Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neurobiol Dis ; 186: 106263, 2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37591465

RESUMO

The R47H variant of triggering receptor expressed on myeloid cells 2 (TREM2) increases the risk of Alzheimer's disease (AD). To investigate potential mechanisms, we analyzed knockin mice expressing human TREM2-R47H from one mutant mouse Trem2 allele. TREM2-R47H mice showed increased seizure activity in response to an acute excitotoxin challenge, compared to wildtype controls or knockin mice expressing the common variant of human TREM2. TREM2-R47H also increased spontaneous thalamocortical epileptiform activity in App knockin mice expressing amyloid precursor proteins bearing autosomal dominant AD mutations and a humanized amyloid-ß sequence. In mice with or without such App modifications, TREM2-R47H increased the density of putative synapses in cortical regions without amyloid plaques. TREM2-R47H did not affect synaptic density in hippocampal regions with or without plaques. We conclude that TREM2-R47H increases AD-related network hyperexcitability and that it may do so, at least in part, by causing an imbalance in synaptic densities across brain regions.


Assuntos
Doença de Alzheimer , Humanos , Animais , Camundongos , Doença de Alzheimer/genética , Alelos , Convulsões , Peptídeos beta-Amiloides , Modelos Animais de Doenças , Placa Amiloide , Sinapses , Glicoproteínas de Membrana/genética , Receptores Imunológicos/genética
2.
Sci Transl Med ; 14(642): eabm5527, 2022 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-35476595

RESUMO

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.


Assuntos
Transtorno Autístico , Epilepsias Mioclônicas , Epilepsia , Morte Súbita Inesperada na Epilepsia , Proteínas tau , Animais , Transtorno Autístico/complicações , Transtorno Autístico/genética , Modelos Animais de Doenças , Epilepsias Mioclônicas/complicações , Epilepsias Mioclônicas/genética , Epilepsia/complicações , Epilepsia/genética , Epilepsia/metabolismo , Síndromes Epilépticas , Humanos , Lactente , Camundongos , Neurônios/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Espasmos Infantis , Proteínas tau/metabolismo
3.
iScience ; 24(11): 103245, 2021 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-34755090

RESUMO

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.

4.
Cell Rep ; 37(3): 109855, 2021 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-34686344

RESUMO

The protein tau has been implicated in many brain disorders. In animal models, tau reduction suppresses epileptogenesis of diverse causes and ameliorates synaptic and behavioral abnormalities in various conditions associated with excessive excitation-inhibition (E/I) ratios. However, the underlying mechanisms are unknown. Global genetic ablation of tau in mice reduces the action potential (AP) firing and E/I ratio of pyramidal cells in acute cortical slices without affecting the excitability of these cells. Tau ablation reduces the excitatory inputs to inhibitory neurons, increases the excitability of these cells, and structurally alters their axon initial segments (AISs). In primary neuronal cultures subjected to prolonged overstimulation, tau ablation diminishes the homeostatic response of AISs in inhibitory neurons, promotes inhibition, and suppresses hypersynchrony. Together, these differential alterations in excitatory and inhibitory neurons help explain how tau reduction prevents network hypersynchrony and counteracts brain disorders causing abnormally increased E/I ratios.


Assuntos
Potenciais Pós-Sinápticos Excitadores , Potenciais Pós-Sinápticos Inibidores , Interneurônios/metabolismo , Inibição Neural , Vias Neurais/metabolismo , Células Piramidais/metabolismo , Córtex Somatossensorial/metabolismo , Proteínas tau/deficiência , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Animais , Células Cultivadas , Epilepsia/genética , Epilepsia/metabolismo , Epilepsia/fisiopatologia , Feminino , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Vias Neurais/citologia , Plasticidade Neuronal , Córtex Somatossensorial/citologia , Fatores de Tempo , Proteínas tau/genética
5.
J Genet Genomics ; 48(8): 695-705, 2021 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-34315684

RESUMO

Coordination between the sporophytic tissue and the gametic pollen within anthers is tightly controlled to achieve the optimal pollen fitness. Glucose-6-phosphate/phosphate translocator (GPT) transports glucose-6-phosphate, a key precursor of starch and/or fatty acid biosynthesis, into plastids. Here, we report the functional characterization of OsGPT1 in the rice anther development and pollen fertility. Pollen grains from homozygous osgpt1 mutant plants fail to accumulate starch granules, resulting in pollen sterility. Genetic analyses reveal a sporophytic effect for this mutation. OsGPT1 is highly expressed in the tapetal layer of rice anther. Degeneration of the tapetum, an important process to provide cellular contents to support pollen development, is impeded in osgpt1 plants. In addition, defective intine and exine are observed in the pollen from osgpt1 plants. Expression levels of multiple genes that are important to tapetum degeneration or pollen wall formation are significantly decreased in osgpt1 anthers. Previously, we reported that AtGPT1 plays a gametic function in the accumulation of lipid bodies in Arabidopsis pollen. This report highlights a sporophytic role of OsGPT1 in the tapetum degeneration and pollen development. The divergent functions of OsGPT1 and AtGPT1 in pollen development might be a result of their independent evolution after monocots and dicots diverged.


Assuntos
Glucose-6-Fosfato
6.
eNeuro ; 8(3)2021.
Artigo em Inglês | MEDLINE | ID: mdl-33833046

RESUMO

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.


Assuntos
Doença de Alzheimer , Precursor de Proteína beta-Amiloide , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fenótipo , Fatores de Transcrição
7.
Mol Neurodegener ; 15(1): 53, 2020 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-32921309

RESUMO

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.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Comportamento Animal/fisiologia , Encéfalo/metabolismo , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Rede Nervosa/metabolismo , Rede Nervosa/patologia
8.
Mol Plant ; 13(11): 1608-1623, 2020 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-32916336

RESUMO

The mitotic activity of root apical meristem (RAM) is critical to primary root growth and development. Previous studies have identified the roles of ROOT GROWTH FACTOR 1 (RGF1), a peptide ligand, and its receptors, RGF1 INSENSITIVEs (RGIs), a clade of five leucine-rich-repeat receptor-like kinases, in promoting cell division in the RAM, which determines the primary root length. However, the downstream signaling components remain elusive. In this study, we identify a complete mitogen-activated protein kinase (MAPK or MPK) cascade, composed of YDA, MKK4/MKK5, and MPK3/MPK6, that functions downstream of the RGF1-RGI ligand-receptor pair. Similar to the rgi1/2/3/4/5 quintuple mutant, loss-of-function mutants of MPK3 and MPK6, MKK4 and MKK5, or YDA show a short-root phenotype, which is associated with reduced mitotic activity and lower expression of PLETHORA 1 (PLT1)/PLT2 in the RAM. Furthermore, MPK3/MPK6 activation in response to exogenous RGF1 treatment is impaired in the rgi1/2/3/4/5 quintuple, yda single, and mkk4 mkk5 double mutants. Epistatic analyses demonstrated that the expression of constitutively active MKK4, MKK5, or YDA driven by the RGI2 promoter can rescue the short-root phenotype of the rgi1/2/3/4/5 mutant. Taken together, these results suggest that the YDA-MKK4/MKK5-MPK3/MPK6 cascade functions downstream of the RGF1-RGI ligand-receptor pair and upstream of PLT1/PLT2 to modulate the stem cell population and primary root growth in Arabidopsis.


Assuntos
Proteínas de Arabidopsis/metabolismo , Arabidopsis/fisiologia , Sistema de Sinalização das MAP Quinases , Meristema/fisiologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mitose/fisiologia , Peptídeos/metabolismo , Raízes de Plantas/fisiologia , Arabidopsis/genética , Arabidopsis/metabolismo , Ligantes , MAP Quinase Quinase Quinases/metabolismo , Mutação , Raízes de Plantas/metabolismo , Fatores de Transcrição/metabolismo
9.
Neuron ; 106(3): 421-437.e11, 2020 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-32126198

RESUMO

Autism is characterized by repetitive behaviors, impaired social interactions, and communication deficits. It is a prevalent neurodevelopmental disorder, and available treatments offer little benefit. Here, we show that genetically reducing the protein tau prevents behavioral signs of autism in two mouse models simulating distinct causes of this condition. Similar to a proportion of people with autism, both models have epilepsy, abnormally enlarged brains, and overactivation of the phosphatidylinositol 3-kinase (PI3K)/Akt (protein kinase B)/ mammalian target of rapamycin (mTOR) signaling pathway. All of these abnormalities were prevented or markedly diminished by partial or complete genetic removal of tau. We identify disinhibition of phosphatase and tensin homolog deleted on chromosome 10 (PTEN), a negative PI3K regulator that tau controls, as a plausible mechanism and demonstrate that tau interacts with PTEN via tau's proline-rich domain. Our findings suggest an enabling role of tau in the pathogenesis of autism and identify tau reduction as a potential therapeutic strategy for some of the disorders that cause this condition.


Assuntos
Transtorno Autístico/genética , Megalencefalia/genética , Proteínas tau/genética , Animais , Transtorno Autístico/metabolismo , Sítios de Ligação , Encéfalo/metabolismo , Encéfalo/patologia , Células Cultivadas , Células HEK293 , Humanos , Megalencefalia/metabolismo , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , PTEN Fosfo-Hidrolase/química , PTEN Fosfo-Hidrolase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Domínios Proteicos Ricos em Prolina , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Proteínas tau/metabolismo
10.
Neurobiol Dis ; 117: 181-188, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29859869

RESUMO

Neural network dysfunction may contribute to functional decline and disease progression in neurodegenerative disorders. Diverse lines of evidence suggest that neuronal accumulation of tau promotes network dysfunction and cognitive decline. The A152T-variant of human tau (hTau-A152T) increases the risk of Alzheimer's disease (AD) and several other tauopathies. When overexpressed in neurons of transgenic mice, it causes age-dependent neuronal loss and cognitive decline, as well as non-convulsive epileptic activity, which is also seen in patients with AD. Using intracranial EEG recordings with electrodes implanted over the parietal cortex, we demonstrate that hTau-A152T increases the power of brain oscillations in the 0.5-6 Hz range more than wildtype human tau in transgenic lines with comparable levels of human tau protein in brain, and that genetic ablation of endogenous tau in Mapt-/- mice decreases the power of these oscillations as compared to wildtype controls. Suppression of hTau-A152T production in doxycycline-regulatable transgenic mice reversed their abnormal network activity. Treatment of hTau-A152T mice with the antiepileptic drug levetiracetam also rapidly and persistently reversed their brain dysrhythmia and network hypersynchrony. These findings suggest that both the level and the sequence of tau modulate the power of specific brain oscillations. The potential of EEG spectral changes as a biomarker deserves to be explored in clinical trials of tau-lowering therapeutics. Our results also suggest that levetiracetam treatment is able to counteract tau-dependent neural network dysfunction. Tau reduction and levetiracetam treatment may be of benefit in AD and other conditions associated with brain dysrhythmias and network hypersynchrony.


Assuntos
Encéfalo/metabolismo , Ritmo Delta/fisiologia , Neurônios/metabolismo , Ritmo Teta/fisiologia , Proteínas tau/metabolismo , Animais , Encéfalo/patologia , Ondas Encefálicas/fisiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/patologia
11.
Cell Metab ; 26(3): 547-557.e8, 2017 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-28877458

RESUMO

Ketogenic diets recapitulate certain metabolic aspects of dietary restriction such as reliance on fatty acid metabolism and production of ketone bodies. We investigated whether an isoprotein ketogenic diet (KD) might, like dietary restriction, affect longevity and healthspan in C57BL/6 male mice. We find that Cyclic KD, KD alternated weekly with the Control diet to prevent obesity, reduces midlife mortality but does not affect maximum lifespan. A non-ketogenic high-fat diet (HF) fed similarly may have an intermediate effect on mortality. Cyclic KD improves memory performance in old age, while modestly improving composite healthspan measures. Gene expression analysis identifies downregulation of insulin, protein synthesis, and fatty acid synthesis pathways as mechanisms common to KD and HF. However, upregulation of PPARα target genes is unique to KD, consistent across tissues, and preserved in old age. In all, we show that a non-obesogenic ketogenic diet improves survival, memory, and healthspan in aging mice.


Assuntos
Envelhecimento/fisiologia , Dieta Cetogênica , Memória/fisiologia , Mortalidade , Animais , Dieta Hiperlipídica , Jejum , Regulação da Expressão Gênica , Fígado/metabolismo , Masculino , Camundongos Endogâmicos C57BL , PPAR alfa/genética , PPAR alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...