Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Circ Res ; 131(2): 168-183, 2022 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-35642564

RESUMO

BACKGROUND: Insulin resistance (IR) can increase atherosclerotic and cardiovascular risk by inducing endothelial dysfunction, decreasing nitric oxide (NO) production, and accelerating arterial inflammation. The aim is to determine the mechanism by which insulin action and NO production in endothelial cells can improve systemic bioenergetics and decrease atherosclerosis via differentiation of perivascular progenitor cells (PPCs) into brown adipocytes (BAT). METHODS: Studies used various endothelial transgenic and deletion mutant ApoE-/- mice of insulin receptors, eNOS (endothelial NO synthase) and ETBR (endothelin receptor type B) receptors for assessments of atherosclerosis. Cells were isolated from perivascular fat and micro-vessels for studies on differentiation and signaling mechanisms in responses to NO, insulin, and lipokines from BAT. RESULTS: Enhancing insulin's actions on endothelial cells and NO production in ECIRS1 transgenic mice reduced body weight and increased systemic energy expenditure and BAT mass and activity by inducing differentiation of PPCs into beige/BAT even with high-fat diet. However, positive changes in bioenergetics, BAT differentiation from PPCs and weight loss were inhibited by N(gamma)-nitro-L-arginine methyl ester (L-NAME), an inhibitor of eNOS, in ECIRS1 mice and eNOSKO mice. The mechanism mediating NO's action on PPC differentiation into BAT was identified as the activation of solubilized guanylate cyclase/PKGIα (cGMP protein-dependent kinase Iα)/GSK3ß (glycogen synthase kinase 3ß) pathways. Plasma lipidomics from ECIRS1 mice with NO-induced increased BAT mass revealed elevated 12,13-diHOME production. Infusion of 12,13-diHOME improved endothelial dysfunction and decreased atherosclerosis, whereas its reduction had opposite effects in ApoE-/-mice. CONCLUSIONS: Activation of eNOS and endothelial cells by insulin enhanced the differentiation of PPC to BAT and its lipokines and improved systemic bioenergetics and atherosclerosis, suggesting that endothelial dysfunction is a major contributor of energy disequilibrium in obesity.


Assuntos
Tecido Adiposo Marrom , Aterosclerose , Tecido Adiposo Marrom/metabolismo , Animais , Apolipoproteínas E/metabolismo , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/prevenção & controle , Células Endoteliais/metabolismo , Insulina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico/metabolismo
2.
Blood ; 129(16): 2280-2290, 2017 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-28130211

RESUMO

Thrombolytic therapy using tissue plasminogen activator (tPA) in acute stroke is associated with increased risks of cerebral hemorrhagic transformation and angioedema. Although plasma kallikrein (PKal) has been implicated in contributing to both hematoma expansion and thrombosis in stroke, its role in the complications associated with the therapeutic use of tPA in stroke is not yet available. We investigated the effects of tPA on plasma prekallikrein (PPK) activation and the role of PKal on cerebral outcomes in a murine thrombotic stroke model treated with tPA. We show that tPA increases PKal activity in vitro in both murine and human plasma, via a factor XII (FXII)-dependent mechanism. Intravenous administration of tPA increased circulating PKal activity in mice. In mice with thrombotic occlusion of the middle cerebral artery, tPA administration increased brain hemorrhage transformation, infarct volume, and edema. These adverse effects of tPA were ameliorated in PPK (Klkb1)-deficient and FXII-deficient mice and in wild-type (WT) mice pretreated with a PKal inhibitor prior to tPA. tPA-induced brain hemisphere reperfusion after photothrombolic middle cerebral artery occlusion was increased in Klkb1-/- mice compared with WT mice. In addition, PKal inhibition reduced matrix metalloproteinase-9 activity in brain following stroke and tPA therapy. These data demonstrate that tPA activates PPK in plasma and PKal inhibition reduces cerebral complications associated with tPA-mediated thrombolysis in stroke.


Assuntos
Angioedema/induzido quimicamente , Hemorragia Cerebral/induzido quimicamente , Fibrinolíticos/efeitos adversos , Calicreína Plasmática/metabolismo , Acidente Vascular Cerebral/tratamento farmacológico , Trombose/tratamento farmacológico , Ativador de Plasminogênio Tecidual/efeitos adversos , Administração Intravenosa , Angioedema/sangue , Angioedema/genética , Animais , Hemorragia Cerebral/sangue , Hemorragia Cerebral/genética , Modelos Animais de Doenças , Fator XII/genética , Fator XII/metabolismo , Expressão Gênica , Humanos , Infarto da Artéria Cerebral Média/sangue , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/genética , Infarto da Artéria Cerebral Média/patologia , Masculino , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Knockout , Calicreína Plasmática/genética , Acidente Vascular Cerebral/sangue , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/patologia , Terapia Trombolítica , Trombose/sangue , Trombose/genética , Trombose/patologia
3.
Exp Eye Res ; 186: 107744, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31351056

RESUMO

The plasma kallikrein-stimulated generation of bradykinin (BK) has been implicated in diabetic macular edema (DME). This study characterizes the effects of BK on the ultrastructure and proteome of the rat retina. The effects of intravitreal injection of BK on retinal thickness and vascular ultrastructure in Sprague Dawley rats were analyzed and compared with the effects of VEGF using spectral-domain optical coherence tomography. At 24 h post intravitreal injection of BK or saline vehicle retina were harvested and solubilized proteins were analyzed by mass spectrometry-based proteomics. Proteins were identified using X!Tandem and spectral counts were used as a semiquantitative measurement of protein abundance. Proteins identified from retinal extracts were annotated by Gene Ontology (GO) slim terms and compared with a human DME vitreous proteome. Intravitreal injection of BK and VEGF induced transient increases in retinal thickness of 46 µm (24.6%, p = 0.015) and 39 µm (20.3%, p = 0.004), respectively at 24 h, which were resolved to baseline thicknesses at 96 h post injection. BK and VEGF also increased retinal vessel diameters and tortuosity at 24 h post intravitreal injection. Proteomic analyses identified 1757 non-redundant proteins in the rat retina, including 1739 and 1725 proteins from BK- and saline control-injected eyes, respectively. Eighteen proteins, including two proteins associated with intercellular junctions, filamin A and actinin alpha 4, were decreased by at least 50% (p < 0.05) in retina from BK-injected eyes compare with retina from eyes injected with saline. In addition, 32 proteins were increased by > 2-fold (p < 0.05) in retina from BK-injected eyes. Eight proteins, including complement C3, were identified to be increased in both BK-stimulated rat retina and in human DME vitreous. Western blot analysis showed that Complement 3 levels in vitreous from BK-injected eyes in rats and clinical DME samples were increased by 6.6-fold (p = 0.039) and 4.3-fold (p = 0.02), compared with their respective controls. In summary, this study identifies protein changes in rat retina that are associated with BK-induced retinal thickening, including 8 proteins that were previously reported to be increased in the human DME vitreous proteome.


Assuntos
Bradicinina/farmacologia , Edema Macular/metabolismo , Proteoma/metabolismo , Retina/metabolismo , Vasodilatadores/farmacologia , Animais , Western Blotting , Injeções Intravítreas , Edema Macular/induzido quimicamente , Masculino , Calicreína Plasmática , Proteômica , Ratos , Ratos Sprague-Dawley , Retina/diagnóstico por imagem , Vasos Retinianos/metabolismo , Tomografia de Coerência Óptica , Fator A de Crescimento do Endotélio Vascular/farmacologia
4.
Nat Med ; 13(2): 181-8, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17259996

RESUMO

Excessive retinal vascular permeability contributes to the pathogenesis of proliferative diabetic retinopathy and diabetic macular edema, leading causes of vision loss in working-age adults. Using mass spectroscopy-based proteomics, we detected 117 proteins in human vitreous and elevated levels of extracellular carbonic anhydrase-I (CA-I) in vitreous from individuals with diabetic retinopathy, suggesting that retinal hemorrhage and erythrocyte lysis contribute to the diabetic vitreous proteome. Intravitreous injection of CA-I in rats increased retinal vessel leakage and caused intraretinal edema. CA-I-induced alkalinization of vitreous increased kallikrein activity and its generation of factor XIIa, revealing a new pathway for contact system activation. CA-I-induced retinal edema was decreased by complement 1 inhibitor, neutralizing antibody to prekallikrein and bradykinin receptor antagonism. Subdural infusion of CA-I in rats induced cerebral vascular permeability, suggesting that extracellular CA-I could have broad relevance to neurovascular edema. Inhibition of extracellular CA-I and kallikrein-mediated innate inflammation could provide new therapeutic opportunities for the treatment of hemorrhage-induced retinal and cerebral edema.


Assuntos
Permeabilidade Capilar/efeitos dos fármacos , Inibidores da Anidrase Carbônica/uso terapêutico , Anidrases Carbônicas/metabolismo , Retinopatia Diabética/tratamento farmacológico , Proteínas do Olho/metabolismo , Sistema Calicreína-Cinina/fisiologia , Corpo Vítreo/enzimologia , Acetazolamida/farmacologia , Animais , Western Blotting , Antagonistas dos Receptores da Bradicinina , Inibidores da Anidrase Carbônica/farmacologia , Anidrases Carbônicas/toxicidade , Complemento C1/antagonistas & inibidores , Fator XIIa/metabolismo , Humanos , Espectrometria de Massas , Papiledema/induzido quimicamente , Proteômica , Ratos , Ratos Sprague-Dawley , Estatísticas não Paramétricas
5.
Arterioscler Thromb Vasc Biol ; 32(5): 1124-31, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22426129

RESUMO

OBJECTIVE: To determine the contribution of hyperinsulinemia to atherosclerosis development. METHODS AND RESULTS: Apolipoprotein E (Apoe) null mice that had knockout of a single allele of the insulin receptor (Insr) gene were compared with littermate Apoe null mice with intact insulin receptors. Plasma insulin levels in Insr haploinsufficient/Apoe null mice were 50% higher in the fasting state and up to 69% higher during a glucose tolerance test, but glucose tolerance was not different in the 2 groups. C-peptide levels, insulin sensitivity, and postreceptor insulin signaling in muscle, liver, fat, and aorta were not different between groups, whereas disappearance in plasma of an injected insulin analog was delayed in Insr haploinsufficient/Apoe null mice, indicating that impaired insulin clearance was the primary cause of hyperinsulinemia. No differences were observed in plasma lipids or blood pressure. Despite the hyperinsulinemia, atherosclerotic lesion size was not different between the 2 groups at time points up to 52 weeks of age when measured as en face lesion area in the aorta, cross-sectional plaque area in the aortic sinus, and cholesterol abundance in the brachiocephalic artery. CONCLUSIONS: Hyperinsulinemia, without substantial vascular or whole-body insulin resistance and without changes in plasma lipids or blood pressure, does not change susceptibility to atherosclerosis.


Assuntos
Apolipoproteínas E/genética , Aterosclerose/genética , Hiperinsulinismo/complicações , Resistência à Insulina , Animais , Apolipoproteínas E/sangue , Aterosclerose/sangue , Aterosclerose/etiologia , Progressão da Doença , Feminino , Regulação da Expressão Gênica , Hiperinsulinismo/sangue , Hiperinsulinismo/genética , Proteínas Substratos do Receptor de Insulina/biossíntese , Proteínas Substratos do Receptor de Insulina/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais
6.
Front Pharmacol ; 14: 1287487, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38178859

RESUMO

Background: The kallikrein kinin system (KKS) is an established pharmacological target for the treatment and prevention of attacks in hereditary angioedema (HAE). Proteolytic activities of FXIIa and single-chain Factor XII (FXII) zymogen contribute to KKS activation and thereby may play roles in both initiating and propagating HAE attacks. In this report, we investigated the effects of potent small molecule FXIIa inhibitors on FXIIa and single chain FXII enzymatic activities, KKS activation, and angioedema in mice. Methods: We examined the effects of 29 structurally distinct FXIIa inhibitors on enzymatic activities of FXIIa and a mutant single chain FXII with R334A, R343A and R353A substitutions (rFXII-T), that does not undergo zymogen conversion to FXIIa, using kinetic fluorogenic substrate assays. We examined the effects of a representative FXIIa inhibitor, KV998086, on KKS activation and both carrageenan- and captopril-induced angioedema in mice. Results: FXIIa inhibitors designed to target its catalytic domain also potently inhibited the enzymatic activity of rFXII-T and the pIC50s of these compounds linearly correlated for rFXIIa and rFXII-T (R 2 = 0.93). KV998086, a potent oral FXIIa inhibitor (IC50 = 7.2 nM) inhibited dextran sulfate (DXS)-stimulated generation of plasma kallikrein and FXIIa, and the cleavage of high molecular weight kininogen (HK) in human plasma. KV998086 also inhibited rFXII-T mediated HK cleavage (p < 0.005) in plasma from FXII knockout mice supplemented with rFXII-T and stimulated with polyphosphate or DXS. Orally administered KV998086 protected mice from 1) captopril-induced Evans blue leakage in colon and laryngotracheal tissues and 2) blocked carrageenan-induced plasma HK consumption and paw edema. Conclusion: These findings show that small molecule FXIIa inhibitors, designed to target its active site, also inhibit the enzymatic activity of FXII zymogen. Combined inhibition of FXII zymogen and FXIIa may thereby suppress both the initiation and amplification of KKS activation that contribute to hereditary angioedema attacks and other FXII-mediated diseases.

7.
Transl Stroke Res ; 13(2): 287-299, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34241810

RESUMO

Plasma kallikrein (PKa) has been implicated in contributing to hemorrhage following thrombolytic therapy; however, its role in spontaneous intracerebral hemorrhage is currently not available. This report investigates the role of PKa on hemorrhage and hypertension in stroke-prone spontaneously hypertensive rats (SHRSP). SHRSP were fed with a high salt-containing stroke-prone diet to increase blood pressure and induce intracerebral hemorrhage. The roles of PKa on blood pressure, hemorrhage, and survival in SHRSP were examined in rats receiving a PKa inhibitor or plasma prekallikrein antisense oligonucleotide (PK ASO) compared with rats receiving control ASO. Effects on PKa on the proteolytic cleavage of atrial natriuretic peptide (ANP) were analyzed by tandem mass spectrometry. We show that SHRSP on high-salt diet displayed increased levels of PKa activity compared with control rats. Cleaved kininogen was increased in plasma during stroke compared to SHRSP without stroke. Systemic administration of a PKa inhibitor or PK ASO to SHRSP reduced hemorrhage and blood pressure, and improved neurological function and survival compared with SHRSP receiving control ASO. Since PKa inhibition was associated with reduced blood pressure in hypertensive rats, we investigated the effects of PKa on the cleavage of ANP. Incubation of PKa with ANP resulted in the generation fragment ANP5-28, which displayed reduced effects on blood pressure lowering compared with full length ANP. PKa contributes to increased blood pressure in SHRSP, which is associated with hemorrhage and reduced survival. PKa-mediated cleavage of ANP reduces its blood pressure lowering effects and thereby may contribute to hypertension-induced intracerebral hemorrhage.


Assuntos
Hipertensão , Acidente Vascular Cerebral , Animais , Fator Natriurético Atrial , Pressão Sanguínea/fisiologia , Hemorragia Cerebral/complicações , Hipertensão/complicações , Calicreína Plasmática , Ratos , Ratos Endogâmicos SHR , Acidente Vascular Cerebral/complicações
8.
Invest Ophthalmol Vis Sci ; 61(10): 53, 2020 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-32866267

RESUMO

Purpose: The purpose of this study was to evaluate differences in optical coherence tomography angiography (OCTA) metrics in the superficial (SCP), intermediate (ICP), and deep (DCP) vascular plexuses across diabetic retinopathy (DR) severity levels. Methods: This was a cross sectional observational retrospective chart review study. Eligible patients with diabetes who underwent same day RTVue XR Avanti OCTA, spectral-domain optical coherence tomography (SD-OCT), and 200-degree Optos ultrawide field color imaging. SCP, ICP, and DCP vessel density (VD) and vessel length density (VLD) were assessed using 3-D projection artifact removal software (PAROCTA) software. Results: Of 396 eyes (237 patients), 16.1% had no DR, 26.9% mild nonproliferative DR (NPDR), 21.1% moderate NPDR, 12.1% severe NPDR, 10.1% proliferative DR (PDR) without panretinal photocoagulation (PRP), and 13.4% PDR with PRP. When comparing mild NPDR to no DR eyes, ICP and DCP VD and VLD were significantly lower, but there was no difference for SCP metrics. In eyes with more severe DR, there were significant differences in SCP VD and VLD between DR severity levels (mild versus moderate NPDR: VD 35.45 ± 3.31 vs. 34.14 ± 3.38, P = 0.008 and VLD 17.59 ± 1.83 vs. 16.80 ± 1.83, P = 0.003; moderate versus severe NPDR: VLD 16.80 ± 1.83 vs. 15.79 ± 1.84, P = 0.019), but no significant differences in ICP or DCP. Conclusions: Although VD of each of the three individual layers decreases with increasing DR severity, DR severity has a substantially different effect on OCTA parameters within each layer. Vascular changes in eyes with no to early DR were present primarily in the deeper vascular layers, whereas in eyes with advanced DR the opposite was observed. This study highlights the effects of ICP and the importance of assessing SCP and DCP changes independently across each DR severity level.


Assuntos
Retinopatia Diabética/patologia , Vasos Retinianos/patologia , Estudos Transversais , Retinopatia Diabética/diagnóstico por imagem , Feminino , Fundo de Olho , Humanos , Masculino , Pessoa de Meia-Idade , Vasos Retinianos/diagnóstico por imagem , Estudos Retrospectivos , Índice de Gravidade de Doença , Tomografia de Coerência Óptica
9.
J Hazard Mater ; 386: 121659, 2020 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-31776080

RESUMO

Although in-vivo exposure of PM2.5 has been suggested to initiate a disorder on vascular permeability, the effects and related mechanism has not been well defined. In this work, an obvious increase on vascular permeability has been confirmed in vivo by vein injection of PM2.5 into Balb/c mouse. Human umbilical vein vascular endothelial cells and the consisted ex-vivo vascular endothelium were used as model to investigate the effects of PM2.5 on the vascular permeability and the underlying molecular mechanism. Upon PM2.5 exposure, the vascular endothelial growth factor receptor 2 on cell membrane phosphorylates and activates the downstream mitogen-activated protein kinase (MAPK)/ERK signaling. The adherens junction protein VE-cadherin sheds and the intercellular junction opens, damaging the integrity of vascular endothelium via paracellular pathway. Besides, PM2.5 induces the intracellular reactive oxygen species (ROS) production and triggers the oxidative stress including activity decrease of superoxide dismutase, lactate dehydrogenase release and permeability increase of cell membrane. Taken together, the paracellular and transcellular permeability enhancement jointly contributes to the significant increase of endothelium permeability and thus vascular permeability upon PM2.5 exposure. This work provides an insight into molecular mechanism of PM2.5 associated cardiovascular disease and offered a real-time screening method for the health risk of PM2.5.


Assuntos
Permeabilidade Capilar/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Material Particulado/toxicidade , Espécies Reativas de Oxigênio/metabolismo , Acetilcisteína/farmacologia , Junções Aderentes/efeitos dos fármacos , Animais , Antígenos CD/metabolismo , Butadienos/farmacologia , Caderinas/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Junções Intercelulares/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Nitrilas/farmacologia , Estresse Oxidativo/efeitos dos fármacos
10.
Cell Metab ; 32(2): 215-228.e7, 2020 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-32663458

RESUMO

Rapid alterations in cellular metabolism allow tissues to maintain homeostasis during changes in energy availability. The central metabolic regulator acetyl-CoA carboxylase 2 (ACC2) is robustly phosphorylated during cellular energy stress by AMP-activated protein kinase (AMPK) to relieve its suppression of fat oxidation. While ACC2 can also be hydroxylated by prolyl hydroxylase 3 (PHD3), the physiological consequence thereof is poorly understood. We find that ACC2 phosphorylation and hydroxylation occur in an inverse fashion. ACC2 hydroxylation occurs in conditions of high energy and represses fatty acid oxidation. PHD3-null mice demonstrate loss of ACC2 hydroxylation in heart and skeletal muscle and display elevated fatty acid oxidation. Whole body or skeletal muscle-specific PHD3 loss enhances exercise capacity during an endurance exercise challenge. In sum, these data identify an unexpected link between AMPK and PHD3, and a role for PHD3 in acute exercise endurance capacity and skeletal muscle metabolism.


Assuntos
Gorduras/metabolismo , Prolina Dioxigenases do Fator Induzível por Hipóxia/metabolismo , Músculo Esquelético/metabolismo , Acetil-CoA Carboxilase/metabolismo , Animais , Linhagem Celular , Tolerância ao Exercício , Feminino , Humanos , Prolina Dioxigenases do Fator Induzível por Hipóxia/deficiência , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Oxirredução
11.
J Proteome Res ; 8(12): 5541-9, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19845401

RESUMO

Diabetic retinopathy is the most common microvascular complication caused by diabetes mellitus and is a leading cause of vision loss among working-age adults in developed countries. Understanding the effects of diabetes on the retinal proteome may provide insights into factors and mechanisms responsible for this disease. We have performed a comprehensive proteomic analysis and comparison of retina from C57BL/6 mice with 2 months of streptozotocin-induced diabetes and age-matched nondiabetic control mice. To explore the role of the angiotensin AT1 receptor in the retinal proteome in diabetes, a subgroup of mice were treated with the AT1 antagonist candesartan. We identified 1792 proteins from retinal lysates, of which 65 proteins were differentially changed more than 2-fold in diabetic mice compared with nondiabetic mice. A majority (72%) of these protein changes were normalized by candesartan treatment. Most of the significantly changed proteins were associated with metabolism, oxidative phosphorylation, and apoptotic pathways. An analysis of the proteomics data revealed metabolic and apoptotic abnormalities in the retina from diabetic mice that were ameliorated with candesartan treatment. These results provide insight into the effects of diabetes on the retina and the role of the AT1 receptor in modulating this response.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Retinopatia Diabética/metabolismo , Proteínas do Olho/análise , Proteoma/efeitos dos fármacos , Retina/química , Bloqueadores do Receptor Tipo 1 de Angiotensina II/uso terapêutico , Animais , Apoptose , Benzimidazóis/farmacologia , Benzimidazóis/uso terapêutico , Compostos de Bifenilo , Diabetes Mellitus/metabolismo , Retinopatia Diabética/tratamento farmacológico , Proteínas do Olho/metabolismo , Metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação Oxidativa , Proteômica , Estreptozocina , Tetrazóis/farmacologia , Tetrazóis/uso terapêutico
12.
Sci Transl Med ; 11(499)2019 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-31270273

RESUMO

The Joslin Medalist Study characterized people affected with type 1 diabetes for 50 years or longer. More than 35% of these individuals exhibit no to mild diabetic retinopathy (DR), independent of glycemic control, suggesting the presence of endogenous protective factors against DR in a subpopulation of patients. Proteomic analysis of retina and vitreous identified retinol binding protein 3 (RBP3), a retinol transport protein secreted mainly by the photoreceptors, as elevated in Medalist patients protected from advanced DR. Mass spectrometry and protein expression analysis identified an inverse association between vitreous RBP3 concentration and DR severity. Intravitreal injection and photoreceptor-specific overexpression of RBP3 in rodents inhibited the detrimental effects of vascular endothelial growth factor (VEGF). Mechanistically, our results showed that recombinant RBP3 exerted the therapeutic effects by binding and inhibiting VEGF receptor tyrosine phosphorylation. In addition, by binding to glucose transporter 1 (GLUT1) and decreasing glucose uptake, RBP3 blocked the detrimental effects of hyperglycemia in inducing inflammatory cytokines in retinal endothelial and Müller cells. Elevated expression of photoreceptor-secreted RBP3 may have a role in protection against the progression of DR due to hyperglycemia by inhibiting glucose uptake via GLUT1 and decreasing the expression of inflammatory cytokines and VEGF.


Assuntos
Diabetes Mellitus/metabolismo , Diabetes Mellitus/patologia , Retinopatia Diabética/metabolismo , Retinopatia Diabética/patologia , Proteínas do Olho/metabolismo , Retina/metabolismo , Retina/patologia , Proteínas de Ligação ao Retinol/metabolismo , 3-O-Metilglucose/metabolismo , Ácidos/metabolismo , Animais , Movimento Celular/efeitos dos fármacos , Desoxiglucose/metabolismo , Diabetes Mellitus/fisiopatologia , Retinopatia Diabética/fisiopatologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Células Ependimogliais/efeitos dos fármacos , Células Ependimogliais/metabolismo , Proteínas do Olho/administração & dosagem , Proteínas do Olho/sangue , Proteínas do Olho/química , Glicólise/efeitos dos fármacos , Humanos , Injeções Intravítreas , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patologia , Substâncias Protetoras/farmacologia , Domínios Proteicos , Ratos Endogâmicos Lew , Proteínas Recombinantes/farmacologia , Reprodutibilidade dos Testes , Retina/fisiopatologia , Proteínas de Ligação ao Retinol/administração & dosagem , Proteínas de Ligação ao Retinol/química , Transdução de Sinais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Corpo Vítreo/efeitos dos fármacos , Corpo Vítreo/metabolismo
13.
Diabetes ; 55(11): 3112-20, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17065350

RESUMO

Diabetes induces the activation of several protein kinase C (PKC) isoforms in the renal glomeruli. We used PKC-beta(-/-) mice to examine the action of PKC-beta isoforms in diabetes-induced oxidative stress and renal injury at 8 and 24 weeks of disease. Diabetes increased PKC activity in renal cortex of wild-type mice and was significantly reduced (<50% of wild-type) in diabetic PKC-beta(-/-) mice. In wild-type mice, diabetes increased the translocation of PKC-alpha and -beta1 to the membrane, whereas only PKC-alpha was elevated in PKC-beta(-/-) mice. Increases in urinary isoprostane and 8-hydroxydeoxyguanosine, parameters of oxidative stress, in diabetic PKC-beta(-/-) mice were significantly reduced compared with diabetic wild-type mice. Diabetes increased NADPH oxidase activity and the expressions of p47(phox), Nox2, and Nox4 mRNA levels in the renal cortex and were unchanged in diabetic PKC-beta(-/-) mice. Increased expression of endothelin-1 (ET-1), vascular endothelial growth factor (VEGF), transforming growth factor (TGF)-beta, connective tissue growth factor (CTGF), and collagens IV and VI found in diabetic wild-type mice was attenuated in diabetic PKC-beta(-/-) mice. Diabetic PKC-beta(-/-) mice were protected from renal hypertrophy, glomerular enlargement, and hyperfiltration observed in diabetic wild-type mice and had less proteinuria. Lack of PKC-beta can protect against diabetes-induced renal dysfunction, fibrosis, and increased expressions of Nox2 and -4, ET-1, VEGF, TGF-beta, CTGF, and oxidant production.


Assuntos
Citocinas/genética , Diabetes Mellitus Experimental/fisiopatologia , Estresse Oxidativo/fisiologia , Proteína Quinase C/genética , Animais , Glicemia/metabolismo , Pressão Sanguínea , Peso Corporal , Diabetes Mellitus Experimental/genética , Nefropatias Diabéticas/prevenção & controle , Ativação Enzimática , Fibrose/prevenção & controle , Regulação da Expressão Gênica , Taxa de Filtração Glomerular , Córtex Renal/enzimologia , Córtex Renal/fisiologia , Córtex Renal/fisiopatologia , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase , Proteína Quinase C/deficiência , Proteína Quinase C/metabolismo , Proteína Quinase C beta , Valores de Referência
14.
Diabetes ; 55(3): 691-8, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16505232

RESUMO

Activation of protein kinase C (PKC) in vascular tissue is associated with endothelial dysfunction and insulin resistance. However, the effect of vascular PKC activation on insulin-stimulated endothelial nitric oxide (NO) synthase (eNOS) regulation has not been characterized in obesity-associated insulin resistance. Diacylglycerol (DAG) concentration and PKC activity were increased in the aorta of Zucker fatty compared with Zucker lean rats. Insulin-stimulated increases in Akt phosphorylation and cGMP concentration (a measure of NO bioavailability) after euglycemic-hyperinsulinemic clamp were blunted in the aorta of fatty compared with lean rats but were partly normalized after 2 weeks of treatment with the PKCbeta inhibitor ruboxistaurin (LY333531). In endothelial cell culture, overexpression of PKCbeta1 and -beta2, but not PKCalpha, -delta, or -zeta, decreased insulin-stimulated Akt phosphorylation and eNOS expression. Overexpression of PKCbeta1 and -beta2, but not PKCalpha or -delta, also decreased Akt phosphorylation stimulated by vascular endothelial growth factor (VEGF). In microvessels isolated from transgenic mice overexpressing PKCbeta2 only in vascular cells, Akt phosphorylation stimulated by insulin was decreased compared with wild-type mice. Thus, activation of PKCbeta in endothelial cells and vascular tissue inhibits Akt activation by insulin and VEGF, inhibits Akt-dependent eNOS regulation by insulin, and causes endothelial dysfunction in obesity-associated insulin resistance.


Assuntos
Vasos Sanguíneos/enzimologia , Resistência à Insulina , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Obesidade/metabolismo , Proteína Quinase C/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Animais , Aterosclerose/etiologia , Bovinos , Células Cultivadas , GMP Cíclico/biossíntese , Diglicerídeos/sangue , Ativação Enzimática , Masculino , Camundongos , Camundongos Transgênicos , Óxido Nítrico Sintase Tipo III/fisiologia , Obesidade/complicações , Fosforilação , Proteína Quinase C beta , Ratos , Ratos Zucker , Fator A de Crescimento do Endotélio Vascular/farmacologia
15.
Invest Ophthalmol Vis Sci ; 48(12): 5750-5, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18055828

RESUMO

PURPOSE: Originally identified as a lipopolysaccharide binding protein with Gram-negative bactericidal activity in the leukocytes, bactericidal/permeability-increasing protein (BPI) has been shown to induce various effects in retinal cells in vivo and in vitro. METHODS: The authors recently reported that BPI can induce ERK1/2 and Akt activity and that it increases DNA synthesis in the bovine retinal pigment epithelial (RPE) and pericyte cells. The authors have extended the characterization of BPI interaction with membrane proteins from bovine RPE. Crude membrane pools from RPE were isolated, solubilized, and bound to rBPI(21) affinity column. Bound proteins were separated by SDS-PAGE and stained with Coomassie blue, which showed an intense band at 36 kDa consistently displaced by rBPI(21). RESULTS: Tandem mass spectrometry of the 36-kDa band suggested that cell surface protein glypican 4 (GPC4) serves as a putative BPI-binding protein. Heparitinase, phosphatidylinositol-specific phospholipase C, and anti-GPC4 antibody suppressed BPI-induced ERK and Akt phosphorylation in bovine RPE. Moreover, heparitinase also inhibited BPI actions on VEGF and PDGF-B mRNA expression induced by H(2)O(2). CONCLUSIONS: These new findings suggest that GPC4 is a specific binding protein for BPI on RPE to mediate the activation of ERK1/2, Akt, and the mRNA expressions of PDGF-B and VEGF.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , Proteínas Sanguíneas/metabolismo , Glipicanas/metabolismo , Proteínas de Membrana/metabolismo , Epitélio Pigmentado Ocular/metabolismo , Transdução de Sinais/fisiologia , Animais , Peptídeos Catiônicos Antimicrobianos/química , Proteínas Sanguíneas/química , Bovinos , Células Cultivadas , Eletroforese em Gel de Poliacrilamida , Glipicanas/química , Immunoblotting , Proteínas de Membrana/química , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação , Epitélio Pigmentado Ocular/efeitos dos fármacos , Polissacarídeo-Liases/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-sis/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espectrometria de Massas em Tandem , Fosfolipases Tipo C/farmacologia , Fator A de Crescimento do Endotélio Vascular/genética
16.
FASEB J ; 20(8): 1203-5, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16672634

RESUMO

PPARgamma agonists, thiazolidinediones, cause fluid retention and edema due to unknown mechanisms. We characterized the effect of rosiglitazone (RSG), a thiazolidinedione, to induce vascular permeability, vascular endothelial growth factor (VEGF) expression, and protein kinase C (PKC) activation with edema and wt gain. In lean, fatty and diabetic Zucker rats, and endothelial insulin receptor knockout mice, RSG increased wt and vascular permeability, selectively in fat and retina, but not in heart or skeletal muscle. H2O content and wt of epididymal fat were increased by RSG and correlated to increases in capillary permeability in fat and body wt. RSG induced VEGF mRNA expression and PKC activation in fat and retina up to 2.5-fold. Ruboxistaurin, a PKCbeta isoform inhibitor, in the latter 2 wk of a 4-wk study, normalized vascular permeability in fat and decreased total wt gain, H2O content, and wt of fat vs. RSG alone but did not decrease VEGF expression, basal permeability, or food intake. Finally, RSG did not increase wt or vascular permeability in PKCbeta knockout vs. control mice. Thus, thiazolidinedione's effects on edema and wt are partially due to an adipose tissue-selective activation of PKC and vascular permeability that may be prevented by PKCbeta inhibition.


Assuntos
Tecido Adiposo/efeitos dos fármacos , Permeabilidade Capilar/efeitos dos fármacos , Edema/induzido quimicamente , Hipoglicemiantes/toxicidade , PPAR gama/agonistas , Proteína Quinase C/metabolismo , Tiazolidinedionas/toxicidade , Aumento de Peso/efeitos dos fármacos , Tecido Adiposo/irrigação sanguínea , Tecido Adiposo/enzimologia , Animais , Diabetes Mellitus/metabolismo , Diabetes Mellitus/fisiopatologia , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Hipoglicemiantes/farmacologia , Indóis/farmacologia , Masculino , Maleimidas/farmacologia , Camundongos , Camundongos Knockout , Obesidade/metabolismo , Obesidade/fisiopatologia , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/genética , Proteína Quinase C beta , Ratos , Ratos Zucker , Receptor de Insulina/genética , Receptor de Insulina/fisiologia , Rosiglitazona , Tiazolidinedionas/farmacologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
17.
FASEB J ; 20(12): 2058-67, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17012258

RESUMO

Bactericidal/permeability-increasing protein (BPI) was originally identified as a lipopolysaccharide (LPS) binding protein with gram-negative bactericidal activity in the leukocytes. In this study, we characterized the previously unknown effects of BPI in the eye and the molecular mechanisms involved in its action. BPI mRNA was detected in bovine retina; retinal pigment epithelium; and primary cultures of bovine retinal pigment epithelial cells (RPE), pericytes (RPC), and endothelial cells (REC); while BPI protein was measured in human vitreous and plasma. BPI, but not control protein thaumatin, activated extracellular regulated kinase (ERK) and AKT, and increased DNA synthesis in RPE and RPC but not in REC. A human recombinant 21 kDa modified amino-terminal fragment of BPI (rBPI21) reduced H2O2-induced apoptosis in RPE and inhibited vascular endothelial growth factor (VEGF)-stimulated ERK phosphorylation in REC when preincubated with VEGF. Intraperitoneal (i.p.)-injected rBPI21 reduced ischemia-induced retinal neovascularization and diabetes-induced retinal permeability. Since BPI has unusual dual properties of promoting RPC and RPE growth while suppressing VEGF-induced REC growth and vascular permeability, the mechanistic understanding of BPI's action may provide novel therapeutic opportunities for diabetic retinopathy and age-related macular degeneration.


Assuntos
Inibidores da Angiogênese/farmacologia , Peptídeos Catiônicos Antimicrobianos/farmacologia , Proteínas Sanguíneas/farmacologia , Proteínas de Membrana/farmacologia , Retina/citologia , Vasos Retinianos/efeitos dos fármacos , Transdução de Sinais , Animais , Peptídeos Catiônicos Antimicrobianos/análise , Apoptose/efeitos dos fármacos , Proteínas Sanguíneas/análise , Permeabilidade Capilar/efeitos dos fármacos , Bovinos , Células Cultivadas , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Humanos , Proteínas de Membrana/análise , Neovascularização Patológica/tratamento farmacológico , Pericitos/citologia , Pericitos/efeitos dos fármacos , Pericitos/metabolismo , Epitélio Pigmentado Ocular/citologia , Epitélio Pigmentado Ocular/efeitos dos fármacos , Epitélio Pigmentado Ocular/metabolismo , Plasma/química , Proteínas Recombinantes/farmacologia , Retina/efeitos dos fármacos , Retina/metabolismo , Vasos Retinianos/crescimento & desenvolvimento , Fator A de Crescimento do Endotélio Vascular/farmacologia , Corpo Vítreo/química
18.
Invest Ophthalmol Vis Sci ; 58(7): 3100-3106, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28632845

RESUMO

Purpose: To determine the ocular consequences of a dominant-negative mutation in the p85α subunit of phosphatidylinositol 3-kinase (PIK3R1) using a knock-in mouse model of SHORT syndrome, a syndrome associated with short stature, lipodystrophy, diabetes, and Rieger anomaly in humans. Methods: We investigated knock-in mice heterozygous for the SHORT syndrome mutation changing arginine 649 to tryptophan in p85α (PIK3R1) using physical examination, optical coherence tomography (OCT), tonometry, and histopathologic sections from paraffin-embedded eyes, and compared the findings to similar investigations in two human subjects with SHORT syndrome heterozygous for the same mutation. Results: While overall eye development was normal with clear cornea and lens, normal anterior chamber volume, normal intraocular pressure, and no changes in the retinal structure, OCT images of the knock-in mouse eyes revealed a significant decrease in thickness and width of the iris resulting in increased pupil area and irregularity of shape. Both human subjects had Rieger anomaly with similar defects including thin irides and irregular pupils, as well as a prominent ring of Schwalbe, goniosynechiae, early cataract formation, and glaucoma. Although the two subjects had had diabetes for more than 30 years, there were no signs of diabetic retinopathy. Conclusions: A dominant-negative mutation in the p85α regulatory subunit of PI3K affects development of the iris, and contributes to changes consistent with anterior segment dysgenesis in both humans and mice.


Assuntos
Segmento Anterior do Olho/anormalidades , DNA/genética , Anormalidades do Olho/genética , Iris/anormalidades , Mutação , Fosfatidilinositol 3-Quinases/genética , Animais , Segmento Anterior do Olho/diagnóstico por imagem , Segmento Anterior do Olho/enzimologia , Classe Ia de Fosfatidilinositol 3-Quinase , Análise Mutacional de DNA , Modelos Animais de Doenças , Anormalidades do Olho/diagnóstico , Anormalidades do Olho/enzimologia , Oftalmopatias Hereditárias , Feminino , Humanos , Pressão Intraocular , Iris/diagnóstico por imagem , Masculino , Camundongos , Camundongos Knockout , Fosfatidilinositol 3-Quinases/metabolismo , Tomografia de Coerência Óptica
19.
J Biomed Opt ; 22(1): 16005, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-28056146

RESUMO

The purpose of this study is to evaluate the suitability of five different anesthetic protocols (isoflurane, isoflurane­xylazine, pentobarbital, ketamine­xylazine, and ketamine­xylazine­vecuronium) for functional blood flow imaging in the rat eye. Total retinal blood flow was measured at a series of time points using an ultrahigh-speed Doppler OCT system. Additionally, each anesthetic protocol was qualitatively evaluated according to the following criteria: (1) time-stability of blood flow, (2) overall rate of blood flow, (3) ocular immobilization, and (4) simplicity. We observed that different anesthetic protocols produced markedly different blood flows. Different anesthetic protocols also varied with respect to the four evaluated criteria. These findings suggest that the choice of anesthetic protocol should be carefully considered when designing and interpreting functional blood flow studies in the rat eye.


Assuntos
Anestesia/métodos , Anestésicos , Olho/irrigação sanguínea , Fluxo Sanguíneo Regional/efeitos dos fármacos , Animais , Isoflurano , Ketamina , Pentobarbital , Ratos , Retina , Brometo de Vecurônio , Xilazina
20.
Invest Ophthalmol Vis Sci ; 47(1): 86-92, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16384948

RESUMO

PURPOSE: To assess ocular and systemic safety and pharmacodynamic effects of the oral PKC beta selective inhibitor ruboxistaurin (RBX; LY333531) mesylate in patients with diabetes. METHODS: This was a double-masked, placebo-controlled, parallel, randomized, single-center clinical study evaluating the effect of oral administration of RBX (8 mg twice a day, 16 mg per day, or 16 mg twice a day) or placebo for 28 days in patients with no or very mild diabetic retinopathy on mean retinal circulation time (RCT), retinal blood flow (RBF), treatment-emergent adverse events, and other safety parameters. RESULTS: Twenty-nine persons aged 18 to 65 years with type 1 or 2 diabetes were evaluated. The only treatment-emergent adverse event with a statistically significant difference among treatment groups was abdominal pain, which was more common in placebo-treated subjects (P = 0.049). Statistically significant effects of RBX were observed on several hematologic and laboratory parameters, but values were within the normal reference range and none of the changes was deemed clinically meaningful. In patients receiving 16 mg RBX twice daily, the diabetes-induced increase in RCT was ameliorated, with a baseline-to-endpoint difference of -0.84 seconds (P = 0.046) relative to placebo. Increasing RBX dose was linearly associated with greater effect on RCT (P = 0.03). Similar results were observed with RBF. CONCLUSIONS: RBX was well tolerated at doses up to 16 mg twice daily for 28 days in patients with diabetes. It ameliorated diabetes-induced RCT abnormalities. No serious safety problems were identified in this patient population. Compared with prior published data, these findings represent the first direct human evidence of both bioavailability of RBX to retinal vessels and amelioration of diabetes-induced retinal hemodynamic abnormalities by an oral PKC beta inhibitor.


Assuntos
Diabetes Mellitus Tipo 1/complicações , Diabetes Mellitus Tipo 2/complicações , Retinopatia Diabética/prevenção & controle , Inibidores Enzimáticos/administração & dosagem , Indóis/administração & dosagem , Maleimidas/administração & dosagem , Proteína Quinase C/antagonistas & inibidores , Vasos Retinianos/efeitos dos fármacos , Administração Oral , Adolescente , Adulto , Idoso , Velocidade do Fluxo Sanguíneo/efeitos dos fármacos , Barreira Hematorretiniana/efeitos dos fármacos , Diabetes Mellitus Tipo 1/fisiopatologia , Diabetes Mellitus Tipo 2/fisiopatologia , Retinopatia Diabética/etiologia , Retinopatia Diabética/fisiopatologia , Método Duplo-Cego , Inibidores Enzimáticos/efeitos adversos , Feminino , Hemodinâmica/efeitos dos fármacos , Humanos , Indóis/efeitos adversos , Masculino , Maleimidas/efeitos adversos , Pessoa de Meia-Idade , Proteína Quinase C beta , Vasos Retinianos/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA