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













Base de datos
Intervalo de año de publicación
1.
Cell Death Dis ; 13(11): 959, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36379916

RESUMEN

Caspase-2 (Casp2) is a promising therapeutic target in several human diseases, including nonalcoholic steatohepatitis (NASH) and Alzheimer's disease (AD). However, the design of an active-site-directed inhibitor selective to individual caspase family members is challenging because caspases have extremely similar active sites. Here we present new peptidomimetics derived from the VDVAD pentapeptide structure, harboring non-natural modifications at the P2 position and an irreversible warhead. Enzyme kinetics show that these new compounds, such as LJ2 or its specific isomers LJ2a, and LJ3a, strongly and irreversibly inhibit Casp2 with genuine selectivity. In agreement with the established role of Casp2 in cellular stress responses, LJ2 inhibits cell death induced by microtubule destabilization or hydroxamic acid-based deacetylase inhibition. The most potent peptidomimetic, LJ2a, inhibits human Casp2 with a remarkably high inactivation rate (k3/Ki ~5,500,000 M-1 s-1), and the most selective inhibitor, LJ3a, has close to a 1000 times higher inactivation rate on Casp2 as compared to Casp3. Structural analysis of LJ3a shows that the spatial configuration of Cα at the P2 position determines inhibitor efficacy. In transfected human cell lines overexpressing site-1 protease (S1P), sterol regulatory element-binding protein 2 (SREBP2) and Casp2, LJ2a and LJ3a fully inhibit Casp2-mediated S1P cleavage and thus SREBP2 activation, suggesting a potential to prevent NASH development. Furthermore, in primary hippocampal neurons treated with ß-amyloid oligomers, submicromolar concentrations of LJ2a and of LJ3a prevent synapse loss, indicating a potential for further investigations in AD treatment.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Peptidomiméticos , Humanos , Caspasa 2/metabolismo , Caspasa 3/metabolismo , Neuronas/metabolismo , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Peptidomiméticos/farmacología , Peptidomiméticos/metabolismo
2.
J Neurosci ; 38(27): 6102-6113, 2018 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-29875265

RESUMEN

Activating Transcription Factor 4 (ATF4) has been postulated as a key regulator of learning and memory. We previously reported that specific hippocampal ATF4 downregulation causes deficits in synaptic plasticity and memory and reduction of glutamatergic functionality. Here we extend our studies to address ATF4's role in neuronal excitability. We find that long-term ATF4 knockdown in cultured rat hippocampal neurons significantly increases the frequency of spontaneous action potentials. This effect is associated with decreased functionality of metabotropic GABAB receptors (GABABRs). Knocking down ATF4 results in significant reduction of GABABR-induced GIRK currents and increased mIPSC frequency. Furthermore, reducing ATF4 significantly decreases expression of membrane-exposed, but not total, GABABR 1a and 1b subunits, indicating that ATF4 regulates GABABR trafficking. In contrast, ATF4 knockdown has no effect on surface expression of GABABR2s, several GABABR-coupled ion channels or ß2 and γ2 GABAARs. Pharmacologic manipulations confirmed the relationship between GABABR functionality and action potential frequency in our cultures. Specifically, the effects of ATF4 downregulation cited above are fully rescued by transcriptionally active, but not by transcriptionally inactive, shRNA-resistant, ATF4. We previously reported that ATF4 promotes stabilization of the actin-regulatory protein Cdc42 by a transcription-dependent mechanism. To test the hypothesis that this action underlies the mechanism by which ATF4 loss affects neuronal firing rates and GABABR trafficking, we downregulated Cdc42 and found that this phenocopies the effects of ATF4 knockdown on these properties. In conclusion, our data favor a model in which ATF4, by regulating Cdc42 expression, affects trafficking of GABABRs, which in turn modulates the excitability properties of neurons.SIGNIFICANCE STATEMENT GABAB receptors (GABABRs), the metabotropic receptors for the inhibitory neurotransmitter GABA, have crucial roles in controlling the firing rate of neurons. Deficits in trafficking/functionality of GABABRs have been linked to a variety of neurological and psychiatric conditions, including epilepsy, anxiety, depression, schizophrenia, addiction, and pain. Here we show that GABABRs trafficking is influenced by Activating Transcription Factor 4 (ATF4), a protein that has a pivotal role in hippocampal memory processes. We found that ATF4 downregulation in hippocampal neurons reduces membrane-bound GABABR levels and thereby increases intrinsic excitability. These effects are mediated by loss of the small GTPase Cdc42 following ATF4 downregulation. These findings reveal a critical role for ATF4 in regulating the modulation of neuronal excitability by GABABRs.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Receptores de GABA-B/metabolismo , Animales , Femenino , Hipocampo/metabolismo , Masculino , Neuronas/metabolismo , Transporte de Proteínas/fisiología , Ratas , Proteína de Unión al GTP cdc42/metabolismo
3.
Front Mol Neurosci ; 11: 62, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29599707

RESUMEN

Activating transcription factor 4 (ATF4) plays important physiologic roles in the brain including regulation of learning and memory as well as neuronal survival and death. Yet, outside of translational regulation by the eIF2α-dependent stress response pathway, there is little information about how its levels are controlled in neurons. Here, we show that brain-derived neurotrophic factor (BDNF) promotes a rapid and sustained increase in neuronal ATF4 transcripts and protein levels. This increase is dependent on tropomyosin receptor kinase (TrkB) signaling, but independent of levels of phosphorylated eIF2α. The elevation in ATF4 protein occurs both in nuclei and processes. Transcriptome analysis revealed that ATF4 mediates BDNF-promoted induction of Sesn2 which encodes Sestrin2, a protector against oxidative and genotoxic stresses and a mTor complex 1 inhibitor. In contrast, BDNF-elevated ATF4 did not affect expression of a number of other known ATF4 targets including several with pro-apoptotic activity. The capacity of BDNF to elevate neuronal ATF4 may thus represent a means to maintain this transcription factor at levels that provide neuroprotection and optimal brain function without risk of triggering neurodegeneration.

4.
J Cell Biol ; 216(10): 3161-3178, 2017 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-28877993

RESUMEN

Oligomeric Amyloid ß1-42 (Aß) plays a crucial synaptotoxic role in Alzheimer's disease, and hyperphosphorylated tau facilitates Aß toxicity. The link between Aß and tau, however, remains controversial. In this study, we find that in hippocampal neurons, Aß acutely induces tubulin posttranslational modifications (PTMs) and stabilizes dynamic microtubules (MTs) by reducing their catastrophe frequency. Silencing or acute inhibition of the formin mDia1 suppresses these activities and corrects the synaptotoxicity and deficits of axonal transport induced by Aß. We explored the mechanism of rescue and found that stabilization of dynamic MTs promotes tau-dependent loss of dendritic spines and tau hyperphosphorylation. Collectively, these results uncover a novel role for mDia1 in Aß-mediated synaptotoxicity and demonstrate that inhibition of MT dynamics and accumulation of PTMs are driving factors for the induction of tau-mediated neuronal damage.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Axones/metabolismo , Proteínas Portadoras/metabolismo , Citocromo-B(5) Reductasa/metabolismo , Espinas Dendríticas/metabolismo , Microtúbulos/metabolismo , Fragmentos de Péptidos/metabolismo , Sinapsis/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/genética , Animales , Proteínas Portadoras/genética , Citocromo-B(5) Reductasa/genética , Forminas , Ratones , Ratones Noqueados , Microtúbulos/genética , Fragmentos de Péptidos/genética , Procesamiento Proteico-Postraduccional/genética , Transporte de Proteínas/genética , Ratas , Ratas Sprague-Dawley , Sinapsis/genética , Proteínas tau/genética
5.
Acta Neuropathol ; 134(5): 749-767, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28631094

RESUMEN

The mechanisms underlying ryanodine receptor (RyR) dysfunction associated with Alzheimer disease (AD) are still not well understood. Here, we show that neuronal RyR2 channels undergo post-translational remodeling (PKA phosphorylation, oxidation, and nitrosylation) in brains of AD patients, and in two murine models of AD (3 × Tg-AD, APP +/- /PS1 +/-). RyR2 is depleted of calstabin2 (KFBP12.6) in the channel complex, resulting in endoplasmic reticular (ER) calcium (Ca2+) leak. RyR-mediated ER Ca2+ leak activates Ca2+-dependent signaling pathways, contributing to AD pathogenesis. Pharmacological (using a novel RyR stabilizing drug Rycal) or genetic rescue of the RyR2-mediated intracellular Ca2+ leak improved synaptic plasticity, normalized behavioral and cognitive functions and reduced Aß load. Genetically altered mice with congenitally leaky RyR2 exhibited premature and severe defects in synaptic plasticity, behavior and cognitive function. These data provide a mechanism underlying leaky RyR2 channels, which could be considered as potential AD therapeutic targets.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Calcio/metabolismo , Trastornos del Conocimiento/metabolismo , Procesamiento Proteico-Postraduccional , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Enfermedad de Alzheimer/patología , Animales , Señalización del Calcio , Trastornos del Conocimiento/patología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Femenino , Humanos , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Transgénicos , Estrés Oxidativo/fisiología , Fosforilación , Reconocimiento en Psicología/fisiología , Retículo Sarcoplasmático/metabolismo
6.
Nat Med ; 22(11): 1207-1208, 2016 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-27824824
7.
Cell Rep ; 11(2): 183-91, 2015 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-25865882

RESUMEN

Prior studies suggested that the transcription factor ATF4 negatively regulates synaptic plastic and memory. By contrast, we provide evidence from direct in vitro and in vivo knockdown of ATF4 in rodent hippocampal neurons and from ATF4-null mice that implicate ATF4 as essential for normal synaptic plasticity and memory. In particular, hippocampal ATF4 downregulation produces deficits in long-term spatial memory and behavioral flexibility without affecting associative memory or anxiety-like behavior. ATF4 knockdown or loss also causes profound impairment of both long-term potentiation (LTP) and long-term depression (LTD) as well as decreased glutamatergic function. We conclude that ATF4 is a key regulator of the physiological state necessary for neuronal plasticity and memory.


Asunto(s)
Factor de Transcripción Activador 4/genética , Memoria/fisiología , Plasticidad Neuronal/fisiología , Neuronas/fisiología , Factor de Transcripción Activador 4/biosíntesis , Animales , Hipocampo/fisiología , Ratones , Ratones Noqueados , Plasticidad Neuronal/genética , Sinapsis/genética , Sinapsis/fisiología
8.
PLoS One ; 10(3): e0120352, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25781952

RESUMEN

Alzheimer's disease (AD) is a complex multifactorial disorder with poorly characterized pathogenesis. Our understanding of this disease would thus benefit from an approach that addresses this complexity by elucidating the regulatory networks that are dysregulated in the neural compartment of AD patients, across distinct brain regions. Here, we use a Systems Biology (SB) approach, which has been highly successful in the dissection of cancer related phenotypes, to reverse engineer the transcriptional regulation layer of human neuronal cells and interrogate it to infer candidate Master Regulators (MRs) responsible for disease progression. Analysis of gene expression profiles from laser-captured neurons from AD and controls subjects, using the Algorithm for the Reconstruction of Accurate Cellular Networks (ARACNe), yielded an interactome consisting of 488,353 transcription-factor/target interactions. Interrogation of this interactome, using the Master Regulator INference algorithm (MARINa), identified an unbiased set of candidate MRs causally responsible for regulating the transcriptional signature of AD progression. Experimental assays in autopsy-derived human brain tissue showed that three of the top candidate MRs (YY1, p300 and ZMYM3) are indeed biochemically and histopathologically dysregulated in AD brains compared to controls. Our results additionally implicate p53 and loss of acetylation homeostasis in the neurodegenerative process. This study suggests that an integrative, SB approach can be applied to AD and other neurodegenerative diseases, and provide significant novel insight on the disease progression.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Proteínas del Tejido Nervioso/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Animales , Humanos , Proteínas del Tejido Nervioso/genética , Ratas
9.
J Clin Invest ; 125(2): 681-6, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25574843

RESUMEN

Tau is a highly abundant and multifunctional brain protein that accumulates in neurofibrillary tangles (NFTs), most commonly in Alzheimer's disease (AD) and primary age-related tauopathy. Recently, microRNAs (miRNAs) have been linked to neurodegeneration; however, it is not clear whether miRNA dysregulation contributes to tau neurotoxicity. Here, we determined that the highly conserved brain miRNA miR-219 is downregulated in brain tissue taken at autopsy from patients with AD and from those with severe primary age-related tauopathy. In a Drosophila model that produces human tau, reduction of miR-219 exacerbated tau toxicity, while overexpression of miR-219 partially abrogated toxic effects. Moreover, we observed a bidirectional modulation of tau levels in the Drosophila model that was dependent on miR-219 expression or neutralization, demonstrating that miR-219 regulates tau in vivo. In mammalian cellular models, we found that miR-219 binds directly to the 3'-UTR of the tau mRNA and represses tau synthesis at the post-transcriptional level. Together, our data indicate that silencing of tau by miR-219 is an ancient regulatory mechanism that may become perturbed during neurofibrillary degeneration and suggest that this regulatory pathway may be useful for developing therapeutics for tauopathies.


Asunto(s)
Regiones no Traducidas 3' , Enfermedad de Alzheimer/metabolismo , MicroARNs/metabolismo , Biosíntesis de Proteínas , Proteínas tau/biosíntesis , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Animales , Modelos Animales de Enfermedad , Drosophila melanogaster , Humanos , MicroARNs/genética , Proteínas tau/genética
10.
J Biol Chem ; 290(9): 5267-79, 2015 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-25568312

RESUMEN

KCl withdrawal-induced apoptosis in cerebellar granule neurons is associated with aberrant cell cycle activation, and treatment with cyclin-dependent kinase (Cdk) inhibitors protects cells from undergoing apoptosis. Because the Cdk inhibitor flavopiridol is known to inhibit RNA polymerase II (Pol II)-dependent transcription elongation by inhibiting the positive transcription elongation factor b (P-TEFb, a complex of CDK9 and cyclin T), we examined whether inhibition of RNA Pol II protects neurons from apoptosis. Treatment of neurons with 5, 6-dichloro-1-ß-D-ribobenzimidazole (DRB), an RNA Pol II-dependent transcription elongation inhibitor, and flavopiridol inhibited phosphorylation and activation of Pol II and protected neurons from undergoing apoptosis. In addition to Pol II, neurons subjected to KCl withdrawal showed increased phosphorylation and activation of p70 S6 kinase, which was inhibited by both DRB and flavopiridol. Immunostaining analysis of the neurons deprived of KCl showed increased nuclear levels of phospho-p70 S6 kinase, and neurons protected with DRB and flavopiridol showed accumulation of the kinase into large spliceosome assembly factor-positive speckle domains within the nuclei. The formation of these foci corresponded with cell survival, and removal of the inhibitors resulted in dispersal of the speckles into smaller foci with subsequent apoptosis induction. Because p70 S6 kinase is known to induce translation of mRNAs containing a 5'-terminal oligopyrimidine tract, our data suggest that transcription and translation of this subset of mRNAs may contribute to KCl withdrawal-induced apoptosis in neurons.


Asunto(s)
Apoptosis , Flavonoides , Neuronas/metabolismo , Piperidinas , ARN Polimerasa II/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Animales , Western Blotting , Células Cultivadas , Cerebelo/citología , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/genética , Quinasas Ciclina-Dependientes/metabolismo , Diclororribofuranosil Benzoimidazol/farmacología , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Inmunohistoquímica , Neuronas/citología , Neuronas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Piperidinas/farmacología , Cloruro de Potasio/metabolismo , Cloruro de Potasio/farmacología , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Elongación de la Transcripción Genética/efectos de los fármacos
11.
Acta Neuropathol ; 128(6): 755-66, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25348064

RESUMEN

We recommend a new term, "primary age-related tauopathy" (PART), to describe a pathology that is commonly observed in the brains of aged individuals. Many autopsy studies have reported brains with neurofibrillary tangles (NFTs) that are indistinguishable from those of Alzheimer's disease (AD), in the absence of amyloid (Aß) plaques. For these "NFT+/Aß-" brains, for which formal criteria for AD neuropathologic changes are not met, the NFTs are mostly restricted to structures in the medial temporal lobe, basal forebrain, brainstem, and olfactory areas (bulb and cortex). Symptoms in persons with PART usually range from normal to amnestic cognitive changes, with only a minority exhibiting profound impairment. Because cognitive impairment is often mild, existing clinicopathologic designations, such as "tangle-only dementia" and "tangle-predominant senile dementia", are imprecise and not appropriate for most subjects. PART is almost universally detectable at autopsy among elderly individuals, yet this pathological process cannot be specifically identified pre-mortem at the present time. Improved biomarkers and tau imaging may enable diagnosis of PART in clinical settings in the future. Indeed, recent studies have identified a common biomarker profile consisting of temporal lobe atrophy and tauopathy without evidence of Aß accumulation. For both researchers and clinicians, a revised nomenclature will raise awareness of this extremely common pathologic change while providing a conceptual foundation for future studies. Prior reports that have elucidated features of the pathologic entity we refer to as PART are discussed, and working neuropathological diagnostic criteria are proposed.


Asunto(s)
Envejecimiento/patología , Encéfalo/patología , Tauopatías/patología , Diagnóstico Diferencial , Humanos , Placa Amiloide/patología , Tauopatías/clasificación , Tauopatías/diagnóstico , Terminología como Asunto
12.
Front Cell Neurosci ; 8: 177, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25071442

RESUMEN

The ubiquitously expressed activating transcription factor 4 (ATF4) has been variably reported to either promote or inhibit neuronal plasticity and memory. However, the potential cellular bases for these and other actions of ATF4 in brain are not well-defined. In this report, we focus on ATF4's role in post-synaptic synapse development and dendritic spine morphology. shRNA-mediated silencing of ATF4 significantly reduces the densities of PSD-95 and GluR1 puncta (presumed markers of excitatory synapses) in long-term cultures of cortical and hippocampal neurons. ATF4 knockdown also decreases the density of mushroom spines and increases formation of abnormally-long dendritic filopodia in such cultures. In vivo knockdown of ATF4 in adult mouse hippocampal neurons also reduces mushroom spine density. In contrast, ATF4 over-expression does not affect the densities of PSD-95 puncta or mushrooom spines. Regulation of synaptic puncta and spine densities by ATF4 requires its transcriptional activity and is mediated at least in part by indirectly controlling the stability and expression of the total and active forms of the actin regulatory protein Cdc42. In support of such a mechanism, ATF4 silencing decreases the half-life of Cdc42 in cultured cortical neurons from 31.5 to 18.5 h while knockdown of Cdc42, like ATF4 knockdown, reduces the densities of mushroom spines and PSD-95 puncta. Thus, ATF4 appears to participate in neuronal development and plasticity by regulating the post-synaptic development of synapses and dendritic mushroom spines via a mechanism that includes regulation of Cdc42 levels.

13.
Acta Neuropathol Commun ; 2: 4, 2014 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-24398250

RESUMEN

BACKGROUND: Induced pluripotent stem cells (iPSCs) derived from patients with neurodegenerative disease generally lack neuropathological confirmation, the gold standard for disease classification and grading of severity. The use of tissue with a definitive neuropathological diagnosis would be an ideal source for iPSCs. The challenge to this approach is that the majority of biobanked brain tissue was not meant for growing live cells, and thus was not frozen in the presence of cryoprotectants such as DMSO. RESULTS: We report the generation of iPSCs from frozen non-cryoprotected dural tissue stored at -80°C for up to 11 years. This autopsy cohort included subjects with Alzheimer's disease and four other neurodegenerative diseases. CONCLUSIONS: Disease-specific iPSCs can be generated from readily available, archival biobanked tissue. This allows for rapid expansion of generating iPSCs with confirmed pathology as well as allowing access to rare patient variants that have been banked.


Asunto(s)
Duramadre/patología , Células Madre Pluripotentes Inducidas/fisiología , Enfermedad de Alzheimer/patología , Animales , Antígenos de Superficie/metabolismo , Diferenciación Celular , Línea Celular Transformada/patología , Proliferación Celular , Bases de Datos como Asunto , Fibroblastos/metabolismo , Fibroblastos/virología , Proteínas de Homeodominio/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/patología , Ratones , Proteína Homeótica Nanog , Enfermedades Neurodegenerativas/patología , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Cambios Post Mortem , Proteoglicanos/metabolismo , Piel/citología , Antígenos Embrionarios Específico de Estadio/metabolismo
14.
J Neurosci ; 31(9): 3186-96, 2011 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-21368030

RESUMEN

The generation, differentiation, and migration of newborn neurons are critical features of normal brain development that are subject to both extracellular and intracellular regulation. However, the means of such control are only partially understood. Here, we show that expression of RTP801/REDD1, an inhibitor of mTOR (mammalian target of rapamycin) activation, is regulated during neuronal differentiation and that RTP801 functions to influence the timing of both neurogenesis and neuron migration. RTP801 levels are high in embryonic cortical neuroprogenitors, diminished in newborn neurons, and low in mature neurons. Knockdown of RTP801 in vitro and in vivo accelerates cell cycle exit by neuroprogenitors and their differentiation into neurons. It also disrupts migration of rat newborn neurons to the cortical plate and results in the ectopic localization of mature neurons. On the other hand, RTP801 overexpression delays neuronal differentiation. These findings suggest that endogenous RTP801 plays an essential role in temporal control of cortical development and in cortical patterning.


Asunto(s)
Movimiento Celular/fisiología , Corteza Cerebral/fisiología , Neurogénesis/fisiología , Neuronas/citología , Neuronas/fisiología , Proteínas Represoras/fisiología , Animales , Tipificación del Cuerpo/genética , Tipificación del Cuerpo/fisiología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Movimiento Celular/genética , Células Cultivadas , Corteza Cerebral/citología , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Endogámicos C57BL , Neurogénesis/genética , Células PC12 , Ratas , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Células Madre/citología , Células Madre/fisiología , Factores de Tiempo , Factores de Transcripción
15.
J Alzheimers Dis ; 12(3): 229-40, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18057556

RESUMEN

APP overexpressing mice have been widely used in the study of Alzheimer's disease (AD), focusing mainly at older ages, with higher accumulation of amyloid-beta peptide (Abeta). A decrease in hippocampal adult neurogenesis has been described in these models and proposed to be a consequence of Abeta accumulation. Only one study demonstrates increased neurogenesis in the hippocampus of APP-overexpressing J20 mice, and suggests it is a compensatory effect due to a subtle Abeta-induced damage. We have previously reported that a specific aggregation of Abeta has neurogenic potential on neural stem cells (NSC) in vitro. In order to clarify the contradicting data reported in vivo, we investigated NSC proliferation and neuronal differentiation in the hippocampi of J20 mice at a broader range of ages. Using immunohistochemistry, we show increased proliferation and neuronal differentiation in the hippocampi of 3 month-old J20 mice that reverted when animals became older. The increase in neurogenesis correlated with detectable levels of oligomeric Abeta, measured by ELISA and western blot. We suggest that oligomeric Abeta directly induces neurogenesis in vivo as has been demonstrated in vitro. Understanding the mechanisms underlying these changes could lead to treatments to control the neuronal differentiation of endogenous precursors through the progress of AD.


Asunto(s)
Precursor de Proteína beta-Amiloide/genética , Factores de Edad , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/inmunología , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Cromatografía Líquida de Alta Presión , Modelos Animales de Enfermedad , Hipocampo/patología , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Ratones , Ratones Transgénicos , Molécula L1 de Adhesión de Célula Nerviosa/inmunología , Neuronas/inmunología , Neuronas/metabolismo , Neuronas/patología , Ácidos Siálicos/inmunología
16.
Dev Neurobiol ; 67(6): 818-26, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17443827

RESUMEN

Purkinje cells are vulnerable to a number of physical, chemical, and genetic insults during development and maturity. Normal development of these cells depends on the cell-cell interactions between granule and astroglial cell populations. Apoptotic death in Purkinje neurons had been shown to be associated with cell cycle activation, and new DNA synthesis is associated with Purkinje cell death in staggerer and lurcher mutant mice. Here using an in vitro organotypic slice culture model from 9 (P9) and 4 days (P4) old postnatal rats we show that the cyclin dependent kinase (cdk) inhibitors (roscovitine, olomoucine, and flavopiridol) protect the Purkinje cells from cell death. The results are more pronounced in the cerebellar sections from P4 rats. Analysis of Purkinje neurons in sections from P4 rats after 1 week of culturing showed that while there were very limited calbindin positive neurons in the untreated sections the cdk inhibitor treated sections had a notably higher number. Although treatment with cdk inhibitors inhibited Purkinje cell loss significantly, the morphology of these neurons was abnormal, with stunted dendrites and axons. Since the retinoblastoma protein (Rb) is the major pocket protein involved in determining the differentiated state of neurons we examined the effect of over-expressing Rb in the organotypic cultures. Rb overexpression significantly inhibited the Purkinje cell death and these neurons maintained their normal morphology. Thus our studies show that the cell death in Purkinje neurons observed in organotypic cultures is cell cycle dependent and the optimal survival requires Rb.


Asunto(s)
Ciclo Celular/fisiología , Muerte Celular/fisiología , Células de Purkinje/citología , Células de Purkinje/metabolismo , Proteína de Retinoblastoma/biosíntesis , Animales , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Inmunohistoquímica , Técnicas de Cultivo de Órganos , Células de Purkinje/patología , Ratas
17.
J Neurosci ; 24(45): 10040-6, 2004 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-15537872

RESUMEN

The study of protein function in neurons has been hindered by the lack of highly efficient, nontoxic methods of inducing RNA interference in such cells. Here we show that application of synthetic small interfering RNA (siRNA) linked to the vector peptide Penetratin1 results in rapid, highly efficient uptake of siRNA by entire populations of cultured primary mammalian hippocampal and sympathetic neurons. This treatment leads to specific knock-down of targeted proteins within hours without the toxicity associated with transfection. In contrast to current methods, our technique permits study of protein function across entire populations with minimal disturbance of complex cellular networks. Using this technique, we found that protein knock-down (evident after 6 hr) precedes any decrease in targeted message (evident after 24 hr), suggesting an early, translational repression by perfectly targeted siRNAs.


Asunto(s)
Marcación de Gen/métodos , Neuronas/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología , Animales , Proteínas Portadoras/administración & dosificación , Caspasa 3 , Caspasas/análisis , Caspasas/genética , Péptidos de Penetración Celular , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Medio de Cultivo Libre de Suero/farmacología , Hipocampo/citología , Ratones , Neuronas/metabolismo , Ratas , Ganglio Cervical Superior/citología , Superóxido Dismutasa/análisis , Superóxido Dismutasa/genética , Superóxido Dismutasa-1 , Transfección
18.
J Neurocytol ; 33(3): 377-87, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15475691

RESUMEN

Synaptic damage and loss are factors that affect the degree of dementia experienced in Alzheimer disease (AD) patients. Multicolor DiOlistic labeling of the hippocampus has been undertaken which allows the full dendritic arbor of targeted neurons to be imaged. Using this labeling technique the neuronal morphology of two transgenic mouse lines (J20 and APP/PS1) expressing mutant forms of the Amyloid Precursor Protein (APP), at various ages, have been visualized and compared to Wild Type (WT) littermate controls. Swollen bulbous dystrophic neurites with loss of spines were apparent in the transgenic animals. Upon quantification, statistically significant reductions in the number of spines and total dendrite area was observed in both transgenic mouse lines at 11 months of age. Similar morphological abnormalities were seen in human AD hippocampal tissue both qualitatively and quantitatively. Immunohistochemistry and DiOlistic labeling was combined so that Abeta plaques were imaged in relation to the dendritic trees. No preferential localization of these abnormal dystrophic neurites was seen in regions with plaques. DiI labeled reative astrocytes were often apparent in close proximity to A beta plaques.


Asunto(s)
Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Dendritas/patología , Espinas Dendríticas/patología , Hipocampo/patología , Envejecimiento/metabolismo , Envejecimiento/patología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Astrocitos/metabolismo , Astrocitos/patología , Biolística , Carbocianinas , Dendritas/metabolismo , Espinas Dendríticas/metabolismo , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Humanos , Inmunohistoquímica , Ratones , Ratones Transgénicos , Mutación/genética , Placa Amiloide/genética , Placa Amiloide/metabolismo , Placa Amiloide/patología , Células Piramidales/metabolismo , Células Piramidales/patología , Coloración y Etiquetado/métodos
19.
J Neurosci ; 24(23): 5439-44, 2004 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-15190117

RESUMEN

The adult mammalian brain contains neural stem cells (NSCs) with self-renewal and multilineage potential in the hippocampus and subventricular zone. However, neurogenesis from these areas does not compensate for neuronal loss in age-related neurodegenerative disorders such as Alzheimer's disease (AD). To test whether an impairment of neurogenesis could contribute to the pathogenesis of AD, we examined the effects of amyloid-beta peptide (Abeta) on the survival and neuronal differentiation of cultured NSCs from striatum and hippocampus. We show that Abeta peptide does not impair the neurogenic rate in NSC progeny, but that it increases the total number of neurons in vitro in a dose-dependent manner. The neurogenic effect of Abeta peptide is not dependent on soluble factors released from the NSC progeny. Neurogenesis is induced by Abeta42 and not Abeta40 or Abeta 25-35, and the activity appears to be a property of Abeta oligomers and not fibrils. These results suggest that Abeta may have positive as well as deleterious actions, and that a knowledge of the mechanisms involved in the former could be valuable in exploiting the regenerative and plastic potential of the brain in preventing and treating Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/metabolismo , Cuerpo Estriado/patología , Cuerpo Estriado/fisiopatología , Hipocampo/patología , Hipocampo/fisiopatología , Neuronas/metabolismo , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/efectos adversos , Animales , Animales Recién Nacidos , Técnicas de Cultivo de Célula , Diferenciación Celular , Supervivencia Celular , Cuerpo Estriado/citología , Cuerpo Estriado/metabolismo , Embrión de Mamíferos , Hipocampo/citología , Hipocampo/metabolismo , Ratones , Ratones Endogámicos , Regeneración Nerviosa , Plasticidad Neuronal , Neuronas/patología , Placa Amiloide/metabolismo , Ratas , Ratas Sprague-Dawley , Células Madre
20.
Neurobiol Aging ; 25(8): 1057-66, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15212831

RESUMEN

In this paper, we examine the hypothesis that 4-hydroxynonenal (HNE), a product of lipid peroxidation, is a key mediator of cell death resulting from beta-amyloid exposure. We revisit the effects of HNE on different neuronal cell types to determine which caspase or caspases are required for HNE-induced death, and to compare these results with the known caspase requirements in other death paradigms. We have previously shown that in a given neuronal cell type different death stimuli can evoke stimulus-specific apoptotic pathways. We now show that HNE treatment of neuronal cells induced dose-dependent death and caspase activity which were blocked by inhibition of caspases. Antisense down-regulation of caspases-3, -7 or -9 provided complete protection from HNE-induced death, as did down-regulation of the caspase regulators APAF-1 and DIABLO. Conversely, this work and our previous studies of three other death paradigms show that caspase-3 is not required for death induced by beta-amyloid, SOD1 down-regulation, or trophic factor deprivation. We also show that HNE accumulated in settings where death does not ensue. We conclude that HNE toxicity is mediated via a caspase-9-dependent pathway but that HNE accumulation need not induce cell death nor is it an obligate mediator of Abeta-induced cell death.


Asunto(s)
Aldehídos/metabolismo , Péptidos beta-Amiloides/metabolismo , Apoptosis/fisiología , Sistema Nervioso Central/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Neuronas/metabolismo , Aldehídos/toxicidad , Péptidos beta-Amiloides/toxicidad , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis , Factor Apoptótico 1 Activador de Proteasas , Proteínas Portadoras/efectos de los fármacos , Proteínas Portadoras/metabolismo , Caspasa 3 , Caspasa 9 , Inhibidores de Caspasas , Caspasas/metabolismo , Sistema Nervioso Central/fisiopatología , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Inhibidores Enzimáticos/farmacología , Ratones , Proteínas Mitocondriales/efectos de los fármacos , Proteínas Mitocondriales/metabolismo , Factores de Crecimiento Nervioso/deficiencia , Enfermedades Neurodegenerativas/fisiopatología , Neuronas/efectos de los fármacos , Neuronas/patología , Células PC12 , Proteínas/metabolismo , Ratas , Superóxido Dismutasa/deficiencia , Superóxido Dismutasa-1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA