Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Brain Commun ; 6(1): fcae014, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38347943

RESUMO

This scientific commentary refers to 'Path integration deficits are associated with phosphorylated tau accumulation in the entorhinal cortex', by Koike et al. (https://doi.org/10.1093/braincomms/fcad359).

2.
Neuroscience ; 479: 91-106, 2021 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-34762981

RESUMO

Like other members of the superfamily of nuclear receptors, the peroxisome proliferator-activated receptor γ (PPARγ), is a ligand-activated transcription factor known for its insulin-sensitizing actions in the periphery. Despite only sparse evidence for PPARγ in the CNS, many reports suggest direct PPARγ-mediated actions in the brain. This study aimed to (i) map PPARγ expression in rodent brain areas, involved in the regulation of cognitive, motivational, and emotional functions, (ii) examine the regulation of central PPARγ by physiological variables (age, sex, obesity); (iii) chemotypically identify PPARγ-expressing cells in the frontal cortex (FC) and hippocampus (HP); (iv) study whether activation of PPARγ by pioglitazone (Pio) in FC and HP cells can induce target gene expression; and (v) demonstrate the impact of activated PPARγ on learning behavior and motivation. Immunoreactive PPARγ was detectable in specific sub-nuclei/subfields of the FC, HP, nucleus accumbens, amygdala, hypothalamus, thalamus, and granular layers of the cerebellum. PPARγ protein levels were upregulated during aging and in high fat diet-induced obesity. PPARγ mRNA expression was upregulated in the amygdala of females (but not males) that were made obese. Neural precursor cells, mature neurons, and astrocytes in primary FC and HP cultures were shown to express PPARγ. Pioglitazone dose-dependently upregulated PPARγ target genes in manner that was specific to the origin (FC or HP) of the cultures. Lastly, administration of Pio impaired motivation and associative learning. Collectively, we provide evidence for the presence of regulatable PPARγ in the brain and demonstrate their participation the regulation of key behaviors.


Assuntos
Células-Tronco Neurais , Tiazolidinedionas , Encéfalo/metabolismo , Feminino , Humanos , Masculino , Motivação , Células-Tronco Neurais/metabolismo , PPAR gama/metabolismo , Pioglitazona/farmacologia , Tiazolidinedionas/farmacologia
3.
Front Behav Neurosci ; 15: 812184, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35295248

RESUMO

Labels serve as identifiers and convenient descriptors of inanimate and animate objects. In humans, given labels can easily become part of an individual's self-perceived identity. Negative labels ascribed to a person can result in internalized stigma, a state that will shape the subject's biography. This can ultimately impact the person's mental and physical health since perceived and/or anticipated stigma discourages the use of social and health services. Per definition, stigma involves labeling of persons with physical, mental, or social characteristics that do not match the observer's arbitrarily conditioned and calibrated sense of norms (public stigma); such labeling may eventually become embedded in rules, regulations, and laws (structural stigma). Internalized stigma projects onto a person's emotions and actions. Public (enacted) stigma results from stereotyping (collectively agreed-upon notions about a group of persons that are used to categorize these people) and devaluation, which subsequently leads to social distancing, discrimination, and blatant abuse of human rights. Much of what we know about stigma results from research in the psychosocial sciences and, more recently, from social neuroscience. The stigma around mental health has generated much attention in the field of psychiatry where, to date, most research has focussed on epidemiology and anti-stigma interventions. This essay intends to stimulate thought, debate, and research within the behavioral neuroscience community and, therefore, to inform evidence-based design and implementation of neuroscience-based approaches by other professionals working towards the elimination of the stigma attached to mental illness. The article starts by considering the concept of stigma and the psychological processes that give rise to the phenomenon; it also considers how projected and perceived stigma are multiplied. Finally, after a brief review of the few existing neuroscientific explorations of stigma, gaps in our knowledge of the neurobiological basis of stigma are identified and discussed.

4.
Aging Brain ; 1: 100006, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-36911508
6.
Acta Physiol (Oxf) ; 228(2): e13345, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31310704

RESUMO

AIM: Since foods with high hedonic value are often consumed in excess of energetic needs, this study was designed to identify the mechanisms that may counter anorexigenic signalling in the presence of hedonic foods in lean animals. METHODS: Mice, in different states of satiety (fed/fasted, or fed/fasted and treated with ghrelin or leptin, respectively), were allowed to choose between high-fat/high-sucrose and standard foods. Intake of each food type and the activity of hypothalamic neuropetidergic neurons that regulate appetite were monitored. In some cases, food choice was monitored in leptin-injected fasted mice that received microinjections of galanin receptor agonists into the lateral hypothalamus. RESULTS: Appetite-stimulating orexin neurons in the lateral hypothalamus are rapidly activated when lean, satiated mice consume a highly palatable food (PF); such activation (upregulated c-Fos expression) occurred even after administration of the anorexigenic hormone leptin and despite intact leptin signalling in the hypothalamus. The ability of leptin to restrain PF eating is restored when a galanin receptor 2 (Gal2R) agonist is injected into the lateral hypothalamus. CONCLUSION: Hedonically-loaded foods interrupt the inhibitory actions of leptin on orexin neurons and interfere with the homeostatic control of feeding. Overeating of palatable foods can be curtailed in lean animals by activating Gal2R in the lateral hypothalamus.


Assuntos
Ingestão de Alimentos/fisiologia , Hiperfagia/prevenção & controle , Região Hipotalâmica Lateral/efeitos dos fármacos , Leptina/farmacologia , Neurônios/metabolismo , Receptor Tipo 2 de Galanina/agonistas , Animais , Modelos Animais de Doenças , Ingestão de Alimentos/efeitos dos fármacos , Galanina/farmacologia , Grelina/metabolismo , Hiperfagia/metabolismo , Hiperfagia/patologia , Região Hipotalâmica Lateral/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Orexinas/metabolismo , Receptor Tipo 2 de Galanina/metabolismo
7.
Front Cell Neurosci ; 13: 239, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31263400

RESUMO

Type 2 diabetes increases the risk for dementia, including Alzheimer's disease (AD). Pioglitazone (Pio), a pharmacological agonist of the peroxisome proliferator-activated receptor γ (PPARγ), improves insulin sensitivity and has been suggested to have potential in the management of AD symptoms, albeit through mostly unknown mechanisms. We here investigated the potential of Pio to counter synaptic malfunction and loss, a characteristic of AD pathology and its accompanying cognitive deficits. Results from experiments on primary mouse neuronal cultures and a human neural cell line (SH-SY5Y) show that Pio treatment attenuates amyloid ß (Aß)-triggered the pathological (mis-) processing of amyloid precursor protein (APP) and inhibits Aß-induced accumulation and hyperphosphorylation of Tau. These events are accompanied by increased glutamatergic receptor 2B subunit (GluN2B) levels that are causally linked with neuronal death. Further, Pio treatment blocks Aß-triggered missorting of hyperphosphorylated Tau to synapses and the subsequent loss of PSD95-positive synapses. These latter effects of Pio are PPARγ-mediated since they are blocked in the presence of GW9662, a selective PPARγ inhibitor. Collectively, these data show that activated PPARγ buffer neurons against APP misprocessing, Tau hyperphosphorylation and its missorting to synapses and subsequently, synaptic loss. These first insights into the mechanisms through which PPARγ influences synaptic loss make a case for further exploration of the potential usefulness of PPARγ agonists in the prevention and treatment of synaptic pathology in AD.

8.
Eur J Neurosci ; 50(5): 2773-2785, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31231836

RESUMO

Chronic stress is a major risk factor for developing Alzheimer's disease (AD) and promotes the processing of amyloid precursor protein (APP) to ß-amyloid (Aß). However, the precise relationship of stress and disease-typical cognitive decline is presently not well understood. The aim of this study was to investigate how early life stress may affect cognition in adult mice with and without soluble Aß pathology typical for the early stages of the disease. We focussed on sustained attention and response control, aspects of cognition mediated by the prefrontal cortex that are consistently impaired both in early AD and after chronic stress exposure. Young wild-type mice as well as transgenic arcAß mice overexpressing the hAPParc/swe transgene were exposed to a chronic unpredictable stress paradigm (age 3-8 weeks). At 15 weeks, these mice were tested on the 5-choice serial reaction time task, a test of sustained attention and executive control. We found that, expectedly, chronic stress increased impulsive choices and impaired sustained attention in wild-type mice. However, the same treatment reduced impulsivity and did not interfere with sustained attention in arcAß mice. These findings suggest an unexpected interaction between chronic stress and Aß whereby Aß-pathology caused by the hAPParc/swe mutation prevented and/or reversed stress-induced cognitive changes through mechanisms that deserve further investigation. They also indicate that Aß, in modest amounts, may have a beneficial role for cognitive stability, for example by protecting neural networks from the impact of further physiological or behavioural stress.


Assuntos
Precursor de Proteína beta-Amiloide/genética , Cognição/fisiologia , Função Executiva/fisiologia , Estresse Psicológico/genética , Doença de Alzheimer/genética , Animais , Atenção/fisiologia , Comportamento Animal/fisiologia , Modelos Animais de Doenças , Comportamento Impulsivo/fisiologia , Masculino , Camundongos , Mutação , Tempo de Reação/fisiologia
9.
Adv Exp Med Biol ; 1184: 241-257, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-32096043

RESUMO

Alzheimer's disease (AD) is a multifactorial neurodegenerative disorder with a complex physiopathology whose initiators are poorly defined. Accumulating clinical and experimental evidence suggests a causal role of lifetime stress in AD. This chapter summarizes current knowledge about how chronic stress and its accompanying high levels of glucocorticoid (GC) secretion, trigger the two main pathomechanisms of AD: (i) misprocessing of amyloid precursor protein (APP) and the generation of amyloid beta (Aß) and (ii) Tau hyperphosphorylation and aggregation. Given that depression is a well-known stress-related illness, and the evidence that depression may precede AD, this chapter also explores neurobiological mechanisms that may be common to depressive and AD pathologies. This review also discusses emerging insights into the role of Tau and its malfunction in disrupting neuronal cascades and neuroplasticity and, thus triggering brain pathology.


Assuntos
Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Depressão/complicações , Depressão/etiologia , Estresse Psicológico/complicações , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/metabolismo , Depressão/metabolismo , Depressão/patologia , Humanos , Fosforilação , Proteínas tau/química , Proteínas tau/metabolismo
10.
Front Neurosci ; 12: 124, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29545742

RESUMO

Hippocampal hyperactivity, ascribed to amyloid ß (Aß)-induced imbalances in neural excitation and inhibition, is found in patients with mild cognitive impairment, a prodromal stage of Alzheimer's disease (AD). To better understand the relationship between hippocampal hyperactivity and the molecular triggers of behavioral impairments in AD, we used Mn-enhanced MRI (MEMRI) to assess neuronal activity after subjecting mice to a task requiring spatial learning and memory. Depletion of endogenous tau in an amyloid precursor protein (APP) transgenic (J20) mouse line was shown to ameliorate hippocampal hyperactivity in J20 animals, tau depletion failed to reverse memory deficits associated with APP/Aß overproduction. On the other hand, deletion of tau alleviated the hyperlocomotion displayed by APP transgenics, suggesting that the functional effects of Aß-tau interactions reflect the temporal appearance of these molecules in individual brain areas.

11.
J Neurosci ; 38(2): 441-451, 2018 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-29196316

RESUMO

Early-life obesity predisposes to obesity in adulthood, a condition with broad medical implications including sleep disorders, which can exacerbate metabolic disturbances and disrupt cognitive and affective behaviors. In this study, we examined the long-term impact of transient peripubertal diet-induced obesity (ppDIO, induced between 4 and 10 weeks of age) on sleep-wake behavior in male mice. EEG and EMG recordings revealed that ppDIO increases sleep during the active phase but reduces resting-phase sleep quality. This impaired sleep phenotype persisted for up to 1 year, although animals were returned to a non-obesiogenic diet from postnatal week 11 onwards. To better understand the mechanisms responsible for the ppDIO-induced alterations in sleep, we focused on the lateral hypothalamus (LH). Mice exposed to ppDIO did not show altered mRNA expression levels of orexin and melanin-concentrating hormone, two peptides that are important for sleep-wake behavior and food intake. Conversely, the LH of ppDIO-exposed mice had reduced contents of serotonin (5-hydroxytryptamine, 5-HT), a neurotransmitter involved in both sleep-wake and satiety regulation. Interestingly, an acute peripheral injection of the satiety-signaling peptide YY 3-36 increased 5-HT turnover in the LH and ameliorated the ppDIO-induced sleep disturbances, suggesting the therapeutic potential of this peptide. These findings provide new insights into how sleep-wake behavior is programmed during early life and how peripheral and central signals are integrated to coordinate sleep.SIGNIFICANCE STATEMENT Adult physiology and behavior are strongly influenced by dynamic reorganization of the brain during puberty. The present work shows that obesity during puberty leads to persistently dysregulated patterns of sleep and wakefulness by blunting serotonergic signaling in the lateral hypothalamus. It also shows that pharmacological mimicry of satiety with peptide YY3-36 can reverse this neurochemical imbalance and acutely restore sleep composition. These findings add insight into how innate behaviors such as feeding and sleep are integrated and suggest a novel mechanism through which diet-induced obesity during puberty imposes its long-lasting effects on sleep-wake behavior.


Assuntos
Região Hipotalâmica Lateral/metabolismo , Obesidade/complicações , Serotonina/metabolismo , Transtornos do Sono-Vigília/etiologia , Animais , Homeostase/fisiologia , Região Hipotalâmica Lateral/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Fragmentos de Peptídeos/farmacologia , Peptídeo YY/farmacologia , Transtornos do Sono-Vigília/metabolismo
12.
EBioMedicine ; 20: 120-126, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28566250

RESUMO

Tau is a major component of the neurofibrillary tangles (NFT) that represent a pathological hallmark of Alzheimer's disease (AD). Although generally considered an axonal protein, Tau is found in the somato-dendritic compartment of degenerating neurons and this redistribution is thought to be a trigger of neurodegeneration in AD. Here, we show the presence of tau mRNA in a dendritic ribonucleoprotein (RNP) complex that includes Ca2+-calmodulin dependent protein kinase (CaMK)IIα mRNA and that is translated locally in response to glutamate stimulation. Further, we show that Tau mRNA is a component of mRNP granules that contain RNA-binding proteins, and that it interacts with Myosin Va, a postsynaptic motor protein; these findings suggest that tau mRNA is transported into dendritic spines. We also report that tau mRNA localized in the somato-dendritic component of primary hippocampal cells and that a sub-toxic concentration of glutamate enhances local translation and hyperphosphorylation of tau, effects that are blocked by the gluatamatergic antagonists MK801 and NBQX. These data thus demonstrate that alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) and N-methyl-d-aspartate (NMDA) stimulation redistributes tau to the somato-dendritic region of neurons where it may trigger neurodegeneration.


Assuntos
Neurônios/metabolismo , Biossíntese de Proteínas , Receptores de AMPA/agonistas , Receptores de N-Metil-D-Aspartato/agonistas , Proteínas tau/genética , Proteínas tau/metabolismo , Animais , Córtex Cerebral/metabolismo , Dendritos/metabolismo , Expressão Gênica , Ácido Glutâmico/metabolismo , Ácido Glutâmico/farmacologia , Hipocampo/metabolismo , Humanos , Masculino , Camundongos , Camundongos Knockout , Fosforilação , Transporte Proteico , Receptores de AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Ribonucleoproteínas/metabolismo
13.
Cereb Cortex ; 27(4): 2580-2591, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-27073221

RESUMO

Tau protein in dendrites and synapses has been recently implicated in synaptic degeneration and neuronal malfunction. Chronic stress, a well-known inducer of neuronal/synaptic atrophy, triggers hyperphosphorylation of Tau protein and cognitive deficits. However, the cause-effect relationship between these events remains to be established. To test the involvement of Tau in stress-induced impairments of cognition, we investigated the impact of stress on cognitive behavior, neuronal structure, and the synaptic proteome in the prefrontal cortex (PFC) of Tau knock-out (Tau-KO) and wild-type (WT) mice. Whereas exposure to chronic stress resulted in atrophy of apical dendrites and spine loss in PFC neurons as well as significant impairments in working memory in WT mice, such changes were absent in Tau-KO animals. Quantitative proteomic analysis of PFC synaptosomal fractions, combined with transmission electron microscopy analysis, suggested a prominent role for mitochondria in the regulation of the effects of stress. Specifically, chronically stressed animals exhibit Tau-dependent alterations in the levels of proteins involved in mitochondrial transport and oxidative phosphorylation as well as in the synaptic localization of mitochondria in PFC. These findings provide evidence for a causal role of Tau in mediating stress-elicited neuronal atrophy and cognitive impairment and indicate that Tau may exert its effects through synaptic mitochondria.


Assuntos
Mitocôndrias/patologia , Córtex Pré-Frontal/patologia , Estresse Psicológico/complicações , Sinapses/patologia , Proteínas tau/metabolismo , Animais , Atrofia , Cromatografia Líquida de Alta Pressão , Dendritos/patologia , Dendritos/ultraestrutura , Modelos Animais de Doenças , Masculino , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Proteômica
14.
J Alzheimers Dis ; 54(1): 135-48, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27497478

RESUMO

Early-stage Alzheimer's disease (AD) is characterized by synaptic dysfunction, a phenomenon in which soluble oligomers of amyloid-beta (Aß) and N-methyl-D-aspartate receptor (NMDAR) are implicated. Here, we demonstrated that astrocytes express NMDARs and therefore have the potential to modulate the synaptotoxic actions of Aß. We found that specific pharmacological antagonism of two of the major NMDAR subunits, GluN2A and GluN2B, exacerbates Aß-induced synaptotoxicity suggesting, for the first time, that astrocytic GluN2A and GluN2B mediate synaptoprotection. From the perspective of the pathogenic mechanisms of Alzheimer's disease, in which Aß and NMDAR play significant roles, these observations are striking since neuronal GluN2A and GluN2B are well known modulators of neurodegeneration. We did initial studies to understand the basis for the differential effects of astrocytic and neuronal GluN2A and GluN2B in the promotion of synapse survival, and identified a neurotrophin produced by astrocytes, nerve growth factor ß (ß-NGF), as a likely mediator of the synaptoprotective effects of astrocytic GluN2A and GluN2B activation. The results presented suggest that astrocytes may be suitable druggable targets for the prevention and/or delay of the synaptic loss that occurs during early stages of AD.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Astrócitos/metabolismo , Hipocampo/metabolismo , Fragmentos de Peptídeos/toxicidade , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapses/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Comunicação Celular/efeitos dos fármacos , Comunicação Celular/fisiologia , Células Cultivadas , Técnicas de Cocultura , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/metabolismo , Espinhas Dendríticas/patologia , Fármacos Atuantes sobre Aminoácidos Excitatórios/farmacologia , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , N-Metilaspartato/metabolismo , N-Metilaspartato/farmacologia , Fator de Crescimento Neural/metabolismo , Neuroproteção/efeitos dos fármacos , Neuroproteção/fisiologia , Ratos Wistar , Receptores de N-Metil-D-Aspartato/agonistas , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Sinapses/efeitos dos fármacos , Sinapses/patologia
15.
Cardiovasc Diabetol ; 15(1): 114, 2016 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-27538526

RESUMO

BACKGROUND: Chronic stress is associated with increased risk of glucose intolerance and cardiovascular diseases, albeit through undefined mechanisms. With the aim of gaining insights into the latter, this study examined the metabolic profile of young adult male rats that were exposed to chronic unpredictable stress. METHODS: Young adult male rats were submitted to 4 weeks of chronic unpredictable stress and allowed to recover for 5 weeks. An extensive analysis including of morphologic, biochemical and molecular parameters was carried out both after chronic unpredictable stress and after recovery from stress. RESULTS: After 28 days of chronic unpredictable stress (CUS) the animals submitted to this protocol displayed less weight gain than control animals. After 5 weeks of recovery the weight gain rebounded to similar values of controls. In addition, following CUS, fasting insulin levels were increased and were accompanied by signs of impaired glucose tolerance and elevated serum corticosteroid levels. This biochemical profile persisted into the post-stress recovery period, despite the restoration of baseline corticosteroid levels. The mRNA expression levels of peroxisome proliferator-activated receptor (PPAR)-γ and lipocalin-2 in white adipose tissue were, respectively, down- and up-regulated. CONCLUSIONS: Reduction of PPAR-γ expression and generation of a pro-inflammatory environment by increased lipocalin-2 expression in white adipose tissue may contribute to stress-induced glucose intolerance.


Assuntos
Intolerância à Glucose/etiologia , Gordura Intra-Abdominal/metabolismo , PPAR gama/metabolismo , Estresse Psicológico/complicações , Animais , Biomarcadores/sangue , Glicemia/metabolismo , Doença Crônica , Corticosterona/sangue , Modelos Animais de Doenças , Regulação para Baixo , Intolerância à Glucose/sangue , Intolerância à Glucose/genética , Intolerância à Glucose/fisiopatologia , Mediadores da Inflamação/metabolismo , Insulina/sangue , Lipocalina-2/genética , Lipocalina-2/metabolismo , Masculino , PPAR gama/genética , Fenótipo , Ratos Wistar , Transdução de Sinais , Estresse Psicológico/sangue , Estresse Psicológico/genética , Estresse Psicológico/fisiopatologia , Fatores de Tempo , Aumento de Peso
16.
Proc Natl Acad Sci U S A ; 113(26): E3755-63, 2016 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-27274066

RESUMO

Exposure to chronic stress is frequently accompanied by cognitive and affective disorders in association with neurostructural adaptations. Chronic stress was previously shown to trigger Alzheimer's-like neuropathology, which is characterized by Tau hyperphosphorylation and missorting into dendritic spines followed by memory deficits. Here, we demonstrate that stress-driven hippocampal deficits in wild-type mice are accompanied by synaptic missorting of Tau and enhanced Fyn/GluN2B-driven synaptic signaling. In contrast, mice lacking Tau [Tau knockout (Tau-KO) mice] do not exhibit stress-induced pathological behaviors and atrophy of hippocampal dendrites or deficits of hippocampal connectivity. These findings implicate Tau as an essential mediator of the adverse effects of stress on brain structure and function.


Assuntos
Doença de Alzheimer/metabolismo , Hipocampo/metabolismo , Proteínas tau/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Espinhas Dendríticas/metabolismo , Modelos Animais de Doenças , Hipocampo/patologia , Humanos , Masculino , Memória , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Estresse Fisiológico , Sinapses/metabolismo , Proteínas tau/genética
17.
Neural Plast ; 2016: 6391686, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27034847

RESUMO

Stress and stress hormones, glucocorticoids (GCs), exert widespread actions in central nervous system, ranging from the regulation of gene transcription, cellular signaling, modulation of synaptic structure, and transmission and glial function to behavior. Their actions are mediated by glucocorticoid and mineralocorticoid receptors which are nuclear receptors/transcription factors. While GCs primarily act to maintain homeostasis by inducing physiological and behavioral adaptation, prolonged exposure to stress and elevated GC levels may result in neuro- and psychopathology. There is now ample evidence for cause-effect relationships between prolonged stress, elevated GC levels, and cognitive and mood disorders while the evidence for a link between chronic stress/GC and neurodegenerative disorders such as Alzheimer's (AD) and Parkinson's (PD) diseases is growing. This brief review considers some of the cellular mechanisms through which stress and GC may contribute to the pathogenesis of AD and PD.


Assuntos
Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Glucocorticoides/metabolismo , Plasticidade Neuronal , Doença de Parkinson/metabolismo , Estresse Psicológico/metabolismo , Doença de Alzheimer/etiologia , Animais , Humanos , Inflamação/complicações , Inflamação/metabolismo , Doença de Parkinson/etiologia , Fatores de Risco , Estresse Psicológico/complicações
18.
J Alzheimers Dis ; 51(1): 197-212, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26836185

RESUMO

Synaptic dysfunction during early stages of Alzheimer's disease (AD) is triggered by soluble amyloid-ß (Aß) oligomers that interact with NMDA receptors (NMDARs). We previously showed that Aß induces synaptic protein loss through NMDARs, albeit through undefined mechanisms. Accordingly, we here examined the contribution of individual NMDAR subunits to synaptotoxicity and demonstrate that Aß exerts differential effects on the levels and distribution of GluN2A and GluN2B subunits of NMDAR in dendrites. Treatment of cultured hippocampal neurons with Aß1-40 (10 µM, 1 h) induced a significant increase of dendritic and synaptic GluN2B puncta densities with parallel decreases in the puncta densities of denritic and synaptic pTyr1472-GluN2B. Conversely, Aß significantly decreased dendritic and synaptic GluN2A and dendritic pTyr1325-GluN2A puncta densities and increased synaptic pTyr1325-GluN2A puncta densities. Unexpectedly, Aß treatment resulted in a significant reduction of GluN2B and pTyr1472-GluN2B protein levels but did not influence GluN2A and pTyr1325-GluN2A levels. These results show that Aß exerts complex and distinct regulatory effects on the trafficking and phosphorylation of GluN2A and GluN2B, as well as on their localization within synaptic and non-synaptic sites. Increased understanding of these early events in Aß-induced synaptic dysfunction is likely to be important for the development of timely preventive and therapeutic interventions.


Assuntos
Peptídeos beta-Amiloides/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/citologia , Neurônios/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Animais Recém-Nascidos , Células Cultivadas , Dendritos/efeitos dos fármacos , Dendritos/metabolismo , Masculino , Neurônios/citologia , Fosforilação/efeitos dos fármacos , Ratos , Ratos Wistar
19.
Nat Commun ; 6: 10216, 2015 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-26671725

RESUMO

Neurofibrillary tangles, composed of hyperphosphorylated tau fibrils, are a pathological hallmark of Alzheimer's disease; the neurofibrillary tangle load correlates strongly with clinical progression of the disease. A growing body of evidence indicates that tau oligomer formation precedes the appearance of neurofibrillary tangles and contributes to neuronal loss. Here we show that tau oligomer formation can be inhibited by compounds whose chemical backbone includes 1,2-dihydroxybenzene. Specifically, we demonstrate that 1,2-dihydroxybenzene-containing compounds bind to and cap cysteine residues of tau and prevent its aggregation by hindering interactions between tau molecules. Further, we show that orally administered DL-isoproterenol, an adrenergic receptor agonist whose skeleton includes 1,2-dihydroxybenzene and which penetrates the brain, reduces the levels of detergent-insoluble tau, neuronal loss and reverses neurofibrillary tangle-associated brain dysfunction. Thus, compounds that target the cysteine residues of tau may prove useful in halting the progression of Alzheimer's disease and other tauopathies.


Assuntos
Agonistas Adrenérgicos beta/farmacologia , Doença de Alzheimer/metabolismo , Catecóis/farmacologia , Cisteína/efeitos dos fármacos , Isoproterenol/farmacologia , Emaranhados Neurofibrilares/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Proteínas tau/efeitos dos fármacos , Agonistas Adrenérgicos beta/química , Animais , Comportamento Animal/efeitos dos fármacos , Western Blotting , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Catecóis/química , Catecóis/metabolismo , Linhagem Celular Tumoral , Cisteína/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Avaliação Pré-Clínica de Medicamentos , Isoproterenol/química , Camundongos , Camundongos Transgênicos , Emaranhados Neurofibrilares/metabolismo , Neurônios/patologia , Polimerização , Proteínas tau/genética , Proteínas tau/metabolismo
20.
J Clin Med ; 4(5): 1051-62, 2015 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-26239465

RESUMO

Endocrine dysfunction is a common effect of traumatic brain injury (TBI). In addition to affecting the regulation of important body functions, the disruption of endocrine physiology can significantly impair mental functions, such as attention, memory, executive function, and mood. This mini-review focuses on alterations in mental functioning that are associated with neuroendocrine disturbances in adults who suffered TBI. It summarizes the contribution of hormones to the regulation of mental functions, the consequences of TBI on mental health and neuroendocrine homeostasis, and the effects of hormone substitution on mental dysfunction caused by TBI. The available empirical evidence suggests that comprehensive assessment of mental functions should be standard in TBI subjects presenting with hormone deficiency and that hormone replacement therapy should be accompanied by pre- and post-assessments.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...