Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Cell ; 145(6): 813-4, 2011 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-21663784

RESUMEN

Abnormalities in the kynurenine pathway are associated with neurodegenerative disorders. Zwilling et al. (2011) show that inhibition of kynurenine 3-monooxygenase in the body's periphery leads to an increase in kyneuric acid, a neuroprotective compound, in the brain. This intervention ameliorates neurodegeneration in mouse models of Alzheimer's disease and Huntington's disease.

2.
J Neurosci ; 33(16): 7020-6, 2013 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-23595759

RESUMEN

BACE1 is the rate-limiting enzyme that cleaves amyloid precursor protein (APP) to produce the amyloid ß peptides that accumulate in Alzheimer's disease (AD). BACE1, which is elevated in AD patients and APP transgenic mice, also cleaves the ß2-subunit of voltage-gated sodium channels (Navß2). Although increased BACE1 levels are associated with Navß2 cleavage in AD patients, whether Navß2 cleavage occurs in APP mice had not yet been examined. Such a finding would be of interest because of its potential impact on neuronal activity: previous studies demonstrated that BACE1-overexpressing mice exhibit excessive cleavage of Navß2 and reduced sodium current density, but the phenotype associated with loss of function mutations in either Navß-subunits or pore-forming α-subunits is epilepsy. Because mounting evidence suggests that epileptiform activity may play an important role in the development of AD-related cognitive deficits, we examined whether enhanced cleavage of Navß2 occurs in APP transgenic mice, and whether it is associated with aberrant neuronal activity and cognitive deficits. We found increased levels of BACE1 expression and Navß2 cleavage fragments in cortical lysates from APP transgenic mice, as well as associated alterations in Nav1.1α expression and localization. Both pyramidal neurons and inhibitory interneurons exhibited evidence of increased Navß2 cleavage. Moreover, the magnitude of alterations in sodium channel subunits was associated with aberrant EEG activity and impairments in the Morris water maze. Together, these results suggest that altered processing of voltage-gated sodium channels may contribute to aberrant neuronal activity and cognitive deficits in AD.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/patología , Neuronas/metabolismo , Canales de Sodio/metabolismo , Enfermedad de Alzheimer/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Ácido Aspártico Endopeptidasas/metabolismo , Biotinilación , Modelos Animales de Enfermedad , Electroencefalografía , Regulación de la Expresión Génica/genética , Glutamato Descarboxilasa/metabolismo , Humanos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Transgénicos , Mutación/genética , Canal de Sodio Activado por Voltaje NAV1.1/genética , Canal de Sodio Activado por Voltaje NAV1.1/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/patología , Neuropéptido Y/genética , Neuropéptido Y/metabolismo
3.
Nat Neurosci ; 11(3): 334-43, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18297067

RESUMEN

Estrogens have long been implicated in influencing cognitive processes, yet the molecular mechanisms underlying these effects and the roles of the estrogen receptors alpha (ERalpha) and beta (ERbeta) remain unclear. Using pharmacological, biochemical and behavioral techniques, we demonstrate that the effects of estrogen on hippocampal synaptic plasticity and memory are mediated through ERbeta. Selective ERbeta agonists increased key synaptic proteins in vivo, including PSD-95, synaptophysin and the AMPA-receptor subunit GluR1. These effects were absent in ERbeta knockout mice. In hippocampal slices, ERbeta activation enhanced long-term potentiation, an effect that was absent in slices from ERbeta knockout mice. ERbeta activation induced morphological changes in hippocampal neurons in vivo, including increased dendritic branching and increased density of mushroom-type spines. An ERbeta agonist, but not an ERalpha agonist, also improved performance in hippocampus-dependent memory tasks. Our data suggest that activation of ERbeta can regulate hippocampal synaptic plasticity and improve hippocampus-dependent cognition.


Asunto(s)
Receptor beta de Estrógeno/metabolismo , Estrógenos/metabolismo , Hipocampo/metabolismo , Memoria/fisiología , Plasticidad Neuronal/fisiología , Neuronas/metabolismo , Animales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/efectos de los fármacos , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/metabolismo , Estradiol/metabolismo , Estradiol/farmacología , Receptor beta de Estrógeno/agonistas , Receptor beta de Estrógeno/genética , Estrógenos/agonistas , Estrógenos/farmacología , Femenino , Hipocampo/citología , Hipocampo/efectos de los fármacos , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/fisiología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plasticidad Neuronal/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos , Técnicas de Cultivo de Órganos , Ovariectomía , Fosforilación/efectos de los fármacos , Ratas , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología
4.
Neurobiol Dis ; 43(1): 248-56, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21458569

RESUMEN

Huntington's disease (HD) is a late-onset, neurodegenerative disease for which there are currently no cures nor disease-modifying treatments. Here we report the identification of several potential anti-inflammatory targets for HD using an ex vivo model of HD that involves the acute transfection of human mutant huntingtin-based constructs into rat brain slices. This model recapitulates key components of the human disease, including the formation of intracellular huntingtin protein (HTT)-containing inclusions and the progressive neurodegeneration of striatal neurons-both occurring within the native tissue context of these neurons. Using this "high-throughput biology" screening platform, we conducted a hypothesis-neutral screen of a collection of drug-like compounds which identified several anti-inflammatory targets that provided neuroprotection against HTT fragment-induced neurodegeneration. The nature of these targets provide further support for non-cell autonomous mechanisms mediating significant aspects of neuropathogenesis induced by mutant HTT fragment proteins.


Asunto(s)
Antiinflamatorios no Esteroideos/metabolismo , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Enfermedad de Huntington/tratamiento farmacológico , Degeneración Nerviosa/tratamiento farmacológico , Animales , Animales Recién Nacidos , Antiinflamatorios no Esteroideos/farmacología , Cuerpo Estriado/patología , Evaluación Preclínica de Medicamentos/métodos , Humanos , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/patología , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Fármacos Neuroprotectores/metabolismo , Fármacos Neuroprotectores/farmacología , Técnicas de Cultivo de Órganos , Ratas , Ratas Sprague-Dawley
5.
Proc Natl Acad Sci U S A ; 105(1): 33-8, 2008 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-18162540

RESUMEN

Rapamycin is an immunosuppressive immunophilin ligand reported as having neurotrophic activity. We show that modification of rapamycin at the mammalian target of rapamycin (mTOR) binding region yields immunophilin ligands, WYE-592 and ILS-920, with potent neurotrophic activities in cortical neuronal cultures, efficacy in a rodent model for ischemic stroke, and significantly reduced immunosuppressive activity. Surprisingly, both compounds showed higher binding selectivity for FKBP52 versus FKBP12, in contrast to previously reported immunophilin ligands. Affinity purification revealed two key binding proteins, the immunophilin FKBP52 and the beta1-subunit of L-type voltage-dependent Ca(2+) channels (CACNB1). Electrophysiological analysis indicated that both compounds can inhibit L-type Ca(2+) channels in rat hippocampal neurons and F-11 dorsal root ganglia (DRG)/neuroblastoma cells. We propose that these immunophilin ligands can protect neurons from Ca(2+)-induced cell death by modulating Ca(2+) channels and promote neurite outgrowth via FKBP52 binding.


Asunto(s)
Canales de Calcio/química , Sirolimus/química , Proteínas de Unión a Tacrolimus/química , Animales , Calcio/metabolismo , Electrofisiología/métodos , Humanos , Inmunofilinas/metabolismo , Inmunosupresores/farmacología , Ligandos , Modelos Químicos , Neuritas/metabolismo , Neuroblastoma/metabolismo , Neuronas/metabolismo , Técnicas de Placa-Clamp , Unión Proteica , Ratas , Accidente Cerebrovascular/metabolismo
6.
Proc Natl Acad Sci U S A ; 105(25): 8754-9, 2008 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-18559859

RESUMEN

The amyloid hypothesis states that a variety of neurotoxic beta-amyloid (Abeta) species contribute to the pathogenesis of Alzheimer's disease. Accordingly, a key determinant of disease onset and progression is the appropriate balance between Abeta production and clearance. Enzymes responsible for the degradation of Abeta are not well understood, and, thus far, it has not been possible to enhance Abeta catabolism by pharmacological manipulation. We provide evidence that Abeta catabolism is increased after inhibition of plasminogen activator inhibitor-1 (PAI-1) and may constitute a viable therapeutic approach for lowering brain Abeta levels. PAI-1 inhibits the activity of tissue plasminogen activator (tPA), an enzyme that cleaves plasminogen to generate plasmin, a protease that degrades Abeta oligomers and monomers. Because tPA, plasminogen and PAI-1 are expressed in the brain, we tested the hypothesis that inhibitors of PAI-1 will enhance the proteolytic clearance of brain Abeta. Our data demonstrate that PAI-1 inhibitors augment the activity of tPA and plasmin in hippocampus, significantly lower plasma and brain Abeta levels, restore long-term potentiation deficits in hippocampal slices from transgenic Abeta-producing mice, and reverse cognitive deficits in these mice.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Fibrinolisina/metabolismo , Fibrinolíticos/metabolismo , Animales , Humanos , Inmunohistoquímica , Ratones , Ratones Transgénicos , Inactivadores Plasminogénicos/metabolismo , Activador de Tejido Plasminógeno/antagonistas & inhibidores , Activador de Tejido Plasminógeno/metabolismo
7.
Neurobiol Dis ; 39(3): 311-7, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20451607

RESUMEN

The c-Jun N-terminal kinase (JNK) pathway potentially links together the three major pathological hallmarks of Alzheimer's disease (AD): development of amyloid plaques, neurofibrillary tangles, and brain atrophy. As activation of the JNK pathway has been observed in amyloid models of AD in association with peri-plaque regions and neuritic dystrophy, as we confirm here for Tg2576/PS(M146L) transgenic mice, we directly tested whether JNK inhibition could provide neuroprotection in a novel brain slice model for amyloid precursor protein (APP)-induced neurodegeneration. We found that APP/amyloid beta (Abeta)-induced neurodegeneration is blocked by both small molecule and peptide inhibitors of JNK, and provide evidence that this neuroprotection occurs downstream of APP/Abeta production and processing. Our findings demonstrate that Abeta can induce neurodegeneration, at least in part, through the JNK pathway and suggest that inhibition of JNK may be of therapeutic utility in the treatment of AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Degeneración Nerviosa/prevención & control , Neuronas/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Análisis de Varianza , Animales , Western Blotting , Encéfalo/patología , Modelos Animales de Enfermedad , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Ratones Transgénicos , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Ovillos Neurofibrilares/metabolismo , Ovillos Neurofibrilares/patología , Neuronas/patología , Ratas , Ratas Sprague-Dawley , Transducción de Señal
9.
Neurodegener Dis ; 7(1-3): 153-9, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20197696

RESUMEN

BACKGROUND: Parkinson's disease (PD) is a progressive neurodegenerative condition characterized by an increasing loss of dopaminergic neurons resulting in motor dysfunction. However, cognitive impairments in PD patients are a common clinical feature that has gained increased attention. OBJECTIVE: The purpose of the current study was to evaluate the effects of an MPTP-induced dopaminergic lesion in mice on social odor recognition (SOR) memory. METHODS: Mice were acutely treated with MPTP and evaluated for memory impairments in the SOR assay and characterized using biochemical and immunohistochemical methods approximately 2 weeks later. RESULTS: Here we demonstrate that SOR memory is sensitive to MPTP treatment and that it correlates with multiple measures of nigrostriatal integrity. MPTP treatment of C57BL/6N mice produced a profound decrease in dopamine levels, dopamine transporter binding and tyrosine hydroxylase immunoreactivity in the striatum. These impairments in stratial dopaminergic function were blocked by pretreatment with the MAO-B inhibitor deprenyl. Changes in the dopaminergic system parallel those observed in SOR with MPTP treatment impairing recognition memory in the absence of a deficit in odor discrimination during learning. Deprenyl pretreatment blocked the MPTP-induced impairment of SOR memory. CONCLUSION: The use of the SOR memory model may provide a preclinical method for evaluating cognitive therapies for PD.


Asunto(s)
Intoxicación por MPTP/complicaciones , Intoxicación por MPTP/psicología , Trastornos de la Memoria/etiología , Reconocimiento en Psicología/fisiología , Predominio Social , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacología , Análisis de Varianza , Animales , Modelos Animales de Enfermedad , Dopamina/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Conducta Exploratoria/efectos de los fármacos , Intoxicación por MPTP/inducido químicamente , Intoxicación por MPTP/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Sustancia Negra/efectos de los fármacos , Tirosina 3-Monooxigenasa/metabolismo
10.
J Neurosci ; 28(45): 11445-53, 2008 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-18987181

RESUMEN

Inheritance of the apoE4 allele (epsilon4) increases the risk of developing Alzheimer's disease; however, the mechanisms underlying this association remain elusive. Recent data suggest that inheritance of epsilon4 may lead to reduced apoE protein levels in the CNS. We therefore examined apoE protein levels in the brains, CSF and plasma of epsilon2/2, epsilon3/3, and epsilon4/4 targeted replacement mice. These apoE mice showed a genotype-dependent decrease in apoE levels; epsilon2/2 >epsilon3/3 >epsilon4/4. Next, we sought to examine the relative contributions of apoE4 and apoE3 in the epsilon3/4 mouse brains. ApoE4 represented 30-40% of the total apoE. Moreover, the absolute amount of apoE3 per allele was similar between epsilon3/3 and epsilon3/4 mice, implying that the reduced levels of total apoE in epsilon3/4 mice can be explained by the reduction in apoE4 levels. In culture medium from epsilon3/4 human astrocytoma or epsilon3/3, epsilon4/4 and epsilon3/4 primary astrocytes, apoE4 levels were consistently lower than apoE3. Secreted cholesterol levels were also lower from epsilon4/4 astrocytes. Pulse-chase experiments showed an enhanced degradation and reduced half-life of newly synthesized apoE4 compared with apoE3. Together, these data suggest that astrocytes preferentially degrade apoE4, leading to reduced apoE4 secretion and ultimately to reduced brain apoE levels. Moreover, the genotype-dependent decrease in CNS apoE levels, mirror the relative risk of developing AD, and suggest that low levels of total apoE exhibited by epsilon4 carriers may directly contribute to the disease progression, perhaps by reducing the capacity of apoE to promote synaptic repair and/or Abeta clearance.


Asunto(s)
Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Encéfalo/metabolismo , Polimorfismo Genético , Análisis de Varianza , Animales , Animales Recién Nacidos , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Análisis Mutacional de ADN , Relación Dosis-Respuesta a Droga , Humanos , Hidrocarburos Fluorados/farmacología , Masculino , Ratones , Ratones Transgénicos , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , ARN Mensajero/metabolismo , Sulfonamidas/farmacología , Factores de Tiempo
11.
J Neurochem ; 108(6): 1539-49, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19183261

RESUMEN

The molecular determinants of Sonic Hedgehog (Shh) signaling in mammalian cells and, in particular, those of the CNS are unclear. Here we report that primary cortical astrocyte cultures are highly responsive to both Shh protein and Hh Agonist 1.6, a selective, small molecule Smoothened agonist. Both agonists produced increases in mRNA expression of Shh-regulated gene targets, Gli-1 and Patched in a cyclopamine- and forskolin-sensitive manner. Using this model we show for the first time that Shh pathway activation mediates rapid increases in p38 MAPK phosphorylation, without altering phosphorylation of either extracellular-signal-regulated kinases or c-jun N-terminal kinases. Selective inhibition of p38 MAPK significantly attenuated Shh-dependent up-regulation of Gli-1, inter-alpha trypsin inhibitor and thrombomodulin mRNA, however did not affect expression of insulin-like growth factor 2 or a novel Shh target, membrane-associated guanylate kinase p55 subfamily member 6. Using RNAi and a constitutively-active mutant we show that Shh signaling to p38 MAPK and subsequent Gli-1 transcription requires G-protein receptor kinase 2. Taken together, these findings provide evidence for a central role of G-protein receptor kinase 2-dependent p38 MAPK activity in regulating Shh-mediated gene transcription in astrocytes.


Asunto(s)
Astrocitos/metabolismo , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Proteínas Hedgehog/metabolismo , Transducción de Señal/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Ratas , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transfección/métodos
12.
J Pharmacol Exp Ther ; 331(2): 598-608, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19671883

RESUMEN

The presenilin containing gamma-secretase complex is responsible for the regulated intramembraneous proteolysis of the amyloid precursor protein (APP), the Notch receptor, and a multitude of other substrates. gamma-Secretase catalyzes the final step in the generation of Abeta(40) and Abeta(42) peptides from APP. Amyloid beta-peptides (Abeta peptides) aggregate to form neurotoxic oligomers, senile plaques, and congophilic angiopathy, some of the cardinal pathologies associated with Alzheimer's disease. Although inhibition of this protease acting on APP may result in potentially therapeutic reductions of neurotoxic Abeta peptides, nonselective inhibition of the enzyme may cause severe adverse events as a result of impaired Notch receptor processing. Here, we report the preclinical pharmacological profile of GSI-953 (begacestat), a novel thiophene sulfonamide gamma-secretase inhibitor (GSI) that selectively inhibits cleavage of APP over Notch. This GSI inhibits Abeta production with low nanomolar potency in cellular and cell-free assays of gamma-secretase function, and displaces a tritiated analog of GSI-953 from enriched gamma-secretase enzyme complexes with similar potency. Cellular assays of Notch cleavage reveal that this compound is approximately 16-fold selective for the inhibition of APP cleavage. In the human APP-overexpressing Tg2576 transgenic mouse, treatment with this orally active compound results in a robust reduction in brain, plasma, and cerebral spinal fluid Abeta levels, and a reversal of contextual fear-conditioning deficits that are correlated with Abeta load. In healthy human volunteers, oral administration of a single dose of GSI-953 produces dose-dependent changes in plasma Abeta levels, confirming pharmacodynamic activity of GSI-953 in humans.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Sulfonamidas/farmacología , Tiofenos/farmacología , Adolescente , Adulto , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Unión Competitiva , Células CHO , Línea Celular , Cricetinae , Cricetulus , Perros , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/toxicidad , Miedo/psicología , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Ratas , Ratas Sprague-Dawley , Receptores Notch/fisiología , Transducción de Señal/efectos de los fármacos , Sulfonamidas/farmacocinética , Sulfonamidas/toxicidad , Tiofenos/farmacocinética , Tiofenos/toxicidad , Adulto Joven
13.
Cell Chem Biol ; 25(11): 1389-1402.e9, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30197194

RESUMEN

α-Synuclein (αSN) aggregation is central to the etiology of Parkinson's disease (PD). Large-scale screening of compounds to identify aggregation inhibitors is challenged by stochastic αSN aggregation and difficulties in detecting early-stage oligomers (αSOs). We developed a high-throughput screening assay combining SDS-stimulated αSN aggregation with FRET to reproducibly detect initial stages in αSN aggregation. We screened 746,000 compounds, leading to 58 hits that markedly inhibit αSN aggregation and reduce αSOs' membrane permeabilization activity. The most effective aggregation inhibitors were derivatives of (4-hydroxynaphthalen-1-yl)sulfonamide. They interacted strongly with the N-terminal part of monomeric αSN and reduced αSO-membrane interactions, possibly by affecting electrostatic interactions. Several compounds reduced αSO toxicity toward neuronal cell lines. The inhibitors introduced chemical modifications of αSN that were, however, not a prerequisite for inhibitory activity. We also identified several phenyl-benzoxazol compounds that promoted αSN aggregation (proaggregators). These compounds may be useful tools to modulate αSN aggregation in cellula.


Asunto(s)
Amiloide/química , Benzoxazoles/química , Benzoxazoles/farmacología , Agregado de Proteínas/efectos de los fármacos , alfa-Sinucleína/química , Amiloide/antagonistas & inhibidores , Amiloide/ultraestructura , Transferencia Resonante de Energía de Fluorescencia/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Conformación Proteica/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , alfa-Sinucleína/antagonistas & inhibidores , alfa-Sinucleína/ultraestructura
14.
J Neurosci ; 22(5): 1840-9, 2002 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-11880513

RESUMEN

Voltage-dependent large-conductance Ca2+-activated K+ channels (BK channels) are widely expressed in excitable and nonexcitable cells. BK channels exhibit diverse electrophysiological properties, which are attributable in part to alternative splicing of their alpha-subunits. BK currents have been implicated in the growth control of glial cells, and BK channels with novel biophysical properties have recently been characterized in human glioma cells. Here we report the isolation, cloning, and functional characterization of glioma BK (gBK), a novel splice isoform of hSlo, the gene that encodes the alpha-subunits of human BK channels. The primary sequence of gBK is 97% identical to its closest homolog hbr5, but it contains an additional 34-amino-acid exon at splice site 2 in the C-terminal tail of BK channels. hSlo transcripts containing this novel exon are expressed ubiquitously in various normal tissues as well as in neoplasmic samples, suggesting that the novel exon may modulate important physiological functions of BK channels. Expression of gBK in Xenopus oocytes gives rise to iberiotoxin-sensitive (IbTX) currents, with an IC(50) for IbTX of 5.7 nm and a Hill coefficient of 0.76. Single gBK channels have a unitary conductance of similar250 pS, and the currents show significantly slower activation and higher Ca2+ sensitivity than hbr5. Ca2+ sensitivity was enhanced specifically at physiologically relevant [Ca2+]i (100-500 nm). Examination of biopsies from patients with malignant gliomas has revealed specific overexpression of BK channels in gliomas compared with nonmalignant human cortical tissues. Importantly, tumor malignancy grades have correlated positively with BK channel expression, suggesting an important role for the gBK channel in glioma biology.


Asunto(s)
Astrocitoma/metabolismo , Glioblastoma/metabolismo , Glioma/metabolismo , Canales de Potasio Calcio-Activados/genética , Empalme Alternativo , Animales , Calcio/metabolismo , Clonación Molecular , Glioma/patología , Humanos , Immunoblotting , Cinética , Canales de Potasio de Gran Conductancia Activados por el Calcio , Microinyecciones , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Especificidad de Órganos , Técnicas de Placa-Clamp , Péptidos/farmacología , Reacción en Cadena de la Polimerasa , Canales de Potasio Calcio-Activados/antagonistas & inhibidores , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , ARN Complementario/genética , ARN Complementario/metabolismo , Análisis de Secuencia de ADN , Células Tumorales Cultivadas , Regulación hacia Arriba , Xenopus
15.
J Neurosci ; 24(37): 7999-8008, 2004 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-15371500

RESUMEN

Transcriptional dysregulation by mutant huntingtin (Htt) protein has been implicated in the pathogenesis of Huntington's disease (HD). We find that cultured cells expressing mutant Htt and striatal cells from HD transgenic mice have elevated nuclear factor-kappaB (NF-kappaB) activity. Furthermore, NF-kappaB is concentrated in the nucleus of neurons in the brains of HD transgenic mice. In inducible PC12 cells and in HD transgenic mice, mutant Htt activates the IkappaB kinase complex (IKK), a key regulator of NF-kappaB. Activation of IKK is likely mediated by direct interaction with mutant Htt, because the expanded polyglutamine stretch and adjacent proline-rich motifs in mutant Htt interact with IKKgamma, a regulatory subunit of IKK. Activation of IKK may also influence the toxicity of mutant Htt, because expression of IKKgamma promotes aggregation and nuclear localization of mutant Htt exon-1. Moreover, in acute striatal slice cultures, inhibition of IKK activity with an N-terminally truncated form of IKKgamma blocks mutant Htt-induced toxicity in medium-sized spiny neurons (MSNs). In addition, blocking degradation of NF-kappaB inhibitors with a dominant-negative ubiquitin ligase beta-transducin repeat-containing protein also reduces the toxicity of mutant Htt in MSNs. Therefore, aberrant NF-kappaB activation may contribute to the neurodegeneration induced by mutant Htt.


Asunto(s)
Regulación de la Expresión Génica/fisiología , FN-kappa B/fisiología , Proteínas del Tejido Nervioso/fisiología , Proteínas Nucleares/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Secuencias de Aminoácidos , Animales , Biolística , Línea Celular , Línea Celular Transformada , Cuerpo Estriado/citología , Activación Enzimática , Exones/genética , Regulación de la Expresión Génica/efectos de los fármacos , Genes Reporteros , Humanos , Proteína Huntingtina , Quinasa I-kappa B , Interleucina-1/farmacología , Riñón , Ratones , Ratones Transgénicos , Repeticiones de Minisatélite , FN-kappa B/antagonistas & inhibidores , Degeneración Nerviosa , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/química , Proteínas Nucleares/genética , Células PC12 , Fosforilación , Unión Proteica , Mapeo de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/genética , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes de Fusión/análisis , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/fisiología , Transfección , Ubiquitina-Proteína Ligasas/fisiología
16.
Int J Alzheimers Dis ; 2010: 864625, 2010 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-20981353

RESUMEN

Mice transgenic for production of excessive or mutant forms of beta-amyloid differ from patients with Alzheimer's disease in the degree of inflammation, oxidative damage, and alteration of intermediary metabolism, as well as the paucity or absence of neuronal atrophy and cognitive impairment. Previous observers have suggested that differences in inflammatory response reflect a discrepancy in the state of the locus coeruleus (LC), loss of which is an early change in Alzheimer's disease but which is preserved in the transgenic mice. In this paper, we extend these observations by examining the effects of the LC on markers of oxidative stress and intermediary metabolism. We compare four groups: wild-type or Tg2576 Aß transgenic mice injected with DSP4 or vehicle. Of greatest interest were metabolites different between ablated and intact transgenics, but not between ablated and intact wild-type animals. The Tg2576_DSP4 mice were distinguished from the other three groups by oxidative stress and altered energy metabolism. These observations provide further support for the hypothesis that Tg2576 Aß transgenic mice with this ablation may be a more congruent model of Alzheimer's disease than are transgenics with an intact LC.

17.
PLoS One ; 5(10): e13672, 2010 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-21060682

RESUMEN

Mutations in LRRK2 (leucine-rich repeat kinase 2) have been identified as major genetic determinants of Parkinson's disease (PD). The most prevalent mutation, G2019S, increases LRRK2's kinase activity, therefore understanding the sites and substrates that LRRK2 phosphorylates is critical to understanding its role in disease aetiology. Since the physiological substrates of this kinase are unknown, we set out to reveal potential targets of LRRK2 G2019S by identifying its favored phosphorylation motif. A non-biased screen of an oriented peptide library elucidated F/Y-x-T-x-R/K as the core dependent substrate sequence. Bioinformatic analysis of the consensus phosphorylation motif identified several novel candidate substrates that potentially function in neuronal pathophysiology. Peptides corresponding to the most PD relevant proteins were efficiently phosphorylated by LRRK2 in vitro. Interestingly, the phosphomotif was also identified within LRRK2 itself. Autophosphorylation was detected by mass spectrometry and biochemical means at the only F-x-T-x-R site (Thr 1410) within LRRK2. The relevance of this site was assessed by measuring effects of mutations on autophosphorylation, kinase activity, GTP binding, GTP hydrolysis, and LRRK2 multimerization. These studies indicate that modification of Thr1410 subtly regulates GTP hydrolysis by LRRK2, but with minimal effects on other parameters measured. Together the identification of LRRK2's phosphorylation consensus motif, and the functional consequences of its phosphorylation, provide insights into downstream LRRK2-signaling pathways.


Asunto(s)
Proteínas Serina-Treonina Quinasas/metabolismo , Línea Celular , Cromatografía Liquida , Electroforesis en Gel de Poliacrilamida , Guanosina Trifosfato/metabolismo , Humanos , Hidrólisis , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Fosforilación , Unión Proteica , Proteínas Serina-Treonina Quinasas/química , Transducción de Señal , Espectrometría de Masas en Tándem
18.
FEBS J ; 276(2): 466-78, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19076219

RESUMEN

Mutations in leucine-rich repeat kinase 2 (LRRK2) comprise the leading cause of autosomal dominant Parkinson's disease, with age of onset and symptoms identical to those of idiopathic forms of the disorder. Several of these pathogenic mutations are thought to affect its kinase activity, so understanding the roles of LRRK2, and modulation of its kinase activity,may lead to novel therapeutic strategies for treating Parkinson's disease. In this study, highly purified, baculovirus-expressed proteins have been used,for the first time providing large amounts of protein that enable a thorough enzymatic characterization of the kinase activity of LRRK2.Although LRRK2 undergoes weak autophosphorylation, it exhibits high activity towards the peptidic substrate LRRKtide, suggesting that it is a catalytically efficient kinase. We have also utilized a time-resolved fluorescence resonance energy transfer (TR-FRET) assay format (Lantha-ScreenTM) to characterize LRRK2 and test the effects of nonselective kinase inhibitors. Finally, we have used both radiometric and TR-FRETassays to assess the role of clinical mutations affecting LRRK2's kinase activity. Our results suggest that only the most prevalent clinical mutation,G2019S, results in a robust enhancement of kinase activity with LRRKtideas the substrate. This mutation also affects binding of ATP to LRRK2,with wild-type binding being tighter (Km,app of 57 lm) than with theG2019S mutant (Km,app of 134 lm). Overall, these studies delineate the catalytic efficiency of LRRK2 as a kinase and provide strategies by which a therapeutic agent for Parkinson's disease may be identified.


Asunto(s)
Proteínas Serina-Treonina Quinasas/análisis , Proteínas Serina-Treonina Quinasas/metabolismo , Baculoviridae/genética , Activación Enzimática , Expresión Génica , Concentración 50 Inhibidora , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Mutación/genética , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Espectrometría de Fluorescencia , Factores de Tiempo
19.
Mol Cell Neurosci ; 34(4): 621-8, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17336088

RESUMEN

Recent studies show that intracellular cholesterol levels can modulate the processing of amyloid precursor protein to Abeta peptide. Moreover, cholesterol-rich apoE-containing lipoproteins may also promote Abeta clearance. Agonists of the liver X receptor (LXR) transcriptionally induce genes involved in intracellular lipid efflux and transport, including apoE. Thus, LXR agonists have the potential to both inhibit APP processing and promote Abeta clearance. Here we show that LXR agonist, TO901317, increased hippocampal ABCA1 and apoE and decreased Abeta42 levels in APP transgenic mice. TO901317 had no significant effects on levels of Abeta40, full length APP, or the APP processing products. Next, we examined the effects of TO901317 in the contextual fear conditioning paradigm; TO901317 completely reversed the contextual memory deficit in these mice. These data demonstrate that LXR agonists do not directly inhibit APP processing but rather facilitate the clearance of Abeta42 and may represent a novel therapeutic approach to Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/metabolismo , Proteínas de Unión al ADN/agonistas , Hipocampo/metabolismo , Memoria/efectos de los fármacos , Fragmentos de Péptidos/metabolismo , Receptores Citoplasmáticos y Nucleares/agonistas , Sulfonamidas/farmacología , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Apolipoproteínas E/metabolismo , Hipocampo/efectos de los fármacos , Humanos , Hidrocarburos Fluorados , Receptores X del Hígado , Masculino , Memoria/fisiología , Ratones , Ratones Transgénicos , Receptores Nucleares Huérfanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Proc Natl Acad Sci U S A ; 103(13): 5161-6, 2006 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-16549764

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative disorder for which numerous mouse models have been generated. In both AD patients and mouse models, there is increasing evidence that neuronal dysfunction occurs before the accumulation of beta-amyloid (Abeta)-containing plaques and neurodegeneration. Characterization of the timing and nature of preplaque dysfunction is important for understanding the progression of this disease and to identify pathways and molecular targets for therapeutic intervention. Hence, we have examined the progression of dysfunction at the morphological, functional, and behavioral levels in the Tg2576 mouse model of AD. Our data show that decreased dendritic spine density, impaired long-term potentiation (LTP), and behavioral deficits occurred months before plaque deposition, which was first detectable at 18 months of age. We detected a decrease in spine density in the outer molecular layer of the dentate gyrus (DG) beginning as early as 4 months of age. Furthermore, by 5 months, there was a decline in LTP in the DG after perforant path stimulation and impairment in contextual fear conditioning. Moreover, an increase in the Abeta42/Abeta40 ratio was first observed at these early ages. However, total amyloid levels did not significantly increase until approximately 18 months of age, at which time significant increases in reactive astrocytes and microglia could be observed. Overall, these data show that the perforant path input from the entorhinal cortex to the DG is compromised both structurally and functionally, and this pathology is manifested in memory defects long before significant plaque deposition.


Asunto(s)
Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Conducta Animal/fisiología , Sinapsis/patología , Envejecimiento/fisiología , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Miedo , Memoria , Ratones , Ratones Transgénicos , Tamaño de los Órganos , Columna Vertebral/patología , Sinapsis/metabolismo , Sinaptofisina/metabolismo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA