Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 139
Filtrar
1.
Int J Mol Sci ; 22(21)2021 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-34769465

RESUMO

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.


Assuntos
Dermatite/fisiopatologia , Neovascularização Patológica , Angiopoietinas/genética , Angiopoietinas/fisiologia , Animais , Doença Crônica , Dermatite/complicações , Dermatite/genética , Dermatite/patologia , Dermatite Atópica/etiologia , Dermatite Atópica/patologia , Dermatite Atópica/fisiopatologia , Humanos , Neovascularização Patológica/complicações , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Neovascularização Patológica/fisiopatologia , Psoríase/etiologia , Psoríase/patologia , Psoríase/fisiopatologia , Rosácea/etiologia , Rosácea/patologia , Rosácea/fisiopatologia , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/fisiologia
2.
Sci Rep ; 11(1): 419, 2021 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-33432093

RESUMO

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.


Assuntos
Angiopoietinas/fisiologia , Vasos Sanguíneos/crescimento & desenvolvimento , Modelos Teóricos , Neoplasias/irrigação sanguínea , Neovascularização Patológica/patologia , Angiopoietinas/genética , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia , Simulação por Computador , Progressão da Doença , Humanos , Regressão Neoplásica Espontânea/genética , Regressão Neoplásica Espontânea/patologia , Neoplasias/patologia , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Indução de Remissão , Fator A de Crescimento do Endotélio Vascular/fisiologia
3.
Angiogenesis ; 23(2): 105-117, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31707538

RESUMO

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.


Assuntos
Indutores da Angiogênese/farmacologia , Inibidores da Angiogênese/fisiologia , Placenta/irrigação sanguínea , Circulação Placentária , Complicações na Gravidez , Inibidores da Angiogênese/farmacologia , Angiopoietinas/fisiologia , Endoglina/fisiologia , Feminino , Humanos , Placenta/efeitos dos fármacos , Placenta/fisiologia , Fator de Crescimento Placentário/fisiologia , Circulação Placentária/efeitos dos fármacos , Circulação Placentária/genética , Gravidez , Complicações na Gravidez/genética , Complicações na Gravidez/metabolismo , Complicações na Gravidez/fisiopatologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/fisiologia
4.
J Diabetes Res ; 2019: 5140521, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31485452

RESUMO

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.


Assuntos
Angiopoietinas/fisiologia , Retinopatia Diabética/etiologia , Angiopoietinas/sangue , Animais , Complicações do Diabetes/sangue , Complicações do Diabetes/tratamento farmacológico , Diabetes Mellitus/sangue , Diabetes Mellitus/tratamento farmacológico , Retinopatia Diabética/sangue , Retinopatia Diabética/prevenção & controle , Humanos , Hipoglicemiantes/uso terapêutico , Edema Macular/sangue , Edema Macular/etiologia , Edema Macular/prevenção & controle , Transdução de Sinais/efeitos dos fármacos
5.
J Am Soc Nephrol ; 28(7): 1973-1982, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28465380

RESUMO

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.


Assuntos
Angiopoietinas/fisiologia , Inflamação/etiologia , Receptor TIE-2/fisiologia , Transdução de Sinais , Animais , Humanos
6.
Metabolism ; 69: 157-170, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28285646

RESUMO

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.


Assuntos
Angiopoietinas/genética , Angiopoietinas/fisiologia , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Osteoblastos/fisiologia , Adipócitos/fisiologia , Proteína 2 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina , Animais , Comunicação Autócrina/genética , Comunicação Autócrina/fisiologia , Células Cultivadas , Técnicas de Silenciamento de Genes , Inflamação/genética , Inflamação/patologia , Sistema de Sinalização das MAP Quinases/genética , Sistema de Sinalização das MAP Quinases/fisiologia , Células-Tronco Mesenquimais , Camundongos , RNA Interferente Pequeno , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
7.
Actas Dermosifiliogr ; 108(6): 515-523, 2017.
Artigo em Inglês, Espanhol | MEDLINE | ID: mdl-28162227

RESUMO

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.


Assuntos
Neovascularização Patológica/etiologia , Neovascularização Fisiológica , Dermatopatias/complicações , Inibidores da Angiogênese/efeitos adversos , Inibidores da Angiogênese/uso terapêutico , Proteínas Angiogênicas/fisiologia , Angiopoietinas/antagonistas & inibidores , Angiopoietinas/fisiologia , Resistencia a Medicamentos Antineoplásicos , Hemangioma/tratamento farmacológico , Hemangioma/fisiopatologia , Humanos , Melanoma/complicações , Melanoma/tratamento farmacológico , Melanoma/imunologia , Melanoma/fisiopatologia , Terapia de Alvo Molecular , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/fisiologia , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/fisiopatologia , Psoríase/tratamento farmacológico , Psoríase/fisiopatologia , Receptor TIE-2/antagonistas & inibidores , Receptor TIE-2/fisiologia , Pele/irrigação sanguínea , Dermatopatias/imunologia , Dermatopatias/fisiopatologia , Neoplasias Cutâneas/irrigação sanguínea , Neoplasias Cutâneas/complicações , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/fisiopatologia
8.
Hepatology ; 64(5): 1637-1651, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27530187

RESUMO

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).


Assuntos
Angiopoietinas/fisiologia , Antineoplásicos/uso terapêutico , Carcinoma Hepatocelular/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos , Neoplasias Hepáticas/tratamento farmacológico , Niacinamida/análogos & derivados , Compostos de Fenilureia/uso terapêutico , Proteínas Proto-Oncogênicas c-met/fisiologia , Proteína 1 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina , Animais , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Células-Tronco Neoplásicas , Niacinamida/uso terapêutico , Sorafenibe
9.
Inflammation ; 39(3): 974-85, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26973239

RESUMO

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.


Assuntos
Angiopoietinas/fisiologia , Inflamação/tratamento farmacológico , Macrófagos/patologia , Transdução de Sinais/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteína 7 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina , Animais , Inflamação/induzido quimicamente , Camundongos , Fosforilação , Células RAW 264.7 , Fator de Transcrição RelA/metabolismo
10.
Endocrine ; 52(2): 187-93, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26754661

RESUMO

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.


Assuntos
Angiopoietinas/fisiologia , Hipobetalipoproteinemias/genética , Metabolismo dos Lipídeos/genética , Lipoproteínas/metabolismo , Adulto , Idoso , Proteína 3 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina , Feminino , Humanos , Hipobetalipoproteinemias/metabolismo , Masculino , Pessoa de Meia-Idade
12.
Nefrologia ; 35(2): 131-8, 2015.
Artigo em Inglês, Espanhol | MEDLINE | ID: mdl-26300505

RESUMO

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.


Assuntos
Nefropatias Diabéticas/metabolismo , Fator A de Crescimento do Endotélio Vascular/fisiologia , Angiopoietinas/fisiologia , Nefropatias Diabéticas/epidemiologia , Nefropatias Diabéticas/patologia , Nefropatias Diabéticas/fisiopatologia , Saúde Global , Humanos , Resistência à Insulina , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Proteínas de Membrana/fisiologia , Óxido Nítrico/fisiologia , Óxido Nítrico Sintase Tipo III/fisiologia , Receptor de Insulina/fisiologia , Receptores de Fatores de Crescimento do Endotélio Vascular/fisiologia , Transdução de Sinais
13.
Proc Natl Acad Sci U S A ; 112(23): E3030-9, 2015 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-26039997

RESUMO

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.


Assuntos
Angiopoietinas/fisiologia , Retinopatia Diabética/tratamento farmacológico , Adulto , Idoso , Idoso de 80 Anos ou mais , Angiopoietinas/metabolismo , Retinopatia Diabética/metabolismo , Olho/irrigação sanguínea , Olho/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neovascularização Patológica , Fator A de Crescimento do Endotélio Vascular/sangue
14.
J Vasc Res ; 52(1): 22-31, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25896412

RESUMO

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.


Assuntos
Isquemia Encefálica/fisiopatologia , Encéfalo/irrigação sanguínea , Circulação Cerebrovascular/fisiologia , Microcirculação/fisiologia , Traumatismo por Reperfusão/fisiopatologia , Remodelação Vascular/fisiologia , Angiopoietinas/fisiologia , Animais , Arteríolas/fisiologia , Artérias Cerebrais/fisiopatologia , Circulação Colateral , Endotélio Vascular/fisiopatologia , Humanos , Infarto da Artéria Cerebral Média/fisiopatologia , Camundongos , Neovascularização Fisiológica/fisiologia , Óxido Nítrico/fisiologia , Pia-Máter/irrigação sanguínea , Ratos , Fator A de Crescimento do Endotélio Vascular/fisiologia
15.
PLoS One ; 10(1): e0116838, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25622036

RESUMO

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.


Assuntos
Angiopoietinas/fisiologia , Neovascularização da Córnea/metabolismo , Proteína 7 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina , Animais , Células Cultivadas , Técnicas de Cocultura , Fibroblastos/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Queratinócitos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Transcriptoma
16.
Oncol Rep ; 33(1): 58-66, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25370833

RESUMO

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.


Assuntos
Angiopoietinas/fisiologia , Neoplasias de Próstata Resistentes à Castração/metabolismo , Proteína 2 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina , Proteínas Reguladoras de Apoptose/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Transição Epitelial-Mesenquimal , Expressão Gênica , Humanos , Integrina alfa5beta1/metabolismo , Masculino , Neoplasias de Próstata Resistentes à Castração/patologia
17.
J Oral Pathol Med ; 44(2): 126-33, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25060575

RESUMO

Lymph node metastasis is a major factor for poor prognosis in oral squamous cell carcinoma (OSCC). However, the molecular mechanisms of lymph node metastasis are unclear. We determined that angiopoietin-like protein 4 (ANGPTL4) mRNA and protein expression were increased in OSCC cells established from the primary site in metastatic cases. In addition, ANGPTL4 expression in biopsy specimens was correlated with the presence of lymph node metastasis. Therefore, our initial findings suggest that OSCC cells expressing ANGPTL4 may possess metastatic ability. Furthermore, cell culture supernatants from OSCC cells that metastasized to the lymph node contain ANGPTL4 and promote invasive ability. These findings suggest that secreted ANGPTL4 may affect the invasive ability of OSCC. Moreover, the rates of positive ANGPTL4 expression at the primary site were significantly higher in the lymph node metastasis group. These results demonstrate that ANGPTL4 contributes to OSCC metastasis by stimulating cell invasion. Therefore, ANGPTL4 is a potential therapeutic target for preventing cancer metastasis.


Assuntos
Angiopoietinas/fisiologia , Carcinoma de Células Escamosas/secundário , Metástase Linfática/patologia , Neoplasias Bucais/patologia , Proteína 4 Semelhante a Angiopoietina , Angiopoietinas/genética , Biomarcadores Tumorais/análise , Biópsia , Carcinoma de Células Escamosas/química , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Meios de Cultivo Condicionados , Feminino , Técnicas de Silenciamento de Genes , Inativação Gênica , Humanos , Imuno-Histoquímica , Masculino , Neoplasias Bucais/química , Gradação de Tumores , RNA Interferente Pequeno/genética
18.
J Gastroenterol Hepatol ; 30(2): 396-404, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25090954

RESUMO

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.


Assuntos
Angiopoietinas/fisiologia , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Proteína 2 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina , Animais , Linhagem Celular Tumoral , Movimento Celular/genética , Modelos Animais de Doenças , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Masculino , Camundongos Endogâmicos BALB C , Terapia de Alvo Molecular , Invasividade Neoplásica/genética , Metástase Neoplásica/genética , Transplante de Neoplasias
19.
J Pharmacol Sci ; 125(4): 333-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25030744

RESUMO

Cerebral ischemia, a pathological condition in which brain tissue experiences a shortage of cerebral blood flow, is associated with cerebrovascular disease, brain trauma, epilepsy, and cardiac arrest. A reduction in blood flow leaves the brain tissue unsupplied with oxygen and glucose, thus leading to cell death in the ischemic core as well as subsequent peripheral injury in the penumbra. Neurons in the penumbra, where reperfusion occurs, are functionally inactive but still viable. Many biochemical changes, which may lead to neuronal cell death, thereby induce dysfunction of the central nervous system. However, the mechanisms responsible for ischemic stroke-induced cell damage remain to be determined. Protein phosphorylation has been implicated in the regulation of diverse cellular responses in the brain. Initially, tyrosine phosphorylation was considered to be involved in the regulation of cell growth and development. In addition, a variety of synaptic and cellular functions mediated by tyrosine phosphorylation in the brain were found to be associated with relatively high levels of protein tyrosine kinase activity. However, the involvement of this protein tyrosine kinase activity in ischemic cell death is still not fully understood. This review summarizes recent advances dealing with the possible implications of protein tyrosine phosphorylation in the ischemic brain.


Assuntos
Isquemia Encefálica/metabolismo , Tirosina/metabolismo , Angiopoietinas/fisiologia , Animais , Isquemia Encefálica/etiologia , Proteínas de Transporte , Efrinas/metabolismo , Proteínas de Arcabouço Homer , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Terapia de Alvo Molecular , Fatores de Crescimento Neural/fisiologia , Ocludina/metabolismo , Fosforilação , Proteínas Tirosina Quinases/fisiologia , Receptores da Família Eph/metabolismo , Receptores de Glutamato/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA