Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 141(1): 178-90, 2010 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-20371353

RESUMEN

Our findings that PlGF is a cancer target and anti-PlGF is useful for anticancer treatment have been challenged by Bais et al. Here we take advantage of carcinogen-induced and transgenic tumor models as well as ocular neovascularization to report further evidence in support of our original findings of PlGF as a promising target for anticancer therapies. We present evidence for the efficacy of additional anti-PlGF antibodies and their ability to phenocopy genetic deficiency or silencing of PlGF in cancer and ocular disease but also show that not all anti-PlGF antibodies are effective. We also provide additional evidence for the specificity of our anti-PlGF antibody and experiments to suggest that anti-PlGF treatment will not be effective for all tumors and why. Further, we show that PlGF blockage inhibits vessel abnormalization rather than density in certain tumors while enhancing VEGF-targeted inhibition in ocular disease. Our findings warrant further testing of anti-PlGF therapies.


Asunto(s)
Neovascularización Fisiológica/efectos de los fármacos , Proteínas Gestacionales/antagonistas & inhibidores , Proteínas Gestacionales/metabolismo , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Anticuerpos Monoclonales/uso terapéutico , Carcinoma Hepatocelular/irrigación sanguínea , Carcinoma Hepatocelular/prevención & control , Coroides/irrigación sanguínea , Modelos Animales de Enfermedad , Oftalmopatías/patología , Humanos , Neoplasias Hepáticas Experimentales/irrigación sanguínea , Neoplasias Hepáticas Experimentales/prevención & control , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Papiloma/irrigación sanguínea , Papiloma/inducido químicamente , Papiloma/prevención & control , Factor de Crecimiento Placentario , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/prevención & control
2.
Exp Eye Res ; 207: 108608, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33930400

RESUMEN

The progressive and sight-threatening disease, age-related macular degeneration (AMD), is a growing public health concern due to ageing demographics, with the highest unmet medical need for the advanced stage of dry AMD, geographic atrophy. The pathogenesis underlying AMD is driven by a complex interplay of genetic and environmental factors. There is ample evidence that inflammation is strongly involved in AMD development. Interleukin-33 (IL-33) has been proposed to be critically involved in retinal degeneration, but a protective role in eye pathophysiology was also demonstrated. The current study investigated the therapeutic potential of IL-33trap, a novel IL-33-neutralizing biologic, in dry AMD/geographic atrophy and, based on controversial data regarding the protective versus detrimental functions of IL-33 in neovascularization, evaluated the risk of progression to wet AMD by IL-33 neutralization. Repeated intravitreal (IVT) injections of IL-33trap in the mouse laser-induced choroidal neovascularization model did not exacerbate neovascularization or leakage, while it significantly inhibited inflammatory cell infiltration in the retinal pigment epithelium and choroid. On the contrary, IVT treatment with IL-33trap significantly induced retinal inflammation and could not prevent retinopathy induction in the mouse sodium iodate (NaIO3) model. Overall, these data suggest a complex and dichotomous role of IL-33 in eye pathology and indicate that IL-33 neutralization is not able to prevent onset and progression of dry AMD pathogenesis.


Asunto(s)
Neovascularización Coroidal/tratamiento farmacológico , Modelos Animales de Enfermedad , Atrofia Geográfica/tratamiento farmacológico , Interleucina-33/uso terapéutico , Animales , Neovascularización Coroidal/diagnóstico , Neovascularización Coroidal/fisiopatología , Electrorretinografía , Angiografía con Fluoresceína , Atrofia Geográfica/diagnóstico , Atrofia Geográfica/fisiopatología , Inmunohistoquímica , Inflamación/prevención & control , Coagulación con Láser , Masculino , Ratones , Ratones Endogámicos C57BL , Tomografía de Coherencia Óptica
3.
J Pharmacokinet Pharmacodyn ; 48(6): 825-836, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34302260

RESUMEN

Intravitreal (IVT) injection of pharmacological agents is an established and widely used procedure for the treatment of many posterior segment of the eye diseases. IVT injections permit drugs to reach high concentrations in the retina whilst limiting systemic exposure. Beyond the risk of secondary complications such as intraocular infection, the potential of systemic adverse events cannot be neglected. Therefore, a detailed understanding of the rules governing systemic exposure following IVT drug administration remains a prerequisite for the evaluation and development of new pharmacological agents intended for eye delivery. We present here a novel mathematical model to describe and predict circulating drug levels following IVT in the rabbit eye, a species which is widely used for drug delivery, pharmacokinetic, and pharmacodynamic studies. The mathematical expression was derived from a pharmacokinetic model that assumes the existence of a compartment between the vitreous humor compartment itself and the systemic compartment. We show that the model accurately describes circulating levels of THR-149, a plasma kallikrein inhibitor in development for the treatment of diabetic macular edema. We hypothesize that the model based on the rabbit eye has broader relevance to the human eye and can be used to analyze systemic exposure of a variety of drugs delivered in the eye.


Asunto(s)
Retinopatía Diabética , Edema Macular , Animales , Retinopatía Diabética/tratamiento farmacológico , Edema Macular/tratamiento farmacológico , Edema Macular/metabolismo , Preparaciones Farmacéuticas/metabolismo , Conejos , Retina/metabolismo , Cuerpo Vítreo/metabolismo
4.
Diabetologia ; 63(10): 2235-2248, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32734440

RESUMEN

AIMS/HYPOTHESIS: Diabetic retinopathy is a common complication of diabetes and a leading cause of visual impairment and blindness. Despite recent advances, our understanding of its pathophysiology remains incomplete. The aim of this study was to provide deeper insight into the complex network of molecular and cellular changes that underlie diabetic retinopathy by systematically mapping the transcriptional changes that occur in the different cellular compartments of the degenerating diabetic mouse retina. METHODS: Single-cell RNA sequencing was performed on retinal tissue from 12-week-old wild-type and Akimba (Ins2Akita×Vegfa+/-) mice, which are known to replicate features of clinical diabetic retinopathy. This resulted in transcriptome data for 9474 retinal cells, which could be annotated to eight distinct retinal cell types. Using STRING analysis, we studied differentially expressed gene networks in neuronal, glial and immune cell compartments to create a comprehensive view on the pathological changes that occur in the Akimba retina. Using subclustering analysis, we further characterised macroglial and inflammatory cell subpopulations. Prominent findings were confirmed at the protein level using immunohistochemistry, western blotting and ELISA. RESULTS: At 12 weeks, the Akimba retina was found to display degeneration of rod photoreceptors and presence of inflammatory cells, identified by subclustering analysis as monocyte, macrophage and microglial populations. Analysis of differentially expressed genes in the rod, cone, bipolar cell and macroglial compartments indicated changes in cell metabolism and ribosomal gene expression, gliosis, activation of immune system pathways and redox and metal ion dyshomeostasis. Experiments at the protein level supported a metabolic shift from glycolysis to oxidative phosphorylation (glyceraldehyde 3-phosphate dehydrogenase), activation of microglia/macrophages (isolectin-B4), metal ion and oxidative stress response (metallothionein and haem oxygenase-1) and reactive macroglia (glial fibrillary acidic protein and S100) in the Akimba retina, compared with wild-type mice. Our single-cell approach also indicates macroglial subpopulations with distinct fibrotic, inflammatory and gliotic profiles. CONCLUSIONS/INTERPRETATION: Our study identifies molecular pathways underlying inflammatory, metabolic and oxidative stress-mediated changes in the Akimba mouse model of diabetic retinopathy and distinguishes distinct functional subtypes of inflammatory and macroglial cells. DATA AVAILABILITY: RNA-seq data have been deposited in the ArrayExpress database at EMBL-EBI ( www.ebi.ac.uk/arrayexpress ) under accession number E-MTAB-9061. Graphical abstract.


Asunto(s)
Retinopatía Diabética/genética , Perfilación de la Expresión Génica , Retina/metabolismo , Animales , Retinopatía Diabética/metabolismo , Glucólisis/genética , Insulina/genética , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Ratones Transgénicos , Microglía/citología , Microglía/metabolismo , Monocitos/citología , Monocitos/metabolismo , Fosforilación Oxidativa , Estrés Oxidativo/genética , RNA-Seq , Retina/citología , Células Bipolares de la Retina/citología , Células Bipolares de la Retina/metabolismo , Células Fotorreceptoras Retinianas Conos/citología , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Fotorreceptoras Retinianas Bastones/citología , Células Fotorreceptoras Retinianas Bastones/metabolismo , Análisis de la Célula Individual , Estrés Fisiológico/genética , Factor A de Crecimiento Endotelial Vascular/genética
5.
Glia ; 68(12): 2643-2660, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32645232

RESUMEN

Increasing evidence suggests that functional impairments at the level of the neurovascular unit (NVU) underlie many neurodegenerative and neuroinflammatory diseases. While being part of the NVU, astrocytes have been largely overlooked in this context and only recently, tightening of the glia limitans has been put forward as an important neuroprotective response to limit these injurious processes. In this study, using the retina as a central nervous system (CNS) model organ, we investigated the structure and function of the glia limitans, and reveal that the blood-retina barrier and glia limitans function as a coordinated double barrier to limit infiltration of leukocytes and immune molecules. We provide in vitro and in vivo evidence for a protective response at the NVU upon CNS injury, which evokes inflammation-induced glia limitans tightening. Matrix metalloproteinase-3 (MMP-3) was found to be a crucial regulator of this process, thereby revealing its beneficial and immunomodulatory role in the CNS. in vivo experiments in which MMP-3 activity was deleted via genetic and pharmacological approaches, combined with a comprehensive study of tight junction molecules, glial end feet markers, myeloid cell infiltration, cytokine expression and neurodegeneration, show that MMP-3 attenuates neuroinflammation and neurodegeneration by tightening the glia limitans, thereby pointing to a prominent role of MMP-3 in preserving the integrity of the NVU upon injury. Finally, we gathered promising evidence to suggest that IL1b, which is also regulated by MMP-3, is at least one of the molecular messengers that induces glia limitans tightening in the injured CNS.


Asunto(s)
Traumatismos del Nervio Óptico , Astrocitos , Humanos , Metaloproteinasa 3 de la Matriz , Neuroglía , Retina
6.
Exp Eye Res ; 180: 43-52, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30472075

RESUMEN

Integrins are associated with various eye diseases such as diabetic retinopathy (DR) and wet age-related macular degeneration (AMD) and implicated in main pathologic disease hallmarks like neovascularization, inflammation, fibrosis and vascular leakage. Targeting integrins has the potential to attenuate these vision-threatening processes, independent of anti-vascular endothelial growth factor (VEGF) responsiveness. The current investigation characterized THR-687 as a novel pan RGD (arginylglycylaspartic acid) integrin receptor antagonist able to compete for binding with the natural ligand with nanomolar potency (e.g. αvß3 (IC50 of 4.4 ±â€¯2.7 nM), αvß5 (IC50 of 1.3 ±â€¯0.5 nM) and α5ß1 (IC50 of 6.8 ±â€¯3.2 nM)). THR-687 prevented the migration of human umbilical vein endothelial cells (HUVECs) into a cell-free area (IC50 of 258 ±â€¯113 nM) as well as vessel sprouting in an ex vivo mouse choroidal explant model (IC50 of 236 ±â€¯173 nM), and was able to induce the regression of pre-existing vascular sprouts. Moreover, combined intravitreal and intraperitoneal administration of THR-687 potently inhibited VEGF-induced leakage in the mouse retina. In addition, THR-687 injected intravitreally at 3 different dose levels (0.45 mg, 2.25 mg or 4.5 mg/eye) potently inhibited neovascularization-induced leakage in the cynomolgus laser-induced choroidal neovascularization (CNV) model. These data suggest that THR-687 is a promising drug candidate for the treatment of vision-threatening retinal vascular eye diseases such as DR and wet AMD.


Asunto(s)
Neovascularización Coroidal/tratamiento farmacológico , Retinopatía Diabética/tratamiento farmacológico , Compuestos Orgánicos/farmacología , Receptores Inmunológicos/antagonistas & inhibidores , Receptores de Péptidos/antagonistas & inhibidores , Vasos Retinianos/efectos de los fármacos , Degeneración Macular Húmeda/tratamiento farmacológico , Animales , Permeabilidad Capilar/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Angiografía con Fluoresceína , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inyecciones Intraperitoneales , Inyecciones Intravítreas , Macaca fascicularis , Masculino , Ratones , Ratones Endogámicos C57BL , Compuestos Orgánicos/uso terapéutico , Conejos , Tomografía de Coherencia Óptica , Factor A de Crecimiento Endotelial Vascular/farmacología
7.
Exp Eye Res ; 165: 136-150, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28965804

RESUMEN

The current standard of care in clinical practice for diabetic retinopathy (DR), anti-vascular endothelial growth factor (VEGF) therapy, has shown a significant improvement in visual acuity. However, treatment response can be variable and might be associated with potential side effects. This study was designed to investigate inhibition of placental growth factor (PlGF) as a possible alternative therapy for DR. The effect of the anti-PlGF antibody (PL5D11D4) was preclinically evaluated in various animal models by investigating different DR hallmarks, including inflammation, neurodegeneration, vascular leakage and fibrosis. The in vivo efficacy was tested in diabetic streptozotocin (STZ) and Akimba models and in the laser induced choroidal neovascularization (CNV) mouse model. Intravitreal (IVT) administration of the anti-PlGF antibody was compared to anti-VEGFR-2 antibody (DC101), anti-VEGF antibody (B20), VEGF-Trap (aflibercept) and triamcinolone acetonide (TAAC). Vascular leakage was investigated in the mouse STZ model by fluorescein isothiocyanate labeled bovine serum albumin (FITC-BSA) perfusion and in the Akimba model by fluorescein angiography (FA). Repeated IVT administration of the anti-PlGF antibody reduced vascular leakage, which was comparable to a single administration of VEGFR-2 inhibition in the mouse STZ model. PL5D11D4 treatment did not alter retinal ganglion cell (RGC) density, as demonstrated by Brn3a staining, whereas DC101 significantly reduced RGC number with 20%. Immunohistological stainings were performed to investigate inflammation (CD45, F4/80) and fibrosis (collagen type 1a). In the CNV model, IVT injection(s) of PL5D11D4 dose-dependently reduced inflammation and fibrosis, as compared to PBS treatment. Equimolar single administration of the anti-PlGF antibody and aflibercept (21 nM) and TAAC decreased leukocyte and macrophage infiltration with 50%, whereas DC101 and B20 (21 nM) had no effect on the inflammatory response. Similar results were observed in the mouse STZ model on the number of microglia and macrophages in the retina. Repeated administration of PL5D11D4 (21 nM) and TAAC similarly reduced fibrosis, while no effect was observed after equimolar DC101, B20 nor aflibercept administration (21 nM). In summary, the anti-PlGF antibody showed comparable efficacy as well-characterized VEGF-inhibitor on the process of vascular leakage, but differentiates itself by also reducing inflammation and fibrosis, without triggering a neurodegenerative response.


Asunto(s)
Anticuerpos Bloqueadores/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Retinopatía Diabética/tratamiento farmacológico , Factor de Crecimiento Placentario/antagonistas & inhibidores , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Modelos Animales de Enfermedad , Inyecciones Intravítreas , Ratones , Ratones Endogámicos C57BL
8.
J Cell Mol Med ; 17(12): 1632-43, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24118824

RESUMEN

Excessive post-operative wound healing with subsequent scarring frequently leads to surgical failure of glaucoma filtration surgery (trabeculectomy). We investigated the hypothesis that placental growth factor (PlGF) plays a role in post-operative scar formation, and that it therefore may be a target for improvement of filtration surgery outcome. ELISA experiments showed that PlGF levels were significantly increased in aqueous humour of glaucoma patients and after VEGF treatment, which may indicate an important contribution of this growth factor to wound healing after trabeculectomy. Using a mouse model of glaucoma filtration surgery, we were able to show that intracameral injection of a previously characterized anti-PlGF antibody (ThromboGenics NV) significantly improved surgical outcome by increasing bleb survival and bleb area. This was associated with a significant reduction in post-operative proliferation, inflammation and angiogenesis during the first post-operative days after surgery, and with a decrease in collagen deposition at later stages. Furthermore, inhibition of PlGF seemed to be more effective than anti-VEGF-R2 treatment in improving surgical outcome, possibly via its additional effect on inflammation. These results render PlGF an appealing target for ocular wound healing and point to potential therapeutic benefits of PlGF inhibition for the prevention of surgical failure.


Asunto(s)
Glaucoma/metabolismo , Glaucoma/cirugía , Proteínas Gestacionales/antagonistas & inhibidores , Animales , Anticuerpos/administración & dosificación , Anticuerpos/farmacología , Humor Acuoso/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cicatriz/patología , Modelos Animales de Enfermedad , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Glaucoma/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Factor de Crecimiento Placentario , Proteínas Gestacionales/metabolismo , Conejos , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Cápsula de Tenon/patología , Trabeculectomía , Resultado del Tratamiento , Regulación hacia Arriba/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo
9.
Aging Cell ; 21(1): e13537, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34927348

RESUMEN

As the mammalian central nervous system matures, its regenerative ability decreases, leading to incomplete or non-recovery from the neurodegenerative diseases and central nervous system insults that we are increasingly facing in our aging world population. Current neuroregenerative research is largely directed toward identifying the molecular and cellular players that underlie central nervous system repair, yet it repeatedly ignores the aging context in which many of these diseases appear. Using an optic nerve crush model in a novel biogerontology model, that is, the short-living African turquoise killifish, the impact of aging on injury-induced optic nerve repair was investigated. This work reveals an age-related decline in axonal regeneration in female killifish, with different phases of the repair process being affected depending on the age. Interestingly, as in mammals, both a reduced intrinsic growth potential and a non-supportive cellular environment seem to lie at the basis of this impairment. Overall, we introduce the killifish visual system and its age-dependent regenerative ability as a model to identify new targets for neurorepair in non-regenerating individuals, thereby also considering the effects of aging on neurorepair.


Asunto(s)
Regeneración Nerviosa/fisiología , Nervio Óptico/fisiopatología , Factores de Edad , Animales , Fundulidae
10.
Trends Endocrinol Metab ; 33(1): 50-71, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34794851

RESUMEN

Type 2 diabetes (T2D) is associated with multiple comorbidities, including diabetic retinopathy (DR) and cognitive decline, and T2D patients have a significantly higher risk of developing Alzheimer's disease (AD). Both DR and AD are characterized by a number of pathological mechanisms that coalesce around the neurovascular unit, including neuroinflammation and degeneration, vascular degeneration, and glial activation. Chronic hyperglycemia and insulin resistance also play a significant role, leading to activation of pathological mechanisms such as increased oxidative stress and the accumulation of advanced glycation end-products (AGEs). Understanding these common pathways and the degree to which they occur simultaneously in the brain and retina during diabetes will provide avenues to identify T2D patients at risk of cognitive decline.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Diabetes Mellitus Tipo 2 , Retinopatía Diabética , Enfermedad de Alzheimer/metabolismo , Disfunción Cognitiva/etiología , Diabetes Mellitus Tipo 2/complicaciones , Retinopatía Diabética/metabolismo , Retinopatía Diabética/patología , Productos Finales de Glicación Avanzada/metabolismo , Humanos
11.
Exp Eye Res ; 93(5): 689-99, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21907194

RESUMEN

Vascular endothelial growth factor (VEGF) plays an important role in both physiological and pathological angiogenesis. Our previous studies showed a differential role of VEGF isoforms in retinal physiological angiogenesis. We also demonstrated that non-selective inhibition of VEGF by bevacizumab had a beneficial effect on surgical outcome after glaucoma filtration surgery by reducing angiogenesis as well as fibrosis. However, the function of the VEGF isoforms in pathological angiogenesis and wound healing in the eye still remains unidentified. This study was designed to elucidate the differential roles of VEGF isoforms in scar formation after trabeculectomy. Furthermore, we also investigated whether pegaptanib (Macugen™, Pfizer), an aptamer which specifically blocks VEGF(165), could improve surgical outcome by reducing postoperative scarring. VEGF-R2 and neuropilin-1 (NRP-1) expression was analyzed in vitro by RT-PCR, and were found to be expressed at higher levels in human umbilical vein endothelial cells (HUVEC) as compared to Tenon fibroblasts (TF). The effect of the different VEGF isoforms (VEGF(121), VEGF(165) and VEGF(189)) and pegaptanib on cell proliferation was determined via WST-1 assay. Endothelial cell proliferation was stimulated after addition of VEGF(121) and VEGF(165), whereas VEGF(121) and VEGF(189) increased fibroblast growth. These effects on proliferation were associated with an activation of the ERK pathway, as revealed using the TransAM c-Myc assay. Inhibition of the ERK pathway, by PD98059 administration, significantly reduced VEGF isoform induced cell growth. A dose-dependent reduction of endothelial cell proliferation was observed after pegaptanib administration, while only the highest dose was able to inhibit fibroblast growth. Next, the in vivo effect of pegaptanib was investigated in a rabbit model of trabeculectomy. The surgical outcome was evaluated by performing clinical investigations (IOP, bleb area, height and survival), as well as histomorphometric analyses of angiogenesis (CD31), inflammation (CD45) and fibrosis (Sirius Red). A single postoperative application of pegaptanib had a beneficial impact on surgical outcome, mainly by reducing angiogenesis, but not inflammation or collagen deposition. Repeated injections slightly improved surgical outcome, but again solely by reducing angiogenesis. In summary, our results revealed that the VEGF isoforms play a differential role in ocular wound healing: VEGF(165) and VEGF(121) predominantly affect blood vessel growth, whereas VEGF(189) is rather involved in fibrosis, an important process in wound healing.


Asunto(s)
Glaucoma/cirugía , Malla Trabecular/patología , Trabeculectomía , Factor A de Crecimiento Endotelial Vascular/fisiología , Animales , Aptámeros de Nucleótidos/farmacología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Fibrosis , Humanos , Presión Intraocular/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Neuropilina-1/genética , Isoformas de Proteínas/fisiología , ARN Mensajero/metabolismo , Conejos , Reacción en Cadena en Tiempo Real de la Polimerasa , Cápsula de Tenon/citología , Malla Trabecular/metabolismo , Venas Umbilicales/citología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Cicatrización de Heridas/fisiología
12.
Curr Eye Res ; 46(8): 1166-1170, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33372561

RESUMEN

PURPOSE: Pentosan polysulfate sodium (PPS; Elmiron) is a FDA-approved heparanase inhibitor for the treatment of bladder pain and interstitial cystitis. The chronic use of PPS has been associated with a novel pigmentary maculopathy, associated with discrete vitelliform deposits that exhibit hyperfluorescence, macular hyper-pigmentary spots, and foci of nodular RPE enlargement. Therefore, this study aimed to investigate the retinal morphology of heparanase knockout mice. MATERIAL AND METHODS: The retinal morphology of heparanase knock-out and age-matched control wild type mice of 3-, 9- and 15-weeks old was characterized by means of histological evaluation. Immuno-histological stains for RPE65, F4/80 and Ki67 were performed for investigating the RPE, inflammatory and proliferating cells, respectively. RESULTS: Histological analysis showed no changes in age-matched wild-type controls, whereas the eyes of heparanase null mice were characterized by alterations in RPE and neural retina, as manifest by RPE folds and choroidal thickening, detached RPE cells, thickening of the photoreceptor layer and retinal disorganization. The presence of discrete hyperfluorescent foci, however, was absent. The prevalence of the RPE/choroidal changes or protrusions seemed to progress over time and were correlated with more RPE65 signal rather than influx of F4/80- or Ki67-positive cells. These results indicate that the subretinal alterations were mostly RPE driven, without influx of inflammatory or proliferating cells. CONCLUSIONS: Our results indicate that heparanase deficiency in the mice leads to RPE folds, choroidal thickening, and retinal disorganization. The presence of discrete hyperfluorescent foci, a key characteristic of the human disease, was not observed. However, it can be concluded that some of the observations in mice are similar to those seen after chronic use of PPS in humans. These findings indicate that the toxicity observed in the presence of heparanase inhibitors is target-related and will preclude the clinical use of heparanase inhibition as a therapeutic intervention.


Asunto(s)
Enfermedades de la Coroides/enzimología , Glucuronidasa/deficiencia , Desprendimiento de Retina/enzimología , Epitelio Pigmentado de la Retina/enzimología , Animales , Anticoagulantes , Proteínas de Unión al Calcio/metabolismo , Enfermedades de la Coroides/diagnóstico , Enfermedades de la Coroides/metabolismo , Angiografía con Fluoresceína , Glucuronidasa/genética , Antígeno Ki-67/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Poliéster Pentosan Sulfúrico , Receptores Acoplados a Proteínas G/metabolismo , Desprendimiento de Retina/diagnóstico , Desprendimiento de Retina/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología , Tomografía de Coherencia Óptica , cis-trans-Isomerasas/metabolismo
13.
Invest Ophthalmol Vis Sci ; 62(13): 18, 2021 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-34677569

RESUMEN

Purpose: To investigate the effect of plasma kallikrein (PKal)-inhibition by THR-149 on preventing key pathologies associated with diabetic macular edema (DME) in a rat model. Methods: Following streptozotocin-induced diabetes, THR-149 or its vehicle was administered in the rat via either a single intravitreal injection or three consecutive intravitreal injections (with a 1-week interval; both, 12.5 µg/eye). At 4 weeks post-diabetes, the effect of all groups was compared by histological analysis of Iba1-positive retinal inflammatory cells, inflammatory cytokines, vimentin-positive Müller cells, inwardly rectifying potassium and water homeostasis-related channels (Kir4.1 and AQP4, respectively), vascular leakage (fluorescein isothiocyanate-labeled bovine serum albumin), and retinal thickness. Results: Single or repeated THR-149 injections resulted in reduced inflammation, as depicted by decreasing numbers and activation state of immune cells and IL-6 cytokine levels in the diabetic retina. The processes of reactive gliosis, vessel leakage, and retinal thickening were only significantly reduced after multiple THR-149 administrations. Individual retinal layer analysis showed that repeated THR-149 injections significantly decreased diabetes-induced thickening of the inner plexiform, inner nuclear, outer nuclear, and photoreceptor layers. At the glial-vascular interface, reduced Kir4.1-channel levels in the diabetic retina were restored to control non-diabetic levels in the presence of THR-149. In contrast, little or no effect of THR-149 was observed on the AQP4-channel levels. Conclusions: These data demonstrate that repeated THR-149 administration reduces several DME-related key pathologies such as retinal thickening and neuropil disruption in the diabetic rat. These observations indicate that modulation of the PKal pathway using THR-149 has clinical potential to treat patients with DME.


Asunto(s)
Anticoagulantes/administración & dosificación , Retinopatía Diabética/sangre , Calicreína Plasmática/antagonistas & inhibidores , Retina/patología , Tomografía de Coherencia Óptica/métodos , Animales , Biomarcadores/sangre , Diabetes Mellitus Experimental , Retinopatía Diabética/patología , Inyecciones Intravítreas , Masculino , Calicreína Plasmática/metabolismo , Ratas , Ratas Endogámicas BN , Retina/metabolismo
14.
Prog Retin Eye Res ; 85: 100966, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33775825

RESUMEN

Integrins are a class of transmembrane receptors that are involved in a wide range of biological functions. Dysregulation of integrins has been implicated in many pathological processes and consequently, they are attractive therapeutic targets. In the ophthalmology arena, there is extensive evidence suggesting that integrins play an important role in diabetic retinopathy (DR), age-related macular degeneration (AMD), glaucoma, dry eye disease and retinal vein occlusion. For example, there is extensive evidence that arginyl-glycyl-aspartic acid (Arg-Gly-Asp; RGD)-binding integrins are involved in key disease hallmarks of DR and neovascular AMD (nvAMD), specifically inflammation, vascular leakage, angiogenesis and fibrosis. Based on such evidence, drugs that engage integrin-linked pathways have received attention for their potential to block all these vision-threatening pathways. This review focuses on the pathophysiological role that RGD-binding integrins can have in complex multifactorial retinal disorders like DR, diabetic macular edema (DME) and nvAMD, which are leading causes of blindness in developed countries. Special emphasis will be given on how RGD-binding integrins can modulate the intricate molecular pathways and regulate the underlying pathological mechanisms. For instance, the interplay between integrins and key molecular players such as growth factors, cytokines and enzymes will be summarized. In addition, recent clinical advances linked to targeting RGD-binding integrins in the context of DME and nvAMD will be discussed alongside future potential for limiting progression of these diseases.


Asunto(s)
Diabetes Mellitus , Retinopatía Diabética , Edema Macular , Degeneración Macular Húmeda , Inhibidores de la Angiogénesis/uso terapéutico , Retinopatía Diabética/tratamiento farmacológico , Humanos , Integrinas/uso terapéutico , Oligopéptidos/uso terapéutico , Factor A de Crecimiento Endotelial Vascular , Agudeza Visual
15.
Cell Metab ; 31(4): 862-877.e14, 2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32268117

RESUMEN

Endothelial cell (EC) metabolism is an emerging target for anti-angiogenic therapy in tumor angiogenesis and choroidal neovascularization (CNV), but little is known about individual EC metabolic transcriptomes. By single-cell RNA sequencing 28,337 murine choroidal ECs (CECs) and sprouting CNV-ECs, we constructed a taxonomy to characterize their heterogeneity. Comparison with murine lung tumor ECs (TECs) revealed congruent marker gene expression by distinct EC phenotypes across tissues and diseases, suggesting similar angiogenic mechanisms. Trajectory inference predicted that differentiation of venous to angiogenic ECs was accompanied by metabolic transcriptome plasticity. ECs displayed metabolic transcriptome heterogeneity during cell-cycle progression and in quiescence. Hypothesizing that conserved genes are important, we used an integrated analysis, based on congruent transcriptome analysis, CEC-tailored genome-scale metabolic modeling, and gene expression meta-analysis in cross-species datasets, followed by in vitro and in vivo validation, to identify SQLE and ALDH18A1 as previously unknown metabolic angiogenic targets.


Asunto(s)
Células Endoteliales/metabolismo , Neoplasias Pulmonares/metabolismo , Degeneración Macular/metabolismo , Neovascularización Patológica/metabolismo , Transcriptoma , Animales , Células Endoteliales/citología , Células Endoteliales/patología , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia de ARN , Análisis de la Célula Individual
16.
Prog Retin Eye Res ; 69: 116-136, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30385175

RESUMEN

Placental growth factor (PlGF) is a member of the vascular endothelial growth factor (VEGF) family. Upon binding to VEGF- and neuropilin-receptor sub-types, PlGF modulates a range of neural, glial and vascular cell responses that are distinct from VEGF-A. As PlGF expression is selectively associated with pathological angiogenesis and inflammation, its blockade does not affect the healthy vasculature. PlGF actions have been extensively described in tumor biology but more recently there has been accumulating preclinical evidence that indicates that this growth factor could have an important role in retinal diseases. High levels of PlGF have been found in aqueous humor, vitreous and/or retina of patients exhibiting retinopathies, especially those with diabetic retinopathy (DR) and neovascular age-related macular degeneration (nvAMD). Expression of this growth factor seems to correlate closely with many of the key pathogenic features of early and late retinopathy in preclinical models. For example, studies using genetic modification and/or pharmacological treatment to block PlGF in the laser-induced choroidal neovascularization (CNV) model, oxygen-induced retinopathy model, as well as various murine diabetic models, have shown that PlGF deletion or inhibition can reduce neovascularization, retinal leakage, inflammation and gliosis, without affecting vascular development or inducing neuronal degeneration. Moreover, an inhibitory effect of PlGF blockade on retinal scarring in the mouse CNV model has also been recently demonstrated and was found to be unique for PlGF inhibition, as compared to various VEGF inhibition strategies. Together, these preclinical results suggest that anti-PlGF therapy might have advantages over anti-VEGF treatment, and that it may have clinical applications as a standalone treatment or in combination with anti-VEGF. Additional clinical studies are clearly needed to further elucidate the role of PlGF and its potential as a therapeutic target in ocular diseases.


Asunto(s)
Neuropilinas/fisiología , Factor de Crecimiento Placentario/fisiología , Enfermedades de la Retina/metabolismo , Animales , Neovascularización Coroidal/metabolismo , Retinopatía Diabética/metabolismo , Modelos Animales de Enfermedad , Humanos , Degeneración Macular/metabolismo , Factores de Crecimiento Endotelial Vascular/fisiología
17.
Curr Eye Res ; 44(8): 813-822, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31055948

RESUMEN

There is growing evidence that placental growth factor (PlGF) is an important player in multiple pathologies, including tumorigenesis, inflammatory disorders and degenerative retinopathies. PlGF is a member of the vascular endothelial growth factor (VEGF) family and in the retina, binding of this growth factor to specific receptors is associated with pathological angiogenesis, vascular leakage, neurodegeneration and inflammation. Although they share some receptor signalling pathways, many of the actions of PlGF are distinct from VEGF and this has revealed the enticing prospect that it could be a useful therapeutic target for treating early and late stages of diabetic retinopathy (DR) and neovascular age-related macular degeneration (AMD). Recent research suggests that modulation of PlGF could also be important in the geographic atrophy (GA) form of late AMD by protecting the outer retina and the retinal pigment epithelium (RPE). This review discusses PlGF and its signalling pathways and highlights the potential of blocking the bioactivity of this growth factor to treat irreversible visual loss due to the two main forms of AMD.


Asunto(s)
Degeneración Macular/fisiopatología , Factor de Crecimiento Placentario/fisiología , Transducción de Señal/fisiología , Células Epiteliales Alveolares/fisiología , Humanos , Epitelio Pigmentado de la Retina/fisiología
18.
Invest Ophthalmol Vis Sci ; 60(2): 807-822, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30811545

RESUMEN

Purpose: The goal of this study was to perform an extensive temporal characterization of the early pathologic processes in the streptozotocin (STZ)-induced diabetic retinopathy (DR) mouse model, beyond the vascular phenotype, and to investigate the potential of clinically relevant compounds in attenuating these processes. Methods: Visual acuity and contrast sensitivity (CS) were studied in the mouse STZ model until 24 weeks postdiabetes onset. ERG, spectral domain optical coherence tomography (SD-OCT), leukostasis, and immunohistochemistry were applied to investigate neurodegeneration, inflammation, and gliosis during early-, mid- and late-phase diabetes. Aflibercept or triamcinolone acetonide (TAAC) was administered to investigate their efficacy on the aforementioned processes. Results: Visual acuity and CS loss started at 4 and 18 weeks postdiabetes onset, respectively, and progressively declined over time. ERG amplitudes were diminished and OP latencies increased after 6 weeks, whereas SD-OCT revealed retinal thinning from 4 weeks postdiabetes. Immunohistochemical analyses linked these findings to retinal ganglion and cholinergic amacrine cell loss at 4 and 8 weeks postdiabetes onset, respectively, which was further decreased after aflibercept administration. The number of adherent leukocytes was augmented after 2 weeks, whereas increased micro- and macroglia reactivity was present from 4 weeks postdiabetes. Aflibercept or TAAC showed improved efficacy on inflammation and gliosis. Conclusions: STZ-induced diabetic mice developed early pathologic DR hallmarks, from which inflammation seemed the initial trigger, leading to further development of functional and morphologic retinal changes. These findings indicate that the mouse STZ model is suitable to study novel integrative non-vascular therapies to treat early DR.


Asunto(s)
Sensibilidad de Contraste/fisiología , Diabetes Mellitus Experimental/patología , Retinopatía Diabética/patología , Modelos Animales de Enfermedad , Retina/fisiopatología , Agudeza Visual/fisiología , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Diabetes Mellitus Experimental/tratamiento farmacológico , Retinopatía Diabética/tratamiento farmacológico , Electrorretinografía , Estudios de Seguimiento , Glucocorticoides/uso terapéutico , Inmunohistoquímica , Leucostasis , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Factores de Crecimiento Endotelial Vascular/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Estreptozocina , Tomografía de Coherencia Óptica , Resultado del Tratamiento , Triamcinolona Acetonida/uso terapéutico
19.
J Med Chem ; 61(7): 2823-2836, 2018 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-29517911

RESUMEN

Plasma kallikrein, a member of the kallikrein-kinin system, catalyzes the release of the bioactive peptide bradykinin, which induces inflammation, vasodilation, vessel permeability, and pain. Preclinical evidence implicates the activity of plasma kallikrein in diabetic retinopathy, which is a leading cause of visual loss in patients suffering from diabetes mellitus. Employing a technology based on phage-display combined with chemical cyclization, we have identified highly selective bicyclic peptide inhibitors with nano- and picomolar potencies toward plasma kallikrein. Stability in biological matrices was either intrinsic to the peptide or engineered via the introduction of non-natural amino acids and nonpeptidic bonds. The peptides prevented bradykinin release in vitro, and in vivo efficacy was demonstrated in both a rat paw edema model and in rodent models of diabetes-induced retinal permeability. With a highly extended half-life of ∼40 h in rabbit eyes following intravitreal administration, the bicyclic peptides are promising novel agents for the treatment of diabetic retinopathy and diabetic macular edema.


Asunto(s)
Compuestos Bicíclicos con Puentes/síntesis química , Compuestos Bicíclicos con Puentes/farmacología , Complicaciones de la Diabetes/tratamiento farmacológico , Retinopatía Diabética/tratamiento farmacológico , Edema Macular/tratamiento farmacológico , Edema Macular/etiología , Calicreína Plasmática/antagonistas & inhibidores , Inhibidores de Proteasas/síntesis química , Inhibidores de Proteasas/farmacología , Animales , Bradiquinina/metabolismo , Edema/tratamiento farmacológico , Ojo/metabolismo , Pie/patología , Semivida , Inyecciones Intravítreas , Masculino , Ratones , Ratones Endogámicos C57BL , Permeabilidad , Inhibidores de Proteasas/administración & dosificación , Conejos , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad , Especificidad por Sustrato , Cuerpo Vítreo/química , Cuerpo Vítreo/metabolismo
20.
Invest Ophthalmol Vis Sci ; 57(10): 4347-55, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27556218

RESUMEN

PURPOSE: The complementary effects of mitomycin-C (MMC) and anti-placental growth factor (PlGF) therapy were explored and compared to the combined administration of MMC and aflibercept. Additionally, the effect of PlGF (inhibition) on IOP was investigated, since aqueous PlGF is known to be upregulated in glaucoma patients. METHODS: In the trabeculectomy mouse model, intracameral injection(s) of the PlGF inhibitor (5D11D4) were compared to MMC or aflibercept and to the combination of both compounds. Treatment outcome was studied by bleb investigation and by Sirius Red staining. The effect of subconjunctival PlGF administration and topical 5D11D4 on IOP was investigated in normotensive mice and was compared to topical administration of latanoprost, the gold standard for IOP-lowering. RESULTS: Combination of MMC and 5D11D4 was able to significantly improve surgical outcome compared to monotherapy of MMC or 5D11D4 (n = 20; P < 0.001). Compared to combined treatment of MMC with aflibercept, the simultaneous administration of MMC and 5D11D4 was equally efficacious in improving surgical outcome (n = 15; P = 0.88). In normotensive mice, 5D11D4 was able to significantly reduce the IOP-elevation induced by PlGF (n = 10; P < 0.05), whereas no effect of 5D11D4 was seen in naive mice, which was in contrast to latanoprost. CONCLUSIONS: The current data suggest that application of MMC together with PlGF inhibition may have complementary effects in the improvement of surgical outcome and is equally efficacious as the combined treatment of MMC and aflibercept. Inhibition of PlGF also might open alternative perspectives as IOP-lowering strategy for glaucoma patients with increased aqueous PlGF levels.


Asunto(s)
Proteínas Sanguíneas/administración & dosificación , Cicatriz/prevención & control , Cirugía Filtrante/efectos adversos , Glaucoma/cirugía , Guías como Asunto , Mitomicina/administración & dosificación , Animales , Cámara Anterior , Proteínas Sanguíneas/farmacocinética , Cicatriz/etiología , Cicatriz/metabolismo , Modelos Animales de Enfermedad , Glaucoma/metabolismo , Glaucoma/patología , Inyecciones , Presión Intraocular , Ratones , Ratones Endogámicos C57BL , Mitomicina/farmacocinética , Inhibidores de la Síntesis del Ácido Nucleico/administración & dosificación , Inhibidores de la Síntesis del Ácido Nucleico/farmacocinética , Complicaciones Posoperatorias/prevención & control
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA