Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Tipo de estudio
Intervalo de año de publicación
1.
bioRxiv ; 2024 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-38562805

RESUMEN

Ibudilast, an inhibitor of macrophage migration inhibitory factor (MIF) and phosphodiesterase (PDE), has been recently shown to have neuroprotective effects in a variety of neurologic diseases. We utilize a chick excitotoxic retinal damage model to investigate ibudilast's potential to protect retinal neurons. Using single cell RNA-sequencing (scRNA-seq), we find that MIF, putative MIF receptors CD74 and CD44, and several PDEs are upregulated in different retinal cells during damage. Intravitreal ibudilast is well tolerated in the eye and causes no evidence of toxicity. Ibudilast effectively protects neurons in the inner nuclear layer from NMDA-induced cell death, restores retinal layer thickness on spectral domain optical coherence tomography, and preserves retinal neuron function, particularly for the ON bipolar cells, as assessed by electroretinography. PDE inhibition seems essential for ibudilast's neuroprotection, as AV1013, the analogue that lacks PDE inhibitor activity, is ineffective. scRNA-seq analysis reveals upregulation of multiple signaling pathways, including mTOR, in damaged Müller glia (MG) with ibudilast treatment compared to AV1013. Components of mTORC1 and mTORC2 are upregulated in both bipolar cells and MG with ibudilast. The mTOR inhibitor rapamycin blocked accumulation of pS6 but did not reduce TUNEL positive dying cells. Additionally, through ligand-receptor interaction analysis, crosstalk between bipolar cells and MG may be important for neuroprotection. We have identified several paracrine signaling pathways that are known to contribute to cell survival and neuroprotection and might play essential roles in ibudilast function. These findings highlight ibudilast's potential to protect inner retinal neurons during damage and show promise for future clinical translation.

2.
PLoS One ; 16(9): e0257148, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34492087

RESUMEN

OBJECTIVES: The chick is rapidly becoming a standardized preclinical model in vision research to study mechanisms of ocular disease. We seek to comprehensively evaluate the N-methyl-D-aspartate (NMDA) model of excitotoxic retinal damage using multimodal imaging, functional, and histologic approaches in NMDA-damaged, vehicle-treated, and undamaged chicks. METHODS: Chicks were either left undamaged in both eyes or were injected with NMDA in the left eye and saline (vehicle) in the right eye. TUNEL assay was performed on chicks to assess levels of retinal cell death one day post-injection of NMDA or saline and on age-matched untreated chicks. Spectral domain optical coherence tomography (SD-OCT) was performed weekly on chicks and age-matched controls day 1 (D1) up to D28 post-injection. Light adapted electroretinograms (ERG) were performed alongside SD-OCT measurements on post-injection chicks along with age-matched untreated controls. RESULTS: Untreated and vehicle-treated eyes had no TUNEL positive cells while NMDA-treated eyes accumulated large numbers of TUNEL positive cells in the Inner Nuclear Layer (INL), but not other layers, at D1 post injection. Significant inner retina swelling or edema was found on SD-OCT imaging at D1 post-injection which resolved at subsequent timepoints. Both the INL and the inner plexiform layer significantly thinned by one-week post-injection and did not recover for the duration of the measurements. On ERG, NMDA-treated eyes had significantly reduced amplitudes of all parameters at D1 with all metrics improving over time. The b-wave, oscillatory potentials, and ON/OFF bipolar responses were the most affected with at least 70% reduction immediately after damage compared to the fellow eye control. CONCLUSION: This study establishes a normative baseline on the retinal health and gross functional ability as well as intraocular pressures of undamaged, vehicle-treated, and NMDA-damaged chicks to provide a standard for comparing therapeutic treatment studies in this important animal model.


Asunto(s)
Imagen Multimodal , Retina/patología , Enfermedades de la Retina/inducido químicamente , Enfermedades de la Retina/diagnóstico por imagen , Animales , Pollos , Modelos Animales de Enfermedad , Electrorretinografía , Fondo de Ojo , Imagenología Tridimensional , Presión Intraocular , N-Metilaspartato , Retina/diagnóstico por imagen , Retina/fisiopatología , Enfermedades de la Retina/patología , Tomografía de Coherencia Óptica
5.
Invest Ophthalmol Vis Sci ; 60(1): 349-357, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30682205

RESUMEN

Purpose: Mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) signaling is neuroprotective in some retinal damage models but its role in neuronal survival during retinal detachment (RD) is unclear. In addition, serous RDs are a prevalent side effect of MEK inhibitors (MEKi), blocking MAPK/ERK signaling for treatment of certain cancers. We tested the hypothesis that MEKi treatment in experimental RD would increase photoreceptor death. Methods: The MEKi selumetinib was delivered daily to C57BL/6 mice at a clinically relevant dose (10 mg/mL) starting 1 day prior to creating RD with subretinal hyaluronic acid injection. Photoreceptor TUNEL and outer nuclear layer (ONL) thickness were analyzed. Phospho-ERK1/2 (pERK) distribution, glial fibrillary acidic protein (GFAP) accumulation, and Iba-1 (microglia/macrophages) were evaluated with immunofluorescence. Results: pERK accumulated in the Müller glia in detached retinas, but this was effectively blocked by selumetinib. Selumetinib did not induce serous RDs at day 1 and did not increase TUNEL positive photoreceptors or further decrease ONL thickness compared to controls. Retinal gliosis was not altered, but selumetinib did block the increase in intraretinal microglia/macrophage Iba-1 fluorescence intensity and acquisition of amoeboid morphology. Conclusions: MAPK/ERK is neuroprotective in some retinal damage models; in RD, selumetinib blocked Müller pERK accumulation and changed the retinal microglia/macrophage phenotype but did not alter photoreceptor survival. This is consistent with the relatively good visual acuity seen in patients developing transient retinal detachments on MEK inhibitor therapy. Compensation by other neuroprotective pathways in the retina during retinal detachment may occur in the presence of MEK inhibition.


Asunto(s)
Bencimidazoles/farmacología , Células Fotorreceptoras de Vertebrados/efectos de los fármacos , Desprendimiento de Retina/patología , Administración Oral , Animales , Bencimidazoles/administración & dosificación , Proteínas de Unión al Calcio/metabolismo , Supervivencia Celular/fisiología , Células Ependimogliales/efectos de los fármacos , Células Ependimogliales/metabolismo , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Células Fotorreceptoras de Vertebrados/metabolismo , Desprendimiento de Retina/enzimología
6.
Exp Eye Res ; 176: 121-129, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29959928

RESUMEN

Nuclear co-localization labels are critical to ocular research. Among these, the TUNEL assay has been established as a gold standard of cell death and apoptosis. While several validated computer-based methods exist to quantitate these markers, including ImageJ Retina Analysis (RA) Toolkit and ImagePro, none verify the count with the nuclear counter stain to confirm nuclear co-localization. We established a new ImageJ-based automated multichannel thresholding (MCT) method to quantitate nuclear co-localized labeling. The MCT method was validated by comparing it with the two published TUNEL analysis in TUNEL-positive photoreceptors in an experimental retinal detachment (RD) model. RDs were induced in murine eyes and cross-sectional images of TUNEL and DAPI counter stain were obtained. Images were classified as "typical" or high density "hotspot" TUNEL regions (n = 10/group). Images were analyzed and compared between the MCT method and the published techniques including both "standard" and "high" settings of the RA Toolkit for detecting lower or higher TUNEL densities, respectively. Additional testing of the MCT method with built-in ImageJ thresholding algorithms was performed to produce fully automated measurements. All images were compared with Bland-Altman mean difference plots to assess the difference in counts and linear regression plots to assess correlation. Comparison between the MCT method and the ImagePro method were found to be well correlated (typical: R2 = 0.8972, hotspot: R2 = 0.9000) with minor to non-significant differences. The RA Toolkit settings were found to be mostly well correlated as well (standard/typical: R2 = 0.8036, standard/hotspot: R2 = 0.4309, high/typical: R2 = 0.7895, high/hotspot: R2 = 0.8738) but were often found to have significantly higher counts than the MCT. In conclusion, the MCT method compared favorably with validated computer-based methods of nuclear marker immunofluorescence quantitation and avoids staining artifacts through the incorporation of the nuclear counter stain to confirm positive cells.


Asunto(s)
Apoptosis , Etiquetado Corte-Fin in Situ/métodos , Células Fotorreceptoras de Vertebrados/patología , Desprendimiento de Retina/patología , Animales , Núcleo Celular/patología , ADN/análisis , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Colorantes Fluorescentes/metabolismo , Indoles/metabolismo , Ratones , Ratones Endogámicos C57BL , Células Fotorreceptoras de Vertebrados/metabolismo , Desprendimiento de Retina/metabolismo
7.
J Pharmacol Exp Ther ; 324(3): 1227-33, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18079357

RESUMEN

Erythropoietin (Epo) receptor (EpoR) is expressed in several cancer cell lines, and the functional consequence of this expression is under extensive study. In this study, we used a cervical cancer cell line in which EpoR was first found to be expressed and to correlate with the severity of the disease. We demonstrate that Epo is a chemoattractant for these cancer cells, enhancing their migration under serum-starved conditions. Using a Transwell migration system, we show that Epo enhances cancer cell migration in a dose- and time-dependent manner. The effect of Epo is dependent on the activity of two signaling pathways: the mitogen-activated protein kinase (MAPK) pathway and the RhoA GTPase pathway. We show that Epo activates both pathways in a Janus kinase-dependent manner and that this activation is required for Epo effects on cell migration. Furthermore, we use both pharmacological and genetic inhibitors to demonstrate that the activation of RhoA GTPase is dependent on the activity of the MAPK pathway, providing the first evidence for interaction between these two signaling cascades.


Asunto(s)
Movimiento Celular/fisiología , Eritropoyetina/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neoplasias/enzimología , Proteína de Unión al GTP rhoA/metabolismo , Movimiento Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Eritropoyetina/toxicidad , Células HeLa , Humanos , Neoplasias/inducido químicamente , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
8.
Biochem Pharmacol ; 74(4): 590-600, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17586475

RESUMEN

Erythropoietin (Epo) is a cytokine that is required for the survival of erythroid progenitors through interaction with its receptor on the surface of these cells. Recent studies showed that erythropoietin receptor (EpoR) is expressed on many cancer cells. The factors that govern EpoR expression on the cell surface are poorly understood. Using both biotinlyation and radiolabeled Epo binding experiments, we show here that Epo starvation of the Epo-dependent erythroleukemia cell line, ASE2, leads to a time-dependent increase in both forms of EpoR, the maturing 64 kDa and the mature 66 kDa proteins. Mevalonate depletion inhibits the formation of the highly glycosylated mature form of EpoR without affecting the other form. Treatment of cells with lovastatin, a selective inhibitor of the rate-limiting enzyme in the mevalonate pathway leads to inhibition of cell surface EpoR that is induced by Epo starvation. The effect of lovastatin appears to be the consequence of inhibition of two processes, glycosylation and geranylgeranylation. Adding back geranylgeranyl pyrophosphate to lovastatin-treated cells completely prevents the lovastatin effect on EpoR expression. Dolichol, the sugar carrier in N-linked glycosylation that is derived from the mevalonate pathway, partially reverses lovastatin's effect. The glycosylation inhibitor tunicamycin also partially suppresses EpoR surface expression. Inhibiting protein geranylgeranylation mimics the effect of lovastatin and inhibits EpoR surface expression in a concentration-dependent manner. Finally, lovastatin inhibits Epo's stimulatory effects on cell proliferation. These results indicate that mevalonate derivatives are required for normal EpoR expression on the cell surface through two pathways, glycosylation and geranylgeranylation.


Asunto(s)
Lovastatina/farmacología , Receptores de Eritropoyetina/antagonistas & inhibidores , Antibacterianos/farmacología , Anticolesterolemiantes/farmacología , Antivirales/farmacología , Biotinilación/métodos , Western Blotting , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Dolicoles/antagonistas & inhibidores , Dolicoles/biosíntesis , Relación Dosis-Respuesta a Droga , Eritropoyetina/farmacología , Glicosilación/efectos de los fármacos , Humanos , Radioisótopos de Yodo , Janus Quinasa 2/antagonistas & inhibidores , Janus Quinasa 2/metabolismo , Leucina/análogos & derivados , Leucina/farmacología , Ácido Mevalónico/metabolismo , Ácido Mevalónico/farmacología , Fosforilación/efectos de los fármacos , Fosfatos de Poliisoprenilo/metabolismo , Fosfatos de Poliisoprenilo/farmacología , Prenilación de Proteína/efectos de los fármacos , Receptores de Eritropoyetina/metabolismo , Factor de Transcripción STAT5/antagonistas & inhibidores , Factor de Transcripción STAT5/metabolismo , Tunicamicina/farmacología
9.
J Pharmacol Exp Ther ; 316(1): 403-9, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16203826

RESUMEN

Erythropoietin (Epo) acts through the erythropoietin receptor, a member of the type-1 cytokine receptor family, to influence survival, proliferation, and differentiation of erythroid progenitors. Epo stimulation of factor-dependent 32D cells results in phosphorylation of many proteins, including Janus kinase (Jak) 2, signal transducer and activator of transcription (Stat) 5, and extracellular signal-regulated kinase (Erk). Some of Epo-activated signaling proteins require isoprenylation, either farnesylation or geranylgeranylation, for post-translational modification. In this study, we sought to characterize the interplay between protein isoprenylation and Epo signal transduction. Using two different Epo-responsive cell lines, we found that depletion of mevalonate and its isoprenoid derivatives using the 3-hydroxy-3-methylglutaryl (HMG)-CoA reductase inhibitor lovastatin impairs Epo signaling as assessed by phosphorylation of cellular substrates and inhibition of apoptosis. Interestingly, the effect of mevalonate depletion was prevented by adding back geranylgeranyl pyrophosphate but not farnesyl pyrophosphate. Furthermore, selective inhibition of protein geranylgeranylation mimicked the effect of lovastatin, whereas selective inhibition of farnesylation had no effect. These results indicate that protein geranylgeranylation and not farnesylation is important for proper Epo signal transduction.


Asunto(s)
Fosfatos de Poliisoprenilo/metabolismo , Receptores de Eritropoyetina/fisiología , Transducción de Señal/fisiología , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Línea Celular , Eritropoyetina/metabolismo , Eritropoyetina/fisiología , Glicosilación , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Janus Quinasa 2 , Lovastatina/farmacología , Ácido Mevalónico/metabolismo , Ratones , Proteínas Tirosina Quinasas/metabolismo , Proteínas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Eritropoyetina/biosíntesis , Factor de Transcripción STAT5/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...