Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Neurobiol Dis ; 173: 105857, 2022 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-36075537

RESUMEN

Huntington disease (HD) is a neurodegenerative disorder caused by polyglutamine-encoding CAG repeat expansion in the huntingtin (HTT) gene. HTT is involved in the axonal transport of vesicles containing brain-derived neurotrophic factor (BDNF). In HD, diminished BDNF transport leads to reduced BDNF delivery to the striatum, contributing to striatal and cortical neuronal death. Pridopidine is a selective and potent sigma-1 receptor (S1R) agonist currently in clinical development for HD. The S1R is located at the endoplasmic reticulum (ER)-mitochondria interface, where it regulates key cellular pathways commonly impaired in neurodegenerative diseases. We used a microfluidic device that reconstitutes the corticostriatal network, allowing the investigation of presynaptic dynamics, synaptic morphology and transmission, and postsynaptic signaling. Culturing primary neurons from the HD mouse model HdhCAG140/+ provides a "disease-on-a-chip" platform ideal for investigating pathogenic mechanisms and drug activity. Pridopidine rescued the trafficking of BDNF and TrkB resulting in an increased neurotrophin signaling at the synapse. This increased the capacity of HD neurons to release glutamate and restored homeostasis at the corticostriatal synapse. These data suggest that pridopidine enhances the availability of corticostriatal BDNF via S1R activation, leading to neuroprotective effects.


Asunto(s)
Enfermedad de Huntington , Fármacos Neuroprotectores , Animales , Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Modelos Animales de Enfermedad , Glutamatos/farmacología , Glutamatos/uso terapéutico , Homeostasis , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/genética , Dispositivos Laboratorio en un Chip , Ratones , Fármacos Neuroprotectores/farmacología , Piperidinas , Sinapsis/metabolismo
2.
Sci Rep ; 11(1): 21975, 2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34753986

RESUMEN

Optic neuropathies such as glaucoma are characterized by retinal ganglion cell (RGC) degeneration and death. The sigma-1 receptor (S1R) is an attractive target for treating optic neuropathies as it is highly expressed in RGCs, and its absence causes retinal degeneration. Activation of the S1R exerts neuroprotective effects in models of retinal degeneration. Pridopidine is a highly selective and potent S1R agonist in clinical development. We show that pridopidine exerts neuroprotection of retinal ganglion cells in two different rat models of glaucoma. Pridopidine strongly binds melanin, which is highly expressed in the retina. This feature of pridopidine has implications to its ocular distribution, bioavailability, and effective dose. Mitochondria dysfunction is a key contributor to retinal ganglion cell degeneration. Pridopidine rescues mitochondrial function via activation of the S1R, providing support for the potential mechanism driving its neuroprotective effect in retinal ganglion cells.


Asunto(s)
Glaucoma/prevención & control , Fármacos Neuroprotectores/farmacología , Piperidinas/farmacología , Receptores sigma/agonistas , Células Ganglionares de la Retina/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Glaucoma/metabolismo , Glaucoma/patología , Mitocondrias/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Piperidinas/administración & dosificación , Ratas , Especies Reactivas de Oxígeno/metabolismo , Células Ganglionares de la Retina/metabolismo , Receptor Sigma-1
3.
Neurotherapeutics ; 18(2): 1017-1038, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33797036

RESUMEN

Pridopidine is a selective Sigma-1 receptor (S1R) agonist in clinical development for Huntington disease (HD) and amyotrophic lateral sclerosis. S1R is a chaperone protein localized in mitochondria-associated endoplasmic reticulum (ER) membranes, a signaling platform that regulates Ca2+ signaling, reactive oxygen species (ROS) and mitochondrial fission. Here, we investigate the protective effects of pridopidine on various mitochondrial functions in human and mouse HD models. Pridopidine effects on mitochondrial dynamics were assessed in primary neurons from YAC128 HD mice expressing the mutant human HTT gene. We observe that pridopidine prevents the disruption of mitochondria-ER contact sites and improves the co-localization of inositol 1,4,5-trisphosphate receptor (IP3R) and its chaperone S1R with mitochondria in YAC128 neurons, leading to increased mitochondrial activity, elongation, and motility. Increased mitochondrial respiration is also observed in YAC128 neurons and in pridopidine-treated HD human neural stem cells (hNSCs). ROS levels were assessed after oxidative insult or S1R knockdown in pridopidine-treated YAC128 neurons, HD hNSCs, and human HD lymphoblasts. All HD models show increased ROS levels and deficient antioxidant response, which are efficiently rescued with pridopidine. Importantly, pridopidine treatment before H2O2-induced mitochondrial dysfunction and S1R presence are required for HD cytoprotection. YAC128 mice treated at early/pre-symptomatic age with pridopidine show significant improvement in motor coordination, indicating a delay in symptom onset. Additionally, in vivo pridopidine treatment reduces mitochondrial ROS levels by normalizing mitochondrial complex activity. In conclusion, S1R-mediated enhancement of mitochondrial function contributes to the neuroprotective effects of pridopidine, providing insight into its mechanism of action and therapeutic potential.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedad de Huntington/metabolismo , Mitocondrias/metabolismo , Piperidinas/farmacología , Receptores sigma/agonistas , Receptores sigma/metabolismo , Animales , Técnicas de Cocultivo , Femenino , Humanos , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/patología , Peróxido de Hidrógeno/toxicidad , Masculino , Ratones , Ratones Transgénicos , Mitocondrias/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Piperidinas/uso terapéutico , Embarazo , Receptor Sigma-1
4.
J Neurochem ; 158(2): 467-481, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33871049

RESUMEN

The endoplasmic reticulum (ER)-localized Sigma-1 receptor (S1R) is neuroprotective in models of neurodegenerative diseases, among them Huntington disease (HD). Recent clinical trials in HD patients and preclinical studies in cellular and mouse HD models suggest a therapeutic potential for the high-affinity S1R agonist pridopidine. However, the molecular mechanisms of the cytoprotective effect are unclear. We have previously reported strong induction of ER stress by toxic mutant huntingtin (mHtt) oligomers, which is reduced upon sequestration of these mHtt oligomers into large aggregates. Here, we show that pridopidine significantly ameliorates mHtt-induced ER stress in cellular HD models, starting at low nanomolar concentrations. Pridopidine reduced the levels of markers of the three branches of the unfolded protein response (UPR), showing the strongest effects on the PKR-like endoplasmic reticulum kinase (PERK) branch. The effect is S1R-dependent, as it is abolished in cells expressing mHtt in which the S1R was deleted using CRISPR/Cas9 technology. mHtt increased the level of the detergent-insoluble fraction of S1R, suggesting a compensatory cellular mechanism that responds to increased ER stress. Pridopidine further enhanced the levels of insoluble S1R, suggesting the stabilization of activated S1R oligomers. These S1R oligomeric species appeared in ER-localized patches, and not in the mitochondria-associated membranes nor the ER-derived quality control compartment. The colocalization of S1R with the chaperone BiP was significantly reduced by mHtt, and pridopidine restored this colocalization to normal, unstressed levels. Pridopidine increased toxic oligomeric mHtt recruitment into less toxic large sodium dodecyl sulfate-insoluble aggregates, suggesting that this in turn reduces ER stress and cytotoxicity.


Asunto(s)
Estrés del Retículo Endoplásmico/efectos de los fármacos , Proteína Huntingtina/genética , Proteína Huntingtina/toxicidad , Piperidinas/farmacología , Receptores sigma/efectos de los fármacos , Células 3T3 , Animales , Sistemas CRISPR-Cas , Chaperón BiP del Retículo Endoplásmico , Técnicas de Inactivación de Genes , Células HEK293 , Proteínas de Choque Térmico , Humanos , Ratones , Membranas Mitocondriales/efectos de los fármacos , Respuesta de Proteína Desplegada/efectos de los fármacos , Receptor Sigma-1
5.
Nucleic Acids Res ; 48(6): e32, 2020 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-31974573

RESUMEN

In neurons, the specific spatial and temporal localization of protein synthesis is of great importance for function and survival. Here, we visualized tRNA and protein synthesis events in fixed and live mouse primary cortical culture using fluorescently-labeled tRNAs. We were able to characterize the distribution and transport of tRNAs in different neuronal sub-compartments and to study their association with the ribosome. We found that tRNA mobility in neural processes is lower than in somata and corresponds to patterns of slow transport mechanisms, and that larger tRNA puncta co-localize with translational machinery components and are likely the functional fraction. Furthermore, chemical induction of long-term potentiation (LTP) in culture revealed up-regulation of mRNA translation with a similar effect in dendrites and somata, which appeared to be GluR-dependent 6 h post-activation. Importantly, measurement of protein synthesis in neurons with high resolutions offers new insights into neuronal function in health and disease states.


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia , Neuronas/metabolismo , Biosíntesis de Proteínas , ARN de Transferencia/metabolismo , Animales , Compartimento Celular , Células Cultivadas , Dendritas/metabolismo , Colorantes Fluorescentes/metabolismo , Potenciación a Largo Plazo , Masculino , Ratones Endogámicos C57BL , Neuroglía/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ribosomas/metabolismo
6.
Front Mol Neurosci ; 11: 311, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30233312

RESUMEN

Local protein synthesis in neuronal axons plays an important role in essential spatiotemporal signaling processes; however, the molecular basis for the post-transcriptional regulation controlling this process in axons is still not fully understood. Here we studied the axonal mechanisms underlying the transport and localization of microRNA (miRNA) and the RNAi machinery along the axon. We first identified miRNAs, Dicer, and Argonaute-2 (Ago2) in motor neuron (MN) axons. We then studied the localization of RNAi machinery and demonstrated that mitochondria associate with miR-124 and RNAi proteins in axons. Importantly, this co-localization occurs primarily at axonal branch points and growth cones. Moreover, using live cell imaging of a functional Cy3-tagged miR-124, we revealed that this miRNA is actively transported with acidic compartments in axons, and associates with stalled mitochondria at growth cones and axonal branch points. Finally, we observed enhanced retrograde transport of miR-124-Cy3, and a reduction in its localization to static mitochondria in MNs expressing the ALS causative gene hSOD1G93A. Taken together, our data suggest that mitochondria participate in the axonal localization and transport of RNAi machinery, and further imply that alterations in this mechanism may be associated with neurodegeneration in ALS.

7.
Sci Rep ; 7: 44500, 2017 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-28300211

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a multifactorial lethal motor neuron disease with no known treatment. Although the basic mechanism of its degenerative pathogenesis remains poorly understood, a subcellular spatial alteration in RNA metabolism is thought to play a key role. The nature of these RNAs remains elusive, and a comprehensive characterization of the axonal RNAs involved in maintaining neuronal health has yet to be described. Here, using cultured spinal cord (SC) neurons grown using a compartmented platform followed by next-generation sequencing (NGS) technology, we find that RNA expression differs between the somatic and axonal compartments of the neuron, for both mRNA and microRNA (miRNA). Further, the introduction of SOD1G93A and TDP43A315T, established ALS-related mutations, changed the subcellular expression and localization of RNAs within the neurons, showing a spatial specificity to either the soma or the axon. Altogether, we provide here the first combined inclusive profile of mRNA and miRNA expression in two ALS models at the subcellular level. These data provide an important resource for studies on the roles of local protein synthesis and axon degeneration in ALS and can serve as a possible target pool for ALS treatment.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Axones/metabolismo , Proteínas de Unión al ADN/genética , Superóxido Dismutasa-1/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Axones/patología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/genética , Humanos , Ratones , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Degeneración Nerviosa , ARN no Traducido/genética
8.
Mol Cell Proteomics ; 15(2): 506-22, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26598648

RESUMEN

Synapse disruption takes place in many neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). However, the mechanistic understanding of this process is still limited. We set out to study a possible role for dynein in synapse integrity. Cytoplasmic dynein is a multisubunit intracellular molecule responsible for diverse cellular functions, including long-distance transport of vesicles, organelles, and signaling factors toward the cell center. A less well-characterized role dynein may play is the spatial clustering and anchoring of various factors including mRNAs in distinct cellular domains such as the neuronal synapse. Here, in order to gain insight into dynein functions in synapse integrity and disruption, we performed a screen for novel dynein interactors at the synapse. Dynein immunoprecipitation from synaptic fractions of the ALS model mSOD1(G93A) and wild-type controls, followed by mass spectrometry analysis on synaptic fractions of the ALS model mSOD1(G93A) and wild-type controls, was performed. Using advanced network analysis, we identified Staufen1, an RNA-binding protein required for the transport and localization of neuronal RNAs, as a major mediator of dynein interactions via its interaction with protein phosphatase 1-beta (PP1B). Both in vitro and in vivo validation assays demonstrate the interactions of Staufen1 and PP1B with dynein, and their colocalization with synaptic markers was altered as a result of two separate ALS-linked mutations: mSOD1(G93A) and TDP43(A315T). Taken together, we suggest a model in which dynein's interaction with Staufen1 regulates mRNA localization along the axon and the synapses, and alterations in this process may correlate with synapse disruption and ALS toxicity.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Dineínas Citoplasmáticas/genética , Proteómica , Proteínas de Unión al ARN/biosíntesis , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Axones/metabolismo , Axones/patología , Dineínas Citoplasmáticas/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Mutación , Proteínas de Unión al ARN/genética , Sinapsis/genética , Sinapsis/metabolismo , Sinapsis/patología , Sinaptosomas/metabolismo , Sinaptosomas/patología
9.
Mol Cell Proteomics ; 15(2): 409-25, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26657538

RESUMEN

Neurodegenerative diseases, such as Alzheimer's diseases (AD), are becoming more prevalent as the population ages. However, the mechanisms that lead to synapse destabilization and neuron death remain elusive. The advent of proteomics has allowed for high-throughput screening methods to search for biomarkers that could lead to early diagnosis and treatment and to identify alterations in the cellular proteome that could provide insight into disease etiology and possible treatment avenues. In this review, we have concentrated mainly on the findings that are related to how and whether proteomics studies have contributed to two aspects of AD research, the development of biomarkers for clinical diagnostics, and the recognition of proteins that can help elucidate the pathways leading to AD brain pathology. As a result of these studies, several candidate cerebrospinal fluid biomarkers are now available for further validation in different AD cohorts. Studies in AD brain and AD transgenic models support the notion that oxidative damage results in the alterations of metabolic enzymes and that mitochondrial dysfunction is central to AD neuropathology.


Asunto(s)
Enfermedad de Alzheimer/genética , Degeneración Nerviosa/genética , Proteómica , Sinapsis/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Biomarcadores/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Humanos , Ratones , Ratones Transgénicos , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Neuronas/metabolismo , Neuronas/patología , Proteoma/genética , Sinapsis/patología
10.
Int Rev Cell Mol Biol ; 315: 23-71, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25708461

RESUMEN

Spatiotemporal localization of signals is a fundamental feature impacting cell survival and proper function. The cell needs to respond in an accurate manner in both space and time to both intra- and intercellular environment cues. The regulation of this comprehensive process involves the cytoskeleton and the trafficking machinery, as well as local protein synthesis and ligand-receptor mechanisms. Alterations in such mechanisms can lead to cell dysfunction and disease. Motor neurons that can extend over tens of centimeters are a classic example for the importance of such events. Changes in spatiotemporal localization mechanisms are thought to play a role in motor neuron degeneration that occurs in amyotrophic lateral sclerosis (ALS). In this review we will discuss these mechanisms and argue that possible misregulated factors can lead to motor neuron degeneration in ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Precursor de Proteína beta-Amiloide/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Transporte Axonal , Humanos , Proteínas de Unión al ARN/metabolismo , Transducción de Señal , Factores de Tiempo
11.
Hum Mol Genet ; 22(23): 4720-5, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23836781

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder of motor neurons. Although most cases of ALS are sporadic (sALS) and of unknown etiology, there are also inherited familial ALS (fALS) cases that share a phenotype similar to sALS pathological and clinical phenotype. In this study, we have identified two new potential genetic ALS biomarkers in human bone marrow mesenchymal stem cells (hMSC) obtained from sALS patients, namely the TDP-43 (TAR DNA-binding protein 43) and SLPI (secretory leukocyte protease inhibitor). Together with the previously discovered ones-CyFIP2 and RbBP9, we investigated whether these four potential ALS biomarkers may be differentially expressed in tissues obtained from mutant SOD1(G93A) transgenic mice, a model that is relevant for at least 20% of the fALS cases. Quantitative real-time PCR analysis of brain, spinal cord and muscle tissues of the mSOD1(G93A) and controls at various time points during the progression of the neurological disease showed differential expression of the four identified biomarkers in correlation with (i) the tissue type, (ii) the stage of the disease and (iii) the gender of the animals, creating thus a novel spatiotemporal molecular signature of ALS. The biomarkers detected in the fALS animal model were homologous to those that were identified in hMSC of our sALS cases. These results support the possibility of a molecular link between sALS and fALS and may indicate common pathogenetic mechanisms involved in both types of ALS. Moreover, these results may pave the path for using the mSOD1(G93A) mouse model and these biomarkers as molecular beacons to evaluate the effects of novel drugs/treatments in ALS.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Esclerosis Amiotrófica Lateral/patología , Biomarcadores/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Unión al ADN/genética , Inhibidor Secretorio de Peptidasas Leucocitarias/genética , Superóxido Dismutasa/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adulto , Esclerosis Amiotrófica Lateral/genética , Animales , Encéfalo/metabolismo , Encéfalo/patología , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Humanos , Masculino , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Músculos/metabolismo , Músculos/patología , Inhibidor Secretorio de Peptidasas Leucocitarias/metabolismo , Médula Espinal/metabolismo , Médula Espinal/patología , Superóxido Dismutasa/metabolismo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA