Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Biochim Biophys Acta Bioenerg ; 1859(9): 975-983, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29580805

RESUMO

Succinate is known to act as an inflammatory signal in classically activated macrophages through stabilization of HIF-1α leading to IL-1ß production. Relevant to this, hypoxia is known to drive succinate accumulation and release into the extracellular milieu. The metabolic alterations associated with succinate release during inflammation and under hypoxia are poorly understood. Data are presented showing that Mycoplasma arginini infection of VM-M3 cancer cells enhances the Warburg effect associated with succinate production in mitochondria and eventual release into the extracellular milieu. We investigated how succinate production and release was related to the changes of other soluble metabolites, including itaconate and 2-HG. Furthermore, we found that hypoxia alone could induce succinate release from the VM-M3 cells and that this could occur in the absence of glucose-driven lactate production. Our results elucidate metabolic pathways responsible for succinate accumulation and release in cancer cells, thus identifying potential targets involved in both inflammation and hypoxia. This article is part of a Special Issue entitled 20th European Bioenergetics Conference, edited by László Zimányi and László Tretter.


Assuntos
Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Hipóxia/complicações , Inflamação/complicações , Infecções por Mycoplasma/complicações , Mycoplasma/patogenicidade , Succinatos/metabolismo , Animais , Neoplasias Encefálicas/etiologia , Neoplasias Encefálicas/metabolismo , Metabolismo Energético , Glioblastoma/etiologia , Glioblastoma/metabolismo , Metaboloma , Camundongos , Células Tumorais Cultivadas
2.
J Cell Sci ; 125(Pt 4): 831-43, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22399811

RESUMO

Vascular endothelial growth factor (VEGF) plays a crucial role in developmental and pathological angiogenesis. Expression of VEGF in quiescent adult tissue suggests a potential role in the maintenance of mature blood vessels. We demonstrate, using a Vegf-lacZ reporter mouse model, that VEGF is expressed by arterial but not by venous or capillary endothelial cells (ECs) in vivo. Using an in vitro model, we show that arterial shear stress of human umbilical vein ECs (HUVECs) decreases apoptosis and increases VEGF expression, which is mediated by the induction of Krüppel-like factor 2 (KLF2). Additionally, shear stress stimulates the expression of VEGF receptor 2 (VEGFR2) and is associated with its activation. Knockdown of VEGF in shear stressed HUVECs blocks the protective effect of shear stress, resulting in EC apoptosis equivalent to that in control ECs cultured under static conditions. Similarly, treatment of ECs subjected to arterial shear stress with the VEGF receptor tyrosine kinase inhibitor SU1498, or VEGFR2 neutralizing antiserum, led to increased apoptosis, demonstrating that the mechanoprotection from increased shear is mediated by VEGFR2. Taken together, these studies suggest that arterial flow induces VEGF-VEGFR2 autocrine-juxtacrine signaling, which is a previously unidentified mechanism for vascular EC survival in adult arterial blood vessels.


Assuntos
Células Endoteliais/citologia , Células Endoteliais/metabolismo , Estresse Mecânico , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Artérias/citologia , Capilares/citologia , Sobrevivência Celular , Ativação Enzimática , Feminino , Células Endoteliais da Veia Umbilical Humana , Humanos , Fatores de Transcrição Kruppel-Like/metabolismo , Masculino , Camundongos , Receptores de Fatores de Crescimento do Endotélio Vascular/biossíntese , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/biossíntese , Veias/citologia
3.
FASEB J ; 27(8): 3257-71, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23682123

RESUMO

Vascular endothelial growth factor (VEGF) is critical for angiogenesis, but also has pleiotropic effects on several nonvascular cells. Our aim was to investigate the role of VEGF in brown adipose tissue (BAT). We show that VEGF expression increases 2.5-fold during differentiation of cultured murine brown adipocytes and that VEGF receptor-2 is phosphorylated, indicating VEGF signaling. VEGF increased proliferation in brown preadipocytes in vitro by 70%, and blockade of VEGF signaling using anti-VEGFR2 antibody DC101 increased brown adipocyte apoptosis, as determined by cell number and activation of caspase 3. Systemic VEGF neutralization in mice, accomplished by adenoviral expression of soluble Flt1, resulted in 7-fold increase in brown adipocyte apoptosis, mitochondrial degeneration, and increased mitophagy compared to control mice expressing a null adenovirus. Absence of the heparan sulfate-binding VEGF isoforms, VEGF164 and VEGF188, resulted in abnormal BAT development in mice at E15.5, with fewer brown adipocytes and lower mitochondrial protein compared to wild-type littermates. These results suggest a role for VEGF in brown adipocytes and preadipocytes to promote survival, proliferation, and normal mitochondria and development.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo Marrom/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Tecido Adiposo Marrom/citologia , Tecido Adiposo Marrom/embriologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Western Blotting , Caspase 3/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Células Cultivadas , Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
4.
Arterioscler Thromb Vasc Biol ; 33(11): 2608-17, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23968981

RESUMO

OBJECTIVE: Vascular endothelial cells (ECs) are continuously exposed to blood flow that contributes to the maintenance of vessel structure and function; however, the effect of hemodynamic forces on transforming growth factor-ß (TGF-ß) signaling in the endothelium is poorly described. We examined the potential role of TGF-ß signaling in mediating the protective effects of shear stress on ECs. APPROACH AND RESULTS: Human umbilical vein ECs (HUVECs) exposed to shear stress were compared with cells grown under static conditions. Signaling through the TGF-ß receptor ALK5 was inhibited with SB525334. Cells were examined for morphological changes and harvested for analysis by real-time polymerase chain reaction, Western blot analysis, apoptosis, proliferation, and immunocytochemistry. Shear stress resulted in ALK5-dependent alignment of HUVECs as well as attenuation of apoptosis and proliferation compared with static controls. Shear stress led to an ALK5-dependent increase in TGF-ß3 and Krüppel-like factor 2, phosphorylation of endothelial NO synthase, and NO release. Addition of the NO donor S-nitroso-N-acetylpenicillamine rescued the cells from apoptosis attributable to ALK5 inhibition under shear stress. Knockdown of TGF-ß3, but not TGF-ß1, disrupted the HUVEC monolayer and prevented the induction of Krüppel-like factor 2 by shear. CONCLUSIONS: Shear stress of HUVECs induces TGF-ß3 signaling and subsequent activation of Krüppel-like factor 2 and NO, and represents a novel role for TGF-ß3 in the maintenance of HUVEC homeostasis in a hemodynamic environment.


Assuntos
Endotélio Vascular/metabolismo , Endotélio Vascular/fisiopatologia , Hemodinâmica/fisiologia , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta3/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Imidazóis/farmacologia , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Óxido Nítrico/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Quinoxalinas/farmacologia , RNA Mensageiro/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais/efeitos dos fármacos , Estresse Mecânico
5.
FASEB J ; 26(2): 567-75, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22067481

RESUMO

Age-related macular degeneration (AMD) is the leading cause of blindness in the Western world. In advanced AMD, new vessels from choriocapillaris (CC) invade through the Bruch's membrane (BrM) into the retina, forming choroidal neovascularization (CNV). BrM, an elastic lamina that is located between the retinal pigment epithelium (RPE) and CC, is thought to act as a physical and functional barrier against CNV. The BrM of patients with early AMD are characterized by decreased levels of antiangiogenic factors, including endostatin, thrombospondin-1 (TSP-1), and pigment epithelium-derived factor (PEDF), as well as by degeneration of the elastic layer. Motivated by a previous report that heat increases elastin expression in human skin, we examined the effect of heat on human ARPE-19 cell production of BrM components. Heat treatment stimulated the production of BrM components, including TSP-1, PEDF, and tropoelastin in vitro and increased the antiangiogenic activity of RPE measured in a mouse corneal pocket assay. The effect of heat on experimental CNV was investigated by pretreating the retina with heat via infrared diode laser prior to the induction of CNV. Heat treatment blocked the development of experimental CNV in vivo. These findings suggest that heat treatment may restore BrM integrity and barrier function against new vessel growth.


Assuntos
Neovascularização de Coroide/prevenção & controle , Temperatura Alta/uso terapêutico , Epitélio Pigmentado da Retina/irrigação sanguínea , Inibidores da Angiogênese/genética , Inibidores da Angiogênese/metabolismo , Animais , Lâmina Basilar da Corioide/irrigação sanguínea , Lâmina Basilar da Corioide/metabolismo , Lâmina Basilar da Corioide/patologia , Linhagem Celular , Neovascularização de Coroide/genética , Neovascularização de Coroide/metabolismo , Neovascularização de Coroide/patologia , Tecido Elástico/metabolismo , Tecido Elástico/patologia , Endostatinas/genética , Endostatinas/metabolismo , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Expressão Gênica , Humanos , Lasers Semicondutores/uso terapêutico , Degeneração Macular/genética , Degeneração Macular/metabolismo , Degeneração Macular/patologia , Degeneração Macular/terapia , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia , Serpinas/genética , Serpinas/metabolismo , Trombospondina 1/genética , Trombospondina 1/metabolismo , Tropoelastina/metabolismo , Degeneração Macular Exsudativa/genética , Degeneração Macular Exsudativa/metabolismo , Degeneração Macular Exsudativa/patologia , Degeneração Macular Exsudativa/prevenção & controle
6.
Nat Commun ; 13(1): 3401, 2022 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-35697682

RESUMO

Age-related macular degeneration (AMD) is one of the most common causes of visual impairment in the elderly, with a complex and still poorly understood etiology. Whole-genome association studies have discovered 34 genomic regions associated with AMD. However, the genes and cognate proteins that mediate the risk, are largely unknown. In the current study, we integrate levels of 4782 human serum proteins with all genetic risk loci for AMD in a large population-based study of the elderly, revealing many proteins and pathways linked to the disease. Serum proteins are also found to reflect AMD severity independent of genetics and predict progression from early to advanced AMD after five years in this population. A two-sample Mendelian randomization study identifies several proteins that are causally related to the disease and are directionally consistent with the observational estimates. In this work, we present a robust and unique framework for elucidating the pathobiology of AMD.


Assuntos
Degeneração Macular , Proteogenômica , Idoso , Loci Gênicos , Estudo de Associação Genômica Ampla , Humanos , Degeneração Macular/genética , Degeneração Macular/metabolismo , Análise da Randomização Mendeliana , Fatores de Risco
7.
FASEB J ; 24(9): 3186-95, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20400538

RESUMO

The small GTPase RhoA and its downstream effectors, ROCK1 and ROCK2, regulate a number of cellular processes, including cell motility, proliferation, survival, and permeability. Pharmacological inhibitors of the Rho pathway reportedly block angiogenesis; however, the molecular details of this inhibition are largely unknown. We demonstrate that vascular endothelial growth factor-A (VEGF) rapidly induces RhoA activation in endothelial cells (ECs). Moreover, the pharmacological inhibition of ROCK1/2 using 10 microM Y-27632 (the IC(50) for this compound in ECs) strongly disrupts vasculogenesis in pluripotent embryonic stem cell cultures, VEGF-mediated regenerative angiogenesis in ex vivo retinal explants, and VEGF-mediated in vitro EC tube formation. Furthermore, using small interfering RNA knockdown and mouse heterozygote knockouts of ROCK1 and ROCK2, we provide data indicating that VEGF-driven angiogenesis is largely mediated through ROCK2. These data demonstrate that Rho/ROCK signaling is an important mediator in a number of angiogenic processes, including EC migration, survival, and cell permeability, and suggest that Rho/ROCK inhibition may prove useful for the treatment of angiogenesis-related disorders.


Assuntos
Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/fisiologia , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/farmacologia , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Amidas/farmacologia , Animais , Apoptose , Western Blotting , Bovinos , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Humanos , Camundongos , Microscopia de Fluorescência , Piridinas/farmacologia , RNA Interferente Pequeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Quinases Associadas a rho/genética
8.
Microvasc Res ; 80(1): 166-73, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20226797

RESUMO

Aberrant transforming growth factor-beta (TGF-beta) signaling plays a clear role in a number of pathologies, in particular, fibrotic diseases and cancer. Accumulating evidence also suggests that TGF-beta is required for vascular homeostasis, shedding light on the role of circulating TGF-beta and the expression of TGF-beta receptors in the adult vasculature, in the absence of any pathologic processes. In human pathologies such as hereditary hemorrhagic telangiectasia (HHT) and preeclampsia, TGF-beta signaling is abnormal. Studies from these disorders have helped elucidate the complex and multiple roles of this ubiquitously-expressed growth factor. The aim of this review is to collate some of the recent evidence that demonstrates a non-redundant role for TGF-beta signaling in maintaining vessel structure and function.


Assuntos
Homeostase/fisiologia , Microvasos/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Animais , Humanos
9.
Circ Res ; 103(12): 1370-82, 2008 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-19059839

RESUMO

The Notch signaling pathway is critical for cell fate determination during embryonic development, including many aspects of vascular development. An emerging paradigm suggests that the Notch gene regulatory network is often recapitulated in the context of phenotypic modulation of vascular smooth muscle cells (VSMC), vascular remodeling, and repair in adult vascular disease following injury. Notch ligand receptor interactions lead to cleavage of receptor, translocation of the intracellular receptor (Notch IC), activation of transcriptional CBF-1/RBP-Jkappa-dependent and -independent pathways, and transduction of downstream Notch target gene expression. Hereditary mutations of Notch components are associated with congenital defects of the cardiovascular system in humans such as Alagille syndrome and cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL). Recent loss- or gain-of-function studies have provided insight into novel Notch-mediated CBF-1/RBP-Jkappa-dependent and -independent signaling and cross-regulation to other molecules that may play a critical role in VSMC phenotypic switching. Notch receptors are critical for controlling VSMC differentiation and dictating the phenotypic response following vascular injury through interaction with a triad of transcription factors that act synergistically to regulate VSMC differentiation. This review focuses on the role of Notch receptor ligand interactions in dictating VSMC behavior and phenotype and presents recent findings on the molecular interactions between the Notch components and VSMC-specific genes to further understand the function of Notch signaling in vascular tissue and disease.


Assuntos
Músculo Liso Vascular/fisiologia , Fenótipo , Receptores Notch/fisiologia , Animais , Humanos , Receptores Notch/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
10.
Arterioscler Thromb Vasc Biol ; 29(8): 1185-92, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19461051

RESUMO

OBJECTIVE: Motivated by the central roles that vascular endothelial growth factor (VEGF) and transforming growth factor (TGF)-beta play in the assembly and maintenance of the vasculature, we examined the impact of systemic VEGF or TGF-beta signal inhibition on endothelial activation as detected by leukocyte-endothelial interactions. METHODS AND RESULTS: VEGF or TGF-beta inhibition, accomplished using adenovirus expression of soluble Flt1 (Ad-sFlt1) or soluble endoglin (Ad-sEng), resulted in a significant increase in the number of leukocytes rolling along the mesenteric venous endothelium and a significant decrease in rolling velocity in Ad-sEng mice. Neutralization of VEGF or TGF-beta resulted in endothelial surface expression of P-selectin and impaired peripheral vasodilatation. Neither inhibition of VEGF nor TGF-beta was associated with platelet or leukocyte activation, as detected by the activation markers platelet P-selectin and the active integrin alphaIIbbetaIII, or by leukocyte expression of L-selectin. Soluble vascular cell adhesion molecule (VCAM)-1 and E-selectin were increased in sEng-expressing mice, indicating higher levels of these adhesion receptors. CONCLUSIONS: VEGF or TGF-beta neutralization leads to impaired endothelium-mediated vasodilatation and elevated expression of surface adhesion molecules, resulting in increased leukocyte adhesion. These results indicate an essential role for both VEGF and TGF-beta in maintaining the endothelium in a nonactivated state and have implications for therapeutic approaches that neutralize VEGF or TGF-beta.


Assuntos
Endotélio Vascular/citologia , Leucócitos/metabolismo , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Vasodilatação/fisiologia , Animais , Adesão Celular , Selectina E/biossíntese , Endotélio Vascular/metabolismo , Contagem de Leucócitos , Leucócitos/citologia , Veias Mesentéricas/citologia , Veias Mesentéricas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Selectina-P/biossíntese , Fator de Crescimento Transformador beta/metabolismo , Molécula 1 de Adesão de Célula Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
11.
Endothelium ; 14(1): 17-24, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17364893

RESUMO

Impaired vasoactive release of opposing vasodilator and vasoconstrictor mediators due to endothelial dysfunction is integral to the pathogenesis of diabetic retinopathy. The aim of this study was to determine the effect of hyperglycemia on the expression of endothelial nitric oxide synthase (eNOS) and the release of nitric oxide (NO) in bovine microvascular retinal endothelial cells (BRECs) under both static (basal and acetylcholine stimulated) and flow (laminar shear stress [10 dynes/cm2 and pulsatile flow 0.3 to 23 dynes/cm2) conditions using a laminar shear apparatus and an in vitro perfused transcapillary culture system. The activity and expression of eNOS, measured by nitrate levels and immunoblot, respectively, were determined following exposure of BRECs to varying concentrations of glucose and mannitol (0 to 25 mM). Under static conditions the expression of eNOS decreased significantly following exposure to increasing concentrations of glucose when compared to osmotic mannitol controls and was accompanied by a significant dose-dependent decrease in nitrate levels in conditioned medium. The acetylcholine stimulated increase in NO release (2.0 +/- 0.3-fold) was significantly reduced by 55% +/- 5% and 65% +/- 4.5% following exposure to 16 and 25 mM glucose, respectively, when compared to osmotic controls. In parallel studies, glucose significantly inhibited both laminar shear stress and pulsatile flow-induced activity when compared to mannitol. We conclude that hyperglycemia impairs agonist- and flow-dependent release of NO in retinal microvascular endothelial cells and may thus contribute to the vascular endothelial dysfunction and impaired autoregulation of diabetic retinopathy.


Assuntos
Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Glucose/farmacologia , Hiperglicemia/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Fluxo Pulsátil/efeitos dos fármacos , Vasos Retinianos/citologia , Acetilcolina/farmacologia , Animais , Bovinos , Células Cultivadas , Técnicas de Cocultura , Meios de Cultivo Condicionados/análise , Meios de Cultivo Condicionados/química , Relação Dose-Resposta a Droga , Células Endoteliais/citologia , Fluorometria , Manose/farmacologia , Nitratos/análise , Pressão Osmótica , Estresse Mecânico , Vasodilatadores/farmacologia
12.
Invest Ophthalmol Vis Sci ; 46(1): 375-82, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15623798

RESUMO

PURPOSE: By the development of a novel retinal microvascular endothelial and pericyte cell coculture system, this study determined the effects of pulsatile flow on the activation of the endothelial cell markers nitric oxide (NO), prostacyclin (PGI2), and endothelin (ET)-1. METHODS: Monocultured bovine retinal endothelial cells (BRECs) and cocultured BRECs with bovine retinal pericytes (BRPs) were exposed to low flow (flow rate, 0.3 mL/min; pulse pressure, 6 mmHg; shear stress, 0.5 dyne/cm2) or high flow (flow rate, 25 mL/min; pulse pressure, 56 mmHg; shear stress, 23 dynes/cm2) for 24 hours, by using a novel perfused transcapillary culture system. The cells were characterized by immunohistochemistry and electron and confocal microscopy. Endothelial nitric oxide synthase (eNOS) and phosphorylated-eNOSSer1179 (pp-eNOS) were determined by Western blot analysis. Nitrate, PGI2, and ET-1 levels were quantified in the medium perfusate by using fluorometric and enzyme-linked immunosorbent assays, respectively. Activation of cyclooxygenase (COX)-2 in BRECs was determined by measuring COX-2 promoter activity with a luciferase reporter assay. RESULTS: The presence of BRPs and BRECs was confirmed by Western blot, immunocytochemistry, and scanning electron microscopy. Phosphorylated eNOS (pp-eNOS) protein levels in BRECs were significantly increased from low to high flow in both mono- and cocultures, concomitant with a significant increase in nitrate levels in the conditioned medium after exposure to pulsatile flow. In parallel cultures, PGI2 levels were also significantly enhanced concomitant with an increase in the transactivation of a COX-2 promoter BREC after exposure to pulsatile flow. ET-1 levels were also increased in both mono- and cocultured cells. conclusions. In this study a novel, functioning, in vitro model of retinal microvascular endothelial and pericyte cells that respond to changes in pulsatile flow was established.


Assuntos
Endotelina-1/metabolismo , Endotélio Vascular/metabolismo , Epoprostenol/metabolismo , Óxido Nítrico Sintase/metabolismo , Pericitos/metabolismo , Fluxo Pulsátil/fisiologia , Vasos Retinianos/fisiologia , Animais , Western Blotting , Bovinos , Células Cultivadas , Técnicas de Cocultura , Ciclo-Oxigenase 2 , Eletroforese em Gel de Poliacrilamida , Endotélio Vascular/citologia , Técnica Indireta de Fluorescência para Anticorpo , Isoenzimas/metabolismo , Microscopia Confocal , Microscopia Eletrônica de Varredura , Óxido Nítrico Sintase Tipo III , Pericitos/citologia , Fosforilação , Prostaglandina-Endoperóxido Sintases/metabolismo , Vasos Retinianos/citologia
13.
Diab Vasc Dis Res ; 11(4): 270-280, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24853909

RESUMO

Hyperglycaemia and hypoxia play essential pathophysiological roles in diabetes. We determined whether hyperglycaemia influences endothelial cell growth under hypoxic conditions in vitro. Using a Ruskinn Invivo2 400 Hypoxia Workstation, bovine aortic endothelial cells (BAEC) were exposed to high glucose concentrations (25 mM glucose) under normoxic or hypoxic conditions before cell growth (balance of proliferation and apoptosis) was assessed by fluorescence-activated cell sorting (FACS) analysis, proliferating cell nuclear antigen (pCNA), Bcl-xL and caspase-3 protein expression and activity. Hypoxia increased hypoxia response element (HRE) transactivation and induced hypoxia-inducible factor-1α (HIF-1α) expression when compared to normoxic controls concomitant with a significant decrease in cell growth. High glucose (25 mM) concentrations attenuated HRE transactivation and HIF-1α protein expression while concurrently reducing hypoxia-induced changes in BAEC growth. Knockdown of HIF-1α expression significantly decreased hypoxia-induced changes in growth and attenuated the modulatory effects of glucose. These results provide evidence that hypoxia-induced control of BAEC growth can be altered by the presence of glucose via inhibition of HIF-1α expression and activation.

14.
Invest Ophthalmol Vis Sci ; 53(12): 7520-7, 2012 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-23081980

RESUMO

PURPOSE: The role of VEGF-A in the normal ciliary body is largely unexplored. The ciliary body is similar in many respects to the choroid plexus of the brain, and we demonstrated previously the importance of VEGF-A in maintenance of choroid plexus vasculature and ependymal cells. Therefore, the role of VEGF-A in ciliary body homeostasis was explored. METHODS: Swiss-Webster mice (VEGF-LacZ) were used to determine VEGF-A expression during ciliary body development and in the adult. VEGFR2 expression was determined in adult wild type C56BL/6J mice. Systemic VEGF-A neutralization in vivo was achieved with adenovirus-mediated overexpression of soluble VEGFR1 (sFlt1). Following VEGF-A neutralization, the ciliary epithelium was analyzed by light microscopy and transmission electron microscopy (TEM). The effect of VEGF-A blockade on ciliary body function also was assessed by measuring intraocular pressure. RESULTS: VEGF-A expression was detected at embryonic day 18.5 (E18.5), the onset of ciliary process formation. In the adult ciliary body, VEGF-A was expressed by the pigmented epithelium, whereas VEGFR2 was localized primarily to the capillary endothelium and nonpigmented epithelium. Systemic VEGF-A neutralization led to a thinning of the nonpigmented epithelium, vacuolization of the pigmented epithelium, loss of capillary fenestrations, and thrombosis. These changes were associated with impaired ciliary body function, as evidenced by decreased intraocular pressure in sFlt1-overexpressing animals (15.31 ± 2.06 mm Hg) relative to controls (18.69 ± 1.49 mm Hg). CONCLUSIONS: VEGF-A has an important role in ciliary body homeostasis. Potential for undesired off-target effects should be considered with the chronic use of anti-VEGF-A therapies.


Assuntos
Corpo Ciliar/metabolismo , Prenhez , Fator A de Crescimento do Endotélio Vascular/biossíntese , Animais , Corpo Ciliar/embriologia , Corpo Ciliar/crescimento & desenvolvimento , Ensaio de Imunoadsorção Enzimática , Epitélio/metabolismo , Epitélio/ultraestrutura , Feminino , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão e Varredura , Gravidez
15.
Cardiovasc Res ; 89(3): 661-70, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21030535

RESUMO

AIMS: Aberrant retinal blood flow is a hallmark of various retinopathies and may be a causative factor in the pathology associated with these conditions. We examined the effects of pulsatile flow on bovine retinal endothelial cell (BREC) and bovine retinal pericyte (BRP) apoptosis and proliferation. METHODS AND RESULTS: Co-cultured BRECs and BRPs were exposed to low (0.3 mL/min) or high (25 mL/min) pulsatile flow for 72 h using a perfused transcapillary culture system. Pulsatile flow increased BREC nitric oxide synthase (eNOS) and cyclooxygenase-2 (COX-2) expression and activity concomitant with a significant decrease in pre-pro-endothelin-1 (ET-1) mRNA and peptide. BREC apoptosis was significantly attenuated following exposure to high flow. The inhibition of NOS, COX, and ET receptors significantly reduced the pro-survival effects of flow on BREC. In contrast, BRP apoptosis was significantly enhanced following exposure to high flow. The inhibition of COX and ET receptors significantly attenuated the high flow-induced increase in BRP apoptosis when compared with untreated controls. Treatment of static BREC with NO donor (S-nitroso-N-acetylpenicillamine, SNAP), ET-1, or iloprost inhibited serum deprivation-induced apoptosis, whereas treatment of BRP with ET-1 and iloprost, but not SNAP, was ineffective. High pulsatile flow decreased BRP proliferation, in the absence of any changes in BREC proliferation. CONCLUSION: Increased pulsatile flow promotes BREC survival and enhances BRP apoptosis through the activation of endothelial-derived vasoactive substances. Altered pulsatile flow does not alter BREC proliferation in co-culture with BRP, whereas BRP proliferation was significantly decreased at high flow rates. These interactions have important implications for vessel growth and regression during retinal vascular pathogenesis.


Assuntos
Apoptose/fisiologia , Células Endoteliais/citologia , Microcirculação/fisiologia , Pericitos/citologia , Fluxo Pulsátil/fisiologia , Vasos Retinianos/citologia , Animais , Bovinos , Divisão Celular/fisiologia , Células Cultivadas , Técnicas de Cocultura , Células Endoteliais/fisiologia , Endotelina-1/metabolismo , Epoprostenol/metabolismo , Óxido Nítrico/metabolismo , Pericitos/fisiologia , Fluxo Sanguíneo Regional/fisiologia , Retina/citologia , Retina/fisiologia , Vasos Retinianos/fisiologia , Estresse Mecânico
16.
Invest Ophthalmol Vis Sci ; 52(13): 9478-87, 2011 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-22058334

RESUMO

PURPOSE: Despite a lack of active angiogenesis, VEGF is expressed in nearly every adult tissue, and recent evidence suggests that VEGF may serve as a survival factor for both vascular and nonvascular tissues. VEGF blockade is a widely used treatment for neovascular diseases such as wet age-related macular degeneration (AMD). Therefore, it was sought in this study to evaluate the expression and role of endogenous VEGF in RPE. METHODS: VEGF and VEGFR2 expression in the murine retina were assessed during development. Bevacizumab was used to neutralize VEGF in ARPE-19 cells, and the effects on cell survival and apical microvill were assessed by TUNEL and SEM, respectively. VEGF was systemically neutralized in vivo by adenoviral-mediated overexpression of soluble VEGFR1 (sFlt). RPE and choriocapillaris were analyzed by transmission electron microscopy (TEM). Changes in gene expression were evaluated by quantitative real-time PCR. RESULTS: VEGF expression was detected in the developing RPE as early as embryonic day (E) 9.5, whereas VEGFR2 expression by RPE began nonuniformly between postnatal (P) day 6.5 and P8.5. VEGF neutralization in vitro led to increased apoptosis and reduced microvilli density and length. Systemic VEGF neutralization led to transient degenerative changes; RPE were vacuolated and separated from photoreceptor outer segments, and choriocapillaris fenestrations were decreased. VEGF levels were elevated in RPE of Ad-sFlt1 mice at day 4 postinfection, and there was increased expression of the neurotrophic factor CD59a at day 14. CONCLUSIONS: These results indicate that VEGF plays a critical role in survival and maintenance of RPE integrity. Potential undesired off-target effects should be considered with chronic use of anti-VEGF agents.


Assuntos
Envelhecimento/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , RNA Mensageiro/genética , Epitélio Pigmentado da Retina/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Animais Recém-Nascidos , Apoptose , Células Cultivadas , Feminino , Imuno-Histoquímica , Camundongos , Microscopia Eletrônica de Transmissão e Varredura , Gravidez , Epitélio Pigmentado da Retina/crescimento & desenvolvimento , Epitélio Pigmentado da Retina/ultraestrutura , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator A de Crescimento do Endotélio Vascular/biossíntese , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/biossíntese , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
17.
Invest Ophthalmol Vis Sci ; 52(7): 4472-83, 2011 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-21498615

RESUMO

PURPOSE: Aberrant retinal blood flow is a hallmark of retinopathies and may be a causative factor in their pathophysiology. In this study, the effects of pulsatile flow on hedgehog and Notch control of retinal endothelial cell and pericyte apoptosis were examined. METHODS: The levels of hedgehog and Notch signaling components in bovine retinal endothelial cells (BRECs) and pericytes (BRPs) were examined in vitro under static conditions and after exposure to pulsatile flow, with a perfused transcapillary co-culture system. Notch and hedgehog signaling was examined by immunocytochemistry, immunoblot, and real-time PCR. RESULTS: Notch and hedgehog proteins were present in BRECs and BRPs in vitro and in human retinal vasculature in vivo. Inhibition of hedgehog with cyclopamine and Notch with DAPT decreased hedgehog target gene levels and Notch intracellular receptor expression, respectively, concomitant with an increase in BREC and BRP apoptosis. Sonic hedgehog (Shh) mediated upregulation of Notch1 receptor levels was attenuated after cyclopamine treatment in both cell types. Exposure of co-cultured BRECs and BRPs to pulsatile flow increased apoptosis in the BRPs while concurrently decreasing apoptosis in the BRECs. These changes were concomitant with increased expression of Notch and hedgehog signaling components in the BRECs and reduced expression in the BRPs. The flow-induced decrease in apoptosis in the BRECs was associated with increased Notch receptor expression and was reversed after inhibition of hedgehog signaling with cyclopamine and inhibition of Notch signaling after ectopic expression of the CBF-1/RBP-Jκ-binding protein, RPMS-1. CONCLUSIONS: Pulsatile flow promotes BREC survival and enhances BRP apoptosis through modulation of Notch and hedgehog pathways. These interactions have important implications for the pathogenesis of retinopathies.


Assuntos
Apoptose/fisiologia , DNA/genética , Endotélio Vascular/citologia , Proteínas Hedgehog/genética , Pericitos/citologia , Receptores Notch/genética , Retina/citologia , Animais , Western Blotting , Bovinos , Células Cultivadas , Células Endoteliais/citologia , Endotélio Vascular/metabolismo , Proteínas Hedgehog/biossíntese , Humanos , Imuno-Histoquímica , Pericitos/metabolismo , Reação em Cadeia da Polimerase , Receptores Notch/biossíntese , Retina/metabolismo , Transdução de Sinais
18.
PLoS One ; 4(4): e5149, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19340291

RESUMO

Pericyte-endothelial cell (EC) interactions are critical to both vascular development and vessel stability. We have previously shown that TGF-beta signaling between EC and mural cells participates in vessel stabilization in vitro. We therefore investigated the role of TGF-beta signaling in maintaining microvessel structure and function in the adult mouse retinal microvasculature. TGF-beta signaling was inhibited by systemic expression of soluble endoglin (sEng) and inhibition was demonstrated by reduced phospho-smad2 in the adult retina. Blockade of TGF-beta signaling led to increased vascular and neural cell apoptosis in the retina, which was associated with decreased retinal function, as measured by electroretinogram (ERG). Perfusion of the inner retinal vasculature was impaired and was accompanied by defective autoregulation and loss of capillary integrity. Fundus angiography and Evans blue permeability assay revealed a breakdown of the blood-retinal-barrier that was characterized by decreased association between the tight junction proteins zo-1 and occludin. Inhibition of TGF-beta signaling in cocultures of EC and 10T1/2 cells corroborated the in vivo findings, with impaired EC barrier function, dissociation of EC from 10T1/2 cells, and endothelial cell death, supporting the role of EC-mesenchymal interactions in TGF-beta signaling. These results implicate constitutive TGF-beta signaling in maintaining the integrity and function of the adult microvasculature and shed light on the potential role of TGF-beta signaling in vasoproliferative and vascular degenerative retinal diseases.


Assuntos
Endotélio Vascular/fisiologia , Homeostase , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Animais , DNA Complementar , Endotélio Vascular/ultraestrutura , Marcação In Situ das Extremidades Cortadas , Camundongos , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
19.
Annu Rev Pathol ; 3: 615-43, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18039132

RESUMO

Although the terms ischemia and hypoxia are often used interchangeably, they represent distinct processes that result in different modulatory effects at the cellular level. Hypoxia is a reduction in oxygen delivery below tissue demand, whereas ischemia is a lack of perfusion, characterized not only by hypoxia but also by insufficient nutrient supply. Hypoxia can be either acute or chronic, and both are centrally regulated by hypoxia-inducible factor, a transcription factor that governs the expression of key response genes such as vascular endothelial growth factor and erythropoietin. Whereas severe chronic hypoxia can cause cell death, less-severe hypoxia can protect against subsequent damage, a phenomenon known as hypoxic conditioning. Several important processes are characterized by hypoxia, including ischemia-reperfusion, tumor growth and progression, inflammation, myocardial ischemia, and a number of ocular pathologies.


Assuntos
Hipóxia/patologia , Traumatismo por Reperfusão/patologia , Reperfusão , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Eritropoese/fisiologia , Regulação da Expressão Gênica , Humanos , Hipóxia/metabolismo , Fator 1 Induzível por Hipóxia/genética , Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Neovascularização Fisiológica/fisiologia , Recuperação de Função Fisiológica/fisiologia , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/metabolismo
20.
J Exp Med ; 205(2): 491-501, 2008 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-18268040

RESUMO

Although the role of vascular endothelial growth factor (VEGF) in developmental and pathological angiogenesis is well established, its function in the adult is less clear. Similarly, although transforming growth factor (TGF) beta is involved in angiogenesis, presumably by mediating capillary (endothelial cell [EC]) stability, its involvement in quiescent vasculature is virtually uninvestigated. Given the neurological findings in patients treated with VEGF-neutralizing therapy (bevacizumab) and in patients with severe preeclampsia, which is mediated by soluble VEGF receptor 1/soluble Fms-like tyrosine kinase receptor 1 and soluble endoglin, a TGF-beta signaling inhibitor, we investigated the roles of VEGF and TGF-beta in choroid plexus (CP) integrity and function in adult mice. Receptors for VEGF and TGF-beta were detected in adult CP, as well as on ependymal cells. Inhibition of VEGF led to decreased CP vascular perfusion, which was associated with fibrin deposition. Simultaneous blockade of VEGF and TGF-beta resulted in the loss of fenestrae on CP vasculature and thickening of the otherwise attenuated capillary endothelium, as well as the disappearance of ependymal cell microvilli and the development of periventricular edema. These results provide compelling evidence that both VEGF and TGF-beta are involved in the regulation of EC stability, ependymal cell function, and periventricular permeability.


Assuntos
Plexo Corióideo/metabolismo , Epêndima/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adenoviridae/genética , Animais , Permeabilidade Capilar , Plexo Corióideo/ultraestrutura , Células Endoteliais/metabolismo , Epêndima/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Microscopia Eletrônica de Transmissão , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Fator de Crescimento Transformador beta1/genética , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA