Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 119(48): e2210584119, 2022 11 29.
Article in English | MEDLINE | ID: mdl-36413502

ABSTRACT

Antiretroviral therapy (ART) can attain prolonged undetectable HIV-1 in plasma and cerebrospinal fluid (CSF), but brain injury remains prevalent in people living with HIV-1 infection (PLHIV). We investigated cell-associated (CA)-HIV-1 RNA transcripts in cells in CSF and blood, using the highly sensitive Double-R assay, together with proton Magnetic Resonance Spectroscopy (1H MRS) of major brain metabolites, in sixteen PLHIV. 14/16 CSF cell samples had quantifiable CA-HIV-1 RNA, at levels significantly higher than in their PBMCs (median 9,266 vs 185 copies /106 CD4+ T-cells; p<0.0001). In individual PLHIV, higher levels of HIV-1 transcripts in CSF cells were associated with greater brain injury in the frontal white matter (Std ß=-0.73; p=0.007) and posterior cingulate (Std ß=-0.61; p=0.03). 18-colour flow cytometry revealed that the CSF cells were 91% memory T-cells, equally CD4+ and CD8+ T-cells, but fewer B cells (0.4 %), and monocytes (3.1%). CXCR3+CD49d+integrin ß7-, CCR5+CD4+ T-cells were highly enriched in CSF, compared with PBMC (p <0.001). However, CA-HIV-1 RNA could not be detected in 10/16 preparations of highly purified monocytes from PBMC, and was extremely low in the other six. Our data show that elevated HIV-1 transcripts in CSF cells were associated with brain injury, despite suppressive ART. The cellular source is most likely memory CD4+ T cells from blood, rather than trafficking monocytes. Future research should focus on inhibitors of this transcription to reduce local production of potentially neurotoxic and inflammatory viral products.


Subject(s)
Brain Injuries , HIV Infections , HIV Seropositivity , HIV-1 , Humans , HIV-1/genetics , CD4-Positive T-Lymphocytes , Leukocytes, Mononuclear , HIV Infections/drug therapy
2.
J Neurochem ; 168(5): 910-954, 2024 May.
Article in English | MEDLINE | ID: mdl-38183680

ABSTRACT

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.


Subject(s)
Brain , Energy Metabolism , Animals , Humans , Brain/metabolism
3.
J Neurochem ; 164(4): 451-453, 2023 02.
Article in English | MEDLINE | ID: mdl-36495565

ABSTRACT

The ISMRM study group on magnetic resonance spectroscopy has produced recommendations for reporting methods. The Journal of Neurochemistry has decided to encourage the use of the checklist for these standards by authors and reviewers in order to improve reproducibility and reliability of the science, make it easier for reviewers and to help educate the scientific community. Here, we explain why getting the details right is important.


Subject(s)
Checklist , Reproducibility of Results , Reference Standards , Magnetic Resonance Spectroscopy
4.
J Neurochem ; 2023 Mar 28.
Article in English | MEDLINE | ID: mdl-36977628

ABSTRACT

AMP-activated protein kinase (AMPK) is a key sensor of energy balance playing important roles in the balancing of anabolic and catabolic activities. The high energy demands of the brain and its limited capacity to store energy indicate that AMPK may play a significant role in brain metabolism. Here, we activated AMPK in guinea pig cortical tissue slices, both directly with A769662 and PF 06409577 and indirectly with AICAR and metformin. We studied the resultant metabolism of [1-13 C]glucose and [1,2-13 C]acetate using NMR spectroscopy. We found distinct activator concentration-dependent effects on metabolism, which ranged from decreased metabolic pool sizes at EC50 activator concentrations with no expected stimulation in glycolytic flux to increased aerobic glycolysis and decreased pyruvate metabolism with certain activators. Further, activation with direct versus indirect activators produced distinct metabolic outcomes at both low (EC50 ) and higher (EC50 × 10) concentrations. Specific direct activation of ß1-containing AMPK isoforms with PF 06409577 resulted in increased Krebs cycle activity, restoring pyruvate metabolism while A769662 increased lactate and alanine production, as well as labelling of citrate and glutamine. These results reveal a complex metabolic response to AMPK activators in brain beyond increased aerobic glycolysis and indicate that further research is warranted into their concentration- and mechanism-dependent impact.

5.
J Neurosci Res ; 101(2): 263-277, 2023 02.
Article in English | MEDLINE | ID: mdl-36353842

ABSTRACT

Substantia nigra (SN) hyperechogenicity, viewed with transcranial ultrasound, is a risk marker for Parkinson's disease. We hypothesized that SN hyperechogenicity in healthy adults aged 50-70 years is associated with reduced short-interval intracortical inhibition in primary motor cortex, and that the reduced intracortical inhibition is associated with neurochemical markers of activity in the pre-supplementary motor area (pre-SMA). Short-interval intracortical inhibition and intracortical facilitation in primary motor cortex was assessed with paired-pulse transcranial magnetic stimulation in 23 healthy adults with normal (n = 14; 61 ± 7 yrs) or abnormally enlarged (hyperechogenic; n = 9; 60 ± 6 yrs) area of SN echogenicity. Thirteen of these participants (7 SN- and 6 SN+) also underwent brain magnetic resonance spectroscopy to investigate pre-SMA neurochemistry. There was no relationship between area of SN echogenicity and short-interval intracortical inhibition in the ipsilateral primary motor cortex. There was a significant positive relationship, however, between area of echogenicity in the right SN and the magnitude of intracortical facilitation in the right (ipsilateral) primary motor cortex (p = .005; multivariate regression), evidenced by the amplitude of the conditioned motor evoked potential (MEP) at the 10-12 ms interstimulus interval. This relationship was not present on the left side. Pre-SMA glutamate did not predict primary motor cortex inhibition or facilitation. The results suggest that SN hyperechogenicity in healthy older adults may be associated with changes in excitability of motor cortical circuitry. The results advance understanding of brain changes in healthy older adults at risk of Parkinson's disease.


Subject(s)
Cortical Excitability , Motor Cortex , Parkinson Disease , Humans , Aged , Motor Cortex/diagnostic imaging , Parkinson Disease/diagnostic imaging
6.
J Sleep Res ; 31(2): e13482, 2022 04.
Article in English | MEDLINE | ID: mdl-34528315

ABSTRACT

It is challenging to determine which patients with obstructive sleep apnea (OSA) have impaired driving ability. Vulnerability to this neurobehavioral impairment may be explained by lower brain metabolites levels involved in mitochondrial metabolism. This study compared markers of brain energy metabolism in OSA patients identified as vulnerable vs resistant to driving impairment following extended wakefulness. 44 patients with moderate-severe OSA underwent 28hr extended wakefulness with three 90min driving simulation assessments. Using a two-step cluster analysis, objective driving data (steering deviation and crashes) from the 2nd driving assessment (22.5 h awake) was used to categorise patients into vulnerable (poor driving, n = 21) or resistant groups (good driving, n = 23). 1 H magnetic resonance spectra were acquired at baseline using two scan sequences (short echo PRESS and longer echo-time asymmetric PRESS), focusing on key metabolites, creatine, glutamate, N-acetylaspartate (NAA) in the hippocampus, anterior cingulate cortex and left orbito-frontal cortex. Based on cluster analysis, the vulnerable group had impaired driving performance compared with the resistant group and had lower levels of creatine (PRESS p = ns, APRESS p = 0.039), glutamate, (PRESS p < 0.01, APRESS p < 0.01), NAA (PRESS p = 0.038, APRESS p = 0.035) exclusively in the left orbito-frontal cortex. Adjusted analysis, higher glutamate was associated with a 21% (PRESS) and 36% (APRESS) reduced risk of vulnerable classification. Brain mitochondrial bioenergetics in the frontal brain regions are impaired in OSA patients who are vulnerable to driving impairment following sleep loss. These findings provide a potential way to identify at risk OSA phenotype when assessing fitness to drive, but this requires confirmation in larger future studies.


Subject(s)
Automobile Driving , Sleep Apnea, Obstructive , Brain/diagnostic imaging , Creatine , Glutamates , Humans , Mitochondria
7.
J Neurochem ; 156(1): 13-15, 2021 01.
Article in English | MEDLINE | ID: mdl-33197055

ABSTRACT

The last common ancestor of humans and fruit flies lived about 800 million years ago, yet both of us have nervous systems that share a number of common important features, for example the use of glutamate as a neurotransmitter. We can now possibly add another common feature to the neural tissue of humans and fruit flies which is that of N-acetylaspartylglutamate (NAAG) peptides. This Editorial highlights an article by Kozik and coworkers in the current issue of the Journal of Neurochemistry, in which the authors report the discovery, in Drosophila melanogaster nervous system, of NAA-glutamylglutamate (NAAG2).


Subject(s)
Aspartic Acid , Drosophila , Animals , Dipeptides , Drosophila melanogaster , Nervous System , Pedigree
8.
J Neurosci ; 39(46): 9185-9196, 2019 11 13.
Article in English | MEDLINE | ID: mdl-31570533

ABSTRACT

It has long been thought that severe chronic pain conditions, such as complex regional pain syndrome (CRPS), are not only associated with, but even maintained by a reorganization of the somatotopic representation of the affected limb in primary somatosensory cortex (S1). This notion has driven treatments that aim to restore S1 representations in CRPS patients, such as sensory discrimination training and mirror therapy. However, this notion is based on both indirect and incomplete evidence obtained with imaging methods with low spatial resolution. Here, we used fMRI to characterize the S1 representation of the affected and unaffected hand in humans (of either sex) with unilateral CRPS. The cortical area, location, and geometry of the S1 representation of the CRPS hand were largely comparable with those of both the unaffected hand and healthy controls. We found no differential relation between affected versus unaffected hand map measures and clinical measures (pain severity, upper limb disability, disease duration). Thus, if any map reorganization occurs, it does not appear to be directly related to pain and disease severity. These findings compel us to reconsider the cortical mechanisms underlying CRPS and the rationale for interventions that aim to "restore" somatotopic representations to treat pain.SIGNIFICANCE STATEMENT This study shows that the spatial map of the fingers in somatosensory cortex is largely preserved in chronic complex regional pain syndrome (CRPS). These findings challenge the treatment rationale for restoring somatotopic representations in complex regional pain syndrome patients.


Subject(s)
Complex Regional Pain Syndromes/physiopathology , Neuronal Plasticity , Somatosensory Cortex/physiopathology , Adult , Aged , Brain Mapping , Complex Regional Pain Syndromes/diagnostic imaging , Female , Hand/physiopathology , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Physical Stimulation , Somatosensory Cortex/diagnostic imaging , Young Adult
9.
Neurochem Res ; 45(10): 2527, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32638216

ABSTRACT

The original version of this published article, the bottom right hand panels of Figs. 3-6 were labelled as "Isotopomers formed from [1-13C]D-glucose". This is incorrect and should read "Isotopomers formed from [1,2-13C]acetate". This has been corrected by publishing this correction article.

10.
Neurochem Res ; 45(6): 1438-1450, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32424601

ABSTRACT

L-Ornithine-L-aspartate (LOLA), a crystalline salt, is used primarily in the management of hepatic encephalopathy. The degree to which it might penetrate the brain, and the effects it might have on metabolism in brain are poorly understood. Here, to investigate the effects of LOLA on brain energy metabolism we incubated brain cortical tissue slices from guinea pig (Cavea porcellus) with the constituent amino acids of LOLA, L-ornithine or L-aspartate, as well as LOLA, in the presence of [1-13C]D-glucose and [1,2-13C]acetate; these labelled substrates are useful indicators of brain metabolic activity. L-Ornithine produced significant "sedative" effects on brain slice metabolism, most likely via conversion of ornithine to GABA via the ornithine aminotransferase pathway, while L-aspartate showed concentration-dependent excitatory effects. The metabolic effects of LOLA reflected a mix of these two different processes and were concentration-dependent. We also investigated the effect of an intraperitoneal bolus injection of L-ornithine, L-aspartate or LOLA on levels of metabolites in kidney, liver and brain cortex and brain stem in mice (C57Bl6J) 1 h later. No significant changes in metabolite levels were seen following the bolus injection of L-aspartate, most likely due to rapid metabolism of aspartate before reaching the target tissue. Brain cortex glutamate was decreased by L-ornithine but no other brain effects were observed with any other compound. Kidney levels of aspartate were increased after injection of L-ornithine and LOLA which may be due to interference by ornithine with the kidney urea cycle. It is likely that without optimising chronic intravenous infusion, LOLA has minimal impact on healthy brain energy metabolism due to systemic clearance and the blood - brain barrier.


Subject(s)
Aspartic Acid/metabolism , Brain/metabolism , Dipeptides/metabolism , Energy Metabolism/physiology , Ornithine/metabolism , Animals , Aspartic Acid/pharmacology , Brain/drug effects , Dipeptides/pharmacology , Dose-Response Relationship, Drug , Energy Metabolism/drug effects , Female , Guinea Pigs , Male , Mice , Mice, Inbred C57BL , Ornithine/pharmacology
11.
Neuropsychol Rev ; 29(3): 357-385, 2019 09.
Article in English | MEDLINE | ID: mdl-31512192

ABSTRACT

This review provides the first systematic and quantitative synthesis of the literature examining the relationship between binge drinking, cognition, brain structure and function in youth aged 10 to 24 years. PubMed, EMBASE, Medline, PsychINFO and ProQuest were searched for neuroimaging, neurophysiological, and neuropsychological studies. A total of 58 studies (21 neuroimaging, 16 neurophysiological, 21 neuropsychological) met the eligibility criteria and were included in the review. Overall, abnormal or delayed development of key frontal executive-control regions may predispose youth to binge drink. These abnormalities appear to be further exacerbated by the uptake of binge drinking, in addition to alcohol-related neural aberrations in reward-seeking and incentive salience regions, indexed by cognitive deficits and maladaptive alcohol associations. A meta-analysis of neuropsychological correlates identified that binge drinking in youth was associated with a small overall neurocognitive deficit (g = -0.26) and specific deficits in decision-making (g = -1.70), and inhibition (g = -0.39). Using the Grades of Recommendation, Assessment, Development, and Evaluation (GRADE) Evidence Profile, the certainty in outcomes ranged from very low to low. Future prospective longitudinal studies should address concomitant factors, exposure thresholds, and age-related vulnerabilities of binge drinking, as well as the degree of recovery following discontinuation of use.


Subject(s)
Binge Drinking , Brain/pathology , Brain/physiopathology , Adolescent , Binge Drinking/pathology , Binge Drinking/physiopathology , Binge Drinking/psychology , Decision Making , Executive Function , Humans , Inhibition, Psychological , Neuropsychological Tests
12.
Hum Brain Mapp ; 39(5): 1945-1956, 2018 05.
Article in English | MEDLINE | ID: mdl-29341331

ABSTRACT

Recurrent thalamocortical connections are integral to the generation of brain rhythms and it is thought that the inhibitory action of the thalamic reticular nucleus is critical in setting these rhythms. Our work and others' has suggested that chronic pain that develops following nerve injury, that is, neuropathic pain, results from altered thalamocortical rhythm, although whether this dysrhythmia is associated with thalamic inhibitory function remains unknown. In this investigation, we used electroencephalography and magnetic resonance spectroscopy to investigate cortical power and thalamic GABAergic concentration in 20 patients with neuropathic pain and 20 pain-free controls. First, we found thalamocortical dysrhythmia in chronic orofacial neuropathic pain; patients displayed greater power than controls over the 4-25 Hz frequency range, most marked in the theta and low alpha bands. Furthermore, sensorimotor cortex displayed a strong positive correlation between cortical power and pain intensity. Interestingly, we found no difference in thalamic GABA concentration between pain subjects and control subjects. However, we demonstrated significant linear relationships between thalamic GABA concentration and enhanced cortical power in pain subjects but not controls. Whilst the difference in relationship between thalamic GABA concentration and resting brain rhythm between chronic pain and control subjects does not prove a cause and effect link, it is consistent with a role for thalamic inhibitory neurotransmitter release, possibly from the thalamic reticular nucleus, in altered brain rhythms in individuals with chronic neuropathic pain.


Subject(s)
Brain Waves/physiology , Cerebral Cortex/physiopathology , Neuralgia/pathology , Rest , Thalamus/metabolism , gamma-Aminobutyric Acid/metabolism , Adult , Aged , Brain Mapping , Cerebral Cortex/diagnostic imaging , Electroencephalography , Female , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Male , Middle Aged , Neuralgia/diagnostic imaging , Young Adult
13.
Acta Neuropathol ; 135(1): 95-113, 2018 01.
Article in English | MEDLINE | ID: mdl-29116375

ABSTRACT

N-Acetylaspartate (NAA) is the second most abundant organic metabolite in the brain, but its physiological significance remains enigmatic. Toxic NAA accumulation appears to be the key factor for neurological decline in Canavan disease-a fatal neurometabolic disorder caused by deficiency in the NAA-degrading enzyme aspartoacylase. To date clinical outcome of gene replacement therapy for this spongiform leukodystrophy has not met expectations. To identify the target tissue and cells for maximum anticipated treatment benefit, we employed comprehensive phenotyping of novel mouse models to assess cell type-specific consequences of NAA depletion or elevation. We show that NAA-deficiency causes neurological deficits affecting unconscious defensive reactions aimed at protecting the body from external threat. This finding suggests, while NAA reduction is pivotal to treat Canavan disease, abrogating NAA synthesis should be avoided. At the other end of the spectrum, while predicting pathological severity in Canavan disease mice, increased brain NAA levels are not neurotoxic per se. In fact, in transgenic mice overexpressing the NAA synthesising enzyme Nat8l in neurons, supra-physiological NAA levels were uncoupled from neurological deficits. In contrast, elimination of aspartoacylase expression exclusively in oligodendrocytes elicited Canavan disease like pathology. Although conditional aspartoacylase deletion in oligodendrocytes abolished expression in the entire CNS, the remaining aspartoacylase in peripheral organs was sufficient to lower NAA levels, delay disease onset and ameliorate histopathology. However, comparable endpoints of the conditional and complete aspartoacylase knockout indicate that optimal Canavan disease gene replacement therapies should restore aspartoacylase expression in oligodendrocytes. On the basis of these findings we executed an ASPA gene replacement therapy targeting oligodendrocytes in Canavan disease mice resulting in reversal of pre-existing CNS pathology and lasting neurological benefits. This finding signifies the first successful post-symptomatic treatment of a white matter disorder using an adeno-associated virus vector tailored towards oligodendroglial-restricted transgene expression.


Subject(s)
Aspartic Acid/analogs & derivatives , Brain/metabolism , Brain/pathology , Canavan Disease/metabolism , Canavan Disease/therapy , Acetyltransferases/metabolism , Amidohydrolases/administration & dosage , Amidohydrolases/genetics , Amidohydrolases/metabolism , Animals , Aspartic Acid/metabolism , Brain/diagnostic imaging , Canavan Disease/pathology , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem/physiology , Evoked Potentials, Visual/physiology , Female , Genetic Therapy , Humans , Male , Mice, Transgenic , Neurons/metabolism , Neurons/pathology , Oligodendroglia/metabolism , Oligodendroglia/pathology , Phenotype , RNA, Messenger/metabolism
14.
J Neurochem ; 140(6): 903-918, 2017 03.
Article in English | MEDLINE | ID: mdl-27925207

ABSTRACT

[13 C]Acetate is known to label metabolites preferentially in astrocytes rather than neurons and it has consequently been used as a marker for astrocytic activity. Recent discoveries suggest that control of acetate metabolism and its contributions to the synthesis of metabolites in brain is not as simple as first thought. Here, using a Guinea pig brain cortical tissue slice model metabolizing [1-13 C]D-glucose and [1,2-13 C]acetate, we investigated control of acetate metabolism and the degree to which it reflects astrocytic activity. Using a range of [1,2-13 C]acetate concentrations, we found that acetate is a poor substrate for metabolism and will inhibit metabolism of itself and of glucose at concentrations in excess of 2 mmol/L. By activating astrocytes using potassium depolarization, we found that use of [1,2-13 C]acetate to synthesize glutamine decreases significantly under these conditions showing that acetate metabolism does not necessarily reflect astrocytic activity. By blocking synthesis of glutamine using methionine sulfoximine, we found that significant amount of [1,2-13 C]acetate are still incorporated into GABA and its metabolic precursors in neurons, with around 30% of the GABA synthesized from [1,2-13 C]acetate likely to be made directly in neurons rather than from glutamine supplied by astrocytes. Finally, to test whether activity of the acetate metabolizing enzyme acetyl-CoA synthetase is under acetylation control in the brain, we incubated slices with the AceCS1 deacetylase silent information regulator 1 (SIRT1) activator SRT 1720 and showed consequential increased incorporation of [1,2-13 C]acetate into metabolites. Taken together, these data show that acetate metabolism is not directly nor exclusively related to astrocytic metabolic activity, that use of acetate is related to enzyme acetylation and that acetate is directly metabolized to a significant degree in GABAergic neurons. Changes in acetate metabolism should be interpreted as modulation of metabolism through changes in cellular energetic status via altered enzyme acetylation levels rather than simply as an adjustment of glial-neuronal metabolic activity.


Subject(s)
Acetic Acid/metabolism , Astrocytes/metabolism , GABAergic Neurons/metabolism , Sirtuin 1/metabolism , gamma-Aminobutyric Acid/biosynthesis , Animals , Cerebral Cortex/metabolism , Female , Guinea Pigs , Organ Culture Techniques
15.
Anal Biochem ; 529: 127-143, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28034792

ABSTRACT

We review the transport, synthesis and catabolism of glutathione in the brain as well as its compartmentation and biochemistry in different brain cells. The major reactions involving glutathione are reviewed and the factors limiting its availability in brain cells are discussed. We also describe and critique current methods for measuring glutathione in the human brain using magnetic resonance spectroscopy, and review the literature on glutathione measurements in healthy brains and in neurological, psychiatric, neurodegenerative and neurodevelopmental conditions In summary: Healthy human brain glutathione concentration is ∼1-2 mM, but it varies by brain region, with evidence of gender differences and age effects; in neurological disease glutathione appears reduced in multiple sclerosis, motor neurone disease and epilepsy, while being increased in meningiomas; in psychiatric disease the picture is complex and confounded by methodological differences, regional effects, length of disease and drug-treatment. Both increases and decreases in glutathione have been reported in depression and schizophrenia. In Alzheimer's disease and mild cognitive impairment there is evidence for a decrease in glutathione compared to age-matched healthy controls. Improved methods to measure glutathione in vivo will provide better precision in glutathione determination and help resolve the complex biochemistry of this molecule in health and disease.


Subject(s)
Brain/metabolism , Glutathione/metabolism , Magnetic Resonance Imaging/methods , Magnetic Resonance Spectroscopy/methods , Animals , Humans , Models, Biological
16.
Neurochem Res ; 42(1): 35-49, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27826689

ABSTRACT

ß-Hydroxybutyrate (ßOHB), a ketone body, is oxidised as a brain fuel. Although its contribution to energy metabolism in the healthy brain is minimal, it is an interesting metabolite which is not only oxidised but also has other direct and collateral effects which make it a molecule of interest for therapeutic purposes. In brain ßOHB can be produced in astrocytes from oxidation of fatty acids or catabolism of amino acids and is metabolised in the mitochondria of all brain cell types although uptake across the blood brain barrier is a metabolic control point. ßOHB possesses an intrinsic high heat of combustion, making it an efficient mitochondrial fuel, where it can alter the NAD+/NADH and Q/QH2 couples and reduce production of mitochondrial reactive oxygen species. It can directly interact as a signalling molecule influencing opening of K+ channels and regulation of Ca2+ channels. ßOHB is an inhibitor of histone deacetylases resulting in upregulation of genes involved in protection against oxidative stress and regulation of metabolism. It interacts with an inflammasome in immune cells to reduce production of inflammatory cytokines and reduce inflammation. Use of ßOHB as an efficient neurotherapeutic relies on increasing blood ßOHB levels so as to encourage entry of ßOHB to the brain. While use of ßOHB as a sole therapeutic is currently limited, with employment of a ketogenic diet a more widely used approach, recent development and testing of esterified forms of ßOHB have shown great promise, with the approach elevating plasma ßOHB while allowing consumption of normal diet. An improved understanding of the mechanisms by which ßOHB acts will allow better design of both diet and supplemental interventions.


Subject(s)
3-Hydroxybutyric Acid/metabolism , Brain/metabolism , Energy Metabolism/physiology , Oxidative Stress/physiology , Animals , Blood-Brain Barrier/metabolism , Diet, Ketogenic/methods , Humans
17.
Neurochem Res ; 42(6): 1710-1723, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28316020

ABSTRACT

The ketone body, ß-hydroxybutyrate (ßOHB), is metabolised by the brain alongside the mandatory brain fuel glucose. To examine the extent and circumstances by which ßOHB can supplement glucose metabolism, we studied guinea pig cortical brain slices using increasing concentrations of [U-13C]D-ßOHB in conjunction with [1-13C]D-glucose under conditions of normo- and hypoglycaemia, as well as under high potassium (40 mmol/L K+) depolarization in normo- and hypoglycaemic conditions. The contribution of ßOHB to synthesis of GABA was also probed by inhibiting the synthesis of glutamine, a GABA precursor, with methionine sulfoximine (MSO). [U-13C]D-ßOHB at lower concentrations (0.25 and 1.25 mmol/L) stimulated mitochondrial metabolism, producing greater total incorporation of label into glutamate and GABA but did not have a similar effect in the cytosolic compartment where labelling of glutamine was reduced at 1.25 mmol/L [U-13C]D-ßOHB. At higher concentrations (2.5 mmol/L) [U-13C]D-ßOHB inhibited metabolism of [1-13C]D-glucose, and reduced total label incorporation and total metabolite pools. When glucose levels were reduced, ßOHB was able to partially restore the loss of glutamate and GABA caused by hypoglycaemia, but was not able to supplement levels of lactate, glutamine or alanine or to prevent the increase in aspartate. Under depolarizing conditions glucose was the preferred substrate over ßOHB, even in hypoglycaemic conditions where comparatively less ßOHB was incorporated except into aspartate isotopomers. Inhibition of glutamine synthesis with MSO had no significant effect on incorporation of label from [U-13C]D-ßOHB into GABA C2,1 indicating that the majority of this GABA was synthesized in GABAergic neurons from [U-13C]D-ßOHB rather than from Gln C4,5 imported from astrocytes.


Subject(s)
3-Hydroxybutyric Acid/metabolism , 3-Hydroxybutyric Acid/pharmacology , Glucose/metabolism , Mitochondria/metabolism , Neurons/metabolism , Animals , Dose-Response Relationship, Drug , Glutamine/metabolism , Guinea Pigs , Male , Mitochondria/drug effects , Neurons/drug effects
18.
J Neurochem ; 139(5): 806-822, 2016 12.
Article in English | MEDLINE | ID: mdl-27696399

ABSTRACT

Toluene is a commonly abused inhalant that is easily accessible to adolescents. Despite the increasing incidence of use, our understanding of its long-term impact remains limited. Here, we used a range of techniques to examine the acute and chronic effects of toluene exposure on glutameteric and GABAergic function, and on indices of psychological function in adult rats after adolescent exposure. Metabolomics conducted on cortical tissue established that acute exposure to toluene produces alterations in cellular metabolism indicative of a glutamatergic and GABAergic profile. Similarly, in vitro electrophysiology in Xenopus oocytes found that acute toluene exposure reduced NMDA receptor signalling. Finally, in an adolescent rodent model of chronic intermittent exposure to toluene (10 000 ppm), we found that, while toluene exposure did not affect initial learning, it induced a deficit in updating that learning when response-outcome relationships were reversed or degraded in an instrumental conditioning paradigm. There were also group differences when more effort was required to obtain the reward; toluene-exposed animals were less sensitive to progressive ratio schedules and to delayed discounting. These behavioural deficits were accompanied by changes in subunit expression of both NMDA and GABA receptors in adulthood, up to 10 weeks after the final exposure to toluene in the hippocampus, prefrontal cortex and ventromedial striatum; regions with recognized roles in behavioural flexibility and decision-making. Collectively, our data suggest that exposure to toluene is sufficient to induce adaptive changes in glutamatergic and GABAergic systems and in adaptive behaviour that may underlie the deficits observed following adolescent inhalant abuse, including susceptibility to further drug-use.


Subject(s)
Receptors, GABA-A/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Toluene/administration & dosage , Toluene/toxicity , Administration, Inhalation , Age Factors , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Female , Guinea Pigs , Learning/drug effects , Learning/physiology , Male , Organ Culture Techniques , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/physiology , Solvents/administration & dosage , Solvents/toxicity , Xenopus laevis
19.
J Neurochem ; 134(1): 3-6, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26052752

ABSTRACT

This Editorial highlights a study by Gibson et al. published in this issue of JNeurochem, in which the authors reveal a novel role for the α-ketoglutarate dehydrogenase complex (KGDHC) in post-translational modification of proteins. KGDHC may catalyze post-translational modification of itself as well as several other proteins by succinylation of lysine residues. The authors' report of an enzyme responsible for succinylation of key mitochondrial enzymes represents a major step toward our understanding of the complex functional metabolome. TCA, tricarboxylic acid; KG, α-ketoglutarate; KGDHC, α-ketoglutarate dehydrogenase complex; FUM, fumarase; MDH, malate dehydrogenase; ME, malic enzyme; GDH, glutamate dehydrogenase; AAT, aspartate aminotransferase; GS, glutamine synthetase; PAG, phosphate-activated glutaminase; SIRT3, silent information regulator 3; SIRT5, silent information regulator 5.


Subject(s)
Acyl Coenzyme A/metabolism , Ketoglutarate Dehydrogenase Complex/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Animals , Female , Male
20.
J Neurosci Res ; 93(7): 1147-56, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25677687

ABSTRACT

Silent information regulators (SIRTs) have been shown to deacetylate a range of metabolic enzymes, including those in glycolysis and the Krebs cycle, and thus alter their activity. SIRTs require NAD(+) for their activity, linking cellular energy status to enzyme activity. To examine the impact of SIRT1 modulation on oxidative metabolism, this study tests the effect of ligands that are either SIRT-activating compounds (resveratrol and SRT1720) or SIRT inhibitors (EX527) on the metabolism of (13)C-enriched substrates by guinea pig brain cortical tissue slices with (13)C and (1)H nuclear magnetic resonance spectroscopy. Resveratrol increased lactate labeling but decreased incorporation of (13)C into Krebs cycle intermediates, consistent with effects on AMPK and inhibition of the F0/F1-ATPase. By testing with resveratrol that was directly applied to astrocytes with a Seahorse analyzer, increased glycolytic shift and increased mitochondrial proton leak resulting from interactions of resveratrol with the mitochondrial electron transport chain were revealed. SRT1720, by contrast, stimulated incorporation of (13)C into Krebs cycle intermediates and reduced incorporation into lactate, although the inhibitor EX527 paradoxically also increased Krebs cycle (13)C incorporation. In summary, the various SIRT1 modulators show distinct acute effects on oxidative metabolism. The strong effects of resveratrol on the mitochondrial respiratory chain and on glycolysis suggest that caution should be used in attempts to increase bioavailability of this compound in the CNS.


Subject(s)
Antioxidants/pharmacology , Brain , Lactic Acid/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Stilbenes/pharmacology , Animals , Area Under Curve , Astrocytes/drug effects , Astrocytes/metabolism , Brain/drug effects , Brain/metabolism , Brain/ultrastructure , Carbazoles/pharmacology , Carbon Isotopes/metabolism , Dose-Response Relationship, Drug , Female , Guinea Pigs , Heterocyclic Compounds, 4 or More Rings/pharmacology , Magnetic Resonance Imaging , Male , Oxidative Stress/drug effects , Oxygen Consumption/drug effects , Resveratrol , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL