Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Neurobiol Dis ; 62: 543-50, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24269732

RESUMEN

The neuronal ceroid lipofuscinoses (NCLs, Batten disease) are inherited neurodegenerative lysosomal storage diseases caused by mutations in several different genes. Mutations in CLN5 cause a variant late-infantile human disease and some cases of juvenile and adult clinical disease. NCLs also occur in animals, and a flock of New Zealand Borderdale sheep with a CLN5 splice-site mutation has been developed for model studies. Dissociated mixed neural cells from CLN5-deficient foetal sheep brains contained no obvious storage bodies at plating but these accumulated rapidly in culture, mainly in microglial cells and also in neurons and astrocytes. Accumulation was very obvious after a week, as monitored by fluorescent microscopy and immunostaining for subunit c of mitochondrial ATP synthase. Photography at intervals revealed the dynamic nature of the cultures and a flow of storage bodies between cells, specifically the phagocytosis of storage-body containing cells by microglia and incorporation of the storage bodies into the host cells. No storage was observed in cultured control cells. Transduction of cell cultures with a lentiviral vector expressing a C-terminal Myc tagged CLN5 resulted in secretion of post-translationally glycosylated and processed CLN5. Transduction of CLN5-deficient cultures with this construct rapidly reversed storage body accumulation, to less than half in only six days. These results show that storage body accumulation is reversible with enzyme correction and support the use of these cultures for testing of therapeutics prior to whole animal studies.


Asunto(s)
Proteínas de la Membrana/metabolismo , Lipofuscinosis Ceroideas Neuronales/genética , Lipofuscinosis Ceroideas Neuronales/metabolismo , Neuronas/metabolismo , Secuencia de Aminoácidos , Animales , Terapia Genética , Células HEK293 , Humanos , Lentivirus/genética , Proteínas de Membrana de los Lisosomas , Proteínas de la Membrana/genética , Datos de Secuencia Molecular , Lipofuscinosis Ceroideas Neuronales/embriología , Lipofuscinosis Ceroideas Neuronales/patología , Neuronas/patología , Ovinos
2.
Biochem Biophys Res Commun ; 444(2): 189-94, 2014 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-24440698

RESUMEN

Wnt signaling has a crucial role in synaptic function at the central nervous system. Here we evaluate whether Wnts affect nitric oxide (NO) generation in hippocampal neurons. We found that non-canonical Wnt-5a triggers NO production; however, Wnt-3a a canonical ligand did not exert the same effect. Co-administration of Wnt-5a with the soluble Frizzled related protein-2 (sFRP-2) a Wnt antagonist blocked the NO production. Wnt-5a activates the non-canonical Wnt/Ca(2+) signaling through a mechanism that depends on Ca(2+) release from Ryanodine-sensitive internal stores. The increase in NO levels evoked by Wnt-5a promotes the insertion of the GluN2B subunit of the NMDA receptor (NMDAR) into the neuronal cell surface. To the best of our knowledge, this is the first time that Wnt-5a signaling is related to NO production, which in turn increases NMDARs trafficking to the cell surface.


Asunto(s)
Neuronas/metabolismo , Óxido Nítrico/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Proteínas Wnt/metabolismo , Animales , Western Blotting , Calcio/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Células HEK293 , Hipocampo/citología , Hipocampo/embriología , Humanos , Células L , Proteínas de la Membrana/farmacología , Ratones , Modelos Biológicos , Neuronas/citología , Neuronas/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Proteínas Wnt/antagonistas & inhibidores , Proteínas Wnt/farmacología , Proteína Wnt-5a
3.
Mol Cell Neurosci ; 57: 130-43, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23932971

RESUMEN

Polycomb Repressive Complex 2 (PRC2) mediates transcriptional silencing by catalyzing histone H3 lysine 27 trimethylation (H3K27me3), but its role in the maturation of postmitotic mammalian neurons remains largely unknown. We report that the PRC2 paralogs Ezh1 and Ezh2 are differentially expressed during hippocampal development. We show that depletion of Ezh2 leads to increased expression of PSD-95, a critical plasticity gene, and that reduced PSD-95 gene transcription is correlated with enrichment of Ezh2 at the PSD-95 gene promoter; however, the H3K27me3 epigenetic mark is not present at the PSD-95 gene promoter, likely due to the antagonizing effects of the H3S28P and H3K27Ac marks and the activity of the H3K27 demethylases JMJD3 and UTX. In contrast, increased PSD-95 gene transcription is accompanied by the presence of Ezh1 and elongation-engaged RNA Polymerase II complexes at the PSD-95 gene promoter, while knock-down of Ezh1 reduces PSD-95 transcription. These results indicate that Ezh1 and Ezh2 have antagonistic roles in regulating PSD-95 transcription.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Hipocampo/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Neuronas/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Animales , Homólogo 4 de la Proteína Discs Large , Proteína Potenciadora del Homólogo Zeste 2 , Epigénesis Genética , Hipocampo/citología , Hipocampo/crecimiento & desarrollo , Histonas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Complejo Represivo Polycomb 2/genética , Regiones Promotoras Genéticas , Ratas , Ratas Sprague-Dawley , Transcripción Genética
4.
Neurobiol Dis ; 59: 69-79, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23892229

RESUMEN

Wld(S) mutation protects axons from degeneration in diverse experimental models of neurological disorders, suggesting that the mutation might act on a key step shared by different axon degeneration pathways. Here we test the hypothesis that Wld(S) protects axons by preventing energy deficiency commonly encountered in many diseases. We subjected compartmentally cultured, mouse cortical axons to energy deprivation with 6mM azide and zero glucose. In wild-type (WT) culture, the treatment, which reduced axon ATP level ([ATP]axon) by 65%, caused immediate axon depolarization followed by gradual free calcium accumulation and subsequent irreversible axon damage. The calcium accumulation resulted from calcium influx partially via L-type voltage-gated calcium channel (L-VGCC). Blocking L-VGCC with nimodipine reduced calcium accumulation and protected axons. Without altering baseline [ATP]axon, the presence of Wld(S) mutation significantly reduced the axon ATP loss and depolarization, restrained the subsequent calcium accumulation, and protected axons against energy deprivation. Wld(S) neurons possessed higher than normal nicotinamide mononucleotide adenylyltransferase (NMNAT) activity. The intrinsic Wld(S) NMNAT activity was required for the Wld(S)-mediated energy preservation and axon protection during but not prior to energy deprivation. NMNAT catalyzes the reversible reaction that produces nicotinamide adenine dinucleotide (NAD) from nicotinamide mononucleotide (NMN). Interestingly, preventing the production of NAD from NMN with FK866 increased [ATP]axon and protected axons from energy deprivation. These results indicate that the Wld(S) mutation depends on its intrinsic Wld(S) NMNAT activity and the subsequent increase in axon ATP but not NAD to protect axons, implicating a novel role of Wld(S) NMNAT in axon bioenergetics and protection.


Asunto(s)
Corteza Cerebral/patología , Metabolismo Energético/fisiología , Mutación/genética , Proteínas del Tejido Nervioso/genética , Degeneración Walleriana/genética , Degeneración Walleriana/patología , Adenosina Trifosfato/genética , Animales , Axones/patología , Axones/fisiología , Calcio/metabolismo , Modelos Animales de Enfermedad , Embrión de Mamíferos , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Inhibidores Enzimáticos/toxicidad , Glucosa/deficiencia , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Mitocondrias/fisiología , Proteínas del Tejido Nervioso/metabolismo , Técnicas de Cultivo de Órganos , Azida Sódica/toxicidad
5.
Biochem Biophys Res Commun ; 440(3): 359-63, 2013 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-23988450

RESUMEN

Neurodegeneration causes dysfunction and degeneration of neurons and is triggered by various factors including genetic defects, free radicals, injury, and glutamate excitotoxicity. Among those, glutamate excitotoxicity is implicated in chronic disorders including AD and ALS, and in acute insults in the CNS including traumatic brain injury. Neurological disorders show hallmark morphological abnormalities such as axon degeneration and cell body death. The molecular mechanisms underlying excitotoxicity-induced neurodegeneration are complex and deciphering a molecular mechanism from one angle is beneficial to understand the process, however, still difficult to develop strategies to suppress excitotoxicity-induced degeneration due to existence of other mechanisms. Thus, directly identifying compounds that can modulate excitotoxicity-induced neurodegeneration and subsequently clarifiying the molecular mechanism is a valid approach to develop effective strategies to suppress neurodegeneration. We searched for compounds that can suppress excitotoxicity-induced neurodegeneration and found that CP-31398, a known compound that can rescue the structure and function of the tumor suppressor protein p53 mutant form and stabilize the active conformation of the p53 wild-type form, suppresses excitotoxicity-induced axon degeneration and cell body death. Moreover, CP-31398 suppresses mitochondrial dysfunction which has a strong correlation with excitotoxicity. Thus, our findings identify a compound that can serve as a novel modulator of neurodegeneration induced by glutamate excitotoxicity.


Asunto(s)
Apoptosis/efectos de los fármacos , Axones/efectos de los fármacos , Antagonistas de Aminoácidos Excitadores/farmacología , Ácido Glutámico/toxicidad , Degeneración Nerviosa/prevención & control , Pirimidinas/farmacología , Animales , Axones/patología , Células Cultivadas , Hipocampo , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Degeneración Nerviosa/inducido químicamente , Oxidorreductasas/metabolismo , Ratas , Ratas Wistar
6.
Biochem Biophys Res Commun ; 438(1): 1-5, 2013 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-23747423

RESUMEN

The 42-mer amyloid ß-protein (Aß42) oligomers cause neurotoxicity and cognitive impairment in Alzheimer's disease (AD). We previously identified the toxic conformer of Aß42 with a turn at positions 22-23 ("toxic" turn) to form oligomers and to induce toxicity in rat primary neurons, along with the non-toxic conformer with a turn at positions 25-26. G25P-Aß42 and E22V-Aß42 are non-toxic mutants that disfavor the "toxic" turn. Here we hypothesize that these non-toxic mutants of Aß42 could suppress Aß42-induced neurotoxicity, and examined their effects on the neurotoxicity, aggregation, and levels of the toxic conformer, which was evaluated by dot blotting using a monoclonal antibody (11A1) against the toxic conformer. G25P-Aß42 and E22V-Aß42 suppressed the neurotoxicity and aggregation of Aß42 as well as the formation of the toxic conformer. The neurotoxicity induced by Aß42 was also significantly reduced by the treatment of 11A1, but not of Aß-sequence specific antibodies (6E10 and 4G8). Since recent studies indicate that Aß oligomers contain parallel ß-sheet, the present results suggest that the non-toxic mutants of Aß42 without the "toxic" turn could prevent the propagation process of the toxic conformer of Aß42 resulting in suppression of the formation of the toxic oligomers. This could be a promising strategy for AD therapeutics.


Asunto(s)
Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/toxicidad , Neuronas/efectos de los fármacos , Neuronas/patología , Fragmentos de Péptidos/química , Fragmentos de Péptidos/toxicidad , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Animales , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Isomerismo , Ratas , Ratas Wistar
7.
Biochem Biophys Res Commun ; 436(4): 638-44, 2013 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-23770366

RESUMEN

A serine protease, motopsin (prss12), plays a significant role in cognitive function and the development of the brain, since the loss of motopsin function causes severe mental retardation in humans and enhances social behavior in mice. Motopsin is activity-dependently secreted from neuronal cells, is captured around the synaptic cleft, and cleaves a proteoglycan, agrin. The multi-domain structure of motopsin, consisting of a signal peptide, a proline-rich domain, a kringle domain, three scavenger receptor cysteine-rich domains, and a protease domain at the C-terminal, suggests the interaction with other molecules through these domains. To identify a protein interacting with motopsin, we performed yeast two-hybrid screening and found that seizure-related gene 6 (sez-6), a transmembrane protein on the plasma membrane of neuronal cells, bound to the proline-rich/kringle domain of motopsin. Pull-down and immunoprecipitation analyses indicated the interaction between these proteins. Immunocytochemical and immunohistochemical analyses suggested the co-localization of motopsin and sez-6 at neuronal cells in the developmental mouse brain and at motor neurons in the anterior horn of human spinal cords. Transient expression of motopsin in neuro2a cells increased the number and length of neurites as well as the level of neurite branching. Interestingly, co-expression of sez-6 with motopsin restored the effect of motopsin at the basal level, while sez-6 expression alone showed no effects on cell morphology. Our results suggest that the interaction of motopsin and sez-6 modulates the neuronal cell morphology.


Asunto(s)
Discapacidad Intelectual/genética , Proteínas del Tejido Nervioso/metabolismo , Serina Endopeptidasas/genética , Animales , Células COS , Células Cultivadas , Chlorocebus aethiops , Ratones , Serina Endopeptidasas/metabolismo
8.
Cell Calcium ; 101: 102505, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34871934

RESUMEN

Zinc transporter 1 (ZnT1; SLC30A1) is present in the neuronal plasma membrane, critically modulating NMDA receptor function and Zn2+ neurotoxicity. The mechanism mediating Zn2+ transport by ZnT1, however, has remained elusive. Here, we investigated ZnT1-dependent Zn2+ transport by measuring intracellular changes of this ion using the fluorescent indicator FluoZin-3. In primary mouse cortical neurons, which express ZnT1, transient addition of extracellular Zn2+ triggered a rise in cytosolic Zn2+, followed by its removal. Knockdown of ZnT1 by adeno associated viral (AAV)-short hairpin RNA (shZnT1) markedly increased rates of Zn2+ rise, and decreased rates of its removal, suggesting that ZnT1 is a primary route for Zn2+ efflux in neurons. Although Zn2+ transport by other members of the SLC30A family is dependent on pH gradients across cellular membranes, altered H+ gradients were not coupled to ZnT1-dependent transport. Removal of cytoplasmic Zn2+, against a large inward gradient during the initial loading phase, suggests that Zn2+ efflux requires a large driving force. We therefore asked if Ca2+ gradients across the membrane can facilitate Zn2+ efflux. Elimination of extracellular Ca2+ abolished Zn2+ efflux, while increased extracellular Ca2+ levels enhanced Zn2+ efflux. Intracellular Ca2+ rises, measured in GCaMP6 expressing neurons, closely paralleled cytoplasmic Zn2+ removal. Taken together, these results strongly suggest that ZnT1 functions as a Zn2+/Ca2+ exchanger, thereby regulating the transport of two ions of fundamental importance in neuronal signaling.


Asunto(s)
Proteínas de Transporte de Catión , Animales , Transporte Biológico , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Membrana Celular/metabolismo , Ratones , Neuronas/metabolismo , Zinc/metabolismo
9.
IBRO Neurosci Rep ; 12: 411-418, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35746971

RESUMEN

The dopamine mesolimbic system is a major circuit involved in controlling goal-directed behaviors. Dopamine D2 receptors (D2R) and kappa opioid receptors (KOR) are abundant Gi protein-coupled receptors in the mesolimbic system. D2R and KOR share several functions in dopamine mesencephalic neurons, such as regulation of dopamine release and uptake, and firing of dopamine neurons. In addition, KOR and D2R modulate each other functioning. This evidence indicates that both receptors functionally interact, however, their colocalization in the mesostriatal system has not been addressed. Immunofluorescent assays were performed in cultured dopamine neurons and adult mice's brain tissue to answer this question. We observed that KOR and D2R are present in similar density in dendrites and soma of cultured dopamine neurons, but in a segregated manner. Interestingly, KOR immunolabelling was observed in the first part of the axon, colocalizing with Ankyrin in 20% of cultured dopamine neurons, indicative that KOR is present in the axon initial segment (AIS) of a group of dopaminergic neurons. In the adult brain, KOR and D2R are also segregated in striatal tissue. While the KOR label is in fiber tracts such as the striatal streaks, corpus callosum, and anterior commissure, D2R is located mainly within the striatum and nucleus accumbens, surrounding fiber tracts. D2R is also localized in some fibers that are mostly different from those positives for KOR. In conclusion, KOR and D2R are present in the soma and dendrites of mesencephalic dopaminergic neurons, but KOR is also found in the AIS of a subpopulation of these neurons.

10.
JHEP Rep ; 4(8): 100510, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35845295

RESUMEN

Background & Aims: In cirrhosis, astrocytic swelling is believed to be the principal mechanism of ammonia neurotoxicity leading to hepatic encephalopathy (HE). The role of neuronal dysfunction in HE is not clear. We aimed to explore the impact of hyperammonaemia on mitochondrial function in primary co-cultures of neurons and astrocytes and in acute brain slices of cirrhotic rats using live cell imaging. Methods: To primary cocultures of astrocytes and neurons, low concentrations (1 and 5 µM) of NH4Cl were applied. In rats with bile duct ligation (BDL)-induced cirrhosis, a model known to induce hyperammonaemia and minimal HE, acute brain slices were studied. One group of BDL rats was treated twice daily with the ammonia scavenger ornithine phenylacetate (OP; 0.3 g/kg). Fluorescence measurements of changes in mitochondrial membrane potential (Δψm), cytosolic and mitochondrial reactive oxygen species (ROS) production, lipid peroxidation (LP) rates, and cell viability were performed using confocal microscopy. Results: Neuronal cultures treated with NH4Cl exhibited mitochondrial dysfunction, ROS overproduction, and reduced cell viability (27.8 ± 2.3% and 41.5 ± 3.7%, respectively) compared with untreated cultures (15.7 ± 1.0%, both p <0.0001). BDL led to increased cerebral LP (p = 0.0003) and cytosolic ROS generation (p <0.0001), which was restored by OP (both p <0.0001). Mitochondrial function was severely compromised in BDL, resulting in hyperpolarisation of Δψm with consequent overconsumption of adenosine triphosphate and augmentation of mitochondrial ROS production. Administration of OP restored Δψm. In BDL animals, neuronal loss was observed in hippocampal areas, which was partially prevented by OP. Conclusions: Our results elucidate that low-grade hyperammonaemia in cirrhosis can severely impact on brain mitochondrial function. Profound neuronal injury was observed in hyperammonaemic conditions, which was partially reversible by OP. This points towards a novel mechanism of HE development. Lay summary: The impact of hyperammonaemia, a common finding in patients with liver cirrhosis, on brain mitochondrial function was investigated in this study. The results show that ammonia in concentrations commonly seen in patients induces severe mitochondrial dysfunction, overproduction of damaging oxygen molecules, and profound injury and death of neurons in rat brain cells. These findings point towards a novel mechanism of ammonia-induced brain injury in liver failure and potential novel therapeutic targets.

11.
Neuropharmacology ; 77: 249-56, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24157493

RESUMEN

It is established that growth factors support neuronal survival through the phosphoinositide 3-kinase (PI3K)/Akt pathway but little is known about factors that inhibit Akt signaling in neurons. Given that the sst2 type somatostatin receptor exerts pro-apoptotic effects in tumor cells by inhibiting PI3K/Akt, we examined whether neuronal sst2 has similar effects. In primary cortical cultures heterozygously expressing a sst2 knockout/lacZ knockin allele, beta-galactosidase staining revealed expression of the sst2 gene in the vast majority of the cultured neurons. Somatostatin was identified in a subpopulation of neurons by immunocytochemistry. Immunoblots showed a strong reduction of Akt phosphorylation at S473 in wild type cultures undergoing stimulation with the sst2 agonist BIM-23244. While the sst2 agonist did not cause neuronal death under control conditions, it promoted hypoxic/ischemic neuronal death in cortical cultures subjected to oxygen/glucose deprivation. Treatment of wild type cultures with the sst2 antagonist BIM-23627 and homozygous deletion of the sst2 gene were protective in this paradigm, suggesting that endogenous somatostatin signals through sst2 during hypoxia/ischemia. In fact, examination of sst2 phosphorylation and sst2 internalization provided evidence for sst2 activation in neurons subjected to oxygen/glucose deprivation. Thus, somatostatin acts as a sensor of hypoxia/ischemia, inhibits Akt activity through sst2 and aggravates hypoxic/ischemic neuronal death. sst2-selective antagonists are proposed as neuroprotectants in stroke.


Asunto(s)
Muerte Celular/fisiología , Corteza Cerebral/metabolismo , Hipoxia-Isquemia Encefálica/metabolismo , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Somatostatina/metabolismo , Animales , Muerte Celular/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Ratones , Ratones Transgénicos , Neuronas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/genética
12.
Cell Signal ; 26(2): 220-32, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24211626

RESUMEN

Tissue kallikrein (TK) was previously shown to take most of its biological effects through bradykinin receptors. In this study, we assumed that TK mediated neurite outgrowth was independent of bradykinin receptors. To test the hypothesis, we investigated TK-induced neurite outgrowth and its signaling mechanisms in cultured primary neurons and human SH-SY5Y cells. We found that TK stimulation could increase the number of processes and mean process length of primary neurons, which were blocked by epidermal growth factor receptor (EGFR) inhibitor or down-regulation, small interfering RNA for flotillin-2 and extracellular signal-regulated kinase (ERK) 1/2 inhibitor. Moreover, TK-induced neurite outgrowth was associated with EGFR and ERK1/2 activation, which were inhibited by EGFR antagonist or RNA interference and flotillin-2 knockdown. Interestingly, inhibition of bradykinin receptors had no significant effects on EGFR and ERK1/2 phosphorylation. In the present research, our data also suggested that EGFR and flotillin-2 formed constitutive complex that translocated to around the nuclei in the TK stimulation. In sum, our findings provided evidence that TK could promote neurite outgrowth via EGFR, flotillin-2 and ERK1/2 signaling pathway in vitro.


Asunto(s)
Receptores ErbB/metabolismo , Proteínas de la Membrana/metabolismo , Neuritas/efectos de los fármacos , Calicreínas de Tejido/farmacología , Animales , Células Cultivadas , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos BALB C , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neuritas/metabolismo , Neuronas/citología , Neuronas/metabolismo , Fosforilación/efectos de los fármacos , Quinazolinas/farmacología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos , Tirfostinos/farmacología
13.
Neurosci Lett ; 559: 193-8, 2014 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-24342440

RESUMEN

Neural stem cells (NSCs) have been the focus of an intensive effort to direct their differentiation in vitro towards desired neuronal phenotypes for cell replacement therapies. It is thought that NSCs derived from older embryos have limited neurogenic capacity and are restricted towards an astroglial fate. This idea is largely based on studies that typically analysed NSC-derived progeny following one week of in vitro differentiation. In this report, the neurogenic capacity of older ventral midbrain (VM) NSCs was assessed. When the older NSCs were differentiated for three weeks, there were significant increases in the numbers of newly born neurons at 14 and 21 days, as assessed by 5-bromo-2'-deoxyuridine (BrdU) incorporation. Therefore this study demonstrates that older NSCs retain significantly more neurogenic potential than was previously thought. These data have implications for NSC preparatory protocols and the choice of donor age for cell transplantation studies, and contributes to the understanding of NSC behaviour in vitro.


Asunto(s)
Diferenciación Celular/fisiología , Mesencéfalo/citología , Mesencéfalo/fisiología , Células-Madre Neurales/fisiología , Neurogénesis/fisiología , Animales , Células Cultivadas , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
14.
Neuroscience ; 257: 96-110, 2014 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-24200924

RESUMEN

The spiral ganglion conveys afferent auditory information predominantly through a single class of type I neurons that receive signals from inner hair cell sensory receptors. These auditory primary afferents, like in other systems (Puopolo and Belluzzi, 1998; Gascon and Moqrich, 2010; Leao et al., 2012) possess a marked diversity in their electrophysiological features (Taberner and Liberman, 2005). Consistent with these observations, when the auditory primary afferents were assessed in neuronal explants separated from their peripheral and central targets it was found that individual neurons were markedly heterogeneous in their endogenous electrophysiological features. One aspect of this heterogeneity, obvious throughout the ganglion, was their wide range of excitability as assessed by voltage threshold measurements (Liu and Davis, 2007). Thus, while neurons in the base differed significantly from apical and middle neurons in their voltage thresholds, each region showed distinctly wide ranges of values. To determine whether the resting membrane potentials (RMPs) of these neurons correlate with the threshold distribution and to identify the ion channel regulatory elements underlying heterogeneous neuronal excitability in the ganglion, patch-clamp recordings were made from postnatal day (P5-8) murine spiral ganglion neurons in vitro. We found that RMP mirrored the tonotopic threshold distribution, and contributed an additional level of heterogeneity in each cochlear location. Pharmacological experiments further indicated that threshold and RMP was coupled through the Kv1 current, which had a dual impact on both electrophysiological parameters. Whereas, hyperpolarization-activated cationic channels decoupled these two processes by primarily affecting RMP without altering threshold level. Thus, beyond mechanical and synaptic specializations, ion channel regulation of intrinsic membrane properties imbues spiral ganglion neurons with different excitability levels, a feature that contributes to primary auditory afferent diversity.


Asunto(s)
Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Neuronas/metabolismo , Canales de Potasio de la Superfamilia Shaker/metabolismo , Ganglio Espiral de la Cóclea/citología , 4-Aminopiridina/farmacología , Animales , Animales Recién Nacidos , Fenómenos Biofísicos/efectos de los fármacos , Biofisica , Cloruro de Cadmio/farmacología , Venenos Elapídicos/farmacología , Estimulación Eléctrica , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Neurotoxinas/farmacología , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/farmacología , Tetrodotoxina/farmacología
15.
J Neurosci Methods ; 223: 11-9, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24316294

RESUMEN

Adult zebrafish (Danio rerio) have a remarkable ability to restore function after an injury to the brain or spinal cord. The molecular and cellular mechanisms underlying this phenomenon are not fully understood. To enable investigation of these mechanisms we have developed an in vitro model system from the adult zebrafish brainstem, which can be maintained under serum-containing and serum-free conditions. While cultures are predominantly neuronal, they also contain glia and stem progenitor cells. Various stages of cellular differentiation are observed among both neuronal and non-neuronal populations. Quantitative morphological results revealed typical cellular growth over a two-week period. We argue that our novel brainstem culture model offers a powerful tool for the studies of axonal growth, neurogenesis, and regeneration in the adult zebrafish central nervous system.


Asunto(s)
Tronco Encefálico/citología , Neuronas/fisiología , Células Madre Adultas/fisiología , Animales , Bromodesoxiuridina/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Medio de Cultivo Libre de Suero/farmacología , Femenino , Masculino , Proteínas del Tejido Nervioso/metabolismo , Neuroglía/metabolismo , Neuronas/clasificación , Técnicas de Cultivo de Órganos , Factores de Tiempo , Tubulina (Proteína)/metabolismo , Pez Cebra
16.
Neuropharmacology ; 77: 39-48, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24067927

RESUMEN

Axonal degeneration of dopaminergic neurons is one of the pathological features in the early stages of Parkinson disease. Promotion of axonal outgrowth of the remaining dopaminergic neurons leads to the recovery of the nigrostriatal pathway. Staurosporine (STS), a wide-spectrum kinase inhibitor, induces neurite outgrowth in various cell types, although its mechanism of action remains elusive. In this study, we analyzed which protein kinase is involved in STS-induced neurite outgrowth. We have previously established the method to measure the length of dopaminergic neurites that extend from a mesencephalic cell region, which is formed on a coverslip by an isolation wall. By means of this method, we clarified that STS treatment causes dopaminergic axonal outgrowth in mesencephalic primary cultures. Among the specific protein kinase inhibitors we tested, compound C (C.C), an AMP-activated protein kinase (AMPK) inhibitor, promoted dopaminergic neurite outgrowth. STS as well as C.C elevated the phosphorylation level of 70-kDa ribosomal protein S6 kinase, a downstream target of mammalian target of rapamycin (mTOR) signaling pathway. The STS- and C.C-induced dopaminergic neurite outgrowth was suppressed by rapamycin, an mTOR inhibitor. Furthermore, the application of C.C rescued 1-methyl-4-phenylpyridinium ion (MPP(+))-induced dopaminergic neurite degeneration. These results suggest that STS induces dopaminergic axonal outgrowth through mTOR signaling pathway activation as a consequence of AMPK inhibition.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Neuritas/efectos de los fármacos , Estaurosporina/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Neuronas Dopaminérgicas/metabolismo , Neuritas/metabolismo , Neurogénesis/efectos de los fármacos , Células PC12 , Fosforilación/efectos de los fármacos , Ratas , Transducción de Señal/efectos de los fármacos
17.
Stem Cell Res ; 12(1): 241-9, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24280418

RESUMEN

In severe cases of sensorineural hearing loss where the numbers of auditory neurons are significantly depleted, stem cell-derived neurons may provide a potential source of replacement cells. The success of such a therapy relies upon producing a population of functional neurons from stem cells, to enable precise encoding of sound information to the brainstem. Using our established differentiation assay to produce sensory neurons from human stem cells, patch-clamp recordings indicated that all neurons examined generated action potentials and displayed both transient sodium and sustained potassium currents. Stem cell-derived neurons reliably entrained to stimuli up to 20 pulses per second (pps), with 50% entrainment at 50 pps. A comparison with cultured primary auditory neurons indicated similar firing precision during low-frequency stimuli, but significant differences after 50 pps due to differences in action potential latency and width. The firing properties of stem cell-derived neurons were also considered relative to time in culture (31-56 days) and revealed no change in resting membrane potential, threshold or firing latency over time. Thus, while stem cell-derived neurons did not entrain to high frequency stimulation as effectively as mammalian auditory neurons, their electrical phenotype was stable in culture and consistent with that reported for embryonic auditory neurons.


Asunto(s)
Células Madre Embrionarias/citología , Células Receptoras Sensoriales/citología , Células Receptoras Sensoriales/fisiología , Diferenciación Celular , Línea Celular , Fenómenos Electrofisiológicos , Células Madre Embrionarias/fisiología , Humanos , Técnicas de Placa-Clamp , Células Receptoras Sensoriales/metabolismo , Células Madre/citología , Células Madre/fisiología
18.
Neuroscience ; 259: 13-24, 2014 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-24295634

RESUMEN

AMP-activated protein kinase (AMPK) is a serine/threonine kinase that functions as a cellular and whole organism energy sensor to regulate ATP-consuming (anabolic) and ATP-generating (catabolic) pathways. The heterotrimeric AMPK complex consists of a catalytic α-subunit, regulatory ß-subunit, and an AMP/ATP-binding γ-subunit. Several alternate isoforms exist for each subunit (α1, α2, ß1, ß2, γ1, γ2 and γ3). However, little is known of the expression pattern or function of the individual catalytic complexes in regulating neuronal structure. In this study, we examined the role of AMPK subunits in differentiating hippocampal neurons. We found that during development, the expression of AMPK subunits increase and that activation of AMPK by energetic stress inhibits neuronal development at multiple stages, not only during axon outgrowth, but also during dendrite growth and arborization. The presence of a single functional AMPK catalytic complex was sufficient to mediate these inhibitory effects of energetic stress. Activation of AMPK mediates these effects by suppressing both the mTOR and Akt signaling pathways. These findings demonstrate that the energy-sensing AMPK pathway regulates neuronal structure in distinct regions of developing neurons at multiple stages of development.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Dendritas/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Hipocampo/citología , Neuritas/fisiología , Neuronas/ultraestructura , Proteínas Quinasas Activadas por AMP/deficiencia , Proteínas Quinasas Activadas por AMP/genética , Factores de Edad , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Animales Recién Nacidos , Células Cultivadas , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hipoglucemiantes/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/fisiología , Ratas , Ratas Sprague-Dawley , Ribonucleótidos/farmacología
19.
J Chem Neuroanat ; 53: 11-7, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24126226

RESUMEN

Glutamate-induced excitotoxicity is a major contributor to motor neuron (MN) degeneration in disorders such as amyotrophic lateral sclerosis (ALS), stroke and spinal cord injury. Numerous in vitro and in vivo models have been developed to evaluate the efficacy and mode of action of neuroprotective agents. However, the dominance of glutamate receptor-subtype in the different regions of the spinal cord in these models has generally been overlooked. This study first compared the neuroprotective effect of administering glutamate receptor antagonists, (+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine (MK-801) and 6-cyano-7-nitroquinoxaline-2,3-dione (CNQX), into a serum-free excitotoxic organotypic in vitro system, on the survival of MNs located in the lumbar area of spinal cord. The poor neuroprotection provided by MK-801 (NMDA (N-methyl-D-aspartate) antagonist) in comparison to CNQX (AMPA/KA (a-amino-3-hydroxy-5-methyl-4-isoxazole propionate/kainate) antagonist), raised the hypothesis that the extent of engagement by glutamate receptor sub-types in the mechanism of excitotoxicity may differ within different populations of MNs. The consequent examination of MN susceptibility to glutamate-induced excitotoxicity in relation to the rostro-caudal level from which MN originated revealed a differential glutamate receptor sub-type dominance at different spinal cord regions (i.e. cervical, thoracic and lumbar). In the cervical and lumbar regions, the AMPA receptor was the main contributor to MN excitotoxicity, whereas in thoracic regions, the NMDA receptor was the main contributor. This study provides a new way of looking at mechanisms leading to glutamate-induced excitotoxicity in MN and may therefore be important for the development of treatment strategies in protection of spinal MNs in neurodegenerative disease and traumatic injury.


Asunto(s)
Antagonistas de Aminoácidos Excitadores/farmacología , Ácido Glutámico/toxicidad , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/metabolismo , Fármacos Neuroprotectores/farmacología , Animales , Supervivencia Celular/efectos de los fármacos , Vértebras Cervicales , Inmunohistoquímica , Región Lumbosacra , Técnicas de Cultivo de Órganos , Ratas , Ratas Sprague-Dawley , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Vértebras Torácicas
20.
J Neurosci Methods ; 218(2): 164-9, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23774648

RESUMEN

Modelling the complex process of neuromuscular signalling is key to understanding not only normal circuit function but also importantly the mechanisms underpinning a range of degenerative diseases. We describe a novel in vitro model of the lower motor neuron-neuromuscular junction circuit, incorporating primary spinal motor neurons, supporting glia and skeletal muscle. This culture model is designed to spatially mimic the unique anatomical and cellular interactions of this circuit in compartmented microfluidic devices, such that the glial cells are located with motor neuron cell bodies in the cell body chamber and motor neuron axons extend to a distal chamber containing skeletal muscle cells whilst simultaneously allowing targeted intervention. This model is suitable for use in conjunction with a range of downstream experimental approaches and could also be modified to utilise other cellular sources including appropriate immortal cell lines, cells derived from transgenic models of disease and also patient derived stem cells.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Técnicas Analíticas Microfluídicas/métodos , Neuronas Motoras/citología , Células Musculares/citología , Unión Neuromuscular/citología , Animales , Técnicas de Cocultivo/métodos , Inmunohistoquímica , Microscopía Electrónica de Rastreo , Neuroglía/citología , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda