Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Ophthalmology ; 131(9): 1083-1093, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38423215

RESUMEN

PURPOSE: Cotoretigene toliparvovec (BIIB112/AAV8-RPGR) is an investigational vector-based gene therapy designed to provide a full-length, codon-optimized retinitis pigmentosa GTPase regulator (RPGR) protein to individuals with RPGR-associated X-linked retinitis pigmentosa (XLRP). We assessed efficacy and tolerability of cotoretigene toliparvovec subretinal gene therapy. DESIGN: Part 2 of the XIRIUS trial (ClinicalTrials.gov identifier, NCT03116113) was a phase 2/3, 12-month, randomized (1:1:1) dose-expansion study. PARTICIPANTS: Male patients ≥10 years of age with RPGR-associated XLRP were included. METHODS: Participants were randomized 1:1:1 to receive low-dose subretinal cotoretigene toliparvovec (5 × 1010 vector genomes/eye), high-dose cotoretigene toliparvovec (2.5 × 1011 vector genomes/eye) or to be an untreated control participant. MAIN OUTCOME MEASURES: The primary end point was the percentage of participants meeting microperimetry responder criteria (≥ 7-dB improvement at ≥ 5 of 16 central loci). Secondary end points included change from baseline in retinal sensitivity at the central 16 loci and the entire 68 loci at 12 months and change from baseline in low-luminance visual acuity (LLVA) at 12 months, as well as the proportion of eyes with a ≥ 15-Early Treatment Diabetic Retinopathy Study ETDRS letter LLVA and ≥ 10-ETDRS letter LLVA change from baseline at month 12. RESULTS: Because of the impact of the COVID-19 pandemic, enrollment ended before reaching the initial target, leaving the trial underpowered. Twenty-nine participants were included (low-dose group, n = 10; high-dose group, n = 10; control group, n = 9). At month 12, the percentage of participants meeting microperimetry responder criteria was not significantly different between either cotoretigene toliparvovec group (low dose, 37.5% [P = 0.3181]; high dose, 25.0% [P = 0.5177]) and the control group (22.2%). However, the mean change from baseline in microperimetry sensitivity improved significantly with the low-dose group versus the control group at month 12 (P = 0.0350). Significant improvement in LLVA occurred in the low-dose group versus the control group at month 12 (33.3% difference [80% confidence interval, 14.7%-55.2%]; P = 0.0498). Three ocular-related serious adverse events (SAEs) occurred in the low-dose group versus 7 SAEs in the high-dose group. CONCLUSIONS: The primary microperimetry end point was not met. Significant improvements in LLVA and mean microperimetry were observed compared with controls and fewer SAEs occured with low-dose compared with high dose cotoretigene toliparvovec. FINANCIAL DISCLOSURE(S): Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.


Asunto(s)
Proteínas del Ojo , Terapia Genética , Vectores Genéticos , Proteínas Recombinantes , Retinitis Pigmentosa , Agudeza Visual , Adolescente , Adulto , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven , Dependovirus/genética , Electrorretinografía , Proteínas del Ojo/administración & dosificación , Proteínas del Ojo/efectos adversos , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/efectos adversos , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/efectos adversos , Retinitis Pigmentosa/tratamiento farmacológico , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/fisiopatología , Tomografía de Coherencia Óptica , Agudeza Visual/fisiología , Campos Visuales/fisiología
2.
Exp Eye Res ; 238: 109743, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38056550

RESUMEN

Pigment epithelium-derived factor (PEDF) is widely recognized as a neuroprotective factor expressed in the retina and has shown therapeutic potential in several retinal diseases. Our study aimed to identify the neuroprotective fragment in PEDF and investigate its protective activity in retinas under ischemia-reperfusion (IR) condition. We synthesized a series of shorter synthetic peptides, 6-mer (Ser93-Gln98) and its d-form variant (6 dS) derived from the 44-mer (Val78-Thr121; a PEDF neurotrophic fragment), to determine their cytoprotective activity in IR injury, which was induced in rat retinas by injection of saline into the anterior chamber to increase the intraocular pressure (IOP) followed by reperfusion. We found the cytoprotective effect of 6-mer on glutamate-treated Neuro-2a cells and tert-butyl hydroperoxide (tBHP)-treated 661W cells were 2.6-fold and 1.5-fold higher than the 44-mer, respectively. The cytoprotective effect was blocked by a chemical inhibitor atglistatin and blocking antibody targeting PEDF receptor (PEDF-R). IR induced several impairments in retina, including cell apoptosis, activation of microglia/macroglia, degeneration of retinal capillaries, reduction in electroretinography (ERG) amplitudes, and retinal atrophy. Such IR injuries were ameliorated by treatment with 6-mer and 6 dS eye drops. Also, the neuroprotective activity of 6-mer and 6 dS in ischemic retinas were dramatically reversed by atglistatin preconditioning. Taken together, our data demonstrate smallest neuroprotective fragment of PEDF has potential to treat retinal degeneration-related diseases.


Asunto(s)
Proteínas del Ojo , Factores de Crecimiento Nervioso , Daño por Reperfusión , Retina , Retinitis , Serpinas , Animales , Ratas , Conejos , Factores de Crecimiento Nervioso/administración & dosificación , Factores de Crecimiento Nervioso/química , Factores de Crecimiento Nervioso/metabolismo , Proteínas del Ojo/administración & dosificación , Proteínas del Ojo/química , Proteínas del Ojo/metabolismo , Serpinas/administración & dosificación , Serpinas/química , Serpinas/metabolismo , Retina/metabolismo , Retina/patología , Daño por Reperfusión/metabolismo , Citoprotección , Apoptosis , Neuronas/metabolismo , Retinitis/tratamiento farmacológico , Retinitis/metabolismo , Administración Tópica , Péptidos/administración & dosificación , Péptidos/metabolismo
3.
Graefes Arch Clin Exp Ophthalmol ; 257(8): 1709-1717, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31222405

RESUMEN

BACKGROUND: Pigment epithelium-derived factor (PEDF)-derived 34-mer peptide (PEDF34, Asp44-Asn77) has anti-angiogenic activity but has limitations in clinical application because of an inverted bell-shaped dose-effect relationship and a short half-life. In this study, we attempted to mitigate these problems by mixing PEDF34 with type I collagen. METHODS: The anti-angiogenic activity of the PEDF34/atelocollagen mixture was evaluated by HUVEC tube formation assay and in a laser-induced choroidal neovascular (CNV) mouse model. PEDF34 and/or collagen were administrated using intravitreal injections or eye drops. CNV lesion size was quantified using FITC-dextran-perfused retinal whole mounts. Western blot analysis and inhibitor assays were used to define the action mechanisms of PEDF34 and the mixture. RESULTS: Collagen broadened the effective dose range of PEDF34 in the tube formation assay by > 250 times (from 0.2 to 50 nM). In the CNV model, five intravitreal injections of PEDF34 were required for therapeutic effect, whereas the mixture had a significant therapeutic effect following a single injection. Eye drops of the mixture showed significantly stronger CNV-suppressive effects than drops of PEDF34 alone. The anti-angiogenic activity of PEDF34 might be mediated by inhibition of ERK and JNK activation by VEGF, and collagen potentiated these effects. CONCLUSIONS: Collagen can serve as a carrier and reservoir of PEDF34. PEDF peptide/collagen mixture is easy to prepare than conventional methods for maintaining the therapeutic effect of PEDF peptide.


Asunto(s)
Coroides/patología , Neovascularización Coroidal/tratamiento farmacológico , Colágeno Tipo I/administración & dosificación , Proteínas del Ojo/administración & dosificación , Factores de Crecimiento Nervioso/administración & dosificación , Serpinas/administración & dosificación , Animales , Células Cultivadas , Neovascularización Coroidal/metabolismo , Neovascularización Coroidal/patología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Rayos Láser/efectos adversos , Ratones , Ratones Endogámicos C57BL , Soluciones Oftálmicas , Inhibidores de Proteasas/administración & dosificación , Retina/patología
4.
J Biol Chem ; 292(45): 18486-18499, 2017 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-28972155

RESUMEN

The cornea is densely innervated to sustain the integrity of the ocular surface. Corneal nerve damage produced by aging, diabetes, refractive surgeries, and viral or bacterial infections impairs tear production, the blinking reflex, and epithelial wound healing, resulting in loss of transparency and vision. A combination of the known neuroprotective molecule, pigment epithelium-derived factor (PEDF) plus docosahexaenoic acid (DHA), has been shown to stimulate corneal nerve regeneration, but the mechanisms involved are unclear. Here, we sought to define the molecular events of this effect in an in vivo mouse injury model. We first confirmed that PEDF + DHA increased nerve regeneration in the mouse cornea. Treatment with PEDF activates the phospholipase A2 activity of the PEDF-receptor (PEDF-R) leading to the release of DHA; this free DHA led to enhanced docosanoid synthesis and induction of bdnf, ngf, and the axon growth promoter semaphorin 7a (sema7a), and as a consequence, their products appeared in the mouse tears. Surprisingly, corneal injury and treatment with PEDF + DHA induced transcription of neuropeptide y (npy), small proline-rich protein 1a (sprr1a), and vasoactive intestinal peptide (vip) in the trigeminal ganglia (TG). The PEDF-R inhibitor, atglistatin, blocked all of these changes in the cornea and TG. In conclusion, we uncovered here an active cornea-TG axis, driven by PEDF-R activation, that fosters axon outgrowth in the cornea.


Asunto(s)
Córnea/inervación , Ácidos Docosahexaenoicos/uso terapéutico , Proteínas del Ojo/uso terapéutico , Modelos Neurológicos , Factores de Crecimiento Nervioso/uso terapéutico , Regeneración Nerviosa/efectos de los fármacos , Receptores de Neuropéptido/agonistas , Serpinas/uso terapéutico , Nervio Trigémino/efectos de los fármacos , Administración Oftálmica , Animales , Córnea/efectos de los fármacos , Córnea/patología , Córnea/fisiología , Ácidos Docosahexaenoicos/administración & dosificación , Ácidos Docosahexaenoicos/metabolismo , Quimioterapia Combinada , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/farmacología , Proteínas del Ojo/administración & dosificación , Proteínas del Ojo/agonistas , Proteínas del Ojo/antagonistas & inhibidores , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Proteínas del Ojo/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Inyecciones Intraperitoneales , Masculino , Ratones Endogámicos C57BL , Factores de Crecimiento Nervioso/administración & dosificación , Factores de Crecimiento Nervioso/farmacología , Proteínas del Tejido Nervioso/agonistas , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/metabolismo , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Técnicas de Cultivo de Órganos , Compuestos de Fenilurea/administración & dosificación , Compuestos de Fenilurea/farmacología , Receptores de Neuropéptido/antagonistas & inhibidores , Receptores de Neuropéptido/metabolismo , Serpinas/administración & dosificación , Serpinas/farmacología , Ganglio del Trigémino/efectos de los fármacos , Ganglio del Trigémino/patología , Ganglio del Trigémino/fisiología , Nervio Trigémino/patología , Nervio Trigémino/fisiología , Traumatismos del Nervio Trigémino/tratamiento farmacológico
5.
Mol Pharm ; 15(7): 2539-2547, 2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29912566

RESUMEN

In the present study, tissue distribution and the therapeutic effect of topically applied cyclosporine A (CsA)-loaded methoxy-poly(ethylene-glycol)-hexyl substituted poly(lactic acid) (mPEGhexPLA) nanocarriers (ApidSOL) on experimental autoimmune uveitis (EAU) were investigated. The CsA-loaded mPEGhexPLA nanocarrier was tolerated well locally and showed no signs of immediate toxicity after repeated topical application in mice with EAU. Upon unilateral CsA treatment, CsA accumulated predominantly in the corneal and sclera-choroidal tissue of the treated eye and in lymph nodes (LN). This regimen reduced EAU severity in treated eyes compared to PBS-treated controls. This improvement was accompanied by reduced T-cell count, T-cell proliferation, and IL-2 secretion of cells from ipsilateral LN. In conclusion, topical treatment with CsA-loaded mPEGhexPLA nanocarriers significantly improves the outcome of EAU.


Asunto(s)
Enfermedades Autoinmunes/tratamiento farmacológico , Ciclosporina/administración & dosificación , Portadores de Fármacos/química , Inmunosupresores/administración & dosificación , Uveítis/tratamiento farmacológico , Administración Oftálmica , Animales , Enfermedades Autoinmunes/inmunología , Modelos Animales de Enfermedad , Proteínas del Ojo/administración & dosificación , Proteínas del Ojo/inmunología , Femenino , Humanos , Ratones , Nanopartículas/química , Poliésteres/química , Polietilenglicoles/química , Proteínas de Unión al Retinol/administración & dosificación , Proteínas de Unión al Retinol/inmunología , Resultado del Tratamiento , Uveítis/inmunología
6.
Exp Eye Res ; 161: 153-162, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28642110

RESUMEN

Herpes simplex virus type-1 (HSV-1) infection leads to impaired corneal sensation and, in severe cases, to corneal ulceration, melting and perforation. Here, we explore the potential therapeutic action of pigment epithelial-derived factor (PEDF) plus docosahexaenoic acid (DHA) on corneal inflammation and nerve regeneration following HSV-1 infection. Rabbits inoculated with 100,000 PFU/eye of HSV-1 strain 17Syn+ were treated with PEDF + DHA or vehicle. PEDF + DHA treatment resulted in a biphasic immune response with stronger infiltration of CD4+T cells, neutrophils and macrophages at 7-days post-treatment (p.t.) that was significantly decreased by 14 days, compared to the vehicle-treated group. Screening of 14 immune-related genes by q-PCR showed that treatment induced higher expression of IFN-γ and CCL20 and inhibition of IL-18 by 7 days in the cornea. PEDF + DHA-treated animals developed less dendritic corneal lesions, opacity and neovascularization. Corneal nerve density increased at 12-weeks p.t. with functional recovery of corneal sensation. Treatment with PEDF + DHA that was postponed by 3 weeks also showed increased nerve density when compared to vehicle. Our data demonstrate that PEDF + DHA promotes resolution of the inflammatory response to the virus and, most importantly, induces regeneration of damaged corneal nerves vital for maintaining ocular surface homeostasis.


Asunto(s)
Córnea/inervación , Ácidos Docosahexaenoicos/uso terapéutico , Proteínas del Ojo/uso terapéutico , Queratitis Herpética/tratamiento farmacológico , Factores de Crecimiento Nervioso/uso terapéutico , Regeneración Nerviosa/efectos de los fármacos , Serpinas/uso terapéutico , Nervio Trigémino/fisiología , Administración Tópica , Animales , Linfocitos T CD4-Positivos/inmunología , Citocinas/genética , Modelos Animales de Enfermedad , Ácidos Docosahexaenoicos/administración & dosificación , Quimioterapia Combinada , Proteínas del Ojo/administración & dosificación , Femenino , Herpesvirus Humano 1/fisiología , Inflamación , Queratitis Herpética/inmunología , Queratitis Herpética/fisiopatología , Macrófagos/inmunología , Masculino , Factores de Crecimiento Nervioso/administración & dosificación , Neutrófilos/inmunología , Soluciones Oftálmicas , Conejos , Reacción en Cadena en Tiempo Real de la Polimerasa , Serpinas/administración & dosificación
7.
Clin Exp Immunol ; 183(2): 280-93, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26400205

RESUMEN

The aim of this study was to examine whether macrophage migration inhibitory factor (MIF) could exaggerate inflammatory response in a mouse model of experimental autoimmune uveitis (EAU) and to explore the underlying mechanism. Mutant serotype 8 adeno-associated virus (AAV8) (Y733F)-chicken ß-actin (CBA)-MIF or AAV8 (Y733F)-CBA-enhanced green fluorescent protein (eGFP) vector was delivered subretinally into B10.RIII mice, respectively. Three weeks after vector delivery, EAU was induced with a subcutaneous injection of a mixture of interphotoreceptor retinoid binding protein (IRBP) peptide with CFA. The levels of proinflammatory cytokines were detected by real-time polymerase chain reaction (PCR) and enzyme-linked immunosorbent assay (ELISA). Retinal function was evaluated with electroretinography (ERG). We found that the expression of MIF and its two receptors CD74 and CD44 was increased in the EAU mouse retina. Compared to AAV8.CBA.eGFP-injected and untreated EAU mice, the level of proinflammatory cytokines, the expression of Notch1, Notch4, delta-like ligand 4 (Dll4), Notch receptor intracellular domain (NICD) and hairy enhancer of split-1 (Hes-1) increased, but the ERG a- and b-wave amplitudes decreased in AAV8.CBA.MIF-injected EAU mice. The Notch inhibitor N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester (DAPT) reduced the expression of NICD, Hes-1 and proinflammatory cytokines. Further, a MIF antagonist ISO-1 attenuated intraocular inflammation, and inhibited the differentiation of T helper type 1 (Th1) and Th17 in EAU mice. We demonstrated that over-expression of MIF exaggerated ocular inflammation, which was associated with the activation of the Notch signalling. The expression of both MIF and its receptors are elevated in EAU mice. Over-expression of MIF exaggerates ocular inflammation, and this exaggerated inflammation is associated with the activation of the Notch signalling and Notch pathway. Our data suggest that the MIF-Notch axis may play an important role in the pathogenesis of EAU. Both the MIF signalling pathways may be promising targets for developing novel therapeutic interventions for uveitis.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Oxidorreductasas Intramoleculares/fisiología , Factores Inhibidores de la Migración de Macrófagos/fisiología , Receptores Notch/fisiología , Retina/inmunología , Uveítis/inmunología , Animales , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/terapia , Citocinas/genética , Dependovirus/genética , Modelos Animales de Enfermedad , Electrorretinografía , Ensayo de Inmunoadsorción Enzimática , Proteínas del Ojo/administración & dosificación , Femenino , Vectores Genéticos , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/genética , Masculino , Ratones , Ratones Endogámicos , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Notch/inmunología , Retina/fisiopatología , Retina/ultraestructura , Proteínas de Unión al Retinol/administración & dosificación , Transducción de Señal , Células TH1/inmunología , Células Th17/inmunología , Uveítis/metabolismo , Uveítis/fisiopatología , Uveítis/terapia
8.
Nanomedicine ; 12(8): 2251-2260, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27381066

RESUMEN

Retinitis pigmentosa (RP) is the most common cause of inherited blindness in adults. Mutations in the PRPF31 gene produce autosomal dominant RP (adRP). To date there are no effective treatments for this disease. The purpose of this study was to design an efficient non-viral vector for human PRPF31 gene delivery as an approach to treat this form of adRP. Span based nanoparticles were developed to mediate gene transfer in the subretinal space of a mouse model of adRP carrying a point mutation (A216P) in the Prpf31 gene. Funduscopic examination, electroretinogram, optomotor test and optical coherence tomography were conducted to further in vivo evaluate the safety and efficacy of the nanosystems developed. Span-polyarginine (SP-PA) nanoparticles were able to efficiently transfect the GFP and PRPF31 plasmid in mice retinas. Statistically significant improvement in visual acuity and retinal thickness were found in Prpf31A216P/+ mice treated with the SP-PA-PRPF31 nanomedicine.


Asunto(s)
Proteínas del Ojo/administración & dosificación , Terapia Genética/métodos , Nanopartículas , Retinitis Pigmentosa/terapia , Animales , Arginina , Análisis Mutacional de ADN , Genes Dominantes , Humanos , Ratones , Mutación , Linaje
9.
Pharmazie ; 71(7): 382-389, 2016 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-29441913

RESUMEN

The occurrence of bone defects can be due to a variety of factors not limited to bone fractures and tumours. Most diseased bone is removed and the patient fitted with prosthetics, prior to use of certain factors such as bone morphogenetic proteins (BMPs) to aid healing. Recently, the protein pigment epithelium-derived factor (PEDF) and the polysaccharide chitosan have been found to have promising effects on the regeneration of bone, with the major advantage of these agents being their safety to date. A study was performed to determine whether the combination of both chitosan and PEDF would enhance greater bone regeneration effects. Post-formulation, in silico tests (particle sizing and surface charge determination) were followed by several cell-based assays (microparticle cellular uptake, cytotoxicity, mitochondrial abundance, bone mineral formation, colony formation in matrigel, and colony formation in collagen I matrix), and finally in vivo testing where microparticles were injected periosteally in the hindlimb. Collectively, these findings support the idea that PEDF microencapsulated within chitosan promotes bone regeneration, and has potential for bone trauma management. Future studies will examine the ability of this promising bone regeneration microparticle to heal bone in disease states such as fracture and tumour-mediated osteolysis.


Asunto(s)
Regeneración Ósea/efectos de los fármacos , Proteínas del Ojo/farmacología , Factores de Crecimiento Nervioso/farmacología , Serpinas/farmacología , Animales , Densidad Ósea/efectos de los fármacos , Cápsulas , Supervivencia Celular/efectos de los fármacos , Quitosano/farmacología , Colágeno Tipo I/farmacología , Ensayo de Unidades Formadoras de Colonias , Simulación por Computador , Composición de Medicamentos , Proteínas del Ojo/administración & dosificación , Proteínas del Ojo/metabolismo , Miembro Posterior , Humanos , Inyecciones , Masculino , Ratones , Ratones Endogámicos BALB C , Mitocondrias/efectos de los fármacos , Factores de Crecimiento Nervioso/administración & dosificación , Factores de Crecimiento Nervioso/metabolismo , Tamaño de la Partícula , Serpinas/administración & dosificación , Serpinas/metabolismo
10.
Stroke ; 46(2): 529-36, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25550365

RESUMEN

BACKGROUND AND PURPOSE: Norrin and its receptor Frizzled-4 have important roles in the blood-brain barrier development. This study is to investigate a potential role and mechanism of Norrin/Frizzled-4 on protecting blood-brain barrier integrity after subarachnoid hemorrhage (SAH). METHODS: One hundred and seventy-eight male Sprague-Dawley rats were used. SAH model was induced by endovascular perforation. Frizzled-4 small interfering RNA was injected intracerebroventricularly 48 hours before SAH. Norrin was administrated intracerebroventricularly 3 hours after SAH. SAH grade, neurological scores, brain water content, Evans blue extravasation, western blots, and immunofluorescence were used to study the mechanisms of Norrin and its receptor regulation protein TSPAN12, as well as neurological outcome. RESULTS: Endogenous Norrin and TSPAN12 expression were increased after SAH, and Norrin was colocalized with astrocytes marker glial fibrillary acidic protein in cortex. Exogenous Norrin treatment significantly alleviated neurobehavioral dysfunction, reduced brain water content and Evans blue extravasation, promoted ß-catenin nuclear translocation, and increased Occludin, VE-Cadherin, and ZO-1 expressions. These effects were abolished by Frizzled-4 small interfering RNA pretreated before SAH. CONCLUSIONS: Norrin protected blood-brain barrier integrity and improved neurological outcome after SAH, and the action of Norrin appeared mediated by Frizzled-4 receptor activation, which promoted ß-catenin nuclear translocation, which then enhanced Occludin, VE-Cadherin, and ZO-1 expression. Norrin might have potential to protect blood-brain barrier after SAH.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Proteínas del Ojo/biosíntesis , Receptores Frizzled/biosíntesis , Proteínas del Tejido Nervioso/biosíntesis , Hemorragia Subaracnoidea/metabolismo , Hemorragia Subaracnoidea/prevención & control , beta Catenina/biosíntesis , Animales , Barrera Hematoencefálica/efectos de los fármacos , Proteínas del Ojo/administración & dosificación , Inyecciones Intraventriculares , Masculino , Proteínas del Tejido Nervioso/administración & dosificación , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley
11.
Aging Dis ; 15(5): 2003-2007, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-38421833

RESUMEN

Geographic atrophy (GA) is an advanced form of age-related macular degeneration (AMD), that starts with atrophic lesions in the outer retina that expand to cover the macula and fovea, leading to severe vision loss over time. Pigment Epithelium-Derived Factor (PEDF) has a diverse-range of properties, including its ability to promote cell survival, reduce inflammation, inhibit angiogenesis, combat oxidative stress, regulate autophagy, and stimulate anti-apoptotic pathways, making it a promising therapeutic candidate for GA. However, the relatively short half-life of PEDF protein has precluded its potential as a clinical therapy for GA since it would require frequent injections. Therefore, we describe administration of a PEDF gene, comparing and contrasting delivery routes, viral and non-viral vectors, and consider the critical challenges for PEDF as a neuroprotectant for GA.


Asunto(s)
Proteínas del Ojo , Atrofia Geográfica , Factores de Crecimiento Nervioso , Serpinas , Animales , Humanos , Proteínas del Ojo/metabolismo , Proteínas del Ojo/uso terapéutico , Proteínas del Ojo/administración & dosificación , Proteínas del Ojo/genética , Proteínas del Ojo/farmacología , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Atrofia Geográfica/tratamiento farmacológico , Atrofia Geográfica/metabolismo , Factores de Crecimiento Nervioso/administración & dosificación , Factores de Crecimiento Nervioso/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/farmacología , Serpinas/administración & dosificación , Serpinas/uso terapéutico , Serpinas/genética , Serpinas/metabolismo , Serpinas/farmacología
12.
J Neurosci ; 32(35): 12152-64, 2012 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-22933798

RESUMEN

Pigment epithelium-derived factor (PEDF) is a serine protease inhibitor (serpin) protein with well established neuroprotective and anti-angiogenic properties. Recent studies have also shown that PEDF enhances renewal of adult subventricular zone (SVZ) neural precursors. In neurosphere cultures prepared from the SVZ of adult mice, we found that addition of recombinant PEDF to the medium enhanced expressions of oligodendroglial lineage markers (NG2 and PDGFrα) and transcription factors (Olig1, Olig2, and Sox10). Similarly, continuous PEDF administration into the lateral ventricles of adult glial fibrillary acidic protein:green fluorescent protein (GFAP:GFP) transgenic mice increased the proportions of GFAP:GFP+ and GFAP:GFP- SVZ neural precursors coexpressing oligodendroglial lineage markers and transcription factors. Notably, PEDF infusion also resulted in an induction of doublecortin- and Sox10 double-positive cells in the adult SVZ. Immunoreactive PEDF receptor was detectable in multiple cell types in both adult SVZ and corpus callosum. Furthermore, PEDF intracerebral infusion enhanced survival and maturation of newly born oligodendroglial progenitor cells in the normal corpus callosum, and accelerated oligodendroglial regeneration in lysolecithin-induced corpus callosum demyelinative lesions. Western blot analysis showed a robust upregulation of endogenous PEDF in the corpus callosum upon lysolecithin-induced demyelination. Our results document previously unrecognized oligodendrotrophic effects of recombinant PEDF on the adult SVZ and corpus callosum, demonstrate induction of endogenous CNS PEDF production following demyelination, and make PEDF a strong candidate for pharmacological intervention in demyelinative diseases.


Asunto(s)
Cuerpo Calloso/fisiología , Proteínas del Ojo/administración & dosificación , Ventrículos Laterales/fisiología , Morfogénesis/fisiología , Factores de Crecimiento Nervioso/administración & dosificación , Oligodendroglía/fisiología , Serpinas/administración & dosificación , Animales , Células Cultivadas , Cuerpo Calloso/citología , Proteínas del Ojo/genética , Femenino , Infusiones Intraventriculares , Ventrículos Laterales/citología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Morfogénesis/genética , Factores de Crecimiento Nervioso/deficiencia , Factores de Crecimiento Nervioso/genética , Serpinas/deficiencia , Serpinas/genética
13.
Mol Med ; 18: 1161-8, 2012 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-22714715

RESUMEN

Obesity is a major risk factor for insulin resistance, type 2 diabetes mellitus and cardiovascular disease. The pathophysiology of obesity is associated with chronic low-grade inflammation. Adipose tissue in obesity is significantly infiltrated by macrophages that secrete cytokines. The mechanisms of interaction between macrophages and adipocytes, leading to macrophage activation and increased cytokine release, remain to be elucidated. We reasoned that an adipocyte-derived factor might stimulate activation of macrophages. We have identified pigment epithelium-derived factor (PEDF) as a mediator of inflammation that is secreted by adipocytes and mediates macrophage activation. Recombinant PEDF activates macrophages to release tumor necrosis factor (TNF) and interleukin-1 (IL-1). The PEDF receptor adipose triglyceride lipase (ATGL) is required for PEDF-mediated macrophage activation. Selective inhibition of ATGL on macrophages attenuates PEDF-induced TNF production, and PEDF enhances the phosphorylation of p38 and extracellular signal-regulated kinase 1/2 mitogen-activated protein kinases. PEDF administration to rats results in increased serum TNF levels, and insulin resistance. Together, these findings suggest that PEDF secreted by adipocytes contributes to the onset and maintenance of chronic inflammation in obesity, and may be a therapeutic target in ameliorating insulin resistance.


Asunto(s)
Adipocitos/metabolismo , Proteínas del Ojo/metabolismo , Mediadores de Inflamación/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Serpinas/metabolismo , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Animales , Proteínas del Ojo/administración & dosificación , Proteínas del Ojo/farmacología , Humanos , Inflamación/patología , Resistencia a la Insulina , Lipasa/metabolismo , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Factores de Crecimiento Nervioso/administración & dosificación , Factores de Crecimiento Nervioso/farmacología , Ratas , Ratas Sprague-Dawley , Serpinas/administración & dosificación , Serpinas/farmacología , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/metabolismo
14.
J Pharmacol Exp Ther ; 342(1): 131-9, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22495066

RESUMEN

Pathological retinal neovascularization and choroidal neovascularization are major causes of vision loss in a variety of clinical conditions, such as retinopathy of prematurity, age-related macular degeneration, and diabetic retinopathy. Pigment epithelial-derived factor (PEDF) has been found to be the most potent natural, endogenous inhibitor of neovascularization, but its application is restricted because of its instability and short half-life. Polyethylene glycol (PEG) has been used as a drug carrier to slow clearance rate for decades. The present study investigated PEGylated-PEDF for the first time and evaluated its long-term effects on preventing angiogenesis in vitro and in vivo. PEG showed lower cytotoxicity to human umbilical vein endothelial cells (HUVECs). In vitro, PEGylated-PEDF inhibited HUVEC proliferation, migration, tube formation, and vascular endothelium growth factor secretion and induced HUVEC apoptosis in a dose-dependent manner, and it showed a statistically significant difference compared with the PEDF treatment group. In vivo, PEGylated-PEDF had a long-lasting effect in both plasma and retinal concentrations. In an oxygen-induced retinopathy model, one intravitreous injection of PEGylated-PEDF after mouse pups were moved into room air resulted in a significant difference in the inhibition of retinal neovascularization, which decreased the nonperfusion area, compared with the PEDF-treated group. Our present study demonstrated for the first time the long-term inhibitory effects of PEGylated-PEDF on the prevention of neovascularization in vitro and in vivo. These data suggest that PEGylated-PEDF could offer an innovative therapeutic strategy for preventing retinal neovascularization.


Asunto(s)
Proteínas del Ojo/administración & dosificación , Factores de Crecimiento Nervioso/administración & dosificación , Polietilenglicoles/administración & dosificación , Proteínas Recombinantes/administración & dosificación , Retina/efectos de los fármacos , Neovascularización Retiniana/tratamiento farmacológico , Serpinas/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neovascularización Coroidal/metabolismo , Neovascularización Coroidal/prevención & control , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/metabolismo , Neovascularización Patológica/prevención & control , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Ratas , Ratas Wistar , Proteínas Recombinantes/sangre , Proteínas Recombinantes/genética , Retina/metabolismo , Retina/patología , Neovascularización Retiniana/metabolismo , Serpinas/genética , Serpinas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
15.
Am J Pathol ; 178(2): 591-8, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21281791

RESUMEN

Oxidative stress and inflammation are involved in cardiac remodeling after acute myocardial infarction (AMI). We have found that pigment epithelium-derived factor (PEDF) inhibits vascular inflammation through its anti-oxidative properties. However, effects of PEDF on cardiac remodeling after AMI remain unknown. We investigated whether PEDF could inhibit left ventricular remodeling and improve cardiac function in rats with AMI. AMI was induced in 8-week-old Sprague-Dawley rats by ligation of the left ascending coronary artery. Rats were treated intravenously with vehicle or 10 µg PEDF/100 g b.wt. every day for up to 2 weeks after AMI. Each rat was followed until 16 weeks of age. PEDF levels in infarcted areas and serum were significantly decreased at 1 week after AMI and remained low during the observational periods. PEDF administration inhibited apoptotic cell death and oxidative stress generation around the infarcted areas at 2 and 8 weeks after AMI. Further, PEDF injection suppressed cardiac fibrosis by reducing transforming growth factor-ß and type III collagen expression, improved left ventricular ejection fraction, ameliorated diastolic dysfunction, and inhibited the increase in left ventricular mass index at 8 weeks after AMI. The present study demonstrated that PEDF could inhibit tissue remodeling and improve cardiac function in AMI rats. Substitution of PEDF may be a novel therapeutic strategy for cardiac remodeling after AMI.


Asunto(s)
Proteínas del Ojo/administración & dosificación , Proteínas del Ojo/farmacología , Pruebas de Función Cardíaca/efectos de los fármacos , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/fisiopatología , Factores de Crecimiento Nervioso/administración & dosificación , Factores de Crecimiento Nervioso/farmacología , Serpinas/administración & dosificación , Serpinas/farmacología , Remodelación Ventricular/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proteínas del Ojo/metabolismo , Proteínas del Ojo/uso terapéutico , Fibrosis , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/enzimología , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Hemodinámica/efectos de los fármacos , Macrófagos/efectos de los fármacos , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/enzimología , Miocardio/patología , Neovascularización Fisiológica/efectos de los fármacos , Factores de Crecimiento Nervioso/metabolismo , Factores de Crecimiento Nervioso/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Serpinas/metabolismo , Serpinas/uso terapéutico , Ultrasonografía
16.
Mol Vis ; 18: 1858-64, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22815639

RESUMEN

PURPOSE: SWAP 70-like adaptor of T cells (SLAT; aka Def6) is a recently discovered guanine nucleotide exchange factor for Rho guanosine triphosphate (GTP)ases that has been previously shown to play a role in cluster of differentiation(CD)4+ T cell activation, T-helper (Th)1/Th2/Th17 differentiation and development of experimental autoimmune encephalomyelitis. Here, we investigated the role of SLAT/Def6 in the development of experimental autoimmune uveitis (EAU), an animal model for several uveitic conditions in humans. METHODS: SLAT/Def6 deficient ("KO") mice and C57BL/6 controls were immunized with interphotoreceptor retinoid-binding protein (IRBP), along with pertussis toxin. The development of ocular inflammation was determined by both fundoscopy and histological examination. Lymphoid cells from draining lymph nodes were cultured with IRBP to measure lymphocyte proliferation and release of cytokines. Purified dendritic cells were tested for their capacity to present antigen to responding lymphocytes. In addition, the lymphoid cells were tested for the expression of forkhead box P3 (FoxP3), using conventional methods, and the activity of T-regulatory cells was determined by their capacity to inhibit in vitro proliferative responses. Serum anti -IRBP antibody levels were measured by enzyme-linked immunosorbant assay (ELISA). quantitative polymerase chain reaction (qPCR) was used to determine the transcript levels of cytokines in inflamed eyes. RESULTS: SLAT/Def6 KO mice had significantly reduced EAU compared to controls. Cells isolated from draining lymph nodes of SLAT/Def6 KO mice exhibited impaired proliferation and production of Th1 and Th17 signature cytokines (interferon [IFN]-γ and interleukin [IL]-17, respectively) when compared with cells isolated from control mice. qPCR of inflamed eyes detected similar levels of IFN-γ transcript in control and SLAT/Def6 KO mice, whereas the IL-17 transcript levels in eyes of the SLAT/Def6 KO mice were lower than in eyes of the controls. The SLAT/Def6 KO mice resembled their wild type (WT) controls, however, in the levels of their serum antibody against IRBP, the antigen presenting capacity of their dendritic cells, the proportion of cells expressing Foxp3 and the immunosuppressive activity of their T-regulatory cells. CONCLUSIONS: SLAT/Def6 KO mice exhibit reduced capacity to develop ocular inflammation and cellular activity when immunized with IRBP. Our study provides new data showing that SLAT/Def6 plays a major role in the T cell-mediated autoimmune processes that bring about the inflammatory eye disease, EAU.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Proteínas de Unión al ADN/inmunología , Proteínas del Ojo/inmunología , Inflamación/inmunología , Proteínas Nucleares/inmunología , Proteínas de Unión al Retinol/inmunología , Uveítis/inmunología , Animales , Anticuerpos/sangre , Anticuerpos/inmunología , Enfermedades Autoinmunes/inducido químicamente , Enfermedades Autoinmunes/genética , Proliferación Celular/efectos de los fármacos , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Proteínas del Ojo/administración & dosificación , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/inmunología , Factores de Intercambio de Guanina Nucleótido , Inflamación/inducido químicamente , Inflamación/genética , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Interleucina-17/biosíntesis , Interleucina-17/inmunología , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Ratones , Ratones Noqueados , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Toxina del Pertussis/administración & dosificación , Proteínas de Unión al Retinol/administración & dosificación , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología , Células TH1/efectos de los fármacos , Células TH1/inmunología , Células TH1/patología , Células Th17/efectos de los fármacos , Células Th17/inmunología , Células Th17/patología , Células Th2/efectos de los fármacos , Células Th2/inmunología , Células Th2/patología , Uveítis/inducido químicamente , Uveítis/genética
17.
Microvasc Res ; 84(1): 105-8, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22386653

RESUMEN

Pigment epithelium-derived factor (PEDF) a glycoprotein that belongs to the superfamily of serine protease inhibitors, has been recently shown to be the most potent inhibitor of angiogenesis in the mammalian eye. However, which active domain of PEDF protein could be involved in its anti-angiogenic properties remains unknown. Therefore, in this study, we examined which PEDF-derived synthetic peptides could inhibit corneal neovascularization induced by chemical cauterization in vivo. Rats treated with topical application of PEDF protein had 31% less corneal neovascularization at day 7 after the injury than phosphate-buffered saline (PBS)-treated rats. P5-2 and P5-3 peptides (residues 388-393 and 394-400 of PEDF protein, respectively) significantly suppressed the corneal neovascularization after chemical cauterization at day 7, and its anti-angiogenic potential was almost equal to that of full-length PEDF protein. Further, full-length PEDF protein and P5-3 peptide significantly decreased 8-hydroxy-2'-deoxyguanosine and vascular endothelial growth factor (VEGF) levels in the corneal. Our present study suggests that PEDF-derived synthetic peptide, P5-3 could inhibit the corneal neovascularization induced by chemical cauterization in rats by suppressing VEGF expression via its anti-oxidative properties.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Córnea/efectos de los fármacos , Neovascularización de la Córnea/inducido químicamente , Proteínas del Ojo/farmacología , Factores de Crecimiento Nervioso/farmacología , Serpinas/farmacología , Factor A de Crecimiento Endotelial Vascular/metabolismo , 8-Hidroxi-2'-Desoxicoguanosina , Administración Tópica , Inhibidores de la Angiogénesis/administración & dosificación , Animales , Cauterización , Córnea/irrigación sanguínea , Lesiones de la Cornea , Neovascularización de la Córnea/patología , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Modelos Animales de Enfermedad , Proteínas del Ojo/administración & dosificación , Masculino , Factores de Crecimiento Nervioso/administración & dosificación , Ratas , Ratas Sprague-Dawley , Serpinas/administración & dosificación
18.
BMC Cancer ; 12: 129, 2012 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-22462776

RESUMEN

BACKGROUND: Angiogenesis plays an important role in tumor growth and metastasis, therefore antiangiogenic therapy was widely investigated as a promising approach for cancer therapy. Recently, pigment epithelium-derived factor (PEDF) has been shown to be the most potent inhibitor of angiogenesis. Adeno-associated virus (AAV) vectors have been intensively studied due to their wide tropisms, nonpathogenicity, and long-term transgene expression in vivo. The objective of this work was to evaluate the ability of AAV-mediated human PEDF (hPEDF) as a potent tumor suppressor and a potential candidate for cancer gene therapy. METHODS: Recombinant AAV2 encoding hPEDF (rAAV2-hPEDF) was constructed and produced, and then was assigned for in vitro and in vivo experiments. Conditioned medium from cells infected with rAAV2-hPEDF was used for cell proliferation and tube formation tests of human umbilical vein endothelial cells (HUVECs). Subsequently, colorectal peritoneal carcinomatosis (CRPC) mouse model was established and treated with rAAV2-hPEDF. Therapeutic efficacy of rAAV2-hPEDF were investigated, including tumor growth and metastasis, survival time, microvessel density (MVD) and apoptosis index of tumor tissues, and hPEDF levels in serum and ascites. RESULTS: rAAV2-hPEDF was successfully constructed, and transmission electron microscope (TEM) showed that rAAV2-hPEDF particles were non-enveloped icosahedral shape with a diameter of approximately 20 nm. rAAV2-hPEDF-infected cells expressed hPEDF protein, and the conditioned medium from infected cells inhibited proliferation and tube-formation of HUVECs in vitro. Furthermore, in CRPC mouse model, rAAV2-hPEDF significantly suppressed tumor growth and metastasis, and prolonged survival time of treated mice. Immunofluorescence studies indicated that rAAV2-hPEDF could inhibit angiogenesis and induce apoptosis in tumor tissues. Besides, hPEDF levels in serum and ascites of rAAV2-hPEDF-treated mice were significant higher than those in rAAV2-null or normal saline (NS) groups. CONCLUSIONS: Thus, our results suggest that rAAV2-hPEDF may be a potential candidate as an antiangiogenic therapy agent.


Asunto(s)
Antineoplásicos/administración & dosificación , Carcinoma/terapia , Proteínas del Ojo/administración & dosificación , Terapia Genética/métodos , Factores de Crecimiento Nervioso/administración & dosificación , Neoplasias Peritoneales/terapia , Serpinas/administración & dosificación , Adenoviridae/genética , Animales , Western Blotting , Carcinoma/patología , Carcinoma/secundario , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Vectores Genéticos , Ratones , Ratones Endogámicos BALB C , Microvasos/patología , Metástasis de la Neoplasia , Neovascularización Patológica/tratamiento farmacológico , Neoplasias Peritoneales/patología
19.
J Biomed Biotechnol ; 2012: 230298, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22701300

RESUMEN

The potent antiangiogenic pigment epithelium-derived factor (PEDF) has shown promise against osteosarcoma, a tumour that originates in the bone and metastasises to the lungs. Neurotrophic, antiangiogenic, antiproliferative, and antimetastatic properties of PEDF have been attributed to a number of functional epitopes on the PEDF glycoprotein. StVOrth-2 (residues 78-102) and StVOrth-3 (residues 90-114) are two PEDF-derived peptides based on these functional epitopes. StVOrth-2 has previously been shown to inhibit osteosarcoma cell proliferation, while StVOrth-3 increased osteosarcoma cell adhesion to collagen I in vitro. In this paper, we have evaluated systemically and continuously delivered StVOrth-2 and StVOrth-3 using a clinically relevant murine model of osteosarcoma with spontaneous metastasis. Treatment with StVOrth-2 or StVOrth-3 with microosmotic pumps was initiated after primary osteosarcoma was established in the tibia. While treatment with StVOrth-2 and StVOrth-3 did not appear to affect local tumour invasion, tumour necrosis or apoptosis, StVOrth-2 predominantly restricted the growth of primary tumours, while StVOrth-3 restricted the burden of pulmonary metastatic disease. No peptide caused gross toxicity in mouse tissues as assessed by measuring weight of animals, serum biochemistry, and gross tissue observation. The differential effects exhibited by StVOrth-2 and StVOrth-3 in this orthotopic model of osteosarcoma may be related to the functional epitopes on the PEDF glycoprotein that they represent.


Asunto(s)
Neoplasias Óseas/tratamiento farmacológico , Proteínas del Ojo/química , Metástasis de la Neoplasia/tratamiento farmacológico , Factores de Crecimiento Nervioso/química , Osteosarcoma/tratamiento farmacológico , Péptidos/administración & dosificación , Serpinas/química , Animales , Apoptosis/efectos de los fármacos , Neoplasias Óseas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Epítopos/efectos de los fármacos , Epítopos/inmunología , Proteínas del Ojo/administración & dosificación , Humanos , Ratones , Metástasis de la Neoplasia/patología , Neovascularización Patológica/tratamiento farmacológico , Factores de Crecimiento Nervioso/administración & dosificación , Osteosarcoma/patología , Péptidos/síntesis química , Serpinas/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
20.
J Cell Physiol ; 226(11): 2943-52, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21302290

RESUMEN

Current osteoinductive protein therapy utilizes bolus administration of large doses of bone morphogenetic proteins (BMPs), which is costly, and may not replicate normal bone healing. The limited in vivo biologic activity of BMPs requires the investigation of growth factors that may enhance this activity. In this study, we utilized the C3H10T1/2 murine mesenchymal stem cell line to test the hypotheses that osteoactivin (OA) has comparable osteoinductive effects to bone morphogenetic protein-2 (BMP-2), and that sustained administration of either growth factor would result in increased osteoblastic differentiation as compared to bolus administration. Sustained release biodegradable hydrogels were designed, and C3H10T1/2 cells were grown on hydrogels loaded with BMP-2 or OA. Controls were grown on unloaded hydrogels, and positive controls were exposed to bolus growth factor administration. Cells were harvested at several time points to assess osteoblastic differentiation. Alkaline phosphatase (ALP) staining and activity, and gene expression of ALP and osteocalcin were assessed. Treatment with OA or BMP-2 resulted in comparable effects on osteoblastic marker expression. However, cells grown on hydrogels demonstrated osteoblastic differentiation that was not as robust as cells treated with bolus administration. This study shows that OA has comparable effects to BMP-2 on osteoblastic differentiation using both bolus administration and continuous release, and that bolus administration of OA has a more profound effect than administration using hydrogels for sustained release. This study will lead to a better understanding of appropriate delivery methods of osteogenic growth factors like OA for repair of fractures and segmental bone defects.


Asunto(s)
Proteína Morfogenética Ósea 2/administración & dosificación , Diferenciación Celular/efectos de los fármacos , Proteínas del Ojo/administración & dosificación , Glicoproteínas de Membrana/administración & dosificación , Células Madre Mesenquimatosas/efectos de los fármacos , Fosfatasa Alcalina/biosíntesis , Fosfatasa Alcalina/genética , Animales , Diferenciación Celular/genética , Línea Celular , Preparaciones de Acción Retardada , Expresión Génica/efectos de los fármacos , Hidrogel de Polietilenoglicol-Dimetacrilato/administración & dosificación , Células Madre Mesenquimatosas/citología , Ratones , Osteocalcina/biosíntesis , Osteocalcina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA