Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Am J Pathol ; 185(6): 1769-82, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25913075

ABSTRACT

Retinopathy of prematurity is a sight-threatening complication of premature birth caused by nitro-oxidative insult to the developing retinal vasculature during therapeutic hyperoxia exposure and later ischemia-induced neovascularization on supplemental oxygen withdrawal. In the vasodegenerative phase, during hyperoxia, defective endothelial nitric oxide synthase (NOS) produces reactive oxygen and nitrogen free radicals rather than vasoprotective nitric oxide for unclear reasons. Crucially, normal NOS function depends on availability of the cofactor (6R)-5,6,7,8-tetrahydrobiopterin (BH4). Because BH4 synthesis is controlled enzymatically by GTP cyclohydrolase (GTPCH), we used GTPCH-depleted mice [hyperphenylalaninemia strain (hph1)] to investigate the impact of hyperoxia on BH4 bioavailability and retinal vascular pathology in the neonate. Hyperoxia decreased BH4 in retinas, lungs, and aortas in all experimental groups, resulting in a dose-dependent decrease in NOS activity and, in the wild-type group, elevated NOS-derived superoxide. Retinal dopamine levels were similarly diminished, consistent with the dependence of tyrosine hydroxylase on BH4. Despite greater depletion of BH4, the hph(+/-) and hph1(-/-) groups did not show exacerbated hyperoxia-induced vessel closure, but exhibited greater vascular protection and reduced progression to neovascular disease. This vasoprotective effect was independent of enhanced circulating vascular endothelial growth factor (VEGF), which was reduced by hyperoxia, but to local retinal ganglion cell layer-derived VEGF. In conclusion, a constitutively higher level of VEGF expression associated with retinal development protects GTPCH-deficient neonates from oxygen-induced vascular damage.


Subject(s)
Biopterins/analogs & derivatives , Hyperoxia/metabolism , Nitric Oxide Synthase/metabolism , Retina/metabolism , Retinopathy of Prematurity/metabolism , Animals , Biopterins/metabolism , Female , Hyperoxia/pathology , Male , Mice , Retina/pathology , Retinopathy of Prematurity/pathology , Vascular Endothelial Growth Factor A/metabolism
2.
Biochem J ; 448(1): 103-13, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22880819

ABSTRACT

Gene targeting by microRNAs is important in health and disease. We developed a functional assay for identifying microRNA targets and applied it to the K(+) channel K(ir)2.1 [KCNJ2 (potassium inwardly-rectifying channel, subfamily J, member 2)] which is dysregulated in cardiac and vascular disorders. The 3'UTR (untranslated region) was inserted downstream of the mCherry red fluorescent protein coding sequence in a mammalian expression plasmid. MicroRNA sequences were inserted into the pSM30 expression vector which provides enhanced green fluorescent protein as an indicator of microRNA expression. HEK (human embryonic kidney)-293 cells were co-transfected with the mCherry-3'UTR plasmid and a pSM30-based plasmid with a microRNA insert. The principle of the assay is that functional targeting of the 3'UTR by the microRNA results in a decrease in the red/green fluorescence intensity ratio as determined by automated image analysis. The method was validated with miR-1, a known down-regulator of K(ir)2.1 expression, and was used to investigate the targeting of the K(ir)2.1 3'UTR by miR-212. The red/green ratio was lower in miR-212-expressing cells compared with the non-targeting controls, an effect that was attenuated by mutating the predicted target site. miR-212 also reduced inward rectifier current and K(ir)2.1 protein in HeLa cells. This novel assay has several advantages over traditional luciferase-based assays including larger sample size, amenability to time course studies and adaptability to high-throughput screening.


Subject(s)
3' Untranslated Regions/genetics , MicroRNAs/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Base Pairing , Binding Sites , Down-Regulation , Fluorometry/methods , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , HEK293 Cells , HeLa Cells , Humans , Luciferases/analysis , Luciferases/genetics , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Mutagenesis, Site-Directed , Patch-Clamp Techniques , Potassium Channels, Inwardly Rectifying/physiology , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Red Fluorescent Protein
3.
Cell Cycle ; 22(11): 1380-1390, 2023 06.
Article in English | MEDLINE | ID: mdl-37183425

ABSTRACT

FBXW7 is the substrate recognition component of the E3 ubiquitin ligase SCFFBW7 complex which controls the levels of CYCLINE, c-MYC and HIF1α proteins crucial for cell growth and differentiation. Mutations in FBXW7 are frequently associated with tumourigenesis. While examining FBXW7 regulation we were compelled to reevaluate a commonly used anti-FBXW7 antibody. Retinal microvascular endothelial cells (RMEC) were exposed to normoxia (21% oxygen) or hypoxia (1% oxygen) for 24 h or treated with MG132 and protein extracted for western blotting. Flag-tagged FBXW7-α, ß or γ isoforms were transfected into HEK293A cells and processed using denaturing and native extraction protocols for western blotting or immunoprecipitation analysis. Two anti-FBXW7 antibodies were used, one raised to the unique FBXW7α N-terminus and the other to the C-terminus region common to all isoforms. Initial studies showed that the pan-isoform C-terminus antibody detected a single 64kDa band in RMEC rather than any of the predicted sizes for FBXW7. In contrast, expression of the isoform-specific constructs, detected with an anti-Flag antibody, confirmed the expected migratory distance of 110kDa, 68kDa and 65kDa for α, ß and γ respectfully. Similarly, the N-terminus FBXW7α antibody also detected the 110kDa product. Notably, the C-terminus antibody did not recognize any of the isoforms but continued to detect a 64kDa band in all samples, including the non-transfected controls. Immunoprecipitation confirmed this lack of specificity and the inability to detect overexpressed or endogenous FBXW7α in HEK293A cells and RMEC. A commonly used C-terminus FBXW7 antibody does not detect FBXW7 under standard western blotting conditions.


Subject(s)
F-Box Proteins , F-Box Proteins/genetics , F-Box Proteins/metabolism , Endothelial Cells/metabolism , F-Box-WD Repeat-Containing Protein 7/genetics , F-Box-WD Repeat-Containing Protein 7/metabolism , Cell Cycle/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Antibodies
4.
Sci Rep ; 13(1): 21292, 2023 12 02.
Article in English | MEDLINE | ID: mdl-38042898

ABSTRACT

Dysregulation of nitric oxide (NO) production can cause ischaemic retinal injury and result in blindness. How this dysregulation occurs is poorly understood but thought to be due to an impairment in NO synthase function (NOS) and nitro-oxidative stress. Here we investigated the possibility of correcting this defective NOS activity by supplementation with the cofactor tetrahydrobiopterin, BH4. Retinal ischaemia was examined using the oxygen-induced retinopathy model and BH4 deficient Hph-1 mice used to establish the relationship between NOS activity and BH4. Mice were treated with the stable BH4 precursor sepiapterin at the onset of hypoxia and their retinas assessed 48 h later. HPLC analysis confirmed elevated BH4 levels in all sepiapterin supplemented groups and increased NOS activity. Sepiapterin treatment caused a significant decrease in neuronal cell death in the inner nuclear layer that was most notable in WT animals and was associated with significantly diminished superoxide and local peroxynitrite formation. Interestingly, sepiapterin also increased inflammatory cytokine levels but not microglia cell number. BH4 supplementation by sepiapterin improved both redox state and neuronal survival during retinal ischaemia, in spite of a paradoxical increase in inflammatory cytokines. This implicates nitro-oxidative stress in retinal neurones as the cytotoxic element in ischaemia, rather than enhanced pro-inflammatory signalling.


Subject(s)
Biopterins , Retinal Diseases , Mice , Animals , Biopterins/metabolism , Retinal Diseases/drug therapy , Nitric Oxide/metabolism , Cell Death , Dietary Supplements , Ischemia/drug therapy
5.
Article in English | MEDLINE | ID: mdl-38983523

ABSTRACT

Recent studies have provided novel insights of co-development of the neural and vascular elements of the retina. Knowledge of these relationships are crucial to understand the impact of therapeutic measures in Retinopathy of Prematurity (ROP). ROP is imposed by therapeutic oxygen upon immature retinal blood vessels and neural cells causing delayed development and vascular regression. However, the impact of hyperoxia on developing retinal neurons is less understood because some aspects of normal development remain unknown. The metabolic changes during differentiation of retinal progenitor cells to functional neurons is one such aspect. We correlated immunomarkers of hypoxia with markers of metabolic change in developing retinal neurons during the early postnatal period in mice. The same marker proteins were studied in secondary lens fiber differentiation at postnatal day-3 (P3). Nuclear localization of the oxygen-sensitive subunits of hypoxia inducible factor, HIF-1α and HIF-2α was correlated with increasing mitochondrial content in differentiating neurons. Nuclear HIF was also correlated with AMP-dependent protein kinase (AMPK), and the AMPK phosphorylation target PPAR-gamma coactivator-1alpha (PGC-1α), the principal regulator of mitochondrial biogenesis. Expression of AMPK, PGC1α and HIF-2α in secondary fiber differentiation was visible in each profile of the lens equator. Strong nuclear localization for all markers was present at the onset of secondary fiber differentiation, and reflected changes in size, mitochondrial content, and metabolism. We speculate that the 'physiological hypoxia' that drives retinal vascular development is cell-specific and reliant upon neuronal differentiation and mitochondrial biogenesis. We suggest that the onset of differentiation increases energy consumption that is detected by AMPK. In turn AMPK increases mitochondrial biogenesis via PGC-1α. Mitochondrial oxygen consumption may then create intracellular hypoxia that activates HIF. This progression is congruent with the expression of these markers in secondary lens fiber differentiation and nuclear localization of HIF-2α. Nuclear localization of HIF-1α and HIF-2α in the postnatal retina is less defined than in the lens as it may involve the remnant of HIF expression from the embryonic period that is sustained and increased by intracellular hypoxia caused by increasing mitochondrial oxygen consumption. This the first report of the involvement of HIF-2α, AMPK and PGC-1α in lens development.

6.
Cardiovasc Res ; 116(2): 393-405, 2020 02 01.
Article in English | MEDLINE | ID: mdl-30937452

ABSTRACT

AIMS: Cord blood-derived endothelial colony-forming cells (CB-ECFCs) are a defined progenitor population with established roles in vascular homeostasis and angiogenesis, which possess low immunogenicity and high potential for allogeneic therapy and are highly sensitive to regulation by reactive oxygen species (ROS). The aim of this study was to define the precise role of the major ROS-producing enzyme, NOX4 NADPH oxidase, in CB-ECFC vasoreparative function. METHODS AND RESULTS: In vitro CB-ECFC migration (scratch-wound assay) and tubulogenesis (tube length, branch number) was enhanced by phorbol 12-myristate 13-acetate (PMA)-induced superoxide in a NOX-dependent manner. CB-ECFCs highly-expressed NOX4, which was further induced by PMA, whilst NOX4 siRNA and plasmid overexpression reduced and potentiated in vitro function, respectively. Increased ROS generation in NOX4-overexpressing CB-ECFCs (DCF fluorescence, flow cytometry) was specifically reduced by superoxide dismutase, highlighting induction of ROS-specific signalling. Laser Doppler imaging of mouse ischaemic hindlimbs at 7 days indicated that NOX4-knockdown CB-ECFCs inhibited blood flow recovery, which was enhanced by NOX4-overexpressing CB-ECFCs. Tissue analysis at 14 days revealed consistent alterations in vascular density (lectin expression) and eNOS protein despite clearance of injected CB-ECFCs, suggesting NOX4-mediated modulation of host tissue. Indeed, proteome array analysis indicated that NOX4-knockdown CB-ECFCs largely suppressed tissue angiogenesis, whilst NOX4-overexpressing CB-ECFCs up-regulated a number of pro-angiogenic factors specifically-linked with eNOS signalling, in parallel with equivalent modulation of NOX-dependent ROS generation, suggesting that CB-ECFC NOX4 signalling may promote host vascular repair. CONCLUSION: Taken together, these findings indicate a key role for NOX4 in CB-ECFCs, thereby highlighting its potential as a target for enhancing their reparative function through therapeutic priming to support creation of a pro-reparative microenvironment and effective post-ischaemic revascularization.


Subject(s)
Endothelial Progenitor Cells/transplantation , Ischemia/surgery , Muscle, Skeletal/blood supply , NADPH Oxidase 4/metabolism , Neovascularization, Physiologic , Animals , Cell Movement , Cells, Cultured , Cellular Microenvironment , Disease Models, Animal , Endothelial Progenitor Cells/enzymology , Fetal Blood/cytology , Hindlimb , Humans , Ischemia/enzymology , Ischemia/genetics , Ischemia/physiopathology , Mice, Inbred NOD , NADPH Oxidase 4/genetics , Reactive Oxygen Species/metabolism , Recovery of Function , Signal Transduction
7.
Mol Vis ; 15: 1509-20, 2009 Aug 05.
Article in English | MEDLINE | ID: mdl-19668595

ABSTRACT

PURPOSE: Advanced glycation endproduct (AGE) formation on the basement membrane of retinal capillaries has been previously described but the impact of these adducts on capillary endothelial cell function vascular repair remains uncertain. This investigation has evaluated retinal microvascular endothelial cells (RMECs) growing on AGE-modified fibronectin (FN) and determined how this has an impact on cell-substrate interactions and downstream oxidative responses and cell survival. METHODS: RMECs were grown on methylglyoxal-modified FN (AGE-FN) or native FN as a control. RMEC attachment and spreading was quantified. In a separate treatment, the AGE-FN substrate had Arg-Gly-Asp-Ser (RGDS) or scrambled peptide added before seeding. Phosphorylation of focal adhesion kinase (FAK) and alpha5beta1 integrin localization was assessed and apoptosis evaluated. In a subset of RMECs that remained attached to the AGE-FN substrate, the production of superoxide (O(2) (-)) was assayed using dihydroethidium (DHE) fluorescence or lucigenin, in the presence or absence of NADPH. The specificity of the O(2) (-) assays was confirmed by inhibition in the presence of polyethylene-glycol-superoxide dismutase (PEG-SOD). AGE-mediated changes to mRNAs encoding key basement membrane proteins and regulatory enzymes were investigated using real-time RT-PCR. RESULTS: AGE-FN reduced RMEC attachment and spreading when compared to FN controls (p<0.001). RGDS peptide enhanced cell attachment on AGE-FN (p<0.001), while the scrambled peptide had no effect. FAK phosphorylation in AGE-exposed RMECs was reduced in a time-dependent fashion, while alpha5beta1 integrin-immunoreactivity became focal at the basal membrane. AGE-exposure induced apoptosis, a response significantly prevented by RGDS peptide. AGE-exposure caused a significant increase in basal O(2) (-) and NADPH-stimulated production by RMECs (p<0.01), while AGE-FN also increased basement membrane associated mRNA expression (p<0.05). CONCLUSIONS: AGE substrate modifications impair the function of retinal capillary endothelium and their reparative potential in response to diabetes-related insults. Arginine-specific modifications alter vital endothelial cell interactions with the substrate. This phenomenon could play an important role in dysfunction and nonperfusion of retinal capillaries during diabetes.


Subject(s)
Endothelial Cells/pathology , Glycation End Products, Advanced/pharmacology , Microvessels/pathology , Oligopeptides/pharmacology , Retinal Vessels/pathology , Animals , Basement Membrane/drug effects , Caspase 3/metabolism , Cattle , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Enzyme Activation/drug effects , Fibronectins/pharmacology , Gene Expression Regulation/drug effects , Microvessels/drug effects , Microvessels/enzymology , Mitochondria/drug effects , Mitochondria/enzymology , Permeability/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retinal Vessels/drug effects , Retinal Vessels/enzymology , Signal Transduction/drug effects , Superoxides/metabolism
8.
Invest Ophthalmol Vis Sci ; 58(1): 230-241, 2017 01 01.
Article in English | MEDLINE | ID: mdl-28114584

ABSTRACT

Purpose: Endothelial nitric oxide synthase (eNOS)-derived nitric oxide (NO) has important vasoprotective functions that are compromised in the vasodegenerative phase of retinopathy of prematurity, owing to hyperoxia-induced depletion of the essential NOS cofactor BH4. Because modulating eNOS function can be beneficial or detrimental, our aim was to investigate the effect of BH4 supplementation on eNOS function and vascular regression in hyperoxia. Methods: Endothelial-specific eNOS-green fluorescent protein (GFP) overexpressing mice at postnatal day 7 (P7) were exposed to hyperoxia for 48 hours in the presence or absence of supplemental BH4, achieved by administration of sepiapterin, a stable BH4 precursor. Tissue was collected either for retinal flat mounts that were stained with lectin to determine the extent of vessel coverage or for analysis of BH4 by high-performance liquid chromatography, nitrotyrosine (NT) marker by Western blotting, VEGF expression by ELISA, and NOS activity by arginine-to-citrulline conversion. Primary retinal microvascular endothelial cells (RMEC) were similarly treated, and hyperoxia-induced damage was determined. Results: Sepiapterin effectively enhanced BH4 levels in hyperoxia-exposed retinas and brains, elevated NOS activity, and reduced NT-modified protein, leading to reversal of the exacerbated vasoregression observed in the presence of eNOS overexpression. In RMECs, hyperoxia-mediated depletion of BH4 dysregulated the redox balance by reducing nitrite and elevating superoxide and impaired proliferative ability. BH4 supplementation restored normal RMEC proliferation in vitro and also in vivo, providing a mechanistic link with the enhanced vascular coverage in eNOS-GFP retinas. Conclusions: These results demonstrate that BH4 supplementation corrects hyperoxia-induced RMEC dysfunction and preserves vascular integrity by enhancing eNOS function.


Subject(s)
Biopterins/analogs & derivatives , Endothelium, Vascular/enzymology , Hyperoxia/prevention & control , Nitric Oxide Synthase Type III/metabolism , Retinal Vessels/enzymology , Retinopathy of Prematurity/prevention & control , Animals , Animals, Newborn , Biopterins/pharmacology , Blotting, Western , Cells, Cultured , Disease Models, Animal , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Enzyme-Linked Immunosorbent Assay , Humans , Hyperoxia/complications , Hyperoxia/metabolism , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type III/drug effects , Retinal Vessels/drug effects , Retinal Vessels/pathology , Retinopathy of Prematurity/etiology , Retinopathy of Prematurity/pathology
9.
Int J Nanomedicine ; 10: 7183-96, 2015.
Article in English | MEDLINE | ID: mdl-26648722

ABSTRACT

We report the formulation of novel composite nanoparticles that combine the high transfection efficiency of cationic peptide-DNA nanoparticles with the biocompatibility and prolonged delivery of polylactic acid-polyethylene glycol (PLA-PEG). The cationic cell-penetrating peptide RALA was used to condense DNA into nanoparticles that were encapsulated within a range of PLA-PEG copolymers. The composite nanoparticles produced exhibited excellent physicochemical properties including size <200 nm and encapsulation efficiency >80%. Images of the composite nanoparticles obtained with a new transmission electron microscopy staining method revealed the peptide-DNA nanoparticles within the PLA-PEG matrix. Varying the copolymers modulated the DNA release rate >6 weeks in vitro. The best formulation was selected and was able to transfect cells while maintaining viability. The effect of transferrin-appended composite nanoparticles was also studied. Thus, we have demonstrated the manufacture of composite nanoparticles for the controlled delivery of DNA.


Subject(s)
Gene Transfer Techniques , Nanoparticles/chemistry , Peptides/chemistry , Polyethylene Glycols/chemistry , Cations , Cell Line, Tumor , Cell Survival , Cell-Penetrating Peptides/chemistry , DNA/metabolism , Electrophoretic Mobility Shift Assay , Humans , Nanoparticles/ultrastructure , Particle Size , Temperature , Transfection
10.
Pharmacogenetics ; 14(12): 831-9, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15608562

ABSTRACT

The G894T endothelial nitric oxide synthase (eNOS) polymorphism results in a Glu to Asp substitution at position 298. This position is located externally on the protein and as the regulation of eNOS is dependent on its subcellular localization and interaction with modulatory proteins, we aimed to address whether the substitution of Asp at 298 had any effect on these mechanisms. Initially, we developed a novel method to accurately determine molar quantities of each variant by expressing them as green fluorescent protein (GFP) fusion proteins and using recombinant adenoviruses to facilitate transient infection of human microvascular endothelial cells. Sodium dodecyl sulphate-polyacrylamide gel electrophoresis and Western blotting of eNOSAsp revealed a 135-kDa proteolytic fragment which was not present with eNOSGlu. This proteolysis was prevented by using LDS buffer confirming that this differential cleavage is an artefact of sample preparation and unlikely to occur intracellularly. Nitric oxide was measured following stimulation with calcium ionophore or oestrogen in the presence of varying sepiapterin concentrations. GFP fluorescence was used to quantify the amount of fusion protein and calculate intracellular specific activity. There was no significant difference in intracellular specific activity between Glu and Asp eNOS in response to calcium ionophore or oestrogen. Tetrahydrobiopterin supplementation increased eNOS activity of both variants in an identical manner. The presence of the GFP also facilitated the visualization of the variants by confocal microscopy and demonstrated that both localized to the plasma membrane and the Golgi. These findings demonstrate that the Asp substitution at 298 does not have a major effect in modulating eNOS activity in vivo.


Subject(s)
Amino Acid Substitution , Endothelium, Vascular/metabolism , Genetic Variation , Nitric Oxide Synthase/genetics , Polymorphism, Genetic , Adenoviridae/genetics , Aspartic Acid/metabolism , Blotting, Western , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Green Fluorescent Proteins , Humans , Luminescent Proteins/metabolism , Microscopy, Confocal , Mutagenesis, Site-Directed , Nitric Oxide/analysis , Nitric Oxide Synthase/chemistry , Recombinant Fusion Proteins/metabolism , Structure-Activity Relationship
11.
J Pharm Biomed Anal ; 100: 236-242, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25171486

ABSTRACT

Gene therapy has the potential to provide safe and targeted therapies for a variety of diseases. A range of intracellular gene delivery vehicles have been proposed for this purpose. Non-viral vectors are a particularly attractive option and among them cationic peptides have emerged as promising candidates. For the pharmaceutical formulation and application to clinical studies it is necessary to quantify the amount of pDNA condensed with the delivery system. There is a severe deficiency in this area, thus far no methods have been reported specifically for pDNA condensed with cationic peptide to form nanoparticles. The current study seeks to address this and describes the evaluation of a range of disruption agents to extract DNA from nanoparticles formed by condensation with cationic fusogenic peptides RALA and KALA. Only proteinase K exhibited efficient and reproducible results and compatibility with the PicoGreen reagent based quantification assay. Thus we report for the first time a simple and reliable method that can quantify the pDNA content in pDNA cationic peptide nanoparticles.


Subject(s)
DNA-Binding Proteins/chemistry , DNA/isolation & purification , Gene Transfer Techniques , Nanoparticles , Peptides/chemistry , Cations , DNA/chemistry , Endopeptidase K/chemistry , Fluorescent Dyes/chemistry , Nucleic Acid Conformation , Organic Chemicals/chemistry , Particle Size , Protein Conformation , Sodium Dodecyl Sulfate/chemistry , Temperature , Time Factors
12.
Diabetes Care ; 36(4): 998-1005, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23238663

ABSTRACT

OBJECTIVE: Patients with type 2 diabetes mellitus (T2DM) are at increased risk of developing cardiovascular disease, largely as a result of defective production of cardioprotective nitric oxide and a concomitant rise in oxidative stress. Dietary interventions that could reverse this trend would be extremely beneficial. Here we investigated whether dietary n-3 polyunsaturated fatty acid (n-3 PUFA) supplementation positively affected platelet nitroso-redox imbalance. RESEARCH DESIGN AND METHODS: We randomized hypertensive T2DM patients (T2DM HT; n = 22) and age-and-sex matched hypertensive study participants without diabetes (HT alone; n = 23) in a double-blind, crossover fashion to receive 8 weeks of n-3 PUFAs (1.8 g eicosapentaenoic acid and 1.5 g docosahexaenoic acid) or identical olive oil capsules (placebo), with an intervening 8-week washout period. Platelet nitrite and superoxide were measured and compared before and after treatment; 8-isoprostane was determined by ELISA and subcellular compartmentalization of the NAD(P)H oxidase subunit p47-phox examined by Western blotting. RESULTS: The n-3 PUFA supplementation reduced 8-isoprostane and superoxide levels in platelets from T2DM HT, but not HT alone, participants, without effect on nitrite production. This coincided with a significant decrease in p47-phox membrane localization and a similar reduction in superoxide to that achieved with apocynin. At baseline, a subcohort of T2DM HT and HT alone participants showed evidence of nitric oxide synthase (NOS)-derived superoxide production, indicating defective enzymatic activity. This was reversed significantly in T2DM HT participants after treatment, demonstrating improved NOS function. CONCLUSIONS: Our finding that n-3 PUFAs diminish platelet superoxide production in T2DM HT patients in vivo suggests a therapeutic role for these agents in reducing the vascular-derived oxidative stress associated with diabetes.


Subject(s)
Blood Platelets/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/diet therapy , Fatty Acids, Omega-3/therapeutic use , Hypertension/blood , Hypertension/diet therapy , Aged , Cross-Over Studies , Dinoprost/analogs & derivatives , Dinoprost/metabolism , Double-Blind Method , Female , Humans , Male , Middle Aged , Oxidation-Reduction , Superoxides/metabolism
13.
Invest Ophthalmol Vis Sci ; 53(11): 6833-50, 2012 Oct 03.
Article in English | MEDLINE | ID: mdl-22930723

ABSTRACT

PURPOSE: In ischemic retinopathies, the misdirection of reparative angiogenesis away from the hypoxic retina leads to pathologic neovascularization. Thus, therapeutic strategies that reverse this trend would be extremely beneficial. Nitric oxide (NO) produced by endothelial nitric oxide synthase (eNOS) is an important mediator of vascular endothelial growth factor (VEGF) function facilitating vascular growth and maturation. However, in addition to NO, eNOS can also produce superoxide (O(2)(-)), exacerbating pathology. Here, our aim was to investigate the effect of eNOS overexpression on vascular closure and subsequent recovery of the ischemic retina. METHODS: Mice overexpressing eNOS-GFP were subjected to oxygen-induced retinopathy (OIR) and changes in retinal vascularization quantified. Background angiogenic drive was assessed during vascular development and in aortic rings. NOS activity was measured by Griess assay or conversion of radiolabeled arginine to citrulline, nitrotyrosine (NT), and superoxide by immunolabeling and dihydroethidium fluorescence and VEGF by ELISA. RESULTS: In response to hyperoxia, enhanced eNOS expression led to increased NOS-derived superoxide and dysfunctional NO production, NT accumulation, and exacerbated vessel closure associated with tetrahydrobiopterin (BH4) insufficiency. Despite worse vaso-obliteration, eNOS overexpression resulted in elevated hypoxia-induced angiogenic drive, independent of VEGF production. This correlated with increased vascular branching similar to that observed in isolated aortas and during development. Enhanced recovery was also associated with neovascular tuft formation, which showed defective NO production and increased eNOS-derived superoxide and NT levels. CONCLUSIONS: In hyperoxia, reduced BH4 bioavailability causes overexpressed eNOS to become dysfunctional, exacerbating vaso-obliteration. In the proliferative phase, however, eNOS has important prorepair functions enhancing angiogenic growth potential and recovery in ischemia.


Subject(s)
Nitric Oxide Synthase Type III/biosynthesis , Retinal Neovascularization/enzymology , Retinal Vessels/enzymology , Animals , Blotting, Western , Cell Proliferation , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Male , Mice , Mice, Inbred C57BL , Retinal Neovascularization/pathology , Retinal Vessels/pathology
14.
Br J Pharmacol ; 166(1): 327-38, 2012 May.
Article in English | MEDLINE | ID: mdl-22035179

ABSTRACT

BACKGROUND AND PURPOSE: Obestatin is a recently discovered gastrointestinal peptide with established metabolic actions, which is linked to diabetes and may exert cardiovascular benefits. Here we aimed to investigate the specific effects of obestatin on vascular relaxation. EXPERIMENTAL APPROACH: Cumulative relaxation responses to obestatin peptides were assessed in rat isolated aorta and mesenteric artery (n≥ 8) in the presence and absence of selective inhibitors. Complementary studies were performed in cultured bovine aortic endothelial cells (BAEC). KEY RESULTS: Obestatin peptides elicited concentration-dependent relaxation in both aorta and mesenteric artery. Responses to full-length obestatin(1-23) were greater than those to obestatin(1-10) and obestatin(11-23). Obestatin(1-23)-induced relaxation was attenuated by endothelial denudation, l-NAME (NOS inhibitor), high extracellular K(+) , GDP-ß-S (G-protein inhibitor), MDL-12,330A (adenylate cyclase inhibitor), wortmannin (PI3K inhibitor), KN-93 (CaMKII inhibitor), ODQ (guanylate cyclase inhibitor) and iberiotoxin (BK(Ca) blocker), suggesting that it is mediated by an endothelium-dependent NO signalling cascade involving an adenylate cyclase-linked GPCR, PI3K/PKB, Ca(2+) -dependent eNOS activation, soluble guanylate cyclase and modulation of vascular smooth muscle K(+) . Supporting data from BAEC indicated that nitrite production, intracellular Ca(2+) and PKB phosphorylation were increased after exposure to obestatin(1-23). Relaxations to obestatin(1-23) were unaltered by inhibitors of candidate endothelium-derived hyperpolarizing factors (EDHFs) and combined SK(Ca) /IK(Ca) blockade, suggesting that EDHF-mediated pathways were not involved. CONCLUSIONS AND IMPLICATIONS: Obestatin produces significant vascular relaxation via specific activation of endothelium-dependent NO signalling. These actions may be important in normal regulation of vascular function and are clearly relevant to diabetes, a condition characterized by endothelial dysfunction and cardiovascular complications.


Subject(s)
Endothelium, Vascular/drug effects , Nitric Oxide/metabolism , Peptide Hormones/metabolism , Vasodilation/drug effects , Animals , Aorta/drug effects , Aorta/metabolism , Calcium/metabolism , Cattle , Cells, Cultured , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/metabolism , Peptide Hormones/administration & dosage , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
15.
Invest Ophthalmol Vis Sci ; 51(6): 3291-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20107169

ABSTRACT

PURPOSE: Neovascularization occurs in response to tissue ischemia and growth factor stimulation. In ischemic retinopathies, however, new vessels fail to restore the hypoxic tissue; instead, they infiltrate the transparent vitreous. In a model of oxygen-induced retinopathy (OIR), TNFalpha and iNOS, upregulated in response to tissue ischemia, are cytotoxic and inhibit vascular repair. The aim of this study was to investigate the mechanism for this effect. METHODS: Wild-type C57/BL6 (WT) and TNFalpha(-/-) mice were subjected to OIR by exposure to 75% oxygen (postnatal days 7-12). The retinas were removed during the hypoxic phase of the model. Retinal cell death was determined by TUNEL staining, and the microglial cells were quantified after Z-series capture with a confocal microscope. In situ peroxynitrite and superoxide were measured by using the fluorescent dyes DCF and DHE. iNOS, nitrotyrosine, and arginase were analyzed by real-time PCR, Western blot analysis, and activity determined by radiolabeled arginine conversion. Astrocyte coverage was examined after GFAP immunostaining. RESULTS: The TNFalpha(-/-) animals displayed a significant reduction in TUNEL-positive apoptotic cells in the inner nuclear layer of the avascular retina compared with that in the WT control mice. The reduction coincided with enhanced astrocytic survival and an increase in microglial cells actively engaged in phagocytosing apoptotic debris that displayed low ROS, RNS, and NO production and high arginase activity. CONCLUSIONS: Collectively, the results suggest that improved vascular recovery in the absence of TNFalpha is associated with enhanced astrocyte survival and that both phenomena are dependent on preservation of microglial cells that display an anti-inflammatory phenotype during the early ischemic phase of OIR.


Subject(s)
Ischemia/metabolism , Microglia/cytology , Oxidative Stress , Retinal Diseases/metabolism , Retinal Neurons/pathology , Retinal Vessels/metabolism , Tumor Necrosis Factor-alpha/physiology , Animals , Apoptosis , Arginase/metabolism , Blotting, Western , Cell Count , Cell Death , Cell Survival , In Situ Nick-End Labeling , Ischemia/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/metabolism , Nitrosation , Oxygen/toxicity , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Retinal Diseases/pathology , Reverse Transcriptase Polymerase Chain Reaction , Tyrosine/analogs & derivatives , Tyrosine/metabolism
16.
Invest Ophthalmol Vis Sci ; 51(12): 6815-25, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20702831

ABSTRACT

PURPOSE: Disturbances to the cellular production of nitric oxide (NO) and superoxide (O(2)(-)) can have deleterious effects on retinal vascular integrity and angiogenic signaling. Dietary agents that could modulate the production of these signaling molecules from their likely enzymatic sources, endothelial nitric oxide synthase (eNOS) and NADPH oxidase, would therefore have a major beneficial effect on retinal vascular disease. The effect of ω-3 polyunsaturated fatty acids (PUFAs) on angiogenic signaling and NO/superoxide production in retinal microvascular endothelial cells (RMECs) was investigated. METHODS: Primary RMECs were treated with docosahexaenoic acid (DHA) or eicosapentaenoic acid (EPA) for 48 hours. RMEC migration was determined by scratch-wound assay, proliferation by the incorporation of BrdU, and angiogenic sprouting using a three-dimensional model of in vitro angiogenesis. NO production was quantified by Griess assay, and phospho-eNOS accumulation and superoxide were measured using the fluorescent probe dihydroethidine. eNOS localization to caveolin-rich microdomains was determined by Western blot analysis after subfractionation on a linear sucrose gradient. RESULTS: DHA treatment increased nitrite and decreased superoxide production, which correlated with the displacement of eNOS from caveolar subdomains and colocalization with the negative regulator caveolin-1. In addition, both ω-3 PUFAs demonstrated reduced responsiveness to VEGF-stimulated superoxide and nitrite release and significantly impaired endothelial wound healing, proliferation, and angiogenic sprout formation. CONCLUSIONS: DHA improves NO bioavailability, decreases O(2)(-) production, and blunts VEGF-mediated angiogenic signaling. These findings suggest a role for ω-3 PUFAs, particularly DHA, in maintaining vascular integrity while reducing pathologic retinal neovascularization.


Subject(s)
Docosahexaenoic Acids/pharmacology , Endothelium, Vascular/drug effects , Neovascularization, Pathologic/prevention & control , Nitric Oxide/metabolism , Signal Transduction/drug effects , Superoxides/metabolism , Vascular Endothelial Growth Factor A/toxicity , Animals , Apoptosis , Blotting, Western , Cattle , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Eicosapentaenoic Acid/pharmacology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Fluorescent Dyes , In Situ Nick-End Labeling , Neovascularization, Pathologic/pathology , Nitric Oxide Synthase Type III/metabolism , Nitrosation , Oxidation-Reduction , Retinal Vessels/cytology , Wound Healing/drug effects
17.
Am J Pathol ; 166(2): 637-44, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15681845

ABSTRACT

The present study was undertaken to test whether inhibition of the proangiogenic inflammatory cytokine tumor necrosis factor (TNF)-alpha can modulate retinal hypoxia and preretinal neovascularization in a murine model of oxygen-induced retinopathy (OIR). OIR was produced in TNF-alpha-/- and wild-type (WT) control C57B6 neonatal mice by exposure to 75% oxygen between postnatal days 7 and 12 (P7 to P12). Half of each WT litter was treated with the cytokine inhibitor semapimod (formerly known as CNI-1493) (5 mg/kg) by daily intraperitoneal injection from the time of reintroduction to room air at P12 until P17. The extent of preretinal neovascularization and intraretinal revascularization was quantified by image analysis of retinal flat-mounts and retinal hypoxia correlated with vascularization by immunofluorescent localization of the hypoxia-sensitive drug pimonidazole (hypoxyprobe, HP). HP adducts were also characterized by Western analysis and quantified by competitive enzyme-linked immunosorbent assay. TNF-alpha-/- and WT mice showed a similar sensitivity to hyperoxia-induced retinal ischemia at P12. At P13 some delay in early reperfusion was evident in TNF-alpha-/- and WT mice treated with semapimod. However, at P17 both these groups had significantly better vascular recovery with less ischemic/hypoxic retina and preretinal neovascularization compared to untreated retinopathy in WT mice. Immunohistochemistry showed deposition of HP in the avascular inner retina but not in areas underlying preretinal neovascularization, indicating that such aberrant vasculature can reduce retinal hypoxia. Inhibition of TNF-alpha significantly improves vascular recovery within ischemic tissue and reduces pathological neovascularization in OIR. HP provides a useful tool for mapping and quantifying tissue hypoxia in experimental ischemic retinopathy.


Subject(s)
Ischemia , Neovascularization, Pathologic , Retinal Diseases/pathology , Retinal Neovascularization , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Hydrazones/pharmacology , Hyperoxia , Hypoxia , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Retina/pathology , Retinal Vessels/pathology , Time Factors , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL