Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Front Pharmacol ; 14: 1287487, 2023.
Article in English | MEDLINE | ID: mdl-38178859

ABSTRACT

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.

2.
Circ Res ; 131(2): 168-183, 2022 07 08.
Article in English | MEDLINE | ID: mdl-35642564

ABSTRACT

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.


Subject(s)
Adipose Tissue, Brown , Atherosclerosis , Adipose Tissue, Brown/metabolism , Animals , Apolipoproteins E/metabolism , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/prevention & control , Endothelial Cells/metabolism , Insulin/metabolism , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism
3.
Transl Stroke Res ; 13(2): 287-299, 2022 04.
Article in English | MEDLINE | ID: mdl-34241810

ABSTRACT

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.


Subject(s)
Hypertension , Stroke , Animals , Atrial Natriuretic Factor , Blood Pressure/physiology , Cerebral Hemorrhage/complications , Hypertension/complications , Plasma Kallikrein , Rats , Rats, Inbred SHR , Stroke/complications
4.
Invest Ophthalmol Vis Sci ; 61(10): 53, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32866267

ABSTRACT

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.


Subject(s)
Diabetic Retinopathy/pathology , Retinal Vessels/pathology , Cross-Sectional Studies , Diabetic Retinopathy/diagnostic imaging , Female , Fundus Oculi , Humans , Male , Middle Aged , Retinal Vessels/diagnostic imaging , Retrospective Studies , Severity of Illness Index , Tomography, Optical Coherence
5.
Cell Metab ; 32(2): 215-228.e7, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32663458

ABSTRACT

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.


Subject(s)
Fats/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Muscle, Skeletal/metabolism , Acetyl-CoA Carboxylase/metabolism , Animals , Cell Line , Exercise Tolerance , Female , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Oxidation-Reduction
6.
J Hazard Mater ; 386: 121659, 2020 03 15.
Article in English | MEDLINE | ID: mdl-31776080

ABSTRACT

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.


Subject(s)
Capillary Permeability/drug effects , MAP Kinase Signaling System/drug effects , Particulate Matter/toxicity , Reactive Oxygen Species/metabolism , Acetylcysteine/pharmacology , Adherens Junctions/drug effects , Animals , Antigens, CD/metabolism , Butadienes/pharmacology , Cadherins/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Intercellular Junctions/drug effects , Male , Mice , Mice, Inbred BALB C , Nitriles/pharmacology , Oxidative Stress/drug effects
7.
Exp Eye Res ; 186: 107744, 2019 09.
Article in English | MEDLINE | ID: mdl-31351056

ABSTRACT

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.


Subject(s)
Bradykinin/pharmacology , Macular Edema/metabolism , Proteome/metabolism , Retina/metabolism , Vasodilator Agents/pharmacology , Animals , Blotting, Western , Intravitreal Injections , Macular Edema/chemically induced , Male , Plasma Kallikrein , Proteomics , Rats , Rats, Sprague-Dawley , Retina/diagnostic imaging , Retinal Vessels/metabolism , Tomography, Optical Coherence , Vascular Endothelial Growth Factor A/pharmacology
8.
Sci Transl Med ; 11(499)2019 07 03.
Article in English | MEDLINE | ID: mdl-31270273

ABSTRACT

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.


Subject(s)
Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Eye Proteins/metabolism , Retina/metabolism , Retina/pathology , Retinol-Binding Proteins/metabolism , 3-O-Methylglucose/metabolism , Acids/metabolism , Animals , Cell Movement/drug effects , Deoxyglucose/metabolism , Diabetes Mellitus/physiopathology , Diabetic Retinopathy/physiopathology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Ependymoglial Cells/drug effects , Ependymoglial Cells/metabolism , Eye Proteins/administration & dosage , Eye Proteins/blood , Eye Proteins/chemistry , Glycolysis/drug effects , Humans , Intravitreal Injections , Mice, Inbred C57BL , Mice, Transgenic , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , Protective Agents/pharmacology , Protein Domains , Rats, Inbred Lew , Recombinant Proteins/pharmacology , Reproducibility of Results , Retina/physiopathology , Retinol-Binding Proteins/administration & dosage , Retinol-Binding Proteins/chemistry , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vitreous Body/drug effects , Vitreous Body/metabolism
9.
Invest Ophthalmol Vis Sci ; 58(7): 3100-3106, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28632845

ABSTRACT

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.


Subject(s)
Anterior Eye Segment/abnormalities , DNA/genetics , Eye Abnormalities/genetics , Iris/abnormalities , Mutation , Phosphatidylinositol 3-Kinases/genetics , Animals , Anterior Eye Segment/diagnostic imaging , Anterior Eye Segment/enzymology , Class Ia Phosphatidylinositol 3-Kinase , DNA Mutational Analysis , Disease Models, Animal , Eye Abnormalities/diagnosis , Eye Abnormalities/enzymology , Eye Diseases, Hereditary , Female , Humans , Intraocular Pressure , Iris/diagnostic imaging , Male , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , Tomography, Optical Coherence
10.
Blood ; 129(16): 2280-2290, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28130211

ABSTRACT

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.


Subject(s)
Angioedema/chemically induced , Cerebral Hemorrhage/chemically induced , Fibrinolytic Agents/adverse effects , Plasma Kallikrein/metabolism , Stroke/drug therapy , Thrombosis/drug therapy , Tissue Plasminogen Activator/adverse effects , Administration, Intravenous , Angioedema/blood , Angioedema/genetics , Animals , Cerebral Hemorrhage/blood , Cerebral Hemorrhage/genetics , Disease Models, Animal , Factor XII/genetics , Factor XII/metabolism , Gene Expression , Humans , Infarction, Middle Cerebral Artery/blood , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/pathology , Male , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Knockout , Plasma Kallikrein/genetics , Stroke/blood , Stroke/genetics , Stroke/pathology , Thrombolytic Therapy , Thrombosis/blood , Thrombosis/genetics , Thrombosis/pathology
11.
J Biomed Opt ; 22(1): 16005, 2017 01 01.
Article in English | MEDLINE | ID: mdl-28056146

ABSTRACT

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.


Subject(s)
Anesthesia/methods , Anesthetics , Eye/blood supply , Regional Blood Flow/drug effects , Animals , Isoflurane , Ketamine , Pentobarbital , Rats , Retina , Vecuronium Bromide , Xylazine
12.
Invest Ophthalmol Vis Sci ; 57(6): 2390-9, 2016 05 01.
Article in English | MEDLINE | ID: mdl-27138737

ABSTRACT

PURPOSE: Plasma kallikrein is a serine protease and circulating component of inflammation, which exerts clinically significant effects on vasogenic edema. This study examines the role of plasma kallikrein in VEGF-induced retinal edema. METHODS: Intravitreal injections of VEGF and saline vehicle were performed in plasma prekallikrein-deficient (KLKB1-/-) and wild-type (WT) mice, and in both rats and mice receiving a selective plasma kallikrein inhibitor, VA999272. Retinal vascular permeability (RVP) and retinal thickness were measured by Evans blue permeation and optical coherence tomography, respectively. The retinal kallikrein kinin system was examined by Western blotting and immunohistochemistry. Retinal neovascularization was investigated in KLKB1-/- and WT mice subjected to oxygen-induced retinopathy. RESULTS: Vascular endothelial growth factor-induced RVP and retinal thickening were reduced in KLKB1-/- mice by 68% and 47%, respectively, compared to VEGF responses in WT mice. Plasma kallikrein also contributes to TNFα-induced retinal thickening, which was reduced by 52% in KLKB1-/- mice. Systemic administration of VA999272 reduced VEGF-induced retinal thickening by 57% (P < 0.001) in mice and 53% (P < 0.001) in rats, compared to vehicle-treated controls. Intravitreal injection of VEGF in WT mice increased plasma prekallikrein in the retina, which was diffusely distributed throughout the inner and outer retinal layers. Avascular and neovascular areas induced by oxygen-induced retinopathy were similar in WT and KLKB1-/- mice. CONCLUSIONS: Vascular endothelial growth factor increases extravasation of plasma kallikrein into the retina, and plasma kallikrein is required for the full effects of VEGF on RVP and retinal thickening in rodents. Systemic plasma kallikrein inhibition may provide a therapeutic opportunity to treat VEGF-induced retina edema.


Subject(s)
Macular Edema/metabolism , Plasma Kallikrein/metabolism , Retina/pathology , Animals , Blotting, Western , Capillary Permeability , Intravitreal Injections , Macular Edema/chemically induced , Macular Edema/pathology , Male , Mice , Mice, Inbred C57BL , Plasma Kallikrein/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Retina/metabolism , Retina/physiopathology , Tomography, Optical Coherence , Vascular Endothelial Growth Factor A/administration & dosage , Vascular Endothelial Growth Factor A/poisoning
13.
Nanotoxicology ; 10(4): 501-11, 2016.
Article in English | MEDLINE | ID: mdl-26399585

ABSTRACT

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.


Subject(s)
Adherens Junctions/drug effects , Capillary Permeability/drug effects , Metal Nanoparticles/chemistry , Metal Nanoparticles/toxicity , Plasma/metabolism , Retinal Vessels/drug effects , Silver/toxicity , Animals , Antigens, CD/metabolism , Aprotinin/pharmacology , Bradykinin/analogs & derivatives , Bradykinin/blood , Bradykinin/pharmacology , Cadherins/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Factor XII/metabolism , Intravitreal Injections , Kallikreins/blood , Male , Metal Nanoparticles/administration & dosage , Mice , Prekallikrein/metabolism , Silver/administration & dosage , Silver/chemistry , Silver/pharmacokinetics
14.
Diabetes ; 64(10): 3588-99, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25979073

ABSTRACT

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.


Subject(s)
Diabetes Complications/etiology , Kallikrein-Kinin System/physiology , Kallikreins/metabolism , Kinins/metabolism , Macular Edema/etiology , Vascular Endothelial Growth Factor A/metabolism , Animals , Cattle , Cells, Cultured , Diabetes Mellitus, Experimental/complications , Endothelial Cells/physiology , Gene Expression Regulation/physiology , Humans , Mice , Rats , Retinal Vessels/pathology , Vitreous Body/chemistry
15.
Invest Ophthalmol Vis Sci ; 54(2): 1086-94, 2013 Feb 07.
Article in English | MEDLINE | ID: mdl-23299478

ABSTRACT

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.


Subject(s)
Plasma Kallikrein/metabolism , Retinal Diseases/etiology , Retinal Hemorrhage/complications , Retinal Vessels/pathology , Animals , Blood , Blood-Retinal Barrier/drug effects , Bradykinin/pharmacology , Capillary Permeability , Cattle , Cells, Cultured , Collagenases/pharmacology , Concanavalin A/metabolism , Evans Blue/metabolism , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/metabolism , Fluorophotometry , Intravitreal Injections , Leukostasis/etiology , Male , Pericytes/drug effects , Pericytes/metabolism , Plasma Kallikrein/pharmacology , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Retinal Diseases/metabolism , Retinal Hemorrhage/metabolism , Retinal Vessels/metabolism , Tandem Mass Spectrometry , Vitreous Body/drug effects , Vitreous Body/metabolism
16.
Biomed Opt Express ; 3(5): 1047-61, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22567595

ABSTRACT

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.

17.
Arterioscler Thromb Vasc Biol ; 32(5): 1124-31, 2012 May.
Article in English | MEDLINE | ID: mdl-22426129

ABSTRACT

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.


Subject(s)
Apolipoproteins E/genetics , Atherosclerosis/genetics , Hyperinsulinism/complications , Insulin Resistance , Animals , Apolipoproteins E/blood , Atherosclerosis/blood , Atherosclerosis/etiology , Disease Progression , Female , Gene Expression Regulation , Hyperinsulinism/blood , Hyperinsulinism/genetics , Insulin Receptor Substrate Proteins/biosynthesis , Insulin Receptor Substrate Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction
18.
Diabetes ; 60(5): 1590-8, 2011 May.
Article in English | MEDLINE | ID: mdl-21444925

ABSTRACT

OBJECTIVE: Plasma kallikrein (PK) has been identified in vitreous fluid obtained from individuals with diabetic retinopathy and has been implicated in contributing to retinal vascular dysfunction. In this report, we examined the effects of PK on retinal vascular functions and thickness in diabetic rats. RESEARCH DESIGN AND METHODS: We investigated the effects of a selective PK inhibitor, ASP-440, and C1 inhibitor (C1-INH), the primary physiological inhibitor of PK, on retinal vascular permeability (RVP) and hemodynamics in rats with streptozotocin-induced diabetes. The effect of intravitreal PK injection on retinal thickness was examined by spectral domain optical coherence tomography. RESULTS: Systemic continuous administration of ASP-440 for 4 weeks initiated at the time of diabetes onset inhibited RVP by 42% (P = 0.013) and 83% (P < 0.001) at doses of 0.25 and 0.6 mg/kg per day, respectively. Administration of ASP-440 initiated 2 weeks after the onset of diabetes ameliorated both RVP and retinal blood flow abnormalities in diabetic rats measured at 4 weeks' diabetes duration. Intravitreal injection of C1-INH similarly decreased impaired RVP in rats with 2 weeks' diabetes duration. Intravitreal injection of PK increased both acute RVP and sustained focal RVP (24 h postinjection) to a greater extent in diabetic rats compared with nondiabetic control rats. Intravitreal injection of PK increased retinal thickness compared with baseline to a greater extent (P = 0.017) in diabetic rats (from 193 ± 10 µm to 223 ± 13 µm) compared with nondiabetic rats (from 182 ± 8 µm to 193 ± 9 µm). CONCLUSIONS: These results show that PK contributes to retinal vascular dysfunctions in diabetic rats and that the combination of diabetes and intravitreal injection of PK in rats induces retinal thickening.


Subject(s)
Plasma Kallikrein/metabolism , Retina/metabolism , Retina/pathology , Animals , Complement C1 Inhibitor Protein/pharmacology , Enzyme Inhibitors/pharmacology , Fluorescein Angiography , Fluorophotometry , Hemodynamics/drug effects , Humans , Male , Plasma Kallikrein/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Retina/drug effects
19.
Nat Med ; 17(2): 206-10, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21258336

ABSTRACT

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.


Subject(s)
Cerebral Hemorrhage/physiopathology , Hematoma/physiopathology , Hyperglycemia/physiopathology , Plasma Kallikrein/physiology , Animals , Blood-Brain Barrier/physiopathology , Brain/drug effects , Brain/physiopathology , Diabetes Mellitus, Experimental/physiopathology , Injections, Intraventricular , Mice , Mice, Inbred C57BL , Plasma Kallikrein/pharmacology , Plasminogen/physiology , Platelet Aggregation/drug effects , Platelet Aggregation/physiology , Rats , Rats, Sprague-Dawley
20.
Semin Ophthalmol ; 25(5-6): 289-94, 2010.
Article in English | MEDLINE | ID: mdl-21091014

ABSTRACT

Diabetic retinopathy is the major cause of acquired blindness in working-age adults. Studies of the vitreous proteome have provided insights into the etiology of diabetic retinopathy and suggested potential molecular targets for treatments. Further characterization of the protein changes associated with the progression of this disease may suggest additional therapeutic approaches as well as reveal novel factors that may be useful in predicting risk and functional outcomes of interventional therapies. This article provides an overview of the various techniques used for proteomic analysis of the vitreous and details results from various studies evaluating vitreous of diabetic patients using the proteomic approach.


Subject(s)
Diabetic Retinopathy/metabolism , Eye Proteins/metabolism , Proteomics , Vitreous Body/metabolism , Diabetic Retinopathy/etiology , Humans , Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL