Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Neurochem Res ; 42(1): 50-63, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26141225

RESUMO

2-Deoxy-D-[14C]glucose ([14C]DG) is commonly used to determine local glucose utilization rates (CMRglc) in living brain and to estimate CMRglc in cultured brain cells as rates of [14C]DG phosphorylation. Phosphorylation rates of [14C]DG and its metabolizable fluorescent analog, 2-(N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino)-2-deoxyglucose (2-NBDG), however, do not take into account differences in the kinetics of transport and metabolism of [14C]DG or 2-NBDG and glucose in neuronal and astrocytic cells in cultures or in single cells in brain tissue, and conclusions drawn from these data may, therefore, not be correct. As a first step toward the goal of quantitative determination of CMRglc in astrocytes and neurons in cultures, the steady-state intracellular-to-extracellular concentration ratios (distribution spaces) for glucose and [14C]DG were determined in cultured striatal neurons and astrocytes as functions of extracellular glucose concentration. Unexpectedly, the glucose distribution spaces rose during extreme hypoglycemia, exceeding 1.0 in astrocytes, whereas the [14C]DG distribution space fell at the lowest glucose levels. Calculated CMRglc was greatly overestimated in hypoglycemic and normoglycemic cells because the intracellular glucose concentrations were too high. Determination of the distribution space for [14C]glucose revealed compartmentation of intracellular glucose in astrocytes, and probably, also in neurons. A smaller metabolic pool is readily accessible to hexokinase and communicates with extracellular glucose, whereas the larger pool is sequestered from hexokinase activity. A new experimental approach using double-labeled assays with DG and glucose is suggested to avoid the limitations imposed by glucose compartmentation on metabolic assays.


Assuntos
Astrócitos/metabolismo , Radioisótopos de Carbono/metabolismo , Desoxiglucose/metabolismo , Glucose/metabolismo , Líquido Intracelular/metabolismo , Neurônios/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/fisiologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Radioisótopos de Carbono/farmacologia , Células Cultivadas , Desoxiglucose/farmacologia , Relação Dose-Resposta a Droga , Feminino , Líquido Intracelular/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Gravidez , Ratos , Ratos Sprague-Dawley
2.
Neurochem Res ; 42(6): 1683-1696, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27822667

RESUMO

Ammonia is neurotoxic, and chronic hyperammonemia is thought to be a major contributing factor to hepatic encephalopathy in patients with liver disease. Portacaval shunting of rats is used as an animal model to study the detrimental metabolic effects of elevated ammonia levels on body tissues, particularly brain and testes that are deleteriously targeted by high blood ammonia. In normal adult rats, the initial uptake of label (expressed as relative concentration) in these organs was relatively low following a bolus intravenous injection of [13N]ammonia compared with lungs, kidneys, liver, and some other organs. The objective of the present study was to determine the distribution of label following intravenous administration of [13N]ammonia among 14 organs in portacaval-shunted rats at 12 weeks after shunt construction. At an early time point (12 s) following administration of [13N]ammonia the relative concentration of label was highest in lung with lower, but still appreciable relative concentrations in kidney and heart. Clearance of 13N from blood and kidney tended to be slower in portacaval-shunted rats versus normal rats during the 2-10 min interval after the injection. At later times post injection, brain and testes tended to have higher-than-normal 13N levels, whereas many other tissues had similar levels in both groups. Thus, reduced removal of ammonia from circulating blood by the liver diverts more ammonia to extrahepatic tissues, including brain and testes, and alters the nitrogen homeostasis in these tissues. These results emphasize the importance of treatment paradigms designed to reduce blood ammonia levels in patients with liver disease.


Assuntos
Amônia/administração & dosagem , Amônia/metabolismo , Encéfalo/metabolismo , Radioisótopos de Nitrogênio/administração & dosagem , Radioisótopos de Nitrogênio/metabolismo , Derivação Portocava Cirúrgica , Animais , Encéfalo/efeitos dos fármacos , Injeções Intravenosas , Rim/efeitos dos fármacos , Rim/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Masculino , Ratos , Ratos Wistar , Testículo/efeitos dos fármacos , Testículo/metabolismo , Distribuição Tecidual/efeitos dos fármacos , Distribuição Tecidual/fisiologia
3.
J Neurochem ; 138(1): 14-52, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27166428

RESUMO

Aerobic glycolysis occurs during brain activation and is characterized by preferential up-regulation of glucose utilization compared with oxygen consumption even though oxygen level and delivery are adequate. Aerobic glycolysis is a widespread phenomenon that underlies energetics of diverse brain activities, such as alerting, sensory processing, cognition, memory, and pathophysiological conditions, but specific cellular functions fulfilled by aerobic glycolysis are poorly understood. Evaluation of evidence derived from different disciplines reveals that aerobic glycolysis is a complex, regulated phenomenon that is prevented by propranolol, a non-specific ß-adrenoceptor antagonist. The metabolic pathways that contribute to excess utilization of glucose compared with oxygen include glycolysis, the pentose phosphate shunt pathway, the malate-aspartate shuttle, and astrocytic glycogen turnover. Increased lactate production by unidentified cells, and lactate dispersal from activated cells and lactate release from the brain, both facilitated by astrocytes, are major factors underlying aerobic glycolysis in subjects with low blood lactate levels. Astrocyte-neuron lactate shuttling with local oxidation is minor. Blockade of aerobic glycolysis by propranolol implicates adrenergic regulatory processes including adrenal release of epinephrine, signaling to brain via the vagus nerve, and increased norepinephrine release from the locus coeruleus. Norepinephrine has a powerful influence on astrocytic metabolism and glycogen turnover that can stimulate carbohydrate utilization more than oxygen consumption, whereas ß-receptor blockade 're-balances' the stoichiometry of oxygen-glucose or -carbohydrate metabolism by suppressing glucose and glycogen utilization more than oxygen consumption. This conceptual framework may be helpful for design of future studies to elucidate functional roles of preferential non-oxidative glucose utilization and glycogen turnover during brain activation. Aerobic glycolysis, the preferential up-regulation of glucose utilization (CMRglc ) compared with oxygen consumption (CMRO2 ) during brain activation, is blocked by propranolol. Epinephrine release from the adrenal gland stimulates vagus nerve signaling to the locus coeruleus, enhancing norepinephrine release in the brain, and regulation of astrocytic and neuronal metabolism to stimulate CMRglc more than CMRO2 . Propranolol suppresses CMRglc more than CMRO2 .


Assuntos
Astrócitos/metabolismo , Metabolismo Energético/fisiologia , Glicólise/fisiologia , Norepinefrina/metabolismo , Consumo de Oxigênio/fisiologia , Animais , Glicogênio/metabolismo , Humanos
4.
Neurochem Res ; 41(1-2): 16-32, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26224289

RESUMO

Phenylketonuria and hyperphenylalanemia are inborn errors in metabolism of phenylalanine arising from defects in steps to convert phenylalanine to tyrosine. Phe accumulation causes severe mental retardation that can be prevented by timely identification of affected individuals and their placement on a Phe-restricted diet. In spite of many studies in patients and animal models, the basis for acquisition of mental retardation during the critical period of brain development is not adequately understood. All animal models for human disease have advantages and limitations, and characteristics common to different models are most likely to correspond to the disorder. This study established similar levels of Phe exposure in developing rats between 3 and 16 days of age using three models to produce chronic hyperphenylalanemia, and identified changes in brain amino acid levels common to all models that persist for ~16 h of each day. In a representative model, local rates of glucose utilization (CMRglc) were determined at 25-27 days of age, and only selective changes that appeared to depend on Phe exposure were observed. CMRglc was reduced in frontal cortex and thalamus and increased in hippocampus and globus pallidus. Behavioral testing to evaluate neuromuscular competence revealed poor performance in chronically-hyperphenylalanemic rats that persisted for at least 3 weeks after cessation of Phe injections and did not occur with mild or acute hyperphenylalanemia. Thus, the abnormal amino acid environment, including hyperglycinemia, in developing rat brain is associated with selective regional changes in glucose utilization and behavioral abnormalities that are not readily reversed after they are acquired.


Assuntos
Comportamento Animal , Fenilcetonúrias/metabolismo , Animais , Encéfalo/metabolismo , Doença Crônica , Glucose/metabolismo , Fenilalanina/administração & dosagem , Fenilalanina/sangue , Fenilalanina/metabolismo , Fenilcetonúrias/fisiopatologia , Ratos , Ratos Endogâmicos F344
5.
Metab Brain Dis ; 30(1): 281-98, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24515302

RESUMO

Glycogen is the major store of glucose in brain and is mainly in astrocytes. Brain glycogen levels in unstimulated, carefully-handled rats are 10-12 µmol/g, and assuming that astrocytes account for half the brain mass, astrocytic glycogen content is twice as high. Glycogen turnover is slow under basal conditions, but it is mobilized during activation. There is no net increase in incorporation of label from glucose during activation, whereas label release from pre-labeled glycogen exceeds net glycogen consumption, which increases during stronger stimuli. Because glycogen level is restored by non-oxidative metabolism, astrocytes can influence the global ratio of oxygen to glucose utilization. Compensatory increases in utilization of blood glucose during inhibition of glycogen phosphorylase are large and approximate glycogenolysis rates during sensory stimulation. In contrast, glycogenolysis rates during hypoglycemia are low due to continued glucose delivery and oxidation of endogenous substrates; rates that preserve neuronal function in the absence of glucose are also low, probably due to metabolite oxidation. Modeling studies predict that glycogenolysis maintains a high level of glucose-6-phosphate in astrocytes to maintain feedback inhibition of hexokinase, thereby diverting glucose for use by neurons. The fate of glycogen carbon in vivo is not known, but lactate efflux from brain best accounts for the major metabolic characteristics during activation of living brain. Substantial shuttling coupled with oxidation of glycogen-derived lactate is inconsistent with available evidence. Glycogen has important roles in astrocytic energetics, including glucose sparing, control of extracellular K(+) level, oxidative stress management, and memory consolidation; it is a multi-functional compound.


Assuntos
Nível de Alerta/fisiologia , Astrócitos/metabolismo , Encéfalo/metabolismo , Metabolismo Energético , Glicogênio/metabolismo , Transmissão Sináptica/fisiologia , Animais , Artefatos , Carbono/metabolismo , Técnicas de Química Analítica , Glucose/metabolismo , Ácido Glutâmico/metabolismo , Glicogenólise , Humanos , Hipoglicemia/metabolismo , Modelos Neurológicos , Neurônios/metabolismo , Estresse Oxidativo , Oxigênio/metabolismo , Potássio/metabolismo , Ratos , Estresse Fisiológico/fisiologia
6.
Metab Brain Dis ; 29(4): 1041-52, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24154686

RESUMO

Portacaval shunting is a model for hepatic encephalopathy that causes chronic hyperammonemia, disruption of metabolic, signaling, and neurotransmitter systems, and progressive morphological changes. Exposure of cultured cells to ammonia raises intralysosomal pH and inhibits proteolysis, and the present study tested the hypothesis that proteolytic capacity is diminished in portacaval-shunted rats. Proteins were labeled in vivo with tracer doses of diisopropylfluorophosphate (DFP) and clearance of label was assayed. This approach labeled proteins independent of protein synthesis, which is reported to be altered in shunted rats, and avoided complications arising from re-utilization of labeled amino acids that causes underestimation of degradation rate. Characterization of DFP labeling showed that protein labeling was fast, about 50% of the label was released during a 24 h interval, labeling by DFP metabolites was negligible, inhibition of brain acetylcholinesterase was not detectable, and labeling by [(3)H]- and [(14)C]DFP was equivalent. To assay degradative capacity, proteins were first labeled with [(3)H]DFP, followed by labeling with [(14)C]DFP that was given 24 or 72 h later. The (3)H/(14)C ratio in each animal was used as a relative measure of removal of (3)H-labeled proteins. (3)H/(14)C ratios were generally significantly higher in portacaval-shunted rats than in controls, consistent with reduced proteolytic capacity. Assays of amino acid incorporation into brain protein generally replicated literature reports, supporting the conclusion that protein synthesis unlikely to be markedly inhibited and amino acid recycling influences calculated protein synthesis rates in shunted rats. Therapeutic strategies to reduce ammonia level would help normalize lysosomal functions and protein and lipid turnover.


Assuntos
Corantes Fluorescentes/análise , Encefalopatia Hepática/metabolismo , Isoflurofato/análise , Lisossomos/metabolismo , Derivação Portocava Cirúrgica/efeitos adversos , Proteínas/metabolismo , Proteólise , Aminoácidos/metabolismo , Animais , Encefalopatia Hepática/etiologia , Concentração de Íons de Hidrogênio , Hiperamonemia/etiologia , Hiperamonemia/metabolismo , Masculino , Ratos , Ratos Wistar
7.
J Neurochem ; 125(2): 247-59, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23346911

RESUMO

α-Syntrophin is a component of the dystrophin scaffold-protein complex that serves as an adaptor for recruitment of key proteins to the cytoplasmic side of plasma membranes. α-Syntrophin knockout (KO) causes loss of the polarized localization of aquaporin4 (AQP4) at astrocytic endfeet and interferes with water and K(+) homeostasis. During brain activation, release of ions and metabolites from endfeet is anticipated to increase perivascular fluid osmolarity, AQP4-mediated osmotic water flow from endfeet, and metabolite washout from brain. This study tests the hypothesis that reduced levels of endfoot AQP4 increase retention of [(14)C]metabolites during sensory stimulation. Conscious KO and wild-type mice were pulse-labeled with [6-(14)C] glucose during unilateral acoustic stimulation or bilateral acoustic plus whisker stimulation, and label retention was assayed by computer-assisted brain imaging or analysis of [(14)C]metabolites in extracts, respectively. High-resolution autoradiographic assays detected a 17% side-to-side difference (p < 0.05) in inferior colliculus of KO mice, not wild-type mice. However, there were no labeling differences between KO and wild-type mice for five major HPLC fractions from four dissected regions, presumably because of insufficient anatomical resolution. The results suggest a role for AQP4-mediated water flow in support of washout of metabolites, and underscore the need for greater understanding of astrocytic water and metabolite fluxes.


Assuntos
Aquaporina 4/metabolismo , Encéfalo/fisiologia , Proteínas de Ligação ao Cálcio/metabolismo , Glucose/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Musculares/metabolismo , Estimulação Acústica , Animais , Autorradiografia , Proteínas de Ligação ao Cálcio/deficiência , Radioisótopos de Carbono , Cromatografia Líquida de Alta Pressão , Masculino , Proteínas de Membrana/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Musculares/deficiência , Estimulação Física
8.
J Neurosci Res ; 89(12): 2052-67, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21567444

RESUMO

Experimental diabetes increases production of reactive oxygen-nitrogen species and inhibits astrocytic gap junctional communication in tissue culture and brain slices from streptozotocin (STZ)-diabetic rats by unidentified mechanisms. Relative connexin (Cx) protein levels were assessed by Western blotting using extracts from cultured astrocytes grown in high (25 mmol/liter) or low (5.5 mmol/liter) glucose for 2-3 weeks and STZ-diabetic rat brain. Chemiluminescent signals for diabetic samples were normalized to those of controls on the same blot and same protein load. Growth in high glucose did not alter relative Cx26 level, whereas Cx30 and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) were reduced by ∼30%, and Cx43 increased ∼1.9-fold. In the inferior colliculus of STZ-diabetic rats, Cx30 and Cx43 levels in three of four rats were half those of controls, whereas GAPDH and actin were unaffected. Diabetes did not affect levels of Cx30, Cx43, or GAPDH in cerebral cortex, but actin level rose 24%. Cx43 was predominantly phosphorylated in control and diabetic samples, so the reduced dye transfer is not due to overall dephosphorylation of Cx43. Astrocytic growth in high glucose reduced the dye-labeled area by 75%, but 10 min of treatment with dithiothreitol restored normal dye transfer. In contrast, nitric oxide donors inhibited dye transfer among astrocytes grown in low glucose by 50-65% within 1 hr. Thus, modifications arising from oxidative-nitrosative stress, not altered connexin levels, may underlie the reduced dye transfer among severely hyperglycemic cultured astrocytes, whereas both oxidative-nitrosative stress and regionally selective down-regulation of connexin protein content may affect gap junctional communication in the brains of STZ-diabetic rats.


Assuntos
Astrócitos/metabolismo , Encéfalo/metabolismo , Conexinas/metabolismo , Diabetes Mellitus Experimental/fisiopatologia , Junções Comunicantes/metabolismo , Animais , Astrócitos/patologia , Western Blotting , Encéfalo/patologia , Diabetes Mellitus Experimental/metabolismo , Junções Comunicantes/patologia , Estresse Oxidativo/fisiologia , Ratos , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo
9.
J Neurochem ; 109 Suppl 1: 30-7, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19393006

RESUMO

The magnitude of metabolic activation is greatly underestimated in autoradiographic studies using [1- or 6-14C]glucose compared to parallel assays with [14C]deoxyglucose indicating that most of the label corresponding to the additional [14C]glucose consumed during activation compared to rest is quickly released from activated structures. Label could be lost by net release of [14C]lactate from brain or via lactate exchange between blood and brain. These possibilities were distinguished by comparison of glucose and lactate specific activities in arterial blood and brain before, during, and after generalized sensory stimulation and during spreading cortical depression. Over a wide range of brain lactate concentrations, lactate specific activity was close to the theoretical maximum, i.e. half that of [6-14C]glucose, indicating that exchange-mediated dilution of lactate is negligible and that efflux of [14C]lactate probably accounts for most of the label loss. Low lactate dilution also indicates that dilution of glutamate C4 fractional enrichment in [13C]glucose studies, currently ascribed predominantly to lactate exchange, arises from other unidentified pathways or factors. Alternative explanations for glutamate dilution (presented in Supporting Information) include poorly labeled amino acid pools and oxidative metabolism of minor substrates in astrocytes to first dilute the astrocytic glutamine pool, followed by dilution of glutamate via glutamate-glutamine cycling.


Assuntos
Química Encefálica/fisiologia , Ácido Glutâmico/metabolismo , Glutamina/metabolismo , Ácido Láctico/metabolismo , Aminoácidos/metabolismo , Animais , Astrócitos/metabolismo , Autorradiografia , Glicemia/metabolismo , Depressão Alastrante da Atividade Elétrica Cortical/fisiologia , Ácido Glutâmico/sangue , Glutamina/sangue , Ácido Láctico/sangue , Oxirredução , Ratos , Sensação/fisiologia
10.
J Neurochem ; 111(2): 522-36, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19682206

RESUMO

Brain is a highly-oxidative organ, but during activation, glycolytic flux is preferentially up-regulated even though oxygen supply is adequate. The biochemical and cellular basis of metabolic changes during brain activation and the fate of lactate produced within brain are important, unresolved issues central to understanding brain function, brain images, and spectroscopic data. Because in vivo brain imaging studies reveal rapid efflux of labeled glucose metabolites during activation, lactate trafficking among astrocytes and between astrocytes and neurons was examined after devising specific, real-time, sensitive enzymatic fluorescent assays to measure lactate and glucose levels in single cells in adult rat brain slices. Astrocytes have a 2- to 4-fold faster and higher capacity for lactate uptake from extracellular fluid and for lactate dispersal via the astrocytic syncytium compared to neuronal lactate uptake from extracellular fluid or shuttling of lactate to neurons from neighboring astrocytes. Astrocytes can also supply glucose to neurons as well as glucose can be taken up by neurons from extracellular fluid. Astrocytic networks can provide neuronal fuel and quickly remove lactate from activated glycolytic domains, and the lactate can be dispersed widely throughout the syncytium to endfeet along the vasculature for release to blood or other brain regions via perivascular fluid flow.


Assuntos
Astrócitos/metabolismo , Glucose/metabolismo , Colículos Inferiores/metabolismo , Ácido Láctico/metabolismo , Neurônios/metabolismo , Animais , Astrócitos/citologia , Transporte Biológico/fisiologia , Comunicação Celular/fisiologia , Espaço Extracelular/metabolismo , Fluorescência , Junções Comunicantes/metabolismo , Colículos Inferiores/citologia , Masculino , Neurônios/citologia , Técnicas de Cultura de Órgãos , Ratos , Ratos Wistar
11.
J Neurochem ; 110(3): 857-69, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19457076

RESUMO

To assess the specificity of metabolite trafficking among gap junction-coupled astrocytes, we developed novel, real-time, single-cell enzymatic fluorescence assays to assay cell-to-cell transfer of unlabeled glycolytic intermediates and report (i) highly restricted transfer of glucose-6-phosphate (P) and two analogs, deoxyglucose (DG)-6-P, and 2-[N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino]-2-DG-6-P, compared with DG and 2- and 6-[N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino]-2-DG, (ii) extensive junctional diffusion of glyceraldehyde-3-P, NADH, and NADPH plus three anionic fluorescent dyes used as internal standards for transfer assays, and (iii) stimulation of gap junctional communication by increased intracellular Na(+) that also evokes metabolic responses in nearby coupled astrocytes. Thus, dye transfer does not predict gap junctional permeability of endogenous metabolites. Intracellular retention of flux-regulating compounds (e.g. glucose-6-P) may be necessary for local metabolic control, whereas 'syncytial sharing' may dissipate the work load on peri-synaptic astrocytes. Imaging of brain functional activity depends on local accumulation of exogenous or endogenous signals, and DG-6-P is trapped in the cell where it is phosphorylated, whereas rapid dispersal of cytoplasmic NAD(P)H and labeled glucose metabolites throughout the astrocytic syncytium can interfere with cellular assessment of neuron-astrocyte relationships in autoradiographic, fluorescence microscopic, and magnetic resonance spectroscopic studies.


Assuntos
Astrócitos/citologia , Astrócitos/metabolismo , Encéfalo/citologia , Encéfalo/metabolismo , Junções Comunicantes/metabolismo , Glicólise/fisiologia , Animais , Autorradiografia , Células Cultivadas , Espectroscopia de Ressonância Magnética , Masculino , Microscopia de Fluorescência , Transporte Proteico/fisiologia , Ratos , Ratos Wistar
12.
J Neurochem ; 103(4): 1506-22, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17725580

RESUMO

Astrocytes have important roles in control of extracellular environment, de novo synthesis of neurotransmitters, and regulation of neurotransmission and blood flow. All of these functions require energy, suggesting that astrocytic metabolism should rise and fall with changes in neuronal activity and that brain imaging can be used to visualize and quantify astrocytic activation in vivo. A unilateral photic stimulation paradigm was used to test the hypothesis that graded sensory stimuli cause progressive increases in the uptake coefficient of [2-(14)C]acetate, a substrate preferentially oxidized by astrocytes. The acetate uptake coefficient fell in deafferented visual structures and it rose in intact tissue during photic stimulation of conscious rats; the increase was highest in structures with monosynaptic input from the eye and was much smaller in magnitude than the change in glucose utilization (CMR(glc)) by all cells. The acetate uptake coefficient was not proportional to stimulus rate and did not correlate with CMR(glc) in resting or activated structures. Simulation studies support the conclusions that acetate uptake coefficients represent mainly metabolism and respond to changes in metabolism rate, with a lower response at high rates. A model portraying regulation of acetate oxidation illustrates complex relationships among functional activation, cation levels, and astrocytic metabolism.


Assuntos
Astrócitos/metabolismo , Estimulação Luminosa/métodos , Acetatos/metabolismo , Animais , Astrócitos/fisiologia , Metabolismo Energético/fisiologia , Glucose/metabolismo , Glucose/fisiologia , Masculino , Ratos , Ratos Sprague-Dawley
13.
Neurochem Int ; 48(6-7): 586-95, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16513214

RESUMO

Glucose delivered to brain by the cerebral circulation is the major and obligatory fuel for all brain cells, and assays of functional activity in working brain routinely focus on glucose utilization. However, these assays do not take into account the contributions of minor substrates or endogenous fuel consumed by astrocytes during brain activation, and emerging evidence suggests that glycogen, acetate, and, perhaps, glutamate, are metabolized by working astrocytes in vivo to provide physiologically significant amounts of energy in addition to that derived from glucose. Rates of glycogenolysis during sensory stimulation of normal, conscious rats are high enough to support the notion that glycogen can contribute substantially to astrocytic glucose utilization during activation. Oxidative metabolism of glucose provides most of the ATP for cultured astrocytes, and a substantial contribution of respiration to astrocyte energetics is supported by recent in vivo studies. Astrocytes preferentially oxidize acetate taken up into brain from blood, and calculated local rates of acetate utilization in vivo are within the range of calculated rates of glucose oxidation in astrocytes. Glutamate may also serve as an energy source for activated astrocytes in vivo because astrocytes in tissue culture and in adult brain tissue readily oxidize glutamate. Taken together, contributions of minor metabolites derived from endogenous and exogenous sources add substantially to the energy obtained by astrocytes from blood-borne glucose. Because energy-generating reactions from minor substrates are not taken into account by routine assays of functional metabolism, they reflect a "hidden cost" of astrocyte work in vivo.


Assuntos
Ácido Acético/metabolismo , Astrócitos/fisiologia , Encéfalo/metabolismo , Glucose/metabolismo , Ácido Glutâmico/metabolismo , Glicogênio/metabolismo , Trifosfato de Adenosina/biossíntese , Animais , Astrócitos/metabolismo , Encéfalo/citologia , Metabolismo Energético , Ácido Glutâmico/biossíntese , Glicogenólise , Oxirredução
14.
J Cereb Blood Flow Metab ; 22(12): 1476-89, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12468892

RESUMO

The concentration of glycogen, the major brain energy reserve localized mainly in astrocytes, is generally reported as about 2 or 3 micromol/g, but sometimes as high as 3.9 to 8 micromol/g, in normal rat brain. The authors found high but very different glycogen levels in two recent studies in which glycogen was determined by the routine amyloglucosidase procedure in 0.03N HCl digests either of frozen powders (4.8 to 6 micromol/g) or of ethanol-insoluble fractions (8 to 12 micromol/g). To evaluate the basis for these discrepant results, glycogen was assayed in parallel extracts of the same samples. Glycogen levels in ethanol extracts were twice those in 0.03N HCl digests, suggesting incomplete enzyme inactivation even with very careful thawing. The very high glycogen levels were biologically active and responsive to physiologic and pharmacological challenge. Glycogen levels fell after brief sensory stimulation, and metabolic labeling indicated its turnover under resting conditions. About 95% of the glycogen was degraded under in vitro ischemic conditions, and its "carbon equivalents" recovered mainly as glc, glc-P, and lactate. Resting glycogen stores were reduced by about 50% by chronic inhibition of nitric oxide synthase. Because neurotransmitters are known to stimulate glycogenolysis, stress or sensory activation due to animal handling and tissue-sampling procedures may stimulate glycogenolysis during an experiment, and glycogen lability during tissue sampling and extraction can further reduce glycogen levels. The very high glycogen levels in normal rat brain suggest an unrecognized role for astrocytic energy metabolism during brain activation.


Assuntos
Astrócitos/metabolismo , Encéfalo/metabolismo , Metabolismo Energético/fisiologia , Meio Ambiente , Glicogênio/metabolismo , Animais , Encéfalo/citologia , Inibidores Enzimáticos/farmacologia , Glucose/metabolismo , Ácido Láctico/metabolismo , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Pós , Ratos , Descanso/fisiologia
15.
J Cereb Blood Flow Metab ; 22(12): 1490-502, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12468893

RESUMO

Interpretation of functional metabolic brain images requires understanding of metabolic shifts in working brain. Because the disproportionately higher uptake of glucose compared with oxygen ("aerobic glycolysis") during sensory stimulation is not fully explained by changes in levels of lactate or glycogen, metabolic labeling by [6-14C]glucose was used to evaluate utilization of glucose during brief brain activation. Increased labeling of tricarboxylic acid cycle-derived amino acids, mainly glutamate but also gamma-aminobutyric acid, reflects a rise in oxidative metabolism during aerobic glycolysis. The size of the glutamate, lactate, alanine, and aspartate pools changed during stimulation. Brain lactate was derived from blood-borne glucose and its specific activity was twice that of alanine, revealing pyruvate compartmentation. Glycogen labeling doubled during recovery compared with rest and activation; only 4% to 8% of the total 14C was recovered in lactate plus glycogen. Restoration of glycogen levels was slow, and diversion of glucose from oxidative pathways to restore its level could cause a prolonged reduction of the global O2/glucose uptake ratio. The rise in the brain glucose-oxygen uptake ratio during activation does not simply reflect an upward shift of glycolysis under aerobic conditions; instead, it involves altered fluxes into various (oxidative and biosynthetic) pathways with different time courses.


Assuntos
Encéfalo/metabolismo , Metabolismo Energético/fisiologia , Glucose/farmacocinética , Glicólise/fisiologia , Oxigênio/farmacocinética , Aerobiose , Aminoácidos/metabolismo , Animais , Astrócitos/metabolismo , Encéfalo/citologia , Radioisótopos de Carbono , Estado de Consciência , Glicogênio/metabolismo , Lactose/metabolismo , Masculino , Neurônios Aferentes/fisiologia , Oxirredução , Consumo de Oxigênio/fisiologia , Ratos , Ratos Sprague-Dawley , Tato
16.
Neurochem Int ; 43(4-5): 339-54, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12742078

RESUMO

Metabolic responses of brain cells to a stimulus are governed, in part, by their enzymatic specialization and interrelationships with neighboring cells, and local shifts in functional metabolism during brain activation are likely to be influenced by the neurotransmitter system, subcellular compartmentation, and anatomical structure. Selected examples of functional activation illustrate the complexity of metabolic interactions in working brain and of interpretation of changes in brain lactate levels. The major focus of this article is the disproportionately higher metabolism of glucose compared to oxygen in normoxic brain, a phenomenon that occurs during activation in humans and animals. The glucose utilized in excess of oxygen is not fully explained by accumulation of glucose, lactate, or glycogen in brain or by lactate efflux from brain to blood. Thus, any lactate derived from the excess glucose could not have been stoichiometrically exported to and metabolized by neighboring neurons because oxygen consumption would have otherwise increased and matched that of glucose. Metabolic labeling of tricarboxylic acid cycle-derived amino acids increased during brief sensory stimulation, reflecting a rise in oxidative metabolism. Brain glycogen is mainly in astrocytes, and its level falls throughout the stimulus and early post-activation interval. Glycogenolysis cannot be accounted for by lactate accumulation or oxidation; there must be rapid product clearance. Glycogen restoration is slow and diversion of glucose from oxidative pathways for its re-synthesis could reduce the global O(2)/glucose uptake ratio; astrocytes could downshift this ratio for up to an hour after 5 min stimulus. Morphological studies of astrocytes reveal a paucity of cytoplasm and organelles in the fine processes that surround synapses and form gap junction connections with neighboring astrocytes. Specialized regions of astrocytes, e.g. their endfeet and thin peripheral lamellae, are likely to have compartmentalized metabolic activities. Anatomical constraints imposed upon the fine processes might require preferential utilization of glycolysis to satisfy their energy demands, but rapid lactate clearance would then be essential, since its accumulation would inhibit glycolysis. Gap junctional connections between neighboring astrocytes provide a mechanism for rapid metabolite spreading via the astrocytic syncytium and elimination of by-products. Local structure-function relationships need to be incorporated into experimental models of neuron-astrocyte and astrocyte-astrocyte interactions in working brain.


Assuntos
Astrócitos/metabolismo , Barreira Hematoencefálica , Encéfalo/citologia , Encéfalo/metabolismo , Glicogênio/metabolismo , Lactatos/metabolismo , Consumo de Oxigênio
17.
Neurochem Int ; 45(2-3): 321-51, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15145548

RESUMO

Functional activation of astrocytic metabolism is believed, according to one hypothesis, to be closely linked to excitatory neurotransmission and to provide lactate as fuel for oxidative metabolism in neighboring neurons. However, review of emerging evidence suggests that the energetic demands of activated astrocytes are higher and more complex than recognized and much of the lactate presumably produced by astrocytes is not locally oxidized during activation. In vivo activation studies in normal subjects reveal that the rise in consumption of blood-borne glucose usually exceeds that of oxygen, especially in retina compared to brain. When the contribution of glycogen, the brain's major energy reserve located in astrocytes, is taken into account the magnitude of the carbohydrate-oxygen utilization mismatch increases further because the magnitude of glycogenolysis greatly exceeds the incremental increase in utilization of blood-borne glucose. Failure of local oxygen consumption to equal that of glucose plus glycogen in vivo is strong evidence against stoichiometric transfer of lactate from astrocytes to neighboring neurons for oxidation. Thus, astrocytes, not nearby neurons, use the glycogen for energy during physiological activation in normal brain. These findings plus apparent compartmentation of metabolism of glycogen and blood-borne glucose during activation lead to our working hypothesis that activated astrocytes have high energy demands in their fine perisynaptic processes (filopodia) that might be met by glycogenolysis and glycolysis coupled to rapid lactate clearance. Tissue culture studies do not consistently support the lactate shuttle hypothesis because key elements of the model, glutamate-induced increases in glucose utilization and lactate release, are not observed in many astrocyte preparations, suggesting differences in their oxidative capacities that have not been included in the model. In vivo nutritional interactions between working neurons and astrocytes are not as simple as implied by "sweet (glucose-glycogen) and sour (lactate) food for thought."


Assuntos
Encéfalo/fisiologia , Comunicação Celular/fisiologia , Paladar , Anestesia , Animais , Astrócitos/citologia , Astrócitos/fisiologia , Circulação Cerebrovascular/fisiologia , Humanos , Modelos Neurológicos , Fenômenos Fisiológicos da Nutrição , Transmissão Sináptica/fisiologia
18.
Neurochem Int ; 42(5): 359-74, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12510019

RESUMO

Astrocytic proliferation is a hallmark of brain injury, but the biological functions and metabolic activities of reactive astrocytes in vivo are poorly understood. [2-14C]Acetate, which is preferentially transported into and, therefore, metabolized by astrocytes, was used to assess injury- and trophic factor-induced changes in astrocyte metabolic activity. Local rates of net [2-14C]acetate uptake and glucose utilization (CMR(glc)), determined with [14C]deoxyglucose to assay overall metabolic activity of all brain cells, were assayed 7 days after a cannula placement; adjacent brain sections were immunostained to identify glial fibrillary acidic protein-positive (GFAP(+)) astrocytes and microglia plus macrophages (lectin-positive cells). GFAP(+) cells were abundant in tissue surrounding the cannula compared to the contralateral hemisphere, whereas lectin(+) cells were restricted to the wound boundary. CMR(glc) fell 25% in regions enriched in reactive astrocytes compared to the homologous contralateral hemisphere, whereas [14C]acetate uptake increased slightly (6%) but statistically significantly; metabolism of both tracers in 13 other brain structures was unchanged. Injection of basic fibroblast growth factor (b-FGF) into cerebral cortex or superior colliculus produced fiber-rich cell clusters containing both GFAP(+) and lectin(+) cells that had a 37% increase in [14C]acetate uptake; GFAP(+)-cell density rose in the nearby neuropil but the corresponding change in [14C]acetate uptake was small (6-8%). Sensory stimulation did not alter [14C]acetate uptake into the clusters. Thus, [14C]acetate uptake was relatively stable with respect to changes in the density of reactive astrocytes that are dispersed throughout the neuropil and to changes in cellular activity arising from sensory stimulation. In contrast, b-FGF-induced cell clusters that contain mixed cell types and numerous fibers accumulated higher levels of [14C]acetate, raising the possibility that increased uptake might be due to high numbers of activated astrocytes and, perhaps, acetate metabolism by other cell types.


Assuntos
Acetatos/metabolismo , Encéfalo/citologia , Encéfalo/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Animais , Antimetabólitos/metabolismo , Astrócitos/metabolismo , Encéfalo/efeitos dos fármacos , Contagem de Células , Corantes , Desoxiglucose/metabolismo , Endotélio/citologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Corpos Estranhos/metabolismo , Corpos Estranhos/patologia , Imuno-Histoquímica , Lectinas/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Microglia/efeitos dos fármacos , Microglia/metabolismo , Microinjeções , Ratos , Ratos Wistar , Colículos Superiores/fisiologia
19.
Brain Res ; 961(2): 201-12, 2003 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-12531487

RESUMO

Functional neurological outcome after transient ischemia might be improved by timely therapeutic intervention. To determine if restorative behavioral therapy influences damage, improves task learning, or alters astrocyte metabolic activity after ischemia, rats (food-restricted to 85% of free-feeding weight) were (a) first trained to respond on one of two levers under a fixed-ratio 20 schedule of food presentation (FR20), then (b) subjected to sham manipulation of carotid arteries or 10 min ischemia by four-vessel occlusion, followed by (c) 4 days of operant testing or inactivity, (d) then all rats were tested under a FR20 lever reversal task for 4 weeks, and (e) 3 days after the last behavioral session astrocyte metabolism was assayed by local uptake of [2-14C]acetate. Mild loss of hippocampal neurons occurred in ischemic rats with or without training after ischemia. Glial fibrillary acidic protein-positive astrocytes were present in similar numbers throughout brains of sham control and ischemic rats. Mild ischemia did not impair learning, and no changes in FR20 reversal learning were detected in sham vs. ischemic rats. Net [14C]acetate uptake was unaffected by ischemia but [14C]acetate uptake increased 15-24% (P<0.05; n=12-15/group) in specific structures (caudate, primary motor and sensorimotor cortex, CA1 hippocampus, subcortical white matter) in the pooled groups of rats that had 4 days FR20 testing vs. inactivity before reversal learning. 'Behavioral therapy' (operant testing on the 4 days immediately following either sham manipulation or ischemia) did not alter ischemic outcome, but was associated with higher acetate utilization in regions involved in motor activities.


Assuntos
Ácido Acético/metabolismo , Astrócitos/metabolismo , Hipocampo/patologia , Ataque Isquêmico Transitório/reabilitação , Atividade Motora , Neurônios/patologia , Animais , Radioisótopos de Carbono , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/terapia , Condicionamento Operante , Proteína Glial Fibrilar Ácida/metabolismo , Ataque Isquêmico Transitório/complicações , Ataque Isquêmico Transitório/metabolismo , Aprendizagem , Masculino , Ratos , Ratos Wistar
20.
Neurochem Int ; 62(5): 784-95, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23411409

RESUMO

Oxidative-nitrosative stress and inflammatory responses are associated with endoplasmic reticulum (ER) stress in diabetic retinopathy, raising the possibility that disturbances in ER protein processing may contribute to CNS dysfunction in diabetics. Upregulation of the unfolded protein response (UPR) is a homeostatic response to accumulation of abnormal proteins in the ER, and the present study tested the hypothesis that the UPR is upregulated in two models for diabetes, cultured astrocytes grown in 25mmol/L glucose for up to 4weeks and brain of streptozotocin (STZ)-treated rats with diabetes for 1-7months. Markers associated with translational blockade (phospho-eIF2α and apoptosis (CHOP), inflammatory response (inducible nitric oxide synthase, iNOS), and nitrosative stress (nuclear translocation of glyceraldehyde-3-phosphate dehydrogenase, GAPDH) were not detected in either model. Nrf2 was present in nuclei of low- and high-glucose cultures, consistent with oxidative stress. Astrocytic ATF4 expression was not altered by culture glucose concentration, whereas phospho-IRE and ATF6 levels were higher in low- compared with high-glucose cultures. The glucose-regulated chaperones, GRP78 and GRP94, were also expressed at higher levels in low- than high-glucose cultures, probably due to recurrent glucose depletion between feeding cycles. In STZ-rat cerebral cortex, ATF4 level was transiently reduced at 4months, and p-IRE levels were transiently elevated at 3months. However, GRP78 and GRP94 expression was not upregulated, and iNOS, amyloid-ß, and nuclear accumulation of GAPDH were not evident in STZ-diabetic brain. High-glucose cultured astrocytes and STZ-diabetic brain are relatively resistant to diabetes-induced ER stress, in sharp contrast with cultured retinal Müller cells and diabetic rodent retina.


Assuntos
Astrócitos/metabolismo , Encéfalo/metabolismo , Diabetes Mellitus Experimental/metabolismo , Estresse Oxidativo , Resposta a Proteínas não Dobradas , Animais , Células Cultivadas , Masculino , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Estreptozocina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA