Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Eur J Pharm Biopharm ; 129: 267-272, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29906511

RESUMO

Proliferative vitreoretinopathy (PVR) is a challenging pathological condition, often causing failure of retinal detachment surgery. The purpose of this study was to evaluate the feasibility of a delivery system of bioactive proteins using anionic and cationic gelatin microspheres and to establish a new PVR model in rabbits by intraocular sustained delivery of basic fibroblast growth factor (bFGF) and interferon-beta (IFNß). Anionic and cationic gelatin microspheres were prepared and immersed in bFGF and IFNß solution, respectively, to yield a polyion complex between gelatin matrix and a bioactive protein. The bFGF-impregnated microspheres were injected into the subretinal space in rabbit eyes. At week 2, the IFNß-impregnated microspheres also were injected into the same space. Control eyes received gelatin microspheres without bFGF or IFNß, or both. The eyes then were observed for 8 weeks by ophthalmoscopy, fundus photography, and fluorescein angiography. The eyes also were evaluated histologically. In the group with both bFGF and IFNß, the number of eyes with more severe PVR increased over time. Histologic examination showed retinal folds. In contrast, no proliferative changes were seen in any control groups. Subretinal implantation of bFGF and IFNß-impregnated gelatin microspheres induced reproducible PVR in rabbit eyes. This study guaranteed delivery of bioactive proteins with gelatin microspheres.


Assuntos
Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos/métodos , Coelhos , Retina/efeitos dos fármacos , Vitreorretinopatia Proliferativa/induzido quimicamente , Animais , Estudos de Viabilidade , Fator 2 de Crescimento de Fibroblastos/administração & dosagem , Fator 2 de Crescimento de Fibroblastos/toxicidade , Gelatina/química , Humanos , Injeções Intraoculares , Interferon beta/administração & dosagem , Interferon beta/toxicidade , Microesferas , Oftalmoscopia , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/toxicidade , Reprodutibilidade dos Testes , Retina/diagnóstico por imagem , Retina/patologia , Vitreorretinopatia Proliferativa/diagnóstico por imagem , Vitreorretinopatia Proliferativa/patologia
2.
Toxicol Mech Methods ; 24(5): 323-31, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24559483

RESUMO

Basic fibroblast growth factor (bFGF) is a mitogenic cytokine that can stimulate mesoderm-and neuroectoderm-originated cell proliferation. This study was performed to investigate the effects of bFGF on cell differentiation and the expression of specific markers at different embryonic developmental stages. We firstly evaluated the embryotoxic potential of bFGF in vitro using a modified EST protocol. Sequentially, we further investigated how bFGF impact the different tissue-special genes and proteins expressions during the differentiation of murine ES cells in vitro and attempt to reveal the effects of bFGF on differentiation processes. This analysis was focused on key tissue- and stage-specific genes involved in ectodermal, mesodermal, and endodermal differentiation, including ectodermal-specific gene Nestin, Oligo2 and Syn, mesodermal-specific gene MHC and MyoD, and endodermal-specific gene GATA6, TTR and ALB, as well as undifferentiated gene Sox-2 and Oct-4. The results demonstrate that bFGF could promote expression of ectodermal-specific genes and protein, but suppress the expressions of endoderm-specific and some mesoderm-specific gene and protein. A conclusion can be drawn that bFGF exhibits weak embryotoxicity and mainly promotes ES cell differentiation towards the ectodermal lineages but suppress differentiation into endoderm lineages. These opposing effects of bFGF on the embryonic development of the three germ layers may be related to its weak embryotoxic potential. More specifically, inhibition of expression of the endodermal-specific markers transthyretin (TTR), and albumin (ALB) by bFGF may be of more value in detecting the embryotoxic potential of bFGF.


Assuntos
Células-Tronco Embrionárias/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/toxicidade , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Animais , Células 3T3 BALB , Diferenciação Celular/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Camadas Germinativas/metabolismo , Camundongos
3.
Brain Res ; 1547: 25-33, 2014 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-24374066

RESUMO

The cystine/glutamate antiporter (system xc-) transports cystine into cell in exchange for glutamate. Fibroblast growth factor-2 (FGF-2) upregulates system xc- selectively on astrocytes, which leads to increased cystine uptake, the substrate for glutathione production, and increased glutamate release. While increased intracellular glutathione can limit oxidative stress, the increased glutamate release can potentially lead to excitotoxicity to neurons. To test this hypothesis, mixed neuronal and glial cortical cultures were treated with FGF-2. Treatment with FGF-2 for 48 h caused a significant neuronal death in these cultures. Cell death was not observed in neuronal-enriched cultures, or astrocyte-enriched cultures, suggesting the toxicity was the result of neuron-glia interaction. Blocking system xc- eliminated the neuronal death as did the AMPA/kainate receptor antagonist 2,3-dihydroxy-6-nitro-7-sulfamoyl-benzo[f]quinoxaline-2,3-dione (NBQX), but not the NMDA receptor antagonist memantine. When cultures were exposed directly to glutamate, both NBQX and memantine blocked the neuronal toxicity. The mechanism of this altered profile of glutamate receptor mediated toxicity by FGF-2 is unclear. The selective calcium permeable AMPA receptor antagonist 1-naphthyl acetyl spermine (NASPM) failed to offer protection. The most likely explanation for the results is that 48 h FGF-2 treatment induces AMPA/kainate receptor toxicity through increased system xc- function resulting in increased release of glutamate. At the same time, FGF-2 alters the sensitivity of the neurons to glutamate toxicity in a manner that promotes selective AMPA/kainate receptor mediated toxicity.


Assuntos
Sistemas de Transporte de Aminoácidos/metabolismo , Cistina/metabolismo , Fator 2 de Crescimento de Fibroblastos/toxicidade , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Animais , Morte Celular/efeitos dos fármacos , Células Cultivadas , Ácido Glutâmico/toxicidade , Camundongos , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Receptores de AMPA/metabolismo , Receptores de Ácido Caínico/metabolismo , Regulação para Cima
4.
Pain ; 154(10): 2216-2226, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23867734

RESUMO

UNLABELLED: Growth factors such as nerve growth factor and glial cell line-derived neurotrophic factor are known to induce pain sensitization. However, a plethora of other growth factors is released during inflammation and tissue regeneration, and many of them are essential for wound healing. Which wound-healing factors also alter the sensitivity of nociceptive neurons is not well known. We studied the wound-healing factor, basic fibroblast growth factor (bFGF), for its role in pain sensitization. Reverse transcription polymerase chain reaction showed that the receptor of bFGF, FGFR1, is expressed in lumbar rat dorsal root ganglia (DRG). We demonstrated presence of FGFR1 protein in DRG neurons by a recently introduced quantitative automated immunofluorescent microscopic technique. FGFR1 was expressed in all lumbar DRG neurons as quantified by mixture modeling. Corroborating the mRNA and protein expression data, bFGF induced Erk1/2 phosphorylation in nociceptive neurons, which could be blocked by inhibition of FGF receptors. Furthermore, bFGF activated Erk1/2 in a dose- and time-dependent manner. Using single-cell electrophysiological recordings, we found that bFGF treatment of DRG neurons increased the current-density of NaV1.8 channels. Erk1/2 inhibitors abrogated this increase. Importantly, intradermal injection of bFGF in rats induced Erk1/2-dependent mechanical hyperalgesia. PERSPECTIVE: Analyzing intracellular signaling dynamics in nociceptive neurons has proven to be a powerful approach to identify novel modulators of pain. In addition to describing a new sensitizing factor, our findings indicate the potential to investigate wound-healing factors for their role in nociception.


Assuntos
Fator 2 de Crescimento de Fibroblastos/toxicidade , Gânglios Espinais/fisiopatologia , Hiperalgesia/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Cicatrização/fisiologia , Animais , Células Cultivadas , Gânglios Espinais/efeitos dos fármacos , Hiperalgesia/induzido quimicamente , Hiperalgesia/fisiopatologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Ratos , Ratos Sprague-Dawley , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/biossíntese , Cicatrização/efeitos dos fármacos
5.
Exp Eye Res ; 99: 1-16, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22695224

RESUMO

Different growth factors have been shown to influence the development of form-deprivation myopia and lens-induced ametropias. However, growth factors have relatively little effect on the growth of eyes with unrestricted vision. We investigate whether the combination of insulin-like growth factor 1 (IGF1) and fibroblast growth factor 2 (FGF2) influence ocular growth in eyes with unrestricted vision. Different doses of IGF1 and FGF2 were injected into the vitreous chamber of postnatal chicks. Measurements of ocular dimensions and intraocular pressure (IOP) were made during and at the completion of different treatment paradigms. Histological and immunocytochemical analyses were performed to assess cell death, cellular proliferation and integrity of ocular tissues. Treated eyes had significant increases in equatorial diameter and vitreous chamber depth. With significant variability between individuals, IGF1/FGF2-treatment caused hypertrophy of lens and ciliary epithelia, lens thickness was increased, and anterior chamber depth was decreased. Treated eyes developed myopia, in excess of 15 diopters of refractive error. Shortly after treatment, eyes had increased intraocular pressure (IOP), which was increased in a dose-dependent manner. Seven days after treatment with IGF1 and FGF2 changes to anterior chamber depth, lens thickness and elevated IOP were reduced, whereas increases in the vitreous chamber were persistent. Some damage to ganglion cells was detected in peripheral regions of the retina at 7 days after treatment. We conclude that the extreme myopia in IGF1/FGF2-treated eyes results from increased vitreous chamber depth, decreased anterior chamber depth, and changes in the lens. We propose that factor-induced ocular enlargement and myopia result from changes to the sclera, lens and anterior chamber depth.


Assuntos
Modelos Animais de Doenças , Olho/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/toxicidade , Fator de Crescimento Insulin-Like I/toxicidade , Miopia Degenerativa/induzido quimicamente , Animais , Animais Recém-Nascidos , Câmara Anterior/efeitos dos fármacos , Câmara Anterior/patologia , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Galinhas , Corpo Ciliar/efeitos dos fármacos , Corpo Ciliar/patologia , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Olho/crescimento & desenvolvimento , Hipertrofia , Marcação In Situ das Extremidades Cortadas , Pressão Intraocular/efeitos dos fármacos , Injeções Intravítreas , Cristalino/efeitos dos fármacos , Cristalino/patologia , Miopia Degenerativa/patologia , Tamanho do Órgão/efeitos dos fármacos , RNA Mensageiro/metabolismo , Receptor IGF Tipo 1/genética , Receptor de Insulina/genética , Receptores de Fatores de Crescimento de Fibroblastos/genética , Retina/efeitos dos fármacos , Retina/patologia , Retinoscopia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
6.
São Paulo; s.n; s.n; 2012. 162 p. tab, graf, ilus.
Tese em Português | LILACS | ID: biblio-846848

RESUMO

FGF2 (Fibroblast Growth Factor 2) é o membro fundador de uma grande família de fatores de crescimento protéicos. Sua atividade se dá através da ligação e ativação de receptores específicos de membrana (FGFRs) com atividade de tirosina quinase. No organismo adulto, a sinalização de FGF2 está envolvida na indução de processos de sobrevivência, proliferação e diferenciação celular; além de cicatrização e angiogênese. Por atuar como um clássico fator de crescimento, a atividade de FGF2 está freqüentemente implicada em mecanismos pró-tumorais. Entretanto, alguns grupos, incluindo o nosso, têm reportado que FGF2 também pode apresentar efeitos antiproliferativos a até citotóxicos seletivamente em células malignas. Em 2008, publicamos um compreensivo relato mostrando que FGF2 bloqueia irreversivelmente a proliferação de linhagens murinas malignas dependentes de Ras. Alterações que levem a atividade aumentada de proteínas Ras estão presentes em diversos cânceres humanos e, freqüentemente, resultando em problemas no tratamento e prognóstico ruim. No presente trabalho, utilizamos principalmente a linhagem murina maligna dependente de Ras Y1 D1G, que apresenta um controle estrito de quiescência/proliferação em função da presença de soro; e é por isso mesmo um bom modelo para a análise dos efeitos de FGF2 sobre o ciclo celular. Análises por citometria de fluxo mostraram que, nessas células, apesar de disparar a transição G0→G1→S, FGF2 provoca um atraso na fase S seguido de um bloqueio do ciclo em G2. Embora bloqueie a progressão no ciclo (proliferação), FGF2 induz em Y1 D1G o crescimento celular em termos de massa e volume. Assim, nessas células FGF2 "desconecta" crescimento celular de proliferação. Esse desarranjo do ciclo celular provocado por FGF2 nas células Y1 D1G tem como resultado a instabilidade genotípica e morte celular; evidenciada pela perda da integridade de membrana plasmática e altas taxas de fragmentação de DNA observadas após o estímulo por esse fator. Esse efeito tóxico de FGF2 depende da atividade da proteína Src; porque a inibição química dessa proteína apresentou proteção total frente aos efeitos tóxicos de FGF2. Análises por espectrometria de massas mostraram que FGF2 induz aumento dos níveis de proteínas relacionadas à síntese protéica, e também de proteínas relacionadas ao estresse proteotóxico. Sabe-se que células malignas lidam com níveis basais altos de diferentes tipos de estresse; incluindo o estresse proteotóxico. Esse quadro mostra que o efeito tóxico disparado por FGF2 em Y1 D1G está relacionado a um acumulo de proteínas/célula, perda da homeostase de proteínas e estresse proteotóxico. Corrobora essas proposições o fato de que a inibição química de Src, que protege totalmente as células do efeito tóxico de FGF2, impede completamente o acúmulo de proteínas/célula. Além disso, em células Y1 D1G resistentes ao efeito tóxico de FGF2, e que inclusive dependem deste para proliferar em cultura, a atividade de FGF2 tem efeito oposto; ou seja, provoca diminuição dos níveis estacionários de proteínas/célula. Juntos, esses resultados demonstram que FGF2 é capaz de atacar uma vulnerabilidade de células malignas dependentes de Ras; e no caso estudado, essa vulnerabilidade decorre do desequilíbrio na homeostase de proteínas


FGF2 is the first member of a large family of peptide growth factors. It binds and activates specific membrane receptors (FGFRs) belonging to a family of tyrosine kinase receptors (RTK). In adult organisms, FGF2 signaling is involved in the induction of cell surveillance, proliferation and differentiation; and also wound healing and angiogenesis. FGF2 is a bona fide growth factor and, as such, it is often implicated in pro-tumor mechanisms. However, several groups, including ours, have reported that FGF2 can also display antiproliferative and even cytotoxic effects selectively in malignant cells. In 2008, we fully reported that FGF2 irreversibly blocks the proliferation of Ras-driven mouse malignant lineages. Alterations leading to Ras proteins overactivity are present in many human cancers frequently with bad prognosis. In the present work, we used mainly the Ras-driven mouse malignant lineage Y1 D1G that shows a strict control of quiescence/proliferation by serum factors, making it a great model to analyze the FGF2 effects upon cell cycle control. Flow cytometry analyses showed that in these cells, in spite of triggering G0→G1→S transition, FGF2 causes a delay on S phase followed by cell cycle arrest in G2. Despite blocking cell division, FGF2 induces cell growth in terms of mass and volume. Therefore, in these cells FGF2 "disconnects" cell growth from proliferation. This malfunction of cell cycle control caused by FGF2 on Y1 D1G cells leads to genotypic instability and cell death, highlighted by loss of plasma membrane integrity and high rates of DNA fragmentation. This FGF2 toxic effect depends on the activity of Src protein, because Src chemical inhibition completely protects cells from the FGF2 toxic effects. Mass spec analyses showed that FGF2 increases the levels of proteins involved in the protein synthesis machinery, and also of proteins active in proteostasis, indicating proteotoxic stress. It is known that malignant cells deal with high basal levels of different stresses, including the proteotoxic stress. This picture shows that the toxic effects triggered by FGF2 in Y1 D1G involve accumulation of proteins/cell, loss of protein homeostasis and proteotoxic stress. Corroborating these propositions, chemical inhibition of Src, which completely protects the cells from FGF2 toxic effects, totally abrogates the accumulation of proteins/cell. Moreover, in FGF2-resistant Y1 D1G cells, which depend on this factor for proliferation, FGF2 shows the opposite effect, causing decrease in steady state levels of protein/cell. Altogether, these results show that FGF2 causes a severe proteostasis imbalance in these Ras-driven mouse malignant cells


Assuntos
Divisão Celular/genética , Fator 2 de Crescimento de Fibroblastos/toxicidade , Proteínas ras/genética , Ciclo Celular/genética , Proliferação de Células , Proliferação de Células/genética , Vulnerabilidade a Desastres , Dano ao DNA , Citometria de Fluxo/métodos , Neoplasias/complicações
7.
Eur J Ophthalmol ; 20(1): 48-54, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19882518

RESUMO

PURPOSE: To determine the effect of vascular endothelial growth factor (VEGF) TrapR1R2 on bFGF-induced experimental corneal neovascularization (NV). METHODS: Control pellets or pellets containing 80 ng bFGF were surgically implanted into wild-type C57BL/6 and VEGF-LacZ mouse corneas. The corneas were photographed, harvested, and the percentage of corneal NV was calculated. The harvested corneas were evaluated for VEGF expression. VEGF-LacZ mice received tail vein injections of an endothelial-specific lectin after pellet implantation to determine the temporal and spatial relationship between VEGF expression and corneal NV. Intraperitoneal injections of VEGF TrapR1R2 or a human IgG Fc domain control protein were administered, and bFGF pellet-induced corneal NV was evaluated. RESULTS: NV of the corneal stroma began on day 4 and was sustained through day 21 following bFGF pellet implantation. Progression of vascular endothelial cells correlated with increased VEGF-LacZ expression. Western blot analysis showed increased VEGF expression in the corneal NV zone. Following bFGF pellet implantation, the area of corneal NV in untreated controls was 1.05+/-0.12 mm2 and 1.53+/-0.27 mm2 at days 4 and 7, respectively. This was significantly greater than that of mice treated with VEGF Trap (0.24+/-0.11 mm2 and 0.35+/-0.16 mm2 at days 4 and 7, respectively; p<0.05). CONCLUSIONS: Corneal keratocytes express VEGF after bFGF stimulation and bFGF-induced corneal NV is blocked by intraperitoneal VEGF TrapR1R2 administration. Systemic administration of VEGF TrapR1R2 may have potential therapeutic applications in the management of corneal NV.


Assuntos
Neovascularização da Córnea/prevenção & controle , Modelos Animais de Doenças , Receptores de Fatores de Crescimento/administração & dosagem , Proteínas Recombinantes de Fusão/administração & dosagem , Animais , Western Blotting , Córnea/metabolismo , Córnea/patologia , Neovascularização da Córnea/induzido quimicamente , Neovascularização da Córnea/patologia , Fator 2 de Crescimento de Fibroblastos/toxicidade , Injeções Intraperitoneais , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Fator A de Crescimento do Endotélio Vascular/metabolismo
8.
J Control Release ; 129(2): 117-23, 2008 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-18524409

RESUMO

The objective of this study was to evaluate the effect of vector architecture on DNA condensation, particle stability, and gene transfer efficiency. Two recombinant non-viral vectors with the same amino acid compositions but different architectures, composed of lysine-histidine (KH) repeating units fused to fibroblast growth factor, were genetically engineered. In one vector lysine residues were dispersed (KHKHKHKHKK)(6)-FGF2, whereas in the other they were in clusters (KKKHHHHKKK)(6)-FGF2. Organization of lysine residues in this manner was inspired by the sequence of DNA condensing motifs that exist in nature (e.g., histones) where lysine residues are organized in clusters. These two constructs were compared in terms of DNA condensation and gene transfer efficiency. It was observed that the construct with KH units in clusters was able to condense pDNA into more stable particles with sizes <150 nm making them suitable for cellular uptake via receptor mediated endocytosis. This in turn resulted in five times higher transfection efficiency for the cKH-FGF2. This study demonstrates that in targeted non-viral gene transfer, the vector architecture plays as significant a role as its amino acid sequence. Thus, in the design of the non-viral vectors (synthetic or recombinant) this factor should be considered of paramount importance.


Assuntos
DNA/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Transfecção/métodos , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Clonagem Molecular , DNA/química , Relação Dose-Resposta a Droga , Endocitose , Endossomos/metabolismo , Fator 2 de Crescimento de Fibroblastos/química , Fator 2 de Crescimento de Fibroblastos/genética , Fator 2 de Crescimento de Fibroblastos/toxicidade , Engenharia Genética , Histidina/metabolismo , Humanos , Lisina/metabolismo , Camundongos , Dados de Sequência Molecular , Células NIH 3T3 , Conformação de Ácido Nucleico , Tamanho da Partícula , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Proteínas Recombinantes de Fusão/metabolismo
9.
Invest Ophthalmol Vis Sci ; 49(1): 250-4, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18172099

RESUMO

PURPOSE: To evaluate the influence of some widely used antiglaucoma agents on angiogenesis in a novel rat cornea model. METHODS: Angiogenesis was induced in 32 rats by slow-release polymer pellets containing basic fibroblast growth factor (bFGF) placed in a corneal micropocket. Angiogenesis was later measured and compared in groups of rats given one of four antiglaucoma drug therapies and one control group. The drugs were commercially available preparations of prostaglandins, beta-blockers, alpha-2 agonists, and carbonic anhydrase inhibitors given for 7 days in a manner similar to that used in humans. Growth was measured by calculating the maximum linear vessel growth divided by pellet-limbus distance. RESULTS: Biomicroscopic observation disclosed that all tested animals showed an induction of neovascular reactions in their corneal stroma. The growth index results for the control, latanoprost, dorzolamide, brimonidine, and timolol malate groups were 1.65 +/- 0.16, 1.98 +/- 0.18, 1.85 +/- 0.19, 2.03 +/- 0.38, and 1.65 +/- 0.14, respectively, confirming the hypothesis that topically delivered antiglaucoma drugs modify the normal angiogenic response. Of them, the prostaglandins showed the most prominent angiogenic stimulatory effect (P = 0.03). CONCLUSIONS: This modified micropocket assay of corneal angiogenesis in rats demonstrated the stimulatory effect of several widely used topically delivered antiglaucoma medications on the angiogenic process. The results indicate that the selection of drugs for treating different ophthalmic diseases should take into account their influence on angiogenic processes.


Assuntos
Anti-Hipertensivos/toxicidade , Neovascularização da Córnea/fisiopatologia , Substância Própria/irrigação sanguínea , Modelos Animais de Doenças , Animais , Tartarato de Brimonidina , Substância Própria/efeitos dos fármacos , Implantes de Medicamento , Fator 2 de Crescimento de Fibroblastos/toxicidade , Latanoprosta , Prostaglandinas F Sintéticas/toxicidade , Quinoxalinas/toxicidade , Ratos , Ratos Wistar , Sulfonamidas/toxicidade , Tiofenos/toxicidade , Timolol/toxicidade
10.
Invest Ophthalmol Vis Sci ; 48(11): 5184-90, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17962472

RESUMO

PURPOSE: To evaluate the angiogenic inhibitory effects of an alpha(v)beta(3)/alpha(v)beta(5) integrin antagonist, EMD478761, released from a polymeric implant in a chick chorioallantoic membrane (CAM) assay and laser-induced experimental choroidal neovascularization (CNV) in rats. METHODS: Polyvinyl alcohol-based reservoir implants releasing EMD478761 were designed for placement onto a CAM or intravitreally in rats. In vitro release rates of the implants were measured using HPLC. Angiogenesis was induced on 10-day-old chick embryos by basic fibroblast growth factor (bFGF), and areas of neovascularization were measured. Experimental CNV was induced in the Brown-Norway rat with a diode laser. EMD478761 or sham microimplants were placed within the vitreous chamber of Brown-Norway rats. Two weeks later, areas of CNV were determined by FITC-dextran staining of choroidal flatmounts. RESULTS: Sustained delivery of EMD478761 significantly inhibited bFGF-induced angiogenesis in CAM, as determined by a reduction in angiogenesis areas, without drug toxicity to the normal CAM vasculature. In an experimental rat model, intravitreal EMD478761 implants significantly suppressed laser-induced CNV compared with intravitreal sham implants, with the mean area reduced by 63% (P < 0.05). CONCLUSIONS: Sustained delivery of EMD478761demonstrates potent antiangiogenic properties in vivo. These results suggest that an EMD478761 implant may be beneficial in the treatment of neovascular ocular diseases.


Assuntos
Inibidores da Angiogênese/administração & dosagem , Benzoxazinas/administração & dosagem , Membrana Corioalantoide/irrigação sanguínea , Neovascularização de Coroide/prevenção & controle , Integrina alfaVbeta3/antagonistas & inibidores , Integrinas/antagonistas & inibidores , Neovascularização Patológica/tratamento farmacológico , Receptores de Vitronectina/antagonistas & inibidores , Animais , Embrião de Galinha , Neovascularização de Coroide/patologia , Cromatografia Líquida de Alta Pressão , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos , Implantes de Medicamento , Fator 2 de Crescimento de Fibroblastos/toxicidade , Injeções , Terapia a Laser , Masculino , Neovascularização Patológica/induzido quimicamente , Ratos , Ratos Endogâmicos BN , Corpo Vítreo/efeitos dos fármacos
11.
Am J Pathol ; 170(2): 764-73, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17255342

RESUMO

Corneal neovascularization is a vision-threatening condition caused by various ocular pathological conditions. The aim of this study was to evaluate the function of the ephrin ligands and Eph receptors in vitro and in vivo in corneal angiogenesis in a mouse model. The Eph tyrosine kinase receptors and their ligands, ephrins, are expressed on the cell surface. The functions of Eph and ephrins have been shown to regulate axonal guidance, segmentation, cell migration, and angiogenesis. Understanding the roles of Eph and ephrin in corneal angiogenesis may provide a therapeutic intervention for the treatment of angiogenesis-related disorders. Immunohistochemical studies demonstrated that ephrinB1 and EphB1 were expressed in basic fibroblast growth factor (bFGF)-induced vascularized corneas. EphB1 was specifically colocalized with vascular endothelial marker CD31 surrounded by type IV collagen. EphrinB1 was expressed in corneal-resident keratocytes and neutrophils. Recombinant ephrinB1-Fc, which induces EphB receptor activation, enhanced bFGF-induced tube formation in an in vitro aortic ring assay and promoted bFGF-induced corneal angiogenesis in vivo in a corneal pocket assay. Synergistically enhanced and sustained activation of extracellular signal-regulated kinase was noted in vascular endothelial cell lines after stimulation with ephrin B1 and bFGF combinations. These results suggest that ephrinB1 plays a synergistic role in corneal neovascularization.


Assuntos
Neovascularização da Córnea/metabolismo , Efrina-B1/biossíntese , Fator 2 de Crescimento de Fibroblastos/toxicidade , Regulação da Expressão Gênica/efeitos dos fármacos , Receptores da Família Eph/biossíntese , Animais , Aorta/metabolismo , Bovinos , Colágeno Tipo IV/biossíntese , Neovascularização da Córnea/induzido quimicamente , Neovascularização da Córnea/tratamento farmacológico , Neovascularização da Córnea/patologia , Ativação Enzimática/efeitos dos fármacos , Efrina-B1/agonistas , Efrina-B1/farmacologia , Fator 2 de Crescimento de Fibroblastos/agonistas , Humanos , Camundongos , Técnicas de Cultura de Órgãos , Molécula-1 de Adesão Celular Endotelial a Plaquetas/biossíntese
12.
Invest Ophthalmol Vis Sci ; 46(10): 3502-6, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16186326

RESUMO

PURPOSE: Bone-marrow (BM)-derived hematopoietic precursor cells are thought to participate in the growth of blood vessels during postnatal vasculogenesis. In this investigation, multichannel laser scanning confocal microscopy and quantitative image analysis were used to study the fate of BM-derived hematopoietic precursor cells in corneal neovascularization. METHODS: A BM-reconstituted mouse model was used in which the BM from enhanced green fluorescent protein (GFP)-positive mice was transplanted into C57BL/6 mice. Basic fibroblast growth factor (bFGF) was used to induce corneal neovascularization in mice. The vasculogenic potential of adult BM-derived cells and their progeny were tested in this in vivo model. Seventy-two histologic sections selected by systematic random sampling from four mice were immunostained and imaged with a confocal microscope and analyzed with image-analysis software. RESULTS: BM-derived endothelial cells did not contribute to bFGF-induced neovascularization in the cornea. BM-derived periendothelial vascular mural cells (pericytes) were detected at sites of neovascularization, whereas endothelial cells of blood vessels originated from preexisting blood vessels in limbal capillaries. Fifty three percent of all neovascular pericytes originated from BM, and 47% of them originated from preexisting corneoscleral limbus capillaries. Ninety-six percent and 92% of BM-derived pericytes also expressed CD45 and CD11b, respectively, suggesting their hematopoietic origin from the BM. CONCLUSIONS: Pericytes of new corneal vessels have a dual source: BM and preexisting limbal capillaries. These findings establish BM as a significant effector organ in corneal disorders associated with neovascularization.


Assuntos
Neovascularização da Córnea/fisiopatologia , Células-Tronco Hematopoéticas/fisiologia , Pericitos/fisiologia , Animais , Antígeno CD11b/metabolismo , Neovascularização da Córnea/induzido quimicamente , Neovascularização da Córnea/metabolismo , Modelos Animais de Doenças , Endotélio Linfático/fisiologia , Endotélio Vascular/fisiologia , Feminino , Fator 2 de Crescimento de Fibroblastos/toxicidade , Proteínas de Fluorescência Verde/metabolismo , Células-Tronco Hematopoéticas/citologia , Imuno-Histoquímica , Antígenos Comuns de Leucócito/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Pericitos/citologia
13.
Mol Vis ; 11: 28-35, 2005 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-15660022

RESUMO

PURPOSE: This study was designed to investigate the effects of the locally supplied endogenous angiogenesis inhibitor vasostatin (VS) on corneal angiogenesis. METHODS: Recombinant VS was expressed and purified. The effects of VS on the proliferation of endothelial cells were investigated using the methyl thiazolyl tetrazolium (MTT) assay in the absence or presence of angiogenic factors such as basic fibroblast growth factor (bFGF) or vascular endothelial growth factor (VEGF). Corneal neovascularization was induced by implantation of hydron pellets containing bFGF in rat corneal micropockets. The potency of VS to inhibit corneal angiogenesis was investigated by incorporation of VS with bFGF in hydron pellets or topical application of VS containing eye drops to rat eyes implanted with bFGF pellets. The extent of corneal neovascularization was evaluated by microscopic and histological analyses. RESULTS: VS potently inhibited the growth of endothelial cells in the absence or presence of angiogenic factors such as bFGF or VEGF. In the rat corneal micropocket assay, concurrent incorporation of VS abolished the bFGF induced neovascularization. When formulated in a methylcellulose eye drop, VS remained intact and functional in a 4 degrees C solution for more than 7 days. Topical application of VS eye drops potently inhibited bFGF induced neovascularization in rat corneas. CONCLUSIONS: The present study effectively demonstrated the potential feasibility of local application of VS for treatment of corneal angiogenesis.


Assuntos
Inibidores da Angiogênese/administração & dosagem , Calreticulina/administração & dosagem , Neovascularização da Córnea/prevenção & controle , Fragmentos de Peptídeos/administração & dosagem , Administração Tópica , Inibidores da Angiogênese/genética , Inibidores da Angiogênese/uso terapêutico , Animais , Aorta/citologia , Calreticulina/genética , Calreticulina/uso terapêutico , Bovinos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Clonagem Molecular , Neovascularização da Córnea/induzido quimicamente , Neovascularização da Córnea/patologia , Eletroforese em Gel de Poliacrilamida , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/toxicidade , Expressão Gênica , Masculino , Soluções Oftálmicas , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/uso terapêutico , Epitélio Pigmentado Ocular/citologia , Epitélio Pigmentado Ocular/efeitos dos fármacos , Coelhos , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/uso terapêutico , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator A de Crescimento do Endotélio Vascular/toxicidade
14.
Yan Ke Xue Bao ; 21(1): 56-61, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17162918

RESUMO

PURPOSE: To investigate the effects of culture medium of human amniotic membrane (AM) on corneal neovascularization (CNV) induced by basic fibroblast growth factor (bFGF) in mice. METHODS: Culture medium of amniotic membrane was prepared by cultivating AM (with epithelium side up) in EGM basic medium for 3 days, and was collected separately to three groups, e.g. control (EGM only), AM with epithelium (AM) and AM without epithelium (De-AM). Corneal neovascularization was induced in mice by using micropocket assay with Hydron polymer pellets containing 100 ng bFGF. Migration and proliferation of human umbilical cord vein endothelial cells (HUVEC) were performed in Boyden chambers and by using the CyQUANT fluorescence binding assay respectively. The levels of tissue inhibitors of metalloproteinase 1 and 2 (TIMP1, TIMP2) in culture medium were determined by ELISA assay. RESULTS: CNV induced by bFGF was significantly suppressed by culture medium of amniotic membrane. When the medium was applied as an eyedrop 4 times a day for 7 days, the area of CNV was (2.48+/-0.76) mm(2),(0.64+/-0.52) mm(2) and (1.96+/-0.65) mm(2) in control, AM and De-AM group respectively. The migration and proliferation of HUVEC were strongly inhibited by culture medium of AM with epithelium, while the De-AM had no effect on the migration of HUVEC cells. The high level of TIMP2 was found in AM group, but not in De-AM group, while there was no difference in the amount of TIMP1 in medium among three groups. CONCLUSION: Culture medium of amniotic membrane significantly suppresses the corneal nevovascularization induced by bFGF. The mechanism of which at least in part is that high level of TIMP2 protein secreted or released into the culture medium of AM and inhibition of migration and growth of vascular endothelial cells.


Assuntos
Âmnio/metabolismo , Neovascularização da Córnea/prevenção & controle , Meios de Cultura/farmacologia , Neovascularização Patológica/prevenção & controle , Inibidor Tecidual de Metaloproteinase-2/metabolismo , Âmnio/citologia , Animais , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Neovascularização da Córnea/induzido quimicamente , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Endotélio Vascular/patologia , Fator 2 de Crescimento de Fibroblastos/toxicidade , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Veias Umbilicais/patologia
15.
Oncol Rep ; 10(3): 617-21, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12684633

RESUMO

Conjugated linoleic acid (CLA) is a potent inhibitor of mammary carcinogenesis. Cancer cells produce various angiogenic factors which stimulate host vascular endothelial cell mitogenesis and chemotaxis for their growth and metastasis. Basic fibroblast growth factor (bFGF) is a potent angiogenic factor that is expressed in many tumors. In this study, we found that CLA decreased bFGF-induced endothelial cell proliferation and DNA synthesis in a dose-dependent manner. However, CLA did not inhibit endothelial cell migration. Furthermore, CLA showed a potent inhibitory effect on embryonic vasculogenesis and bFGF-induced angiogenesis in vivo. Collectively, these results suggest that CLA selectively inhibits the active proliferating endothelial cells induced by bFGF, which may explain its anti-carcinogenic properties in vivo.


Assuntos
Inibidores da Angiogênese/farmacologia , Endotélio Vascular/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/toxicidade , Ácidos Linoleicos Conjugados/farmacologia , Neovascularização Patológica/tratamento farmacológico , Alantoide/irrigação sanguínea , Animais , Capilares , Divisão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Embrião de Galinha , Colágeno , Replicação do DNA/efeitos dos fármacos , Combinação de Medicamentos , Endotélio Vascular/citologia , Laminina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/induzido quimicamente , Proteoglicanas , Timidina/metabolismo
16.
Clin Exp Metastasis ; 19(2): 145-53, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-11964078

RESUMO

A novel naturally occurring antiangiogenic agent isolated from cartilage, referred to as Neovastat (AE-941), was examined for its efficacy against tumor neovascularization and progression. Exposure to Neovastat results in ex ovo antiangiogenic properties in the chorioallantoid membrane of chicken embryo (71% decrease in the angiogenic index as compared to the basic fibroblast growth factor (bFGF) treated control embryos, P < 0.0001). Oral administration of Neovastat inhibits bFGF-induced angiogenesis in the Matrigel mouse model (87.5% decrease in hemoglobin as compared to the bFGF-treated control implants, P < 0.0001). Neovastat also induces a dose response decrease of lung metastases in the Lewis lung carcinoma model (oral administration; 69.1% of inhibition obtained at the maximal dose of 0.5 ml/day, P < 0.0001). Combined with a sub-optimal dose of cisplatinum (2 mg/kg, i.p.), Neovastat (0.5 ml/day) improved the therapeutic index by increasing the antimetastatic efficacy and by exerting a protective activity against cisplatinum-induced body weight loss and myelosuppression. In summary, our experimental data provide evidence of antiangiogenic and antimetastatic properties of Neovastat, following oral administration.


Assuntos
Inibidores da Angiogênese/farmacologia , Antineoplásicos/farmacologia , Vasos Sanguíneos/efeitos dos fármacos , Neovascularização Patológica/prevenção & controle , Extratos de Tecidos/farmacologia , Administração Oral , Inibidores da Angiogênese/isolamento & purificação , Animais , Antineoplásicos/isolamento & purificação , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Peso Corporal/efeitos dos fármacos , Carcinoma Pulmonar de Lewis/irrigação sanguínea , Carcinoma Pulmonar de Lewis/tratamento farmacológico , Carcinoma Pulmonar de Lewis/patologia , Cartilagem/química , Embrião de Galinha , Cisplatino/administração & dosagem , Colágeno , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Feminino , Fator 2 de Crescimento de Fibroblastos/toxicidade , Humanos , Laminina , Camundongos , Camundongos Endogâmicos BALB C , Proteoglicanas , Extratos de Tecidos/isolamento & purificação
17.
Cornea ; 21(1): 62-7, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11805510

RESUMO

PURPOSE: To examine the applicability of culture supernatant of human amniotic cells on basic fibroblast growth factor (bFGF)-induced corneal neovascularization. METHODS: Human amniotic epithelial and mesenchymal cells (AC) were obtained from human amniotic membranes by digesting with collagenase and maintained in serum-containing medium. The AC preparations predominantly contained cytokeratin-positive cells (91.2 +/- 3.1%, n = 4). The culture supernatant was prepared by cultivating AC in serum-free medium for 24 hours. Neovascularization was obtained by a micropocket assay with Hydron pellets containing bFGF. Migration assay was carried out by a double-chamber method using human umbilical vein endothelial cells (HUVEC). Cell growth assay was done by MTT assay using HUVEC. RESULTS: Basic fibroblast growth factor-induced corneal neovascularization was significantly reduced by administration of AC culture supernatant. When either control or AC culture supernatant was administered three times per day for 10 days, the area with neovascularization was 13.2 +/- 3.2 mm2 and 4.0 +/- 1.4 mm2 for the control and AC culture supernatant-treated eyes, respectively (n = 7, p = 0.021). AC culture supernatant strongly inhibited bFGF-induced migration and cell growth of HUVEC. CONCLUSIONS: Amniotic cell culture supernatant contains potent inhibitors of neovascularization. This effect is explained in part by inhibition of migration and cell growth of vascular endothelial cells. AC culture supernatant may be applicable for treatment of corneal diseases with neovascularization.


Assuntos
Âmnio/citologia , Inibidores da Angiogênese/farmacologia , Córnea/efeitos dos fármacos , Neovascularização da Córnea/prevenção & controle , Meios de Cultura/farmacologia , Animais , Técnicas de Cultura de Células , Divisão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Córnea/patologia , Neovascularização da Córnea/induzido quimicamente , Neovascularização da Córnea/patologia , Endotélio Vascular/citologia , Feminino , Fator 2 de Crescimento de Fibroblastos/toxicidade , Humanos , Coelhos
19.
Curr Eye Res ; 22(2): 140-7, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11402391

RESUMO

PURPOSE: Vascular endothelial growth factor (VEGF) causes widespread retinal vascular dilation, produces breakdown of the blood-retinal barrier, and is implicated in ocular neovascularization (NV). Basic fibroblast growth factor (bFGF) also has been implicated in the production of ocular NV. This study was performed to investigate the ability of simultaneous sustained intravitreal release of both VEGF and bFGF to induce robust retinal NV in the rabbit. METHODS: Intravitreal implantation of sustained-release Hydron polymeric pellets containing both 20 microg of VEGF and 20 microg of bFGF was performed on adult male Dutch belted rabbits. In other animals either 20 microg or 50 microg bFGF-containing pellets was implanted intravitreally; also, either 20 microg VEGF or 50 microg VEGF-containing pellets was implanted. Control rabbits received either blank polymeric pellets or a pellet containing 30 microg bovine serum albumin. Eyes were examined by indirect ophthalmoscopy after surgery at 24 hrs, 48 hrs, 4 days, 7 days, 14 days, 21 days, and 28 days. Findings were documented by color fundus photography and fluorescein angiography (FA). Eyes were enucleated and prepared for histologic analysis at 28 days following intravitreal implantation of the VEGF/bFGF-containing pellets. RESULTS: In all eyes implanted with VEGF/bFGF pellets, dilation and tortuosity of existing blood vessels were observed within 48 hrs after pellet implantation. The progression of retinal vascular changes was rapid and occurred over the entire optic disk and medullary rays between 4 and 7 days. Hemorrhage occurred as early as 14 days after VEGF/bFGF pellet implantation. In eyes with massive hemorrhage, total traction retinal detachment developed after the second week. The presence of abnormal tissues at the vitreo-retinal interface within 28 days was demonstrated by light microscopy while FA showed profuse leakage of dye from anomalous vessels within the first week. Neither bFGF-exposed eyes nor control eyes showed any vascular changes. Eyes that received only VEGF-containing pellets exhibited tortuosity of existing vessels, but neither hemorrhaging nor retinal detachment occurred. CONCLUSIONS: These results demonstrate that retinal vascular changes leading to hemorrhaging is produced rapidly in the rabbit by simultaneous intravitreal release of both VEGF and bFGF. Understanding how these growth factors induce retinal NV may suggest novel therapeutic treatment strategies.


Assuntos
Fatores de Crescimento Endotelial/toxicidade , Fator 2 de Crescimento de Fibroblastos/toxicidade , Linfocinas/toxicidade , Hemorragia Retiniana/induzido quimicamente , Neovascularização Retiniana/induzido quimicamente , Vasos Retinianos/efeitos dos fármacos , Animais , Preparações de Ação Retardada , Combinação de Medicamentos , Implantes de Medicamento , Angiofluoresceinografia , Fundo de Olho , Masculino , Oftalmoscopia , Coelhos , Descolamento Retiniano/induzido quimicamente , Descolamento Retiniano/patologia , Hemorragia Retiniana/patologia , Neovascularização Retiniana/patologia , Vasos Retinianos/patologia , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular , Corpo Vítreo
20.
Teratology ; 62(4): 189-94, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10992260

RESUMO

BACKGROUND: This laboratory previously demonstrated that placement of fibroblast growth factor-2 (FGF-2)-soaked beads adjacent to the developing ventricle at stage 24 caused cardiovascular anomalies by embryonic day 15. We sought to characterize early cellular changes that may suggest mechanisms for the abnormalities observed at day 15. Because levels of both myocyte proliferation and immunohistochemically detectable endogenous FGF-2 begin to decline before stage 24 in untreated embryos, it was of interest to determine whether exogenous FGF-2 might maintain cardiac myocyte proliferation at or near peak levels. METHODS: Chick embryos were incubated to stage 18 (2.8 days), at which time beads soaked in phosphate-buffered saline (PBS) or 100 microg/ml FGF-2 were placed adjacent to the developing ventricle and development was allowed to continue. After 3 days (stage 29), bromodeoxyuridine (BrdU) was applied to mark dividing cells, followed by double fluorescent assessments to detect relative numbers of dividing and nondividing cells. RESULTS: Quantitative image analysis, using Metamorph software, showed that exogenous FGF-2 caused a 62% increase in the overall number of dividing cells (P < 0.01), concomitant with a 25% increase in total cell number (cell density: P < 0.05). Expressed in relative terms, these changes corresponded to a 25% increase in the proliferation labeling index: 30% of all cells were proliferating in FGF-treated hearts, in contrast with only 24% in control hearts. CONCLUSIONS: Taken together, these data suggest that an FGF-induced imbalance in myocardial cell proliferation at early developmental stages of heart development causes cardiovascular anomalies during late embryogenesis.


Assuntos
Anormalidades Induzidas por Medicamentos/etiologia , Anormalidades Cardiovasculares/induzido quimicamente , Embrião de Galinha/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/toxicidade , Coração/embriologia , Miocárdio/patologia , Anormalidades Induzidas por Medicamentos/patologia , Animais , Bromodesoxiuridina/metabolismo , Anormalidades Cardiovasculares/patologia , Contagem de Células , Divisão Celular/efeitos dos fármacos , Técnica Indireta de Fluorescência para Anticorpo , Coração/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA