Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
Article in English | MEDLINE | ID: mdl-38764181

ABSTRACT

OBJECTIVE: Obesity is associated with alterations in eating behavior and neurocognitive function. In this study, we investigate the effect of obesity on brain energy utilization, including brain glucose transport and metabolism. METHODS: A total of 11 lean participants and 7 young healthy participants with obesity (mean age, 27 years) underwent magnetic resonance spectroscopy scanning coupled with a hyperglycemic clamp (target, ~180 mg/dL) using [1-13C] glucose to measure brain glucose uptake and metabolism, as well as peripheral markers of insulin resistance. RESULTS: Individuals with obesity demonstrated an ~20% lower ratio of brain glucose uptake to cerebral glucose metabolic rate (Tmax/CMRglucose) than lean participants (2.12 ± 0.51 vs. 2.67 ± 0.51; p = 0.04). The cerebral tricarboxylic acid cycle flux (VTCA) was similar between the two groups (p = 0.64). There was a negative correlation between total nonesterified fatty acids and Tmax/CMRglucose (r = -0.477; p = 0.045). CONCLUSIONS: We conclude that CMRglucose is unlikely to differ between groups due to similar VTCA, and, therefore, the glucose transport Tmax is lower in individuals with obesity. These human findings suggest that obesity is associated with reduced cerebral glucose transport capacity even at a young age and in the absence of other cardiometabolic comorbidities, which may have implications for long-term brain function and health.

2.
Front Cell Neurosci ; 17: 1130816, 2023.
Article in English | MEDLINE | ID: mdl-37187610

ABSTRACT

Introduction: There is a lack of robust metabolic imaging techniques that can be routinely applied to characterize lesions in patients with brain tumors. Here we explore in an animal model of glioblastoma the feasibility to detect uptake and metabolism of deuterated choline and describe the tumor-to-brain image contrast. Methods: RG2 cells were incubated with choline and the level of intracellular choline and its metabolites measured in cell extracts using high resolution 1H NMR. In rats with orthotopically implanted RG2 tumors deuterium metabolic imaging (DMI) was applied in vivo during, as well as 1 day after, intravenous infusion of 2H9-choline. In parallel experiments, RG2-bearing rats were infused with [1,1',2,2'-2H4]-choline and tissue metabolite extracts analyzed with high resolution 2H NMR to identify molecule-specific 2H-labeling in choline and its metabolites. Results: In vitro experiments indicated high uptake and fast phosphorylation of exogenous choline in RG2 cells. In vivo DMI studies revealed a high signal from the 2H-labeled pool of choline + metabolites (total choline, 2H-tCho) in the tumor lesion but not in normal brain. Quantitative DMI-based metabolic maps of 2H-tCho showed high tumor-to-brain image contrast in maps acquired both during, and 24 h after deuterated choline infusion. High resolution 2H NMR revealed that DMI data acquired during 2H-choline infusion consists of free choline and phosphocholine, while the data acquired 24 h later represent phosphocholine and glycerophosphocholine. Discussion: Uptake and metabolism of exogenous choline was high in RG2 tumors compared to normal brain, resulting in high tumor-to-brain image contrast on DMI-based metabolic maps. By varying the timing of DMI data acquisition relative to the start of the deuterated choline infusion, the metabolic maps can be weighted toward detection of choline uptake or choline metabolism. These proof-of-principle experiments highlight the potential of using deuterated choline combined with DMI to metabolically characterize brain tumors.

3.
J Neurochem ; 2023 May 07.
Article in English | MEDLINE | ID: mdl-37150946

ABSTRACT

During transient brain activation cerebral blood flow (CBF) increases substantially more than cerebral metabolic rate of oxygen consumption (CMRO2 ) resulting in blood hyperoxygenation, the basis of BOLD fMRI contrast. Explanations for the high CBF vs. CMRO2 slope, termed neurovascular coupling (NVC) constant, focused on maintainenance of tissue oxygenation to support mitochondrial ATP production. However, paradoxically the brain has a 3-fold lower oxygen extraction fraction (OEF) than other organs with high energy requirements, like heart and muscle during exercise. Here, we hypothesize that the NVC constant and the capillary oxygen mass transfer coefficient (which in combination determine OEF) are co-regulated during activation to maintain simultaneous homeostasis of pH and partial pressure of CO2 and O2 (pCO2 and pO2 ). To test our hypothesis, we developed an arteriovenous flux balance model for calculating blood and brain pH, pCO2 , and pO2 as a function of baseline OEF (OEF0 ), CBF, CMRO2 , and proton production by nonoxidative metabolism coupled to ATP hydrolysis. Our model was validated against published brain arteriovenous difference studies and then used to calculate pH, pCO2, and pO2 in activated human cortex from published calibrated fMRI and PET measurements. In agreement with our hypothesis, calculated pH, pCO2, and pO2 remained close to constant independently of CMRO2 in correspondence to experimental measurements of NVC and OEF0 . We also found that the optimum values of the NVC constant and OEF0 that ensure simultaneous homeostasis of pH, pCO2, and pO2 were remarkably similar to their experimental values. Thus, the high NVC constant is overall determined by proton removal by CBF due to increases in nonoxidative glycolysis and glycogenolysis. These findings resolve the paradox of the brain's high CBF yet low OEF during activation, and may contribute to explaining the vulnerability of brain function to reductions in blood flow and capillary density with aging and neurovascular disease.

4.
NMR Biomed ; : e4957, 2023 Apr 23.
Article in English | MEDLINE | ID: mdl-37088548

ABSTRACT

The olfactory bulb (OB) plays a fundamental role in the sense of smell and has been implicated in several pathologies, including Alzheimer's disease. Despite its importance, high metabolic activity and unique laminar architecture, the OB is not frequently studied using MRS methods, likely due to the small size and challenging location. Here we present a detailed metabolic characterization of OB metabolism, in terms of both static metabolite concentrations using 1 H MRS and metabolic fluxes associated with neuro-energetics and neurotransmission by tracing the dynamic 13 C flow from intravenously administered [1,6-13 C2 ]-glucose, [2-13 C]-glucose and [2-13 C]-acetate to downstream metabolites, including [4-13 C]-glutamate, [4-13 C]-glutamine and [2-13 C]-GABA. The unique laminar architecture and associated metabolism of the OB, distinctly different from that of the cerebral cortex, is characterized by elevated GABA and glutamine levels, as well as increased GABAergic and astroglial energy metabolism and neurotransmission. The results show that, despite the technical challenges, high-quality 1 H and 1 H-[13 C] MR spectra can be obtained from the rat OB in vivo. The derived metabolite concentrations and metabolic rates demonstrate a unique metabolic profile for the OB. The metabolic model provides a solid basis for future OB studies on functional activation or pathological conditions.

5.
NMR Biomed ; 36(4): e4879, 2023 04.
Article in English | MEDLINE | ID: mdl-36424353

ABSTRACT

This paper provides a brief description of the early use of ex vivo nuclear magnetic resonance (NMR) studies of tissue and tissue extracts performed in the laboratory of Dr. Robert G. Shulman from 1975 through 1995 at Bell Laboratories, then later at Yale University. During that period, ex vivo NMR provided critical information in support of resonance assignments and the quantitation of concentrations for magnetic resonance spectroscopy studies. The period covered saw rapid advances in magnet technology, starting with studies of microorganisms in vertical bore high-resolution NMR studies, then by 1981 studies of small mammals in a horizontal bore magnet, and then studies of humans in 1984. Ex vivo NMR played a critical role in all these studies. A general strategy developed in the lab for using ex vivo NMR to support in vivo studies is presented, as well as illustrative examples.


Subject(s)
Laboratories , Magnetic Resonance Imaging , Animals , Humans , Magnetic Resonance Spectroscopy/methods , Mammals
6.
J Neurochem ; 2022 Sep 11.
Article in English | MEDLINE | ID: mdl-36089566

ABSTRACT

The ~1:1 stoichiometry between the rates of neuronal glucose oxidation (CMRglc-ox-N ) and glutamate (Glu)/γ-aminobutyric acid (GABA)-glutamine (Gln) neurotransmitter (NT) cycling between neurons and astrocytes (VNTcycle ) has been firmly established. However, the mechanistic basis for this relationship is not fully understood, and this knowledge is critical for the interpretation of metabolic and brain imaging studies in normal and diseased brain. The pseudo-malate-aspartate shuttle (pseudo-MAS) model established the requirement for glycolytic metabolism in cultured glutamatergic neurons to produce NADH that is shuttled into mitochondria to support conversion of extracellular Gln (i.e., astrocyte-derived Gln in vivo) into vesicular neurotransmitter Glu. The evaluation of this model revealed that it could explain half of the 1:1 stoichiometry and it has limitations. Modifications of the pseudo-MAS model were, therefore, devised to address major knowledge gaps, that is, submitochondrial glutaminase location, identities of mitochondrial carriers for Gln and other model components, alternative mechanisms to transaminate α-ketoglutarate to form Glu and shuttle glutamine-derived ammonia while maintaining mass balance. All modified models had a similar 0.5 to 1.0 predicted mechanistic stoichiometry between VNTcycle and the rate of glucose oxidation. Based on studies of brain ß-hydroxybutyrate oxidation, about half of CMRglc-ox-N may be linked to glutamatergic neurotransmission and localized in pre-synaptic structures that use pseudo-MAS type mechanisms for Glu-Gln cycling. In contrast, neuronal compartments that do not participate in transmitter cycling may use the MAS to sustain glucose oxidation. The evaluation of subcellular compartmentation of neuronal glucose metabolism in vivo is a critically important topic for future studies to understand glutamatergic and GABAergic neurotransmission.

7.
J Cereb Blood Flow Metab ; 42(5): 844-860, 2022 05.
Article in English | MEDLINE | ID: mdl-34994222

ABSTRACT

Over the last two decades, it has been established that glucose metabolic fluxes in neurons and astrocytes are proportional to the rates of the glutamate/GABA-glutamine neurotransmitter cycles in close to 1:1 stoichiometries across a wide range of functional energy demands. However, there is presently no mechanistic explanation for these relationships. We present here a theoretical meta-analysis that tests whether the brain's unique compartmentation of glycogen metabolism in the astrocyte and the requirement for neuronal glucose homeostasis lead to the observed stoichiometries. We found that blood-brain barrier glucose transport can be limiting during activation and that the energy demand could only be met if glycogenolysis supports neuronal glucose metabolism by replacing the glucose consumed by astrocytes, a mechanism we call Glucose Sparing by Glycogenolysis (GSG). The predictions of the GSG model are in excellent agreement with a wide range of experimental results from rats, mice, tree shrews, and humans, which were previously unexplained. Glycogenolysis and glucose sparing dictate the energy available to support neuronal activity, thus playing a fundamental role in brain function in health and disease.


Subject(s)
Glycogenolysis , Animals , Astrocytes/metabolism , Brain/metabolism , Energy Metabolism/physiology , Glucose/metabolism , Glutamic Acid/metabolism , Glycogenolysis/physiology , Mice , Rats , Synaptic Transmission/physiology
8.
J Cereb Blood Flow Metab ; 42(6): 911-934, 2022 06.
Article in English | MEDLINE | ID: mdl-35078383

ABSTRACT

While functional MRI (fMRI) localizes brain activation and deactivation, functional MRS (fMRS) provides insights into the underlying metabolic conditions. There is much interest in measuring task-induced and resting levels of metabolites implicated in neuroenergetics (e.g., lactate, glucose, or ß-hydroxybutyrate (BHB)) and neurotransmission (e.g., γ-aminobutyric acid (GABA) or pooled glutamate and glutamine (Glx)). Ultra-high magnetic field (e.g., 7T) has boosted the fMRS quantification precision, reliability, and stability of spectroscopic observations using short echo-time (TE) 1H-MRS techniques. While short TE 1H-MRS lacks sensitivity and specificity for fMRS at lower magnetic fields (e.g., 3T or 4T), most of these metabolites can also be detected by J-difference editing (JDE) 1H-MRS with longer TE to filter overlapping resonances. The 1H-MRS studies show that JDE can detect GABA, Glx, lactate, and BHB at 3T, 4T and 7T. Most recently, it has also been demonstrated that JDE 1H-MRS is capable of reliable detection of metabolic changes in different brain areas at various magnetic fields. Combining fMRS measurements with fMRI is important for understanding normal brain function, but also clinically relevant for mechanisms and/or biomarkers of neurological and neuropsychiatric disorders. We provide an up-to-date overview of fMRS research in the last three decades, both in terms of applications and technological advances. Overall the emerging fMRS techniques can be expected to contribute substantially to our understanding of metabolism for brain function and dysfunction.


Subject(s)
Brain , Magnetic Resonance Imaging , 3-Hydroxybutyric Acid/metabolism , Brain/diagnostic imaging , Brain/metabolism , Glutamic Acid/metabolism , Glutamine/metabolism , Humans , Lactic Acid/metabolism , Magnetic Resonance Imaging/methods , Reproducibility of Results , Synaptic Transmission , gamma-Aminobutyric Acid/metabolism
9.
J Cereb Blood Flow Metab ; 42(8): 1507-1523, 2022 08.
Article in English | MEDLINE | ID: mdl-35048735

ABSTRACT

Anaplerosis occurs predominately in astroglia through the action of pyruvate carboxylase (PC). The rate of PC (Vpc) has been reported for cerebral cortex (or whole brain) of awake humans and anesthetized rodents, but regional brain rates remain largely unknown and, hence, were subjected to investigation in the current study. Awake male rats were infused with either [2-13C]glucose or [1-13C]glucose (n = 27/30) for 8, 15, 30, 60 or 120 min, followed by rapid euthanasia with focused-beam microwave irradiation to the brain. Blood plasma and extracts of cerebellum, hippocampus, striatum, and cerebral cortex were analyzed by 1H-[13C]-NMR to establish 13C-enrichment time courses for glutamate-C4,C3,C2, glutamine-C4,C3, GABA-C2,C3,C4 and aspartate-C2,C3. Metabolic rates were determined by fitting a three-compartment metabolic model (glutamatergic and GABAergic neurons and astroglia) to the eighteen time courses. Vpc varied by 44% across brain regions, being lowest in the cerebellum (0.087 ± 0.004 µmol/g/min) and highest in striatum (0.125 ± 0.009) with intermediate values in cerebral cortex (0.106 ± 0.005) and hippocampus (0.114 ± 0.005). Vpc constituted 13-19% of the oxidative glucose consumption rate. Combination of cerebral cortical data with literature values revealed a positive correlation between Vpc and the rates of glutamate/glutamine-cycling and oxidative glucose consumption, respectively, consistent with earlier observations.


Subject(s)
Glutamic Acid , Pyruvate Carboxylase , Animals , Brain/metabolism , Carbon Isotopes/metabolism , Glucose/metabolism , Glutamic Acid/metabolism , Glutamine/metabolism , Male , Neurons/metabolism , Neurotransmitter Agents/metabolism , Pyruvate Carboxylase/metabolism , Rats , Wakefulness , gamma-Aminobutyric Acid/metabolism
10.
Magn Reson Med ; 86(1): 62-68, 2021 07.
Article in English | MEDLINE | ID: mdl-33590529

ABSTRACT

PURPOSE: Deuterium metabolic imaging (DMI) combined with [6,6'-2 H2 ]-glucose has the potential to detect glycogen synthesis in the liver. However, the similar chemical shifts of [6,6'-2 H2 ]-glucose and [6,6'-2 H2 ]-glycogen in the 2 H NMR spectrum make unambiguous detection and separation difficult in vivo, in contrast to comparable approaches using 13 C MRS. Here the NMR visibility of 2 H-labeled glycogen is investigated to better understand its potential contribution to the observed signal in liver following administration of [6,6'-2 H2 ]-glucose. METHODS: Mice were provided drinking water containing 2 H-labeled glucose. High-resolution NMR analyses was performed of isolated liver glycogen in solution, before and after the addition of the glucose-releasing enzyme amyloglucosidase. RESULTS: 2 H-labeled glycogen was barely detectable in solution using 2 H NMR because of the very short T2 (<2 ms) of 2 H-labeled glycogen, giving a spectral line width that is more than five times as broad as that of 13 C-labeled glycogen (T2 = ~10 ms). CONCLUSION: 2 H-labeled glycogen is not detectable with 2 H MRS(I) under in vivo conditions, leaving 13 C MRS as the preferred technique for in vivo detection of glycogen.


Subject(s)
Liver Glycogen , Magnetic Resonance Imaging , Animals , Deuterium , Glucose , Liver/diagnostic imaging , Magnetic Resonance Spectroscopy , Mice
11.
J Cereb Blood Flow Metab ; 41(5): 986-1000, 2021 05.
Article in English | MEDLINE | ID: mdl-33472521

ABSTRACT

Neuroimaging with functional MRI (fMRI) identifies activated and deactivated brain regions in task-based paradigms. These patterns of (de)activation are altered in diseases, motivating research to understand their underlying biochemical/biophysical mechanisms. Essentially, it remains unknown how aerobic metabolism of glucose to lactate (aerobic glycolysis) and excitatory-inhibitory balance of glutamatergic and GABAergic neuronal activities vary in these areas. In healthy volunteers, we investigated metabolic distinctions of activating visual cortex (VC, a task-positive area) using a visual task and deactivating posterior cingulate cortex (PCC, a task-negative area) using a cognitive task. We used fMRI-guided J-edited functional MRS (fMRS) to measure lactate, glutamate plus glutamine (Glx) and γ-aminobutyric acid (GABA), as indicators of aerobic glycolysis and excitatory-inhibitory balance, respectively. Both lactate and Glx increased upon activating VC, but did not change upon deactivating PCC. Basal GABA was negatively correlated with BOLD responses in both brain areas, but during functional tasks GABA decreased in VC upon activation and GABA increased in PCC upon deactivation, suggesting BOLD responses in relation to baseline are impacted oppositely by task-induced inhibition. In summary, opposite relations between BOLD response and GABAergic inhibition, and increases in aerobic glycolysis and glutamatergic activity distinguish the BOLD response in (de)activated areas.


Subject(s)
Brain/metabolism , Glutamic Acid/metabolism , Magnetic Resonance Imaging/methods , Visual Cortex/metabolism , gamma-Aminobutyric Acid/metabolism , 3-Hydroxybutyric Acid/metabolism , Adult , Brain/anatomy & histology , Brain Mapping/instrumentation , Female , Glycolysis/physiology , Gyrus Cinguli/metabolism , Humans , Lactic Acid/metabolism , Magnetic Resonance Imaging/statistics & numerical data , Male , Neurovascular Coupling/physiology , Visual Cortex/physiology
12.
Mol Psychiatry ; 26(9): 5097-5111, 2021 09.
Article in English | MEDLINE | ID: mdl-32488125

ABSTRACT

Both the NMDA receptor (NMDAR) positive allosteric modulator (PAM), and antagonist, can exert rapid antidepressant effects as shown in several animal and human studies. However, how this bidirectional modulation of NMDARs causes similar antidepressant effects remains unknown. Notably, the initial cellular trigger, specific cell-type(s), and subunit(s) of NMDARs mediating the antidepressant-like effects of a PAM or an antagonist have not been identified. Here, we used electrophysiology, microdialysis, and NMR spectroscopy to evaluate the effect of a NMDAR PAM (rapastinel) or NMDAR antagonist, ketamine on NMDAR function and disinhibition-mediated glutamate release. Further, we used cell-type specific knockdown (KD), pharmacological, and behavioral approaches to dissect the cell-type specific role of GluN2B, GluN2A, and dopamine receptor subunits in the actions of NMDAR PAM vs. antagonists. We demonstrate that rapastinel directly enhances NMDAR activity on principal glutamatergic neurons in medial prefrontal cortex (mPFC) without any effect on glutamate efflux, while ketamine blocks NMDAR on GABA interneurons to cause glutamate efflux and indirect activation of excitatory synapses. Behavioral studies using cell-type-specific KD in mPFC demonstrate that NMDAR-GluN2B KD on Camk2a- but not Gad1-expressing neurons blocks the antidepressant effects of rapastinel. In contrast, GluN2B KD on Gad1- but not Camk2a-expressing neurons blocks the actions of ketamine. The results also demonstrate that Drd1-expressing pyramidal neurons in mPFC mediate the rapid antidepressant actions of ketamine and rapastinel. Together, these results demonstrate unique initial cellular triggers as well as converging effects on Drd1-pyramidal cell signaling that underlie the antidepressant actions of NMDAR-positive modulation vs. NMDAR blockade.


Subject(s)
Ketamine , Receptors, N-Methyl-D-Aspartate , Animals , Antidepressive Agents/pharmacology , Humans , Interneurons/metabolism , Ketamine/pharmacology , Prefrontal Cortex/metabolism , Pyramidal Cells/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism
13.
ACS Chem Neurosci ; 12(1): 234-243, 2021 01 06.
Article in English | MEDLINE | ID: mdl-33319987

ABSTRACT

Deuterium metabolic imaging (DMI) is a novel, 3D, magnetic resonance (MR)-based method to map metabolism of deuterated substrates in vivo. The replacement of protons with deuterons could potentially lead to kinetic isotope effects (KIEs) in which metabolic rates of deuterated substrates are reduced due to the presence of a heavier isotope. Knowledge of the extent of KIE in vivo and 2H label loss due to exchange reactions is required for DMI-based measurements of absolute metabolic rates. Here the deuterium KIE and label loss in vivo are investigated for glucose and acetate using a double substrate/double labeling strategy and 1H-decoupled 13C NMR in rat glioma cells and rat brain tissue metabolite extracts. The unique spectral patterns due to extensive 2H-13C and 13C-13C scalar couplings allow the identification of all possible metabolic products. The 2H label loss observed in lactate, glutamate, and glutamine of rat brain was 15.7 ± 2.6, 37.9 ± 1.1, and 41.5 ± 5.2% when using [6,6-2H2]-glucose as the metabolic substrate. For [2-2H3]-acetate, the 2H label loss in glutamate and glutamine was 14.4 ± 3.4 and 13.6 ± 2.2%, respectively, in excellent agreement with predicted values. Steady-state 2H label accumulation in the C4 position of glutamate and glutamine was contrasted by the absence of label accumulation in the C2 or C3 positions, indicating that during a full turn of the tricarboxylic acid cycle all 2H label is lost. The measured KIE was relatively small (4-6%) for both substrates and all measured metabolic products. These results pave the way for further development of quantitative DMI studies to generate metabolic flux maps in vivo.


Subject(s)
Glutamic Acid , Glutamine , Animals , Carbon Isotopes , Deuterium , Isotope Labeling , Kinetics , Magnetic Resonance Spectroscopy , Rats
14.
NMR Biomed ; 34(5): e4393, 2021 05.
Article in English | MEDLINE | ID: mdl-33236818

ABSTRACT

Proton MR spectra of the brain, especially those measured at short and intermediate echo times, contain signals from mobile macromolecules (MM). A description of the main MM is provided in this consensus paper. These broad peaks of MM underlie the narrower peaks of metabolites and often complicate their quantification but they also may have potential importance as biomarkers in specific diseases. Thus, separation of broad MM signals from low molecular weight metabolites enables accurate determination of metabolite concentrations and is of primary interest in many studies. Other studies attempt to understand the origin of the MM spectrum, to decompose it into individual spectral regions or peaks and to use the components of the MM spectrum as markers of various physiological or pathological conditions in biomedical research or clinical practice. The aim of this consensus paper is to provide an overview and some recommendations on how to handle the MM signals in different types of studies together with a list of open issues in the field, which are all summarized at the end of the paper.


Subject(s)
Brain/diagnostic imaging , Consensus , Expert Testimony , Macromolecular Substances/metabolism , Proton Magnetic Resonance Spectroscopy , Adult , Aged , Aged, 80 and over , Humans , Lipids/chemistry , Magnetic Resonance Imaging , Metabolome , Middle Aged , Models, Theoretical , Signal Processing, Computer-Assisted , Young Adult
15.
NMR Biomed ; 32(10): e4172, 2019 10.
Article in English | MEDLINE | ID: mdl-31478594

ABSTRACT

In the last 25 years 13 C MRS has been established as the only noninvasive method for measuring glutamate neurotransmission and cell specific neuroenergetics. Although technically and experimentally challenging 13 C MRS has already provided important new information on the relationship between neuroenergetics and neuronal function, the high energy cost of brain function in the resting state and the role of altered neuroenergetics and neurotransmitter cycling in disease. In this paper we review the metabolic and neurotransmitter pathways that can be measured by 13 C MRS and key findings on the linkage between neuroenergetics, neurotransmitter cycling, and brain function. Applications of 13 C MRS to neurological and psychiatric disease as well as brain cancer are reviewed. Recent technological developments that may help to overcome spatial resolution and brain coverage limitations of 13 C MRS are discussed.


Subject(s)
Brain Neoplasms/metabolism , Carbon Isotopes/chemistry , Magnetic Resonance Spectroscopy , Mental Disorders/metabolism , Neurotransmitter Agents/metabolism , Animals , Brain Neoplasms/physiopathology , Humans , Mental Disorders/physiopathology , Synaptic Transmission
16.
Neurochem Int ; 129: 104508, 2019 10.
Article in English | MEDLINE | ID: mdl-31326460

ABSTRACT

Glutaminase mediates the recycling of neurotransmitter glutamate, supporting most excitatory neurotransmission in the mammalian central nervous system. A constitutive heterozygous reduction in GLS1 engenders in mice a model of schizophrenia resilience and associated increases in Gln, reductions in Glu and activity-dependent attenuation of excitatory synaptic transmission. Hippocampal brain slices from GLS1 heterozygous mice metabolize less Gln to Glu. Whether glutaminase activity is diminished in the intact brain in GLS1 heterozygous mice has not been assessed, nor the regional impact. Moreover, it is not known whether pharmacological inhibition would mimic the genetic reduction. We addressed this using magnetic resonance spectroscopy to assess amino acid content and 13C-acetate loading to assess glutaminase activity, in multiple brain regions. Glutaminase activity was reduced significantly in the hippocampus of GLS1 heterozygous mice, while acute treatment with the putative glutaminase inhibitor ebselen did not impact glutaminase activity, but did significantly increase GABA. This approach identifies a molecular imaging strategy for testing target engagement by comparing genetic and pharmacological inhibition, across brain regions.


Subject(s)
Azoles/pharmacology , Brain/enzymology , Glutaminase/antagonists & inhibitors , Nerve Tissue Proteins/antagonists & inhibitors , Organoselenium Compounds/pharmacology , Amino Acids/analysis , Animals , Brain Chemistry/drug effects , Female , Glutaminase/analysis , Glutaminase/genetics , Heterozygote , Hippocampus/drug effects , Hippocampus/enzymology , Isoindoles , Male , Mice , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/genetics , Nuclear Magnetic Resonance, Biomolecular , Sequence Deletion , gamma-Aminobutyric Acid/analysis
17.
Sci Adv ; 4(8): eaat7314, 2018 08.
Article in English | MEDLINE | ID: mdl-30140744

ABSTRACT

Currently, the only widely available metabolic imaging technique in the clinic is positron emission tomography (PET) detection of the radioactive glucose analog 2-18F-fluoro-2-deoxy-d-glucose (18FDG). However, 18FDG-PET does not inform on metabolism downstream of glucose uptake and often provides ambiguous results in organs with intrinsic high glucose uptake, such as the brain. Deuterium metabolic imaging (DMI) is a novel, noninvasive approach that combines deuterium magnetic resonance spectroscopic imaging with oral intake or intravenous infusion of nonradioactive 2H-labeled substrates to generate three-dimensional metabolic maps. DMI can reveal glucose metabolism beyond mere uptake and can be used with other 2H-labeled substrates as well. We demonstrate DMI by mapping metabolism in the brain and liver of animal models and human subjects using [6,6'-2H2]glucose or [2H3]acetate. In a rat glioma model, DMI revealed pronounced metabolic differences between normal brain and tumor tissue, with high-contrast metabolic maps depicting the Warburg effect. We observed similar metabolic patterns and image contrast in two patients with a high-grade brain tumor after oral intake of 2H-labeled glucose. Further, DMI used in rat and human livers showed [6,6'-2H2]glucose stored as labeled glycogen. DMI is a versatile, robust, and easy-to-implement technique that requires minimal modifications to existing clinical magnetic resonance imaging scanners. DMI has great potential to become a widespread method for metabolic imaging in both (pre)clinical research and the clinic.


Subject(s)
Brain Mapping/methods , Brain/pathology , Deuterium/metabolism , Glioma/pathology , Glucose/metabolism , Magnetic Resonance Imaging/methods , Animals , Brain/metabolism , Case-Control Studies , Glioma/metabolism , Humans , Image Processing, Computer-Assisted/methods , Male , Middle Aged , Rats , Rats, Inbred F344
18.
J Neurochem ; 146(6): 722-734, 2018 09.
Article in English | MEDLINE | ID: mdl-29964293

ABSTRACT

Depression is one of the most debilitating neuropsychiatric disorders. Most of the current antidepressants have long remission time and low recovery rate. This study explores the impact of ketamine on neuronal and astroglial metabolic activity in prefrontal cortex in a social defeat (SD) model of depression. C57BL/6 mice were subjected to a social defeat paradigm for 5 min a day for 10 consecutive days. Ketamine (10 mg/kg, intraperitoneal) was administered to mice for two consecutive days following the last defeat stress. Mice were infused with [1,6-13 C2 ]glucose or [2-13 C]acetate to assess neuronal and astroglial metabolic activity, respectively, together with proton-observed carbon-edited nuclear magnetic resonance spectroscopy in prefrontal cortex tissue extract. The 13 C labeling of amino acids from glucose and acetate was decreased in SD mice. Ketamine treatment in SD mice restored sucrose preference, social interaction and immobility time to control values. Acute subanesthetic ketamine restored the 13 C labeling of brain amino acids from glucose as well as acetate in SD mice to the respective control values, suggesting that rates of neuronal and astroglial tricarboxylic acid (TCA) cycle and neurotransmitter cycling were re-established to normal levels. The finding of improved energy metabolism in SD mice suggests that fast anti-depressant action of ketamine is linked with improved neurotransmitter cycling.


Subject(s)
Analgesics/therapeutic use , Astrocytes/metabolism , Depressive Disorder , Ketamine/therapeutic use , Neurons/metabolism , Stress, Psychological/complications , Acetates/pharmacokinetics , Animals , Astrocytes/drug effects , Carbon Isotopes/pharmacokinetics , Depressive Disorder/drug therapy , Depressive Disorder/etiology , Depressive Disorder/pathology , Depressive Disorder/psychology , Disease Models, Animal , Food Preferences/drug effects , Glucose/pharmacokinetics , Hierarchy, Social , Interpersonal Relations , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Sucrose/administration & dosage , Sweetening Agents/administration & dosage , Swimming/psychology
19.
Neuroscientist ; 24(4): 316-328, 2018 08.
Article in English | MEDLINE | ID: mdl-29276856

ABSTRACT

Ceftriaxone stimulates astrocytic uptake of the excitatory neurotransmitter glutamate, and it is used to treat glutamatergic excitotoxicity that becomes manifest during many brain diseases. Ceftriaxone-stimulated glutamate transport was reported to drive signals underlying [18F]fluorodeoxyglucose-positron emission tomographic ([18F]FDG-PET) metabolic images of brain glucose utilization and interpreted as supportive of the notion of lactate shuttling from astrocytes to neurons. This study draws attention to critical roles of astrocytes in the energetics and imaging of brain activity, but the results are provocative because (1) the method does not have cellular resolution or provide information about downstream pathways of glucose metabolism, (2) neuronal and astrocytic [18F]FDG uptake were not separately measured, and (3) strong evidence against lactate shuttling was not discussed. Evaluation of potential metabolic responses to ceftriaxone suggests lack of astrocytic specificity and significant contributions by pre- and postsynaptic neuronal compartments. Indeed, astrocytic glycolysis may not make a strong contribution to the [18F]FDG-PET signal because partial or complete oxidation of one glutamate molecule on its uptake generates enough ATP to fuel uptake of 3 to 10 more glutamate molecules, diminishing reliance on glycolysis. The influence of ceftriaxone on energetics of glutamate-glutamine cycling must be determined in astrocytes and neurons to elucidate its roles in excitotoxicity treatment.


Subject(s)
Brain/drug effects , Brain/diagnostic imaging , Ceftriaxone/pharmacology , Central Nervous System Agents/pharmacology , Glutamic Acid/metabolism , Positron-Emission Tomography , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Brain/metabolism , Fluorodeoxyglucose F18 , Humans , Neurons/drug effects , Neurons/metabolism , Radiopharmaceuticals
SELECTION OF CITATIONS
SEARCH DETAIL
...