Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
2.
Mol Neurobiol ; 58(11): 5857-5875, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34415487

RESUMEN

Complement component 3 (C3) expression is increased in the cerebellum of aging mice that demonstrate locomotor impairments and increased excitatory synapse density. However, C3 regulation of locomotion, as well as C3 roles in excitatory synapse function, remains poorly understood. Here, we demonstrate that constitutive loss of C3 function in mice evokes a locomotor phenotype characterized by decreased speed, increased active state locomotor probability, and gait ataxia. C3 loss does not alter metabolism or body mass composition. No evidence of significant muscle weakness or degenerative arthritis was found in C3 knockout mice to explain decreased gait speeds. In an enriched primary cerebellar granule cell culture model, loss of C3 protein results in increased excitatory synaptic density and increased response to KCl depolarization. Our analysis of excitatory synaptic density in the cerebellar internal granule cell and molecular layers did not demonstrate increased synaptic density in vivo, suggesting the presence of compensatory mechanisms regulating synaptic development. Functional deficits in C3 knockout mice are therefore more likely to result from altered synaptic function and/or connectivity than gross synaptic deficits. Our data demonstrate a novel role for complement proteins in cerebellar regulation of locomotor output and control.


Asunto(s)
Cerebelo/patología , Complemento C3/deficiencia , Ataxia de la Marcha/etiología , Proteínas del Tejido Nervioso/biosíntesis , Sinapsis/metabolismo , Animales , Apoptosis , Composición Corporal , Calcio/análisis , Calorimetría Indirecta , Células Cultivadas , Cerebelo/metabolismo , Complemento C3/fisiología , Ataxia de la Marcha/metabolismo , Regulación de la Expresión Génica , Fuerza de la Mano , Articulación de la Rodilla/diagnóstico por imagen , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Microtomografía por Rayos X
3.
J Biol Chem ; 295(32): 10988-11001, 2020 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-32554807

RESUMEN

The development of the dendritic arbor in pyramidal neurons is critical for neural circuit function. Here, we uncovered a pathway in which δ-catenin, a component of the cadherin-catenin cell adhesion complex, promotes coordination of growth among individual dendrites and engages the autophagy mechanism to sculpt the developing dendritic arbor. Using a rat primary neuron model, time-lapse imaging, immunohistochemistry, and confocal microscopy, we found that apical and basolateral dendrites are coordinately sculpted during development. Loss or knockdown of δ-catenin uncoupled this coordination, leading to retraction of the apical dendrite without altering basolateral dendrite dynamics. Autophagy is a key cellular pathway that allows degradation of cellular components. We observed that the impairment of the dendritic arbor resulting from δ-catenin knockdown could be reversed by knockdown of autophagy-related 7 (ATG7), a component of the autophagy machinery. We propose that δ-catenin regulates the dendritic arbor by coordinating the dynamics of individual dendrites and that the autophagy mechanism may be leveraged by δ-catenin and other effectors to sculpt the developing dendritic arbor. Our findings have implications for the management of neurological disorders, such as autism and intellectual disability, that are characterized by dendritic aberrations.


Asunto(s)
Autofagia , Cateninas/metabolismo , Células Dendríticas/metabolismo , Animales , Proteína 7 Relacionada con la Autofagia/genética , Cateninas/genética , Células Cultivadas , Técnicas de Silenciamiento del Gen , Hipocampo/citología , Hipocampo/metabolismo , Ratones , Células Piramidales/metabolismo , Ratas , Catenina delta
4.
Neurochem Res ; 44(1): 61-77, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29651654

RESUMEN

N-methyl-D-aspartate receptors (NMDARs) are widely distributed in the brain with high concentrations in the telencephalon where they modulate synaptic plasticity, working memory, and other functions. While the actions of the predominate GluN2 NMDAR subunits, GluN2A and GluN2B are relatively well understood, the function of GluN2C and GluN2D subunits in the telencephalon is largely unknown. To better understand the possible role of GluN2C subunits, we used fluorescence in situ hybridization (FISH) together with multiple cell markers to define the distribution and type of cells expressing GluN2C mRNA. Using a GluN2C-KO mouse as a negative control, GluN2C mRNA expression was only found in non-neuronal cells (NeuN-negative cells) in the hippocampus, striatum, amygdala, and cerebral cortex. For these regions, a significant fraction of GFAP-positive cells also expressed GluN2C mRNA. Overall, for the telencephalon, the globus pallidus and olfactory bulb were the only regions where GluN2C was expressed in neurons. In contrast to GluN2C, GluN2D subunit mRNA colocalized with neuronal and not astrocyte markers or GluN2C mRNA in the telencephalon (except for the globus pallidus). GluN2C mRNA did, however, colocalize with GluN2D in the thalamus where neuronal GluN2C expression is found. These findings strongly suggest that GluN2C has a very distinct function in the telencephalon compared to its role in other brain regions and compared to other GluN2-containing NMDARs. NMDARs containing GluN2C may have a specific role in regulating L-glutamate or D-serine release from astrocytes in response to L-glutamate spillover from synaptic activity.


Asunto(s)
Interneuronas/metabolismo , Neuroglía/metabolismo , ARN Mensajero/biosíntesis , Receptores de N-Metil-D-Aspartato/biosíntesis , Telencéfalo/metabolismo , Animales , Expresión Génica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Subunidades de Proteína/biosíntesis , Subunidades de Proteína/genética , ARN Mensajero/genética , Receptores de N-Metil-D-Aspartato/genética
5.
Matters (Zur) ; 20172017.
Artículo en Inglés | MEDLINE | ID: mdl-28894740

RESUMEN

Mobility and locomotor impairments have high prevalence, morbidity, and significant mortality in older adult populations. Cerebellar functional changes have been implicated in the pathogenesis of these age-related mobility and gait deficits unrelated to stroke, Parkinson's disease, or degenerative joint disease. We thus examined total cerebellar glutamate, glutamine, GABA, glycine, dopamine, norepinephrine, tryptophan, serotonin, alanine, threonine, and asparagine content from male 2-3-month (young, n = 6) and 21-24-month-old (aged, n = 6) C57BL/6 mice. Neurotransmitter and amino acid concentrations were determined by high-performance liquid chromatography followed with mass spectroscopy. We found a significant increase in cerebellar serotonin in aged versus young mice, but otherwise no significant phenotypic differences in measured neurotransmitter concentrations. Applying current thought about cerebellar aging and cerebellar serotonergic systems, we consider how this age-related increase in cerebellar serotonin may contribute to gait ataxia.

6.
Front Aging Neurosci ; 9: 219, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28769782

RESUMEN

Regulatory systems required to maintain behavioral arousal remain incompletely understood. We describe a previously unappreciated role that toll-like receptor 2 (Tlr2, a membrane bound pattern recognition receptor that recognizes specific bacterial, viral, and fungal peptides), contributes toward regulation of behavioral arousal. In 4-4.5 month old mice with constitutive loss of Tlr2 function (Tlr2-/- mice), we note a marked consolidation in the circadian pattern of both active and inactive states. Specifically, Tlr2-/- mice demonstrated significantly fewer but longer duration active states during the circadian dark cycle, and significantly fewer but longer duration inactive states during the circadian light cycle. Tlr2-/- mice also consumed less food and water, and moved less during the circadian light cycle. Analysis of circadian rhythms further suggested that Tlr2-/- mice demonstrated less day-to-day variability in feeding, drinking, and movement behaviors. Reevaluation of this same mouse cohort at age 8-8.5 months revealed a clear blunting of these differences. However, Tlr2-/- mice were still noted to have fewer short-duration active states during the circadian dark cycle, and continued to demonstrate significantly less day-to-day variability in feeding, drinking, and movement behaviors. These results suggest that Tlr2 function may have a role in promoting transitions between active and inactive states. Prior studies have demonstrated that Tlr2 regulates sickness behaviors including hypophagia, hyperthermia, and decreased activity. Our work suggests that Tlr2 function also evokes behavioral fragmentation, another aspect of sickness behavior and a clinically significant problem of older adults.

7.
Aging (Albany NY) ; 8(9): 2153-2181, 2016 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-27689748

RESUMEN

We describe age-related molecular and neuronal changes that disrupt mobility or energy balance based on brain region and genetic background. Compared to young mice, aged C57BL/6 mice exhibit marked locomotor (but not energy balance) impairments. In contrast, aged BALB mice exhibit marked energy balance (but not locomotor) impairments. Age-related changes in cerebellar or hypothalamic gene expression accompany these phenotypes. Aging evokes upregulation of immune pattern recognition receptors and cell adhesion molecules. However, these changes do not localize to microglia, the major CNS immunocyte. Consistent with a neuronal role, there is a marked age-related increase in excitatory synapses over the cerebellum and hypothalamus. Functional imaging of these regions is consistent with age-related synaptic impairments. These studies suggest that aging reactivates a developmental program employed during embryogenesis where immune molecules guide synapse formation and pruning. Renewed activity in this program may disrupt excitatory neurotransmission, causing significant behavioral deficits.


Asunto(s)
Envejecimiento/fisiología , Cerebelo/fisiología , Aminoácidos Excitadores/fisiología , Hipotálamo/fisiología , Sinapsis/fisiología , Transmisión Sináptica/fisiología , Envejecimiento/genética , Envejecimiento/inmunología , Animales , Metabolismo Energético/fisiología , Expresión Génica , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Actividad Motora/fisiología
8.
Brain ; 137(Pt 2): 503-19, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24176979

RESUMEN

The development of neuroprotective strategies to attenuate retinal ganglion cell death could lead to novel therapies for chronic optic neuropathies such as glaucoma. Intravitreal transplantation of mesenchymal stem cells slows retinal ganglion cell death in models of optic nerve injury, but the mechanism of action remains unclear. Here we characterized the neuroprotective effects of mesenchymal stem cells and mesenchymal stem cell-derived factors in organotypic retinal explant culture and an in vivo model of ocular hypertensive glaucoma. Co-culture of rat and human bone marrow-derived mesenchymal stem cells with retinal explants increased retinal ganglion cell survival, after 7 days ex vivo, by ∼2-fold and was associated with reduced apoptosis and increased nerve fibre layer and inner plexiform layer thicknesses. These effects were not demonstrated by co-culture with human or mouse fibroblasts. Conditioned media from mesenchymal stem cells conferred neuroprotection, suggesting that the neuroprotection is mediated, at least partly, by secreted factors. We compared the concentrations of 29 factors in human mesenchymal stem cell and fibroblast conditioned media, and identified 11 enriched in the mesenchymal stem cell secretome. Treatment of retinal explants with a cocktail of these factors conferred retinal ganglion cell neuroprotection, with factors from the platelet-derived growth factor family being the most potent. Blockade of platelet-derived growth factor signalling with neutralizing antibody or with small molecule inhibitors of platelet-derived growth factor receptor kinase or downstream phosphatidylinositol 3 kinase eliminated retinal ganglion cell neuroprotection conferred by mesenchymal stem cell co-culture. Intravitreal injection of platelet-derived growth factor -AA or -AB led to profound optic nerve neuroprotection in vivo following experimental induction of elevated intraocular pressure. These data demonstrate that mesenchymal stem cells secrete a number of neuroprotective proteins and suggest that platelet-derived growth factor secretion in particular may play an important role in mesenchymal stem cell-mediated retinal ganglion cell neuroprotection. Furthermore, platelet-derived growth factor may represent an independent target for achieving retinal ganglion cell neuroprotection.


Asunto(s)
Células Madre Mesenquimatosas/metabolismo , Fármacos Neuroprotectores/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Células Ganglionares de la Retina/metabolismo , Animales , Axotomía/efectos adversos , Técnicas de Cocultivo/métodos , Humanos , Células Madre Mesenquimatosas/patología , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Células Ganglionares de la Retina/patología
9.
Behav Neurosci ; 126(3): 479-87, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22642889

RESUMEN

Huntington's disease (HD) is a neurodegenerative disorder in humans caused by an expansion of a CAG trinucleotide repeat that produces choreic movements, which are preceded by cognitive deficits. The HD transgenic rat (tgHD), which contains the human HD mutation with a 51 CAG repeat allele, exhibits motor deficits that begin when these rats are 12 months of age. However, there are no reports of cognitive dysfunction occurring prior to this. To assess whether cognitive dysfunction might precede motor deficits in tgHD rats, one group of 9-month-old male rats with homozygotic mutated genes and one group of wild-type (WT) rats underwent three testing phases in a unique Spatial Operant Reversal Test (SORT) paradigm, as well as assessment of spontaneous motor activity. After testing, morphological and histological examination of the brains were made. Results indicated that tgHD rats acquired the cued-response (Phase 1) portion of the SORT, but made significantly more errors during the reversal (Phase 2) and during the pseudorandomized reversals (Phase 3) portion of the study, when compared to WT rats. Analysis of the data using mathematical principles of reinforcement revealed no memory, motor, or motivational deficits. These results indicate that early cognitive dysfunction, as measured by the SORT, occur prior to motor deficits, gross anatomical changes, or cell loss in the tgHD rat with 51 CAG repeats, and suggest that this protocol could provide a useful screen for therapeutic studies.


Asunto(s)
Trastornos del Conocimiento/genética , Enfermedad de Huntington/genética , Proteínas del Tejido Nervioso/genética , Repeticiones de Trinucleótidos/genética , Animales , Muerte Celular/genética , Trastornos del Conocimiento/psicología , Condicionamiento Operante/fisiología , Modelos Animales de Enfermedad , Humanos , Proteína Huntingtina , Enfermedad de Huntington/patología , Enfermedad de Huntington/psicología , Masculino , Distribución Aleatoria , Ratas , Ratas Transgénicas
10.
Invest Ophthalmol Vis Sci ; 52(6): 3309-20, 2011 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-21345987

RESUMEN

PURPOSE. To validate an established adult organotypic retinal explant culture system for use as an efficient medium-throughput screening tool to investigate novel retinal ganglion cell (RGC) neuroprotective therapies. METHODS. Optimal culture conditions for detecting RGC neuroprotection in rat retinal explants were identified. Retinal explants were treated with various recognized, or purported, neuroprotective agents and cultured for either 4 or 7 days ex vivo. The number of cells surviving in the RGC layer (RGCL) was quantified using histologic and immunohistochemical techniques, and statistical analyses were applied to detect neuroprotective effects. RESULTS. The ability to replicate previously reported in vivo RGC neuroprotection in retinal explants was verified by demonstrating that caspase inhibition, brain-derived neurotrophic factor treatment, and stem cell transplantation all reduced RGCL cell loss in this model. Further screening of potential neuroprotective pharmacologic agents demonstrated that betaxolol, losartan, tafluprost, and simvastatin all alleviated RGCL cell loss in retinal explants, supporting previous reports. However, treatment with brimonidine did not protect RGCL neurons from death in retinal explant cultures. Explants cultured for 4 days ex vivo proved most sensitive for detecting neuroprotection. CONCLUSIONS. The current adult rat retinal explant culture model offers advantages over other models for screening potential neuroprotective drugs, including maintenance of neurons in situ, control of environmental conditions, and dissociation from other factors such as intraocular pressure. Verification that neuroprotection by previously identified RGC-protective therapies could be replicated in adult retinal explant cultures suggests that this model could be used for efficient medium-throughput screening of novel neuroprotective therapies for retinal neurodegenerative disease.


Asunto(s)
Modelos Animales de Enfermedad , Fármacos Neuroprotectores/farmacología , Hipertensión Ocular/prevención & control , Enfermedades del Nervio Óptico/prevención & control , Células Ganglionares de la Retina/citología , Clorometilcetonas de Aminoácidos/farmacología , Animales , Apoptosis/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/farmacología , Inhibidores de Caspasas , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Colforsina/farmacología , Evaluación Preclínica de Medicamentos , Masculino , Células Madre Mesenquimatosas/citología , Compresión Nerviosa , Nervio Óptico , Técnicas de Cultivo de Órganos , Ratas , Ratas Sprague-Dawley , Células Ganglionares de la Retina/fisiología , Trasplante de Células Madre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...