Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(7)2024 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-38612856

RESUMEN

PURPOSE: Resveratrol is a natural polyphenol which has a very low bioavailability but whose antioxidant, anti-inflammatory and anti-apoptotic properties may have therapeutic potential for the treatment of neurodegenerative diseases such as multiple sclerosis (MS). Previously, we reported the oral administration of resveratrol nanoparticles (RNs) elicited a neuroprotective effect in an experimental autoimmune encephalomyelitis (EAE) mouse model of MS, at significantly lower doses than unconjugated resveratrol (RSV) due to enhanced bioavailability. Furthermore, we demonstrated that the intranasal administration of a cell-derived secretome-based therapy at low concentrations leads to the selective neuroprotection of the optic nerve in EAE mice. The current study sought to assess the potential selective efficacy of lower concentrations of intranasal RNs for attenuating optic nerve damage in EAE mice. METHODS: EAE mice received either a daily intranasal vehicle, RNs or unconjugated resveratrol (RSV) for a period of thirty days beginning on the day of EAE induction. Mice were assessed daily for limb paralysis and weekly for visual function using the optokinetic response (OKR) by observers masked to treatment regimes. After sacrifice at day 30, spinal cords and optic nerves were stained to assess inflammation and demyelination, and retinas were immunostained to quantify retinal ganglion cell (RGC) survival. RESULTS: Intranasal RNs significantly increased RGC survival at half the dose previously shown to be required when given orally, reducing the risk of systemic side effects associated with prolonged use. Both intranasal RSV and RN therapies enhanced RGC survival trends, however, only the effects of intranasal RNs were significant. RGC loss was prevented even in the presence of inflammatory and demyelinating changes induced by EAE in optic nerves. CONCLUSIONS: The intranasal administration of RNs is able to reduce RGC loss independent of the inflammatory and demyelinating effects on the optic nerve and the spinal cord. The concentration of RNs needed to achieve neuroprotection is lower than previously demonstrated with oral administration, suggesting intranasal drug delivery combined with nanoparticle conjugation warrants further exploration as a potential neuroprotective strategy for the treatment of optic neuritis, alone as well as in combination with glucocorticoids.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , Nanopartículas , Animales , Ratones , Resveratrol/farmacología , Neuroprotección , Administración Intranasal , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico
2.
Gene Ther ; 28(5): 256-264, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33589779

RESUMEN

SIRT1 prevents retinal ganglion cell (RGC) loss in models of optic neuropathy following pharmacologic activation or genetic overexpression. The exact mechanism of loss is not known, prior evidence suggests this is through oxidative stress to either neighboring cells or RGC specifically. We investigated the neuroprotective potential of RGC-selective SIRT1 gene therapy in the optic nerve crush (ONC) model. We hypothesized that AAV-mediated overexpression of SIRT1 in RGCs reduces RGC loss, thereby preserving visual function. Cohorts of C57Bl/6J mice received intravitreal injection of experimental or control AAVs using either a ganglion cell promoter or a constitutive promoter and ONC was performed. Visual function was examined by optokinetic response (OKR) for 7 days following ONC. Retina and optic nerves were harvested to investigate RGC survival by immunolabeling. The AAV7m8-SNCG.SIRT1 vector showed 44% transduction efficiency for RGCs compared with 25% (P > 0.05) by AAV2-CAG.SIRT1, and AAV7m8-SNCG.SIRT1 drives expression selectively in RGCs in vivo. Animals modeling ONC demonstrated reduced visual acuity compared to controls. Intravitreal delivery of AAV7m8-SNCG.SIRT1 mediated significant preservation of the OKR and RGC survival compared to AAV7m8-SNCG.eGFP controls, an effect not seen with the AAV2 vector. RGC-selective expression of SIRT1 offers a targeted therapy for an animal model with significant ganglion cell loss. Over-expression of SIRT1 through AAV-mediated gene transduction suggests a RGC selective component of neuro-protection using the ONC model. This study expands our understanding of SIRT1 mediated neuroprotection in the context of compressive or traumatic optic neuropathy, making it a strong therapeutic candidate for testing in all optic neuropathies.


Asunto(s)
Traumatismos del Nervio Óptico , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Compresión Nerviosa , Nervio Óptico , Traumatismos del Nervio Óptico/genética , Traumatismos del Nervio Óptico/terapia , Células Ganglionares de la Retina , Sirtuina 1/genética
4.
J Neuroophthalmol ; 39(2): 191-199, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30829880

RESUMEN

INTRODUCTION: Previous studies have shown that intranasally administered ST266, a novel biological secretome of amnion-derived multipotent progenitor cells containing multiple growth factors and anti-inflammatory cytokines, attenuated visual dysfunction and prevented retinal ganglion cell (RGC) loss in experimental optic neuritis. Long-term effects and dose escalation studies examined here have not been reported previously. METHODS: Optic neuritis was induced in the multiple sclerosis model experimental autoimmune encephalomyelitis (EAE). EAE and control mice were treated once or twice daily with intranasal placebo/vehicle or ST266 beginning after onset of optic neuritis for either 15 days or continuously until sacrifice. Visual function was assessed by optokinetic responses (OKRs). RGC survival and optic nerve inflammation and demyelination were measured. RESULTS: Both once and twice daily continuous intranasal ST266 treatment from disease onset to 56 days after EAE induction significantly increased OKR scores, decreased RGC loss, and reduced optic nerve inflammation and demyelination compared with placebo (saline, nonspecific protein solution, or cell culture media)-treated EAE mice. ST266 treatment given for just 15 days after disease onset, then discontinued, only delayed OKR decreases, and had limited effects on RGC survival and optic nerve inflammation 56 days after disease induction. CONCLUSIONS: ST266 is a potential neuroprotective therapy to prevent RGC damage, and intranasal delivery warrants further study as a novel mechanism to deliver protein therapies for optic neuropathies. Results suggest that once daily ST266 treatment is sufficient to sustain maximal benefits and demonstrate that neuroprotective effects promoted by ST266 are specific to the combination of factors present in this complex biologic therapy.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Neuritis Óptica/prevención & control , Enfermedades de la Retina/prevención & control , Células Ganglionares de la Retina/efectos de los fármacos , Administración Intranasal , Amnios , Animales , Supervivencia Celular/fisiología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/fisiopatología , Femenino , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones , Ratones Endogámicos C57BL , Células Madre Multipotentes/metabolismo , Neuroprotección/efectos de los fármacos , Fármacos Neuroprotectores/metabolismo , Nistagmo Optoquinético/fisiología , Neuritis Óptica/fisiopatología , Enfermedades de la Retina/fisiopatología , Células Ganglionares de la Retina/fisiología , Agudeza Visual/fisiología
5.
Clin Immunol ; 170: 9-19, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27394164

RESUMEN

The presence of immunoglobulin oligoclonal bands in the cerebrospinal fluid of Multiple Sclerosis (MS) patients supports the hypothesis of an infectious etiology, although the antigenic targets remain elusive. Neurotropic mouse hepatitis virus (MHV) infection in mice provides a useful tool for studying mechanisms of demyelination in a virus-induced experimental model of MS. This study uses Affymetrix microarray analysis to compare differential spinal cord mRNA levels between mice infected with demyelinating and non-demyelinating strains of MHV to identify host immune genes expressed in this demyelinating disease model. The study reveals that during the acute stage of infection, both strains induce inflammatory innate immune response genes, whereas upregulation of several immunoglobulin genes during chronic stage infection is unique to infection with the demyelinating strain. Results suggest that the demyelinating strain induced an innate-immune response during acute infection that may promote switching of Ig isotype genes during chronic infection, potentially playing a role in antibody-mediated progressive demyelination even after viral clearance.


Asunto(s)
Inmunidad Adaptativa/genética , Infecciones por Coronavirus/genética , Enfermedades Desmielinizantes/genética , Regulación de la Expresión Génica/genética , Inmunidad Innata/genética , Animales , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Enfermedades Desmielinizantes/inmunología , Enfermedades Desmielinizantes/virología , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/inmunología , Interacciones Huésped-Patógeno/inmunología , Inmunohistoquímica , Ratones Endogámicos C57BL , Virus de la Hepatitis Murina/inmunología , Virus de la Hepatitis Murina/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Médula Espinal/inmunología , Médula Espinal/metabolismo , Médula Espinal/virología
6.
J Neurosci ; 33(8): 3582-7, 2013 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-23426685

RESUMEN

Dexras1, a small G-protein localized predominantly to the brain, is transcriptionally upregulated by the synthetic glucocorticoid dexamethasone. It has close homology to the Ras subfamily but differs in that Dexras1 contains an extended 7 kDa C-terminal tail. Previous studies in our laboratory showed that NMDA receptor activation, via NO and Dexras1, physiologically stimulates DMT1, the major iron importer. A membrane-permeable iron chelator substantially reduces NMDA excitotoxicity, suggesting that Dexras1-mediated iron influx plays a crucial role in NMDA/NO-mediated cell death. We here report that iron influx is elicited by nitric oxide but not by other proapoptotic stimuli, such as H2O2 or staurosporine. Deletion of Dexras1 in mice attenuates NO-mediated cell death in dissociated primary cortical neurons and retinal ganglion cells in vivo. Thus, Dexras1 appears to mediate NMDA-elicited neurotoxicity via NO and iron influx.


Asunto(s)
Corteza Cerebral/fisiología , Ácido Glutámico/toxicidad , N-Metilaspartato/toxicidad , Células Ganglionares de la Retina/fisiología , Proteínas ras/fisiología , Animales , Corteza Cerebral/efectos de los fármacos , Ácido Glutámico/fisiología , Células HEK293 , Humanos , Hierro/metabolismo , Hierro/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Unión al GTP Monoméricas/deficiencia , Proteínas de Unión al GTP Monoméricas/fisiología , N-Metilaspartato/fisiología , Óxido Nítrico/fisiología , Óxido Nítrico/toxicidad , Células PC12 , Ratas , Proteínas ras/deficiencia
7.
Transl Vis Sci Technol ; 13(9): 27, 2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-39330985

RESUMEN

Purpose: Resveratrol (RSV) is a nutraceutical compound known for its therapeutic potential in neurodegenerative and metabolic diseases. RSV promotes survival signals in retinal ganglion cells (RGCs) through activation of SIRT1, an NAD+-dependent deacetylase. RSV and SIRT1 reduce RGC loss induced by direct optic nerve injury, but effects in indirect models of traumatic optic neuropathy remain unknown and are examined in this study. Methods: An electromagnetic stereotaxic impactor device was used to impart five traumatic skull impacts with an inter-concussion interval of 48 hours to wild type (WT) and SIRT1 knock in (KI) C57BL/6J mice overexpressing the SIRT1 gene. A cohort of WT mice also received intranasal administration of RSV (16 mg/kg) throughout the experimental period. Loss of righting reflex (RR), optokinetic response (OKR) scores, and immunolabeled RGC count are determined to assess optic neuropathy in this model of traumatic brain injury (TBI). Results: TBI significantly decreases RGC survival and decreases OKR scores compared with control uninjured mice. Either RSV administration in WT mice, or SIRT1 overexpression in SIRT1 KI mice, significantly increases RGC survival and improves OKR scores. RR time increases after the first few impacts in all groups of mice subjected to TBI, demonstrating that RSV and SIRT1 overexpression are able to attenuate optic neuropathy following similar degrees of TBI. Conclusions: Intranasal RSV is effective in preserving visual function in WT mice following TBI. Constitutive overexpression of SIRT1 recapitulates the neuroprotective effect of RSV. Translational Relevance: Results support future exploration of RSV as a potential therapy for indirect traumatic optic neuropathy.


Asunto(s)
Modelos Animales de Enfermedad , Traumatismos Cerrados de la Cabeza , Ratones Endogámicos C57BL , Traumatismos del Nervio Óptico , Resveratrol , Células Ganglionares de la Retina , Sirtuina 1 , Animales , Sirtuina 1/genética , Sirtuina 1/metabolismo , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/patología , Células Ganglionares de la Retina/metabolismo , Traumatismos del Nervio Óptico/tratamiento farmacológico , Traumatismos del Nervio Óptico/genética , Traumatismos del Nervio Óptico/patología , Ratones , Resveratrol/farmacología , Resveratrol/uso terapéutico , Resveratrol/administración & dosificación , Traumatismos Cerrados de la Cabeza/genética , Traumatismos Cerrados de la Cabeza/patología , Traumatismos Cerrados de la Cabeza/tratamiento farmacológico , Masculino , Administración Intranasal , Supervivencia Celular/efectos de los fármacos , Ratones Transgénicos , Reflejo de Enderezamiento/efectos de los fármacos , Nistagmo Optoquinético/efectos de los fármacos
8.
Neurotherapeutics ; 20(4): 1138-1153, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37160530

RESUMEN

Resveratrol is a natural polyphenol which may be useful for treating neurodegenerative diseases such as multiple sclerosis (MS). To date, current immunomodulatory treatments for MS aim to reduce inflammation with limited effects on the neurodegenerative component of this disease. The purpose of the current study is to develop a novel nanoparticle formulation of resveratrol to increase its solubility, and to assess its ability to prevent optic nerve and spinal cord degeneration in an experimental autoimmune encephalomyelitis (EAE) mouse model of MS. Resveratrol nanoparticles (RNs) were made using a thin rehydration technique. EAE mice received a daily oral administration of vehicle, RNs or unconjugated resveratrol for one month. They were assessed daily for clinical signs of paralysis and weekly for their visual acuity with optokinetic responses (OKR). After one month, their spinal cords and optic nerves were stained for inflammation and demyelination and retinal ganglion cells immunostained for Brn3a. RNs were stable for three months. The administration of RNs did not have any effect on clinical manifestation of EAE and did not preserve OKR scores but reduced the intensity of the disease. It did not reduce inflammation and demyelination in the spinal cord and the optic nerve. However, RNs were able to decrease RGC loss compared to the vehicle. Results demonstrate that resveratrol is neuroprotective by reducing RGC loss. Interestingly, neuroprotective effects and decreased disease severity occurred without reduction of inflammation or demyelination, suggesting this therapy may fill an unmet need to limit the neurodegenerative component of MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , Fármacos Neuroprotectores , Neuritis Óptica , Ratones , Animales , Resveratrol , Fármacos Neuroprotectores/uso terapéutico , Solubilidad , Ratones Endogámicos C57BL , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Esclerosis Múltiple/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Modelos Animales de Enfermedad
9.
Neurotherapeutics ; 20(3): 896-907, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36941497

RESUMEN

SIRT1 prevents retinal ganglion cell (RGC) loss in several acute and subacute optic neuropathy models following pharmacologic activation or genetic overexpression. We hypothesized that adeno-associated virus (AAV)-mediated overexpression of SIRT1 in RGCs in a chronic ocular hypertension model can reduce RGC loss, thereby preserving visual function by sustained therapeutic effect. A control vector AAV-eGFP and therapeutic vector AAV-SIRT1 were constructed and optimized for transduction efficiency. A magnetic microbead mouse model of ocular hypertension was optimized to induce a time-dependent and chronic loss of visual function and RGC degeneration. Mice received intravitreal injection of control or therapeutic AAV in which a codon-optimized human SIRT1 expression is driven by a RGC selective promoter. Intraocular pressure (IOP) was measured, and visual function was examined by optokinetic response (OKR) weekly for 49 days following microbead injection. Visual function, RGC survival, and axon numbers were compared among control and therapeutic AAV-treated animals. AAV-eGFP and AAV-SIRT1 showed transduction efficiency of ~ 40%. AAV-SIRT1 maintains the transduction of SIRT1 over time and is selectively expressed in RGCs. Intravitreal injections of AAV-SIRT1 in a glaucoma model preserved visual function, increased RGC survival, and reduced axonal degeneration compared with the control construct. Over-expression of SIRT1 through AAV-mediated gene transduction indicates a RGC-selective component of neuroprotection in multiple models of acute optic nerve degeneration. Results here show a neuroprotective effect of RGC-selective gene therapy in a chronic glaucoma model characterized by sustained elevation of IOP and subsequent RGC loss. Results suggest that this strategy may be an effective therapeutic approach for treating glaucoma, and warrants evaluation for the treatment of other chronic neurodegenerative diseases.


Asunto(s)
Glaucoma , Hipertensión Ocular , Humanos , Ratones , Animales , Células Ganglionares de la Retina/metabolismo , Presión Intraocular , Sirtuina 1/genética , Sirtuina 1/metabolismo , Glaucoma/genética , Glaucoma/terapia , Hipertensión Ocular/genética , Hipertensión Ocular/terapia , Terapia Genética/métodos , Modelos Animales de Enfermedad , Axones/metabolismo
10.
J Neurosci ; 30(13): 4725-34, 2010 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-20357123

RESUMEN

Interleukin-15 (IL15) is a cytokine produced by normal brain, but the functions of the IL15 system in normal adults are not yet clear. The hypothesis that the hippocampal IL15 system is essential for memory consolidation was tested by use of IL15Ralpha knock-out mice in behavioral, biochemical, immunohistological, and electron microscopic analyses. The knock-out mice showed deficits in memory, determined by the Stone T-maze and fear conditioning. In their hippocampi, the concentration of GABA was significantly lower. There were region-specific changes of the GABA-synthesizing enzyme glutamic acid decarboxylase (GAD), with increased GAD-67-immunopositive interneurons in the stratum oriens of the CA1 region of the hippocampus, accompanied by nonsignificant reduction of GAD-67 synapses in the CA3 region. Western blotting showed an increase of GAD-65, but not GAD-67, in the hippocampal homogenate. The ultrastructure of the hippocampus remained intact in the knock-out mice. To further test the hypothesis that IL15 directly modulates GABA turnover by reuptake mechanisms, the dose-response relationship of IL15 on (3)H-GABA uptake was determined in two neuronal cell lines. The effective and nontoxic dose was further used in the synaptosomal uptake studies. IL15 decreased the uptake of (3)H-GABA in synaptosomes from the forebrain of wild-type mice. Consistent with this, IL15Ralpha knock-out mice had increased synaptosomal uptake of (3)H-GABA. Overall, the results show novel functions of a unique cytokine in normal hippocampal activity by regulating GABA transmission.


Asunto(s)
Hipocampo/fisiología , Subunidad alfa del Receptor de Interleucina-15/fisiología , Memoria/fisiología , Neurotransmisores/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Animales , Células Cultivadas , Condicionamiento Psicológico , Miedo , Glutamato Descarboxilasa/metabolismo , Hipocampo/ultraestructura , Interleucina-15/farmacología , Interleucina-15/fisiología , Subunidad alfa del Receptor de Interleucina-15/genética , Aprendizaje por Laberinto , Ratones , Ratones Noqueados , Neuronas/metabolismo , Sinapsis/metabolismo , Sinapsis/ultraestructura , Sinaptosomas/metabolismo
11.
Neurotherapeutics ; 18(1): 448-459, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33067748

RESUMEN

The human amnion has been used for decades in wound healing, particularly burns. Amnion epithelial cells (AECs) have been the focus of extensive research based on their possible pluripotent differentiation ability. A novel, cultured cell population derived from AECs, termed human amnion-derived multipotent progenitor (AMP) cells, secrete numerous cytokines and growth factors that enhance tissue regeneration and reduce inflammation. This AMP cell secretome, termed ST266, is a unique biological solution that accumulates in eyes and optic nerves following intranasal delivery, resulting in selective suppression of optic neuritis in the experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis, but not myelitis at the administered dose. We tested the hypothesis that systemic AMP cell administration could suppress both optic neuritis and myelitis in EAE. Intravenous and intraperitoneal administration of AMP cells significantly reduced ascending paralysis and attenuated visual dysfunction in EAE mice. AMP cell treatment increased retinal ganglion cell (RGC) survival and decreased optic nerve inflammation, with variable improvement in optic nerve demyelination and spinal cord inflammation and demyelination. Results show systemic AMP cell administration inhibits RGC loss and visual dysfunction similar to previously demonstrated effects of intranasally delivered ST266. Importantly, AMP cells also promote neuroprotective effects in EAE spinal cords, marked by reduced paralysis. Protective effects of systemically administered AMP cells suggest they may serve as a potential novel treatment for multiple sclerosis.


Asunto(s)
Células Madre Multipotentes/trasplante , Mielitis/terapia , Neuritis Óptica/terapia , Amnios/citología , Animales , Enfermedades Desmielinizantes/terapia , Encefalomielitis Autoinmune Experimental/terapia , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Células Ganglionares de la Retina/metabolismo , Médula Espinal/patología
12.
Transl Vis Sci Technol ; 10(1): 8, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33505775

RESUMEN

Purpose: Traumatic optic neuropathy (TON) is often caused by blunt head trauma and has no currently effective treatment. Common animal models of TON induced by surgical crush injury are plagued by variability and do not mimic typical mechanisms of TON injury. Traumatic head impact models have recently shown evidence of TON, but the degree of head impact necessary to consistently induce TON is not well characterized, and it is examined here. Methods: Traumatic skull impacts to C57BL/6J mice were induced using an electromagnetic controlled impact device. One impact performed at two depths (mild and severe), as well as three and five repetitive impacts with an interconcussion interval of 48 hours, were tested. Optokinetic responses (OKRs) and retinal ganglion cell (RGC) loss were measured. Results: Five repetitive mild impacts significantly decreased OKR scores and RGC numbers compared with control mice 10 weeks after initial impact, with maximal pathology observed by 6 weeks and partial but significant loss present by 3 weeks. One severe impact induced similar TON. Three mild impacts also induced early OKR and RGC loss, but one mild impact did not. Equivalent degrees of TON were induced bilaterally, and a significant correlation was observed between OKR scores and RGC numbers. Conclusions: Repetitive, mild closed head trauma in mice induces progressive RGC and vision loss that worsens with increasing impacts. Translational Relevance: Results detail a reproducible model of TON that provides a reliable platform for studying potential treatments over a 3- to 6-week time course.


Asunto(s)
Traumatismos Cerrados de la Cabeza , Traumatismos del Nervio Óptico , Animales , Modelos Animales de Enfermedad , Traumatismos Cerrados de la Cabeza/complicaciones , Ratones , Ratones Endogámicos C57BL , Células Ganglionares de la Retina
13.
PLoS One ; 16(1): e0243862, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33406093

RESUMEN

ST266 is the biological secretome of cultured Amnion-derived Multipotent Progenitor cells containing multiple growth factors and cytokines. While intranasally-administered ST266 improves the phenotype in experimental optic neuritis, specific ST266 components mediating these effects are not known. We compared the effects of ST266 with and without removal of large molecular weight proteins both in vitro and in the multiple sclerosis model experimental autoimmune encephalomyelitis (EAE) in C57BL/6J mice. Mice were treated daily with intranasal vehicle, ST266 or lower molecular weight fraction of ST266. Retinal ganglion cells were counted in isolated retinas, and optic nerves were assessed for inflammation and demyelination. ST266 treatment significantly improved retinal ganglion cell survival and reduced optic nerve demyelination in EAE mice. The lower molecular weight ST266 fraction significantly improved optic nerve demyelination, but only showed a trend towards improved retinal ganglion cell survival. ST266 fractions below 50kDa increased Schwann cell proliferation in vitro, but were less effective than non-fractionated ST266. Demyelination attenuation was partially associated with the lower molecular weight ST266 fraction, but removal of higher molecular weight biomolecules from ST266 diminishes its neuroprotective effects, suggesting at least some high molecular weight proteins play a role in ST266-mediated neuroprotection.


Asunto(s)
Amnios/citología , Células Madre Multipotentes/citología , Neuroprotección , Animales , Proliferación Celular , Enfermedades Desmielinizantes/complicaciones , Enfermedades Desmielinizantes/patología , Encefalomielitis Autoinmune Experimental/complicaciones , Encefalomielitis Autoinmune Experimental/patología , Femenino , Ratones Endogámicos C57BL , Peso Molecular , Glicoproteína Mielina-Oligodendrócito , Nervio Óptico/patología , Neuritis Óptica/complicaciones , Neuritis Óptica/patología , Péptidos , Células Ganglionares de la Retina/patología , Células de Schwann/patología
14.
Epilepsy Behav ; 17(4): 441-7, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20153260

RESUMEN

In the present study, the effects of Bacopa monnieri and its active component, bacoside A, on motor deficit and alterations of GABA receptor functional regulation in the cerebellum of epileptic rats were investigated. Scatchard analysis of [(3)H]GABA and [(3)H]bicuculline in the cerebellum of epileptic rats revealed a significant decrease in B(max) compared with control. Real-time polymerase chain reaction amplification of GABA(A) receptor subunits-GABA(Aalpha1), GABA(Aalpha5,) and GABA(Adelta)-was downregulated (P<0.001) in the cerebellum of epileptic rats compared with control rats. Epileptic rats exhibit deficits in radial arm and Y-maze performance. Treatment with B. monnieri and bacoside A reversed these changes to near-control levels. Our results suggest that changes in GABAergic activity, motor learning, and memory deficit are induced by the occurrence of repetitive seizures. Treatment with B. monnieri and bacoside A prevents the occurrence of seizures thereby reducing the impairment of GABAergic activity, motor learning, and memory deficit.


Asunto(s)
Síntomas Conductuales , Cerebelo/efectos de los fármacos , Epilepsia/complicaciones , Epilepsia/patología , Fitoterapia/métodos , Preparaciones de Plantas/uso terapéutico , Receptores de GABA/metabolismo , Saponinas/uso terapéutico , Triterpenos/uso terapéutico , Animales , Anticonvulsivantes/uso terapéutico , Síntomas Conductuales/tratamiento farmacológico , Síntomas Conductuales/etiología , Bicuculina/farmacocinética , Carbamazepina/uso terapéutico , Modelos Animales de Enfermedad , Epilepsia/inducido químicamente , Epilepsia/tratamiento farmacológico , Antagonistas del GABA/farmacocinética , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Pilocarpina , Unión Proteica/efectos de los fármacos , Ratas , Ratas Wistar , Tritio/farmacocinética , Ácido gamma-Aminobutírico/farmacocinética
15.
J Neurochem ; 111(3): 819-27, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19719822

RESUMEN

The blood-brain barrier is a gatekeeper and modulatory interface for the CNS. Cerebral endothelial cells are the major component of the blood-brain barrier, and they modify inflammatory signals from the circulation to the CNS by production and secretion of secondary substances. The inflammatory mediators induced by tumor necrosis factor alpha (TNF) were determined by microarray analysis of RBE4 cerebral endothelial cells, at 0, 6, 12, or 24 h after TNF treatment. Interleukin (IL)-15 and its receptors were among the most robustly up-regulated genes. This was confirmed by real-time RT-PCR and western blotting. The three subunits of the IL15 receptor complex (IL15Ralpha, IL2Rbeta, and IL2Rgamma) showed differential regulation by TNF in their time course and amplitude of increased expression. Consistent with increased expression of the specific high affinity receptor IL15Ralpha, TNF increased cellular uptake of (125)I-IL15 and enhanced the fluorescent intensity of Alexa568-IL15 in RBE4 cells. TNF treatment in mice also increased the level of expression of IL15 receptors in enriched cerebral microvessels. We conclude that the cerebral microvascular IL15 system is a novel inflammatory mediator that transduces the actions of TNF.


Asunto(s)
Corteza Cerebral/anatomía & histología , Interleucina-15/metabolismo , Microvasos/citología , Factores de Necrosis Tumoral/farmacología , Regulación hacia Arriba/efectos de los fármacos , Animales , Línea Celular Transformada , Células Endoteliales/efectos de los fármacos , Perfilación de la Expresión Génica/métodos , Interleucina-15/genética , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , ARN Mensajero/metabolismo , Ratas , Receptores de Interleucina-15/clasificación , Receptores de Interleucina-15/genética , Receptores de Interleucina-15/metabolismo , Factores de Tiempo
16.
Sci Rep ; 9(1): 11664, 2019 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-31406150

RESUMEN

Dysregulation of iron metabolism, and resultant cytotoxicity, has been implicated in the pathogenesis of multiple sclerosis (MS) and other neurodegenerative processes. Iron accumulation promotes cytotoxicity through various mechanisms including oxidative stress and glutamate toxicity, and occurs in both MS patients and in the experimental autoimmune encephalomyelitis (EAE) model of MS. Divalent Metal Transporter1, a major iron importer in cells, is stimulated by signaling of Dexras1, a small G protein member of the Ras family. Dexras1 is activated by S-nitrosylation by nitric oxide (NO) produced by either inducible nitric oxide synthase in activated microglia/macrophages or neuronal nitric oxide synthase in neurons. Here we show Dexras1 exacerbates oxidative stress-induced neurodegeneration in experimental optic neuritis, an inflammatory demyelinating optic nerve condition that occurs in MS and EAE. Dexras1 deletion, as well as treatment with the iron chelator deferiprone, preserves vision and attenuates retinal ganglion cell (RGC) and axonal loss during EAE optic neuritis. These results suggest that iron entry triggered by NO-activated Dexras1 signaling is a potential mechanism of neuronal death in experimental optic neuritis. The current data suggest modulation of Dexras1 signaling and iron chelation are potential novel treatment strategies for optic neuritis and MS, and possibly other optic neuropathies as well.


Asunto(s)
Encefalomielitis Autoinmune Experimental/complicaciones , Hierro/metabolismo , Esclerosis Múltiple/complicaciones , Neuritis Óptica/prevención & control , Proteínas ras/metabolismo , Animales , Quelantes/administración & dosificación , Deferiprona/administración & dosificación , Encefalomielitis Autoinmune Experimental/patología , Femenino , Humanos , Ratones , Ratones Noqueados , Esclerosis Múltiple/patología , Óxido Nítrico/metabolismo , Neuritis Óptica/etiología , Neuritis Óptica/patología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteínas ras/genética
17.
Artículo en Inglés | MEDLINE | ID: mdl-30234031

RESUMEN

Neurotropic strains of mouse hepatitis virus (MHV) induce acute inflammation and chronic demyelination in the spinal cord and optic nerves mediated by axonal spread following intracranial inoculation in mice, with pathologic features similar to the human demyelinating disease multiple sclerosis. Spinal cord demyelination is also induced following intranasal inoculation with neurotropic MHV strains, however much higher viral doses are required as compared to intracranial inoculation. Recently, it was shown that intranasal administration of low concentrations of proteins leads to significant, rapid accumulation of protein in the optic nerve and in the eye, with only low levels reaching spinal cord and other brain regions. Thus, we examined whether intranasal inoculation with MHV at doses equivalent to those given intracranially could induce optic neuritis-inflammation, demyelination and loss of retinal ganglion cells (RGCs) in the optic nerve with or without inducing spinal cord demyelination. Four week old male C57BL/6J mice were inoculated intracranially with the recombinant demyelinating strain RSA59, or intranasally with RSA59 or the non-demyelinating strain RSMHV2 as control. One month post-inoculation, mice inoculated intracranially with RSA59 had significant myelin loss in both spinal cord and optic nerves, with significant loss of RGCs as well, consistent with prior studies. As expected, intranasal inoculation with RSA59 failed to induce demyelination in spinal cord; however, it also did not induce optic nerve demyelination. No acute inflammation was found, and no viral antigen was detected, in the optic nerve or retina 1 day after inoculation. Results confirm the neurotropic effects of RSA59 following intracranial inoculation, and suggest that direct infection with axonal transport of virus from brain to spinal cord and optic nerve is required to induce demyelinating disease. These studies suggest that MHV does not selectively concentrate in optic nerve and retina to sufficient levels to induce demyelination following intranasal inoculation. Intracranial inoculation should continue to be considered a preferred method for studies of MHV-induced optic neuritis and central nervous system (CNS) demyelinating disease.


Asunto(s)
Modelos Animales de Enfermedad , Esclerosis Múltiple/patología , Virus de la Hepatitis Murina/crecimiento & desarrollo , Neuritis Óptica/patología , Administración Intranasal , Animales , Supervivencia Celular , Inyecciones Intraventriculares , Ratones Endogámicos C57BL , Nervio Óptico/patología , Células Ganglionares de la Retina/patología , Médula Espinal/patología
18.
Invest Ophthalmol Vis Sci ; 59(6): 2470-2477, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29847652

RESUMEN

Purpose: Intranasally delivered ST266, the biological, proteinaceous secretome of amnion-derived multipotent progenitor cells, reduces retinal ganglion cell (RGC) loss, optic nerve inflammation, and demyelination in experimental optic neuritis. This unique therapy and novel administration route delivers numerous cytokines and growth factors to the eye and optic nerve, suggesting a potential to also treat other optic neuropathies. Thus, ST266-mediated neuroprotection was examined following traumatic optic nerve injury. Methods: Optic nerve crush injury was surgically induced in C57BL/6J mice. Mice were treated daily with intranasal PBS or ST266. RGC function was assessed by optokinetic responses (OKRs), RGCs were counted, and optic nerve sections were stained with luxol fast blue and anti-neurofilament antibodies to assess myelin and RGC axon damage. Results: Intranasal ST266 administered daily for 5 days, beginning at the time that a 1-second optic nerve crush was performed, significantly attenuated OKR decreases. Furthermore, ST266 treatment reduced damage to RGC axons and myelin within optic nerves, and blocked RGC loss. Following a 4-second optic nerve crush, intranasal ST266 increased RGC survival and showed a trend toward reduced RGC axon and myelin damage. Ten days following optic nerve crush, ST266 prevented myelin damage, while also inducing a trend toward increased RGC survival and visual function. Conclusions: ST266 significantly attenuates traumatic optic neuropathy. Neuroprotective effects of this unique combination of biologic molecules observed here and previously in optic neuritis suggest potential broad application for preventing neuronal damage in multiple optic nerve disorders. Furthermore, results support intranasal delivery as a novel, noninvasive therapeutic modality for eyes and optic nerves.


Asunto(s)
Amnios/metabolismo , Citocinas/administración & dosificación , Neuroprotección/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Traumatismos del Nervio Óptico/tratamiento farmacológico , Células Ganglionares de la Retina/efectos de los fármacos , Administración Intranasal , Animales , Axones/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Vaina de Mielina/metabolismo , Compresión Nerviosa , Nistagmo Optoquinético/fisiología , Traumatismos del Nervio Óptico/fisiopatología
19.
Oxid Med Cell Longev ; 2017: 7180632, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28680531

RESUMEN

The ability of novel mitochondrial uncoupler prodrug of 2,4-dinitrophenol (DNP), MP201, to prevent neuronal damage and preserve visual function in an experimental autoimmune encephalomyelitis (EAE) model of optic neuritis was evaluated. Optic nerve inflammation, demyelination, and axonal loss are prominent features of optic neuritis, an inflammatory optic neuropathy often associated with the central nervous system demyelinating disease multiple sclerosis. Currently, optic neuritis is frequently treated with high-dose corticosteroids, but treatment fails to prevent permanent neuronal damage and associated vision changes that occur as optic neuritis resolves, thus suggesting that additional therapies are required. MP201 administered orally, once per day, attenuated visual dysfunction, preserved retinal ganglion cells (RGCs), and reduced RGC axonal loss and demyelination in the optic nerves of EAE mice, with limited effects on inflammation. The prominent mild mitochondrial uncoupling properties of MP201, with slow elimination of DNP, may contribute to the neuroprotective effect by modulating the entire mitochondria's physiology directly. Results suggest that MP201 is a potential novel treatment for optic neuritis.


Asunto(s)
2,4-Dinitrofenol/metabolismo , Mitocondrias/metabolismo , Neuritis Óptica/genética , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Neuritis Óptica/metabolismo , Profármacos
20.
Sci Rep ; 7: 41768, 2017 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-28139754

RESUMEN

The ability of a novel intranasally delivered amnion cell derived biologic to suppress inflammation, prevent neuronal damage and preserve neurologic function in the experimental autoimmune encephalomyelitis animal model of multiple sclerosis was assessed. Currently, there are no existing optic nerve treatment methods for disease or trauma that result in permanent vision loss. Demyelinating optic nerve inflammation, termed optic neuritis, induces permanent visual dysfunction due to retinal ganglion cell damage in multiple sclerosis and experimental autoimmune encephalomyelitis. ST266, the biological secretome of Amnion-derived Multipotent Progenitor cells, contains multiple anti-inflammatory cytokines and growth factors. Intranasally administered ST266 accumulated in rodent eyes and optic nerves, attenuated visual dysfunction, and prevented retinal ganglion cell loss in experimental optic neuritis, with reduced inflammation and demyelination. Additionally, ST266 reduced retinal ganglion cell death in vitro. Neuroprotective effects involved oxidative stress reduction, SIRT1-mediated mitochondrial function promotion, and pAKT signaling. Intranasal delivery of neuroprotective ST266 is a potential novel, noninvasive therapeutic modality for the eyes, optic nerves and brain. The unique combination of biologic molecules in ST266 provides an innovative approach with broad implications for suppressing inflammation in autoimmune diseases, and for preventing neuronal damage in acute neuronal injury and chronic neurodegenerative diseases such as multiple sclerosis.


Asunto(s)
Amnios/metabolismo , Factores Biológicos/administración & dosificación , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Administración Intranasal , Animales , Axones/metabolismo , Supervivencia Celular/efectos de los fármacos , Enfermedades Desmielinizantes , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Femenino , Ratones , Neuronas/patología , Fármacos Neuroprotectores/farmacología , Nervio Óptico/metabolismo , Nervio Óptico/patología , Neuritis Óptica/etiología , Neuritis Óptica/metabolismo , Neuritis Óptica/patología , Ratas , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/metabolismo , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA