Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Alzheimers Dement ; 2022 Dec 08.
Article in English | MEDLINE | ID: mdl-36479795

ABSTRACT

Disturbances in the brain's capacity to meet its energy demand increase the risk of synaptic loss, neurodegeneration, and cognitive decline. Nutritional and metabolic interventions that target metabolic pathways combined with diagnostics to identify deficits in cerebral bioenergetics may therefore offer novel therapeutic potential for Alzheimer's disease (AD) prevention and management. Many diet-derived natural bioactive components can govern cellular energy metabolism but their effects on brain aging are not clear. This review examines how nutritional metabolism can regulate brain bioenergetics and mitigate AD risk. We focus on leading mechanisms of cerebral bioenergetic breakdown in the aging brain at the cellular level, as well as the putative causes and consequences of disturbed bioenergetics, particularly at the blood-brain barrier with implications for nutrient brain delivery and nutritional interventions. Novel therapeutic nutrition approaches including diet patterns are provided, integrating studies of the gut microbiome, neuroimaging, and other biomarkers to guide future personalized nutritional interventions.

3.
Eur Geriatr Med ; 12(6): 1285-1292, 2021 12.
Article in English | MEDLINE | ID: mdl-34227053

ABSTRACT

BACKGROUND: Mild cognitive impairment (MCI) is characterized by a decline in cognition and mainly affects older individuals above the age of 60. The global incidence of MCI varies, but it is often underdiagnosed and untreated. There is a distinct lack of approved pharmacologic options to treat MCI. There is, however, evidence to support the efficacy of nutritional interventions, such as ketogenic supplements/diets, which offer ketones as an alternative energy source to brain cells. This article explores the effect of ketones on metabolic activity in the brain and the mechanisms by which ketogenic medium-chain triglycerides (kMCTs) induce ketosis in patients with MCI. KEY TAKEAWAYS: This article reviews the effect of ketogenic supplements/diets on brain metabolism, including evidence supporting the efficacy of ketones as an efficient fuel for the brain. It discusses the use of oral nutritional ketogenic supplements, with particular reference to the 6-month randomized controlled BENEFIC trial, which showed that consumption of a kMCT drink, BrainXpert Energy Complex, improved cognitive performance in individuals with MCI compared with placebo. CONCLUSION: While there is a need for more long-term studies, results from the BENEFIC trial revealed the benefits of a brain-specific ketogenic supplement, as a nutritional intervention, on cognitive performance in individuals with MCI.


Subject(s)
Cognitive Dysfunction , Diet, Ketogenic , Ketosis , Brain/metabolism , Cognition , Cognitive Dysfunction/therapy , Diet, Ketogenic/methods , Humans , Ketosis/metabolism , Randomized Controlled Trials as Topic
4.
PLoS One ; 12(10): e0185926, 2017.
Article in English | MEDLINE | ID: mdl-29016679

ABSTRACT

After advanced age, female sex is the major risk factor for Alzheimer's disease (AD). The biological mechanisms underlying the increased AD risk in women remain largely undetermined. Preclinical studies identified the perimenopause to menopause transition, a neuroendocrine transition state unique to the female, as a sex-specific risk factor for AD. In animals, estrogenic regulation of cerebral glucose metabolism (CMRglc) falters during perimenopause. This is evident in glucose hypometabolism and decline in mitochondrial efficiency which is sustained thereafter. This study bridges basic to clinical science to characterize brain bioenergetics in a cohort of forty-three, 40-60 year-old clinically and cognitively normal women at different endocrine transition stages including premenopause (controls, CNT, n = 15), perimenopause (PERI, n = 14) and postmenopause (MENO, n = 14). All participants received clinical, laboratory and neuropsychological examinations, 18F-fluoro-deoxyglucose (FDG)-Positron Emission Tomography (PET) FDG-PET scans to estimate CMRglc, and platelet mitochondrial cytochrome oxidase (COX) activity measures. Statistical parametric mapping and multiple regression models were used to examine clinical, CMRglc and COX data across groups. As expected, the MENO group was older than PERI and controls. Groups were otherwise comparable for clinical measures and distribution of APOE4 genotype. Both MENO and PERI groups exhibited reduced CMRglc in AD-vulnerable regions which was correlated with decline in mitochondrial COX activity compared to CNT (p's<0.001). A gradient in biomarker abnormalities was most pronounced in MENO, intermediate in PERI, and lowest in CNT (p<0.001). Biomarkers correlated with immediate and delayed memory scores (Pearson's 0.26≤r≤0.32, p≤0.05). These findings validate earlier preclinical findings and indicate emergence of bioenergetic deficits in perimenopausal and postmenopausal women, suggesting that the optimal window of opportunity for therapeutic intervention in women is early in the endocrine aging process.


Subject(s)
Aging/metabolism , Alzheimer Disease/metabolism , Brain/metabolism , Energy Metabolism/physiology , Perimenopause/metabolism , Postmenopause/metabolism , Adult , Aging/pathology , Aging/psychology , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Blood Platelets/metabolism , Blood Platelets/pathology , Brain/diagnostic imaging , Brain/pathology , Brain Mapping , Electron Transport Complex IV/metabolism , Female , Fluorodeoxyglucose F18/administration & dosage , Glucose/metabolism , Humans , Memory/physiology , Middle Aged , Mitochondria/enzymology , Mitochondria/pathology , Neuropsychological Tests , Perimenopause/psychology , Phenotype , Positron-Emission Tomography , Postmenopause/physiology , Radiopharmaceuticals/administration & dosage
5.
Free Radic Biol Med ; 113: 36-47, 2017 12.
Article in English | MEDLINE | ID: mdl-28916476

ABSTRACT

An ascorbate-mediated production of oxidative stress has been shown to retard tumor growth. Subsequent glycolysis inhibition has been suggested. Here, we further define the mechanisms relevant to this observation. Ascorbate was cytotoxic to human neuroblastoma cells through the production of H2O2, which led to ATP depletion, inhibited GAPDH, and non-apoptotic and non-autophagic cell death. The mechanism of cytotoxicity is different when PARP-dependent DNA repair machinery is active or inhibited. Ascorbate-generated H2O2 damaged DNA, activated PARP, depleted NAD+, and reduced glycolysis flux. NAD+ supplementation prevented ATP depletion and cell death, while treatment with a PARP inhibitor, olaparib, preserved NAD+ and ATP levels but led to increased DNA double-strand breakage and did not prevent ascorbate-induced cell death. These data indicate that in cells with an intact PARP-associated DNA repair system, ascorbate-induced cell death is caused by NAD+ and ATP depletion, while in the absence of PARP activation ascorbate-induced cell death still occurs but is a consequence of ROS-induced DNA damage. In a mouse xenograft model, intraperitoneal ascorbate inhibited neuroblastoma tumor growth and prolonged survival. Collectively, these data suggest that ascorbate could be effective in the treatment of glycolysis-dependent tumors. Also, in cancers that use alternative energy metabolism pathways, combining a PARP inhibitor with ascorbate treatment could be useful.


Subject(s)
Ascorbic Acid/toxicity , Cell Death , DNA Damage , Neuroblastoma/metabolism , Oxidative Stress , Poly(ADP-ribose) Polymerases/metabolism , Animals , Ascorbic Acid/pharmacology , DNA/drug effects , DNA/metabolism , DNA Repair , Humans , Hydrogen Peroxide/metabolism , Mice , Neuroblastoma/drug therapy , Neuroblastoma/physiopathology , Xenograft Model Antitumor Assays
6.
J Alzheimers Dis ; 59(1): 291-300, 2017.
Article in English | MEDLINE | ID: mdl-28598847

ABSTRACT

Reductions in bioenergetic fluxes, mitochondrial enzyme activities, and mitochondrial number are observed in Alzheimer's disease (AD). Preclinical work indicates estrogen pathway signaling by either estrogen or selective ß estrogen receptor (ERß) agonists benefits these parameters. To assess whether an ERß agonist could improve mitochondrial function in actual AD subjects, we administered S-equol (10 mg twice daily) to 15 women with AD and determined the platelet mitochondria cytochrome oxidase (COX) activity before initiating S-equol (lead-in), after two weeks of S-equol (active treatment), and two weeks after stopping S-equol (wash-out). Because the intra-individual variation of this enzyme across samples taken at different times was unknown we used a nonparametric, single-arm, dichotomous endpoint that classified subjects whose active treatment COX activity exceeded the average of their lead-in and wash-out measures as positive responders. Eleven positive responses were observed (p < 0.06). The implications of this finding on our null hypothesis (that S-equol does not influence platelet mitochondria COX activity) are discussed. To our knowledge, this is the first time a direct mitochondrial target engagement biomarker has been utilized in an AD clinical study.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Electron Transport Complex IV/metabolism , Equol/administration & dosage , Mitochondria/drug effects , Phytoestrogens/administration & dosage , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Apolipoprotein E4/genetics , Female , Follow-Up Studies , Humans , Middle Aged , Mitochondria/enzymology , Pilot Projects , Treatment Outcome
7.
Oxid Med Cell Longev ; 2017: 9251303, 2017.
Article in English | MEDLINE | ID: mdl-28163822

ABSTRACT

Arsenite is a known carcinogen and its exposure has been implicated in a variety of noncarcinogenic health concerns. Increased oxidative stress is thought to be the primary cause of arsenite toxicity and the toxic effect is thought to be linear with detrimental effects reported at all concentrations of arsenite. But the paradigm of linear dose response in arsenite toxicity is shifting. In the present study we demonstrate that arsenite effects on mitochondrial respiration in primary hepatocytes follow a nonlinear dose response. In vitro exposure of primary hepatocytes to an environmentally relevant, moderate level of arsenite results in increased oxidant production that appears to arise from changes in the expression and activity of respiratory Complex I of the mitochondrial proton circuit. In primary hepatocytes the excess oxidant production appears to elicit adaptive responses that promote resistance to oxidative stress and a propensity to increased proliferation. Taken together, these results suggest a nonlinear dose-response characteristic of arsenite with low-dose arsenite promoting adaptive responses in a process known as mitohormesis, with transient increase in ROS levels acting as transducers of arsenite-induced mitohormesis.


Subject(s)
Arsenites/pharmacology , Hepatocytes/drug effects , Hepatocytes/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Animals , Arsenites/toxicity , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Dose-Response Relationship, Drug , Energy Metabolism , Hep G2 Cells , Humans , Mice , Oxygen Consumption/drug effects , Reactive Oxygen Species/metabolism
8.
J Neurochem ; 139 Suppl 2: 126-135, 2016 10.
Article in English | MEDLINE | ID: mdl-26968700

ABSTRACT

'Metabolism' refers to the vast collection of chemical processes that occur within a living organism. Within this broad designation, one can identify metabolism events that relate specifically to energy homeostasis, whether they occur at the subcellular, cellular, organ, or whole organism level. This review operationally refers to this type of metabolism as 'energy metabolism' or 'bioenergetics.' Changes in energy metabolism/bioenergetics have been linked to brain aging and a number of neurodegenerative diseases, and research suggests mitochondria may uniquely contribute to this. Interventions that manipulate energy metabolism/bioenergetic function and mitochondria may have therapeutic potential and efforts intended to accomplish this are playing out at basic, translational, and clinical levels. This review follows evolving views of energy metabolism's role in neurodegenerative diseases but especially Alzheimer's disease, with an emphasis on the bench-to-bedside process whose ultimate goal is to develop therapeutic interventions. It further considers challenges encountered during this process, which include linking basic concepts to a medical question at the initial research stage, adapting conceptual knowledge gained to a disease-associated application in the translational stage, extending what has been learned to the clinical arena, and maintaining support for the research at each of these fundamentally linked but functionally distinct stages. A bench-to-bedside biomedical research process is discussed that moves through conceptual, basic, translational, and clinical levels. For example, herein a case was made that bioenergetics is a valid Alzheimer's disease therapeutic target. Following this, a fundamental strategy for manipulating bioenergetics was defined, potential implications studied, and the approach extended to the clinical arena. This article is part of the 60th Anniversary special issue.


Subject(s)
Brain/metabolism , Energy Metabolism/physiology , Mitochondria/metabolism , Neurodegenerative Diseases/metabolism , Translational Research, Biomedical/trends , Animals , Brain/pathology , Humans , Mitochondria/pathology , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/therapy , Translational Research, Biomedical/methods
9.
J Neurochem ; 137(1): 76-87, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26811028

ABSTRACT

We tested how the addition of oxaloacetate (OAA) to SH-SY5Y cells affected bioenergetic fluxes and infrastructure, and compared the effects of OAA to malate, pyruvate, and glucose deprivation. OAA displayed pro-glycolysis and pro-respiration effects. OAA pro-glycolysis effects were not a consequence of decarboxylation to pyruvate because unlike OAA, pyruvate lowered the glycolysis flux. Malate did not alter glycolysis flux and reduced mitochondrial respiration. Glucose deprivation essentially eliminated glycolysis and increased mitochondrial respiration. OAA increased, while malate decreased, the cell NAD+/NADH ratio. Cytosolic malate dehydrogenase 1 protein increased with OAA treatment, but not with malate or glucose deprivation. Glucose deprivation increased protein levels of ATP citrate lyase, an enzyme which produces cytosolic OAA, whereas OAA altered neither ATP citrate lyase mRNA nor protein levels. OAA, but not glucose deprivation, increased cytochrome oxidase subunit 2, PGC1α, PGC1ß, and PGC1 related co-activator protein levels. OAA increased total and phosphorylated SIRT1 protein. We conclude that adding OAA to SH-SY5Y cells can support or enhance both glycolysis and respiration fluxes. These effects appear to depend, at least partly, on OAA causing a shift in the cell redox balance to a more oxidized state, that it is not a glycolysis pathway intermediate, and possibly its ability to act in an anaplerotic fashion. We examined how oxaloacetate (OAA) affects bioenergetic fluxes. To advance the understanding of how OAA mediates these changes, we compared the effects of OAA to malate, pyruvate, and glucose deprivation. We further examined how OAA affects levels of enzymes that facilitate its cytosolic metabolism, and found OAA increased the expression of malate dehydrogenase 1 (MDH1-cytosolic). We propose the following: OAA supports both glycolysis and respiration fluxes, shifts the cell redox balance toward a more oxidized state, and acts in an anaplerotic fashion. Abbreviations not defined in the text: MDH2, malate dehydrogenase 2 (mitochondrial).


Subject(s)
Mitochondria/drug effects , Neurons/drug effects , Oxaloacetic Acid/pharmacology , Adenosine Triphosphate/metabolism , Cell Line , Cell Line, Tumor , Cytosol/metabolism , Energy Metabolism/drug effects , Glucose/metabolism , Glucose/pharmacology , Glycolysis/drug effects , Humans , Malate Dehydrogenase/metabolism , Malates/metabolism , Malates/pharmacology , Mitochondria/metabolism , NAD/metabolism , Neuroblastoma/pathology , Neurons/metabolism , Oxygen Consumption , Pyruvic Acid/metabolism , Pyruvic Acid/pharmacology , RNA, Messenger/biosynthesis
10.
Biochem Pharmacol ; 99: 88-100, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26592660

ABSTRACT

Lactate, once considered a metabolic dead-end, has been recently proposed to support neuron bioenergetics. To better understand how lactate specifically influences cell energy metabolism, we studied the effects of lactate supplementation on SH-SY5Y human neuroblastoma cell bioenergetic fluxes. Lactate supplementation increased cell respiration, there was no change in respiratory coupling efficiency, and lactate itself appeared to directly support the respiratory flux increase. Conversely, lactate supplementation reduced the glycolysis flux. This apparent pro-aerobic shift in the respiration:glycolysis ratio was accompanied by post-translational modifications and compartmental redistributions of proteins that respond to and modify bioenergetic fluxes, including cAMP-response element binding protein (CREB), p38 mitogen-activated protein kinases (p38 MAPK), AMP-activated protein kinase (AMPK), peroxisome-proliferator activated receptor gamma coactivator 1 ß (PGC-1ß), Akt, mammalian target of rapamycin (mTOR), and forkhead box protein O1 (FOXO1). mRNA levels for PGC-1ß, nuclear respiratory factor 1 (NRF1), and cytochrome c oxidase subunit 1 (COX1) increased. Some effects depended on the direct presence of lactate, while others were durable and evident several hours after lactate was removed. We conclude lactate can be used to manipulate cell bioenergetics.


Subject(s)
Energy Metabolism/drug effects , Energy Metabolism/physiology , Lactic Acid/pharmacology , Neuroblastoma/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans
11.
J Bioenerg Biomembr ; 47(1-2): 1-11, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25104046

ABSTRACT

Diet composition may affect energy metabolism in a tissue-specific manner. Using C57Bl/6J mice, we tested the effect of ketosis-inducing and non-inducing high fat diets on genes relevant to brain bioenergetic infrastructures, and on proteins that constitute and regulate that infrastructure. At the end of a one-month study period the two high fat diets appeared to differentially affect peripheral insulin signaling, but brain insulin signaling was not obviously altered. Some bioenergetic infrastructure parameters were similarly impacted by both high fat diets, while other parameters were only impacted by the ketogenic diet. For both diets, mRNA levels for CREB, PGC1α, and NRF2 increased while NRF1, TFAM, and COX4I1 mRNA levels decreased. PGC1ß mRNA increased and TNFα mRNA decreased only with the ketogenic diet. Brain mtDNA levels fell in both the ketogenic and non-ketogenic high fat diet groups, although TOMM20 and COX4I1 protein levels were maintained, and mRNA and protein levels of the mtDNA-encoded COX2 subunit were also preserved. Overall, the pattern of changes observed in mice fed ketogenic and non-ketogenic high fat diets over a one month time period suggests these interventions enhance some aspects of the brain's aerobic infrastructure, and may enhance mtDNA transcription efficiency. Further studies to determine which diet effects are due to changes in brain ketone body levels, fatty acid levels, glucose levels, altered brain insulin signaling, or other factors such as adipose tissue-associated hormones are indicated.


Subject(s)
Brain/metabolism , Diet, Ketogenic/adverse effects , Dietary Fats/adverse effects , Energy Metabolism/drug effects , Nerve Tissue Proteins/metabolism , Animals , Brain/pathology , DNA, Mitochondrial/metabolism , Mice , Time Factors
12.
Front Aging Neurosci ; 6: 311, 2014.
Article in English | MEDLINE | ID: mdl-25426068

ABSTRACT

Inflammation is observed in Alzheimer's disease (AD) subject brains. Inflammation-relevant genes are increasingly implicated in AD genetic studies, and inflammatory cytokines to some extent even function as peripheral biomarkers. What underlies AD inflammation is unclear, but no "foreign" agent has been implicated. This suggests that internally produced damage-associated molecular pattern (DAMPs) molecules may drive inflammation in AD. A more complete characterization and understanding of AD-relevant DAMPs could advance our understanding of AD and suggest novel therapeutic strategies. In this review, we consider the possibility that mitochondria, intracellular organelles that resemble bacteria in many ways, trigger and maintain chronic inflammation in AD subjects. Data supporting the possible nexus between AD-associated bioenergetic dysfunction are discussed.

13.
Br J Pharmacol ; 171(8): 1854-69, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24004341

ABSTRACT

Here we discuss a specific therapeutic strategy we call 'bioenergetic medicine'. Bioenergetic medicine refers to the manipulation of bioenergetic fluxes to positively affect health. Bioenergetic medicine approaches rely heavily on the law of mass action, and impact systems that monitor and respond to the manipulated flux. Since classically defined energy metabolism pathways intersect and intertwine, targeting one flux also tends to change other fluxes, which complicates treatment design. Such indirect effects, fortunately, are to some extent predictable, and from a therapeutic perspective may also be desirable. Bioenergetic medicine-based interventions already exist for some diseases, and because bioenergetic medicine interventions are presently feasible, new approaches to treat certain conditions, including some neurodegenerative conditions and cancers, are beginning to transition from the laboratory to the clinic.


Subject(s)
Energy Metabolism/drug effects , Mitochondrial Diseases/drug therapy , Molecular Targeted Therapy/methods , Animals , Energy Metabolism/physiology , Humans , Mitochondrial Diseases/metabolism , Models, Biological , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism
14.
Alzheimers Dement ; 9(4): 452-458.e1, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23809366

ABSTRACT

For decades, researchers have focused primarily on a pathway initiated by amyloid beta aggregation, amyloid deposition, and accumulation in the brain as the key mechanism underlying the disease and the most important treatment target. However, evidence increasingly suggests that amyloid is deposited early during the course of disease, even prior to the onset of clinical symptoms. Thus, targeting amyloid in patients with mild to moderate Alzheimer's disease (AD), as past failed clinical trials have done, may be insufficient to halt further disease progression. Scientists are investigating other molecular and cellular pathways and processes that contribute to AD pathogenesis. Thus, the Alzheimer's Association's Research Roundtable convened a meeting in April 2012 to move beyond amyloid and explore AD as a complex multifactorial disease, with the goal of using a more inclusive perspective to identify novel treatment strategies.


Subject(s)
Alzheimer Disease/drug therapy , Molecular Targeted Therapy , Nootropic Agents/therapeutic use , Aging , Alzheimer Disease/diagnosis , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Animals , Autophagy/drug effects , Biomarkers , Brain/metabolism , Cell Cycle/drug effects , Cooperative Behavior , Diabetes Mellitus, Type 2/complications , Disease Models, Animal , Drug Evaluation, Preclinical , Humans , Inflammation , Insulin Resistance , Lysosomes/drug effects , Lysosomes/physiology , Mice , Mice, Transgenic , Mitochondria/drug effects , Mitochondria/pathology , Models, Neurological , Neuroimaging , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Nootropic Agents/pharmacology , Public-Private Sector Partnerships , Resource Allocation , tau Proteins/drug effects , tau Proteins/physiology
15.
Curr Pharm Des ; 19(34): 6094-111, 2013.
Article in English | MEDLINE | ID: mdl-23448445

ABSTRACT

Slowing aging is a widely shared goal. Plant-derived polyphenols, which are found in commonly consumed food plants such as tea, cocoa, blueberry and grape, have been proposed to have many health benefits, including slowing aging. In-vivo studies have demonstrated the lifespan-extending ability of six polyphenol-containing plants. These include five widely consumed foods (tea, blueberry, cocoa, apple, pomegranate) and a flower commonly used as a folk medicine (betony). These and multiple other plant polyphenols have been shown to have beneficial effects on aging-associated changes across a variety of organisms from worm and fly to rodent and human.


Subject(s)
Aging/drug effects , Disease Models, Animal , Plant Extracts/therapeutic use , Plants, Edible/chemistry , Polyphenols/therapeutic use , Aging/pathology , Animals , Bone Density/drug effects , Cardiovascular Diseases/drug therapy , Cognition Disorders/drug therapy , Humans , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Polyphenols/isolation & purification , Polyphenols/pharmacology
16.
J Alzheimers Dis ; 33(4): 1135-46, 2013.
Article in English | MEDLINE | ID: mdl-23099815

ABSTRACT

Alzheimer's disease (AD) patients have reduced brain acetylcholine and reversing this deficit yields clinical benefits. In this study we explored how increased cholinergic tone impacts cell bioenergetics, which are also perturbed in AD. We treated SH-SY5Y neuroblastoma cells with carbachol, a cholinergic agonist, and tested for bioenergetic flux and bioenergetic infrastructure changes. Carbachol rapidly increased both oxidative phosphorylation and glycolysis fluxes. ATP levels rose slightly, as did cell energy demand, and AMPK phosphorylation occurred. At least some of these effects depended on muscarinic receptor activation, ER calcium release, and ER calcium re-uptake. Our data show that increasing cholinergic signaling enhances cell bioenergetics, and reveal mechanisms that mediate this effect. Phenomena we observed could potentially explain why cholinesterase inhibitor therapy increases AD brain glucose utilization and N-acetyl aspartate levels. The question of whether cholinesterase inhibitors have a disease modifying effect in AD has long been debated; our data suggest a theoretical mechanism through which such an effect could potentially arise.


Subject(s)
Cholinergic Agonists/pharmacology , Energy Metabolism/physiology , Neurons/metabolism , Signal Transduction/physiology , Acetylcholine/metabolism , Carbachol/pharmacology , Cell Line, Tumor , Energy Metabolism/drug effects , Glycolysis/drug effects , Glycolysis/physiology , Humans , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neurons/drug effects , Neurons/pathology , Signal Transduction/drug effects
17.
Biochim Biophys Acta ; 1822(4): 522-6, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22265987

ABSTRACT

Dietary restriction (DR) extends lifespan across multiple species including mouse. Antioxidant plant extracts rich in polyphenols have also been shown to increase lifespan. We hypothesized that polyphenols might potentiate DR-induced lifespan extension. Twenty week old C57BL/6 mice were placed on one of three diets: continuous feeding (control), alternate day chow (Intermittent fed, IF), or IF supplemented with polyphenol antioxidants (PAO) from blueberry, pomegranate, and green tea extracts (IF+PAO). Both IF and IF+PAO groups outlived the control group and the IF+PAO group outlived the IF group (all p<0.001). In the brain, IF induced the expression of inflammatory genes and p38 MAPK phosphorylation, while the addition of PAO reduced brain inflammatory gene expression and p38 MAPK phosphorylation. Our data indicate that while IF overall promotes longevity, some aspects of IF-induced stress may paradoxically lessen this effect. Polyphenol compounds, in turn, may potentiate IF-induced longevity by minimizing specific components of IF-induced cell stress.


Subject(s)
Caloric Restriction , Life Expectancy , Polyphenols/pharmacology , Animals , Female , Male , Mice , Mice, Inbred C57BL
18.
Brain Res ; 1189: 215-8, 2008 Jan 16.
Article in English | MEDLINE | ID: mdl-18061150

ABSTRACT

A study of complex I (NADH:ubiquinone oxidoreductase) activity in Parkinson's disease (PD) brain has identified loss of activity only in substantia nigra although loss of activity of this enzyme has been identified in a number of non-brain tissues. We investigated this paradox by studying complex I and other complexes of the mitochondrial electron transport chain in frontal cortex from PD and aged control brain using a variety of assay conditions and tissue preparations. We found increasingly significant losses of complex I activity in PD frontal cortex as increasingly pure mitochondria were studied. Complexes II, III, and IV were comparable in PD and controls. Inclusion of bovine serum albumin in the assay increased enzyme activity but lessened discrimination between PD and controls. Complex I deficiency in PD brain is not confined to substantia nigra. Methodological issues are critical in demonstrating this loss of activity.


Subject(s)
Brain Diseases, Metabolic/metabolism , Electron Transport Complex I/deficiency , Frontal Lobe/metabolism , Mitochondrial Diseases/complications , Mitochondrial Diseases/metabolism , Parkinson Disease/metabolism , Aged , Aged, 80 and over , Biological Assay/methods , Biological Assay/standards , Biomarkers/analysis , Biomarkers/metabolism , Brain Diseases, Metabolic/physiopathology , Energy Metabolism/physiology , Frontal Lobe/physiopathology , Histocytochemistry/methods , Histocytochemistry/standards , Humans , Mitochondria/metabolism , Mitochondrial Diseases/physiopathology , Neurons/metabolism , Parkinson Disease/physiopathology , Predictive Value of Tests , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL