Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cells ; 9(6)2020 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-32503129

RESUMO

Fenestrae are transcellular plasma membrane pores that mediate blood-tissue exchange in specialised vascular endothelia. The composition and biogenesis of the fenestra remain enigmatic. We isolated and characterised the protein composition of large patches of fenestrated plasma membrane, termed sieve plates. Loss-of-function experiments demonstrated that two components of the sieve plate, moesin and annexin II, were positive and negative regulators of fenestra formation, respectively. Biochemical analyses showed that moesin is involved in the formation of an actin-fodrin submembrane cytoskeleton that was essential for fenestra formation. The link between the fodrin cytoskeleton and the plasma membrane involved the fenestral pore protein PV-1 and Na,K-ATPase, which is a key regulator of signalling during fenestra formation both in vitro and in vivo. These findings provide a conceptual framework for fenestra biogenesis, linking the dynamic changes in plasma membrane remodelling to the formation of a submembrane cytoskeletal signalling complex.


Assuntos
Actinas/metabolismo , Proteínas de Transporte/metabolismo , Membrana Celular/metabolismo , Citoesqueleto/metabolismo , Células Endoteliais/metabolismo , Proteínas dos Microfilamentos/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Anexina A2/metabolismo , Linhagem Celular , Membrana Celular/ultraestrutura , Citoesqueleto/ultraestrutura , Células Endoteliais/ultraestrutura , Masculino , Camundongos , Ouabaína/farmacologia , Ratos Sprague-Dawley
2.
Am J Pathol ; 187(10): 2208-2221, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28739342

RESUMO

Current treatments for choroidal neovascularization, a major cause of blindness for patients with age-related macular degeneration, treat symptoms but not the underlying causes of the disease. Inflammation has been strongly implicated in the pathogenesis of choroidal neovascularization. We examined the inflammatory role of Toll-like receptor 2 (TLR2) in age-related macular degeneration. TLR2 was robustly expressed by the retinal pigment epithelium in mouse and human eyes, both normal and with macular degeneration/choroidal neovascularization. Nuclear localization of NF-κB, a major downstream target of TLR2 signaling, was detected in the retinal pigment epithelium of human eyes, particularly in eyes with advanced stages of age-related macular degeneration. TLR2 antagonism effectively suppressed initiation and growth of spontaneous choroidal neovascularization in a mouse model, and the combination of anti-TLR2 and antivascular endothelial growth factor receptor 2 yielded an additive therapeutic effect on both area and number of spontaneous choroidal neovascularization lesions. Finally, in primary human fetal retinal pigment epithelium cells, ligand binding to TLR2 induced robust expression of proinflammatory cytokines, and end products of lipid oxidation had a synergistic effect on TLR2 activation. Our data illustrate a functional role for TLR2 in the pathogenesis of choroidal neovascularization, likely by promoting inflammation of the retinal pigment epithelium, and validate TLR2 as a novel therapeutic target for reducing choroidal neovascularization.


Assuntos
Neovascularização de Coroide/patologia , Inflamação/patologia , Degeneração Macular/patologia , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia , Receptor 2 Toll-Like/metabolismo , Idoso , Idoso de 80 Anos ou mais , Animais , Anticorpos Neutralizantes/farmacologia , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Núcleo Celular/efeitos da radiação , Chlamydia/efeitos dos fármacos , Chlamydia/efeitos da radiação , Neovascularização de Coroide/complicações , Neovascularização de Coroide/metabolismo , Citocinas/metabolismo , Dipeptídeos/farmacologia , Raios gama , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos da radiação , Humanos , Inflamação/complicações , Inflamação/genética , Lipídeos/química , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/patologia , Degeneração Macular/complicações , Degeneração Macular/metabolismo , Masculino , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Oxirredução , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/efeitos da radiação
3.
Sci Signal ; 8(395): ra94, 2015 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-26396267

RESUMO

Neurons and glial cells in the retina contribute to neovascularization, or the formation of abnormal new blood vessels, in proliferative retinopathy, a condition that can lead to vision loss or blindness. We identified a mechanism by which suppressor of cytokine signaling 3 (SOCS3) in neurons and glial cells prevents neovascularization. We found that Socs3 expression was increased in the retinal ganglion cell and inner nuclear layers after oxygen-induced retinopathy. Mice with Socs3 deficiency in neuronal and glial cells had substantially reduced vaso-obliterated retinal areas and increased pathological retinal neovascularization in response to oxygen-induced retinopathy, suggesting that loss of neuronal/glial SOCS3 increased both retinal vascular regrowth and pathological neovascularization. Furthermore, retinal expression of Vegfa (which encodes vascular endothelial growth factor A) was higher in these mice than in Socs3 flox/flox controls, indicating that neuronal and glial SOCS3 suppressed Vegfa expression during pathological conditions. Lack of neuronal and glial SOCS3 resulted in greater phosphorylation and activation of STAT3, which led to increased expression of its gene target Vegfa, and increased endothelial cell proliferation. In summary, SOCS3 in neurons and glial cells inhibited the STAT3-mediated secretion of VEGF from these cells, which suppresses endothelial cell activation, resulting in decreased endothelial cell proliferation and angiogenesis. These results suggest that neuronal and glial cell SOCS3 limits pathological retinal angiogenesis by suppressing VEGF signaling.


Assuntos
Neuroglia/metabolismo , Neovascularização Retiniana/metabolismo , Neurônios Retinianos/metabolismo , Transdução de Sinais , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Camundongos , Camundongos Knockout , Neuroglia/patologia , Oxigênio/toxicidade , Neovascularização Retiniana/induzido quimicamente , Neovascularização Retiniana/patologia , Neurônios Retinianos/patologia , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/genética , Fator A de Crescimento do Endotélio Vascular/genética
4.
Am J Pathol ; 185(9): 2534-49, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26188133

RESUMO

Choroidal neovascularization (CNV) is a defining feature of wet age-related macular degeneration. We examined the functional role of CCR3 in the development of CNV in mice and primates. CCR3 was associated with spontaneous CNV lesions in the newly described JR5558 mice, whereas CCR3 ligands localized to CNV-associated macrophages and the retinal pigment epithelium/choroid complex. Intravitreal injection of neutralizing antibodies against vascular endothelial growth factor receptor 2, CCR3, CC chemokine ligand 11/eotaxin-1, and CC chemokine ligand 24/eotaxin-2 all reduced CNV area and lesion number in these mice. Systemic administration of the CCR3 antagonists GW766994X and GW782415X reduced spontaneous CNV in JR5558 mice and laser-induced CNV in mouse and primate models in a dose-dependent fashion. Combination treatment with antivascular endothelial growth factor receptor 2 antibody and GW766994X yielded additive reductions in CNV area and hyperpermeability in mice. Interestingly, topical GW766994X and intravitreal anti-CCR3 antibody yielded strong systemic effects, reducing CNV in the untreated, contralateral eye. Contrarily, ocular administration of GW782415X in primates failed to substantially elevate plasma drug levels or to reduce the development of grade IV CNV lesions. These findings suggest that CCR3 signaling may be an attractive therapeutic target for CNV, utilizing a pathway that is at least partly distinct from that of vascular endothelial growth factor receptor. The findings also demonstrate that systemic exposure to CCR3 antagonists may be crucial for CNV-targeted activity.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Permeabilidade Capilar/efeitos dos fármacos , Neovascularização de Coroide/tratamento farmacológico , Receptores CCR3/antagonistas & inibidores , Degeneração Macular Exsudativa/tratamento farmacológico , Animais , Permeabilidade Capilar/imunologia , Corioide/patologia , Neovascularização de Coroide/patologia , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Fator A de Crescimento do Endotélio Vascular/metabolismo , Degeneração Macular Exsudativa/patologia
5.
PLoS One ; 9(1): e85031, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24416337

RESUMO

Proliferative retinopathy is a leading cause of blindness, including retinopathy of prematurity (ROP) in children and diabetic retinopathy in adults. Retinopathy is characterized by an initial phase of vessel loss, leading to tissue ischemia and hypoxia, followed by sight threatening pathologic neovascularization in the second phase. Previously we found that Sirtuin1 (Sirt1), a metabolically dependent protein deacetylase, regulates vascular regeneration in a mouse model of oxygen-induced proliferative retinopathy (OIR), as neuronal depletion of Sirt1 in retina worsens retinopathy. In this study we assessed whether over-expression of Sirtuin1 in retinal neurons and vessels achieved by crossing Sirt1 over-expressing flox mice with Nestin-Cre mice or Tie2-Cre mice, respectively, may protect against retinopathy. We found that over-expression of Sirt1 in Nestin expressing retinal neurons does not impact vaso-obliteration or pathologic neovascularization in OIR, nor does it influence neuronal degeneration in OIR. Similarly, increased expression of Sirt1 in Tie2 expressing vascular endothelial cells and monocytes/macrophages does not protect retinal vessels in OIR. In addition to the genetic approaches, dietary supplement with Sirt1 activators, resveratrol or SRT1720, were fed to wild type mice with OIR. Neither treatment showed significant vaso-protective effects in retinopathy. Together these results indicate that although endogenous Sirt1 is important as a stress-induced protector in retinopathy, over-expression of Sirt1 or treatment with small molecule activators at the examined doses do not provide additional protection against retinopathy in mice. Further studies are needed to examine in depth whether increasing levels of Sirt1 may serve as a potential therapeutic approach to treat or prevent retinopathy.


Assuntos
Degeneração Neural/genética , Neurônios/metabolismo , Retina/metabolismo , Degeneração Retiniana/genética , Sirtuína 1/genética , Animais , Cruzamentos Genéticos , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Expressão Gênica , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Integrases/genética , Integrases/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Neovascularização Patológica , Degeneração Neural/induzido quimicamente , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Nestina/genética , Nestina/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/patologia , Oxigênio/efeitos adversos , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Resveratrol , Retina/efeitos dos fármacos , Retina/patologia , Degeneração Retiniana/induzido quimicamente , Degeneração Retiniana/metabolismo , Degeneração Retiniana/patologia , Sirtuína 1/metabolismo , Estilbenos/farmacologia
6.
Invest Ophthalmol Vis Sci ; 54(2): 1490-500, 2013 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-23385800

RESUMO

PURPOSE: Two noninvasive delivery strategies for VEGF/PDGF receptor tyrosine kinase inhibitors (RTKI) were explored that exploited uveal retention as a means for establishing an ocular drug depot: a single oral "loading" dose and topical administration. METHODS: Melanin binding was confirmed by centrifugation and mass spectrometry. Ocular retention was examined in pigmented and albino rats. Ocular release kinetics were measured 3 to 28 days postdosing in pigmented rats. Microautoradiography was used to demonstrate retention of RTKI in the uveal tract. A uveal drug depot of pazopanib was created by a single oral dose prior to induction of laser choroidal neovascularization (CNV). Choroid/retinal pigmented epithelium (RPE) retention of a related RTKI with enhanced topical bioavailability, GW771806, was confirmed by bioanalytics, and its ability to regress CNV compared with pazopanib. RESULTS: Pazopanib and GW771806 directly bound melanin and were retained within the uveal tract of pigmented rats for weeks following a single oral dose. Pazopanib was undetectable systemically following a single oral administration prior to CNV induction, and reduced CNV as well as twice daily dosing. Topical ocular delivery of GW771806 at 5 mg/mL led to high choroidal/RPE exposure and significantly regressed CNV lesions; 2 mg/mL prevented lesion progression. CONCLUSIONS: Uveal retention of drugs such as pazopanib can be used to create a sustained-release depot. Topical GW771806 regressed CNV. These data indicate that topical or infrequent oral loading dose treatment with VEGF/PDGF RTKI retained in the choroid/RPE might allow noninvasive treatments for ocular neovascular disease.


Assuntos
Inibidores da Angiogênese/administração & dosagem , Neovascularização de Coroide/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Indazóis/administração & dosagem , Pirimidinas/administração & dosagem , Receptores do Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Sulfonamidas/administração & dosagem , Sulfonas/administração & dosagem , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Administração Oral , Administração Tópica , Inibidores da Angiogênese/farmacocinética , Animais , Autorradiografia , Neovascularização de Coroide/diagnóstico , Neovascularização de Coroide/metabolismo , Feminino , Angiofluoresceinografia , Meia-Vida , Indazóis/farmacocinética , Melaninas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Pirimidinas/farmacocinética , Ratos , Ratos Long-Evans , Ratos Sprague-Dawley , Sulfonamidas/farmacocinética , Sulfonas/farmacocinética , Úvea/metabolismo
7.
Invest Ophthalmol Vis Sci ; 49(2): 662-70, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18235012

RESUMO

PURPOSE: To investigate the effect of the combined treatment of photodynamic therapy and specific VEGF165 inhibition with pegaptanib sodium (Macugen; Eyetech Pharmaceuticals, Lexington, MA) on ocular neovascularization. METHODS: Photodynamic therapy's (PDT's) effects on the integrity of pegaptanib sodium were analyzed by HPLC, a VEGF165-binding assay, and a VEGF165-induced tissue factor gene expression assay. The effects of mono- or combined treatment on vessel growth and regression were determined in a murine corneal neovascularization model. The effects of combined treatment on vessel growth were also determined in a murine choroidal neovascularization model. RESULTS: PDT did not affect the chemical composition of pegaptanib sodium nor the efficacy of pegaptanib sodium in the inhibition of VEGF165 binding to Flt-1 and VEGF165-induced gene expression. In an animal model of effects on existing ocular neovascular lesions (corneal neovascularization), PDT monotherapy yielded an initial regression of these vessels, but there followed a rapid regrowth. In contrast, pegaptanib sodium monotherapy yielded little regression but potently abrogated further vessel growth. The combination of pegaptanib sodium and PDT resulted in the regression of the neovascular lesions, as observed with PDT alone, but also prevented significant vessel regrowth, leading to a significantly greater reduction in lesion size than did each monotherapy. In addition, there was a significantly greater effect of the combination of pegaptanib sodium and PDT on lesion size in choroidal neovascularization than with each monotherapy. Pretreatment with pegaptanib sodium appeared to decrease the efficacy of PDT-induced vessel regression in corneal neovascularization, and as such the enhanced efficacy over monotherapy when the agents were delivered simultaneously was not observed. CONCLUSIONS: Although the combined simultaneous treatment of ocular neovascularization with PDT and pegaptanib sodium may provide a more effective approach for the regression and overall treatment of CNV associated with AMD, the order of addition of these treatments may play a role in achieving optimal efficacy.


Assuntos
Aptâmeros de Nucleotídeos/uso terapêutico , Neovascularização de Coroide/tratamento farmacológico , Neovascularização da Córnea/tratamento farmacológico , Modelos Animais de Doenças , Fotoquimioterapia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Neovascularização de Coroide/metabolismo , Neovascularização de Coroide/patologia , Cromatografia Líquida de Alta Pressão , Córnea/irrigação sanguínea , Neovascularização da Córnea/metabolismo , Neovascularização da Córnea/patologia , Quimioterapia Combinada , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fármacos Fotossensibilizantes/uso terapêutico , Porfirinas/uso terapêutico , RNA Mensageiro/metabolismo , Tromboplastina/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Verteporfina
8.
Angiogenesis ; 10(2): 141-8, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17372853

RESUMO

The growth of inappropriately regulated, leaky blood vessels is a prominent component of several debilitating eye diseases, such as age-related macular degeneration (AMD), proliferative diabetic retinopathy (PDR), and retinopathy of prematurity (ROP). New pharmacological therapies that target vascular endothelial growth factor-A (VEGF-A) have significantly enhanced the treatment of AMD by limiting the progression of the disease, and in some cases, by improving vision. Although anti-VEGF therapy will undoubtedly prove valuable in the treatment of other neovascular diseases of the eye, improvements with this type of therapy are still required. At present, anti-VEGF therapy requires intravitreal injection and a relatively frequent dosing regimen (4-6 weeks). Furthermore, in experimental models of neovascularization, anti-VEGF treatment becomes less effective at blocking vessel growth and at regressing vessels as the neovascularization develops over time. As such, the use of anti-VEGF therapy in late-stage AMD may be limited. An important strategy for improved treatment of neovascular diseases of the eye could be combination therapy. Combination therapy of anti-VEGF drugs with established treatments, such as photodynamic therapy with verteporfin (PDT-V), or with newly-developed drugs targeting specific kinases, presents opportunities for increased efficacy and improved therapeutic outcome. In this review, we evaluate the opportunities for combination therapy for the treatment of neovascular diseases of the eye.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Neovascularização Patológica/terapia , Doenças Retinianas/terapia , Terapia Combinada , Retinopatia Diabética/fisiopatologia , Humanos , Recém-Nascido , Degeneração Macular/fisiopatologia , Neovascularização Patológica/etiologia , Retinopatia da Prematuridade/fisiopatologia
9.
Mol Vis ; 12: 1243-9, 2006 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-17110907

RESUMO

PURPOSE: The wet form of age-related macular degeneration (AMD) occurs as a consequence of abnormal blood vessel growth from the choroid into the retina. Pathological angiogenesis during tumor growth and ocular disease has been associated with specific exposure of cryptic extracellular matrix epitopes. We investigated the presence of cryptic collagen IV epitopes in a murine model of choroidal neovascularization (CNV), and tested the effect on blood vessel growth of H8, a humanized antibody directed against a cryptic collagen type IV epitope. METHODS: To induce experimental CNV in adult C57BL/6 mice, Bruch's membrane was ruptured using a diode laser. Subsequently, mice were treated with daily intraperitoneal (i.p.) injections of either H8 (10 mg/kg or 30 mg/kg) or an isotype-matched antibody control. Two weeks postinjection, choroidal flat mounts were immunostained with the blood vessel marker platelet/endothelial cell adhesion molecule-1 (PECAM-1) and H8. CNV was visualized using fluorescence microscopy and the CNV lesion area measured using Open Lab software. RESULTS: Collagen type IV and the cryptic epitope were observed at the site of laser-induced lesions. Staining with H8 was first observed three days post injury, two days after MMP2 expression in CNV lesions, becoming most intense five days following laser injury and extending beyond the area of neovascularization. At 14 days post injury, H8 staining was reduced in intensity, colocalized with the area of CNV, and was nearly absent from the underlying choroidal vessels. In addition, mice treated with H8 had a significant dose-dependent decrease in the area of CNV as compared to isotype-matched antibody controls. CONCLUSIONS: Results suggest that exposure of cryptic collagen type IV epitopes is associated with the incidence of CNV and that the humanized antibody H8 may provide a new treatment for CNV.


Assuntos
Anticorpos/farmacologia , Neovascularização de Coroide/prevenção & controle , Colágeno Tipo IV/imunologia , Epitopos/imunologia , Lasers , Animais , Anticorpos/imunologia , Neovascularização de Coroide/etiologia , Neovascularização de Coroide/imunologia , Epitopos/metabolismo , Matriz Extracelular/imunologia , Olho/efeitos dos fármacos , Olho/metabolismo , Traumatismos Oculares/imunologia , Traumatismos Oculares/metabolismo , Temperatura Alta , Imuno-Histoquímica/métodos , Masculino , Metaloproteinase 2 da Matriz/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Desnaturação Proteica , Lesões por Radiação/imunologia , Lesões por Radiação/metabolismo , Coloração e Rotulagem , Fatores de Tempo
10.
Am J Pathol ; 168(6): 2036-53, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16723717

RESUMO

'Vascular endothelial growth factor-A (VEGF-A) blockade has been recently validated as an effective strategy for the inhibition of new blood vessel growth in cancer and ocular pathologies. However, several studies have also shown that anti-VEGF therapy may not be as effective in the treatment of established unwanted blood vessels, suggesting they may become less dependent on VEGF-A for survival. The VEGF-A dependence of vessels may be related to the presence of vascular mural cells (pericytes or smooth muscle cells). Mural cell recruitment to the growing endothelial tube is regulated by platelet-derived growth factor-B (PDGF-B) signaling, and interference with this pathway causes disruption of endothelial cell-mural cell interactions and loss of mural cells. We have investigated the basis of blood vessel dependence on VEGF-A in models of corneal and choroidal neovascularization using a combination of reagents (an anti-VEGF aptamer and an anti-PDGFR-beta antibody) to inhibit both the VEGF-A and PDGF-B signaling pathways. We demonstrate that neovessels become refractory to VEGF-A deprivation over time. We also show that inhibition of both VEGF-A and PDGF-B signaling is more effective than blocking VEGF-A alone at causing vessel regression in multiple models of neovascular growth. These findings provide insight into blood vessel growth factor dependency and validate a combination therapy strategy for enhancing the current treatments for ocular angiogenic disease.


Assuntos
Endotélio Vascular/patologia , Neovascularização Patológica , Proteínas Proto-Oncogênicas c-sis/metabolismo , Retina/patologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pericitos/metabolismo , Veia Retiniana/metabolismo , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo
11.
Invest Ophthalmol Vis Sci ; 46(8): 2736-47, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16043845

RESUMO

PURPOSE: Pigment epithelium-derived factor (PEDF), a protein secreted by the retinal pigment epithelium (RPE), acts on retinal survival and angiogenesis. Because hypoxia and VEGF regulate matrix metalloproteinases (MMPs), their effects on PEDF proteolysis were explored. METHODS: Mouse models for retinopathy of prematurity (ROP) were used. Cultured monkey RPE cells were exposed to low oxygen and chemical hypoxia mimetics. PEDF and VEGF mRNA levels in RPE were determined by RT-PCR. MMPs were assessed by zymography, DQ-gelatin degradation solution assays, and MMP immunostaining. PEDF proteolysis was assayed in solution and followed by SDS-PAGE and immunostaining. MMP induction by VEGF was performed in baby hamster kidney (BHK) cells. Retinal R28 cell survival, ex vivo chick embryonic aortic vessel sprouting, and directed in vivo angiogenesis assays were performed. RESULTS: Levels of PEDF in RPE/choroid significantly decreased in the ROP model. Hypoxia decreased PEDF levels in the media conditioned by RPE cells, with no significant change in PEDF mRNA. Conversely, PEDF proteolysis, gelatinolytic activities of approximately 57-kDa and approximately 86-kDa zymogens, and MMP-2 immunoreactivities increased with hypoxia. Addition of VEGF to BHK cells caused a time and dose-related upregulation of approximately 57-kDa zymogens and of DQ-gelatinolytic and PEDF-degrading activity. The PEDF-degrading activity and approximately 57-kDa zymogens in the BHK media shared MMP protease inhibition patterns and MMP-2 immunoreactivities with those in the vitreous. Limited proteolysis with MMP-2 and -9 degraded PEDF in a Ca(+2)-dependent fashion. MMP-mediated proteolysis of PEDF abolished the retinal survival and antiangiogenic activities of the PEDF protein. CONCLUSIONS: Hypoxia and VEGF can downregulate PEDF through proteolytic degradation. PEDF is a novel substrate for MMP-2 and -9. These results reveal a novel posttranslational mechanism for downregulating PEDF, and provide an explanation for hypoxia-provoked increases in VEGF/PEDF ratios, in angiogenesis and/or in neuronal death.


Assuntos
Proteínas do Olho/metabolismo , Hipóxia/enzimologia , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Fatores de Crescimento Neural/metabolismo , Retinopatia da Prematuridade/enzimologia , Serpinas/metabolismo , Animais , Animais Recém-Nascidos , Western Blotting , Células Cultivadas , Embrião de Galinha , Cricetinae , Modelos Animais de Doenças , Regulação para Baixo , Eletroforese em Gel de Poliacrilamida , Proteínas do Olho/genética , Humanos , Recém-Nascido , Macaca , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/metabolismo , Fatores de Crescimento Neural/genética , Oxigênio/toxicidade , Epitélio Pigmentado Ocular/citologia , Epitélio Pigmentado Ocular/efeitos dos fármacos , Epitélio Pigmentado Ocular/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Serpinas/genética , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
12.
Yao Xue Xue Bao ; 40(2): 111-6, 2005 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-15875664

RESUMO

AIM: To study the drug metabolizing enzymes involved in the metabolism of bicyclol and identify the major metabolites of bicyclol in rat and human liver microsomes. METHODS: Bicyclol was incubated with rat and human liver microsomes. The metabolites of bicyclol were isolated by HPLC and identified by MS and 1H NMR. RESULTS: The metabolic rate of bicyclol in DEX-induced rat liver microsomes was obviously higher than that in untreated microsomes, while it was much lower in human liver microsomes. Ketoconazole was capable to exhibit strong inhibition ( >90%) on bicyclol metabolism. Two metabolites of bicyclol were identified to be 4-hydroxy-4'-methoxy-6-hydroxy-methyl-6 '-methoxycarbonyl-2,3,2',3'-bis (methylene-dioxy) biphenyl and 4-methoxy-4'-hydroxy-6-hydroxymethyl-6'-methoxycarbonyl-2,3,2',3'-bis (methylene-dioxy) biphenyl. CONCLUSION: CYP3A was considered as the major catalyst involved in bicyclol metabolism in vitro and two metabolites of bicyclol in rats were identified as 4-hydroxy-4'-methoxy-6-hydroxy-methyl-6 '-methoxycarbonyl-2,3,2',3'-bis (methylene-dioxy) biphenyl and 4-methoxy-4'-hydroxy-6-hydroxymethyl-6'-methoxycarbonyl-2,3,2',3'-bis (methylenedioxy) biphenyl.


Assuntos
Compostos de Bifenilo/metabolismo , Microssomos Hepáticos/metabolismo , Adulto , Animais , Compostos de Bifenilo/farmacocinética , Sistema Enzimático do Citocromo P-450/metabolismo , Humanos , Técnicas In Vitro , Cetoconazol/farmacologia , Masculino , Microssomos Hepáticos/efeitos dos fármacos , Ratos , Ratos Wistar
13.
Proc Natl Acad Sci U S A ; 100(26): 15859-64, 2003 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-14657382

RESUMO

Degeneration of vessels precedes and precipitates the devastating ischemia of many diseases, including retinopathy of prematurity and diabetic retinopathy. Ischemia then leads to proliferative retinopathy and blindness. Understanding the mechanisms of blood vessel degeneration is critical to prevention of these diseases. Vessel loss is associated with oxygen-induced suppression of vascular endothelial growth factor (VEGF) and with pericyte (vascular smooth muscle cell) dropout. The molecular mechanism of pericyte protection of the vasculature is unknown. We show that transforming growth factor beta1 (TGF-beta1)-expressing pericytes are specifically found on vessels resistant to oxygen-induced loss. TGF-beta1 potently induces VEGF receptor 1 (VEGFR-1) expression in endothelial cells and thereby prevents oxygen-induced vessel loss in vivo. Vessel survival is further stimulated with a VEGFR-1-specific ligand, placental growth factor 1. TGF-beta1 induction of VEGFR-1 in endothelial cells explains pericyte protection of vessels and the selective vulnerability of neonatal vessels to oxygen. These results implicate induction and activation of VEGFR-1 as critical targets to prevent vessel loss.


Assuntos
Endotélio Vascular/fisiologia , Vasos Retinianos/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Animais , Bovinos , Primers do DNA , Regulação da Expressão Gênica/efeitos dos fármacos , Reação em Cadeia da Polimerase , RNA/genética , RNA/isolamento & purificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/farmacologia , Fator de Crescimento Transformador beta1 , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
14.
J Clin Invest ; 112(1): 50-7, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12840058

RESUMO

Oxygen administration to immature neonates suppresses VEGF-A expression in the retina, resulting in the catastrophic vessel loss that initiates retinopathy of prematurity. To investigate the mechanisms responsible for survival of blood vessels in the developing retina, we characterized two VEGF-A receptors, VEGF receptor-1 (VEGFR-1, also known as Flt-1) and VEGF receptor-2 (VEGFR-2, also known as Flk-1). Surprisingly, these two VEGF-A receptors differed markedly during normal retinal development in mice. At 5 days postpartum (P5), VEGFR-1 protein was colocalized with retinal vessels, whereas VEGFR-2 was detected only in the neural retina. Real-time RT-PCR identified a 60-fold induction of VEGFR-1 mRNA in retina from P3 (early vascularization) to P26 (fully vascularized), and no significant change in VEGFR-2 mRNA expression. Placental growth factor-1 (PlGF-1), which exclusively binds VEGFR-1, decreased hyperoxia-induced retinal vaso-obliteration from 22.2% to 5.1%, whereas VEGF-E, which exclusively binds VEGFR-2, had no effect on blood vessel survival. Importantly, under the same conditions, PlGF-1 did not increase vasoproliferation during (a). normal vessel growth, (b). revascularization following hyperoxia-induced ischemia, or (c). the vasoproliferative phase, indicating a selective function supporting blood vessel survival. We conclude that VEGFR-1 is critical in maintaining the vasculature of the neonatal retina, and that activation of VEGFR-1 by PlGF-1 is a selective strategy for preventing oxygen-induced retinal ischemia without provoking retinal neovascularization.


Assuntos
Oxigênio/toxicidade , Proteínas da Gravidez/uso terapêutico , Retina/patologia , Retinopatia da Prematuridade/tratamento farmacológico , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Animais , Fatores de Crescimento Endotelial/genética , Humanos , Recém-Nascido , Peptídeos e Proteínas de Sinalização Intercelular , Linfocinas , Camundongos , Camundongos Endogâmicos C57BL , Fator de Crescimento Placentário , RNA Mensageiro/análise , Retinopatia da Prematuridade/patologia , Fator A de Crescimento do Endotélio Vascular , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Fatores de Crescimento do Endotélio Vascular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...