Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
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
2.
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
3.
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
4.
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.

5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
Invest Ophthalmol Vis Sci ; 47(6): 2701-8, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16723489

RESUMO

PURPOSE: Although vascular endothelial growth factor (VEGF) is a key mediator of retinal vascular permeability (RVP), there may be additional humoral contributors. Hepatocyte growth factor (HGF) induces endothelial cell separation, regulates expression of cell adhesion molecules and is increased in the vitreous fluid of patients with proliferative diabetic retinopathy. The purpose of this study was to evaluate the in vivo effects of HGF on RVP and retinal hemodynamics and delineate the signaling pathways. METHODS: RVP was assessed by vitreous fluorescein fluorophotometry in rats. Time course and dose-response were determined after intravitreal HGF injection. MAP kinase (MAPK), phosphatidylinositol 3-kinase (PI-3 kinase), and protein kinase C (PKC) involvement were examined by using selective inhibitors. Retinal blood flow (RBF) and mean circulation time (MCT) were evaluated by video fluorescein angiography. RESULTS: HGF increased RVP in a time- and dose-dependent manner. HGF-induced RVP was evident 5 minutes after injection, and reached maximal levels after 25 minutes (+107% versus vehicle, P=0.002). This effect was comparable to that of maximum VEGF stimulation (134%+/-128% at 25 ng/mL). Selective inhibitors of MAPK (PD98059) and PI-3 kinase (LY294002) suppressed HGF-induced RVP by 86%+/-44% (P=0.015) and 97%+/-59% (P=0.021), respectively. Non-isoform-selective inhibition of PKC did not significantly decrease HGF-induced RVP. Although VEGF increases RBF and reduces MCT, HGF did not affect either. CONCLUSIONS: HGF increases RVP in a time- and dose-dependent manner at physiologically relevant concentrations with a magnitude and profile similar to that of VEGF, without affecting retinal hemodynamics. Thus, HGF may represent another clinically significant contributor to retinal edema distinct from the actions of VEGF.


Assuntos
Permeabilidade Capilar/efeitos dos fármacos , Fator de Crescimento de Hepatócito/farmacologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Vasos Retinianos/fisiologia , Animais , Circulação Sanguínea , Western Blotting , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Angiofluoresceinografia , Fluorofotometria , Hemodinâmica/fisiologia , Masculino , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Inibidores de Fosfoinositídeo-3 Quinase , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Ratos , Ratos Sprague-Dawley , Fluxo Sanguíneo Regional , Vasos Retinianos/enzimologia , Fatores de Tempo
14.
Nanotoxicology ; 10(4): 501-11, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26399585

RESUMO

Silver nanoparticles (AgNPs) have been extensively used as antibacterial component in numerous healthcare, biomedical and consumer products. Therefore, their adverse effects to biological systems have become a major concern. AgNPs have been shown to be absorbed into circulation and redistributed into various organs. It is thus of great importance to understand how these nanoparticles affect vascular permeability and uncover the underlying molecular mechanisms. A negatively charged mecaptoundeonic acid-capped silver nanoparticle (MUA@AgNP) was investigated in this work. Ex vivo experiments in mouse plasma revealed that MUA@AgNPs caused plasma prekallikrein cleavage, while positively charged or neutral AgNPs, as well as Ag ions had no effect. In vitro tests revealed that MUA@AgNPs activated the plasma kallikrein-kinin system (KKS) by triggering Hageman factor autoactivation. By using specific inhibitors aprotinin and HOE 140, we demonstrated that KKS activation caused the release of bradykinin, which activated B2 receptors and induced the shedding of adherens junction protein, VE-cadherin. These biological perturbations eventually resulted in endothelial paracellular permeability in mouse retina after intravitreal injection of MUA@AgNPs. The findings from this work provided key insights for toxicity modulation and biomedical applications of AgNPs.


Assuntos
Junções Aderentes/efeitos dos fármacos , Permeabilidade Capilar/efeitos dos fármacos , Nanopartículas Metálicas/química , Nanopartículas Metálicas/toxicidade , Plasma/metabolismo , Vasos Retinianos/efeitos dos fármacos , Prata/toxicidade , Animais , Antígenos CD/metabolismo , Aprotinina/farmacologia , Bradicinina/análogos & derivados , Bradicinina/sangue , Bradicinina/farmacologia , Caderinas/metabolismo , Células Cultivadas , Relação Dose-Resposta a Droga , Fator XII/metabolismo , Injeções Intravítreas , Calicreínas/sangue , Masculino , Nanopartículas Metálicas/administração & dosagem , Camundongos , Pré-Calicreína/metabolismo , Prata/administração & dosagem , Prata/química , Prata/farmacocinética
15.
Circulation ; 105(3): 373-9, 2002 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-11804995

RESUMO

BACKGROUND: Inadequate angiogenic response to ischemia in the myocardium of diabetic patients could result in poor collateral formation. Yet, excessive neovascularization in the retina causes proliferative diabetic retinopathy. Since vascular endothelial growth factor (VEGF) is the major angiogenic factor expressed in response to hypoxia, we have characterized expression of VEGF and its receptors in retina, renal glomeruli, aorta, and myocardium in insulin-resistant and diabetic states. Methods and Results- The expression of mRNA and protein for VEGF and its receptors, VEGF-R1 and VEGF-R2, in the myocardium was decreased significantly by 40% to 70% in both diabetic and insulin-resistant nondiabetic rats. Twofold reductions in VEGF and VEGF-R2 were observed in ventricles from diabetic patients compared with nondiabetic donors. In contrast, expression of VEGF and its receptors were increased 2-fold in retina and glomeruli from diabetic or insulin-resistant rats. Insulin treatment of diabetic rats normalized changes in both cardiac and microvascular tissues. Insulin increased VEGF mRNA expression in cultured rat neonatal cardiac myocytes. CONCLUSIONS: The results documented for the first time that differential regulation of VEGF and its receptors exist between microvascular and cardiac tissues, which can be regulated by insulin. These results provide a potential explanation for concomitant capillary leakage and neovascularization in the retina and inadequate collateral formation in the myocardium of insulin-resistant and diabetic patients.


Assuntos
Diabetes Mellitus/metabolismo , Fatores de Crescimento Endotelial/metabolismo , Resistência à Insulina , Linfocinas/metabolismo , Miocárdio/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Fatores de Crescimento/metabolismo , Animais , Células Cultivadas , Circulação Colateral , Circulação Coronária , Diabetes Mellitus/sangue , Fatores de Crescimento Endotelial/genética , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Insulina/farmacologia , Glomérulos Renais/metabolismo , Linfocinas/genética , Masculino , Pessoa de Meia-Idade , Neovascularização Patológica , Obesidade , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Ratos Zucker , Receptores Proteína Tirosina Quinases/genética , Receptores de Fatores de Crescimento/genética , Receptores de Fatores de Crescimento do Endotélio Vascular , Retina/metabolismo , Transcrição Gênica/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
16.
Diabetes ; 52(3): 829-37, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12606527

RESUMO

Increases in leukostasis/monocyte adhesion to the capillary endothelium (leukostasis) and decreases in retinal blood flow may be causally associated and are implicated in the pathogenesis of diabetic retinopathy. In this study, we demonstrate that increases in leukostasis are observed in insulin-resistant states without diabetes, whereas decreases in retinal blood flow require diabetes and hyperglycemia. Microimpaction studies using beads mimicking retinal capillary obstruction by leukocytes did not affect retinal blood flow. In diabetic rats, treatment with the antioxidant alpha-lipoic acid normalized the amount of leukostasis but not retinal blood flow. In contrast, treatment with D-alpha-tocopherol and protein kinase-C beta-isoform inhibition (LY333531) prevented the increases in leukostasis and decreases in retinal blood flow in diabetic rats. Serum hydroxyperoxide, a marker of oxidative stress, was increased in diabetic rats, but normalized by treatment with antioxidants alpha-lipoic acid and D-alpha-tocopherol and, surprisingly, PKC beta-isoform inhibition. These findings suggest that leukostasis is associated with endothelial dysfunction, insulin resistance, and oxidative stress but is not related to retinal blood flow and is not sufficient to cause diabetic-like retinopathy. Moreover, treatment with PKC beta inhibition is effective to normalize diabetes or hyperglycemia-induced PKC beta-isoform activation and oxidative stress.


Assuntos
Diabetes Mellitus Experimental/fisiopatologia , Hemodinâmica , Resistência à Insulina , Leucostasia , Proteínas Quinases/metabolismo , Retina/patologia , Animais , Antioxidantes/uso terapêutico , Velocidade do Fluxo Sanguíneo , Diabetes Mellitus Experimental/patologia , Retinopatia Diabética/etiologia , Endotélio Vascular/fisiopatologia , Inibidores Enzimáticos/uso terapêutico , Peróxido de Hidrogênio/sangue , Indóis/uso terapêutico , Leucostasia/tratamento farmacológico , Masculino , Maleimidas/uso terapêutico , Microcirculação/fisiopatologia , Microesferas , Oxidantes/metabolismo , Estresse Oxidativo , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C beta , Ratos , Ratos Long-Evans , Ratos Zucker , Vasos Retinianos/patologia , Vasos Retinianos/fisiopatologia , Ácido Tióctico/farmacologia , alfa-Tocoferol/uso terapêutico
17.
Diabetes ; 64(10): 3588-99, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25979073

RESUMO

This study characterizes the kallikrein-kinin system in vitreous from individuals with diabetic macular edema (DME) and examines mechanisms contributing to retinal thickening and retinal vascular permeability (RVP). Plasma prekallikrein (PPK) and plasma kallikrein (PKal) were increased twofold and 11.0-fold (both P < 0.0001), respectively, in vitreous from subjects with DME compared with those with a macular hole (MH). While the vascular endothelial growth factor (VEGF) level was also increased in DME vitreous, PKal and VEGF concentrations do not correlate (r = 0.266, P = 0.112). Using mass spectrometry-based proteomics, we identified 167 vitreous proteins, including 30 that were increased in DME (fourfold or more, P < 0.001 vs. MH). The majority of proteins associated with DME displayed a higher correlation with PPK than with VEGF concentrations. DME vitreous containing relatively high levels of PKal and low VEGF induced RVP when injected into the vitreous of diabetic rats, a response blocked by bradykinin receptor antagonism but not by bevacizumab. Bradykinin-induced retinal thickening in mice was not affected by blockade of VEGF receptor 2. Diabetes-induced RVP was decreased by up to 78% (P < 0.001) in Klkb1 (PPK)-deficient mice compared with wild-type controls. B2- and B1 receptor-induced RVP in diabetic mice was blocked by endothelial nitric oxide synthase (NOS) and inducible NOS deficiency, respectively. These findings implicate the PKal pathway as a VEGF-independent mediator of DME.


Assuntos
Complicações do Diabetes/etiologia , Sistema Calicreína-Cinina/fisiologia , Calicreínas/metabolismo , Cininas/metabolismo , Edema Macular/etiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Bovinos , Células Cultivadas , Diabetes Mellitus Experimental/complicações , Células Endoteliais/fisiologia , Regulação da Expressão Gênica/fisiologia , Humanos , Camundongos , Ratos , Vasos Retinianos/patologia , Corpo Vítreo/química
18.
Invest Ophthalmol Vis Sci ; 54(2): 1086-94, 2013 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-23299478

RESUMO

PURPOSE: Retinal hemorrhages occur in a variety of sight-threatening conditions including ocular trauma, high altitude retinopathy, and chronic diseases such as diabetic and hypertensive retinopathies. The goal of this study is to investigate the effects of blood in the vitreous on retinal vascular function in rats. METHODS: Intravitreal injections of autologous blood, plasma kallikrein (PK), bradykinin, and collagenase were performed in Sprague-Dawley and Long-Evans rats. Retinal vascular permeability was measured using vitreous fluorophotometry and Evans blue dye permeation. Leukostasis was measured by fluorescein isothiocyanate-coupled concanavalin A lectin and acridine orange labeling. Retinal hemorrhage was examined on retinal flatmounts. Primary cultures of bovine retinal pericytes were cultured in the presence of 25 nM PK for 24 hours. The pericyte-conditioned medium was collected and the collagen proteome was analyzed by tandem mass spectrometry. RESULTS: Intravitreal injection of autologous blood induced retinal vascular permeability and retinal leukostasis, and these responses were ameliorated by PK inhibition. Intravitreal injections of exogenous PK induced retinal vascular permeability, leukostasis, and retinal hemorrhage. Proteomic analyses showed that PK increased collagen degradation in pericyte-conditioned medium and purified type IV collagen. Intravitreal injection of collagenase mimicked PK's effect on retinal hemorrhage. CONCLUSIONS: Intraocular hemorrhage increases retinal vascular permeability and leukostasis, and these responses are mediated, in part, via PK. Intravitreal injections of either PK or collagenase, but not bradykinin, induce retinal hemorrhage in rats. PK exerts collagenase-like activity that may contribute to blood-retinal barrier dysfunction.


Assuntos
Calicreína Plasmática/metabolismo , Doenças Retinianas/etiologia , Hemorragia Retiniana/complicações , Vasos Retinianos/patologia , Animais , Sangue , Barreira Hematorretiniana/efeitos dos fármacos , Bradicinina/farmacologia , Permeabilidade Capilar , Bovinos , Células Cultivadas , Colagenases/farmacologia , Concanavalina A/metabolismo , Azul Evans/metabolismo , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/metabolismo , Fluorofotometria , Injeções Intravítreas , Leucostasia/etiologia , Masculino , Pericitos/efeitos dos fármacos , Pericitos/metabolismo , Calicreína Plasmática/farmacologia , Ratos , Ratos Long-Evans , Ratos Sprague-Dawley , Doenças Retinianas/metabolismo , Hemorragia Retiniana/metabolismo , Vasos Retinianos/metabolismo , Espectrometria de Massas em Tandem , Corpo Vítreo/efeitos dos fármacos , Corpo Vítreo/metabolismo
19.
Biomed Opt Express ; 3(5): 1047-61, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22567595

RESUMO

We present an approach to measure pulsatile total retinal arterial blood flow in humans and rats using ultrahigh speed Doppler OCT. The axial blood velocity is measured in an en face plane by raster scanning and the flow is calculated by integrating over the vessel area, without the need to measure the Doppler angle. By measuring flow at the central retinal artery, the scan area can be very small. Combined with ultrahigh speed, this approach enables high volume acquisition rates necessary for pulsatile total flow measurement without modification in the OCT system optics. A spectral domain OCT system at 840nm with an axial scan rate of 244kHz was used for this study. At 244kHz the nominal axial velocity range that could be measured without phase wrapping was ±37.7mm/s. By repeatedly scanning a small area centered at the central retinal artery with high volume acquisition rates, pulsatile flow characteristics, such as systolic, diastolic, and mean total flow values, were measured. Real-time Doppler C-scan preview is proposed as a guidance tool to enable quick and easy alignment necessary for large scale studies. Data processing for flow calculation can be entirely automatic using this approach because of the simple and robust algorithm. Due to the rapid volume acquisition rate and the fact that the measurement is independent of Doppler angle, this approach is inherently less sensitive to involuntary eye motion. This method should be useful for investigation of small animal models of ocular diseases as well as total blood flow measurements in human patients in the clinic.

20.
Nat Med ; 17(2): 206-10, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21258336

RESUMO

Hyperglycemia is associated with greater hematoma expansion and poor clinical outcomes after intracerebral hemorrhage. We show that cerebral hematoma expansion triggered by intracerebral infusion of autologous blood is greater in diabetic rats and mice compared to nondiabetic controls and that this augmented expansion is ameliorated by plasma kallikrein (PK) inhibition or deficiency. Intracerebral injection of purified PK augmented hematoma expansion in both diabetic and acutely hyperglycemic rats, whereas injection of bradykinin, plasmin or tissue plasminogen activator did not elicit such a response. This response, which occurs rapidly, was prevented by co-injection of the glycoprotein VI agonist convulxin and was mimicked by glycoprotein VI inhibition or deficiency, implicating an effect of PK on inhibiting platelet aggregation. We show that PK inhibits collagen-induced platelet aggregation by binding collagen, a response enhanced by elevated glucose concentrations. The effect of hyperglycemia on hematoma expansion and PK-mediated inhibition of platelet aggregation could be mimicked by infusing mannitol. These findings suggest that hyperglycemia augments cerebral hematoma expansion by PK-mediated osmotic-sensitive inhibition of hemostasis.


Assuntos
Hemorragia Cerebral/fisiopatologia , Hematoma/fisiopatologia , Hiperglicemia/fisiopatologia , Calicreína Plasmática/fisiologia , Animais , Barreira Hematoencefálica/fisiopatologia , Encéfalo/efeitos dos fármacos , Encéfalo/fisiopatologia , Diabetes Mellitus Experimental/fisiopatologia , Injeções Intraventriculares , Camundongos , Camundongos Endogâmicos C57BL , Calicreína Plasmática/farmacologia , Plasminogênio/fisiologia , Agregação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/fisiologia , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA