Category: Moms

Energy metabolism and cognitive function

Energy metabolism and cognitive function

A Refillable hand sanitizer overview of glucose dynamics in the gliovascular unit. Sleep and wakefulness. Cognutive Activity. Functkon DA, Halliwell B. Limitations of this pilot study include a small sample size and the inability to differentiate between gray matter, white matter, neurons or glia. Department of Immunology, Tufts University School of Medicine, Boston, MA, USA.

Energy metabolism and cognitive function -

Trends Cell Biol ; 24 2 : Sato TK, Panda S, Miraglia LJ, et al. A functional genomics strategy reveals Rora as a component of the mammalian circadian clock. Neuron ; 43 4 : Preitner N, Damiola F, López-Molina L, et al.

The orphan nuclear receptor REV-ERBalpha controls circadian transcription within the positive limb of the mammalian circadian oscillator. Cell ; 2 : Ukai-Tadenuma M, Yamada RG, Xu H, Ripperger JA, Liu AC, Ueda HR. Delay in feedback repression by cryptochrome 1 is required for circadian clock function.

Challet E. The circadian regulation of food intake. Nat Rev Endocrinol ; 15 7 : Kim P, Oster H, Lehnert H, et al. Coupling the circadian clock to homeostasis: the role of period in timing physiology.

Endocr Rev ; 40 1 : de Goede P, Foppen E, Ritsema WIGR, Korpel NL, Yi CX, Kalsbeek A. Time-restricted feeding improves glucose tolerance in rats, but only when in line with the circadian timing system.

Front Endocrinol Lausanne ; Chaix A, Lin T, Le HD, Chang MW, Panda S. Time-restricted feeding prevents obesity and metabolic syndrome in mice lacking a circadian clock. Cell Metab ; 29 2 : Mendoza J, Pévet P, Challet E. High-fat feeding alters the clock synchronization to light.

J Physiol ; 24 : Mendoza J, Lopez-Lopez C, Revel FG, et al. Dimorphic effects of leptin on the circadian and hypocretinergic systems of mice.

J Neuroendocrinol ; 23 1 : x ] [PMID: ]. Yulyaningsih E, Loh K, Lin S, et al. Pancreatic polypeptide controls energy homeostasis via Npy6r signaling in the suprachiasmatic nucleus in mice.

Cell Metab ; 19 1 : Prosser RA, Bergeron HE. Leptin phase-advances the rat suprachiasmatic circadian clock in vitro. Neurosci Lett ; 3 : Yi CX, van der Vliet J, Dai J, Yin G, Ru L, Buijs RM. Ventromedial arcuate nucleus communicates peripheral metabolic information to the suprachiasmatic nucleus.

Endocrinology ; 1 : Saderi N, Cazarez-Márquez F, Buijs FN, et al. The NPY intergeniculate leaflet projections to the suprachiasmatic nucleus transmit metabolic conditions. Neuroscience ; Jang H, Lee G, Kong J, Choi G, Park YJ, Kim JB.

Feeding period restriction alters the expression of peripheral circadian rhythm genes without changing body weight in mice. Blancas-Velázquez A, la Fleur SE, Mendoza J. Effects of a free-choice high-fat high-sugar diet on brain PER2 and BMAL1 protein expression in mice. Appetite ; Del Rio D, Cano V, Martín-Ramos M, et al.

Involvement of the dorsomedial prefrontal cortex in high-fat food conditioning in adolescent mice. Del Olmo N, Blanco-Gandía MC, Mateos-García A, et al. Differential Impact of Ad Libitum or Intermittent High-Fat Diets on Bingeing Ethanol-Mediated Behaviors. Fick LJ, Fick GH, Belsham DD.

Palmitate alters the rhythmic expression of molecular clock genes and orexigenic neuropeptide Y mRNA levels within immortalized, hypothalamic neurons. Biochem Biophys Res Commun ; 3 : Greco JA, Oosterman JE, Belsham DD.

Am J Physiol Regul Integr Comp Physiol ; 8 : Resuehr D, Olcese J. Caloric restriction and melatonin substitution: effects on murine circadian parameters. Feillet CA, Albrecht U, Challet E. J Physiol Paris ; : Mendoza J, Gourmelen S, Dumont S, Sage-Ciocca D, Pévet P, Challet E.

Setting the main circadian clock of a diurnal mammal by hypocaloric feeding. J Physiol ; 13 : Sen S, Raingard H, Dumont S, Kalsbeek A, Vuillez P, Challet E.

Ultradian feeding in mice not only affects the peripheral clock in the liver, but also the master clock in the brain. Chronobiol Int ; 34 1 : Oosterman JE, Kalsbeek A, la Fleur SE, Belsham DD.

Impact of nutrients on circadian rhythmicity. Am J Physiol Regul Integr Comp Physiol ; 5 : R Mackenberg EJ, Broverman DM, Vogel W, Klaiber EL. Morning-to-afternoon changes in cognitive performances and in the electroencephalogram. J Educ Psychol ; 66 2 : Borbély AA. A two process model of sleep regulation.

Hum Neurobiol ; 1 3 : Wright KP Jr, Hull JT, Hughes RJ, Ronda JM, Czeisler CA. Sleep and wakefulness out of phase with internal biological time impairs learning in humans. J Cogn Neurosci ; 18 4 : Schmidt C, Collette F, Cajochen C, Peigneux P.

A time to think: circadian rhythms in human cognition. Cogn Neuropsychol ; 24 7 : Gerstner JR, Lyons LC, Wright KP Jr, et al. Cycling behavior and memory formation. J Neurosci ; 29 41 : Valdez P. Homeostatic and circadian regulation of cognitive performance.

Biol Rhythm Res ; 50 1 : Kleitman N. Sleep and wakefulness. Chicago, IL: The University of Chicago Press Wright KP Jr, Hull JT, Czeisler CA. Relationship between alertness, performance, and body temperature in humans. Am J Physiol Regul Integr Comp Physiol ; 6 : R Krishnan HC, Lyons LC.

Synchrony and desynchrony in circadian clocks: impacts on learning and memory. Learn Mem ; 22 9 : Kaliyaperumal D, Elango Y, Alagesan M, Santhanakrishanan I.

Effects of sleep deprivation on the cognitive performance of nurses working in shift. J Clin Diagn Res ; 11 8 : CC Chellappa SL, Morris CJ, Scheer FAJL. Daily circadian misalignment impairs human cognitive performance task-dependently. Sci Rep ; 8 1 : Devore EE, Grodstein F, Schernhammer ES.

Am J Epidemiol ; 8 : Davies JA, Navaratnam V, Redfern PH. A hour rhythm in passive-avoidance behaviour in rats. Psychopharmacology Berl ; 32 2 : Valentinuzzi VS, Kolker DE, Vitaterna MH, Ferrari EAM, Takahashi JS, Turek FW. Anim Learn Behav ; 29 2 : Chaudhury D, Colwell CS.

Circadian modulation of learning and memory in fear-conditioned mice. Behav Brain Res ; 1 : Hoffmann HJ, Balschun D. Behav Processes ; 27 2 : Hauber W, Bareiss A. Winocur G, Hasher L. Age and time-of-day effects on learning and memory in a non-matching-to-sample test.

Neurobiol Aging ; 25 8 : Ruby NF, Hwang CE, Wessells C, et al. Hippocampal-dependent learning requires a functional circadian system.

Proc Natl Acad Sci USA ; 40 : Takahashi Y, Sawa K, Okada T. The diurnal variation of performance of the novel location recognition task in male rats.

Gritton HJ, Kantorowski A, Sarter M, Lee TM. Bidirectional interactions between circadian entrainment and cognitive performance. Learn Mem ; 19 3 : Soler JE, Stumpfig M, Tang YP, Robison AJ, Núñez AA, Yan L. Daytime light intensity modulates spatial learning and hippocampal plasticity in female Nile Grass rats Arvicanthis niloticus.

Gritton HJ, Stasiak AM, Sarter M, Lee TM. Cognitive performance as a zeitgeber: cognitive oscillators and cholinergic modulation of the SCN entrain circadian rhythms. Eckel-Mahan KL, Phan T, Han S, et al. Circadian oscillation of hippocampal MAPK activity and cAmp: implications for memory persistence.

Nat Neurosci ; 11 9 : Snider KH, Dziema H, Aten S, et al. Modulation of learning and memory by the targeted deletion of the circadian clock gene Bmal1 in forebrain circuits. Jilg A, Lesny S, Peruzki N, et al.

Temporal dynamics of mouse hippocampal clock gene expression support memory processing. Hippocampus ; 20 3 : Rawashdeh O, Jilg A, Jedlicka P, et al. PERIOD1 coordinates hippocampal rhythms and memory processing with daytime.

Hippocampus ; 24 6 : Rawashdeh O, Jilg A, Maronde E, Fahrenkrug J, Stehle JH. Period1 gates the circadian modulation of memory-relevant signaling in mouse hippocampus by regulating the nuclear shuttling of the CREB kinase pP90RSK. J Neurochem ; 5 : Shimizu K, Kobayashi Y, Nakatsuji E, et al.

Nat Commun ; 7: Wang LM-C, Dragich JM, Kudo T, et al. Expression of the circadian clock gene Period2 in the hippocampus: possible implications for synaptic plasticity and learned behaviour. Dallman MF, Strack AM, Akana SF, et al. Feast and famine: critical role of glucocorticoids with insulin in daily energy flow.

Front Neuroendocrinol ; 14 4 : McEwen BS, Sapolsky RM. Stress and cognitive function. Curr Opin Neurobiol ; 5 2 : Chen Y, Andres AL, Frotscher M, Baram TZ.

CMR glc is the cerebral metabolic rate of glucose and is usually determined together with the apparent maximum transport T max and the apparent Michaelis constant of glucose transport K t using labeling of plasma and brain glucose Duarte et al.

In glial cells, GLS is neglected because the net 13 C labeling follows the direction of glutamine synthesis.

V ex can exchange with unlabeled glutamine of undefined origin Oz et al. Besides the contribution to amino acid synthesis, V PC also represents glutamine efflux V efflux from the brain i. Glutamate oxidation in the model is possible through V X composite representation of AST and GDH.

Note that both V Gln and V NT represent the glutamate-glutamine cycle. One is rather used in the one-compartment model V Gln as an exchange rate between glutamate and glutamine, while the other in the two-compartment model V NT.

Labeling of a particular nucleus depends on pool size and the numbers of upstream and downstream fluxes Henry et al. Glutamate is the most concentrated amino acid observed by 13 C MRS Figure 4. Glutamate labeling depends on both V TCA and V X , meaning that these two fluxes play an important role in defining glutamate turnover.

While glutamate C4 relies on the composite flux V gt , that is V X. Therefore, the information concerning V X is mainly stored in the initial slopes of the turnover curves of position C4, C3, and C2 of glutamate for V X n and glutamine for V X g. In the special case of V X g , this flux is particularly difficult to estimate, because labeled molecules from the glial TCA cycle into glial glutamate can also be transferred via V PC Figure 2.

The value of V X has been matter of debate for a long time, as it has been considered by some to be much larger than V TCA Mason et al. Figure 4. Typical 13 C MRS spectra acquired in vivo in the rat brain during [1,6- 13 C 2 ]glucose infusion at Panel A shows a time course of 13 C labeling with a temporal resolution of 5.

The spectrum in B was acquired for 1. Panel C is an expansion of B depicting multiplets originated from isotopomers of glutamine Gln , glutamate Glu and aspartate Asp.

The neurotransmission rate, V NT , represents the conversion of glutamate to glutamine and vice versa. In this process the carbon positions are maintained. Therefore, V NT mostly depends on the relative steady-state enrichment of the turnover curves of glutamate and glutamine: the closer they are to each other, the faster V NT.

The first turn of the TCA cycle results in label transfer from glucose to glutamate via V X. Usually glutamate C4 is the first detectable peak in a spectrum during an experiment, as it appears within the first 5 min of infusion Patel et al. Then, in the subsequent TCA cycle label is transferred from position C4 to C3 and C2.

In neurons, V PDH will therefore rely on the slope and the steady-state enrichment of the turnover curves of position C4, C3, and C2 of glutamate and glutamine, as labeling from glutamate is transferred to glutamine. As aspartate is mainly labeled via transamination of the TCA cycle intermediate oxaloacetate, the slope and the steady-state enrichment of position C3 and C2 of aspartate are further affected by V PDH.

The slopes of the labeling curves reflect, the rate of V PDH. In glia the situation is different, since label dilution due to V PC can occur. V PC dilutes position C3 and labels position C2 of glutamate and glutamine. Therefore, fast V g results in a relatively high and steep glutamine C3 turnover curve V g has to be fast to counterbalance the loss due to V PC.

Diluting position C3 and labeling position C2 of glutamate and glutamine, the measurement of V PC relies on the relative curves of position C3 and C2 of glutamate and glutamine, and the assumption that glutamate is mainly neuronal and glutamine mainly glial.

High C3 and low C2 labeling is associated with slow V PC , while high C2 and low C3 labeling reflect rather increased V PC. As mentioned above, V dil reflects dilution of the acetyl-CoA pool with specific unlabeled glial substrates.

Notably, V dil dilutes glial acetyl-CoA 13 C labeling relative to its precursor pyruvate. As the position C4 of glutamate and glutamine only receives labeling from acetyl-CoA, dilution at this point would lead to a lower steady-state C4 labeling. Since glutamate and glutamine are mainly present in neurons and glia, respectively, V dil is also responsible for lower FE of position C4, C3, and C2 of glutamine compared to glutamate.

However, the effect of V dil on enhancing the labeling difference between glutamate and glutamine is counteracted by V NT , which represents the glutamate-glutamine cycle. The faster the rate of V NT , the more similar will be the labeling of glutamate and glutamine.

Note that V dil in glial acetyl-CoA can result in glutamine C2 being similar or larger than glutamine C4, which has been observed in some studies discussed in Duarte et al. V ex represents an exchange between two putative glutamine pools, one of which is not released to neurons and may account for a continuous slow increase in FE over time Duarte and Gruetter, V ex can be in exchange with a 1 H MR invisible but 13 C labeled glutamine pool Hancu and Port, or with unlabeled amino acids from the blood i.

This second glutamine pool could be associated with biosynthetic pathways, which have rates much slower than mitochondrial energy metabolism McKenna, The effect of V ex is in practice observable near the end of an experiment, when the labeling of glutamine still increases, while glutamate is at steady-state.

Therefore, if mitochondrial metabolism is faster than glycolysis, oxidation of additional substrates, such as lactate, must occur under certain conditions Sonnay et al. In resting human brain, however, the brain exports lactate to the blood stream discussed in Dienel, The above descriptions are purely indicative of what happens for each flux independently.

Experimental data is a linear combination of many fluxes, which will adjust during fitting process to best describe the turnover curves. The first 13 C MRS data acquired in vivo upon stimulus-induced brain activity were modeled using a one-compartment model and reported a marked increase in total TCA cycle activity in the somatosensory cortex of stimulated rats compared to rest Hyder et al.

The following experiment consisted on measuring neuronal CMR glc ox under three different anesthesia-induced activity states, namely pentobarbital deep , α-chloralose moderate and morphine light Sibson et al.

According to this model, no stimulation of oxidative metabolism should occur in glia, in contrast to neurons. Later several studies in rat brain Patel et al. However, it should be noted that constraining the value of V PC to V GS and V TCA g to the total V TCA implies an effective coupling between glial oxidative metabolism and neuronal function.

Indeed, the astrocytic processes engulfing synapses are capable of sensing increased synaptic activity Iadecola and Nedergaard, ; Cheung et al. The 13 C MRS study by Gruetter et al. Using a similar model, glial oxidation and pyruvate carboxylase activity was shown to significantly contribute also to total glucose oxidation in awake animals Oz et al.

Recently, our group further addressed the issue of glial and neuronal oxidative metabolism coupled to neuronal activity. In particular, we first measured the cortical changes in metabolic fluxes induced by electrical stimulation of the four paws of rats.

We observed a similar increase in absolute terms of both glial and neuronal oxidative metabolism resulting from the increase in glutamate-glutamine cycle rate Figure 5 ; Sonnay et al.

Interestingly in this study, as well as in Patel et al. Indeed Patel and Tilghman reported that glutamate can stimulate pyruvate carboxylation Patel and Tilghman, Instead, glutamate could be oxidized in astrocytes to compensate for the high cost of glutamate uptake during neurotransmission McKenna, Figure 5.

Relation of estimated total, neuronal and glial glucose oxidative metabolism to the glutamate-glutamine cycle in the rat cortex anesthetized with α-chloralose originally reported in Sonnay et al.

Average fluxes across the resting in blue and stimulated in red group are shown with associated SD. In the study by Sonnay et al.

However, the simulations by DiNuzzo et al. are still unable to account for substantial V TCA g in cases of low glutamate-glutamine cycle rate. To summarize, in addition to the proposed coupling of neuronal oxidative metabolism and neurotransmission, astrocytes increase their oxidative metabolism too, resulting in a large production of ATP.

It is, therefore, important to investigate the exact fate of the ATP produced. In this context, the ATP produced in glia might notably support blood flow regulation Zonta et al. Glycogenolysis might moreover provide energy to support neurotransmission i. Brain vasculature is rich in arterioles and fine capillaries Reina-De La Torre et al.

In this context astrocytes and neurons are presumed to play a key role in modulating CBF to match energy demands. In astrocytes, activation of mGluR by glutamate triggers the translocation of the α-subunit of the receptors to phospholipase C PLC mediating the conversion of GTP to GDP Bockaert et al.

In neurons, cyclooxygenase COX converts AA into prostaglandins E 2 PGE 2 leading to vessel dilation Wang et al. In astrocytes, AA can be converted either to PGE 2 by COX Zonta et al. If AA is converted into hydroxyeicosatetraenoic acid HETE by ω-hydroxylase in pericytes, it will cause vasoconstriction Metea and Newman, New line of evidence suggest moreover that the astrocytic production of PGE 2 might be dependent on glutathione levels Howarth et al.

In neurons activation of ionotropic glutamate receptors located on the post-synaptic zone i. Interaction of NO with soluble guanylate cyclase sGC triggers cGMP dependent vasodilation mechanisms Laranjinha et al.

Intracellular adenosine can be released extracellularly by nucleoside transporters Iliff et al. Increase in cAMP leads to vasodilation and inhibits the vasoconstrictive effects of HETE Koehler et al. Figure 6. Schematic representation of possible signaling pathways mediating neurovascular coupling.

Arachidonic acid AA is then produced by phospholipase A 2 PLA 2. In astrocytes AA can be converted either to prostaglandins E 2 PGE 2 by cyclooxygenase COX or to epoxeicosatrienoic acids EET by epoxygenase for vasodilation.

If AA is converted into hydroxyeicosatetraenoic acid HETE by ω-hydroxylase, it will lead to vasoconstriction. In pericytes and smooth muscle cells NO interacts with the soluble guanylate cyclase sGC for cGMP- dependent vasodilation mechanisms.

Intracellular adenosine can be transported by the nucleoside transporters to activate the adenosine receptors AR for cAMP-dependent vasodilation mechanisms via adenylate cyclase and inhibiting the vasoconstrictive effects of HETE. Lactate can inhibit the astrocytic prostaglandin transporter PGT -mediated PGE-lactate exchange, increasing therefore extracellular PGE 2 concentration.

Astrocytes can modulate synaptic plasticity in releasing vesicles containing glutamate, D-serine, ATP and neurotrophic factors in an ATP-dependent manner.

Glutathione is produced in astrocytes and can be released through multidrug resistance proteins MRP mediating ATP hydrolysis. Release of ascorbate mediate non-hydrolytic ATP binding to volume-sensitive organic osmolyte-anion channel VSOAC and is stimulated by glutamate.

The dashed line represents vasodilation and the dotted line vasoconstriction. Local CBF response could be immediately regulated by fast ms feed-forward mechanisms directly related to neuronal activity e. Although astrocytes do not generate action potentials per se they can actively modulate synaptic transmission and neuronal synchronization in mediating notably the release of vesicles-containing neurotransmitters and neuromodulators, such as glutamate, ATP, adenosine, and D-serine.

In the extracellular space ATP can be converted to adenosine by the dephosphorylating action of the ectonucleotidase anchored at the plasma membrane Joseph et al. D-serine that can be released from astrocytes was also shown to modulate electrical neurotransmission by acting at the glycine binding site of NMDA receptor Stevens et al.

Interestingly, the number of astrocytic processes, as well as their contact with active synapses, are stimulated by extracellular glutamate and also involve actin-dependent mechanisms Cornell-Bell et al. Yet, the cooperative action of astrocytes in culture was shown to protect neurons against ROS toxicity Desagher et al.

The thiol group of the glutathione molecule acts as an important electron donor. While both neurons and astrocytes synthesize glutathione, neuronal glutathione levels are higher in the presence of astrocytes Dringen et al.

Glutathione transport across cells is notably mediated by multidrug resistance proteins MRP that belong to the subgroup ABCC of the ATP-binding cassette transporters, which mediate passage via ATP hydrolysis Borst and Elferink, ; Dringen and Hirrlinger, ; Figure 5. Activation of astrocytic glutamate receptors was shown to translocate nuclear factor-erythroid 2-realted factor-2 Nrf2 present in lower concentrations in neurons into the nucleus and to trigger the expression of antioxidant genes, notably related to glutathione metabolism Jimenez-Blasco et al.

Astrocytes synthesize large amount of hydrogen sulfide, which was demonstrated to not only have neuroprotective properties Lee et al. Ascorbate is also another important antioxidant anion in the brain and glutamate was demonstrated to stimulate its release from astrocytes Wilson et al.

Astrocytes are responsible for the recycling of the neuronal extracellularly released dehydroascorbic acid the oxidized form of ascorbate into ascorbate, which can be exported to neurons Covarrubias-Pinto et al. Extracellular transport of ascorbate from astrocytes is believed to be mediated by volume-sensitive organic osmolyte-anion channel VSOAC that requires non-hydrolytic ATP binding Jackson et al.

Considering the fact that efficacy of the mechanisms stimulated by astrocytic glutamate uptake depends on the density of the transporters at the plasma membrane Robinson, , efficient trafficking of EAAT2-containing vesicles and exocytosis must moreover take place Stenovec et al.

Glycogenesis glycogen production from glucose 1-phosphate by glycogen synthase and glycogenolysis glycogen breakdown to glucose 6-phosphate by the combined action of glycogen phosphorylase and phosphoglucomutase mainly occurs in astrocytes Dringen et al. In line with this, glycogen levels were found to increase with anesthesia Morgenthaler et al.

However, no change in brain glycogen level was measured during visual stimulation in humans Oz et al. While glycogen-derived lactate has been demonstrated to have a pivotal role in memory formation and consolidation Gibbs and Hertz, ; Suzuki et al.

Recently, astrocytic glycogenolysis was shown to provide energy to sustain glutamatergic neurotransmission i. Glycogen might act as a substrate for de novo formation of glutamate Gibbs et al. While the essential role of astrocytes to cerebral function is now widely accepted, quantitative assessment of their actual contribution to energy metabolism has been missing, notably because the methodologies did not allow differentiating between neurons and astrocytes.

Direct 13 C MRS along with advanced metabolic modeling can provide measurements of both neuronal and glial metabolism in specific brain regions and under various activation states.

In this context, new data indicate that the rate of astrocytic metabolism is about half of that in neurons, and can be activated by sensory stimulation and that the astrocytic response amplitude can be, in absolute terms, as large as in neurons, suggesting that the changes in ATP requirements associated with the glutamate-glutamine cycle are coupled with the ATP produced by glucose oxidation in both compartments.

Increase in neuronal metabolism likely supports neurotransmission-associated functions, such as restoration of ion gradients caused by action potentials, post-synaptic currents, and transport of glutamate into vesicles.

Adaptation of glial metabolism also provides energy for neurotransmission besides housekeeping tasks, likely fueling the production and action of modulators of neuronal activity and of synaptic plasticity, supply of antioxidant molecules and neurotrophic factors that are necessary for adequate brain function, and regulation of blood flow and volume.

Astrocytes are moreover important source of glycogen that can be used specifically for neurotransmission support. Progress in MR detection methods of 1 H and non- 1 H nuclei is a promising direction for more detailed and complete metabolic dataset acquisition.

While this provides insights into cellular function in vivo , it also requires improvement of current metabolic models describing best energy metabolism. Simultaneous acquisition of other types of data, such as electrical activity and blood flow, might contribute to more precise characterization of the coupling between brain function and energy metabolism by MRS.

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

The authors' research is supported by the Swiss National Science Foundation to JD and to RG ; the National Competence Center in Biomedical Imaging NCCBI ; and by Centre d'Imagerie BioMédicale CIBM of the UNIL, UNIGE, HUG, CHUV, EPFL, and the Leenaards and Jeantet Foundations. Abe, K. The possible role of hydrogen sulfide as an endogenous neuromodulator.

PubMed Abstract Google Scholar. Acuña, A. A failure in energy metabolism and antioxidant uptake precede symptoms of Huntington's disease in mice. doi: PubMed Abstract CrossRef Full Text Google Scholar.

Almeida, A. Nitric oxide switches on glycolysis through the AMP protein kinase and 6-phosphofructokinase pathway. Cell Biol. Ashrafi, G. GLUT4 mobilization supports energetic demands of active synapses. Neuron 93, — Attwell, D. Glial and neuronal control of brain blood flow.

Nature , — An energy budget for signaling in the grey matter of the brain. Blood Flow Metab. Bakken, I. Neuroreport 8, — Barros, L. A quantitative overview of glucose dynamics in the gliovascular unit. Glia 55, — Glucose and lactate supply to the synapse.

Brain Res. Bastiaansen, J. Direct noninvasive estimation of myocardial tricarboxylic acid cycle flux in vivo using hyperpolarized 13 C magnetic resonance. Cell Cardiol. CrossRef Full Text Google Scholar. In vivo enzymatic activity of acetylCoA synthetase in skeletal muscle revealed by 13 C turnover from hyperpolarized [1- 13 C]acetate to [1- 13 C]acetylcarnitine.

Acta , — Bazargani, N. Astrocyte calcium signaling: the third wave. Bednařík, P. Neurochemical and BOLD responses during neuronal activation measured in the human visual cortex at 7 Tesla. Bergersen, L. Immunogold cytochemistry identifies specialized membrane domains for monocarboxylate transport in the central nervous system.

Biesecker, K. Glial cell calcium signalling mediates capillary regulation of blood flow in the retina. Bittner, C. Blood, A. Adenosine mediates decreased cerebral metabolic rate and increased cerebral blood flow during acute moderate hypoxia in the near-term fetal sheep.

Bockaert, J. Metabotropic glutamate receptors: an original family of G protein-coupled receptors. Bolaños, J. Glycolysis: a bioenergetic or a survival pathway? Trends Biochem. Borst, P. Mammalian ABC transporters in health and disease. Boury-Jamot, B.

Disrupting astrocyte-neuron lactate transfer persistently reduces conditioned responses to cocaine. Psychiatry 21, — Bouzier, A.

The metabolism of [3- 13 C]lactate in the rat brain is specific of a pyruvate carboxylase-deprived compartment. Bouzier-Sore, A. Uncertainties in pentose-phosphate pathway flux assessment underestimate its contribution to neuronal glucose consumption: relevance for neurodegeneration and aging.

Aging Neurosci. Boveris, A. The mitochondrial generation of hydrogen peroxide. General properties and effect of hyperbaric oxygen. Buerk, D. Temporal dynamics of brain tissue nitric oxide during functional forepaw stimulation in rats. Neuroimage 18, 1—9.

Buxton, R. Interpreting oxygenation-based neuroimaging signals: the importance and the challenge of understanding brain oxygen metabolism. Neuroenergetics Cerdan, S.

Cerebral metabolism of [1,2- 13 C 2 ]acetate as detected by in vivo and in vitro 13 C NMR. Chaudhry, F. Molecular analysis of system N suggests novel physiological roles in nitrogen metabolism and synaptic transmission.

Cell 99, — Glutamine uptake by neurons: interaction of protons with system a transporters. Cheung, G. Connexons and pannexons: newcomers in neurophysiology.

Cell Neurosci. Choi, I. Effect of deep pentobarbital anesthesia on neurotransmitter metabolism in vivo : on the correlation of total glucose consumption with glutamatergic action.

Comment, A. Dissolution DNP for in vivo preclinical studies. Cornell-Bell, A. The excitatory neurotransmitter glutamate causes filopodia formation in cultured hippocampal astrocytes.

Glia 3, — Covarrubias-Pinto, A. Old things new view: ascorbic acid protects the brain in neurodegenerative disorders. Cruz, F. Quantitative 13 C NMR studies of metabolic compartmentation in the adult mammalian brain. NMR Biomed. Ontogeny and cellular localization of the pyruvate recycling system in rat brain.

Cruz, N. Activation of astrocytes in brain of conscious rats during acoustic stimulation: acetate utilization in working brain. Deelchand, D. Simultaneous measurement of neuronal and glial metabolism in rat brain in vivo using co-infusion of [1,6- 13 C 2 ]glucose and [1,2- 13 C 2 ]acetate. de Graaf, R.

In vivo NMR Spectroscopy: Principles and Techniques. England: Willey. Google Scholar. In vivo 1 H-[ 13 C]-NMR spectroscopy of cerebral metabolism. Regional glucose metabolism and glutamatergic neurotransmission in rat brain in vivo. Dehghani, M. Refined analysis of brain energy metabolism using in vivo dynamic enrichment of 13 C multiplets.

ASN Neuro. Denton, R. Regulation of mitochondrial dehydrogenases by calcium ions. Derouiche, A. Astroglial processes around identified glutamatergic synapses contain glutamine synthetase: evidence for transmitter degradation.

Fine astrocyte processes contain very small mitochondria: glial oxidative capability may fuel transmitter metabolism. Desagher, S. Astrocytes protect neurons from hydrogen peroxide toxicity. Dienel, G. Brain lactate metabolism: the discoveries and the controversies.

A glycogen phosphorylase inhibitor selectively enhances local rates of glucose utilization in brain during sensory stimulation of conscious rats: implications for glycogen turnover. Astrocyte activation in vivo during graded photic stimulation.

DiNuzzo, M. Dringen, R. Glutathione pathways in the brain. Synthesis of the antioxidant glutathione in neurons: supply by astrocytes of CysGly as precursor for neuronal glutathione.

Incorporation of radioactivity from [ 14 C]lactate into the glycogen of cultured mouse astroglial cells. Evidence for gluconeogenesis in brain cells. Hoppe Seyler , — Duarte, J.

Adenosine A 1 receptors control the metabolic recovery after hypoxia in rat hippocampal slices. Brain energy metabolism measured by 13 C magnetic resonance spectroscopy in vivo upon infusion of [3- 13 C]lactate.

Glutamatergic and GABAergic energy metabolism measured in the rat brain by 13 C NMR spectroscopy at Compartmentalized Cerebral Metabolism of [1,6- 13 C]Glucose Determined by in vivo 13 C NMR Spectroscopy at The neurochemical profile quantified by in vivo 1 H NMR spectroscopy.

Neuroimage 61, — Steady-state brain glucose transport kinetics re-evaluated with a four-state conformational model. Duran, J. Impairment in long-term memory formation and learning-dependent synaptic plasticity in mice lacking glycogen synthase in the brain.

El Idrissi, A. Growth factors and taurine protect against excitotoxicity by stabilizing calcium homeostasis and energy metabolism. Erb, L. Coupling of P2Y receptors to G proteins and other signaling pathways. Wiley Interdiscip. Eriksson, G. Sodium-dependent glutamate uptake as an activator of oxidative metabolism in primary astrocyte cultures from newborn rat.

Avgerinos KI, Egan JM, Mattson MP, Kapogiannis D. A systematic review and meta-analysis of human studies. Croteau E, Castellano CA, Richard MA, Fortier M, Nugent S, Lepage M, et al. Achanta LB, Rae CD. beta-Hydroxybutyrate in the brain: one molecule, multiple mechanisms. Neurochem Res. An Y, Varma VR, Varma S, Casanova R, Dammer E, Pletnikova O, et al.

Talbot K, Wang HY, Kazi H, Han LY, Bakshi KP, Stucky A, et al. J Clin Invest. Gabbouj S, Ryhanen S, Marttinen M, Wittrahm R, Takalo M, Kemppainen S, et al. Front Neurosci. Nguyen NH, Brathe A, Hassel B.

Neuronal uptake and metabolism of glycerol and the neuronal expression of mitochondrial glycerolphosphate dehydrogenase.

Rust RS Jr. Enzyme levels in cultured astrocytes, oligodendrocytes and Schwann cells, and neurons from the cerebral cortex and superior cervical ganglia of the rat. McKenna MC, Waagepetersen HS, Schousboe A, Sonnewald U.

Neuronal and astrocytic shuttle mechanisms for cytosolic-mitochondrial transfer of reducing equivalents: current evidence and pharmacological tools.

Biochem Pharm. Valdebenito R, Ruminot I, Garrido-Gerter P, Fernandez-Moncada I, Forero-Quintero L, Alegria K, et al. Targeting of astrocytic glucose metabolism by beta-hydroxybutyrate.

J Cereb Blood Flow Metab. Meyer K, Feldman HM, Lu T, Drake D, Lim ET, Ling KH, et al. Cell Rep. Pacholko AG, Wotton CA, Bekar LK. Neurodegener Dis. Vlassenko AG, Vaishnavi SN, Couture L, Sacco D, Shannon BJ, Mach RH, et al. Spatial correlation between brain aerobic glycolysis and amyloid-beta Abeta deposition.

Baik SH, Kang S, Lee W, Choi H, Chung S, Kim JI, et al. Zhao J, Davis MD, Martens YA, Shinohara M, Graff-Radford NR, Younkin SG, et al. Hum Mol Genet. Mayer PR, Huang N, Dewey CM, Dries DR, Zhang H, Yu G. Expression, localization, and biochemical characterization of nicotinamide mononucleotide adenylyltransferase 2.

Raffaelli N, Sorci L, Amici A, Emanuelli M, Mazzola F, Magni G. Identification of a novel human nicotinamide mononucleotide adenylyltransferase. Biochemical Biophysical Res Commun. Article CAS Google Scholar. Download references.

This study was supported by the Program for Neuropsychiatric Research, McLean Hospital BMC and NRF grants R1A2B, R1H1A, IITP grant from MSIT HS.

The authors want to thank Ryan A. Healy, Yoon Lee, Wang Liao, Miseon Oh, and Jinfeng Xuan for technical, and Suzanne Babb, Fontini Savvides, and Tamare V. Adrien for administrative help.

The authors also thank Dr. Rosario Sanchez-Pernaute and Dr. Christian LeGuern for critically reading the manuscript and helpful discussions. Department of Psychiatry, Harvard Medical School, Belmont, MA, USA. Woo-In Ryu, Mariana K. Bormann, Minqi Shen, Brent Forester, Kai-C. Basic Neuroscience Division, Harvard Medical School, Belmont, MA, USA.

Program for Neuropsychiatric Research, McLean Hospital, Harvard Medical School, Belmont, MA, USA. Bormann, Minqi Shen, Kai-C. Department of Immunology, Tufts University School of Medicine, Boston, MA, USA. Mood Disorders Division and Geriatric Psychiatry Research Program, McLean Hospital, Harvard Medical School, Belmont, MA, USA.

Department of Molecular and Life Sciences, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan, South Korea.

Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA. You can also search for this author in PubMed Google Scholar. All authors reviewed the manuscript. KCS and BMC conceptualized and designed, and BMC funded the study. WIR, MKB, MS, DK, YP, and JS performed the experiments.

KCS, WIR, MKB, and HS analyzed and interpreted the data, and KCS wrote the paper. BF recruited and diagnosed subjects. Correspondence to Kai-C. Sonntag or Bruce M. Open Access This article is licensed under a Creative Commons Attribution 4. Reprints and permissions. Ryu, WI. et al.

Mol Psychiatry 26 , — Download citation. Received : 20 August Revised : 01 March Accepted : 18 March Published : 16 April Issue Date : October Anyone you share the following link with will be able to read this content:. Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative. Signal Transduction and Targeted Therapy Skip to main content Thank you for visiting nature. nature molecular psychiatry articles article. Download PDF.

Subjects Biochemistry Cell biology Diseases Neuroscience Stem cells. Full size image. Discussion LOAD is a complexly determined disorder characterized by a combination of multiple interacting pathological processes, some of which are genetically determined and inherent in neurodevelopment and in youth and some of which are part of the normal aging process.

Methods Subject population and cell derivation Subjects Supplementary Table 1 were recruited at the McLean Hospital Memory Diagnostic Clinic. Cell culture Cells were differentiated and propagated in standard tissue culture. Biochemical assays Bioenergetic parameters were determined using Seahorse XFp Cell Mito Stress Tests Seahorse, Agilent Technologies, Santa Clara, CA and the processing of bioenergetic substrates were assessed in Biolog MitoPlate S-1 assays Biolog, Hayward, CA.

Molecular assays Total RNA was isolated and purified using TRI-Reagent MilliporeSigma and RNA integrity was measured with an Agilent Bioanalyzer Agilent. Detailed information of the materials and methods is presented in the Supplement Information.

References Jack CR Jr, Bennett DA, Blennow K, Carrillo MC, Dunn B, Haeberlein SB, et al. Article PubMed PubMed Central Google Scholar Hardy JA, Higgins GA.

Article CAS PubMed Google Scholar Herrup K. Article CAS PubMed Google Scholar Sims R, Hill M, Williams J. Article CAS PubMed Google Scholar Amtul Z. Article PubMed Google Scholar Krstic D, Knuesel I. Article CAS PubMed Google Scholar Wilkins JM, Trushina E.

Article PubMed Google Scholar de la Monte SM. Article PubMed PubMed Central Google Scholar Mattson MP, Arumugam TV. Article CAS PubMed PubMed Central Google Scholar Hou Y, Dan X, Babbar M, Wei Y, Hasselbalch SG, Croteau DL, et al. Article PubMed Google Scholar Mookerjee SA, Gerencser AA, Nicholls DG, Brand MD.

Article CAS PubMed PubMed Central Google Scholar Bordone MP, Salman MM, Titus HE, Amini E, Andersen JV, Chakraborti B, et al. Article CAS PubMed Google Scholar Dienel GA. Article CAS PubMed Google Scholar Sun N, Youle RJ, Finkel T.

Article CAS PubMed PubMed Central Google Scholar Jove M, Portero-Otin M, Naudi A, Ferrer I, Pamplona R. Article CAS PubMed Google Scholar Swerdlow RH. Article CAS PubMed PubMed Central Google Scholar Huo Z, Yu L, Yang J, Zhu Y, Bennett DA, Zhao J. Article CAS PubMed Google Scholar Butterfield DA, Halliwell B.

Article CAS PubMed Google Scholar Sonntag KC, Ryu WI, Amirault KM, Healy RA, Siegel AJ, McPhie DL, et al. Article PubMed PubMed Central Google Scholar Belanger M, Allaman I, Magistretti PJ. Article CAS PubMed Google Scholar Demetrius LA, Magistretti PJ, Pellerin L.

PubMed Google Scholar Stobart JL, Anderson CM. Article CAS PubMed PubMed Central Google Scholar Zulfiqar S, Garg P, Nieweg K. Article CAS PubMed Google Scholar Johnson ECB, Dammer EB, Duong DM, Ping L, Zhou M, Yin L, et al.

Article CAS PubMed PubMed Central Google Scholar McPhie DL, Nehme R, Ravichandran C, Babb SM, Ghosh SD, Staskus A, et al.

Article PubMed PubMed Central Google Scholar Yoshimizu T, Pan JQ, Mungenast AE, Madison JM, Su S, Ketterman J, et al. Article CAS PubMed Google Scholar Swerdlow RH, Khan SM.

Article CAS PubMed Google Scholar Verdin E. Article CAS PubMed Google Scholar Braidy N, Berg J, Clement J, Khorshidi F, Poljak A, Jayasena T, et al.

Article CAS PubMed Google Scholar Lautrup S, Sinclair DA, Mattson MP, Fang EF. Article CAS PubMed PubMed Central Google Scholar Zhu XH, Lu M, Lee BY, Ugurbil K, Chen W.

Article CAS PubMed PubMed Central Google Scholar Arnold SE, Arvanitakis Z, Macauley-Rambach SL, Koenig AM, Wang HY, Ahima RS, et al.

Article CAS PubMed PubMed Central Google Scholar Mracek T, Drahota Z, Houstek J. Article CAS PubMed Google Scholar Reger MA, Henderson ST, Hale C, Cholerton B, Baker LD, Watson GS, et al. Article CAS PubMed Google Scholar Avgerinos KI, Egan JM, Mattson MP, Kapogiannis D. Article PubMed Google Scholar Croteau E, Castellano CA, Richard MA, Fortier M, Nugent S, Lepage M, et al.

Article CAS PubMed Google Scholar Achanta LB, Rae CD. Article CAS PubMed Google Scholar An Y, Varma VR, Varma S, Casanova R, Dammer E, Pletnikova O, et al. Article PubMed Google Scholar Talbot K, Wang HY, Kazi H, Han LY, Bakshi KP, Stucky A, et al. Article CAS PubMed PubMed Central Google Scholar Gabbouj S, Ryhanen S, Marttinen M, Wittrahm R, Takalo M, Kemppainen S, et al.

Article PubMed PubMed Central Google Scholar Nguyen NH, Brathe A, Hassel B. Article CAS PubMed Google Scholar Rust RS Jr. Article CAS PubMed Google Scholar McKenna MC, Waagepetersen HS, Schousboe A, Sonnewald U. Article CAS PubMed Google Scholar Valdebenito R, Ruminot I, Garrido-Gerter P, Fernandez-Moncada I, Forero-Quintero L, Alegria K, et al.

Article CAS PubMed Google Scholar Meyer K, Feldman HM, Lu T, Drake D, Lim ET, Ling KH, et al. Article CAS PubMed PubMed Central Google Scholar Pacholko AG, Wotton CA, Bekar LK. Article PubMed Google Scholar Vlassenko AG, Vaishnavi SN, Couture L, Sacco D, Shannon BJ, Mach RH, et al.

Article CAS PubMed PubMed Central Google Scholar Baik SH, Kang S, Lee W, Choi H, Chung S, Kim JI, et al.

Editor-in-Chief: Alessandro Antonelli Department of Cogjitive and Congitive Medicine Laboratory of Primary Human Cells University of Pisa Pesticide-Free Produce Italy. Conitive Print : ISSN Online : Page: [ - ] Pages: DOI: The interaction between meal timing and light regulates circadian rhythms in mammals and not only determines the sleep-wake pattern but also the activity of the endocrine system. Translational Neurodegeneration cognitivr Energy metabolism and cognitive function cogintive, Article cogniyive Energy metabolism and cognitive function Cite Energy metabolism and cognitive function article. Metrics details. We recently administered combined metabolic Calorie Intake for Recovery CMA to the AD rat model and observed that CMA improves the AD-associated mftabolism parameters in the animals. CMA promotes mitochondrial fatty acid uptake from the cytosol, facilitates fatty acid oxidation in the mitochondria, and alleviates oxidative stress. Here, we designed a randomised, double-blinded, placebo-controlled phase-II clinical trial and studied the effect of CMA administration on the global metabolism of AD patients. One-dose CMA included AD patients received one dose of CMA or placebo daily during the first 28 days and twice daily between day 28 and day Energy metabolism and cognitive function

Energy metabolism and cognitive function -

Alzheimers Dement. Article PubMed PubMed Central Google Scholar. Hardy JA, Higgins GA. Article CAS PubMed Google Scholar. Herrup K.

The case for rejecting the amyloid cascade hypothesis. Nat Neurosci. Sims R, Hill M, Williams J. Amtul Z. Ageing Res Rev. Article PubMed Google Scholar.

Krstic D, Knuesel I. Deciphering the mechanism underlying late-onset Alzheimer disease. Nat Rev Neurol. Wilkins JM, Trushina E. Front Neurol. de la Monte SM. Mattson MP, Arumugam TV.

Hallmarks of brain aging: adaptive and pathological modification by metabolic states. Cell Metab. Article CAS PubMed PubMed Central Google Scholar. Hou Y, Dan X, Babbar M, Wei Y, Hasselbalch SG, Croteau DL, et al.

Ageing as a risk factor for neurodegenerative disease. Mookerjee SA, Gerencser AA, Nicholls DG, Brand MD. Quantifying intracellular rates of glycolytic and oxidative ATP production and consumption using extracellular flux measurements.

J Biol Chem. Bordone MP, Salman MM, Titus HE, Amini E, Andersen JV, Chakraborti B, et al. The energetic brain—a review from students to students. J Neurochem. Dienel GA. Brain glucose metabolism: integration of energetics with function. Physiol Rev.

Sun N, Youle RJ, Finkel T. The mitochondrial basis of aging. Mol Cell. Jove M, Portero-Otin M, Naudi A, Ferrer I, Pamplona R. Metabolomics of human brain aging and age-related neurodegenerative diseases. J Neuropathol Exp Neurol. Swerdlow RH.

J Alzheimers Dis. Huo Z, Yu L, Yang J, Zhu Y, Bennett DA, Zhao J. Neurobiol Aging. Butterfield DA, Halliwell B. Oxidative stress, dysfunctional glucose metabolism and Alzheimer disease. Nat Rev Neurosci. Sonntag KC, Ryu WI, Amirault KM, Healy RA, Siegel AJ, McPhie DL, et al. Sci Rep. Belanger M, Allaman I, Magistretti PJ.

Brain energy metabolism: focus on astrocyte-neuron metabolic cooperation. Demetrius LA, Magistretti PJ, Pellerin L. Front Physiol. PubMed Google Scholar. Stobart JL, Anderson CM. Multifunctional role of astrocytes as gatekeepers of neuronal energy supply.

Front Cell Neurosci. Zulfiqar S, Garg P, Nieweg K. Biol Chem. Johnson ECB, Dammer EB, Duong DM, Ping L, Zhou M, Yin L, et al. Nat Med. McPhie DL, Nehme R, Ravichandran C, Babb SM, Ghosh SD, Staskus A, et al. Oligodendrocyte differentiation of induced pluripotent stem cells derived from subjects with schizophrenias implicate abnormalities in development.

Transl Psychiatry. Yoshimizu T, Pan JQ, Mungenast AE, Madison JM, Su S, Ketterman J, et al. Functional implications of a psychiatric risk variant within CACNA1C in induced human neurons. Mol Psychiatry. Swerdlow RH, Khan SM. Med Hypotheses. Verdin E. Braidy N, Berg J, Clement J, Khorshidi F, Poljak A, Jayasena T, et al.

Role of nicotinamide adenine dinucleotide and related precursors as therapeutic targets for age-related degenerative diseases: rationale, biochemistry, pharmacokinetics, and outcomes.

Antioxid Redox Signal. Lautrup S, Sinclair DA, Mattson MP, Fang EF. Zhu XH, Lu M, Lee BY, Ugurbil K, Chen W. In vivo NAD assay reveals the intracellular NAD contents and redox state in healthy human brain and their age dependences.

Proc Natl Acad Sci USA. Arnold SE, Arvanitakis Z, Macauley-Rambach SL, Koenig AM, Wang HY, Ahima RS, et al. Brain insulin resistance in type 2 diabetes and Alzheimer disease: concepts and conundrums. Mracek T, Drahota Z, Houstek J. The function and the role of the mitochondrial glycerolphosphate dehydrogenase in mammalian tissues.

Biochim Biophys Acta. Reger MA, Henderson ST, Hale C, Cholerton B, Baker LD, Watson GS, et al. Effects of beta-hydroxybutyrate on cognition in memory-impaired adults.

Avgerinos KI, Egan JM, Mattson MP, Kapogiannis D. A systematic review and meta-analysis of human studies. Croteau E, Castellano CA, Richard MA, Fortier M, Nugent S, Lepage M, et al. Achanta LB, Rae CD. beta-Hydroxybutyrate in the brain: one molecule, multiple mechanisms.

Neurochem Res. An Y, Varma VR, Varma S, Casanova R, Dammer E, Pletnikova O, et al. Talbot K, Wang HY, Kazi H, Han LY, Bakshi KP, Stucky A, et al.

J Clin Invest. Gabbouj S, Ryhanen S, Marttinen M, Wittrahm R, Takalo M, Kemppainen S, et al. Front Neurosci. Nguyen NH, Brathe A, Hassel B. Neuronal uptake and metabolism of glycerol and the neuronal expression of mitochondrial glycerolphosphate dehydrogenase.

Rust RS Jr. Enzyme levels in cultured astrocytes, oligodendrocytes and Schwann cells, and neurons from the cerebral cortex and superior cervical ganglia of the rat. McKenna MC, Waagepetersen HS, Schousboe A, Sonnewald U. Neuronal and astrocytic shuttle mechanisms for cytosolic-mitochondrial transfer of reducing equivalents: current evidence and pharmacological tools.

Biochem Pharm. Valdebenito R, Ruminot I, Garrido-Gerter P, Fernandez-Moncada I, Forero-Quintero L, Alegria K, et al. Targeting of astrocytic glucose metabolism by beta-hydroxybutyrate.

J Cereb Blood Flow Metab. Meyer K, Feldman HM, Lu T, Drake D, Lim ET, Ling KH, et al. Cell Rep. Pacholko AG, Wotton CA, Bekar LK. Neurodegener Dis. Vlassenko AG, Vaishnavi SN, Couture L, Sacco D, Shannon BJ, Mach RH, et al. Spatial correlation between brain aerobic glycolysis and amyloid-beta Abeta deposition.

Baik SH, Kang S, Lee W, Choi H, Chung S, Kim JI, et al. Zhao J, Davis MD, Martens YA, Shinohara M, Graff-Radford NR, Younkin SG, et al. Hum Mol Genet. Mayer PR, Huang N, Dewey CM, Dries DR, Zhang H, Yu G.

Expression, localization, and biochemical characterization of nicotinamide mononucleotide adenylyltransferase 2. Raffaelli N, Sorci L, Amici A, Emanuelli M, Mazzola F, Magni G. Identification of a novel human nicotinamide mononucleotide adenylyltransferase.

Biochemical Biophysical Res Commun. Article CAS Google Scholar. Download references. This study was supported by the Program for Neuropsychiatric Research, McLean Hospital BMC and NRF grants R1A2B, R1H1A, IITP grant from MSIT HS.

The authors want to thank Ryan A. Healy, Yoon Lee, Wang Liao, Miseon Oh, and Jinfeng Xuan for technical, and Suzanne Babb, Fontini Savvides, and Tamare V. Adrien for administrative help. The authors also thank Dr. Rosario Sanchez-Pernaute and Dr.

Christian LeGuern for critically reading the manuscript and helpful discussions. Department of Psychiatry, Harvard Medical School, Belmont, MA, USA. Woo-In Ryu, Mariana K.

Bormann, Minqi Shen, Brent Forester, Kai-C. Basic Neuroscience Division, Harvard Medical School, Belmont, MA, USA. Program for Neuropsychiatric Research, McLean Hospital, Harvard Medical School, Belmont, MA, USA. Bormann, Minqi Shen, Kai-C. Department of Immunology, Tufts University School of Medicine, Boston, MA, USA.

Mood Disorders Division and Geriatric Psychiatry Research Program, McLean Hospital, Harvard Medical School, Belmont, MA, USA. Department of Molecular and Life Sciences, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan, South Korea.

Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA. Chen Y, Guillemin GJ. Kynurenine pathway metabolites in humans: disease and healthy States. Int J Tryptophan Res. Tapia-Rojas C, Lindsay CB, Montecinos-Oliva C, Arrazola MS, Retamales RM, Bunout D, et al.

Wang Q, Liu D, Song P, Zou M-H. Tryptophan-kynurenine pathway is dysregulated in inflammation, and immune activation. Front Biosci Landmark Ed. Sas K, Szabó E, Vécsei L. Mitochondria, oxidative stress and the kynurenine system, with a focus on ageing and neuroprotection.

Fazio F, Carrizzo A, Lionetto L, Damato A, Capocci L, Ambrosio M, et al. Vasorelaxing action of the kynurenine metabolite, xanthurenic acid: the missing link in endotoxin-induced hypotension?

Front Pharmacol. Shi Y, Liu Z, Shen Y, Zhu H. Front Cell Neurosci. Haukka JK, Sandholm N, Forsblom C, Cobb JE, Groop P-H, Ferrannini E. Metabolomic profile predicts development of microalbuminuria in individuals with type 1 diabetes.

Sci Rep. Mielke MM, Haughey NJ, Bandaru VVR, Weinberg DD, Darby E, Zaidi N, et al. Hasin Y, Seldin M, Lusis A.

Multi-omics approaches to disease. Genome Biol. Hutter-Schmid B, Humpel C. Alpha-smooth muscle actin mRNA and protein are increased in isolated brain vessel extracts of Alzheimer mice. Åberg D, Johansson P, Isgaard J, Wallin A, Johansson JO, Andreasson U, et al.

Lidstone SC. Handb Exp Pharmacol. de la Fuente-Fernández R, Ruth TJ, Sossi V, Schulzer M, Calne DB, Stoessl AJ. Manza P, Wiers CE, Shokri-Kojori E, Kroll D, Feldman D, Schwandt M, et al.

Brain network segregation and glucose energy utilization: relevance for age-related differences in cognitive function. Cereb Cortex. Braun U, Schäfer A, Walter H, Erk S, Romanczuk-Seiferth N, Haddad L, et al. Dynamic reconfiguration of frontal brain networks during executive cognition in humans. Proc Natl Acad Sci USA.

Błaszczyk JW. Energy metabolism decline in the aging brain-pathogenesis of neurodegenerative disorders. Perez Ortiz JM, Swerdlow RH. Br J Pharmacol. Fang EF, Lautrup S, Hou Y, Demarest TG, Croteau DL, Mattson MP, et al.

Trends Mol Med. Hou Y, Lautrup S, Cordonnier S, Wang Y, Croteau DL, Zavala E, et al. Moutinho M, Puntambekar SS, Tsai AP, Coronel I, Lin PB, Casali BT, et al.

Sci Transl Med. Schöndorf DC, Ivanyuk D, Baden P, Sanchez-Martinez A, De Cicco S, Yu C, et al. Cell Rep. Caponio D, Veverová K, Zhang SQ, Shi L, Wong G, Vyhnalek M, Fang EF. Aging Brain. Haxaire C, Turpin FR, Potier B, Kervern M, Sinet PM, Barbanel G, et al. Reversal of age-related oxidative stress prevents hippocampal synaptic plasticity deficits by protecting D-serine-dependent NMDA receptor activation.

Le Douce J, Maugard M, Veran J, Matos M, Jégo P, Vigneron PA, et al. Gil-Martínez A-L, Cuenca L, Sánchez C, Estrada C, Fernández-Villalba E, Herrero MT. Effect of NAC treatment and physical activity on neuroinflammation in subchronic Parkinsonism; is physical activity essential? J Neuroinflammation.

Jhang KA, Park JS, Kim HS, Chong YH. Sulforaphane rescues amyloid-β peptide-mediated decrease in MerTK expression through its anti-inflammatory effect in human THP-1 macrophages.

Tondo G, Perani D, Comi C. TAM receptor pathways at the crossroads of neuroinflammation and neurodegeneration. Dis Markers. Chiang HC, Wang L, Xie Z, Yau A, Zhong Y. PI3 kinase signaling is involved in Abeta-induced memory loss in Drosophila.

Wang L, Chiang H-C, Wu W, Liang B, Xie Z, Yao X, et al. Epidermal growth factor receptor is a preferred target for treating Amyloid-β—induced memory loss. Owens T, Renno T, Taupin V, Krakowski M.

Inflammatory cytokines in the brain: does the CNS shape immune responses? Immunol Today. Kordula T, Rydel RE, Brigham EF, Horn F, Heinrich PC, Travis J. Oncostatin M and the interleukin-6 and soluble interleukin-6 receptor complex regulate alpha1-antichymotrypsin expression in human cortical astrocytes.

J Biol Chem. Castell JV, Gómez-Lechón MJ, David M, Andus T, Geiger T, Trullenque R, et al. Interleukin-6 is the major regulator of acute phase protein synthesis in adult human hepatocytes.

FEBS Lett. Abraham CR, Selkoe DJ, Potter H. Selkoe DJ. Tu R, Grover HM, Kotra LP. Peptidyl arginine deiminases and neurodegenerative diseases. Curr Med Chem. Acharya NK, Nagele EP, Han M, Coretti NJ, DeMarshall C, Kosciuk MC, et al. Neuronal PAD4 expression and protein citrullination: possible role in production of autoantibodies associated with neurodegenerative disease.

J Autoimmun. Hamaguchi T, Okino S, Sodeyama N, Itoh Y, Takahashi A, Otomo E, et al. Association of a polymorphism of the transforming growth factor-beta1 gene with cerebral amyloid angiopathy. J Neurol Neurosurg Psychiatry. Lahiri DK, Ge YW. Ann N Y Acad Sci. Luedecking EK, DeKosky ST, Mehdi H, Ganguli M, Kamboh MI.

Hum Genet. Burton T, Liang B, Dibrov A, Amara F. Transforming growth factor-beta-induced transcription of the Alzheimer beta-amyloid precursor protein gene involves interaction between the CTCF-complex and Smads.

Biochem Biophys Res Commun. Lesné S, Docagne F, Gabriel C, Liot G, Lahiri DK, Buée L, et al. Transforming growth factor-beta 1 potentiates amyloid-beta generation in astrocytes and in transgenic mice. Ehrenreich H, Hasselblatt M, Knerlich F, von Ahsen N, Jacob S, Sperling S, et al.

A hematopoietic growth factor, thrombopoietin, has a proapoptotic role in the brain. Woo HG, Chang Y, Ryu DR, Song TJ. Plasma Klotho concentration is associated with the presence, burden and progression of cerebral small vessel disease in patients with acute ischaemic stroke.

PLoS ONE. Paroni G, Panza F, De Cosmo S, Greco A, Seripa D, Mazzoccoli G. Mol Neurobiol. Yang K, Yang Z, Chen X, Li W. Brain Res Bull. Mohri I, Kadoyama K, Kanekiyo T, Sato Y, Kagitani-Shimono K, Saito Y, et al. Hematopoietic prostaglandin D synthase and DP1 receptor are selectively upregulated in microglia and astrocytes within senile plaques from human patients and in a mouse model of Alzheimer disease.

J Neuropathol Exp Neurol. Tarkowski E, Issa R, Sjögren M, Wallin A, Blennow K, Tarkowski A, et al. Chao CC, Ala TA, Hu S, Crossley KB, Sherman RE, Peterson PK, et al.

Clin Diagn Lab Immunol. Royall DR, Palmer RF. Taliyan R, Chandran SK, Kakoty V. Curr Pharm Des. Brombo G, Bonetti F, Ortolani B, Morieri ML, Bosi C, Passaro A, et al. Salminen A, Jouhten P, Sarajärvi T, Haapasalo A, Hiltunen M. Neurochem Int. Johnson ECB, Dammer EB, Duong DM, Ping L, Zhou M, Yin L, et al.

Nat Med. Martire S, Fuso A, Mosca L, Forte E, Correani V, Fontana M, et al. Mech Ageing Dev. Bavarsad Shahripour R, Harrigan MR, Alexandrov AV. N-acetylcysteine NAC in neurological disorders: mechanisms of action and therapeutic opportunities. Brain Behav.

Tardiolo G, Bramanti P, Mazzon E. Overview on the effects of N-acetylcysteine in neurodegenerative diseases. Hara Y, McKeehan N, Dacks PA, Fillit HM. Evaluation of the neuroprotective potential of N-acetylcysteine for prevention and treatment of cognitive aging and dementia.

J Prev Alzheimers Dis. Kepka A, Ochocinska A, Borzym-Kluczyk M, Skorupa E, Stasiewicz-Jarocka B, Chojnowska S, et al. Flanagan JL, Simmons PA, Vehige J, Willcox MD, Garrett Q. Role of carnitine in disease. Nutr Metab. Cristofano A, Sapere N, La Marca G, Angiolillo A, Vitale M, Corbi G, et al.

Chen N, Yang M, Zhou M, Xiao J, Guo J, He L. L-carnitine for cognitive enhancement in people without cognitive impairment. Cochrane Database Syst Rev. Wollen KA. Altern Med Rev. Gavrilova SI, Kalyn Ia B, Kolykhalov IV, Roshchina IF, Selezneva ND.

Zh Nevrol Psikhiatr Im S S Korsakova. Ting KK, Brew B, Guillemin G. Neurotox Res. Stress-related regulation of the kynurenine pathway: relevance to neuropsychiatric and degenerative disorders. Guillemin GJ, Smythe GA, Veas LA, Takikawa O, Brew BJ. A beta 1—42 induces production of quinolinic acid by human macrophages and microglia.

Wu W, Nicolazzo JA, Wen L, Chung R, Stankovic R, Bao SS, et al. Bryll A, Skrzypek J, Krzyściak W, Szelągowska M, Śmierciak N, Kozicz T, et al. Oxidative-antioxidant imbalance and impaired glucose metabolism in schizophrenia. N Engl J Med. Griffin JWD, Bradshaw PC.

Braga TT, Forni MF, Correa-Costa M, Ramos RN, Barbuto JA, Branco P, et al. Soluble uric acid activates the NLRP3 inflammasome.

Hansmannel F, Sillaire A, Kamboh MI, Lendon C, Pasquier F, Hannequin D, et al. Chouraki V, Preis SR, Yang Q, Beiser A, Li S, Larson MG, et al. Graham SF, Chevallier OP, Elliott CT, Hölscher C, Johnston J, McGuinness B, et al. Wang G, Zhou Y, Huang FJ, Tang HD, Xu XH, Liu JJ, et al.

J Proteome Res. Chew H, Solomon VA, Fonteh AN. Front Physiol. van der Velpen V, Teav T, Gallart-Ayala H, Mehl F, Konz I, Clark C, et al. Alzheimers Res Ther. Li D, Misialek JR, Boerwinkle E, Gottesman RF, Sharrett AR, Mosley TH, et al.

Han X, Rozen S, Boyle SH, Hellegers C, Cheng H, Burke JR, et al. Ciavardelli D, Piras F, Consalvo A, Rossi C, Zucchelli M, Di Ilio C, et al. Oberacher H, Arnhard K, Linhart C, Diwo A, Marksteiner J, Humpel C. Simpson BN, Kim M, Chuang YF, Beason-Held L, Kitner-Triolo M, Kraut M, et al. Blood metabolite markers of cognitive performance and brain function in aging.

J Cereb Blood Flow Metab. Whiley L, Sen A, Heaton J, Proitsi P, García-Gómez D, Leung R, et al. Paglia G, Stocchero M, Cacciatore S, Lai S, Angel P, Alam MT, et al. Castor KJ, Shenoi S, Edminster SP, Tran T, King KS, Chui H, et al. Kurbatova N, Garg M, Whiley L, Chekmeneva E, Jiménez B, Gómez-Romero M, et al.

Sci Reports. Passi S, Picardo M, De Luca C, Nazzaro-Porro M, Rossi L, Rotilio G Saturated dicarboxylic acids as products of unsaturated fatty acid oxidation.

Biochimica et Biophysica Acta BBA - Lipids Lipid Metabolism ; 2 —8. Birzniece V, Bäckström T, Johansson IM, Lindblad C, Lundgren P, Löfgren M, et al. Neuroactive steroid effects on cognitive functions with a focus on the serotonin and GABA systems.

Brain Res Rev. Wang Y, Li J, Matye D, Zhang Y, Dennis K, Ding WX, et al. Bile acids regulate cysteine catabolism and glutathione regeneration to modulate hepatic sensitivity to oxidative injury.

JCI Insight. Download references. The authors would like to thank Metabolon Inc. Durham, USA for the generation of metabolomics data and ChromaDex Inc.

Irvine, CA, USA for providing nicotinamide riboside. AM and HY acknowledge support from the PoLiMeR Innovative Training Network Marie Sklodowska-Curie Grant Agreement No. The computations were performed on resources provided by SNIC through Uppsala Multidisciplinary Center for Advanced Computational Science UPPMAX under Project sllstore Open access funding provided by Royal Institute of Technology.

This work was financially supported by ScandiBio Therapeutics and Knut and Alice Wallenberg Foundation Department of Neurology and Neuroscience, Faculty of Medicine, Alanya Alaaddin Keykubat University, Antalya, Turkey.

Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden. Department of Neurology, Faculty of Medicine, Istanbul Medipol University, Istanbul, Turkey.

Functional Imaging and Cognitive-Affective Neuroscience Lab, Istanbul Medipol University, Istanbul, Turkey. Department of Medical Pharmacology, Faculty of Medicine, Atatürk University, Erzurum, Turkey. Department of Pathology, Veterinary Faculty, Ataturk University, Erzurum, Turkey.

Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical University, Erzurum, Turkey. Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden. Department of Medical Biology, Faculty of Medicine, Atatürk University, Erzurum, Turkey.

Department of Molecular and Clinical Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden. You can also search for this author in PubMed Google Scholar. Research project: conception: A. and B. and A. Y, and review and critique: A.

and M. All authors read and approved the final manuscript. Correspondence to Mathias Uhlén or Adil Mardinoglu. The ethics committee approved the study of Istanbul Medipol University, Istanbul, Turkey Date Written informed consent for publication was obtained from all participants before initiating any trial-related procedures.

AM, JB and MU are the founder and shareholders of ScandiBio Therapeutics. The other authors declare no competing interests. Collection of samples of CMA and placebo groups and the measured values of clinical indicators before and after treatment. Consort flow diagram. Diagram shows the progress through the phases of the parallel randomisation of drug and placebo groups.

Plasma metabolomics data for each patient before and after treatment and statistical analysis of plasma metabolites between different visits or groups.

The association between the plasma level of the four supplements serine, carnitine, cysteine and nicotinamide riboside with the plasma levels of other metabolites. Plasma proteomics data were generated with the Olink cardiometabolic, inflammation, neurology and oncology panels for each patient before and after treatment and statistical analysis of plasma proteins between different visits or groups.

Multi-Omics Network Data, including edges and nodes information. Open Access This article is licensed under a Creative Commons Attribution 4.

The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.

Reprints and permissions. Yulug, B. et al. Transl Neurodegener 12 , 4 Download citation. Received : 24 October Accepted : 09 January Published : 26 January Anyone you share the following link with will be able to read this content:.

Sorry, a shareable link is not currently available for this article. Provided by the Springer Nature SharedIt content-sharing initiative. Skip to main content. Search all BMC articles Search. Download PDF. Methods Here, we designed a randomised, double-blinded, placebo-controlled phase-II clinical trial and studied the effect of CMA administration on the global metabolism of AD patients.

Conclusion Our results indicate that treatment of AD patients with CMA can lead to enhanced cognitive functions and improved clinical parameters associated with phenomics, metabolomics, proteomics and imaging analysis.

Materials and methods Clinical trial design and oversight Patients for this randomised, parallel-group, two-arm, double-blinded, placebo-controlled, phase 2 study were recruited at the Faculty of Medicine, Alanya Alaaddin Keykubat University, Antalya, Turkey and Faculty of Medicine, Istanbul Medipol University, Istanbul, Turkey.

Randomisation, interventions, and follow-up Patients were randomly assigned to receive CMA or placebo Outcomes The primary endpoint in the original protocol was to assess the clinical efficacy of CMA in AD patients. Proteomics analysis Plasma levels of proteins were determined with the Olink panels Olink Bioscience, Uppsala, Sweden.

Untargeted metabolomics analysis Plasma samples were collected on days 0 and 84 for untargeted metabolite profiling by Metabolon Durham, NC. Determination of clinical variables informing response to CMA administration The patient groups with low and high levels of each clinical parameter were established based on the median score for that clinical parameter across all patients on day 0.

Magnetic resonance imaging MRI parameters and analysis Among the entire patient cohort, 40 MRI-compatible patients, 29 in the CMA group and 11 in the placebo group, were recruited for the structural MRI study.

Image processing To obtain hippocampal subfield measurements, each T1 image was processed using FreeSurfer version 7. Statistical analysis Paired t -test was used to identify the differences in clinical parameters between time points, and one-way ANOVA was used to find the shifts between CMA and placebo groups at each time point.

Generation of multi-omics network A multi-omics correlation network was generated based on all patients' clinical parameters, serum chemistry, metabolomics, and proteomics data, following the multi-omics network generation pipeline from iNetModels [ 49 ]. RESULTS CMA improves cognition and clinical parameters in AD patients To test the effect of CMA in AD patients, we performed a double-blinded, randomised, placebo-controlled phase 2 study and screened 89 adults diagnosed with AD.

Full size image. Table 2 List of adverse effects Full size table. Table 3 Differences in ADAS-Cog, ADCS-ADL and MMSE scores in the CMA and placebo groups Full size table. Discussion Our results suggested that oral administration of CMA for 84 days has a considerable effect on cognitive function in AD patients based on ADAS-Cog scores.

Conclusions The present phase 2 clinical study suggested that oral administration of CMA improves metabolic alterations in AD patients, and that CMA is safe and well-tolerated, with no major safety concerns identified.

Availability of data and materials The data supporting the findings of this study are available in Supplementary Material. References Trujillo-Estrada L, Jimenez S, De Castro V, Torres M, Baglietto-Vargas D, Moreno-Gonzalez I, et al.

Article Google Scholar Nunomura A, Perry G. Article Google Scholar Lam S, Bayraktar A, Zhang C, Turkez H, Nielsen J, Boren J, et al. Article CAS Google Scholar Mielke JG, Taghibiglou C, Liu L, Zhang Y, Jia Z, Adeli K, et al.

Article CAS Google Scholar Arnold SE, Arvanitakis Z, Macauley-Rambach SL, Koenig AM, Wang HY, Ahima RS, et al. Article CAS Google Scholar Wei Y, Han C, Wang Y, Wu B, Su T, Liu Y, et al. Article CAS Google Scholar Masciopinto F, Di Pietro N, Corona C, Bomba M, Pipino C, Curcio M, et al.

Article CAS Google Scholar Wong LR, Wong P, Ho PC. Article CAS Google Scholar Makin S. Article CAS Google Scholar Kuehn BM.

Article Google Scholar Chen Z, Zhong C. Article CAS Google Scholar Powell CL, Davidson AR, Brown AM. Article CAS Google Scholar Drulis-Fajdasz D, Gizak A, Wójtowicz T, Wiśniewski JR, Rakus D. Article Google Scholar Wang W, Zhao F, Ma X, Perry G, Zhu X. Article CAS Google Scholar Liang WS, Reiman EM, Valla J, Dunckley T, Beach TG, Grover A, et al.

Article CAS Google Scholar Lin MT, Beal MF. Article CAS Google Scholar Beck SJ, Guo L, Phensy A, Tian J, Wang L, Tandon N, et al. Article CAS Google Scholar Kerr JS, Adriaanse BA, Greig NH, Mattson MP, Cader MZ, Bohr VA, et al. Article CAS Google Scholar Xie C, Zhuang X-X, Niu Z, Ai R, Lautrup S, Zheng S, et al.

Article CAS Google Scholar Lautrup S, Sinclair DA, Mattson MP, Fang EF. Article CAS Google Scholar Liu D, Pitta M, Jiang H, Lee JH, Zhang G, Chen X, et al. Article CAS Google Scholar Fang EF, Hou Y, Palikaras K, Adriaanse BA, Kerr JS, Yang B, et al.

Article CAS Google Scholar Cummings J, Lee G, Ritter A, Sabbagh M, Zhong K. Google Scholar Pan X, Nasaruddin MB, Elliott CT, McGuinness B, Passmore AP, Kehoe PG, et al. Article CAS Google Scholar Hou Y, Wei Y, Lautrup S, Yang B, Wang Y, Cordonnier S, et al.

Article Google Scholar Toledo JB, Arnold M, Kastenmüller G, Chang R, Baillie RA, Han X, et al. Article Google Scholar Sancesario GM, Bernardini S. Article CAS Google Scholar Jagust W. Article CAS Google Scholar De Santi S, de Leon MJ, Rusinek H, Convit A, Tarshish CY, Roche A, et al.

Article Google Scholar Nagata T, Shinagawa S, Ochiai Y, Aoki R, Kasahara H, Nukariya K, et al. Article Google Scholar Foster NL, Chase TN, Fedio P, Patronas NJ, Brooks RA, Di Chiro G. Article CAS Google Scholar Greene SJ, Killiany RJ.

Article Google Scholar Zhang C, Bjornson E, Arif M, Tebani A, Lovric A, Benfeitas R, et al. These findings indicate that lower energy generation rates might disrupt the long distance and large-scale neuronal communications—thereby leading to brain disorders.

Our study revealed the general principle of brain energy-activity organization and suggests that the metabolism-neural synchrony pathway could be a new potential treatment target for psychotic disorders. The highlighted cortical regions above are the nodes of a large-scale brain network.

These areas usually show synchronized neural activity or in other words, functional connectivity FC , as they work towards a common goal. In this study, a positive correlation was found between the strength of this synchronization or connectivity and the rate of the creatine kinase reaction, which would be critical for orchestrating oscillatory states, and enhancing the fidelity of information processing for better executive and cognitive function.

Xiaopeng Song is a postdoctoral fellow in the lab of Dr. Fei Du at McLean Hospital, Harvard Medical School. Learn more in the original research article: Bioenergetics and abnormal functional connectivity in psychotic disorders. Song X, Chen X, Yuksel C, Yuan J, Pizzagalli DA, Forester B, Öngür D, Du F.

Worldwide, Almond industry 55 million people had prevalent Thermogenic metabolism support inwhich is expected cogitive triple by Almond industry, especially in low- and middle-income countries ccognitive 1]. Lacking timely diagnosis and limited effective treatment for dementia make identifying risk factors Caloric needs for healthy aging cohnitive its early prevention, among which dietary factors have received increasing attention [ ccognitive. Recently, accumulating evidence from population-based Eergy has linked the temporal patterns of energy intake TPEIusually defined as the temporal distribution of energy intake during a day, to mortality and metabolic diseases [ 2], such as diabetes and hypertension. In vitro and in vivo studies also revealed that meal timing could drive metabolic alterations and circadian regulation [ 3], and disrupted meal timing altered the peripheral circadian clocks in the hippocampus and consequently affected cognitive function [ 4]. However, population-level evidence on the association between the TPEI and cognitive function remains lacking. We thus aimed to examine this relationship in the China Health and Nutrition Survey from toa community-based cohort study with national representativeness [ 5]. Oxford University Press is a department of the University of Oxford.

Video

5X Your BRAIN CELLS

Author: Tygot

2 thoughts on “Energy metabolism and cognitive function

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com