Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Transl Med ; 12(558)2020 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-32848093

RESUMO

A major sex difference in Alzheimer's disease (AD) is that men with the disease die earlier than do women. In aging and preclinical AD, men also show more cognitive deficits. Here, we show that the X chromosome affects AD-related vulnerability in mice expressing the human amyloid precursor protein (hAPP), a model of AD. XY-hAPP mice genetically modified to develop testicles or ovaries showed worse mortality and deficits than did XX-hAPP mice with either gonad, indicating a sex chromosome effect. To dissect whether the absence of a second X chromosome or the presence of a Y chromosome conferred a disadvantage on male mice, we varied sex chromosome dosage. With or without a Y chromosome, hAPP mice with one X chromosome showed worse mortality and deficits than did those with two X chromosomes. Thus, adding a second X chromosome conferred resilience to XY males and XO females. In addition, the Y chromosome, its sex-determining region Y gene (Sry), or testicular development modified mortality in hAPP mice with one X chromosome such that XY males with testicles survived longer than did XY or XO females with ovaries. Furthermore, a second X chromosome conferred resilience potentially through the candidate gene Kdm6a, which does not undergo X-linked inactivation. In humans, genetic variation in KDM6A was linked to higher brain expression and associated with less cognitive decline in aging and preclinical AD, suggesting its relevance to human brain health. Our study suggests a potential role for sex chromosomes in modulating disease vulnerability related to AD.


Assuntos
Doença de Alzheimer , Doença de Alzheimer/genética , Animais , Feminino , Masculino , Camundongos , Caracteres Sexuais , Testículo , Cromossomo X/genética , Cromossomo Y
2.
J Neurosci ; 38(15): 3680-3688, 2018 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-29540553

RESUMO

Hyperacetylation of tau has been implicated in neurodegeneration and cognitive decline in tauopathy brains. The nicotinamide adenosine dinucleotide-dependent class-III protein deacetylase SIRT1 is one of the major enzymes involved in removal of acetyl groups from tau in vitro However, whether SIRT1 regulates acetylation of pathogenic tau and ameliorates tau-mediated pathogenesis remains unclear. Here, we report deacetylating activity of SIRT1 for acetylated Lys174 (K174) of tau in tauP301S transgenic mice with a brain-specific SIRT1 deletion. We show that SIRT1 deficiency leads to exacerbation of premature mortality, synapse loss, and behavioral disinhibition in tauP301S transgenic mice of both sexes. By contrast, SIRT1 overexpression by stereotaxic delivery of adeno-associated virus that encodes SIRT1 into the hippocampus reduces acetylated K174 tau. Furthermore, SIRT1 overexpression significantly attenuates the spread of tau pathology into anatomically connected brain regions of tauP301S transgenic mice of both sexes. These findings suggest the functional importance of SIRT1 in regulating pathogenic tau acetylation and in suppressing the spread of tau pathology in vivoSIGNIFICANCE STATEMENT In neurodegenerative disorders with inclusions of microtubule-associated protein tau, aberrant lysine acetylation of tau plays critical roles in promoting tau accumulation and toxicity. Identifying strategies to deacetylate tau could interfere with disease progression; however, little is known about how pathogenic tau is deacetylated in vivo Here we show that the protein deacetylase SIRT1 reduces tau acetylation in a mouse model of neurodegeneration. SIRT1 deficiency in the brain aggravates synapse loss and behavioral disinhibition, and SIRT1 overexpression ameliorates propagation of tau pathology.


Assuntos
Sirtuína 1/metabolismo , Tauopatias/metabolismo , Proteínas tau/metabolismo , Acetilação , Animais , Feminino , Células HEK293 , Hipocampo/metabolismo , Hipocampo/patologia , Hipocampo/fisiopatologia , Humanos , Masculino , Aprendizagem em Labirinto , Camundongos , Sirtuína 1/genética , Transmissão Sináptica , Tauopatias/patologia , Tauopatias/fisiopatologia
3.
Neuromuscul Disord ; 27(7): 635-645, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28554556

RESUMO

Enzyme-linked and electrochemiluminescence immunoassays were developed for quantification of amino (N-) terminal fragments of the skeletal muscle protein titin (N-ter titin) and qualified for use in detection of urinary N-ter titin excretion. Urine from normal subjects contained a small but measurable level of N-ter titin (1.0 ± 0.4 ng/ml). A 365-fold increase (365.4 ± 65.0, P = 0.0001) in urinary N-ter titin excretion was seen in Duchene muscular dystrophy (DMD) patients. Urinary N-ter titin was also evaluated in dystrophin deficient rodent models. Mdx mice exhibited low urinary N-ter titin levels at 2 weeks of age followed by a robust and sustained elevation starting at 3 weeks of age, coincident with the development of systemic skeletal muscle damage in this model; fold elevation could not be determined because urinary N-ter titin was not detected in age-matched wild type mice. Levels of serum creatine kinase and serum skeletal muscle troponin I (TnI) were also low at 2 weeks, elevated at later time points and were significantly correlated with urinary N-ter titin excretion in mdx mice. Corticosteroid treatment of mdx mice resulted in improved exercise performance and lowering of both urinary N-ter titin and serum skeletal muscle TnI concentrations. Low urinary N-ter titin levels were detected in wild type rats (3.0 ± 0.6 ng/ml), while Dmdmdx rats exhibited a 556-fold increase (1652.5 ± 405.7 ng/ml, P = 0.002) (both at 5 months of age). These results suggest that urinary N-ter titin is present at low basal concentrations in normal urine and increases dramatically coincident with muscle damage produced by dystrophin deficiency. Urinary N-ter titin has potential as a facile, non-invasive and translational biomarker for DMD.


Assuntos
Conectina/urina , Distrofia Muscular de Duchenne/urina , Adolescente , Corticosteroides/uso terapêutico , Fatores Etários , Animais , Estudos de Casos e Controles , Criança , Pré-Escolar , Conectina/sangue , Creatina Quinase/sangue , Estudos Transversais , Humanos , Técnicas Imunoenzimáticas , Camundongos , Camundongos Endogâmicos mdx , Distrofia Muscular Animal/sangue , Distrofia Muscular Animal/tratamento farmacológico , Distrofia Muscular Animal/urina , Distrofia Muscular de Duchenne/sangue , Distrofia Muscular de Duchenne/genética
4.
Nat Commun ; 6: 8897, 2015 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-26615780

RESUMO

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.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/psicologia , Proteína BRCA1/deficiência , Encéfalo/metabolismo , Reparo do DNA , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/genética , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Proteína BRCA1/genética , Encéfalo/fisiopatologia , Cognição , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/metabolismo
5.
PLoS One ; 10(5): e0125614, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25933020

RESUMO

In Alzheimer's disease (AD), an extensive accumulation of extracellular amyloid plaques and intraneuronal tau tangles, along with neuronal loss, is evident in distinct brain regions. Staging of tau pathology by postmortem analysis of AD subjects suggests a sequence of initiation and subsequent spread of neurofibrillary tau tangles along defined brain anatomical pathways. Further, the severity of cognitive deficits correlates with the degree and extent of tau pathology. In this study, we demonstrate that phospho-tau (p-tau) antibodies, PHF6 and PHF13, can prevent the induction of tau pathology in primary neuron cultures. The impact of passive immunotherapy on the formation and spread of tau pathology, as well as functional deficits, was subsequently evaluated with these antibodies in two distinct transgenic mouse tauopathy models. The rTg4510 transgenic mouse is characterized by inducible over-expression of P301L mutant tau, and exhibits robust age-dependent brain tau pathology. Systemic treatment with PHF6 and PHF13 from 3 to 6 months of age led to a significant decline in brain and CSF p-tau levels. In a second model, injection of preformed tau fibrils (PFFs) comprised of recombinant tau protein encompassing the microtubule-repeat domains into the cortex and hippocampus of young P301S mutant tau over-expressing mice (PS19) led to robust tau pathology on the ipsilateral side with evidence of spread to distant sites, including the contralateral hippocampus and bilateral entorhinal cortex 4 weeks post-injection. Systemic treatment with PHF13 led to a significant decline in the spread of tau pathology in this model. The reduction in tau species after p-tau antibody treatment was associated with an improvement in novel-object recognition memory test in both models. These studies provide evidence supporting the use of tau immunotherapy as a potential treatment option for AD and other tauopathies.


Assuntos
Doença de Alzheimer/terapia , Anticorpos Monoclonais/farmacologia , Transtornos Cognitivos/terapia , Imunização Passiva , Fosfoproteínas/farmacologia , Proteínas tau/antagonistas & inibidores , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/imunologia , Doença de Alzheimer/patologia , Animais , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/imunologia , Córtex Cerebral/patologia , Transtornos Cognitivos/induzido quimicamente , Transtornos Cognitivos/imunologia , Transtornos Cognitivos/patologia , Modelos Animais de Doenças , Comportamento Exploratório/efeitos dos fármacos , Regulação da Expressão Gênica , Hipocampo/efeitos dos fármacos , Hipocampo/imunologia , Hipocampo/patologia , Masculino , Camundongos , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurônios/imunologia , Neurônios/patologia , Cultura Primária de Células , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/efeitos adversos , Transdução de Sinais , Resultado do Tratamento , Proteínas tau/genética , Proteínas tau/imunologia
6.
Nat Neurosci ; 18(3): 423-34, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25622143

RESUMO

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.


Assuntos
Astrócitos/metabolismo , Regulação da Expressão Gênica/fisiologia , Memória de Longo Prazo/fisiologia , Receptor A2A de Adenosina/metabolismo , Receptores 5-HT4 de Serotonina/metabolismo , Transdução de Sinais/fisiologia , Doença de Alzheimer/patologia , Animais , Animais Recém-Nascidos , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Comportamento Exploratório/efeitos dos fármacos , Comportamento Exploratório/fisiologia , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Indóis/farmacologia , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Receptor A2A de Adenosina/genética , Receptores 5-HT4 de Serotonina/genética , Reconhecimento Psicológico/efeitos dos fármacos , Reconhecimento Psicológico/fisiologia , Antagonistas da Serotonina/farmacologia , Sulfonamidas/farmacologia
7.
J Neurosci ; 35(2): 807-18, 2015 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-25589773

RESUMO

Aging is the predominant risk factor for neurodegenerative diseases. One key phenotype as the brain ages is an aberrant innate immune response characterized by proinflammation. However, the molecular mechanisms underlying aging-associated proinflammation are poorly defined. Whether chronic inflammation plays a causal role in cognitive decline in aging and neurodegeneration has not been established. Here we report a mechanistic link between chronic inflammation and aging microglia and a causal role of aging microglia in neurodegenerative cognitive deficits. We showed that SIRT1 is reduced with the aging of microglia and that microglial SIRT1 deficiency has a causative role in aging- or tau-mediated memory deficits via IL-1ß upregulation in mice. Interestingly, the selective activation of IL-1ß transcription by SIRT1 deficiency is likely mediated through hypomethylating the specific CpG sites on IL-1ß proximal promoter. In humans, hypomethylation of IL-1ß is strongly associated with chronological age and with elevated IL-1ß transcription. Our findings reveal a novel epigenetic mechanism in aging microglia that contributes to cognitive deficits in aging and neurodegenerative diseases.


Assuntos
Envelhecimento/metabolismo , Cognição , Epigênese Genética , Interleucina-1beta/metabolismo , Microglia/metabolismo , Sirtuína 1/metabolismo , Animais , Estudos de Casos e Controles , Metilação de DNA , Humanos , Interleucina-1beta/genética , Camundongos , Sirtuína 1/deficiência , Sirtuína 1/genética , Tauopatias/metabolismo , Regulação para Cima
8.
PLoS One ; 9(8): e106050, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25153994

RESUMO

Filamentous inclusions of the microtubule-associated protein, tau, define a variety of neurodegenerative diseases known as tauopathies, including Alzheimer's disease (AD). To better understand the role of tau-mediated effects on pathophysiology and global central nervous system function, we extensively characterized gene expression, pathology and behavior of the rTg4510 mouse model, which overexpresses a mutant form of human tau that causes Frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17). We found that the most predominantly altered gene expression pathways in rTg4510 mice were in inflammatory processes. These results closely matched the causal immune function and microglial gene-regulatory network recently identified in AD. We identified additional gene expression changes by laser microdissecting specific regions of the hippocampus, which highlighted alterations in neuronal network activity. Expression of inflammatory genes and markers of neuronal activity changed as a function of age in rTg4510 mice and coincided with behavioral deficits. Inflammatory changes were tau-dependent, as they were reversed by suppression of the tau transgene. Our results suggest that the alterations in microglial phenotypes that appear to contribute to the pathogenesis of Alzheimer's disease may be driven by tau dysfunction, in addition to the direct effects of beta-amyloid.


Assuntos
Doença de Alzheimer/genética , Expressão Gênica/genética , Redes Reguladoras de Genes/genética , Inflamação/genética , Proteínas tau/genética , Animais , Cromossomos Humanos Par 17/genética , Modelos Animais de Doenças , Feminino , Demência Frontotemporal/genética , Hipocampo/metabolismo , Humanos , Camundongos , Microglia/metabolismo , Proteínas Associadas aos Microtúbulos/genética , Doenças Neurodegenerativas/genética , Neurônios/metabolismo , Transtornos Parkinsonianos/genética
9.
Nat Neurosci ; 16(5): 613-21, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23525040

RESUMO

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.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Quebras de DNA de Cadeia Dupla , Neurônios/fisiologia , Precursor de Proteína beta-Amiloide/genética , Animais , Animais Recém-Nascidos , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Channelrhodopsins , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Corticosterona/sangue , Modelos Animais de Doenças , Antagonistas de Aminoácidos Excitatórios/farmacologia , Comportamento Exploratório/efeitos dos fármacos , Comportamento Exploratório/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Histonas/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Estimulação Luminosa , Quinoxalinas/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Estresse Fisiológico/fisiologia , Valina/análogos & derivados , Valina/farmacologia , Proteínas tau/genética
10.
Neurobiol Aging ; 34(6): 1523-9, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23332171

RESUMO

The microtubule-associated protein tau is expressed throughout the nervous system, most highly in neurons but also in glial cells. Its functions in adult and aging mammals remain to be defined. Previous studies in mouse models found either protective or detrimental effects of genetic tau ablation. Though tau ablation prevented synaptic, network, and cognitive dysfunctions in several models of Alzheimer's disease and made mice more resistant to epileptic seizures, a recent study described a parkinsonian phenotype in aging Tau knockout mice. Here we tested cognition and motor functions in Tau(+/+), Tau(+/-), and Tau(-/-) mice at approximately 1 and 2 years of age. Tau ablation did not impair cognition and caused only minor motor deficits that were much more subtle than those associated with the aging process. Tau ablation caused a mild increase in body weight, which correlated with and might have contributed to some of the motor deficits. However, tau ablation did not cause significant dopaminergic impairments, and dopamine treatment did not improve the motor deficits, suggesting that they do not reflect extrapyramidal dysfunction.


Assuntos
Envelhecimento/fisiologia , Envelhecimento/psicologia , Cognição/fisiologia , Dopamina , Transtornos das Habilidades Motoras/metabolismo , Proteínas tau/deficiência , Animais , Dopamina/fisiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transtornos das Habilidades Motoras/genética , Proteínas tau/genética
11.
PLoS One ; 7(9): e45881, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23029293

RESUMO

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.


Assuntos
Córtex Entorrinal/metabolismo , Hipocampo/metabolismo , Mutação de Sentido Incorreto , Via Perfurante/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Disfunção Cognitiva/genética , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/patologia , Giro Denteado , Córtex Entorrinal/patologia , Córtex Entorrinal/fisiopatologia , Feminino , Expressão Gênica , Hipocampo/patologia , Hipocampo/fisiopatologia , Humanos , Masculino , Aprendizagem em Labirinto , Memória , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Reconhecimento Visual de Modelos , Via Perfurante/patologia , Via Perfurante/fisiopatologia , Reconhecimento Psicológico , Sinapses/metabolismo , Sinapses/patologia , Proteínas tau/genética
12.
Proc Natl Acad Sci U S A ; 109(42): E2895-903, 2012 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-22869752

RESUMO

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.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Anticonvulsivantes/farmacologia , Transtornos Cognitivos/tratamento farmacológico , Cognição/efeitos dos fármacos , Rede Nervosa/efeitos dos fármacos , Piracetam/análogos & derivados , Sinapses/efeitos dos fármacos , Doença de Alzheimer/complicações , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Análise de Variância , Animais , Anticonvulsivantes/sangue , Anticonvulsivantes/uso terapêutico , Western Blotting , Transtornos Cognitivos/etiologia , Eletroencefalografia , Humanos , Imuno-Histoquímica , Levetiracetam , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Rede Nervosa/fisiopatologia , Piracetam/sangue , Piracetam/farmacologia , Piracetam/uso terapêutico
13.
PLoS One ; 7(7): e40555, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22792368

RESUMO

BACKGROUND: Although extensive research has demonstrated the importance of excitatory granule neurons in the dentate gyrus of the hippocampus in normal learning and memory and in the pathogenesis of amnesia in Alzheimer's disease (AD), the role of hilar GABAergic inhibitory interneurons, which control the granule neuron activity, remains unclear. METHODOLOGY AND PRINCIPAL FINDINGS: We explored the function of hilar GABAergic interneurons in spatial learning and memory by inhibiting their activity through Cre-dependent viral expression of enhanced halorhodopsin (eNpHR3.0)--a light-driven chloride pump. Hilar GABAergic interneuron-specific expression of eNpHR3.0 was achieved by bilaterally injecting adeno-associated virus containing a double-floxed inverted open-reading frame encoding eNpHR3.0 into the hilus of the dentate gyrus of mice expressing Cre recombinase under the control of an enhancer specific for GABAergic interneurons. In vitro and in vivo illumination with a yellow laser elicited inhibition of hilar GABAergic interneurons and consequent activation of dentate granule neurons, without affecting pyramidal neurons in the CA3 and CA1 regions of the hippocampus. We found that optogenetic inhibition of hilar GABAergic interneuron activity impaired spatial learning and memory retrieval, without affecting memory retention, as determined in the Morris water maze test. Importantly, optogenetic inhibition of hilar GABAergic interneuron activity did not alter short-term working memory, motor coordination, or exploratory activity. CONCLUSIONS AND SIGNIFICANCE: Our findings establish a critical role for hilar GABAergic interneuron activity in controlling spatial learning and memory retrieval and provide evidence for the potential contribution of GABAergic interneuron impairment to the pathogenesis of amnesia in AD.


Assuntos
Giro Denteado/metabolismo , Neurônios GABAérgicos/metabolismo , Interneurônios/metabolismo , Aprendizagem/fisiologia , Memória/fisiologia , Animais , Giro Denteado/citologia , Dependovirus/genética , Genes Reporter , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Memória de Curto Prazo/fisiologia , Camundongos , Camundongos Transgênicos , Desempenho Psicomotor
14.
Cell ; 149(3): 708-21, 2012 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-22541439

RESUMO

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.


Assuntos
Doença de Alzheimer/fisiopatologia , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Modelos Animais de Doenças , Hipocampo/metabolismo , Humanos , Técnicas In Vitro , Interneurônios/metabolismo , Aprendizagem , Memória , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Canal de Sódio Disparado por Voltagem NAV1.1 , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Canais de Sódio/metabolismo , Sinapses
15.
Physiol Behav ; 105(4): 915-24, 2012 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-22079582

RESUMO

Inter- and intra-species differences in social behavior and recognition-related hormones and receptors suggest that different distribution and/or expression patterns may relate to social recognition. We used qRT-PCR to investigate naturally occurring differences in expression of estrogen receptor-alpha (ERα), ER-beta (ERß), progesterone receptor (PR), oxytocin (OT) and receptor, and vasopressin (AVP) and receptors in proestrous female mice. Following four 5 min exposures to the same two conspecifics, one was replaced with a novel mouse in the final trial (T5). Gene expression was examined in mice showing high (85-100%) and low (40-60%) social recognition scores (i.e., preferential novel mouse investigation in T5) in eight socially-relevant brain regions. Results supported OT and AVP involvement in social recognition, and suggest that in the medial preoptic area, increased OT and AVP mRNA, together with ERα and ERß gene activation, relate to improved social recognition. Initial social investigation correlated with ERs, PR and OTR in the dorsolateral septum, suggesting that these receptors may modulate social interest without affecting social recognition. Finally, increased lateral amygdala gene activation in the LR mice may be associated with general learning impairments, while decreased lateral amygdala activity may indicate more efficient cognitive mechanisms in the HR mice.


Assuntos
Receptor alfa de Estrogênio/biossíntese , Receptor beta de Estrogênio/biossíntese , Ocitocina/biossíntese , Receptores de Ocitocina/biossíntese , Receptores de Vasopressinas/biossíntese , Reconhecimento Psicológico/fisiologia , Comportamento Social , Vasopressinas/biossíntese , Animais , Animais não Endogâmicos , Comportamento Animal/fisiologia , Encéfalo/metabolismo , Feminino , Expressão Gênica , Camundongos , Receptores de Progesterona/biossíntese
16.
J Neurosci ; 31(2): 700-11, 2011 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-21228179

RESUMO

Alzheimer's disease (AD), the most common neurodegenerative disorder, is a growing public health problem and still lacks effective treatments. Recent evidence suggests that microtubule-associated protein tau may mediate amyloid-ß peptide (Aß) toxicity by modulating the tyrosine kinase Fyn. We showed previously that tau reduction prevents, and Fyn overexpression exacerbates, cognitive deficits in human amyloid precursor protein (hAPP) transgenic mice overexpressing Aß. However, the mechanisms by which Aß, tau, and Fyn cooperate in AD-related pathogenesis remain to be fully elucidated. Here we examined the synaptic and network effects of this pathogenic triad. Tau reduction prevented cognitive decline induced by synergistic effects of Aß and Fyn. Tau reduction also prevented synaptic transmission and plasticity deficits in hAPP mice. Using electroencephalography to examine network effects, we found that tau reduction prevented spontaneous epileptiform activity in multiple lines of hAPP mice. Tau reduction also reduced the severity of spontaneous and chemically induced seizures in mice overexpressing both Aß and Fyn. To better understand these protective effects, we recorded whole-cell currents in acute hippocampal slices from hAPP mice with and without tau. hAPP mice with tau had increased spontaneous and evoked excitatory currents, reduced inhibitory currents, and NMDA receptor dysfunction. Tau reduction increased inhibitory currents and normalized excitation/inhibition balance and NMDA receptor-mediated currents in hAPP mice. Our results indicate that Aß, tau, and Fyn jointly impair synaptic and network function and suggest that disrupting the copathogenic relationship between these factors could be of therapeutic benefit.


Assuntos
Doença de Alzheimer/fisiopatologia , Doença de Alzheimer/psicologia , Peptídeos beta-Amiloides/fisiologia , Transtornos Cognitivos/fisiopatologia , Rede Nervosa/fisiologia , Proteínas Proto-Oncogênicas c-fyn/fisiologia , Sinapses/fisiologia , Proteínas tau/metabolismo , Doença de Alzheimer/metabolismo , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/mortalidade , Animais , Transtornos Cognitivos/metabolismo , Transtornos Cognitivos/psicologia , Modelos Animais de Doenças , Eletroencefalografia , Feminino , Hipocampo/fisiopatologia , Técnicas In Vitro , Masculino , Camundongos , Camundongos Mutantes , Plasticidade Neuronal , Convulsões/metabolismo , Convulsões/fisiopatologia , Especificidade da Espécie , Transmissão Sináptica , Proteínas tau/genética
17.
Nature ; 469(7328): 47-52, 2011 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-21113149

RESUMO

Amyloid-ß oligomers may cause cognitive deficits in Alzheimer's disease by impairing neuronal NMDA-type glutamate receptors, whose function is regulated by the receptor tyrosine kinase EphB2. Here we show that amyloid-ß oligomers bind to the fibronectin repeats domain of EphB2 and trigger EphB2 degradation in the proteasome. To determine the pathogenic importance of EphB2 depletions in Alzheimer's disease and related models, we used lentiviral constructs to reduce or increase neuronal expression of EphB2 in memory centres of the mouse brain. In nontransgenic mice, knockdown of EphB2 mediated by short hairpin RNA reduced NMDA receptor currents and impaired long-term potentiation in the dentate gyrus, which are important for memory formation. Increasing EphB2 expression in the dentate gyrus of human amyloid precursor protein transgenic mice reversed deficits in NMDA receptor-dependent long-term potentiation and memory impairments. Thus, depletion of EphB2 is critical in amyloid-ß-induced neuronal dysfunction. Increasing EphB2 levels or function could be beneficial in Alzheimer's disease.


Assuntos
Doença de Alzheimer/fisiopatologia , Doença de Alzheimer/terapia , Cognição/fisiologia , Receptor EphB2/deficiência , Receptor EphB2/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Giro Denteado/metabolismo , Modelos Animais de Doenças , Humanos , Potenciação de Longa Duração , Memória/fisiologia , Camundongos , Camundongos Transgênicos , Plasticidade Neuronal , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Ratos , Receptor EphB2/química , Receptor EphB2/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapses/metabolismo
18.
Neuron ; 68(3): 428-41, 2010 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-21040845

RESUMO

The entorhinal cortex (EC) is one of the earliest affected, most vulnerable brain regions in Alzheimer's disease (AD), which is associated with amyloid-ß (Aß) accumulation in many brain areas. Selective overexpression of mutant amyloid precursor protein (APP) predominantly in layer II/III neurons of the EC caused cognitive and behavioral abnormalities characteristic of mouse models with widespread neuronal APP overexpression, including hyperactivity, disinhibition, and spatial learning and memory deficits. APP/Aß overexpression in the EC elicited abnormalities in synaptic functions and activity-related molecules in the dentate gyrus and CA1 and epileptiform activity in parietal cortex. Soluble Aß was observed in the dentate gyrus, and Aß deposits in the hippocampus were localized to perforant pathway terminal fields. Thus, APP/Aß expression in EC neurons causes transsynaptic deficits that could initiate the cortical-hippocampal network dysfunction in mouse models and human patients with AD.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Córtex Entorrinal/patologia , Hipocampo/patologia , Rede Nervosa/patologia , Neurônios/efeitos dos fármacos , Sinapses/patologia , Doença de Alzheimer/psicologia , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/fisiologia , Animais , Comportamento Animal/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Transtornos Cognitivos/psicologia , Progressão da Doença , Eletroencefalografia , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Aprendizagem em Labirinto/fisiologia , Memória/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo , Placa Amiloide/patologia
19.
Neurosci Lett ; 474(1): 17-21, 2010 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-20193744

RESUMO

In the CNS, lipocalin-type prostaglandin D synthase (L-PGDS) is predominantly a non-neuronal enzyme responsible for the production of PGD(2), an endogenous sleep promoting substance. We have previously demonstrated that estradiol differentially regulates L-PGDS transcript levels in the rodent brain. In hypothalamic nuclei, estradiol increases L-PGDS transcript expression, whereas in the ventrolateral preoptic area L-PGDS gene expression is reduced after estradiol treatment. In the present study, we have used an immortalized glioma cell line transfected with a L-PGDS reporter construct and estrogen receptor (ER) alpha and ERbeta expression plasmids to further elucidate the mechanisms underlying estradiol regulation of L-PGDS gene expression. We found that physiologically relevant concentrations of estradiol evoked an inverted U response in cells expressing ERalpha. The most effective concentration of estradiol (10(-11)M) increased the promoter activity 3-fold over baseline. Expression of ERbeta did not increase activity over control and when ERbeta was co-expressed with ERalpha there was a significant attenuation of the promoter activity. While ERalpha significantly increased L-PGDS promoter activity, our previous in vivo studies demonstrate a greater magnitude of change in L-PGDS gene expression in the presences of estradiol. This led us to ask whether estradiol is signaling via a paracrine factor released by the neighboring neurons. Conditioned media from estradiol treated neurons applied to the glioma cell line resulted in a significant 7-fold increase in L-PGDS promoter activity supporting the possibility that neuronal-glial interactions are involved in estradiol regulation of L-PGDS.


Assuntos
Estradiol/farmacologia , Oxirredutases Intramoleculares/biossíntese , Lipocalinas/biossíntese , Fatores Biológicos/metabolismo , Linhagem Celular Tumoral , Meios de Cultivo Condicionados , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/fisiologia , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/fisiologia , Regulação Enzimológica da Expressão Gênica , Humanos , Oxirredutases Intramoleculares/genética , Lipocalinas/genética , Neuroglia/metabolismo , Neurônios/metabolismo , Comunicação Parácrina , Transcrição Gênica
20.
J Neurosci ; 30(1): 372-81, 2010 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-20053918

RESUMO

Previous studies suggested that cleavage of the amyloid precursor protein (APP) at aspartate residue 664 by caspases may play a key role in the pathogenesis of Alzheimer's disease. Mutation of this site (D664A) prevents caspase cleavage and the generation of the C-terminal APP fragments C31 and Jcasp, which have been proposed to mediate amyloid-beta (Abeta) neurotoxicity. Here we compared human APP transgenic mice with (B254) and without (J20) the D664A mutation in a battery of tests. Before Abeta deposition, hAPP-B254 and hAPP-J20 mice had comparable hippocampal levels of Abeta(1-42). At 2-3 or 5-7 months of age, hAPP-B254 and hAPP-J20 mice had similar abnormalities relative to nontransgenic mice in spatial and nonspatial learning and memory, elevated plus maze performance, electrophysiological measures of synaptic transmission and plasticity, and levels of synaptic activity-related proteins. Thus, caspase cleavage of APP at position D664 and generation of C31 do not play a critical role in the development of these abnormalities.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Caspases/metabolismo , Modelos Animais de Doenças , Neurônios/metabolismo , Desempenho Psicomotor/fisiologia , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Animais , Humanos , Hidrólise , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Neurônios/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...