Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
BMC Biol ; 18(1): 58, 2020 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-32466798

RESUMEN

BACKGROUND: Network alterations underlying neurodegenerative diseases often precede symptoms and functional deficits. Thus, their early identification is central for improved prognosis. In Huntington's disease (HD), the cortico-striatal networks, involved in motor function processing, are the most compromised neural substrate. However, whether the network alterations are intrinsic of the striatum or the cortex is not fully understood. RESULTS: In order to identify early HD neural deficits, we characterized neuronal ensemble calcium activity and network topology of HD striatal and cortical cultures. We used large-scale calcium imaging combined with activity-based network inference analysis. We extracted collective activity events and inferred the topology of the neuronal network in cortical and striatal primary cultures from wild-type and R6/1 mouse model of HD. Striatal, but not cortical, HD networks displayed lower activity and a lessened ability to integrate information. GABAA receptor blockade in healthy and HD striatal cultures generated similar coordinated ensemble activity and network topology, highlighting that the excitatory component of striatal system is spared in HD. Conversely, NMDA receptor activation increased individual neuronal activity while coordinated activity became highly variable and undefined. Interestingly, by boosting NMDA activity, we rectified striatal HD network alterations. CONCLUSIONS: Overall, our integrative approach highlights striatal defective network integration capacity as a major contributor of basal ganglia dysfunction in HD and suggests that increased excitatory drive may serve as a potential intervention. In addition, our work provides a valuable tool to evaluate in vitro network recovery after treatment intervention in basal ganglia disorders.


Asunto(s)
Cuerpo Estriado/fisiopatología , Enfermedad de Huntington/fisiopatología , Neuronas/fisiología , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos
2.
Mol Neurobiol ; 55(11): 8263-8277, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29526016

RESUMEN

Deficits in hippocampal synaptic plasticity result in cognitive impairment in Huntington's disease (HD). Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide that exerts neuroprotective actions, mainly through the PAC1 receptor. However, the role of PACAP in cognition is poorly understood, and no data exists in the context of Huntington's disease (HD). Here, we investigated the ability of PACAP receptor stimulation to enhance memory development in HD. First, we observed a hippocampal decline of all three PACAP receptor expressions, i.e., PAC1, VPAC1, and VPAC2, in two different HD mouse models, R6/1 and HdhQ7/Q111, from the onset of cognitive dysfunction. In hippocampal post-mortem human samples, we found a specific decrease of PAC1, without changes in VPAC1 and VPAC2 receptors. To determine whether activation of PACAP receptors could contribute to improve memory performance, we conducted daily intranasal administration of PACAP38 to R6/1 mice at the onset of cognitive impairment for seven days. We found that PACAP treatment rescued PAC1 level in R6/1 mice, promoted expression of the hippocampal brain-derived neurotrophic factor, and reduced the formation of mutant huntingtin aggregates. Furthermore, PACAP administration counteracted R6/1 mice memory deficits as analyzed by the novel object recognition test and the T-maze spontaneous alternation task. Importantly, the effect of PACAP on cognitive performance was associated with an increase of VGlut-1 and PSD95 immunolabeling in hippocampus of R6/1 mice. Taken together, these results suggest that PACAP, acting through stimulation of PAC1 receptor, may have a therapeutic potential to counteract cognitive deficits induced in HD.


Asunto(s)
Hipocampo/fisiopatología , Enfermedad de Huntington/fisiopatología , Memoria/fisiología , Plasticidad Neuronal/efectos de los fármacos , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Administración Intranasal , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Trastornos del Conocimiento/fisiopatología , Modelos Animales de Enfermedad , Homólogo 4 de la Proteína Discs Large/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/patología , Humanos , Proteína Huntingtina/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/administración & dosificación , Agregado de Proteínas , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo
3.
Cell Death Dis ; 5: e1364, 2014 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-25101677

RESUMEN

Mutations in the PARK2 gene are associated with an autosomal recessive form of juvenile parkinsonism (AR-JP). These mutations affect parkin solubility and impair its E3 ligase activity, leading to a toxic accumulation of proteins within susceptible neurons that results in a slow but progressive neuronal degeneration and cell death. Here, we report that RTP801/REDD1, a pro-apoptotic negative regulator of survival kinases mTOR and Akt, is one of such parkin substrates. We observed that parkin knockdown elevated RTP801 in sympathetic neurons and neuronal PC12 cells, whereas ectopic parkin enhanced RTP801 poly-ubiquitination and proteasomal degradation. In parkin knockout mouse brains and in human fibroblasts from AR-JP patients with parkin mutations, RTP801 levels were elevated. Moreover, in human postmortem PD brains with mutated parkin, nigral neurons were highly positive for RTP801. Further consistent with the idea that RTP801 is a substrate for parkin, the two endogenous proteins interacted in reciprocal co-immunoprecipitates of cell lysates. A potential physiological role for parkin-mediated RTP801 degradation is indicated by observations that parkin protects neuronal cells from death caused by RTP801 overexpression by mediating its degradation, whereas parkin knockdown exacerbates such death. Similarly, parkin knockdown enhanced RTP801 induction in neuronal cells exposed to the Parkinson's disease mimetic 6-hydroxydopamine and increased sensitivity to this toxin. This response to parkin loss of function appeared to be mediated by RTP801 as it was abolished by RTP801 knockdown. Taken together these results indicate that RTP801 is a novel parkin substrate that may contribute to neurodegeneration caused by loss of parkin expression or activity.


Asunto(s)
Proteínas Represoras/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Encéfalo/metabolismo , Células HEK293 , Humanos , Leupeptinas/farmacología , Ratones , Ratones Noqueados , Neuronas/metabolismo , Oxidopamina/toxicidad , Células PC12 , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Complejo de la Endopetidasa Proteasomal/química , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Proteolisis/efectos de los fármacos , ARN Interferente Pequeño/metabolismo , Ratas , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Factores de Transcripción , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
4.
Cell Death Dis ; 4: e595, 2013 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-23598407

RESUMEN

Neuroprotective therapies based on brain-derived neurotrophic factor (BDNF) administration have been proposed for Huntington's disease (HD) treatment. However, our group has recently reported reduced levels of TrkB in HD mouse models and HD human brain suggesting that besides a decrease on BDNF levels a reduction of TrkB expression could also contribute to diminished neurotrophic support in HD. BDNF can also bind to p75 neurotrophin receptor (p75(NTR)) modulating TrkB signaling. Therefore, in this study we have analyzed the levels of p75(NTR) in several HD models, as well as in HD human brain. Our data demonstrates a p75(NTR)/TrkB imbalance in the striatum of two different HD mouse models, Hdh(Q111/111) homozygous knockin mice and R6/1 mice that was also manifested in the putamen of HD patients. The imbalance between TrkB and p75(NTR) levels in a HD cellular model did not affect BDNF-mediated TrkB activation of prosurvival pathways but induced activation of apoptotic cascades as demonstrated by increased JNK phosphorylation. Moreover, BDNF failed to protect mutant huntingtin striatal cells transfected with p75(NTR) against NMDA-mediated excitotoxicity, which was associated with decreased Akt phosphorylation. Interestingly, lack of Akt activation following BDNF and NMDA treatment correlated with increased PP1 levels. Accordingly, pharmacological inhibition of PP1 by okadaic acid (OA) prevented mutant huntingtin striatal cell death induced by NMDA and BDNF. Altogether, our findings demonstrate that the p75(NTR)/TrkB imbalance induced by mutant huntingtin in striatal cells associated with the aberrant activity of PP1 disturbs BDNF neuroprotection likely contributing to increasing striatal vulnerability in HD. On the basis of this data we hypothesize that normalization of p75(NTR) and/or TrkB expression or their signaling will improve BDNF neuroprotective therapies in HD.


Asunto(s)
Enfermedad de Huntington/metabolismo , Receptor de Factor de Crecimiento Nervioso/metabolismo , Receptor trkB/metabolismo , Animales , Apoptosis/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/farmacología , Factor Neurotrófico Derivado del Encéfalo/uso terapéutico , Línea Celular , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Técnicas de Sustitución del Gen , Humanos , Proteína Huntingtina , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/patología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , N-Metilaspartato/farmacología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Ácido Ocadaico/farmacología , Fosforilación , Unión Proteica , Proteína Fosfatasa 1/antagonistas & inhibidores , Proteína Fosfatasa 1/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Putamen/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptor de Factor de Crecimiento Nervioso/antagonistas & inhibidores , Receptor de Factor de Crecimiento Nervioso/genética , Transducción de Señal
5.
Brain Res ; 1459: 100-12, 2012 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-22560595

RESUMEN

Huntington's disease (HD) is a neurodegenerative disease characterized by progressive motor, cognitive and psychiatric deficits, associated with predominant loss of striatal neurons and caused by a polyglutamine expansion in the huntingtin protein. There is so far neither cure nor approved disease-slowing therapy for HD, though recent clinical studies have shown a beneficial long-term effect of pridopidine in patients with HD. The nature of this effect, purely symptomatic or, in addition, neuroprotective, is difficult to elucidate in clinical trials. Pridopidine and (-)-OSU6162 are members of a new family of compounds referred to as dopaminergic stabilizers, which normalize abnormal dopamine neurotransmission. We investigated the effects of (-)-OSU6162 on huntingtin knocked-in striatal neurons in culture. Control neurons had normal full-length huntingtin with 7 glutamines while "mutant" neurons had large expansions (Q=111). We studied the dose-effect curves of (-)-OSU6162 on mitochondrial activity, LDH levels, necrosis and apoptosis in untreated Q7 and Q111 cells. In addition, we investigated the effects of (-)-OSU6162 on Q7 and Q111 neurons challenged with different neurotoxins such as sodium glutamate, H(2)O(2), rotenone and 3-nitropropionic acid (3NP). As we found prevention of toxicity of some of these neurotoxins, we investigated the putative neuroprotective mechanisms of action of (-)-OSU6162 measuring the effects of this dopaminergic stabilizer on expression and release of BDNF, the ratios of Bcl2/Bax proteins and of p-ERK/ERK, the levels of chaperones and GSH, and the effects of (-)-OSU6162 on dopamine uptake and release. We found that (-)-OSU6162, 3-150 µM, produces a dose dependent increase of mitochondrial activity and a reduction of cell death. (-)-OSU6162 does not change glutamate toxicity, but it partially prevents that of H(2)O(2), rotenone and 3-nitropropionic acid. (-)-OSU6162 increases the intracellular levels of BDNF and Bcl2/Bax and decreases those of p-ERK/ERK and CHIP in Q111 cells. (-)-OSU6162 increased (3)H-dopamine uptake and amphetamine-induced (3)H-dopamine release in E13 mouse mid brain neurons. Our studies demonstrate that (-)-OSU6162 improves survival and mitochondrial function in striatal Q111 neurons and the resistance of these cells to several striatal neurotoxins, suggesting that (-)-OSU6162 and related compounds should be tested for neuroprotection in animal models and, eventually, in patients with HD.


Asunto(s)
Cuerpo Estriado/citología , Dopaminérgicos/farmacología , Peróxido de Hidrógeno/toxicidad , Proteínas del Tejido Nervioso/genética , Neuronas/efectos de los fármacos , Péptidos/metabolismo , Piperidinas/farmacología , Rotenona/toxicidad , Análisis de Varianza , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Dopamina/metabolismo , Relación Dosis-Respuesta a Droga , Embrión de Mamíferos , Ensayo de Inmunoadsorción Enzimática , Ácido Glutámico/toxicidad , Glutatión/metabolismo , Humanos , Proteína Huntingtina , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Proteínas del Tejido Nervioso/química , Péptidos/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transfección/métodos , Tritio/metabolismo
6.
Hum Mol Genet ; 21(6): 1203-16, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22116937

RESUMEN

Huntington's disease (HD) is an autosomal dominant progressive neurodegenerative disorder caused by an expanded CAG/polyglutamine repeat in the coding region of the huntingtin (htt) gene. Although HD is classically considered a motor disorder, there is now considerable evidence that early cognitive deficits appear in patients before the onset of motor disturbances. Here we demonstrate early impairment of long-term spatial and recognition memory in heterozygous HD knock-in mutant mice (Hdh(Q7/Q111)), a genetically accurate HD mouse model. Cognitive deficits are associated with reduced hippocampal expression of CREB-binding protein (CBP) and diminished levels of histone H3 acetylation. In agreement with reduced CBP, the expression of CREB/CBP target genes related to memory, such c-fos, Arc and Nr4a2, was significantly reduced in the hippocampus of Hdh(Q7/Q111) mice compared with wild-type mice. Finally, and consistent with a role of CBP in cognitive impairment in Hdh(Q7/Q111) mice, administration of the histone deacetylase inhibitor trichostatin A rescues recognition memory deficits and transcription of selective CREB/CBP target genes in Hdh(Q7/Q111) mice. These findings demonstrate an important role for CBP in cognitive dysfunction in HD and suggest the use of histone deacetylase inhibitors as a novel therapeutic strategy for the treatment of memory deficits in this disease.


Asunto(s)
Proteína de Unión a CREB/fisiología , Modelos Animales de Enfermedad , Histona Acetiltransferasas/deficiencia , Enfermedad de Huntington/enzimología , Enfermedad de Huntington/patología , Memoria a Largo Plazo , Acetilación , Animales , Conducta Animal , Western Blotting , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/patología , Femenino , Genes fos , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Técnicas para Inmunoenzimas , Inmunoprecipitación , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Gene Ther ; 17(10): 1294-308, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20463759

RESUMEN

Brain-derived neurotrophic factor (BDNF) is the main candidate for neuroprotective therapeutic strategies for Huntington's disease. However, the administration system and the control over the dosage are still important problems to be solved. Here we generated transgenic mice overexpressing BDNF under the promoter of the glial fibrillary acidic protein (GFAP) (pGFAP-BDNF mice). These mice are viable and have a normal phenotype. However, intrastriatal administration of quinolinate increased the number of reactive astrocytes and enhanced the release of BDNF in pGFAP-BDNF mice compared with wild-type mice. Coincidentally, pGFAP-BDNF mice are more resistant to quinolinate than wild-type mice, suggesting a protective effect of astrocyte-derived BDNF. To verify this, we next cultured astrocytes from pGFAP-BDNF and wild-type mice for grafting. Wild-type and pGFAP-BDNF-derived astrocytes behave similarly in nonlesioned mice. However, pGFAP-BDNF-derived astrocytes showed higher levels of BDNF and larger neuroprotective effects than the wild-type ones when quinolinate was injected 30 days after grafting. Interestingly, mice grafted with pGFAP-BDNF astrocytes showed important and sustained behavioral improvements over time after quinolinate administration as compared with mice grafted with wild-type astrocytes. These findings show that astrocytes engineered to release BDNF can constitute a therapeutic approach for Huntington's disease.


Asunto(s)
Astrocitos/metabolismo , Factor Neurotrófico Derivado del Encéfalo/genética , Proteína Ácida Fibrilar de la Glía/genética , Enfermedad de Huntington/terapia , Fármacos Neuroprotectores/metabolismo , Regiones Promotoras Genéticas , Animales , Astrocitos/citología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Ratones , Ratones Transgénicos , Fenotipo , Ácido Quinolínico/administración & dosificación , Ácido Quinolínico/farmacología
9.
Cell Death Differ ; 17(2): 324-35, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19745829

RESUMEN

Dysregulation of gene expression is one of the mechanisms involved in the pathophysiology of Huntington's disease (HD). Here, we examined whether mutant huntingtin regulates the levels of PH domain leucine-rich repeat protein phosphatase 1 (PHLPP1), a phosphatase that specifically dephosphorylates Akt at Ser473. Our results show decreased PHLPP1 protein levels in knock-in models (Hdh(Q111/Q111) mouse striatum and STHdh(Q111/Q111) cells), in the striatum of N-terminal exon-1 mutant huntingtin transgenic mouse models (R6/1; R6/1 : BDNF + or - , R6/2 and Tet/HD94) and in the putamen of HD patients. Quantitative PCR analysis revealed a reduction in PHLPP1 mRNA levels in the striatum of R6/1 compared with wild-type mice. Coincident with reduced PHLPP1 protein levels, we observed increased phosphorylated Akt (Ser473) levels specifically in the striatum. The analysis of the conditional mouse model Tet/HD94 disclosed that after mutant huntingtin shutdown PHLPP1 levels returned to wild-type levels whereas phospho-Akt levels were partially reduced. In conclusion, our results show that mutant huntingtin downregulates PHLPP1 expression. In the striatum, these reduced levels of PHLPP1 can contribute to maintain high levels of activated Akt that may delay cell death and allow the recovery of neuronal viability after mutant huntingtin silencing.


Asunto(s)
Cuerpo Estriado/enzimología , Enfermedad de Huntington/enzimología , Proteínas Nucleares/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Adulto , Anciano , Animales , Muerte Celular/fisiología , Línea Celular Transformada , Núcleo Celular/metabolismo , Cuerpo Estriado/patología , Citosol/metabolismo , Modelos Animales de Enfermedad , Exones/genética , Femenino , Técnicas de Sustitución del Gen , Humanos , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Neurotoxinas/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/genética , Fosfoproteínas Fosfatasas/química , Fosfoproteínas Fosfatasas/genética , Fosforilación/fisiología , Estructura Terciaria de Proteína
10.
Neuroscience ; 158(4): 1234-50, 2009 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-19121372

RESUMEN

The involvement of brain-derived neurotrophic factor (BDNF) in cognitive processes and the decrease in its expression in Huntington's disease suggest that this neurotrophin may play a role in learning impairment during the disease progression. We therefore analyzed the onset and severity of cognitive deficits in two different mouse models with the same mutant huntingtin but with different levels of BDNF (R6/1 and R6/1:BDNF+/- mice). We observed that BDNF modulates cognitive function in different learning tasks, even before the onset of motor symptoms. R6/1:BDNF+/- mice showed earlier and more accentuated cognitive impairment than R6/1 mice at 5 weeks of age in discrimination learning; at 5 weeks of age in procedural learning; and at 9 weeks of age in alternation learning. At the earliest age at which cognitive impairment was detected, electrophysiological analysis was performed in the hippocampus. All mutant genotypes showed reduced hippocampal long term potentiation (LTP) with respect to wild type but did not show differences between them. Thus, we evaluated the involvement of BDNF-trkB signaling and glutamate receptor expression in the hippocampus of these mice. We observed a decrease in phospholipaseCgamma activity, but not ERK, in R61, BDNF+/- and R6/1:BDNF+/- hippocampus at the age when LTP was altered. However, a specific decrease in the expression of glutamate receptors NR1, NR2A and GluR1 was detected only in R6/1:BDNF+/- hippocampus. Therefore, these results show that BDNF modulates the learning and memory alterations induced by mutant huntingtin. This interaction leads to intracellular changes, such as specific changes in glutamate receptors and in BDNF-trkB signaling through phospholipaseCgamma.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Trastornos del Conocimiento/fisiopatología , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Fosfolipasa C gamma/metabolismo , Receptores de Glutamato/metabolismo , Factores de Edad , Análisis de Varianza , Animales , Biofisica , Factor Neurotrófico Derivado del Encéfalo/genética , Trastornos del Conocimiento/genética , Aprendizaje Discriminativo/fisiología , Modelos Animales de Enfermedad , Estimulación Eléctrica/métodos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , Proteína Huntingtina , Técnicas In Vitro , Potenciación a Largo Plazo/genética , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos CBA , Ratones Transgénicos , Mutación , Técnicas de Placa-Clamp/métodos , Receptores AMPA/genética , Receptores AMPA/metabolismo , Receptores de Glutamato/genética , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Natación
11.
J Neuroimmunol ; 196(1-2): 188-91, 2008 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-18462809

RESUMEN

Antibodies against neuronal surface antigens (NSA-ab) have been described in pediatric opsoclonus-myoclonus syndrome (OMS). We analyzed the presence of NSA-ab by flow cytometry and immunocytochemistry of live cerebellar granular neurons (CGN) in the serum of 25 adult patients with idiopathic (14) and paraneoplastic (11) OMS. Paraneoplastic, but not idiopathic, OMS sera showed a CGN surface binding by flow cytometry higher than that of controls (mean MFI (median fluorescence intensity): 29+/-6.9 vs. 20+/-5.8; p=0.001) but only one serum had a binding greater than three standard deviations of controls. OMS sera did not label live CGN by immunocytochemistry. Unlike pediatric OMS, NSA-ab were not detected in adult cases suggesting that the immunity to NSA in OMS is heterogeneous.


Asunto(s)
Anticuerpos/metabolismo , Antígenos de Superficie/inmunología , Neuronas/inmunología , Síndrome de Opsoclonía-Mioclonía/inmunología , Adulto , Anciano , Animales , Células Cultivadas , Femenino , Citometría de Flujo/métodos , Humanos , Masculino , Persona de Mediana Edad , Ratas , Ratas Wistar
12.
Mol Cell Neurosci ; 37(4): 663-72, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18272391

RESUMEN

In this study we analyzed whether other members of the Bcl-2 family are regulated in the absence of Bax during the postnatal development of the striatum and cortex and after striatal excitotoxic lesion. Compared with wild-type animals, Bax knockout mice showed region- and time-dependent increases in pro-apoptotic proteins Bak and Bim(EL). Excitotoxicity induced in the adult striatum increased Bim(EL) in both genotypes whereas Bak and Bcl-x(L) were only increased in Bax knockout mice. However, translocation of Bim(EL) protein to the mitochondrial fraction, cytochrome c release and caspase-3 activation were only observed in wild-type striata. Furthermore, analysis of Bim null mutant mice showed that this protein is not essential to excitotoxicity-induced striatal cell death. In conclusion, our results show that in Bax deficient mice Bim(EL) and Bak are specifically regulated during postnatal development, suggesting that these proteins may participate in the compensatory mechanisms triggered in the absence of Bax. In contrast, Bax is required to induce apoptosis after excitotoxicity in the adult striatum.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/biosíntesis , Corteza Cerebral/metabolismo , Cuerpo Estriado/metabolismo , Proteínas de la Membrana/biosíntesis , Proteínas Proto-Oncogénicas/biosíntesis , Regulación hacia Arriba , Proteína Destructora del Antagonista Homólogo bcl-2/biosíntesis , Proteína X Asociada a bcl-2/deficiencia , Animales , Animales Recién Nacidos , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/genética , Proteína 11 Similar a Bcl2 , Muerte Celular/genética , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/fisiología , Cuerpo Estriado/crecimiento & desarrollo , Cuerpo Estriado/fisiología , Femenino , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas/genética , Ácido Quinolínico/toxicidad , Regulación hacia Arriba/genética , Proteína Destructora del Antagonista Homólogo bcl-2/genética , Proteína X Asociada a bcl-2/genética
13.
Neuroscience ; 144(2): 462-71, 2007 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-17081696

RESUMEN

The striatum is one of the brain areas most vulnerable to excitotoxicity, a lesion that can be prevented by neurotrophins. In the present study, intrastriatal injection of the N-methyl-d-aspartate receptor (NMDAR) agonist quinolinate (QUIN) was performed in mice heterozygous for neurotrophin-3 (NT3 +/-) or brain-derived neurotrophic factor (BDNF +/-) to analyze the role of endogenous neurotrophins on the regulation of striatal neurons susceptibility to excitotoxic injury. QUIN injection induced a decrease in dopamine- and cyclic AMP-regulated phosphoprotein of 32 kDa (DARPP-32) protein levels that was higher in NT-3 +/- than in BDNF+/- or wild type animals. This enhanced susceptibility was specific for enkephalin- and tachykinin-positive projection neurons, and also for parvalbumin-positive interneurons. However the excitotoxic damage in large interneurons was not modified in NT-3 +/- mice compared with wild type animals. This effect can be related to the regulation of NMDARs by endogenous NT-3. Thus, our results show that there is an age-dependent regulation of NMDAR subunits NR1 and NR2A, but not NR2B, in NT-3 +/- mice. The deficit of endogenous NT-3 induced a decrease in NR1 and NR2A subunits at postnatal day (P) 0 and P3 mice respectively, whereas an upregulation was observed in 12 week old NT-3 +/- mice. This differential effect was also observed after administration of exogenous NT-3. In primary striatal cultures, NT-3 treatment induced an enhancement in NR2A, but not NR2B, protein levels. However, intrastriatal grafting of NT-3 secreting-cells in adult wild type mice produced a down-regulation of NR2A subunit. In conclusion, NT-3 regulates the expression of NMDAR subunits modifying striatal neuronal properties that confers the differential vulnerability to excitotoxicity in projection neurons and interneurons in the striatum.


Asunto(s)
Cuerpo Estriado/metabolismo , Regulación de la Expresión Génica/fisiología , Neurotrofina 3/fisiología , Receptores de N-Metil-D-Aspartato/metabolismo , Análisis de Varianza , Animales , Factor Neurotrófico Derivado del Encéfalo/deficiencia , Recuento de Células/métodos , Trasplante de Células , Células Cultivadas , Cuerpo Estriado/lesiones , Cuerpo Estriado/patología , Aminoácidos Excitadores/toxicidad , Fibroblastos/metabolismo , Fibroblastos/trasplante , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neurotrofina 3/deficiencia , Ácido Quinolínico/toxicidad , Ratas , Ratas Endogámicas F344 , Receptores de N-Metil-D-Aspartato/genética , Transfección/métodos , Trasplante Heterólogo , Ácido gamma-Aminobutírico/metabolismo
14.
Gene Ther ; 14(2): 118-28, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16943855

RESUMEN

The use of stem cells for reconstructive or neuroprotective strategies can benefit from new advances in neuroimaging techniques to track grafted cells. In the present work, we analyze the potential of a neural stem cell (NSC) line, which stably expresses the glial cell line-derived neurotrophic factor (GDNF) and the firefly luciferase gene (GDNF/Luc-NSC), for cell therapy in a Huntington's disease mouse model. Our results show that detection of light photons is an effective method to quantify the proliferation rate and to characterize the migration pathways of transplanted NSCs. Intravenous administration of luciferine, the luciferase substract, into the grafted animals allowed the detection of implanted cells in real time by an optical neuroimaging methodology, overpassing the limits of serial histological analyses. We observed that transplanted GDNF/Luc-NSCs survive after grafting and expand more when transplanted in quinolinate-lesioned nude mouse striata than when transplanted in non-lesioned mice. We also demonstrate that GDNF/Luc-NSCs prevent the degeneration of striatal neurons in the excitotoxic mouse model of Huntington's disease and reduce the amphetamine-induced rotational behavior in mice bearing unilateral lesions.


Asunto(s)
Cuerpo Estriado/patología , Terapia Genética/métodos , Factor Neurotrófico Derivado de la Línea Celular Glial/uso terapéutico , Enfermedad de Huntington/terapia , Fármacos Neuroprotectores/uso terapéutico , Trasplante de Células Madre/métodos , Animales , Conducta Animal/efectos de los fármacos , Recuento de Células , Cuerpo Estriado/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Luciferasas/administración & dosificación , Ratones , Ratones Desnudos , Células Madre/metabolismo , Células Madre/patología , Transducción Genética/métodos
15.
Neurosci Lett ; 406(3): 270-5, 2006 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-16934924

RESUMEN

We assessed the effect of glatiramer acetate (GA) on the immunophenotypic and cytokine profile and the BDNF production by peripheral blood mononuclear cells, and their association with the clinical response in 19 naïve-treated MS patients prospectively followed-up after GA therapy. Two patients withdrew the therapy. After a median follow-up of 21 months, twelve were considered responders and five as non-responders. Non-responder patients had significant longer disease duration and a higher EDSS score at baseline. In the responder group, a significant decrease in the percentage of INF-gamma producing total lymphocytes, CD4+ and CD8+ T cells, and reduced percentage of IL-2 producing CD4+ and CD8+ T cells were observed at 12, 18 and 24 months. These changes were associated with a significant increase in the percentage of CD3+, CD4+ and CD4(+) CD45RA(+) T cells, and BDNF production from month 6 that remained significant throughout the study. We did not observe significant changes in the nonresponder group for any of the parameters studied. Our data suggest that GA treatment induces a downmodulation of proinflammatory cytokines associated with the regulation of the peripheral T cell compartment and with increased production of BDNF that might be related to the clinical response.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Citocinas/metabolismo , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/metabolismo , Péptidos/farmacología , Adyuvantes Inmunológicos/uso terapéutico , Adulto , Femenino , Citometría de Flujo/métodos , Acetato de Glatiramer , Humanos , Estudios Longitudinales , Activación de Linfocitos/efectos de los fármacos , Masculino , Persona de Mediana Edad , Esclerosis Múltiple/tratamiento farmacológico , Péptidos/uso terapéutico , Subgrupos de Linfocitos T/efectos de los fármacos , Factores de Tiempo
16.
Histol Histopathol ; 21(11): 1217-32, 2006 11.
Artículo en Inglés | MEDLINE | ID: mdl-16874665

RESUMEN

Neurodegenerative disorders affecting the central nervous system, such as Alzheimer's disease, Parkinson's disease, Huntington's chorea (HD) and amyotrophic lateral sclerosis are characterized by the loss of selected neuronal populations. Another striking feature shared by these diseases is the deposition of proteinaceous inclusion bodies in the brain, which may be intracytoplasmatic or intranuclear, or even extracellular. However, the density and prevalence of aggregates are not always directly related to neurodegeneration. Although some of these diseases are the result of mutations in known proteins, with HD a clear example, the expression and location of the affected protein do not explain the selective neurodegeneration. Therefore, other intrinsic mechanisms, characteristic of each neuronal population, might be involved in the neurodegenerative process. In this review we focus on several proposed mechanisms such as excitotoxicity, mitochondrial dysfunction and altered expression of trophic factors, which could account for the pathogenesis of HD.


Asunto(s)
Enfermedad de Huntington/patología , Interneuronas/patología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Cuerpo Estriado/patología , Citoplasma/metabolismo , Humanos , Proteína Huntingtina , Mitocondrias/metabolismo , Mitocondrias/patología , Modelos Biológicos , Mutación , Degeneración Nerviosa/patología , Proteínas del Tejido Nervioso/genética , Enfermedades Neurodegenerativas/patología , Neuronas/metabolismo , Proteínas Nucleares/genética
18.
Mol Ther ; 12(2): 283-9, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15935736

RESUMEN

The present paper investigates the long-term functionality of an ex vivo gene therapy approach based on cell microencapsulation for the continuous delivery of erythropoietin (EPO) without implementation of immunosuppressive protocols. Polymer microcapsules (0.5 ml) loaded with EPO-secreting C(2)C(12) myoblasts and releasing 15,490 +/- 600 IU EPO/24 h were implanted in the peritoneum and subcutaneous tissue of syngeneic and allogeneic mice. High and constant hematocrit levels were maintained for more than 100 days in all implanted mice. Capsules retrieved from the peritoneum were free-floating or forming small capsule clusters, and we detected only a weak fibroblast outgrowth in capsules adhered to organs, whereas capsules explanted from the subcutaneous region appeared altogether as a richly vascularized structure with no signs of major host reaction. Interestingly, the functionality of capsules implanted in the allogeneic mice persisted until day 210 after implantation. These results highlight the feasibility of cell encapsulation technology for the long-term delivery of EPO independent of the method of administration and the mouse strain.


Asunto(s)
Materiales Biocompatibles Revestidos , Eritropoyetina/administración & dosificación , Mioblastos/trasplante , Animales , Cápsulas , Línea Celular , Eritropoyetina/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H
19.
Neurosci Lett ; 380(1-2): 122-6, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15854763

RESUMEN

A neuroprotective role of inflammation has been suggested based on that immune cells are the main source of brain-derived neurotrophic factor (BDNF). We investigated the 3-year evolution of BDNF levels in serum, CSF and culture supernatant of peripheral blood mononuclear cells (PBMC), unstimulated and stimulated with anti-CD3 and soluble anti-CD28 antibodies, in 14 multiple sclerosis patients who underwent an autologous hematopoietic stem cell transplantation (AHSCT). BDNF levels were correlated with previously reported MRI measures that showed a reduction of T2 lesion load and increased brain atrophy, mainly at first year post-transplant. A significant decrease of serum BDNF levels was seen at 12 months post-transplant. BDNF values were found significantly lower in stimulated but not in unstimulated PBMC supernatants during the follow-up, supporting that AHSCT may induce a down-regulation of BDNF production. The only significant correlation was found between CSF BDNF levels and T2 lesion load before and 1 year after AHSCT, suggesting that BDNF reflects the past and ongoing inflammatory activity and demyelination of these highly active patients. Our study suggests that AHSCT can reduce BDNF levels to values associated with lower activity. This decrease does not seem to correlate with the brain atrophy measures observed in the MRI.


Asunto(s)
Evolución Biológica , Factor Neurotrófico Derivado del Encéfalo , Encéfalo/metabolismo , Trasplante de Células Madre Hematopoyéticas , Esclerosis Múltiple/terapia , Anticuerpos/farmacología , Encéfalo/patología , Factor Neurotrófico Derivado del Encéfalo/sangre , Factor Neurotrófico Derivado del Encéfalo/líquido cefalorraquídeo , Antígenos CD28/inmunología , Complejo CD3/inmunología , Células Cultivadas , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática/métodos , Estudios de Seguimiento , Humanos , Leucocitos/efectos de los fármacos , Leucocitos/metabolismo , Imagen por Resonancia Magnética/métodos , Esclerosis Múltiple/sangre , Esclerosis Múltiple/líquido cefalorraquídeo , Estadística como Asunto , Factores de Tiempo
20.
J Neurosci Res ; 70(5): 638-44, 2002 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-12424731

RESUMEN

Bone morphogenetic proteins (BMPs) are a set of members of the transforming growth factor-beta superfamily recently described as promoting the differentiation of several neuronal populations within the basal ganglia. This study examined whether a member of this family, BMP-6, could exert neurotrophic effects on the neurons of the striatum, in which BMP-6 mRNA had been previously detected during development. Here we show that BMP-6 increases the number and differentiation of calbindin-positive neurons in vitro. Indeed, BMP-6 increased the total area, the perimeter, and the degree of arborization of this neuronal population. This trophic factor promoted dendritic growth without modifying axonal length or soma area. Furthermore, BMP-6 increased the number of glial fibrillary acidic protein-positive cells while decreasing the number of nestin-positive cells. The suppression of cell proliferation or glial development by the antimitotic fluorodeoxyuridine removed the effects on striatal neurons, suggesting the involvement of astroglial cells in the differentiation induced by BMP-6. The current results confirm the relevance of BMPs in the development of the striatum and emphasize the crucial importance of the trophic interaction between glial and neuronal cells.


Asunto(s)
Proteínas Morfogenéticas Óseas/farmacología , Neuronas/química , Neuronas/efectos de los fármacos , Proteína G de Unión al Calcio S100/análisis , Animales , Astrocitos/citología , Biomarcadores , Proteína Morfogenética Ósea 6 , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Calbindinas , Comunicación Celular , División Celular/efectos de los fármacos , Células Cultivadas , Cuerpo Estriado/citología , Dendritas/efectos de los fármacos , Dendritas/fisiología , Femenino , Técnicas In Vitro , Neuronas/ultraestructura , Embarazo , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA