Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
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
2.
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.

3.
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
4.
Invest Ophthalmol Vis Sci ; 56(13): 8008-18, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26720448

ABSTRACT

PURPOSE: It is widely held that neurons of the central nervous system do not store glycogen and that accumulation of the polysaccharide may cause neurodegeneration. Since primary neural injury occurs in diabetic retinopathy, we examined neuronal glycogen status in the retina of streptozotocin-induced diabetic and control rats. METHODS: Glycogen was localized in eyes of streptozotocin-induced diabetic and control rats using light microscopic histochemistry and electron microscopy, and correlated with immunohistochemical staining for glycogen phosphorylase and phosphorylated glycogen synthase (pGS). RESULTS: Electron microscopy of 2-month-old diabetic rats (n = 6) showed massive accumulations of glycogen in the perinuclear cytoplasm of many amacrine neurons. In 4-month-old diabetic rats (n = 11), quantification of glycogen-engorged amacrine cells showed a mean of 26 cells/mm of central retina (SD ± 5), compared to 0.5 (SD ± 0.2) in controls (n = 8). Immunohistochemical staining for glycogen phosphorylase revealed strong expression in amacrine and ganglion cells of control retina, and increased staining in cell processes of the inner plexiform layer in diabetic retina. In control retina, the inactive pGS was consistently sequestered within the cell nuclei of all retinal neurons and the retinal pigment epithelium (RPE), but in diabetics nuclear pGS was reduced or lost in all classes of retinal cell except the ganglion cells and cone photoreceptors. CONCLUSIONS: The present study identifies a large population of retinal neurons that normally utilize glycogen metabolism but show pathologic storage of the polysaccharide during uncontrolled diabetes.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Glycogen/metabolism , Retinal Neurons/metabolism , Amacrine Cells/metabolism , Animals , Cytoplasm/metabolism , Glycogen Phosphorylase/metabolism , Immunohistochemistry , Male , Microscopy/methods , Rats
5.
Invest Ophthalmol Vis Sci ; 55(4): 2157-65, 2014 Apr 07.
Article in English | MEDLINE | ID: mdl-24609622

ABSTRACT

PURPOSE: This study tested the role of K(+) and Cl(-) channels in the regulation of retinal blood flow. METHODS: Studies were carried out in adult Male Hooded Lister rats. Selectivity of ion-channel blockers was established using electrophysiological recordings from smooth muscle in isolated arterioles under voltage clamp conditions. Leukocyte velocity and retinal arteriolar diameter were measured in anesthetized animals using leukocyte fluorography and fluorescein angiography imaging with a confocal scanning laser ophthalmoscope. These values were used to estimate volumetric flow, which was compared between control conditions and following intravitreal injections of ion channel blockers, either alone or in combination with the potent vasoconstrictor Endothelin 1 (Et1). RESULTS: Voltage-activated K(+) current (IKv) was inhibited by correolide, large conductance (BK) Ca(2+)-activated K(+) current (IKCa) by Penitrem A, and Ca(2+)-activated Cl(-) current (IClCa) by disodium 4,4'-diisothiocyanatostilbene-2,2'-disulfonate (DIDS). Intravitreal injections (10 µL) of DIDS (estimated intraocular concentration 10 mM) increased flow by 22%, whereas the BK-blockers Penitrem A (1 µM) and iberiotoxin (4 µM), and the IKv-inhibitor correolide (40 µM) all decreased resting flow by approximately 10%. Endothelin 1 (104 nM) reduced flow by approximately 65%. This effect was completely reversed by DIDS, but was unaffected by Penitrem A, iberiotoxin, or correolide. CONCLUSIONS: These results suggest that Cl(-) channels in retinal arteriolar smooth muscle limit resting blood flow and play an obligatory role in Et1 responses. K(+)-channel activity promotes basal flow but exerts little modifying effect on the Et1 response. Cl(-) channels may be appropriate molecular targets in retinal pathologies characterized by increased Et1 activity and reduced blood flow.


Subject(s)
Arterioles/physiology , Blood Flow Velocity/physiology , Chloride Channels/metabolism , Potassium Channels/metabolism , Retinal Artery/physiology , Animals , Fluorescein Angiography , Fundus Oculi , Male , Muscle, Smooth, Vascular/physiology , Ophthalmoscopy , Patch-Clamp Techniques , Rats
6.
Invest Ophthalmol Vis Sci ; 53(6): 3059-66, 2012 May 17.
Article in English | MEDLINE | ID: mdl-22427579

ABSTRACT

PURPOSE: To investigate the role of feedback by Ca²âº-sensitive plasma-membrane ion channels in endothelin 1 (Et1) signaling in vitro and in vivo. Methods. Et1 responses were imaged from Fluo-4-loaded smooth muscle in isolated segments of rat retinal arteriole using two-dimensional (2-D) confocal laser microscopy. Vasoconstrictor responses to intravitreal injections of Et1 were recorded in the absence and presence of appropriate ion channel blockers using fluorescein angiograms imaged using a confocal scanning laser ophthalmoscope. Results. Et1 (10 nM) increased both basal [Ca²âº](i) and the amplitude and frequency of Ca²âº-waves in retinal arterioles. The Ca²âº-activated Cl⁻-channel blockers DIDS and 9-anthracene carboxylic acid (9AC) blocked Et1-induced increases in wave frequency, and 9AC also inhibited the increase in amplitude. Iberiotoxin, an inhibitor of large conductance (BK) Ca²âº-activated K⁺-channels, increased wave amplitude in the presence of Et1 but had no effect on frequency. None of these drugs affected basal [Ca²âº](i). The voltage-operated Ca²âº-channel inhibitor nimodipine inhibited wave frequency and amplitude and also lowered basal [Ca²âº](i) in the presence of Et1. Intravitreal injection of Et1 caused retinal arteriolar vasoconstriction. This was inhibited by DIDS but not by iberiotoxin or penitrem A, another BK-channel inhibitor. Conclusions. Et1 evokes increases in the frequency of arteriolar Ca²âº-waves in vitro, resulting in vasoconstriction in vivo. These responses, initiated by release of stored Ca²âº, also require positive feedback via Ca²âº-activated Cl⁻-channels and L-type Ca²âº-channels.


Subject(s)
Calcium/metabolism , Endothelin-1/pharmacology , Muscle, Smooth, Vascular/metabolism , Retinal Artery/metabolism , Signal Transduction , Animals , Arterioles/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels/physiology , Cell Membrane/metabolism , Fluorescein Angiography , In Vitro Techniques , Intravitreal Injections , Male , Microscopy, Confocal , Rats , Rats, Sprague-Dawley , Vasoconstriction
7.
Diabetes ; 60(11): 2995-3005, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21911748

ABSTRACT

OBJECTIVE: Erythropoietin (EPO) may be protective for early stage diabetic retinopathy, although there are concerns that it could exacerbate retinal angiogenesis and thrombosis. A peptide based on the EPO helix-B domain (helix B-surface peptide [pHBSP]) is nonerythrogenic but retains tissue-protective properties, and this study evaluates its therapeutic potential in diabetic retinopathy. RESEARCH DESIGN AND METHODS: After 6 months of streptozotocin-induced diabetes, rats (n = 12) and age-matched nondiabetic controls (n = 12) were evenly split into pHBSP and scrambled peptide groups and injected daily (10 µg/kg per day) for 1 month. The retina was investigated for glial dysfunction, microglial activation, and neuronal DNA damage. The vasculature was dual stained with isolectin and collagen IV. Retinal cytokine expression was quantified using real-time RT-PCR. In parallel, oxygen-induced retinopathy (OIR) was used to evaluate the effects of pHBSP on retinal ischemia and neovascularization (1-30 µg/kg pHBSP or control peptide). RESULTS: pHBSP or scrambled peptide treatment did not alter hematocrit. In the diabetic retina, Müller glial expression of glial fibrillary acidic protein was increased when compared with nondiabetic controls, but pHBSP significantly reduced this stress-related response (P < 0.001). CD11b+ microglia and proinflammatory cytokines were elevated in diabetic retina responses, and some of these responses were attenuated by pHBSP (P < 0.01-0.001). pHBSP significantly reduced diabetes-linked DNA damage as determined by 8-hydroxydeoxyguanosine and transferase-mediated dUTP nick-end labeling positivity and also prevented acellular capillary formation (P < 0.05). In OIR, pHBSP had no effect on preretinal neovascularization at any dose. CONCLUSIONS: Treatment with an EPO-derived peptide after diabetes is fully established can significantly protect against neuroglial and vascular degenerative pathology without altering hematocrit or exacerbating neovascularization. These findings have therapeutic implications for disorders such as diabetic retinopathy.


Subject(s)
Diabetic Retinopathy/drug therapy , Erythropoietin/chemistry , Nerve Degeneration/prevention & control , Neuroglia/drug effects , Peptide Fragments/therapeutic use , Retinal Degeneration/prevention & control , Retinal Vessels/drug effects , Animals , Animals, Newborn , Apoptosis/drug effects , Cytokines/genetics , Cytokines/metabolism , DNA Damage/drug effects , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Gene Expression Regulation/drug effects , Ischemia/drug therapy , Ischemia/metabolism , Ischemia/pathology , Male , Mice , Mice, Inbred C57BL , Neuroglia/pathology , Peptide Fragments/adverse effects , Peptide Fragments/chemistry , Protein Interaction Domains and Motifs , Random Allocation , Rats , Rats, Sprague-Dawley , Retina/drug effects , Retina/metabolism , Retina/pathology , Retinal Vessels/pathology
8.
Mol Med ; 17(9-10): 1045-55, 2011.
Article in English | MEDLINE | ID: mdl-21670847

ABSTRACT

Endothelial progenitor cells (EPCs) promote angiogenesis, and clinical trials have shown such cell therapy to be feasible for treating ischemic disease. However, clinical outcomes have been contradictory owing to the diverse range of EPC types used. We recently characterized two EPC subtypes, and identified outgrowth endothelial cells as the only EPC type with true progenitor and endothelial characteristics. By contrast, myeloid angiogenic cells (MACs) were shown to be monocytic cells without endothelial characteristics despite being widely described as "EPCs." In the current study we demonstrated that although MACs do not become endothelial cells or directly incorporate into a microvascular network, they can significantly induce endothelial tube formation in vitro and vascular repair in vivo. MAC-derived interleukin-8 (IL-8) was identified as a key paracrine factor, and blockade of IL-8 but not vascular endothelial growth factor (VEGF) prevented MAC-induced angiogenesis. Extracellular IL-8 transactivates VEGFR2 and induces phosphorylation of extracellular signal-regulated kinases. Further transcriptomic and immunophenotypic analysis indicates that MACs represent alternative activated M2 macrophages. Our findings demonstrate an unequivocal role for MACs in angiogenesis, which is linked to paracrine release of cytokines such as IL-8. We also show, for the first time, the true identity of these cells as alternative M2 macrophages with proangiogenic, antiinflammatory and pro-tissue-repair properties.


Subject(s)
Endothelial Cells/physiology , Interleukin-8/metabolism , Macrophages/physiology , Myeloid Cells/physiology , Neovascularization, Physiologic/physiology , Adult , Animals , Cattle , Cells, Cultured , Endothelial Cells/metabolism , Gene Expression Profiling/methods , Humans , Immunoblotting , Interleukin-8/genetics , Ischemia/physiopathology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Myeloid Cells/metabolism , Oligonucleotide Array Sequence Analysis , Proteomics/methods , Retinal Vessels/metabolism , Retinal Vessels/physiology , Stem Cells/metabolism , Stem Cells/physiology , Transcriptome , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
9.
Prog Retin Eye Res ; 30(3): 149-66, 2011 May.
Article in English | MEDLINE | ID: mdl-21352947

ABSTRACT

Retinal ischaemic disorders such as diabetic retinopathy and retinal vein occlusion are common. The hypoxia-related stimuli from oxygen-deprived neural and glial networks can drive expression of growth factors and cytokines which induce leakage from the surviving vasculature and/or pre-retinal and papillary neovascularisation. If left untreated, retinal vascular stasis, hypoxia or ischaemia can lead to macular oedema or fibro-vascular scar formation which are associated with severe visual impairment, and even blindness. Current therapies for ischaemic retinopathies include laser photocoagulation, injection of corticosteroids or VEGF-antibodies and vitreoretinal surgery, however they carry significant side effects. As an alternative approach, we propose that if reparative intra-retinal angiogenesis can be harnessed at the appropriate stage, ischaemia could be contained or reversed. This review provides evidence that reperfusion of ischaemic retina and suppression of sight-threatening sequelae is possible in both experimental and clinical settings. In particular, there is emphasis on the clinical potential for endothelial progenitor cells (EPCs) to promote vascular repair and reversal of ischaemic injury in various tissues including retina. Gathering evidence from an extensive published literature, we outline the molecular and phenotypic nature of EPCs, how they are altered in disease and provide a rationale for harnessing the vascular reparative properties of various cell sub-types. When some of the remaining questions surrounding the clinical use of EPCs are addressed, they may provide an exciting new therapeutic option for treating ischaemic retinopathies.


Subject(s)
Diabetic Retinopathy/therapy , Endothelium, Vascular/cytology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Reperfusion Injury/therapy , Retinal Vein Occlusion/therapy , Animals , Humans , Retinal Vessels/cytology
10.
Am J Pathol ; 178(4): 1517-28, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21435441

ABSTRACT

This study was conducted to determine the perivascular cell responses to increased endothelial cell expression of insulin-like growth factor binding protein-3 (IGFBP-3) in mouse retina. The contribution of bone marrow cells in the IGFBP-3-mediated response was examined using green fluorescent protein-positive (GFP(+)) adult chimeric mice subjected to laser-induced retinal vessel occlusion injury. Intravitreal injection of an endothelial-specific IGFBP-3-expressing plasmid resulted in increased differentiation of GFP(+) hematopoietic stem cells (HSCs) into pericytes and astrocytes as determined by immunohistochemical analysis. Administration of IGFBP-3 plasmid to mouse pups that underwent the oxygen-induced retinopathy model resulted in increased pericyte ensheathment and reduced pericyte apoptosis in the developing retina. Increased IGFBP-3 expression reduced the number of activated microglial cells and decreased apoptosis of neuronal cells in the oxygen-induced retinopathy model. In summary, IGFBP-3 increased differentiation of GFP(+) HSCs into pericytes and astrocytes while increasing vascular ensheathment of pericytes and decreasing apoptosis of pericytes and retinal neurons. All of these cytoprotective effects exhibited by IGFBP-3 overexpression can result in a more stable retinal vascular bed. Thus, endothelial expression of IGFBP-3 may represent a physiologic response to injury and may represent a therapeutic strategy for the treatment of ischemic vascular eye diseases, such as diabetic retinopathy and retinopathy of prematurity.


Subject(s)
Apoptosis , Insulin-Like Growth Factor Binding Protein 3/metabolism , Ischemia/pathology , Microglia/metabolism , Neurons/metabolism , Pericytes/metabolism , Retina/injuries , Animals , Astrocytes/cytology , Cell Death , Cell Differentiation , Eye Diseases/pathology , Female , Green Fluorescent Proteins/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence/methods , Pericytes/cytology , Retina/metabolism
11.
PLoS One ; 5(7): e11870, 2010 Jul 29.
Article in English | MEDLINE | ID: mdl-20686695

ABSTRACT

BACKGROUND: Erythropoiesis stimulating agents (ESAs) are widely used to treat anaemia but concerns exist about their potential to promote pathological angiogenesis in some clinical scenarios. In the current study we have assessed the angiogenic potential of three ESAs; epoetin delta, darbepoetin alfa and epoetin beta using in vitro and in vivo models. METHODOLOGY/PRINCIPAL FINDINGS: The epoetins induced angiogenesis in human microvascular endothelial cells at high doses, although darbepoetin alfa was pro-angiogenic at low-doses (1-20 IU/ml). ESA-induced angiogenesis was VEGF-mediated. In a mouse model of ischaemia-induced retinopathy, all ESAs induced generation of reticulocytes but only epoetin beta exacerbated pathological (pre-retinal) neovascularisation in comparison to controls (p<0.05). Only epoetin delta induced a significant revascularisation response which enhanced normality of the vasculature (p<0.05). This was associated with mobilisation of haematopoietic stem cells and their localisation to the retinal vasculature. Darbepoetin alfa also increased the number of active microglia in the ischaemic retina relative to other ESAs (p<0.05). Darbepoetin alfa induced retinal TNFalpha and VEGF mRNA expression which were up to 4 fold higher than with epoetin delta (p<0.001). CONCLUSIONS: This study has implications for treatment of patients as there are clear differences in the angiogenic potential of the different ESAs.


Subject(s)
Hematinics/therapeutic use , Ischemia/complications , Neovascularization, Pathologic/drug therapy , Retinal Diseases/drug therapy , Animals , Cells, Cultured , Chromatography, High Pressure Liquid , Disease Models, Animal , Erythropoietin/therapeutic use , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Neovascularization, Pathologic/etiology , Recombinant Proteins , Retina/pathology , Retina/ultrastructure , Retinal Diseases/etiology , Reverse Transcriptase Polymerase Chain Reaction
12.
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
13.
Invest Ophthalmol Vis Sci ; 51(11): 5906-13, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20554606

ABSTRACT

PURPOSE: Endothelial progenitor cells (EPCs) have potential for promoting vascular repair and revascularization of ischemic retina. However, the highly heterogeneous nature of these cells causes confusion when assessing their biological functions. The purpose of this study was to provide a comprehensive comparison between the two main EPC subtypes, early EPCs (eEPCs) and outgrowth endothelial cells (OECs), and to establish the potential of OECs as a novel cell therapy for ischemic retinopathy. METHODS: Two types of human blood-derived EPCs were isolated and compared using immunophenotyping and multiple in vitro functional assays to assess interaction with retinal capillary endothelial cells and angiogenic activity. OECs were delivered intravitreally in a mouse model of ischemic retinopathy, and flat mounted retinas were examined using confocal microscopy. RESULTS: These data indicate that eEPCs are hematopoietic cells with minimal proliferative capacity that lack tube-forming capacity. By contrast, OECs are committed to an endothelial lineage and have significant proliferative and de novo tubulogenic potential. Furthermore, only OECs are able to closely interact with endothelial cells through adherens and tight junctions and to integrate into retinal vascular networks in vitro. The authors subsequently chose OECs to test a novel cell therapy approach for ischemic retinopathy. Using a murine model of retinal ischemia, they demonstrated that OECs directly incorporate into the resident vasculature, significantly decreasing avascular areas, concomitantly increasing normovascular areas, and preventing pathologic preretinal neovascularization. CONCLUSIONS: As a distinct EPC population, OECs have potential as therapeutic cells to vascularize the ischemic retina.


Subject(s)
Disease Models, Animal , Endothelial Cells/cytology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Reperfusion Injury/therapy , Retinal Diseases/therapy , Animals , Biomarkers/metabolism , Capillaries/cytology , Cell Separation , Cells, Cultured , Coculture Techniques , Endothelial Cells/physiology , Flow Cytometry , Fluorescent Antibody Technique, Indirect , Hematopoietic Stem Cells/physiology , Humans , Immunophenotyping , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Reperfusion Injury/pathology , Retinal Diseases/pathology , Retinal Vessels/cytology
14.
BMC Med Genomics ; 3: 18, 2010 May 13.
Article in English | MEDLINE | ID: mdl-20465783

ABSTRACT

BACKGROUND: The term endothelial progenitor cells (EPCs) is currently used to refer to cell populations which are quite dissimilar in terms of biological properties. This study provides a detailed molecular fingerprint for two EPC subtypes: early EPCs (eEPCs) and outgrowth endothelial cells (OECs). METHODS: Human blood-derived eEPCs and OECs were characterised by using genome-wide transcriptional profiling, 2D protein electrophoresis, and electron microscopy. Comparative analysis at the transcript and protein level included monocytes and mature endothelial cells as reference cell types. RESULTS: Our data show that eEPCs and OECs have strikingly different gene expression signatures. Many highly expressed transcripts in eEPCs are haematopoietic specific (RUNX1, WAS, LYN) with links to immunity and inflammation (TLRs, CD14, HLAs), whereas many transcripts involved in vascular development and angiogenesis-related signalling pathways (Tie2, eNOS, Ephrins) are highly expressed in OECs. Comparative analysis with monocytes and mature endothelial cells clusters eEPCs with monocytes, while OECs segment with endothelial cells. Similarly, proteomic analysis revealed that 90% of spots identified by 2-D gel analysis are common between OECs and endothelial cells while eEPCs share 77% with monocytes. In line with the expression pattern of caveolins and cadherins identified by microarray analysis, ultrastructural evaluation highlighted the presence of caveolae and adherens junctions only in OECs. CONCLUSIONS: This study provides evidence that eEPCs are haematopoietic cells with a molecular phenotype linked to monocytes; whereas OECs exhibit commitment to the endothelial lineage. These findings indicate that OECs might be an attractive cell candidate for inducing therapeutic angiogenesis, while eEPC should be used with caution because of their monocytic nature.


Subject(s)
Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Hematopoietic Stem Cells/metabolism , Stem Cells/metabolism , Adult , Cadherins/metabolism , Caveolins/metabolism , Cell Lineage , Cells, Cultured , Electrophoresis, Gel, Two-Dimensional , Endothelial Cells/ultrastructure , Female , Gene Expression Profiling , Humans , Monocytes/immunology , Monocytes/metabolism , Principal Component Analysis , Signal Transduction , Stem Cells/classification
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.
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
17.
Invest Ophthalmol Vis Sci ; 50(10): 4967-73, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19474402

ABSTRACT

PURPOSE: Bone marrow-derived endothelial progenitor cells (EPCs) contribute to vascular repair although it is uncertain how local endothelial cell apoptosis influences their reparative function. This study was conducted to determine how the presence of apoptotic bodies at sites of endothelial damage may influence participation of EPCs in retinal microvascular repair. METHODS: Microlesions of apoptotic cell death were created in monolayers of retinal microvascular endothelial cells (RMECs) by using the photodynamic drug verteporfin. The adhesion of early-EPCs to these lesions was studied before detachment of the apoptotic cells or after their removal from the wound site. Apoptotic bodies were fed to normal RMECs and mRNA levels for adhesion molecules were analyzed. RESULTS: Endothelial lesions where apoptotic bodies were left attached at the wound site showed a fivefold enhancement in EPC recruitment (P < 0.05) compared with lesions where the apoptotic cells had been removed. In intact RMEC monolayers exposed to apoptotic bodies, expression of ICAM, VCAM, and E-selectin was upregulated by 5- to 15-fold (P < 0.05-0.001). EPCs showed a characteristic chemotactic response (P < 0.05) to conditioned medium obtained from apoptotic bodies, whereas analysis of the medium showed significantly increased levels of VEGF, IL-8, IL-6, and TNF-alpha when compared to control medium; SDF-1 remained unchanged. CONCLUSIONS: The data indicate that apoptotic bodies derived from retinal capillary endothelium mediate release of proangiogenic cytokines and chemokines and induce adhesion molecule expression in a manner that facilitates EPC recruitment.


Subject(s)
Apoptosis , Cell Movement/physiology , Endothelium, Vascular/physiology , Retinal Vessels/cytology , Stem Cells/physiology , Animals , Capillaries/cytology , Cattle , Cell Adhesion/physiology , Cells, Cultured , Chemotaxis/physiology , Cytokines/metabolism , E-Selectin/genetics , E-Selectin/metabolism , Endothelium, Vascular/drug effects , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique, Indirect , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Photosensitizing Agents/toxicity , Porphyrins/toxicity , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism , Vascular Endothelial Growth Factor A/metabolism , Verteporfin , Wound Healing/physiology
18.
Oncol Res ; 17(3): 93-101, 2008.
Article in English | MEDLINE | ID: mdl-18669161

ABSTRACT

The contribution of endothelial cell growth to angiogenesis has been widely studied; however, the involvement of pericytes is less well documented, especially in human tumors. In this study we aimed to quantify and assess the prognostic significance of pericyte coverage, the extent of hypoxia, and microvessel density (MVD) in normal bladder mucosa and urothelial carcinoma. Antibody to alpha-smooth muscle actin was used to assess the distribution of pericytes (mural/smooth muscle cells) in the microvessels of normal human bladder (n = 4) mucosa and in urothelial carcinoma (n = 47) samples; this was quantitated using microvessel pericyte index (MPI). The MVD was measured using two different methods (n = 47) and hypoxia was assessed using glucose transporter-1 (Glut-1) staining (n = 30). There was a 70% reduction in MPI in urothelial carcinomas compared to normal bladder mucosa (p < 0.0012); MPI did not correlate with tumor stage or grade. Ta and T1 superficial tumors were divided into two groups with a MPI of <15% or >15%. Progression-free survival was significantly shorter for tumors with MPI >15% (p = 0.0036). MVD had no prognostic value using either evaluation method. Glut-1 immunoreactivity was not prognostic in superficial urothelial carcinoma samples. Tumors with a higher MPI showed a greater Glut-1 immunoreactivity (p = 0.0051). Microvessels in urothelial carcinoma have a considerable loss of pericyte coverage compared to normal bladder mucosa. The data from this preliminary study indicate that progression-free survival was shorter in patients whose superficial tumors had higher pericyte coverage of the microvessels. This may be due to increased levels of hypoxia, as demonstrated by a significant increase in Glut-1 staining.


Subject(s)
Carcinoma/pathology , Neovascularization, Pathologic/pathology , Pericytes/pathology , Urinary Bladder Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Capillary Permeability , Carcinoma/blood supply , Carcinoma/physiopathology , Cell Count , Female , Glucose Transporter Type 1/analysis , Humans , Hypoxia/etiology , Hypoxia/pathology , Male , Middle Aged , Neoplasm Recurrence, Local/blood supply , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/physiopathology , Neoplasm Staging , Neovascularization, Pathologic/physiopathology , Prognosis , Urinary Bladder/blood supply , Urinary Bladder Neoplasms/blood supply , Urinary Bladder Neoplasms/physiopathology
19.
PLoS One ; 3(7): e2584, 2008 Jul 09.
Article in English | MEDLINE | ID: mdl-18612412

ABSTRACT

BACKGROUND: Current guidelines encourage the use of statins to reduce the risk of cardiovascular disease in diabetic patients; however the impact of these drugs on diabetic retinopathy is not well defined. Moreover, pleiotropic effects of statins on the highly specialised retinal microvascular endothelium remain largely unknown. The objective of this study was to investigate the effects of clinically relevant concentrations of simvastatin on retinal endothelium in vitro and in vivo. METHODS AND FINDINGS: Retinal microvascular endothelial cells (RMECs) were treated with 0.01-10 microM simvastatin and a biphasic dose-related response was observed. Low concentrations enhanced microvascular repair with 0.1 microM simvastatin significantly increasing proliferation (p<0.05), and 0.01 microM simvastatin significantly promoting migration (p<0.05), sprouting (p<0.001), and tubulogenesis (p<0.001). High concentration of simvastatin (10 microM) had the opposite effect, significantly inhibiting proliferation (p<0.01), migration (p<0.01), sprouting (p<0.001), and tubulogenesis (p<0.05). Furthermore, simvastatin concentrations higher than 1 microM induced cell death. The mouse model of oxygen-induced retinopathy was used to investigate the possible effects of simvastatin treatment on ischaemic retinopathy. Low dose simvastatin (0.2 mg/Kg) promoted retinal microvascular repair in response to ischaemia by promoting intra-retinal re-vascularisation (p<0.01). By contrast, high dose simvastatin(20 mg/Kg) significantly prevented re-vascularisation (p<0.01) and concomitantly increased pathological neovascularisation (p<0.01). We also demonstrated that the pro-vascular repair mechanism of simvastatin involves VEGF stimulation, Akt phosphorylation, and nitric oxide production; and the anti-vascular repair mechanism is driven by marked intracellular cholesterol depletion and related disorganisation of key intracellular structures. CONCLUSIONS: A beneficial effect of low-dose simvastatin on ischaemic retinopathy is linked to angiogenic repair reducing ischaemia, thereby preventing pathological neovascularisation. High-dose simvastatin may be harmful by inhibiting reparative processes and inducing premature death of retinal microvascular endothelium which increases ischaemia-induced neovascular pathology. Statin dosage should be judiciously monitored in patients who are diabetic or are at risk of developing other forms of proliferative retinopathy.


Subject(s)
Anticholesteremic Agents/pharmacology , Endothelium, Vascular/drug effects , Retinal Neovascularization/drug therapy , Retinal Vessels/drug effects , Simvastatin/pharmacology , Actins/metabolism , Animals , Anticholesteremic Agents/therapeutic use , Cattle , Cell Movement , Cell Proliferation , Cholesterol/metabolism , Diabetic Retinopathy/drug therapy , Endothelial Cells/drug effects , Ischemia/drug therapy , Nitric Oxide/biosynthesis , Oncogene Protein v-akt/metabolism , Phosphorylation , Simvastatin/therapeutic use
20.
Invest Ophthalmol Vis Sci ; 49(3): 1232-41, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18326753

ABSTRACT

PURPOSE: Vascular repair by marrow-derived endothelial progenitor cells (EPCs) is impaired during diabetes, although the precise mechanism of this dysfunction remains unknown. The hypothesis for the study was that progressive basement membrane (BM) modification by advanced glycation end products (AGEs) contributes to impairment of EPC reparative function after diabetes-related endothelial injury. METHODS: EPCs isolated from peripheral blood were characterized by immunocytochemistry and flow cytometry. EPC interactions on native or AGE-modified fibronectin (AGE-FN) were studied for attachment and spreading, whereas chemotaxis to SDF-1 was assessed with the Dunn chamber assay. In addition, photoreactive agent-treated monolayers of retinal microvascular endothelial cells (RMECs) produced circumscribed areas of apoptosis and the ability of EPCs to "endothelialize" these wounds was evaluated. RESULTS: EPC attachment and spreading on AGE-FN was reduced compared with control cells (P < 0.05-0.01) but was significantly restored by pretreatment with Arg-Gly-Asp (RGD). Chemotaxis of EPCs was abolished on AGE-FN but was reversed by treatment with exogenous RGD. On wounded RMEC monolayers, EPCs showed clustering at the wound site, compared with untreated regions (P < 0.001); AGE-FN significantly reduced this targeting response (P < 0.05). RGD supplementation enhanced EPC incorporation in the monolayer, as determined by EPC participation in tight junction formation and restoration of transendothelial electric resistance (TEER). CONCLUSIONS: AGE-modification of vascular substrates impairs EPC adhesion, spreading, and migration; and alteration of the RGD integrin recognition motif plays a key role in these responses. The presence of AGE adducts on BM compromises repair by EPC with implications for vasodegeneration during diabetic microvasculopathy.


Subject(s)
Diabetic Retinopathy/metabolism , Endothelium, Vascular/metabolism , Fibronectins/metabolism , Glycation End Products, Advanced/metabolism , Stem Cells/metabolism , Wound Healing , Animals , Cattle , Cell Adhesion/physiology , Cell Movement , Chemokine CXCL12/pharmacology , Chemotaxis/drug effects , Endothelium, Vascular/drug effects , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunohistochemistry , Oligopeptides/pharmacology , Pyruvaldehyde/toxicity , Retinal Vessels/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...