Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 8(1): 16393, 2018 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-30401824

RESUMEN

Most human neurodegenerative diseases share a phenotype of neuronal protein aggregation. In Amyotrophic Lateral Sclerosis (ALS), the abundant protein superoxide dismutase (SOD1) or the TAR-DNA binding protein TDP-43 can aggregate in motor neurons. Recently, numerous studies have highlighted the ability of aggregates to spread from neuron to neuron in a prion-like fashion. These studies have typically focused on the use of neuron-like cell lines or neurons that are not normally affected by the specific aggregated protein being studied. Here, we have investigated the uptake of pre-formed SOD1 aggregates by cultures containing pluripotent stem cell-derived human motor neurons. We found that all cells take up aggregates by a process resembling fluid-phase endocytosis, just as found in earlier studies. However, motor neurons, despite taking up smaller amounts of SOD1, were much more vulnerable to the accumulating aggregates. Thus, the propagation of disease pathology depends less on selective uptake than on selective response to intracellular aggregates. We further demonstrate that anti-SOD1 antibodies, being considered as ALS therapeutics, can act by blocking the uptake of SOD1, but also by blocking the toxic effects of intracellular SOD1. This work demonstrates the importance of using disease relevant cells even in studying phenomena such as aggregate propagation.


Asunto(s)
Muerte Celular , Neuronas Motoras/citología , Agregado de Proteínas , Superóxido Dismutasa-1/química , Superóxido Dismutasa-1/metabolismo , Línea Celular , Humanos , Neuronas Motoras/metabolismo , Transporte de Proteínas
2.
Stem Cell Reports ; 6(6): 993-1008, 2016 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-27304920

RESUMEN

Human pluripotent stem cells (hPSCs) offer a renewable source of cells that can be expanded indefinitely and differentiated into virtually any type of cell in the human body, including neurons. This opens up unprecedented possibilities to study neuronal cell and developmental biology and cellular pathology of the nervous system, provides a platform for the screening of chemical libraries that affect these processes, and offers a potential source of transplantable cells for regenerative approaches to neurological disease. However, defining protocols that permit a large number and high yield of neurons has proved difficult. We present differentiation protocols for the generation of distinct subtypes of neurons in a highly reproducible manner, with minimal experiment-to-experiment variation. These neurons form synapses with neighboring cells, exhibit spontaneous electrical activity, and respond appropriately to depolarization. hPSC-derived neurons exhibit a high degree of maturation and survive in culture for up to 4-5 months, even without astrocyte feeder layers.


Asunto(s)
Técnicas de Cultivo de Célula , Red Nerviosa/citología , Neurogénesis/efectos de los fármacos , Neuronas/efectos de los fármacos , Células Madre Pluripotentes/efectos de los fármacos , Biomarcadores/metabolismo , Factor Neurotrófico Derivado del Encéfalo/farmacología , Diferenciación Celular/efectos de los fármacos , Factor Neurotrófico Ciliar/farmacología , Factor Neurotrófico Derivado de la Línea Celular Glial/farmacología , Humanos , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Red Nerviosa/fisiología , Neurogénesis/genética , Neuronas/clasificación , Neuronas/citología , Neuronas/metabolismo , Variaciones Dependientes del Observador , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Reproducibilidad de los Resultados , Proteínas Smad/antagonistas & inhibidores , Proteínas Smad/genética , Proteínas Smad/metabolismo , Esferoides Celulares/citología , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
3.
J Mol Neurosci ; 58(1): 46-58, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26691332

RESUMEN

The 150-year-long search for treatments of amyotrophic lateral sclerosis (ALS) is still fueled by frustration over the shortcomings of available therapeutics. Contributing to the therapeutic limitations might be the targeting of a single aspect of this multifactorial-multisystemic disease. In an attempt to overcome this, we devised a novel multifactorial-cocktail treatment, using lentiviruses encoding: EAAT2, GDH2, and NRF2, that act synergistically to address the band and width of the effected excito-oxidative axis, reducing extracellular-glutamate and glutamate availability while improving the metabolic state and the anti-oxidant response. This strategy yielded particularly impressive results, as all three genes together but not separately prolonged survival in ALS mice by an average of 19-22 days. This was accompanied by improvement in every parameter evaluated, including body-weight loss, reflex score, neurologic score, and motor performance. We hope to provide a novel strategy to slow down disease progression and alleviate symptoms of patients suffering from ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/terapia , Terapia Genética , Proteínas de Transporte de Glutamato en la Membrana Plasmática/genética , Ácido Glutámico/metabolismo , Estrés Oxidativo , Superóxido Dismutasa/genética , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Células Cultivadas , Proteínas de Transporte de Glutamato en la Membrana Plasmática/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Mutación Missense , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Deshidrogenasas del Alcohol de Azúcar/genética , Deshidrogenasas del Alcohol de Azúcar/metabolismo , Superóxido Dismutasa-1
4.
Glia ; 61(3): 312-26, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23280929

RESUMEN

Amyotrophic Lateral Sclerosis (ALS) is a fatal, rapidly progressive, neurodegenerative disease caused by motor neuron degeneration. Despite extensive efforts, the underlying cause of ALS and the path of neurodegeneration remain elusive. Astrocyte activation occurs in response to central nervous system (CNS) insult and is considered a double edged sword in many pathological conditions. We propose that reduced glutamatergic and trophic response of astrocytes to activation may, over time, lead to accumulative CNS damage, thus facilitating neurodegeneration. We found that astrocytes derived from the SOD1(G93A) ALS mouse model exhibit a reduced glutamatergic and trophic response to specific activations compared to their wild-type counterparts. Wild-type astrocytes exhibited a robust response when activated with lipopolysaccharide (LPS), G5 or treated with ceftriaxone in many parameters evaluated. These parameters include increased expression of GLT-1 and GLAST the two major astrocytic glutamate transporters, accompanied by a marked increase in the astrocytic glutamate clearance and up-regulation of neurtrophic factor expression. However, not only do un-treated SOD1(G93A) astrocytes take up glutamate less efficiently, but in response to activation they show no further increase in any of the glutamatergic parameters evaluated. Furthermore, activation of wild-type astrocytes, but not SOD1(G93A) astrocytes, improved their ability to protect the motor neuron cell line NSC-34 from glutamate induced excitotoxicity. Our data indicates that altered astrocyte activation may well be pivotal to the pathogenesis of ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Astrocitos/metabolismo , Corteza Cerebral/metabolismo , Superóxido Dismutasa/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/patología , Ceftriaxona/farmacología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Lipopolisacáridos/farmacología , Ratones , Ratones Transgénicos , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1 , Regulación hacia Arriba/efectos de los fármacos
5.
J Mol Neurosci ; 48(1): 176-84, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22638856

RESUMEN

Stem cell-based regenerative medicine raises great hope for the treatment of multiple sclerosis (MS). Bone marrow-derived mesenchymal stem cells (BM-MSCs) are being tested in clinical trials. Bone marrow is the traditional source of human MSCs, but human term placenta appears to be an excellent alternative because of its availability, without ethical issues. In this study, the therapeutic effect of human placental MSCs (PL-MSCs) was evaluated in experimental autoimmune encephalomyelitis (EAE), the mice model of MS. EAE mice were transplanted intra-cerebrally with PL-MSCs or with the vehicle saline 5 or 10 days after first MOG injection. The mice were monitored for a month after therapy. A daily EAE score revealed a decrease in disease severity in the transplanted animals when compared to saline. Survival was significantly higher in the transplanted animals. In vitro experiments demonstrated that conditioned media from LPS-activated astrocytes stimulated PL-MSCs to express the gene TNF-α-stimulated gene/protein 6 (TSG-6). The same mRNA expression was obtained when PL-MSCs were exposed to TNF-α or IL1-ß. These results demonstrate that PL-MSCs have a therapeutic effect in the EAE mice model. We assume that this effect is caused by reduction of the anti-inflammatory protein, TSG-6, of the inflammatory damage.


Asunto(s)
Encefalomielitis Autoinmune Experimental/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Esclerosis Múltiple/terapia , Placenta/citología , Animales , Animales Recién Nacidos , Astrocitos/citología , Encéfalo/citología , Ventrículos Cerebrales/citología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/patología , Femenino , Humanos , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Esclerosis Múltiple/patología , Embarazo , Cultivo Primario de Células , Índice de Severidad de la Enfermedad , Médula Espinal/citología , Bazo/citología , Trasplante Heterólogo
6.
EPMA J ; 1(2): 343-61, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23199069

RESUMEN

Treatment of amyotrophic lateral sclerosis (ALS) has been fueled, in part, by frustration over the shortcomings of the symptomatic drugs available, since these do not impede the progression of this disease. Currently, over 150 different potential therapeutic agents or strategies have been tested in preclinical models of ALS. Unfortunately, therapeutic modifiers of murine ALS have failed to be successfully translated into strategies for patients, probably because of differences in pharmacokinetics of the therapeutic agents, route of delivery, inefficiency of the agents to affect the distinct pathologies of the disease or inherent limitations of the available animal models. Given the multiplicity of the pathological mechanisms implicated in ALS, new therapies should consider the simultaneous manipulation of multiple targets. Additionally, a better management of ALS therapy should include understanding the interactions between potential risk factors, biomarkers and heterogeneous clinical features of the patients, aiming to manage their adverse events or personalize the safety profile of these agents. This review will discuss novel pharmacological approaches concerning adjusted therapy for ALS patients: iron-binding brain permeable multimodal compounds, genetic manipulation and cell-based treatment.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...