Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Sci Transl Med ; 14(642): eabm5527, 2022 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-35476595

RESUMEN

Intracellular accumulation of TAU aggregates is a hallmark of several neurodegenerative diseases. However, global genetic reduction of TAU is beneficial also in models of other brain disorders that lack such TAU pathology, suggesting a pathogenic role of nonaggregated TAU. Here, conditional ablation of TAU in excitatory, but not inhibitory, neurons reduced epilepsy, sudden unexpected death in epilepsy, overactivation of the phosphoinositide 3-kinase-AKT-mammalian target of rapamycin pathway, brain overgrowth (megalencephaly), and autism-like behaviors in a mouse model of Dravet syndrome, a severe epileptic encephalopathy of early childhood. Furthermore, treatment with a TAU-lowering antisense oligonucleotide, initiated on postnatal day 10, had similar therapeutic effects in this mouse model. Our findings suggest that excitatory neurons are the critical cell type in which TAU has to be reduced to counteract brain dysfunctions associated with Dravet syndrome and that overall cerebral TAU reduction could have similar benefits, even when initiated postnatally.


Asunto(s)
Trastorno Autístico , Epilepsias Mioclónicas , Epilepsia , Muerte Súbita e Inesperada en la Epilepsia , Proteínas tau , Animales , Trastorno Autístico/complicaciones , Trastorno Autístico/genética , Modelos Animales de Enfermedad , Epilepsias Mioclónicas/complicaciones , Epilepsias Mioclónicas/genética , Epilepsia/complicaciones , Epilepsia/genética , Epilepsia/metabolismo , Síndromes Epilépticos , Humanos , Lactante , Ratones , Neuronas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Espasmos Infantiles , Proteínas tau/metabolismo
2.
iScience ; 24(11): 103245, 2021 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-34755090

RESUMEN

Nonconvulsive epileptiform activity and microglial alterations have been detected in people with Alzheimer's disease (AD) and related mouse models. However, the relationship between these abnormalities remains to be elucidated. We suppressed epileptiform activity by treatment with the antiepileptic drug levetiracetam or by genetic ablation of tau and found that these interventions reversed or prevented aberrant microglial gene expression in brain tissues of aged human amyloid precursor protein transgenic mice, which simulate several key aspects of AD. The most robustly modulated genes included multiple factors previously implicated in AD pathogenesis, including TREM2, the hypofunction of which increases disease risk. Genetic reduction of TREM2 exacerbated epileptiform activity after mice were injected with kainate. We conclude that AD-related epileptiform activity markedly changes the molecular profile of microglia, inducing both maladaptive and adaptive alterations in their activities. Increased expression of TREM2 seems to support microglial activities that counteract this type of network dysfunction.

3.
eNeuro ; 8(3)2021.
Artículo en Inglés | MEDLINE | ID: mdl-33833046

RESUMEN

Diverse gene products contribute to the pathogenesis of Alzheimer's disease (AD). Experimental models have helped elucidate their mechanisms and impact on brain functions. Human amyloid precursor protein (hAPP) transgenic mice from line J20 (hAPP-J20 mice) are widely used to simulate key aspects of AD. However, they also carry an insertional mutation in noncoding sequence of one Zbtb20 allele, a gene involved in neural development. We demonstrate that heterozygous hAPP-J20 mice have reduced Zbtb20 expression in some AD-relevant brain regions, but not others, and that Zbtb20 levels are higher in hAPP-J20 mice than heterozygous Zbtb20 knock-out (Zbtb20+/-) mice. Whereas hAPP-J20 mice have premature mortality, severe deficits in learning and memory, other behavioral alterations, and prominent nonconvulsive epileptiform activity, Zbtb20+/- mice do not. Thus, the insertional mutation in hAPP-J20 mice does not ablate the affected Zbtb20 allele and is unlikely to account for the AD-like phenotype of this model.


Asunto(s)
Enfermedad de Alzheimer , Precursor de Proteína beta-Amiloide , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , Ratones Transgénicos , Fenotipo , Factores de Transcripción
4.
J Fish Biol ; 98(1): 17-32, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32964432

RESUMEN

The elasmoid scales of anadromous sea trout Salmo trutta L. represent a significant internal reservoir of Ca2+ . Although more is known about long-term remodelling of scales in response to calciotropic challenges encountered during smoltification and migration, very little is known about the contribution made by scales to the short-term, minute-to-minute regulation of Ca2+ homeostasis in the extracellular fluid (ECF) during these phases of the life cycle. This gap in the knowledge is partly due to the technical challenges involved in measuring small Ca2+ fluxes around the scales of live fish in real time. Here, this study describes exfoliating, mounting and culturing scales and their resident cells from parr, smolt and adult sea trout from a freshwater environment, as well as from adult sea trout caught in sea or brackish water. All the scales were then examined using an extracellular, non-invasive, surface-scanning Ca2+ -sensitive microelectrode. The authors quantified the Ca2+ fluxes, in the absence of any systemic or local regulators, into and out of scales on both the episquamal and hyposquamal sides under different extracellular calcemic challenges set to mimic a variety of ECF-Ca2+ concentrations. Scales from the life-cycle stages as well as from adult fish taken from sea, brackish or fresh water all showed a consistent efflux or influx of Ca2+ under hypo- or hypercalcemic conditions, respectively. What were considered to be isocalcemic conditions resulted in minimal flux of Ca2+ in either direction, or in the case of adult scales, a consistent but small influx. Indeed, adult scales appeared to display the largest flux densities in either direction. These new data extend the current understanding of the role played by fish scales in the short-term, minute-to-minute homeostatic regulation of ECF-Ca2+ concentration, and are similar to those recently reported from zebrafish Danio rerio scales. This suggests that this short-term regulatory response might be a common feature of teleost scales.


Asunto(s)
Migración Animal/fisiología , Escamas de Animales/metabolismo , Calcio/metabolismo , Líquido Extracelular/química , Homeostasis , Trucha/fisiología , Animales , Calcio/sangre , Agua Dulce , Agua de Mar , Trucha/sangre
5.
Mol Neurodegener ; 15(1): 53, 2020 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-32921309

RESUMEN

BACKGROUND: Alzheimer's disease (AD) is the most frequent and costly neurodegenerative disorder. Although diverse lines of evidence suggest that the amyloid precursor protein (APP) is involved in its causation, the precise mechanisms remain unknown and no treatments are available to prevent or halt the disease. A favorite hypothesis has been that APP contributes to AD pathogenesis through the cerebral accumulation of the amyloid-ß peptide (Aß), which is derived from APP through sequential proteolytic cleavage by BACE1 and γ-secretase. However, inhibitors of these enzymes have failed in clinical trials despite clear evidence for target engagement. METHODS: To further elucidate the roles of APP and its metabolites in AD pathogenesis, we analyzed transgenic mice overexpressing wildtype human APP (hAPP) or hAPP carrying mutations that cause autosomal dominant familial AD (FAD), as well as App knock-in mice that do not overexpress hAPP but have two mouse App alleles with FAD mutations and a humanized Aß sequence. RESULTS: Although these lines of mice had marked differences in cortical and hippocampal levels of APP, APP C-terminal fragments, soluble Aß, Aß oligomers and age-dependent amyloid deposition, they all developed cognitive deficits as well as non-convulsive epileptiform activity, a type of network dysfunction that also occurs in a substantive proportion of humans with AD. Pharmacological inhibition of BACE1 effectively reduced levels of amyloidogenic APP C-terminal fragments (C99), soluble Aß, Aß oligomers, and amyloid deposits in transgenic mice expressing FAD-mutant hAPP, but did not improve their network dysfunction and behavioral abnormalities, even when initiated at early stages before amyloid deposits were detectable. CONCLUSIONS: hAPP transgenic and App knock-in mice develop similar pathophysiological alterations. APP and its metabolites contribute to AD-related functional alterations through complex combinatorial mechanisms that may be difficult to block with BACE inhibitors and, possibly, also with other anti-Aß treatments.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Conducta Animal/fisiología , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Red Nerviosa/metabolismo , Red Nerviosa/patología
6.
Proc Natl Acad Sci U S A ; 115(48): E11388-E11396, 2018 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-30413620

RESUMEN

Located within the brain's ventricles, the choroid plexus produces cerebrospinal fluid and forms an important barrier between the central nervous system and the blood. For unknown reasons, the choroid plexus produces high levels of the protein klotho. Here, we show that these levels naturally decline with aging. Depleting klotho selectively from the choroid plexus via targeted viral vector-induced knockout in Klothoflox/flox mice increased the expression of multiple proinflammatory factors and triggered macrophage infiltration of this structure in young mice, simulating changes in unmanipulated old mice. Wild-type mice infected with the same Cre recombinase-expressing virus did not show such alterations. Experimental depletion of klotho from the choroid plexus enhanced microglial activation in the hippocampus after peripheral injection of mice with lipopolysaccharide. In primary cultures, klotho suppressed thioredoxin-interacting protein-dependent activation of the NLRP3 inflammasome in macrophages by enhancing fibroblast growth factor 23 signaling. We conclude that klotho functions as a gatekeeper at the interface between the brain and immune system in the choroid plexus. Klotho depletion in aging or disease may weaken this barrier and promote immune-mediated neuropathogenesis.


Asunto(s)
Envejecimiento/inmunología , Encéfalo/inmunología , Plexo Coroideo/inmunología , Glucuronidasa/inmunología , Envejecimiento/genética , Animales , Femenino , Glucuronidasa/genética , Hipocampo/inmunología , Humanos , Proteínas Klotho , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología
7.
Neuron ; 98(1): 75-89.e5, 2018 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-29551491

RESUMEN

Inhibitory interneurons regulate the oscillatory rhythms and network synchrony that are required for cognitive functions and disrupted in Alzheimer's disease (AD). Network dysrhythmias in AD and multiple neuropsychiatric disorders are associated with hypofunction of Nav1.1, a voltage-gated sodium channel subunit predominantly expressed in interneurons. We show that Nav1.1-overexpressing, but not wild-type, interneuron transplants derived from the embryonic medial ganglionic eminence (MGE) enhance behavior-dependent gamma oscillatory activity, reduce network hypersynchrony, and improve cognitive functions in human amyloid precursor protein (hAPP)-transgenic mice, which simulate key aspects of AD. Increased Nav1.1 levels accelerated action potential kinetics of transplanted fast-spiking and non-fast-spiking interneurons. Nav1.1-deficient interneuron transplants were sufficient to cause behavioral abnormalities in wild-type mice. We conclude that the efficacy of interneuron transplantation and the function of transplanted cells in an AD-relevant context depend on their Nav1.1 levels. Disease-specific molecular optimization of cell transplants may be required to ensure therapeutic benefits in different conditions.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Ondas Encefálicas/fisiología , Encéfalo/metabolismo , Cognición/fisiología , Interneuronas/metabolismo , Canal de Sodio Activado por Voltaje NAV1.1/biosíntesis , Potenciales de Acción/fisiología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/terapia , Animales , Encéfalo/cirugía , Modelos Animales de Enfermedad , Expresión Génica , Hipocampo/metabolismo , Hipocampo/cirugía , Humanos , Interneuronas/trasplante , Locomoción/fisiología , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Transgénicos , Canal de Sodio Activado por Voltaje NAV1.1/genética
8.
Neurobiol Dis ; 110: 29-36, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29100987

RESUMEN

Adenosine A2A receptors are putative therapeutic targets for neurological disorders. The adenosine A2A receptor antagonist istradefylline is approved in Japan for Parkinson's disease and is being tested in clinical trials for this condition elsewhere. A2A receptors on neurons and astrocytes may contribute to Alzheimer's disease (AD) by impairing memory. However, it is not known whether istradefylline enhances cognitive function in aging animals with AD-like amyloid plaque pathology. Here, we show that elevated levels of Aß, C-terminal fragments of the amyloid precursor protein (APP), or amyloid plaques, but not overexpression of APP per se, increase astrocytic A2A receptor levels in the hippocampus and neocortex of aging mice. Moreover, in amyloid plaque-bearing mice, low-dose istradefylline treatment enhanced spatial memory and habituation, supporting the conclusion that, within a well-defined dose range, A2A receptor blockers might help counteract memory problems in patients with Alzheimer's disease.


Asunto(s)
Antagonistas del Receptor de Adenosina A2/farmacología , Astrocitos/metabolismo , Encéfalo/efectos de los fármacos , Trastornos de la Memoria , Purinas/farmacología , Envejecimiento , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides , Animales , Encéfalo/patología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Placa Amiloide/patología , Receptor de Adenosina A2A
9.
Nat Commun ; 6: 8897, 2015 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-26615780

RESUMEN

Maintaining DNA integrity is vital for all cells and organisms. Defective DNA repair may contribute to neurological disorders, including Alzheimer's disease (AD). We found reduced levels of BRCA1, but not of other DNA repair factors, in the brains of AD patients and human amyloid precursor protein (hAPP) transgenic mice. Amyloid-ß oligomers reduced BRCA1 levels in primary neuronal cultures. In wild-type mice, knocking down neuronal BRCA1 in the dentate gyrus caused increased DNA double-strand breaks, neuronal shrinkage, synaptic plasticity impairments, and learning and memory deficits, but not apoptosis. Low levels of hAPP/Amyloid-ß overexpression exacerbated these effects. Physiological neuronal activation increased BRCA1 levels, whereas stimulating predominantly extrasynaptic N-methyl-D-aspartate receptors promoted the proteasomal degradation of BRCA1. We conclude that BRCA1 is regulated by neuronal activity, protects the neuronal genome, and critically supports neuronal integrity and cognitive functions. Pathological accumulation of Aß depletes neuronal BRCA1, which may contribute to cognitive deficits in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/psicología , Proteína BRCA1/deficiencia , Encéfalo/metabolismo , Reparación del ADN , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Animales , Proteína BRCA1/genética , Encéfalo/fisiopatología , Cognición , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/metabolismo
10.
J Cell Biol ; 209(3): 419-33, 2015 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-25963821

RESUMEN

Axonal transport deficits in Alzheimer's disease (AD) are attributed to amyloid ß (Aß) peptides and pathological forms of the microtubule-associated protein tau. Genetic ablation of tau prevents neuronal overexcitation and axonal transport deficits caused by recombinant Aß oligomers. Relevance of these findings to naturally secreted Aß and mechanisms underlying tau's enabling effect are unknown. Here we demonstrate deficits in anterograde axonal transport of mitochondria in primary neurons from transgenic mice expressing familial AD-linked forms of human amyloid precursor protein. We show that these deficits depend on Aß1-42 production and are prevented by tau reduction. The copathogenic effect of tau did not depend on its microtubule binding, interactions with Fyn, or potential role in neuronal development. Inhibition of neuronal activity, N-methyl-d-aspartate receptor function, or glycogen synthase kinase 3ß (GSK3ß) activity or expression also abolished Aß-induced transport deficits. Tau ablation prevented Aß-induced GSK3ß activation. Thus, tau allows Aß oligomers to inhibit axonal transport through activation of GSK3ß, possibly by facilitating aberrant neuronal activity.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Transporte Axonal , Glucógeno Sintasa Quinasa 3/metabolismo , Fragmentos de Péptidos/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/genética , Animales , Activación Enzimática/genética , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Humanos , Ratones , Ratones Noqueados , Fragmentos de Péptidos/genética , Proteínas Proto-Oncogénicas c-fyn/genética , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Proteínas tau/genética
11.
J Neurosci ; 35(6): 2358-71, 2015 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-25673831

RESUMEN

Aging is the principal demographic risk factor for Alzheimer disease (AD), the most common neurodegenerative disorder. Klotho is a key modulator of the aging process and, when overexpressed, extends mammalian lifespan, increases synaptic plasticity, and enhances cognition. Whether klotho can counteract deficits related to neurodegenerative diseases, such as AD, is unknown. Here we show that elevating klotho expression decreases premature mortality and network dysfunction in human amyloid precursor protein (hAPP) transgenic mice, which simulate key aspects of AD. Increasing klotho levels prevented depletion of NMDA receptor (NMDAR) subunits in the hippocampus and enhanced spatial learning and memory in hAPP mice. Klotho elevation in hAPP mice increased the abundance of the GluN2B subunit of NMDAR in postsynaptic densities and NMDAR-dependent long-term potentiation, which is critical for learning and memory. Thus, increasing wild-type klotho levels or activities improves synaptic and cognitive functions, and may be of therapeutic benefit in AD and other cognitive disorders.


Asunto(s)
Precursor de Proteína beta-Amiloide/fisiología , Cognición/fisiología , Glucuronidasa/fisiología , Longevidad/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Conducta Animal/fisiología , Trastornos del Conocimiento/genética , Trastornos del Conocimiento/psicología , Humanos , Proteínas Klotho , Longevidad/fisiología , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Transgénicos , Red Nerviosa/patología , Receptores de N-Metil-D-Aspartato/fisiología , Sinapsis/patología , Proteínas tau/metabolismo
12.
Nat Neurosci ; 18(3): 423-34, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25622143

RESUMEN

Astrocytes express a variety of G protein-coupled receptors and might influence cognitive functions, such as learning and memory. However, the roles of astrocytic Gs-coupled receptors in cognitive function are not known. We found that humans with Alzheimer's disease (AD) had increased levels of the Gs-coupled adenosine receptor A2A in astrocytes. Conditional genetic removal of these receptors enhanced long-term memory in young and aging mice and increased the levels of Arc (also known as Arg3.1), an immediate-early gene that is required for long-term memory. Chemogenetic activation of astrocytic Gs-coupled signaling reduced long-term memory in mice without affecting learning. Like humans with AD, aging mice expressing human amyloid precursor protein (hAPP) showed increased levels of astrocytic A2A receptors. Conditional genetic removal of these receptors enhanced memory in aging hAPP mice. Together, these findings establish a regulatory role for astrocytic Gs-coupled receptors in memory and suggest that AD-linked increases in astrocytic A2A receptor levels contribute to memory loss.


Asunto(s)
Astrocitos/metabolismo , Regulación de la Expresión Génica/fisiología , Memoria a Largo Plazo/fisiología , Receptor de Adenosina A2A/metabolismo , Receptores de Serotonina 5-HT4/metabolismo , Transducción de Señal/fisiología , Enfermedad de Alzheimer/patología , Animales , Animales Recién Nacidos , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Conducta Exploratoria/efectos de los fármacos , Conducta Exploratoria/fisiología , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Indoles/farmacología , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Receptor de Adenosina A2A/genética , Receptores de Serotonina 5-HT4/genética , Reconocimiento en Psicología/efectos de los fármacos , Reconocimiento en Psicología/fisiología , Antagonistas de la Serotonina/farmacología , Sulfonamidas/farmacología
13.
Ann Clin Transl Neurol ; 2(11): 1012-28, 2015 11.
Artículo en Inglés | MEDLINE | ID: mdl-26732627

RESUMEN

OBJECTIVE: Dementia with Lewy bodies (DLB) is associated with the accumulation of wild-type human α-synuclein (SYN) in neurons and with prominent slowing of brain oscillations on electroencephalography (EEG). However, it remains uncertain whether the EEG abnormalities are actually caused by SYN. METHODS: To determine whether SYN can cause neural network abnormalities, we performed EEG recordings and analyzed the expression of neuronal activity-dependent gene products in SYN transgenic mice. We also carried out comparative analyses in humans with DLB. RESULTS: We demonstrate that neuronal expression of SYN in transgenic mice causes a left shift in spectral power that closely resembles the EEG slowing observed in DLB patients. Surprisingly, SYN mice also had seizures and showed molecular hippocampal alterations indicative of aberrant network excitability, including calbindin depletion in the dentate gyrus. In postmortem brain tissues from DLB patients, we found reduced levels of calbindin mRNA in the dentate gyrus. Furthermore, nearly one quarter of DLB patients showed myoclonus, a clinical sign of aberrant network excitability that was associated with an earlier age of onset of cognitive impairments. In SYN mice, partial suppression of epileptiform activity did not alter their shift in spectral power. Furthermore, epileptiform activity in human amyloid precursor protein transgenic mice was not associated with a left shift in spectral power. INTERPRETATION: We conclude that neuronal accumulation of SYN slows brain oscillations and, in parallel, causes aberrant network excitability that can escalate into seizure activity. The potential role of aberrant network excitability in DLB merits further investigation.

14.
Ann Neurol ; 76(3): 443-56, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25042160

RESUMEN

OBJECTIVE: Reducing levels of the microtubule-associated protein tau has shown promise as a potential treatment strategy for diseases with secondary epileptic features such as Alzheimer disease. We wanted to determine whether tau reduction may also be of benefit in intractable genetic epilepsies. METHODS: We studied a mouse model of Dravet syndrome, a severe childhood epilepsy caused by mutations in the human SCN1A gene encoding the voltage-gated sodium channel subunit Nav 1.1. We genetically deleted 1 or 2 Tau alleles in mice carrying an Nav 1.1 truncation mutation (R1407X) that causes Dravet syndrome in humans, and examined their survival, epileptic activity, related hippocampal alterations, and behavioral abnormalities using observation, electroencephalographic recordings, acute slice electrophysiology, immunohistochemistry, and behavioral assays. RESULTS: Tau ablation prevented the high mortality of Dravet mice and reduced the frequency of spontaneous and febrile seizures. It reduced interictal epileptic spikes in vivo and drug-induced epileptic activity in brain slices ex vivo. Tau ablation also prevented biochemical changes in the hippocampus indicative of epileptic activity and ameliorated abnormalities in learning and memory, nest building, and open field behaviors in Dravet mice. Deletion of only 1 Tau allele was sufficient to suppress epileptic activity and improve survival and nesting performance. INTERPRETATION: Tau reduction may be of therapeutic benefit in Dravet syndrome and other intractable genetic epilepsies.


Asunto(s)
Epilepsias Mioclónicas/metabolismo , Hipocampo/metabolismo , Convulsiones/metabolismo , Proteínas tau/metabolismo , Alelos , Animales , Conducta Animal/fisiología , Modelos Animales de Enfermedad , Electroencefalografía , Epilepsias Mioclónicas/fisiopatología , Epilepsias Mioclónicas/terapia , Femenino , Hipocampo/patología , Hipocampo/fisiopatología , Aprendizaje/fisiología , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , Canal de Sodio Activado por Voltaje NAV1.1/genética , Convulsiones/etiología , Convulsiones/fisiopatología , Proteínas tau/genética
15.
Cell Rep ; 7(4): 1065-76, 2014 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-24813892

RESUMEN

Aging is the primary risk factor for cognitive decline, an emerging health threat to aging societies worldwide. Whether anti-aging factors such as klotho can counteract cognitive decline is unknown. We show that a lifespan-extending variant of the human KLOTHO gene, KL-VS, is associated with enhanced cognition in heterozygous carriers. Because this allele increased klotho levels in serum, we analyzed transgenic mice with systemic overexpression of klotho. They performed better than controls in multiple tests of learning and memory. Elevating klotho in mice also enhanced long-term potentiation, a form of synaptic plasticity, and enriched synaptic GluN2B, an N-methyl-D-aspartate receptor (NMDAR) subunit with key functions in learning and memory. Blockade of GluN2B abolished klotho-mediated effects. Surprisingly, klotho effects were evident also in young mice and did not correlate with age in humans, suggesting independence from the aging process. Augmenting klotho or its effects may enhance cognition and counteract cognitive deficits at different life stages.


Asunto(s)
Cognición/fisiología , Glucuronidasa/fisiología , Factores de Edad , Anciano , Anciano de 80 o más Años , Animales , Estudios de Cohortes , Femenino , Glucuronidasa/genética , Glucuronidasa/metabolismo , Humanos , Proteínas Klotho , Esperanza de Vida , Masculino , Memoria/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/metabolismo
16.
PLoS One ; 9(12): e115765, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25551452

RESUMEN

OBJECTIVE: Because reduction of the microtubule-associated protein Tau has beneficial effects in mouse models of Alzheimer's disease and epilepsy, we wanted to determine whether this strategy can also improve the outcome of mild traumatic brain injury (TBI). METHODS: We adapted a mild frontal impact model of TBI for wildtype C57Bl/6J mice and characterized the behavioral deficits it causes in these animals. The Barnes maze, Y maze, contextual and cued fear conditioning, elevated plus maze, open field, balance beam, and forced swim test were used to assess different behavioral functions. Magnetic resonance imaging (MRI, 7 Tesla) and histological analysis of brain sections were used to look for neuropathological alterations. We also compared the functional effects of this TBI model and of controlled cortical impact in mice with two, one or no Tau alleles. RESULTS: Repeated (2-hit), but not single (1-hit), mild frontal impact impaired spatial learning and memory in wildtype mice as determined by testing of mice in the Barnes maze one month after the injury. Locomotor activity, anxiety, depression and fear related behaviors did not differ between injured and sham-injured mice. MRI imaging did not reveal focal injury or mass lesions shortly after the injury. Complete ablation or partial reduction of tau prevented deficits in spatial learning and memory after repeated mild frontal impact. Complete tau ablation also showed a trend towards protection after a single controlled cortical impact. Complete or partial reduction of tau also reduced the level of axonopathy in the corpus callosum after repeated mild frontal impact. INTERPRETATION: Tau promotes or enables the development of learning and memory deficits and of axonopathy after mild TBI, and tau reduction counteracts these adverse effects.


Asunto(s)
Lesiones Encefálicas/fisiopatología , Trastornos de la Memoria/genética , Aprendizaje Espacial/fisiología , Proteínas tau/genética , Animales , Axones/patología , Cuerpo Calloso/patología , Modelos Animales de Enfermedad , Femenino , Imagen por Resonancia Magnética , Masculino , Aprendizaje por Laberinto/fisiología , Memoria , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
17.
Nat Neurosci ; 16(5): 613-21, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23525040

RESUMEN

We show that a natural behavior, exploration of a novel environment, causes DNA double-strand breaks (DSBs) in neurons of young adult wild-type mice. DSBs occurred in multiple brain regions, were most abundant in the dentate gyrus, which is involved in learning and memory, and were repaired within 24 h. Increasing neuronal activity by sensory or optogenetic stimulation increased neuronal DSBs in relevant but not irrelevant networks. Mice transgenic for human amyloid precursor protein (hAPP), which simulate key aspects of Alzheimer's disease, had increased neuronal DSBs at baseline and more severe and prolonged DSBs after exploration. Interventions that suppress aberrant neuronal activity and improve learning and memory in hAPP mice normalized their levels of DSBs. Blocking extrasynaptic NMDA-type glutamate receptors prevented amyloid-ß (Aß)-induced DSBs in neuronal cultures. Thus, transient increases in neuronal DSBs occur as a result of physiological brain activity, and Aß exacerbates DNA damage, most likely by eliciting synaptic dysfunction.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Roturas del ADN de Doble Cadena , Neuronas/fisiología , Precursor de Proteína beta-Amiloide/genética , Animales , Animales Recién Nacidos , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Channelrhodopsins , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Corticosterona/sangre , Modelos Animales de Enfermedad , Antagonistas de Aminoácidos Excitadores/farmacología , Conducta Exploratoria/efectos de los fármacos , Conducta Exploratoria/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/citología , Hipocampo/efectos de los fármacos , Histonas/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/efectos de los fármacos , Estimulación Luminosa , Quinoxalinas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Estrés Fisiológico/fisiología , Valina/análogos & derivados , Valina/farmacología , Proteínas tau/genética
18.
PLoS One ; 7(9): e45881, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23029293

RESUMEN

Accumulation of hyperphosphorylated tau in the entorhinal cortex (EC) is one of the earliest pathological hallmarks in patients with Alzheimer's disease (AD). It can occur before significant Aß deposition and appears to "spread" into anatomically connected brain regions. To determine whether this early-stage pathology is sufficient to cause disease progression and cognitive decline in experimental models, we overexpressed mutant human tau (hTauP301L) predominantly in layer II/III neurons of the mouse EC. Cognitive functions remained normal in mice at 4, 8, 12 and 16 months of age, despite early and extensive tau accumulation in the EC. Perforant path (PP) axon terminals within the dentate gyrus (DG) contained abnormal conformations of tau even in young EC-hTau mice, and phosphorylated tau increased with age in both the EC and PP. In old mice, ultrastructural alterations in presynaptic terminals were observed at PP-to-granule cell synapses. Phosphorylated tau was more abundant in presynaptic than postsynaptic elements. Human and pathological tau was also detected within hippocampal neurons of this mouse model. Thus, hTauP301L accumulation predominantly in the EC and related presynaptic pathology in hippocampal circuits was not sufficient to cause robust cognitive deficits within the age range analyzed here.


Asunto(s)
Corteza Entorrinal/metabolismo , Hipocampo/metabolismo , Mutación Missense , Vía Perforante/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Disfunción Cognitiva/genética , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/patología , Giro Dentado , Corteza Entorrinal/patología , Corteza Entorrinal/fisiopatología , Femenino , Expresión Génica , Hipocampo/patología , Hipocampo/fisiopatología , Humanos , Masculino , Aprendizaje por Laberinto , Memoria , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Reconocimiento Visual de Modelos , Vía Perforante/patología , Vía Perforante/fisiopatología , Reconocimiento en Psicología , Sinapsis/metabolismo , Sinapsis/patología , Proteínas tau/genética
19.
Proc Natl Acad Sci U S A ; 109(42): E2895-903, 2012 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-22869752

RESUMEN

In light of the rising prevalence of Alzheimer's disease (AD), new strategies to prevent, halt, and reverse this condition are needed urgently. Perturbations of brain network activity are observed in AD patients and in conditions that increase the risk of developing AD, suggesting that aberrant network activity might contribute to AD-related cognitive decline. Human amyloid precursor protein (hAPP) transgenic mice simulate key aspects of AD, including pathologically elevated levels of amyloid-ß peptides in brain, aberrant neural network activity, remodeling of hippocampal circuits, synaptic deficits, and behavioral abnormalities. Whether these alterations are linked in a causal chain remains unknown. To explore whether hAPP/amyloid-ß-induced aberrant network activity contributes to synaptic and cognitive deficits, we treated hAPP mice with different antiepileptic drugs. Among the drugs tested, only levetiracetam (LEV) effectively reduced abnormal spike activity detected by electroencephalography. Chronic treatment with LEV also reversed hippocampal remodeling, behavioral abnormalities, synaptic dysfunction, and deficits in learning and memory in hAPP mice. Our findings support the hypothesis that aberrant network activity contributes causally to synaptic and cognitive deficits in hAPP mice. LEV might also help ameliorate related abnormalities in people who have or are at risk for AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Anticonvulsivantes/farmacología , Trastornos del Conocimiento/tratamiento farmacológico , Cognición/efectos de los fármacos , Red Nerviosa/efectos de los fármacos , Piracetam/análogos & derivados , Sinapsis/efectos de los fármacos , Enfermedad de Alzheimer/complicaciones , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Análisis de Varianza , Animales , Anticonvulsivantes/sangre , Anticonvulsivantes/uso terapéutico , Western Blotting , Trastornos del Conocimiento/etiología , Electroencefalografía , Humanos , Inmunohistoquímica , Levetiracetam , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Transgénicos , Red Nerviosa/fisiopatología , Piracetam/sangre , Piracetam/farmacología , Piracetam/uso terapéutico
20.
Cell ; 149(3): 708-21, 2012 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-22541439

RESUMEN

Alzheimer's disease (AD) results in cognitive decline and altered network activity, but the mechanisms are unknown. We studied human amyloid precursor protein (hAPP) transgenic mice, which simulate key aspects of AD. Electroencephalographic recordings in hAPP mice revealed spontaneous epileptiform discharges, indicating network hypersynchrony, primarily during reduced gamma oscillatory activity. Because this oscillatory rhythm is generated by inhibitory parvalbumin (PV) cells, network dysfunction in hAPP mice might arise from impaired PV cells. Supporting this hypothesis, hAPP mice and AD patients had decreased levels of the interneuron-specific and PV cell-predominant voltage-gated sodium channel subunit Nav1.1. Restoring Nav1.1 levels in hAPP mice by Nav1.1-BAC expression increased inhibitory synaptic activity and gamma oscillations and reduced hypersynchrony, memory deficits, and premature mortality. We conclude that reduced Nav1.1 levels and PV cell dysfunction critically contribute to abnormalities in oscillatory rhythms, network synchrony, and memory in hAPP mice and possibly in AD.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Humanos , Técnicas In Vitro , Interneuronas/metabolismo , Aprendizaje , Memoria , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Canal de Sodio Activado por Voltaje NAV1.1 , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Canales de Sodio/metabolismo , Sinapsis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...