Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(14)2023 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-37511316

RESUMEN

Oxidative stress (OS)-induced mitochondrial damage is a risk factor for primary open-angle glaucoma (POAG). Mitochondria-targeted novel antioxidant therapies could unearth promising drug candidates for the management of POAG. Previously, our dual-acting hybrid molecule SA-2 with nitric oxide-donating and antioxidant activity reduced intraocular pressure and improved aqueous humor outflow in rodent eyes. Here, we examined the mechanistic role of SA-2 in trabecular meshwork (TM) cells in vitro and measured the activity of intracellular antioxidant enzymes during OS. Primary human TM cells isolated from normal (hNTM) or glaucomatous (hGTM) post-mortem donors and transformed glaucomatous TM cells (GTM-3) were used for in vitro assays. We examined the effect of SA-2 on oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) in vitro using Seahorse Analyzer with or without the oxidant, tert-butyl hydroperoxide (TBHP) treatment. Concentrations of total antioxidant enzymes, catalase (CAT), malondialdehyde (MDA), and glutathione peroxidase (GPx) were measured. We observed significant protection of both hNTM and hGTM cells from TBHP-induced cell death by SA-2. Antioxidant enzymes were elevated in SA-2-treated cells compared to TBHP-treated cells. In addition, SA-2 demonstrated an increase in mitochondrial metabolic parameters. Altogether, SA-2 protected both normal and glaucomatous TM cells from OS via increasing mitochondrial energy parameters and the activity of antioxidant enzymes.


Asunto(s)
Glaucoma de Ángulo Abierto , Glaucoma , Humanos , Antioxidantes/metabolismo , Malla Trabecular/metabolismo , Glaucoma de Ángulo Abierto/tratamiento farmacológico , Glaucoma de Ángulo Abierto/metabolismo , Glaucoma/tratamiento farmacológico , Glaucoma/metabolismo , Mitocondrias/metabolismo
2.
Invest Ophthalmol Vis Sci ; 62(10): 17, 2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34406331

RESUMEN

Purpose: The purpose of this study was to determine the effects of the Sigma-1R (σ-1r) on retinal ganglion cell (RGC) survival following optic nerve crush (ONC) and the signaling mechanism involved in the σ-1r protection. Methods: The overall strategy was to induce injury by ONC and mitigate RGC death by increasing σ-1r expression and/or activate σ-1r activity in σ-1r K/O mice and wild type (WT) mice. AAV2-σ-1r vector was used to increase σ-1r expression and σ-1r agonist used to activate the σ-1r and RGCs were counted. Immunohistochemical and Western blot analysis determined phosphorylated (p)-c-Jun, c-Jun, and Caspase-3. Pattern electroretinography (PERG) determined RGC activity. Results: RGC counts and function were similar in pentazocine-treated WT mice when compared to untreated mice and in WT mice when compared with σ-1r K/O mice. Pentazocine-induced effects and the effects of σ-1r K/O were only observable after ONC. ONC resulted in decreased RGC counts and activity in both WT and σ-1r K/O mice, with σ-1r K/O mice experiencing significant decreases compared with WT mice. The σ-1r transgenic expression resulted in increased RGC counts and activity following ONC. In WT mice, treatment with σ-1r agonist pentazocine resulted in increased RGC counts and increased activity when compared with untreated WT mice. There were time-dependent increases in c-jun, p-c-jun, and caspase-3 expression in ONC mice that were mitigated with pentazocine-treatment. Conclusions: These findings suggest that the apoptotic pathway is involved in RGC losses seen in an ONC model. The σ-1r offers neuroprotection, as activation and/or transgenic expression of σ-1r attenuated the apoptotic pathway and restored RGCs number and function following ONC.


Asunto(s)
Glaucoma/genética , Traumatismos del Nervio Óptico/genética , Receptores sigma/genética , Células Ganglionares de la Retina/patología , Animales , Apoptosis , Modelos Animales de Enfermedad , Electrorretinografía , Glaucoma/complicaciones , Glaucoma/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Compresión Nerviosa/métodos , Traumatismos del Nervio Óptico/etiología , Traumatismos del Nervio Óptico/patología , Receptores sigma/biosíntesis , Células Ganglionares de la Retina/metabolismo , Transducción de Señal , Receptor Sigma-1
3.
Antioxidants (Basel) ; 10(4)2021 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-33917924

RESUMEN

Oxidative stress induced death and dysregulation of trabecular meshwork (TM) cells contribute to the increased intraocular pressure (IOP) in primary open angle (POAG) glaucoma patients. POAG is one of the major causes of irreversible vision loss worldwide. Nitric oxide (NO), a small gas molecule, has demonstrated IOP lowering activity in glaucoma by increasing aqueous humor outflow and relaxing TM. Glaucomatous pathology is associated with decreased antioxidant enzyme levels in ocular tissues causing increased reactive oxygen species (ROS) production that reduce the bioavailability of NO. Here, we designed, synthesized, and conducted in vitro studies of novel second-generation sulfur containing hybrid NO donor-antioxidants SA-9 and its active metabolite SA-10 to scavenge broad-spectrum ROS as well as provide efficient protection from t-butyl hydrogen peroxide (TBHP) induced oxidative stress while maintaining NO bioavailability in TM cells. To allow a better drug delivery, a slow release nanosuspension SA-9 nanoparticles (SA-9 NPs) was prepared, characterized, and tested in dexamethasone induced ocular hypertensive (OHT) mice model for IOP lowering activity. A single topical eye drop of SA-9 NPs significantly lowered IOP (61%) at 3 h post-dose, with the effect lasting up to 72 h. This class of molecule has high potential to be useful for treatment of glaucoma.

4.
Proc Natl Acad Sci U S A ; 118(16)2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33853948

RESUMEN

Primary Open Angle Glaucoma (POAG) is the most common form of glaucoma that leads to irreversible vision loss. Dysfunction of trabecular meshwork (TM) tissue, a major regulator of aqueous humor (AH) outflow resistance, is associated with intraocular pressure (IOP) elevation in POAG. However, the underlying pathological mechanisms of TM dysfunction in POAG remain elusive. In this regard, transient receptor potential vanilloid 4 (TRPV4) cation channels are known to be important Ca2+ entry pathways in multiple cell types. Here, we provide direct evidence supporting Ca2+ entry through TRPV4 channels in human TM cells and show that TRPV4 channels in TM cells can be activated by increased fluid flow/shear stress. TM-specific TRPV4 channel knockout in mice elevated IOP, supporting a crucial role for TRPV4 channels in IOP regulation. Pharmacological activation of TRPV4 channels in mouse eyes also improved AH outflow facility and lowered IOP. Importantly, TRPV4 channels activated endothelial nitric oxide synthase (eNOS) in TM cells, and loss of eNOS abrogated TRPV4-induced lowering of IOP. Remarkably, TRPV4-eNOS signaling was significantly more pronounced in TM cells compared to Schlemm's canal cells. Furthermore, glaucomatous human TM cells show impaired activity of TRPV4 channels and disrupted TRPV4-eNOS signaling. Flow/shear stress activation of TRPV4 channels and subsequent NO release were also impaired in glaucomatous primary human TM cells. Together, our studies demonstrate a central role for TRPV4-eNOS signaling in IOP regulation. Our results also provide evidence that impaired TRPV4 channel activity in TM cells contributes to TM dysfunction and elevated IOP in glaucoma.


Asunto(s)
Glaucoma de Ángulo Abierto/fisiopatología , Canales Catiónicos TRPV/metabolismo , Animales , Humor Acuoso/fisiología , Canales de Calcio/metabolismo , Femenino , Glaucoma/metabolismo , Glaucoma/fisiopatología , Glaucoma de Ángulo Abierto/metabolismo , Humanos , Presión Intraocular/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo III/metabolismo , Esclerótica/metabolismo , Transducción de Señal/fisiología , Canales Catiónicos TRPV/fisiología , Malla Trabecular/fisiología
5.
Mol Vis ; 27: 37-49, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33633438

RESUMEN

Purpose: Glaucoma is a neurodegenerative disease of the eye with an estimated prevalence of more than 111.8 million patients worldwide by 2040, with at least 6 to 8 million projected to become bilaterally blind. Clinically, the current method of slowing glaucomatous vision loss is to reduce intraocular pressure (IOP). In this manuscript, we describe the in vitro cytoprotective and in vivo long lasting IOP-lowering activity of the poly D, L-lactic-co-glycolic acid (PLGA) nanoparticle-encapsulated hybrid compound SA-2, possessing nitric oxide (NO) donating and superoxide radical scavenging functionalities. Methods: Previously characterized primary human trabecular meshwork (hTM) cells were used for the study. hTM cells were treated with SA-2 (100 µM, 200 µM, and 1,000 µM), SA-2 PLGA-loaded nanosuspension (SA-2 NPs, 0.1%), or vehicle for 30 min. Cyclic guanosine monophosphate (cGMP) and super oxide dismutase (SOD) levels were analyzed using commercial kits. In another experiment, hTM cells were pretreated with tert-butyl hydrogen peroxide (TBHP, 300 µM) for 30 min followed by treatment with escalating doses of SA-2 for 24 h, and CellTiter 96 cell proliferation assay was performed. For the biodistribution study, the cornea, aqueous humor, vitreous humor, retina, choroid, and sclera were collected after 1 h of administration of a single eye drop (30 µl) of SA-2 NPs (1% w/v) formulated in PBS to rat (n = 6) eyes. Compound SA-2 was quantified using high performance liquid chromatography /mass spectrometry (HPLC/MS). For the IOP-lowering activity study, a single SA-2 NPs (1%) eye drop was instilled in normotensive rats eyes and in the IOP-elevated rat eyes (n = 3/group, in the Morrison model of glaucoma), or Ad5TGFß2-induced ocular hypertensive (OHT) mouse eyes (n = 5/group). IOP was measured at various time points up to 72 h, and the experiment was repeated in triplicate. Mouse aqueous humor outflow facility was determined with multiple flow-rate infusion and episcleral venous pressure estimated with manometry. Results: SA-2 upregulated cGMP levels (six- to ten-fold) with an half maximal effective concentration (EC50) of 20.3 µM in the hTM cells and simultaneously upregulated (40-fold) the SOD enzyme when compared with the vehicle-treated hTM cells. SA-2 also protected hTM cells from TBHP-induced decrease in cell survival with an EC50 of 0.38 µM. A single dose of slow-release SA-2 NPs (1% w/v) delivered as an eye drop significantly lowered IOP (by 30%) in normotensive and OHT rodent eyes after 3 h post-dose, with the effect lasting up to 72 h. A statistically significant increase in aqueous outflow facility and a decrease in episcleral venous pressure was observed in rodents at this dose at 54 h. Conclusions: Hybrid compound SA-2 upregulated cGMP in hTM cells, increased outflow facility and decreased IOP in rodent models of OHT. Compound SA-2 possessing an antioxidant moiety provided additive cytoprotective activity to oxidatively stressed hTM cells by scavenging reactive oxygen species (ROS) and increasing SOD enzyme activity. Additionally, the PLGA nanosuspension formulation (SA-2 NPs) provided longer duration of IOP-lowering activity (up to 3 days) in comparison with the free non-encapsulated SA-2 drug. The data have implications for developing novel, non-prostaglandin therapeutics for IOP-lowering and cytoprotective effects with the possibility of an eye drop dosing regimen of once every 3 days for patients with glaucoma.


Asunto(s)
Antihipertensivos/uso terapéutico , Modelos Animales de Enfermedad , Presión Intraocular/efectos de los fármacos , Hipertensión Ocular/tratamiento farmacológico , Piperidinas/uso terapéutico , Malla Trabecular/efectos de los fármacos , Administración Oftálmica , Adulto , Anciano de 80 o más Años , Animales , Antihipertensivos/farmacocinética , Antihipertensivos/farmacología , Humor Acuoso/fisiología , Disponibilidad Biológica , Células Cultivadas , GMP Cíclico/metabolismo , Portadores de Fármacos , Femenino , Depuradores de Radicales Libres/farmacocinética , Depuradores de Radicales Libres/farmacología , Depuradores de Radicales Libres/uso terapéutico , Glicolatos/química , Humanos , Masculino , Ratones Endogámicos C57BL , Donantes de Óxido Nítrico/farmacocinética , Donantes de Óxido Nítrico/farmacología , Donantes de Óxido Nítrico/uso terapéutico , Hipertensión Ocular/metabolismo , Soluciones Oftálmicas , Piperidinas/farmacocinética , Piperidinas/farmacología , Ratas , Ratas Endogámicas BN , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Esclerótica/irrigación sanguínea , Superóxido Dismutasa/metabolismo , Distribución Tisular , Malla Trabecular/metabolismo , Presión Venosa/fisiología
6.
Invest Ophthalmol Vis Sci ; 60(8): 3064-3073, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31348824

RESUMEN

Purpose: Determine the toxicity, bioavailability in the retina, and neuroprotective effects of a hybrid antioxidant-nitric oxide donor compound SA-2 against oxidative stress-induced retinal ganglion cell (RGC) death in neurodegenerative animal models. Methods: Optic nerve crush (ONC) and ischemia reperfusion (I/R) injury models were used in 12-week-old C57BL/6J mice to mimic conditions of glaucomatous neurodegeneration. Mice were treated intravitreally with either vehicle or SA-2. Retinal thickness was measured by spectral-domain optical coherence tomography (SD-OCT). The electroretinogram and pattern ERG (PERG) were used to assess retinal function. RGC survival was determined by counting RBPMS-positive RGCs and immunohistochemical analysis of superoxide dismutase 1 (SOD1) levels was carried out in the retina sections. Concentrations of SA-2 in the retina and choroid were determined using HPLC and MS. In addition, the direct effect of SA-2 treatment on RGC survival was assessed in ex vivo rat retinal explants under hypoxic (0.5% O2) conditions. Results: Compound SA-2 did not induce any appreciable change in retinal thickness, or in a- or b-wave amplitude in naive animals. SA-2 was found to be bioavailable in both the retina and choroid after a single intravitreal injection (2% wt/vol). An increase in SOD1 levels in the retina of mice subjected to ONC and SA-2 treatment, suggests an enhancement in antioxidant activity. SA-2 provided significant (P < 0.05) RGC protection in all three of the tested RGC injury models in rodents. PERG amplitudes were significantly higher in both I/R and ONC mouse eyes following SA-2 treatment (P ≤ 0.001) in comparison with the vehicle and control groups. Conclusions: Compound SA-2 was effective in preventing RGC death and loss of function in three different rodent models of acute RGC injury: ONC, I/R, and hypoxia.


Asunto(s)
Neuroprotección/efectos de los fármacos , Donantes de Óxido Nítrico/farmacocinética , Estrés Oxidativo , Degeneración Retiniana/tratamiento farmacológico , Células Ganglionares de la Retina/patología , Animales , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Electrorretinografía , Femenino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Células Ganglionares de la Retina/metabolismo , Tomografía de Coherencia Óptica
7.
J Ocul Pharmacol Ther ; 34(1-2): 85-106, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28820649

RESUMEN

Progressive neurodegeneration of the optic nerve and the loss of retinal ganglion cells is a hallmark of glaucoma, the leading cause of irreversible blindness worldwide, with primary open-angle glaucoma (POAG) being the most frequent form of glaucoma in the Western world. While some genetic mutations have been identified for some glaucomas, those associated with POAG are limited and for most POAG patients, the etiology is still unclear. Unfortunately, treatment of this neurodegenerative disease and other retinal degenerative diseases is lacking. For POAG, most of the treatments focus on reducing aqueous humor formation, enhancing uveoscleral or conventional outflow, or lowering intraocular pressure through surgical means. These efforts, in some cases, do not always lead to a prevention of vision loss and therefore other strategies are needed to reduce or reverse the progressive neurodegeneration. In this review, we will highlight some of the ocular pharmacological approaches that are being tested to reduce neurodegeneration and provide some form of neuroprotection.


Asunto(s)
Glaucoma/tratamiento farmacológico , Enfermedades Neurodegenerativas/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Soluciones Oftálmicas/farmacología , Animales , Glaucoma/cirugía , Humanos , Presión Intraocular/efectos de los fármacos , Enfermedades Neurodegenerativas/cirugía
8.
Invest Ophthalmol Vis Sci ; 58(5): 2755-2764, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28549090

RESUMEN

Purpose: Understanding the role of mitochondria in retinal ganglion cells (RGCs) is relevant to human disease as studies have shown mitochondrial abnormalities in primary open-angle glaucoma patients. This study seeks to determine the effects of the sigma-1 receptor (σ-1r) and its agonists on mitochondrial function in oxygen- and glucose- deprived (OGD) purified neonatal RGCs. Methods: Retinal ganglion cells were isolated from rat pups and subjected to OGD in varying conditions in the presence or absence of σ-1r agonist and antagonist and following addition of an AAV2-σ-1r vector that was used to increase σ-1r expression. Western blots and immunofluorescence microscopy validated findings. Mitochondrial function was determined by measuring mitochondrial membrane potential (Δψm) using the dye, fluorescence tetraethylbenzimidazolylcarbocyanineiodide (JC-1), and determination of cytochrome c oxidase activity using a cytochrome c oxidase assay kit. Caspase 3 and 7 activities were also measured using a luminescent assay kit. Results: Oxygen and glucose deprivation in RGCs resulted in decreased mitochondrial membrane potential and cytochrome c oxidase activity when compared with normoxic RGCs. σ-1r agonists or overexpression of the σ-1r restored the mitochondrial membrane potential comparable to normoxic conditions, while σ-1r antagonists abolished these effects. Oxygen and glucose depreavtation induced decreases in cytochrome c activity were partially restored by overexpression or activation of σ-1r. Caspase activity was increased in response to OGD and was decreased by the addition of σ-1r agonist, pentazocine, and following σ-1r overexpression. Conclusions: These data suggest that activation and/or overexpression of σ-1r restores RGCs mitochondrial function following OGD and that mitochondrial function is vital to the function of RGCs.


Asunto(s)
Glucosa/metabolismo , Mitocondrias/fisiología , Oxígeno/metabolismo , Receptores sigma/metabolismo , Células Ganglionares de la Retina/metabolismo , Animales , Animales Recién Nacidos , Bencimidazoles/farmacología , Western Blotting , Carbocianinas/farmacología , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Hipoxia de la Célula/fisiología , Dependovirus/genética , Complejo IV de Transporte de Electrones/metabolismo , Vectores Genéticos , Potencial de la Membrana Mitocondrial/fisiología , Microscopía Fluorescente , Pentazocina/farmacología , Ratas , Ratas Sprague-Dawley , Receptores sigma/agonistas , Receptores sigma/antagonistas & inhibidores , Células Ganglionares de la Retina/efectos de los fármacos , Receptor Sigma-1
9.
Nutr Neurosci ; 20(5): 273-283, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26651837

RESUMEN

OBJECTIVES: Alzheimer's disease is a progressive neurodegenerative disease characterized by loss of hippocampal neurons leading to memory deficits and cognitive decline. Studies suggest that levels of the vasoactive peptide endothelin-1 (ET-1) are increased in the brain tissue of Alzheimer's patients. Curcumin, the main ingredient of the spice turmeric, has been shown to have anti-inflammatory, anti-cancer, and neuroprotective effects. However, the mechanisms underlying some of these beneficial effects are not completely understood. The objective of this study was to determine if curcumin could protect hippocampal neurons from ET-1 mediated cell death and examine the involvement of c-Jun in this pathway. METHODS: Primary hippocampal neurons from rat pups were isolated using a previously published protocol. Viability of the cells was measured by the live/dead assay. Immunoblot and immunohistochemical analyses were performed to analyze c-Jun levels in hippocampal neurons treated with either ET-1 or a combination of ET-1 and curcumin. Apoptotic changes were evaluated by immunoblot detection of cleaved caspase-3, cleaved fodrin, and a caspase 3/7 activation assay. RESULTS: ET-1 treatment produced a 2-fold increase in the levels of c-Jun as determined by an immunoblot analysis in hippocampal neurons. Co-treatment with curcumin significantly attenuated the ET-1 mediated increase in c-Jun levels. ET-1 caused increased neuronal cell death of hippocampal neurons indicated by elevation of cleaved caspase-3, cleaved fodrin and an increased activity of caspases 3 and 7 which was attenuated by co-treatment with curcumin. Blockade of JNK, an upstream effector of c-Jun by specific inhibitor SP600125 did not fully protect from ET-1 mediated activation of pro-apoptotic enzymes in primary hippocampal cells. DISCUSSION: Our data suggests that one mechanism by which curcumin protects against ET-1-mediated cell death is through blocking an increase in c-Jun levels. Other possible mechanisms include decreasing pro-apoptotic signaling activated by ET-1 in primary hippocampal neurons.


Asunto(s)
Muerte Celular/efectos de los fármacos , Curcumina/farmacología , Endotelina-1/farmacología , Hipocampo/citología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores , Enfermedad de Alzheimer , Animales , Apoptosis/efectos de los fármacos , Proteínas Portadoras/análisis , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Células Cultivadas , Hipocampo/química , Proteínas de Microfilamentos/análisis , Neuronas/química , Proteínas Proto-Oncogénicas c-jun/análisis , Proteínas Proto-Oncogénicas c-jun/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley , Transducción de Señal
10.
Int J Med Sci ; 12(5): 432-40, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26078703

RESUMEN

Intermittent hypoxia preconditioning (IHP) has been shown to protect neurons against ischemic stroke injury. Studying how proteins respond to IHP may identify targets that can help fight stroke. The objective of the present study was to investigate whether mitochondrial dihydrolipoamide dehydrogenase (DLDH) would respond to IHP and if so, whether such a response could be linked to neuroprotection in ischemic stroke injury. To do this, we subjected male rats to IHP for 20 days and measured the content and activity of DLDH as well as the three α-keto acid dehydrogenase complexes that contain DLDH. We also measured mitochondrial electron transport chain enzyme activities. Results show that DLDH content was indeed upregulated by IHP and this upregulation did not alter the activities of the three α-keto acid dehydrogenase complexes. Results also show that the activities of the five mitochondrial complexes (I-V) were not altered either by IHP. To investigate whether IHP-induced DLDH upregulation is linked to neuroprotection against ischemic stroke injury, we subjected both DLDH deficient mouse and DLDH transgenic mouse to stroke surgery followed by measurement of brain infarction volume. Results indicate that while mouse deficient in DLDH had exacerbated brain injury after stroke, mouse overexpressing human DLDH also showed increased brain injury after stroke. Therefore, the physiological significance of IHP-induced DLDH upregulation remains to be further investigated.


Asunto(s)
Isquemia Encefálica/metabolismo , Dihidrolipoamida Deshidrogenasa/metabolismo , Mitocondrias/metabolismo , Animales , Isquemia Encefálica/patología , Hipoxia de la Célula , Dihidrolipoamida Deshidrogenasa/genética , Modelos Animales de Enfermedad , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Humanos , Precondicionamiento Isquémico , Ratones Transgénicos , Ratas , Regulación hacia Arriba
11.
Exp Eye Res ; 128: 156-69, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25305575

RESUMEN

Sigma-1 receptor (σ-1) activation and mitogen-activated protein kinases (MAPKs) have been shown to protect retinal ganglion cells (RGCs) from cell death. The purpose of this study was to determine if σ-1 receptor stimulation with pentazocine could promote neuroprotection under conditions of an ischemia-like insult (oxygen glucose deprivation (OGD)) through the phosphorylation of extracellular signal regulated kinase (pERK)1/2. Primary RGCs were isolated from P3-P7 Sprague-Dawley rats and purified by sequential immunopanning using Thy1.1 antibodies. RGCs were cultured for 7 days before subjecting the cells to an OGD insult (0.5% oxygen in glucose-free medium) for 6 h. During the OGD, RGCs were treated with pentazocine (σ-1 receptor agonist) with or without BD 1047 (σ-1 receptor antagonist). In other experiments, primary RGCs were treated with pentazocine in the presence or absence of an MEK1/2 inhibitor, PD098059. Cell survival/death was assessed by staining with the calcein-AM/ethidium homodimer reagent. Levels of pERK1/2, total ERK1/2, and beta tubulin expression were determined by immunoblotting and immunofluorescence staining. RGCs subjected to OGD for 6 h induced 50% cell death in primary RGCs (p < 0.001) and inhibited pERK1/2 expression by 65% (p < 0.001). Cell death was attenuated when RGCs were treated with pentazocine under OGD (p < 0.001) and pERK1/2 expression was increased by 1.6 fold (p < 0.05) compared to OGD treated RGCs without pentazocine treatment. The co-treatment of PD098059 (MEK1/2 inhibitor) with pentazocine significantly abolished the protective effects of pentazocine on the RGCs during this OGD insult. Activation of the σ-1 receptor is a neuroprotective target that can protect RGCs from an ischemia-like insult. These results also established a direct relationship between σ-1 receptor stimulation and the neuroprotective effects of the ERK1/2 pathway in purified RGCs subjected to OGD. These findings suggest that activation of the σ-1 receptor may be a therapeutic target for neuroprotection particularly relevant to ocular neurodegenerative diseases that effect RGCs.


Asunto(s)
Analgésicos Opioides/farmacología , Isquemia/prevención & control , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Pentazocina/farmacología , Receptores sigma/metabolismo , Células Ganglionares de la Retina/efectos de los fármacos , Animales , Western Blotting , Supervivencia Celular , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Técnica del Anticuerpo Fluorescente Indirecta , Glucosa/metabolismo , Isquemia/enzimología , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Oxígeno/metabolismo , Fosforilación , Ratas , Ratas Sprague-Dawley , Células Ganglionares de la Retina/enzimología , Receptor Sigma-1
12.
Cell Physiol Biochem ; 28(6): 1145-54, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22179003

RESUMEN

Glaucoma afflicts millions of people worldwide and is a major cause of blindness. The risk to develop glaucoma is enhanced by increases in IOP, which result from deranged flow of aqueous humor. Aqueous humor is a fluid located in the front of the eye that gives the eye its buoyancy and supplies nutrients to other eye tissues. Aqueous humor is secreted by a tissue called ciliary processes and exits the eye via two tissues; the trabecular meshwork (TM) and Schlemm's canal. Because the spaces through which the fluid flows get smaller as the TM joins the area of the Schlemm's canal, there is resistance to aqueous humor outflow and this resistance creates IOP. There is a correlation between changes in TM and Schlemm's canal cell volume and rates of aqueous humor outflow; agents that decrease TM and Schlemm's canal cell volume, increase the rate of aqueous humor outflow, thus decreasing IOP. IOP is regulated by guanylate cyclase activators as shown in humans, rabbits and monkeys. There are two distinct groups of guanylate cyclases, membrane guanylate cyclase and soluble guanylate cyclase (sGC); activation of both have been shown to decrease IOP. Members of the membrane guanylate cyclase family of receptors bind to peptide ligands, while the sGC responds to gases (such as NO and CO(2)) and compounds (such as YC1, [3-(5'-hydroxymethyl-2'furyl)-1-benzyl indazole), a benzyl indazole derivative, and BAY-58-2667); activation of either results in formation of cyclic GMP (cGMP) and activation of protein kinase G (PKG) and subsequent phosphorylation of target proteins, including the high conductance calcium activated potassium channel (BKca channel). While activators of both membrane guanylate cyclase and sGC have the ability to lower IOP, the IOP lowering effects of sGC are noteworthy because sGC activators can be topically applied to the eye to achieve an effect. We have demonstrated that activators of sGC increase the rate at which aqueous humor exits the eye in a time course that correlates with the time course for sGC-induced decreases in TM and Schlemm's canal cell volume. Additionally, sGC-induced decrease in cell volume is accompanied by both K(+) and Cl(-) efflux induced by activation of K(+) and Cl(-) channels, including the BKca channel and/or K(+)Cl(-) symport. This suggests that parallel K(+)Cl(-) efflux, and resultant H(2)O efflux result in decreases in cell volume. These observations suggest a functional role for sGC activators, and suggest that the sGC/cGMP/PKG systems are potential therapeutic targets in the treatment of glaucoma.


Asunto(s)
Humor Acuoso , Guanilato Ciclasa/metabolismo , Presión Intraocular/fisiología , Animales , Humor Acuoso/citología , Humor Acuoso/enzimología , Humor Acuoso/fisiología , Tamaño de la Célula , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Glaucoma/enzimología , Glaucoma/metabolismo , Óxido Nítrico/metabolismo , Malla Trabecular/metabolismo , Malla Trabecular/fisiología
13.
Invest Ophthalmol Vis Sci ; 51(11): 5817-24, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20484594

RESUMEN

PURPOSE: There is a time-course correlation between nitric oxide (NO)-induced decreases in trabecular meshwork (TM) cell volume and NO-induced increases in outflow facility. The Schlemm's canal (SC) cells may also provide resistance to aqueous humor outflow; therefore, this study tests the involvement of the nitric oxide synthase (NOS) and NO signaling pathway and the BK(Ca)-channel in mediating SC cell volume decreases. METHODS: Cell volume was measured in low-passage human SC cells using calcein AM fluorescent dye; images were captured with a confocal microscope, and data were quantified using NIH ImageJ software. RESULTS: Inhibition of endogenous NOS resulted in a 7% increase in SC cell volume. Exposure of SC cells to DETA-NO resulted in a 12% to 16% decrease in cell volume that was abolished by the soluble guanylyl cyclase (sGC) inhibitor 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ) (5 µM), the protein kinase G (PKG) inhibitor (RP)-8-Br-PET-cGMP-S (50 µM), and the high-conductance calcium-activated potassium channel (BK(Ca) channel) inhibitor iberiotoxin (50 nM). Hypertonic media significantly decreased SC cell volume by 14%, whereas hypotonic media significantly increased cell volume by 11.2%. CONCLUSIONS: These data suggest that endogenous NOS regulates steady state cell volume and the involvement of the NOS/NO/sGC/cGMP/PKG pathway and the BK(Ca)-channel in mediating NO-induced reductions in SC cell volume. These decreases in cell volume correlated with the time-course for NO-induced increases in outflow facility, suggesting that the NO-induced reduction in SC cell volume may also influence outflow facility.


Asunto(s)
Tamaño de la Célula , Óxido Nítrico Sintasa/fisiología , Óxido Nítrico/metabolismo , Transducción de Señal/fisiología , Malla Trabecular/citología , Células Cultivadas , GMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Inhibidores Enzimáticos/farmacología , Fluoresceínas/metabolismo , Colorantes Fluorescentes/metabolismo , Guanilato Ciclasa/antagonistas & inhibidores , Guanilato Ciclasa/metabolismo , Humanos , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Concentración Osmolar , Donantes de Tejidos , Triazenos/farmacología
14.
J Ocul Pharmacol Ther ; 25(4): 309-14, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19552602

RESUMEN

PURPOSE: Inhibition of the BK(Ca) channel attenuated the nitric oxide-induced increase in outflow facility and decrease in trabecular meshwork (TM) cell volume suggesting the involvement of the BK(Ca) channel in TM cell function. This study examined the effects of activation of the BK(Ca) channel on outflow facility and TM cell volume and determined if the effects of NO and BK(Ca) channel activation on TM cell volume were additive. METHODS: Porcine eyes were used to measure outflow facility using the anterior segment organ culture perfusion system. Cell volume was measured using Calcein AM fluorescent dye, detected by confocal microscopy, and quantified using NIH ImageJ software. RESULTS: NS1619 increased outflow facility 86% over baseline. Additionally, there was a concentration-dependent decrease in TM cell volume in response to NS1619, which was abolished by iberiotoxin (IBTX). While NS1619 alone and DETA-NO alone decreased TM cell volume, together their effects were not additive. The time course for NS1619-induced increases in outflow facility correlated with the time course for NS1619-induced decreases in cell volume. CONCLUSIONS: BK(Ca) channel activation increases outflow facility and decreases cell volume suggesting that K(+) efflux regulates TM cell function.


Asunto(s)
Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , Óxido Nítrico/metabolismo , Malla Trabecular/metabolismo , Animales , Bencimidazoles/administración & dosificación , Bencimidazoles/farmacología , Línea Celular , Tamaño de la Célula , Relación Dosis-Respuesta a Droga , Fluoresceínas , Colorantes Fluorescentes , Humanos , Microscopía Confocal , Péptidos/farmacología , Porcinos , Factores de Tiempo , Malla Trabecular/efectos de los fármacos , Triazenos/farmacología
15.
Invest Ophthalmol Vis Sci ; 50(7): 3353-9, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19234350

RESUMEN

PURPOSE: There is a correlation between cell volume changes and changes in the rate of aqueous humor outflow; agents that decrease trabecular meshwork (TM) cell volume increase the rate of aqueous humor outflow. This study investigated the effects of the nitric oxide (NO)-independent activators of soluble guanylate cyclase (sGC), YC-1, and BAY-58-2667 on TM cell volume and the signal transduction pathways and ion channel involved. METHODS: Cell volume was measured with the use of calcein AM fluorescent dye, detected by confocal microscopy. Inhibitors and activators of sGC, 3',5'-cyclic guanosine monophosphate (cGMP), protein kinase G (PKG), and the BK(Ca) channel were used to characterize their involvement in the YC-1- and BAY-58-2667-induced regulation of TM cell volume. cGMP was assayed by an enzyme immunoassay. RESULTS: YC-1 (10 nM-200 microM) and BAY-58-2667 (10 nM-100 microM) each elicited a biphasic effect on TM cell volume. YC-1 (1 microM) increased TM cell volume, but higher concentrations decreased TM cell volume. Similarly, BAY-58-2667 (100 nM) increased TM cell volume, but higher concentrations decreased cell volume. The YC-1-induced cell volume decrease was mimicked by 8-Br-cGMP and abolished by the sGC inhibitor ODQ, the PKG inhibitor (RP)-8-Br-PET-cGMP-S, and the BK(Ca) channel inhibitor IBTX. The BAY-58-2667-induced cell volume decrease was mimicked by 8-Br-cGMP and was abolished by the PKG inhibitor and the BK(Ca) channel inhibitor. Unlike the YC-1 response, ODQ potentiated the BAY-58-2667-induced decreases in cell volume. CONCLUSIONS: These data suggest that the NO-independent decrease in TM cell volume is mediated by the sGC/cGMP/PKG pathway and involves K(+) efflux.


Asunto(s)
Benzoatos/farmacología , Tamaño de la Célula/efectos de los fármacos , Guanilato Ciclasa/metabolismo , Indazoles/farmacología , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Óxido Nítrico/metabolismo , Malla Trabecular/citología , Adulto , Anciano , Anciano de 80 o más Años , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , GMP Cíclico/farmacología , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Activadores de Enzimas , Inhibidores Enzimáticos/farmacología , Fluoresceínas/metabolismo , Colorantes Fluorescentes/metabolismo , Humanos , Microscopía Confocal , Oxadiazoles/farmacología , Quinoxalinas/farmacología , Malla Trabecular/metabolismo
16.
Invest Ophthalmol Vis Sci ; 50(4): 1808-13, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19074804

RESUMEN

PURPOSE: Nitric oxide (NO) increases the rate at which aqueous humor exits the eye; however, the involvement of soluble guanylate cyclase (sGC) is unknown. This study investigated the role of sGC in mediating the NO-induced increases in outflow facility. METHODS: Outflow facility was measured in porcine eyes using the anterior segment organ culture perfusion system. sGC activity was assessed by cGMP production in low-passage porcine and human trabecular meshwork (TM) cells and transformed human TM cells, as measured by enzyme immunoassay. sGCalpha and sGCbeta isoform expression were determined using Western blot analysis. RESULTS: Activation of sGC is necessary for the NO-induced increases in outflow facility (0.3215 microL/min per mm Hg [baseline outflow facility]+/-0.0837 [SEM]). NO resulted in increased sGC activity that was abolished by 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-1 (ODQ). Western blot analysis of total protein demonstrated an equivalent ratio of sGCalpha and sGCbeta subunit expression. In transformed cell fractions, however, the level of cytoplasmic sGCalpha subunit expression was decreased compared with low-passage human TM cells. CONCLUSIONS: Activation of sGC is involved in the NO-induced increases in outflow facility. The expression of alpha and beta sGC subunits in an equivalent ratio would suggest a functional sGC heterodimer because DETA-NO increased cGMP levels in low-passage human and porcine TM cells. However, the inability of DETA-NO to cause increases in cGMP levels in transformed TM cells suggests that though the sGC heterodimer is necessary, it is not sufficient and may require other factors not present in transformed cells.


Asunto(s)
Humor Acuoso/metabolismo , Guanilato Ciclasa/fisiología , Receptores Citoplasmáticos y Nucleares/fisiología , Malla Trabecular/efectos de los fármacos , Triazenos/farmacología , Adulto , Anciano de 80 o más Años , Animales , Western Blotting , Técnicas de Cultivo de Célula , GMP Cíclico/metabolismo , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Guanilato Ciclasa/antagonistas & inhibidores , Humanos , Isoenzimas/fisiología , Técnicas de Cultivo de Órganos , Oxadiazoles/farmacología , Quinoxalinas/farmacología , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Guanilil Ciclasa Soluble , Porcinos , Malla Trabecular/metabolismo
17.
Am J Physiol Cell Physiol ; 294(6): C1378-86, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18385281

RESUMEN

Nitric oxide (NO) donors decrease intraocular pressure (IOP) by increasing aqueous outflow facility in the trabecular meshwork (TM) and/or Schlemm's canal. However, the cellular mechanisms are unknown. Cellular mechanisms known to regulate outflow facility include changes in cell volume and cellular contractility. In this study, we investigated the effects of NO donors on outflow facility and NO-induced effects on TM cell volume. We tested the involvement of soluble guanylate cyclase (sGC), cGMP, PKG, and the large-conductance Ca2+-activated K+ (BKCa) channel using inhibitors and activators. Cell volume was measured using calcein AM fluorescent dye, detected by confocal microscopy, and quantified using NIH ImageJ software. An anterior segment organ perfusion system measured outflow facility. NO increased outflow facility in porcine eye anterior segments (0.4884-1.3956 microl.min(-1).mmHg(-1)) over baseline (0.2373-0.5220 microl.min(-1).mmHg(-1)) within 10 min of drug application. These NO-induced increases in outflow facility were inhibited by the the BKCa channel inhibitor IBTX. Exposure of TM cells to NO resulted in a 10% decrease in cell volume, and these decreases were abolished by the sGC inhibitor 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one and IBTX, suggesting the involvement of sGC and K+ eflux, respectively. NO-induced decreases in cell volume were mimicked by 8-Br-cGMP and abolished by the PKG inhibitor (RP)-8-Br-PET-cGMP-S, suggesting the involvement cGMP and PKG. Additionally, the time course for NO-induced decreases in TM cell volume correlated with NO-induced increases in outflow facility, suggesting that the NO-induced alterations in cell volume may influence outflow facility.


Asunto(s)
Humor Acuoso/metabolismo , Tamaño de la Célula , Presión Intraocular , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , Óxido Nítrico/metabolismo , Malla Trabecular/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Línea Celular , Tamaño de la Célula/efectos de los fármacos , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , GMP Cíclico/farmacología , Proteínas Quinasas Dependientes de GMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Fluoresceínas , Colorantes Fluorescentes , Guanilato Ciclasa/antagonistas & inhibidores , Guanilato Ciclasa/metabolismo , Humanos , Presión Intraocular/efectos de los fármacos , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/antagonistas & inhibidores , Microscopía Confocal , Persona de Mediana Edad , Donantes de Óxido Nítrico/farmacología , Concentración Osmolar , Oxadiazoles/farmacología , Péptidos/farmacología , Perfusión , Bloqueadores de los Canales de Potasio/farmacología , Quinoxalinas/farmacología , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/metabolismo , Guanilil Ciclasa Soluble , Porcinos , Tionucleótidos/farmacología , Factores de Tiempo , Técnicas de Cultivo de Tejidos , Malla Trabecular/citología , Malla Trabecular/efectos de los fármacos , Malla Trabecular/enzimología
19.
J Neurosci ; 23(1): 43-51, 2003 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-12514200

RESUMEN

Na,K-ATPase plays a critical role in energy metabolism and ion fluxes. Its loss was investigated in the G93A mouse model of amyotrophic lateral sclerosis (ALS) in which the mutation of Cu/Zn superoxide dismutase (SOD1) is thought to lead to aberrant oxidative damage. Observed losses in spinal cord Na,K-ATPase activity exceeded all expectations. All three catalytic subunit isoforms (alpha1, alpha2, alpha3) were reduced, and the global alpha subunit loss affected not just neurons, glia, and myelinated axon tracts but even ependymal and pial membranes. Decreases in Na,K-ATPase activity were greater than losses of protein, and there were losses of Na,K-ATPase alpha, but not beta, subunits. Together, these observations are consistent with selective degradation of the alpha subunit after damage. Overexpression of normal SOD1 does not cause ALS-like symptoms, but it has other known pathological effects. In transgenic mice overexpressed normal human SOD1 had a smaller but still considerable effect on Na,K-ATPase. Furthermore, the nitric oxide-mediated regulatory pathway for Na,K-ATPase inhibition was undetectable in spinal cord tissue slices from mice overexpressing either mutant or normal human SOD1. Na,K-ATPase activity did not respond to nitric oxide donors, and the free radical-dependent step of the pathway could not be bypassed by the addition of the downstream protein kinase G activator, 8-Br-cGMP. The data demonstrate that Na,K-ATPase is vulnerable to aberrant SOD1 activity, making it a potential contributing factor in disease pathology. Moreover, the global cellular distribution of Na,K-ATPase loss indicates that SOD1 overexpression is far-reaching in its pathological effects.


Asunto(s)
Esclerosis Amiotrófica Lateral/enzimología , Óxido Nítrico/fisiología , ATPasa Intercambiadora de Sodio-Potasio/análisis , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Superóxido Dismutasa/genética , Animales , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Radicales Libres/metabolismo , Humanos , Ratones , Ratones Transgénicos , Mutación Missense , Ouabaína/farmacología , Isoformas de Proteínas/análisis , Subunidades de Proteína , ATPasa Intercambiadora de Sodio-Potasio/inmunología , Médula Espinal/efectos de los fármacos , Médula Espinal/enzimología , Superóxido Dismutasa-1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...