angiopoyetina 2/antagonistas & inhibidores , Angiopoyetina 2/fisiología , Factores de Transcripción Forkhead/fisiología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Sepsis/tratamiento farmacológico , Simvastatina/uso terapéutico , Anciano , Animales , Estudios de Casos y Controles , Reposicionamiento de Medicamentos , Femenino , Proteína Forkhead Box O1 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...