Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Gerontol A Biol Sci Med Sci ; 75(6): 1021-1030, 2020 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-31180116

RESUMO

Intranasal insulin is a safe and effective method for ameliorating memory deficits associated with pathological brain aging. However, the impact of different formulations and the duration of treatment on insulin's efficacy and the cellular processes targeted by the treatment remain unclear. Here, we tested whether intranasal insulin aspart, a short-acting insulin formulation, could alleviate memory decline associated with aging and whether long-term treatment affected regulation of insulin receptors and other potential targets. Outcome variables included measures of spatial learning and memory, autoradiography and immunohistochemistry of the insulin receptor, and hippocampal microarray analyses. Aged Fischer 344 rats receiving long-term (3 months) intranasal insulin did not show significant memory enhancement on the Morris water maze task. Autoradiography results showed that long-term treatment reduced insulin binding in the thalamus but not the hippocampus. Results from hippocampal immunofluorescence revealed age-related decreases in insulin immunoreactivity that were partially offset by intranasal administration. Microarray analyses highlighted numerous insulin-sensitive genes, suggesting insulin aspart was able to enter the brain and alter hippocampal RNA expression patterns including those associated with tumor suppression. Our work provides insights into potential mechanisms of intranasal insulin and insulin resistance, and highlights the importance of treatment duration and the brain regions targeted.


Assuntos
Envelhecimento/fisiologia , Insulina Aspart/administração & dosagem , Transtornos da Memória/tratamento farmacológico , Receptor de Insulina/metabolismo , Administração Intranasal , Animais , Expressão Gênica , Hipocampo/metabolismo , Insulina Aspart/genética , Insulina Aspart/farmacologia , Masculino , Aprendizagem em Labirinto , Modelos Animais , Ratos , Ratos Endogâmicos F344
2.
Exp Neurol ; 313: 79-87, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30576640

RESUMO

It has been >20 years since studies first revealed that the brain is insulin sensitive, highlighted by the expression of insulin receptors in neurons and glia, the presence of circulating brain insulin, and even localized insulin production. Following these discoveries, evidence of decreased brain insulin receptor number and function was reported in both clinical samples and animal models of aging and Alzheimer's disease, setting the stage for the hypothesis that neuronal insulin resistance may underlie memory loss in these conditions. The development of therapeutic insulin delivery to the brain using intranasal insulin administration has been shown to improve aspects of memory or learning in both humans and animal models. However, whether this approach functions by compensating for poorly signaling insulin receptors, for reduced insulin levels in the brain, or for reduced trafficking of insulin into the brain remains unclear. Direct measures of insulin's impact on cellular physiology and metabolism in the brain have been sparse in models of Alzheimer's disease, and even fewer studies have analyzed these processes in the aged brain. Nevertheless, recent evidence supports the role of brain insulin as a mediator of glucose metabolism through several means, including altering glucose transporters. Here, we provide a review of contemporary literature on brain insulin resistance, highlight the rationale for improving memory function using intranasal insulin, and describe initial results from experiments using a molecular approach to more directly measure the impact of insulin receptor activation and signaling on glucose uptake in neurons.


Assuntos
Envelhecimento/fisiologia , Doença de Alzheimer/fisiopatologia , Encéfalo/fisiologia , Encéfalo/fisiopatologia , Resistência à Insulina/fisiologia , Insulina/fisiologia , Idoso , Idoso de 80 Anos ou mais , Humanos
3.
J Alzheimers Dis ; 66(4): 1371-1378, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30412490

RESUMO

Aging is the leading risk factor for idiopathic Alzheimer's disease (AD), indicating that normal aging processes promote AD and likely are present in the neurons in which AD pathogenesis originates. In AD, neurofibrillary tangles (NFTs) appear first in entorhinal cortex, implying that aging processes in entorhinal neurons promote NFT pathogenesis. Using electrophysiology and immunohistochemistry, we find pronounced aging-related Ca2 + dysregulation in rat entorhinal neurons homologous with the human neurons in which NFTs originate. Considering that humans recapitulate many aspects of animal brain aging, these results support the hypothesis that aging-related Ca2 + dysregulation occurs in human entorhinal neurons and promotes NFT pathogenesis.


Assuntos
Envelhecimento/metabolismo , Doença de Alzheimer/metabolismo , Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Córtex Entorrinal/metabolismo , Neurônios/metabolismo , Doença de Alzheimer/patologia , Animais , Córtex Entorrinal/patologia , Masculino , Emaranhados Neurofibrilares/metabolismo , Emaranhados Neurofibrilares/patologia , Neurônios/patologia , Ratos , Ratos Endogâmicos F344
4.
J Neurosci ; 38(4): 1030-1041, 2018 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-29255009

RESUMO

Hippocampal overexpression of FK506-binding protein 12.6/1b (FKBP1b), a negative regulator of ryanodine receptor Ca2+ release, reverses aging-induced memory impairment and neuronal Ca2+ dysregulation. Here, we tested the hypothesis that FKBP1b also can protect downstream transcriptional networks from aging-induced dysregulation. We gave hippocampal microinjections of FKBP1b-expressing viral vector to male rats at either 13 months of age (long-term, LT) or 19 months of age (short-term, ST) and tested memory performance in the Morris water maze at 21 months of age. Aged rats treated ST or LT with FKBP1b substantially outperformed age-matched vector controls and performed similarly to each other and young controls (YCs). Transcriptional profiling in the same animals identified 2342 genes with hippocampal expression that was upregulated/downregulated in aged controls (ACs) compared with YCs (the aging effect). Of these aging-dependent genes, 876 (37%) also showed altered expression in aged FKBP1b-treated rats compared with ACs, with FKBP1b restoring expression of essentially all such genes (872/876, 99.5%) in the direction opposite the aging effect and closer to levels in YCs. This inverse relationship between the aging and FKBP1b effects suggests that the aging effects arise from FKBP1b deficiency. Functional category analysis revealed that genes downregulated with aging and restored by FKBP1b were associated predominantly with diverse brain structure categories, including cytoskeleton, membrane channels, and extracellular region. Conversely, genes upregulated with aging but not restored by FKBP1b associated primarily with glial-neuroinflammatory, ribosomal, and lysosomal categories. Immunohistochemistry confirmed aging-induced rarefaction and FKBP1b-mediated restoration of neuronal microtubular structure. Therefore, a previously unrecognized genomic network modulating diverse brain structural processes is dysregulated by aging and restored by FKBP1b overexpression.SIGNIFICANCE STATEMENT Previously, we found that hippocampal overexpression of FK506-binding protein 12.6/1b (FKBP1b), a negative regulator of intracellular Ca2+ responses, reverses both aging-related Ca2+ dysregulation and cognitive impairment. Here, we tested whether hippocampal FKBP1b overexpression also counteracts aging changes in gene transcriptional networks. In addition to reducing memory deficits in aged rats, FKBP1b selectively counteracted aging-induced expression changes in 37% of aging-dependent genes, with cytoskeletal and extracellular structure categories highly associated with the FKBP1b-rescued genes. Our results indicate that, in parallel with cognitive processes, a novel transcriptional network coordinating brain structural organization is dysregulated with aging and restored by FKBP1b.


Assuntos
Envelhecimento/fisiologia , Regulação da Expressão Gênica/fisiologia , Hipocampo/metabolismo , Memória/fisiologia , Proteínas de Ligação a Tacrolimo/metabolismo , Animais , Sinalização do Cálcio/fisiologia , Hipocampo/fisiopatologia , Masculino , Transtornos da Memória/fisiopatologia , Ratos , Ratos Endogâmicos F344 , Ratos Transgênicos
5.
Neuroscience ; 364: 130-142, 2017 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-28939258

RESUMO

Both insulin signaling disruption and Ca2+ dysregulation are closely related to memory loss during aging and increase the vulnerability to Alzheimer's disease (AD). In hippocampal neurons, aging-related changes in calcium regulatory pathways have been shown to lead to higher intracellular calcium levels and an increase in the Ca2+-dependent afterhyperpolarization (AHP), which is associated with cognitive decline. Recent studies suggest that insulin reduces the Ca2+-dependent AHP. Given the sensitivity of neurons to insulin and evidence that brain insulin signaling is reduced with age, insulin-mediated alterations in calcium homeostasis may underlie the beneficial actions of insulin in the brain. Indeed, increasing insulin signaling in the brain via intranasal delivery has yielded promising results such as improving memory in both clinical and animal studies. However, while several mechanisms have been proposed, few have focused on regulation on intracellular Ca2+. In the present study, we further examined the effects of acute insulin on calcium pathways in primary hippocampal neurons in culture. Using the whole-cell patch-clamp technique, we found that acute insulin delivery reduced voltage-gated calcium currents. Fura-2 imaging was used to also address acute insulin effects on spontaneous and depolarization-mediated Ca2+ transients. Results indicate that insulin reduced Ca2+ transients, which appears to have involved a reduction in ryanodine receptor function. Together, these results suggest insulin regulates pathways that control intracellular Ca2+ which may reduce the AHP and improve memory. This may be one mechanism contributing to improved memory recall in response to intranasal insulin therapy in the clinic.


Assuntos
Envelhecimento/metabolismo , Cálcio/metabolismo , Hipocampo/metabolismo , Insulina/metabolismo , Insulina/farmacologia , Neurônios/metabolismo , Transdução de Sinais , Animais , Células Cultivadas , Insulina/administração & dosagem , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos
6.
J Gerontol A Biol Sci Med Sci ; 72(2): 189-197, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27069097

RESUMO

Novel therapies have turned to delivering compounds to the brain using nasal sprays, bypassing the blood brain barrier, and enriching treatment options for brain aging and/or Alzheimer's disease. We conducted a series of in vivo experiments to test the impact of intranasal Apidra, a zinc-free insulin formulation, on the brain of young and aged F344 rats. Both single acute and repeated daily doses were compared to test the hypothesis that insulin could improve memory recall in aged memory-deficient animals. We quantified insulin signaling in different brain regions and at different times following delivery. We measured cerebral blood flow (CBF) using MRI and also characterized several brain metabolite levels using MR spectroscopy. We show that neither acute nor chronic Apidra improved memory or recall in young or aged animals. Within 2 hours of a single dose, increased insulin signaling was seen in ventral areas of the aged brains only. Although chronic Apidra was able to offset reduced CBF with aging, it also caused significant reductions in markers of neuronal integrity. Our data suggest that this zinc-free insulin formulation may actually hasten cognitive decline with age when used chronically.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Cognição/efeitos dos fármacos , Insulina/análogos & derivados , Transdução de Sinais/efeitos dos fármacos , Administração Intranasal , Fatores Etários , Animais , Circulação Cerebrovascular , Insulina/administração & dosagem , Insulina/farmacologia , Masculino , Ratos , Ratos Endogâmicos F344 , Zinco
7.
Biochem Biophys Res Commun ; 483(4): 981-987, 2017 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-27553276

RESUMO

Neuroscientists studying normal brain aging, spinal cord injury, Alzheimer's disease (AD) and other neurodegenerative diseases have focused considerable effort on carefully characterizing intracellular perturbations in calcium dynamics or levels. At the cellular level, calcium is known for controlling life and death and orchestrating most events in between. For many years, intracellular calcium has been recognized as an essential ion associated with nearly all cellular functions from cell growth to degeneration. Often the emphasis is on the negative impact of calcium dysregulation and the typical worse-case-scenario leading inevitably to cell death. However, even high amplitude calcium transients, when executed acutely, can alter neuronal communication and synaptic strength in positive ways, without necessarily killing neurons. Here, we focus on the evidence that calcium has a subtle and distinctive role in shaping and controlling synaptic events that underpin neuronal communication and that these subtle changes in aging or AD may contribute to cognitive decline. We emphasize that calcium imaging in dendritic components is ultimately necessary to directly test for the presence of age- or disease-associated alterations during periods of synaptic activation.


Assuntos
Encéfalo/fisiologia , Cálcio/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Humanos , Modelos Biológicos
8.
J Gerontol A Biol Sci Med Sci ; 71(1): 30-9, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25659889

RESUMO

Peripheral insulin resistance is a key component of metabolic syndrome associated with obesity, dyslipidemia, hypertension, and type 2 diabetes. While the impact of insulin resistance is well recognized in the periphery, it is also becoming apparent in the brain. Recent studies suggest that insulin resistance may be a factor in brain aging and Alzheimer's disease (AD) whereby intranasal insulin therapy, which delivers insulin to the brain, improves cognition and memory in AD patients. Here, we tested a clinically relevant delivery method to determine the impact of two forms of insulin, short-acting insulin lispro (Humalog) or long-acting insulin detemir (Levemir), on cognitive functions in aged F344 rats. We also explored insulin effects on the Ca(2+)-dependent hippocampal afterhyperpolarization (AHP), a well-characterized neurophysiological marker of aging which is increased in the aged, memory impaired animal. Low-dose intranasal insulin improved memory recall in aged animals such that their performance was similar to that seen in younger animals. Further, because ex vivo insulin also reduced the AHP, our results suggest that the AHP may be a novel cellular target of insulin in the brain, and improved cognitive performance following intranasal insulin therapy may be the result of insulin actions on the AHP.


Assuntos
Envelhecimento , Encéfalo , Senescência Celular/fisiologia , Cognição , Insulina Detemir , Insulina Lispro , Administração Intranasal , Envelhecimento/metabolismo , Envelhecimento/psicologia , Animais , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Senescência Celular/efeitos dos fármacos , Cognição/efeitos dos fármacos , Cognição/fisiologia , Transtornos Cognitivos/metabolismo , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Hipoglicemiantes/administração & dosagem , Hipoglicemiantes/metabolismo , Insulina Detemir/administração & dosagem , Insulina Detemir/metabolismo , Insulina Lispro/administração & dosagem , Insulina Lispro/metabolismo , Resistência à Insulina , Memória/efeitos dos fármacos , Ratos , Resultado do Tratamento
9.
J Neurosci ; 35(30): 10878-87, 2015 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-26224869

RESUMO

Brain Ca2+ regulatory processes are altered during aging, disrupting neuronal, and cognitive functions. In hippocampal pyramidal neurons, the Ca2+ -dependent slow afterhyperpolarization (sAHP) exhibits an increase with aging, which correlates with memory impairment. The increased sAHP results from elevated L-type Ca2+ channel activity and ryanodine receptor (RyR)-mediated Ca2+ release, but underlying molecular mechanisms are poorly understood. Previously, we found that expression of the gene encoding FK506-binding protein 12.6/1b (FKBP1b), a small immunophilin that stabilizes RyR-mediated Ca2+ release in cardiomyocytes, declines in hippocampus of aged rats and Alzheimer's disease subjects. Additionally, knockdown/disruption of hippocampal FKBP1b in young rats augments neuronal Ca2+ responses. Here, we test the hypothesis that declining FKBP1b underlies aging-related hippocampal Ca2+ dysregulation. Using microinjection of adeno-associated viral vector bearing a transgene encoding FKBP1b into the hippocampus of aged male rats, we assessed the critical prediction that overexpressing FKBP1b should reverse Ca2+ -mediated manifestations of brain aging. Immunohistochemistry and qRT-PCR confirmed hippocampal FKBP1b overexpression 4-6 weeks after injection. Compared to aged vector controls, aged rats overexpressing FKBP1b showed dramatic enhancement of spatial memory, which correlated with marked reduction of sAHP magnitude. Furthermore, simultaneous electrophysiological recording and Ca2+ imaging in hippocampal neurons revealed that the sAHP reduction was associated with a decrease in parallel RyR-mediated Ca2+ transients. Thus, hippocampal FKBP1b overexpression reversed key aspects of Ca2+ dysregulation and cognitive impairment in aging rats, supporting the novel hypothesis that declining FKBP1b is a molecular mechanism underlying aging-related Ca2+ dysregulation and unhealthy brain aging and pointing to FKBP1b as a potential therapeutic target. SIGNIFICANCE STATEMENT: This paper reports critical tests of a novel hypothesis that proposes a molecular mechanism of unhealthy brain aging and possibly, Alzheimer's disease. For more than 30 years, evidence has been accumulating that brain aging is associated with dysregulation of calcium in neurons. Recently, we found that FK506-binding protein 12.6/1b (FKBP1b), a small protein that regulates calcium, declines with aging in the hippocampus, a brain region important for memory. Here we used gene therapy approaches and found that raising FKBP1b reversed calcium dysregulation and memory impairment in aging rats, allowing them to perform a memory task as well as young rats. These studies identify a potential molecular mechanism of brain aging and may also have implications for treatment of Alzheimer's disease.


Assuntos
Envelhecimento/fisiologia , Cálcio/metabolismo , Cognição/fisiologia , Neurônios/metabolismo , Proteínas de Ligação a Tacrolimo/metabolismo , Animais , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/metabolismo , Hipocampo/metabolismo , Imuno-Histoquímica , Masculino , Técnicas de Patch-Clamp , Ratos , Ratos Endogâmicos F344 , Ratos Transgênicos , Reação em Cadeia da Polimerase em Tempo Real , Transgenes
10.
Brain Res ; 1621: 214-21, 2015 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-25451110

RESUMO

Results from clinical studies provide evidence that cognitive changes relatively late in life may be traced to antecedent conditions including diabetes, obesity, a sedentary lifestyle, and an atherogenic diet. As such, several traits of Type 2 diabetes (T2DM) could be considered pathogenic factors of aging, contributing to age-dependent cognitive decline and our susceptibility to Alzheimer's disease. It appears that both the duration of metabolic condition and the age of the individual, together can contribute to the potential impact on peripheral as well as brain health. Because of robust evidence that in animal models of aging, Ca(2+) dysregulation alters neuronal health, synaptic plasticity, and learning and memory processes, we tested the hypothesis that peripheral metabolic dysregulation could exacerbate Ca(2+) dysfunction in hippocampal CA1 neurons. Using intracellular/ extracellular electrophysiological and Ca(2+) imaging techniques, we show that Ca(2+)levels at rest or during synaptic stimulation, the Ca(2+)-dependent afterhyperpolarization, baseline field potentials, and short-term synaptic plasticity were not significantly altered in young-adult male Zucker diabetic fatty rats compare to their lean counterparts. Our observations suggest that early phases of T2DM characterized by high levels of glucose and insulin may be too transient to alter hippocampal CA1 physiology in this animal model of diabetes. These results are supported by clinical data showing that longer T2DM duration can have greater negative impact on cognitive functions. This article is part of a Special Issue entitled SI: Brain and Memory.


Assuntos
Envelhecimento , Região CA1 Hipocampal/metabolismo , Cálcio/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Neurônios/metabolismo , Potenciais de Ação , Animais , Biomarcadores/metabolismo , Glicemia/metabolismo , Região CA1 Hipocampal/fisiologia , Diabetes Mellitus Tipo 2/fisiopatologia , Modelos Animais de Doenças , Potenciais Pós-Sinápticos Excitadores , Hemoglobinas Glicadas/metabolismo , Insulina/metabolismo , Masculino , Ratos
11.
Proc Natl Acad Sci U S A ; 111(41): E4359-66, 2014 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-25267625

RESUMO

Vitamin D is an important calcium-regulating hormone with diverse functions in numerous tissues, including the brain. Increasing evidence suggests that vitamin D may play a role in maintaining cognitive function and that vitamin D deficiency may accelerate age-related cognitive decline. Using aging rodents, we attempted to model the range of human serum vitamin D levels, from deficient to sufficient, to test whether vitamin D could preserve or improve cognitive function with aging. For 5-6 mo, middle-aged F344 rats were fed diets containing low, medium (typical amount), or high (100, 1,000, or 10,000 international units/kg diet, respectively) vitamin D3, and hippocampal-dependent learning and memory were then tested in the Morris water maze. Rats on high vitamin D achieved the highest blood levels (in the sufficient range) and significantly outperformed low and medium groups on maze reversal, a particularly challenging task that detects more subtle changes in memory. In addition to calcium-related processes, hippocampal gene expression microarrays identified pathways pertaining to synaptic transmission, cell communication, and G protein function as being up-regulated with high vitamin D. Basal synaptic transmission also was enhanced, corroborating observed effects on gene expression and learning and memory. Our studies demonstrate a causal relationship between vitamin D status and cognitive function, and they suggest that vitamin D-mediated changes in hippocampal gene expression may improve the likelihood of successful brain aging.


Assuntos
Envelhecimento/patologia , Transtornos Cognitivos/prevenção & controle , Transtornos Cognitivos/fisiopatologia , Hipocampo/fisiopatologia , Transmissão Sináptica , Vitamina D/uso terapêutico , Envelhecimento/efeitos dos fármacos , Animais , Transtornos Cognitivos/tratamento farmacológico , Dieta , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Humanos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Modelos Neurológicos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Ratos Endogâmicos F344 , Elementos de Resposta/genética , Software , Transmissão Sináptica/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Vitamina D/sangue , Vitamina D/farmacologia
12.
Acta Neuropathol Commun ; 2: 64, 2014 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-24916066

RESUMO

Mid-life obesity and type 2 diabetes mellitus (T2DM) confer a modest, increased risk for Alzheimer's disease (AD), though the underlying mechanisms are unknown. We have created a novel mouse model that recapitulates features of T2DM and AD by crossing morbidly obese and diabetic db/db mice with APPΔNL/ΔNLx PS1P264L/P264L knock-in mice. These mice (db/AD) retain many features of the parental lines (e.g. extreme obesity, diabetes, and parenchymal deposition of ß-amyloid (Aß)). The combination of the two diseases led to additional pathologies-perhaps most striking of which was the presence of severe cerebrovascular pathology, including aneurysms and small strokes. Cortical Aß deposition was not significantly increased in the diabetic mice, though overall expression of presenilin was elevated. Surprisingly, Aß was not deposited in the vasculature or removed to the plasma, and there was no stimulation of activity or expression of major Aß-clearing enzymes (neprilysin, insulin degrading enzyme, or endothelin-converting enzyme). The db/AD mice displayed marked cognitive impairment in the Morris Water Maze, compared to either db/db or APPΔNLx PS1P264L mice. We conclude that the diabetes and/or obesity in these mice leads to a destabilization of the vasculature, leading to strokes and that this, in turn, leads to a profound cognitive impairment and that this is unlikely to be directly dependent on Aß deposition. This model of mixed or vascular dementia provides an exciting new avenue of research into the mechanisms underlying the obesity-related risk for age-related dementia, and will provide a useful tool for the future development of therapeutics.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Transtornos Cognitivos/etiologia , Demência Vascular/complicações , Diabetes Mellitus/fisiopatologia , Obesidade Mórbida/complicações , Precursor de Proteína beta-Amiloide/genética , Animais , Pressão Sanguínea/genética , Transtornos Cognitivos/sangue , Transtornos Cognitivos/genética , Demência Vascular/sangue , Demência Vascular/genética , Diabetes Mellitus/sangue , Diabetes Mellitus/genética , Modelos Animais de Doenças , Teste de Tolerância a Glucose , Humanos , Insulina/metabolismo , Leptina/sangue , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Transgênicos , Mutação/genética , Neprilisina/metabolismo , Obesidade Mórbida/sangue , Obesidade Mórbida/genética , Presenilina-1/genética , Presenilina-1/metabolismo , Receptores para Leptina/genética
13.
Eur J Pharmacol ; 719(1-3): 34-43, 2013 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-23872402

RESUMO

Recently it has become clear that conditions of insulin resistance/metabolic syndrome, obesity and diabetes, are linked with moderate cognitive impairment in normal aging and elevated risk of Alzheimer's disease. It appears that a common feature of these conditions is impaired insulin signaling, affecting the brain as well as peripheral target tissues. A number of studies have documented that insulin directly affects brain processes and that reduced insulin signaling results in impaired learning and memory. Several studies have also shown that diabetes induces Ca(2+) dysregulation in neurons. Because brain aging is associated with substantial Ca(2+) dyshomeostasis, it has been proposed that impaired insulin signaling exacerbates or accelerates aging-related Ca(2+) dyshomeostasis. However, there have been few studies examining insulin interactions with Ca(2+) regulation in aging animals. We have been testing predictions of the Ca(2+) dysregulation/diabetes/brain aging hypothesis and have found that insulin and insulin-sensitizers (thiazolidinediones) target several hippocampal Ca(2+)-related processes affected by aging. The drugs appear able to reduce the age-dependent increase in Ca(2+) transients and the Ca(2+) -sensitive afterhyperpolarization. Thus, while additional testing is needed, the results to date are consistent with the view that strategies that enhance insulin signaling can counteract the effect of aging on Ca(2+) dysregulation.


Assuntos
Envelhecimento/metabolismo , Cálcio/metabolismo , Diabetes Mellitus/metabolismo , Hipocampo/metabolismo , Envelhecimento/fisiologia , Animais , Cognição , Diabetes Mellitus/fisiopatologia , Hipocampo/fisiopatologia , Humanos , Insulina/metabolismo
14.
Free Radic Biol Med ; 65: 324-334, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23872023

RESUMO

In addition to the well-known effects of vitamin D (VitD) in maintaining bone health, there is increasing appreciation that this vitamin may serve important roles in other organs and tissues, including the brain. Given that VitD deficiency is especially widespread among the elderly, it is important to understand how the range of serum VitD levels that mimic those found in humans (from low to high) affects the brain during aging from middle age to old age. To address this issue, 27 male F344 rats were split into three groups and fed isocaloric diets containing low (100 IU/kg food), control (1000 IU/kg food), or high (10,000 IU/kg food) VitD beginning at middle age (12 months) and continued for a period of 4-5 months. We compared the effects of these dietary VitD manipulations on oxidative and nitrosative stress measures in posterior brain cortices. The low-VitD group showed global elevation of 3-nitrotyrosine compared to control and high-VitD-treated groups. Further investigation showed that this elevation may involve dysregulation of the nuclear factor κ-light-chain enhancer of activated B cells (NF-κB) pathway and NF-κB-mediated transcription of inducible nitric oxide synthase (iNOS) as indicated by translocation of NF-κB to the nucleus and elevation of iNOS levels. Proteomics techniques were used to provide insight into potential mechanisms underlying these effects. Several brain proteins were found at significantly elevated levels in the low-VitD group compared to the control and high-VitD groups. Three of these proteins, 6-phosphofructokinase, triose phosphate isomerase, and pyruvate kinase, are involved directly in glycolysis. Two others, peroxiredoxin-3 and DJ-1/PARK7, have peroxidase activity and are found in mitochondria. Peptidyl-prolyl cis-trans isomerase A (cyclophilin A) has been shown to have multiple roles, including protein folding, regulation of protein kinases and phosphatases, immunoregulation, cell signaling, and redox status. Together, these results suggest that dietary VitD deficiency contributes to significant nitrosative stress in brain and may promote cognitive decline in middle-aged and elderly adults.


Assuntos
Encéfalo/metabolismo , Transtornos Cognitivos/etiologia , Tirosina/metabolismo , Deficiência de Vitamina D/complicações , Deficiência de Vitamina D/metabolismo , Envelhecimento/metabolismo , Animais , Western Blotting , Transtornos Cognitivos/metabolismo , Dieta , Modelos Animais de Doenças , Focalização Isoelétrica , Masculino , Espectrometria de Massas , Nitrosação , Proteômica , Ratos , Ratos Endogâmicos F344 , Tirosina/análogos & derivados
15.
Endocrinology ; 154(8): 2807-20, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23736296

RESUMO

Although glucocorticoids (GCs) are known to exert numerous effects in the hippocampus, their chronic regulatory functions remain poorly understood. Moreover, evidence is inconsistent regarding the long-standing hypothesis that chronic GC exposure promotes brain aging/Alzheimer disease. Here, we adrenalectomized male F344 rats at 15 months of age, maintained them for 3 months with implanted corticosterone (CORT) pellets producing low or intermediate (glucocorticoid receptor-activating) blood levels of CORT, and performed microarray/pathway analyses in hippocampal CA1. We defined the chronic GC-dependent transcriptome as 393 genes that exhibited differential expression between intermediate and low CORT groups. Short-term CORT (4 days) did not recapitulate this transcriptome. Functional processes/pathways overrepresented by chronic CORT-up-regulated genes included learning/plasticity, differentiation, glucose metabolism, and cholesterol biosynthesis, whereas processes overrepresented by CORT-down-regulated genes included inflammatory/immune/glial responses and extracellular structure. These profiles indicate that GCs chronically activate neuronal/metabolic processes while coordinately repressing a glial axis of reactivity/inflammation. We then compared the GC transcriptome with a previously defined hippocampal aging transcriptome, revealing a high proportion of common genes. Although CORT and aging moved expression of some common genes in the same direction, the majority were shifted in opposite directions by CORT and aging (eg, glial inflammatory genes down-regulated by CORT are up-regulated with aging). These results contradict the hypothesis that GCs simply promote brain aging and also suggest that the opposite direction shifts during aging reflect resistance to CORT regulation. Therefore, we propose a new model in which aging-related GC resistance develops in some target pathways, whereas GC overstimulation develops in others, together generating much of the brain aging phenotype.


Assuntos
Região CA1 Hipocampal/metabolismo , Corticosterona/farmacologia , Glucocorticoides/farmacologia , Transcriptoma/efeitos dos fármacos , Adrenalectomia , Envelhecimento/genética , Animais , Peso Corporal/efeitos dos fármacos , Corticosterona/sangue , Ingestão de Líquidos/efeitos dos fármacos , Implantes de Medicamento , Ingestão de Alimentos/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Imuno-Histoquímica , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Ratos , Ratos Endogâmicos F344 , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética
16.
Neurobiol Aging ; 34(8): 1977-87, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23545425

RESUMO

The prevalence of obesity and type 2 diabetes increases with age. Despite this, few studies have examined these conditions simultaneously in aged animals, and fewer studies have measured the impact of these conditions on brain function. Using an established animal model of brain aging (F344 rats), we investigated whether a high-fat diet (HFD) exacerbates cognitive decline and the hippocampal calcium-dependent afterhyperpolarization (a marker of age-dependent calcium dysregulation). Young and mid-aged animals were maintained on control or HFD for 4.5 months, and peripheral metabolic variables, cognitive function, and electrophysiological responses to insulin in the hippocampus were measured. HFD increased lipid accumulation in the periphery, although overt diabetes did not develop, nor were spatial learning and memory altered. Hippocampal adiponectin levels were reduced in aging animals but were unaffected by HFD. For the first time, however, we show that the AHP is sensitive to insulin, and that this sensitivity is reduced by HFD. Interestingly, although peripheral glucose regulation was relatively insensitive to HFD, the brain appeared to show greater sensitivity to HFD in F344 rats.


Assuntos
Envelhecimento/metabolismo , Envelhecimento/fisiologia , Cognição , Dieta Hiperlipídica/efeitos adversos , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Transtornos da Memória/etiologia , Transtornos da Memória/psicologia , Adiponectina/metabolismo , Envelhecimento/psicologia , Animais , Cálcio/metabolismo , Diabetes Mellitus Tipo 2/etiologia , Progressão da Doença , Masculino , Transtornos da Memória/metabolismo , Obesidade/etiologia , Ratos , Ratos Endogâmicos F344 , Fatores de Risco
17.
PLoS One ; 7(7): e40128, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22792227

RESUMO

BACKGROUND: Many aging changes seem similar to those elicited by sleep-deprivation and psychosocial stress. Further, sleep architecture changes with age suggest an age-related loss of sleep. Here, we hypothesized that sleep deprivation in young subjects would elicit both stress and aging-like transcriptional responses. METHODOLOGY/PRINCIPAL FINDINGS: F344 rats were divided into control and sleep deprivation groups. Body weight, adrenal weight, corticosterone level and hippocampal CA1 transcriptional profiles were measured. A second group of animals was exposed to novel environment stress (NES), and their hippocampal transcriptional profiles measured. A third cohort exposed to control or SD was used to validate transcriptional results with Western blots. Microarray results were statistically contrasted with prior transcriptional studies. Microarray results pointed to sleep pressure signaling and macromolecular synthesis disruptions in the hippocampal CA1 region. Animals exposed to NES recapitulated nearly one third of the SD transcriptional profile. However, the SD-aging relationship was more complex. Compared to aging, SD profiles influenced a significant subset of genes. mRNA associated with neurogenesis and energy pathways showed agreement between aging and SD, while immune, glial, and macromolecular synthesis pathways showed SD profiles that opposed those seen in aging. CONCLUSIONS/SIGNIFICANCE: We conclude that although NES and SD exert similar transcriptional changes, selective presynaptic release machinery and Homer1 expression changes are seen in SD. Among other changes, the marked decrease in Homer1 expression with age may represent an important divergence between young and aged brain response to SD. Based on this, it seems reasonable to conclude that therapeutic strategies designed to promote sleep in young subjects may have off-target effects in the aged. Finally, this work identifies presynaptic vesicular release and intercellular adhesion molecular signatures as novel therapeutic targets to counter effects of SD in young subjects.


Assuntos
Envelhecimento/genética , Região CA1 Hipocampal/metabolismo , Privação do Sono/genética , Privação do Sono/metabolismo , Estresse Fisiológico/genética , Transcriptoma , Animais , Análise por Conglomerados , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Masculino , Proteômica , Ratos , Transdução de Sinais
18.
J Alzheimers Dis ; 30(4): 943-61, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22495349

RESUMO

Thiazolidinediones (TZDs) are agonists at peroxisome proliferator-activated gamma-type (PPAR-γ) receptors and are used clinically for the treatment of type 2 diabetes where they have been shown to reestablish insulin sensitivity, improve lipid profiles, and reduce inflammation. Recent work also suggests that TZDs may be beneficial in Alzheimer's disease (AD), ameliorating cognitive decline early in the disease process. However, there have been only a few studies identifying mechanisms through which cognitive benefits may be exerted. Starting at 10 months of age, the triple transgenic mouse model of AD (3xTg-AD) with accelerated amyloid-ß (Aß) deposition and tau pathology was treated with the TZD pioglitazone (PIO-Actos) at 18 mg/Kg body weight/day. After four months, PIO-treated animals showed multiple beneficial effects, including improved learning on the active avoidance task, reduced serum cholesterol, decreased hippocampal amyloid-ß and tau deposits, and enhanced short- and long-term plasticity. Electrophysiological membrane properties and post-treatment blood glucose levels were unchanged by PIO. Gene microarray analyses of hippocampal tissue identified predicted transcriptional responses following TZD treatment as well as potentially novel targets of TZDs, including facilitation of estrogenic processes and decreases in glutamatergic and lipid metabolic/cholesterol dependent processes. Taken together, these results confirm prior animal studies showing that TZDs can ameliorate cognitive deficits associated with AD-related pathology, but also extend these findings by pointing to novel molecular targets in the brain.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/patologia , Aprendizagem/efeitos dos fármacos , Tiazolidinedionas/administração & dosagem , Doença de Alzheimer/psicologia , Animais , Biomarcadores/sangue , Química Encefálica/efeitos dos fármacos , Química Encefálica/fisiologia , Modelos Animais de Doenças , Esquema de Medicação , Feminino , Aprendizagem/fisiologia , Camundongos , Camundongos Transgênicos , Pioglitazona , Fatores de Tempo
19.
PLoS One ; 6(10): e26812, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22046366

RESUMO

Healthy brain aging and cognitive function are promoted by exercise. The benefits of exercise are attributed to several mechanisms, many which highlight its neuroprotective role via actions that enhance neurogenesis, neuronal morphology and/or neurotrophin release. However, the brain is also composed of glial and vascular elements, and comparatively less is known regarding the effects of exercise on these components in the aging brain. Here, we show that aerobic exercise at mid-age decreased markers of unhealthy brain aging including astrocyte hypertrophy, a hallmark of brain aging. Middle-aged female mice were assigned to a sedentary group or provided a running wheel for six weeks. Exercise decreased hippocampal astrocyte and myelin markers of aging but increased VEGF, a marker of angiogenesis. Brain vascular casts revealed exercise-induced structural modifications associated with improved endothelial function in the periphery. Our results suggest that age-related astrocyte hypertrophy/reactivity and myelin dysregulation are aggravated by a sedentary lifestyle and accompanying reductions in vascular function. However, these effects appear reversible with exercise initiated at mid-age. As this period of the lifespan coincides with the appearance of multiple markers of brain aging, including initial signs of cognitive decline, it may represent a window of opportunity for intervention as the brain appears to still possess significant vascular plasticity. These results may also have particular implications for aging females who are more susceptible than males to certain risk factors which contribute to vascular aging.


Assuntos
Envelhecimento/fisiologia , Encéfalo/fisiopatologia , Fenômenos Fisiológicos Cardiovasculares , Neuroglia/patologia , Condicionamento Físico Animal/fisiologia , Animais , Biomarcadores , Encéfalo/irrigação sanguínea , Encéfalo/patologia , Feminino , Camundongos
20.
Cell Calcium ; 50(6): 548-58, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21978418

RESUMO

Neuronal Ca(2+) dyshomeostasis associated with cognitive impairment and mediated by changes in several Ca(2+) sources has been seen in animal models of both aging and diabetes. In the periphery, dysregulation of intracellular Ca(2+) signals may contribute to the development of insulin resistance. In the brain, while it is well-established that type 2 diabetes mellitus is a risk factor for the development of dementia in the elderly, it is not clear whether Ca(2+) dysregulation might also affect insulin sensitivity and glucose utilization. Here we present a combination of imaging techniques testing the disappearance of the fluorescent glucose analog 2-(N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino)-2-deoxyglucose (2-NBDG) as an indication of glycolytic activity in neurons and astrocytes. Our work shows that glucose utilization at rest is greater in neurons compared to astrocytes, and ceases upon activation in neurons with little change in astrocytes. Pretreatment of hippocampal cultures with pioglitazone, a drug used in the treatment of type 2 diabetes, significantly reduced glycolytic activity in neurons and enhanced it in astrocytes. This series of experiments, including Fura-2 and NADH imaging, provides results that are consistent with the idea that Ca(2+) levels may rapidly alter glycolytic activity, and that downstream events beyond Ca(2+) dysregulation with aging, may alter cellular metabolism in the brain.


Assuntos
4-Cloro-7-nitrobenzofurazano/análogos & derivados , Astrócitos/metabolismo , Cálcio/metabolismo , Desoxiglucose/análogos & derivados , Corantes Fluorescentes/análise , NAD/metabolismo , Neurônios/metabolismo , Tiazolidinedionas/farmacologia , 4-Cloro-7-nitrobenzofurazano/análise , Animais , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Sinalização do Cálcio/efeitos dos fármacos , Células Cultivadas , Desoxiglucose/análise , Fura-2 , Glucose/metabolismo , Glicólise/efeitos dos fármacos , Hipocampo/citologia , NAD/efeitos dos fármacos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Pioglitazona , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...