Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
J Neurochem ; 168(5): 781-800, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38317494

RESUMO

Hormone-sensitive lipase (HSL) is active throughout the brain and its genetic ablation impacts brain function. Its activity in the brain was proposed to regulate bioactive lipid availability, namely eicosanoids that are inflammatory mediators and regulate cerebral blood flow (CBF). We aimed at testing whether HSL deletion increases susceptibility to neuroinflammation and impaired brain perfusion upon diet-induced obesity. HSL-/-, HSL+/-, and HSL+/+ mice of either sex were fed high-fat diet (HFD) or control diet for 8 weeks, and then assessed in behavior tests (object recognition, open field, and elevated plus maze), metabolic tests (insulin and glucose tolerance tests and indirect calorimetry in metabolic cages), and CBF determination by arterial spin labeling (ASL) magnetic resonance imaging (MRI). Immunofluorescence microscopy was used to determine coverage of blood vessels, and morphology of astrocytes and microglia in brain slices. HSL deletion reduced CBF, most prominently in cortex and hippocampus, while HFD feeding only lowered CBF in the hippocampus of wild-type mice. CBF was positively correlated with lectin-stained vessel density. HSL deletion did not exacerbate HFD-induced microgliosis in the hippocampus and hypothalamus. HSL-/- mice showed preserved memory performance when compared to wild-type mice, and HSL deletion did not significantly aggravate HFD-induced memory impairment in object recognition tests. In contrast, HSL deletion conferred protection against HFD-induced obesity, glucose intolerance, and insulin resistance. Altogether, this study points to distinct roles of HSL in periphery and brain during diet-induced obesity. While HSL-/- mice were protected against metabolic syndrome development, HSL deletion reduced brain perfusion without leading to aggravated HFD-induced neuroinflammation and memory dysfunction.


Assuntos
Circulação Cerebrovascular , Dieta Hiperlipídica , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade , Animais , Obesidade/genética , Camundongos , Dieta Hiperlipídica/efeitos adversos , Circulação Cerebrovascular/fisiologia , Masculino , Feminino , Esterol Esterase/genética , Esterol Esterase/metabolismo , Memória/fisiologia , Deleção de Genes , Transtornos da Memória/etiologia , Transtornos da Memória/genética , Encéfalo/patologia , Encéfalo/metabolismo
2.
J Neurochem ; 168(5): 910-954, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38183680

RESUMO

Although we have learned much about how the brain fuels its functions over the last decades, there remains much still to discover in an organ that is so complex. This article lays out major gaps in our knowledge of interrelationships between brain metabolism and brain function, including biochemical, cellular, and subcellular aspects of functional metabolism and its imaging in adult brain, as well as during development, aging, and disease. The focus is on unknowns in metabolism of major brain substrates and associated transporters, the roles of insulin and of lipid droplets, the emerging role of metabolism in microglia, mysteries about the major brain cofactor and signaling molecule NAD+, as well as unsolved problems underlying brain metabolism in pathologies such as traumatic brain injury, epilepsy, and metabolic downregulation during hibernation. It describes our current level of understanding of these facets of brain energy metabolism as well as a roadmap for future research.


Assuntos
Encéfalo , Metabolismo Energético , Animais , Humanos , Encéfalo/metabolismo
3.
J Neuroinflammation ; 21(1): 173, 2024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-39014461

RESUMO

Dietary patterns that include an excess of foods rich in saturated fat are associated with brain dysfunction. Although microgliosis has been proposed to play a key role in the development of brain dysfunction in diet-induced obesity (DIO), neuroinflammation with cytokine over-expression is not always observed. Thus, mechanisms by which microglia contribute to brain impairment in DIO are uncertain. Using the BV2 cell model, we investigated the gliosis profile of microglia exposed to palmitate (200 µmol/L), a saturated fatty acid abundant in high-fat diet and in the brain of obese individuals. We observed that microglia respond to a 24-hour palmitate exposure with increased proliferation, and with a metabolic network rearrangement that favors energy production from glycolysis rather than oxidative metabolism, despite stimulated mitochondria biogenesis. In addition, while palmitate did not induce increased cytokine expression, it modified the protein cargo of released extracellular vesicles (EVs). When administered intra-cerebroventricularly to mice, EVs secreted from palmitate-exposed microglia in vitro led to memory impairment, depression-like behavior, and glucose intolerance, when compared to mice receiving EVs from vehicle-treated microglia. We conclude that microglia exposed to palmitate can mediate brain dysfunction through the cargo of shed EVs.


Assuntos
Vesículas Extracelulares , Camundongos Endogâmicos C57BL , Microglia , Palmitatos , Animais , Microglia/efeitos dos fármacos , Microglia/metabolismo , Camundongos , Vesículas Extracelulares/efeitos dos fármacos , Vesículas Extracelulares/metabolismo , Palmitatos/toxicidade , Palmitatos/farmacologia , Masculino , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Dieta Hiperlipídica/efeitos adversos , Citocinas/metabolismo
4.
Neurochem Res ; 49(2): 338-347, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37794263

RESUMO

Sphingosine-1-phosphate (S1P) is a phosphosphingolipid with pleiotropic biological functions. S1P acts as an intracellular second messenger, as well as extracellular ligand to five G-protein coupled receptors (S1PR1-5). In the brain, S1P regulates neuronal proliferation, apoptosis, synaptic activity and neuroglia activation. Moreover, S1P metabolism alterations have been reported in neurodegenerative disorders. We have previously reported that S1PRs are present in nerve terminals, exhibiting distinct sub-synaptic localization and neuromodulation actions. Since type 2 diabetes (T2D) causes synaptic dysfunction, we hypothesized that S1P signaling is modified in nerve terminals. In this study, we determined the density of S1PRs in cortical synaptosomes from insulin-resistant Goto-Kakizaki (GK) rats and Wistar controls, and from mice fed a high-fat diet (HFD) and low-fat-fed controls. Relative to their controls, GK rats showed similar cortical S1P concentration despite higher S1P levels in plasma, yet lower density of S1PR1, S1PR2 and S1PR4 in nerve-terminal-enriched membranes. HFD-fed mice exhibited increased plasma and cortical concentrations of S1P, and decreased density of S1PR1 and S1PR4. These findings point towards altered S1P signaling in synapses of insulin resistance and diet-induced obesity models, suggesting a role of S1P signaling in T2D-associated synaptic dysfunction.


Assuntos
Diabetes Mellitus Tipo 2 , Receptores de Lisoesfingolipídeo , Ratos , Camundongos , Animais , Receptores de Esfingosina-1-Fosfato , Receptores de Lisoesfingolipídeo/metabolismo , Camundongos Obesos , Insulina , Ratos Wistar , Esfingosina/metabolismo , Dieta Hiperlipídica/efeitos adversos , Lisofosfolipídeos/metabolismo
5.
Biochem Soc Trans ; 51(1): 287-301, 2023 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-36606696

RESUMO

The pathophysiological mechanisms intersecting metabolic and neurodegenerative disorders include insulin resistance, which has a strong involvement of environmental factors. Besides central regulation of whole-body homeostasis, insulin in the central nervous system controls molecular signalling that is critical for cognitive performance, namely signalling through pathways that modulate synaptic transmission and plasticity, and metabolism in neurons and astrocytes. This review provides an overview on how insulin signalling in the brain might regulate brain energy metabolism, and further identified molecular mechanisms by which brain insulin resistance might impair synaptic fuelling, and lead to cognitive deterioration.


Assuntos
Resistência à Insulina , Humanos , Insulina/metabolismo , Encéfalo/metabolismo , Sistema Nervoso Central/metabolismo , Metabolismo Energético
6.
Nutr Neurosci ; 26(11): 1090-1102, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36222315

RESUMO

Background: Obesity constitutes a risk factor for cognitive impairment. In rodent models, long-term exposure to obesogenic diets leads to hippocampal taurine accumulation. Since taurine has putative cyto-protective effects, hippocampal taurine accumulation in obese and diabetic models might constitute a counteracting response to metabolic stress. Objective: We tested the hypothesis that treatment with taurine or with N-acetylcysteine (NAC), which provides cysteine for the synthesis of taurine and glutathione, prevent high-fat diet (HFD)-associated hippocampal alterations and memory impairment. Methods: Female mice were fed either a regular diet or HFD. Some mice had access to 3%(w/v) taurine or 3%(w/v) NAC in the drinking water. After 2 months, magnetic resonance spectroscopy (MRS) was used to measure metabolite profiles. Memory was assessed in novel object and novel location recognition tests. Results: HFD feeding caused memory impairment in both tests, and reduced concentration of lactate, phosphocreatine-to-creatine ratio, and the neuronal marker N-acetylaspartate in the hippocampus. Taurine and NAC prevented HFD-induced memory impairment and N-acetylaspartate reduction. NAC, but not taurine, prevented the reduction of lactate and phosphocreatine-to-creatine ratio. MRS revealed NAC/taurine-induced increase of hippocampal glutamate and GABA levels. Conclusion: NAC and taurine can prevent memory impairment, while only NAC prevents alterations of metabolite concentrations in HFD-exposed female mice.


Assuntos
Acetilcisteína , Dieta Hiperlipídica , Camundongos , Animais , Feminino , Acetilcisteína/uso terapêutico , Acetilcisteína/farmacologia , Dieta Hiperlipídica/efeitos adversos , Creatina/metabolismo , Fosfocreatina/metabolismo , Obesidade/metabolismo , Transtornos da Memória/etiologia , Transtornos da Memória/prevenção & controle , Hipocampo/metabolismo , Lactatos/metabolismo , Camundongos Endogâmicos C57BL
7.
Int J Mol Sci ; 24(12)2023 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-37373212

RESUMO

Epidemiological studies have associated plasma galectin-4 (Gal-4) levels with prevalent and incident diabetes, and with an increased risk of coronary artery disease. To date, data regarding possible associations between plasma Gal-4 and stroke are lacking. Using linear and logistic regression analyses, we tested Gal-4 association with prevalent stroke in a population-based cohort. Additionally, in mice fed a high-fat diet (HFD), we investigated whether plasma Gal-4 increases in response to ischemic stroke. Plasma Gal-4 was higher in subjects with prevalent ischemic stroke, and was associated with prevalent ischemic stroke (odds ratio 1.52; 95% confidence interval 1.01-2.30; p = 0.048) adjusted for age, sex, and covariates of cardiometabolic health. Plasma Gal-4 increased after experimental stroke in both controls and HFD-fed mice. HFD exposure was devoid of impact on Gal-4 levels. This study demonstrates higher plasma Gal-4 levels in both experimental stroke and in humans that experienced ischemic stroke.


Assuntos
AVC Isquêmico , Acidente Vascular Cerebral , Humanos , Animais , Camundongos , Galectina 4 , Galectinas , Galectina 3 , Biomarcadores
8.
J Lipid Res ; 63(5): 100195, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35300984

RESUMO

Hormone-sensitive lipase (HSL) is mainly present in adipose tissue where it hydrolyzes diacylglycerol. Although expression of HSL has also been reported in the brain, its presence in different cellular compartments is uncertain, and its role in regulating brain lipid metabolism remains hitherto unexplored. We hypothesized that HSL might play a role in regulating the availability of bioactive lipids necessary for neuronal function and therefore investigated whether dampening HSL activity could lead to brain dysfunction. In mice, we found HSL protein and enzymatic activity throughout the brain, localized within neurons and enriched in synapses. HSL-null mice were then analyzed using a battery of behavioral tests. Relative to wild-type littermates, HSL-null mice showed impaired short-term and long-term memory, yet preserved exploratory behaviors. Molecular analysis of the cortex and hippocampus showed increased expression of genes involved in glucose utilization in the hippocampus, but not cortex, of HSL-null mice compared with controls. Furthermore, lipidomics analyses indicated an impact of HSL deletion on the profile of bioactive lipids, including a decrease in endocannabinoids and eicosanoids that are known to modulate neuronal activity, cerebral blood flow, and inflammation processes. Accordingly, mild increases in the expression of proinflammatory cytokines in HSL mice compared with littermates were suggestive of low-grade inflammation. We conclude that HSL has a homeostatic role in maintaining pools of lipids required for normal brain function. It remains to be tested, however, whether the recruitment of HSL for the synthesis of these lipids occurs during increased neuronal activity or whether HSL participates in neuroinflammatory responses.


Assuntos
Lipídeos , Esterol Esterase , Animais , Inflamação , Camundongos , Camundongos Knockout , Esterol Esterase/genética , Esterol Esterase/metabolismo , Sinapses/metabolismo
9.
J Neurochem ; 163(1): 6-7, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35996309

RESUMO

This Editorial highlights an interesting study in the current issue of the Journal of Neurochemistry in which Zhou et al. report new data showing that the ablation of serine racemase increases local insulin production in neurons of the hippocampus. The authors explored some of the possible mechanisms mediating the interaction between dampening production of D-serine and the local synthesis of insulin, and they further propose that stimulating insulin production could counteract hippocampal insulin resistance in Alzheimer's disease (AD). Most importantly, they leave open a number of questions that need to be experimentally addressed to ascertain whether D-serine modulation of neuronal insulin expression can effectively improve insulin sensitivity in AD, as well as in metabolic disease with neurological impact.


Assuntos
Doença de Alzheimer , Resistência à Insulina , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Hipocampo/metabolismo , Humanos , Insulina/farmacologia , Racemases e Epimerases , Serina/metabolismo
10.
Neurochem Res ; 47(10): 3114-3125, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35781853

RESUMO

Sphingosine-1-phosphate (S1P) is best known for its roles as vascular and immune regulator. Besides, it is also present in the central nervous system (CNS) where it can act as neuromodulator via five S1P receptors (S1PRs), and thus control neurotransmitter release. The distribution of S1PRs in the active zone and postsynaptic density of CNS synapses remains unknown. In the current study, we investigated the localization of S1PR1-5 in synapses of the mouse cortex. Cortical nerve terminals purified in a sucrose gradient were endowed with all five S1PRs. Further subcellular fractionation of cortical nerve terminals revealed S1PR2 and S1PR4 immunoreactivity in the active zone of presynaptic nerve terminals. Interestingly, only S1PR2 and S1PR3 immunoreactivity was found in the postsynaptic density. All receptors were present outside the active zone of nerve terminals. Neurons in the mouse cortex and primary neurons in culture showed immunoreactivity against all five S1PRs, and Ca2+ imaging revealed that S1P inhibits spontaneous neuronal activity in a dose-dependent fashion. When testing selective agonists for each of the receptors, we found that only S1PR1, S1PR2 and S1PR4 control spontaneous neuronal activity. We conclude that S1PR2 and S1PR4 are located in the active zone of nerve terminals and inhibit neuronal activity. Future studies need to test whether these receptors modulate stimulation-induced neurotransmitter release.


Assuntos
Receptores de Lisoesfingolipídeo , Esfingosina , Animais , Lisofosfolipídeos , Camundongos , Neurônios , Esfingosina/análogos & derivados , Sinapses
11.
Int J Mol Sci ; 24(1)2022 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-36613742

RESUMO

Excess dietary salt reduces resting cerebral blood flow (CBF) and vascular reactivity, which can limit the fueling of neuronal metabolism. It is hitherto unknown whether metabolic derangements induced by high-salt-diet (HSD) exposure during adulthood are reversed by reducing salt intake. In this study, male and female mice were fed an HSD from 9 to 16 months of age, followed by a normal-salt diet (ND) thereafter until 23 months of age. Controls were continuously fed either ND or HSD. CBF and metabolite profiles were determined longitudinally by arterial spin labeling magnetic resonance imaging and magnetic resonance spectroscopy, respectively. HSD reduced cortical and hippocampal CBF, which recovered after dietary salt normalization, and affected hippocampal but not cortical metabolite profiles. Compared to ND, HSD increased hippocampal glutamine and phosphocreatine levels and decreased creatine and choline levels. Dietary reversal only allowed recovery of glutamine levels. Histology analyses revealed that HSD reduced the dendritic arborization and spine density of cortical and hippocampal neurons, which were not recovered after dietary salt normalization. We conclude that sustained HSD exposure throughout adulthood causes permanent structural and metabolic alterations to the mouse brain that are not fully normalized by lowering dietary salt during aging.


Assuntos
Glutamina , Cloreto de Sódio na Dieta , Camundongos , Masculino , Feminino , Animais , Cloreto de Sódio na Dieta/metabolismo , Glutamina/metabolismo , Hipocampo/metabolismo , Dieta , Circulação Cerebrovascular/fisiologia
12.
J Neurosci Res ; 97(8): 1004-1017, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31044444

RESUMO

Diabetes impacts the central nervous system predisposing to cognitive decline. While glucose is the main source of energy fueling the adult brain, brain glycogen is necessary for adequate neuronal function, synaptic plasticity and memory. In this study, we tested the hypothesis that brain glycogen metabolism is impaired in type 2 diabetes (T2D). 13 C magnetic resonance spectroscopy (MRS) during [1-13 C]glucose i.v. infusion was employed to detect 13 C incorporation into whole-brain glycogen in male Goto-Kakizaki (GK) rats, a lean model of T2D, and control Wistar rats. Labeling from [1-13 C]glucose into brain glycogen occurred at a rate of 0.25 ± 0.12 and 0.48 ± 0.22 µmol/g/h in GK and Wistar rats, respectively (p = 0.028), despite similar brain glycogen concentrations. In addition, the appearance of [1-13 C]glucose in the brain was used to evaluate glucose transport and consumption. T2D caused a 31% reduction (p = 0.031) of the apparent maximum transport rate (Tmax ) and a tendency for reduced cerebral metabolic rate of glucose (CMRglc ; -29%, p = 0.062), indicating impaired glucose utilization in T2D. After MRS in vivo, gas chromatography-mass spectrometry was employed to measure regional 13 C fractional enrichment of glucose and glycogen in the cortex, hippocampus, striatum, and hypothalamus. The diabetes-induced reduction in glycogen labeling was most prominent in the hippocampus and hypothalamus, which are crucial for memory and energy homeostasis, respectively. These findings were further supported by changes in the phosphorylation rate of glycogen synthase, as analyzed by Western blotting. Altogether, the present results indicate that T2D is associated with impaired brain glycogen metabolism.


Assuntos
Encéfalo/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Glicogênio/metabolismo , Animais , Modelos Animais de Doenças , Transportador de Glucose Tipo 1/metabolismo , Espectroscopia de Ressonância Magnética , Masculino , Ratos Wistar
13.
Int J Obes (Lond) ; 43(6): 1295-1304, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30301962

RESUMO

BACKGROUND/OBJECTIVES: High-fat diet consumption is known to trigger an inflammatory response in the hypothalamus, which has been characterized by an initial expression of pro-inflammatory genes followed by hypothalamic astrocytosis, microgliosis, and the appearance of neuronal injury markers. The specific effects of high-fat diet on hypothalamic energy metabolism and neurotransmission are however not yet known and have not been investigated before. SUBJECTS/METHODS: We used 1H and 13C magnetic resonance spectroscopy (MRS) and immunofluorescence techniques to evaluate in vivo the consequences of high-saturated fat diet administration to mice, and explored the effects on hypothalamic metabolism in three mouse cohorts at different time points for up to 4 months. RESULTS: We found that high-fat diet increases significantly the hypothalamic levels of glucose (P < 0.001), osmolytes (P < 0.001), and neurotransmitters (P < 0.05) from 2 months of diet, and alters the rates of metabolic (P < 0.05) and neurotransmission fluxes (P < 0.001), and the contribution of non-glycolytic substrates to hypothalamic metabolism (P < 0.05) after 10 weeks of high-fat feeding. CONCLUSIONS/INTERPRETATION: We report changes that reveal a high-fat diet-induced alteration of hypothalamic metabolism and neurotransmission that is quantifiable by 1H and 13C MRS in vivo, and present the first evidence of the extension of the inflammation pathology to a localized metabolic imbalance.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Gorduras na Dieta/farmacologia , Metabolismo Energético/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Animais , Gorduras na Dieta/administração & dosagem , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Hipotálamo/fisiopatologia , Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo
14.
Neurochem Res ; 44(1): 102-116, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29616444

RESUMO

In the past couple of decades, major efforts were made to increase reliability of metabolic assessments by magnetic resonance methods. Magnetic resonance spectroscopy (MRS) has been valuable for providing in vivo evidence and investigating biomarkers in neuropsychiatric disorders, namely schizophrenia. Alterations of glutamate and glutamine levels in brains of schizophrenia patients relative to healthy subjects are generally interpreted as markers of glutamatergic dysfunction. However, only a small fraction of MRS-detectable glutamate is involved in neurotransmission. Here we review and discuss brain metabolic processes that involve glutamate and that are likely to be implicated in neuropsychiatric disorders.


Assuntos
Encéfalo/metabolismo , Metabolismo Energético/fisiologia , Ácido Glutâmico/metabolismo , Espectroscopia de Ressonância Magnética/métodos , Esquizofrenia/metabolismo , Animais , Encéfalo/diagnóstico por imagem , Humanos , Esquizofrenia/diagnóstico por imagem
15.
Glia ; 66(3): 477-491, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29120073

RESUMO

Astrocytes play an important role in glutamatergic neurotransmission, namely by clearing synaptic glutamate and converting it into glutamine that is transferred back to neurons. The rate of this glutamate-glutamine cycle (VNT ) has been proposed to couple to that of glucose utilization and of neuronal tricarboxylic acid (TCA) cycle. In this study, we tested the hypothesis that glutamatergic neurotransmission is also coupled to the TCA cycle rate in astrocytes. For that we investigated energy metabolism by means of magnetic resonance spectroscopy (MRS) in the primary visual cortex of tree shrews (Tupaia belangeri) under light isoflurane anesthesia at rest and during continuous visual stimulation. After identifying the activated cortical volume by blood oxygenation level-dependent functional magnetic resonance imaging, 1 H MRS was performed to measure stimulation-induced variations in metabolite concentrations. Relative to baseline, stimulation of cortical activity for 20 min caused a reduction of glucose concentration by -0.34 ± 0.09 µmol/g (p < 0.001), as well as a -9% ± 1% decrease of the ratio of phosphocreatine-to-creatine (p < 0.05). Then 13 C MRS during [1,6-13 C]glucose infusion was employed to measure fluxes of energy metabolism. Stimulation of glutamatergic activity, as indicated by a 20% increase of VNT , resulted in increased TCA cycle rates in neurons by 12% ( VTCAn, p < 0.001) and in astrocytes by 24% ( VTCAg, p = 0.007). We further observed linear relationships between VNT and both VTCAn and VTCAg. Altogether, these results suggest that in the tree shrew primary visual cortex glutamatergic neurotransmission is linked to overall glucose oxidation and to mitochondrial metabolism in both neurons and astrocytes.


Assuntos
Astrócitos/metabolismo , Ácido Glutâmico/metabolismo , Glutamina/metabolismo , Neurônios/metabolismo , Córtex Visual/metabolismo , Animais , Mapeamento Encefálico , Espectroscopia de Ressonância Magnética Nuclear de Carbono-13 , Ciclo do Ácido Cítrico/fisiologia , Feminino , Glucose/metabolismo , Imageamento por Ressonância Magnética , Masculino , Mitocôndrias/metabolismo , Oxirredução , Oxigênio/metabolismo , Espectroscopia de Prótons por Ressonância Magnética , Distribuição Aleatória , Tupaiidae , Córtex Visual/diagnóstico por imagem , Percepção Visual/fisiologia
16.
MAGMA ; 31(2): 341-354, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29027041

RESUMO

OBJECTIVE: We monitored hepatic lipid content (HLC) and fatty acid (FA) composition in the context of enhanced lipid handling induced by a metabolic high-fat diet (HFD) challenge and fasting. MATERIALS AND METHODS: Mice received a control diet (10% of kilocalories from fat, N = 14) or an HFD (45% or 60% of kilocalories from fat, N = 10 and N = 16, respectively) for 26 weeks. A subset of five mice receiving an HFD (60% of kilocalories from fat) were switched to the control diet for the final 7 weeks. At nine time points, magnetic resonance spectroscopy was performed in vivo at 14.1 T, interleaved with glucose tolerance tests. RESULTS: Glucose intolerance promptly developed with the HFD, followed by a progressive increase of fasting insulin level, simultaneously with that of HLC. These metabolic defects were normalized by dietary reversal. HFD feeding immediately increased polyunsaturation of hepatic FA, before lipid accumulation. Fasting-induced changes in hepatic lipids (increased HLC and FA polyunsaturation, decreased FA monounsaturation) in control-diet-fed mice were not completely reproduced in HFD-fed mice, not even after dietary reversal. CONCLUSION: A similar adaptation of hepatic lipids to both fasting and an HFD suggests common mechanisms of lipid trafficking from adipose tissue to the liver. Altered hepatic lipid handling with fasting indicates imperfect metabolic recovery from HFD exposure.


Assuntos
Dieta Hiperlipídica , Ácidos Graxos Insaturados/química , Ácidos Graxos/química , Lipídeos/química , Fígado/metabolismo , Tecido Adiposo/metabolismo , Animais , Teste de Tolerância a Glucose , Insulina/sangue , Espectroscopia de Ressonância Magnética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Fenótipo
17.
J Neurochem ; 142(5): 767-775, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28664650

RESUMO

Environmental stress can interact with genetic predisposition to increase the risk of developing psychopathology. In this work, we tested the hypothesis that social isolation stress interacts with impaired glutathione synthesis and have cumulative effects on the neurochemical profile of the frontal cortex. A mouse model with chronic glutathione deficit induced by knockout (-/-) of the glutamate-cysteine ligase modulatory subunit (Gclm) was exposed to social isolation stress from weaning to post-natal day 65. Using magnetic resonance methods at high-field (14.1 T), we analysed the neurochemical profile in the frontal cortex, brain size and ventricular volume of adult animals. Glutathione deficit was accompanied by elevated concentrations of N-acetylaspartate, alanine, and glutamine, as well as the ratio of glutamine-to-glutamate (Gln/Glu), and by a reduction in levels of myo-inositol and choline-containing compounds in the frontal cortex of -/- animals with respect to wild-type littermates. Although there was no significant interaction between social isolation stress and glutathione deficiency, mice reared in isolation displayed lower myo-inositol concentration (-8.4%, p < 0.05) and larger Gln/Glu (+7.6%, p < 0.05), relative to those in group housing. Furthermore, glutathione deficiency caused a reduction in whole brain volume and enlargement of ventricles, but social isolation had no effect on these parameters. We conclude that social isolation caused neurochemical alterations that may add to those associated to impaired glutathione synthesis.


Assuntos
Lobo Frontal/metabolismo , Ácido Glutâmico/metabolismo , Glutamina/metabolismo , Glutationa/deficiência , Inositol/metabolismo , Isolamento Social , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Isolamento Social/psicologia , Estresse Psicológico/metabolismo , Estresse Psicológico/psicologia
18.
J Neurosci Res ; 95(11): 2297-2306, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28316083

RESUMO

Barbiturates, commonly used as general anaesthetics, depress neuronal activity and thus cerebral metabolism. Moreover, they are likely to disrupt the metabolic support of astrocytes to neurons, as well as the uptake of nutrients from circulation. By employing 13 C magnetic resonance spectroscopy (MRS) in vivo at high magnetic field, we characterized neuronal and astrocytic pathways of energy metabolism in the rat cortex under thiopental anaesthesia. The neuronal tricarboxylic acid (TCA) cycle rate was 0.46 ± 0.02 µmol/g/min, and the rate of the glutamate-glutamine cycle was 0.09 ± 0.02 µmol/g/min. In astrocytes, the TCA cycle rate was 0.16 ± 0.02 µmol/g/min, accounting for a quarter of whole brain glucose oxidation, pyruvate carboxylase rate was 0.02 ± 0.01 µmol/g/min, and glutamine synthetase was 0.12 ± 0.01 µmol/g/min. Relative to previous experiments under light α-chloralose anaesthesia, thiopental reduced oxidative metabolism in neurons and even more so in astrocytes. Interestingly, total oxidative metabolism in the cortex under thiopental anaesthesia surpassed the rate of pyruvate production by glycolysis, indicating substantial utilisation of substrates other than glucose, likely plasma lactate. © 2017 Wiley Periodicals, Inc.


Assuntos
Anestésicos Intravenosos/farmacologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Metabolismo Energético/efeitos dos fármacos , Espectroscopia de Ressonância Magnética/métodos , Tiopental/farmacologia , Animais , Metabolismo Energético/fisiologia , Masculino , Ratos , Ratos Sprague-Dawley
19.
Neurochem Res ; 42(6): 1629-1635, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28083850

RESUMO

Patients with diabetes display a progressive decay in the physiological counter-regulatory response to hypoglycemia, resulting in hypoglycemia unawareness. The mechanism through which the brain adapts to hypoglycemia may involve brain glycogen. We tested the hypothesis that brain glycogen supercompensation following hypoglycemia depends on blood glucose levels during recovery. Conscious rats were submitted to hypoglycemia of 2 mmol/L for 90 min and allowed to recover at different glycemia, controlled by means of i.v. glucose infusion. Brain glycogen concentration was elevated above control levels after 24 h of recovery in the cortex, hippocampus and striatum. This glycogen supercompensation was independent of blood glucose levels in the post-hypoglycemia period. In the absence of a preceding hypoglycemia insult, brain glycogen concentrations were unaltered after 24 h under hyperglycemia. In the hypothalamus, which controls peripheral glucose homeostasis, glycogen levels were unaltered. Overall, we conclude that post-hypoglycemia glycogen supercompensation occurs in several brain areas and its magnitude is independent of plasma glucose levels. By supporting brain metabolism during recurrent hypoglycemia periods, glycogen may have a role in the development of hypoglycemia unawareness.


Assuntos
Encéfalo/metabolismo , Glucose/metabolismo , Glicogênio/metabolismo , Hipoglicemia/metabolismo , Recuperação de Função Fisiológica/fisiologia , Doença Aguda , Animais , Masculino , Ratos , Ratos Sprague-Dawley
20.
J Neurochem ; 136(5): 947-57, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26709861

RESUMO

Adenosine is a neuromodulator that protects neurons from hypoxia. This effect is attributed to the ability of adenosine A1 receptors (A1 R) to inhibit excitatory synaptic transmission. However, A1 R activation also protects non-brain tissues from hypoxic insults by controlling metabolism. Thus, we now tested the hypothesis that A1 R-mediated neuroprotection after a hypoxic insult in superfused hippocampal slices also involves the control of neuronal and astrocytic metabolism. A 90-min hypoxia insult increased lactate, alanine, and pyruvate levels and decreased energy charge (EC), phosphocreatine/creatine ratio, and glutamine content. These metabolic modifications were fully recovered after reoxygenation for 3 h. The presence of the A1 R-selective antagonist 1,3-dipropyl-8-cyclopentylxanthine stimulated glycolysis, prevented the hypoxia-induced decrease of EC, and increased the levels of GABA. A1 R blockade further blunted the recovery of metabolism on reoxygenation after hypoxia, as typified by a sustained decreased EC and an increased mitochondrial metabolism, as confirmed by a greater [U-(13) C]glucose oxidation through the tricarboxylic acid cycle. These results demonstrate that A1 R blockade prevents the recovery of hypoxia-induced metabolic alterations during reoxygenation, which indicates that the ability of A1 R to control primary metabolism in the brain tissue may be a hitherto unrecognized mechanism of A1 R-mediated neuroprotection. This study demonstrates that tonic activation of adenosine A1 receptors (A1 R) plays an important role in the reoxygenation recovery of the metabolic alterations caused by transient hypoxia in rat hippocampal slices. This ability of A1 R to inhibit neuronal metabolism may be a key mechanism by which adenosine affords neuroprotection upon acute hypoxia, thus preventing the long-term impairment of neuronal circuits.


Assuntos
Hipocampo/efeitos dos fármacos , Hipóxia/tratamento farmacológico , Neurônios/metabolismo , Receptor A1 de Adenosina/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Xantinas/farmacologia , Adenosina/metabolismo , Animais , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Hipocampo/metabolismo , Hipóxia/metabolismo , Masculino , Receptor A1 de Adenosina/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA