Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 142
Filtrar
1.
Int J Mol Sci ; 22(21)2021 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-34769465

RESUMEN

Angiogenesis, the growth of new blood vessels from preexisting vessels, is associated with inflammation in various pathological conditions. Well-known angiogenetic factors include vascular endothelial growth factor (VEGF), angiopoietins, platelet-derived growth factor, transforming growth factor-ß, and basic fibroblast growth factor. Yes-associated protein 1 (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) have recently been added to an important angiogenic factor. Accumulating evidence indicates associations between angiogenesis and chronic inflammatory skin diseases. Angiogenesis is deeply involved in the pathogenesis of psoriasis. VEGF, angiopoietins, tumor necrosis factor-a, interleukin-8, and interleukin-17 are unregulated in psoriasis and induce angiogenesis. Angiogenesis may be involved in the pathogenesis of atopic dermatitis, and in particular, mast cells are a major source of VEGF expression. Angiogenesis is an essential process in rosacea, which is induced by LL-37 from a signal cascade by microorganisms, VEGF, and MMP-3 from mast cells. In addition, angiogenesis by increased VEGF has been reported in chronic urticaria and hidradenitis suppurativa. The finding that VEGF is expressed in inflammatory skin lesions indicates that inhibition of angiogenesis is a useful strategy for treatment of chronic, inflammatory skin disorders.


Asunto(s)
Dermatitis/fisiopatología , Neovascularización Patológica , Angiopoyetinas/genética , Angiopoyetinas/fisiología , Animales , Enfermedad Crónica , Dermatitis/complicaciones , Dermatitis/genética , Dermatitis/patología , Dermatitis Atópica/etiología , Dermatitis Atópica/patología , Dermatitis Atópica/fisiopatología , Humanos , Neovascularización Patológica/complicaciones , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Neovascularización Patológica/fisiopatología , Psoriasis/etiología , Psoriasis/patología , Psoriasis/fisiopatología , Rosácea/etiología , Rosácea/patología , Rosácea/fisiopatología , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/fisiología
2.
Sci Rep ; 11(1): 419, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33432093

RESUMEN

Excessive tumour growth results in a hypoxic environment around cancer cells, thus inducing tumour angiogenesis, which refers to the generation of new blood vessels from pre-existing vessels. This mechanism is biologically and physically complex, with various mathematical simulation models proposing to reproduce its formation. However, although temporary vessel regression is clinically known, few models succeed in reproducing this phenomenon. Here, we developed a three-dimensional simulation model encompassing both angiogenesis and tumour growth, specifically including angiopoietin. Angiopoietin regulates both adhesion and migration between vascular endothelial cells and wall cells, thus inhibiting the cell-to-cell adhesion required for angiogenesis initiation. Simulation results showed a regression, i.e. transient decrease, in the overall length of new vessels during vascular network formation. Using our model, we also evaluated the efficacy of administering the drug bevacizumab. The results highlighted differences in treatment efficacy: (1) earlier administration showed higher efficacy in inhibiting tumour growth, and (2) efficacy depended on the treatment interval even with the administration of the same dose. After thorough validation in the future, these results will contribute to the design of angiogenesis treatment protocols.


Asunto(s)
Angiopoyetinas/fisiología , Vasos Sanguíneos/crecimiento & desarrollo , Modelos Teóricos , Neoplasias/irrigación sanguínea , Neovascularización Patológica/patología , Angiopoyetinas/genética , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patología , Simulación por Computador , Progresión de la Enfermedad , Humanos , Regresión Neoplásica Espontánea/genética , Regresión Neoplásica Espontánea/patología , Neoplasias/patología , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Inducción de Remisión , Factor A de Crecimiento Endotelial Vascular/fisiología
3.
Angiogenesis ; 23(2): 105-117, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31707538

RESUMEN

A branched vascular network is crucial to placental development and is dependent on factors such as vascular endothelial growth factor (VEGF), placental growth factor (PlGF), angiopoietin-1 (Ang-1), angiopoietin-2 (Ang-2), soluble fms-like tyrosine kinase-1 (sFlt-1) and soluble endoglin (sEng) to regulate blood vessel growth. Imbalances in these factors can lead to aberrant placental vascular development. Throughout pregnancy, these factors are also released into the maternal circulation to aid in adapting the maternal cardiovascular system to pregnancy. Increased secretion of anti-angiogenic factors can lead to the development of an anti-angiogenic state in the mother and contribute to the development of pregnancy pathologies such as pre-eclampsia and foetal growth restriction (FGR). Thus, what are commonly referred to as 'angiogenic factors' have distinct functions in the maternal and placental circulations making this a misnomer. Indeed, technical issues in this field such as assay methodology and lack of data considering different placental cell types mean that the physiological roles of these factors in the maternal and placental circulations are frequently muddled in the literature. This review aims to (1) unpick the distinct roles of factors that influence placental vascular development and separate these from the roles of the same factors within the maternal circulation in normal pregnancy and (2) critically assess how imbalances may contribute to the distinct pathophysiological mechanisms underlying pregnancy disorders. Together, this critical assessment of the field endeavours to improve our ability to accurately use these factors as predictive/diagnostic biomarkers in the future.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/fisiología , Placenta/irrigación sanguínea , Circulación Placentaria , Complicaciones del Embarazo , Inhibidores de la Angiogénesis/farmacología , Angiopoyetinas/fisiología , Endoglina/fisiología , Femenino , Humanos , Placenta/efectos de los fármacos , Placenta/fisiología , Factor de Crecimiento Placentario/fisiología , Circulación Placentaria/efectos de los fármacos , Circulación Placentaria/genética , Embarazo , Complicaciones del Embarazo/genética , Complicaciones del Embarazo/metabolismo , Complicaciones del Embarazo/fisiopatología , Factor A de Crecimiento Endotelial Vascular/fisiología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/fisiología
4.
J. investig. allergol. clin. immunol ; 30(4): 272-280, 2020. tab, graf
Artículo en Inglés | IBECS | ID: ibc-194935

RESUMEN

BACKGROUND: Angiotensin-converting enzyme inhibitor-associated angioedema (ACEI-AAE) affects 0.1%-0.7% of patients treated with ACEIs. While previous research suggests that angioedema attacks result from increased vascular permeability, the pathogenesis is not completely understood. OBJECTIVE: This study aimed to describe the clinical, genetic, and laboratory parameters of ACEI-AAE patients and to investigate the role of vascular endothelial growth factors A and C (VEGF-A and VEGF-C), angiopoietins 1 and 2 (Ang1/Ang2), and secretory phospholipase A2 (sPLA2) in the pathogenesis of ACEI-AAE. METHODS: The clinical and laboratory data of ACEI-AAE patients were collected from 2 angioedema reference centers. Healthy volunteers and ACEI-treated patients without angioedema were enrolled to compare laboratory parameters. Genetic analyses to detect mutations in the genes SERPING1, ANGPT1, PLG, and F12 were performed in a subset of patients. RESULTS: A total of 51 patients (57% male) were diagnosed with ACEI-AAE. The average time to onset of symptoms from the start of ACEI therapy was 3 years (range, 30 days-20 years). The most commonly affected sites were the lips (74.5%), tongue (51.9%), and face (41.2%). Switching from ACEIs to sartans was not associated with an increased risk of angioedema in patients with a history of ACEIAAE. VEGF-A, VEGF-C, and sPLA2 plasma levels were higher in ACEI-AAE patients than in the controls. Ang1/2 concentrations remained unchanged. No mutations were detected in the genes analyzed. CONCLUSIONS: Our data suggest that sartans are a safe therapeutic alternative in ACEI-AAE patients. Increased concentrations of VEGF-A, VEGF-C, and sPLA2 in ACEI-AAE patients suggest a possible role of these mediators in the pathogenesis of ACEI-AAE


ANTECEDENTES: El angioedema asociado al consumo de inhibidores de la enzima convertidora de angiotensina (IECA-AAE) ocurre en el 0,1%-0,7% de los pacientes tratados con IECA. Aunque se sugiere que los ataques de angioedema son el resultado de una mayor permeabilidad vascular, la patogénesis de este proceso no está plenamente esclarecida. OBJETIVO: En este trabajo se estudiaron los parámetros clínicos, genéticos y de laboratorio de pacientes con IECA-AAE, así como el papel de los factores de crecimiento endotelial vascular A y C (VEGF-A y VEGF-C), las angiopoyetinas 1 y 2 (Ang1/Ang2) y la fosfolipasa secretora A2 (sPLA2). MÉTODOS: Se recogieron datos clínicos y de laboratorio de pacientes con IECA-AAE procedentes de dos centros de referencia en angioedema. Se utilizaron pacientes control, que incluyeron a voluntarios sanos y a pacientes tratados con IECA sin angioedema, para comparar las concentraciones de los parámetros de laboratorio. Finalmente, se realizó un análisis genético en un subconjunto de pacientes para detectar mutaciones en los genes SERPING1, ANGPT1, PLG y F12. RESULTADOS: Se diagnosticaron a 51 pacientes (57% hombres) con IECA-AAE. El tiempo promedio para el inicio de los síntomas desde el comienzo del tratamiento con IECA fue de 3 años (rango de 30 días a 20 años). Los lugares más comúnmente afectados fueron: labios (74,5%), lengua (51,9%) y cara (41,2%). El cambio de IECA a ARA-II no se asoció con un mayor riesgo de angioedema en pacientes con antecedentes de IECA-AAE. Los niveles plasmáticos de VEGF-A, VEGF-C y sPLA2 fueron más altos en pacientes con IECA-AAE que en los controles. No se detectaron cambios en las concentraciones de Ang1/Ang2, ni se detectaron mutaciones en los genes analizados. CONCLUSIONES: Nuestros datos sugieren que los ARA-II pueden ser una alternativa terapéutica segura en pacientes con IECA-AAE. El aumento de las concentraciones de VEGF-A, VEGF-C y sPLA2 en pacientes con ACEI-AAE sugiere un posible papel de estos mediadores en la patogénesis de esta enfermedad


Asunto(s)
Humanos , Masculino , Femenino , Adulto , Persona de Mediana Edad , Anciano , Anciano de 80 o más Años , Inhibidores de la Enzima Convertidora de Angiotensina/efectos adversos , Angioedema/inducido químicamente , Angioedema/genética , Factores de Crecimiento Endotelial/fisiología , Angiopoyetinas/fisiología , Fosfolipasas/fisiología , Mutación , Angioedema/fisiopatología , Estudios de Cohortes
5.
J Diabetes Res ; 2019: 5140521, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31485452

RESUMEN

Diabetic retinopathy (DR) is the commonest cause of blindness in the working-age population of the developed world. The molecular pathophysiology of DR is complex, and a complete spatiotemporal model of the disease is still being elucidated. Recently, a role for angiopoietin (Ang) proteins in the pathophysiology of DR has been proposed by several research groups, and several aspects of Ang signalling are being explored as novel therapeutic strategies. Here, we review the role of the Ang proteins in two important forms of DR, diabetic macular oedema and proliferative diabetic retinopathy. The function of the Ang proteins in regulating blood vessel permeability and neovascularisation is discussed, and we also evaluate recent preclinical and clinical studies highlighting the potential benefits of modulating Ang signalling as a treatment for DR.


Asunto(s)
Angiopoyetinas/fisiología , Retinopatía Diabética/etiología , Angiopoyetinas/sangre , Animales , Complicaciones de la Diabetes/sangre , Complicaciones de la Diabetes/tratamiento farmacológico , Diabetes Mellitus/sangre , Diabetes Mellitus/tratamiento farmacológico , Retinopatía Diabética/sangre , Retinopatía Diabética/prevención & control , Humanos , Hipoglucemiantes/uso terapéutico , Edema Macular/sangre , Edema Macular/etiología , Edema Macular/prevención & control , Transducción de Señal/efectos de los fármacos
6.
Actas dermo-sifiliogr. (Ed. impr.) ; 108(6): 515-523, jul.-ago. 2017. ilus, tab
Artículo en Inglés | IBECS | ID: ibc-164505

RESUMEN

Angiogenesis is the growth of new blood vessels from pre-existing vessels. It is a biological process essential in physiological wound healing or pathological inflammation and tumor growth, which underlies a complex interplay of stimulating and inhibiting signals. Extracellular matrix, cells of innate and adaptive immunity and endothelial cells itself are a major source of angiogenic factors that activate or inhibit specific receptors and consequently influence intracellular signaling pathways. Most inflammatory and neoplastic diseases in dermatology are characterized by excessive angiogenesis, such as psoriasis, atopic dermatitis, as well as melanoma, non-melanoma skin cancer, but also benign vascular neoplasia. In this article we describe current knowledge of angiogenesis and its most relevant mechanisms in different dermatological disorders with particular emphasis on the angiogenic factors (vascular endothelial growth factor) and angiopoietins as a target of current and future directions of anti-angiogenic therapy (AU)


La angiogénesis es el desarrollo de nuevos vasos a partir de estructuras vasculares preexistentes. Es un proceso biológico esencial en la cicatrización de las heridas, pero también en la inflamación y el crecimiento tumoral y es controlado por una compleja red de factores inhibitorios y estimulantes. La matriz extracelular, las células del sistema inmune innato y adaptativo así como las células endoteliales son fuente de factores angiogénicos que pueden estimular o inhibir receptores específicos y modificar la respuesta de distintas vías de señalización intracelular. La mayoría de las enfermedades inflamatorias y neoplásicas dermatológicas se caracterizan en general por un exceso de angiogénesis, como por ejemplo en la psoriasis, la dermatitis atópica, o el melanoma, así como en el cáncer cutáneo no melanocítico pero también en las neoplasias vasculares benignas. En este artículo de revisión describimos los conocimientos actuales del proceso de angiogénesis y sus mecanismos más relevantes en las diferentes enfermedades dermatológicas haciendo especial énfasis en los factores proangiogénicos como el factor de crecimiento vascular endotelial y las angiopoietinas como potenciales dianas terapéuticas (AU)


Asunto(s)
Humanos , Neovascularización Patológica/patología , Psoriasis/fisiopatología , Melanoma/fisiopatología , Angiopoyetinas/fisiología , Factores de Crecimiento Endotelial Vascular/fisiología , Inhibidores de la Angiogénesis/fisiología , Patología Molecular
7.
J Am Soc Nephrol ; 28(7): 1973-1982, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28465380

RESUMEN

Systemic inflammation is a hallmark of commonly encountered diseases ranging from bacterial sepsis to sterile syndromes such as major trauma. Derangements in the host vasculature contribute to the cardinal manifestations of sepsis in profound ways. Recent studies of control pathways regulating the vascular endothelium have illuminated how this single cell layer toggles between quiescence and activation to affect the development of shock and multiorgan dysfunction. This article focuses on one such control pathway, the Tie2 receptor and its ligands the angiopoietins, to describe a growing body of genetic, biochemical, mechanistic, and human studies that implicate Tie2 as a critical switch. In health, activated Tie2 maintains the endothelium in a quiescent state characterized by dynamic barrier function and antiadhesion against circulating leukocytes. In sepsis and related diseases, expression of the angiopoietins becomes markedly imbalanced and Tie2 signaling is greatly attenuated. These rapid molecular changes potentiate pathophysiologic responses throughout the body, resulting in injurious vascular leakage and organ inflammation. The Tie2 axis, therefore, may be a promising avenue for future translational studies.


Asunto(s)
Angiopoyetinas/fisiología , Inflamación/etiología , Receptor TIE-2/fisiología , Transducción de Señal , Animales , Humanos
8.
Metabolism ; 69: 157-170, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28285646

RESUMEN

INTRODUCTION AND AIMS: Several studies have reported that angiopoietin-like protein 2 (Angptl2) is expressed abundantly in adipocytes and is associated with adipose tissue inflammation. In the present study, we found that osteoblasts and mesenchymal stem cells also expressed Angptl2 at high levels. The aim of this study was to understand the role of Angptl2 in osteoblastic cell differentiation. METHODS: Angptl2 expression was examined during osteoblast and adipocyte differentiation. The role of Angptl2 on cell differentiation and associated signaling was analyzed by gene knockdown using Angptl2 small interfering ribonucleic acid (siRNA). RESULTS: Angptl2 was highly expressed in MC3T3-E1 cells, ST2 cells and primary osteoblasts, but not in RAW264 cells. Inhibition of Angptl2 expression using siRNA markedly inhibited alkaline phosphatase (ALP) expression and osteoblastic differentiation in MC3T3-E1, ST2 cells and primary osteoblasts. Angptl2 siRNA also inhibited adipocyte differentiation in ST2 cells. Treatment of MC3T3-E1 cells with Angptl2 siRNA led to the down-regulation of the activities of several cell signaling pathways, including extracellular signal-regulated kinase (ERK), Jun amino-terminal kinase (JNK), Akt, and nuclear factor kappa B (NF-κB) signals. It also down-regulated the expression of Osterix, but not that of runt-related transcription factor 2 (Runx2), suggesting that Angptl2 is a positive activator of Osterix and its down-stream signals. Treatment of MC3T3-E1 cells with anti-Angptl2 antibodies suppressed ALP gene expression. In addition, treatment of Angptl2 siRNA-treated cells with culture supernatants of normal MC3T3-E1 cells restored ALP gene expression, indicating that Angptl2 acts in an autocrine manner. CONCLUSIONS: The results suggest that Angptl2 is an autocrine positive regulator of cell differentiation. Thus, it is suggested that Angptl2 regulates not only adipose tissue metabolism but also bone metabolism.


Asunto(s)
Angiopoyetinas/genética , Angiopoyetinas/fisiología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Osteoblastos/fisiología , Adipocitos/fisiología , Proteína 2 Similar a la Angiopoyetina , Proteínas Similares a la Angiopoyetina , Animales , Comunicación Autocrina/genética , Comunicación Autocrina/fisiología , Células Cultivadas , Técnicas de Silenciamiento del Gen , Inflamación/genética , Inflamación/patología , Sistema de Señalización de MAP Quinasas/genética , Sistema de Señalización de MAP Quinasas/fisiología , Células Madre Mesenquimatosas , Ratones , ARN Interferente Pequeño , Transducción de Señal/genética , Transducción de Señal/fisiología
9.
Actas Dermosifiliogr ; 108(6): 515-523, 2017.
Artículo en Inglés, Español | MEDLINE | ID: mdl-28162227

RESUMEN

Angiogenesis is the growth of new blood vessels from pre-existing vessels. It is a biological process essential in physiological wound healing or pathological inflammation and tumor growth, which underlies a complex interplay of stimulating and inhibiting signals. Extracellular matrix, cells of innate and adaptive immunity and endothelial cells itself are a major source of angiogenic factors that activate or inhibit specific receptors and consequently influence intracellular signaling pathways. Most inflammatory and neoplastic diseases in dermatology are characterized by excessive angiogenesis, such as psoriasis, atopic dermatitis, as well as melanoma, non-melanoma skin cancer, but also benign vascular neoplasia. In this article we describe current knowledge of angiogenesis and its most relevant mechanisms in different dermatological disorders with particular emphasis on the angiogenic factors (vascular endothelial growth factor) and angiopoietins as a target of current and future directions of anti-angiogenic therapy.


Asunto(s)
Neovascularización Patológica/etiología , Neovascularización Fisiológica , Enfermedades de la Piel/complicaciones , Inhibidores de la Angiogénesis/efectos adversos , Inhibidores de la Angiogénesis/uso terapéutico , Proteínas Angiogénicas/fisiología , Angiopoyetinas/antagonistas & inhibidores , Angiopoyetinas/fisiología , Resistencia a Antineoplásicos , Hemangioma/tratamiento farmacológico , Hemangioma/fisiopatología , Humanos , Melanoma/complicaciones , Melanoma/tratamiento farmacológico , Melanoma/inmunología , Melanoma/fisiopatología , Terapia Molecular Dirigida , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/fisiología , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/fisiopatología , Psoriasis/tratamiento farmacológico , Psoriasis/fisiopatología , Receptor TIE-2/antagonistas & inhibidores , Receptor TIE-2/fisiología , Piel/irrigación sanguínea , Enfermedades de la Piel/inmunología , Enfermedades de la Piel/fisiopatología , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/complicaciones , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/fisiopatología
10.
Hepatology ; 64(5): 1637-1651, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27530187

RESUMEN

Angiopoietin-like protein 1 (ANGPTL1) has been shown to act as a tumor suppressor by inhibiting angiogenesis, cancer invasion, and metastasis. However, little is known about the effects of ANGPTL1 on sorafenib resistance and cancer stem cell properties in hepatocellular carcinoma (HCC) and the mechanism underlying these effects. Here, we show that ANGPTL1 expression positively correlates with sorafenib sensitivity in HCC cells and human HCC tissues. ANGPTL1 significantly decreases epithelial-mesenchymal transition (EMT)-driven sorafenib resistance, cancer stemness, and tumor growth of HCC cells by repressing Slug expression. ANGPTL1 directly interacts with and inactivates MET receptor, which contributes to Slug suppression through inhibition of the extracellular receptor kinase/protein kinase B (ERK/AKT)-dependent early growth response protein 1 (Egr-1) pathway. ANGPTL1 expression inversely correlates with Slug expression, poor sorafenib responsiveness, and poor clinical outcomes in HCC patients. CONCLUSION: ANGPTL1 inhibits sorafenib resistance and cancer stemness in HCC cells by repressing EMT through inhibition of the MET receptor-AKT/ERK-Egr-1-Slug signaling cascade. ANGPTL1 may serve as a novel MET receptor inhibitor for advanced HCC therapy. (Hepatology 2016;64:1637-1651).


Asunto(s)
Angiopoyetinas/fisiología , Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/tratamiento farmacológico , Resistencia a Antineoplásicos , Neoplasias Hepáticas/tratamiento farmacológico , Niacinamida/análogos & derivados , Compuestos de Fenilurea/uso terapéutico , Proteínas Proto-Oncogénicas c-met/fisiología , Proteína 1 Similar a la Angiopoyetina , Proteínas Similares a la Angiopoyetina , Animales , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Células Madre Neoplásicas , Niacinamida/uso terapéutico , Sorafenib
11.
Inflammation ; 39(3): 974-85, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26973239

RESUMEN

Angiopoietin-like protein 7 (Angptl7) has been extensively studied for decades, but its potential immune functions have not been characterized. Hence, we investigated the relationship between Angptl7 and inflammation by using RAW264.7 monocyte/macrophage cells. The expression of genes encoding inflammation-associated factors cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), tumor necrosis factor alpha (TNF-α), interleukin-1 beta (IL-1ß), IL-6, IL-10, and transforming growth factor beta 1 (TGF-ß1)) decreased after RAW264.7 cells were treated with anti-Angptl7 polyclonal antibody but increased after the cells were transfected with an Angptl7-expressing plasmid. Angptl7 overexpression enhanced phagocytosis and inhibited the proliferation of RAW264.7 cells. In addition, Angptl7 antagonized the anti-inflammatory effects of TGF-ß1 and dexamethasone. Pathway analysis showed that Angptl7 promoted the phosphorylation of both p65 and p38, but only the P38 mitogen-activated protein kinase (MAPK) signaling pathway mediated Angptl7-associated inflammatory functions. Additionally, after 1 week of daily intraperitoneal injections of recombinant TNF-α in a mouse model of peripheral inflammation, Angptl7 expression increased in the mouse eyes. Thus, Angptl7 is a factor that promotes pro-inflammatory responses in macrophages through the P38 MAPK signaling pathway and represents a potential therapeutic target for treatment of inflammatory diseases.


Asunto(s)
Angiopoyetinas/fisiología , Inflamación/tratamiento farmacológico , Macrófagos/patología , Transducción de Señal/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteína 7 Similar a la Angiopoyetina , Proteínas Similares a la Angiopoyetina , Animales , Inflamación/inducido químicamente , Ratones , Fosforilación , Células RAW 264.7 , Factor de Transcripción ReIA/metabolismo
12.
Endocrine ; 52(2): 187-93, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26754661

RESUMEN

Angiopoietin-like protein 3 (ANGPTL3) is a secretory protein regulating plasma lipid levels via affecting lipoprotein lipase- and endothelial lipase-mediated hydrolysis of triglycerides and phospholipids. ANGPTL3-deficiency due to loss-of-function mutations in the ANGPTL3 gene causes familial combined hypobetalipoproteinemia (FHBL2, OMIM # 605019), a phenotype characterized by low concentration of all major lipoprotein classes in circulation. ANGPTL3 is therefore a potential therapeutic target to treat combined hyperlipidemia, a major risk factor for atherosclerotic coronary heart disease. This review focuses on the mechanisms behind ANGPTL3-deficiency induced FHBL2.


Asunto(s)
Angiopoyetinas/fisiología , Hipobetalipoproteinemias/genética , Metabolismo de los Lípidos/genética , Lipoproteínas/metabolismo , Adulto , Anciano , Proteína 3 Similar a la Angiopoyetina , Proteínas Similares a la Angiopoyetina , Femenino , Humanos , Hipobetalipoproteinemias/metabolismo , Masculino , Persona de Mediana Edad
14.
Nefrologia ; 35(2): 131-8, 2015.
Artículo en Inglés, Español | MEDLINE | ID: mdl-26300505

RESUMEN

The prevalence of diabetes mellitus increased during the last century and it is estimated that 45% of the patients are not diagnosed. In South America the prevalence of diabetes and chronic kidney disease (CKD) increased, with a great disparity among the countries with respect to access to dialysis. In Ecuador it is one of the main causes of mortality, principally in the provinces located on the coast of the Pacific Ocean. The greatest single cause of beginning dialysis is diabetic nephropathy (DN). Even using the best therapeutic options for DN, the residual risk of proteinuria and of terminal CKD remains high. In this review we indicate the importance of the problem globally and in our region. We analyse relevant cellular and molecular studies that illustrate the crucial significance of glomerular events in DN development and evolution and in insulin resistance. We include basic anatomical, pathophysiological and clinical concepts, with special attention to the role of angiogenic factors such as the vascular endothelial growth factor (VEGF-A) and their relationship to the insulin receptor, endothelial isoform of nitric oxide synthase (eNOS) and angiopoietins. We also propose various pathways that have therapeutic potential in our opinion. Greater in-depth study of VEGF-A and angiopoietins, the state of glomerular VEGF resistance, the relationship of VEGF receptor 2/nephrin, VEGF/insulin receptors/nephrin and the relationship of VEGF/eNOS-NO at glomerular level could provide solutions to the pressing world problem of DN and generate new treatment alternatives.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Factor A de Crecimiento Endotelial Vascular/fisiología , Angiopoyetinas/fisiología , Nefropatías Diabéticas/epidemiología , Nefropatías Diabéticas/patología , Nefropatías Diabéticas/fisiopatología , Salud Global , Humanos , Resistencia a la Insulina , Glomérulos Renales/metabolismo , Glomérulos Renales/patología , Proteínas de la Membrana/fisiología , Óxido Nítrico/fisiología , Óxido Nítrico Sintasa de Tipo III/fisiología , Receptor de Insulina/fisiología , Receptores de Factores de Crecimiento Endotelial Vascular/fisiología , Transducción de Señal
15.
Proc Natl Acad Sci U S A ; 112(23): E3030-9, 2015 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-26039997

RESUMEN

Diabetic eye disease is the most common cause of severe vision loss in the working-age population in the developed world, and proliferative diabetic retinopathy (PDR) is its most vision-threatening sequela. In PDR, retinal ischemia leads to the up-regulation of angiogenic factors that promote neovascularization. Therapies targeting vascular endothelial growth factor (VEGF) delay the development of neovascularization in some, but not all, diabetic patients, implicating additional factor(s) in PDR pathogenesis. Here we demonstrate that the angiogenic potential of aqueous fluid from PDR patients is independent of VEGF concentration, providing an opportunity to evaluate the contribution of other angiogenic factor(s) to PDR development. We identify angiopoietin-like 4 (ANGPTL4) as a potent angiogenic factor whose expression is up-regulated in hypoxic retinal Müller cells in vitro and the ischemic retina in vivo. Expression of ANGPTL4 was increased in the aqueous and vitreous of PDR patients, independent of VEGF levels, correlated with the presence of diabetic eye disease, and localized to areas of retinal neovascularization. Inhibition of ANGPTL4 expression reduced the angiogenic potential of hypoxic Müller cells; this effect was additive with inhibition of VEGF expression. An ANGPTL4 neutralizing antibody inhibited the angiogenic effect of aqueous fluid from PDR patients, including samples from patients with low VEGF levels or receiving anti-VEGF therapy. Collectively, our results suggest that targeting both ANGPTL4 and VEGF may be necessary for effective treatment or prevention of PDR and provide the foundation for studies evaluating aqueous ANGPTL4 as a biomarker to help guide individualized therapy for diabetic eye disease.


Asunto(s)
Angiopoyetinas/fisiología , Retinopatía Diabética/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Angiopoyetinas/metabolismo , Retinopatía Diabética/metabolismo , Ojo/irrigación sanguínea , Ojo/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neovascularización Patológica , Factor A de Crecimiento Endotelial Vascular/sangre
16.
J Vasc Res ; 52(1): 22-31, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25896412

RESUMEN

Clinical and experimental studies have been focused on the pathophysiological mechanisms induced by brain ischemia-reperfusion injury. Recovery events, such as neurogenesis, angiogenesis and the growth of new blood vessels from the preexisting vascular tree, have been intensively studied in the last decades to clarify the vascular remodeling crucial for stroke outcome. This review aims to discuss the cerebral microcirculation remodeling induced by ischemia-reperfusion and the mechanisms involved in angiogenesis and vasculogenesis. The first in vivo observations were focused on anastomotic shunting of cerebral blood flow (CBF) in experimental and clinical models. Thereafter, vascular remodeling induced by cerebral ischemia-reperfusion was reported in mice and rats. Successively, other studies have assessed that within 30 days of middle cerebral artery (MCA) occlusion in rats, there is an increase in CBF and recovery from stroke. Recently, rats submitted to transient MCA occlusion showed pial microcirculation remodeling with the formation of new arterioles sprouting from penumbra vessels and overlapping the ischemic core. This review focuses on the production and/or activation of vasculotrophic factors able to trigger and facilitate microvascular remodeling. Vascular endothelial growth factor and endothelium-released nitric oxide appear to be the main factors involved in the formation of new vessels during microvascular remodeling. These studies are fundamental for consequent interventions on molecular targets, useful for fostering vascular remodeling and the recovery of functions.


Asunto(s)
Isquemia Encefálica/fisiopatología , Encéfalo/irrigación sanguínea , Circulación Cerebrovascular/fisiología , Microcirculación/fisiología , Daño por Reperfusión/fisiopatología , Remodelación Vascular/fisiología , Angiopoyetinas/fisiología , Animales , Arteriolas/fisiología , Arterias Cerebrales/fisiopatología , Circulación Colateral , Endotelio Vascular/fisiopatología , Humanos , Infarto de la Arteria Cerebral Media/fisiopatología , Ratones , Neovascularización Fisiológica/fisiología , Óxido Nítrico/fisiología , Piamadre/irrigación sanguínea , Ratas , Factor A de Crecimiento Endotelial Vascular/fisiología
17.
Nefrología (Madr.) ; 35(2): 131-138, mar.-abr. 2015. ilus
Artículo en Español | IBECS | ID: ibc-139278

RESUMEN

La prevalencia de diabetes mellitus aumentó en el último siglo y se estima que el 45%de los pacientes, no estarían diagnosticados. En Sudamérica la prevalencia de diabetes y de enfermedad renal crónica (ERC) incrementó, existiendo gran disparidad entre los países respecto al acceso a diálisis. En Ecuador es una de las principales causas de mortalidad, principalmente en las provincias ubicadas en la costa del océano Pacífico. La mayor causa aislada de ingreso a diálisis es la nefropatía diabética (ND). Aun utilizando las mejores opciones terapéuticas para la ND, el riesgo residual de proteinuria y de ERC terminal permanece elevado. En esta revisión describimos la importancia del problema en el mundo y en nuestra región Analizamos estudios moleculares y celulares relevantes que indican la crucial importancia de eventos glomerulares en el desarrollo y en la evolución de la ND y en la insulinorresistencia. Incluimos conceptos anatómicos, fisiopatológicos y clínicos básicos, desarrollando especial énfasis en el rol de factores angiogénicos como el factor de crecimiento vascular endotelial(VEGF-A) y su relación con el receptor de insulina, la sintasa endotelial de óxido nítrico-óxidonítrico (eNOS) y las angiopoietinas. En el transcurso del texto proponemos diversas vías, que a nuestro entender tienen potencial terapéutico. Profundizar en el estudio del VEGF-A y la sangiopoietinas, el estado de VEGF resistencia glomerular, la relación del receptor 2 de VEGF/nefrina, VEGF/receptores de insulina/nefrina, la relación VEGF/eNOS-ON a nivel glomerular podría aportar soluciones al acuciante problema de la ND en el mundo y generar nuevas alternativas de tratamiento (AU)


The prevalence of diabetes mellitus increased during the last century and it is estimated that45% of the patients are not diagnosed. In South America the prevalence of diabetes and chronic kidney disease (CKD) increased, with a great disparity among the countries with respect to access to dialysis. In Ecuador it is one of the main causes of mortality, principally in the provinces located on the coast of the Pacific Ocean. The greatest single cause of beginning dialysis is diabetic nephropathy (DN). Even using the best therapeutic options for DN, the residual risk of proteinuria and of terminal CKD remains high. In this review we indicate the importance of the problem globally and in our region. We analyse relevant cellular and molecular studies that illustrate the crucial significance of glomerular events in DN development and evolution and in insulin resistance. We include basic anatomical, pathophysiological and clinical concepts, with special attention to the role of angiogenic factors such as the vascular endothelial growth factor (VEGF-A) and their relationship to the insulin receptor, endothelial isoform of nitric oxide synthase (eNOS) and angiopoietins. We also propose various pathways that have therapeutic potential in our opinion. Greater in-depth study of VEGF-A and angiopoietins, the state of glomerular VEGF resistance, the relationship of VEGF receptor 2/nephrin, VEGF/insulin receptors/nephrin and the relationship of VEGF/eNOS-NO at glomerular level could provide solutions to the pressing world problem of DN and generate new treatment alternatives (AU)


Asunto(s)
Humanos , Nefropatías Diabéticas/fisiopatología , Receptores de Factores de Crecimiento Endotelial Vascular/fisiología , Receptor de Insulina/fisiología , Angiopoyetinas/fisiología , Podocitos/fisiología , Resistencia a la Insulina/fisiología , Óxido Nítrico/fisiología , Tasa de Filtración Glomerular/fisiología
18.
PLoS One ; 10(1): e0116838, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25622036

RESUMEN

PURPOSE: We sought to identify the anti-angiogenic molecule expressed in corneal keratocytes that is responsible for maintaining the avascularity of the cornea. METHODS: Human umbilical vein endothelial cells (HUVECs) were cultured with either human dermal fibroblasts or with human corneal keratocytes under serum-free conditions. The areas that exhibited blood vessel formation were estimated by immunostaining the cultures with an antitibody against CD31, a blood vessel marker. We also performed microarray gene-expression analysis and selected one molecule, angiopoietin-like 7 (ANGPTL7) for further functional studies conducted with the keratocytes and in vivo in mice. RESULTS: Areas showing blood vessel formation in normal serum-free medium were conditions were markedly smaller when HUVECs were co-cultured with corneal keratocytes than when they were co-cultured with the dermal fibroblasts under the same conditions. Microarray analysis revealed that ANGPTL7 expression was higher in keratocytes than in dermal fibroblasts. In vitro, inhibiting ANGPTL7 expression by using a specific siRNA led to greater tube formation than did the transfection of cells with a control siRNA, and this increase in tube formation was abolished when recombinant ANGPTL7 protein was added to the cultures. In vivo, intrastromal injections of an ANGPTL7 PshRNA into the avascular corneal stroma of mice resulted in the growth of blood vessels. CONCLUSIONS: ANGPTL7, which is abundantly expressed in keratocytes, plays a major role in maintaining corneal avascularity and transparency.


Asunto(s)
Angiopoyetinas/fisiología , Neovascularización de la Córnea/metabolismo , Proteína 7 Similar a la Angiopoyetina , Proteínas Similares a la Angiopoyetina , Animales , Células Cultivadas , Técnicas de Cocultivo , Fibroblastos/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Queratinocitos/metabolismo , Masculino , Ratones Endogámicos C57BL , Transcriptoma
19.
J Gastroenterol Hepatol ; 30(2): 396-404, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25090954

RESUMEN

BACKGROUND AND AIM: Angiopoietin-like protein 2 (ANGPTL2) plays various roles in metabolism, vascular biology, inflammation, and tumor metastasis, but little is known about its function in human hepatocellular carcinoma (HCC) metastasis. This study aimed to further explore the function of ANGPTL2 on migration and invasion of liver cancer cells. METHODS: Quantitative real-time polymerase chain reaction, Western blotting, immunohistochemistry, transwell migration, and invasion assays were performed to clarify the function of ANGPTL2 in the regulation of cell migration and invasion in human HCC. RESULTS: In HCC patients, ANGPTL2 expression was higher in HCC tissues compared with matched noncancerous liver tissues. And the ANGPTL2 levels of HCC tissues positively correlated with intrahepatic metastasis in HCC patients. Overexpression of ANGPTL2 significantly increased migration and invasion of HCC cells in vitro, and promoted intrahepatic and distal pulmonary metastasis in vivo, while knockdown of endogenous ANGPTL2 resulted in a reduced migration and invasion in vitro. Colony formation assay and 3-(4,5-Dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay showed ANGPTL2 did not affect cell proliferation in vitro, whereas overexpression of ANGPTL2 promoted tumor formation in xenograft animal model. CONCLUSIONS: Our findings show that ANGPTL2 drives human HCC metastasis and provides a potential therapeutic target for HCC treatment.


Asunto(s)
Angiopoyetinas/fisiología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Proteína 2 Similar a la Angiopoyetina , Proteínas Similares a la Angiopoyetina , Animales , Línea Celular Tumoral , Movimiento Celular/genética , Modelos Animales de Enfermedad , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Masculino , Ratones Endogámicos BALB C , Terapia Molecular Dirigida , Invasividad Neoplásica/genética , Metástasis de la Neoplasia/genética , Trasplante de Neoplasias
20.
Oncol Rep ; 33(1): 58-66, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25370833

RESUMEN

Angiopoietin-like proteins (ANGPTLs), which comprise 7 members (ANGPTL1-ANGPTL7), structurally resemble angiopoietins. We investigated the roles of ANGPTLs in the acquisition of androgen independence and the malignant behavior of human prostate cancer cells. Expression of ANGPTL messenger RNA (mRNA) and proteins were ascertained using RT-qPCR and western blot analysis in human prostate cancer cell lines. Androgen­dependent LNCaP and androgen-independent LNCaP/AI cells, respectively, were cultured in fetal bovine and charcoal-stripped medium. Cell proliferation, androgen dependence, migration and invasion, respectively, were examined under the overexpression and knockdown of ANGPTL2 by transfection of ANGPTL2 cDNA and its small­interfering RNA (siRNA). The effects of exogenous ANGPTL2 and blocking of its receptor, integrin α5ß1, were also investigated. Human prostate cancer cell lines predominantly expressed ANGPTL2 among the members. Interrupting ANGPTL2 expression with siRNA suppressed the proliferation, migration and invasion of LNCaP cells. LNCaP/AI cells showed a higher ANGPTL2 expression than that of LNCaP cells. Furthermore, siRNA led to apoptosis of LNCaP/AI cells. The ANGPTL2-overexpressing LNCaP cells markedly increased proliferation, epithelial-to-mesenchymal transition (EMT) and malignant behavior in androgen­deprived medium. The migration rates were increased depending on the concentration of ANGPTL2 recombinant protein and were inhibited by anti-integrin α5ß1 antibodies. To the best of our knowledge, this is the first study to elucidate the expression of ANGPTL2 in human prostate cancer cells. ANGPTL2 may be important in the acquisition of androgen independency and tumor progression of prostate cancer in an autocrine and/or paracrine manner via the integrin α5ß1 receptor. Targeting ANGPTL2 may therefore be an efficacious therapeutic modality for prostate cancer.


Asunto(s)
Angiopoyetinas/fisiología , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Proteína 2 Similar a la Angiopoyetina , Proteínas Similares a la Angiopoyetina , Proteínas Reguladoras de la Apoptosis/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Transición Epitelial-Mesenquimal , Expresión Génica , Humanos , Integrina alfa5beta1/metabolismo , Masculino , Neoplasias de la Próstata Resistentes a la Castración/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...