Recommended related articles

Home
Brain metabolism--seminal article, 2017
BDNF-3-hydroxybuytrate-regulation-Seminal-2016
Fasting promotes-mitophagy-hepatocytes-etc-2998
Page Title
New Page Title
BDNF-3-hydroxybuytrate-regulation-Seminal-2016

Article on fatty acids producing 3-hydroxybuytrate which causes adaptive responses of neurons to fasting, exercise, and ketogenic diets.

 

 

3-Hydroxybutyrate regulates energy metabolism and induces BDNF expression in cerebral cortical neurons

Authors

Krisztina Marosi, Sang Woo Kim, Keelin Moehl, Morten Scheibye-Knudsen, Aiwu Cheng, Roy Cutler, Simonetta Camandola, Mark P. Mattson  -references of his articles 2004 and 2014

Published Date

14 November 2016

 

Abstract

During fasting and vigorous exercise, a shift of brain cell energy substrate utilization from glucose to the ketone 3-hydroxybutyrate (3OHB) occurs. Studies have shown that 3OHB can protect neurons against excitotoxicity and oxidative stress, but the underlying mechanisms remain unclear.  Neurons maintained in the presence of 3OHB exhibited increased oxygen consumption and ATP production, and an elevated NAD+/NADH ratio. We found that 3OHB metabolism increases mitochondrial respiration which drives changes in expression of brain-derived neurotrophic factor (BDNF) in cultured cerebral cortical neurons. The mechanism by which 3OHB induces Bdnf gene expression involves generation of reactive oxygen species, activation of the transcription factor NF-κB, and activity of the histone acetyltransferase p300/EP300. Because BDNF plays important roles in synaptic plasticity and neuronal stress resistance, our findings suggest cellular signaling mechanisms by which 3OHB may mediate adaptive responses of neurons to fasting, exercise, and ketogenic diets.

 

image

 

Abbreviations used

  3OHB

  3-hydroxybutyrate

  AD

  Alzheimer's disease

  BDNF

  brain-derived neurotrophic factor

  CREB

  cAMP response element-binding protein

  CSA

  cyclosporin A

  ECAR

  extracellular acidification rate

  ETC.

  electron transport chain

  HAT

  histone acetyltransferase

  HD

  Huntington's disease

  KA

  kainic acid

  MCT2

  monocarboxylic acid transporter 2

  NAC

  N-acetylcysteine

  NAD+

  nicotinamide adenine dinucleotide oxidized

  NADH

  nicotinamide adenine dinucleotide reduced

  NF-kB

  nuclear factor kappa B

  OCR

  oxygen consumption rate

  PD

  Parkinson's disease

  ROS

  reactive oxygen species

  SIRT1

  sirtuin 1

  SIRT3

  sirtuin 3

  SOD2

  superoxide dismutase 2

Bioenergetic challenges such as vigorous exercise and intermittent fasting have been shown to enhance synaptic plasticity and neuronal stress resistance, and can protect neurons against dysfunction and degeneration in animal models of stroke, Alzheimer's disease (AD), Huntington's disease (HD) and Parkinson's disease (PD) (for review see Mattson 2012; Voss et al. 2013; Longo and Mattson 2014; Neufer et al. 2015). Studies of responses of brain cells to exercise and intermittent fasting suggest that both types of energetic challenge up-regulate signaling pathways involved in cellular adaptations to stress and neuroplasticity. Examples include increased expression of genes encoding brain-derived neurotrophic factor (BDNF) (Marosi and Mattson 2014), DNA repair enzymes (Yang et al. 2014), protein deacetylases sirtuin 1 and sirtuin 3 (Cheng et al. 2012; Cheng et al. 2016; Wang et al. 2013), and antioxidant enzymes (Marosi et al. 2012).

Emerging evidence suggests that beneficial effects of exercise and fasting on the brain are mediated, at least in part, by signaling molecules released into the blood from peripheral organs. For example, levels of circulating insulin-like growth factor 1 increase in response to exercise, and insulin-like growth factor 1 can enter the brain where it contributes to enhancement of cognitive function (Trejo et al. 2008). Recently, it was reported that a protein called irisin is released into the blood from muscle cells in response to exercise, and can then cross the blood–brain barrier and induce BDNF expression (Wrann et al. 2013). BDNF plays roles in the development, maintenance, and plasticity of the central and peripheral nervous systems (Chao 2006). It promotes neurogenesis, enhances neurite outgrowth and synaptogenesis, and can prevent apoptosis (Mattson et al. 2004; Marosi and Mattson 2014). Decrements in BDNF levels occur in vulnerable brain regions in several disorders including Alzheimer's disease (AD), HD, and major depression (Zuccato and Cattaneo 2009). Interventions that increase BDNF levels or activate TrkB have been shown to ameliorate clinical symptoms and underlying cellular and molecular neuropathologies in mouse models of AD, PD, HD, stroke, and depression (Levivier et al. 1995; Gobbo and O'Mara 2004; Spires et al. 2004; Duman et al. 2008; Devi 2012).

Whether factors produced in the periphery mediate beneficial effects of fasting on neuroplasticity and neuronal stress resistance remains unknown. A major physiological response to fasting and vigorous exercise is the mobilization of fatty acids from adipose cells and the hepatic production of ketone bodies from those fatty acids (Longo and Mattson 2014). The major ketone, 3-hydroxybutyrate (3OHB), provides an energy source for neurons to sustain their function when glucose levels are reduced (Maalouf et al. 2009; Chowdhury et al. 2014). Ketogenic diets (McNally and Hartman 2012) and fasting (Bruce-Keller et al., 1999) can protect hippocampal neurons against seizure-induced damage. Fasting and 3OHB can also counteract disease processes and improve functional outcome in animal models of Alzheimer's and Parkinson's diseases, stroke, and traumatic brain injury (Duan and Mattson 1999; Anson et al. 2003; Arumugam et al. 2010; Kashiwaya et al. 2013; Prins and Matsumoto 2014; Greco et al. 2015; Yin et al. 2015). Clinical data also suggest that ketone bodies can have a therapeutic benefit in neurodegenerative diseases such as AD and PD (Reger et al. 2004; Vanitallie et al. 2005).

In addition to being a source of acetyl coenzyme A for neuronal energy metabolism, recent findings suggest that 3OHB influences certain cellular signaling pathways. For example, 3OHB can inhibit or activate protein deacetylases (Newman and Verdin 2014; Scheibye-Knudsen et al. 2014), and can inhibit mitochondrial membrane permeability transition pore opening (Kim et al. 2015). While multiple effects of energy restriction and exercise on neuroplasticity and neuroprotection have been linked to BDNF signaling (Marosi and Mattson 2014), the mechanisms by which these bioenergetic challenges induce BDNF signaling and adaptive stress response pathways remain unknown. Here, we show that 3OHB changes neuronal bioenergetics by increasing mitochondrial respiration that results in increased BDNF expression.

Methods

Animals

All animal procedures were approved by the Animal Care and Use Committee of the National Institute on Aging Intramural Research Program. Male C57BL/6 mice (purchased from Jackson Laboratories, Bar Harbor, ME, USA) were housed individually with (n = 8) or without (n = 8) running wheels beginning at 4 months of age. The mice were provided food and water ad libitum, and were maintained on a 12-h light/12-h dark cycle. After 6 weeks, mice in each group were killed, their brains were removed, flash frozen, and stored at −80°C. The BDNF protein levels in the brain were detected using a BDNF ELISA (Promega, Madison, WI, USA). Glucose and 3-hydroxybutyrate concentrations in the plasma were quantified using a Roche Cobas Fara II analyzer (Roche Diagnostic Systems; Montclair, NJ, USA).

Cell culture and experimental treatments

Cultures of cerebral cortical or hippocampal neurons were prepared from Sprague–Dawley rat embryos (male and female) at 18 days of gestation using methods similar to those described previously (Glazner et al. 2000). Dissociated cells were seeded into polyethyleneimine-coated plastic dishes or glass coverslips in MEM medium supplemented with 10% fetal bovine serum at a density of 80 000 cells/cm2. After cells attached to the substrate, the medium was replaced with Neurobasal medium containing 5% B27 minus antioxidants, 1% GlutaMAX™, and 1% Anti-Anti (Gibco, Rockville, MD, USA). Experiments were initiated on culture day 10. 3-D-hydroxybutyrate sodium salt and N-acetyl cysteine, cyclosporin A, and sodium butyrate were obtained from Sigma, St Louis, MO, USA. AR-C155858 was obtained from Tocris Bioscience (Bristol, United Kingdom). SN-50 was purchased from Santa Cruz Biotechnology, Santa Cruz, CA, USA.

Western blotting and immunoprecipitation

Cells were lysed in radioimmunoprecipitation assay buffer-containing protease inhibitors (Roche) and phosphatase inhibitors (Sigma). Lysates were sonicated for 1 min and centrifuged at 14 000 g for 10 min at 4°C. Twenty micrograms of proteins were resolved in 4–10% NuPAGE Bis-Tris Mini gels (Invitrogen, Carlsbad, CA, USA) and electrophoretically transferred to a nitrocellulose membrane (Invitrogen). The unspecific binding sites were blocked in blocking solution containing 5% milk or bovine serum albumin for 1 h at 23°C. Then, the membranes were incubated overnight in primary antibodies followed by incubation in secondary antibodies for 1 h at 23°C. The reaction products in the membranes were visualized using an enhanced chemiluminescence Western Blot Detection Kit (Thermo Scientific, Grand Island, NY USA). For immunoprecipitation experiments, 500 μg of total protein lysates was incubated overnight at 4°C with anti-Flag agarose beads (Santa Cruz). The primary antibodies those against NF-κB p65 (Thermo Scientific); p300 (Abcam); superoxide dismutase 2 (SOD2) (Santa Cruz); total OXPHOS (Abcam, Cambridge, MA USA); and β-actin (Sigma-Aldrich, St. Louis, MO. USA). The densities of protein bands were quantified using ImageJ software (National Institutes of Health, Bethesda, MD, USA) and normalized to the β-actin band intensity for the same sample.

BDNF ELISA

The BDNF concentrations in the primary neuronal samples were determined by enzyme-linked immunosorbent assay (ELISA) (Promega). The assay was performed according to the manufacturer's instructions.

Quantitative reverse transcriptase PCR amplification

Total RNAs were isolated from lysates of cultured neurons using the RNAeasy purification kit (Qiagen, Valencia, CA, USA). Isolated RNA was reverse transcribed into cDNA by following the protocol from SuperScript III First-Strand Synthesis System (Invitrogen). Real-time polymerase chain reaction (PCR) was performed using a PTC-200 Thermal Cycler (Bio-Rad Laboratories, Hercules, CA, USA) using 2X SYBR Green Master Mix (Applied Biosystems, ThermoFisher Scientific, Grand Island, NY USA). Cycle parameters for all amplifications were as follows: 1 cycle of 95°C for 5 min, 40 cycles of 95°C for 30 s, and 72°C for 1 min, followed by melt-curve analysis (55°C+ for 10 s × 80 cycles). The mRNA level of the targeted genes was normalized to the level of β-actin mRNA. Primers specific for the following mRNAs were used: mct2: sense, 5′-GGCCTTCGGTAGGATTAATAG-3′, antisense, 5′-ATGCCTGATGATAACACGACT-3′; bdnf: sense, 5′- CTGCGCCCATGAAAGAAG-3′, antisense, 5′-CCAGCAGCTCTTCGATCA-3′; β-actin sense, 5′- TCATGAAGTGTGACGTTGACATCCGTAAAG-3′, antisense, 5′-CCTAGAACGATTTCGGGTGCACGATGGAGG-3′, tfam, sense: 5' GAAAGCACAAATCAAGAGGAG-3′, antisense: 5′-ATCATGAGCTGCTTTTCACAG-3′, pgc1α, sense: 5′-GTGCAGCCAAGACTCTGTATGG-3′, antisense: 5′GTCCAGGTCATTCACTGCTTTTCCATCAAGTTC-3′, nrf1, sense: 5′-TTACTCTCTGCTGTGGCTGATGG-3′, antisense: 5′-CCTCTGATGCTTGCGTCGTCT-3′, nrf2, sense: 5′-TTCCTCTGCTGCCATTAGTCAGTC-3′, antisense: 3′-GCTCTTCCATTTCCGAGTCACTG-3′

Mitochondrial DNA relative copy number

DNA from primary cortical neurons was extracted using a QIAamp DNA blood extraction kit (Qiagen). Mitochondrial DNA (mtDNA) copy number was measured using real-time quantitative PCR. One primer pair specific for the mtDNA (12sR sense: 5′CTCAAGACGCCTTCGCTAG′, antisense: 5′-CGTATGACCGCGGTGGCT-3′,) designed for relative quantification for mtDNA copy number. Twenty ng of DNA was added to each reaction. Threshold cycle value differences were used to quantify mtDNA copy number relative to the actin gene sense: 5-GAAATCGTGCGTGACATTAAAG-3′, antisense: 5′-ATCGGAACCGCTCATTG-3′.

Measurement of metabolic activity

Oxygen consumption and glycolytic rate were measured using the Seahorse XF-96 instrument (Seahorse Biosciences, North Billerica, MA, USA). Neurons in 96-well Seahorse plates (40 000/well) were incubated in the treatment conditions for 1 and 24 h and then the assay was performed in serum-free unbuffered XF assay medium-containing 1 mM pyruvate and 2 mM glutamax (pH 7.4). Respiration was measured in four consecutive 3-min time periods. The basal respiration rate was measured during the first 3-min period. Next, 1 μM oligomycin was added to evaluate the coupling efficiency. Maximal respiration was then initiated with 1 μM carbonyl cyanide 4-(trifluoromethoxy) phenylhydrazone. Finally, 0.51 μM antimycin and rotenone were added and the last measurements were performed.

Luciferase assay

Cell transfections were performed on HEK293 cells that were maintained in Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum. The cells were co-transfected with 2 μg Bdnf promoter sequences to drive luciferase expression and 2 μg pRL-TK vector expressing Renilla luciferase (Promega) using Fugene 6 (Roche) according to the manufacturer's instructions. Forty-eight h after transfection, cells were incubated in the presence of experimental treatments for 6 h. Luciferase activity was quantified using a Dual-Luciferase-Reporter System (Promega). Luciferase activity in each sample was normalized to the internal control renilla luciferase activity. Luminescence was expressed in an arbitrary scale as relative light units.

ROS and NAD+/NADH measurements

Cellular reactive oxygen species (ROS) were measured in neurons loaded with the fluorescent probe 2, 7-dichlorofluorescein diacetate (DCF), and dihydrorhodamine 123 (DHR123) using methods described previously (Wang and Joseph 1999). In brief, DCF and DHR123 were added to cultures in a final concentration of 5 μM and then incubated at 37°C for 20 min. After washing the cultures with phosphate-buffered saline (PBS), the fluorescent intensity was measured by microplate reader. The excitation and emission wavelengths for DCF were 488 and 510 nm, for DHR:500 and 536 nm. The NAD+/NADH ratio was measured using NAD+/NADH-Glo™ bioluminescent assay using the provided protocol (Promega). The assay uses NAD Cycling Enzyme that converts NAD+ into NADH. In the presence of NADH, a reductase enzymatically reduces a proluciferin reductase substrate to luciferin. The amount of light produced is proportional to the amount of NAD+ and NADH in a sample. Briefly, the neurons were washed with PBS and the pellets were collected for the separate measurements of NAD+ and NADH. The neurons were homogenized in PBS and were split into two samples: One sample is treated with acid (0.4 N HCl) to quantify NAD+, and the other is treated with base to quantify NADH (0.5 M Trizma®base, Sigma-Aldrich, St. Louis, MO. USA). The oxidized form (NAD+) is selectively destroyed by heating in basic solution, while the reduced form (NADH) is not stable in acidic solution. Thus, luminescence from acid-treated samples is proportional to the amount of NAD+, the luminescence from base-treated samples is proportional to the amount of NADH.

Histone acetyltransferase (HAT) activity

The histone acetyltransferase (HAT) activity was measured using the colorimetric HAT activity assay kit (Abcam) according to the manufacturer's instructions. Briefly, neuronal lysates were incubated with HAT substrates and NADH-generating enzyme in HAT assay buffer with the presence and absence of 8 mM 3OHB for 1 h at 37°C. Absorbance was determined at 450 nm in an ELISA plate reader, with active nuclear extract used as a positive control and standard. The HAT activity was normalized to the protein content of the cell lysates.

ATP content

The adenosine triphosphate (ATP) content in the neurons was measured using Molecular Probes® (Eugene, OR USA) ATP Determination Kit bioluminescence according to the manufacturer's instructions. The assay is based on luciferases requirement for ATP in producing light. The assay was performed on a 96-well plate, the number of neurons was 40 000 per well.

Cell viability assay

Cell survival was monitored 24 h after 100 μM kainic acid treatment by the determination of 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) reduction. After 22 h of kainic acid the neurons were incubated with MTS for 1 h. Formazan absorbance was measured spectrophotometrically at 570 nm. Cell viability is expressed as percentage of cell viability relative to the untreated control.

Chromatin immunoprecipitation

ChIP assays were performed using a Pierce Magnetic ChIP Kit according to the manufacturer's instructions. Briefly, rat primary neurons were treated with 3OHB for 6 h followed by fixing in 1% formaldehyde and quenching with glycine. The cells were pelleted, lysed, and subjected to MNase digestion. The lysates were sonicated to shear lengths of 200–1000 base pair DNA fragments using 15 sets of 10 s pulses of sonication. The length of the fragments was verified by running in a 1% agarose gel. Ten percent of the digested chromatin was saved as total input sample and the remainder was incubated overnight under rotation at 4°C with 5 μg of anti-p300 or normal IgG followed by incubation with ChIP Grade Protein A/G Magnetic Beads for 2 h at 4°C with mixing. Beads were then washed and incubated at 65°C for 30 min. A total of 20 mg/mL Proteinase K was added to all IP and total input samples and incubated for 1.5 h at 65°C. Then, the DNA was purified on DNA Clean-UpColumns. DNA pellets were re-suspended in 50 μL of sterile water. The purified DNA was used directly as a template for PCR. The following exon-specific BDNF primers were used in the qPCR reaction: exon I, sense, 5′-TGAGAGCTTGGCTTACACCG-3′, and antisense, 5′-GATGACTAGGCGAGAGGCAC-3′; exon II, sense, 5′-CTGCGTGGAACAAACTTGGG-3′, and antisense, 5′-TTAACCCCCTTGCGGATGTC-3′; exon III, sense, 5′-CGGTGTCGCCCTTAAAAAGC-3′, and antisense, 5′-ACCCAGTATACCAACCCGGA-3′; exon IV, sense, 5′-GCGCGGAATTCTGATTCTGG-3′, and antisense, 5′-CTGCCTTGACGTGAGCTGTC-3′. The qRT-PCR was performed using SYBR GREEN MasterMix (Applied Biosystems). Cycling parameters for all amplifications were as follows: 1 cycle of 95°C for 15 min, 40 cycles of 95°C for 15 s, and 65°C for 1 min, followed by melt-curve analysis (55°C+ for 10 s × 80 cycles). Data were normalized to input and non-specific IgG, and fold increase versus control was calculated.

Plasma chemistry

Glucose and 3-hydroxybutyrate concentrations were quantified in plasma using a Roche Cobas Fara II analyzer (Roche Diagnostic Systems; Montclair, NJ, USA).

Statistical analyses

Data are presented as mean ± SEM. Statistical analyses were performed using one- or two-way anova for comparisons among the different treatment groups at different time points, and Tukey post hoc tests were performed for pairwise comparisons among treatment groups. For comparisons involving only two groups, Student's t-test was performed. Significant differences of p < 0.05 are identified with an asterisk (*p < 0.05, **p < 0.01, ***p < 0.005). All experiments were repeated at least three times; fold changes to the untreated control were calculated and subjected to data analysis.

Results

3OHB is utilized by cortical neurons and increases oxidative metabolism

The primary cortical neurons were incubated in the presence and absence of glucose for 1 h and the basal oxygen consumption rate (OCR) and extracellular acidification rate were measured with Seahorse XF-96 extracellular flux analyzer. Acute, 1 h incubation with 1 mM 3OHB increased the basal oxygen consumption rate (Fig. 1a) and the ATP content (Fig. 1c), although it did not affect the glycolysis in the neurons (Fig. 1b), indicating that 3OHB provides an energy substrate that shifts the neuronal metabolism toward oxidative state.

image

Figure 1

Open in figure viewerPowerPoint

3-hydroxybutyrate (3OHB) increases basal oxygen consumption and ATP production without altering glycolytic rate. (a) 3OHB (1 mM) elevates the basal oxygen consumption rate (OCR) in primary cortical neurons in the presence and absence of 1 mM glucose (**p < 0.01). (b) The extracellular acidification rates (ECAR) were not altered by 1 mM 3OHB. (c) Acute, 1 h incubation with 1 mM 3OHB increases cellular ATP content (*p < 0.05).

3OHB increases metabolic rate by increasing functional changes in the mitochondria

To further examine the effect of 3OHB on neuronal energy metabolism, we measured oxygen consumption in cortical neurons treated with 3OHB for 24 h. After the pre-incubation in the treatment conditions, the metabolic assays were performed in serum-free unbuffered XF assay medium which contained equal amounts of energy substrates in each condition (5 mM glucose, 1 mM pyruvate, and 2 mM glutamax). The basal OCR normalized to neuronal numbers was elevated in the 3OHB-treated neurons (Fig. 2a and b). The ATP production was estimated from the OCR rates following the 1 μM oligomycin treatment. ATP production was higher in the 3OHB-treated neurons, which was confirmed by the direct measurement of cellular ATP content (Fig. 2c). The carbonyl cyanide 4-(trifluoromethoxy) phenylhydrazone (2.5 mM)-stimulated maximal OCR was significantly elevated with 3OHB treatment showing an increase in mitochondrial oxidative capacity (Fig. 2b). The stimulation of maximal respiration by 3OHB was absent in high-glucose conditions (Fig. S1). The NAD+/NADH ratio was significantly increased in 3OHB-treated neurons (Fig. 2d), suggesting enhanced mitochondrial electron transport chain (ETC) activity. Next, we investigated whether the enhanced metabolic rate of 3OHB-treated neurons is related to increased numbers of mitochondria or altered expression of the ETC proteins. We found that the total mtDNA content and the mRNA levels of transcription factors that regulate mitochondrial biogenesis remained unchanged after 3OHB treatment for 24 h (Fig. 2e and f), indicating that 3OHB treatment did not affect the number of mitochondria/neuron. 3OHB-treated neurons exhibited an increase in the level of the mitochondrial complex I protein NDUFB8, but not three other mitochondrial complex proteins, UQCRC2, MTCO1, and succinate dehydrogenase (Fig. 2g), further supporting no effect of 3OHB on mitochondrial numbers. NDUFB8 is an accessory subunit of the mitochondrial membrane respiratory chain NADH dehydrogenase (Complex I). The increase in levels of NDUFB8 in response to 3OHB may therefore lead to a higher NAD+ turnover rate (Fig. 2d) and an increase in ETC-mediated cellular respiration (Fig. 2b).

image

Figure 2

Open in figure viewerPowerPoint

3-hydroxybutyrate (3OHB) stimulates mitochondrial respiration. The neurons were pre-treated with control medium containing 1 mM glucose (CTRL), or medium containing 8 mM 3OHB and 1 mM glucose for 24 h, and then incubated with unbuffered Seahorse XF medium for 1 h. (a) Key parameters of mitochondrial function were determined by measuring the oxygen consumption rate (OCR) of the neurons in the presence of modulators of respiration that target components of the electron transport chain. The compounds [oligomycin, carbonyl cyanide 4-(trifluoromethoxy) phenylhydrazone (FCCP), and a mix of rotenone and antimycin A] were serially applied. The neurons and the recorded OCRs were used to quantify relative levels of ATP production and maximal respiration, respectively. Spare respiratory capacity was calculated using these parameters and basal respiration. Basal and FCCP-induced oxygen consumption rates (OCR) were elevated in primary cortical neurons treated with 8 mM 3OHB for 24 h compared to control cultures (CTRL). Representative graph shows the basal oxygen consumption rates (OCR) and the OCR values after oligomycin (1 μM), FCCP (1 μM), and rotenone plus antimycin A (0.5 μM) treatments measured using a Seahorse XF-96 instrument. p < 0.05 versus CTRL. (b) Values for basal OCR, maximal OCR, spare respiratory capacity (RC), and ATP production in cortical neurons that had been exposed for 24 h to vehicle (CTRL) or 8 mM 3OHB (n = 5 separate cultures). p < 0.05, p < 0.01, p < 0.001 versus CTRL. (c and d) Cortical neurons exhibited higher ATP levels (c) and NAD+/NADH ratio (d) after incubation with 8 mM 3OHB for 6 h compared to CTRL (n = 3 separate cultures). p < 0.05. (e) Results of quantitative polymerase chain reaction (PCR) analysis reveal that 8 mM 3OHB treatment does not affect the expression of genes involved in the regulation of mitochondrial biogenesis (n = 3 separate cultures). (f) Results of quantitative PCR analysis show that the mitochondrial DNA copy number is not affected by 3OHB (24 h treatment with 8 mM 3OHB) (n = 3 separate cultures). Mitochondrial DNA copy number was quantified as described in the Methods section. (g) Immunoblots showing relative levels of the indicated mitochondrial proteins in cortical neurons that were treated with vehicle (CTRL) or 8 mM 3OHB for 24 h.*p < 0.05, **p < 0.01.

3OHB increases Bdnf gene and protein expression

Primary neuronal cultures are typically maintained in medium containing a supraphysiological concentration of glucose (10–25 mM), whereas levels of extracellular glucose in the brain in vivo normally ranges from 0.2 to 2.5 mM in accord with changes in plasma glucose levels from hypoglycemia to hyperglycemic conditions (Silver and Erecinska 1994). We found that when cortical and hippocampal neurons were incubated in medium containing 1 mM glucose and treated for 24 h with 0.1–8 mM 3OHB, a concentration range that includes 3OHB concentrations ranging from those occurring during non-fasting conditions (0.1 mM) to a concentration achieved during extended fasting (8 mM) (Owen et al. 1967), Bdnf mRNA levels were significantly increased by approximately 3-fold within 6 h (Fig. 3a and b, and Fig. S2). In contrast, Bdnf expression was unaffected by 3OHB in cortical neurons incubated in medium containing a high-glucose concentration (10 mM). Similarly, there was a significant increase in the BDNF protein levels in the 3OHB-treated neurons incubated in medium containing 1 mM glucose, but not in those incubated in medium containing 10 mM glucose (Fig. 3b). There were no significant differences in the Bdnf gene expression in neurons incubated in low- compared to high-glucose conditions. (Fig. 3a and b). In a concentration–response study, we found that when cortical neurons were incubated in medium containing 1 mM glucose and treated for 24 h with 0.1–8 mM 3OHB, low concentrations of 3OHB (0.1 and 0.5 mM) had no significant effect on Bdnf mRNA levels, whereas 1 mM 3OHB significantly increased Bdnf mRNA levels (Fig. 3c). Previous studies have shown that, similar to butyrate, 3OHB is an endogenous and specific inhibitor of class I histone deacetylases (Shimazu et al. 2013). We investigated whether sodium–butyrate (Na-butyrate) can similarly regulate the expression of Bdnf. No induction of Bdnf by Na-butyrate was found (Fig. 3d), indicating that changes in Bdnf expression are not driven by the HDAC1 inhibitory properties of 3OHB.

image

Figure 3

Open in figure viewerPowerPoint

3-hydroxybutyrate (3OHB) treatment increases brain-derived neurotrophic factor (BDNF) expression in cerebral cortical neurons. Cortical neurons were incubated in medium containing either a low (1 mM) or high (10 mM) concentration of glucose (GL) and were then exposed to 8 mM 3OHB or vehicle control (CTRL). Neurons were then harvested after either 6 h of 3OHB treatment for measurement of Bdnf mRNA levels (a) or 24 h for measurement of BDNF protein levels (b). Values are the mean and SEM of determinations made in five separate experiments. **p < 0.01, ***p < 0.001. (c) Bdnf gene expression levels were increased by 3OHB in primary cortical neurons in a concentration-dependent manner in 1 mM glucose condition. ***p < 0.001. (d) No induction of Bdnf gene by Na-butyrate (8 mM) was detected in 1 mM glucose condition.*p < 0.05.

The monocarboxylic acid transporter 2 mediates the uptake of 3OHB into neurons (Martin et al. 2006; Chiry et al. 2008). We found that mct2 mRNA levels were increased significantly in response to 3OHB (Fig. 4a). Pre-incubation with an inhibitor of the monocarboxylate transporters [1 μM AR-C155858 (AR-C)] abolished the enhancement in Bdnf mRNA levels by 3OHB (Fig. 4b), consistent with a requirement for cellular uptake of 3OHB in the mechanism by which 3OHB induces BDNF expression.

image

Figure 4

Open in figure viewerPowerPoint

3-hydroxybutyrate (3OHB) treatment increases brain-derived neurotrophic factor (BDNF) expression in cerebral cortical neurons in a MCT2-dependent manner. (a) Cortical neurons were incubated in medium containing 1 mM glucose and were then exposed to 8 mM 3OHB for 6 h. Levels of mct2 mRNA were quantified (n = 5 separate cultures). ***p < 0.001. (b) Cortical neurons (in medium containing 1 mM glucose) were pre-incubated with or without the MCT2 inhibitor AR-C155858 (1 μM) for 1 h. The neurons were then treated with 8 mM 3OHB or vehicle control for 6 h and were then processed for measurement of Bdnf mRNA levels (n = 4 separate cultures). *p < 0.05.

NF-κB mediates 3OHB-induced Bdnf gene transcription

The rat Bdnf gene was reported to have four different promoters that regulate expression of distinct exons (Timmusk et al. 1993) that have been shown to be responsive to various stimuli including electrical activity, exercise, stress, and calcium influx (Shieh and Ghosh 1999; Sakata et al. 2010). To determine if and how 3OHB induces Bdnf gene expression, we transfected HEK293 cells with the luciferase gene driven by Bdnf promoter I, II, III, or IV. We found that 3OHB induces expression from Bdnf promoter IV, but not from promoters I, II, or III (Fig. S3). There was a slight increase in luciferase activity of promoter I as well so it is likely that the promoter I might also contribute to the effect of 3OHB on Bdnf gene expression. Two transcription factors that have been shown to induce Bdnf expression are cAMP response element-binding protein (CREB) and nuclear factor-κB (NF-κB) (Tao et al. 1998; Marini et al. 2004). We tested the phosphorylation of CREB and the nuclear translocation of NF-κB in response to 3OHB treatment. When neurons were treated with 3OHB for 6 h there was an increased amount of the NF-κB subunit p65 in the nucleus compared to vehicle-treated neurons (Fig. 5a). Treatment of neurons with the NF-κB inhibitor SN-50 (10 μM) completely blocked the ability of 3OHB to increase the level of nuclear p65 (Fig. 5a). Inhibition of NF-κB using SN-50 significantly suppressed 3OHB-induced Bdnf gene expression (Fig. 5b).

image

Figure 5

Open in figure viewerPowerPoint

Evidence for the involvement of NF-κB and p300 in 3-hydroxybutyrate (3OHB)-induced Bdnf gene transcription. (a) Cortical neurons were pre-treated for 1 h without or with the NF-κB inhibitor SN50, were then treated with vehicle (CTRL) or 8 mM 3OHB for 6 h, and proteins in nuclei were isolated from the neurons and were subjected to immunoblot analysis with antibodies against the p65 subunit of NF-κB or the nuclear lamin A/B proteins. (b) NF-κB inhibition using 10 μM SN-50 suppressed the induction of Bdnf mRNA expression by 8 mM 3OHB in primary cortical neurons (n = 3 separate cultures). *p < 0.05. (c) Global histone acetyltransferase activity (HAT) was measured in primary cortical neurons at the indicated time points after exposure to vehicle (CTRL) or 8 mM 3OHB (n = 3 separate cultures). *p < 0.05. (d) Cortical neurons were exposed to vehicle (CTRL) or 8 mM 3OHB for 6 h and then the extracts were subjected to immunoprecipitation with an antibody against the p65 NF-κB protein and blotted with p300 antibody. Total NF-κB levels were used as the loading control. (e) Chromatin immunoprecipitation performed on cortical neurons that had been exposed to vehicle (CTRL) or 8 mM 3OHB for 6 h. Bdnf promoter targets (I, II, III, and IV) were evaluated by quantitative polymerase chain reaction (PCR). Quantification of immunoprecipitated material relative to its input level is represented as the fold induction (n = 3 separate cultures). *p < 0.05. Graph B shows that inhibition of p300 blocks the effect of 3OHB on Bdnf expression. Cortical neurons were pre-incubated with or without C646 (2.5 μM), a specific inhibitor of p300, for 1 h and the neurons were then treated with 3OHB or vehicle (CTRL) for 6 h (n = 3 separate cultures), *p < 0.05, **p < 0.01.

p300 interacts with NF-κB to regulate bdnf expression

It was reported that 3OHB can inhibit histone deacetylases (Shimazu et al. 2013) which might alter the dynamic balance between histone acetylation and deacetylation toward acetylation. Because histone acetylation can regulate gene transcription, we evaluated global HAT activity in nuclear extracts from primary neurons that had been exposed to 3OHB for 1 h. Compared to neurons in control cultures, neurons exposed to 3OHB exhibited significantly elevated nuclear HAT activity (Fig. 5c). p300 is a histone acetyltransferase and transcriptional co-activator that can interact with NF-κB to promote neuronal survival (Culmsee et al. 2003; Greene and Chen 2004). A co-immunoprecipitation assay revealed the binding of p300 to p65 in neurons treated with 3OHB for 6 h (Fig. 5d). We next tested the binding of p300 at promoters I–IV of the Bdnf gene. 3OHB significantly enhanced the recruitment of p 300 to the Bdnf promoter IV sequence (Fig. 5e). We then used a specific inhibitor of p300 (C646, 2.5 μM) to demonstrate a requirement for p300 in the stimulation of Bdnf gene expression by 3OHB (Fig 5b).

3OHB enhances mitochondrial oxidative metabolism which plays a role in the regulation of Bdnf gene expression

We next investigated the possible mechanism by which 3OHB triggers NF-κB activation and Bdnf gene transcription. Because metabolism of non-glycolytic substrates is accompanied by elevated levels of mitochondrial ROS (Forsberg et al. 1998), and because NF-κB and p300 are known to be redox-sensitive transcription regulators (Li et al. 1999; Dansen et al. 2009; Chen et al. 2011), we hypothesized that 3OHB-induced Bdnf gene transcription is mediated by mitochondrial ROS production. We found that exposure of cortical neurons to 3OHB resulted in an increase in ROS levels that was evident within 6 h (Fig. 6a and b). Consistent with an adaptive redox response to 3OHB, levels of the antioxidant enzyme SOD2 were elevated in 3OHB-treated neurons (Fig. 6c). Next, we determined whether the suppression of ROS production can affect 3OHB-induced Bdnf expression. We incubated cells with or without 3OHB and an antioxidant for 6 h and then measured Bdnf promoter IV activity. The antioxidants included the glutathione precursor N-acetylcysteine (10 μM) and the superoxide scavenger mitoTempo (5 μM); and cyclosporin A (0.5 μM) that prevents the opening of the mitochondria permeability transition pore in response to toxic insults. The antioxidants prevented the induction of Bdnf gene (Fig. 6d), although cyclosporin A did not have similar effect indicating that 3OHB does not cause alteration in the mitochondria permeability transition pore.

image

Figure 6

Open in figure viewerPowerPoint

Evidence that 3-hydroxybutyrate (3OHB) induces Bdnf expression by a mechanism involving mitochondrial reactive oxygen species. (a and b) Relative levels of reactive oxygen species (ROS) [dichlorofluorescein diacetate (DCF) and dihydrorhodamine 123 (DHR123) fluorescence] in cortical neurons incubated without (CTRL) or with 8 mM 3OHB for 6 h. Values for 3OHB-treated cultures were expressed as fold of the value for CTRL cultures (= 4 separate cultures). *< 0.01. (c) Immunoblots showing relative levels of the SOD2 protein in samples from cultured cortical neurons that had been exposed to 8 mM 3OHB for 24 h. These blots are representative of results obtained in three experiments. (d) Stimulation of Bdnf gene expression by 3OHB is blocked by mitochondrial antioxidants but not cyclosporin A (CysA). Cortical neurons were incubated in medium lacking or containing mitoTempo, N-acetylcistein, and cyclosporin A and were then exposed for 6 h to vehicle (CTRL) or 8 mM 3OHB. Neurons were then harvested and levels of Bdnf mRNA were quantified (= 3 separate cultures), *< 0.05, ***p < 0.001.

BDNF mediates the excitoprotective effect of 3OHB

Next, we investigated the functional role of the induction of BDNF by 3OHB. Neurons were pre-incubated with or without 3OHB for 6 h in the presence or absence of TrkB inhibitor ANA-12 (2 μM). 3OHB treatment slightly increased the survival of the neurons in the presence of kainic acid (Fig. S4). Although there was no difference in the neuronal survival between the groups when the TrkB receptor was blocked indicating that BDNF plays a role in the neuroprotective effect of 3OHB.

Exercise decreases plasma glucose levels, increases 3OHB levels, and induces BDNF in the brain

Aerobic exercise has been known to enhance brain BDNF levels. We investigated whether exercise induces changes in the concentration of the circulating energy sources for the neurons that might be associated with BDNF production in the brain. We tested the levels of the main energy sources, glucose, and 3OHB levels in the sedentary mice and in the mice subjected to 6 weeks of voluntary exercise. We also measured the BDNF levels in the hippocampus. We found that exercise decreased basal glucose levels and increased the 3OHB levels in the plasma (Fig. 7a and b). Hippocampal BDNF levels were also induced by aerobic exercise (Fig. 7c), which corresponds to previous data. In addition, we found a correlation between the circulating levels of 3OHB and the hippocampal BDNF levels, suggesting that 3OHB might regulate BDNF levels in vivo (Fig. S5).

image

Figure 7

Open in figure viewerPowerPoint

Voluntary exercise induces 3-hydroxybutyrate (3OHB) levels in the plasma and brain-derived neurotrophic factor (BDNF) expression in the brain. (a and b). Exercise decreased plasma basal glucose levels (a) and increased 3OHB levels (b). (c) Hippocampal BDNF levels were elevated in mice subjected to aerobic exercise compared to the sedentary mice (C), p < 0.001,***p < 0.001.

Discussion

Our findings suggest that 3OHB induces expression of the Bdnf gene and increases BDNF protein levels in cerebral cortical neurons via activation of the Bdnf gene promoter IV by a mechanism involving the transcription factor NF-κB and the histone acetyltransferase p300. Inhibition of monocarboxylic acid transporter 2 prevented 3OHB-induced Bdnf expression indicating a requirement for cellular uptake of 3OHB for stimulation of Bdnf expression. Cultured primary neurons are typically maintained in media containing concentrations of glucose (10–25 mM) that would be considered pathologically hyperglycemic (i.e., diabetic) in vivo (e.g., Brewer et al. 1993). We found that whereas 3OHB stimulated Bdnf expression in neurons maintained in a relatively low concentration of glucose (1 mM), it did not increase Bdnf expression in neurons maintained in a high concentration of glucose (10 mM). During prolonged fasting, plasma glucose concentrations are maintained low (3–5 mM) while 3OHB concentrations are elevated greatly (5–10 mM). However, under the latter conditions the extracellular glucose concentration in the brain is believed to be considerably lower that plasma glucose concentration (de Vries et al. 2003). Previous studies have shown that BDNF levels are reduced in the hippocampus in rodent models of type 2 diabetes including leptin receptor mutant mice (Stranahan et al. 2009) and rats maintained on a high-fat plus glucose diet (Stranahan et al. 2008). Conversely, intermittent fasting induces BDNF expression in the hippocampus, which may mediate beneficial effects of intermittent fasting on hippocampal neurogenesis, synaptic plasticity, and neuroprotection (Lee et al. 2002; Arumugam et al. 2010). Our findings therefore suggest the possibility that 3OHB contributes to increased BDNF expression in response to fasting.

It was previously reported that NF-κB induces N-methyl-D-apartate receptor-mediated Bdnf gene transcription in cultured cerebellar granule cells (Lipsky et al. 2001; Marini et al. 2004). We found that 3OHB induces the nuclear translocation of the NF-κB p65 subunit and increases the interaction of the transcriptional co-activator p300 with p65 in cortical neurons. Consistent with our findings in neurons, it was recently reported that 3OHB increases NF-κB activation in calf hepatocytes (Shi et al. 2014). We found that cortical neurons treated with 3OHB exhibited enhanced recruitment of p300 to a Bdnf promoter IV sequence, and inhibitors of NF-κB and p300 blocked the ability of 3OHB to induce Bdnf gene expression. Increased mitochondrial ROS generation appears to mediate 3OHB-induced Bdnf expression because we found that agents that reduced mitochondrial superoxide levels prevented 3OHB-induced Bdnf promoter activity. Interestingly, we found that levels of two antioxidant enzymes (SOD2 and heme oxygenase 1) that are encoded by genes previously shown to be induced by NF-κB (Kiningham et al. 2001; Naidu et al. 2008) were increased in 3OHB-treated neurons. We found that 3OHB increased mitochondrial respiration in cortical neurons suggesting that up-regulation of antioxidant defenses in response to 3OHB may represent an adaptive response to cope with elevated mitochondrial ROS generation.

We found that neurons treated with BDNF exhibited an increased mitochondrial respiration rate, and that a BDNF blocking antibody prevented 3OHB-induced increases in mitochondrial respiration. It was previously reported that BDNF can increase mitochondrial respiratory coupling in rat brain mitochondria (Markham et al. 2004) and increases respiratory coupling efficiency in mouse brain synaptosomes (Markham et al. 2012). It has also been shown that when cultured rat cortical neurons are treated with 3OHB, their respiratory capacity increases and they are better able to sustain mitochondrial function when exposed to high levels of glutamate (Laird et al. 2013). Our findings suggest the neuroprotective actions of 3OHB are mediated, at least in part, by BDNF signaling. Numerous studies have shown that fasting is neuroprotective in animal models relevant to AD, PD, and HD (Duan and Mattson 1999; Duan et al. 2003; Halagappa et al., 2007; Griffioen et al. 2013), as well as stroke (Arumugam et al. 2010) and traumatic brain injury (Davis et al. 2008). If and to what extent 3OHB contributes to such neuroprotection remains to be established. However, the ability of ketogenic diets (Gasior et al. 2006; Maalouf et al. 2009) and 3OHB supplementation (Kashiwaya et al. 2013) to protect neurons and improve functional outcome in animal models of neurodegenerative conditions is consistent with an important role for 3OHB in the neuroprotective effects of fasting.

Evolutionary considerations and experimental findings suggest that cognitive function is bolstered by vigorous physical activity and food scarcity/fasting (Mattson 2015). The ability to outsmart one's competitors in the battle for limited food resources has been fundamental to the evolution of the brains of most species including humans. The integration of signaling pathways by which the brain and other organ systems respond adaptively to evolutionarily fundamental bioenergetics challenges are undoubtedly complex involving activation of CNS neural circuits, neuroendocrine pathways, and signals from peripheral organs to the brain. Emerging evidence suggests that BDNF is key mediator of adaptive responses of the brain and peripheral organ systems to bioenergetic challenges (Marosi and Mattson 2014). Our findings suggest a role for 3OHB in up-regulation of BDNF signaling in brain cells in response to exercise and fasting. We found that voluntary exercise increases plasma 3OHB levels and that there is a significant positive correlation between the concentration of circulating 3OHB and levels of BDNF in the hippocampus. Consistent with a role for 3OHB-induced BDNF expression in the neuroprotective effects of fasting and exercise in vivo, it was reported that fasting engages TrkB signaling, promotes neuroplasticity, and improves behavioral recovery after spinal cord injury (Plunet et al., 2008). For example, BDNF signaling in the CNS regulates appetite (Kernie et al. 2000), enhances peripheral insulin sensitivity (Nakagawa et al. 2000), and enhances parasympathetic regulation of heart rate (Wan et al. 2014). Because BDNF expression in multiple brain regions is increased in response to energetic challenges that elevate 3OHB levels, our finding that 3OHB acts directly on neurons to stimulate BDNF expression suggests potential roles for 3OHB in up-regulating BDNF expression under such conditions and could, by this mechanism, contribute to the beneficial effects of fasting and vigorous exercise on cognitive performance, and to improved peripheral energy metabolism and cardiovascular fitness (Wan et al. 2003; van Praag et al. 2014).

Acknowledgments and conflict of interest disclosure

We thank Erez Eitan for providing assistance with some of the experiments. This research was supported, in its entirety, by the Intramural Research Program of the National Institute on Aging. None of the authors have any conflicts of interest to declare.

All experiments were conducted in compliance with the ARRIVE guidelines.

 

REFERENCES

Anson R. M., Guo Z., de Cabo R., Iyun T., Rios M., Hagepanos A., Ingram D. K., Lane M. A. and Mattson M. P. (2003) Intermittent fasting dissociates beneficial effects of dietary restriction on glucose metabolism and neuronal resistance to injury from calorie intake. Proc. Natl Acad. Sci. USA 100, 6216– 6220.

CrossrefCASPubMedWeb of Science®Google Scholar

Arumugam T. V., Phillips T. M., Cheng A., Morrell C. H., Mattson M. P. and Wan R. (2010) Age and energy intake interact to modify cell stress pathways and stroke outcome. Ann. Neurol. 67, 41– 52.

Wiley Online LibraryCASPubMedWeb of Science®Google Scholar

Brewer G. J., Torricelli J. R., Evege E. K. and Price P. J. (1993) Optimized survival of hippocampal neurons in B27-supplemented Neurobasal, a new serum-free medium combination. J. Neurosci. Res. 35, 567– 576.

Wiley Online LibraryGoogle Scholar

Bruce-Keller A. J., Umberger G., McFall R. and Mattson M. P. (1999) Food restriction reduces brain damage and improves behavioral outcome following excitotoxic and metabolic insults. Ann Neurol. 45, 8– 15.

Wiley Online LibraryCASPubMedWeb of Science®Google Scholar

Chao M. V. (2006) Neurotrophin signalling in health and disease. Clin. Sci. (Lond.) 110, 167– 173.

CrossrefCASPubMedWeb of Science®Google Scholar

Chen A. C., Arany P. R., Huang Y. Y. et al. (2011) Low-level laser therapy activates NF-kB via generation of reactive oxygen species in mouse embryonic fibroblasts. PLoS ONE 6, e22453.

CrossrefCASPubMedWeb of Science®Google Scholar

Cheng A., Wan R., Yang J. L., Kamimura N., Son T. G., Ouyang X., Luo Y., Okun E. and Mattson M. P. (2012) Involvement of PGC-1α in the formation and maintenance of neuronal dendritic spines. Nat. Commun. 3, 1250.

CrossrefPubMedWeb of Science®Google Scholar

Cheng A., Yang Y., Zhou Y., Maharana C., Lu D., Peng W., Liu Y., Wan R., Marosi K., Misiak M., Bohr V. A. and Mattson M. P. (2016) Mitochondrial SIRT3 Mediates Adaptive Responses of Neurons to Exercise and Metabolic and Excitatory Challenges. Cell Metab. 23, 128- 142.

CrossrefCASPubMedWeb of Science®Google Scholar

Chiry O., Fishbein W. N., Merezhinskaya N., Clarke S., Galuske R., Magistretti P. J. and Pellerin L. (2008) Distribution of the monocarboxylate transporter MCT2 in human cerebral cortex: an immunohistochemical study. Brain Res. 1226, 61– 69.

CrossrefCASPubMedWeb of Science®Google Scholar

Chowdhury G. M., Jiang L., Rothman D. L. and Behar K. L. (2014) The contribution of ketone bodies to basal and activity-dependent neuronal oxidation in vivo. J. Cereb. Blood Flow Metab. 34, 1233– 1242.

CrossrefCASPubMedWeb of Science®Google Scholar

Culmsee C., Siewe J., Junker V., Retiounskaia M., Schwarz S., Camandola S., El-Metainy S., Behnke H., Mattson M. P. and Krieglstein J. (2003) Reciprocal inhibition of p53 and nuclear factor-kappaB transcriptional activities determines cell survival or death in neurons. J. Neurosci. 23, 8586– 8595.

CrossrefCASPubMedWeb of Science®Google Scholar

Dansen T. B., Smits L. M., vanTriest M. H. et al. Redox-sensitive cysteines bridge p300/CBP-mediated acetylation and FoxO4 activity. Nat. Chem. Biol. 2009; 5: 664– 672.

CrossrefCASPubMedWeb of Science®Google Scholar

Davis L. M., Pauly J. R., Readnower R. D., Rho J. M. and Sullivan P. G. (2008) Fasting is neuroprotective following traumatic brain injury. J. Neurosci. Res. 86, 1812– 1822.

Wiley Online LibraryCASPubMedWeb of Science®Google Scholar

Devi L. (2012) Ohno M 7,8-Dihydroxyflavone, a small-molecule TrkB agonist, reverses memory deficits and BACE1 elevation in a mouse model of Alzheimer's disease. Neuropsychopharmacology 37, 434– 444.

CrossrefCASPubMedWeb of Science®Google Scholar

Duan W. and Mattson M. P. (1999) Dietary restriction and 2-deoxyglucose administration improve behavioral outcome and reduce degeneration of dopaminergic neurons in models of Parkinson's disease. J. Neurosci. Res. 57, 195– 206.

Wiley Online LibraryCASPubMedWeb of Science®Google Scholar

Duan W., Guo Z., Jiang H., Ware M. and Li X. J. (2003) Mattson MP Dietary restriction normalizes glucose metabolism and BDNF levels, slows disease progression, and increases survival in huntingtin mutant mice. Proc. Natl Acad. Sci. USA 100, 2911– 2916.

CrossrefCASPubMedWeb of Science®Google Scholar

Duman C. H., Schlesinger L., Russell D. S. and Duman RS (2008) Voluntary exercise produces antidepressant and anxiolytic behavioral effects in mice. Brain Res. 1199, 148– 158.

CrossrefCASPubMedWeb of Science®Google Scholar

Forsberg H., Eriksson U. J., Melefors O. and Welsh N. (1998) Beta-hydroxybutyrate increases reactive oxygen species in late but not in early postimplantation embryonic cells in vitro. Diabetes 47, 255– 262.

CrossrefCASPubMedWeb of Science®Google Scholar

Gasior M., Rogawski M. A. and Hartman A. L. (2006) Neuroprotective and disease-modifying effects of the ketogenic diet. Behav. Pharmacol. 17, 431– 439.

CrossrefCASPubMedWeb of Science®Google Scholar

Glazner G. W., Chan S. L., Lu C. and Mattson M. P. (2000) Caspase-mediated degradation of AMPA receptor subunits: a mechanism for preventing excitotoxic necrosis and ensuring apoptosis. J. Neurosci. 20, 3641– 3649.

CASPubMedWeb of Science®Google Scholar

Gobbo O. L. and O'Mara SM (2004) Impact of enriched-environment housing on brain-derived neurotrophic factor and on cognitive performance after a transient global ischemia. Behav. Brain Res. 152, 231– 241.

CrossrefCASPubMedWeb of Science®Google Scholar

Greco T., Glenn T. C., Hovda D. A. and Prins M. L. (2015) Ketogenic diet decreases oxidative stress and improves mitochondrial respiratory complex activity. J. Cereb. Blood Flow Metab. 13. pii: 0271678X15610584. [Epub ahead of print].

Google Scholar

Greene W. C. and Chen L. F. (2004) Regulation of NF-kappaB action by reversible acetylation. Novartis Found. Symp. 259, 208– 217.

Wiley Online LibraryCASPubMedGoogle Scholar

Griffioen K. J., Rothman S. M., Ladenheim B., Wan R., Vranis N., Hutchison E., Okun E., Cadet J. L. and Mattson M. P. (2013) Dietary energy intake modifies brainstem autonomic dysfunction caused by mutant α-synuclein. Neurobiol. Aging 34, 928– 935.

CrossrefCASPubMedWeb of Science®Google Scholar

Halagappa V. K., Guo Z., Pearson M., Matsuoka Y., Cutler R. G., Laferla F. M. and Mattson M. P. (2007) Intermittent fasting and caloric restriction ameliorate age-related behavioral deficits in the triple-transgenic mouse model of Alzheimer's disease. Neurobiol Dis. 26, 212- 220.

CrossrefCASPubMedWeb of Science®Google Scholar

Kashiwaya Y., Bergman C., Lee J. H., Wan R., King M. T., Mughal M. R., Okun E., Clarke K., Mattson M. P. and Veech R. L. (2013) A ketone ester diet exhibits anxiolytic and cognition-sparing properties, and lessens amyloid and tau pathologies in a mouse model of Alzheimer's disease. Neurobiol. Aging 34, 1530– 1539.

CrossrefCASPubMedWeb of Science®Google Scholar

Kernie S. G., Liebl D. J. and Parada L. F. (2000) BDNF regulates eating behavior and locomotor activity in mice. EMBO J. 19, 1290– 1300.

Wiley Online LibraryCASPubMedWeb of Science®Google Scholar

Kim do. Y., Simeone K. A., Simeone T. A., Pandya J. D., Wilke J. C., Ahn Y., Geddes J. W., Sullivan P. G. and Rho J. M.. Ketone bodies mediate antiseizure effects through mitochondrial permeability transition. Ann. Neurol. 2015; 78: 77– 87.

Wiley Online LibraryCASWeb of Science®Google Scholar

Kiningham K. K., Xu Y., Daosukho C. and Popova B. (2001) St Clair DK. Nuclear factor kappaB-dependent mechanisms coordinate the synergistic effect of PMA and cytokines on the induction of superoxide dismutase 2. Biochem. J. 353, 147– 156.

CrossrefCASPubMedWeb of Science®Google Scholar

Laird M. D., Clerc P., Polster B. M. and Fiskum G. (2013) Augmentation of normal and glutamate-impaired neuronal respiratory capacity by exogenous alternative biofuels. Transl. Stroke Res. 4, 643– 651.

CrossrefCASPubMedWeb of Science®Google Scholar

Lee J., Duan W. and Mattson M. P. (2002) Evidence that brain-derived neurotrophic factor is required for basal neurogenesis and mediates, in part, the enhancement of neurogenesis by dietary restriction in the hippocampus of adult mice. J. Neurochem. 82, 1367– 1375.

Wiley Online LibraryCASPubMedWeb of Science®Google Scholar

Levivier M., Przedborski S., Bencsics C. and Kang U. J. (1995) Intrastriatal implantation of fibroblasts genetically engineered to produce brain-derived neurotrophic factor prevents degeneration of dopaminergic neurons in a rat model of Parkinson's disease. J. Neurosci. 15, 7810– 7820.

CrossrefCASPubMedWeb of Science®Google Scholar

Li Y. P., Atkins C. M., Sweatt J. D. and Reid M. B. (1999) Mitochondria mediate tumor necrosis factor-alpha/NF-kappaB signaling in skeletal muscle myotubes. Antioxid. Redox Signal. 1, 97– 104.

CrossrefCASPubMedWeb of Science®Google Scholar

Lipsky R. H., Xu K., Zhu D., Kelly C., Terhakopian A., Novelli A. and Marini A. M. (2001) Nuclear factor kappaB is a critical determinant in N-methyl-D-aspartate receptor-mediated neuroprotection. J. Neurochem. 78, 254– 264.

Wiley Online LibraryCASPubMedWeb of Science®Google Scholar

Longo V. D. and Mattson M. P. (2014) Fasting: molecular mechanisms and clinical applications. Cell Metab. 19, 181– 192.

CrossrefCASPubMedWeb of Science®Google Scholar

Maalouf M., Rho J. M. and Mattson M. P. (2009) The neuroprotective properties of calorie restriction, the ketogenic diet, and ketone bodies. Brain Res. Rev. 59, 293– 315.

CrossrefCASPubMedWeb of Science®Google Scholar

Marini A. M., Jiang X., Wu X., Tian F., Zhu D., Okagaki P. and Lipsky R. H. (2004) Role of brain-derived neurotrophic factor andNF-kappaB in neuronal plasticity and survival: from genes to phenotype. Restor Neurol. Neurosci. 22, 121– 130.

CASPubMedWeb of Science®Google Scholar

Markham A., Cameron I., Franklin P. and Spedding M. (2004) BDNF increases rat brain mitochondrial respiratory coupling at complex I, but not complex II. Eur. J. Neurosci. 20, 1189– 1196.

Wiley Online LibraryCASPubMedWeb of Science®Google Scholar

Markham A., Cameron I., Bains R., Franklin P., Kiss J. P., Schwendimann L., Gressens P. and Spedding M. (2012) Brain-derived neurotrophic factor-mediated effects on mitochondrial respiratory coupling and neuroprotection share the same molecular signalling pathways. Eur. J. Neurosci. 35, 366– 374.

Wiley Online LibraryCASPubMedWeb of Science®Google Scholar

Marosi K. and Mattson M. P. (2014) BDNF mediates adaptive brain and body responses to energetic challenges. Trends Endocrinol. Metab. 25, 89– 98.

CrossrefCASPubMedWeb of Science®Google Scholar

Marosi K., Bori Z., Hart N., Sárga L., Koltai E., Radák Z. and Nyakas C. (2012) Long-term exercise treatment reduces oxidative stress in the hippocampus of aging rats. Neuroscience 226, 21– 28.

CrossrefCASPubMedWeb of Science®Google Scholar

Martin P. M., Gopal E., Ananth S., Zhuang L., Itagaki S., Prasad B. M., Smith S. B., Prasad P. D. and Ganapathy V. (2006) Identity of SMCT1 (SLC5A8) as a neuron-specific Na+-coupled transporter for active uptake of L-lactate and ketone bodies in the brain. J. Neurochem. 98, 279– 288.

Wiley Online LibraryCASPubMedWeb of Science®Google Scholar

Mattson M. P. (2012) Energy intake and exercise as determinants of brain health and vulnerability to injury and disease. Cell Metab. 16, 706– 722.

CrossrefCASPubMedWeb of Science®Google Scholar

Mattson M. P. (2015) Lifelong brain health is a lifelong challenge: from evolutionary principles to empirical evidence. Ageing Res. Rev. 20, 37– 45.

CrossrefPubMedWeb of Science®Google Scholar

Mattson M. P., Duan W., Wan Z. and Guo Z. (2004) Prophylactic activation of neuroprotective stress response pathways by dietary and behavioral manipulations. NeuroRx 1, 111– 116.

CrossrefPubMedGoogle Scholar

McNally M. A. and Hartman A. L. (2012) Ketone bodies in epilepsy. J. Neurochem. 121, 28– 35.

Wiley Online LibraryCASPubMedWeb of Science®Google Scholar

Naidu S., Wijayanti N., Santoso S., Kietzmann T. and Immenschuh S. (2008) An atypical NF-kappa B-regulated pathway mediates phorbol ester-dependent heme oxygenase-1 gene activation in monocytes. J. Immunol. 181, 4113– 4123.

CrossrefCASPubMedWeb of Science®Google Scholar

Nakagawa T., Tsuchida A., Itakura Y., Nonomura T., Ono M., Hirota F., Inoue T., Nakayama C., Taiji M. and Noguchi H. (2000) Brain-derived neurotrophic factor regulates glucose metabolism by modulating energy balance in diabetic mice. Diabetes 49, 436– 444.

CrossrefCASPubMedWeb of Science®Google Scholar

Neufer P. D., Bamman M. M., Muoio D. M. et al. (2015) Understanding the cellular and molecular mechanisms of physical activity-induced health benefits. Cell Metab. 22, 4– 11.

CrossrefCASPubMedWeb of Science®Google Scholar

Newman J. C. and Verdin E. (2014) Ketone bodies as signaling metabolites. Trends Endocrinol. Metab. 25, 42– 52.

CrossrefCASPubMedWeb of Science®Google Scholar

Owen O. E., Morgan A. P., Kemp H. G., Sullivan J. M., Herrera M. G. and Cahill G. F. (1967) Brain metabolism during fasting. J. Clin. Invest. 46, 1589– 1595.

CrossrefCASPubMedWeb of Science®Google Scholar

Plunet W. T., Streijger F., Lam C. K., Lee J. H., Liu J. and Tetzlaff W. (2008) Dietary restriction started after spinal cord injury improves functional recovery. Exp Neurol. 213, 28- 35.

CrossrefPubMedWeb of Science®Google Scholar

van Praag H., Fleshner M. and Schwartz M. W. (2014) Mattson MP Exercise, energy intake, glucose homeostasis, and the brain. J. Neurosci. 34, 15139– 15149.

CrossrefCASPubMedWeb of Science®Google Scholar

Prins M. L. and Matsumoto J. H. (2014) The collective therapeutic potential of cerebral ketone metabolism in traumatic brain injury. J. Lipid Res. 55, 2450– 2457.

CrossrefCASPubMedWeb of Science®Google Scholar

Reger M. A., Henderson S. T., Hale C., Cholerton B., Baker L. D., Watson G. S., Hyde K., Chapman D. and Craft S. (2004) Effects of beta-hydroxybutyrate on cognition in memory-impaired adults. Neurobiol. Aging 25, 311– 314.

CrossrefCASPubMedWeb of Science®Google Scholar

Sakata K., Jin L. and Jha S. (2010) Lack of promoter IV-driven BDNF transcription results in depression-like behavior. Genes Brain Behav. 9, 712– 721.

Wiley Online LibraryCASPubMedWeb of Science®Google Scholar

Scheibye-Knudsen M., Mitchell S. J., Fang E. F. et al. (2014) A high-fat diet and NAD(+) activate Sirt1 to rescue premature aging in cockayne syndrome. Cell Metab. 20, 840– 855.

CrossrefCASPubMedWeb of Science®Google Scholar

Shi X., Li X., Li D. et al. (2014) β-Hydroxybutyrate activates the NF-κB signaling pathway to promote the expression of pro-inflammatory factors in calf hepatocytes. Cell. Physiol. Biochem. 33, 920– 932.

CrossrefCASPubMedWeb of Science®Google Scholar

Shieh P. B. and Ghosh A. (1999) Molecular mechanisms underlying activity-dependent regulation of BDNF expression. J. Neurobiol. 41, 127– 134.

Wiley Online LibraryCASPubMedWeb of Science®Google Scholar

Shimazu T., Hirschey M. D., Newman J. et al. (2013) Suppression of oxidative stress by β-hydroxybutyrate, an endogenous histone deacetylase inhibitor. Science 339, 211– 214.

CrossrefCASPubMedWeb of Science®Google Scholar

Silver I. A. and Erecinska M. (1994) Extracellular glucose concentration in mammalian brain: continuous monitoring of changes during increased neuronal activity and upon limitation in oxygen supply in normo-, hypo-, and hyperglycemic animals. J. Neurosci. 14, 5068– 5076.

CrossrefCASPubMedWeb of Science®Google Scholar

Spires T. L., Grote H. E., Varshney N. K., Cordery P. M., van Dellen A. and Hannan AJ (2004) Blakemore C. Environmental enrichment rescues protein deficits in a mouse model of Huntington's disease, indicating a possible disease mechanism. J. Neurosci. 24, 2270– 2276.

CrossrefCASPubMedWeb of Science®Google Scholar

Stranahan A. M., Norman E. D., Lee K., Cutler R. G., Telljohann R. S., Egan J. M. and Mattson M. P. (2008) Diet-induced insulin resistance impairs hippocampal synaptic plasticity and cognition in middle-aged rats. Hippocampus 18, 1085– 1108.

Wiley Online LibraryPubMedWeb of Science®Google Scholar

Stranahan A. M., Lee K., Martin B., Maudsley S., Golden E., Cutler R. G. and Mattson M. P. (2009) Voluntary exercise and caloric restriction enhance hippocampal dendritic spine density and BDNF levels in diabetic mice. Hippocampus 19, 951– 961.

Wiley Online LibraryCASPubMedWeb of Science®Google Scholar

Tao X., Finkbeiner S., Arnold D. B., Shaywitz A. J. and Greenberg M. E. (1998) Ca2 +  influx regulates BDNF transcription by a CREB family transcription factor-dependent mechanism. Neuron 20, 709– 726.

CrossrefCASPubMedWeb of Science®Google Scholar

Timmusk T., Palm K., Metsis M., Reintam T., Paalme V., Saarma M. and Persson H. (1993) Multiple promoters direct tissue-specific expression of the rat BDNF gene. Neuron 10, 475– 489.

CrossrefCASPubMedWeb of Science®Google Scholar

Trejo J. L., Llorens-Martín M. V. and Torres-Alemán I. (2008) The effects of exercise on spatial learning and anxiety-like behavior are mediated by an IGF-I-dependent mechanism related to hippocampal neurogenesis. Mol. Cell Neurosci. 37, 402– 411.

CrossrefCASPubMedWeb of Science®Google Scholar

Vanitallie T. B., Nonas C., Di Rocco A., Boyar K., Hyams K. and Heymsfield S. B. (2005) Treatment of Parkinson disease with diet-induced hyperketonemia: a feasibility study. Neurology 64, 728– 730.

CrossrefCASPubMedWeb of Science®Google Scholar

Voss M. W., Vivar C., Kramer A. F. and van Praag H. (2013) Bridging animal and human models of exercise-induced brain plasticity. Trends Cogn. Sci. 17, 525– 544.

CrossrefPubMedWeb of Science®Google Scholar

de Vries M. G., Arseneau L. M., Lawson M. E. and Beverly J. L. (2003) Extracellular glucose in rat ventromedial hypothalamus during acute and recurrent hypoglycemia. Diabetes 52, 2767– 2773.

CrossrefCASPubMedWeb of Science®Google Scholar

Wan R., Camandola S. and Mattson M. P. (2003) Intermittent food deprivation improves cardiovascular and neuroendocrine responses to stress in rats. J. Nutr. 133, 1921– 1929.

CrossrefCASPubMedWeb of Science®Google Scholar

Wan R., Weigand L. A., Bateman R., Griffioen K., Mendelowitz D. and Mattson M. P. (2014) Evidence that BDNF regulates heart rate by a mechanism involving increased brainstem parasympathetic neuron excitability. J. Neurochem. 129, 573– 580.

Wiley Online LibraryCASPubMedWeb of Science®Google Scholar

Wang H. and Joseph J. A. (1999) Quantifying cellular oxidative stress by dichlorofluorescein assay using microplate reader. Free Radic. Biol. Med. 27, 612– 616.

CrossrefCASPubMedWeb of Science®Google Scholar

Wang R., Li J. J., Diao S. et al. (2013) Metabolic stress modulates Alzheimer's β-secretase gene transcription via SIRT1-PPARγ-PGC-1 in neurons. Cell Metab. 17, 685– 694.

CrossrefCASPubMedWeb of Science®Google Scholar

Wrann C. D., White J. P., Salogiannnis J., Laznik-Bogoslavski D., Wu J., Ma D., Lin J. D., Greenberg M. E. and Spiegelman B. M. (2013) Exercise induces hippocampal BDNF through a PGC-1α/FNDC5 pathway. Cell Metab. 18, 649– 659.

CrossrefCASPubMedWeb of Science®Google Scholar

Yang J. L., Lin Y. T., Chuang P. C., Bohr V. A. and Mattson M. P. (2014) BDNF and exercise enhance neuronal DNA repair by stimulating CREB-mediated production of apurinic/apyrimidinic endonuclease 1. Neuromolecular Med. 16, 161– 174.

CrossrefCASPubMedWeb of Science®Google Scholar

Yin J., Han P., Tang Z., Liu Q. and Shi J. (2015) Sirtuin 3 mediates neuroprotection of ketones against ischemic stroke. J. Cereb. Blood Flow Metab. 35, 1783– 1789.

CrossrefCASPubMedWeb of Science®Google Scholar

Zuccato C. and Cattaneo E. (2009) Brain-derived neurotrophic factor in neurodegenerative diseases. Nat. Rev. Neurol. 5, 311– 322.

CrossrefCASPubMedWeb of Science®Google Scholar

 

References to this article 94 of them as of 9/19/21

Number of times cited according to CrossRef: 94

Giselli Scaini, Taylor Andrews, Camila N.C. Lima, Deborah Benevenuto, Emilio L. Streck, João Quevedo, Mitochondrial dysfunction as a critical event in the pathophysiology of bipolar disorder, Mitochondrion, 10.1016/j.mito.2020.12.002, 57, (23-36), (2021).

Crossref

Maria M. Bayliak, Nadia M. Mosiichuk, Oksana M. Sorochynska, Oksana V. Kuzniak, Lesia O. Sishchuk, Anastasiia O. Hrushchenko, Alina O. Semchuk, Taras V. Pryimak, Yulia V. Vasylyk, Dmytro V. Gospodaryov, Kenneth B. Storey, Olga Garaschuk, Volodymyr I. Lushchak, Middle aged turn point in parameters of oxidative stress and glucose catabolism in mouse cerebellum during lifespan: minor effects of every-other-day fasting, Biogerontology, 10.1007/s10522-021-09918-x, 22, 3, (315-328), (2021).

Crossref

undefined Mahtabuddin, Azmatulla Shaik, Khizer Hussain Afroze M, Rinku Garg, Impact of intermittent fasting on human health, Indian Journal of Clinical Anatomy and Physiology, 10.18231/j.ijcap.2020.081, 7, 4, (388-393), (2021).

Crossref

Scott J. Koppel, Dong Pei, Heather M. Wilkins, Ian W. Weidling, Xiaowan Wang, Blaise W. Menta, Judit Perez‐Ortiz, Anuradha Kalani, Sharon Manley, Lesya Novikova, Devin C. Koestler, Russell H. Swerdlow, A ketogenic diet differentially affects neuron and astrocyte transcription, Journal of Neurochemistry, 10.1111/jnc.15313, 157, 6, (1930-1945), (2021).

Wiley Online Library

Ewa Ogłodek, Wiesław Pilis, Prof., Is Water-Only Fasting Safe?, Global Advances in Health and Medicine, 10.1177/21649561211031178, 10, (216495612110311), (2021).

Crossref

Alinny R. Isaac, Ricardo Lima-Filho, Mychael V. Lourenco, How does the skeletal muscle contribute to brain function?, Neuropharmacology, 10.1016/j.neuropharm.2021.108744, (108744), (2021).

Crossref

Karlien Veldscholte, Arnout B.G. Cramer, Koen F.M. Joosten, Sascha C.A.T. Verbruggen, Intermittent fasting in paediatric critical illness: The properties and potential beneficial effects of an overnight fast in the PICU, Clinical Nutrition, 10.1016/j.clnu.2021.07.030, (2021).

Crossref

Jamasb Joshua Sayadi, Lohrasb Sayadi, Ellen Satteson, Mustafa Chopan, Nerve injury and repair in a ketogenic milieu: A systematic review of traumatic injuries to the spinal cord and peripheral nervous tissue, PLOS ONE, 10.1371/journal.pone.0244244, 16, 1, (e0244244), (2021).

Crossref

Jin Yu, Hong Zhu, Saeid Taheri, William Mondy, Stephen Perry, Mark S. Kindy, Plant-Based Nutritional Supplementation Attenuates LPS-Induced Low-Grade Systemic Activation, International Journal of Molecular Sciences, 10.3390/ijms22020573, 22, 2, (573), (2021).

Crossref

Antonio Paoli, Lorenzo Cenci, PierLuigi Pompei, Nese Sahin, Antonino Bianco, Marco Neri, Massimiliano Caprio, Tatiana Moro, Effects of Two Months of Very Low Carbohydrate Ketogenic Diet on Body Composition, Muscle Strength, Muscle Area, and Blood Parameters in Competitive Natural Body Builders, Nutrients, 10.3390/nu13020374, 13, 2, (374), (2021).

Crossref

Basem H. Elesawy, Bassem M. Raafat, Aya Al Muqbali, Amr M. Abbas, Hussein F. Sakr, The Impact of Intermittent Fasting on Brain-Derived Neurotrophic Factor, Neurotrophin 3, and Rat Behavior in a Rat Model of Type 2 Diabetes Mellitus, Brain Sciences, 10.3390/brainsci11020242, 11, 2, (242), (2021).

Crossref

Patrick C. Bradshaw, Acetyl-CoA Metabolism and Histone Acetylation in the Regulation of Aging and Lifespan, Antioxidants, 10.3390/antiox10040572, 10, 4, (572), (2021).

Crossref

Kévin Nay, William J. Smiles, Jacqueline Kaiser, Luke M. McAloon, Kim Loh, Sandra Galic, Jonathan S. Oakhill, Andrew L. Gundlach, John W. Scott, Molecular Mechanisms Underlying the Beneficial Effects of Exercise on Brain Function and Neurological Disorders, International Journal of Molecular Sciences, 10.3390/ijms22084052, 22, 8, (4052), (2021).

Crossref

Zsolt Kovács, Brigitta Brunner, Csilla Ari, Beneficial Effects of Exogenous Ketogenic Supplements on Aging Processes and Age-Related Neurodegenerative Diseases, Nutrients, 10.3390/nu13072197, 13, 7, (2197), (2021).

Crossref

Alexander Choi, Mark Hallett, Debra Ehrlich, Nutritional Ketosis in Parkinson’s Disease — a Review of Remaining Questions and Insights, Neurotherapeutics, 10.1007/s13311-021-01067-w, (2021).

Crossref

Yixuan Fang, Yue Gu, Chen Zhao, Yaqi Lv, Jiawei Qian, Lingjiang Zhu, Na Yuan, Suping Zhang, Li Wang, Mengli Li, Qing Zhang, Li Xu, Wen Wei, Lei Li, Li Ji, Xueqin Gao, Jingyi Zhang, Yueping Shen, Zixing Chen, Guanghui Wang, Kesheng Dai, Jianrong Wang, Impact of supervised beego, a traditional Chinese water-only fasting, on thrombosis and haemostasis, BMJ Nutrition, Prevention & Health, 10.1136/bmjnph-2020-000183, (bmjnph-2020-000183), (2021).

Crossref

Maria Assunta Potenza, Luca Sgarra, Vanessa Desantis, Carmela Nacci, Monica Montagnani, Diabetes and Alzheimer’s Disease: Might Mitochondrial Dysfunction Help Deciphering the Common Path?, Antioxidants, 10.3390/antiox10081257, 10, 8, (1257), (2021).

Crossref

Jakob Norgren, Makrina Daniilidou, Ingemar Kåreholt, Shireen Sindi, Ulrika Akenine, Karin Nordin, Staffan Rosenborg, Tiia Ngandu, Miia Kivipelto, Anna Sandebring-Matton, Serum proBDNF Is Associated With Changes in the Ketone Body β-Hydroxybutyrate and Shows Superior Repeatability Over Mature BDNF: Secondary Outcomes From a Cross-Over Trial in Healthy Older Adults, Frontiers in Aging Neuroscience, 10.3389/fnagi.2021.716594, 13, (2021).

Crossref

Lirong Hu, Luiz F. Brito, Zaheer Abbas, Abdul Sammad, Ling Kang, Dongsheng Wang, Hongjun Wu, Airong Liu, Guiqiang Qi, Man Zhao, Yachun Wang, Qing Xu, Investigating the Short-Term Effects of Cold Stress on Metabolite Responses and Metabolic Pathways in Inner-Mongolia Sanhe Cattle, Animals, 10.3390/ani11092493, 11, 9, (2493), (2021).

Crossref

Darío García-Rodríguez, Alfredo Giménez-Cassina, Ketone Bodies in the Brain Beyond Fuel Metabolism: From Excitability to Gene Expression and Cell Signaling, Frontiers in Molecular Neuroscience, 10.3389/fnmol.2021.732120, 14, (2021).

Crossref

Shingo Nakajima, Hiroshi Kunugi, Lauric acid promotes neuronal maturation mediated by astrocytes in primary cortical cultures, Heliyon, 10.1016/j.heliyon.2020.e03892, 6, 5, (e03892), (2020).

Crossref

Jeremy J. Walsh, Étienne Myette-Côté, Helena Neudorf, Jonathan P. Little, Potential Therapeutic Effects of Exogenous Ketone Supplementation for Type 2 Diabetes: A Review, Current Pharmaceutical Design, 10.2174/1381612826666200203120540, 26, 9, (958-969), (2020).

Crossref

Guilherme B. Freitas, Mychael V. Lourenco, Fernanda G. De Felice, Protective actions of exercise‐related FNDC5/Irisin in memory and Alzheimer’s disease, Journal of Neurochemistry, 10.1111/jnc.15039, 155, 6, (602-611), (2020).

Wiley Online Library

Teresa Montiel, Luis A. Montes-Ortega, Susana Flores-Yáñez, Lourdes Massieu, Treatment with the Ketone Body D-β-hydroxybutyrate Attenuates Autophagy Activated by NMDA and Reduces Excitotoxic Neuronal Damage in the Rat Striatum In Vivo, Current Pharmaceutical Design, 10.2174/1381612826666200115103646, 26, 12, (1377-1387), (2020).

Crossref

Nicholas G. Norwitz, Javier Gilbert Jaramillo, Kieran Clarke, Adrian Soto, Ketotherapeutics for neurodegenerative diseases, 10.1016/bs.irn.2020.02.003, (2020).

Crossref

Sarah K. Jachim, Ayumi E. Sakamoto, Xu Zhang, Vesselina M. Pearsall, Marissa J. Schafer, Nathan K. LeBrasseur, Harnessing the Effects of Endurance Exercise to Optimize Cognitive Health: Fundamental Insights from Dr. Mark P. Mattson, Ageing Research Reviews, 10.1016/j.arr.2020.101147, (101147), (2020).

Crossref

Madhuvika Murugan, Detlev Boison, Ketogenic Diet, Neuroprotection, and Antiepileptogenesis, Epilepsy Research, 10.1016/j.eplepsyres.2020.106444, (106444), (2020).

Crossref

Jeremy J. Walsh, Étienne Myette-Côté, Jonathan P. Little, The Effect of Exogenous Ketone Monoester Ingestion on Plasma BDNF During an Oral Glucose Tolerance Test, Frontiers in Physiology, 10.3389/fphys.2020.01094, 11, (2020).

Crossref

Marwa Elamin, David N. Ruskin, Paola Sacchetti, Susan A. Masino, A Unifying Mechanism of Ketogenic Diet Action: The Multiple Roles of Nicotinamide Adenine Dinucleotide, Epilepsy Research, 10.1016/j.eplepsyres.2020.106469, (106469), (2020).

Crossref

Shengying Pan, Peili Hu, Qingsheng You, Jinliang Chen, Jingjing Wu, Yaru Zhang, Zixuan Cai, Ting Ye, Xing Xu, Zhuo Chen, Lijuan Tong, Chao Huang, Haiyan He, Evaluation of the antidepressive property of β-hydroxybutyrate in mice, Behavioural Pharmacology, 10.1097/FBP.0000000000000535, 31, 4, (322-332), (2020).

Crossref

Miren Ettcheto, Oriol Busquets, Amanda Cano, Elena Sánchez-Lopez, Patricia R. Manzine, Triana Espinosa-Jimenez, Ester Verdaguer, Francesc X. Sureda, Jordi Olloquequi, Ruben D. Castro-Torres, Carme Auladell, Jaume Folch, Gemma Casadesús, Antoni Camins, Pharmacological Strategies to Improve Dendritic Spines in Alzheimer’s Disease, Journal of Alzheimer's Disease, 10.3233/JAD-201106, (1-17), (2020).

Crossref

Martina-Annette Thaele-Franz, Nico Steckhan, Andreas Michalsen, Rainer Stange, Ketosis in patients undergoing medically supervised therapeutic fasting—results from an observational trial, European Journal of Clinical Nutrition, 10.1038/s41430-020-0694-4, 74, S1, (43-47), (2020).

Crossref

Justyna Mierziak, Wioleta Wojtasik, Anna Kulma, Mariusz Dziadas, Kamil Kostyn, Lucyna Dymińska, Jerzy Hanuza, Magdalena Żuk, Jan Szopa, 3-Hydroxybutyrate Is Active Compound in Flax That Upregulates Genes Involved in DNA Methylation, International Journal of Molecular Sciences, 10.3390/ijms21082887, 21, 8, (2887), (2020).

Crossref

Françoise Wilhelmi de Toledo, Franziska Grundler, Cesare R. Sirtori, Massimiliano Ruscica, Unravelling the health effects of fasting: a long road from obesity treatment to healthy life span increase and improved cognition, Annals of Medicine, 10.1080/07853890.2020.1770849, (1-15), (2020).

Crossref

B. Mahalakshmi, Nancy Maurya, Shin-Da Lee, V. Bharath Kumar, Possible Neuroprotective Mechanisms of Physical Exercise in Neurodegeneration, International Journal of Molecular Sciences, 10.3390/ijms21165895, 21, 16, (5895), (2020).

Crossref

Csilla Ari, Cem Murdun, Craig Goldhagen, Andrew P. Koutnik, Sahil R. Bharwani, David M. Diamond, Mark Kindy, Dominic P. D’Agostino, Zsolt Kovacs, Exogenous Ketone Supplements Improved Motor Performance in Preclinical Rodent Models, Nutrients, 10.3390/nu12082459, 12, 8, (2459), (2020).

Crossref

Shou-Zen Fan, Chih-Wei Sung, Yi-Hsuan Tsai, Sheng-Rong Yeh, Wei-Sheng Lin, Pei-Yu Wang, Nervous System Deletion of Mammalian INDY in Mice Mimics Dietary Restriction-Induced Memory Enhancement, The Journals of Gerontology: Series A, 10.1093/gerona/glaa203, (2020).

Crossref

Carmen Torres-Esquivel, Teresa Montiel, Marco Flores-Méndez, Lourdes Massieu, Effect of β-Hydroxybutyrate on Autophagy Dynamics During Severe Hypoglycemia and the Hypoglycemic Coma, Frontiers in Cellular Neuroscience, 10.3389/fncel.2020.547215, 14, (2020).

Crossref

Nicholas G. Norwitz, David J. Dearlove, Meng Lu, Kieran Clarke, Helen Dawes, Michele T. Hu, A Ketone Ester Drink Enhances Endurance Exercise Performance in Parkinson’s Disease, Frontiers in Neuroscience, 10.3389/fnins.2020.584130, 14, (2020).

Crossref

Lionel Carneiro, Corinne Leloup, Mens sana in corpore sano: Does the Glycemic Index Have a Role to Play?, Nutrients, 10.3390/nu12102989, 12, 10, (2989), (2020).

Crossref

Thomas Ho-yin Lee, Suk-yu Yau, From Obesity to Hippocampal Neurodegeneration: Pathogenesis and Non-Pharmacological Interventions, International Journal of Molecular Sciences, 10.3390/ijms22010201, 22, 1, (201), (2020).

Crossref

Inês Marques-Aleixo, Jorge Beleza, Arnaldina Sampaio, Jelena Stevanovic, Pedro Coxito, Inês Gonçalves, António Ascensão, José Magalhães, Preventive and therapeutic potential of physical exercise in neurodegenerative diseases, Antioxidants & Redox Signaling, 10.1089/ars.2020.8075, (2020).

Crossref

Nancy Tseng, Scott C Lambie, Christopher Q Huynh, Bridget Sanford, Manisha Patel, Paco S Herson, D Ryan Ormond, Mitochondrial transfer from mesenchymal stem cells improves neuronal metabolism after oxidant injury in vitro: The role of Miro1, Journal of Cerebral Blood Flow & Metabolism, 10.1177/0271678X20928147, (0271678X2092814), (2020).

Crossref

Pedro L Valenzuela, Adrián Castillo-García, Javier S Morales, Alejandro Lucia, Perspective: Ketone Supplementation in Sports—Does It Work?, Advances in Nutrition, 10.1093/advances/nmaa130, (2020).

Crossref

Weiliang He, Haoyuan Wang, Congying Zhao, Xiaochao Tian, Litao Li, Hebo Wang, Role of liraglutide in brain repair promotion through Sirt1‐mediated mitochondrial improvement in stroke, Journal of Cellular Physiology, 10.1002/jcp.29204, 235, 3, (2986-3001), (2019).

Wiley Online Library

M.P. Mattson, Excitotoxicity, Stress: Physiology, Biochemistry, and Pathology, 10.1016/B978-0-12-813146-6.00011-4, (125-134), (2019).

Crossref

Mark P. Mattson, An Evolutionary Perspective on Why Food Overconsumption Impairs Cognition, Trends in Cognitive Sciences, 10.1016/j.tics.2019.01.003, (2019).

Crossref

Kun Liu, Fangzhou Li, Qianqian Sun, Ning Lin, Haichao Han, Kaiqiang You, Feng Tian, Zebin Mao, Tingting Li, Tanjun Tong, Meiyu Geng, Yingming Zhao, Wei Gu, Wenhui Zhao, p53 β-hydroxybutyrylation attenuates p53 activity, Cell Death & Disease, 10.1038/s41419-019-1463-y, 10, 3, (2019).

Crossref

Ettore Bergamini, Gabriella Cavallini, Primary Prevention of Alzheimer’s Disease (AD), Geriatric Medicine and Gerontology [Working Title], 10.5772/intechopen.77654, (2019).

Crossref

Susan Westfall, Umar Iqbal, Maria Sebastian, Giulio Maria Pasinetti, Gut microbiota mediated allostasis prevents stress-induced neuroinflammatory risk factors of Alzheimer's disease, 10.1016/bs.pmbts.2019.06.013, (2019).

Crossref

Xin Feng, Han Xu, Jian-Feng Chen, Ling-Yu Ruan, Wen-Long Zhao, Hui-Hui Meng, Wen-Ya Liu, Wen-Li Zhao, Qi Zheng, Zhi-Chao Liu, Zhao-Ben Kan, Ge-Jia Zhong, Jun-Song Wang, Potential hepatoxicity risk of the shell of Herpetospermum caudigerum Wall in rats based on 1H-NMR metabonomics, Journal of Pharmaceutical and Biomedical Analysis, 10.1016/j.jpba.2019.112800, (112800), (2019).

Crossref

Aileen Rivell, Mark P. Mattson, Intergenerational Metabolic Syndrome and Neuronal Network Hyperexcitability in Autism, Trends in Neurosciences, 10.1016/j.tins.2019.08.006, (2019).

Crossref

Sofie Lautrup, David A. Sinclair, Mark P. Mattson, Evandro F. Fang, NAD+ in Brain Aging and Neurodegenerative Disorders, Cell Metabolism, 10.1016/j.cmet.2019.09.001, 30, 4, (630-655), (2019).

Crossref

Lucy Camberos-Luna, Lourdes Massieu, Therapeutic strategies for ketosis induction and their potential efficacy for the treatment of acute brain injury and neurodegenerative diseases, Neurochemistry International, 10.1016/j.neuint.2019.104614, (104614), (2019).

Crossref

Konstantinos I. Avgerinos, Josephine M. Egan, Mark P. Mattson, Dimitrios Kapogiannis, Medium Chain Triglycerides induce mild ketosis and may improve cognition in Alzheimer’s disease. A systematic review and meta-analysis of human studies, Ageing Research Reviews, 10.1016/j.arr.2019.101001, (101001), (2019).

Crossref

Robin Mesnage, Franziska Grundler, Andreas Schwiertz, Yvon Le Maho, Françoise Wilhelmi de Toledo, Changes in human gut microbiota composition are linked to the energy metabolic switch during 10 d of Buchinger fasting, Journal of Nutritional Science, 10.1017/jns.2019.33, 8, (2019).

Crossref

Aftab Ahmed, Farhan Saeed, Muhammad Umair Arshad, Muhammad Afzaal, Ali Imran, Shinawar Waseem Ali, Bushra Niaz, Awais Ahmad, Muhammad Imran, Impact of intermittent fasting on human health: an extended review of metabolic cascades, International Journal of Food Properties, 10.1080/10942912.2018.1560312, 21, 1, (2700-2713), (2019).

Crossref

Françoise Wilhelmi de Toledo, Franziska Grundler, Audrey Bergouignan, Stefan Drinda, Andreas Michalsen, Safety, health improvement and well-being during a 4 to 21-day fasting period in an observational study including 1422 subjects, PLOS ONE, 10.1371/journal.pone.0209353, 14, 1, (e0209353), (2019).

Crossref

Nicholas G. Norwitz, Michele T. Hu, Kieran Clarke, The Mechanisms by Which the Ketone Body D-β-Hydroxybutyrate May Improve the Multiple Cellular Pathologies of Parkinson's Disease, Frontiers in Nutrition, 10.3389/fnut.2019.00063, 6, (2019).

Crossref

Zsolt Kovács, Dominic P. D’Agostino, David Diamond, Mark S. Kindy, Christopher Rogers, Csilla Ari, Therapeutic Potential of Exogenous Ketone Supplement Induced Ketosis in the Treatment of Psychiatric Disorders: Review of Current Literature, Frontiers in Psychiatry, 10.3389/fpsyt.2019.00363, 10, (2019).

Crossref

Huajun Yang, Wei Shan, Fei Zhu, Jianping Wu, Qun Wang, Ketone Bodies in Neurological Diseases: Focus on Neuroprotection and Underlying Mechanisms, Frontiers in Neurology, 10.3389/fneur.2019.00585, 10, (2019).

Crossref

Matthew K. Taylor, Russell H. Swerdlow, Debra K. Sullivan, Dietary Neuroketotherapeutics for Alzheimer’s Disease: An Evidence Update and the Potential Role for Diet Quality, Nutrients, 10.3390/nu11081910, 11, 8, (1910), (2019).

Crossref

Xinhui Li, Zhipeng Zhan, Jingzhu Zhang, Fuyuan Zhou, Li An, β-Hydroxybutyrate Ameliorates Aβ-Induced Downregulation of TrkA Expression by Inhibiting HDAC1/3 in SH-SY5Y Cells, American Journal of Alzheimer's Disease & Other Dementias®, 10.1177/1533317519883496, (153331751988349), (2019).

Crossref

Tyler Hill, John D. Polk, BDNF, endurance activity, and mechanisms underlying the evolution of hominin brains, American Journal of Physical Anthropology, 10.1002/ajpa.23762, 168, S67, (47-62), (2018).

Wiley Online Library

Nina Klimova, Aaron Long, Susana Scafidi, Tibor Kristian, Interplay between NAD+ and acetyl‑CoA metabolism in ischemia-induced mitochondrial pathophysiology, Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease, 10.1016/j.bbadis.2018.09.025, (2018).

Crossref

Owoyele Victor Bamidele, Ayilara Gideon Opeyemi, Coconut oil protects against light-induced retina degeneration in male Wistar rats, Pathophysiology, 10.1016/j.pathophys.2018.10.001, (2018).

Crossref

Rosy Wang, R.M. Damian Holsinger, Exercise-Induced Brain-Derived Neurotrophic Factor Expression: Therapeutic Implications for Alzheimer’s Dementia, Ageing Research Reviews, 10.1016/j.arr.2018.10.002, (2018).

Crossref

Erling Hu, Huan Du, Xinliang Zhu, Leilei Wang, Sen Shang, Xingjuan Wu, Haixia Lu, Xiaoyun Lu, Beta-hydroxybutyrate Promotes the Expression of BDNF in Hippocampal Neurons under Adequate Glucose Supply, Neuroscience, 10.1016/j.neuroscience.2018.06.036, 386, (315-325), (2018).

Crossref

Anna Hernández-Aguilera, Salvador Fernández-Arroyo, Noemí Cabre, Fedra Luciano-Mateo, Gerard Baiges-Gaya, Montserrat Fibla, Vicente Martín-Paredero, Javier A. Menendez, Jordi Camps, Jorge Joven, Plasma Energy-Balance Metabolites Discriminate Asymptomatic Patients with Peripheral Artery Disease, Mediators of Inflammation, 10.1155/2018/2760272, 2018, (1-12), (2018).

Crossref

Joonki Kim, Sung-Wook Kang, Karthik Mallilankaraman, Sang-Ha Baik, James C Lim, Priyanka Balaganapathy, David T She, Ker-Zhing Lok, David Y Fann, Uma Thambiayah, Sung-Chun Tang, Alexis M Stranahan, S Thameem Dheen, Mathias Gelderblom, Raymond C Seet, Vardan T Karamyan, Raghu Vemuganti, Christopher G Sobey, Mark P Mattson, Dong-Gyu Jo, Thiruma V Arumugam, Transcriptome analysis reveals intermittent fasting-induced genetic changes in ischemic stroke, Human Molecular Genetics, 10.1093/hmg/ddy057, 27, 9, (1497-1513), (2018).

Crossref

Udo Bonnet, Dieter Bingmann, Erwin-Josef Speckmann, Martin Wiemann, Small intraneuronal acidification via short-chain monocarboxylates: First evidence of an inhibitory action on over-excited human neocortical neurons, Life Sciences, 10.1016/j.lfs.2018.05.005, 204, (65-70), (2018).

Crossref

Rehana K. Leak, Edward J. Calabrese, Walter J. Kozumbo, Jeffrey M. Gidday, Thomas E. Johnson, James R. Mitchell, C. Keith Ozaki, Reinhard Wetzker, Aalt Bast, Regina G. Belz, Hans E. Bøtker, Sebastian Koch, Mark P. Mattson, Roger P. Simon, Randy L. Jirtle, Melvin E. Andersen, Enhancing and Extending Biological Performance and Resilience, Dose-Response, 10.1177/1559325818784501, 16, 3, (155932581878450), (2018).

Crossref

Mark P. Mattson, Thiruma V. Arumugam, Hallmarks of Brain Aging: Adaptive and Pathological Modification by Metabolic States, Cell Metabolism, 10.1016/j.cmet.2018.05.011, 27, 6, (1176-1199), (2018).

Crossref

Stephen Malunga Manchishi, Ran Ji Cui, Xiao Han Zou, Zi Qian Cheng, Bing jin Li, Effect of caloric restriction on depression, Journal of Cellular and Molecular Medicine, 10.1111/jcmm.13418, 22, 5, (2528-2535), (2018).

Wiley Online Library

Mark P. Mattson, Keelin Moehl, Nathaniel Ghena, Maggie Schmaedick, Aiwu Cheng, Intermittent metabolic switching, neuroplasticity and brain health, Nature Reviews Neuroscience, 10.1038/nrn.2017.156, 19, 2, (63-80), (2018).

Crossref

Yongsheng Lan, Zhaoyuan Huang, Yanjie Jiang, Xuehua Zhou, Jingyu Zhang, Dianyu Zhang, Bo Wang, Guangqing Hou, Strength exercise weakens aerobic exercise-induced cognitive improvements in rats, PLOS ONE, 10.1371/journal.pone.0205562, 13, 10, (e0205562), (2018).

Crossref

Md Mahdi Hasan-Olive, Knut H. Lauritzen, Mohammad Ali, Lene Juel Rasmussen, Jon Storm-Mathisen, Linda H. Bergersen, A Ketogenic Diet Improves Mitochondrial Biogenesis and Bioenergetics via the PGC1α-SIRT3-UCP2 Axis, Neurochemical Research, 10.1007/s11064-018-2588-6, (2018).

Crossref

Anna Ronowska, Andrzej Szutowicz, Hanna Bielarczyk, Sylwia Gul-Hinc, Joanna Klimaszewska-Łata, Aleksandra Dyś, Marlena Zyśk, Agnieszka Jankowska-Kulawy, The Regulatory Effects of Acetyl-CoA Distribution in the Healthy and Diseased Brain, Frontiers in Cellular Neuroscience, 10.3389/fncel.2018.00169, 12, (2018).

Crossref

Mads Svart, Lars C. Gormsen, Jakob Hansen, Dora Zeidler, Michael Gejl, Kim Vang, Joel Aanerud, Niels Moeller, Regional cerebral effects of ketone body infusion with 3-hydroxybutyrate in humans: Reduced glucose uptake, unchanged oxygen consumption and increased blood flow by positron emission tomography. A randomized, controlled trial, PLOS ONE, 10.1371/journal.pone.0190556, 13, 2, (e0190556), (2018).

Crossref

C’iana Cooper, Hyo Youl Moon, Henriette van Praag, On the Run for Hippocampal Plasticity, Cold Spring Harbor Perspectives in Medicine, 10.1101/cshperspect.a029736, 8, 4, (a029736), (2017).

Crossref

Chao Huang, Peng Wang, Xing Xu, Yaru Zhang, Yu Gong, Wenfeng Hu, Minhui Gao, Yue Wu, Yong Ling, Xi Zhao, Yibin Qin, Rongrong Yang, Wei Zhang, The ketone body metabolite β‐hydroxybutyrate induces an antidepression‐associated ramification of microglia via HDACs inhibition‐triggered Akt‐small RhoGTPase activation, Glia, 10.1002/glia.23241, 66, 2, (256-278), (2017).

Wiley Online Library

Stephen D. Anton, Keelin Moehl, William T. Donahoo, Krisztina Marosi, Stephanie A. Lee, Arch G. Mainous, Christiaan Leeuwenburgh, Mark P. Mattson, Flipping the Metabolic Switch: Understanding and Applying the Health Benefits of Fasting, Obesity, 10.1002/oby.22065, 26, 2, (254-268), (2017).

Wiley Online Library

Li Chen, Zhigang Miao, Xingshun Xu, β-hydroxybutyrate alleviates depressive behaviors in mice possibly by increasing the histone3-lysine9-β-hydroxybutyrylation, Biochemical and Biophysical Research Communications, 10.1016/j.bbrc.2017.05.184, 490, 2, (117-122), (2017).

Crossref

Lavanya B. Achanta, Benjamin D. Rowlands, Donald S. Thomas, Gary D. Housley, Caroline D. Rae, β-Hydroxybutyrate Boosts Mitochondrial and Neuronal Metabolism but is not Preferred Over Glucose Under Activated Conditions, Neurochemical Research, 10.1007/s11064-017-2228-6, 42, 6, (1710-1723), (2017).

Crossref

Adele Romano, Justyna Barbara Koczwara, Cristina Anna Gallelli, Daniele Vergara, Maria Vittoria Micioni Di Bonaventura, Silvana Gaetani, Anna Maria Giudetti, Fats for thoughts: An update on brain fatty acid metabolism, The International Journal of Biochemistry & Cell Biology, 10.1016/j.biocel.2016.12.015, 84, (40-45), (2017).

Crossref

Patrycja Puchalska, Peter A. Crawford, Multi-dimensional Roles of Ketone Bodies in Fuel Metabolism, Signaling, and Therapeutics, Cell Metabolism, 10.1016/j.cmet.2016.12.022, 25, 2, (262-284), (2017).

Crossref

Chan Ho Park, Yi-Sub Kwak, Analysis of energy restriction and physical activity on brain function: the role of ketone body and brain-derived neurotrophic factor, Journal of Exercise Rehabilitation, 10.12965/jer.1735028.514, 13, 4, (378-380), (2017).

Crossref

Xue Shao, Yunxuan Tang, Hailei Long, Hui Gu, Jie Zhang, Pengchi Deng, Yinglan Zhao, Xiaobo Cen, HMG-CoA synthase 2 drives brain metabolic reprogramming in cocaine exposure, Neuropharmacology, 10.1016/j.neuropharm.2017.10.001, (2017).

Crossref

Boaz Kim, Alice Brandli, John Mitrofanis, Jonathan Stone, Sivaraman Purushothuman, Daniel M. Johnstone, Remote tissue conditioning — An emerging approach for inducing body-wide protection against diseases of ageing, Ageing Research Reviews, 10.1016/j.arr.2017.05.005, 37, (69-78), (2017).

Crossref

Shunjie Bai, Qingchuan Hu, Zhi Chen, Zihong Liang, Wei Wang, Peng Shen, Ting Wang, Haiyang Wang, Peng Xie, Brain region-specific metabolite networks regulate antidepressant effects of venlafaxine, RSC Adv., 10.1039/C7RA08726H, 7, 73, (46358-46369), (2017).

Crossref

Angélique D. Ducray, Andrea Felser, Jana Zielinski, Aniela Bittner, Julia V. Bürgi, Jean-Marc Nuoffer, Martin Frenz, Meike Mevissen, Effects of silica nanoparticle exposure on mitochondrial function during neuronal differentiation, Journal of Nanobiotechnology, 10.1186/s12951-017-0284-3, 15, 1, (2017).

Crossref

Scott J. Koppel, Russell H. Swerdlow, Neuroketotherapeutics: A modern review of a century-old therapy, Neurochemistry International, 10.1016/j.neuint.2017.05.019, (2017).

Crossref

Milene L. Brownlow, Seung H. Jung, Raquel J. Moore, Naomi Bechmann, Ryan Jankord, Nutritional Ketosis Affects Metabolism and Behavior in Sprague-Dawley Rats in Both Control and Chronic Stress Environments, Frontiers in Molecular Neuroscience, 10.3389/fnmol.2017.00129, 10, (2017).

Crossref

Lavanya B. Achanta, Caroline D. Rae, β-Hydroxybutyrate in the Brain: One Molecule, Multiple Mechanisms, Neurochemical Research, 10.1007/s11064-016-2099-2, 42, 1, (35-49), (2016).

Crossref

Oliver Wendel Holmes Senior said:  If you throw all the medicines in the ocean it would be better for mankind and worse for the fish.  He also wrote:  drugs are what you take while you wait for your body to heal. 
That which made drugs bad in 1885, the profit incentive is still the same, only the percentage take drugs has increased