Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Am J Pathol ; 185(6): 1769-82, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25913075

RESUMO

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.


Assuntos
Biopterinas/análogos & derivados , Hiperóxia/metabolismo , Óxido Nítrico Sintase/metabolismo , Retina/metabolismo , Retinopatia da Prematuridade/metabolismo , Animais , Biopterinas/metabolismo , Feminino , Hiperóxia/patologia , Masculino , Camundongos , Retina/patologia , Retinopatia da Prematuridade/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
2.
Sci Rep ; 13(1): 21292, 2023 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-38042898

RESUMO

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.


Assuntos
Biopterinas , Doenças Retinianas , Camundongos , Animais , Biopterinas/metabolismo , Doenças Retinianas/tratamento farmacológico , Óxido Nítrico/metabolismo , Morte Celular , Suplementos Nutricionais , Isquemia/tratamento farmacológico
3.
Am J Pathol ; 178(4): 1517-28, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21435441

RESUMO

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.


Assuntos
Apoptose , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Isquemia/patologia , Microglia/metabolismo , Neurônios/metabolismo , Pericitos/metabolismo , Retina/lesões , Animais , Astrócitos/citologia , Morte Celular , Diferenciação Celular , Oftalmopatias/patologia , Feminino , Proteínas de Fluorescência Verde/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência/métodos , Pericitos/citologia , Retina/metabolismo
4.
Mol Med ; 17(9-10): 1045-55, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21670847

RESUMO

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.


Assuntos
Células Endoteliais/fisiologia , Interleucina-8/metabolismo , Macrófagos/fisiologia , Células Mieloides/fisiologia , Neovascularização Fisiológica/fisiologia , Adulto , Animais , Bovinos , Células Cultivadas , Células Endoteliais/metabolismo , Perfilação da Expressão Gênica/métodos , Humanos , Immunoblotting , Interleucina-8/genética , Isquemia/fisiopatologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células Mieloides/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Proteômica/métodos , Vasos Retinianos/metabolismo , Vasos Retinianos/fisiologia , Células-Tronco/metabolismo , Células-Tronco/fisiologia , Transcriptoma , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
5.
Mol Vis ; 15: 1509-20, 2009 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-19668595

RESUMO

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.


Assuntos
Células Endoteliais/patologia , Produtos Finais de Glicação Avançada/farmacologia , Microvasos/patologia , Oligopeptídeos/farmacologia , Vasos Retinianos/patologia , Animais , Membrana Basal/efeitos dos fármacos , Caspase 3/metabolismo , Bovinos , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Ativação Enzimática/efeitos dos fármacos , Fibronectinas/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Microvasos/efeitos dos fármacos , Microvasos/enzimologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/enzimologia , Permeabilidade/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Vasos Retinianos/efeitos dos fármacos , Vasos Retinianos/enzimologia , Transdução de Sinais/efeitos dos fármacos , Superóxidos/metabolismo
6.
Invest Ophthalmol Vis Sci ; 49(3): 1232-41, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18326753

RESUMO

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.


Assuntos
Retinopatia Diabética/metabolismo , Endotélio Vascular/metabolismo , Fibronectinas/metabolismo , Produtos Finais de Glicação Avançada/metabolismo , Células-Tronco/metabolismo , Cicatrização , Animais , Bovinos , Adesão Celular/fisiologia , Movimento Celular , Quimiocina CXCL12/farmacologia , Quimiotaxia/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Oligopeptídeos/farmacologia , Aldeído Pirúvico/toxicidade , Vasos Retinianos/citologia
7.
Oncol Res ; 17(3): 93-101, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18669161

RESUMO

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.


Assuntos
Carcinoma/patologia , Neovascularização Patológica/patologia , Pericitos/patologia , Neoplasias da Bexiga Urinária/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Permeabilidade Capilar , Carcinoma/irrigação sanguínea , Carcinoma/fisiopatologia , Contagem de Células , Feminino , Transportador de Glucose Tipo 1/análise , Humanos , Hipóxia/etiologia , Hipóxia/patologia , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/irrigação sanguínea , Recidiva Local de Neoplasia/patologia , Recidiva Local de Neoplasia/fisiopatologia , Estadiamento de Neoplasias , Neovascularização Patológica/fisiopatologia , Prognóstico , Bexiga Urinária/irrigação sanguínea , Neoplasias da Bexiga Urinária/irrigação sanguínea , Neoplasias da Bexiga Urinária/fisiopatologia
8.
Diabetes ; 54(3): 785-94, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15734857

RESUMO

Suppression of angiogenesis during diabetes is a recognized phenomenon but is less appreciated within the context of diabetic retinopathy. The current study has investigated regulation of retinal angiogenesis by diabetic serum and determined if advanced glycation end products (AGEs) could modulate this response, possibly via AGE-receptor interactions. A novel in vitro model of retinal angiogenesis was developed and the ability of diabetic sera to regulate this process was quantified. AGE-modified serum albumin was prepared according to a range of protocols, and these were also analyzed along with neutralization of the AGE receptors galectin-3 and RAGE. Retinal ischemia and neovascularization were also studied in a murine model of oxygen-induced proliferative retinopathy (OIR) in wild-type and galectin-3 knockout mice (gal3(-/-)) after perfusion of preformed AGEs. Serum from nondiabetic patients showed significantly more angiogenic potential than diabetic serum (P < 0.0001) and within the diabetic group, poor glycemic control resulted in more AGEs but less angiogenic potential than tight control (P < 0.01). AGE-modified albumin caused a dose-dependent inhibition of angiogenesis (P < 0.001), and AGE receptor neutralization significantly reversed the AGE-mediated suppression of angiogenesis (P < 0.01). AGE-treated wild-type mice showed a significant increase in inner retinal ischemia and a reduction in neovascularization compared with non-AGE controls (P < 0.001). However, ablation of galectin-3 abolished the AGE-mediated increase in retinal ischemia and restored the neovascular response to that seen in controls. The data suggest a significant suppression of angiogenesis by the retinal microvasculature during diabetes and implicate AGEs and AGE-receptor interactions in its causation.


Assuntos
Diabetes Mellitus/sangue , Diabetes Mellitus/fisiopatologia , Galectina 3/fisiologia , Produtos Finais de Glicação Avançada/fisiologia , Neovascularização Fisiológica/fisiologia , Vasos Retinianos/fisiopatologia , Adulto , Albuminas , Animais , Retinopatia Diabética/fisiopatologia , Galectina 3/sangue , Galectina 3/genética , Produtos Finais de Glicação Avançada/sangue , Humanos , Imunoglobulina G/sangue , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/fisiologia , Fator A de Crescimento do Endotélio Vascular/sangue
9.
Invest Ophthalmol Vis Sci ; 47(12): 5561-8, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17122149

RESUMO

PURPOSE: Diabetic patients who also have retinitis pigmentosa (RP) appear to have fewer and less severe retinal microvascular lesions. Diabetic retinopathy may be linked to increased inner retinal hypoxia, with the possibility that this is exacerbated by oxygen usage during the dark-adaptation response. Therefore, patients with RP with depleted rod photoreceptors may encounter proportionately less retinal hypoxia, and, when diabetes is also present, there may be fewer retinopathic lesions. This hypothesis was tested in rhodopsin knockout mice (Rho-/-) as an RP model in which the diabetic milieu is superimposed. The study was designed to investigate whether degeneration of the outer retina has any impact on hypoxia, to examine diabetes-related retinal gene expression responses, and to assess lesions of diabetic retinopathy. METHODS: Streptozotocin-induced diabetes was created in male C57Bl6 (wild-type; WT) and Rho-/- mice, and hyperglycemia was maintained for 5 months. The extent of diabetes was confirmed by measurement of glycated hemoglobin (%GHb) and accumulation of advanced glycation end products (AGEs). Retinal hypoxia was assessed using the bioreductive drug pimonidazole. The retinal microvasculature was studied in retinal flatmounts stained by the ADPase reaction, and the outer retina was evaluated histologically in paraffin-embedded sections. Retinal gene expression of VEGF-A, TNF-alpha, and mRNAs encoding basement membrane component proteins were quantified by real-time RT-PCR. RESULTS: The percentage GHb increased significantly in the presence of diabetes (P < 0.001) and was not different between WT or Rho-/- mice. Hypoxia increased in the retina of WT diabetic animals when compared with controls (P < 0.001) but this diabetes-induced change was absent in Rho-/- mice. Retinal gene expression of VEGF-A was significantly increased in WT mice with diabetes (P < 0.05), but was unchanged in Rho-/- mice. TNF-alpha gene expression significantly increased (4.9-fold) in WT mice with diabetes (P < 0.05) and also increased appreciably in Rho-/- mice but to a reduced extent (1.5 fold; P < 0.05). The outer nuclear layer in nondiabetic Rho-/- mice was reduced to a single layer after 6 months, but when diabetes was superimposed on this model, there was less degeneration of photoreceptors (P < 0.05). Vascular density was attenuated in diabetic WT mice compared with the nondiabetic control (P < 0.001); however, this diabetes-related disease was not observed in Rho-/- mice. CONCLUSIONS: Loss of the outer retina reduces the severity of diabetic retinopathy in a murine model. Oxygen usage by the photoreceptors during dark adaptation may contribute to retinal hypoxia and exacerbate the progression of diabetic retinopathy.


Assuntos
Diabetes Mellitus Experimental/fisiopatologia , Retinopatia Diabética/fisiopatologia , Retinose Pigmentar/fisiopatologia , Rodopsina/fisiologia , Animais , Animais Geneticamente Modificados , Apirase/metabolismo , Membrana Basal/metabolismo , Diabetes Mellitus Experimental/metabolismo , Retinopatia Diabética/metabolismo , Ensaio de Imunoadsorção Enzimática , Hemoglobinas Glicadas/metabolismo , Produtos Finais de Glicação Avançada/metabolismo , Histocitoquímica , Hipóxia/metabolismo , Hipóxia/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/metabolismo , Vasos Retinianos/patologia , Retinose Pigmentar/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
10.
Invest Ophthalmol Vis Sci ; 47(12): 5553-60, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17122148

RESUMO

PURPOSE: This study was conducted to evaluate whether regions of the retinal neuropile become hypoxic during periods of high oxygen consumption and whether depletion of the outer retina reduces hypoxia and related changes in gene expression. METHODS: Retinas from rhodopsin knockout (Rho-/-) mice were evaluated along with those of wild-type (WT) control animals. Retinas were also examined at the end of 12-hour dark or light periods, and a separate group was treated with l-cis-diltiazem at the beginning of a 12-hour dark period. Hypoxia was assessed by deposition of hypoxyprobe (HP) and HP-protein adducts were localized by immunohistochemistry and quantified using ELISA. Also, hypoxia-regulated gene expression and transcriptional activity were assessed alongside vascular density. RESULTS: Hypoxia was observed in the inner nuclear and ganglion cell layers in WT retina and was significantly reduced in Rho-/- mice (P < 0.05). Retinal hypoxia was significantly increased during dark adaptation in WT mice (P < 0.05), whereas no change was observed in Rho-/- or with l-cis-diltiazem-treated WT mice. Hypoxia-inducible factor (HIF)-1alpha DNA-binding and VEGF mRNA expression in Rho-/- retina was significantly reduced in unison with outer retinal depletion (P < 0.05). Retina from the Rho-/- mice displayed an extensive intraretinal vascular network after 6 months, although there was evidence that capillary density was depleted in comparison with that in WT retinas. CONCLUSIONS: Relative hypoxia occurs in the inner retina especially during dark adaptation. Photoreceptor loss reduces retinal oxygen usage and hypoxia which corresponds with attenuation of the retinal microvasculature. These studies suggest that in normal physiological conditions and diurnal cycles the adult retina exists in a state of borderline hypoxia, making this tissue particularly susceptible to even subtle reductions in perfusion.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Hipóxia/metabolismo , Degeneração Retiniana/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Rodopsina/fisiologia , Ativação Transcricional/fisiologia , Animais , Apirase/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 6 , Adaptação à Escuridão , Diltiazem/farmacologia , Ensaio de Imunoadsorção Enzimática , Hipóxia/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Luz , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nitroimidazóis/farmacologia , Consumo de Oxigênio , Diester Fosfórico Hidrolases/metabolismo , RNA Mensageiro/metabolismo , Degeneração Retiniana/patologia , Vasos Retinianos/enzimologia , Vasos Retinianos/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
11.
Curr Eye Res ; 31(12): 1033-45, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17169842

RESUMO

PURPOSE: This study aimed to evaluate the effects of endostatin on tight junction (TJ) integrity in retinal microvascular endothelial cells (RMECs) in vitro and in vivo. Moreover, it was hypothesized that endostatin-induced occludin upregulation regulated VEGF165-mediated increases in endothelial cell permeability and involved activation of the MAPK signaling cascade. Endostatin is a 20-kDa fragment of collagen XVIII that has been shown to be efficacious in the eye by preventing retinal neovascularization. Endostatin is a specific inhibitor of endothelial cell proliferation, migration, and angiogenesis and has been reported to reverse VEGF-mediated increases in vasopermeability and to promote integrity of the blood-retinal barrier (BRB). In order to determine the mechanism of endostatin action on BRB integrity, we have examined the effects of endostatin on a number of intracellular pathways implicated in endothelial cell physiology. METHODS: C57/Bl6 mice were injected with VEGF165 and/or endostatin, and the distribution of occludin staining was determined using retinal flatmounts. Western blot analysis of RMECs treated with VEGF165 and/or endostatin was used to determine changes in occludin expression and p38 MAPK and extracellular regulated kinase (ERK1/ERK2 MAPK) activation, while FD-4 flux across the RMEC monolayer was used to determine changes in paracellular permeability. RESULTS: Endostatin prevented the discontinuous pattern of occludin staining observed at the retinal blood vessels of mice administered an intraocular injection of VEGF165. It was shown that endostatin activated p38 MAPK 5 min after addition to RMECs and continued to do so for approximately 30 min. Endostatin was also shown to activate ERK1/ERK2 5 min after addition and continued to do so, albeit with less potency, up to and including 15 min after addition. Inhibition of p38 MAPK and ERK1/ERK2 prevented endostatin's ability to upregulate levels of occludin expression. Inhibition of these key signaling molecules was shown to prevent endostatin's ability to protect against VEGF165-mediated increases in paracellular permeability in vitro. However, it appears that p38 MAPK may play a more important role in VEGF-mediated permeability, as inhibition of ERK1/ERK2 will not prevent VEGF165-mediated permeability compared with control (untreated) cells or cells treated with both a p38 MAPK inhibitor and VEGF165. CONCLUSIONS: Occludin is important for the maintenance of tight junction integrity in vivo. In a p38 MAPK and ERK1/ERK2 dependent manner, endostatin was shown to upregulate the levels of expression of the tight junction protein occludin. Inhibition of these key MAPK components may prevent endostatin's ability to decrease VEGF165-induced paracellular permeability.


Assuntos
Inibidores da Angiogênese/farmacologia , Barreira Hematorretiniana/efeitos dos fármacos , Endostatinas/farmacologia , Endotélio Vascular/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Western Blotting , Bovinos , Endotélio Vascular/metabolismo , Inibidores Enzimáticos/farmacologia , Técnica Indireta de Fluorescência para Anticorpo , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Ocludina , Vasos Retinianos/citologia , Junções Íntimas , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
12.
Invest Ophthalmol Vis Sci ; 56(13): 8008-18, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26720448

RESUMO

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.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Glicogênio/metabolismo , Neurônios Retinianos/metabolismo , Células Amácrinas/metabolismo , Animais , Citoplasma/metabolismo , Glicogênio Fosforilase/metabolismo , Imuno-Histoquímica , Masculino , Microscopia/métodos , Ratos
13.
Invest Ophthalmol Vis Sci ; 44(10): 4457-64, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14507893

RESUMO

PURPOSE: A critical event in the pathogenesis of diabetic retinopathy is the inappropriate adherence of leukocytes to the retinal capillaries. Advanced glycation end-products (AGEs) are known to play a role in chronic inflammatory processes, and the authors postulated that these adducts may play a role in promoting pathogenic increases in proinflammatory pathways within the retinal microvasculature. METHODS: Retinal microvascular endothelial cells (RMECs) were treated with glycoaldehyde-modified albumin (AGE-Alb) or unmodified albumin (Alb). NFkappaB DNA binding was measured by electromobility shift assay (EMSA) and quantified with an ELISA: In addition, the effect of AGEs on leukocyte adhesion to endothelial cell monolayers was investigated. Further studies were performed in an attempt to confirm that this was AGE-induced adhesion by co-incubation of AGE-treated cells with soluble receptor for AGE (sRAGE). Parallel in vivo studies of nondiabetic mice assessed the effect of intraperitoneal delivery of AGE-Alb on ICAM-1 mRNA expression, NFkappaB DNA-binding activity, leukostasis, and blood-retinal barrier breakdown. RESULTS: Treatment with AGE-Alb significantly enhanced the DNA-binding activity of NFkappaB (P = 0.0045) in retinal endothelial cells (RMECs) and increased the adhesion of leukocytes to RMEC monolayers (P = 0.04). The latter was significantly reduced by co-incubation with sRAGE (P < 0.01). Mice infused with AGE-Alb demonstrated a 1.8-fold increase in ICAM-1 mRNA when compared with control animals (P < 0.001, n = 20) as early as 48 hours, and this response remained for 7 days of treatment. Quantification of retinal NFkappaB demonstrated a threefold increase with AGE-Alb infusion in comparison to control levels (AGE Alb versus Alb, 0.23 vs. 0.076, P < 0.001, n = 10 mice). AGE-Alb treatment of mice also caused a significant increase in leukostasis in the retina (AGE-Alb versus Alb, 6.89 vs. 2.53, n = 12, P < 0.05) and a statistically significant increase in breakdown of the blood-retinal barrier (AGE Alb versus Alb, 8.2 vs. 1.6 n = 10, P < 0.001). CONCLUSIONS: AGEs caused upregulation of NFkappaB in the retinal microvascular endothelium and an AGE-specific increase in leukocyte adhesion in vitro was also observed. In addition, increased leukocyte adherence in vivo was demonstrated that was accompanied by blood-retinal barrier dysfunction. These findings add further evidence to the thinking that AGEs may play an important role in the pathogenesis of diabetic retinopathy.


Assuntos
Endotélio Vascular/metabolismo , Produtos Finais de Glicação Avançada/farmacologia , Neutrófilos/fisiologia , Vasos Retinianos/metabolismo , Animais , Barreira Hematorretiniana/efeitos dos fármacos , Capilares , Permeabilidade Capilar , Bovinos , Adesão Celular/efeitos dos fármacos , DNA/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Ensaio de Imunoadsorção Enzimática , Humanos , Molécula 1 de Adesão Intercelular/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima
14.
Mol Vis ; 10: 432-8, 2004 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-15235573

RESUMO

PURPOSE: Perforation injury to the eye can protect against retinal degeneration and pigment epithelial derived factor (PEDF) may play a role in this neuro-protective effect. PEDF has also been shown to possess potent anti-angiogenic properties. The current study has investigated a possible anti-angiogenic effect of penetrating ocular injury in a murine model of oxygen induced proliferative retinopathy (OIR) and determined if such a procedure alters PEDF expression in the retina. METHODS: OIR was produced by exposure of neonatal mice to 75% oxygen between postnatal days 7 and 12 (P7-P12). Mice were separated into various groups, with one group receiving a penetrating injury in a single eye. Pre-retinal neovascularization and intra-retinal ischaemia was quantified at P17 and PEDF protein expression was determined using immunofluorescence in retinal flatmounts and sections. PEDF mRNA was quantified using real-time RT-PCR. RESULTS: Punctured eyes showed less pre-retinal neovascularization at P17 when compared to the non-punctured eyes (p<0.001) although there was no effect on retinal ischaemia. PEDF immunreactivity was strongest in the ganglion cells of the retina, and intensity increased in punctured eyes at P13. There was more immunoreactive PEDF in ganglion cells adjacent to retinal venules than arterioles. At P13, retinal PEDF mRNA was also increased in punctured eyes compared to non-punctured controls (p<0.05), although there was no differential at P17. CONCLUSIONS: Penetrating ocular injury suppresses retinal neovascularization and modulates expression of PEDF. These findings have implications for intra-vitreal delivery of angiostatic agents since ocular perforation may provoke an acute, endogenous anti-angiogenic response that should be taken into account.


Assuntos
Ferimentos Oculares Penetrantes/complicações , Proteínas do Olho , Fatores de Crescimento Neural , Proteínas/metabolismo , Neovascularização Retiniana/prevenção & controle , Serpinas/metabolismo , Animais , Animais Recém-Nascidos , Corpo Ciliar/lesões , Modelos Animais de Doenças , Ferimentos Oculares Penetrantes/metabolismo , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Hiperóxia/complicações , Recém-Nascido , Camundongos , Camundongos Endogâmicos C57BL , Oxigênio/toxicidade , Proteínas/genética , RNA Mensageiro/metabolismo , Retina/metabolismo , Neovascularização Retiniana/etiologia , Neovascularização Retiniana/metabolismo , Retinopatia da Prematuridade/etiologia , Retinopatia da Prematuridade/metabolismo , Retinopatia da Prematuridade/prevenção & controle , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Serpinas/genética , Regulação para Cima , Corpo Vítreo/lesões
15.
Mol Vis ; 9: 665-72, 2003 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-14685146

RESUMO

PURPOSE: Progressive dysfunction and death of vascular smooth muscle cells and pericytes is a pathophysiological hallmark of diabetic retinopathy, although the underlying mechanisms behind this process remain ill-defined. The multifunctional peptide platelet-derived growth factor (PDGF) is known to act as an important survival factor for both of these vascular cell-types at times of physiological stress. The retinal cell source(s) of PDGF remain unknown. It is important to understand how diabetes alters expression of this important growth factor. METHODS: Streptozotocin-diabetes was established in C57 mice. Following 8 weeks of sustained diabetes, the eyes were enucleated and in situ hybridization was used to localize expression of PDGF-A and PDGF-B chains in retina from both diabetic and non-diabetic controls. mRNA levels for both forms of PDGF, and their cognate PDGF-alpha and PDGF-beta receptors, were also quantified using real-time PCR. RESULTS: In situ hybridization demonstrated that PDGF-A and PDGF-B were predominantly expressed by the retinal ganglion cells/nerve fibre layer in both normal and diabetic mice, and this localization pattern did not alter in diabetes. PDGF-A receptor was expressed exclusively in the ganglion cell layer of the retina while PDGF-B receptor was mostly localized to the Muller cell end-feet at the internal limiting membrane with lesser immunoreactivity in the ganglion cells, inner plexiform layer, and inner nuclear layer. PDGF-A and PDGF-alpha receptor mRNA expression levels remained unaltered between treatment groups, although retinal immunolocalization patterns between both receptors was distinct. However, there was a significant decrease of PDGF-B mRNA levels in diabetic retina when compared to non-diabetic controls (p<0.001), although there was no significant difference in PDGF-alpha receptor(insert space) expression. CONCLUSIONS: Previous studies have shown PDGF expression in a range of cell-types during retinal development, but these results confirm ganglion cells as the principal PDGF source in mature retina. It may be significant that diabetes can reduce PDGF-B mRNA expression since this may have serious implications for vascular survival during diabetic retinopathy progression.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Retinopatia Diabética/metabolismo , Fibras Nervosas/metabolismo , Fator de Crescimento Derivado de Plaquetas/genética , Proteínas Proto-Oncogênicas c-sis/genética , Células Ganglionares da Retina/metabolismo , Animais , Diabetes Mellitus Experimental/patologia , Retinopatia Diabética/patologia , Imuno-Histoquímica , Hibridização In Situ , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogênicas c-sis/metabolismo , RNA Mensageiro/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
Mol Vis ; 10: 996-1004, 2004 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-15635291

RESUMO

PURPOSE: To assess the effects of advanced glycation endproduct (AGE) modification of vascular basement membrane (BM) on endothelin-1 (Et-1) induced intracellular [Ca2+] ([Ca2+]i) homeostasis and contraction in retinal microvascular pericytes (RMP). METHODS: RMPs were isolated from bovine retinal capillaries and propagated on AGE modified BM extract (AGE-BM) or non-modified native BM. Cytosolic Ca2+ was estimated using fura-2 microfluorimetry and cellular contraction determined by measurement of planimetric cell surface area. ETA receptor mRNA and protein expression was assessed by real time RT-PCR and western blotting, respectively. RESULTS: Exogenous endothelin-1 (Et-1) evoked rises in [Ca2+]i and contraction in RMPs were found to be mediated entirely through ETA receptor (ETAR) activation. Both peak and plateau phases of the Et-1 induced [Ca2+]i response and contraction were impaired in RMPs propagated on AGE modified BM. ETAR mRNA expression remained unchanged in RMPs exposed to native or AGE-BM, but protein expression for ETAR (66 kDa) was lower in the AGE exposed cells. CONCLUSIONS: These results suggest that substrate derived AGE crosslinks can influence RMP physiology by mechanisms which include disruption of ETA receptor signalling. AGE modification of vascular BMs may contribute to the retinal hemodynamic abnormalities observed during diabetes.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Contratura/metabolismo , Endotelina-1/farmacologia , Produtos Finais de Glicação Avançada/farmacologia , Pericitos/fisiologia , Vasos Retinianos/fisiologia , Animais , Membrana Basal/efeitos dos fármacos , Membrana Basal/metabolismo , Western Blotting , Cálcio/metabolismo , Capilares , Bovinos , Eletroforese em Gel de Poliacrilamida , Fura-2/metabolismo , Microscopia de Fluorescência , Pericitos/citologia , RNA Mensageiro/metabolismo , Receptor de Endotelina A/genética , Vasos Retinianos/citologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
Invest Ophthalmol Vis Sci ; 55(4): 2157-65, 2014 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-24609622

RESUMO

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.


Assuntos
Arteríolas/fisiologia , Velocidade do Fluxo Sanguíneo/fisiologia , Canais de Cloreto/metabolismo , Canais de Potássio/metabolismo , Artéria Retiniana/fisiologia , Animais , Angiofluoresceinografia , Fundo de Olho , Masculino , Músculo Liso Vascular/fisiologia , Oftalmoscopia , Técnicas de Patch-Clamp , Ratos
18.
Invest Ophthalmol Vis Sci ; 53(6): 3059-66, 2012 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-22427579

RESUMO

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.


Assuntos
Cálcio/metabolismo , Endotelina-1/farmacologia , Músculo Liso Vascular/metabolismo , Artéria Retiniana/metabolismo , Transdução de Sinais , Animais , Arteríolas/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio/fisiologia , Membrana Celular/metabolismo , Angiofluoresceinografia , Técnicas In Vitro , Injeções Intravítreas , Masculino , Microscopia Confocal , Ratos , Ratos Sprague-Dawley , Vasoconstrição
19.
Prog Retin Eye Res ; 30(3): 149-66, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21352947

RESUMO

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.


Assuntos
Retinopatia Diabética/terapia , Endotélio Vascular/citologia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Traumatismo por Reperfusão/terapia , Oclusão da Veia Retiniana/terapia , Animais , Humanos , Vasos Retinianos/citologia
20.
Diabetes ; 60(11): 2995-3005, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21911748

RESUMO

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.


Assuntos
Retinopatia Diabética/tratamento farmacológico , Eritropoetina/química , Degeneração Neural/prevenção & controle , Neuroglia/efeitos dos fármacos , Fragmentos de Peptídeos/uso terapêutico , Degeneração Retiniana/prevenção & controle , Vasos Retinianos/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Citocinas/genética , Citocinas/metabolismo , Dano ao DNA/efeitos dos fármacos , Retinopatia Diabética/metabolismo , Retinopatia Diabética/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Isquemia/tratamento farmacológico , Isquemia/metabolismo , Isquemia/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuroglia/patologia , Fragmentos de Peptídeos/efeitos adversos , Fragmentos de Peptídeos/química , Domínios e Motivos de Interação entre Proteínas , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Retina/efeitos dos fármacos , Retina/metabolismo , Retina/patologia , Vasos Retinianos/patologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa